Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Immunol ; 21(11): 1408-1420, 2020 11.
Article in English | MEDLINE | ID: mdl-32868930

ABSTRACT

B lymphocyte development and selection are central to adaptive immunity and self-tolerance. These processes require B cell receptor (BCR) signaling and occur in bone marrow, an environment with variable hypoxia, but whether hypoxia-inducible factor (HIF) is involved is unknown. We show that HIF activity is high in human and murine bone marrow pro-B and pre-B cells and decreases at the immature B cell stage. This stage-specific HIF suppression is required for normal B cell development because genetic activation of HIF-1α in murine B cells led to reduced repertoire diversity, decreased BCR editing and developmental arrest of immature B cells, resulting in reduced peripheral B cell numbers. HIF-1α activation lowered surface BCR, CD19 and B cell-activating factor receptor and increased expression of proapoptotic BIM. BIM deletion rescued the developmental block. Administration of a HIF activator in clinical use markedly reduced bone marrow and transitional B cells, which has therapeutic implications. Together, our work demonstrates that dynamic regulation of HIF-1α is essential for normal B cell development.


Subject(s)
B-Lymphocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphopoiesis/genetics , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Biomarkers , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoglobulin Light Chains/genetics , Immunophenotyping , Mice , Mice, Knockout , RNA Editing , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Transcriptional Activation
2.
EMBO J ; 43(11): 2127-2165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38580776

ABSTRACT

The in vitro oxygen microenvironment profoundly affects the capacity of cell cultures to model physiological and pathophysiological states. Cell culture is often considered to be hyperoxic, but pericellular oxygen levels, which are affected by oxygen diffusivity and consumption, are rarely reported. Here, we provide evidence that several cell types in culture actually experience local hypoxia, with important implications for cell metabolism and function. We focused initially on adipocytes, as adipose tissue hypoxia is frequently observed in obesity and precedes diminished adipocyte function. Under standard conditions, cultured adipocytes are highly glycolytic and exhibit a transcriptional profile indicative of physiological hypoxia. Increasing pericellular oxygen diverted glucose flux toward mitochondria, lowered HIF1α activity, and resulted in widespread transcriptional rewiring. Functionally, adipocytes increased adipokine secretion and sensitivity to insulin and lipolytic stimuli, recapitulating a healthier adipocyte model. The functional benefits of increasing pericellular oxygen were also observed in macrophages, hPSC-derived hepatocytes and cardiac organoids. Our findings demonstrate that oxygen is limiting in many terminally-differentiated cell types, and that considering pericellular oxygen improves the quality, reproducibility and translatability of culture models.


Subject(s)
Adipocytes , Cell Differentiation , Oxygen , Oxygen/metabolism , Adipocytes/metabolism , Adipocytes/cytology , Humans , Cell Culture Techniques/methods , Animals , Glycolysis , Hepatocytes/metabolism , Cell Hypoxia , Mitochondria/metabolism , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cells, Cultured , Glucose/metabolism , Macrophages/metabolism
4.
Cell ; 152(5): 1008-20, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23452850

ABSTRACT

Metazoan evolution involves increasing protein domain complexity, but how this relates to control of biological decisions remains uncertain. The Ras guanine nucleotide exchange factor (RasGEF) Sos1 and its adaptor Grb2 are multidomain proteins that couple fibroblast growth factor (FGF) signaling to activation of the Ras-Erk pathway during mammalian development and drive embryonic stem cells toward the primitive endoderm (PrE) lineage. We show that the ability of Sos1/Grb2 to appropriately regulate pluripotency and differentiation factors and to initiate PrE development requires collective binding of multiple Sos1/Grb2 domains to their protein and phospholipid ligands. This provides a cooperative system that only allows lineage commitment when all ligand-binding domains are occupied. Furthermore, our results indicate that the interaction domains of Sos1 and Grb2 have evolved so as to bind ligands not with maximal strength but with specificities and affinities that maintain cooperativity. This optimized system ensures that PrE lineage commitment occurs in a timely and selective manner during embryogenesis.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , GRB2 Adaptor Protein/metabolism , SOS1 Protein/metabolism , Amino Acid Sequence , Animals , Cell Lineage , Endoderm/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , ras Guanine Nucleotide Exchange Factors/metabolism
5.
Cell ; 147(6): 1340-54, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153077

ABSTRACT

The poly(ADP-ribose)polymerases Tankyrase 1/2 (TNKS/TNKS2) catalyze the covalent linkage of ADP-ribose polymer chains onto target proteins, regulating their ubiquitylation, stability, and function. Dysregulation of substrate recognition by Tankyrases underlies the human disease cherubism. Tankyrases recruit specific motifs (often called RxxPDG "hexapeptides") in their substrates via an N-terminal region of ankyrin repeats. These ankyrin repeats form five domains termed ankyrin repeat clusters (ARCs), each predicted to bind substrate. Here we report crystal structures of a representative ARC of TNKS2 bound to targeting peptides from six substrates. Using a solution-based peptide library screen, we derive a rule-based consensus for Tankyrase substrates common to four functionally conserved ARCs. This 8-residue consensus allows us to rationalize all known Tankyrase substrates and explains the basis for cherubism-causing mutations in the Tankyrase substrate 3BP2. Structural and sequence information allows us to also predict and validate other Tankyrase targets, including Disc1, Striatin, Fat4, RAD54, BCR, and MERIT40.


Subject(s)
Cherubism/metabolism , Tankyrases/chemistry , Tankyrases/metabolism , Amino Acid Sequence , Animals , Ankyrin Repeat , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Molecular Sequence Data , Sequence Alignment
6.
Mol Cell Proteomics ; 23(6): 100771, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642805

ABSTRACT

Signaling networks are critical for virtually all cell functions. Our current knowledge of cell signaling has been summarized in signaling pathway databases, which, while useful, are highly biased toward well-studied processes, and do not capture context specific network wiring or pathway cross-talk. Mass spectrometry-based phosphoproteomics data can provide a more unbiased view of active cell signaling processes in a given context, however, it suffers from low signal-to-noise ratio and poor reproducibility across experiments. While progress in methods to extract active signaling signatures from such data has been made, there are still limitations with respect to balancing bias and interpretability. Here we present phuEGO, which combines up-to-three-layer network propagation with ego network decomposition to provide small networks comprising active functional signaling modules. PhuEGO boosts the signal-to-noise ratio from global phosphoproteomics datasets, enriches the resulting networks for functional phosphosites and allows the improved comparison and integration across datasets. We applied phuEGO to five phosphoproteomics data sets from cell lines collected upon infection with SARS CoV2. PhuEGO was better able to identify common active functions across datasets and to point to a subnetwork enriched for known COVID-19 targets. Overall, phuEGO provides a flexible tool to the community for the improved functional interpretation of global phosphoproteomics datasets.


Subject(s)
Phosphoproteins , Proteomics , Signal Transduction , Proteomics/methods , Humans , Phosphoproteins/metabolism , SARS-CoV-2/metabolism , COVID-19/metabolism , COVID-19/virology , Software , Phosphorylation , Mass Spectrometry/methods
7.
Genome Res ; 32(4): 750-765, 2022 04.
Article in English | MEDLINE | ID: mdl-35197309

ABSTRACT

The morphology of breast cancer cells is often used as an indicator of tumor severity and prognosis. Additionally, morphology can be used to identify more fine-grained, molecular developments within a cancer cell, such as transcriptomic changes and signaling pathway activity. Delineating the interface between morphology and signaling is important to understand the mechanical cues that a cell processes in order to undergo epithelial-to-mesenchymal transition and consequently metastasize. However, the exact regulatory systems that define these changes remain poorly characterized. In this study, we used a network-systems approach to integrate imaging data and RNA-seq expression data. Our workflow allowed the discovery of unbiased and context-specific gene expression signatures and cell signaling subnetworks relevant to the regulation of cell shape, rather than focusing on the identification of previously known, but not always representative, pathways. By constructing a cell-shape signaling network from shape-correlated gene expression modules and their upstream regulators, we found central roles for developmental pathways such as WNT and Notch, as well as evidence for the fine control of NF-kB signaling by numerous kinase and transcriptional regulators. Further analysis of our network implicates a gene expression module enriched in the RAP1 signaling pathway as a mediator between the sensing of mechanical stimuli and regulation of NF-kB activity, with specific relevance to cell shape in breast cancer.


Subject(s)
Breast Neoplasms , NF-kappa B , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Shape , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Transcriptome
8.
Mol Syst Biol ; 19(3): e10631, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36688815

ABSTRACT

Genetic alterations in cancer cells trigger oncogenic transformation, a process largely mediated by the dysregulation of kinase and transcription factor (TF) activities. While the mutational profiles of thousands of tumours have been extensively characterised, the measurements of protein activities have been technically limited until recently. We compiled public data of matched genomics and (phospho)proteomics measurements for 1,110 tumours and 77 cell lines that we used to estimate activity changes in 218 kinases and 292 TFs. Co-regulation of kinase and TF activities reflects previously known regulatory relationships and allows us to dissect genetic drivers of signalling changes in cancer. We find that loss-of-function mutations are not often associated with the dysregulation of downstream targets, suggesting frequent compensatory mechanisms. Finally, we identified the activities most differentially regulated in cancer subtypes and showed how these can be linked to differences in patient survival. Our results provide broad insights into the dysregulation of protein activities in cancer and their contribution to disease severity.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Signal Transduction , Genomics , Proteomics/methods , Gene Expression Regulation
9.
Mol Syst Biol ; 19(12): e11462, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38031960

ABSTRACT

Endothelial dysfunction (ED) is critical in the development and progression of cardiovascular (CV) disorders, yet effective therapeutic targets for ED remain elusive due to limited understanding of its underlying molecular mechanisms. To address this gap, we employed a systems biology approach to identify potential targets for ED. Our study combined multi omics data integration, with siRNA screening, high content imaging and network analysis to prioritise key ED genes and identify a pro- and anti-ED network. We found 26 genes that, upon silencing, exacerbated the ED phenotypes tested, and network propagation identified a pro-ED network enriched in functions associated with inflammatory responses. Conversely, 31 genes ameliorated ED phenotypes, pointing to potential ED targets, and the respective anti-ED network was enriched in hypoxia, angiogenesis and cancer-related processes. An independent screen with 17 drugs found general agreement with the trends from our siRNA screen and further highlighted DUSP1, IL6 and CCL2 as potential candidates for targeting ED. Overall, our results demonstrate the potential of integrated system biology approaches in discovering disease-specific candidate drug targets for endothelial dysfunction.


Subject(s)
Systems Biology , RNA, Small Interfering
10.
Mol Cell ; 57(5): 936-947, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25747659

ABSTRACT

Cells chemically isolate molecules in compartments to both facilitate and regulate their interactions. In addition to membrane-encapsulated compartments, cells can form proteinaceous and membraneless organelles, including nucleoli, Cajal and PML bodies, and stress granules. The principles that determine when and why these structures form have remained elusive. Here, we demonstrate that the disordered tails of Ddx4, a primary constituent of nuage or germ granules, form phase-separated organelles both in live cells and in vitro. These bodies are stabilized by patterned electrostatic interactions that are highly sensitive to temperature, ionic strength, arginine methylation, and splicing. Sequence determinants are used to identify proteins found in both membraneless organelles and cell adhesion. Moreover, the bodies provide an alternative solvent environment that can concentrate single-stranded DNA but largely exclude double-stranded DNA. We propose that phase separation of disordered proteins containing weakly interacting blocks is a general mechanism for forming regulated, membraneless organelles.


Subject(s)
Cytoplasmic Granules/chemistry , DEAD-box RNA Helicases/chemistry , Organelles/chemistry , Phase Transition , Amino Acid Sequence , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cytoplasmic Granules/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA/chemistry , DNA/metabolism , HeLa Cells , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Methylation , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Organelles/metabolism , Osmolar Concentration , Sequence Homology, Amino Acid , Static Electricity , Time-Lapse Imaging , Transition Temperature
11.
Proteomics ; 21(23-24): e2000034, 2021 12.
Article in English | MEDLINE | ID: mdl-34314098

ABSTRACT

Transcription factors (TFs) are key regulators of intrinsic cellular processes, such as differentiation and development, and of the cellular response to external perturbation through signaling pathways. In this review we focus on the role of TFs as a link between signaling pathways and gene regulation. Cell signaling tends to result in the modulation of a set of TFs that then lead to changes in the cell's transcriptional program. We highlight the molecular layers at which TF activity can be measured and the associated technical and conceptual challenges. These layers include post-translational modifications (PTMs) of the TF, regulation of TF binding to DNA through chromatin accessibility and epigenetics, and expression of target genes. We highlight that a large number of TFs are understudied in both signaling and gene regulation studies, and that our knowledge about known TF targets has a strong literature bias. We argue that TFs serve as a perfect bridge between the fields of gene regulation and signaling, and that separating these fields hinders our understanding of cell functions. Multi-omics approaches that measure multiple dimensions of TF activity are ideally suited to study the interplay of cell signaling and gene regulation using TFs as the anchor to link the two fields.


Subject(s)
Gene Expression Regulation , Transcription Factors , Chromatin , Gene Regulatory Networks , Protein Binding , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Brief Bioinform ; 20(3): 767-777, 2019 05 21.
Article in English | MEDLINE | ID: mdl-29077858

ABSTRACT

Recent advances in proteomics allow the accurate measurement of abundances for thousands of proteins and phosphoproteins from multiple samples in parallel. Therefore, for the first time, we have the opportunity to measure the proteomic profiles of thousands of patient samples or disease model cell lines in a systematic way, to identify the precise underlying molecular mechanism and discover personalized biomarkers, networks and treatments. Here, we review examples of successful use of proteomics and phosphoproteomics data sets in as well as their integration other omics data sets with the aim of precision medicine. We will discuss the bioinformatics challenges posed by the generation, analysis and integration of such large data sets and present potential reasons why proteomics profiling and biomarkers are not currently widely used in the clinical setting. We will finally discuss ways to contribute to the better use of proteomics data in precision medicine and the clinical setting.


Subject(s)
Phosphoproteins/metabolism , Precision Medicine , Proteomics , Biomarkers/metabolism , Computational Biology , Humans
13.
Mol Syst Biol ; 16(10): e9698, 2020 10.
Article in English | MEDLINE | ID: mdl-33073517

ABSTRACT

An emerging theme from large-scale genetic screens that identify genes essential for cell fitness is that essentiality of a given gene is highly context-specific. Identification of such contexts could be the key to defining gene function and also to develop novel therapeutic interventions. Here, we present Context-specific Essentiality Network-tools (CEN-tools), a website and python package, in which users can interrogate the essentiality of a gene from large-scale genome-scale CRISPR screens in a number of biological contexts including tissue of origin, mutation profiles, expression levels and drug responses. We show that CEN-tools is suitable for the systematic identification of genetic dependencies and for more targeted queries. The associations between genes and a given context are represented as dependency networks (CENs), and we demonstrate the utility of these networks in elucidating novel gene functions. In addition, we integrate the dependency networks with existing protein-protein interaction networks to reveal context-dependent essential cellular pathways in cancer cells. Together, we demonstrate the applicability of CEN-tools in aiding the current efforts to define the human cellular dependency map.


Subject(s)
Computational Biology/methods , Genes, Essential , Melanoma/genetics , Melanoma/metabolism , Metabolomics/methods , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Gene Regulatory Networks , Humans , Melanoma/pathology , Mutation , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction/genetics , Skin Neoplasms/pathology , Software
14.
PLoS Comput Biol ; 13(3): e1005462, 2017 03.
Article in English | MEDLINE | ID: mdl-28346509

ABSTRACT

Proteomics techniques can identify thousands of phosphorylation sites in a single experiment, the majority of which are new and lack precise information about function or molecular mechanism. Here we present a fast method to predict potential phosphorylation switches by mapping phosphorylation sites to protein-protein interactions of known structure and analysing the properties of the protein interface. We predict 1024 sites that could potentially enable or disable particular interactions. We tested a selection of these switches and showed that phosphomimetic mutations indeed affect interactions. We estimate that there are likely thousands of phosphorylation mediated switches yet to be uncovered, even among existing phosphorylation datasets. The results suggest that phosphorylation sites on globular, as distinct from disordered, parts of the proteome frequently function as switches, which might be one of the ancient roles for kinase phosphorylation.


Subject(s)
Models, Chemical , Phosphotransferases/chemistry , Protein Interaction Mapping/methods , Proteome/chemistry , Sequence Analysis, Protein/methods , Binding Sites , Computer Simulation , Models, Molecular , Phosphorylation , Phosphotransferases/ultrastructure , Protein Binding , Protein Conformation , Proteome/ultrastructure , Structure-Activity Relationship
15.
Proc Natl Acad Sci U S A ; 112(13): E1594-603, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25829543

ABSTRACT

Systematic characterization of intercellular signaling approximating the physiological conditions of stimulation that involve direct cell-cell contact is challenging. We describe a proteomic strategy to analyze physiological signaling mediated by the T-cell costimulatory receptor CD28. We identified signaling pathways activated by CD28 during direct cell-cell contact by global analysis of protein phosphorylation. To define immediate CD28 targets, we used phosphorylated forms of the CD28 cytoplasmic region to obtain the CD28 interactome. The interaction profiles of selected CD28-interacting proteins were further characterized in vivo for amplifying the CD28 interactome. The combination of the global phosphorylation and interactome analyses revealed broad regulation of CD28 and its interactome by phosphorylation. Among the cellular phosphoproteins influenced by CD28 signaling, CapZ-interacting protein (CapZIP), a regulator of the actin cytoskeleton, was implicated by functional studies. The combinatorial approach applied herein is widely applicable for characterizing signaling networks associated with membrane receptors with short cytoplasmic tails.


Subject(s)
CD28 Antigens/metabolism , Cell Communication , Gene Expression Regulation , Receptors, Peptide/metabolism , Actins/metabolism , Cell Line, Tumor , Cytoskeleton/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Mass Spectrometry , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proteomics , Signal Transduction
16.
Int J Mol Sci ; 19(4)2018 Mar 21.
Article in English | MEDLINE | ID: mdl-29561791

ABSTRACT

The cellular signalling process is a highly complex mechanism, involving multiple players, which together orchestrate the cell's response to environmental changes and perturbations. Given the multitude of genes that participate in the process of cellular signalling, its study in a genome-wide manner has proven challenging. Recent advances in gene editing technologies, including clustered regularly-interspaced short palindromic repeats/Cas9 (CRISPR/Cas9) approaches, have opened new opportunities to investigate global regulatory signalling programs of cells in an unbiased manner. In this review, we focus on how the application of pooled genetic screening approaches using the CRISPR/Cas9 system has contributed to a systematic understanding of cellular signalling processes in normal and disease contexts.


Subject(s)
CRISPR-Cas Systems/genetics , Cells/metabolism , Genetic Testing , Genome , Signal Transduction
17.
Mol Syst Biol ; 12(4): 863, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27107012

ABSTRACT

High-throughput binary protein interaction mapping is continuing to extend our understanding of cellular function and disease mechanisms. However, we remain one or two orders of magnitude away from a complete interaction map for humans and other major model organisms. Completion will require screening at substantially larger scales with many complementary assays, requiring further efficiency gains in proteome-scale interaction mapping. Here, we report Barcode Fusion Genetics-Yeast Two-Hybrid (BFG-Y2H), by which a full matrix of protein pairs can be screened in a single multiplexed strain pool. BFG-Y2H uses Cre recombination to fuse DNA barcodes from distinct plasmids, generating chimeric protein-pair barcodes that can be quantified via next-generation sequencing. We applied BFG-Y2H to four different matrices ranging in scale from ~25 K to 2.5 M protein pairs. The results show that BFG-Y2H increases the efficiency of protein matrix screening, with quality that is on par with state-of-the-art Y2H methods.


Subject(s)
Centrosome/metabolism , Protein Interaction Mapping/methods , Proteome/metabolism , Saccharomyces cerevisiae/genetics , Chromosomes, Human/metabolism , Gene Library , High-Throughput Nucleotide Sequencing , Humans , Protein Binding , Two-Hybrid System Techniques
18.
Nucleic Acids Res ; 43(W1): W276-82, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25948583

ABSTRACT

While phospho-proteomics studies have shed light on the dynamics of cellular signaling, they mainly describe global effects and rarely explore mechanistic details, such as kinase/substrate relationships. Tools and databases, such as NetworKIN and PhosphoSitePlus, provide valuable regulatory details on signaling networks but rely on prior knowledge. They therefore provide limited information on less studied kinases and fewer unexpected relationships given that better studied signaling events can mask condition- or cell-specific 'network wiring'. SELPHI is a web-based tool providing in-depth analysis of phospho-proteomics data that is intuitive and accessible to non-bioinformatics experts. It uses correlation analysis of phospho-sites to extract kinase/phosphatase and phospho-peptide associations, and highlights the potential flow of signaling in the system under study. We illustrate SELPHI via analysis of phospho-proteomics data acquired in the presence of erlotinib-a tyrosine kinase inhibitor (TKI)-in cancer cells expressing TKI-resistant and -sensitive variants of the Epidermal Growth Factor Receptor. In this data set, SELPHI revealed information overlooked by the reporting study, including the known role of MET and EPHA2 kinases in conferring resistance to erlotinib in TKI sensitive strains. SELPHI can significantly enhance the analysis of phospho-proteomics data contributing to improved understanding of sample-specific signaling networks. SELPHI is freely available via http://llama.mshri.on.ca/SELPHI.


Subject(s)
Protein Kinases/metabolism , Proteomics/methods , Signal Transduction , Software , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Internet , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Peptides/chemistry , Peptides/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology
19.
PLoS Comput Biol ; 11(3): e1004147, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25794154

ABSTRACT

Identifying driver mutations and their functional consequences is critical to our understanding of cancer. Towards this goal, and because domains are the functional units of a protein, we explored the protein domain-level landscape of cancer-type-specific somatic mutations. Specifically, we systematically examined tumor genomes from 21 cancer types to identify domains with high mutational density in specific tissues, the positions of mutational hotspots within these domains, and the functional and structural context where possible. While hotspots corresponding to specific gain-of-function mutations are expected for oncoproteins, we found that tumor suppressor proteins also exhibit strong biases toward being mutated in particular domains. Within domains, however, we observed the expected patterns of mutation, with recurrently mutated positions for oncogenes and evenly distributed mutations for tumor suppressors. For example, we identified both known and new endometrial cancer hotspots in the tyrosine kinase domain of the FGFR2 protein, one of which is also a hotspot in breast cancer, and found new two hotspots in the Immunoglobulin I-set domain in colon cancer. Thus, to prioritize cancer mutations for further functional studies aimed at more precise cancer treatments, we have systematically correlated mutations and cancer types at the protein domain level.


Subject(s)
Mutation/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Protein Structure, Tertiary/genetics , Cluster Analysis , Computational Biology , DNA Mutational Analysis , Humans , Models, Molecular , Sequence Analysis, Protein
20.
J Biol Chem ; 289(51): 35397-408, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25331951

ABSTRACT

SH2D5 is a mammalian-specific, uncharacterized adaptor-like protein that contains an N-terminal phosphotyrosine-binding domain and a C-terminal Src homology 2 (SH2) domain. We show that SH2D5 is highly enriched in adult mouse brain, particularly in Purkinjie cells in the cerebellum and the cornu ammonis of the hippocampus. Despite harboring two potential phosphotyrosine (Tyr(P)) recognition domains, SH2D5 binds minimally to Tyr(P) ligands, consistent with the absence of a conserved Tyr(P)-binding arginine residue in the SH2 domain. Immunoprecipitation coupled to mass spectrometry (IP-MS) from cultured cells revealed a prominent association of SH2D5 with breakpoint cluster region protein, a RacGAP that is also highly expressed in brain. This interaction occurred between the phosphotyrosine-binding domain of SH2D5 and an NxxF motif located within the N-terminal region of the breakpoint cluster region. siRNA-mediated depletion of SH2D5 in a neuroblastoma cell line, B35, induced a cell rounding phenotype correlated with low levels of activated Rac1-GTP, suggesting that SH2D5 affects Rac1-GTP levels. Taken together, our data provide the first characterization of the SH2D5 signaling protein.


Subject(s)
GTPase-Activating Proteins/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Shc Signaling Adaptor Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Brain/cytology , Brain/metabolism , Cell Line, Tumor , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , K562 Cells , Male , Mice, Inbred C57BL , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Phosphotyrosine/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcr/genetics , RNA Interference , Rats , Shc Signaling Adaptor Proteins/genetics , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL