Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci Alliance ; 3(10)2020 10.
Article in English | MEDLINE | ID: mdl-32843533

ABSTRACT

The activated B-cell (ABC) to plasmablast transition encompasses the cusp of antibody-secreting cell (ASC) differentiation. We explore this transition with integrated analysis in human cells, focusing on changes that follow removal from CD40-mediated signals. Within hours of input signal loss, cell growth programs shift toward enhanced proliferation, accompanied by ER-stress response, and up-regulation of ASC features. Clustering of genomic occupancy for IRF4, BLIMP1, XBP1, and CTCF with histone marks identifies a dichotomy: XBP1 and IRF4 link to induced but not repressed gene modules in plasmablasts, whereas BLIMP1 links to modules of ABC genes that are repressed, but not to activated genes. Between ABC and plasmablast states, IRF4 shifts away from AP1/IRF composite elements while maintaining occupancy at IRF and ETS/IRF elements. This parallels the loss of BATF expression, which is identified as a potential BLIMP1 target. In plasmablasts, IRF4 acquires an association with CTCF, a feature maintained in plasma cell myeloma lines. Thus, shifting occupancy links IRF4 to both ABC and ASC gene expression, whereas BLIMP1 occupancy links to repression of the activation state.


Subject(s)
B-Lymphocytes/cytology , Gene Regulatory Networks/genetics , Plasma Cells/cytology , Adult , B-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Female , Humans , Interferon Regulatory Factors/metabolism , Lymphocyte Activation/physiology , Male , Positive Regulatory Domain I-Binding Factor 1/metabolism , Signal Transduction , Transcriptional Activation/physiology , X-Box Binding Protein 1/metabolism
3.
Nature ; 523(7560): 313-7, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26153859

ABSTRACT

Progesterone receptor (PR) expression is used as a biomarker of oestrogen receptor-α (ERα) function and breast cancer prognosis. Here we show that PR is not merely an ERα-induced gene target, but is also an ERα-associated protein that modulates its behaviour. In the presence of agonist ligands, PR associates with ERα to direct ERα chromatin binding events within breast cancer cells, resulting in a unique gene expression programme that is associated with good clinical outcome. Progesterone inhibited oestrogen-mediated growth of ERα(+) cell line xenografts and primary ERα(+) breast tumour explants, and had increased anti-proliferative effects when coupled with an ERα antagonist. Copy number loss of PGR, the gene coding for PR, is a common feature in ERα(+) breast cancers, explaining lower PR levels in a subset of cases. Our findings indicate that PR functions as a molecular rheostat to control ERα chromatin binding and transcriptional activity, which has important implications for prognosis and therapeutic interventions.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , DNA Copy Number Variations/genetics , Disease Progression , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Progesterone/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Receptors, Progesterone/genetics , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
4.
Cell Rep ; 3(2): 342-9, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23403292

ABSTRACT

Estrogen receptor-α (ER) is the driving transcription factor in most breast cancers, and its associated proteins can influence drug response, but direct methods for identifying interacting proteins have been limited. We purified endogenous ER using an approach termed RIME (rapid immunoprecipitation mass spectrometry of endogenous proteins) and discovered the interactome under agonist- and antagonist-liganded conditions in breast cancer cells, revealing transcriptional networks in breast cancer. The most estrogen-enriched ER interactor is GREB1, a potential clinical biomarker with no known function. GREB1 is shown to be a chromatin-bound ER coactivator and is essential for ER-mediated transcription, because it stabilizes interactions between ER and additional cofactors. We show a GREB1-ER interaction in three xenograft tumors, and using a directed protein-protein approach, we find GREB1-ER interactions in half of ER(+) primary breast cancers. This finding is supported by histological expression of GREB1, which shows that GREB1 is expressed in half of ER(+) cancers, and predicts good clinical outcome. These findings reveal an unexpected role for GREB1 as an estrogen-specific ER cofactor that is expressed in drug-sensitive contexts.


Subject(s)
Estrogen Receptor alpha/metabolism , Neoplasm Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein Interaction Maps , RNA Interference , RNA, Small Interfering/metabolism , Transcription, Genetic , Transplantation, Heterologous
5.
Nat Commun ; 3: 1055, 2012.
Article in English | MEDLINE | ID: mdl-22968701

ABSTRACT

The role of transforming growth factor-beta (TGFß) in the progression of different molecular subtypes of breast cancer has not been clarified. Here we show that TGFß increases breast tumour-initiating cell (BTIC) numbers but only in claudin(low) breast cancer cell lines by orchestrating a specific gene signature enriched in stem cell processes that predicts worse clinical outcome in breast cancer patients. NEDD9, a member of the Cas family of integrin scaffold proteins, is necessary to mediate these TGFß-specific effects through a positive feedback loop that integrates TGFß/Smad and Rho-actin-SRF-dependent signals. In normal human mammary epithelium, TGFß induces progenitor activity only in the basal/stem cell compartment, where claudin(low) cancers are presumed to arise. These data show opposing responses to TGFß in both breast malignant cell subtypes and normal mammary epithelial cell subpopulations and suggest therapeutic strategies for a subset of human breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Claudins/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chromatin Immunoprecipitation , Claudins/genetics , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mice , Neoplastic Stem Cells/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Cells, Cultured
6.
Proc Natl Acad Sci U S A ; 109(23): 9077-82, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22603795

ABSTRACT

Barrett esophagus (BE) is a human metaplastic condition that is the only known precursor to esophageal adenocarcinoma. BE is characterized by a posterior intestinal-like phenotype in an anterior organ and therefore it is reminiscent of homeotic transformations, which can occur in transgenic animal models during embryonic development as a consequence of mutations in HOX genes. In humans, acquired deregulation of HOX genes during adulthood has been linked to carcinogenesis; however, little is known about their role in the pathogenesis of premalignant conditions. We hypothesized that HOX genes may be implicated in the development of BE. We demonstrated that three midcluster HOXB genes (HOXB5, HOXB6, and HOXB7) are overexpressed in BE, compared with the anatomically adjacent normal esophagus and gastric cardia. The midcluster HOXB gene signature in BE is identical to that seen in normal colonic epithelium. Ectopic expression of these three genes in normal squamous esophageal cells in vitro induces markers of intestinal differentiation, such as KRT20, MUC2, and VILLIN. In BE-associated adenocarcinoma, the activation midcluster HOXB gene is associated with loss of H3K27me3 and gain of AcH3, compared with normal esophagus. These changes in histone posttranslational modifications correlate with specific chromatin decompaction at the HOXB locus. We suggest that epigenetically regulated alterations of HOX gene expression can trigger changes in the transcriptional program of adult esophageal cells, with implications for the early stages of carcinogenesis.


Subject(s)
Adenocarcinoma/metabolism , Barrett Esophagus/metabolism , Epigenesis, Genetic/genetics , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, Homeobox/genetics , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Adult , Barrett Esophagus/complications , Barrett Esophagus/genetics , Blotting, Western , Chromatin Immunoprecipitation , Colon/cytology , Colon/metabolism , DNA Primers/genetics , Esophageal Neoplasms/etiology , Esophageal Neoplasms/genetics , Esophagus/cytology , Esophagus/metabolism , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Microarray Analysis , Polymerase Chain Reaction
7.
Carcinogenesis ; 31(9): 1669-75, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584750

ABSTRACT

Nitric oxide (NO) has been shown to induce double strand DNA breaks in Barrett's oesophagus (BO) and in other cancers has a role in invasion. The specific aims of this study were to investigate whether NO can induce invasion in cells representative of different stages of Barrett's progression and to determine possible underlying mechanisms. Physiological concentrations of NO that mimic luminal production of NO from dietary sources enhanced invasion in cell lines from high-grade dysplasia (GihTERT) and oesophageal adenocarcinoma (FLO) but not a non-dysplastic Barrett's cell line (QhTERT). Real-time reverse transcription-polymerase chain reaction revealed that NO induced expression of matrix metalloproteinase (MMP)-1, -3, -7, -9 and -10 and tissue inhibitor of metalloproteinase (TIMP)-1, -2 and -3 in these cell lines. Furthermore, ex vivo treatment of Barrett's biopsy samples with NO induced increases in MMP-1 and TIMP-1 expression, suggesting that NO enhances invasion through deregulating MMP and TIMP expression in epithelial cells. In keeping with these findings, microarray analysis and immunohistochemistry performed on biopsy samples showed enhanced expression of MMP-1, -3, -7 and -10 and TIMP-1 in the progression from non-dysplastic BO to adenocarcinoma, although this could not be directly attributed to the effect of NO. Thus, NO may play a role in Barrett's carcinogenesis through deregulating MMP and TIMP expression to enhance invasive potential.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Free Radical Scavengers/pharmacology , Nitric Oxide/pharmacology , Precancerous Conditions/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Disease Progression , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Gene Expression Profiling , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/drug therapy , Precancerous Conditions/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Cells, Cultured
8.
Proc Natl Acad Sci U S A ; 107(5): 2177-82, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080664

ABSTRACT

The stromal compartment is increasingly recognized to play a role in cancer. However, its role in the transition from preinvasive to invasive disease is unknown. Most gastrointestinal tumors have clearly defined premalignant stages, and Barrett's esophagus (BE) is an ideal research model. Supervised clustering of gene expression profiles from microdissected stroma identified a gene signature that could distinguish between BE metaplasia, dysplasia, and esophageal adenocarcinoma (EAC). EAC patients overexpressing any of the five genes (TMEPAI, JMY, TSP1, FAPalpha, and BCL6) identified from this stromal signature had a significantly poorer outcome. Gene ontology analysis identified a strong inflammatory component in BE disease progression, and key pathways included cytokine-cytokine receptor interactions and TGF-beta. Increased protein levels of inflammatory-related genes significantly up-regulated in EAC compared with preinvasive stages were confirmed in the stroma of independent samples, and in vitro assays confirmed functional relevance of these genes. Gene set enrichment analysis of external datasets demonstrated that the stromal signature was also relevant in the preinvasive to invasive transition of the stomach, colon, and pancreas. These data implicate inflammatory pathways in the genesis of gastrointestinal tract cancers, which can affect prognosis.


Subject(s)
Digestive System Neoplasms/genetics , Digestive System Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/immunology , Barrett Esophagus/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cytokines/genetics , DNA-Binding Proteins/genetics , Digestive System Neoplasms/immunology , Endopeptidases , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Gelatinases/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Membrane Proteins/genetics , Metaplasia , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Nuclear Proteins/genetics , Oncogenes , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Prognosis , Proto-Oncogene Proteins c-bcl-6 , Receptors, Cytokine/genetics , Serine Endopeptidases/genetics , Stromal Cells/immunology , Stromal Cells/pathology , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...