Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Sci Rep ; 12(1): 855, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039581

ABSTRACT

The proto-oncogene MYC is important for development and cell growth, however, its abnormal regulation causes cancer. Recent studies identified distinct enhancers of MYC in various cancers, but any MYC enhancer(s) in hepatocellular carcinoma (HCC) remain(s) elusive. By analyzing H3K27ac enrichment and enhancer RNA (eRNA) expression in cultured HCC cells, we identified six putative MYC enhancer regions. Amongst these, two highly active enhancers, located ~ 800 kb downstream of the MYC gene, were identified by qRT-PCR and reporter assays. We functionally confirmed these enhancers by demonstrating a significantly reduced MYC expression and cell proliferation upon CRISPR/Cas9-based deletion and/or antisense oligonucleotide (ASO)-mediated inhibition. In conclusion, we identified potential MYC enhancers of HCC and propose that the associated eRNAs may be suitable targets for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/physiology , Genes, myc/drug effects , Genes, myc/physiology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , RNA, Untranslated , Cell Line, Tumor , Humans
2.
Clin Transl Med ; 12(1): e703, 2022 01.
Article in English | MEDLINE | ID: mdl-35073459

ABSTRACT

BACKGROUND: Chemoresistance to cisplatin (DDP) remains a major challenge in advanced gastric cancer (GC) treatment. Although accumulating evidence suggests an association between dysregulation of long non-coding RNAs (lncRNAs) and chemoresistance, the regulatory functions and complexities of lncRNAs in modulating DDP-based chemotherapy in GC remain under-investigated. This study was designed to explore the critical chemoresistance-related lncRNAs in GC and identify novel therapeutic targets for patients with chemoresistant GC. METHODS: Chemoresistance-related lncRNAs were identified through microarray and verified through a quantitative real-time polymerase chain reaction (qRT-PCR). Proteins bound by lncRNAs were identified through a human proteome array and validated through RNA immunoprecipitation (RIP) and RNA pull-down assays. Co-immunoprecipitation and ubiquitination assays were performed to explore the molecular mechanisms of the Musashi2 (MSI2) post-modification. The effects of LINC00942 (LNC942) and MSI2 on DDP-based chemotherapy were investigated through MTS, apoptosis assays and xenograft tumour formation in vivo. RESULTS: LNC942 was found to be up-regulated in chemoresistant GC cells, and its high expression was positively correlated with the poor prognosis of patients with GC. Functional studies indicated that LNC942 confers chemoresistance to GC cells by impairing apoptosis and inducing stemness. Mechanically, LNC942 up-regulated the MSI2 expression by preventing its interaction with SCFß-TRCP E3 ubiquitin ligase, eventually inhibiting ubiquitination. Then, LNC942 stabilized c-Myc mRNA in an N6-methyladenosine (m6 A)-dependent manner. As a potential m6 A recognition protein, MSI2 stabilized c-Myc mRNA with m6 A modifications. Moreover, inhibition of the LNC942-MSI2-c-Myc axis was found to restore chemosensitivity both in vitro and in vivo. CONCLUSIONS: These results uncover a chemoresistant accelerating function of LNC942 in GC, and disrupting the LNC942-MSI2-c-Myc axis could be a novel therapeutic strategy for GC patients undergoing chemoresistance.


Subject(s)
Cisplatin/metabolism , Drug Resistance/drug effects , Genes, myc/drug effects , RNA, Long Noncoding/agonists , RNA-Binding Proteins/antagonists & inhibitors , Cisplatin/therapeutic use , Genes, myc/physiology , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/therapeutic use , RNA-Binding Proteins/genetics , RNA-Binding Proteins/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
3.
Cell Rep ; 38(4): 110292, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081348

ABSTRACT

The MYC oncogene has been studied for decades, yet there is still intense debate over how this transcription factor controls gene expression. Here, we seek to answer these questions with an in vivo readout of discrete events of gene expression in single cells. We engineered an optogenetic variant of MYC (Pi-MYC) and combined this tool with single-molecule RNA and protein imaging techniques to investigate the role of MYC in modulating transcriptional bursting and transcription factor binding dynamics in human cells. We find that the immediate consequence of MYC overexpression is an increase in the duration rather than in the frequency of bursts, a functional role that is different from the majority of human transcription factors. We further propose that the mechanism by which MYC exerts global effects on the active period of genes is by altering the binding dynamics of transcription factors involved in RNA polymerase II complex assembly and productive elongation.


Subject(s)
Gene Expression Regulation/genetics , Genes, myc/physiology , Transcription, Genetic/physiology , Animals , Cell Line , Humans , Mice , Transcription Factors/metabolism
5.
Clin Transl Oncol ; 23(11): 2253-2268, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34023970

ABSTRACT

Glutamine metabolism is one of the hallmarks of cancers which is described as an essential role in serving as a major energy and building blocks supply to cell proliferation in cancer cells. Many malignant tumor cells always display glutamine addiction. The "kidney-type" glutaminase (GLS1) is a metabolism enzyme which plays a significant part in glutaminolysis. Interestingly, GLS1 is often overexpressed in highly proliferative cancer cells to fulfill enhanced glutamine demand. So far, GLS1 has been proved to be a significant target during the carcinogenesis process, and emerging evidence reveals that its inhibitors could provide a benefit strategy for cancer therapy. Herein, we summarize the prognostic value of GLS1 in multiple cancer type and its related regulatory factors which are associated with antitumor activity. Moreover, this review article highlights the remarkable reform of discovery and development for GLS1 inhibitors. On the basis of case studies, our perspectives for targeting GLS1 and development of GLS1 antagonist are discussed in the final part.


Subject(s)
Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Glutamine/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Apoptosis/physiology , Benzophenanthridines/pharmacology , Cell Proliferation/physiology , Diazooxonorleucine/metabolism , Disease Progression , Drug Resistance, Neoplasm , Genes, myc/physiology , Humans , MicroRNAs/physiology , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/mortality , Oxidation-Reduction , Phosphates/metabolism , Prognosis , Retinoblastoma Protein/metabolism , Sulfides/pharmacology , Thiadiazoles/pharmacology
6.
Methods Mol Biol ; 2318: 21-43, 2021.
Article in English | MEDLINE | ID: mdl-34019285

ABSTRACT

The C-terminal region of the c-MYC transcription factor consists of approximately 100 amino acids that in its native state does not adopt a stable structure. When this region binds to the obligatory partner MAX via a coupled folding-and-binding mechanism, it forms a basic-helix-loop-helix-leucine zipper (bHLHZip) heterodimeric complex. The C-terminal region of MYC is the target for numerous drug discovery programs for direct MYC inhibition via blocking the dimerization event and/or binding to DNA, and a proper understanding of the partially folded, dynamic nature of the heterodimeric complex is essential to these efforts. The bHLHZip motif also drives protein-protein interactions with cofactors that are crucial for both transcriptional repression and activation of MYC target genes. Targeting these interactions could potentially provide a means of developing alternative approaches to halt MYC functions; however, the molecular mechanism of these regulatory interactions is poorly understood. Herein we provide methods to produce high-quality human c-MYC C-terminal by itself and in complex MAX, and how to study them using Nuclear Magnetic Resonance spectroscopy and X-ray crystallography. Our protein expression and purification protocols have already been used to study interactions with cofactors.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/isolation & purification , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/isolation & purification , Amino Acid Sequence/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Crystallography, X-Ray/methods , DNA/chemistry , DNA/genetics , Dimerization , Genes, myc/genetics , Genes, myc/physiology , Helix-Loop-Helix Motifs/genetics , Helix-Loop-Helix Motifs/physiology , Humans , Leucine Zippers/genetics , Leucine Zippers/physiology , Magnetic Resonance Spectroscopy/methods , Protein Binding , Protein Domains/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism
7.
Methods Mol Biol ; 2318: 45-67, 2021.
Article in English | MEDLINE | ID: mdl-34019286

ABSTRACT

By identifying MYC protein-protein interactors, we aim to gain a deeper mechanistic understanding of MYC as a regulator of gene transcription and potent oncoprotein. This information can then be used to devise strategies for disrupting critical MYC protein-protein interactions to inhibit MYC-driven tumorigenesis. In this chapter, we discuss four techniques to identify and validate MYC-interacting partners. First, we highlight BioID, a powerful discovery method used to identify high-confidence proximal interactors in living cells. We also discuss bioinformatic prioritization strategies for the BioID-derived MYC-proximal complexes. Next, we discuss how protein interactions can be validated using techniques such as in vivo-in vitro pull-down assays and the proximity ligation assay (PLA). We conclude with an overview of biolayer interferometry (BLI), a quantitative method used to characterize direct interactions between two proteins in vitro. Overall, we highlight the principles of each assay and provide methodology necessary to conduct these experiments and adapt them to the study of interactors of additional proteins of interest.


Subject(s)
Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/isolation & purification , Amino Acid Sequence/genetics , Binding Sites , Computational Biology/methods , DNA/chemistry , DNA/genetics , Dimerization , Genes, myc/genetics , Genes, myc/physiology , Humans , Protein Binding , Protein Domains , Protein Interaction Mapping/methods , Proto-Oncogene Proteins c-myc/metabolism
8.
Methods Mol Biol ; 2318: 69-85, 2021.
Article in English | MEDLINE | ID: mdl-34019287

ABSTRACT

Detection of post-translational modifications in c-Myc is an invaluable tool in assessing Myc status, particularly in cancer. However, it can be challenging to detect these modifications. The evaluation of phosphorylation status of c-Myc can also be challenging with the current commercially available phosphorylation sensitive antibodies. Here, we describe protocols for the immunoprecipitation of endogenous c-Myc to probe for phosphorylation status, as well as the detection of ubiquitination and SUMOylation on c-Myc. We will also discuss the challenges of detecting phosphorylated c-Myc in formalin-fixed paraffin-embedded tissues by immunofluorescence and describe a protocol using a new rat monoclonal antibody we have generated suitable for this purpose.


Subject(s)
Immunoprecipitation/methods , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins c-myc/genetics , Fluorescent Antibody Technique , Genes, myc/genetics , Genes, myc/physiology , Humans , Phosphorylation , Protein Processing, Post-Translational/physiology , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sumoylation , Ubiquitination
9.
Methods Mol Biol ; 2318: 87-117, 2021.
Article in English | MEDLINE | ID: mdl-34019288

ABSTRACT

The MYC oncogene was originally identified as a transduced allele (v-myc) in the genome of the highly oncogenic avian retrovirus MC29. The protein product (MYC) of the cellular MYC (c-myc) protooncogene represents the key component of a transcription factor network controlling the expression of a large fraction of all human genes. MYC regulates fundamental cellular processes like growth control, metabolism, proliferation, differentiation, and apoptosis. Mutational deregulation of MYC, leading to increased levels of the MYC protein, is a frequent event in the etiology of human cancers. In this chapter, we describe cell systems and experimental strategies to quantify the oncogenic potential of MYC alleles, to test MYC inhibitors, and to monitor MYC-specific protein-protein interactions that are relevant for the cell transformation process. We also describe experimental procedures to study the evolutionary origin of MYC and to analyze structure, function, and regulation of the ancestral MYC proto-oncogenes.


Subject(s)
Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Alleles , Amino Acid Sequence , Animals , Carcinogenesis , Cell Transformation, Neoplastic/genetics , Evolution, Molecular , Genes, myc/genetics , Genes, myc/physiology , Humans , Mutation , Neoplasms/pathology , Oncogenes , Protein Interaction Mapping/methods , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics
10.
Methods Mol Biol ; 2318: 187-208, 2021.
Article in English | MEDLINE | ID: mdl-34019291

ABSTRACT

MYC is a transcription factor playing multiple functions both in physiological and pathological settings. Biochemical characterizations, combined with the analyses of MYC chromatin binding, have shown that its pleiotropic activity depends on the chromatin context and its protein-protein interactions with different cofactors. In order to determine the contribution of MYC in a certain biological condition, it would be relevant to analyze the concomitant binding of MYC and its associated proteins, in relationship to the chromatin environment. To this end, we here provide a simple method to parallel map the genome-wide binding of MYC-associated proteins, together with the chromatin profiling of multiple histone modifications. We detail the procedure to perform high-throughput ChIP-seq (HT-ChIP-seq) with a variety of biological samples. In addition, we describe simple bioinformatic steps to determine the distribution of MYC binding with respect to the chromatin context and the association of its cofactors. The described approach will permit the reproducible characterization of MYC activity in different biological contexts.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Epigenomics/methods , Proto-Oncogene Proteins c-myc/genetics , Chromatin/genetics , Chromatin Immunoprecipitation/methods , Computational Biology/methods , DNA/genetics , Epigenesis, Genetic/genetics , Genes, myc/genetics , Genes, myc/physiology , High-Throughput Nucleotide Sequencing/methods , Histone Code/genetics , Histones/metabolism , Humans , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins c-myc/metabolism , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
11.
Methods Mol Biol ; 2318: 231-239, 2021.
Article in English | MEDLINE | ID: mdl-34019293

ABSTRACT

The MYC gene regulates normal cell growth and is deregulated in many human cancers, contributing to tumor growth and progression. The MYC transcription factor activates RNA polymerases I, II, and III target genes that are considered housekeeping genes. These target genes are largely involved in ribosome biogenesis, fatty acid, protein and nucleotide synthesis, nutrient influx or metabolic waste efflux, glycolysis, and glutamine metabolism. MYC's function as a driver of cell growth has been revealed through RNA sequencing, genome-wide chromatin immunoprecipitation, proteomics, and importantly metabolomics, which is highlighted in this chapter.


Subject(s)
Carcinogenesis/metabolism , Metabolomics/methods , Proto-Oncogene Proteins c-myc/metabolism , Animals , Carcinogenesis/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , DNA/genetics , Genes, myc/genetics , Genes, myc/physiology , Glucose/metabolism , Glycolysis , Humans , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase I/metabolism , Ribosomes/metabolism
12.
Methods Mol Biol ; 2318: 241-254, 2021.
Article in English | MEDLINE | ID: mdl-34019294

ABSTRACT

Cellular senescence plays a role in several physiological processes including aging, embryonic development, tissue remodeling, and wound healing and is considered one of the main barriers against tumor development. Studies of normal and tumor cells both in culture and in vivo suggest that MYC plays an important role in regulating senescence, thereby contributing to tumor development. We have previously described different common methods to measure senescence in cell cultures and in tissues. Unfortunately, there is no unique marker that unambiguously defines a senescent state, and it is therefore necessary to combine measurements of several different markers in order to assure the correct identification of senescent cells. Here we describe protocols for simultaneous detection of multiple senescence markers in situ, a quantitative fluorogenic method to measure senescence-associated ß-galactosidase activity (SA-ß-gal), and a new method to detect senescent cells based on the Sudan Black B (SBB) analogue GL13, which is applicable to formalin-fixed paraffin-embedded tissues. The application of these methods in various systems will hopefully shed further light on the role of MYC in regulation of senescence, and how that impacts normal physiological processes as well as diseases and in particular cancer development.


Subject(s)
Cellular Senescence/physiology , Proto-Oncogene Proteins c-myc/metabolism , Staining and Labeling/methods , Aging , Azo Compounds/chemistry , Biomarkers , Cells, Cultured , Cellular Senescence/genetics , DNA/genetics , Fluorescent Dyes/chemistry , Genes, myc/genetics , Genes, myc/physiology , Humans , Naphthalenes/chemistry , Proto-Oncogene Proteins c-myc/genetics , beta-Galactosidase/analysis , beta-Galactosidase/metabolism
13.
Methods Mol Biol ; 2318: 255-266, 2021.
Article in English | MEDLINE | ID: mdl-34019295

ABSTRACT

A central component of Myc's role as a master coordinator of energy metabolism and biomass accumulation is its ability to increase the rate of protein synthesis, driving cell cycle progression, and proliferation. Importantly, Myc-induced alterations in both global and specific mRNA translation is a key determinant of Myc's oncogenic function. Herein, we provide five assays to enable researchers to measure global protein synthesis changes, to identify the translatome uniquely regulated by Myc and to investigate the mechanisms generating the tailored Myc translation network. Metabolic labeling of cells with 35S-containing methionine and cysteine in culture and O-propargyl-puromycin (OP-Puro) incorporation in vivo are presented as methods to measure the overall rate of global protein synthesis. Isolation of polysome-associated mRNAs followed by quantitative real-time PCR (qRT-PCR) and the toeprint assay enable the detection of altered translation of specific mRNAs and isoforms, and visualization of differential ribosomal engagement at start codons uniquely mediated by Myc activation, respectively. Finally, the translation initiation reporter assay is utilized to uncover the molecular mechanism mediating altered translation initiation of a specific mRNA. Together, the protocols detailed in this chapter can be used to illuminate how and to what degree Myc-dependent regulation of translation influences homeostatic cellular functions as well as tumorigenesis.


Subject(s)
Metabolomics/methods , Protein Biosynthesis/physiology , Proto-Oncogene Proteins c-myc/metabolism , Cell Transformation, Neoplastic/metabolism , DNA/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Genes, myc/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Neoplasms/genetics , Neoplasms/metabolism , Protein Biosynthesis/genetics , Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
14.
Methods Mol Biol ; 2318: 267-279, 2021.
Article in English | MEDLINE | ID: mdl-34019296

ABSTRACT

Cellular reprogramming is a process by which adult differentiated cells lose their identity and are converted into pluripotent stem cells, known as induced pluripotent stem (iPS) cells. This process can be achieved in vitro and in vivo and is relevant for many fields including regenerative medicine and cancer. Cellular reprogramming is commonly induced by the ectopic expression of a transcription factor cocktail composed by Oct4, Sox2, Klf4, and Myc (abbreviated as OSKM), and its efficiency and kinetics are strongly dependent on the presence of Myc. Here, we describe a versatile method to study reprogramming in vivo based on the use of adeno-associated viral (AAV) vectors, which allows the targeting of specific organs and cell types. This method can be used to test Myc mutations or genes that may replace Myc, or be combined with different Myc regulators. In vivo reprogramming can be scored by the presence of teratomas and the isolation of in vivo iPS, thereby providing a simple surrogate for the function of Myc in dedifferentiation and stemness. Our protocol can be divided into five steps: (1) intravenous inoculation of AAV vectors; (2) monitoring the animals until the appearance of teratomas; (3) analysis of teratomas; (4) histopathological analysis of mouse organs; and (5) isolation of in vivo-generated iPS cells from teratomas, blood, and bone marrow. The information obtained by this in vivo testing platform may provide relevant information on the role of Myc in tissue regeneration, stemness, and cancer.


Subject(s)
Cellular Reprogramming Techniques/methods , Cellular Reprogramming/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Cellular Reprogramming/physiology , DNA/genetics , Dependovirus/genetics , Fibroblasts/cytology , Genes, myc/genetics , Genes, myc/physiology , Genetic Engineering/methods , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/metabolism , Transduction, Genetic
15.
Methods Mol Biol ; 2318: 297-312, 2021.
Article in English | MEDLINE | ID: mdl-34019298

ABSTRACT

The tetracycline regulatory system provides a tractable strategy to interrogate the role of oncogenes in the initiation, maintenance, and regression of tumors through both spatial and temporal control of expression. This approach has several potential advantages over conventional methods to generate genetically engineered mouse models. First, continuous constitutive overexpression of an oncogene can be lethal to the host impeding further study. Second, constitutive overexpression fails to model adult onset of disease. Third, constitutive deletion does not permit, whereas conditional overexpression of an oncogene enables the study of the consequences of restoring expression of an oncogene back to endogenous levels. Fourth, the conditional activation of oncogenes enables examination of specific and/or developmental state-specific consequences.Hence, by allowing precise control of when and where a gene is expressed, the tetracycline regulatory system provides an ideal approach for the study of putative oncogenes in the initiation as well as the maintenance of tumorigenesis and the examination of the mechanisms of oncogene addiction. In this protocol, we describe the methods involved in the development of a conditional mouse model of MYC-induced T-cell acute lymphoblastic leukemia.


Subject(s)
Genetic Engineering/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Animals, Genetically Modified/genetics , Apoptosis , Carcinogenesis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Genes, myc/genetics , Genes, myc/physiology , Humans , Mice , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Synthesis Inhibitors , Proto-Oncogene Proteins c-myc/genetics , T-Lymphocytes/metabolism , Tetracycline/pharmacology
16.
Methods Mol Biol ; 2318: 281-295, 2021.
Article in English | MEDLINE | ID: mdl-34019297

ABSTRACT

Myc is deregulated in most-if not all-cancers, and it not only promotes tumor progression by inducing cell proliferation but is also responsible for tumor immune evasion. In a nutshell, MYC promotes the development of tumor-associated macrophages, impairs the cellular response to interferons, induces the expression of immunosuppressive molecules, and excludes tumor infiltrating lymphocytes (TILs) from the tumor site. Based on the insights into the role of MYC in promoting and regulating immune evasion by cancer cells, it is of special interest to study the different immune cell populations infiltrating the tumors. MYC inhibition has emerged as a potential new strategy for the treatment of cancer, directly inhibiting tumor progression while also counteracting the immunosuppressive tumor microenvironment, allowing an optimal anti-tumor immune response. Hence, this chapter describes a flow cytometry-based method to study the different immune cell subsets infiltrating the tumor by combining surface, cytoplasmic, and nuclear multicolor protein stainings.


Subject(s)
Flow Cytometry/methods , Proto-Oncogene Proteins c-myc/metabolism , Tumor Microenvironment/immunology , Cell Proliferation , DNA/genetics , Genes, myc/genetics , Genes, myc/physiology , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/genetics , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-myc/genetics , Tumor Microenvironment/physiology
17.
Methods Mol Biol ; 2318: 313-320, 2021.
Article in English | MEDLINE | ID: mdl-34019299

ABSTRACT

In situ hybridization (ISH) allows evaluation of genetic abnormalities, such as changes in chromosome number, chromosome translocations, or gene amplifications, by hybridization of tagged DNA (or RNA) probes with complementary DNA (or RNA) sequences in interphase nuclei of target tissue. However, chromogenic in situ hybridization (CISH) is also applicable to formalin-fixed, paraffin-embedded (FFPE ) tissues, besides metaphase chromosome spreads. CISH is similar to fluorescent in situ hybridization (FISH) regarding pretreatments and hybridization protocols but differs in the way of visualization. Indeed, CISH signal detection is similar to that used in immunohistochemistry, making use of a peroxidase-based chromogenic reaction instead of fluorescent dyes. In particular, tagged DNA probes are indirectly detected using an enzyme-conjugated antibody targeting the tags. The enzymatic reaction of the chromogenic substrate leads to the formation of strong permanent brown signals that can be visualized by bright-field microscopy at 40× magnification. The advantage of CISH is that it allows the simultaneous observation of gene amplification and tissue morphology, and the slides can be stored for a long time.


Subject(s)
In Situ Hybridization, Fluorescence/methods , In Situ Hybridization/methods , Proto-Oncogene Proteins c-myc/immunology , Chromogenic Compounds/chemistry , DNA/genetics , DNA Probes , Gene Amplification , Genes, myc/genetics , Genes, myc/physiology , Humans , Immunohistochemistry/methods , Neoplasms , Paraffin Embedding/methods , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Translocation, Genetic
18.
Methods Mol Biol ; 2318: 321-336, 2021.
Article in English | MEDLINE | ID: mdl-34019300

ABSTRACT

Clinical tumor specimens are routinely formalin-fixed and paraffin-embedded (FFPE) in Pathology departments worldwide. FFPE blocks are convenient, long-term stable, and easy to archive and manipulate. However, nucleic acids extracted from FFPE tissues generally show a high degree of fragmentation as well as chemical modifications, mainly due to the fixation process. Methods to determine copy number alterations (CNAs) from FFPE clinical samples have proven challenging, in the fact that they are low-plex, only able to profile single genes or gene clusters (such as in situ hybridization-based methods), and/or show a low degree of robustness with partially degraded samples (array-based, NGS-based) as well as being time-consuming, costly, and with limitations in resolution. The NanoString nCounter® System is a medium-plex, extremely FFPE-robust system, that overcomes several of the frequent issues when dealing with clinical samples. The technique is based on hybridization of molecular barcoded probes directly to FFPE-derived DNA, followed by single molecule imaging to detect hundreds of unique molecules in a single reaction without any amplification steps that might introduce undesired biases. Here we describe nCounter v2 Cancer Copy Number Assay, a robust and highly reproducible method for detecting the copy number status of 87 genes commonly amplified or deleted in cancer, including the MYC proto-oncogene.


Subject(s)
DNA Copy Number Variations , Nucleic Acid Hybridization/methods , Proto-Oncogene Proteins c-myc/genetics , DNA/genetics , DNA Probes , Formaldehyde/chemistry , Genes, myc/genetics , Genes, myc/physiology , Humans , In Situ Hybridization, Fluorescence/methods , Neoplasms/genetics , Paraffin Embedding/methods , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , Tissue Fixation/methods
19.
Methods Mol Biol ; 2318: 337-346, 2021.
Article in English | MEDLINE | ID: mdl-34019301

ABSTRACT

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of the Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Subject(s)
Drug Screening Assays, Antitumor/methods , Genes, myc/drug effects , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Cell Line, Tumor , Genes, myc/genetics , Genes, myc/physiology , Humans , Oncogene Proteins/drug effects , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Small Molecule Libraries/pharmacology , Transcription Factors/drug effects , Transcription Factors/metabolism
20.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33653688

ABSTRACT

The huge cadre of genes regulated by Myc has obstructed the identification of critical effectors that are essential for Myc-driven tumorigenesis. Here, we describe how only the lack of the receptor Fzd9, previously identified as a Myc transcriptional target, impairs sustained tumor expansion and ß-cell dedifferentiation in a mouse model of Myc-driven insulinoma, allows pancreatic islets to maintain their physiological structure and affects Myc-related global gene expression. Importantly, Wnt signaling inhibition in Fzd9-competent mice largely recapitulates the suppression of proliferation caused by Fzd9 deficiency upon Myc activation. Together, our results indicate that the Wnt signaling receptor Fzd9 is essential for Myc-induced tumorigenesis in pancreatic islets.


Subject(s)
Adenoma, Islet Cell/physiopathology , Carcinogenesis/metabolism , Frizzled Receptors/metabolism , Adenoma, Islet Cell/metabolism , Animals , Cell Movement , Cell Proliferation , Female , Frizzled Receptors/genetics , Frizzled Receptors/physiology , Genes, myc/genetics , Genes, myc/physiology , Islets of Langerhans/metabolism , Male , Mice , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...