Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
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
2.
Nat Immunol ; 11(1): 63-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19915568

ABSTRACT

Interleukin 1 beta (IL-1 beta) is a potent proinflammatory factor during viral infection. Its production is tightly controlled by transcription of Il1b dependent on the transcription factor NF-kappaB and subsequent processing of pro-IL-1 beta by an inflammasome. However, the sensors and mechanisms that facilitate RNA virus-induced production of IL-1 beta are not well defined. Here we report a dual role for the RNA helicase RIG-I in RNA virus-induced proinflammatory responses. Whereas RIG-I-mediated activation of NF-kappaB required the signaling adaptor MAVS and a complex of the adaptors CARD9 and Bcl-10, RIG-I also bound to the adaptor ASC to trigger caspase-1-dependent inflammasome activation by a mechanism independent of MAVS, CARD9 and the Nod-like receptor protein NLRP3. Our results identify the CARD9-Bcl-10 module as an essential component of the RIG-I-dependent proinflammatory response and establish RIG-I as a sensor able to activate the inflammasome in response to certain RNA viruses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Inflammation/physiopathology , Interleukin-1beta/metabolism , RNA Viruses/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cell Line , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/physiology , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Host-Pathogen Interactions , Humans , Immunoblotting , Inflammation/immunology , Inflammation/virology , Interferon-Induced Helicase, IFIH1 , Mice , Mice, Knockout , Models, Biological , RNA Virus Infections/immunology , RNA Virus Infections/physiopathology , RNA Virus Infections/virology , RNA Viruses/immunology , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/physiology , bcl-X Protein/genetics , bcl-X Protein/metabolism
3.
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
4.
Nature ; 519(7544): 477-81, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25561175

ABSTRACT

Cell growth and proliferation are tightly linked to nutrient availability. The mechanistic target of rapamycin complex 1 (mTORC1) integrates the presence of growth factors, energy levels, glucose and amino acids to modulate metabolic status and cellular responses. mTORC1 is activated at the surface of lysosomes by the RAG GTPases and the Ragulator complex through a not fully understood mechanism monitoring amino acid availability in the lysosomal lumen and involving the vacuolar H(+)-ATPase. Here we describe the uncharacterized human member 9 of the solute carrier family 38 (SLC38A9) as a lysosomal membrane-resident protein competent in amino acid transport. Extensive functional proteomic analysis established SLC38A9 as an integral part of the Ragulator-RAG GTPases machinery. Gain of SLC38A9 function rendered cells resistant to amino acid withdrawal, whereas loss of SLC38A9 expression impaired amino-acid-induced mTORC1 activation. Thus SLC38A9 is a physical and functional component of the amino acid sensing machinery that controls the activation of mTOR.


Subject(s)
Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/metabolism , Nucleotides/metabolism
5.
Mol Cell Proteomics ; 15(3): 1139-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26933192

ABSTRACT

Tandem affinity purification-mass spectrometry (TAP-MS) is a popular strategy for the identification of protein-protein interactions, characterization of protein complexes, and entire networks. Its employment in cellular settings best fitting the relevant physiology is limited by convenient expression vector systems. We developed an easy-to-handle, inducible, dually selectable retroviral expression vector allowing dose- and time-dependent control of bait proteins bearing the efficient streptavidin-hemagglutinin (SH)-tag at their N- or C termini. Concomitant expression of a reporter fluorophore allows to monitor bait-expressing cells by flow cytometry or microscopy and enables high-throughput phenotypic assays. We used the system to successfully characterize the interactome of the neuroblastoma RAS viral oncogene homolog (NRAS) Gly12Asp (G12D) mutant and exploited the advantage of reporter fluorophore expression by tracking cytokine-independent cell growth using flow cytometry. Moreover, we tested the feasibility of studying cytotoxicity-mediating proteins with the vector system on the cell death-inducing mixed lineage kinase domain-like protein (MLKL) Ser358Asp (S358D) mutant. Interaction proteomics analysis of MLKL Ser358Asp (S358D) identified heat shock protein 90 (HSP90) as a high-confidence interacting protein. Further phenotypic characterization established MLKL as a novel HSP90 client. In summary, this novel inducible expression system enables SH-tag-based interaction studies in the cell line proficient for the respective phenotypic or signaling context and constitutes a valuable tool for experimental approaches requiring inducible or traceable protein expression.


Subject(s)
Chromatography, Affinity/methods , HSP90 Heat-Shock Proteins/metabolism , Mutation , Protein Kinases/metabolism , Proteomics/methods , Retroviridae/genetics , Tandem Mass Spectrometry/methods , Animals , Cell Line , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Genes, Reporter , HEK293 Cells , HT29 Cells , Humans , K562 Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Kinases/genetics
6.
Immunity ; 28(5): 651-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18439848

ABSTRACT

Upon detection of viral RNA, the helicases RIG-I and/or MDA5 trigger, via their adaptor Cardif (also known as IPS-1, MAVS, or VISA), the activation of the transcription factors NF-kappaB and IRF3, which collaborate to induce an antiviral type I interferon (IFN) response. FADD and RIP1, known as mediators of death-receptor signaling, are implicated in this antiviral pathway; however, the link between death-receptor and antiviral signaling is not known. Here we showed that TRADD, a crucial adaptor of tumor necrosis factor receptor (TNFRI), was important in RIG-like helicase (RLH)-mediated signal transduction. TRADD is recruited to Cardif and orchestrated complex formation with the E3 ubiquitin ligase TRAF3 and TANK and with FADD and RIP1, leading to the activation of IRF3 and NF-kappaB. Loss of TRADD prevented Cardif-dependent activation of IFN-beta, reduced the production of IFN-beta in response to RNA viruses, and enhanced vesicular stomatitis virus replication. Thus, TRADD is not only an essential component of proinflammatory TNFRI signaling, but is also required for RLH-Cardif-dependent antiviral immune responses.


Subject(s)
DNA Helicases/metabolism , Interferon Regulatory Factor-3/metabolism , Rhabdoviridae Infections/immunology , TNF Receptor-Associated Death Domain Protein/metabolism , Vesiculovirus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Fas-Associated Death Domain Protein/metabolism , GTPase-Activating Proteins/metabolism , Humans , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , Rhabdoviridae Infections/virology , Signal Transduction , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , Vesiculovirus/physiology
7.
Nat Methods ; 10(10): 965-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24161985

ABSTRACT

Knockout collections are invaluable tools for studying model organisms such as yeast. However, there are no large-scale knockout collections of human cells. Using gene-trap mutagenesis in near-haploid human cells, we established a platform to generate and isolate individual 'gene-trapped cells' and used it to prepare a collection of human cell lines carrying single gene-trap insertions. In most cases, the insertion can be reversed. This growing library covers 3,396 genes, one-third of the expressed genome, is DNA-barcoded and allows systematic screens for a wide variety of cellular phenotypes. We examined cellular responses to TNF-α, TGF-ß, IFN-γ and TNF-related apoptosis-inducing ligand (TRAIL), to illustrate the value of this unique collection of isogenic human cell lines.


Subject(s)
Gene Library , Haploidy , Mutagenesis, Insertional/methods , Reverse Genetics/methods , Cell Line, Tumor , Genome, Human , Humans , Molecular Sequence Data
8.
Trends Immunol ; 34(12): 610-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23827258

ABSTRACT

The immune response to pathogens is controlled by complex and tightly regulated molecular networks. Recent technological advances have empowered approaches to investigate innate immune signaling and monitor host-pathogen interactions at a systems level. Protein complexes are key players in pathogen recognition and integrate much of the host molecular responses that occur at the transcriptional and translational level. The ability to monitor protein complex abundance, dynamics, and composition is therefore important to understand the ability of cells to mount the appropriate immune response. Here, we focus on current proteomics technologies applied to identify the protein complexes involved, and highlight recent studies illustrating the power of these approaches to unravel how the dedicated molecular machinery is integrated with other cellular processes to safeguard homeostasis.


Subject(s)
Immunity, Innate/genetics , Immunity, Innate/immunology , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , Proteome/genetics , Proteome/immunology , Humans , Proteomics/methods , Signal Transduction/genetics , Signal Transduction/immunology
9.
J Immunol ; 188(8): 3820-8, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22412192

ABSTRACT

Nucleotide-binding oligomerization domain-like receptors (NLRs) are intracellular proteins involved in innate-driven inflammatory responses. The function of the family member NLR caspase recruitment domain containing protein 5 (NLRC5) remains a matter of debate, particularly with respect to NF-κB activation, type I IFN, and MHC I expression. To address the role of NLRC5, we generated Nlrc5-deficient mice (Nlrc5(Δ/Δ)). In this article we show that these animals exhibit slightly decreased CD8(+) T cell percentages, a phenotype compatible with deregulated MHC I expression. Of interest, NLRC5 ablation only mildly affected MHC I expression on APCs and, accordingly, Nlrc5(Δ/Δ) macrophages efficiently primed CD8(+) T cells. In contrast, NLRC5 deficiency dramatically impaired basal expression of MHC I in T, NKT, and NK lymphocytes. NLRC5 was sufficient to induce MHC I expression in a human lymphoid cell line, requiring both caspase recruitment and LRR domains. Moreover, endogenous NLRC5 localized to the nucleus and occupied the proximal promoter region of H-2 genes. Consistent with downregulated MHC I expression, the elimination of Nlrc5(Δ/Δ) lymphocytes by cytotoxic T cells was markedly reduced and, in addition, we observed low NLRC5 expression in several murine and human lymphoid-derived tumor cell lines. Hence, loss of NLRC5 expression represents an advantage for evading CD8(+) T cell-mediated elimination by downmodulation of MHC I levels-a mechanism that may be exploited by transformed cells. Our data show that NLRC5 acts as a key transcriptional regulator of MHC I in lymphocytes and support an essential role for NLRs in directing not only innate but also adaptive immune responses.


Subject(s)
Genes, MHC Class I , Intracellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Bone Marrow/immunology , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/immunology , Cell Proliferation , Gene Expression Regulation , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , T-Lymphocytes, Cytotoxic/cytology
10.
Cell Rep ; 42(8): 112916, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37527038

ABSTRACT

Endolysosomal Toll-like receptors (TLRs) play crucial roles in immune responses to pathogens, while aberrant activation of these pathways is associated with autoimmune diseases, including systemic lupus erythematosus (SLE). The endolysosomal solute carrier family 15 member 4 (SLC15A4) is required for TLR7/8/9-induced responses and disease development in SLE models. SLC15A4 has been proposed to affect TLR7-9 activation through its transport activity, as well as by assembling an IRF5-activating complex with TASL, but the relative contribution of these functions remains unclear. Here, we show that the essential role of SLC15A4 is to recruit TASL to endolysosomes, while its transport activity is dispensable when TASL is tethered to this compartment. Endolysosomal-localized TASL rescues TLR7-9-induced IRF5 activation as well as interferon ß and cytokine production in SLC15A4-deficient cells. SLC15A4 acts as signaling scaffold, and this function is essential to control TLR7-9-mediated inflammatory responses. These findings support targeting the SLC15A4-TASL complex as a potential therapeutic strategy for SLE and related diseases.


