Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Proteomics ; 17(3-4)2017 02.
Article in English | MEDLINE | ID: mdl-27682984

ABSTRACT

Genomic instability is a critical driver in the process of cancer formation. At the same time, inducing DNA damage by irradiation or genotoxic compounds constitutes a key therapeutic strategy to kill fast-dividing cancer cells. Sensing of DNA lesions initiates a complex set of signalling pathways, collectively known as the DNA damage response (DDR). Deciphering DDR signalling pathways with high-throughput technologies could provide insights into oncogenic transformation, metastasis formation and therapy responses, and could build a basis for better therapeutic interventions in cancer treatment. Mass spectrometry (MS)-based proteomics emerged as a method of choice for global studies of proteins and their posttranslational modifications (PTMs). MS-based studies of the DDR have aided in delineating DNA damage-induced signalling responses. Those studies identified changes in abundance, interactions and modification of proteins in the context of genotoxic stress. Here we review ground-breaking MS-based proteomics studies, which analysed changes in protein abundance, protein-protein and protein-DNA interactions, phosphorylation, acetylation, ubiquitylation, SUMOylation and Poly(ADP-ribose)ylation (PARylation) in the DDR. Finally, we provide an outlook on how proteomics studies of the DDR could aid clinical developments on multiple levels.


Subject(s)
DNA Damage , DNA Repair , Protein Processing, Post-Translational , Proteomics/methods , Genomic Instability , Humans , Mass Spectrometry/methods , Neoplasms/genetics , Neoplasms/therapy , Phosphorylation
2.
Expert Rev Proteomics ; 12(5): 469-87, 2015.
Article in English | MEDLINE | ID: mdl-26400465

ABSTRACT

Site-specific phosphorylation is a fast and reversible covalent post-translational modification that is tightly regulated in cells. The cellular machinery of enzymes that write, erase and read these modifications (kinases, phosphatases and phospho-binding proteins) is frequently deregulated in different diseases, including cancer. Large-scale studies of phosphoproteins - termed phosphoproteomics - strongly rely on the use of high-performance mass spectrometric instrumentation. This powerful technology has been applied to study a great number of phosphorylation-based phenotypes. Nevertheless, many technical and biological challenges have to be overcome to identify biologically relevant phosphorylation sites in cells and tissues. This review describes different technological strategies to identify and quantify phosphorylation sites with high accuracy, without significant loss of analysis speed and reproducibility in tissues and cells. Moreover, computational tools for analysis, integration and biological interpretation of phosphorylation events are discussed.


Subject(s)
Mass Spectrometry/methods , Protein Processing, Post-Translational , Proteome/metabolism , Proteomics/methods , Animals , Humans , Organ Specificity , Phosphorylation
3.
Arch Toxicol ; 87(9): 1635-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23943208

ABSTRACT

Genotoxic perturbation holds a central place in cancer formation and aging, but also is key to cancer therapy by irradiation or chemotherapeutic drugs. Sensing of DNA lesions initiates a highly complex DNA damage response (DDR). This response involves signaling cascades that activate appropriate damage repair pathways, arrest the cell cycle, and ultimately determine cell survival or death. The DDR must be integrated with ongoing signaling and housekeeping processes. With the emergence of high-throughput omics technologies, it has become clear that DNA damage-mediated responses penetrate far deeper than previously appreciated into virtually all cellular signaling pathways. Advances in the last decade have revealed a plethora of early DNA damage-induced changes in posttranslational modifications and subsequent alterations in gene expression profiles, and have provided a glimpse into the assorted rewiring of signal transduction cascades providing biomarkers for chemo- or radiosensitivity. At the same time, genome-wide RNAi screening has provided mechanistic insights into DDR signaling cascades and identified genes involved in mechanisms of cancer resistance to genotoxic therapies. Most recently, distinct omics datasets have been integrated, and sophisticated mathematical models have been applied to the DDR. Here, we review such recent advances that have widened and, in some cases, deepened our knowledge of DDR signaling.


Subject(s)
DNA Damage , Models, Biological , Signal Transduction , Systems Theory , Animals , Computational Biology , Humans , Mutagenesis/drug effects , Mutagenesis/radiation effects , RNA Interference , Signal Transduction/drug effects , Signal Transduction/radiation effects
4.
Brain ; 133(Pt 4): 983-95, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20375133

ABSTRACT

Glioma growth and progression depend on a specialized subpopulation of tumour cells, termed tumour stem cells. Thus, tumour stem cells represent a critical therapeutic target, but the molecular mechanisms that regulate them are poorly understood. Hypoxia plays a key role in tumour progression and in this study we provide evidence that the hypoxic tumour microenvironment also controls tumour stem cells. We define a detailed molecular signature of tumour stem cell genes, which are overexpressed by tumour cells in vascular and perinecrotic/hypoxic niches. Mechanistically, we show that hypoxia plays a key role in the regulation of the tumour stem cell phenotype through hypoxia-inducible factor 2alpha and subsequent induction of specific tumour stem cell signature genes, including mastermind-like protein 3 (Notch pathway), nuclear factor of activated T cells 2 (calcineurin pathway) and aspartate beta-hydroxylase domain-containing protein 2. Notably, a number of these genes belong to pathways regulating the stem cell phenotype. Consistently, tumour stem cell signature genes are overexpressed in newly formed gliomas and are associated with worse clinical prognosis. We propose that tumour stem cells are maintained within a hypoxic niche, providing a functional link between the well-established role of hypoxia in stem cell and tumour biology. The identification of molecular regulators of tumour stem cells in the hypoxic niche points to specific signalling mechanisms that may be used to target the glioblastoma stem cell population.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Hypoxia/physiology , Cell Line, Tumor , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Humans
5.
Nat Commun ; 9(1): 1045, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29535314

ABSTRACT

Comprehensive mass spectrometry (MS)-based proteomics is now feasible, but reproducible quantification remains challenging, especially for post-translational modifications such as phosphorylation. Here, we compare the most popular quantification techniques for global phosphoproteomics: label-free quantification (LFQ), stable isotope labeling by amino acids in cell culture (SILAC) and MS2- and MS3-measured tandem mass tags (TMT). In a mixed species comparison with fixed phosphopeptide ratios, we find LFQ and SILAC to be the most accurate techniques. MS2-based TMT yields the highest precision but lowest accuracy due to ratio compression, which MS3-based TMT can partly rescue. However, MS2-based TMT outperforms MS3-based TMT when analyzing phosphoproteome changes in the DNA damage response, since its higher precision and larger identification numbers allow detection of a greater number of significantly regulated phosphopeptides. Finally, we utilize the TMT multiplexing capabilities to develop an algorithm for determining phosphorylation site stoichiometry, showing that such applications benefit from the high accuracy of MS3-based TMT.


Subject(s)
Phosphopeptides/analysis , Proteomics/methods , Algorithms , Isotope Labeling , Mass Spectrometry/methods , Phosphorylation , Protein Processing, Post-Translational , Tandem Mass Spectrometry
6.
Cell Rep ; 21(2): 546-558, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020638

ABSTRACT

The mechanisms that protect eukaryotic DNA during the cumbersome task of replication depend on the precise coordination of several post-translational modification (PTM)-based signaling networks. Phosphorylation is a well-known regulator of the replication stress response, and recently an essential role for SUMOs (small ubiquitin-like modifiers) has also been established. Here, we investigate the global interplay between phosphorylation and SUMOylation in response to replication stress. Using SUMO and phosphoproteomic technologies, we identify thousands of regulated modification sites. We find co-regulation of central DNA damage and replication stress responders, of which the ATR-activating factor TOPBP1 is the most highly regulated. Using pharmacological inhibition of the DNA damage response kinases ATR and ATM, we find that these factors regulate global protein SUMOylation in the protein networks that protect DNA upon replication stress and fork breakage, pointing to integration between phosphorylation and SUMOylation in the cellular systems that protect DNA integrity.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Replication , Proteome/metabolism , Sumoylation , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Stress, Physiological
7.
Mol Cell Biol ; 35(7): 1254-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624349

ABSTRACT

DNA damage response signaling is crucial for genome maintenance in all organisms and is corrupted in cancer. In an RNA interference (RNAi) screen for (de)ubiquitinases and sumoylases modulating the apoptotic response of embryonic stem (ES) cells to DNA damage, we identified the E3 ubiquitin ligase/ISGylase, ariadne homologue 1 (ARIH1). Silencing ARIH1 sensitized ES and cancer cells to genotoxic compounds and ionizing radiation, irrespective of their p53 or caspase-3 status. Expression of wild-type but not ubiquitinase-defective ARIH1 constructs prevented sensitization caused by ARIH1 knockdown. ARIH1 protein abundance increased after DNA damage through attenuation of proteasomal degradation that required ATM signaling. Accumulated ARIH1 associated with 4EHP, and in turn, this competitive inhibitor of the eukaryotic translation initiation factor 4E (eIF4E) underwent increased nondegradative ubiquitination upon DNA damage. Genotoxic stress led to an enrichment of ARIH1 in perinuclear, ribosome-containing regions and triggered 4EHP association with the mRNA 5' cap as well as mRNA translation arrest in an ARIH1-dependent manner. Finally, restoration of DNA damage-induced translation arrest in ARIH1-depleted cells by means of an eIF2 inhibitor was sufficient to reinstate resistance to genotoxic stress. These findings identify ARIH1 as a potent mediator of DNA damage-induced translation arrest that protects stem and cancer cells against genotoxic stress.


Subject(s)
Carrier Proteins/metabolism , DNA Damage , Eukaryotic Initiation Factor-4E/metabolism , RNA Cap-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/genetics , Caspase 3/metabolism , Cell Line , Cell Line, Tumor , Embryonic Stem Cells/metabolism , Humans , Mice , Protein Biosynthesis , RNA Interference , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
8.
PLoS One ; 8(10): e76476, 2013.
Article in English | MEDLINE | ID: mdl-24146875

ABSTRACT

The chemotherapeutic compound, cisplatin causes various kinds of DNA lesions but also triggers other pertubations, such as ER and oxidative stress. We and others have shown that treatment of pluripotent stem cells with cisplatin causes a plethora of transcriptional and post-translational alterations that, to a major extent, point to DNA damage response (DDR) signaling. The orchestrated DDR signaling network is important to arrest the cell cycle and repair the lesions or, in case of damage beyond repair, eliminate affected cells. Failure to properly balance the various aspects of the DDR in stem cells contributes to ageing and cancer. Here, we performed metabolic profiling by mass spectrometry of embryonic stem (ES) cells treated for different time periods with cisplatin. We then integrated metabolomics with transcriptomics analyses and connected cisplatin-regulated metabolites with regulated metabolic enzymes to identify enriched metabolic pathways. These included nucleotide metabolism, urea cycle and arginine and proline metabolism. Silencing of identified proline metabolic and catabolic enzymes indicated that altered proline metabolism serves as an adaptive, rather than a toxic response. A group of enriched metabolic pathways clustered around the metabolite S-adenosylmethionine, which is a hub for methylation and transsulfuration reactions and polyamine metabolism. Enzymes and metabolites with pro- or anti-oxidant functions were also enriched but enhanced levels of reactive oxygen species were not measured in cisplatin-treated ES cells. Lastly, a number of the differentially regulated metabolic enzymes were identified as target genes of the transcription factor p53, pointing to p53-mediated alterations in metabolism in response to genotoxic stress. Altogether, our findings reveal interconnecting metabolic pathways that are responsive to cisplatin and may serve as signaling modules in the DDR in pluripotent stem cells.


Subject(s)
Cisplatin/pharmacology , Metabolic Networks and Pathways/drug effects , Pluripotent Stem Cells/metabolism , Animals , Arginine/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Metabolome/drug effects , Metabolomics , Mice , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/enzymology , Proline/metabolism , Purines/metabolism , Pyrimidines/metabolism , Signal Transduction/drug effects , Transcriptome/drug effects , Transcriptome/genetics
9.
Sci Signal ; 6(259): ra5, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23354688

ABSTRACT

In pluripotent stem cells, DNA damage triggers loss of pluripotency and apoptosis as a safeguard to exclude damaged DNA from the lineage. An intricate DNA damage response (DDR) signaling network ensures that the response is proportional to the severity of the damage. We combined an RNA interference screen targeting all kinases, phosphatases, and transcription factors with global transcriptomics and phosphoproteomics to map the DDR in mouse embryonic stem cells treated with the DNA cross-linker cisplatin. Networks derived from canonical pathways shared in all three data sets were implicated in DNA damage repair, cell cycle and survival, and differentiation. Experimental probing of these networks identified a mode of DNA damage-induced Wnt signaling that limited apoptosis. Silencing or deleting the p53 gene demonstrated that genotoxic stress elicited Wnt signaling in a p53-independent manner. Instead, this response occurred through reduced abundance of Csnk1a1 (CK1α), a kinase that inhibits ß-catenin. Together, our findings reveal a balance between p53-mediated elimination of stem cells (through loss of pluripotency and apoptosis) and Wnt signaling that attenuates this response to tune the outcome of the DDR.


Subject(s)
Casein Kinase I/metabolism , DNA Damage , Embryonic Stem Cells/enzymology , Pluripotent Stem Cells/enzymology , Systems Biology , Wnt Signaling Pathway , Animals , Apoptosis/genetics , Casein Kinase I/genetics , Cell Line , Embryonic Stem Cells/cytology , Mice , Pluripotent Stem Cells/cytology , RNA Interference , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Mol Cell Biol ; 31(24): 4964-77, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22006019

ABSTRACT

Cellular responses to DNA-damaging agents involve the activation of various DNA damage signaling and transduction pathways. Using quantitative and high-resolution tandem mass spectrometry, we determined global changes in protein level and phosphorylation site profiles following treatment of SILAC (stable isotope labeling by amino acids in cell culture)-labeled murine embryonic stem cells with the anticancer drug cisplatin. Network and pathway analyses indicated that processes related to the DNA damage response and cytoskeleton organization were significantly affected. Although the ATM (ataxia telangiectasia mutated) and ATR (ATM and Rad3-related) consensus sequence (S/T-Q motif) was significantly overrepresented among hyperphosphorylated peptides, about half of the >2-fold-upregulated phosphorylation sites based on the consensus sequence were not direct substrates of ATM and ATR. Eleven protein kinases mainly belonging to the mitogen-activated protein kinase (MAPK) family were identified as being regulated in their kinase domain activation loop. The biological importance of three of these kinases (cyclin-dependent kinase 7 [CDK7], Plk1, and KPCD1) in the protection against cisplatin-induced cytotoxicity was demonstrated by small interfering RNA (siRNA)-mediated knockdown. Our results indicate that the cellular response to cisplatin involves a variety of kinases and phosphatases not only acting in the nucleus but also regulating cytoplasmic targets, resulting in extensive cytoskeletal rearrangements. Integration of transcriptomic and proteomic data revealed a poor correlation between changes in the relative levels of transcripts and their corresponding proteins, but a large overlap in affected pathways at the levels of mRNA, protein, and phosphoprotein. This study provides an integrated view of pathways activated by genotoxic stress and deciphers kinases that play a pivotal role in regulating cellular processes other than the DNA damage response.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Damage , Gene Expression Profiling/methods , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Embryonic Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteomics/methods , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL