Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
iScience ; 21: 188-204, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31670074

ABSTRACT

In pediatric neuroblastoma, MYCN-amplification correlates to poor clinical outcome and new treatment options are needed for these patients. Identifying the metabolic adaptations crucial for tumor progression may be a promising strategy to discover novel therapeutic targets. Here, we have combined proteomics, gene expression profiling, functional analysis, and metabolic tracing to decipher the impact of MYCN on neuroblastoma cell metabolism. We found that high MYCN levels are correlated with altered expression of proteins involved in multiple metabolic processes, including enhanced glycolysis and increased oxidative phosphorylation. Unexpectedly, we discovered that MYCN-amplified cells showed de novo glutamine synthesis. Furthermore, inhibition of ß-oxidation reduced the viability of MYCN-amplified cells in vitro and decreased tumor burden in vivo, while not affecting non-MYCN-amplified tumors. Our data provide information on metabolic processes in MYCN expressing tumors, which could be exploited for the development of novel targeted therapies.

2.
Proc Natl Acad Sci U S A ; 112(1): 220-5, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535357

ABSTRACT

The p53-family member TAp73 is known to function as a tumor suppressor and regulates genomic integrity, cellular proliferation, and apoptosis; however, its role in tumor angiogenesis is poorly understood. Here we demonstrate that TAp73 regulates tumor angiogenesis through repression of proangiogenic and proinflammatory cytokines. Importantly, loss of TAp73 results in highly vascularized tumors, as well as an increase in vessel permeability resulting from disruption of vascular endothelial-cadherin junctions between endothelial cells. In contrast, loss of the oncogenic p73 isoform ΔNp73 leads to reduced blood vessel formation in tumors. Furthermore, we show that up-regulated ΔNp73 levels are associated with increased angiogenesis in human breast cancer and that inhibition of TAp73 results in an accumulation of HIF-1α and up-regulation of HIF-1α target genes. Taken together, our data demonstrate that loss of TAp73 or ΔNp73 up-regulation activates the angiogenic switch that stimulates tumor growth and progression.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Hypoxia , Cell Line, Transformed , Cell Proliferation , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Permeability , Protein Isoforms/metabolism , Tumor Protein p73 , Zebrafish
3.
PLoS One ; 9(5): e97285, 2014.
Article in English | MEDLINE | ID: mdl-24859015

ABSTRACT

Members of the MYC family are the most frequently deregulated oncogenes in human cancer and are often correlated with aggressive disease and/or poorly differentiated tumors. Since patients with MYCN-amplified neuroblastoma have a poor prognosis, targeting MYCN using small molecule inhibitors could represent a promising therapeutic approach. We have previously demonstrated that the small molecule 10058-F4, known to bind to the c-MYC bHLHZip dimerization domain and inhibiting the c-MYC/MAX interaction, also interferes with the MYCN/MAX dimerization in vitro and imparts anti-tumorigenic effects in neuroblastoma tumor models with MYCN overexpression. Our previous work also revealed that MYCN-inhibition leads to mitochondrial dysfunction resulting in accumulation of lipid droplets in neuroblastoma cells. To expand our understanding of how small molecules interfere with MYCN, we have now analyzed the direct binding of 10058-F4, as well as three of its analogs; #474, #764 and 10058-F4(7RH), one metabolite C-m/z 232, and a structurally unrelated c-MYC inhibitor 10074-G5, to the bHLHZip domain of MYCN. We also assessed their ability to induce apoptosis, neurite outgrowth and lipid accumulation in neuroblastoma cells. Interestingly, all c-MYC binding molecules tested also bind MYCN as assayed by surface plasmon resonance. Using a proximity ligation assay, we found reduced interaction between MYCN and MAX after treatment with all molecules except for the 10058-F4 metabolite C-m/z 232 and the non-binder 10058-F4(7RH). Importantly, 10074-G5 and 10058-F4 were the most efficient in inducing neuronal differentiation and lipid accumulation in MYCN-amplified neuroblastoma cells. Together our data demonstrate MYCN-binding properties for a selection of small molecules, and provide functional information that could be of importance for future development of targeted therapies against MYCN-amplified neuroblastoma.


Subject(s)
Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lipid Metabolism/drug effects , Molecular Sequence Data , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Nuclear Proteins/chemistry , Oncogene Proteins/chemistry , Oxadiazoles/pharmacology , Protein Folding/drug effects , Protein Structure, Tertiary , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/metabolism , Small Molecule Libraries/metabolism
4.
Proc Natl Acad Sci U S A ; 110(25): 10258-63, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23733953

ABSTRACT

The MYC genes are the most frequently activated oncogenes in human tumors and are hence attractive therapeutic targets. MYCN amplification leads to poor clinical outcome in childhood neuroblastoma, yet strategies to modulate the function of MYCN do not exist. Here we show that 10058-F4, a characterized c-MYC/Max inhibitor, also targets the MYCN/Max interaction, leading to cell cycle arrest, apoptosis, and neuronal differentiation in MYCN-amplified neuroblastoma cells and to increased survival of MYCN transgenic mice. We also report the discovery that inhibition of MYC is accompanied by accumulation of intracellular lipid droplets in tumor cells as a direct consequence of mitochondrial dysfunction. This study expands on the current knowledge of how MYC proteins control the metabolic reprogramming of cancer cells, especially highlighting lipid metabolism and the respiratory chain as important pathways involved in neuroblastoma pathogenesis. Together our data support direct MYC inhibition as a promising strategy for the treatment of MYC-driven tumors.


Subject(s)
Apoptosis/drug effects , Lipid Metabolism/drug effects , Neuroblastoma/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Electron Transport/drug effects , Fatty Acids/metabolism , Humans , Mice , Mice, Nude , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins c-myc/metabolism , Receptor, trkA/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
PLoS One ; 6(11): e27988, 2011.
Article in English | MEDLINE | ID: mdl-22132187

ABSTRACT

Expression of MYC is deregulated in a wide range of human cancers, and is often associated with aggressive disease and poorly differentiated tumor cells. Identification of compounds with selectivity for cells overexpressing MYC would hence be beneficial for the treatment of these tumors. For this purpose we used cell lines with conditional MYCN or c-MYC expression, to screen a library of 80 conventional cytotoxic compounds for their ability to reduce tumor cell viability and/or growth in a MYC dependent way. We found that 25% of the studied compounds induced apoptosis and/or inhibited proliferation in a MYC-specific manner. The activities of the majority of these were enhanced both by c-MYC or MYCN over-expression. Interestingly, these compounds were acting on distinct cellular targets, including microtubules (paclitaxel, podophyllotoxin, vinblastine) and topoisomerases (10-hydroxycamptothecin, camptothecin, daunorubicin, doxorubicin, etoposide) as well as DNA, RNA and protein synthesis and turnover (anisomycin, aphidicholin, gliotoxin, MG132, methotrexate, mitomycin C). Our data indicate that MYC overexpression sensitizes cells to disruption of specific pathways and that in most cases c-MYC and MYCN overexpression have similar effects on the responses to cytotoxic compounds. Treatment of the cells with topoisomerase I inhibitors led to down-regulation of MYC protein levels, while doxorubicin and the small molecule MYRA-A was found to disrupt MYC-Max interaction. We conclude that the MYC pathway is only targeted by a subset of conventional cytotoxic drugs currently used in the clinic. Elucidating the mechanisms underlying their specificity towards MYC may be of importance for optimizing treatment of tumors with MYC deregulation. Our data also underscores that MYC is an attractive target for novel therapies and that cellular screenings of chemical libraries can be a powerful tool for identifying compounds with a desired biological activity.


Subject(s)
Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Doxorubicin/pharmacology , Humans , Inhibitory Concentration 50 , Neuroblastoma/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Topoisomerase I Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...