Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Cell ; 159(4): 869-83, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417162

ABSTRACT

X chromosome inactivation (XCI) depends on the long noncoding RNA Xist and its recruitment of Polycomb Repressive Complex 2 (PRC2). PRC2 is also targeted to other sites throughout the genome to effect transcriptional repression. Using XCI as a model, we apply an unbiased proteomics approach to isolate Xist and PRC2 regulators and identified ATRX. ATRX unexpectedly functions as a high-affinity RNA-binding protein that directly interacts with RepA/Xist RNA to promote loading of PRC2 in vivo. Without ATRX, PRC2 cannot load onto Xist RNA nor spread in cis along the X chromosome. Moreover, epigenomic profiling reveals that genome-wide targeting of PRC2 depends on ATRX, as loss of ATRX leads to spatial redistribution of PRC2 and derepression of Polycomb responsive genes. Thus, ATRX is a required specificity determinant for PRC2 targeting and function.


Subject(s)
DNA Helicases/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation , Animals , DNA Helicases/isolation & purification , Embryonic Stem Cells/metabolism , Female , Male , Mice , Nuclear Proteins/isolation & purification , X-linked Nuclear Protein
2.
Nature ; 607(7917): 149-155, 2022 07.
Article in English | MEDLINE | ID: mdl-35705813

ABSTRACT

Immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex class I (MHC-I) molecules1-5. Current approaches to profiling of MHC-I-associated peptides, collectively known as the immunopeptidome, are limited to in vitro investigation or bulk tumour lysates, which limits our understanding of cancer-specific patterns of antigen presentation in vivo6. To overcome these limitations, we engineered an inducible affinity tag into the mouse MHC-I gene (H2-K1) and targeted this allele to the KrasLSL-G12D/+Trp53fl/fl mouse model (KP/KbStrep)7. This approach enabled us to precisely isolate MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma and from lung adenocarcinoma (LUAD) in vivo. In addition, we profiled the LUAD immunopeptidome from the alveolar type 2 cell of origin up to late-stage disease. Differential peptide presentation in LUAD was not predictable by mRNA expression or translation efficiency and is probably driven by post-translational mechanisms. Vaccination with peptides presented by LUAD in vivo induced CD8+ T cell responses in naive mice and tumour-bearing mice. Many peptides specific to LUAD, including immunogenic peptides, exhibited minimal expression of the cognate mRNA, which prompts the reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance8. Beyond cancer, the KbStrep allele is compatible with other Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease and autoimmunity.


Subject(s)
Antigens, Neoplasm , Peptides , Proteomics , Alveolar Epithelial Cells/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/analysis , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Lung Neoplasms/chemistry , Lung Neoplasms/immunology , Mice , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/immunology , Peptides/analysis , Peptides/chemistry , Peptides/immunology , RNA, Messenger
3.
Semin Immunol ; 66: 101733, 2023 03.
Article in English | MEDLINE | ID: mdl-36841147

ABSTRACT

Central to successful cancer immunotherapy is effective T cell antitumor immunity. Multiple targeted immunotherapies engineered to invigorate T cell-driven antitumor immunity rely on identifying the repertoire of T cell antigens expressed on the tumor cell surface. Mass spectrometry-based survey of such antigens ("immunopeptidomics") combined with other omics platforms and computational algorithms has been instrumental in identifying and quantifying tumor-derived T cell antigens. In this review, we discuss the types of tumor antigens that have emerged for targeted cancer immunotherapy and the immunopeptidomics methods that are central in MHC peptide identification and quantification. We provide an overview of the strength and limitations of mass spectrometry-driven approaches and how they have been integrated with other technologies to discover targetable T cell antigens for cancer immunotherapy. We highlight some of the emerging cancer immunotherapies that successfully capitalized on immunopeptidomics, their challenges, and mass spectrometry-based strategies that can support their development.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Antigens, Neoplasm , Immunotherapy , T-Lymphocytes , Peptides
4.
Proc Natl Acad Sci U S A ; 119(49): e2208900119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36454758

ABSTRACT

Combining multiple therapeutic strategies in NRAS/BRAF mutant melanoma-namely MEK/BRAF kinase inhibitors, immune checkpoint inhibitors (ICIs), and targeted immunotherapies-may offer an improved survival benefit by overcoming limitations associated with any individual therapy. Still, optimal combination, order, and timing of administration remains under investigation. Here, we measure how MEK inhibition (MEKi) alters anti-tumor immunity by utilizing quantitative immunopeptidomics to profile changes in the peptide major histocompatibility molecules (pMHC) repertoire. These data reveal a collection of tumor antigens whose presentation levels are selectively augmented following therapy, including several epitopes present at over 1,000 copies per cell. We leveraged the tunable abundance of MEKi-modulated antigens by targeting four epitopes with pMHC-specific T cell engagers and antibody drug conjugates, enhancing cell killing in tumor cells following MEK inhibition. These results highlight drug treatment as a means to enhance immunotherapy efficacy by targeting specific upregulated pMHCs and provide a methodological framework for identifying, quantifying, and therapeutically targeting additional epitopes of interest.


Subject(s)
Melanoma , Mitogen-Activated Protein Kinase Kinases , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Antigens, Neoplasm/genetics , Melanoma/drug therapy , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Epitopes
5.
Mol Cancer ; 23(1): 17, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38229082

ABSTRACT

Triple negative breast cancer (TNBC) is a heterogeneous group of tumors which lack estrogen receptor, progesterone receptor, and HER2 expression. Targeted therapies have limited success in treating TNBC, thus a strategy enabling effective targeted combinations is an unmet need. To tackle these challenges and discover individualized targeted combination therapies for TNBC, we integrated phosphoproteomic analysis of altered signaling networks with patient-specific signaling signature (PaSSS) analysis using an information-theoretic, thermodynamic-based approach. Using this method on a large number of TNBC patient-derived tumors (PDX), we were able to thoroughly characterize each PDX by computing a patient-specific set of unbalanced signaling processes and assigning a personalized therapy based on them. We discovered that each tumor has an average of two separate processes, and that, consistent with prior research, EGFR is a major core target in at least one of them in half of the tumors analyzed. However, anti-EGFR monotherapies were predicted to be ineffective, thus we developed personalized combination treatments based on PaSSS. These were predicted to induce anti-EGFR responses or to be used to develop an alternative therapy if EGFR was not present.In-vivo experimental validation of the predicted therapy showed that PaSSS predictions were more accurate than other therapies. Thus, we suggest that a detailed identification of molecular imbalances is necessary to tailor therapy for each TNBC. In summary, we propose a new strategy to design personalized therapy for TNBC using pY proteomics and PaSSS analysis. This method can be applied to different cancer types to improve response to the biomarker-based treatment.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Signal Transduction
6.
Am J Med Genet A ; 194(4): e63477, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37969032

ABSTRACT

Germline pathogenic variants in the RAS/mitogen-activated protein kinase (MAPK) signaling pathway are the molecular cause of RASopathies, a group of clinically overlapping genetic syndromes. RASopathies constitute a wide clinical spectrum characterized by distinct facial features, short stature, predisposition to cancer, and variable anomalies in nearly all the major body systems. With increasing global recognition of these conditions, the 8th International RASopathies Symposium spotlighted global perspectives on clinical care and research, including strategies for building international collaborations and developing diverse patient cohorts in anticipation of interventional trials. This biannual meeting, organized by RASopathies Network, was held in a hybrid virtual/in-person format. The agenda featured emerging discoveries and case findings as well as progress in preclinical and therapeutic pipelines. Stakeholders including basic scientists, clinician-scientists, practitioners, industry representatives, patients, and family advocates gathered to discuss cutting edge science, recognize current gaps in knowledge, and hear from people with RASopathies about the experience of daily living. Presentations by RASopathy self-advocates and early-stage investigators were featured throughout the program to encourage a sustainable, diverse, long-term research and advocacy partnership focused on improving health and bringing treatments to people with RASopathies.


Subject(s)
Costello Syndrome , Ectodermal Dysplasia , Heart Defects, Congenital , Neoplasms , Noonan Syndrome , Humans , ras Proteins/genetics , MAP Kinase Signaling System/genetics , Costello Syndrome/genetics , Neoplasms/genetics , Ectodermal Dysplasia/genetics , Noonan Syndrome/genetics , Heart Defects, Congenital/genetics
8.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34497125

ABSTRACT

Absolute quantification measurements (copies per cell) of peptide major histocompatibility complex (pMHC) antigens are necessary to inform targeted immunotherapy drug design; however, existing methods for absolute quantification have critical limitations. Here, we present a platform termed SureQuant-IsoMHC, utilizing a series of pMHC isotopologues and internal standard-triggered targeted mass spectrometry to generate an embedded multipoint calibration curve to determine endogenous pMHC concentrations for a panel of 18 tumor antigens. We apply SureQuant-IsoMHC to measure changes in expression of our target panel in a melanoma cell line treated with a MEK inhibitor and translate this approach to estimate antigen concentrations in melanoma tumor biopsies.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/analysis , Benzimidazoles/pharmacology , Histocompatibility Antigens Class I/immunology , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/immunology , Antigen Presentation/drug effects , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/metabolism , Tumor Cells, Cultured
9.
Mol Cell Proteomics ; 20: 100104, 2021.
Article in English | MEDLINE | ID: mdl-34052394

ABSTRACT

Utilizing a protein carrier in combination with isobaric labeling to "boost" the signal of other low-level samples in multiplexed analyses has emerged as an attractive strategy to enhance data quantity while minimizing protein input in mass spectrometry analyses. Recent applications of this approach include pMHC profiling and tyrosine phosphoproteomics, two applications that are often limited by large sample requirements. While including a protein carrier has been shown to increase the number of identifiable peptides in both applications, the impact of a protein carrier on quantitative accuracy remains to be thoroughly explored, particularly in relevant biological contexts where samples exhibit dynamic changes in abundance across peptides. Here, we describe two sets of analyses comparing MS2-based quantitation using a 20× protein carrier in pMHC analyses and a high (~100×) and low (~9×) protein carrier in pTyr analyses, using CDK4/6 inhibitors and EGF stimulation to drive dynamic changes in the immunopeptidome and phosphoproteome, respectively. In both applications, inclusion of a protein carrier resulted in an increased number of MHC peptide or phosphopeptide identifications, as expected. At the same time, quantitative accuracy was adversely affected by the presence of the protein carrier, altering interpretation of the underlying biological response to perturbation. Moreover, for tyrosine phosphoproteomics, the presence of high levels of protein carrier led to a large number of missing values for endogenous phosphopeptides, leading to fewer quantifiable peptides relative to the "no-boost" condition. These data highlight the unique limitations and future experimental considerations for both analysis types and provide a framework for assessing quantitative accuracy in protein carrier experiments moving forward.


Subject(s)
Phosphopeptides/metabolism , Tyrosine/metabolism , Cell Line, Tumor , Humans , Phosphorylation , Proteomics
10.
Expert Rev Proteomics ; 18(8): 661-674, 2021 08.
Article in English | MEDLINE | ID: mdl-34468274

ABSTRACT

INTRODUCTION: Many pathologies, including cancer, have been associated with aberrant phosphorylation-mediated signaling networks that drive altered cell proliferation, migration, metabolic regulation, and can lead to systemic inflammation. Phosphoproteomics, the large-scale analysis of protein phosphorylation sites, has emerged as a powerful tool to define signaling network regulation and dysregulation in normal and pathological conditions. AREAS COVERED: We provide an overview of methodology for global phosphoproteomics as well as enrichment of specific subsets of the phosphoproteome, including phosphotyrosine and phospho-motif enrichment of kinase substrates. We review quantitative methods, advantages and limitations of different mass spectrometry acquisition formats, and computational approaches to extract biological insight from phosphoproteomics data. Throughout, we discuss various applications and their challenges in implementation. EXPERT OPINION: Over the past 20 years the field of phosphoproteomics has advanced to enable deep biological and clinical insight through the quantitative analysis of signaling networks. Future areas of development include Clinical Laboratory Improvement Amendments (CLIA)-approved methods for analysis of clinical samples, continued improvements in sensitivity to enable analysis of small numbers of rare cells and tissue microarrays, and computational methods to integrate data resulting from multiple systems-level quantitative analytical methods.


Subject(s)
Neoplasms , Proteomics , Mass Spectrometry , Phosphoproteins/metabolism , Phosphorylation , Signal Transduction
11.
Mol Syst Biol ; 16(12): e9819, 2020 12.
Article in English | MEDLINE | ID: mdl-33289969

ABSTRACT

Alzheimer's disease (AD) is characterized by the appearance of amyloid-ß plaques, neurofibrillary tangles, and inflammation in brain regions involved in memory. Using mass spectrometry, we have quantified the phosphoproteome of the CK-p25, 5XFAD, and Tau P301S mouse models of neurodegeneration. We identified a shared response involving Siglec-F which was upregulated on a subset of reactive microglia. The human paralog Siglec-8 was also upregulated on microglia in AD. Siglec-F and Siglec-8 were upregulated following microglial activation with interferon gamma (IFNγ) in BV-2 cell line and human stem cell-derived microglia models. Siglec-F overexpression activates an endocytic and pyroptotic inflammatory response in BV-2 cells, dependent on its sialic acid substrates and immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation sites. Related human Siglecs induced a similar response in BV-2 cells. Collectively, our results point to an important role for mouse Siglec-F and human Siglec-8 in regulating microglial activation during neurodegeneration.


Subject(s)
Inflammation/pathology , Microglia/metabolism , Nerve Degeneration/pathology , Phosphoproteins/metabolism , Proteomics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Antibodies/metabolism , Cell Death , Cell Line , Humans , Inflammation/metabolism , Interferon-gamma/metabolism , Mice, Transgenic , Microglia/pathology , Nerve Degeneration/metabolism , Peptides/metabolism , Phagocytosis , Phosphotyrosine/metabolism , Proteome/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/chemistry , Signal Transduction , Up-Regulation
12.
Genes Dev ; 27(19): 2147-63, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24115771

ABSTRACT

Production of haploid gametes from diploid progenitor cells is mediated by a specialized cell division, meiosis, where two divisions, meiosis I and II, follow a single S phase. Errors in progression from meiosis I to meiosis II lead to aneuploid and polyploid gametes, but the regulatory mechanisms controlling this transition are poorly understood. Here, we demonstrate that the conserved kinase Ime2 regulates the timing and order of the meiotic divisions by controlling translation. Ime2 coordinates translational activation of a cluster of genes at the meiosis I-meiosis II transition, including the critical determinant of the meiotic chromosome segregation pattern CLB3. We further show that Ime2 mediates translational control through the meiosis-specific RNA-binding protein Rim4. Rim4 inhibits translation of CLB3 during meiosis I by interacting with the 5' untranslated region (UTR) of CLB3. At the onset of meiosis II, Ime2 kinase activity rises and triggers a decrease in Rim4 protein levels, thereby alleviating translational repression. Our results elucidate a novel developmentally regulated translational control pathway that establishes the meiotic chromosome segregation pattern.


Subject(s)
Chromosome Segregation/genetics , Gene Expression Regulation, Fungal , Meiosis/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , 5' Untranslated Regions/genetics , Intracellular Signaling Peptides and Proteins , Multigene Family/genetics , Protein Binding , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
13.
Mol Syst Biol ; 15(8): e8849, 2019 08.
Article in English | MEDLINE | ID: mdl-31464373

ABSTRACT

Obesity-associated type 2 diabetes and accompanying diseases have developed into a leading human health risk across industrialized and developing countries. The complex molecular underpinnings of how lipid overload and lipid metabolites lead to the deregulation of metabolic processes are incompletely understood. We assessed hepatic post-translational alterations in response to treatment of cells with saturated and unsaturated free fatty acids and the consumption of a high-fat diet by mice. These data revealed widespread tyrosine phosphorylation changes affecting a large number of enzymes involved in metabolic processes as well as canonical receptor-mediated signal transduction networks. Targeting two of the most prominently affected molecular features in our data, SRC-family kinase activity and elevated reactive oxygen species, significantly abrogated the effects of saturated fat exposure in vitro and high-fat diet in vivo. In summary, we present a comprehensive view of diet-induced alterations of tyrosine signaling networks, including proteins involved in fundamental metabolic pathways.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Liver/drug effects , Obesity/metabolism , Phosphotyrosine/metabolism , Protein Processing, Post-Translational , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Fatty Acids/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology , Obesity/genetics , Obesity/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Rats , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , src-Family Kinases/genetics , src-Family Kinases/metabolism
14.
Nat Chem Biol ; 14(3): 206-214, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29443976

ABSTRACT

Despite decades of accumulated knowledge about proteins and their post-translational modifications (PTMs), numerous questions remain regarding their molecular composition and biological function. One of the most fundamental queries is the extent to which the combinations of DNA-, RNA- and PTM-level variations explode the complexity of the human proteome. Here, we outline what we know from current databases and measurement strategies including mass spectrometry-based proteomics. In doing so, we examine prevailing notions about the number of modifications displayed on human proteins and how they combine to generate the protein diversity underlying health and disease. We frame central issues regarding determination of protein-level variation and PTMs, including some paradoxes present in the field today. We use this framework to assess existing data and to ask the question, "How many distinct primary structures of proteins (proteoforms) are created from the 20,300 human genes?" We also explore prospects for improving measurements to better regularize protein-level biology and efficiently associate PTMs to function and phenotype.


Subject(s)
Genome, Human , Protein Processing, Post-Translational , Proteins/chemistry , Proteome/chemistry , Proteomics/methods , Databases, Protein , Humans , Mass Spectrometry , Phenotype , Protein Biosynthesis , Protein Isoforms/chemistry , Ubiquitin/chemistry
15.
J Biol Chem ; 293(2): 623-637, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29162725

ABSTRACT

Phosphotyrosine (pTyr)-dependent signaling is critical for many cellular processes. It is highly dynamic, as signal output depends not only on phosphorylation and dephosphorylation rates but also on the rates of binding and dissociation of effectors containing phosphotyrosine-dependent binding modules such as Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains. Previous in vitro studies suggested that binding of SH2 and PTB domains can enhance protein phosphorylation by protecting the sites bound by these domains from phosphatase-mediated dephosphorylation. To test whether this occurs in vivo, we used the binding of growth factor receptor bound 2 (GRB2) to phosphorylated epidermal growth factor receptor (EGFR) as a model system. We analyzed the effects of SH2 domain overexpression on protein tyrosine phosphorylation by quantitative Western and far-Western blotting, mass spectrometry, and computational modeling. We found that SH2 overexpression results in a significant, dose-dependent increase in EGFR tyrosine phosphorylation, particularly of sites corresponding to the binding specificity of the overexpressed SH2 domain. Computational models using experimentally determined EGFR phosphorylation and dephosphorylation rates, and pTyr-EGFR and GRB2 concentrations, recapitulated the experimental findings. Surprisingly, both modeling and biochemical analyses suggested that SH2 domain overexpression does not result in a major decrease in the number of unbound phosphorylated SH2 domain-binding sites. Our results suggest that signaling via SH2 domain binding is buffered over a relatively wide range of effector concentrations and that SH2 domain proteins with overlapping binding specificities are unlikely to compete with one another for phosphosites in vivo.


Subject(s)
ErbB Receptors/metabolism , GRB2 Adaptor Protein/metabolism , Binding Sites , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Signal Transduction
16.
Nat Methods ; 13(9): 770-6, 2016 09.
Article in English | MEDLINE | ID: mdl-27479327

ABSTRACT

Uncovering the molecular context of dysregulated metabolites is crucial to understand pathogenic pathways. However, their system-level analysis has been limited owing to challenges in global metabolite identification. Most metabolite features detected by untargeted metabolomics carried out by liquid-chromatography-mass spectrometry cannot be uniquely identified without additional, time-consuming experiments. We report a network-based approach, prize-collecting Steiner forest algorithm for integrative analysis of untargeted metabolomics (PIUMet), that infers molecular pathways and components via integrative analysis of metabolite features, without requiring their identification. We demonstrated PIUMet by analyzing changes in metabolism of sphingolipids, fatty acids and steroids in a Huntington's disease model. Additionally, PIUMet enabled us to elucidate putative identities of altered metabolite features in diseased cells, and infer experimentally undetected, disease-associated metabolites and dysregulated proteins. Finally, we established PIUMet's ability for integrative analysis of untargeted metabolomics data with proteomics data, demonstrating that this approach elicits disease-associated metabolites and proteins that cannot be inferred by individual analysis of these data.


Subject(s)
Algorithms , Huntington Disease/metabolism , Metabolic Networks and Pathways , Metabolomics/methods , Neural Networks, Computer , Databases, Protein , Fatty Acids/metabolism , Humans , Machine Learning , Metabolomics/instrumentation , Sphingolipids/metabolism , Steroids/metabolism
17.
Nature ; 498(7453): 246-50, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23728299

ABSTRACT

DNA damage activates a signalling network that blocks cell-cycle progression, recruits DNA repair factors and/or triggers senescence or programmed cell death. Alterations in chromatin structure are implicated in the initiation and propagation of the DNA damage response. Here we further investigate the role of chromatin structure in the DNA damage response by monitoring ionizing-radiation-induced signalling and response events with a high-content multiplex RNA-mediated interference screen of chromatin-modifying and -interacting genes. We discover that an isoform of Brd4, a bromodomain and extra-terminal (BET) family member, functions as an endogenous inhibitor of DNA damage response signalling by recruiting the condensin II chromatin remodelling complex to acetylated histones through bromodomain interactions. Loss of this isoform results in relaxed chromatin structure, rapid cell-cycle checkpoint recovery and enhanced survival after irradiation, whereas functional gain of this isoform compacted chromatin, attenuated DNA damage response signalling and enhanced radiation-induced lethality. These data implicate Brd4, previously known for its role in transcriptional control, as an insulator of chromatin that can modulate the signalling response to DNA damage.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Damage , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Acetylation , Adenosine Triphosphatases/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/radiation effects , Chromatin/chemistry , Chromatin/radiation effects , Chromatin Assembly and Disassembly/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphorylation/radiation effects , Positive Transcriptional Elongation Factor B/metabolism , Protein Isoforms/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics
18.
Proc Natl Acad Sci U S A ; 113(11): 3114-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929352

ABSTRACT

Despite extensive study of the EGF receptor (EGFR) signaling network, the immediate posttranslational changes that occur in response to growth factor stimulation remain poorly characterized; as a result, the biological mechanisms underlying signaling initiation remain obscured. To address this deficiency, we have used a mass spectrometry-based approach to measure system-wide phosphorylation changes throughout the network with 10-s resolution in the 80 s after stimulation in response to a range of eight growth factor concentrations. Significant changes were observed on proteins far downstream in the network as early as 10 s after stimulation, indicating a system capable of transmitting information quickly. Meanwhile, canonical members of the EGFR signaling network fall into clusters with distinct activation patterns. Src homology 2 domain containing transforming protein (Shc) and phosphoinositol 3-kinase (PI3K) phosphorylation levels increase rapidly, but equilibrate within 20 s, whereas proteins such as Grb2-associated binder-1 (Gab1) and SH2-containing tyrosine phosphatase (SHP2) show slower, sustained increases. Proximity ligation assays reveal that Shc and Gab1 phosphorylation patterns are representative of separate timescales for physical association with the receptor. Inhibition of phosphatases with vanadate reveals site-specific regulatory mechanisms and also uncovers primed activating components in the network, including Src family kinases, whose inhibition affects only a subset of proteins within the network. The results presented highlight the complexity of signaling initiation and provide a window into exploring mechanistic hypotheses about receptor tyrosine kinase (RTK) biology.


Subject(s)
Breast/enzymology , ErbB Receptors/physiology , Signal Transduction/physiology , Breast/cytology , Cell Division , Cell Line , Culture Media, Serum-Free/pharmacology , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , ErbB Receptors/agonists , Female , Humans , Multiprotein Complexes , Phosphoprotein Phosphatases/physiology , Phosphoproteins/analysis , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Time Factors , src-Family Kinases/physiology
19.
Development ; 141(18): 3495-504, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25142464

ABSTRACT

Sirtuins are NAD(+)-dependent deacylases that regulate numerous biological processes in response to the environment. SirT1 is the mammalian ortholog of yeast Sir2, and is involved in many metabolic pathways in somatic tissues. Whole body deletion of SirT1 alters reproductive function in oocytes and the testes, in part caused by defects in central neuro-endocrine control. To study the function of SirT1 specifically in the male germ line, we deleted this sirtuin in male germ cells and found that mutant mice had smaller testes, a delay in differentiation of pre-meiotic germ cells, decreased spermatozoa number, an increased proportion of abnormal spermatozoa and reduced fertility. At the molecular level, mutants do not have the characteristic increase in acetylation of histone H4 at residues K5, K8 and K12 during spermiogenesis and demonstrate corresponding defects in the histone to protamine transition. Our findings thus reveal a germ cell-autonomous role of SirT1 in spermatogenesis.


Subject(s)
Cell Differentiation/genetics , Fertility/genetics , Germ Cells/physiology , Sirtuin 1/metabolism , Spermatogenesis/genetics , Acetylation , Animals , Cell Differentiation/physiology , Chromatin Assembly and Disassembly/genetics , Chromatography, Liquid , Female , Fertility/physiology , Fluorescent Antibody Technique , Histones/metabolism , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/genetics , Sirtuin 1/deficiency , Tandem Mass Spectrometry , Testis/metabolism
20.
J Immunol ; 192(1): 123-35, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24298013

ABSTRACT

IL-2 and IL-15 are common γ-chain family cytokines involved in regulation of T cell differentiation and homeostasis. Despite signaling through the same receptors, IL-2 and IL-15 have non-redundant roles in T cell biology, both physiologically and at the cellular level. The mechanisms by which IL-2 and IL-15 trigger distinct phenotypes in T cells remain elusive. To elucidate these mechanisms, we performed a quantitative comparison of the phosphotyrosine signaling network and resulting phenotypes triggered by IL-2 and IL-15. This study revealed that the signaling networks activated by IL-2 or IL-15 are highly similar and that T cell proliferation and metabolism are controlled in a quantitatively distinct manner through IL-2/15R signal strength independent of the cytokine identity. Distinct phenotypes associated with IL-2 or IL-15 stimulation therefore arise through differential regulation of IL-2/15R signal strength and duration because of differences in cytokine-receptor binding affinity, receptor expression levels, physiological cytokine levels, and cytokine-receptor intracellular trafficking kinetics. These results provide important insights into the function of other shared cytokine and growth factor receptors, quantitative regulation of cell proliferation and metabolism through signal transduction, and improved design of cytokine based clinical immunomodulatory therapies for cancer and infectious diseases.


Subject(s)
Interleukin-15/pharmacology , Interleukin-2/pharmacology , Phenotype , Receptors, Cytokine/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cell Line , Cell Proliferation/drug effects , Cluster Analysis , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Phosphorylation , Receptors, Interleukin-12/metabolism , Receptors, Interleukin-2/metabolism , T-Lymphocytes/immunology , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL