Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Cancer Discov ; 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058094

ABSTRACT

The Hippo signaling pathway is commonly dysregulated in human cancer, which leads to a powerful tumor dependency on the YAP/TAZ transcriptional coactivators. Here, we used paralog co-targeting CRISPR screens to identify the kinases MARK2/3 as absolute catalytic requirements for YAP/TAZ function in diverse carcinoma and sarcoma contexts. Underlying this observation is direct MARK2/3-dependent phosphorylation of NF2 and YAP/TAZ, which effectively reverses the tumor suppressive activity of the Hippo module kinases LATS1/2. To simulate targeting of MARK2/3, we adapted the CagA protein from H. pylori as a catalytic inhibitor of MARK2/3, which we show can regress established tumors in vivo. Together, these findings reveal MARK2/3 as powerful co-dependencies of YAP/TAZ in human cancer; targets that may allow for pharmacology that restores Hippo pathway-mediated tumor suppression.

2.
Gut ; 73(6): 941-954, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38262672

ABSTRACT

OBJECTIVE: The optimal therapeutic response in cancer patients is highly dependent upon the differentiation state of their tumours. Pancreatic ductal adenocarcinoma (PDA) is a lethal cancer that harbours distinct phenotypic subtypes with preferential sensitivities to standard therapies. This study aimed to investigate intratumour heterogeneity and plasticity of cancer cell states in PDA in order to reveal cell state-specific regulators. DESIGN: We analysed single-cell expression profiling of mouse PDAs, revealing intratumour heterogeneity and cell plasticity and identified pathways activated in the different cell states. We performed comparative analysis of murine and human expression states and confirmed their phenotypic diversity in specimens by immunolabeling. We assessed the function of phenotypic regulators using mouse models of PDA, organoids, cell lines and orthotopically grafted tumour models. RESULTS: Our expression analysis and immunolabeling analysis show that a mucus production programme regulated by the transcription factor SPDEF is highly active in precancerous lesions and the classical subtype of PDA - the most common differentiation state. SPDEF maintains the classical differentiation and supports PDA transformation in vivo. The SPDEF tumour-promoting function is mediated by its target genes AGR2 and ERN2/IRE1ß that regulate mucus production, and inactivation of the SPDEF programme impairs tumour growth and facilitates subtype interconversion from classical towards basal-like differentiation. CONCLUSIONS: Our findings expand our understanding of the transcriptional programmes active in precancerous lesions and PDAs of classical differentiation, determine the regulators of mucus production as specific vulnerabilities in these cell states and reveal phenotype switching as a response mechanism to inactivation of differentiation states determinants.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Mice , Humans , Mucus/metabolism , Mucoproteins/metabolism , Mucoproteins/genetics , Cell Line, Tumor , Cell Differentiation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/metabolism , Proteins/genetics , Organoids/pathology , Organoids/metabolism , Cell Plasticity , Gene Expression Regulation, Neoplastic , Disease Models, Animal , Oncogene Proteins
3.
Adv Sci (Weinh) ; 11(6): e2308537, 2024 02.
Article in English | MEDLINE | ID: mdl-38110836

ABSTRACT

Engrailed-1 (EN1) is a critical homeodomain transcription factor (TF) required for neuronal survival, and EN1 expression has been shown to promote aggressive forms of triple negative breast cancer. Here, it is reported that EN1 is aberrantly expressed in a subset of pancreatic ductal adenocarcinoma (PDA) patients with poor outcomes. EN1 predominantly repressed its target genes through direct binding to gene enhancers and promoters, implicating roles in the activation of MAPK pathways and the acquisition of mesenchymal cell properties. Gain- and loss-of-function experiments demonstrated that EN1 promoted PDA transformation and metastasis in vitro and in vivo. The findings nominate the targeting of EN1 and downstream pathways in aggressive PDA.


Subject(s)
Carcinoma, Pancreatic Ductal , Homeodomain Proteins , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation , Pancreatic Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
4.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32633781

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, and new therapies are needed. Altered metabolism is a cancer vulnerability, and several metabolic pathways have been shown to promote PDAC. However, the changes in cholesterol metabolism and their role during PDAC progression remain largely unknown. Here we used organoid and mouse models to determine the drivers of altered cholesterol metabolism in PDAC and the consequences of its disruption on tumor progression. We identified sterol O-acyltransferase 1 (SOAT1) as a key player in sustaining the mevalonate pathway by converting cholesterol to inert cholesterol esters, thereby preventing the negative feedback elicited by unesterified cholesterol. Genetic targeting of Soat1 impairs cell proliferation in vitro and tumor progression in vivo and reveals a mevalonate pathway dependency in p53 mutant PDAC cells that have undergone p53 loss of heterozygosity (LOH). In contrast, pancreatic organoids lacking p53 mutation and p53 LOH are insensitive to SOAT1 loss, indicating a potential therapeutic window for inhibiting SOAT1 in PDAC.


Subject(s)
Mevalonic Acid/metabolism , Pancreatic Neoplasms/enzymology , Sterol O-Acyltransferase/metabolism , Animals , Cell Line, Tumor , Cholesterol/metabolism , Disease Progression , Humans , Loss of Heterozygosity/genetics , Mice, Inbred C57BL , Models, Biological , Pancreatic Neoplasms/pathology , Sterol O-Acyltransferase/deficiency , Tumor Suppressor Protein p53/metabolism
5.
Cell Rep ; 29(5): 1287-1298.e6, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665640

ABSTRACT

Glutamine is thought to play an important role in cancer cells by being deaminated via glutaminolysis to α-ketoglutarate (aKG) to fuel the tricarboxylic acid (TCA) cycle. Supporting this notion, aKG supplementation can restore growth/survival of glutamine-deprived cells. However, pancreatic cancers are often poorly vascularized and limited in glutamine supply, in alignment with recent concerns on the significance of glutaminolysis in pancreatic cancer. Here, we show that aKG-mediated rescue of glutamine-deprived pancreatic ductal carcinoma (PDAC) cells requires glutamate ammonia ligase (GLUL), the enzyme responsible for de novo glutamine synthesis. GLUL-deficient PDAC cells are capable of the TCA cycle but defective in aKG-coupled glutamine biosynthesis and subsequent nitrogen anabolic processes. Importantly, GLUL expression is elevated in pancreatic cancer patient samples and in mouse PDAC models. GLUL ablation suppresses the development of KrasG12D-driven murine PDAC. Therefore, GLUL-mediated glutamine biosynthesis couples the TCA cycle with nitrogen anabolism and plays a critical role in PDAC.


Subject(s)
Carbon/metabolism , Glutamine/metabolism , Nitrogen/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Deletion , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/metabolism , Humans , Ketoglutaric Acids/metabolism , Male , Mice, Inbred C57BL , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology
6.
Antioxid Redox Signal ; 29(17): 1727-1745, 2018 12 10.
Article in English | MEDLINE | ID: mdl-28899199

ABSTRACT

SIGNIFICANCE: Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor that coordinates the basal and stress-inducible activation of a vast array of cytoprotective genes. Understanding the regulation of Nrf2 activity and downstream pathways has major implications for human health. Recent Advances: Nrf2 regulates the transcription of components of the glutathione and thioredoxin antioxidant systems, as well as enzymes involved in phase I and phase II detoxification of exogenous and endogenous products, NADPH regeneration, and heme metabolism. It therefore represents a crucial regulator of the cellular defense mechanisms against xenobiotic and oxidative stress. In addition to antioxidant responses, Nrf2 is involved in other cellular processes, such as autophagy, intermediary metabolism, stem cell quiescence, and unfolded protein response. Given the wide range of processes that Nrf2 controls, its activity is tightly regulated at multiple levels. Here, we review the different modes of regulation of Nrf2 activity and the current knowledge of Nrf2-mediated transcriptional control. CRITICAL ISSUES: It is now clear that Nrf2 lies at the center of a complex regulatory network. A full comprehension of the Nrf2 program will require an integrated consideration of all the different factors determining Nrf2 activity. FUTURE DIRECTIONS: Additional computational and experimental studies are needed to obtain a more dynamic global view of Nrf2-mediated gene regulation. In particular, studies comparing how the Nrf2-dependent network changes from a physiological to a pathological condition can provide insight into mechanisms of disease and instruct new treatment strategies.


Subject(s)
Gene Expression Regulation , NF-E2-Related Factor 2/metabolism , Transcription, Genetic , Animals , Humans , Oxidative Stress/genetics
7.
Nat Cell Biol ; 18(8): 897-909, 2016 08.
Article in English | MEDLINE | ID: mdl-27347849

ABSTRACT

In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53-proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP-microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53.


Subject(s)
Mutant Proteins/drug effects , Mutation, Missense/drug effects , Proteasome Endopeptidase Complex/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Humans , Mice , MicroRNAs/genetics , Mutant Proteins/genetics , Mutation, Missense/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteome/drug effects , Quinuclidines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
8.
Genom Data ; 7: 29-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26981355

ABSTRACT

The transcriptional programs activated by p53 in B cells in vivo following exposure to ionizing radiation were studied through the integrated analysis of various types of next-generation sequencing data: genome-wide profiling of p53 binding sites, mapping of histone marks and open chromatin regions and quantification of gene expression. Moreover, the binding of p53 was associated to a series of specific motifs on the DNA, which were directly inferred from the data. Here, we describe in detail the computational analysis of the datasets associated with our study (Tonelli et al., Oncotarget 6 (2015), 24611-26), deposited in the GEO archive (accession code GSE71180), and we provide the R scripts needed to generated the figures of the paper.

9.
Oncotarget ; 6(28): 24611-26, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26372730

ABSTRACT

The tumor suppressor p53 is a transcription factor that coordinates the cellular response to DNA damage. Here we provide an integrated analysis of p53 genomic occupancy and p53-dependent gene regulation in the splenic B and non-B cell compartments of mice exposed to whole-body ionizing radiation, providing insight into general principles of p53 activity in vivo. In unstressed conditions, p53 bound few genomic targets; induction of p53 by ionizing radiation increased the number of p53 bound sites, leading to highly overlapping profiles in the different cell types. Comparison of these profiles with chromatin features in unstressed B cells revealed that, upon activation, p53 localized at active promoters, distal enhancers, and a smaller set of unmarked distal regions. At promoters, recognition of the canonical p53 motif as well as binding strength were associated with p53-dependent transcriptional activation, but not repression, indicating that the latter was most likely indirect. p53-activated targets constituted the core of a cell type-independent response, superimposed onto a cell type-specific program. Core response genes included most of the known p53-regulated genes, as well as many new ones. Our data represent a unique characterization of the p53-regulated response to ionizing radiation in vivo.


Subject(s)
B-Lymphocytes/radiation effects , DNA Damage , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/metabolism , Whole-Body Irradiation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Binding Sites , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Genome-Wide Association Study , Genotype , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
10.
Nature ; 511(7510): 488-492, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043028

ABSTRACT

The c-myc proto-oncogene product, Myc, is a transcription factor that binds thousands of genomic loci. Recent work suggested that rather than up- and downregulating selected groups of genes, Myc targets all active promoters and enhancers in the genome (a phenomenon termed 'invasion') and acts as a general amplifier of transcription. However, the available data did not readily discriminate between direct and indirect effects of Myc on RNA biogenesis. We addressed this issue with genome-wide chromatin immunoprecipitation and RNA expression profiles during B-cell lymphomagenesis in mice, in cultured B cells and fibroblasts. Consistent with long-standing observations, we detected general increases in total RNA or messenger RNA copies per cell (hereby termed 'amplification') when comparing actively proliferating cells with control quiescent cells: this was true whether cells were stimulated by mitogens (requiring endogenous Myc for a proliferative response) or by deregulated, oncogenic Myc activity. RNA amplification and promoter/enhancer invasion by Myc were separable phenomena that could occur without one another. Moreover, whether or not associated with RNA amplification, Myc drove the differential expression of distinct subsets of target genes. Hence, although having the potential to interact with all active or poised regulatory elements in the genome, Myc does not directly act as a global transcriptional amplifier. Instead, our results indicate that Myc activates and represses transcription of discrete gene sets, leading to changes in cellular state that can in turn feed back on global RNA production and turnover.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Disease Progression , Down-Regulation/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genome/genetics , Lymphoma, B-Cell/metabolism , Male , Mice , Mitogens/pharmacology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL