Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 41(10): 1817-1828.e9, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37683639

ABSTRACT

The dysregulated expression of immune checkpoint molecules enables cancer cells to evade immune destruction. While blockade of inhibitory immune checkpoints like PD-L1 forms the basis of current cancer immunotherapies, a deficiency in costimulatory signals can render these therapies futile. CD58, a costimulatory ligand, plays a crucial role in antitumor immune responses, but the mechanisms controlling its expression remain unclear. Using two systematic approaches, we reveal that CMTM6 positively regulates CD58 expression. Notably, CMTM6 interacts with both CD58 and PD-L1, maintaining the expression of these two immune checkpoint ligands with opposing functions. Functionally, the presence of CMTM6 and CD58 on tumor cells significantly affects T cell-tumor interactions and response to PD-L1-PD-1 blockade. Collectively, these findings provide fundamental insights into CD58 regulation, uncover a shared regulator of stimulatory and inhibitory immune checkpoints, and highlight the importance of tumor-intrinsic CMTM6 and CD58 expression in antitumor immune responses.


Subject(s)
B7-H1 Antigen , MARVEL Domain-Containing Proteins , Myelin Proteins , Neoplasms , T-Lymphocytes , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Immunity , Immunotherapy , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/immunology , Myelin Proteins/metabolism , MARVEL Domain-Containing Proteins/metabolism
2.
Nat Struct Mol Biol ; 29(6): 586-591, 2022 06.
Article in English | MEDLINE | ID: mdl-35710836

ABSTRACT

Cohesin structures the genome through the formation of chromatin loops and by holding together the sister chromatids. The acetylation of cohesin's SMC3 subunit is a dynamic process that involves the acetyltransferase ESCO1 and deacetylase HDAC8. Here we show that this cohesin acetylation cycle controls the three-dimensional genome in human cells. ESCO1 restricts the length of chromatin loops, and of architectural stripes emanating from CTCF sites. HDAC8 conversely promotes the extension of such loops and stripes. This role in controlling loop length turns out to be distinct from the canonical role of cohesin acetylation that protects against WAPL-mediated DNA release. We reveal that acetylation controls the interaction of cohesin with PDS5A to restrict chromatin loop length. Our data support a model in which this PDS5A-bound state acts as a brake that enables the pausing and restart of loop enlargement. The cohesin acetylation cycle hereby provides punctuation in the process of genome folding.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Acetylation , Cell Cycle Proteins/metabolism , Chromatids/metabolism , Chromatin , Chromosomal Proteins, Non-Histone/metabolism , Histone Deacetylases/genetics , Humans , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Cohesins
3.
Nat Med ; 25(4): 612-619, 2019 04.
Article in English | MEDLINE | ID: mdl-30833751

ABSTRACT

Cancer cells can evade immune surveillance through the expression of inhibitory ligands that bind their cognate receptors on immune effector cells. Expression of programmed death ligand 1 in tumor microenvironments is a major immune checkpoint for tumor-specific T cell responses as it binds to programmed cell death protein-1 on activated and dysfunctional T cells1. The activity of myeloid cells such as macrophages and neutrophils is likewise regulated by a balance between stimulatory and inhibitory signals. In particular, cell surface expression of the CD47 protein creates a 'don't eat me' signal on tumor cells by binding to SIRPα expressed on myeloid cells2-5. Using a haploid genetic screen, we here identify glutaminyl-peptide cyclotransferase-like protein (QPCTL) as a major component of the CD47-SIRPα checkpoint. Biochemical analysis demonstrates that QPCTL is critical for pyroglutamate formation on CD47 at the SIRPα binding site shortly after biosynthesis. Genetic and pharmacological interference with QPCTL activity enhances antibody-dependent cellular phagocytosis and cellular cytotoxicity of tumor cells. Furthermore, interference with QPCTL expression leads to a major increase in neutrophil-mediated killing of tumor cells in vivo. These data identify QPCTL as a novel target to interfere with the CD47 pathway and thereby augment antibody therapy of cancer.


Subject(s)
Aminoacyltransferases/metabolism , Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Receptors, Immunologic/metabolism , Aminoacyltransferases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Mice, Transgenic , Neoplasms/pathology , Opsonin Proteins/metabolism , Pyrrolidonecarboxylic Acid/metabolism
4.
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
5.
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
6.
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
7.
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
8.
Cell Rep ; 11(12): 1919-28, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095358

ABSTRACT

Lipid metabolism and receptor-mediated signaling are highly intertwined processes that cooperate to fulfill cellular functions and safeguard cellular homeostasis. Activation of Toll-like receptors (TLRs) leads to a complex cellular response, orchestrating a diverse range of inflammatory events that need to be tightly controlled. Here, we identified the GPI-anchored Sphingomyelin Phosphodiesterase, Acid-Like 3B (SMPDL3B) in a mass spectrometry screening campaign for membrane proteins co-purifying with TLRs. Deficiency of Smpdl3b in macrophages enhanced responsiveness to TLR stimulation and profoundly changed the cellular lipid composition and membrane fluidity. Increased cellular responses could be reverted by re-introducing affected ceramides, functionally linking membrane lipid composition and innate immune signaling. Finally, Smpdl3b-deficient mice displayed an intensified inflammatory response in TLR-dependent peritonitis models, establishing its negative regulatory role in vivo. Taken together, our results identify the membrane-modulating enzyme SMPDL3B as a negative regulator of TLR signaling that functions at the interface of membrane biology and innate immunity.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Immunity, Innate/genetics , Inflammation/genetics , Peritonitis/genetics , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Lipids/immunology , Macrophages/immunology , Mice , Peritonitis/immunology , Peritonitis/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
9.
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
10.
Nature ; 487(7408): 486-90, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22810585

ABSTRACT

Viruses must enter host cells to replicate, assemble and propagate. Because of the restricted size of their genomes, viruses have had to evolve efficient ways of exploiting host cell processes to promote their own life cycles and also to escape host immune defence mechanisms. Many viral open reading frames (viORFs) with immune-modulating functions essential for productive viral growth have been identified across a range of viral classes. However, there has been no comprehensive study to identify the host factors with which these viORFs interact for a global perspective of viral perturbation strategies. Here we show that different viral perturbation patterns of the host molecular defence network can be deduced from a mass-spectrometry-based host-factor survey in a defined human cellular system by using 70 innate immune-modulating viORFs from 30 viral species. The 579 host proteins targeted by the viORFs mapped to an unexpectedly large number of signalling pathways and cellular processes, suggesting yet unknown mechanisms of antiviral immunity. We further experimentally verified the targets heterogeneous nuclear ribonucleoprotein U, phosphatidylinositol-3-OH kinase, the WNK (with-no-lysine) kinase family and USP19 (ubiquitin-specific peptidase 19) as vulnerable nodes in the host cellular defence system. Evaluation of the impact of viral immune modulators on the host molecular network revealed perturbation strategies used by individual viruses and by viral classes. Our data are also valuable for the design of broad and specific antiviral therapies.


Subject(s)
Host-Pathogen Interactions/immunology , Viruses/immunology , Endopeptidases/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Host-Pathogen Interactions/physiology , Humans , Immunity, Innate/immunology , Mass Spectrometry , Open Reading Frames/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Reproducibility of Results , Signal Transduction , Substrate Specificity , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Viruses/metabolism
11.
J Bacteriol ; 193(3): 744-58, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115658

ABSTRACT

We report here the first demonstration of intra- and interspecies conjugative plasmid DNA transfer for Campylobacter fetus. Gene regions carried by a Campylobacter coli plasmid were identified that are sufficient for conjugative mobilization to Escherichia coli and C. fetus recipients. A broader functional range is predicted. Efficient DNA transfer involves the virB9 and virD4 genes of the type IV bacterial secretion system encoded by a pathogenicity island of C. fetus subsp. venerealis. Complementation of these phenotypes from expression constructions based on the promoter of the C. fetus surface antigen protein (sap) locus was temperature dependent, and a temperature regulation of the sap promoter was subsequently confirmed under laboratory conditions. Gene transfer was sensitive to surface or entry exclusion functions in potential recipient cells carrying IncPα plasmid RP4 implying functional relatedness to C. fetus proteins. The virB/virD4 locus is also known to be involved in bacterial invasion and killing of cultured human cells in vitro. Whether specifically secreted effector proteins contribute to host colonization and infection activities is currently unknown. Two putative effector proteins carrying an FIC domain conserved in a few bacterial type III and type IV secreted proteins of pathogens were analyzed for secretion by the C. fetus or heterologous conjugative systems. No evidence for interbacterial translocation of the Fic proteins was found.


Subject(s)
Campylobacter fetus/metabolism , Cell Surface Extensions/metabolism , Macromolecular Substances/metabolism , Membrane Transport Proteins/metabolism , Campylobacter fetus/genetics , Conjugation, Genetic , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genomic Islands , Molecular Sequence Data , Plasmids , Sequence Analysis, DNA , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...