Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2018, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440627

ABSTRACT

The ubiquitin ligase NEDD4 promotes neural crest cell (NCC) survival and stem-cell like properties to regulate craniofacial and peripheral nervous system development. However, how ubiquitination and NEDD4 control NCC development remains unknown. Here we combine quantitative analysis of the proteome, transcriptome and ubiquitinome to identify key developmental signalling pathways that are regulated by NEDD4. We report 276 NEDD4 targets in NCCs and show that loss of NEDD4 leads to a pronounced global reduction in specific ubiquitin lysine linkages. We further show that NEDD4 contributes to the regulation of the NCC actin cytoskeleton by controlling ubiquitination and turnover of Profilin 1 to modulate filamentous actin polymerization. Taken together, our data provide insights into how NEDD4-mediated ubiquitination coordinates key regulatory processes during NCC development.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Neural Crest , Actins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Neural Crest/metabolism , Profilins/genetics , Profilins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
Arch Biochem Biophys ; 687: 108386, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32360748

ABSTRACT

Growth factor receptor bound protein 7 (Grb7) is a mammalian adaptor protein participating in signaling pathways implicated in cell migration, metastatic invasion, cell proliferation and tumor-associated angiogenesis. We expressed tagged versions of wild type Grb7 and the mutant Grb7Δ, lacking its calmodulin-binding domain (CaM-BD), in human embryonic kidney (HEK) 293 cells and rat glioma C6 cells to identify novel binding partners using shot-gun proteomics. Among the new identified proteins, we validated the ubiquitin-ligase Nedd4 (neural precursor cell expressed developmentally down-regulated protein 4), the heat-shock protein Hsc70/HSPA8 (heat shock cognate protein 70) and the cell cycle regulatory protein caprin-1 (cytoplasmic activation/proliferation-associated protein 1) in rat glioma C6 cells. Our results suggest a role of Grb7 in pathways where these proteins are implicated. These include protein trafficking and degradation, stress-response, chaperone-mediated autophagy, apoptosis and cell proliferation.


Subject(s)
Cell Cycle Proteins/metabolism , GRB7 Adaptor Protein/metabolism , HSC70 Heat-Shock Proteins/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Animals , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Line, Tumor , GRB7 Adaptor Protein/genetics , HEK293 Cells , Humans , Mutation , Protein Binding , Protein Domains/genetics , Protein Structure, Secondary , Proteomics , Rats
3.
Immunol Cell Biol ; 94(10): 994-999, 2016 11.
Article in English | MEDLINE | ID: mdl-27377765

ABSTRACT

Mevalonate kinase deficiency (MKD) is caused by mutations in a key enzyme of the mevalonate-cholesterol biosynthesis pathway, leading to recurrent autoinflammatory disease characterised by enhanced release of interleukin-1ß (IL-1ß). It is currently believed that the inflammatory phenotype of MKD is triggered by temperature-sensitive loss of mevalonate kinase activity and reduced biosynthesis of isoprenoid lipids required for the prenylation of small GTPase proteins. However, previous studies have not clearly shown any change in protein prenylation in patient cells under normal conditions. With lymphoblast cell lines from two compound heterozygous MKD patients, we used a highly sensitive in vitro prenylation assay, together with quantitative mass spectrometry, to reveal a subtle accumulation of unprenylated Rab GTPases in cells cultured for 3 days or more at 40 °C compared with 37 °C. This included a 200% increase in unprenylated Rab7A, Rab14 and Rab1A. Inhibition of sterol regulatory element-binding protein (SREBP) activation by fatostatin led to more pronounced accumulation of unprenylated Rab proteins in MKD cells but not parent cells, suggesting that cultured MKD cells may partially overcome the loss of isoprenoid lipids by SREBP-mediated upregulation of enzymes required for isoprenoid biosynthesis. Furthermore, while inhibition of Rho/Rac/Rap prenylation promoted the release of IL-1ß, specific inhibition of Rab prenylation by NE10790 had no effect in human peripheral blood mononuclear cells or human THP-1 monocytic cells. These studies demonstrate for the first time that mutations in mevalonate kinase can lead to a mild, temperature-induced defect in the prenylation of small GTPases, but that loss of prenylated Rab GTPases is not the cause of enhanced IL-1ß release in MKD.


Subject(s)
Mevalonate Kinase Deficiency/enzymology , Protein Prenylation , rab GTP-Binding Proteins/metabolism , Cell Line , Child , Child, Preschool , Female , Humans , Interleukin-1beta/metabolism , Isotope Labeling , Leukocytes, Mononuclear/metabolism , Male , Mevalonate Kinase Deficiency/pathology , Pyridines/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Temperature , Thiazoles/pharmacology
4.
Small GTPases ; 6(4): 202-11, 2015 10 02.
Article in English | MEDLINE | ID: mdl-26399387

ABSTRACT

Bisphosphonate drugs such as zoledronic acid (ZOL), used for the treatment of common bone disorders, target the skeleton and inhibit bone resorption by preventing the prenylation of small GTPases in bone-destroying osteoclasts. Increasing evidence indicates that bisphosphonates also have pleiotropic effects outside the skeleton, most likely via cells of the monocyte/macrophage lineage exposed to nanomolar circulating drug concentrations. However, no effects of such low concentrations of ZOL have been reported using existing approaches. We have optimized a highly sensitive in vitro prenylation assay utilizing recombinant geranylgeranyltransferases to enable the detection of subtle effects of ZOL on the prenylation of Rab- and Rho-family GTPases. Using this assay, we found for the first time that concentrations of ZOL as low as 10nM caused inhibition of Rab prenylation in J774 macrophages following prolonged cell culture. By combining the assay with quantitative mass spectrometry we identified an accumulation of 18 different unprenylated Rab proteins in J774 cells after nanomolar ZOL treatment, with a >7-fold increase in the unprenylated form of Rab proteins associated with the endophagosome pathway (Rab1, Rab5, Rab6, Rab7, Rab11, Rab14 and Rab21). Finally, we also detected a clear effect of subcutaneous ZOL administration in vivo on the prenylation of Rab1A, Rab5B, Rab7A and Rab14 in mouse peritoneal macrophages, confirming that systemic treatment with bisphosphonate drug can inhibit prenylation in myeloid cells in vivo outside the skeleton. These observations begin a new era in defining the precise pharmacological actions of bisphosphonate drugs on the prenylation of small GTPases in vivo.


Subject(s)
Diphosphonates/pharmacology , Imidazoles/pharmacology , Macrophages, Peritoneal/metabolism , Protein Prenylation/drug effects , rab GTP-Binding Proteins/metabolism , Animals , Cell Line , Mice , Zoledronic Acid
5.
Mol Cell Proteomics ; 14(8): 2194-212, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26055452

ABSTRACT

Entry into mitosis is driven by the coordinated phosphorylation of thousands of proteins. For the cell to complete mitosis and divide into two identical daughter cells it must regulate dephosphorylation of these proteins in a highly ordered, temporal manner. There is currently a lack of a complete understanding of the phosphorylation changes that occur during the initial stages of mitotic exit in human cells. Therefore, we performed a large unbiased, global analysis to map the very first dephosphorylation events that occur as cells exit mitosis. We identified and quantified the modification of >16,000 phosphosites on >3300 unique proteins during early mitotic exit, providing up to eightfold greater resolution than previous studies. The data have been deposited to the ProteomeXchange with identifier PXD001559. Only a small fraction (∼ 10%) of phosphorylation sites were dephosphorylated during early mitotic exit and these occurred on proteins involved in critical early exit events, including organization of the mitotic spindle, the spindle assembly checkpoint, and reformation of the nuclear envelope. Surprisingly this enrichment was observed across all kinase consensus motifs, indicating that it is independent of the upstream phosphorylating kinase. Therefore, dephosphorylation of these sites is likely determined by the specificity of phosphatase/s rather than the activity of kinase/s. Dephosphorylation was significantly affected by the amino acids at and surrounding the phosphorylation site, with several unique evolutionarily conserved amino acids correlating strongly with phosphorylation status. These data provide a potential mechanism for the specificity of phosphatases, and how they co-ordinate the ordered events of mitotic exit. In summary, our results provide a global overview of the phosphorylation changes that occur during the very first stages of mitotic exit, providing novel mechanistic insight into how phosphatase/s specifically regulate this critical transition.


Subject(s)
Mitosis , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proteomics/methods , Amino Acid Motifs , Amino Acid Sequence , Amino Acids/metabolism , Anaphase , Conserved Sequence , Evolution, Molecular , HeLa Cells , Humans , Metaphase , Models, Biological , Molecular Sequence Data , Phosphopeptides/metabolism , Phosphorylation , Protein Kinases/metabolism , Reproducibility of Results , Substrate Specificity
6.
Sci Signal ; 8(364): ra18, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25690013

ABSTRACT

Factors secreted by tumor cells shape the local microenvironment to promote invasion and metastasis, as well as condition the premetastatic niche to enable secondary-site colonization and growth. In addition to this secretome, tumor cells have increased abundance of growth-promoting receptors at the cell surface. We found that the tyrosine phosphatase PTPN14 (also called Pez, which is mutated in various cancers) suppressed metastasis by reducing intracellular protein trafficking through the secretory pathway. Knocking down PTPN14 in tumor cells or injecting the peritoneum of mice with conditioned medium from PTPN14-deficient cell cultures promoted the growth and metastasis of breast cancer xenografts. Loss of catalytically functional PTPN14 increased the secretion of growth factors and cytokines, such as IL-8 (interleukin-8), and increased the abundance of EGFR (epidermal growth factor receptor) at the cell surface of breast cancer cells and of FLT4 (vascular endothelial growth factor receptor 3) at the cell surface of primary lymphatic endothelial cells. We identified RIN1 (Ras and Rab interactor 1) and PRKCD (protein kinase C-δ) as binding partners and substrates of PTPN14. Similar to cells overexpressing PTPN14, receptor trafficking to the cell surface was inhibited in cells that lacked PRKCD or RIN1 or expressed a nonphosphorylatable RIN1 mutant, and cytokine secretion was decreased in cells treated with PRKCD inhibitors. Invasive breast cancer tissue had decreased expression of PTPN14, and patient survival was worse when tumors had increased expression of the genes encoding RIN1 or PRKCD. Thus, PTPN14 prevents metastasis by restricting the trafficking of both soluble and membrane-bound proteins.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Metastasis/physiopathology , Protein Transport/physiology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Tumor Microenvironment/physiology , Animals , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Heterografts/metabolism , Heterografts/physiopathology , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Isotope Labeling , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/prevention & control , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/pharmacology , Tandem Mass Spectrometry , rab GTP-Binding Proteins/metabolism
7.
Cancer Discov ; 5(1): 35-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25312016

ABSTRACT

UNLABELLED: Recent clinical trials have shown that bisphosphonate drugs improve breast cancer patient survival independent of their antiresorptive effects on the skeleton. However, because bisphosphonates bind rapidly to bone mineral, the exact mechanisms of their antitumor action, particularly on cells outside of bone, remain unknown. Here, we used real-time intravital two-photon microscopy to show extensive leakage of fluorescent bisphosphonate from the vasculature in 4T1 mouse mammary tumors, where it initially binds to areas of small, granular microcalcifications that are engulfed by tumor-associated macrophages (TAM), but not tumor cells. Importantly, we also observed uptake of radiolabeled bisphosphonate in the primary breast tumor of a patient and showed the resected tumor to be infiltrated with TAMs and to contain similar granular microcalcifications. These data represent the first compelling in vivo evidence that bisphosphonates can target cells in tumors outside the skeleton and that their antitumor activity is likely to be mediated via TAMs. SIGNIFICANCE: Bisphosphonates are assumed to act solely in bone. However, mouse models and clinical trials show that they have surprising antitumor effects outside bone. We provide unequivocal evidence that bisphosphonates target TAMs, but not tumor cells, to exert their extraskeletal effects, offering a rationale for use in patients with early disease.


Subject(s)
Bone Density Conservation Agents/metabolism , Diphosphonates/metabolism , Macrophages/metabolism , Neoplasms/diagnosis , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Animals , Bone Density Conservation Agents/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Calcinosis , Carbocyanines , Diphosphonates/therapeutic use , Disease Models, Animal , Female , Humans , Macrophages/drug effects , Macrophages/immunology , Mice , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasms/drug therapy , Phagocytosis/immunology , Xenograft Model Antitumor Assays
8.
Breast Cancer Res ; 16(5): 437, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25200860

ABSTRACT

INTRODUCTION: Although aberrant tyrosine kinase signalling characterises particular breast cancer subtypes, a global analysis of tyrosine phosphorylation in mouse models of breast cancer has not been undertaken to date. This may identify conserved oncogenic pathways and potential therapeutic targets. METHODS: We applied an immunoaffinity/mass spectrometry workflow to three mouse models: murine stem cell virus-Neu, expressing truncated Neu, the rat orthologue of human epidermal growth factor receptor 2, Her2 (HER2); mouse mammary tumour virus-polyoma virus middle T antigen (PyMT); and the p53-/- transplant model (p53). Pathways and protein-protein interaction networks were identified by bioinformatics analysis. Molecular mechanisms underpinning differences in tyrosine phosphorylation were characterised by Western blot analysis and array comparative genomic hybridisation. The functional role of mesenchymal-epithelial transition factor (Met) in a subset of p53-null tumours was interrogated using a selective tyrosine kinase inhibitor (TKI), small interfering RNA (siRNA)-mediated knockdown and cell proliferation assays. RESULTS: The three models could be distinguished on the basis of tyrosine phosphorylation signatures and signalling networks. HER2 tumours exhibited a protein-protein interaction network centred on avian erythroblastic leukaemia viral oncogene homologue 2 (Erbb2), epidermal growth factor receptor and platelet-derived growth factor receptor α, and they displayed enhanced tyrosine phosphorylation of ERBB receptor feedback inhibitor 1. In contrast, the PyMT network displayed significant enrichment for components of the phosphatidylinositol 3-kinase signalling pathway, whereas p53 tumours exhibited increased tyrosine phosphorylation of Met and components or regulators of the cytoskeleton and shared signalling network characteristics with basal and claudin-low breast cancer cells. A subset of p53 tumours displayed markedly elevated cellular tyrosine phosphorylation and Met expression, as well as Met gene amplification. Treatment of cultured p53-null cells exhibiting Met amplification with a selective Met TKI abrogated aberrant tyrosine phosphorylation and blocked cell proliferation. The effects on proliferation were recapitulated when Met was knocked down using siRNA. Additional subtypes of p53 tumours exhibited increased tyrosine phosphorylation of other oncogenes, including Peak1/SgK269 and Prex2. CONCLUSION: This study provides network-level insights into signalling in the breast cancer models utilised and demonstrates that comparative phosphoproteomics can identify conserved oncogenic signalling pathways. The Met-amplified, p53-null tumours provide a new preclinical model for a subset of triple-negative breast cancers.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Phosphoproteins/metabolism , Phosphotyrosine/metabolism , Proteome/metabolism , Animals , Female , Gene Dosage , Humans , Indoles/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Oncogenes , Protein Interaction Maps , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Sulfones/pharmacology
9.
FEBS J ; 280(21): 5237-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23876235

ABSTRACT

Acquired resistance to the anti-estrogen tamoxifen remains a significant challenge in breast cancer management. In this study, we used an integrative approach to characterize global protein expression and tyrosine phosphorylation events in tamoxifen-resistant MCF7 breast cancer cells (TamR) compared with parental controls. Quantitative mass spectrometry and computational approaches were combined to identify perturbed signalling networks, and candidate regulatory proteins were functionally interrogated by siRNA-mediated knockdown. Network analysis revealed that cellular metabolism was perturbed in TamR cells, together with pathways enriched for proteins associated with growth factor, cell-cell and cell matrix-initiated signalling. Consistent with known roles for Ras/MAPK and PI3-kinase signalling in tamoxifen resistance, tyrosine-phosphorylated MAPK1, SHC1 and PIK3R2 were elevated in TamR cells. Phosphorylation of the tyrosine kinase Yes and expression of the actin-binding protein myristoylated alanine-rich C-kinase substrate (MARCKS) were increased two- and eightfold in TamR cells respectively, and these proteins were selected for further analysis. Knockdown of either protein in TamR cells had no effect on anti-estrogen sensitivity, but significantly decreased cell motility. MARCKS expression was significantly higher in breast cancer cell lines than normal mammary epithelial cells and in ER-negative versus ER-positive breast cancer cell lines. In primary breast cancers, cytoplasmic MARCKS staining was significantly higher in basal-like and HER2 cancers than in luminal cancers, and was independently predictive of poor survival in multivariate analyses of the whole cohort (P < 0.0001) and in ER-positive patients (P = 0.0005). These findings provide network-level insights into the molecular alterations associated with the tamoxifen-resistant phenotype, and identify MARCKS as a potential biomarker of therapeutic responsiveness that may assist in stratification of patients for optimal therapy.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Drug Resistance, Neoplasm , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Cell Adhesion , Cell Cycle , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Middle Aged , Myristoylated Alanine-Rich C Kinase Substrate , Phosphorylation/drug effects , Protein Interaction Maps , Proteomics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Array Analysis , Tumor Cells, Cultured
10.
J Proteome Res ; 12(7): 3104-16, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23692254

ABSTRACT

Kinase enrichment utilizing broad-spectrum kinase inhibitors enables the identification of large proportions of the expressed kinome by mass spectrometry. However, the existing inhibitors are still inadequate in covering the entire kinome. Here, we identified a novel bisanilino pyrimidine, CTx-0294885, exhibiting inhibitory activity against a broad range of kinases in vitro, and further developed it into a Sepharose-supported kinase capture reagent. Use of a quantitative proteomics approach confirmed the selectivity of CTx-0294885-bound beads for kinase enrichment. Large-scale CTx-0294885-based affinity purification followed by LC-MS/MS led to the identification of 235 protein kinases from MDA-MB-231 cells, including all members of the AKT family that had not been previously detected by other broad-spectrum kinase inhibitors. Addition of CTx-0294885 to a mixture of three kinase inhibitors commonly used for kinase-enrichment increased the number of kinase identifications to 261, representing the largest kinome coverage from a single cell line reported to date. Coupling phosphopeptide enrichment with affinity purification using the four inhibitors enabled the identification of 799 high-confidence phosphosites on 183 kinases, ∼10% of which were localized to the activation loop, and included previously unreported phosphosites on BMP2K, MELK, HIPK2, and PRKDC. Therefore, CTx-0294885 represents a powerful new reagent for analysis of kinome signaling networks that may facilitate development of targeted therapeutic strategies. Proteomics data have been deposited to the ProteomeXchange Consortium ( http://proteomecentral.proteomexchange.org ) via the PRIDE partner repository with the data set identifier PXD000239.


Subject(s)
Phosphotransferases/isolation & purification , Protein Kinase Inhibitors/pharmacology , Proteomics , Pyrimidines/chemistry , ortho-Aminobenzoates/chemistry , Cell Line , Chromatography, Liquid/methods , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Tandem Mass Spectrometry/methods
11.
Cancer Res ; 73(6): 1969-80, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378338

ABSTRACT

Basal breast cancer cells feature high expression of the Src family kinase Lyn that has been implicated in the pathogenicity of this disease. In this study, we identified novel Lyn kinase substrates, the most prominent of which was the atypical kinase SgK269 (PEAK1). In breast cancer cells, SgK269 expression associated with the basal phenotype. In primary breast tumors, SgK269 overexpression was detected in a subset of basal, HER2-positive, and luminal cancers. In immortalized MCF-10A mammary epithelial cells, SgK269 promoted transition to a mesenchymal phenotype and increased cell motility and invasion. Growth of MCF-10A acini in three-dimensional (3D) culture was enhanced upon SgK269 overexpression, which induced an abnormal, multilobular acinar morphology and promoted extracellular signal-regulated kinase (Erk) and Stat3 activation. SgK269 Y635F, mutated at a major Lyn phosphorylation site, did not enhance acinar size or cellular invasion. We show that Y635 represents a Grb2-binding site that promotes both Stat3 and Erk activation in 3D culture. RNA interference-mediated attenuation of SgK269 in basal breast cancer cells promoted acquisition of epithelial characteristics and decreased anchorage-independent growth. Together, our results define a novel signaling pathway in basal breast cancer involving Lyn and SgK269 that offers clinical opportunities for therapeutic intervention.


Subject(s)
Breast Neoplasms/enzymology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , src-Family Kinases/metabolism , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , Female , Humans , Microscopy, Fluorescence , Neoplasm Invasiveness , Phosphorylation , Polymerase Chain Reaction
12.
Proteomics ; 11(16): 3390-401, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21751366

ABSTRACT

The transforming growth factor-ß (TGF-ß) signaling pathway progresses through a series of protein phosphorylation regulated steps. Smad4 is a key mediator of the classical TGF-ß signaling pathway; however, reports suggest that TGF-ß can activate other cellular pathways independent of Smad4. By investigating the TGF-ß-regulated phosphoproteome, we aimed to uncover new functions controlled by TGF-ß. We applied titanium dioxide to enrich phosphopeptides from stable isotope labeling with amino acids in cell culture (SILAC)-labeled SW480 cells stably expressing Smad4 and profiled them by mass spectrometry. TGF-ß stimulation for 30 min resulted in the induction of 17 phosphopeptides and the repression of 8 from a total of 149 unique phosphopeptides. Proteins previously not known to be phosphorylated by TGF-ß including programmed cell death protein 4, nuclear ubiquitous casein and cyclin-dependent kinases substrate, hepatoma-derived growth factor and cell division kinases amongst others were induced following TGF-ß stimulation, while the phosphorylation of TRAF2 and NCK-interacting protein kinase are examples of proteins whose phosphorylation status was repressed. This phosphoproteomic screen has identified new TGF-ß-modulated phosphorylation responses in colon carcinoma cells.


Subject(s)
Colonic Neoplasms/metabolism , Phosphoproteins/metabolism , Proteomics/methods , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Cell Line, Tumor , Colonic Neoplasms/genetics , Germinal Center Kinases , Humans , Isotope Labeling , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/metabolism , Phosphoproteins/analysis , Phosphoproteins/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteome/analysis , Proteome/chemistry , Proteome/metabolism , Smad4 Protein/biosynthesis , Smad4 Protein/genetics , Smad4 Protein/metabolism , TNF Receptor-Associated Factor 2/metabolism , Titanium/chemistry
13.
Proteomics ; 11(4): 535-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21243637

ABSTRACT

In this review we examine techniques, software, and statistical analyses used in label-free quantitative proteomics studies for area under the curve and spectral counting approaches. Recent advances in the field are discussed in an order that reflects a logical workflow design. Examples of studies that follow this design are presented to highlight the requirement for statistical assessment and further experiments to validate results from label-free quantitation. Limitations of label-free approaches are considered, label-free approaches are compared with labelling techniques, and forward-looking applications for label-free quantitative data are presented. We conclude that label-free quantitative proteomics is a reliable, versatile, and cost-effective alternative to labelled quantitation.


Subject(s)
Mass Spectrometry/methods , Peptide Mapping/methods , Proteomics/methods
14.
Mol Biosyst ; 6(11): 2332-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20862427

ABSTRACT

TGF-ß signalling can play a paradoxical cell type specific role in cancer progression. Smad4 is a key mediator of the TGF-ß pathway, and is mutated and/or deleted in many cancers. To investigate Smad4 regulated TGF-ß signalling in colon cancer we conducted an iTRAQ mass spectrometry quantitative screen using wild type SW480 (Smad4 negative) colon carcinoma cells and stably restored Smad4 positive SW480 cells. In cells possessing a restored canonical TGF-ß signalling pathway, 48 h TGF-ß stimulation induced the expression of 15 proteins and repressed 1 protein, while in Smad4 wild type cells, TGF-ß induced 7 proteins and repressed 2 proteins. The expression of several S100 protein family members (A2, A4, A10, and A11), transgelin-2 and AKAP12, amongst others, were shown to be regulated by TGF-ß in a Smad4 dependent manner. We observed that S100 A4 could be repressed by TGF-ß, independently of Smad4 expression, while other Smad4 independent TGF-ß responses were restricted to induction of ribosomes and cytoskeletal proteins. Our proteomic screen has identified new Smad4 dependent and independent TGF-ß responses in colon carcinoma cells.


Subject(s)
Colonic Neoplasms/metabolism , Proteomics/methods , Signal Transduction , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , Immunoblotting , Isotope Labeling , Mass Spectrometry , Models, Biological , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...