Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Nucleic Acids Res ; 44(17): 8153-64, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27257062

ABSTRACT

Forkhead box A1 (FOXA1) is an FKHD family protein that plays pioneering roles in lineage-specific enhancer activation and gene transcription. Through genome-wide location analyses, here we show that FOXA1 expression and occupancy are, in turn, required for the maintenance of these epigenetic signatures, namely DNA hypomethylation and histone 3 lysine 4 methylation. Mechanistically, this involves TET1, a 5-methylcytosine dioxygenase. We found that FOXA1 induces TET1 expression via direct binding to its cis-regulatory elements. Further, FOXA1 physically interacts with the TET1 protein through its CXXC domain. TET1 thus co-occupies FOXA1-dependent enhancers and mediates local DNA demethylation and concomitant histone 3 lysine 4 methylation, further potentiating FOXA1 recruitment. Consequently, FOXA1 binding events are markedly reduced following TET1 depletion. Together, our results suggest that FOXA1 is not only able to recognize but also remodel the epigenetic signatures at lineage-specific enhancers, which is mediated, at least in part, by a feed-forward regulatory loop between FOXA1 and TET1.


Subject(s)
Cell Lineage/genetics , Enhancer Elements, Genetic , Hepatocyte Nuclear Factor 3-alpha/metabolism , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Line, Tumor , Epigenesis, Genetic , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Mixed Function Oxygenases/genetics , Models, Biological , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Transcription, Genetic
2.
J Clin Invest ; 129(2): 569-582, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30511964

ABSTRACT

Prostate cancer (PC) progressed to castration resistance (CRPC) is a fatal disease. CRPC tumors develop resistance to new-generation antiandrogen enzalutamide through lineage plasticity, characterized by epithelial-mesenchymal transition (EMT) and a basal-like phenotype. FOXA1 is a transcription factor essential for epithelial lineage differentiation. Here, we demonstrate that FOXA1 loss leads to remarkable upregulation of transforming growth factor beta 3 (TGFB3), which encodes a ligand of the TGF-ß pathway. Mechanistically, this is due to genomic occupancy of FOXA1 on an upstream enhancer of the TGFB3 gene to directly inhibit its transcription. Functionally, FOXA1 downregulation induces TGF-ß signaling, EMT, and cell motility, which is effectively blocked by the TGF-ß receptor I inhibitor galunisertib (LY2157299). Tissue microarray analysis confirmed reduced levels of FOXA1 protein and a concordant increase in TGF-ß signaling, indicated by SMAD2 phosphorylation, in CRPC as compared with primary tumors. Importantly, combinatorial LY2157299 treatment sensitized PC cells to enzalutamide, leading to synergistic effects in inhibiting cell invasion in vitro and xenograft CRPC tumor growth and metastasis in vivo. Therefore, our study establishes FOXA1 as an important regulator of lineage plasticity mediated in part by TGF-ß signaling, and supports a novel therapeutic strategy to control lineage switching and potentially extend clinical response to antiandrogen therapies.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha , Neoplasm Proteins , Prostatic Neoplasms, Castration-Resistant , Pyrazoles/pharmacology , Quinolines/pharmacology , Signal Transduction , Transforming Growth Factor beta3 , Animals , Cell Line, Tumor , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-alpha/antagonists & inhibitors , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Male , Mice , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Xenograft Model Antitumor Assays
3.
Cancer Res ; 77(2): 412-422, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27815387

ABSTRACT

The lethal phenotype of castration-resistant prostate cancer (CRPC) is generally caused by augmented signaling from the androgen receptor (AR). Here, we report that the AR-repressed gene CCN3/NOV inhibits AR signaling and acts in a negative feedback loop to block AR function. Mechanistically, a cytoplasmic form of CCN3 interacted with the AR N-terminal domain to sequester AR in the cytoplasm of prostate cancer cells, thereby reducing AR transcriptional activity and inhibiting cell growth. However, constitutive repression of CCN3 by the Polycomb group protein EZH2 disrupted this negative feedback loop in both CRPC and enzalutamide-resistant prostate cancer cells. Notably, restoring CCN3 was sufficient to effectively reduce CPRC cell proliferation in vitro and to abolish xenograft tumor growth in vivo Taken together, our findings establish CCN3 as a pivotal regulator of AR signaling and prostate cancer progression and suggest a functional intersection between Polycomb and AR signaling in CRPC. Cancer Res; 77(2); 412-22. ©2016 AACR.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Feedback, Physiological/physiology , Heterografts , Humans , Immunoprecipitation , Male , Mice , Polymerase Chain Reaction , Prostatic Neoplasms, Castration-Resistant/metabolism
4.
Biol Psychiatry ; 77(3): 246-255, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25108803

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) exhibits numerous clinical and molecular features that are consistent with putative epigenetic misregulation. Despite growing interest in epigenetic studies of psychiatric diseases, the methodologies guiding such studies have not been well defined. METHODS: We performed DNA modification analysis in white blood cells from monozygotic twins discordant for MDD, in brain prefrontal cortex, and germline (sperm) samples from affected individuals and control subjects (total N = 304) using 8.1K CpG island microarrays and fine mapping. In addition to the traditional locus-by-locus comparisons, we explored the potential of new analytical approaches in epigenomic studies. RESULTS: In the microarray experiment, we detected a number of nominally significant DNA modification differences in MDD and validated selected targets using bisulfite pyrosequencing. Some MDD epigenetic changes, however, overlapped across brain, blood, and sperm more often than expected by chance. We also demonstrated that stratification for disease severity and age may increase the statistical power of epimutation detection. Finally, a series of new analytical approaches, such as DNA modification networks and machine-learning algorithms using binary and quantitative depression phenotypes, provided additional insights on the epigenetic contributions to MDD. CONCLUSIONS: Mapping epigenetic differences in MDD (and other psychiatric diseases) is a complex task. However, combining traditional and innovative analytical strategies may lead to identification of disease-specific etiopathogenic epimutations.


Subject(s)
Depressive Disorder, Major/genetics , Epigenesis, Genetic , Adolescent , Adult , Aged , CpG Islands , Female , Humans , Leukocytes , Male , Microarray Analysis , Middle Aged , Prefrontal Cortex , Spermatozoa , Twins, Monozygotic , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL