Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
J Gastroenterol Hepatol ; 37(5): 898-907, 2022 May.
Article in English | MEDLINE | ID: mdl-35244298

ABSTRACT

BACKGROUND AND AIM: Colorectal cancer (CRC) is the second leading cause of cancer death worldwide. To improve outcomes for these patients, we need to develop new treatment strategies. Personalized cancer medicine, where patients are treated based on the characteristics of their own tumor, has gained significant interest for its promise to improve outcomes and reduce unnecessary side effects. The purpose of this study was to examine the potential utility of patient-derived colorectal cancer organoids (PDCOs) in a personalized cancer medicine setting. METHODS: Patient-derived colorectal cancer organoids were derived from tissue obtained from treatment-naïve patients undergoing surgical resection for the treatment of CRC. We examined the recapitulation of key histopathological, molecular, and phenotypic characteristics of the primary tumor. RESULTS: We created a bio-resource of PDCOs from primary and metastatic CRCs. Key histopathological features were retained in PDCOs when compared with the primary tumor. Additionally, a cohort of 12 PDCOs, and their corresponding primary tumors and normal sample, were characterized through whole exome sequencing and somatic variant calling. These PDCOs exhibited a high level of concordance in key driver mutations when compared with the primary tumor. CONCLUSIONS: Patient-derived colorectal cancer organoids recapitulate characteristics of the tissue from which they are derived and are a powerful tool for cancer research. Further research will determine their utility for predicting patient outcomes in a personalized cancer medicine setting.


Subject(s)
Colorectal Neoplasms , Organoids , Cohort Studies , Colorectal Neoplasms/pathology , Humans , Organoids/pathology , Precision Medicine
2.
Analyst ; 146(7): 2277-2291, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33617612

ABSTRACT

Quantitative hyperspectral coherent Raman scattering microscopy merges imaging with spectroscopy and utilises quantitative data analysis algorithms to extract physically meaningful chemical components, spectrally and spatially-resolved, with sub-cellular resolution. This label-free non-invasive method has the potential to significantly advance our understanding of the complexity of living multicellular systems. Here, we have applied an in-house developed hyperspectral coherent anti-Stokes Raman scattering (CARS) microscope, combined with a quantitative data analysis pipeline, to imaging living mouse liver organoids as well as fixed mouse brain tissue sections xenografted with glioblastoma cells. We show that the method is capable of discriminating different cellular sub-populations, on the basis of their chemical content which is obtained from an unsupervised analysis, i.e. without prior knowledge. Specifically, in the organoids, we identify sub-populations of cells at different phases in the cell cycle, while in the brain tissue, we distinguish normal tissue from cancer cells, and, notably, tumours derived from transplanted cancer stem cells versus non-stem glioblastoma cells. The ability of the method to identify different sub-populations was validated by correlative fluorescence microscopy using fluorescent protein markers. These examples expand the application portfolio of quantitative chemical imaging by hyperspectral CARS microscopy to multicellular systems of significant biomedical relevance, pointing the way to new opportunities in non-invasive disease diagnostics.


Subject(s)
Glioblastoma , Spectrum Analysis, Raman , Algorithms , Animals , Glioblastoma/diagnostic imaging , Mice , Microscopy, Fluorescence , Proteins
3.
PLoS One ; 15(8): e0235319, 2020.
Article in English | MEDLINE | ID: mdl-32810173

ABSTRACT

Aberrant activation of the Wnt signalling pathway is required for tumour initiation and survival in the majority of colorectal cancers. The development of inhibitors of Wnt signalling has been the focus of multiple drug discovery programs targeting colorectal cancer and other malignancies associated with aberrant pathway activation. However, progression of new clinical entities targeting the Wnt pathway has been slow. One challenge lies with the limited predictive power of 2D cancer cell lines because they fail to fully recapitulate intratumoural phenotypic heterogeneity. In particular, the relationship between 2D cancer cell biology and cancer stem cell function is poorly understood. By contrast, 3D tumour organoids provide a platform in which complex cell-cell interactions can be studied. However, complex 3D models provide a challenging platform for the quantitative analysis of drug responses of therapies that have differential effects on tumour cell subpopulations. Here, we generated tumour organoids from colorectal cancer patients and tested their responses to inhibitors of Tankyrase (TNKSi) which are known to modulate Wnt signalling. Using compounds with 3 orders of magnitude difference in cellular mechanistic potency together with image-based assays, we demonstrate that morphometric analyses can capture subtle alterations in organoid responses to Wnt inhibitors that are consistent with activity against a cancer stem cell subpopulation. Overall our study highlights the value of phenotypic readouts as a quantitative method to asses drug-induced effects in a relevant preclinical model.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Organoids/drug effects , Tankyrases/antagonists & inhibitors , Adult , Animals , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/pathology , Enzyme Inhibitors/therapeutic use , Female , Humans , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Organoids/pathology
4.
Curr Opin Cell Biol ; 62: 17-25, 2020 02.
Article in English | MEDLINE | ID: mdl-31509785

ABSTRACT

In the liver, the tight spatiotemporal regulation of Wnt/ß-catenin signaling is required to establish and maintain a metabolic form of tissue polarity termed zonation. In this review, we discuss the latest technologies applied in the study of liver zonation and provide a summary of the Wnt ligand and receptor expression patterns in the hepatic lobule. We further discuss the mechanisms, by which Wnt instructive cues might be spatially confined and propagated along the central vein-portal triad axis.


Subject(s)
Gene Expression/genetics , Liver/physiopathology , Wnt Signaling Pathway/physiology , Animals , Humans
5.
Elife ; 82019 12 12.
Article in English | MEDLINE | ID: mdl-31829936

ABSTRACT

Tcf7l2 mediates Wnt/ß-Catenin signalling during development and is implicated in cancer and type-2 diabetes. The mechanisms by which Tcf7l2 and Wnt/ß-Catenin signalling elicit such a diversity of biological outcomes are poorly understood. Here, we study the function of zebrafish tcf7l2alternative splice variants and show that only variants that include exon five or an analogous human tcf7l2 variant can effectively provide compensatory repressor function to restore eye formation in embryos lacking tcf7l1a/tcf7l1b function. Knockdown of exon five specific tcf7l2 variants in tcf7l1a mutants also compromises eye formation, and these variants can effectively repress Wnt pathway activity in reporter assays using Wnt target gene promoters. We show that the repressive activities of exon5-coded variants are likely explained by their interaction with Tle co-repressors. Furthermore, phosphorylated residues in Tcf7l2 coded exon5 facilitate repressor activity. Our studies suggest that developmentally regulated splicing of tcf7l2 can influence the transcriptional output of the Wnt pathway.


Subject(s)
Eye/embryology , Gene Expression Regulation, Developmental , Protein Isoforms/biosynthesis , RNA Splicing , Transcription Factor 7-Like 2 Protein/biosynthesis , Transcription, Genetic , Zebrafish Proteins/biosynthesis , Animals , HEK293 Cells , Humans , Protein Isoforms/genetics , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway , Zebrafish , Zebrafish Proteins/genetics
6.
Cell Chem Biol ; 24(8): 958-968.e5, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28757184

ABSTRACT

The Wnt signaling pathway plays a critical role in cell proliferation and differentiation, thus it is often associated with diseases such as cancers. Unfortunately, although attractive, developing anti-cancer strategy targeting Wnt signaling has been challenging given that the most attractive targets are involved in protein-protein interactions (PPIs). Here, we develop a stapled peptide inhibitor that targets the interaction between ß-catenin and T cell factor/lymphoid enhancer-binding factor transcription factors, which are crucially involved in Wnt signaling. Our integrative approach combines peptide stapling to optimize proteolytic stability, with lessons learned from cell-penetrating peptide (CPP) design to maximize cellular uptake resulting in NLS-StAx-h, a selective, cell permeable, stapled peptide inhibitor of oncogenic Wnt signaling that efficiently inhibits ß-catenin-transcription factor interactions. We expect that this type of integrative strategy that endows stapled peptides with CPP features will be generally useful for developing inhibitors of intracellular PPIs.


Subject(s)
Cell-Penetrating Peptides/metabolism , beta Catenin/metabolism , Amino Acid Sequence , Axin Protein/genetics , Axin Protein/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Movement , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Gene Expression/drug effects , Genes, Reporter , HeLa Cells , Humans , Microscopy, Confocal , Protein Interaction Domains and Motifs , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors
7.
PLoS Comput Biol ; 13(2): e1005400, 2017 02.
Article in English | MEDLINE | ID: mdl-28245235

ABSTRACT

The Notch pathway plays a vital role in determining whether cells in the intestinal epithelium adopt a secretory or an absorptive phenotype. Cell fate specification is coordinated via Notch's interaction with the canonical Wnt pathway. Here, we propose a new mathematical model of the Notch and Wnt pathways, in which the Hes1 promoter acts as a hub for pathway crosstalk. Computational simulations of the model can assist in understanding how healthy intestinal tissue is maintained, and predict the likely consequences of biochemical knockouts upon cell fate selection processes. Chemical reaction network theory (CRNT) is a powerful, generalised framework which assesses the capacity of our model for monostability or multistability, by analysing properties of the underlying network structure without recourse to specific parameter values or functional forms for reaction rates. CRNT highlights the role of ß-catenin in stabilising the Notch pathway and damping oscillations, demonstrating that Wnt-mediated actions on the Hes1 promoter can induce dynamic transitions in the Notch system, from multistability to monostability. Time-dependent model simulations of cell pairs reveal the stabilising influence of Wnt upon the Notch pathway, in which ß-catenin- and Dsh-mediated action on the Hes1 promoter are key in shaping the subcellular dynamics. Where Notch-mediated transcription of Hes1 dominates, there is Notch oscillation and maintenance of fate flexibility; Wnt-mediated transcription of Hes1 favours bistability akin to cell fate selection. Cells could therefore regulate the proportion of Wnt- and Notch-mediated control of the Hes1 promoter to coordinate the timing of cell fate selection as they migrate through the intestinal epithelium and are subject to reduced Wnt stimuli. Furthermore, mutant cells characterised by hyperstimulation of the Wnt pathway may, through coupling with Notch, invert cell fate in neighbouring healthy cells, enabling an aberrant cell to maintain its neighbours in mitotically active states.


Subject(s)
Intestinal Mucosa/metabolism , Models, Biological , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factor HES-1/metabolism , Wnt Signaling Pathway/physiology , Biological Clocks/physiology , Cells, Cultured , Computer Simulation , Humans , Receptor Cross-Talk/physiology
8.
Gut ; 66(7): 1268-1277, 2017 07.
Article in English | MEDLINE | ID: mdl-27618837

ABSTRACT

OBJECTIVE: Transcribed-ultraconserved regions (T-UCR) are long non-coding RNAs which are conserved across species and are involved in carcinogenesis. We studied T-UCRs downstream of the Wnt/ß-catenin pathway in liver cancer. DESIGN: Hypomorphic Apc mice (Apcfl/fl) and thiocetamide (TAA)-treated rats developed Wnt/ß-catenin dependent hepatocarcinoma (HCC) and cholangiocarcinoma (CCA), respectively. T-UCR expression was assessed by microarray, real-time PCR and in situ hybridisation. RESULTS: Overexpression of the T-UCR uc.158- could differentiate Wnt/ß-catenin dependent HCC from normal liver and from ß-catenin negative diethylnitrosamine (DEN)-induced HCC. uc.158- was overexpressed in human HepG2 versus Huh7 cells in line with activation of the Wnt pathway. In vitro modulation of ß-catenin altered uc.158- expression in human malignant hepatocytes. uc.158- expression was increased in CTNNB1-mutated human HCCs compared with non-mutated human HCCs, and in human HCC with nuclear localisation of ß-catenin. uc.158- was increased in TAA rat CCA and reduced after treatment with Wnt/ß-catenin inhibitors. uc.158- expression was negative in human normal liver and biliary epithelia, while it was increased in human CCA in two different cohorts. Locked nucleic acid-mediated inhibition of uc.158- reduced anchorage cell growth, 3D-spheroid formation and spheroid-based cell migration, and increased apoptosis in HepG2 and SW1 cells. miR-193b was predicted to have binding sites within the uc.158- sequence. Modulation of uc.158- changed miR-193b expression in human malignant hepatocytes. Co-transfection of uc.158- inhibitor and anti-miR-193b rescued the effect of uc.158- inhibition on cell viability. CONCLUSIONS: We showed that uc.158- is activated by the Wnt pathway in liver cancers and drives their growth. Thus, it may represent a promising target for the development of novel therapeutics.


Subject(s)
Bile Duct Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , Cholangiocarcinoma/metabolism , Conserved Sequence/genetics , Liver Neoplasms/metabolism , RNA, Untranslated/genetics , Wnt Signaling Pathway , Animals , Bile Duct Neoplasms/genetics , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Hepatocytes/metabolism , Humans , Liver Neoplasms/genetics , Mice, Knockout , MicroRNAs/metabolism , Neoplasms, Experimental , Transfection , beta Catenin/genetics , beta Catenin/metabolism
9.
Elife ; 52016 12 09.
Article in English | MEDLINE | ID: mdl-27935476

ABSTRACT

Mediator-associated kinases CDK8/19 are context-dependent drivers or suppressors of tumorigenesis. Their inhibition is predicted to have pleiotropic effects, but it is unclear whether this will impact on the clinical utility of CDK8/19 inhibitors. We discovered two series of potent chemical probes with high selectivity for CDK8/19. Despite pharmacodynamic evidence for robust on-target activity, the compounds exhibited modest, though significant, efficacy against human tumor lines and patient-derived xenografts. Altered gene expression was consistent with CDK8/19 inhibition, including profiles associated with super-enhancers, immune and inflammatory responses and stem cell function. In a mouse model expressing oncogenic beta-catenin, treatment shifted cells within hyperplastic intestinal crypts from a stem cell to a transit amplifying phenotype. In two species, neither probe was tolerated at therapeutically-relevant exposures. The complex nature of the toxicity observed with two structurally-differentiated chemical series is consistent with on-target effects posing significant challenges to the clinical development of CDK8/19 inhibitors.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Mediator Complex/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/toxicity , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Disease Models, Animal , Heterografts , Humans , Hyperplasia/drug therapy , Mice , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/toxicity , Treatment Outcome
10.
Nat Commun ; 7: 13207, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27782124

ABSTRACT

The development of in vitro culture systems quantitatively and qualitatively recapitulating normal breast biology is key to the understanding of mammary gland biology. Current three-dimensional mammary culture systems have not demonstrated concurrent proliferation and functional differentiation ex vivo in any system for longer than 2 weeks. Here, we identify conditions including Neuregulin1 and R-spondin 1, allowing maintenance and expansion of mammary organoids for 2.5 months in culture. The organoids comprise distinct basal and luminal compartments complete with functional steroid receptors and stem/progenitor cells able to reconstitute a complete mammary gland in vivo. Alternative conditions are also described that promote enrichment of basal cells organized into multiple layers surrounding a keratinous core, reminiscent of structures observed in MMTV-Wnt1 tumours. These conditions comprise a unique tool that should further understanding of normal mammary gland development, the molecular mechanism of hormone action and signalling events whose deregulation leads to breast tumourigenesis.


Subject(s)
Mammary Glands, Animal/metabolism , Neuregulin-1/metabolism , Organoids/metabolism , Receptor, ErbB-3/metabolism , Receptor, ErbB-4/metabolism , Wnt Signaling Pathway , Animals , Female , Gene Expression Regulation, Developmental , Karyotyping , Mammary Glands, Animal/growth & development , Mice, Inbred C57BL , Microscopy, Confocal , Neuregulin-1/genetics , Organoids/growth & development , Receptor, ErbB-3/genetics , Receptor, ErbB-4/genetics , Time-Lapse Imaging/methods , Tissue Culture Techniques/methods
11.
J Med Chem ; 59(20): 9337-9349, 2016 Oct 27.
Article in English | MEDLINE | ID: mdl-27490956

ABSTRACT

The mediator complex-associated cyclin dependent kinase CDK8 regulates ß-catenin-dependent transcription following activation of WNT signaling. Multiple lines of evidence suggest CDK8 may act as an oncogene in the development of colorectal cancer. Here we describe the successful optimization of an imidazo-thiadiazole series of CDK8 inhibitors that was identified in a high-throughput screening campaign and further progressed by structure-based design. In several optimization cycles, we improved the microsomal stability, potency, and kinase selectivity. The initial imidazo-thiadiazole scaffold was replaced by a 3-methyl-1H-pyrazolo[3,4-b]-pyridine which resulted in compound 25 (MSC2530818) that displayed excellent kinase selectivity, biochemical and cellular potency, microsomal stability, and is orally bioavailable. Furthermore, we demonstrated modulation of phospho-STAT1, a pharmacodynamic biomarker of CDK8 activity, and tumor growth inhibition in an APC mutant SW620 human colorectal carcinoma xenograft model after oral administration. Compound 25 demonstrated suitable potency and selectivity to progress into preclinical in vivo efficacy and safety studies.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Drug Discovery , High-Throughput Screening Assays , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Thiadiazoles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 8/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry
12.
J Med Chem ; 59(3): 1078-101, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26796641

ABSTRACT

The Mediator complex-associated cyclin-dependent kinase CDK8 has been implicated in human disease, particularly in colorectal cancer where it has been reported as a putative oncogene. Here we report the discovery of 109 (CCT251921), a potent, selective, and orally bioavailable inhibitor of CDK8 with equipotent affinity for CDK19. We describe a structure-based design approach leading to the discovery of a 3,4,5-trisubstituted-2-aminopyridine series and present the application of physicochemical property analyses to successfully reduce in vivo metabolic clearance, minimize transporter-mediated biliary elimination while maintaining acceptable aqueous solubility. Compound 109 affords the optimal compromise of in vitro biochemical, pharmacokinetic, and physicochemical properties and is suitable for progression to animal models of cancer.


Subject(s)
Aminopyridines/pharmacology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Discovery , Small Molecule Libraries/pharmacology , Administration, Oral , Aminopyridines/administration & dosage , Aminopyridines/chemistry , Animals , Biological Availability , Caco-2 Cells , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Dogs , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Rats , Rats, Wistar , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Solubility , Structure-Activity Relationship , Xenograft Model Antitumor Assays
13.
Mol Cancer ; 14: 206, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643252

ABSTRACT

BACKGROUND: Wnt/ß-catenin signaling is often portrayed as a simple pathway that is initiated by Wnt ligand at the cell surface leading, via linear series of interactions between 'core pathway' members, to the induction of nuclear transcription from genes flanked by ß-catenin/TCF transcription factor binding sites. Wnt/ß-catenin signaling is also regulated by a much larger set of 'non-core regulators'. However the relationship between 'non-core regulators' is currently not well understood. Aberrant activation of the pathway has been shown to drive tumorgenesis in a number of different tissues. METHODS: Mammalian cells engineered to have a partially-active level of Wnt/ß-catenin signaling were screened by transfection for proteins that up or down-regulated a mid-level of TCF-dependent transcription induced by transient expression of an activated LRP6 Wnt co-receptor (∆NLRP). RESULTS: 141 novel regulators of TCF-dependent transcription were identified. Surprisingly, when tested without ∆NLRP activation, most up-regulators failed to alter TCF-dependent transcription. However, when expressed in pairs, 27 % (466/1170) functionally interacted to alter levels of TCF-dependent transcription. When proteins were displayed as nodes connected by their ability to co-operate in the regulation of TCF-dependent transcription, a network of functional interactions was revealed. In this network, 'core pathway' components (Eg. ß-catenin, GSK-3, Dsh) were found to be the most highly connected nodes. Activation of different nodes in this network impacted on the sensitivity to Wnt pathway small molecule antagonists. CONCLUSIONS: The 'functional connectome' identified here strongly supports an alternative model of the Wnt pathway as a complex context-dependent network. The network further suggests that mutational activation of highly connected Wnt signaling nodes predisposed cells to further context-dependent alterations in levels of TCF-dependent transcription that may be important during tumor progression and treatment.


Subject(s)
Antineoplastic Agents/pharmacology , TCF Transcription Factors/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway/drug effects , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , HEK293 Cells , Humans , Protein Interaction Maps , Transcription, Genetic , Xenopus laevis
14.
Nat Chem Biol ; 11(12): 973-980, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502155

ABSTRACT

There is unmet need for chemical tools to explore the role of the Mediator complex in human pathologies ranging from cancer to cardiovascular disease. Here we determine that CCT251545, a small-molecule inhibitor of the WNT pathway discovered through cell-based screening, is a potent and selective chemical probe for the human Mediator complex-associated protein kinases CDK8 and CDK19 with >100-fold selectivity over 291 other kinases. X-ray crystallography demonstrates a type 1 binding mode involving insertion of the CDK8 C terminus into the ligand binding site. In contrast to type II inhibitors of CDK8 and CDK19, CCT251545 displays potent cell-based activity. We show that CCT251545 and close analogs alter WNT pathway-regulated gene expression and other on-target effects of modulating CDK8 and CDK19, including expression of genes regulated by STAT1. Consistent with this, we find that phosphorylation of STAT1(SER727) is a biomarker of CDK8 kinase activity in vitro and in vivo. Finally, we demonstrate in vivo activity of CCT251545 in WNT-dependent tumors.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Molecular Probes/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Spiro Compounds/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Humans , Models, Molecular , Molecular Probes/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Spiro Compounds/chemistry
16.
J Med Chem ; 58(4): 1717-35, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25680029

ABSTRACT

WNT signaling is frequently deregulated in malignancy, particularly in colon cancer, and plays a key role in the generation and maintenance of cancer stem cells. We report the discovery and optimization of a 3,4,5-trisubstituted pyridine 9 using a high-throughput cell-based reporter assay of WNT pathway activity. We demonstrate a twisted conformation about the pyridine-piperidine bond of 9 by small-molecule X-ray crystallography. Medicinal chemistry optimization to maintain this twisted conformation, cognisant of physicochemical properties likely to maintain good cell permeability, led to 74 (CCT251545), a potent small-molecule inhibitor of WNT signaling with good oral pharmacokinetics. We demonstrate inhibition of WNT pathway activity in a solid human tumor xenograft model with evidence for tumor growth inhibition following oral dosing. This work provides a successful example of hypothesis-driven medicinal chemistry optimization from a singleton hit against a cell-based pathway assay without knowledge of the biochemical target.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Evaluation, Preclinical/methods , Luciferases/antagonists & inhibitors , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Spiro Compounds/pharmacology , Wnt Signaling Pathway/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biological Assay/methods , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Crystallography, X-Ray , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Luciferases/metabolism , Mice , Models, Molecular , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Spiro Compounds/administration & dosage , Spiro Compounds/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
17.
Acta Neurochir (Wien) ; 157(3): 487-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25585835

ABSTRACT

BACKGROUND: We previously reported on a series of side errors in cranial neurosurgery that occurred around the UK before the year 2006. That survey was prompted by a cluster of six cranial and spinal side errors that occurred in the neurosurgery department in Newcastle upon Tyne during the year 2006. The report was part of our investigation into the problem and how to solve it. METHODS: A human factors training programme was run in the department in response to a further side error. All 125 members of the neurosurgical theatre staff attended 1 of 5 training days. Fifteen days of professional observation and coaching were held within the theatre suite. Time between errors was recorded. The success or otherwise of human factors measures such as checking and briefing was observed. RESULTS: A side checking system was adopted and became universal. Pre-list briefing meetings were adopted and quickly became widely used but took several years to become universal. Post-list debriefing meetings were introduced but were not widely adopted and quickly fell out of use. Mean time between side errors was 2 months pre-intervention, 18 months after introducing a standardised checking system and 82 error free months had passed since the human factors training programme. CONCLUSIONS: Side errors in neurosurgery can be reduced by a combination of systematic checking and education. We suspect that education is useful in reducing error rates from low to very low but, as is generally true of human factor interventions, the evidence for this is soft.


Subject(s)
Medical Errors/prevention & control , Neurosurgical Procedures/standards , Clinical Protocols , Humans , Medical Errors/psychology , Medical Errors/statistics & numerical data , Neurosurgical Procedures/education , Neurosurgical Procedures/methods , Risk Factors , Surgery Department, Hospital/standards , Surgery Department, Hospital/statistics & numerical data , United Kingdom
18.
Antioxid Redox Signal ; 22(7): 555-71, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25336178

ABSTRACT

AIMS: Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a master regulator of oxidant and xenobiotic metabolism, but it is unknown how it is regulated to provide basal expression of this defense system. Here, we studied the putative connection between NRF2 and the canonical WNT pathway, which modulates hepatocyte metabolism. RESULTS: WNT-3A increased the levels of NRF2 and its transcriptional signature in mouse hepatocytes and HEK293T cells. The use of short interfering RNAs in hepatocytes and mouse embryonic fibroblasts which are deficient in the redox sensor Kelch-like ECH-associated protein 1 (KEAP1) indicated that WNT-3A activates NRF2 in a ß-Catenin- and KEAP1-independent manner. WNT-3A stabilized NRF2 by preventing its GSK-3-dependent phosphorylation and subsequent SCF/ß-TrCP-dependent ubiquitination and proteasomal degradation. Axin1 and NRF2 were physically associated in a protein complex that was regulated by WNT-3A, involving the central region of Axin1 and the Neh4/Neh5 domains of NRF2. Axin1 knockdown increased NRF2 protein levels, while Axin1 stabilization with Tankyrase inhibitors blocked WNT/NRF2 signaling. The relevance of this novel pathway was assessed in mice with a conditional deletion of Axin1 in the liver, which showed upregulation of the NRF2 signature in hepatocytes and disruption of liver zonation of antioxidant metabolism. INNOVATION: NRF2 takes part in a protein complex with Axin1 that is regulated by the canonical WNT pathway. This new WNT-NRF2 axis controls the antioxidant metabolism of hepatocytes. CONCLUSION: These results uncover the participation of NRF2 in a WNT-regulated signalosome that participates in basal maintenance of hepatic antioxidant metabolism.


Subject(s)
Antioxidants/metabolism , Axin Protein/genetics , Axin Protein/metabolism , Hepatocytes/metabolism , Wnt3A Protein/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Liver/cytology , Liver/metabolism , Mice , Mice, Transgenic
19.
Oncotarget ; 5(14): 5736-49, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25026278

ABSTRACT

H-Prune hydrolyzes short-chain polyphosphates (PPase activity) together with an hitherto cAMP-phosphodiesterase (PDE), the latest influencing different human cancers by its overexpression. H-Prune promotes cell migration in cooperation with glycogen synthase kinase-3 (Gsk-3ß). Gsk-3ß is a negative regulator of canonical WNT/ß-catenin signaling. Here, we investigate the role of Gsk-3ß/h-Prune complex in the regulation of WNT/ß-catenin signaling, demonstrating the h-Prune capability to activate WNT signaling also in a paracrine manner, through Wnt3a secretion. In vivo study demonstrates that h-Prune silencing inhibits lung metastasis formation, increasing mouse survival. We assessed h-Prune levels in peripheral blood of lung cancer patients using ELISA assay, showing that h-Prune is an early diagnostic marker for lung cancer. Our study dissects out the mechanism of action of h-Prune in tumorigenic cells and also sheds light on the identification of a new therapeutic target in non-small-cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/blood , Glycogen Synthase Kinase 3/metabolism , Lung Neoplasms/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins/genetics , Disease Progression , Female , Glycogen Synthase Kinase 3 beta , Heterografts , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Phosphoric Monoester Hydrolases , beta Catenin/genetics
20.
Sci Signal ; 7(317): ra26, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24643799

ABSTRACT

Wnt signaling plays a central role in development, adult tissue homeostasis, and cancer. Several steps in the canonical Wnt/ß-catenin signaling cascade are regulated by ubiquitylation, a protein modification that influences the stability, subcellular localization, or interactions of target proteins. To identify regulators of the Wnt/ß-catenin pathway, we performed an RNA interference screen in Caenorhabditis elegans and identified the HECT domain-containing ubiquitin ligase EEL-1 as an inhibitor of Wnt signaling. In human embryonic kidney 293T cells, knockdown of the EEL-1 homolog Huwe1 enhanced the activity of a Wnt reporter in cells stimulated with Wnt3a or in cells that overexpressed casein kinase 1 (CK1) or a constitutively active mutant of the Wnt co-receptor low-density lipoprotein receptor-related protein 6 (LRP6). However, knockdown of Huwe1 had no effect on reporter gene expression in cells expressing constitutively active ß-catenin, suggesting that Huwe1 inhibited Wnt signaling upstream of ß-catenin and downstream of CK1 and LRP6. Huwe1 bound to and ubiquitylated the cytoplasmic Wnt pathway component Dishevelled (Dvl) in a Wnt3a- and CK1ε-dependent manner. Mass spectrometric analysis showed that Huwe1 promoted K63-linked, but not K48-linked, polyubiquitination of Dvl. Instead of targeting Dvl for degradation, ubiquitylation of the DIX domain of Dvl by Huwe1 inhibited Dvl multimerization, which is necessary for its function. Our findings indicate that Huwe1 is part of an evolutionarily conserved negative feedback loop in the Wnt/ß-catenin pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dishevelled Proteins , HEK293 Cells , Humans , Mass Spectrometry , RNA Interference , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitination , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...