Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
2.
J Proteome Res ; 23(6): 2160-2168, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38767394

ABSTRACT

Resistance is a major problem with effective cancer treatment and the stroma forms a significant portion of the tumor mass but traditional drug screens involve cancer cells alone. Cancer-associated fibroblasts (CAFs) are a major tumor stroma component and its secreted proteins may influence the function of cancer cells. The majority of secretome studies compare different cancer or CAF cell lines exclusively. Here, we present the direct characterization of the secreted protein profiles between CAFs and KRAS mutant-cancer cell lines from colorectal, lung, and pancreatic tissues using multiplexed mass spectrometry. 2573 secreted proteins were annotated, and differential analysis highlighted understudied CAF-enriched secreted proteins, including Wnt family member 5B (WNT5B), in addition to established CAF markers, such as collagens. The functional role of CAF secreted proteins was explored by assessing its effect on the response to 97 anticancer drugs since stromal cells may cause a differing cancer drug response, which may be missed on routine drug screening using cancer cells alone. CAF secreted proteins caused specific effects on each of the cancer cell lines, which highlights the complexity and challenges in cancer treatment and so the importance to consider stromal elements.


Subject(s)
Cancer-Associated Fibroblasts , Secretome , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Secretome/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Mass Spectrometry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proteomics/methods , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics
3.
J Proteomics ; 288: 104973, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37481068

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a key component of tumors. We aimed to profile the proteome of cancer cell lines representing three common cancer types (lung, colorectal and pancreatic) and a representative CAF cell line from each tumor type to gain insight into CAF function and novel CAF biomarkers. We used isobaric labeling, liquid chromatography and mass spectrometry to evaluate the proteome of 9 cancer and 3 CAF cell lines. Of the 9460 proteins evaluated, functional enrichment analysis revealed an upregulation of N-glycan biosynthesis and extracellular matrix proteins in CAFs. 85 proteins had 16-fold higher expression in CAFs compared to cancer cells, including previously known CAF markers like fibroblast activation protein (FAP). Novel overexpressed CAF biomarkers included heat shock protein ß-6 (HSPB6/HSP20) and cyclooxygenase 1 (PTGS1/COX1). SiRNA knockdown of the genes encoding these proteins did not reduce contractility in lung CAFs, suggesting they were not crucial to this function. Immunohistochemical analysis of 30 tumor samples (10 lung, 10 colorectal and 10 pancreatic) showed restricted HSPB6 and PTGS1 expression in the stroma. Therefore, we describe an unbiased differential proteome analysis of CAFs compared to cancer cells, which revealed higher expression of HSPB6 and PTGS1 in CAFs. Data are available via ProteomeXchange (PXD040360). SIGNIFICANCE: Cancer-associated fibroblasts (CAFs) are highly abundant stromal cells present in tumors. CAFs are known to influence tumor progression and drug resistance. Characterizing the proteome of CAFs could give potential insights into new stromal drug targets and biomarkers. Mass spectrometry-based analysis comparing proteomic profiles of CAFs and cancers characterized 9460 proteins of which 85 proteins had 16-fold higher expression in CAFs compared to cancer cells. Further interrogation of this rich resource could provide insight into the function of CAFs and could reveal putative stromal targets. We describe for the first time that heat shock protein ß-6 (HSPB6/HSP20) and cyclooxygenase 1 (PTGS1/COX1) are overexpressed in CAFs compared to cancer cells.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Cyclooxygenase 1 , Proteome/metabolism , Proteomics , Cell Line , Biomarkers/analysis , Colorectal Neoplasms/metabolism , Heat-Shock Proteins/metabolism , Fibroblasts/metabolism , Tumor Microenvironment/physiology
4.
Sci Rep ; 12(1): 18633, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36329085

ABSTRACT

By suppressing gene transcription through the recruitment of corepressor proteins, B-cell lymphoma 6 (BCL6) protein controls a transcriptional network required for the formation and maintenance of B-cell germinal centres. As BCL6 deregulation is implicated in the development of Diffuse Large B-Cell Lymphoma, we sought to discover novel small molecule inhibitors that disrupt the BCL6-corepressor protein-protein interaction (PPI). Here we report our hit finding and compound optimisation strategies, which provide insight into the multi-faceted orthogonal approaches that are needed to tackle this challenging PPI with small molecule inhibitors. Using a 1536-well plate fluorescence polarisation high throughput screen we identified multiple hit series, which were followed up by hit confirmation using a thermal shift assay, surface plasmon resonance and ligand-observed NMR. We determined X-ray structures of BCL6 bound to compounds from nine different series, enabling a structure-based drug design approach to improve their weak biochemical potency. We developed a time-resolved fluorescence energy transfer biochemical assay and a nano bioluminescence resonance energy transfer cellular assay to monitor cellular activity during compound optimisation. This workflow led to the discovery of novel inhibitors with respective biochemical and cellular potencies (IC50s) in the sub-micromolar and low micromolar range.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Humans , Crystallography, X-Ray , Proto-Oncogene Proteins c-bcl-6/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Drug Design , Ligands
5.
Mol Cancer Ther ; 21(6): 1020-1029, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35368084

ABSTRACT

We hypothesize that the study of acute protein perturbation in signal transduction by targeted anticancer drugs can predict drug sensitivity of these agents used as single agents and rational combination therapy. We assayed dynamic changes in 52 phosphoproteins caused by an acute exposure (1 hour) to clinically relevant concentrations of seven targeted anticancer drugs in 35 non-small cell lung cancer (NSCLC) cell lines and 16 samples of NSCLC cells isolated from pleural effusions. We studied drug sensitivities across 35 cell lines and synergy of combinations of all drugs in six cell lines (252 combinations). We developed orthogonal machine-learning approaches to predict drug response and rational combination therapy. Our methods predicted the most and least sensitive quartiles of drug sensitivity with an AUC of 0.79 and 0.78, respectively, whereas predictions based on mutations in three genes commonly known to predict response to the drug studied, for example, EGFR, PIK3CA, and KRAS, did not predict sensitivity (AUC of 0.5 across all quartiles). The machine-learning predictions of combinations that were compared with experimentally generated data showed a bias to the highest quartile of Bliss synergy scores (P = 0.0243). We confirmed feasibility of running such assays on 16 patient samples of freshly isolated NSCLC cells from pleural effusions. We have provided proof of concept for novel methods of using acute ex vivo exposure of cancer cells to targeted anticancer drugs to predict response as single agents or combinations. These approaches could complement current approaches using gene mutations/amplifications/rearrangements as biomarkers and demonstrate the utility of proteomics data to inform treatment selection in the clinic.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Pleural Effusion , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Artificial Intelligence , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation
6.
Cell Oncol (Dordr) ; 44(5): 1197-1206, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34462871

ABSTRACT

PURPOSE: We hypothesised that plasticity in signal transduction may be a mechanism of drug resistance and tested this hypothesis in the setting of cetuximab resistance in patients with KRAS/NRAS/BRAFV600 wild-type colorectal cancer (CRC). METHODS: A multiplex antibody-based platform was used to study simultaneous changes in signal transduction of 55 phospho-proteins in 12 KRAS/NRAS/BRAFV600 wild-type CRC cell lines (6 cetuximab sensitive versus 6 cetuximab resistant) following 1 and 4 h in vitro cetuximab exposure. We validated our results in CRC patient samples (n = 4) using ex vivo exposure to cetuximab in KRAS/NRAS/BRAFV600 cells that were immunomagnetically separated from the serous effusions of patients with known cetuximab resistance. RESULTS: Differences in levels of phospho-proteins in cetuximab sensitive and resistant cell lines included reductions in phospho-RPS6 and phospho-PRAS40 in cetuximab sensitive, but not cetuximab resistant cell lines at 1 and 4 h, respectively. In addition, phospho-AKT levels were found to be elevated in 3/4 patient samples following ex vivo incubation with cetuximab for 1 h. We further explored these findings by studying the effects of combinations of cetuximab and two PI3K pathway inhibitors in 3 cetuximab resistant cell lines. The addition of PI3K pathway inhibitors to cetuximab led to a significantly higher reduction in colony formation capacity compared to cetuximab alone. CONCLUSION: Our findings suggest activation of the PI3K pathway as a mechanism of cetuximab resistance in KRAS/NRAS/BRAFV600 wild-type CRC.


Subject(s)
Cetuximab/pharmacology , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Proteomics/methods , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Adult , Aged , Antineoplastic Agents, Immunological/pharmacology , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects
7.
J Med Chem ; 62(5): 2447-2465, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30779566

ABSTRACT

A series of imidazo[1,2- b]pyridazin-8-amine kinase inhibitors were discovered to allosterically inhibit the endoribonuclease function of the dual kinase-endoribonuclease inositol-requiring enzyme 1α (IRE1α), a key component of the unfolded protein response in mammalian cells and a potential drug target in multiple human diseases. Inhibitor optimization gave compounds with high kinome selectivity that prevented endoplasmic reticulum stress-induced IRE1α oligomerization and phosphorylation, and inhibited endoribonuclease activity in human cells. X-ray crystallography showed the inhibitors to bind to a previously unreported and unusually disordered conformation of the IRE1α kinase domain that would be incompatible with back-to-back dimerization of the IRE1α protein and activation of the endoribonuclease function. These findings increase the repertoire of known IRE1α protein conformations and can guide the discovery of highly selective ligands for the IRE1α kinase site that allosterically inhibit the endoribonuclease.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Allosteric Regulation , Biopolymers/metabolism , Crystallography, X-Ray , Dimerization , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/chemistry , HEK293 Cells , Humans , Phosphorylation , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/chemistry
8.
Oncotarget ; 4(10): 1647-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24072592

ABSTRACT

The ribosomal P70 S6 kinases play a crucial role in PI3K/mTOR regulated signalling pathways and are therefore potential targets for the treatment of a variety of diseases including diabetes and cancer. In this study we describe the identification of three series of chemically distinct S6K1 inhibitors. In addition, we report a novel PKA-S6K1 chimeric protein with five mutations in or near its ATP-binding site, which was used to determine the binding mode of two of the three inhibitor series, and provided a robust system to aid the optimisation of the oxadiazole-substituted benzimidazole inhibitor series. We show that the resulting oxadiazole-substituted aza-benzimidazole is a potent and ligand efficient S6 kinase inhibitor, which blocks the phosphorylation of RPS6 at Ser235/236 in TSC negative HCV29 human bladder cancer cells by inhibiting S6 kinase activity and thus provides a useful tool compound to investigate the function of S6 kinases.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Design , High-Throughput Screening Assays/methods , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Models, Molecular , Phosphorylation , Protein Kinase Inhibitors/chemistry , Recombinant Fusion Proteins/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Structure-Activity Relationship , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/enzymology
9.
J Med Chem ; 55(7): 3228-41, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22404346
10.
Mol Cancer Ther ; 10(2): 360-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21191045

ABSTRACT

AKT is frequently deregulated in cancer, making it an attractive anticancer drug target. CCT128930 is a novel ATP-competitive AKT inhibitor discovered using fragment- and structure-based approaches. It is a potent, advanced lead pyrrolopyrimidine compound exhibiting selectivity for AKT over PKA, achieved by targeting a single amino acid difference. CCT128930 exhibited marked antiproliferative activity and inhibited the phosphorylation of a range of AKT substrates in multiple tumor cell lines in vitro, consistent with AKT inhibition. CCT128930 caused a G(1) arrest in PTEN-null U87MG human glioblastoma cells, consistent with AKT pathway blockade. Pharmacokinetic studies established that potentially active concentrations of CCT128930 could be achieved in human tumor xenografts. Furthermore, CCT128930 also blocked the phosphorylation of several downstream AKT biomarkers in U87MG tumor xenografts, indicating AKT inhibition in vivo. Antitumor activity was observed with CCT128930 in U87MG and HER2-positive, PIK3CA-mutant BT474 human breast cancer xenografts, consistent with its pharmacokinetic and pharmacodynamic properties. A quantitative immunofluorescence assay to measure the phosphorylation and total protein expression of the AKT substrate PRAS40 in hair follicles is presented. Significant decreases in pThr246 PRAS40 occurred in CCT128930-treated mouse whisker follicles in vivo and human hair follicles treated ex vivo, with minimal changes in total PRAS40. In conclusion, CCT128930 is a novel, selective, and potent AKT inhibitor that blocks AKT activity in vitro and in vivo and induces marked antitumor responses. We have also developed a novel biomarker assay for the inhibition of AKT in human hair follicles, which is currently being used in clinical trials.


Subject(s)
Antineoplastic Agents , Biomarkers, Pharmacological/analysis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Pyrroles/pharmacology , Pyrroles/pharmacokinetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Hair Follicle/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Xenograft Model Antitumor Assays
11.
J Biomol Screen ; 15(8): 918-927, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664067

ABSTRACT

Nek2 is a serine/threonine protein kinase that localizes to the centrosome and is implicated in mitotic regulation. Overexpression of Nek2 induces premature centrosome separation and nuclear defects indicative of mitotic errors, whereas depletion of Nek2 interferes with cell growth. As Nek2 expression is upregulated in a range of cancer cell lines and primary human tumors, inhibitors of Nek2 may have therapeutic value in cancer treatment. The authors used a radiometric proximity assay in a high-throughput screen to identify small-molecule inhibitors of Nek2 kinase activity. The assay was based on the measurement of the radiolabeled phosphorylated product of the kinase reaction brought into contact with the surface of wells of solid scintillant-coated microplates. Seventy nonaggregating hits were identified from approximately 73,000 compounds screened and included a number of toxoflavins and a series of viridin/wortmannin-like compounds. The viridin-like compounds were >70-fold selective for Nek2 over Nek6 and Nek7 and inhibited the growth of human tumor cell lines at concentrations consistent with their biochemical potencies. An automated mechanism-based microscopy assay in which centrosomes were visualized using pericentrin antibodies confirmed that 2 of the viridin inhibitors reduced centrosome separation in a human tumor cell line. The data presented show that pharmacological inhibition of Nek2 kinase results in the expected phenotype of disruption to centrosome function associated with growth inhibition and further supports Nek2 as a target for cancer drug discovery.


Subject(s)
Androstenes/chemistry , Bacteriocins/chemistry , Cells/drug effects , High-Throughput Screening Assays/methods , Protein Kinase Inhibitors/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Androstenes/analysis , Androstenes/pharmacology , Bacteriocins/analysis , Bacteriocins/pharmacology , Biochemical Phenomena/drug effects , Calibration , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cells/metabolism , Drug Screening Assays, Antitumor/methods , HeLa Cells , High-Throughput Screening Assays/standards , Humans , Inhibitory Concentration 50 , Models, Biological , NIMA-Related Kinases , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
12.
Neuroscientist ; 8(2): 143-53, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11954559

ABSTRACT

Glutamatergic synapses are the primary source of excitatory transmission in the central nervous system (CNS), and their formation is critical in the establishment of neuronal connections. The refinement of these connections occurs during development and also it is postulated during learning and memory. Recent progress in understanding the molecular components of synaptic junctions, together with advances in imaging techniques, has started to offer new insights into the development of excitatory synapses. Studies performed on low-density primary neuronal cultures have enabled dissection of the temporal sequence of events, which have lead to the differentiation of pre- and postsynaptic components. A central feature of the development of excitatory synapses is the accumulation of glutamatergic receptors (GluRs) at the postsynaptic site. These receptors need to be localized and fixed opposite nerve terminals that release glutamate. But for this to occur, neurons require intracellular anchoring molecules, as well as mechanisms that ensure the efficient turnover and transport of receptor proteins. This review focuses on some of the developmental changes observed in the subcellular distribution and molecular organization of AMPA and NMDA type ionotropic GluRs (iGluRs), which mediate the majority of fast excitatory neurotransmission in the CNS.


Subject(s)
Hippocampus/growth & development , Hippocampus/physiology , Receptors, Glutamate/physiology , Synapses/physiology , Animals , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Hippocampus/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL