Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Pathol ; 240(1): 84-95, 2016 09.
Article in English | MEDLINE | ID: mdl-27287813

ABSTRACT

Sebaceous carcinoma (SC) is a rare but aggressive malignancy with frequent recurrence and metastases. Surgery is the mainstay of therapy, but effective systemic therapies are lacking because the molecular alterations driving SC remain poorly understood. To identify these, we performed whole-exome next-generation sequencing of 409 cancer-associated genes on 27 SCs (18 primary/locally recurrent ocular, 5 paired metastatic ocular, and 4 primary extraocular) from 20 patients. In ocular SC, we identified 139 non-synonymous somatic mutations (median/lesion 3; range 0-23). Twenty-five of 139 mutations (18%) occurred in potentially clinically actionable genes in 6 of 16 patients. The most common mutations were mutations in TP53 (n = 9), RB1 (n = 6), PIK3CA (n = 2), PTEN (n = 2), ERBB2 (n = 2), and NF1 (n = 2). TP53 and RB1 mutations were restricted to ocular SC and correlated with aberrant TP53 and RB protein expression. Systematic pathway analyses demonstrated convergence of these mutations to activation of the PI3K signalling cascade, and PI3K pathway activation was confirmed in tumours with PTEN and/or PIK3CA mutations. Considerable inter-tumoural heterogeneity was observed between paired primary and metastatic ocular SCs. In primary extraocular SC, we identified 77 non-synonymous somatic mutations (median/lesion 22.5; range 3-29). This overall higher mutational load was attributed to a microsatellite instability phenotype in three of four patients and somatically acquired mutations in mismatch repair genes in two of four patients. Eighteen of 77 mutations (23%) were in potentially clinically actionable genes in three of four patients, including BTK, FGFR2, PDGFRB, HRAS, and NF1 mutations. Identification of potentially clinically actionable mutations in 9 of 20 SC patients (45%) underscores the importance of next-generation sequencing to expand the spectrum of genotype-matched targeted therapies. Frequent activation of PI3K signalling pathways provides a strong rationale for application of mTOR inhibitors in the management of this disease. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adenocarcinoma, Sebaceous/genetics , DNA Mutational Analysis/methods , Eye Neoplasms/genetics , Sebaceous Gland Neoplasms/genetics , Adenocarcinoma, Sebaceous/pathology , Class I Phosphatidylinositol 3-Kinases , Eye Neoplasms/pathology , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Microsatellite Instability , Mutation , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Retinoblastoma Binding Proteins/genetics , Sebaceous Gland Neoplasms/pathology , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
2.
Nat Commun ; 7: 11169, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27041221

ABSTRACT

A deeper mechanistic understanding of tumour angiogenesis regulation is needed to improve current anti-angiogenic therapies. Here we present evidence from systems-based miRNA analyses of large-scale patient data sets along with in vitro and in vivo experiments that miR-192 is a key regulator of angiogenesis. The potent anti-angiogenic effect of miR-192 stems from its ability to globally downregulate angiogenic pathways in cancer cells through regulation of EGR1 and HOXB9. Low miR-192 expression in human tumours is predictive of poor clinical outcome in several cancer types. Using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes, we show that miR-192 delivery leads to inhibition of tumour angiogenesis in multiple ovarian and renal tumour models, resulting in tumour regression and growth inhibition. This anti-angiogenic and anti-tumour effect is more robust than that observed with an anti-VEGF antibody. Collectively, these data identify miR-192 as a central node in tumour angiogenesis and support the use of miR-192 in an anti-angiogenesis therapy.


Subject(s)
Early Growth Response Protein 1/physiology , Gene Regulatory Networks , Homeodomain Proteins/physiology , Kidney Neoplasms/genetics , MicroRNAs/physiology , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Animals , Cell Line, Tumor , Down-Regulation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Genetic Therapy , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/therapy , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/therapy , Phosphatidylcholines , Tumor Burden
3.
Elife ; 5: e10250, 2016 Feb 27.
Article in English | MEDLINE | ID: mdl-26920219

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions.


Subject(s)
Exosomes , Fibroblasts/metabolism , Glucose/metabolism , Neoplasms/physiopathology , Tumor Microenvironment , Exosomes/metabolism , Fermentation , Glycolysis , Lactic Acid/metabolism , Oxidative Phosphorylation
4.
PLoS One ; 10(10): e0140072, 2015.
Article in English | MEDLINE | ID: mdl-26505200

ABSTRACT

MicroRNAs (miRNAs) play a crucial role in the maintenance of cellular homeostasis by regulating the expression of their target genes. As such, the dysregulation of miRNA expression has been frequently linked to cancer. With rapidly accumulating molecular data linked to patient outcome, the need for identification of robust multi-omic molecular markers is critical in order to provide clinical impact. While previous bioinformatic tools have been developed to identify potential biomarkers in cancer, these methods do not allow for rapid classification of oncogenes versus tumor suppressors taking into account robust differential expression, cutoffs, p-values and non-normality of the data. Here, we propose a methodology, Robust Selection Algorithm (RSA) that addresses these important problems in big data omics analysis. The robustness of the survival analysis is ensured by identification of optimal cutoff values of omics expression, strengthened by p-value computed through intensive random resampling taking into account any non-normality in the data and integration into multi-omic functional networks. Here we have analyzed pan-cancer miRNA patient data to identify functional pathways involved in cancer progression that are associated with selected miRNA identified by RSA. Our approach demonstrates the way in which existing survival analysis techniques can be integrated with a functional network analysis framework to efficiently identify promising biomarkers and novel therapeutic candidates across diseases.


Subject(s)
Biomarkers, Tumor/genetics , Computational Biology , MicroRNAs/genetics , Neoplasms/genetics , Algorithms , Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis
6.
NPJ Syst Biol Appl ; 1: 15001, 2015.
Article in English | MEDLINE | ID: mdl-28725457

ABSTRACT

BACKGROUND: Regulation of gene expression by microRNAs (miRNAs) is critical for determining cellular fate and function. Dysregulation of miRNA expression contributes to the development and progression of multiple diseases. miRNA can target multiple mRNAs, making deconvolution of the effects of miRNA challenging and the complexity of regulation of cellular pathways by miRNAs at the functional protein level remains to be elucidated. Therefore, we sought to determine the effects of expression of miRNAs in breast and ovarian cancer cells on cellular pathways by measuring systems-wide miRNA perturbations to protein and phosphoproteins. METHODS: We measure protein level changes by reverse-phase protein array (RPPA) in MDA-MB-231, SKOV3.ip1 and HEYA8 cancer cell lines transfected by a library of 879 human miRNA mimics. RESULTS: The effects of multiple miRNAs-protein networks converged in five broad functional clusters of miRNA, suggesting a broad overlap of miRNA action on cellular pathways. Detailed analysis of miRNA clusters revealed novel miRNA/cell cycle protein networks, which we functionally validated. De novo phosphoprotein network estimation using Gaussian graphical modeling, using no priors, revealed known and novel protein interplay, which we also observed in patient ovarian tumor proteomic data. We identified several miRNAs that have pluripotent activities across multiple cellular pathways. In particular we studied miR-365a whose expression is associated with poor survival across several cancer types and demonstrated that anti-miR-365 significantly reduced tumor formation in animal models. CONCLUSIONS: Mapping of miRNA-induced protein and phosphoprotein changes onto pathways revealed new miRNA-cellular pathway connectivity, paving the way for targeting of dysregulated pathways with potential miRNA-based therapeutics.

7.
Genome Res ; 25(2): 257-67, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25378249

ABSTRACT

We introduce a method for simultaneous prediction of microRNA-target interactions and their mediated competitive endogenous RNA (ceRNA) interactions. Using high-throughput validation assays in breast cancer cell lines, we show that our integrative approach significantly improves on microRNA-target prediction accuracy as assessed by both mRNA and protein level measurements. Our biochemical assays support nearly 500 microRNA-target interactions with evidence for regulation in breast cancer tumors. Moreover, these assays constitute the most extensive validation platform for computationally inferred networks of microRNA-target interactions in breast cancer tumors, providing a useful benchmark to ascertain future improvements.


Subject(s)
Computational Biology/methods , Epistasis, Genetic , Gene Regulatory Networks , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , 3' Untranslated Regions , Algorithms , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cluster Analysis , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/chemistry , RNA, Messenger/chemistry
8.
Nat Commun ; 5: 3887, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24871328

ABSTRACT

Protein levels and function are poorly predicted by genomic and transcriptomic analysis of patient tumours. Therefore, direct study of the functional proteome has the potential to provide a wealth of information that complements and extends genomic, epigenomic and transcriptomic analysis in The Cancer Genome Atlas (TCGA) projects. Here we use reverse-phase protein arrays to analyse 3,467 patient samples from 11 TCGA 'Pan-Cancer' diseases, using 181 high-quality antibodies that target 128 total proteins and 53 post-translationally modified proteins. The resultant proteomic data are integrated with genomic and transcriptomic analyses of the same samples to identify commonalities, differences, emergent pathways and network biology within and across tumour lineages. In addition, tissue-specific signals are reduced computationally to enhance biomarker and target discovery spanning multiple tumour lineages. This integrative analysis, with an emphasis on pathways and potentially actionable proteins, provides a framework for determining the prognostic, predictive and therapeutic relevance of the functional proteome.


Subject(s)
Genome, Human , Neoplasm Proteins/metabolism , Neoplasms/genetics , Proteomics , Cluster Analysis , Gene Dosage , Humans , Neoplasm Proteins/genetics , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Statistics, Nonparametric
9.
Mol Syst Biol ; 8: 596, 2012.
Article in English | MEDLINE | ID: mdl-22864381

ABSTRACT

Dynamic interactions between intracellular networks regulate cellular homeostasis and responses to perturbations. Targeted therapy is aimed at perturbing oncogene addiction pathways in cancer, however, development of acquired resistance to these drugs is a significant clinical problem. A network-based computational analysis of global gene expression data from matched sensitive and acquired drug-resistant cells to lapatinib, an EGFR/ErbB2 inhibitor, revealed an increased expression of the glucose deprivation response network, including glucagon signaling, glucose uptake, gluconeogenesis and unfolded protein response in the resistant cells. Importantly, the glucose deprivation response markers correlated significantly with high clinical relapse rates in ErbB2-positive breast cancer patients. Further, forcing drug-sensitive cells into glucose deprivation rendered them more resistant to lapatinib. Using a chemical genomics bioinformatics mining of the CMAP database, we identified drugs that specifically target the glucose deprivation response networks to overcome the resistant phenotype and reduced survival of resistant cells. This study implicates the chronic activation of cellular compensatory networks in response to targeted therapy and suggests novel combinations targeting signaling and metabolic networks in tumors with acquired resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling/methods , Quinazolines/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Flow Cytometry , Genomics/methods , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Lapatinib , Macrolides/pharmacology , Metformin/pharmacology , Models, Biological , Molecular Targeted Therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction/genetics
10.
Cancer Lett ; 307(1): 6-17, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21543155

ABSTRACT

OBJECTIVES: Obesity has been reported to increase the risk of colorectal cancer, which may due to aberrant lipid metabolism. And recently findings of monoacylglycerol lipase provide a novel evidence in the correlation of obesity and cancer. So in this study, we investigated the effect of MAGL in regulation of tumor growth in colorectal cancer. METHODS: MAGL expression in tumor tissues was estimated, and then JZL184 and siRNA were used to knockdown the expression of MAGL in colorectal cancer cells. Cell viability and invasion were detected to estimate the influence of MAGL knocked down in vitro and vivo. Then cell proliferation, apoptosis, cell cycle transition and screening of candidate genes were performed for further exploring of the effect mediated by MAGL knocked down. RESULTS: It was noted that the expression of MAGL was highly elevated in tumor tissues, however, it was found only significantly correlated with the BMI index. Tumor cells' growth and invasion was significantly inhibited in vitro and in vivo induced by pharmacological and siRNA mediated MAGL knocked down. Cell proliferation was reduced and apoptosis was increased. And two target genes Cyclin D1 and Bcl-2 seemed to be repressed by MAGL knocked down. CONCLUSIONS: This study demonstrated colorectal cancer cells growth can be inhibited via knockdown of MAGL, which manipulate tumor cells proliferation and apoptosis by downregulation of Cyclin D1 and Bcl-2. It provides a novel therapeutic target in treatment of colorectal cancer and a further support for the correlation of obesity and colorectal cancer.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/prevention & control , Monoacylglycerol Lipases/genetics , RNA Interference/physiology , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Movement , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Gene Knockdown Techniques , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Middle Aged , Monoacylglycerol Lipases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Aging (Albany NY) ; 2(12): 900-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21191148

ABSTRACT

Defects in the DNA damage response often lead to an increased susceptibility to cancer, and so the DDR presents an interesting set of novel therapeutic targets. The maintenance of genomic integrity by the DDR has also been found to be involved in the process of organismal ageing. While the removal of cells containing damaged DNA can be beneficial in the prevention of cancer, it may contribute to both normal and pathological ageing.


Subject(s)
Aging/genetics , DNA Damage , DNA Repair , Neoplasms/genetics , Age Factors , Aging/metabolism , Aging/pathology , Animals , Genomic Instability , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...