Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Cell ; 84(10): 1904-1916.e7, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759626

ABSTRACT

Many types of human cancers suppress the expression of argininosuccinate synthase 1 (ASS1), a rate-limiting enzyme for arginine production. Although dependency on exogenous arginine can be harnessed by arginine-deprivation therapies, the impact of ASS1 suppression on the quality of the tumor proteome is unknown. We therefore interrogated proteomes of cancer patients for arginine codon reassignments (substitutants) and surprisingly identified a strong enrichment for cysteine (R>C) in lung tumors specifically. Most R>C events did not coincide with genetically encoded R>C mutations but were likely products of tRNA misalignments. The expression of R>C substitutants was highly associated with oncogenic kelch-like epichlorohydrin (ECH)-associated protein 1 (KEAP1)-pathway mutations and suppressed by intact-KEAP1 in KEAP1-mutated cancer cells. Finally, functional interrogation indicated a key role for R>C substitutants in cell survival to cisplatin, suggesting that regulatory codon reassignments endow cancer cells with more resilience to stress. Thus, we present a mechanism for enriching lung cancer proteomes with cysteines that may affect therapeutic decisions.


Subject(s)
Arginine , Cysteine , Kelch-Like ECH-Associated Protein 1 , Lung Neoplasms , Proteome , Humans , Cysteine/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Proteome/metabolism , Arginine/metabolism , Mutation , Argininosuccinate Synthase/metabolism , Argininosuccinate Synthase/genetics , Cisplatin/pharmacology , Cell Line, Tumor , Proteomics/methods , Gene Expression Regulation, Neoplastic , Cell Survival/drug effects , RNA, Transfer/metabolism , RNA, Transfer/genetics
2.
Mol Cell ; 81(22): 4709-4721.e9, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34562372

ABSTRACT

mRNA translation is a highly conserved and tightly controlled mechanism for protein synthesis. Despite protein quality control mechanisms, amino acid shortage in melanoma induces aberrant proteins by ribosomal frameshifting. The extent and the underlying mechanisms related to this phenomenon are yet unknown. Here, we show that tryptophan depletion-induced ribosomal frameshifting is a widespread phenomenon in cancer. We termed this event sloppiness and strikingly observed its association with MAPK pathway hyperactivation. Sloppiness is stimulated by RAS activation in primary cells, suppressed by pharmacological inhibition of the oncogenic MAPK pathway in sloppy cells, and restored in cells with acquired resistance to MAPK pathway inhibition. Interestingly, sloppiness causes aberrant peptide presentation at the cell surface, allowing recognition and specific killing of drug-resistant cancer cells by T lymphocytes. Thus, while oncogenes empower cancer progression and aggressiveness, they also expose a vulnerability by provoking the production of aberrant peptides through sloppiness.


Subject(s)
Neoplasms/genetics , Oncogenes , Protein Biosynthesis , RNA, Messenger/metabolism , T-Lymphocytes/cytology , Animals , Carcinogenesis , Cell Membrane/metabolism , Disease Progression , Drug Resistance, Neoplasm , Frameshift Mutation , Frameshifting, Ribosomal , Humans , Immunotherapy/methods , MAP Kinase Signaling System , Melanoma/metabolism , Mice , Neoplasms/metabolism , Peptides/chemistry , Protein Kinase Inhibitors , Ribosomes/metabolism , T-Lymphocytes/metabolism , Tryptophan/chemistry , Tryptophan/metabolism
3.
Nature ; 603(7902): 721-727, 2022 03.
Article in English | MEDLINE | ID: mdl-35264796

ABSTRACT

Activated T cells secrete interferon-γ, which triggers intracellular tryptophan shortage by upregulating the indoleamine 2,3-dioxygenase 1 (IDO1) enzyme1-4. Here we show that despite tryptophan depletion, in-frame protein synthesis continues across tryptophan codons. We identified tryptophan-to-phenylalanine codon reassignment (W>F) as the major event facilitating this process, and pinpointed tryptophanyl-tRNA synthetase (WARS1) as its source. We call these W>F peptides 'substitutants' to distinguish them from genetically encoded mutants. Using large-scale proteomics analyses, we demonstrate W>F substitutants to be highly abundant in multiple cancer types. W>F substitutants were enriched in tumours relative to matching adjacent normal tissues, and were associated with increased IDO1 expression, oncogenic signalling and the tumour-immune microenvironment. Functionally, W>F substitutants can impair protein activity, but also expand the landscape of antigens presented at the cell surface to activate T cell responses. Thus, substitutants are generated by an alternative decoding mechanism with potential effects on gene function and tumour immunoreactivity.


Subject(s)
Tryptophan-tRNA Ligase , Tryptophan , Codon/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma , Neoplasms/immunology , Phenylalanine , T-Lymphocytes , Tryptophan/metabolism , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism , Tryptophan-tRNA Ligase/genetics , Tryptophan-tRNA Ligase/metabolism
4.
Nature ; 590(7845): 332-337, 2021 02.
Article in English | MEDLINE | ID: mdl-33328638

ABSTRACT

Extensive tumour inflammation, which is reflected by high levels of infiltrating T cells and interferon-γ (IFNγ) signalling, improves the response of patients with melanoma to checkpoint immunotherapy1,2. Many tumours, however, escape by activating cellular pathways that lead to immunosuppression. One such mechanism is the production of tryptophan metabolites along the kynurenine pathway by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which is induced by IFNγ3-5. However, clinical trials using inhibition of IDO1 in combination with blockade of the PD1 pathway in patients with melanoma did not improve the efficacy of treatment compared to PD1 pathway blockade alone6,7, pointing to an incomplete understanding of the role of IDO1 and the consequent degradation of tryptophan in mRNA translation and cancer progression. Here we used ribosome profiling in melanoma cells to investigate the effects of prolonged IFNγ treatment on mRNA translation. Notably, we observed accumulations of ribosomes downstream of tryptophan codons, along with their expected stalling at the tryptophan codon. This suggested that ribosomes bypass tryptophan codons in the absence of tryptophan. A detailed examination of these tryptophan-associated accumulations of ribosomes-which we term 'W-bumps'-showed that they were characterized by ribosomal frameshifting events. Consistently, reporter assays combined with proteomic and immunopeptidomic analyses demonstrated the induction of ribosomal frameshifting, and the generation and presentation of aberrant trans-frame peptides at the cell surface after treatment with IFNγ. Priming of naive T cells from healthy donors with aberrant peptides induced peptide-specific T cells. Together, our results suggest that IDO1-mediated depletion of tryptophan, which is induced by IFNγ, has a role in the immune recognition of melanoma cells by contributing to diversification of the peptidome landscape.


Subject(s)
Antigen Presentation , Frameshift Mutation , Melanoma/immunology , Peptides/genetics , Peptides/immunology , Protein Biosynthesis/immunology , T-Lymphocytes/immunology , Cell Line , Codon/genetics , Frameshifting, Ribosomal/drug effects , Frameshifting, Ribosomal/genetics , Frameshifting, Ribosomal/immunology , Histocompatibility Antigens Class I/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Melanoma/pathology , Peptides/chemistry , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Proteome , Ribosomes/drug effects , Ribosomes/metabolism , Tryptophan/deficiency , Tryptophan/genetics , Tryptophan/metabolism
5.
EMBO Rep ; 25(5): 2220-2238, 2024 May.
Article in English | MEDLINE | ID: mdl-38600345

ABSTRACT

Perturbation of protein phosphorylation represents an attractive approach to cancer treatment. Besides kinase inhibitors, protein phosphatase inhibitors have been shown to have anti-cancer activity. A prime example is the small molecule LB-100, an inhibitor of protein phosphatases 2A/5 (PP2A/PP5), enzymes that affect cellular physiology. LB-100 has proven effective in pre-clinical models in combination with immunotherapy, but the molecular underpinnings of this synergy remain understood poorly. We report here a sensitivity of the mRNA splicing machinery to phosphorylation changes in response to LB-100 in colorectal adenocarcinoma. We observe enrichment for differentially phosphorylated sites within cancer-critical splicing nodes of U2 snRNP, SRSF and hnRNP proteins. Altered phosphorylation endows LB-100-treated colorectal adenocarcinoma cells with differential splicing patterns. In PP2A-inhibited cells, over 1000 events of exon skipping and intron retention affect regulators of genomic integrity. Finally, we show that LB-100-evoked alternative splicing leads to neoantigens that are presented by MHC class 1 at the cell surface. Our findings provide a potential explanation for the pre-clinical and clinical observations that LB-100 sensitizes cancer cells to immune checkpoint blockade.


Subject(s)
Colonic Neoplasms , RNA Splicing , Humans , Alternative Splicing/drug effects , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Phosphorylation , Protein Phosphatase 2/metabolism , RNA Splicing/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Piperazines/pharmacology
6.
Mol Cell Proteomics ; 22(6): 100548, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37059365

ABSTRACT

Ubiquitination has crucial roles in many cellular processes, and dysregulation of ubiquitin machinery enzymes can result in various forms of pathogenesis. Cells only have a limited set of ubiquitin-conjugating (E2) enzymes to support the ubiquitination of many cellular targets. As individual E2 enzymes have many different substrates and interactions between E2 enzymes and their substrates can be transient, it is challenging to define all in vivo substrates of an individual E2 and the cellular processes it affects. Particularly challenging in this respect is UBE2D3, an E2 enzyme with promiscuous activity in vitro but less defined roles in vivo. Here, we set out to identify in vivo targets of UBE2D3 by using stable isotope labeling by amino acids in cell culture-based and label-free quantitative ubiquitin diGly proteomics to study global proteome and ubiquitinome changes associated with UBE2D3 depletion. UBE2D3 depletion changed the global proteome, with the levels of proteins from metabolic pathways, in particular retinol metabolism, being the most affected. However, the impact of UBE2D3 depletion on the ubiquitinome was much more prominent. Interestingly, molecular pathways related to mRNA translation were the most affected. Indeed, we find that ubiquitination of the ribosomal proteins RPS10 and RPS20, critical for ribosome-associated protein quality control, is dependent on UBE2D3. We show by Targets of Ubiquitin Ligases Identified by Proteomics 2 methodology that RPS10 and RPS20 are direct targets of UBE2D3 and demonstrate that the catalytic activity of UBE2D3 is required to ubiquitinate RPS10 in vivo. In addition, our data suggest that UBE2D3 acts at multiple levels in autophagic protein quality control. Collectively, our findings show that depletion of an E2 enzyme in combination with quantitative diGly-based ubiquitinome profiling is a powerful tool to identify new in vivo E2 substrates, as we have done here for UBE2D3. Our work provides an important resource for further studies on the in vivo functions of UBE2D3.


Subject(s)
Proteome , Ubiquitin , Proteome/metabolism , Ubiquitination , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
J Cell Sci ; 135(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35532004

ABSTRACT

The vitronectin receptor integrin αVß5 can reside in two distinct adhesion structures - focal adhesions (FAs) and flat clathrin lattices (FCLs). Here, we investigate the mechanism that regulates the subcellular distribution of ß5 in keratinocytes and show that ß5 has approximately 7- and 5-fold higher affinity for the clathrin adaptors ARH (also known as LDLRAP1) and Numb, respectively, than for the talin 1 (TLN1); all proteins that bind to the membrane-proximal NPxY motif of the ß5 cytoplasmic domain. Using mass spectrometry, we identified ß5 interactors, including the Rho GEFs p115Rho-GEF and GEF-H1 (also known as ARHGEF1 and ARHGEF2, respectively), and the serine protein kinase MARK2, depletion of which diminishes the clustering of ß5 in FCLs. Replacement of two serine residues (S759 and S762) in the ß5 cytoplasmic domain with phospho-mimetic glutamate residues causes a shift in the localization of ß5 from FAs into FCLs without affecting the interactions with MARK2, p115Rho-GEF or GEF-H1. Instead, we demonstrate that changes in the actomyosin-based cellular contractility by ectopic expression of activated Rho or disruption of microtubules regulates ß5 localization. Finally, we present evidence that ß5 in either FAs or FCLs functions to promote adhesion to vitronectin, cell spreading, and proliferation.


Subject(s)
Clathrin , Receptors, Vitronectin , Cell Adhesion/physiology , Cell Proliferation , Clathrin/metabolism , Focal Adhesions/metabolism , Receptors, Vitronectin/metabolism , Serine/metabolism
8.
Nature ; 546(7657): 307-311, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28562590

ABSTRACT

As key executers of biological functions, the activity and abundance of proteins are subjected to extensive regulation. Deciphering the genetic architecture underlying this regulation is critical for understanding cellular signalling events and responses to environmental cues. Using random mutagenesis in haploid human cells, we apply a sensitive approach to directly couple genomic mutations to protein measurements in individual cells. Here we use this to examine a suite of cellular processes, such as transcriptional induction, regulation of protein abundance and splicing, signalling cascades (mitogen-activated protein kinase (MAPK), G-protein-coupled receptor (GPCR), protein kinase B (AKT), interferon, and Wingless and Int-related protein (WNT) pathways) and epigenetic modifications (histone crotonylation and methylation). This scalable, sequencing-based procedure elucidates the genetic landscapes that control protein states, identifying genes that cause very narrow phenotypic effects and genes that lead to broad phenotypic consequences. The resulting genetic wiring map identifies the E3-ligase substrate adaptor KCTD5 (ref. 1) as a negative regulator of the AKT pathway, a key signalling cascade frequently deregulated in cancer. KCTD5-deficient cells show elevated levels of phospho-AKT at S473 that could not be attributed to effects on canonical pathway components. To reveal the genetic requirements for this phenotype, we iteratively analysed the regulatory network linked to AKT activity in the knockout background. This genetic modifier screen exposes suppressors of the KCTD5 phenotype and mechanistically demonstrates that KCTD5 acts as an off-switch for GPCR signalling by triggering proteolysis of Gßγ heterodimers dissociated from the Gα subunit. Although biological networks have previously been constructed on the basis of gene expression, protein-protein associations, or genetic interaction profiles, we foresee that the approach described here will enable the generation of a comprehensive genetic wiring map for human cells on the basis of quantitative protein states.


Subject(s)
Potassium Channels/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Single-Cell Analysis/methods , Cells, Cultured , Haploidy , Heterotrimeric GTP-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Humans , Interferons/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Phenotype , Phosphorylation/genetics , Potassium Channels/deficiency , Potassium Channels/genetics , Proteolysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway
10.
J Cell Sci ; 131(21)2018 11 05.
Article in English | MEDLINE | ID: mdl-30301780

ABSTRACT

The family of integrin transmembrane receptors is essential for the normal function of multicellular organisms by facilitating cell-extracellular matrix adhesion. The vitronectin-binding integrin αVß5 localizes to focal adhesions (FAs) as well as poorly characterized flat clathrin lattices (FCLs). Here, we show that, in human keratinocytes, αVß5 is predominantly found in FCLs, and formation of the αVß5-containing FCLs requires the presence of vitronectin as ligand, Ca2+, and the clathrin adaptor proteins ARH (also known as LDLRAP1), Numb and EPS15/EPS15L1. Integrin chimeras, containing the extracellular and transmembrane domains of ß5 and the cytoplasmic domains of ß1 or ß3, almost exclusively localize in FAs. Interestingly, lowering actomyosin-mediated contractility promotes integrin redistribution to FLCs in an integrin tail-dependent manner, while increasing cellular tension favors αVß5 clustering in FAs. Our findings strongly indicate that clustering of integrin αVß5 in FCLs is dictated by the ß5 subunit cytoplasmic domain, cellular tension and recruitment of specific adaptor proteins to the ß5 subunit cytoplasmic domains.


Subject(s)
Clathrin/metabolism , Receptors, Vitronectin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Calcium/metabolism , Cells, Cultured , Focal Adhesions/metabolism , Humans , Keratinocytes/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Vitronectin/metabolism
11.
Proc Natl Acad Sci U S A ; 112(21): 6601-6, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25964343

ABSTRACT

Despite technological advances in metabolomics, large parts of the human metabolome are still unexplored. In an untargeted metabolomics screen aiming to identify substrates of the orphan transporter ATP-binding cassette subfamily C member 5 (ABCC5), we identified a class of mammalian metabolites, N-lactoyl-amino acids. Using parallel protein fractionation in conjunction with shotgun proteomics on fractions containing N-lactoyl-Phe-forming activity, we unexpectedly found that a protease, cytosolic nonspecific dipeptidase 2 (CNDP2), catalyzes their formation. N-lactoyl-amino acids are ubiquitous pseudodipeptides of lactic acid and amino acids that are rapidly formed by reverse proteolysis, a process previously considered to be negligible in vivo. The plasma levels of these metabolites strongly correlate with plasma levels of lactate and amino acid, as shown by increased levels after physical exercise and in patients with phenylketonuria who suffer from elevated Phe levels. Our approach to identify unknown metabolites and their biosynthesis has general applicability in the further exploration of the human metabolome.


Subject(s)
Amino Acids/metabolism , Dipeptidases/metabolism , Lactates/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Amino Acids/blood , Cytosol/metabolism , Exercise/physiology , HEK293 Cells , Humans , Lactates/blood , Metabolome , Proteolysis
12.
EMBO Rep ; 16(12): 1723-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26538417

ABSTRACT

c-Myc is one of the major human proto-oncogenes and is often associated with tumor aggression and poor clinical outcome. Paradoxically, Myc was also reported as a suppressor of cell motility, invasiveness, and metastasis. Among the direct targets of Myc are many components of the protein synthesis machinery whose induction results in an overall increase in protein synthesis that empowers tumor cell growth. At present, it is largely unknown whether beyond the global enhancement of protein synthesis, Myc activation results in translation modulation of specific genes. Here, we measured Myc-induced global changes in gene expression at the transcription, translation, and protein levels and uncovered extensive transcript-specific regulation of protein translation. Particularly, we detected a broad coordination between regulation of transcription and translation upon modulation of Myc activity and showed the connection of these responses to mTOR signaling to enhance oncogenic transformation and to the TGFß pathway to modulate cell migration and invasiveness. Our results elucidate novel facets of Myc-induced cellular responses and provide a more comprehensive view of the consequences of its activation in cancer cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Apoptosis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Profiling , Genes, myc , Humans , Protein Biosynthesis , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
13.
J Proteome Res ; 15(12): 4624-4637, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27769112

ABSTRACT

Formin mDia2 is a cytoskeleton-regulatory protein that switches reversibly between a closed, autoinhibited and an open, active conformation. Although the open conformation of mDia2 induces actin assembly thereby controlling many cellular processes, mDia2 possesses also actin-independent and conformation-insensitive scaffolding roles related to microtubules and p53, respectively. Thus, we hypothesize that mDia2 may have other unappreciated functions and regulatory modes. Here we identify and validate proteasome and Ubiquitin as mDia2-interacting partners using stable isotope labeling with amino acids in cell culture-based quantitative proteomics and biochemistry, respectively. Although mDia2 is ubiquitinated, binds ubiquitinated proteins and free Ubiquitin, it is not a proteasome substrate. Surprisingly, knockdown of mDia2 increases the activity of the proteasome in vitro, whereas mDia2 overexpression has opposite effects only when it adopts the open conformation and cannot induce actin assembly. Consistently, a combination of candidate and unbiased proteome-wide analyses indicates that mDia2 regulates the cellular levels of proteasome substrate ß-catenin and a number of ubiquitinated actin-regulatory proteins. Hence, these findings add more complexity to the mDia2 activity cycle by showing that the open conformation may control actin dynamics also through actin-independent regulation of the proteasome.


Subject(s)
Microtubule-Associated Proteins/metabolism , NADPH Dehydrogenase/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics/methods , Actins/metabolism , Animals , Isotope Labeling , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/physiology , NADPH Dehydrogenase/chemistry , NADPH Dehydrogenase/physiology , Protein Conformation , Protein Interaction Mapping , Ubiquitin/metabolism
14.
Arterioscler Thromb Vasc Biol ; 33(7): 1635-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640497

ABSTRACT

OBJECTIVE: Platelet activation and subsequent protein release play an important role in healthy hemostasis and inflammatory responses, yet the identity and quantity of proteins in the platelet releasate are still debated. Here, we present a reversed releasate proteomics approach to determine unambiguously and quantitatively proteins released from activated platelets. APPROACH AND RESULTS: Isolated platelets were mock and fully stimulated after which the released proteins in the supernatant were removed. Using high-end proteomics technology (2D chromatography, stable isotope labeling, electron transfer dissociation, and high collision dissociation fragmentation) allowed us to quantitatively discriminate the released proteins from uncontrolled lysis products. Monitoring the copy numbers of ≈ 4500 platelet proteins, we observed that after stimulation via thrombin and collagen, only 124 (<3%) proteins were significantly released (P<0.05). The released proteins span a concentration range of ≥ 5 orders, as confirmed by ELISA. The released proteins were highly enriched in secretion tags and contained all known factors at high concentrations (>100 ng/mL, eg, thrombospondin, von Willebrand factor, and platelet factor 4). Interestingly, in the lower concentration range of the releasate many novel factors were identified. CONCLUSIONS: Our reversed releasate dataset forms the first unambiguous, in depth repository for molecular factors released by platelets.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Proteins/metabolism , Proteomics/methods , Chromatography, Ion Exchange , Chromatography, Liquid , Collagen/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Isotope Labeling , Tandem Mass Spectrometry , Thrombin/metabolism
15.
Arterioscler Thromb Vasc Biol ; 33(7): 1538-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23619296

ABSTRACT

OBJECTIVE: Platelet adhesion to subendothelial collagen is dependent on the integrin α2ß1 and glycoprotein VI (GPVI) receptors. The major signaling routes in collagen-dependent platelet activation are outlined; however, crucial detailed knowledge of the actual phosphorylation events mediating them is still limited. Here, we explore phosphotyrosine signaling events downstream of GPVI with site-specific detail. APPROACH AND RESULTS: Immunoprecipitations of phosphotyrosine-modified peptides from protein digests of GPVI-activated and resting human platelets were compared by stable isotope-based quantitative mass spectrometry. We surveyed 214 unique phosphotyrosine sites over 2 time points, of which 28 showed a significant increase in phosphorylation on GPVI activation. Among these was Tyr370 of oligophrenin-1 (OPHN1), a Rho GTPase-activating protein. To elucidate the function of OPHN1 in platelets, we performed an array of functional platelet analyses within a small cohort of patients with rare oligophrenia. Because of germline mutations in the OPHN1 gene locus, these patients lack OPHN1 expression entirely and are in essence a human knockout model. Our studies revealed that among other unaltered properties, patients with oligophrenia show normal P-selectin exposure and αIIbß3 activation in response to GPVI, as well as normal aggregate formation on collagen under shear conditions. Finally, the major difference in OPHN1-deficient platelets turned out to be a significantly reduced collagen-induced filopodia formation. CONCLUSIONS: In-depth phosphotyrosine screening revealed many novel signaling recipients downstream of GPVI activation uncovering a new level of detail within this important pathway. To illustrate the strength of such data, functional follow-up of OPHN1 in human platelets deficient in this protein showed reduced filopodia formation on collagen, an important parameter of platelet hemostatic function.


Subject(s)
Blood Platelets/metabolism , Cytoskeletal Proteins/blood , GTPase-Activating Proteins/blood , Metabolism, Inborn Errors/blood , Nuclear Proteins/blood , Platelet Membrane Glycoproteins/metabolism , Pseudopodia/metabolism , Signal Transduction , Case-Control Studies , Child , Collagen/metabolism , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Hemostasis , Humans , Immunoprecipitation , Male , Mass Spectrometry , Metabolism, Inborn Errors/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , P-Selectin/blood , Phosphorylation , Platelet Adhesiveness , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Proteomics/methods , Stress, Mechanical , Time Factors , Tyrosine
16.
Nat Commun ; 15(1): 5032, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866770

ABSTRACT

Maintenance of genome integrity requires tight control of DNA damage response (DDR) signalling and repair, with phosphorylation and ubiquitination representing key elements. How these events are coordinated to achieve productive DNA repair remains elusive. Here we identify the ubiquitin-conjugating enzyme UBE2D3 as a regulator of ATM kinase-induced DDR that promotes non-homologous end-joining (NHEJ) at telomeres. UBE2D3 contributes to DDR-induced chromatin ubiquitination and recruitment of the NHEJ-promoting factor 53BP1, both mediated by RNF168 upon ATM activation. Additionally, UBE2D3 promotes NHEJ by limiting RNF168 accumulation and facilitating ATM-mediated phosphorylation of KAP1-S824. Mechanistically, defective KAP1-S824 phosphorylation and telomeric NHEJ upon UBE2D3-deficiency are linked to RNF168 hyperaccumulation and aberrant PP2A phosphatase activity. Together, our results identify UBE2D3 as a multi-level regulator of NHEJ that orchestrates ATM and RNF168 activities. Moreover, they reveal a negative regulatory circuit in the DDR that is constrained by UBE2D3 and consists of RNF168- and phosphatase-mediated restriction of KAP1 phosphorylation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA End-Joining Repair , Signal Transduction , Tripartite Motif-Containing Protein 28 , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Phosphorylation , Tripartite Motif-Containing Protein 28/metabolism , Tripartite Motif-Containing Protein 28/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , HEK293 Cells , Telomere/metabolism , DNA Damage , Chromatin/metabolism , Animals
17.
Cancer Cell ; 42(4): 623-645.e10, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38490212

ABSTRACT

Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Neoplasms/genetics
18.
Biomed Pharmacother ; 164: 114956, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37267638

ABSTRACT

Mammalian carboxylesterase 1 enzymes can hydrolyze many xenobiotic chemicals and endogenous lipids. We here identified and characterized a mouse strain (FVB/NKI) in which three of the eight Ces1 genes were spontaneously deleted, removing Ces1c and Ces1e partly, and Ces1d entirely. We studied the impact of this Ces1c/d/e deficiency on drug and lipid metabolism and homeostasis. Ces1c/d/e-/- mice showed strongly impaired conversion of the anticancer prodrug irinotecan to its active metabolite SN-38 in plasma, spleen and lung. Plasma hydrolysis of the oral anticancer prodrug capecitabine to 5-DFCR was also profoundly reduced in Ces1c/d/e-/- mice. Our findings resolved previously unexplained FVB/NKI pharmacokinetic anomalies. On a medium-fat diet, Ces1c/d/e-/- female mice exhibited moderately higher body weight, mild inflammation in gonadal white adipose tissue (gWAT), and increased lipid load in brown adipose tissue (BAT). Ces1c/d/e-/- males showed more pronounced inflammation in gWAT and an increased lipid load in BAT. On a 5-week high-fat diet exposure, Ces1c/d/e deficiency predisposed to developing obesity, enlarged and fatty liver, glucose intolerance and insulin resistance, with severe inflammation in gWAT and increased lipid load in BAT. Hepatic proteomics analysis revealed that the acute phase response, involved in the dynamic cycle of immunometabolism, was activated in these Ces1c/d/e-/- mice. This may contribute to the obesity-related chronic inflammation and adverse metabolic disease in this strain. While Ces1c/d/e deficiency clearly exacerbated metabolic syndrome development, long-term (18-week) high-fat diet exposure overwhelmed many, albeit not all, observed phenotypic differences.


Subject(s)
Carboxylic Ester Hydrolases , Metabolic Syndrome , Prodrugs , Animals , Female , Male , Mice , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Inflammation , Irinotecan , Lipids , Mammals , Obesity/metabolism
19.
J Proteome Res ; 11(11): 5235-44, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23020738

ABSTRACT

Coronary atherosclerosis represents the major cause of death in Western societies. As atherosclerosis typically progresses over years without giving rise to clinical symptoms, biomarkers are urgently needed to identify patients at risk. Over the past decade, evidence has accumulated suggesting cross-talk between the diseased vasculature and cells of the innate immune system. We therefore employed proteomics to search for biomarkers associated with severe atherosclerotic coronary lumen stenosis in circulating leukocytes. In a two-phase approach, we first performed in-depth quantitative profiling of the granulocyte proteome on a small pooled cohort of patients suffering from chronic (sub)total coronary occlusion and matched control patients using stable isotope peptide labeling, two-dimensional LC-MS/MS and data-dependent decision tree fragmentation. Over 3000 proteins were quantified, among which 57 candidate biomarker proteins remained after stringent filtering. The most promising biomarker candidates were subsequently verified in the individual samples of the discovery cohort using label-free, single-run LC-MS/MS analysis, as well as in an independent verification cohort of 25 patients with total coronary occlusion (CTO) and 19 matched controls. Our data reveal bactericidal/permeability-increasing protein (BPI) as a promising biomarker for severe atherosclerotic coronary stenosis, being down-regulated in circulating granulocytes of CTO patients.


Subject(s)
Antimicrobial Cationic Peptides/blood , Biomarkers/blood , Coronary Artery Disease/blood , Coronary Stenosis/blood , Granulocytes/metabolism , Proteome , Blood Proteins , Cell Separation , Cohort Studies , Humans
20.
J Cell Biol ; 221(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35687021

ABSTRACT

Integrins mediate cell adhesion by connecting the extracellular matrix to the intracellular cytoskeleton and orchestrate signal transduction in response to chemical and mechanical stimuli by interacting with many cytoplasmic proteins. We used BioID to interrogate the interactomes of ß1 and ß3 integrins in epithelial cells and identified PEAK1 as an interactor of the RGD-binding integrins α5ß1, αVß3, and αVß5 in focal adhesions. We demonstrate that the interaction between integrins and PEAK1 occurs indirectly through Tensin3, requiring both the membrane-proximal NPxY motif on the integrin ß tail and binding of the SH2 domain of Tensin3 to phosphorylated Tyr-635 on PEAK1. Phosphorylation of Tyr-635 is mediated by Src and regulates cell migration. Additionally, we found that Shc1 localizes in focal adhesions in a PEAK1 phosphorylated Tyr-1188-dependent fashion. Besides binding Shc1, PEAK1 also associates with a protein cluster that mediates late EGFR/Shc1 signaling. We propose a model in which PEAK1 binds Tensin3 and Shc1 to converge integrin and growth factor receptor signal transduction.


Subject(s)
Cell Adhesion , Integrins , Protein-Tyrosine Kinases , Tensins , Cell Movement , Focal Adhesions/metabolism , Humans , Integrin beta3/metabolism , Integrins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tensins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL