Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 7: 10238, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728094

ABSTRACT

In view of recent reports documenting pervasive translation outside of canonical protein-coding sequences, we wished to determine the proportion of major histocompatibility complex (MHC) class I-associated peptides (MAPs) derived from non-canonical reading frames. Here we perform proteogenomic analyses of MAPs eluted from human B cells using high-throughput mass spectrometry to probe the six-frame translation of the B-cell transcriptome. We report that ∼ 10% of MAPs originate from allegedly noncoding genomic sequences or exonic out-of-frame translation. The biogenesis and properties of these 'cryptic MAPs' differ from those of conventional MAPs. Cryptic MAPs come from very short proteins with atypical C termini, and are coded by transcripts bearing long 3'UTRs enriched in destabilizing elements. Relative to conventional MAPs, cryptic MAPs display different MHC class I-binding preferences and harbour more genomic polymorphisms, some of which are immunogenic. Cryptic MAPs increase the complexity of the MAP repertoire and enhance the scope of CD8 T-cell immunosurveillance.


Subject(s)
Genes, MHC Class I/genetics , Proteomics/methods , Gene Expression Regulation , Genotype , Humans , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reading Frames
2.
Nat Commun ; 6: 8648, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26468920

ABSTRACT

Protein post-translational modifications (PTMs) play important roles in the control of various biological processes including protein-protein interactions, epigenetics and cell cycle regulation. Mass spectrometry-based proteomics approaches enable comprehensive identification and quantitation of numerous types of PTMs. However, the analysis of PTMs is complicated by the presence of indistinguishable co-eluting isomeric peptides that result in composite spectra with overlapping features that prevent the identification of individual components. In this study, we present Iso-PeptidAce, a novel software tool that enables deconvolution of composite MS/MS spectra of isomeric peptides based on features associated with their characteristic fragment ion patterns. We benchmark Iso-PeptidAce using dilution series prepared from mixtures of known amounts of synthetic acetylated isomers. We also demonstrate its applicability to different biological problems such as the identification of site-specific acetylation patterns in histones bound to chromatin assembly factor-1 and profiling of histone acetylation in cells treated with different classes of HDAC inhibitors.


Subject(s)
Mass Spectrometry , Protein Processing, Post-Translational , Software , Acetylation , Histone Acetyltransferases/metabolism , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , K562 Cells , Ribonucleases , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins
3.
Nat Commun ; 5: 5409, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25391492

ABSTRACT

Small ubiquitin-related modifiers (SUMO) are evolutionarily conserved ubiquitin-like proteins that regulate several cellular processes including cell cycle progression, intracellular trafficking, protein degradation and apoptosis. Despite the importance of protein SUMOylation in different biological pathways, the global identification of acceptor sites in complex cell extracts remains a challenge. Here we generate a monoclonal antibody that enriches for peptides containing SUMO remnant chains following tryptic digestion. We identify 954 SUMO3-modified lysine residues on 538 proteins and profile by quantitative proteomics the dynamic changes of protein SUMOylation following proteasome inhibition. More than 86% of these SUMOylation sites have not been reported previously, including 5 sites on the tumour suppressor parafibromin (CDC73). The modification of CDC73 at K136 affects its nuclear retention within PML nuclear bodies on proteasome inhibition. In contrast, a CDC73 K136R mutant translocates to the cytoplasm under the same conditions, further demonstrating the effectiveness of our method to characterize the dynamics of lysine SUMOylation.


Subject(s)
Lysine/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Animals , Antibodies, Monoclonal/immunology , Chromatography, Affinity , Gas Chromatography-Mass Spectrometry , HEK293 Cells , Humans , Hybridomas/metabolism , Peptides/immunology , Peptides/metabolism , Proteome/metabolism , Rabbits , Small Ubiquitin-Related Modifier Proteins/immunology , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/metabolism
4.
Nat Commun ; 5: 3600, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24714562

ABSTRACT

For decades, the global impact of genomic polymorphisms on the repertoire of peptides presented by major histocompatibility complex (MHC) has remained a matter of speculation. Here we present a novel approach that enables high-throughput discovery of polymorphic MHC class I-associated peptides (MIPs), which play a major role in allorecognition. On the basis of comprehensive analyses of the genomic landscape of MIPs eluted from B lymphoblasts of two MHC-identical siblings, we show that 0.5% of non-synonymous single nucleotide variations are represented in the MIP repertoire. The 34 polymorphic MIPs found in our subjects are encoded by bi-allelic loci with dominant and recessive alleles. Our analyses show that, at the population level, 12% of the MIP-coding exome is polymorphic. Our method provides fundamental insights into the relationship between the genomic self and the immune self and accelerates the discovery of polymorphic MIPs (also known as minor histocompatibility antigens).


Subject(s)
Major Histocompatibility Complex/genetics , Peptides/chemistry , Polymorphism, Genetic/genetics , Alleles , Humans
5.
Mol Cell Proteomics ; 12(9): 2536-50, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23750026

ABSTRACT

Protein modification by small ubiquitin-like modifier (SUMO) modulates the activities of numerous proteins involved in different cellular functions such as gene transcription, cell cycle, and DNA repair. Comprehensive identification of SUMOylated sites is a prerequisite to determine how SUMOylation regulates protein function. However, mapping SUMOylated Lys residues by mass spectrometry (MS) is challenging because of the dynamic nature of this modification, the existence of three functionally distinct human SUMO paralogs, and the large SUMO chain remnant that remains attached to tryptic peptides. To overcome these problems, we created HEK293 cell lines that stably express functional SUMO paralogs with an N-terminal His6-tag and an Arg residue near the C terminus that leave a short five amino acid SUMO remnant upon tryptic digestion. We determined the fragmentation patterns of our short SUMO remnant peptides by collisional activation and electron transfer dissociation using synthetic peptide libraries. Activation using higher energy collisional dissociation on the LTQ-Orbitrap Elite identified SUMO paralog-specific fragment ions and neutral losses of the SUMO remnant with high mass accuracy (< 5 ppm). We exploited these features to detect SUMO modified tryptic peptides in complex cell extracts by correlating mass measurements of precursor and fragment ions using a data independent acquisition method. We also generated bioinformatics tools to retrieve MS/MS spectra containing characteristic fragment ions to the identification of SUMOylated peptide by conventional Mascot database searches. In HEK293 cell extracts, this MS approach uncovered low abundance SUMOylated peptides and 37 SUMO3-modified Lys residues in target proteins, most of which were previously unknown. Interestingly, we identified mixed SUMO-ubiquitin chains with ubiquitylated SUMO proteins (K20 and K32) and SUMOylated ubiquitin (K63), suggesting a complex crosstalk between these two modifications.


Subject(s)
Chromatography, Affinity/methods , Proteins/metabolism , Sumoylation , Amino Acid Sequence , Chromatography, Liquid , Databases, Protein , HEK293 Cells , Humans , Ions , Mass Spectrometry , Molecular Sequence Data , Molecular Weight , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Proteins/chemistry , Search Engine , Sequence Homology, Amino Acid , Time Factors , Trypsin/metabolism
6.
Mol Cell Proteomics ; 12(9): 2394-407, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23674617

ABSTRACT

Macrophages play an important role in innate and adaptive immunity as professional phagocytes capable of internalizing and degrading pathogens to derive antigens for presentation to T cells. They also produce pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) that mediate local and systemic responses and direct the development of adaptive immunity. The present work describes the use of label-free quantitative proteomics to profile the dynamic changes of proteins from resting and TNF-α-activated mouse macrophages. These analyses revealed that TNF-α activation of macrophages led to the down-regulation of mitochondrial proteins and the differential regulation of several proteins involved in vesicle trafficking and immune response. Importantly, we found that the down-regulation of mitochondria proteins occurred through mitophagy and was specific to TNF-α, as other cytokines such as IL-1ß and IFN-γ had no effect on mitochondria degradation. Furthermore, using a novel antigen presentation system, we observed that the induction of mitophagy by TNF-α enabled the processing and presentation of mitochondrial antigens at the cell surface by MHC class I molecules. These findings highlight an unsuspected role of TNF-α in mitophagy and expanded our understanding of the mechanisms responsible for MHC presentation of self-antigens.


Subject(s)
Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mitophagy/drug effects , Proteomics/methods , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigen Presentation/drug effects , Antigens/metabolism , Autophagy/drug effects , Cell Line , Computational Biology , Down-Regulation/drug effects , Histocompatibility Antigens Class I/metabolism , Macrophages/drug effects , Macrophages/ultrastructure , Membrane Proteins/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phospholipases A2, Cytosolic/metabolism , Protein Interaction Maps/drug effects , Proteome/metabolism
7.
J Proteome Res ; 11(2): 927-40, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22059388

ABSTRACT

This report examines the analytical benefits of high-field asymmetric waveform ion mobility spectrometry (FAIMS) coupled to liquid chromatography mass spectrometry (LC-MS) for phosphoproteomics analyses. The ability of FAIMS to separate multiply charged peptide ions from chemical interferences confers a unique advantage in phosphoproteomics by enhancing the detection of low abundance phosphopeptides. LC-FAIMS-MS experiments performed on TiO(2)-enriched tryptic digests from Drosophila melanogaster provided a 50% increase in phosphopeptide identification compared to conventional LC-MS analysis. Also, FAIMS can be used to select different population of multiply charged phosphopeptide ions prior to their activation with either collision activated dissociation (CAD) or electron transfer dissociation (ETD). Importantly, FAIMS enabled the resolution of coeluting phosphoisomers of different abundances to facilitate their unambiguous identification using conventional database search engines. The benefits of FAIMS in large-scale phosphoproteomics of D. melanogaster are further investigated using label-free quantitation to identify differentially regulated phosphoproteins in response to insulin stimulation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Insulin/metabolism , Phosphopeptides/analysis , Proteome/analysis , Proteomics/methods , Algorithms , Amino Acid Sequence , Animals , Cell Line , Chromatography, Liquid , Cluster Analysis , Decision Trees , Drosophila Proteins/analysis , Drosophila Proteins/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Molecular Sequence Data , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphoproteins/analysis , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Signal Transduction , Tandem Mass Spectrometry/methods
8.
Cell ; 143(2): 251-62, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20946983

ABSTRACT

Signaling pathways are controlled by a vast array of posttranslational mechanisms. By contrast, little is known regarding the mechanisms that regulate the expression of their core components. We conducted an RNAi screen in Drosophila for factors modulating RAS/MAPK signaling and identified the Exon Junction Complex (EJC) as a key element of this pathway. The EJC binds the exon-exon junctions of mRNAs and thus far, has been linked exclusively to postsplicing events. Here, we report that the EJC is required for proper splicing of mapk transcripts by a mechanism that apparently controls exon definition. Moreover, whole transcriptome and RT-PCR analyses of EJC-depleted cells revealed that the splicing of long intron-containing genes, which includes mapk, is sensitive to EJC activity. These results identify a role for the EJC in the splicing of a subset of transcripts and suggest that RAS/MAPK signaling depends on the regulation of MAPK levels by the EJC.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Exons , Introns , Mitogen-Activated Protein Kinases/genetics , RNA Splicing , Animals , Cell Line , Drosophila melanogaster/metabolism , RNA Precursors/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...