Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Gastroenterology ; 152(6): 1507-1520.e15, 2017 05.
Article in English | MEDLINE | ID: mdl-28188746

ABSTRACT

BACKGROUND & AIMS: The ability of exocrine pancreatic cells to change the cellular phenotype is required for tissue regeneration upon injury, but also contributes to their malignant transformation and tumor progression. We investigated context-dependent signaling and transcription mechanisms that determine pancreatic cell fate decisions toward regeneration and malignancy. In particular, we studied the function and regulation of the inflammatory transcription factor nuclear factor of activated T cells 1 (NFATC1) in pancreatic cell plasticity and tissue adaptation. METHODS: We analyzed cell plasticity during pancreatic regeneration and transformation in mice with pancreas-specific expression of a constitutively active form of NFATC1, or depletion of enhancer of zeste 2 homologue 2 (EZH2), in the context of wild-type or constitutively activate Kras, respectively. Acute and chronic pancreatitis were induced by intraperitoneal injection of caerulein. EZH2-dependent regulation of NFATC1 expression was studied in mouse in human pancreatic tissue and cells by immunohistochemistry, immunoblotting, and quantitative reverse transcription polymerase chain reaction. We used genetic and pharmacologic approaches of EZH2 and NFATC1 inhibition to study the consequences of pathway disruption on pancreatic morphology and function. Epigenetic modifications on the NFATC1 gene were investigated by chromatin immunoprecipitation assays. RESULTS: NFATC1 was rapidly and transiently induced in early adaptation to acinar cell injury in human samples and in mice, where it promoted acinar cell transdifferentiation and blocked proliferation of metaplastic pancreatic cells. However, in late stages of regeneration, Nfatc1 was epigenetically silenced by EZH2-dependent histone methylation, to enable acinar cell redifferentiation and prevent organ atrophy and exocrine insufficiency. In contrast, oncogenic activation of KRAS signaling in pancreatic ductal adenocarcinoma cells reversed the EZH2-dependent effects on the NFATC1 gene and was required for EZH2-mediated transcriptional activation of NFATC1. CONCLUSIONS: In studies of human and mouse pancreatic cells and tissue, we identified context-specific epigenetic regulation of NFATc1 activity as an important mechanism of pancreatic cell plasticity. Inhibitors of EZH2 might therefore interfere with oncogenic activity of NFATC1 and be used in treatment of pancreatic ductal adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Plasticity/genetics , Cell Transformation, Neoplastic/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation , NFATC Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Regeneration/genetics , Acinar Cells/physiology , Animals , Carcinoma, Pancreatic Ductal/chemistry , Cell Proliferation/genetics , Cell Transdifferentiation/genetics , Ceruletide , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enhancer of Zeste Homolog 2 Protein/analysis , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Silencing , Histones/metabolism , Humans , Methylation , Mice , NFATC Transcription Factors/analysis , NFATC Transcription Factors/metabolism , Pancreas/physiology , Pancreatic Neoplasms/chemistry , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/physiopathology , Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/genetics , Transcription, Genetic
2.
Cells ; 10(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34943970

ABSTRACT

BACKGROUND: The Nuclear Factor of Activated T-cells 1 (NFATc1) transcription factor and the methyltransferase Enhancer of Zeste Homolog 2 (EZH2) significantly contribute to the aggressive phenotype of pancreatic ductal adenocarcinoma (PDAC). Herein, we aimed at dissecting the mechanistic background of their interplay in PDAC progression. METHODS: NFATc1 and EZH2 mRNA and protein expression and complex formation were determined in transgenic PDAC models and human PDAC specimens. NFATc1 binding on the Ezh2 gene and the consequences of perturbed NFATc1 expression on Ezh2 transcription were explored by Chromatin Immunoprecipitation (ChIP) and upon transgenic or siRNA-mediated interference with NFATc1 expression, respectively. Integrative analyses of RNA- and ChIP-seq data was performed to explore NFATc1-/EZH2-dependent gene signatures. RESULTS: NFATc1 targets the Ezh2 gene for transcriptional activation and biochemically interacts with the methyltransferase in murine and human PDAC. Surprisingly, our genome-wide binding and expression analyses do not link the protein complex to joint gene regulation. In contrast, our findings provide evidence for chromatin-independent functions of the NFATc1:EZH2 complex and reveal posttranslational EZH2 phosphorylation at serine 21 as a prerequisite for robust complex formation. CONCLUSION: Our findings disclose a previously unknown NFATc1-EZH2 axis operational in the pancreas and provide mechanistic insights into the conditions fostering NFATc1:EZH2 complex formation in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , NFATC Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/genetics , Chromatin/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Mice , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL