Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nature ; 618(7963): 159-168, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225977

ABSTRACT

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Subject(s)
Nerve Regeneration , Humans , Neoplasms/drug therapy , Nerve Regeneration/drug effects , Protein Isoforms/agonists , Signal Transduction/drug effects , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/drug effects , Cardiotonic Agents/pharmacology , Animals , Biocatalysis/drug effects , Protein Conformation/drug effects , Neurites/drug effects , Reperfusion Injury/prevention & control , Nerve Crush , Cell Proliferation/drug effects
2.
Elife ; 122023 04 03.
Article in English | MEDLINE | ID: mdl-37010886

ABSTRACT

Most studies of cohesin function consider the Stromalin Antigen (STAG/SA) proteins as core complex members given their ubiquitous interaction with the cohesin ring. Here, we provide functional data to support the notion that the SA subunit is not a mere passenger in this structure, but instead plays a key role in the localization of cohesin to diverse biological processes and promotes loading of the complex at these sites. We show that in cells acutely depleted for RAD21, SA proteins remain bound to chromatin, cluster in 3D and interact with CTCF, as well as with a wide range of RNA binding proteins involved in multiple RNA processing mechanisms. Accordingly, SA proteins interact with RNA, and R-loops, even in the absence of cohesin. Our results place SA1 on chromatin upstream of the cohesin ring and reveal a role for SA1 in cohesin loading which is independent of NIPBL, the canonical cohesin loader. We propose that SA1 takes advantage of structural R-loop platforms to link cohesin loading and chromatin structure with diverse functions. Since SA proteins are pan-cancer targets, and R-loops play an increasingly prevalent role in cancer biology, our results have important implications for the mechanistic understanding of SA proteins in cancer and disease.


Subject(s)
R-Loop Structures , RNA , RNA/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/metabolism , Chromatin , CCCTC-Binding Factor/metabolism , Cohesins
3.
PLoS One ; 16(8): e0254697, 2021.
Article in English | MEDLINE | ID: mdl-34424918

ABSTRACT

The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.


Subject(s)
Cell Adhesion/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Neurofibromin 2/genetics , Tumor Suppressor Proteins/genetics , Cell Proliferation/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Contact Inhibition/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Neoplasms/pathology , Protein Binding/genetics , Protein Interaction Maps/genetics , Signal Transduction/genetics
4.
Mol Cell ; 81(14): 2944-2959.e10, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34166609

ABSTRACT

A number of regulatory factors are recruited to chromatin by specialized RNAs. Whether RNA has a more general role in regulating the interaction of proteins with chromatin has not been determined. We used proteomics methods to measure the global impact of nascent RNA on chromatin in embryonic stem cells. Surprisingly, we found that nascent RNA primarily antagonized the interaction of chromatin modifiers and transcriptional regulators with chromatin. Transcriptional inhibition and RNA degradation induced recruitment of a set of transcriptional regulators, chromatin modifiers, nucleosome remodelers, and regulators of higher-order structure. RNA directly bound to factors, including BAF, NuRD, EHMT1, and INO80 and inhibited their interaction with nucleosomes. The transcriptional elongation factor P-TEFb directly bound pre-mRNA, and its recruitment to chromatin upon Pol II inhibition was regulated by the 7SK ribonucleoprotein complex. We postulate that by antagonizing the interaction of regulatory proteins with chromatin, nascent RNA links transcriptional output with chromatin composition.


Subject(s)
Chromatin/metabolism , RNA/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Male , Mice , Nucleosomes/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/physiology , Proteomics/methods , RNA Polymerase II/metabolism , Transcription, Genetic/physiology , Transcriptional Elongation Factors/metabolism
5.
Sci Rep ; 10(1): 663, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959764

ABSTRACT

Antibody combinations targeting cell surface receptors are a new modality of cancer therapy. The trafficking and signalling mechanisms regulated by such therapeutics are not fully understood but could underlie differential tumour responses. We explored EGFR trafficking upon treatment with the antibody combination Sym004 which has shown promise clinically. Sym004 promoted EGFR endocytosis distinctly from EGF: it was asynchronous, not accompanied by canonical signalling events and involved EGFR clustering within detergent-insoluble plasma mebrane-associated tubules. Sym004 induced lysosomal degradation independently of EGFR ubiquitylation but dependent upon Hrs/Tsg101 that are required for the formation of intraluminal vesicles (ILVs) within late endosomes. We propose Sym004 cross-links EGFR physically triggering EGFR endocytosis and incorporation onto ILVs and so Sym004 sensitivity correlates with EGFR numbers available for binding, rather than specific signalling events. Consistently Sym004 efficacy and potentiation of cisplatin responses correlated with EGFR surface expression in head and neck cancer cells. These findings will have implications in understanding the mode of action of this new class of cancer therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies , Antineoplastic Agents , Endocytosis/drug effects , Protein Transport , Cell Membrane/metabolism , Cells, Cultured , DNA-Binding Proteins , Endosomal Sorting Complexes Required for Transport , Endosomes/metabolism , ErbB Receptors/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/etiology , Head and Neck Neoplasms/metabolism , Humans , Lysosomes/metabolism , Phosphoproteins , Receptors, Cell Surface , Transcription Factors
6.
Mol Cell Proteomics ; 18(9): 1836-1850, 2019 09.
Article in English | MEDLINE | ID: mdl-31289117

ABSTRACT

Protein biomarkers for epithelial ovarian cancer are critical for the early detection of the cancer to improve patient prognosis and for the clinical management of the disease to monitor treatment response and to detect recurrences. Unfortunately, the discovery of protein biomarkers is hampered by the limited availability of reliable and sensitive assays needed for the reproducible quantification of proteins in complex biological matrices such as blood plasma. In recent years, targeted mass spectrometry, exemplified by selected reaction monitoring (SRM) has emerged as a method, capable of overcoming this limitation. Here, we present a comprehensive SRM-based strategy for developing plasma-based protein biomarkers for epithelial ovarian cancer and illustrate how the SRM platform, when combined with rigorous experimental design and statistical analysis, can result in detection of predictive analytes.Our biomarker development strategy first involved a discovery-driven proteomic effort to derive potential N-glycoprotein biomarker candidates for plasma-based detection of human ovarian cancer from a genetically engineered mouse model of endometrioid ovarian cancer, which accurately recapitulates the human disease. Next, 65 candidate markers selected from proteins of different abundance in the discovery dataset were reproducibly quantified with SRM assays across a large cohort of over 200 plasma samples from ovarian cancer patients and healthy controls. Finally, these measurements were used to derive a 5-protein signature for distinguishing individuals with epithelial ovarian cancer from healthy controls. The sensitivity of the candidate biomarker signature in combination with CA125 ELISA-based measurements currently used in clinic, exceeded that of CA125 ELISA-based measurements alone. The SRM-based strategy in this study is broadly applicable. It can be used in any study that requires accurate and reproducible quantification of selected proteins in a high-throughput and multiplexed fashion.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Ovarian Epithelial/blood , Mass Spectrometry/methods , Ovarian Neoplasms/blood , Proteomics/methods , Animals , Antigens, Neoplasm/blood , Blood Proteins/analysis , CA-125 Antigen/blood , Case-Control Studies , Cohort Studies , Desmoglein 2/blood , Female , Heavy Chain Disease/blood , Humans , Immunoglobulin mu-Chains/blood , Membrane Proteins/blood , Mice, Transgenic , Neural Cell Adhesion Molecule L1/blood , Sensitivity and Specificity , Thrombospondin 1/blood
7.
Proc Natl Acad Sci U S A ; 116(27): 13330-13339, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31213532

ABSTRACT

Despite the crucial role of RAF kinases in cell signaling and disease, we still lack a complete understanding of their regulation. Heterodimerization of RAF kinases as well as dephosphorylation of a conserved "S259" inhibitory site are important steps for RAF activation but the precise mechanisms and dynamics remain unclear. A ternary complex comprised of SHOC2, MRAS, and PP1 (SHOC2 complex) functions as a RAF S259 holophosphatase and gain-of-function mutations in SHOC2, MRAS, and PP1 that promote complex formation are found in Noonan syndrome. Here we show that SHOC2 complex-mediated S259 RAF dephosphorylation is critically required for growth factor-induced RAF heterodimerization as well as for MEK dissociation from BRAF. We also uncover SHOC2-independent mechanisms of RAF and ERK pathway activation that rely on N-region phosphorylation of CRAF. In DLD-1 cells stimulated with EGF, SHOC2 function is essential for a rapid transient phase of ERK activation, but is not required for a slow, sustained phase that is instead driven by palmitoylated H/N-RAS proteins and CRAF. Whereas redundant SHOC2-dependent and -independent mechanisms of RAF and ERK activation make SHOC2 dispensable for proliferation in 2D, KRAS mutant cells preferentially rely on SHOC2 for ERK signaling under anchorage-independent conditions. Our study highlights a context-dependent contribution of SHOC2 to ERK pathway dynamics that is preferentially engaged by KRAS oncogenic signaling and provides a biochemical framework for selective ERK pathway inhibition by targeting the SHOC2 holophosphatase.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , raf Kinases/chemistry , raf Kinases/metabolism , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Cell Line, Tumor , Gene Editing , Gene Knockout Techniques , Humans , Phosphorylation , Protein Multimerization , ras Proteins/metabolism
8.
Nat Cell Biol ; 20(12): 1389-1399, 2018 12.
Article in English | MEDLINE | ID: mdl-30420664

ABSTRACT

The linear-ubiquitin chain assembly complex (LUBAC) modulates signalling via various immune receptors. In tumour necrosis factor (TNF) signalling, linear (also known as M1) ubiquitin enables full gene activation and prevents cell death. However, the mechanisms underlying cell death prevention remain ill-defined. Here, we show that LUBAC activity enables TBK1 and IKKε recruitment to and activation at the TNF receptor 1 signalling complex (TNFR1-SC). While exerting only limited effects on TNF-induced gene activation, TBK1 and IKKε are essential to prevent TNF-induced cell death. Mechanistically, TBK1 and IKKε phosphorylate the kinase RIPK1 in the TNFR1-SC, thereby preventing RIPK1-dependent cell death. This activity is essential in vivo, as it prevents TNF-induced lethal shock. Strikingly, NEMO (also known as IKKγ), which mostly, but not exclusively, binds the TNFR1-SC via M1 ubiquitin, mediates the recruitment of the adaptors TANK and NAP1 (also known as AZI2). TANK is constitutively associated with both TBK1 and IKKε, while NAP1 is associated with TBK1. We discovered a previously unrecognized cell death checkpoint that is mediated by TBK1 and IKKε, and uncovered an essential survival function for NEMO, whereby it enables the recruitment and activation of these non-canonical IKKs to prevent TNF-induced cell death.


Subject(s)
I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , A549 Cells , Animals , Cell Death/drug effects , Cells, Cultured , HeLa Cells , Humans , Mice, Knockout , Phosphorylation/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
9.
Cell Rep ; 23(9): 2819-2831.e5, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847809

ABSTRACT

Cancer is mostly incurable when diagnosed at a metastatic stage, making its early detection via blood proteins of immense clinical interest. Proteomic changes in tumor tissue may lead to changes detectable in the protein composition of circulating blood plasma. Using a proteomic workflow combining N-glycosite enrichment and SWATH mass spectrometry, we generate a data resource of 284 blood samples derived from patients with different types of localized-stage carcinomas and from matched controls. We observe whether the changes in the patient's plasma are specific to a particular carcinoma or represent a generic signature of proteins modified uniformly in a common, systemic response to many cancers. A quantitative comparison of the resulting N-glycosite profiles discovers that proteins related to blood platelets are common to several cancers (e.g., THBS1), whereas others are highly cancer-type specific. Available proteomics data, including a SWATH library to study N-glycoproteins, will facilitate follow-up biomarker research into early cancer detection.


Subject(s)
Carcinoma/blood , Carcinoma/pathology , Glycoproteins/blood , Mass Spectrometry/methods , Algorithms , Blood Platelets/metabolism , Carcinoma/genetics , Cohort Studies , Humans , Neoplasm Staging , Oncogenes , Proteome/metabolism , ROC Curve
10.
Mol Cell ; 65(4): 730-742.e5, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28212753

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is known for specifically killing cancer cells, whereas in resistant cancers, TRAIL/TRAIL-R can promote metastasis via Rac1 and PI3K. It remains unknown, however, whether and to what extent TRAIL/TRAIL-R signaling in cancer cells can affect the immune microenvironment. Here we show that TRAIL-triggered cytokine secretion from TRAIL-resistant cancer cells is FADD dependent and identify the TRAIL-induced secretome to drive monocyte polarization to myeloid-derived suppressor cells (MDSCs) and M2-like macrophages. TRAIL-R suppression in tumor cells impaired CCL2 production and diminished both lung MDSC presence and tumor growth. In accordance, the receptor of CCL2, CCR2, is required to facilitate increased MDSC presence and tumor growth. Finally, TRAIL and CCL2 are co-regulated with MDSC/M2 markers in lung adenocarcinoma patients. Collectively, endogenous TRAIL/TRAIL-R-mediated CCL2 secretion promotes accumulation of tumor-supportive immune cells in the cancer microenvironment, thereby revealing a tumor-supportive immune-modulatory role of the TRAIL/TRAIL-R system in cancer biology.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cytokines/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Receptors, CCR2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Microenvironment , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 8/genetics , Caspase 8/metabolism , Cell Proliferation , Chemokine CCL2/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Mice, Inbred C57BL , Mice, SCID , Phenotype , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Burden
11.
Sci Rep ; 7: 39985, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28059163

ABSTRACT

To identify novel effectors and processes regulated by PI3K pathway activation, we performed an unbiased phosphoproteomic screen comparing two common events of PI3K deregulation in cancer: oncogenic Pik3ca mutation (Pik3caH1047R) and deletion of Pten. Using mouse embryonic fibroblast (MEF) models that generate inducible, low-level pathway activation as observed in cancer, we quantified 7566 unique phosphopeptides from 3279 proteins. A number of proteins were found to be differentially-regulated by Pik3caH1047R and Pten loss, suggesting unique roles for these two events in processes such as vesicular trafficking, DNA damage repair and RNA splicing. We also identified novel PI3K effectors that were commonly-regulated, including putative AKT substrates. Validation of one of these hits, confirmed NT5C (5',3'-Nucleotidase, Cytosolic) as a novel AKT substrate, with an unexpected role in actin cytoskeleton regulation via an interaction with the ARP2/3 complex. This study has produced a comprehensive data resource and identified a new link between PI3K pathway activation and actin regulation.


Subject(s)
5'-Nucleotidase/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/analysis , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Gene Deletion , Gene Expression Regulation , Mice , Mouse Embryonic Stem Cells , Mutation , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
12.
Cell Rep ; 16(9): 2271-80, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545878

ABSTRACT

Recruitment of the deubiquitinase CYLD to signaling complexes is mediated by its interaction with HOIP, the catalytically active component of the linear ubiquitin chain assembly complex (LUBAC). Here, we identify SPATA2 as a constitutive direct binding partner of HOIP that bridges the interaction between CYLD and HOIP. SPATA2 recruitment to TNFR1- and NOD2-signaling complexes is dependent on HOIP, and loss of SPATA2 abolishes CYLD recruitment. Deficiency in SPATA2 exerts limited effects on gene activation pathways but diminishes necroptosis induced by tumor necrosis factor (TNF), resembling loss of CYLD. In summary, we describe SPATA2 as a previously unrecognized factor in LUBAC-dependent signaling pathways that serves as an adaptor between HOIP and CYLD, thereby enabling recruitment of CYLD to signaling complexes.


Subject(s)
Macrophages/metabolism , Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Binding Sites , Cloning, Molecular , Deubiquitinating Enzyme CYLD , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Macrophages/cytology , Macrophages/drug effects , Mice , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Plasmids , Primary Cell Culture , Protein Binding , Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
13.
Cell Rep ; 13(10): 2258-72, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26670046

ABSTRACT

Ubiquitination and deubiquitination are crucial for assembly and disassembly of signaling complexes. LUBAC-generated linear (M1) ubiquitin is important for signaling via various immune receptors. We show here that the deubiquitinases CYLD and A20, but not OTULIN, are recruited to the TNFR1- and NOD2-associated signaling complexes (TNF-RSC and NOD2-SC), at which they cooperate to limit gene activation. Whereas CYLD recruitment depends on its interaction with LUBAC, but not on LUBAC's M1-chain-forming capacity, A20 recruitment requires this activity. Intriguingly, CYLD and A20 exert opposing effects on M1 chain stability in the TNF-RSC and NOD2-SC. While CYLD cleaves M1 chains, and thereby sensitizes cells to TNF-induced death, A20 binding to them prevents their removal and, consequently, inhibits cell death. Thus, CYLD and A20 cooperatively restrict gene activation and regulate cell death via their respective activities on M1 chains. Hence, the interplay between LUBAC, M1-ubiquitin, CYLD, and A20 is central for physiological signaling through innate immune receptors.


Subject(s)
Cell Death/physiology , Transcriptional Activation/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Cell Line , DNA-Binding Proteins/metabolism , Deubiquitinating Enzyme CYLD , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction/physiology , Transduction, Genetic , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Suppressor Proteins/metabolism
14.
EMBO Mol Med ; 7(9): 1153-65, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26253080

ABSTRACT

The current management of colorectal cancer (CRC) would greatly benefit from non-invasive prognostic biomarkers indicative of clinicopathological tumor characteristics. Here, we employed targeted proteomic profiling of 80 glycoprotein biomarker candidates across plasma samples of a well-annotated patient cohort with comprehensive CRC characteristics. Clinical data included 8-year overall survival, tumor staging, histological grading, regional localization, and molecular tumor characteristics. The acquired quantitative proteomic dataset was subjected to the development of biomarker signatures predicting prognostic clinical endpoints. Protein candidates were selected into the signatures based on significance testing and a stepwise protein selection, each within 10-fold cross-validation. A six-protein biomarker signature of patient outcome could predict survival beyond clinical stage and was able to stratify patients into groups of better and worse prognosis. We further evaluated the performance of the signature on the mRNA level and assessed its prognostic value in the context of previously published transcriptional signatures. Additional signatures predicting regional tumor localization and disease dissemination were also identified. The integration of rich clinical data, quantitative proteomic technologies, and tailored computational modeling facilitated the characterization of these signatures in patient circulation. These findings highlight the value of a simultaneous assessment of important prognostic disease characteristics within a single measurement.


Subject(s)
Biomarkers, Tumor/blood , Clinical Laboratory Techniques/methods , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Plasma/chemistry , Proteomics/methods , Humans , Prognosis
15.
EMBO Mol Med ; 7(9): 1166-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26253081

ABSTRACT

Non-invasive detection of colorectal cancer with blood-based markers is a critical clinical need. Here we describe a phased mass spectrometry-based approach for the discovery, screening, and validation of circulating protein biomarkers with diagnostic value. Initially, we profiled human primary tumor tissue epithelia and characterized about 300 secreted and cell surface candidate glycoproteins. These candidates were then screened in patient systemic circulation to identify detectable candidates in blood plasma. An 88-plex targeting method was established to systematically monitor these proteins in two large and independent cohorts of plasma samples, which generated quantitative clinical datasets at an unprecedented scale. The data were deployed to develop and evaluate a five-protein biomarker signature for colorectal cancer detection.


Subject(s)
Biomarkers, Tumor/blood , Clinical Laboratory Techniques/methods , Colorectal Neoplasms/diagnosis , Mass Spectrometry/methods , Plasma/chemistry , Humans
16.
Nat Protoc ; 8(8): 1602-19, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23887179

ABSTRACT

Targeted proteomics based on selected reaction monitoring (SRM) mass spectrometry is commonly used for accurate and reproducible quantification of protein analytes in complex biological mixtures. Strictly hypothesis-driven, SRM assays quantify each targeted protein by collecting measurements on its peptide fragment ions, called transitions. To achieve sensitive and accurate quantitative results, experimental design and data analysis must consistently account for the variability of the quantified transitions. This consistency is especially important in large experiments, which increasingly require profiling up to hundreds of proteins over hundreds of samples. Here we describe a robust and automated workflow for the analysis of large quantitative SRM data sets that integrates data processing, statistical protein identification and quantification, and dissemination of the results. The integrated workflow combines three software tools: mProphet for peptide identification via probabilistic scoring; SRMstats for protein significance analysis with linear mixed-effect models; and PASSEL, a public repository for storage, retrieval and query of SRM data. The input requirements for the protocol are files with SRM traces in mzXML format, and a file with a list of transitions in a text tab-separated format. The protocol is especially suited for data with heavy isotope-labeled peptide internal standards. We demonstrate the protocol on a clinical data set in which the abundances of 35 biomarker candidates were profiled in 83 blood plasma samples of subjects with ovarian cancer or benign ovarian tumors. The time frame to realize the protocol is 1-2 weeks, depending on the number of replicates used in the experiment.


Subject(s)
Mass Spectrometry/methods , Proteomics/methods , Biomarkers/blood , Data Interpretation, Statistical , Electronic Data Processing/methods , Female , Humans , Ovarian Neoplasms/blood , Plasma/metabolism , Software
17.
Mol Cell Proteomics ; 12(4): 1005-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23408683

ABSTRACT

Protein biomarkers have the potential to transform medicine as they are clinically used to diagnose diseases, stratify patients, and follow disease states. Even though a large number of potential biomarkers have been proposed over the past few years, almost none of them have been implemented so far in the clinic. One of the reasons for this limited success is the lack of technologies to validate proposed biomarker candidates in larger patient cohorts. This limitation could be alleviated by the use of antibody-independent validation methods such as selected reaction monitoring (SRM). Similar to measurements based on affinity reagents, SRM-based targeted mass spectrometry also requires the generation of definitive assays for each targeted analyte. Here, we present a library of SRM assays for 5568 N-glycosites enabling the multiplexed evaluation of clinically relevant N-glycoproteins as biomarker candidates. We demonstrate that this resource can be utilized to select SRM assay sets for cancer-associated N-glycoproteins for their subsequent multiplexed and consistent quantification in 120 human plasma samples. We show that N-glycoproteins spanning 5 orders of magnitude in abundance can be quantified and that previously reported abundance differences in various cancer types can be recapitulated. Together, the established N-glycoprotein SRMAtlas resource facilitates parallel, efficient, consistent, and sensitive evaluation of proposed biomarker candidates in large clinical sample cohorts.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/blood , Glycoproteins/blood , Neoplasm Proteins/blood , Neoplasms/blood , Animals , Antigens, Tumor-Associated, Carbohydrate/chemistry , Case-Control Studies , Glycoproteins/chemistry , Humans , Mice , Molecular Sequence Annotation , Neoplasm Proteins/chemistry , Peptide Library , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry/methods
18.
Proteomics ; 13(8): 1247-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23322582

ABSTRACT

SWATH-MS is a data-independent acquisition method that generates, in a single measurement, a complete recording of the fragment ion spectra of all the analytes in a biological sample for which the precursor ions are within a predetermined m/z versus retention time window. To assess the performance and suitability of SWATH-MS-based protein quantification for clinical use, we compared SWATH-MS and SRM-MS-based quantification of N-linked glycoproteins in human plasma, a commonly used sample for biomarker discovery. Using dilution series of isotopically labeled heavy peptides representing biomarker candidates, the LOQ of SWATH-MS was determined to reach 0.0456 fmol at peptide level by targeted data analysis, which corresponds to a concentration of 5-10 ng protein/mL in plasma, while SRM reached a peptide LOQ of 0.0152 fmol. Moreover, the quantification of endogenous glycoproteins using SWATH-MS showed a high degree of reproducibility, with the mean CV of 14.90%, correlating well with SRM results (R(2) = 0.9784). Overall, SWATH-MS measurements showed a slightly lower sensitivity and a comparable reproducibility to state-of-the-art SRM measurements for targeted quantification of the N-glycosites in human blood. However, a significantly larger number of peptides can be quantified per analysis. We suggest that SWATH-MS analysis combined with N-glycoproteome enrichment in plasma samples is a promising integrative proteomic approach for biomarker discovery and verification.


Subject(s)
Glycoproteins/blood , Mass Spectrometry/methods , Amino Acid Sequence , Biomarkers/blood , Female , Glycoproteins/chemistry , Glycosylation , Humans , Ions , Male , Mass Spectrometry/standards , Molecular Sequence Data , Peptides/chemistry , Proteomics/methods , Reproducibility of Results , Sensitivity and Specificity
19.
J Proteome Res ; 10(1): 5-16, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21142170

ABSTRACT

The development of plasma biomarkers has proven to be more challenging than initially anticipated. Many studies have reported lists of candidate proteins rather than validated candidate markers with an assigned performance to a specific clinical objective. Biomarker research necessitates a clear rational framework with requirements on a multitude of levels. On the technological front, the platform needs to be effective to detect low abundant plasma proteins and be able to measure them in a high throughput manner over a large amount of samples reproducibly. At a conceptual level, the choice of the technological platform and available samples should be part of an overall clinical study design that depends on a joint effort between basic and clinical research. Solutions to these needs are likely to facilitate more feasible studies. Targeted proteomic workflows based on SRM mass spectrometry show the potential of fast verification of biomarker candidates in plasma and thereby closing the gap between discovery and validation in the biomarker development pipeline. Biological samples need to be carefully chosen based on well-established guidelines either for candidate discovery in the form of disease models with optimal fidelity to human disease or for candidate evaluation as well-designed and annotated clinical cohort groups. Most importantly, they should be representative of the target population and directly address the investigated clinical question. A conceptual structure of a biomarker study can be provided in the form of several sequential phases, each having clear objectives and predefined goals. Furthermore, guidelines for reporting the outcome of biomarker studies are critical to adequately assess the quality of the research, interpretation and generalization of the results. By being attentive to and applying these considerations, biomarker research should become more efficient and lead to directly translatable biomarker candidates into clinical evaluation.


Subject(s)
Biomarkers/blood , Blood Proteins/analysis , Diagnostic Techniques and Procedures , Proteomics , Animals , Humans , Mass Spectrometry
20.
J Proteomics ; 73(4): 806-14, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19944197

ABSTRACT

The role of cardiolipin acyl chain composition in assembly/stabilization of mitochondrial complexes was investigated using three yeast deletion mutants (acb1Delta strain; taz1Delta strain; and acb1Deltataz1Delta strain). Deletion of the TAZ1 gene, involved in cardiolipin acyl chain remodeling, is known to increase the content of monolyso-cardiolipin (MLCL) at the expense of CL, and to decrease the unsaturation of the remaining CL. Deletion of the ACB1 gene encoding the acyl-CoA-binding protein, involved in fatty acid elongation, decreases the average length of the CL acyl chains. Furthermore, a TAZ1ACB1 double deletion mutant strain was used in this study which has both a decrease in the length of the CL acyl chains and an increase in MLCL. BN/SDS PAGE analysis revealed that cardiolipin is important for the prohibitin-m-AAA protease complex, the alpha-ketoglutarate dehydrogenase complex and respiratory chain supercomplexes. The results indicate that the decreased level of complexes in taz1Delta and acb1Deltataz1Delta mitochondria is due to a decreased content of CL or the presence of MLCL.


Subject(s)
Cardiolipins/chemistry , Cardiolipins/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/chemistry , Mitochondrial Proteins/metabolism , Repressor Proteins/metabolism , Acylation , Chromatography, Liquid , Diazepam Binding Inhibitor/chemistry , Diazepam Binding Inhibitor/metabolism , Electron Transport/physiology , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutarate Dehydrogenase Complex/genetics , Mass Spectrometry , Metalloendopeptidases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Prohibitins , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...