Subject(s)
Lupus Erythematosus, Systemic , Toll-Like Receptor 7 , Humans , Toll-Like Receptor 7/metabolism , Toll-Like Receptors/metabolism , Interferon Regulatory Factors/metabolism , Immunity, Innate , Nerve Tissue Proteins/metabolism , Membrane Transport Proteins/metabolism
11.
Nat Commun ; 14(1): 6626, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863876

ABSTRACT

Dysregulation of pathogen-recognition pathways of the innate immune system is associated with multiple autoimmune disorders. Due to the intricacies of the molecular network involved, the identification of pathway- and disease-specific therapeutics has been challenging. Using a phenotypic assay monitoring the degradation of the immune adapter TASL, we identify feeblin, a chemical entity which inhibits the nucleic acid-sensing TLR7/8 pathway activating IRF5 by disrupting the SLC15A4-TASL adapter module. A high-resolution cryo-EM structure of feeblin with SLC15A4 reveals that the inhibitor binds a lysosomal outward-open conformation incompatible with TASL binding on the cytoplasmic side, leading to degradation of TASL. This mechanism of action exploits a conformational switch and converts a target-binding event into proteostatic regulation of the effector protein TASL, interrupting the TLR7/8-IRF5 signaling pathway and preventing downstream proinflammatory responses. Considering that all components involved have been genetically associated with systemic lupus erythematosus and that feeblin blocks responses in disease-relevant human immune cells from patients, the study represents a proof-of-concept for the development of therapeutics against this disease.


Subject(s)
Lupus Erythematosus, Systemic , Toll-Like Receptor 7 , Humans , Toll-Like Receptor 7/metabolism , Interferon Regulatory Factors/metabolism , Signal Transduction , Anti-Inflammatory Agents , Nerve Tissue Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism
12.
Nat Commun ; 14(1): 6627, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863913

ABSTRACT

Toll-like receptors (TLRs) are a class of proteins that play critical roles in recognizing pathogens and initiating innate immune responses. TASL, a recently identified innate immune adaptor protein for endolysosomal TLR7/8/9 signaling, is recruited by the lysosomal proton-coupled amino-acid transporter SLC15A4, and then activates IRF5, which in turn triggers the transcription of type I interferons and cytokines. Here, we report three cryo-electron microscopy (cryo-EM) structures of human SLC15A4 in the apo monomeric and dimeric state and as a TASL-bound complex. The apo forms are in an outward-facing conformation, with the dimeric form showing an extensive interface involving four cholesterol molecules. The structure of the TASL-bound complex reveals an unprecedented interaction mode with solute carriers. During the recruitment of TASL, SLC15A4 undergoes a conformational change from an outward-facing, lysosomal lumen-exposed state to an inward-facing state to form a binding pocket, allowing the N-terminal helix of TASL to be inserted into. Our findings provide insights into the molecular basis of regulatory switch involving a human solute carrier and offers an important framework for structure-guided drug discovery targeting SLC15A4-TASL-related human autoimmune diseases.


Subject(s)
Signal Transduction , Toll-Like Receptors , Humans , Cryoelectron Microscopy , Toll-Like Receptors/metabolism , Immunity, Innate , Lysosomes/metabolism , Nerve Tissue Proteins/metabolism , Membrane Transport Proteins/metabolism
13.
Life Sci Alliance ; 5(11)2022 11.
Article in English | MEDLINE | ID: mdl-36114003

ABSTRACT

Solute carrier (SLC) transporters control fluxes of nutrients and metabolites across membranes and thereby represent a critical interface between the microenvironment and cellular and subcellular metabolism. Because of substantial functional overlap, the interplay and relative contributions of SLCs in response to environmental stresses remain poorly elucidated. To infer functional relationships between SLCs and metabolites, we developed a strategy to identify SLCs able to sustain cell viability and proliferation under growth-limiting concentrations of essential nutrients. One-by-one depletion of 13 amino acids required for cell proliferation enabled gain-of-function genetic screens using a SLC-focused CRISPR/Cas9-based transcriptional activation approach to uncover transporters relieving cells from growth-limiting metabolic bottlenecks. Among the transporters identified, we characterized the cationic amino acid transporter SLC7A3 as a gene that, when up-regulated, overcame low availability of arginine and lysine by increasing their uptake, whereas SLC7A5 was able to sustain cellular fitness upon deprivation of several neutral amino acids. Moreover, we identified metabolic compensation mediated by the glutamate/aspartate transporters SLC1A2 and SLC1A3 under glutamine-limiting conditions. Overall, this gain-of-function approach using human cells uncovered functional transporter-nutrient relationships and revealed that transport activity up-regulation may be sufficient to overcome environmental metabolic restrictions.


Subject(s)
Membrane Transport Proteins , Nutrients , Amino Acid Transport Systems, Basic/genetics , Amino Acids/metabolism , Arginine/metabolism , Aspartic Acid/metabolism , Gain of Function Mutation , Glutamates/metabolism , Glutamine/metabolism , Humans , Large Neutral Amino Acid-Transporter 1 , Lysine/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Nutrients/metabolism
14.
EMBO Rep ; 10(8): 916-22, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590578

ABSTRACT

Detection of viral nucleic acids is central to antiviral immunity. Recently, DAI/ZBP1 (DNA-dependent activator of IRFs/Z-DNA binding protein 1) was identified as a cytoplasmic DNA sensor and shown to activate the interferon regulatory factor (IRF) and nuclear factor-kappa B (NF-kappaB) transcription factors, leading to type-I interferon production. DAI-induced IRF activation depends on TANK-binding kinase 1 (TBK1), whereas signalling pathways and molecular components involved in NF-kappaB activation remain elusive. Here, we report the identification of two receptor-interacting protein (RIP) homotypic interaction motifs (RHIMs) in the DAI protein sequence, and show that these domains relay DAI-induced NF-kappaB signals through the recruitment of the RHIM-containing kinases RIP1 and RIP3. We show that knockdown of not only RIP1, but also RIP3 affects DAI-induced NF-kappaB activation. Importantly, RIP recruitment to DAI is inhibited by the RHIM-containing murine cytomegalovirus (MCMV) protein M45. These findings delineate the DAI signalling pathway to NF-kappaB and suggest a possible new immune modulation strategy of the MCMV.


Subject(s)
DNA-Binding Proteins/metabolism , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mice , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Polymerase Chain Reaction , Protein Binding/genetics , Protein Binding/physiology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
15.
Nat Commun ; 11(1): 6145, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33262325

ABSTRACT

About a thousand genes in the human genome encode for membrane transporters. Among these, several solute carrier proteins (SLCs), representing the largest group of transporters, are still orphan and lack functional characterization. We reasoned that assessing genetic interactions among SLCs may be an efficient way to obtain functional information allowing their deorphanization. Here we describe a network of strong genetic interactions indicating a contribution to mitochondrial respiration and redox metabolism for SLC25A51/MCART1, an uncharacterized member of the SLC25 family of transporters. Through a combination of metabolomics, genomics and genetics approaches, we demonstrate a role for SLC25A51 as enabler of mitochondrial import of NAD, showcasing the potential of genetic interaction-driven functional gene deorphanization.


Subject(s)
Epistasis, Genetic , Mitochondria/metabolism , NAD/metabolism , Uncoupling Protein 1/metabolism , Biological Transport , Humans , Mitochondria/genetics , Oxidation-Reduction , Uncoupling Protein 1/genetics
16.
Biochim Biophys Acta Biomembr ; 1861(9): 1558-1567, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31295473

ABSTRACT

The lysosomal amino acid transporter SLC38A9 is referred to as transceptor, i.e. a transporter with a receptor function. The protein is responsible for coupling amino acid transport across the lysosomal membrane according to the substrate availability to mTORC1 signal transduction. This process allows cells to sense amino acid level responding to growth stimuli in physiological and pathological conditions triggering mTOR regulation. The main substrates underlying this function are glutamine and arginine. The functional and kinetic characterization of glutamine and arginine transport was performed using human SLC38A9 produced in E. coli, purified by affinity chromatography and reconstituted in liposomes. A cooperative behaviour for the wild type protein was revealed for both the substrates. A novel Na+ binding site, namely T453, was described by combined approaches of bioinformatics, site-directed mutagenesis and transport assay. Stimulation by cholesterol of glutamine and arginine transport was observed. The biological function of SLC38A9 relies on the interaction between its N-terminus and components of the mTOR complex; a deletion mutant of the N-terminus tail was produced and transport of glutamine was assayed revealing that this portion does not play any role in the intrinsic transport function of the human SLC38A9. Different features for glutamine and arginine transport were revealed: human SLC38A9 is competent for glutamine efflux, while that of arginine is negligible. In line with these results, imposed ∆pH stimulated glutamine, not arginine transport. Arginine plays, on the contrary, a modulatory function and is able to stimulate glutamine efflux. Interestingly, reciprocal inhibition experiments also supported by bioinformatics, suggested that glutamine and arginine may bind to different sites in the human SLC38A9 transporter.


Subject(s)
Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/physiology , Amino Acids/metabolism , Arginine/metabolism , Binding Sites , Biological Transport , Cholesterol/metabolism , Glutamine/metabolism , Humans , Ion Transport , Kinetics , Lysosomes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
17.
Cell Death Differ ; 26(6): 1138-1155, 2019 06.
Article in English | MEDLINE | ID: mdl-30237509

ABSTRACT

Regulation of cell and tissue homeostasis by programmed cell death is a fundamental process with wide physiological and pathological implications. The advent of scalable somatic cell genetic technologies creates the opportunity to functionally map such essential pathways, thereby identifying potential disease-relevant components. We investigated the genetic basis underlying necroptotic cell death by performing a complementary set of loss-of-function and gain-of-function genetic screens. To this end, we established FADD-deficient haploid human KBM7 cells, which specifically and efficiently undergo necroptosis after a single treatment with either TNFα or the SMAC mimetic compound birinapant. A series of unbiased gene-trap screens identified key signaling mediators, such as TNFR1, RIPK1, RIPK3, and MLKL. Among the novel components, we focused on the zinc transporter SLC39A7, whose knock-out led to necroptosis resistance by affecting TNF receptor surface levels. Orthogonal, solute carrier (SLC)-focused CRISPR/Cas9-based genetic screens revealed the exquisite specificity of SLC39A7, among ~400 SLC genes, for TNFR1-mediated and FAS-mediated but not TRAIL-R1-mediated responses. Mechanistically, we demonstrate that loss of SLC39A7 resulted in augmented ER stress and impaired receptor trafficking, thereby globally affecting downstream signaling. The newly established cellular model also allowed genome-wide gain-of-function screening for genes conferring resistance to necroptosis via the CRISPR/Cas9-based synergistic activation mediator approach. Among these, we found cIAP1 and cIAP2, and characterized the role of TNIP1, which prevented pathway activation in a ubiquitin-binding dependent manner. Altogether, the gain-of-function and loss-of-function screens described here provide a global genetic chart of the molecular factors involved in necroptosis and death receptor signaling, prompting further investigation of their individual contribution and potential role in pathological conditions.


Subject(s)
Cation Transport Proteins/genetics , Chromosome Mapping , Necroptosis/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , CRISPR-Cas Systems/genetics , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cell Death , Cell Line , Cell Survival , HEK293 Cells , Humans , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Cell Host Microbe ; 23(6): 766-774.e5, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29779931

ABSTRACT

Macrophages represent the first line of immune defense against pathogens, and phagosome acidification is a necessary step in pathogen clearance. Here, we identified the bicarbonate transporter SLC4A7, which is strongly induced upon macrophage differentiation, as critical for phagosome acidification. Loss of SLC4A7 reduced acidification of phagocytosed beads or bacteria and impaired the intracellular microbicidal capacity in human macrophage cell lines. The phenotype was rescued by wild-type SLC4A7, but not by SLC4A7 mutants, affecting transport capacity or cell surface localization. Loss of SLC4A7 resulted in increased cytoplasmic acidification during phagocytosis, suggesting that SLC4A7-mediated, bicarbonate-driven maintenance of cytoplasmic pH is necessary for phagosome acidification. Altogether, we identify SLC4A7 and bicarbonate-driven cytoplasmic pH homeostasis as an important element of phagocytosis and the associated microbicidal functions in macrophages.


Subject(s)
Bicarbonates/metabolism , Macrophages/metabolism , Phagosomes/metabolism , Sodium-Bicarbonate Symporters/physiology , CRISPR-Cas Systems , Cation Transport Proteins/metabolism , Cytoplasm/metabolism , Gene Knockout Techniques , Homeostasis , Humans , Hydrogen-Ion Concentration , Phagocytosis , Sodium-Bicarbonate Symporters/genetics , THP-1 Cells , Transcriptome , U937 Cells
19.
Science ; 362(6419): 1171-1177, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30442766

ABSTRACT

In genetic screens aimed at understanding drug resistance mechanisms in chronic myeloid leukemia cells, inactivation of the cullin 3 adapter protein-encoding leucine zipper-like transcription regulator 1 (LZTR1) gene led to enhanced mitogen-activated protein kinase (MAPK) pathway activity and reduced sensitivity to tyrosine kinase inhibitors. Knockdown of the Drosophila LZTR1 ortholog CG3711 resulted in a Ras-dependent gain-of-function phenotype. Endogenous human LZTR1 associates with the main RAS isoforms. Inactivation of LZTR1 led to decreased ubiquitination and enhanced plasma membrane localization of endogenous KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). We propose that LZTR1 acts as a conserved regulator of RAS ubiquitination and MAPK pathway activation. Because LZTR1 disease mutations failed to revert loss-of-function phenotypes, our findings provide a molecular rationale for LZTR1 involvement in a variety of inherited and acquired human disorders.


Subject(s)
Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/physiology , Ubiquitination , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drosophila melanogaster , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gain of Function Mutation , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Loss of Function Mutation , MAP Kinase Signaling System/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Signal Transduction , Transcription Factors/genetics , Ubiquitination/genetics
20.
J Cell Biol ; 216(12): 4199-4215, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28993467

ABSTRACT

Signaling from lysosomes controls cellular clearance and energy metabolism. Lysosomal malfunction has been implicated in several pathologies, including neurodegeneration, cancer, infection, immunodeficiency, and obesity. Interestingly, many functions are dependent on the organelle position. Lysosomal motility requires the integration of extracellular and intracellular signals that converge on a competition between motor proteins that ultimately control lysosomal movement on microtubules. Here, we identify a novel upstream control mechanism of Arl8b-dependent lysosomal movement toward the periphery of the cell. We show that the C-terminal domain of lyspersin, a subunit of BLOC-1-related complex (BORC), is essential and sufficient for BORC-dependent recruitment of Arl8b to lysosomes. In addition, we establish lyspersin as the linker between BORC and late endosomal/lysosomal adaptor and mitogen activated protein kinase and mechanistic target of rapamycin activator (LAMTOR) complexes and show that epidermal growth factor stimulation decreases LAMTOR/BORC association, thereby promoting BORC- and Arl8b-dependent lysosomal centrifugal transport.


Subject(s)
ADP-Ribosylation Factors/metabolism , Carrier Proteins/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , ADP-Ribosylation Factors/genetics , Carrier Proteins/genetics , Endosomes/drug effects , Endosomes/ultrastructure , Epidermal Growth Factor/pharmacology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/drug effects , Lysosomes/ultrastructure , Microtubules/drug effects , Microtubules/metabolism , Microtubules/ultrastructure , Movement , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL