Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916275

ABSTRACT

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Subject(s)
Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
2.
Cell ; 162(1): 170-83, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095250

ABSTRACT

Lipid composition affects the biophysical properties of membranes that provide a platform for receptor-mediated cellular signaling. To study the regulatory role of membrane lipid composition, we combined genetic perturbations of sphingolipid metabolism with the quantification of diverse steps in Toll-like receptor (TLR) signaling and mass spectrometry-based lipidomics. Membrane lipid composition was broadly affected by these perturbations, revealing a circular network of coregulated sphingolipids and glycerophospholipids. This evolutionarily conserved network architecture simultaneously reflected membrane lipid metabolism, subcellular localization, and adaptation mechanisms. Integration of the diverse TLR-induced inflammatory phenotypes with changes in lipid abundance assigned distinct functional roles to individual lipid species organized across the network. This functional annotation accurately predicted the inflammatory response of cells derived from patients suffering from lipid storage disorders, based solely on their altered membrane lipid composition. The analytical strategy described here empowers the understanding of higher-level organization of membrane lipid function in diverse biological systems.


Subject(s)
Immunity, Innate , Lipids/immunology , Animals , Cell Membrane/chemistry , Fibroblasts/metabolism , Gaucher Disease/immunology , Humans , Interleukin-6/immunology , Leukodystrophy, Globoid Cell/immunology , Metabolic Networks and Pathways , Mice , Sphingolipids/metabolism , Toll-Like Receptors/immunology
3.
Cell ; 162(3): 478-87, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232220

ABSTRACT

Solute carrier (SLC) membrane transport proteins control essential physiological functions, including nutrient uptake, ion transport, and waste removal. SLCs interact with several important drugs, and a quarter of the more than 400 SLC genes are associated with human diseases. Yet, compared to other gene families of similar stature, SLCs are relatively understudied. The time is right for a systematic attack on SLC structure, specificity, and function, taking into account kinship and expression, as well as the dependencies that arise from the common metabolic space.


Subject(s)
Membrane Transport Proteins/metabolism , Animals , Biomedical Research , Drug Discovery , Gene Expression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics
4.
Cell ; 161(2): 333-47, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25860612

ABSTRACT

NF-κB is a key transcriptional regulator involved in inflammation and cell proliferation, survival, and transformation. Several key steps in its activation are mediated by the ubiquitin (Ub) system. One uncharacterized step is limited proteasomal processing of the NF-κB1 precursor p105 to the p50 active subunit. Here, we identify KPC1 as the Ub ligase (E3) that binds to the ankyrin repeats domain of p105, ubiquitinates it, and mediates its processing both under basal conditions and following signaling. Overexpression of KPC1 inhibits tumor growth likely mediated via excessive generation of p50. Also, overabundance of p50 downregulates p65, suggesting that a p50-p50 homodimer may modulate transcription in place of the tumorigenic p50-p65. Transcript analysis reveals increased expression of genes associated with tumor-suppressive signals. Overall, KPC1 regulation of NF-κB1 processing appears to constitute an important balancing step among the stimulatory and inhibitory activities of the transcription factor in cell growth control.


Subject(s)
NF-kappa B p50 Subunit/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cell-Free System , Humans , Intracellular Signaling Peptides and Proteins , NF-kappa B p50 Subunit/chemistry , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction , Ubiquitin-Protein Ligases/isolation & purification , Ubiquitination
5.
Nat Immunol ; 17(12): 1361-1372, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798618

ABSTRACT

Hemolysis drives susceptibility to bacterial infections and predicts poor outcome from sepsis. These detrimental effects are commonly considered to be a consequence of heme-iron serving as a nutrient for bacteria. We employed a Gram-negative sepsis model and found that elevated heme levels impaired the control of bacterial proliferation independently of heme-iron acquisition by pathogens. Heme strongly inhibited phagocytosis and the migration of human and mouse phagocytes by disrupting actin cytoskeletal dynamics via activation of the GTP-binding Rho family protein Cdc42 by the guanine nucleotide exchange factor DOCK8. A chemical screening approach revealed that quinine effectively prevented heme effects on the cytoskeleton, restored phagocytosis and improved survival in sepsis. These mechanistic insights provide potential therapeutic targets for patients with sepsis or hemolytic disorders.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Guanine Nucleotide Exchange Factors/metabolism , Heme/metabolism , Hemolysis/immunology , Macrophages/immunology , Phagocytosis , Sepsis/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Cytoskeleton/metabolism , Female , Gram-Negative Bacterial Infections/drug therapy , Guanine Nucleotide Exchange Factors/genetics , Heme Oxygenase-1/genetics , Hemolysis/drug effects , Humans , Immune Evasion , Macrophages/drug effects , Macrophages/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Quinine/therapeutic use , RAW 264.7 Cells , Sepsis/drug therapy , cdc42 GTP-Binding Protein/metabolism
6.
Mol Cell ; 76(3): 485-499.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31495563

ABSTRACT

Transcriptional responses to external stimuli remain poorly understood. Using global nuclear run-on followed by sequencing (GRO-seq) and precision nuclear run-on sequencing (PRO-seq), we show that CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance. The Mediator kinase module contains CDK8 or CDK19, which are presumed to be functionally redundant. We implemented cortistatin A, chemical genetics, transcriptomics, and other methods to decouple their function while assessing enzymatic versus structural roles. Unexpectedly, CDK8 and CDK19 regulated different gene sets via distinct mechanisms. CDK8-dependent regulation required its kinase activity, whereas CDK19 governed IFN-γ responses through its scaffolding function (i.e., it was kinase independent). Accordingly, CDK8, not CDK19, phosphorylates the STAT1 transcription factor (TF) during IFN-γ stimulation, and CDK8 kinase inhibition blocked activation of JAK-STAT pathway TFs. Cytokines such as IFN-γ rapidly mobilize TFs to "reprogram" cellular transcription; our results implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Fibroblasts/drug effects , Interferon-gamma/pharmacology , Transcription, Genetic/drug effects , Animals , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Fibroblasts/enzymology , Fibroblasts/virology , HCT116 Cells , Host-Pathogen Interactions , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA Polymerase II/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Vesiculovirus/pathogenicity
7.
Nat Chem Biol ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907113

ABSTRACT

Metabolic alterations in cancer precipitate in associated dependencies that can be therapeutically exploited. To meet this goal, natural product-inspired small molecules can provide a resource of invaluable chemotypes. Here, we identify orpinolide, a synthetic withanolide analog with pronounced antileukemic properties, via orthogonal chemical screening. Through multiomics profiling and genome-scale CRISPR-Cas9 screens, we identify that orpinolide disrupts Golgi homeostasis via a mechanism that requires active phosphatidylinositol 4-phosphate signaling at the endoplasmic reticulum-Golgi membrane interface. Thermal proteome profiling and genetic validation studies reveal the oxysterol-binding protein OSBP as the direct and phenotypically relevant target of orpinolide. Collectively, these data reaffirm sterol transport as a therapeutically actionable dependency in leukemia and motivate ensuing translational investigation via the probe-like compound orpinolide.

8.
Cell ; 147(2): 306-19, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-22000011

ABSTRACT

Chronic myelogenous leukemia (CML) is caused by the constitutively active tyrosine kinase Bcr-Abl and treated with the tyrosine kinase inhibitor (TKI) imatinib. However, emerging TKI resistance prevents complete cure. Therefore, alternative strategies targeting regulatory modules of Bcr-Abl in addition to the kinase active site are strongly desirable. Here, we show that an intramolecular interaction between the SH2 and kinase domains in Bcr-Abl is both necessary and sufficient for high catalytic activity of the enzyme. Disruption of this interface led to inhibition of downstream events critical for CML signaling and, importantly, completely abolished leukemia formation in mice. Furthermore, disruption of the SH2-kinase interface increased sensitivity of imatinib-resistant Bcr-Abl mutants to TKI inhibition. An engineered Abl SH2-binding fibronectin type III monobody inhibited Bcr-Abl kinase activity both in vitro and in primary CML cells, where it induced apoptosis. This work validates the SH2-kinase interface as an allosteric target for therapeutic intervention.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Amino Acid Sequence , Animals , Base Sequence , Benzamides , Cells, Cultured , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Isoleucine/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacology , Signal Transduction , src Homology Domains
9.
Nature ; 581(7808): 316-322, 2020 05.
Article in English | MEDLINE | ID: mdl-32433612

ABSTRACT

Toll-like receptors (TLRs) have a crucial role in the recognition of pathogens and initiation of immune responses1-3. Here we show that a previously uncharacterized protein encoded by CXorf21-a gene that is associated with systemic lupus erythematosus4,5-interacts with the endolysosomal transporter SLC15A4, an essential but poorly understood component of the endolysosomal TLR machinery also linked to autoimmune disease4,6-9. Loss of this type-I-interferon-inducible protein, which we refer to as 'TLR adaptor interacting with SLC15A4 on the lysosome' (TASL), abrogated responses to endolysosomal TLR agonists in both primary and transformed human immune cells. Deletion of SLC15A4 or TASL specifically impaired the activation of the IRF pathway without affecting NF-κB and MAPK signalling, which indicates that ligand recognition and TLR engagement in the endolysosome occurred normally. Extensive mutagenesis of TASL demonstrated that its localization and function relies on the interaction with SLC15A4. TASL contains a conserved pLxIS motif (in which p denotes a hydrophilic residue and x denotes any residue) that mediates the recruitment and activation of IRF5. This finding shows that TASL is an innate immune adaptor for TLR7, TLR8 and TLR9 signalling, revealing a clear mechanistic analogy with the IRF3 adaptors STING, MAVS and TRIF10,11. The identification of TASL as the component that links endolysosomal TLRs to the IRF5 transcription factor via SLC15A4 provides a mechanistic explanation for the involvement of these proteins in systemic lupus erythematosus12-14.


Subject(s)
Interferon Regulatory Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 9/metabolism , Amino Acid Motifs , Animals , Female , Humans , Immunity, Innate , Interferon Type I/immunology , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lupus Erythematosus, Systemic/metabolism , Male , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Protein Binding , Signal Transduction
10.
Cell ; 141(4): 668-81, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20451243

ABSTRACT

Peroxisomes have long been established to play a central role in regulating various metabolic activities in mammalian cells. These organelles act in concert with mitochondria to control the metabolism of lipids and reactive oxygen species. However, while mitochondria have emerged as an important site of antiviral signal transduction, a role for peroxisomes in immune defense is unknown. Here, we report that the RIG-I-like receptor (RLR) adaptor protein MAVS is located on peroxisomes and mitochondria. We find that peroxisomal and mitochondrial MAVS act sequentially to create an antiviral cellular state. Upon viral infection, peroxisomal MAVS induces the rapid interferon-independent expression of defense factors that provide short-term protection, whereas mitochondrial MAVS activates an interferon-dependent signaling pathway with delayed kinetics, which amplifies and stabilizes the antiviral response. The interferon regulatory factor IRF1 plays a crucial role in regulating MAVS-dependent signaling from peroxisomes. These results establish that peroxisomes are an important site of antiviral signal transduction.


Subject(s)
Immunity, Innate , Peroxisomes/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Chlorocebus aethiops , Fibroblasts/metabolism , Hepatocytes/metabolism , Humans , Interferons/metabolism , Mice , Mitochondria/metabolism , Vero Cells
11.
Nucleic Acids Res ; 51(17): 9248-9265, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37587695

ABSTRACT

Though the effect of the recently identified mitochondrial NAD+ transporter SLC25A51 on glucose metabolism has been described, its contribution to other NAD+-dependent processes throughout the cell such as ADP-ribosylation remains elusive. Here, we report that absence of SLC25A51 leads to increased NAD+ concentration not only in the cytoplasm and but also in the nucleus. The increase is not associated with upregulation of the salvage pathway, implying an accumulation of constitutively synthesized NAD+ in the cytoplasm and nucleus. This results in an increase of PARP1-mediated nuclear ADP-ribosylation, as well as faster repair of DNA lesions induced by different single-strand DNA damaging agents. Lastly, absence of SLC25A51 reduces both MMS/Olaparib induced PARP1 chromatin retention and the sensitivity of different breast cancer cells to PARP1 inhibition. Together these results provide evidence that SLC25A51 might be a novel target to improve PARP1 inhibitor based therapies by changing subcellular NAD+ redistribution.


Subject(s)
NAD , Chromatin , DNA Repair , Mitochondria/metabolism , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Humans
12.
Immunity ; 43(5): 974-86, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588782

ABSTRACT

Tissue damage caused by viral hepatitis is a major cause of morbidity and mortality worldwide. Using a mouse model of viral hepatitis, we identified virus-induced early transcriptional changes in the redox pathways in the liver, including downregulation of superoxide dismutase 1 (Sod1). Sod1(-/-) mice exhibited increased inflammation and aggravated liver damage upon viral infection, which was independent of T and NK cells and could be ameliorated by antioxidant treatment. Type I interferon (IFN-I) led to a downregulation of Sod1 and caused oxidative liver damage in Sod1(-/-) and wild-type mice. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against virus-induced liver damage. These results delineate IFN-I mediated oxidative stress as a key mediator of virus-induced liver damage and describe a mechanism of innate-immunity-driven pathology, linking IFN-I signaling with antioxidant host defense and infection-associated tissue damage. VIDEO ABSTRACT.


Subject(s)
Hepatocytes/immunology , Interferon Type I/immunology , Oxidative Stress/immunology , Superoxide Dismutase/immunology , Animals , Antioxidants/metabolism , Hepatitis, Viral, Animal/immunology , Immunity, Innate/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Liver/immunology , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Signal Transduction/immunology , Superoxide Dismutase-1 , T-Lymphocytes/immunology , Transcription, Genetic/immunology
13.
Ann Rheum Dis ; 82(9): 1142-1152, 2023 09.
Article in English | MEDLINE | ID: mdl-37344156

ABSTRACT

INTRODUCTION: Structural reorganisation of the synovium with expansion of fibroblast-like synoviocytes (FLS) and influx of immune cells is a hallmark of rheumatoid arthritis (RA). Activated FLS are increasingly recognised as a critical component driving synovial tissue remodelling by interacting with immune cells resulting in distinct synovial pathotypes of RA. METHODS: Automated high-content fluorescence microscopy of co-cultured cytokine-activated FLS and autologous peripheral CD4+ T cells from patients with RA was established to quantify cell-cell interactions. Phenotypic profiling of cytokine-treated FLS and co-cultured T cells was done by flow cytometry and RNA-Seq, which were integrated with publicly available transcriptomic data from patients with different histological synovial pathotypes. Computational prediction and knock-down experiments were performed in FLS to identify adhesion molecules for cell-cell interaction. RESULTS: Cytokine stimulation, especially with TNF-α, led to enhanced FLS-T cell interaction resulting in cell-cell contact-dependent activation, proliferation and differentiation of T cells. Signatures of cytokine-activated FLS were significantly enriched in RA synovial tissues defined as lymphoid-rich or leucocyte-rich pathotypes, with the most prominent effects for TNF-α. FLS cytokine signatures correlated with the number of infiltrating CD4+ T cells in synovial tissue of patients with RA. Ligand-receptor pair interaction analysis identified ICAM1 on FLS as an important mediator in TNF-mediated FLS-T cell interaction. Both, ICAM1 and its receptors were overexpressed in TNF-treated FLS and co-cultured T cells. Knock-down of ICAM1 in FLS resulted in reduced TNF-mediated FLS-T cell interaction. CONCLUSION: Our study highlights the role of cytokine-activated FLS in orchestrating inflammation-associated synovial pathotypes providing novel insights into disease mechanisms of RA.


Subject(s)
Arthritis, Rheumatoid , Synoviocytes , Humans , Cytokines , Tumor Necrosis Factor-alpha/pharmacology , Synovial Membrane/pathology , Synoviocytes/pathology , Fibroblasts/pathology , Cells, Cultured
14.
Nat Immunol ; 12(7): 624-30, 2011 Jun 05.
Article in English | MEDLINE | ID: mdl-21642987

ABSTRACT

Antiviral innate immunity relies on the recognition of microbial structures. One such structure is viral RNA that carries a triphosphate group on its 5' terminus (PPP-RNA). By an affinity proteomics approach with PPP-RNA as the 'bait', we found that the antiviral protein IFIT1 (interferon-induced protein with tetratricopeptide repeats 1) mediated binding of a larger protein complex containing other IFIT family members. IFIT1 bound PPP-RNA with nanomolar affinity and required the arginine at position 187 in a highly charged carboxy-terminal groove of the protein. In the absence of IFIT1, the growth and pathogenicity of viruses containing PPP-RNA was much greater. In contrast, IFIT proteins were dispensable for the clearance of pathogens that did not generate PPP-RNA. On the basis of this specificity and the great abundance of IFIT proteins after infection, we propose that the IFIT complex antagonizes viruses by sequestering specific viral nucleic acids.


Subject(s)
Arginine/immunology , Carrier Proteins/immunology , RNA, Viral/immunology , Viruses/immunology , Adaptor Proteins, Signal Transducing , Animals , Arginine/chemistry , Arginine/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , RNA-Binding Proteins
15.
Cell ; 134(5): 793-803, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18775312

ABSTRACT

The SH2 domain of cytoplasmic tyrosine kinases can enhance catalytic activity and substrate recognition, but the molecular mechanisms by which this is achieved are poorly understood. We have solved the structure of the prototypic SH2-kinase unit of the human Fes tyrosine kinase, which appears specialized for positive signaling. In its active conformation, the SH2 domain tightly interacts with the kinase N-terminal lobe and positions the kinase alphaC helix in an active configuration through essential packing and electrostatic interactions. This interaction is stabilized by ligand binding to the SH2 domain. Our data indicate that Fes kinase activation is closely coupled to substrate recognition through cooperative SH2-kinase-substrate interactions. Similarly, we find that the SH2 domain of the active Abl kinase stimulates catalytic activity and substrate phosphorylation through a distinct SH2-kinase interface. Thus, the SH2 and catalytic domains of active Fes and Abl pro-oncogenic kinases form integrated structures essential for effective tyrosine kinase signaling.


Subject(s)
Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-fes/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Enzyme Activation , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-fes/metabolism
16.
Nat Chem Biol ; 16(4): 469-478, 2020 04.
Article in English | MEDLINE | ID: mdl-32152546

ABSTRACT

Solute carriers (SLCs) are the largest family of transmembrane transporters in humans and are major determinants of cellular metabolism. Several SLCs have been shown to be required for the uptake of chemical compounds into cellular systems, but systematic surveys of transporter-drug relationships in human cells are currently lacking. We performed a series of genetic screens in a haploid human cell line against 60 cytotoxic compounds representative of the chemical space populated by approved drugs. By using an SLC-focused CRISPR-Cas9 library, we identified transporters whose absence induced resistance to the drugs tested. This included dependencies involving the transporters SLC11A2/SLC16A1 for artemisinin derivatives and SLC35A2/SLC38A5 for cisplatin. The functional dependence on SLCs observed for a significant proportion of the screened compounds suggests a widespread role for SLCs in the uptake and cellular activity of cytotoxic drugs and provides an experimentally validated set of SLC-drug associations for a number of clinically relevant compounds.


Subject(s)
Drug Resistance/genetics , Solute Carrier Proteins/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Antineoplastic Agents , Biochemical Phenomena , Biological Transport/genetics , Biological Transport/physiology , CRISPR-Cas Systems , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Drug Resistance/physiology , Genetic Testing , Humans , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Protein Transport/physiology , Solute Carrier Proteins/physiology , Symporters/genetics , Symporters/metabolism
17.
Mol Syst Biol ; 16(7): e9652, 2020 07.
Article in English | MEDLINE | ID: mdl-32697042

ABSTRACT

Solute carriers (SLCs) are the largest family of transmembrane transporters in the human genome with more than 400 members. Despite the fact that SLCs mediate critical biological functions and several are important pharmacological targets, a large proportion of them is poorly characterized and present no assigned substrate. A major limitation to systems-level de-orphanization campaigns is the absence of a structured, language-controlled chemical annotation. Here we describe a thorough manual annotation of SLCs based on literature. The annotation of substrates, transport mechanism, coupled ions, and subcellular localization for 446 human SLCs confirmed that ~30% of these were still functionally orphan and lacked known substrates. Application of a substrate-based ontology to transcriptomic datasets identified SLC-specific responses to external perturbations, while a machine-learning approach based on the annotation allowed us to identify potential substrates for several orphan SLCs. The annotation is available at https://opendata.cemm.at/gsflab/slcontology/. Given the increasing availability of large biological datasets and the growing interest in transporters, we expect that the effort presented here will be critical to provide novel insights into the functions of SLCs.


Subject(s)
Biological Transport, Active/genetics , Computational Biology/methods , Membrane Transport Proteins/metabolism , Amino Acids/metabolism , Amino Acids/pharmacology , Biological Ontologies , Cell Line , Gene Expression Profiling , Genome, Human , Humans , Machine Learning
18.
Nat Immunol ; 10(3): 266-72, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19158679

ABSTRACT

Cytoplasmic DNA triggers activation of the innate immune system. Although 'downstream' signaling components have been characterized, the DNA-sensing components remain elusive. Here we present a systematic proteomics screen for proteins that associate with DNA, 'crossed' to a screen for transcripts induced by interferon-beta, which identified AIM2 as a candidate cytoplasmic DNA sensor. AIM2 showed specificity for double-stranded DNA. It also recruited the inflammasome adaptor ASC and localized to ASC 'speckles'. A decrease in AIM2 expression produced by RNA-mediated interference impaired DNA-induced maturation of interleukin 1beta in THP-1 human monocytic cells, which indicated that endogenous AIM2 is required for DNA recognition. Reconstitution of unresponsive HEK293 cells with AIM2, ASC, caspase-1 and interleukin 1beta showed that AIM2 was sufficient for inflammasome activation. Our data suggest that AIM2 is a cytoplasmic DNA sensor for the inflammasome.


Subject(s)
DNA/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Caspase 1/immunology , Caspase 1/metabolism , Cytosol/metabolism , DNA/immunology , DNA-Binding Proteins , Gene Expression Profiling , Genomics/methods , Humans , Immunity, Innate , Interferon-beta/immunology , Interferon-beta/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , NIH 3T3 Cells , Nuclear Proteins/immunology , Proteomics/methods
19.
Nat Chem Biol ; 15(3): 232-240, 2019 03.
Article in English | MEDLINE | ID: mdl-30692684

ABSTRACT

The Bruton tyrosine kinase (BTK) inhibitor ibrutinib has substantially improved therapeutic options for chronic lymphocytic leukemia (CLL). Although ibrutinib is not curative, it has a profound effect on CLL cells and may create new pharmacologically exploitable vulnerabilities. To identify such vulnerabilities, we developed a systematic approach that combines epigenome profiling (charting the gene-regulatory basis of cell state) with single-cell chemosensitivity profiling (quantifying cell-type-specific drug response) and bioinformatic data integration. By applying our method to a cohort of matched patient samples collected before and during ibrutinib therapy, we identified characteristic ibrutinib-induced changes that provide a starting point for the rational design of ibrutinib combination therapies. Specifically, we observed and validated preferential sensitivity to proteasome, PLK1, and mTOR inhibitors during ibrutinib treatment. More generally, our study establishes a broadly applicable method for investigating treatment-specific vulnerabilities by integrating the complementary perspectives of epigenetic cell states and phenotypic drug responses in primary patient samples.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Chromatin/physiology , Drug Combinations , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Piperidines , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Single-Cell Analysis/methods , TOR Serine-Threonine Kinases/metabolism , Polo-Like Kinase 1
20.
Nat Rev Mol Cell Biol ; 10(12): 884-7, 2009 12.
Article in English | MEDLINE | ID: mdl-19859061

ABSTRACT

An explosion of scientific and technological advances has broadened the field of biomedicine. Traditional boundaries between the public and private research sectors are now blurred by multidisciplinary projects and the necessity for new and more efficient models of the translational process. This allows the adventurous scientist to boldly and consciously sample selected skills during periods of secondment in different institutions and organizations, and to assemble a personal and unique blend of competences to help them manage their career.


Subject(s)
Biomedical Research , Industry , Occupations , Universities , Humans , Industry/economics , Occupations/economics , Universities/economics
SELECTION OF CITATIONS
SEARCH DETAIL