Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 358
Filter
1.
Nat Immunol ; 18(1): 26-35, 2017 01.
Article in English | MEDLINE | ID: mdl-27893701

ABSTRACT

TRAF1 is a signaling adaptor known for its role in tumor necrosis factor receptor-induced cell survival. Here we show that monocytes from healthy human subjects with a rheumatoid arthritis-associated single-nucleotide polymorphism (SNP) in the TRAF1 gene express less TRAF1 protein but greater amounts of inflammatory cytokines in response to lipopolysaccharide (LPS). The TRAF1 MATH domain binds directly to three components of the linear ubiquitination (LUBAC) complex, SHARPIN, HOIP and HOIL-1, to interfere with the recruitment and linear ubiquitination of NEMO. This results in decreased NF-κB activation and cytokine production, independently of tumor necrosis factor. Consistent with this, Traf1-/- mice show increased susceptibility to LPS-induced septic shock. These findings reveal an unexpected role for TRAF1 in negatively regulating Toll-like receptor signaling, providing a mechanistic explanation for the increased inflammation seen with a disease-associated TRAF1 SNP.


Subject(s)
Arthritis, Rheumatoid/genetics , Leukocytes, Mononuclear/immunology , Monocytes/immunology , Signal Transduction , TNF Receptor-Associated Factor 1/metabolism , Animals , Cytokines/metabolism , Genetic Predisposition to Disease , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide , RNA, Small Interfering/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 1/genetics , Toll-Like Receptors/metabolism
2.
Cell ; 154(4): 859-74, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23953116

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) controls growth and survival in response to metabolic cues. Oxidative stress affects mTORC1 via inhibitory and stimulatory inputs. Whereas downregulation of TSC1-TSC2 activates mTORC1 upon oxidative stress, the molecular mechanism of mTORC1 inhibition remains unknown. Here, we identify astrin as an essential negative mTORC1 regulator in the cellular stress response. Upon stress, astrin inhibits mTORC1 association and recruits the mTORC1 component raptor to stress granules (SGs), thereby preventing mTORC1-hyperactivation-induced apoptosis. In turn, balanced mTORC1 activity enables expression of stress factors. By identifying astrin as a direct molecular link between mTORC1, SG assembly, and the stress response, we establish a unifying model of mTORC1 inhibition and activation upon stress. Importantly, we show that in cancer cells, apoptosis suppression during stress depends on astrin. Being frequently upregulated in tumors, astrin is a potential clinically relevant target to sensitize tumors to apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cytoplasmic Granules/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Oxidative Stress , Regulatory-Associated Protein of mTOR
3.
Nat Immunol ; 15(11): 1079-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25282160

ABSTRACT

Humoral autoimmunity paralleled by the accumulation of follicular helper T cells (T(FH) cells) is linked to mutation of the gene encoding the RNA-binding protein roquin-1. Here we found that T cells lacking roquin caused pathology in the lung and accumulated as cells of the T(H)17 subset of helper T cells in the lungs. Roquin inhibited T(H)17 cell differentiation and acted together with the endoribonuclease regnase-1 to repress target mRNA encoding the T(H)17 cell-promoting factors IL-6, ICOS, c-Rel, IRF4, IκBNS and IκBζ. This cooperation required binding of RNA by roquin and the nuclease activity of regnase-1. Upon recognition of antigen by the T cell antigen receptor (TCR), roquin and regnase-1 proteins were cleaved by the paracaspase MALT1. Thus, this pathway acts as a 'rheostat' by translating TCR signal strength via graded inactivation of post-transcriptional repressors and differential derepression of targets to enhance T(H)17 differentiation.


Subject(s)
Caspases/metabolism , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/immunology , Ribonucleases/metabolism , Th17 Cells/cytology , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Binding Sites/immunology , Cell Differentiation/immunology , Cell Line , Genes, rel/genetics , HEK293 Cells , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Interferon Regulatory Factors/genetics , Interleukin-6/genetics , Intracellular Signaling Peptides and Proteins , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Nuclear Proteins/genetics , Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Sequence Alignment , Th17 Cells/immunology , Ubiquitin-Protein Ligases/genetics
4.
EMBO J ; 40(13): e106777, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33999432

ABSTRACT

The p14ARF protein is a well-known regulator of p53-dependent and p53-independent tumor-suppressive activities. In unstressed cells, p14ARF is predominantly sequestered in the nucleoli, bound to its nucleolar interaction partner NPM. Upon genotoxic stress, p14ARF undergoes an immediate redistribution to the nucleo- and cytoplasm, where it promotes activation of cell cycle arrest and apoptosis. Here, we identify p14ARF as a novel interaction partner and substrate of PRMT1 (protein arginine methyltransferase 1). PRMT1 methylates several arginine residues in the C-terminal nuclear/nucleolar localization sequence (NLS/NoLS) of p14ARF . In the absence of cellular stress, these arginines are crucial for nucleolar localization of p14ARF . Genotoxic stress causes augmented interaction between PRMT1 and p14ARF , accompanied by arginine methylation of p14ARF . PRMT1-dependent NLS/NoLS methylation promotes the release of p14ARF from NPM and nucleolar sequestration, subsequently leading to p53-independent apoptosis. This PRMT1-p14ARF cooperation is cancer-relevant and indicative for PDAC (pancreatic ductal adenocarcinoma) prognosis and chemotherapy response of pancreatic tumor cells. Our data reveal that PRMT1-mediated arginine methylation is an important trigger for p14ARF 's stress-induced tumor-suppressive function.


Subject(s)
Pancreatic Neoplasms/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , HEK293 Cells , HeLa Cells , Humans , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Sf9 Cells , Tumor Suppressor Protein p53/metabolism , Pancreatic Neoplasms
5.
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
6.
Cell ; 136(3): 496-507, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19167051

ABSTRACT

Small regulatory RNAs including small interfering RNAs (siRNAs) and microRNAs (miRNAs) guide Argonaute (Ago) proteins to specific target RNAs leading to mRNA destabilization or translational repression. Here, we report the identification of Importin 8 (Imp8) as a component of miRNA-guided regulatory pathways. We show that Imp8 interacts with Ago proteins and localizes to cytoplasmic processing bodies (P bodies), structures involved in RNA metabolism. Furthermore, we detect Ago2 in the nucleus of HeLa cells, and knockdown of Imp8 reduces the nuclear Ago2 pool. Using immunoprecipitations of Ago2-associated mRNAs followed by microarray analysis, we further demonstrate that Imp8 is required for the recruitment of Ago protein complexes to a large set of Ago2-associated target mRNAs, allowing for efficient and specific gene silencing. Therefore, we provide evidence that Imp8 is required for cytoplasmic miRNA-guided gene silencing and affects nuclear localization of Ago proteins.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , RNA, Messenger/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Argonaute Proteins , Cell Line , Cytoplasmic Granules/metabolism , Gene Silencing , HeLa Cells , Humans , Intranuclear Inclusion Bodies/metabolism , MicroRNAs/metabolism
7.
Development ; 147(21)2020 06 15.
Article in English | MEDLINE | ID: mdl-32376681

ABSTRACT

Cilia are complex cellular protrusions consisting of hundreds of proteins. Defects in ciliary structure and function, many of which have not been characterised molecularly, cause ciliopathies: a heterogeneous group of human syndromes. Here, we report on the FOXJ1 target gene Cfap206, orthologues of which so far have only been studied in Chlamydomonas and Tetrahymena In mouse and Xenopus, Cfap206 was co-expressed with and dependent on Foxj1 CFAP206 protein localised to the basal body and to the axoneme of motile cilia. In Xenopus crispant larvae, the ciliary beat frequency of skin multiciliated cells was enhanced and bead transport across the epidermal mucociliary epithelium was reduced. Likewise, Cfap206 knockout mice revealed ciliary phenotypes. Electron tomography of immotile knockout mouse sperm flagella indicated a role in radial spoke formation reminiscent of FAP206 function in Tetrahymena Male infertility, hydrocephalus and impaired mucociliary clearance of the airways in the absence of laterality defects in Cfap206 mutant mice suggests that Cfap206 may represent a candidate for the subgroup of human primary ciliary dyskinesias caused by radial spoke defects.


Subject(s)
Brain/embryology , Brain/metabolism , Cytoskeletal Proteins/metabolism , Forkhead Transcription Factors/metabolism , Lung/metabolism , Mucociliary Clearance , Sperm Motility , Animals , Axoneme/metabolism , Basal Bodies/metabolism , Cilia/metabolism , Cytoskeletal Proteins/chemistry , Embryonic Development , Epithelial Cells/metabolism , Fluorescence , Hydrocephalus/pathology , Infertility, Male/pathology , Male , Mice, Knockout , Mucus/metabolism , Mutation/genetics , Protein Transport , Spermatozoa/metabolism , Spermatozoa/ultrastructure , Xenopus laevis/embryology , Xenopus laevis/metabolism
8.
Nat Immunol ; 11(8): 725-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20639877

ABSTRACT

The molecular mechanism by which roquin controls the expression of inducible costimulator (ICOS) to prevent autoimmunity remains unsolved. Here we show that in helper T cells, roquin localized to processing (P) bodies and downregulated ICOS expression. The repression was dependent on the RNA helicase Rck, and roquin interacted with Rck and the enhancer of decapping Edc4, which act together in mRNA decapping. Sequences in roquin that confer P-body localization were essential for roquin-mediated ICOS repression. However, this process did not require microRNAs or the RNA-induced silencing complex (RISC). Instead, roquin bound ICOS mRNA directly, showing an intrinsic preference for a previously unrecognized sequence in the 3' untranslated region (3' UTR). Our results support a model in which roquin controls ICOS expression through binding to the 3' UTR of ICOS mRNA and by interacting with proteins that confer post-transcriptional repression.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , DEAD-box RNA Helicases/immunology , MicroRNAs/genetics , Proto-Oncogene Proteins/immunology , RNA, Messenger/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Amino Acid Sequence , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Autoimmunity/genetics , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression Regulation , Inducible T-Cell Co-Stimulator Protein , Mice , Mice, Mutant Strains , Mice, Transgenic , MicroRNAs/immunology , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Ubiquitin-Protein Ligases/immunology
9.
Immunity ; 38(4): 655-68, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23583643

ABSTRACT

The Roquin-1 protein binds to messenger RNAs (mRNAs) and regulates gene expression posttranscriptionally. A single point mutation in Roquin-1, but not gene ablation, increases follicular helper T (Tfh) cell numbers and causes lupus-like autoimmune disease in mice. In T cells, we did not identify a unique role for the much lower expressed paralog Roquin-2. However, combined ablation of both genes induced accumulation of T cells with an effector and follicular helper phenotype. We showed that Roquin-1 and Roquin-2 proteins redundantly repressed the mRNA of inducible costimulator (Icos) and identified the Ox40 costimulatory receptor as another shared mRNA target. Combined acute deletion increased Ox40 signaling, as well as Irf4 expression, and imposed Tfh differentiation on CD4(+) T cells. These data imply that both proteins maintain tolerance by preventing inappropriate T cell activation and Tfh cell differentiation, and that Roquin-2 compensates in the absence of Roquin-1, but not in the presence of its mutated form.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/metabolism , RNA, Messenger/metabolism , Receptors, OX40/metabolism , Repressor Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , CD4 Antigens/metabolism , Cell Differentiation/genetics , HEK293 Cells , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Protein Binding , Receptors, OX40/genetics , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
10.
Mol Cell ; 56(5): 630-40, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25454947

ABSTRACT

Proteolysis by aspartyl intramembrane proteases such as presenilin and signal peptide peptidase (SPP) underlies many cellular processes in health and disease. Saccharomyces cerevisiae encodes a homolog that we named yeast presenilin fold 1 (Ypf1), which we verify to be an SPP-type protease that localizes to the endoplasmic reticulum (ER). Our work shows that Ypf1 functionally interacts with the ER-associated degradation (ERAD) factors Dfm1 and Doa10 to regulate the abundance of nutrient transporters by degradation. We demonstrate how this noncanonical branch of the ERAD pathway, which we termed "ERAD regulatory" (ERAD-R), responds to ligand-mediated sensing as a trigger. More generally, we show that Ypf1-mediated posttranslational regulation of plasma membrane transporters is indispensible for early sensing and adaptation to nutrient depletion. The combination of systematic analysis alongside mechanistic details uncovers a broad role of intramembrane proteolysis in regulating secretome dynamics.


Subject(s)
Endoplasmic Reticulum/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Cell Membrane/metabolism , Endoplasmic Reticulum-Associated Degradation , Gene Expression Regulation, Fungal , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Phylogeny , Saccharomyces cerevisiae/physiology , Sequence Alignment , Ubiquitin-Protein Ligases/metabolism , Zinc/metabolism
11.
Dev Biol ; 459(2): 109-125, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31884020

ABSTRACT

Malfunctions of motile cilia cause a variety of developmental defects and diseases in humans and animal model organisms. Defects include impaired mucociliary clearance of the airways, sperm immotility, hydrocephalus and organ laterality. Here, we characterize the evolutionary conserved Cfap43 gene by loss-of-function experiments in the mouse and the frog Xenopus laevis. Cfap43 is expressed in tissues carrying motile cilia and acts as a target gene of the transcription factor FOXJ1, which is essential for the induction of motile ciliogenesis. We show that CFAP43, a protein of unknown biochemical function, localizes to the ciliary axoneme. CFAP43 is involved in the regulation of the beating frequency of tracheal cilia and loss of CFAP43 causes severe mucus accumulation in the nasal cavity. Likewise, morphant and crispant frog embryos revealed impaired function of motile cilia of the larval epidermis, a model for airway mucociliary epithelia. CFAP43 participates in the formation of flagellar axonemes during spermatogenesis as mice mutant for Cfap43 display male infertility, consistent with observations in male sterile patients. In addition, mice mutant for Cfap43 display early onset hydrocephalus. Together, these results confirm the role of CFAP43 in the male reproductive tract and pinpoint additional functions in airway epithelia mucus clearance and brain development.


Subject(s)
Cilia/metabolism , Cytoskeletal Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Cytoskeletal Proteins/genetics , Epidermal Cells/metabolism , Forkhead Transcription Factors/metabolism , Hydrocephalus/genetics , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Sperm Tail/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Trachea/cytology , Xenopus Proteins/genetics , Xenopus laevis
12.
PLoS Pathog ; 15(5): e1007743, 2019 05.
Article in English | MEDLINE | ID: mdl-31059555

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) belongs to the subfamily of Gammaherpesvirinae and is the etiological agent of Kaposi's sarcoma as well as of two lymphoproliferative diseases: primary effusion lymphoma and multicentric Castleman disease. The KSHV life cycle is divided into a latent and a lytic phase and is highly regulated by viral immunomodulatory proteins which control the host antiviral immune response. Among them is a group of proteins with homology to cellular interferon regulatory factors, the viral interferon regulatory factors 1-4. The KSHV vIRFs are known as inhibitors of cellular interferon signaling and are involved in different oncogenic pathways. Here we characterized the role of the second vIRF protein, vIRF2, during the KSHV life cycle. We found the vIRF2 protein to be expressed in different KSHV positive cells with early lytic kinetics. Importantly, we observed that vIRF2 suppresses the expression of viral early lytic genes in both newly infected and reactivated persistently infected endothelial cells. This vIRF2-dependent regulation of the KSHV life cycle might involve the increased expression of cellular interferon-induced genes such as the IFIT proteins 1, 2 and 3, which antagonize the expression of early KSHV lytic proteins. Our findings suggest a model in which the viral protein vIRF2 allows KSHV to harness an IFN-dependent pathway to regulate KSHV early gene expression.


Subject(s)
Endothelium, Vascular/virology , Gene Expression Regulation, Viral , Herpesvirus 8, Human/physiology , Immediate-Early Proteins/metabolism , Interferon Regulatory Factors/metabolism , Interferons/metabolism , Sarcoma, Kaposi/virology , Viral Proteins/metabolism , Virus Activation , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Immediate-Early Proteins/genetics , Interferon Regulatory Factors/genetics , Interferons/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/metabolism , Viral Proteins/genetics , Virus Latency
13.
Nature ; 526(7573): 443-7, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26322584

ABSTRACT

Alzheimer disease (AD) is characterized by the accumulation of amyloid plaques, which are predominantly composed of amyloid-ß peptide. Two principal physiological pathways either prevent or promote amyloid-ß generation from its precursor, ß-amyloid precursor protein (APP), in a competitive manner. Although APP processing has been studied in great detail, unknown proteolytic events seem to hinder stoichiometric analyses of APP metabolism in vivo. Here we describe a new physiological APP processing pathway, which generates proteolytic fragments capable of inhibiting neuronal activity within the hippocampus. We identify higher molecular mass carboxy-terminal fragments (CTFs) of APP, termed CTF-η, in addition to the long-known CTF-α and CTF-ß fragments generated by the α- and ß-secretases ADAM10 (a disintegrin and metalloproteinase 10) and BACE1 (ß-site APP cleaving enzyme 1), respectively. CTF-η generation is mediated in part by membrane-bound matrix metalloproteinases such as MT5-MMP, referred to as η-secretase activity. η-Secretase cleavage occurs primarily at amino acids 504-505 of APP695, releasing a truncated ectodomain. After shedding of this ectodomain, CTF-η is further processed by ADAM10 and BACE1 to release long and short Aη peptides (termed Aη-α and Aη-ß). CTFs produced by η-secretase are enriched in dystrophic neurites in an AD mouse model and in human AD brains. Genetic and pharmacological inhibition of BACE1 activity results in robust accumulation of CTF-η and Aη-α. In mice treated with a potent BACE1 inhibitor, hippocampal long-term potentiation was reduced. Notably, when recombinant or synthetic Aη-α was applied on hippocampal slices ex vivo, long-term potentiation was lowered. Furthermore, in vivo single-cell two-photon calcium imaging showed that hippocampal neuronal activity was attenuated by Aη-α. These findings not only demonstrate a major functionally relevant APP processing pathway, but may also indicate potential translational relevance for therapeutic strategies targeting APP processing.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Hippocampus/cytology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Neurons/physiology , Proteolysis , ADAM Proteins/metabolism , ADAM10 Protein , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Calcium Signaling , Disease Models, Animal , Female , Hippocampus/enzymology , Hippocampus/physiology , Humans , In Vitro Techniques , Long-Term Potentiation , Male , Matrix Metalloproteinases, Membrane-Associated/deficiency , Membrane Proteins/metabolism , Mice , Molecular Weight , Neurites/enzymology , Neurites/metabolism , Neurons/enzymology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plaque, Amyloid , Protein Processing, Post-Translational , Single-Cell Analysis
14.
Mol Cell ; 50(2): 236-49, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23562326

ABSTRACT

Centromere clustering during interphase is a phenomenon known to occur in many different organisms and cell types, yet neither the factors involved nor their physiological relevance is well understood. Using Drosophila tissue culture cells and flies, we identified a network of proteins, including the nucleoplasmin-like protein (NLP), the insulator protein CTCF, and the nucleolus protein Modulo, to be essential for the positioning of centromeres. Artificial targeting further demonstrated that NLP and CTCF are sufficient for clustering, while Modulo serves as the anchor to the nucleolus. Centromere clustering was found to depend on centric chromatin rather than specific DNA sequences. Moreover, unclustering of centromeres results in the spatial destabilization of pericentric heterochromatin organization, leading to partial defects in the silencing of repetitive elements, defects during chromosome segregation, and genome instability.


Subject(s)
Cell Nucleolus/metabolism , Centromere/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nucleoplasmins/metabolism , Animals , CCCTC-Binding Factor , Cell Line , Chromosomes, Insect , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Gene Knockdown Techniques , Gene Silencing , Genomic Instability , Hemocytes/metabolism , Heterochromatin/metabolism , Histones/metabolism , Interphase , Nucleoplasmins/genetics , Protein Binding , Protein Interaction Maps , Protein Stability , Protein Transport , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism
15.
Mol Cell ; 49(4): 668-79, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23317503

ABSTRACT

The HIV Nef protein recruits the polycomb protein Eed and mimics an integrin receptor signal for reasons that are not entirely clear. Here we demonstrate that Nef and Eed complex with the integrin effector paxillin to recruit and activate TNFα converting enzyme (TACE alias ADAM 17) and its close relative ADAM10. The activated proteases cleaved proTNFα and were shuttled into extracellular vesicles (EVs). Peripheral blood mononuclear cells that ingested these EVs released TNFα. Analyzing the mechanism, we found that Pak2, an established host cell effector of Nef, phosphorylated paxillin on Ser272/274 to induce TACE-paxillin association and shuttling into EVs via lipid rafts. Conversely, Pak1 phosphorylated paxillin on Ser258, which inhibited TACE association and lipid raft transfer. Interestingly, melanoma cells used an identical mechanism to shuttle predominantly ADAM10 into EVs. We conclude that HIV-1 and cancer cells exploit a paxillin/integrin-controlled mechanism to release TACE/ADAM10-containing vesicles, ensuring better proliferation/growth conditions in their microenvironment.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Paxillin/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology , p21-Activated Kinases/physiology , ADAM Proteins/blood , ADAM10 Protein , ADAM17 Protein , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Amyloid Precursor Protein Secretases/blood , Case-Control Studies , Enzyme Activation , HEK293 Cells , HIV Infections/blood , HIV Infections/enzymology , Heterogeneous-Nuclear Ribonucleoprotein K , Humans , Melanoma/blood , Melanoma/enzymology , Membrane Microdomains/enzymology , Membrane Proteins/blood , Mutagenesis, Site-Directed , Paxillin/genetics , Paxillin/metabolism , Phosphorylation , Polycomb Repressive Complex 2/metabolism , Protein Binding , Protein Kinase C-delta/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Protein Transport , Ribonucleoproteins/metabolism , Secretory Vesicles/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , p21-Activated Kinases/metabolism
16.
Nucleic Acids Res ; 47(14): 7444-7459, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31147711

ABSTRACT

Preblastoderm Drosophila embryo development is characterized by fast cycles of nuclear divisions. Extracts from these embryos can be used to reconstitute complex chromatin with high efficiency. We now discovered that this chromatin assembly system contains activities that recognize unprotected DNA ends and signal DNA damage through phosphorylation. DNA ends are initially bound by Ku and MRN complexes. Within minutes, the phosphorylation of H2A.V (homologous to γH2A.X) initiates from DNA breaks and spreads over tens of thousands DNA base pairs. The γH2A.V phosphorylation remains tightly associated with the damaged DNA and does not spread to undamaged DNA in the same reaction. This first observation of long-range γH2A.X spreading along damaged chromatin in an in vitro system provides a unique opportunity for mechanistic dissection. Upon further incubation, DNA ends are rendered single-stranded and bound by the RPA complex. Phosphoproteome analyses reveal damage-dependent phosphorylation of numerous DNA-end-associated proteins including Ku70, RPA2, CHRAC16, the exonuclease Rrp1 and the telomer capping complex. Phosphorylation of spindle assembly checkpoint components and of microtubule-associated proteins required for centrosome integrity suggests this cell-free system recapitulates processes involved in the regulated elimination of fatally damaged syncytial nuclei.


Subject(s)
Cell-Free System/metabolism , DNA Breaks , Drosophila/genetics , Signal Transduction , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA Repair , Drosophila/cytology , Drosophila/embryology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Histones/genetics , Histones/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Phosphorylation , Proteome/genetics , Proteome/metabolism , Proteomics/methods
17.
Int J Cancer ; 146(5): 1396-1408, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31525266

ABSTRACT

Chitinase-like proteins (CLP) are chitin-binding proteins that lack chitin hydrolyzing activity, but possess cytokine-like and growth factor-like properties, and play crucial role in intercellular crosstalk. Both human and mice express two members of CLP family: YKL-40 and stabilin-1 interacting chitinase-like protein (SI-CLP). Despite numerous reports indicating the role of YKL-40 in the support of angiogenesis, tumor cell proliferation, invasion and metastasis, the role of its structurally related protein SI-CLP in cancer was not reported. Using gain-of-function approach, we demonstrate in the current study that the expression of recombinant SI-CLP in mouse TS/A mammary adenocarcinoma cells results in significant and persistent inhibition of in vivo tumor growth. Using quantitative immunohistochemistry, we show that on the cellular level this phenomenon is associated with reduced infiltration of tumor-associated macrophages (TAMs), CD4+ and FoxP3+ cells in SI-CLP expressing tumors. Gene expression analysis in TAM isolated from SI-CLP-expressing and control tumors demonstrated that SI-CLP does not affect macrophage phenotype. However, SI-CLP significantly inhibited migration of murine bone-marrow derived macrophages and human primary monocytes toward monocyte-recruiting chemokine CCL2 produced in the tumor microenvironment (TME). Mechanistically, SI-CLP did not affect CCL2/CCR2 interaction, but suppressed cytoskeletal rearrangements in response to CCL2. Altogether, our data indicate that SI-CLP functions as a tumor growth inhibitor in mouse breast cancer by altering cellular composition of TME and blocking cytokine-induced TAM recruitment. Taking into consideration weak to absent expression of SI-CLP in human breast cancer, it can be considered as a therapeutic protein to block TAM-mediated support of breast tumor growth.


Subject(s)
Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Growth Processes/immunology , Cell Movement/immunology , Female , HEK293 Cells , Humans , Macrophage Activation , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Middle Aged
18.
Immunity ; 35(6): 945-57, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195748

ABSTRACT

Little is known about mechanisms determining the homeostasis of lymphocytes within lymphoid organs. Applying different mouse models, including conditionally proficient Ccr7 gene-targeted mice, we now show that semimature steady state dendritic cells (sDCs) constitutively trafficking into lymph nodes (LNs) were essential contributors to T cell homeostasis in these organs. sDCs provided vascular endothelial growth factor known to support high endothelial venule formation, thus facilitating enhanced homing of T cells to LNs. The presence of sDCs led to increased CCL21 production in T-zone fibroblastic reticular cells. CCL21 is a ligand for CCR7 known to regulate homing as well as retention of T cells in LNs. In addition, we provide evidence that CCL21 binds to the surface of DCs via its heparin-binding domain, further explaining why T cells leave LNs more rapidly in the absence of sDCs. Together, these data reveal multiple roles for sDCs in regulating T cell homeostasis in LNs.


Subject(s)
Dendritic Cells/immunology , Lymph Nodes/immunology , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/metabolism , Cell Movement/immunology , Chemokine CCL21/metabolism , Chimerism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dendritic Cells/metabolism , Gene Targeting , Homeostasis/genetics , Homeostasis/immunology , Humans , Lymph Nodes/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Stromal Cells/metabolism , T-Lymphocytes/metabolism
19.
J Immunol ; 200(2): 558-564, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29222166

ABSTRACT

IL-7 therapy has been evaluated in patients who do not regain normal CD4 T cell counts after virologically successful antiretroviral therapy. IL-7 increases total circulating CD4 and CD8 T cell counts; however, its effect on HIV-specific CD8 T cells has not been fully examined. TRAF1, a prosurvival signaling adaptor required for 4-1BB-mediated costimulation, is lost from chronically stimulated virus-specific CD8 T cells with progression of HIV infection in humans and during chronic lymphocytic choriomeningitis infection in mice. Previous results showed that IL-7 can restore TRAF1 expression in virus-specific CD8 T cells in mice, rendering them sensitive to anti-4-1BB agonist therapy. In this article, we show that IL-7 therapy in humans increases the number of circulating HIV-specific CD8 T cells. For a subset of patients, we also observed an increased frequency of TRAF1+ HIV-specific CD8 T cells 10 wk after completion of IL-7 treatment. IL-7 treatment increased levels of phospho-ribosomal protein S6 in HIV-specific CD8 T cells, suggesting increased activation of the metabolic checkpoint kinase mTORC1. Thus, IL-7 therapy in antiretroviral therapy-treated patients induces sustained changes in the number and phenotype of HIV-specific T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , Ribosomal Protein S6/metabolism , TNF Receptor-Associated Factor 1/metabolism , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , Cytokines/biosynthesis , Gene Expression , HIV Infections/drug therapy , HIV Infections/virology , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Interleukin-7/pharmacology , Interleukin-7/therapeutic use , Lymphocyte Count , Mechanistic Target of Rapamycin Complex 1/metabolism , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Ribosomal Protein S6/genetics , TNF Receptor-Associated Factor 1/genetics , Viral Load
20.
Mol Cell ; 48(3): 434-44, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23022379

ABSTRACT

Packaging of DNA into nucleosomes and the formation of higher-order chromatin structures determine DNA accessibility and activity of genome domains. We identified an RNA-dependent mechanism maintaining the open chromatin structure within euchromatic regions in Drosophila cells. The mechanism of reversible chromatin opening, reconstituted in vitro, depends on the Drosophila decondensation factor 31 (Df31) that specifically binds to RNA and localizes to euchromatic regions. Df31 is capable to tether a heterogeneous pool of short, single-stranded RNAs to chromatin. This class of chromatin-associated RNA (caRNA) is stably linked to chromatin and is largely composed of snoRNAs, which are preferentially bound by Df31. We suggest that the Df31-mediated linkage of snoRNAs and chromatin, forms a RNA-chromatin network resulting in the establishment of open chromatin domains. Analysis of caRNAs in human cells also reveals a strong enrichment of snoRNAs, implying a conserved role for these molecules in higher-order structures of chromatin.


Subject(s)
Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , RNA, Small Nucleolar/genetics , Animals , Base Sequence , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , HeLa Cells , Histones/metabolism , Humans , Immunoprecipitation , Molecular Sequence Data , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , RNA, Small Nucleolar/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL