Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Sci Rep ; 10(1): 14898, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913274

ABSTRACT

While blocking the renin angiotensin aldosterone system (RAAS) has been the main therapeutic strategy to control diabetic kidney disease (DKD) for many years, 25-30% of diabetic patients still develop the disease. In the present work we adopted a systems biology strategy to analyze glomerular protein signatures to identify drugs with potential therapeutic properties in DKD acting through a RAAS-independent mechanism. Glomeruli were isolated from wild type and type 1 diabetic (Ins2Akita) mice treated or not with the angiotensin-converting enzyme inhibitor (ACEi) ramipril. Ramipril efficiently reduced the urinary albumin/creatine ratio (ACR) of Ins2Akita mice without modifying DKD-associated renal-injuries. Large scale quantitative proteomics was used to identify the DKD-associated glomerular proteins (DKD-GPs) that were ramipril-insensitive (RI-DKD-GPs). The raw data are publicly available via ProteomeXchange with identifier PXD018728. We then applied an in silico drug repurposing approach using a pattern-matching algorithm (Connectivity Mapping) to compare the RI-DKD-GPs's signature with a collection of thousands of transcriptional signatures of bioactive compounds. The sesquiterpene lactone parthenolide was identified as one of the top compounds predicted to reverse the RI-DKD-GPs's signature. Oral treatment of 2 months old Ins2Akita mice with dimethylaminoparthenolide (DMAPT, a water-soluble analogue of parthenolide) for two months at 10 mg/kg/d by gavage significantly reduced urinary ACR. However, in contrast to ramipril, DMAPT also significantly reduced glomerulosclerosis and tubulointerstitial fibrosis. Using a system biology approach, we identified DMAPT, as a compound with a potential add-on value to standard-of-care ACEi-treatment in DKD.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetic Nephropathies/drug therapy , Sesquiterpenes/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Connectome/methods , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Gene Expression Regulation/drug effects , Glomerular Filtration Rate , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred C57BL , Renin-Angiotensin System/drug effects
2.
Sci Signal ; 11(538)2018 07 10.
Article in English | MEDLINE | ID: mdl-29991650

ABSTRACT

The activation of T cells requires the guanine nucleotide exchange factor VAV1. Using mice in which a tag for affinity purification was attached to endogenous VAV1 molecules, we analyzed by quantitative mass spectrometry the signaling complex that assembles around activated VAV1. Fifty VAV1-binding partners were identified, most of which had not been previously reported to participate in VAV1 signaling. Among these was CD226, a costimulatory molecule of immune cells. Engagement of CD226 induced the tyrosine phosphorylation of VAV1 and synergized with T cell receptor (TCR) signals to specifically enhance the production of interleukin-17 (IL-17) by primary human CD4+ T cells. Moreover, co-engagement of the TCR and a risk variant of CD226 that is associated with autoimmunity (rs763361) further enhanced VAV1 activation and IL-17 production. Thus, our study reveals that a VAV1-based, synergistic cross-talk exists between the TCR and CD226 during both physiological and pathological T cell responses and provides a rational basis for targeting CD226 for the management of autoimmune diseases.

4.
Proteomics ; 17(7)2017 04.
Article in English | MEDLINE | ID: mdl-27709814

ABSTRACT

The ubiquitin-proteasome pathway (UPP) plays a critical role in the degradation of proteins implicated in cell cycle control, signal transduction, DNA damage response, apoptosis and immune response. Proteasome inhibitors can inhibit the growth of a broad spectrum of human cancer cells by altering the balance of intracellular proteins. However, the targets of these compounds in acute myeloid leukemia (AML) cells have not been fully characterized. Herein, we combined large-scale quantitative analysis by SILAC-MS and targeted quantitative proteomic analysis in order to identify proteins regulated upon proteasome inhibition in two AML cell lines displaying different stages of maturation: immature KG1a cells and mature U937 cells. In-depth data analysis enabled accurate quantification of more than 7000 proteins in these two cell lines. Several candidates were validated by selected reaction monitoring (SRM) measurements in a large number of samples. Despite the broad range of proteins known to be affected by proteasome inhibition, such as heat shock (HSP) and cell cycle proteins, our analysis identified new differentially regulated proteins, including IL-32, MORF family mortality factors and apoptosis inducing factor SIVA, a target of p53. It could explain why proteasome inhibitors induce stronger apoptotic responses in immature AML cells.


Subject(s)
Computational Biology , Gene Expression Regulation, Leukemic/drug effects , Leukocytes/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bortezomib/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Gene Ontology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , Leukocytes/metabolism , Leukocytes/pathology , Leupeptins/pharmacology , Molecular Sequence Annotation , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Sci Rep ; 6: 34255, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27694941

ABSTRACT

IL-33 is a nuclear cytokine from the IL-1 family that plays important roles in health and disease. Extracellular IL-33 activates a growing number of target cells, including group 2 innate lymphoid cells, mast cells and regulatory T cells, but it remains unclear whether intracellular nuclear IL-33 has additional functions in the nucleus. Here, we used a global proteomic approach based on high-resolution mass spectrometry to compare the extracellular and intracellular roles of IL-33 in primary human endothelial cells, a major source of IL-33 protein in human tissues. We found that exogenous extracellular IL-33 cytokine induced expression of a distinct set of proteins associated with inflammatory responses in endothelial cells. In contrast, knockdown of endogenous nuclear IL-33 expression using two independent RNA silencing strategies had no reproducible effect on the endothelial cell proteome. These results suggest that IL-33 acts as a cytokine but not as a nuclear factor regulating gene expression in endothelial cells.


Subject(s)
Cell Nucleus/metabolism , Endothelium, Vascular/metabolism , Extracellular Space/metabolism , Interleukin-33/physiology , Gene Knockdown Techniques , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , RNA/genetics , Tandem Mass Spectrometry
6.
Nat Commun ; 7: 10230, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26756352

ABSTRACT

Obesity favours the occurrence of locally disseminated prostate cancer in the periprostatic adipose tissue (PPAT) surrounding the prostate gland. Here we show that adipocytes from PPAT support the directed migration of prostate cancer cells and that this event is strongly promoted by obesity. This process is dependent on the secretion of the chemokine CCL7 by adipocytes, which diffuses from PPAT to the peripheral zone of the prostate, stimulating the migration of CCR3 expressing tumour cells. In obesity, higher secretion of CCL7 by adipocytes facilitates extraprostatic extension. The observed increase in migration associated with obesity is totally abrogated when the CCR3/CCL7 axis is inhibited. In human prostate cancer tumours, expression of the CCR3 receptor is associated with the occurrence of aggressive disease with extended local dissemination and a higher risk of biochemical recurrence, highlighting the potential benefit of CCR3 antagonists in the treatment of prostate cancer.


Subject(s)
Adenocarcinoma/metabolism , Adipocytes/metabolism , Cell Movement , Chemokine CCL7/metabolism , Neoplasm Recurrence, Local/metabolism , Obesity/metabolism , Prostatic Neoplasms/metabolism , Receptors, CCR3/metabolism , Adenocarcinoma/pathology , Adipose Tissue/cytology , Adult , Aged , Animals , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Male , Mass Spectrometry , Mice , Middle Aged , Prognosis , Prostate , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
7.
J Immunol ; 195(11): 5472-81, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26512138

ABSTRACT

The protein tyrosine kinase LCK plays a key role in TCR signaling, and its activity is dynamically controlled by the tyrosine kinase C-terminal Src kinase (CSK) and the tyrosine phosphatase CD45. CSK is brought in contiguity to LCK via binding to a transmembrane adaptor known as phosphoprotein associated with glycosphingolipid-enriched microdomains (PAG). The lack of a blatant phenotype in PAG-deficient mice has impeded our understanding of the mechanisms through which PAG exerts its negative-regulatory role in TCR signaling. We used quantitative mass spectrometry and both thymocytes and CD4(+) T cells from mice in which a tag for affinity purification was knocked in the gene coding for PAG to determine the composition and dynamics of the multiprotein complexes that are found around PAG over 5 min of activation. Most of the high-confidence interactions that we observed were previously unknown. Using phosphoproteomic analysis, PAG showed low levels of tyrosine phosphorylation in resting primary mouse CD4(+) T cells; the levels of tyrosine phosphorylation increased and reached a maximum 2 min after stimulation. Analysis of the dynamics of association of the protein tyrosine phosphatase PTPN22 and lipid phosphatase SHIP-1 with PAG following T cell activation suggests that both cooperate with CSK to terminate T cell activation. Our findings provide a model of the role for PAG in mouse primary CD4(+) T cells that is consistent with recent phosphoproteomic studies of the Jurkat T cell line but difficult to reconcile with former biochemical studies indicating that PAG is constitutively phosphorylated in resting T cells and rapidly dephosphorylated once the TCR is engaged.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Thymocytes/immunology , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cells, Cultured , Embryonic Stem Cells , Enzyme Activation , Gene Knock-In Techniques , Inositol Polyphosphate 5-Phosphatases , Intercellular Signaling Peptides and Proteins , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoproteins/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Proteomics , Receptors, Antigen, T-Cell/immunology , Tandem Mass Spectrometry
8.
Int J Nanomedicine ; 10: 1869-83, 2015.
Article in English | MEDLINE | ID: mdl-25834426

ABSTRACT

Nanoparticle (NP)-protein interactions in complex samples have not yet been clearly understood. Nevertheless, several studies demonstrated that NP's physicochemical features significantly impact on the protein corona composition. Taking advantage of the NP potential to harvest different subsets of proteins, we assessed for the first time the capacity of three kinds of superparamagnetic NPs to highlight the erythrocyte minor proteome. Using both qualitative and quantitative proteomics approaches, nano-liquid chromatography-tandem mass spectrometry allowed the identification of 893 different proteins, confirming the reproducible capacity of NPs to increase the number of identified proteins, through a reduction of the sample concentration range and the capture of specific proteins on the three different surfaces. These NP-specific protein signatures revealed significant differences in their isoelectric point and molecular weight. Moreover, this NP strategy offered a deeper access to the erythrocyte proteome highlighting several signaling pathways implicated in important erythrocyte functions. The automated potentiality, the reproducibility, and the low-consuming sample demonstrate the strong compatibility of our strategy for large-scale clinical studies and may become a standardized sample preparation in future erythrocyte-associated proteomics studies.


Subject(s)
Blood Proteins/analysis , Erythrocytes/metabolism , Magnetite Nanoparticles/chemistry , Adult , Blood Proteins/metabolism , Chromatography, Liquid/methods , Electrophoresis, Polyacrylamide Gel/methods , Erythrocytes/chemistry , Female , Humans , Male , Middle Aged , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Reproducibility of Results , Surface Properties , Tandem Mass Spectrometry/methods
9.
Mol Syst Biol ; 11(1): 771, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25561571

ABSTRACT

In eukaryotic cells, intracellular protein breakdown is mainly performed by the ubiquitin-proteasome system. Proteasomes are supramolecular protein complexes formed by the association of multiple sub-complexes and interacting proteins. Therefore, they exhibit a very high heterogeneity whose function is still not well understood. Here, using a newly developed method based on the combination of affinity purification and protein correlation profiling associated with high-resolution mass spectrometry, we comprehensively characterized proteasome heterogeneity and identified previously unknown preferential associations within proteasome sub-complexes. In particular, we showed for the first time that the two main proteasome subtypes, standard proteasome and immunoproteasome, interact with a different subset of important regulators. This trend was observed in very diverse human cell types and was confirmed by changing the relative proportions of both 20S proteasome forms using interferon-γ. The new method developed here constitutes an innovative and powerful strategy that could be broadly applied for unraveling the dynamic and heterogeneous nature of other biologically relevant supramolecular protein complexes.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Interaction Domains and Motifs , Cell Line, Tumor , Chromatography, Affinity , Chromatography, Liquid , Gene Expression Profiling , HEK293 Cells , Humans , Mass Spectrometry , Proteomics/methods , Tandem Mass Spectrometry , U937 Cells
10.
Mol Cell Proteomics ; 14(3): 771-81, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25505153

ABSTRACT

The analysis and management of MS data, especially those generated by data independent MS acquisition, exemplified by SWATH-MS, pose significant challenges for proteomics bioinformatics. The large size and vast amount of information inherent to these data sets need to be properly structured to enable an efficient and straightforward extraction of the signals used to identify specific target peptides. Standard XML based formats are not well suited to large MS data files, for example, those generated by SWATH-MS, and compromise high-throughput data processing and storing. We developed mzDB, an efficient file format for large MS data sets. It relies on the SQLite software library and consists of a standardized and portable server-less single-file database. An optimized 3D indexing approach is adopted, where the LC-MS coordinates (retention time and m/z), along with the precursor m/z for SWATH-MS data, are used to query the database for data extraction. In comparison with XML formats, mzDB saves ∼25% of storage space and improves access times by a factor of twofold up to even 2000-fold, depending on the particular data access. Similarly, mzDB shows also slightly to significantly lower access times in comparison with other formats like mz5. Both C++ and Java implementations, converting raw or XML formats to mzDB and providing access methods, will be released under permissive license. mzDB can be easily accessed by the SQLite C library and its drivers for all major languages, and browsed with existing dedicated GUIs. The mzDB described here can boost existing mass spectrometry data analysis pipelines, offering unprecedented performance in terms of efficiency, portability, compactness, and flexibility.


Subject(s)
Database Management Systems , Mass Spectrometry/methods , Datasets as Topic , Epithelial Cells/metabolism , Humans , Proteome/analysis
11.
Mol Cell Proteomics ; 13(12): 3421-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205225

ABSTRACT

Obstructive nephropathy is a frequently encountered situation in newborns. In previous studies, the urinary peptidome has been analyzed for the identification of clinically useful biomarkers of obstructive nephropathy. However, the urinary proteome has not been explored yet and should allow additional insight into the pathophysiology of the disease. We have analyzed the urinary proteome of newborns (n = 5/group) with obstructive nephropathy using label free quantitative nanoLC-MS/MS allowing the identification and quantification of 970 urinary proteins. We next focused on proteins exclusively regulated in severe obstructive nephropathy and identified Arginase 1 as a potential candidate molecule involved in the development of obstructive nephropathy, located at the crossroad of pro- and antifibrotic pathways. The reduced urinary abundance of Arginase 1 in obstructive nephropathy was verified in independent clinical samples using both Western blot and MRM analysis. These data were confirmed in situ in kidneys obtained from a mouse obstructive nephropathy model. In addition, we also observed increased expression of Arginase 2 and increased total arginase activity in obstructed mouse kidneys. mRNA expression analysis of the related arginase pathways indicated that the pro-fibrotic arginase-related pathway is activated during obstructive nephropathy. Taken together we have identified a new actor in the development of obstructive nephropathy in newborns using quantitative urinary proteomics and shown its involvement in an in vivo model of disease. The present study demonstrates the relevance of such a quantitative urinary proteomics approach with clinical samples for a better understanding of the pathophysiology and for the discovery of potential therapeutic targets.


Subject(s)
Arginase/urine , Hydronephrosis/urine , Kidney/metabolism , Proteome/metabolism , Renal Insufficiency/urine , Animals , Arginase/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Hydronephrosis/congenital , Hydronephrosis/pathology , Infant , Infant, Newborn , Kidney/pathology , Male , Mice, Inbred C57BL , Proteome/genetics , Proteomics/methods , Renal Insufficiency/congenital , Renal Insufficiency/pathology , Signal Transduction
12.
PLoS One ; 9(8): e104563, 2014.
Article in English | MEDLINE | ID: mdl-25116916

ABSTRACT

BACKGROUND: Lactoferrins exhibit antitumoral activities either as a secretory lactoferrin or an intracellular delta-lactoferrin isoform. These activities involve processes such as regulation of the cell cycle and apoptosis. While lactoferrin has been shown to exert its function by activating different transduction pathways, delta-lactoferrin has been proven to act as a transcription factor. Like many tumor suppressors, these two proteins are under-expressed in several types of cancer, particularly in breast cancer. METHODOLOGY/PRINCIPAL FINDINGS: In order to compare the differential effects of the re-introduction of lactoferrin isoforms in breast cancer cells we chose the cancerous mammary gland MDA-MB-231 cell line as a model. We produced a cell line stably expressing delta-lactoferrin. We also treated these cells with fresh purified human breast lactoferrin. We performed two quantitative proteomic studies in parallel using SILAC coupled to mass spectrometry in order to compare the effects of different doses of the two lactoferrin isoforms. The proteome of untreated, delta-lactoferrin expressing and human lactoferrin treated MDA-MB-231 cells were compared. Overall, around 5300 proteins were identified and quantified using the in-house developed MFPaQ software. Among these, expression was increased by 1.5-fold or more for around 300 proteins in delta-lactoferrin expressing cells and 190 proteins in lactoferrin treated cells. At the same time, about 200 and 40 proteins were found to be downregulated (0-0.7-fold) in response to delta-lactoferrin and lactoferrin, respectively. CONCLUSIONS/SIGNIFICANCE: Re-introduction of delta-lactoferrin and lactoferrin expression in MDA-MB-231 mainly leads to modifications of protein profiles involved in processes such as proliferation, apoptosis, oxidative stress, the ubiquitin pathway, translation and mRNA quality control. Moreover, this study identified new target genes of delta-lactoferrin transcriptional activity such as SelH, GTF2F2 and UBE2E1.


Subject(s)
Lactoferrin/metabolism , Proteomics , Alternative Splicing , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Isotope Labeling , Lactoferrin/genetics , Lactoferrin/pharmacology , Neoplasm Metastasis , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Proteome , Proteomics/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reproducibility of Results , Response Elements , Selenoproteins/genetics , Transcription Factors/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
13.
Proteomics Clin Appl ; 8(7-8): 620-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24888668

ABSTRACT

PURPOSE: To find potential biomarkers of abdominal aortic aneurysms (AAA), we performed a differential proteomic study based on human plasma-derived microvesicles. EXPERIMENTAL DESIGN: Exosomes and microparticles isolated from plasma of AAA patients and control subjects (n = 10 each group) were analyzed by a label-free quantitative MS-based strategy. Homemade and publicly available software packages have been used for MS data analysis. RESULTS: The application of two kinds of bioinformatic tools allowed us to find differential protein profiles from AAA patients. Some of these proteins found by the two analysis methods belong to main pathological mechanisms of AAA such as oxidative stress, immune-inflammation, and thrombosis. CONCLUSIONS AND CLINICAL RELEVANCE: Data analysis from label-free MS-based experiments requires the use of sophisticated bioinformatic approaches to perform quantitative studies from complex protein mixtures. The application of two of these bioinformatic tools provided us a preliminary list of differential proteins found in plasma-derived microvesicles not previously associated to AAA, which could help us to understand the pathological mechanisms related to this disease.


Subject(s)
Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/metabolism , Exosomes/metabolism , Proteomics/methods , Aortic Aneurysm, Abdominal/pathology , Humans
14.
J Proteome Res ; 13(6): 3027-37, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24804812

ABSTRACT

The proteasome is the main proteolytic system involved in intracellular proteins homeostasis in eukaryotes. Although the structure of proteasome complexes has been well characterized, the distribution of its activators and associated proteins are less studied. Here, we determine the composition and the stoichiometry of proteasome complexes and their associated proteins in a wide range of human cell lines using a one-step affinity purification method and a label-free quantitative proteomic approach. We show that proteasome complexes are highly dynamic protein assemblies, the activity of which being regulated at different levels by variations in the stoichiometry of bound regulators, in the composition of catalytic subunits and associated proteins, and in the rate of the 20S catalytic core complex assembly.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Subunits/metabolism , Cell Line , Homeostasis , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Weight , Proteasome Endopeptidase Complex/chemistry , Protein Interaction Maps , Protein Structure, Quaternary , Protein Subunits/chemistry , Proteomics
15.
Mol Cell Biol ; 34(6): 1046-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24396066

ABSTRACT

The simultaneous interaction of poly(A)-binding protein (PABP) with eukaryotic translation initiation factor 4G (eIF4G) and the mRNA 3' poly(A) tail promotes translation initiation. We previously showed that the interaction of PABP-interacting protein 1 (Paip1) with PABP and eukaryotic translation initiation factor 3 (eIF3; via the eIF3g subunit) further stimulates translation. Here, we demonstrate that the interaction of eIF3 with Paip1 is regulated by amino acids through the mTORC1 signaling pathway. The Paip1-eIF3 interaction is impaired by the mTORC1 inhibitors, rapamycin and PP242. We show that ribosomal protein S6 kinases 1 and 2 (S6K1/2) promote the interaction of eIF3 with Paip1. The enhancement of Paip1-eIF3 interaction by amino acids is abrogated by an S6K inhibitor or shRNA against S6K1/2. S6K1 interacts with eIF3f and, in vitro, phosphorylates eIF3. Finally, we show that S6K inhibition leads to a reduction in translation by Paip1. We propose that S6K1/2 phosphorylate eIF3 to stimulate Paip1-eIF3 interaction and consequent translation initiation. Taken together, these data demonstrate that eIF3 is a new translation target of the mTOR/S6K pathway.


Subject(s)
Eukaryotic Initiation Factor-3/genetics , Eukaryotic Initiation Factor-3/metabolism , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Amino Acids/genetics , Amino Acids/metabolism , Cell Line , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , Phosphorylation/genetics , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors
16.
Proc Natl Acad Sci U S A ; 110(44): 17927-32, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127601

ABSTRACT

DNA lesions that block transcription may cause cell death even when repaired, if transcription does not restart to reestablish cellular metabolism. However, transcription resumption after individual DNA-lesion repair remains poorly described in mechanistic terms and its players are largely unknown. The general transcription factor II H (TFIIH) is a major actor of both nucleotide excision repair subpathways of which transcription-coupled repair highlights the interplay between DNA repair and transcription. Using an unbiased proteomic approach, we have identified the protein eleven-nineteen lysine-rich leukemia (ELL) as a TFIIH partner. Here we show that ELL is recruited to UV-damaged chromatin in a Cdk7- dependent manner (a component of the cyclin-dependent activating kinase subcomplex of TFIIH). We demonstrate that depletion of ELL strongly hinders RNA polymerase II (RNA Pol II) transcription resumption after lesion removal and DNA gap filling. Lack of ELL was also observed to increase RNA Pol II retention to the chromatin during this process. Identifying ELL as an essential player for RNA Pol II restart during cellular DNA damage response opens the way to obtaining a mechanistic description of transcription resumption after DNA repair.


Subject(s)
DNA Repair/physiology , RNA Polymerase II/metabolism , Transcription Factor TFIIH/metabolism , Transcriptional Activation/physiology , Transcriptional Elongation Factors/metabolism , Base Sequence , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Cloning, Molecular , DNA Primers/genetics , Fluorescence Recovery After Photobleaching , Humans , Mass Spectrometry , Molecular Sequence Data , RNA Interference , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
17.
Cell Signal ; 25(12): 2823-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24044920

ABSTRACT

ASB proteins are the specificity subunits of cullin5-RING E3 ubiquitin ligases (CRL5) that play roles in ubiquitin-mediated protein degradation. However, how their activity is regulated remains poorly understood. Here, we unravel a novel mechanism of regulation of a CRL5 through phosphorylation of its specificity subunit ASB2α. Indeed, using mass spectrometry, we showed for the first time that ASB2α is phosphorylated and that phosphorylation of serine-323 (Ser-323) of ASB2α is crucial for the targeting of the actin-binding protein filamin A (FLNa) to degradation. Mutation of ASB2α Ser-323 to Ala had no effect on intrinsic E3 ubiquitin ligase activity of ASB2α but abolished the ability of ASB2α to induce degradation of FLNa. In contrast, the ASB2α Ser-323 to Asp phosphomimetic mutant induced acute degradation of FLNa. Moreover, inhibition of the extracellular signal-regulated kinases 1 and 2 (Erk1/2) activity reduced ASB2α-mediated FLNa degradation. We further showed that the subcellular localization of ASB2α to actin-rich structures is dependent on ASB2α Ser-323 phosphorylation and propose that the interaction with FLNa depends on the electrostatic potential redistribution induced by the Ser-323 phosphate group. Taken together, these data unravel an important mechanism by which ASB2α-mediated FLNa degradation can be regulated.


Subject(s)
Filamins/metabolism , Proteolysis , Serine/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Amino Acid Sequence , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Serine/analysis , Suppressor of Cytokine Signaling Proteins/chemistry , Ubiquitin-Protein Ligases/metabolism
18.
Sci Transl Med ; 5(198): 198ra106, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946195

ABSTRACT

Bilateral congenital abnormalities of the kidney and urinary tract (CAKUT), although are individually rare diseases, remain the main cause of chronic kidney disease in infants worldwide. Bilateral CAKUT display a wide spectrum of pre- and postnatal outcomes ranging from death in utero to normal postnatal renal function. Methods to predict these outcomes in utero are controversial and, in several cases, lead to unjustified termination of pregnancy. Using capillary electrophoresis coupled with mass spectrometry, we have analyzed the urinary proteome of fetuses with posterior urethral valves (PUV), the prototypic bilateral CAKUT, for the presence of biomarkers predicting postnatal renal function. Among more than 4000 fetal urinary peptide candidates, 26 peptides were identified that were specifically associated with PUV in 13 patients with early end-stage renal disease (ESRD) compared to 15 patients with absence of ESRD before the age of 2. A classifier based on these peptides correctly predicted postnatal renal function with 88% sensitivity and 95% specificity in an independent blinded validation cohort of 38 PUV patients, outperforming classical methods, including fetal urine biochemistry and fetal ultrasound. This study demonstrates that fetal urine is an important pool of peptides that can predict postnatal renal function and thus be used to make clinical decisions regarding pregnancy.


Subject(s)
Kidney Diseases/diagnosis , Peptides/urine , Electrophoresis, Capillary , Female , Fetus , Humans , Infant , Kidney Diseases/urine , Male , Mass Spectrometry , Pregnancy , Ultrasonography, Prenatal
19.
Mol Cell Proteomics ; 12(8): 2293-312, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23674615

ABSTRACT

In order to map the extracellular or membrane proteome associated with the vasculature and the stroma in an embryonic organism in vivo, we developed a biotinylation technique for chicken embryo and combined it with mass spectrometry and bioinformatic analysis. We also applied this procedure to implanted tumors growing on the chorioallantoic membrane or after the induction of granulation tissue. Membrane and extracellular matrix proteins were the most abundant components identified. Relative quantitative analysis revealed differential protein expression patterns in several tissues. Through a bioinformatic approach, we determined endothelial cell protein expression signatures, which allowed us to identify several proteins not yet reported to be associated with endothelial cells or the vasculature. This is the first study reported so far that applies in vivo biotinylation, in combination with robust label-free quantitative proteomics approaches and bioinformatic analysis, to an embryonic organism. It also provides the first description of the vascular and matrix proteome of the embryo that might constitute the starting point for further developments.


Subject(s)
Avian Proteins/metabolism , Chick Embryo/metabolism , Chorioallantoic Membrane/metabolism , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Animals , Biotinylation , Cell Line, Tumor , Chorioallantoic Membrane/injuries , Humans , Intestine, Small/embryology , Intestine, Small/metabolism , Kidney/embryology , Kidney/metabolism , Liver/embryology , Liver/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Proteome
20.
Mol Cell Proteomics ; 12(3): 736-48, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275444

ABSTRACT

Hypoxia-inducible factors (HIFs) are heterodimeric transcription factors that play a key role in cellular adaptation to hypoxia. HIF proteins are composed of an α subunit regulated by oxygen pressure (essentially HIF1α or HIF2α) and a constitutively expressed ß subunit. These proteins are often overexpressed in cancer cells, and HIF overexpression frequently correlates with poor prognosis, making HIF proteins promising therapeutic targets. HIF proteins are involved in melanoma initiation and progression; however, the specific function of HIF2 in melanoma has not yet been studied comprehensively. Identifying protein complexes is a valuable way to uncover protein function, and affinity purification coupled with mass spectrometry and label-free quantification is a reliable method for this approach. We therefore applied quantitative interaction proteomics to identify exhaustively the nuclear complexes containing HIF2α in a human melanoma cell line, 501mel. We report, for the first time, a high-throughput analysis of the interactome of an HIF subunit. Seventy proteins were identified that interact with HIF2α, including some well-known HIF partners and some new interactors. The new HIF2α partners microphthalmia-associated transcription factor, SOX10, and AP2α, which are master actors of melanoma development, were confirmed via co-immunoprecipitation experiments. Their ability to bind to HIF1α was also tested: microphthalmia-associated transcription factor and SOX10 were confirmed as HIF1α partners, but the transcription factor AP2α was not. AP2α expression correlates with low invasive capacities. Interestingly, we demonstrated that when HIF2α was overexpressed, only cells expressing large amounts of AP2α exhibited decreased invasive capacities in hypoxia relative to normoxia. The simultaneous presence of both transcription factors therefore reduces cells' invasive properties. Knowledge of the HIF2α interactome is thus a useful resource for investigating the general mechanisms of HIF function and regulation, and here we reveal unexpected, distinct roles for the HIF1 and HIF2 isoforms in melanoma progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Proteome/metabolism , Proteomics/methods , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoprecipitation , Mass Spectrometry/methods , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping/methods , Proteome/genetics , RNA Interference , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Sequence Homology, Amino Acid , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL