Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
EMBO J ; 43(6): 904-930, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38337057

ABSTRACT

Mitochondrial outer membrane permeabilisation (MOMP) is often essential for apoptosis, by enabling cytochrome c release that leads to caspase activation and rapid cell death. Recently, MOMP has been shown to be inherently pro-inflammatory with emerging cellular roles, including its ability to elicit anti-tumour immunity. Nonetheless, how MOMP triggers inflammation and how the cell regulates this remains poorly defined. We find that upon MOMP, many proteins localised either to inner or outer mitochondrial membranes are ubiquitylated in a promiscuous manner. This extensive ubiquitylation serves to recruit the essential adaptor molecule NEMO, leading to the activation of pro-inflammatory NF-κB signalling. We show that disruption of mitochondrial outer membrane integrity through different means leads to the engagement of a similar pro-inflammatory signalling platform. Therefore, mitochondrial integrity directly controls inflammation, such that permeabilised mitochondria initiate NF-κB signalling.


Subject(s)
NF-kappa B , Ubiquitin , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Ubiquitin/metabolism , Mitochondrial Membranes/metabolism , Mitochondria/metabolism , Apoptosis/physiology , Inflammation/metabolism
2.
EMBO J ; 42(18): e113987, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37577760

ABSTRACT

Dysregulation of the PI3K/AKT pathway is a common occurrence in high-grade serous ovarian carcinoma (HGSOC), with the loss of the tumour suppressor PTEN in HGSOC being associated with poor prognosis. The cellular mechanisms of how PTEN loss contributes to HGSOC are largely unknown. We here utilise time-lapse imaging of HGSOC spheroids coupled to a machine learning approach to classify the phenotype of PTEN loss. PTEN deficiency induces PI(3,4,5)P3 -rich and -dependent membrane protrusions into the extracellular matrix (ECM), resulting in a collective invasion phenotype. We identify the small GTPase ARF6 as a crucial vulnerability of HGSOC cells upon PTEN loss. Through a functional proteomic CRISPR screen of ARF6 interactors, we identify the ARF GTPase-activating protein (GAP) AGAP1 and the ECM receptor ß1-integrin (ITGB1) as key ARF6 interactors in HGSOC regulating PTEN loss-associated invasion. ARF6 functions to promote invasion by controlling the recycling of internalised, active ß1-integrin to maintain invasive activity into the ECM. The expression of the CYTH2-ARF6-AGAP1 complex in HGSOC patients is inversely associated with outcome, allowing the identification of patient groups with improved versus poor outcome. ARF6 may represent a therapeutic vulnerability in PTEN-depleted HGSOC.


Subject(s)
Monomeric GTP-Binding Proteins , Ovarian Neoplasms , Humans , Female , Integrins/metabolism , Proteomics , Phosphatidylinositol 3-Kinases/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Monomeric GTP-Binding Proteins/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
3.
Nucleic Acids Res ; 52(5): 2389-2415, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38224453

ABSTRACT

DNA damage represents a challenge for cells, as this damage must be eliminated to preserve cell viability and the transmission of genetic information. To reduce or eliminate unscheduled chemical modifications in genomic DNA, an extensive signaling network, known as the DNA damage response (DDR) pathway, ensures this repair. In this work, and by means of a proteomic analysis aimed at studying the STIM1 protein interactome, we have found that STIM1 is closely related to the protection from endogenous DNA damage, replicative stress, as well as to the response to interstrand crosslinks (ICLs). Here we show that STIM1 has a nuclear localization signal that mediates its translocation to the nucleus, and that this translocation and the association of STIM1 to chromatin increases in response to mitomycin-C (MMC), an ICL-inducing agent. Consequently, STIM1-deficient cell lines show higher levels of basal DNA damage, replicative stress, and increased sensitivity to MMC. We show that STIM1 normalizes FANCD2 protein levels in the nucleus, which explains the increased sensitivity of STIM1-KO cells to MMC. This study not only unveils a previously unknown nuclear function for the endoplasmic reticulum protein STIM1 but also expands our understanding of the genes involved in DNA repair.


Subject(s)
Cell Nucleus , DNA Damage , Stromal Interaction Molecule 1 , Chromatin/genetics , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Mitomycin/pharmacology , Proteomics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Humans , Cell Nucleus/metabolism , Neoplasm Proteins/metabolism
4.
Circ Res ; 132(7): 828-848, 2023 03 31.
Article in English | MEDLINE | ID: mdl-36883446

ABSTRACT

BACKGROUND: Signaling by cAMP is organized in multiple distinct subcellular nanodomains regulated by cAMP-hydrolyzing PDEs (phosphodiesterases). Cardiac ß-adrenergic signaling has served as the prototypical system to elucidate cAMP compartmentalization. Although studies in cardiac myocytes have provided an understanding of the location and properties of a handful of cAMP subcellular compartments, an overall view of the cellular landscape of cAMP nanodomains is missing. METHODS: Here, we combined an integrated phosphoproteomics approach that takes advantage of the unique role that individual PDEs play in the control of local cAMP, with network analysis to identify previously unrecognized cAMP nanodomains associated with ß-adrenergic stimulation. We then validated the composition and function of one of these nanodomains using biochemical, pharmacological, and genetic approaches and cardiac myocytes from both rodents and humans. RESULTS: We demonstrate the validity of the integrated phosphoproteomic strategy to pinpoint the location and provide critical cues to determine the function of previously unknown cAMP nanodomains. We characterize in detail one such compartment and demonstrate that the PDE3A2 isoform operates in a nuclear nanodomain that involves SMAD4 (SMAD family member 4) and HDAC-1 (histone deacetylase 1). Inhibition of PDE3 results in increased HDAC-1 phosphorylation, leading to inhibition of its deacetylase activity, derepression of gene transcription, and cardiac myocyte hypertrophic growth. CONCLUSIONS: We developed a strategy for detailed mapping of subcellular PDE-specific cAMP nanodomains. Our findings reveal a mechanism that explains the negative long-term clinical outcome observed in patients with heart failure treated with PDE3 inhibitors.


Subject(s)
Cyclic AMP , Myocytes, Cardiac , Humans , Proteomics , Phosphoric Diester Hydrolases , Hypertrophy , Adrenergic Agents
6.
Nucleic Acids Res ; 51(4): 1859-1879, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36727461

ABSTRACT

Altered eIF4A1 activity promotes translation of highly structured, eIF4A1-dependent oncogene mRNAs at root of oncogenic translational programmes. It remains unclear how these mRNAs recruit and activate eIF4A1 unwinding specifically to facilitate their preferential translation. Here, we show that single-stranded RNA sequence motifs specifically activate eIF4A1 unwinding allowing local RNA structural rearrangement and translation of eIF4A1-dependent mRNAs in cells. Our data demonstrate that eIF4A1-dependent mRNAs contain AG-rich motifs within their 5'UTR which specifically activate eIF4A1 unwinding of local RNA structure to facilitate translation. This mode of eIF4A1 regulation is used by mRNAs encoding components of mTORC-signalling and cell cycle progression, and renders these mRNAs particularly sensitive to eIF4A1-inhibition. Mechanistically, we show that binding of eIF4A1 to AG-rich sequences leads to multimerization of eIF4A1 with eIF4A1 subunits performing distinct enzymatic activities. Our structural data suggest that RNA-binding of multimeric eIF4A1 induces conformational changes in the RNA resulting in an optimal positioning of eIF4A1 proximal to the RNA duplex enabling efficient unwinding. Our data proposes a model in which AG-motifs in the 5'UTR of eIF4A1-dependent mRNAs specifically activate eIF4A1, enabling assembly of the helicase-competent multimeric eIF4A1 complex, and positioning these complexes proximal to stable localised RNA structure allowing ribosomal subunit scanning.


Subject(s)
Eukaryotic Initiation Factor-4A , Protein Biosynthesis , 5' Untranslated Regions , Purines , RNA, Messenger/metabolism , Humans , Eukaryotic Initiation Factor-4A/metabolism
7.
Proc Natl Acad Sci U S A ; 119(26): e2111506119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35737835

ABSTRACT

Macroautophagy promotes cellular homeostasis by delivering cytoplasmic constituents to lysosomes for degradation [Mizushima, Nat. Cell Biol. 20, 521-527 (2018)]. However, while most studies have focused on the mechanisms of protein degradation during this process, we report here that macroautophagy also depends on glycan degradation via the glycosidase, α-l-fucosidase 1 (FUCA1), which removes fucose from glycans. We show that cells lacking FUCA1 accumulate lysosomal glycans, which is associated with impaired autophagic flux. Moreover, in a mouse model of fucosidosis-a disease characterized by inactivating mutations in FUCA1 [Stepien et al., Genes (Basel) 11, E1383 (2020)]-glycan and autophagosome/autolysosome accumulation accompanies tissue destruction. Mechanistically, using lectin capture and mass spectrometry, we identified several lysosomal enzymes with altered fucosylation in FUCA1-null cells. Moreover, we show that the activity of some of these enzymes in the absence of FUCA1 can no longer be induced upon autophagy stimulation, causing retardation of autophagic flux, which involves impaired autophagosome-lysosome fusion. These findings therefore show that dysregulated glycan degradation leads to defective autophagy, which is likely a contributing factor in the etiology of fucosidosis.


Subject(s)
Fucosidosis , Macroautophagy , Polysaccharides , Animals , Fucosidosis/genetics , Fucosidosis/metabolism , Lysosomes/metabolism , Macroautophagy/physiology , Mice , Polysaccharides/metabolism , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/metabolism
8.
Cell ; 134(2): 353-64, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18662549

ABSTRACT

Stable isotope labeling by amino acids in cell culture (SILAC) has become a versatile tool for quantitative, mass spectrometry (MS)-based proteomics. Here, we completely label mice with a diet containing either the natural or the (13)C(6)-substituted version of lysine. Mice were labeled over four generations with the heavy diet, and development, growth, and behavior were not affected. MS analysis of incorporation levels allowed for the determination of incorporation rates of proteins from blood cells and organs. The F2 generation was completely labeled in all organs tested. SILAC analysis from various organs lacking expression of beta1 integrin, beta-Parvin, or the integrin tail-binding protein Kindlin-3 confirmed their absence and disclosed a structural defect of the red blood cell membrane skeleton in Kindlin-3-deficient erythrocytes. The SILAC-mouse approach is a versatile tool by which to quantitatively compare proteomes from knockout mice and thereby determine protein functions under complex in vivo conditions.


Subject(s)
Cytoskeletal Proteins/metabolism , Erythrocytes/metabolism , Proteomics/methods , Actinin/metabolism , Animal Feed , Animals , Blood Platelets/metabolism , Cell Membrane/chemistry , Cytoskeletal Proteins/analysis , Erythrocytes/chemistry , Female , Integrin beta1/metabolism , Isotope Labeling , Male , Mass Spectrometry , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism
9.
J Cell Sci ; 133(8)2020 04 30.
Article in English | MEDLINE | ID: mdl-32184267

ABSTRACT

Folding of proteins entering the mammalian secretory pathway requires the insertion of the correct disulfides. Disulfide formation involves both an oxidative pathway for their insertion and a reductive pathway to remove incorrectly formed disulfides. Reduction of these disulfides is crucial for correct folding and degradation of misfolded proteins. Previously, we showed that the reductive pathway is driven by NADPH generated in the cytosol. Here, by reconstituting the pathway using purified proteins and ER microsomal membranes, we demonstrate that the thioredoxin reductase system provides the minimal cytosolic components required for reducing proteins within the ER lumen. In particular, saturation of the pathway and its protease sensitivity demonstrates the requirement for a membrane protein to shuttle electrons from the cytosol to the ER. These results provide compelling evidence for the crucial role of the cytosol in regulating ER redox homeostasis, ensuring correct protein folding and facilitating the degradation of misfolded ER proteins.


Subject(s)
Membrane Proteins , Thioredoxin-Disulfide Reductase , Animals , Cytosol , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oxidation-Reduction , Protein Folding , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism
10.
EMBO J ; 36(16): 2373-2389, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28694244

ABSTRACT

Tumor progression alters the composition and physical properties of the extracellular matrix. Particularly, increased matrix stiffness has profound effects on tumor growth and metastasis. While endothelial cells are key players in cancer progression, the influence of tumor stiffness on the endothelium and the impact on metastasis is unknown. Through quantitative mass spectrometry, we find that the matricellular protein CCN1/CYR61 is highly regulated by stiffness in endothelial cells. We show that stiffness-induced CCN1 activates ß-catenin nuclear translocation and signaling and that this contributes to upregulate N-cadherin levels on the surface of the endothelium, in vitro This facilitates N-cadherin-dependent cancer cell-endothelium interaction. Using intravital imaging, we show that knockout of Ccn1 in endothelial cells inhibits melanoma cancer cell binding to the blood vessels, a critical step in cancer cell transit through the vasculature to metastasize. Targeting stiffness-induced changes in the vasculature, such as CCN1, is therefore a potential yet unappreciated mechanism to impair metastasis.


Subject(s)
Cell Communication , Endothelial Cells/physiology , Melanocytes/physiology , Cadherins/analysis , Cell Line , Cysteine-Rich Protein 61/analysis , Gene Expression Regulation , Humans , Mass Spectrometry , beta Catenin/analysis
11.
J Cell Sci ; 130(4): 697-711, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28062852

ABSTRACT

We have previously shown that Rab17, a small GTPase associated with epithelial polarity, is specifically suppressed by ERK2 (also known as MAPK1) signalling to promote an invasive phenotype. However, the mechanisms through which Rab17 loss permits invasiveness, and the endosomal cargoes that are responsible for mediating this, are unknown. Using quantitative mass spectrometry-based proteomics, we have found that knockdown of Rab17 leads to a highly selective reduction in the cellular levels of a v-SNARE (Vamp8). Moreover, proteomics and immunofluorescence indicate that Vamp8 is associated with Rab17 at late endosomes. Reduced levels of Vamp8 promote transition between ductal carcinoma in situ (DCIS) and a more invasive phenotype. We developed an unbiased proteomic approach to elucidate the complement of receptors that redistributes between endosomes and the plasma membrane, and have pin-pointed neuropilin-2 (NRP2) as a key pro-invasive cargo of Rab17- and Vamp8-regulated trafficking. Indeed, reduced Rab17 or Vamp8 levels lead to increased mobilisation of NRP2-containing late endosomes and upregulated cell surface expression of NRP2. Finally, we show that NRP2 is required for the basement membrane disruption that accompanies the transition between DCIS and a more invasive phenotype.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endosomes/metabolism , Proteomics/methods , Amino Acids/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Intracellular Membranes/metabolism , Isotope Labeling , Models, Biological , Neoplasm Grading , Neoplasm Invasiveness , Neuropilin-2/metabolism , Protein Binding , Protein Transport , R-SNARE Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , SNARE Proteins/metabolism , Survival Analysis , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
12.
BMC Cancer ; 19(1): 312, 2019 Apr 04.
Article in English | MEDLINE | ID: mdl-30947697

ABSTRACT

BACKGROUND: The tumour microenvironment is a critical regulator of malignant cancer progression. While endothelial cells have been widely studied in the context of tumour angiogenesis, their role as modulators of cancer cell invasion and migration is poorly understood. METHODS: We have investigated the influence of endothelial cells on the invasive and migratory behaviour of human cancer cells in vitro. RESULTS: Upon exposure to culture supernatants of endothelial cells, distinct cancer cells, such as SK-BR-3 cells, showed significantly increased invasion and cell migration concomitant with changes in cell morphology and gene expression reminiscent of an epithelial-mesenchymal transition (EMT). Interestingly, the pro-migratory effect on SK-BR-3 cells was significantly enhanced by supernatants obtained from subconfluent, proliferative endothelial cells rather than from confluent, quiescent endothelial cells. Systematically comparing the supernatants of subconfluent and confluent endothelial cells by quantitative MS proteomics revealed eight candidate proteins that were secreted at significantly higher levels by confluent endothelial cells representing potential inhibitors of cancer cell migration. Among these proteins, nidogen-1 was exclusively expressed in confluent endothelial cells and was found to be necessary and sufficient for the inhibition of SK-BR-3 cell migration. Indeed, SK-BR-3 cells exposed to nidogen-1-depleted endothelial supernatants showed increased promigratory STAT3 phosphorylation along with increased cell migration. This reflects the situation of enhanced SK-BR-3 migration upon stimulation with conditioned medium from subconfluent endothelial cells with inherent absence of nidogen-1 expression. CONCLUSION: The identification of nidogen-1 as an endothelial-derived inhibitor of migration of distinct cancer cell types reveals a novel mechanism of endothelial control over cancer progression.


Subject(s)
Breast Neoplasms/pathology , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Tumor Microenvironment , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism
13.
Proteomics ; 18(5-6): e1700167, 2018 03.
Article in English | MEDLINE | ID: mdl-29280568

ABSTRACT

Fibroblasts have exceptional phenotypic plasticity and capability to secrete vast amount of soluble factors, extracellular matrix components and extracellular vesicles. While in physiological conditions this makes fibroblasts master regulators of tissue homeostasis and healing of injured tissues, in solid tumors cancer associated fibroblasts (CAFs) co-evolve with the disease, and alter the biochemical and physical structure of the tumor microenvironment, as well as the behavior of the surrounding stromal and cancer cells. Thus CAFs are fundamental regulators of tumor progression and influence response to therapeutic treatments. Increasing efforts are devoted to better understand the biology of CAFs to bring insights to develop complementary strategies to target this cell type in cancer. Here we highlight components of the tumor microenvironment that play key roles in cancer progression and invasion, and provide an extensive overview of past and emerging understanding of CAF biology as well as the contribution that MS-based proteomics has made to this field.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Neoplasms/pathology , Stromal Cells/pathology , Tumor Microenvironment , Animals , Humans
14.
J Proteome Res ; 17(1): 108-118, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29043805

ABSTRACT

The pathogenic protozoan T. brucei alternates into distinct developmental stages in the mammalian and insect hosts. The mitogen-activated protein kinase (MAPK) signaling pathways transduce extracellular stimuli into a range of cellular responses, which ultimately lead to the adaptation to the external environment. Here, we combined a loss of function approach with stable isotope labeling with amino acids in cell culture (SILAC)-based mass spectrometry (MS) to investigate the role of the mitogen-activated protein kinase kinase 5 (MKK5) in T. brucei. The silencing of MKK5 significantly decreased the proliferation of procyclic forms of T. brucei. To shed light on the molecular alterations associated with this phenotype, we measured the total proteome and phosphoproteome of cells silenced for MKK5. In the total proteome, we observed a general decrease in proteins related to ribosome and translation as well as down-regulation of several components of the fatty acids biosynthesis pathway. In addition, we observed alterations in the protein levels and phosphorylation of key metabolic enzymes, which point toward a suppression of the oxidative metabolism. Taken together, our findings show that the silencing of MKK5 alters cell growth, energy metabolism, protein and fatty acids biosynthesis in procyclic T. brucei.


Subject(s)
MAP Kinase Kinase 5/physiology , Trypanosoma brucei brucei/growth & development , Cell Proliferation , Energy Metabolism , Fatty Acids/biosynthesis , Gene Silencing , MAP Kinase Kinase 5/genetics , Mass Spectrometry , Protein Biosynthesis , Trypanosoma brucei brucei/enzymology
15.
Arterioscler Thromb Vasc Biol ; 37(10): 1891-1902, 2017 10.
Article in English | MEDLINE | ID: mdl-28818855

ABSTRACT

OBJECTIVE: Thrombin is the key serine protease of the coagulation cascade and mediates cellular responses by activation of PARs (protease-activated receptors). The predominant thrombin receptor is PAR1, and in endothelial cells (ECs), thrombin dynamically regulates a plethora of phosphorylation events. However, it has remained unclear whether thrombin signaling is exclusively mediated through PAR1. Furthermore, mechanistic insight into activation and inhibition of PAR1-mediated EC signaling is lacking. In addition, signaling networks of biased PAR1 activation after differential cleavage of the PAR1 N terminus have remained an unresolved issue. APPROACH AND RESULTS: Here, we used a quantitative phosphoproteomics approach to show that classical and peptide activation of PAR1 induce highly similar signaling, that low thrombin concentrations initiate only limited phosphoregulation, and that the PAR1 inhibitors vorapaxar and parmodulin-2 demonstrate distinct antagonistic properties. Subsequent analysis of the thrombin-regulated phosphosites in the presence of PAR1 inhibitors revealed that biased activation of PAR1 is not solely linked to a specific G-protein downstream of PAR1. In addition, we showed that only the canonical thrombin PAR1 tethered ligand induces extensive early phosphoregulation in ECs. CONCLUSIONS: Our study provides detailed insight in the signaling mechanisms downstream of PAR1. Our data demonstrate that thrombin-induced EC phosphoregulation is mediated exclusively through PAR1, that thrombin and thrombin-tethered ligand peptide induce similar phosphoregulation, and that only canonical PAR1 cleavage by thrombin generates a tethered ligand that potently induces early signaling. Furthermore, platelet PAR1 inhibitors directly affect EC signaling, indicating that it will be a challenge to design a PAR1 antagonist that will target only those pathways responsible for tissue pathology.


Subject(s)
Endothelial Cells/physiology , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/physiology , Humans , Lactones/pharmacology , Phosphorylation , Proteomics , Pyridines/pharmacology , Signal Transduction
16.
Mol Cell Proteomics ; 14(3): 621-34, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25573745

ABSTRACT

Endothelial cells (ECs) play a key role to maintain the functionality of blood vessels. Altered EC permeability causes severe impairment in vessel stability and is a hallmark of pathologies such as cancer and thrombosis. Integrating label-free quantitative proteomics data into genome-wide metabolic modeling, we built up a model that predicts the metabolic fluxes in ECs when cultured on a tridimensional matrix and organize into a vascular-like network. We discovered how fatty acid oxidation increases when ECs are assembled into a fully formed network that can be disrupted by inhibiting CPT1A, the fatty acid oxidation rate-limiting enzyme. Acute CPT1A inhibition reduces cellular ATP levels and oxygen consumption, which are restored by replenishing the tricarboxylic acid cycle. Remarkably, global phosphoproteomic changes measured upon acute CPT1A inhibition pinpointed altered calcium signaling. Indeed, CPT1A inhibition increases intracellular calcium oscillations. Finally, inhibiting CPT1A induces hyperpermeability in vitro and leakage of blood vessel in vivo, which were restored blocking calcium influx or replenishing the tricarboxylic acid cycle. Fatty acid oxidation emerges as central regulator of endothelial functions and blood vessel stability and druggable pathway to control pathological vascular permeability.


Subject(s)
Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Endothelial Cells/metabolism , Fatty Acids/metabolism , Metabolome , Models, Biological , Proteomics/methods , Adenosine Triphosphate/metabolism , Animals , Endothelial Cells/cytology , Epoxy Compounds/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Mice , Oxidation-Reduction , Oxygen Consumption , Permeability
17.
J Proteome Res ; 15(7): 2187-97, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27246970

ABSTRACT

Endothelial cells (ECs) form the inner layer of blood vessels and physically separate the blood from the surrounding tissue. To support tissues with nutrients and oxygen, the endothelial monolayer is semipermeable. When EC permeability is altered, blood vessels are not functional, and this is associated with disease. A comprehensive knowledge of the mechanisms regulating EC permeability is key in developing strategies to target this mechanism in pathologies. Here we have used an in vitro model of human umbilical vein endothelial cells mimicking the formation of a physiologically permeable vessel and performed time-resolved in-depth molecular profiling using stable isotope labeling by amino acids in cell culture mass spectrometry (MS)-proteomics. Autophagy is induced when ECs are assembled into a physiologically permeable monolayer. By using siRNA and drug treatment to block autophagy in combination with functional assays and MS proteomics, we show that ECs require autophagy flux to maintain intracellular reactive oxygen species levels, and this is required to maintain the physiological permeability of the cells.


Subject(s)
Autophagy/physiology , Reactive Oxygen Species/metabolism , Cells, Cultured , Endothelium, Vascular/physiology , Humans , Mass Spectrometry , Models, Biological , Permeability , Proteomics/methods , Reactive Oxygen Species/analysis
18.
Blood ; 123(12): e22-36, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24501219

ABSTRACT

Thrombin is the key serine protease of the coagulation cascade and a potent trigger of protease-activated receptor 1 (PAR1)-mediated platelet aggregation. In recent years, PAR1 has become an appealing target for anticoagulant therapies. However, the inhibitors that have been developed so far increase bleeding risk in patients, likely because they interfere with endogenous PAR1 signaling in the endothelium. Because of its complexity, thrombin-induced signaling in endothelial cells has remained incompletely understood. Here, we have combined stable isotope amino acids in cell culture, affinity-based phosphopeptide enrichment, and high-resolution mass spectrometry and performed a time-resolved analysis of the thrombin-induced signaling in human primary endothelial cells. We identified 2224 thrombin-regulated phosphorylation sites, the majority of which have not been previously related to thrombin. Those sites were localized on proteins that are novel to thrombin signaling, but also on well-known players such as PAR1, Rho-associated kinase 2, phospholipase C, and proteins related to actin cytoskeleton, cell-cell junctions, and Weibel-Palade body release. Our study provides a unique resource of phosphoproteins and phosphorylation sites that may generate novel insights into an intimate understanding of thrombin-mediated PAR signaling and the development of improved PAR1 antagonists that affect platelet but not endothelial cell function.


Subject(s)
Endothelial Cells/metabolism , Thrombin/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Humans , Mass Spectrometry , Models, Biological , Phosphoproteins/metabolism , Proteomics , Receptor, PAR-1/metabolism , Signal Transduction , Tight Junctions/metabolism , Weibel-Palade Bodies/metabolism , rhoA GTP-Binding Protein/metabolism
19.
Mol Cell Proteomics ; 12(12): 3599-611, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23979707

ABSTRACT

Proteomics has been successfully used for cell culture on dishes, but more complex cellular systems have proven to be challenging and so far poorly approached with proteomics. Because of the complexity of the angiogenic program, we still do not have a complete understanding of the molecular mechanisms involved in this process, and there have been no in depth quantitative proteomic studies. Plating endothelial cells on matrigel recapitulates aspects of vessel growth, and here we investigate this mechanism by using a spike-in SILAC quantitative proteomic approach. By comparing proteomic changes in primary human endothelial cells morphogenesis on matrigel to general adhesion mechanisms in cells spreading on culture dish, we pinpoint pathways and proteins modulated by endothelial cells. The cell-extracellular matrix adhesion proteome depends on the adhesion substrate, and a detailed proteomic profile of the extracellular matrix secreted by endothelial cells identified CLEC14A as a matrix component, which binds to MMRN2. We verify deregulated levels of these proteins during tumor angiogenesis in models of multistage carcinogenesis. This is the most in depth quantitative proteomic study of endothelial cell morphogenesis, which shows the potential of applying high accuracy quantitative proteomics to in vitro models of vessel growth to shed new light on mechanisms that accompany pathological angiogenesis. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000359.


Subject(s)
Antigens, Surface/genetics , Biomarkers, Tumor/genetics , Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Animals , Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carbon Isotopes , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Collagen/chemistry , Drug Combinations , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Isotope Labeling , Laminin/chemistry , Lectins, C-Type/metabolism , Mass Spectrometry , Membrane Glycoproteins/metabolism , Mice , Morphogenesis/genetics , Neovascularization, Pathologic , Primary Cell Culture , Protein Binding , Proteoglycans/chemistry , Proteomics , Signal Transduction
20.
J Cell Sci ; 125(Pt 2): 435-48, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22357970

ABSTRACT

Integrin and receptor tyrosine kinase signalling networks cooperate to regulate various biological functions. The molecular details underlying the integration of both signalling networks remain largely uncharacterized. Here we identify a signalling module composed of a fibronectin-α5ß1-integrin-integrin-linked-kinase (ILK) complex that, in concert with epidermal growth factor (EGF) cues, cooperatively controls the formation of transient actin-based circular dorsal ruffles (DRs) in fibroblasts. DR formation depends on the precise spatial activation of Src at focal adhesions by integrin and EGF receptor signals, in an ILK-dependent manner. In a SILAC-based phosphoproteomics screen we identified the tumour-suppressor Cyld as being required for DR formation induced by α5ß1 integrin and EGF receptor co-signalling. Furthermore, EGF-induced Cyld tyrosine phosphorylation is controlled by integrin-ILK and Src as a prerequisite for DR formation. This study provides evidence for a novel function of integrin-ILK and EGF signalling crosstalk in mediating Cyld tyrosine phosphorylation and fast actin-based cytoskeletal rearrangements.


Subject(s)
ErbB Receptors/metabolism , Integrin alpha5beta1/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Cell Membrane Structures/enzymology , Cell Membrane Structures/physiology , Cells, Cultured , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL