Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Oncol ; 18(6): 1510-1530, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459621

ABSTRACT

The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Nuclear Receptor Interacting Protein 1 , Transcription Factor HES-1 , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Nuclear Receptor Interacting Protein 1/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics
2.
Cancer Drug Resist ; 5(2): 401-414, 2022.
Article in English | MEDLINE | ID: mdl-35800380

ABSTRACT

Aim: The transcription factor RIP140 (receptor interacting protein of 140 kDa) is involved in intestinal tumorigenesis. It plays a role in the control of microsatellite instability (MSI), through the regulation of MSH2 and MSH6 gene expression. The aim of this study was to explore its effect on the expression of POLK, the gene encoding the specialized translesion synthesis (TLS) DNA polymerase κ known to perform accurate DNA synthesis at microsatellites. Methods: Different mouse models and engineered human colorectal cancer (CRC) cell lines were used to analyze by RT-qPCR, while Western blotting and luciferase assays were used to elucidate the role of RIP140 on POLK gene expression. Published DNA microarray datasets were reanalyzed. The in vitro sensitivity of CRC cells to methyl methane sulfonate and cisplatin was determined. Results: RIP140 positively regulates, at the transcriptional level, the expression of the POLK gene, and this effect involves, at least partly, the p53 tumor suppressor. In different cohorts of CRC biopsies (with or without MSI), a strong positive correlation was observed between RIP140 and POLK gene expression. In connection with its effect on POLK levels and the TLS function of this polymerase, the cellular response to methyl methane sulfonate was increased in cells lacking the Rip140 gene. Finally, the association of RIP140 expression with better overall survival of CRC patients was observed only when the corresponding tumors exhibited low levels of POLK, thus strengthening the functional link between the two genes in human CRC. Conclusion: The regulation of POLK gene expression by RIP140 could thus contribute to the maintenance of microsatellite stability, and more generally to the control of genome integrity.

3.
Cell Mol Life Sci ; 79(5): 270, 2022 May 03.
Article in English | MEDLINE | ID: mdl-35501580

ABSTRACT

Glycolysis is essential to support cancer cell proliferation, even in the presence of oxygen. The transcriptional co-regulator RIP140 represses the activity of transcription factors that drive cell proliferation and metabolism and plays a role in mammary tumorigenesis. Here we use cell proliferation and metabolic assays to demonstrate that RIP140-deficiency causes a glycolysis-dependent increase in breast tumor growth. We further demonstrate that RIP140 reduces the transcription of the glucose transporter GLUT3 gene, by inhibiting the transcriptional activity of hypoxia inducible factor HIF-2α in cooperation with p53. Interestingly, RIP140 expression was significantly associated with good prognosis only for breast cancer patients with tumors expressing low GLUT3, low HIF-2α and high p53, thus confirming the mechanism of RIP140 anti-tumor activity provided by our experimental data. Overall, our work establishes RIP140 as a critical modulator of the p53/HIF cross-talk to inhibit breast cancer cell glycolysis and proliferation.


Subject(s)
Breast Neoplasms , Tumor Suppressor Protein p53 , Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/genetics , Female , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/genetics , Humans , Hypoxia , Nuclear Receptor Interacting Protein 1 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Cancers (Basel) ; 13(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34503257

ABSTRACT

Microsatellite instability (MSI) is related to the alteration of mismatch repair (MMR) genes and plays a key role in colorectal cancer (CRC) pathogenesis. We previously reported that the transcription factor Nuclear Receptor Interacting Protein 1 (NRIP1) is involved in sporadic intestinal tumorigenesis. The aim of this study was to decipher its role in MSI CRC. By using different mouse models and engineered cell lines, we demonstrated that NRIP1 increased MSH2 and MSH6 MMR gene transcription and mRNA/protein levels. In human CRC cells, NRIP1 expression was associated with decreased MSI and the hypermutator phenotype, and with resistance to chemotherapy drugs. Using a cohort of 194 CRC patients, we detected in 22% of the cases a MSI-induced frameshift mutation in the NRIP1 coding sequence. This genetic alteration generates a truncated protein with a dominant negative activity that increased human CRC cell proliferation and impaired the regulation of MSH2 and MSH6 gene expression. Moreover, the NRIP1 mutant correlated with a decreased overall survival of patients with advanced CRC, especially when MLH1-deficient. By decreasing the expression of MSH2 and MSH6 gene expression, the NRIP1 variant may amplify MLH1-dependent CRC progression and behave as a new prognostic marker of advanced MSI CRC.

5.
Sci Rep ; 11(1): 7272, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33790340

ABSTRACT

Modular response analysis (MRA) is a widely used inference technique developed to uncover directions and strengths of connections in molecular networks under a steady-state condition by means of perturbation experiments. We devised several extensions of this methodology to search genomic data for new associations with a biological network inferred by MRA, to improve the predictive accuracy of MRA-inferred networks, and to estimate confidence intervals of MRA parameters from datasets with low numbers of replicates. The classical MRA computations and their extensions were implemented in a freely available R package called aiMeRA ( https://github.com/bioinfo-ircm/aiMeRA/ ). We illustrated the application of our package by assessing the crosstalk between estrogen and retinoic acid receptors, two nuclear receptors implicated in several hormone-driven cancers, such as breast cancer. Based on new data generated for this study, our analysis revealed potential cross-inhibition mediated by the shared corepressors NRIP1 and LCoR. We designed aiMeRA for non-specialists and to allow biologists to perform their own analyses.


Subject(s)
Algorithms , Breast Neoplasms , Gene Regulatory Networks , Neoplasm Proteins , Receptors, Retinoic Acid , Software , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism
6.
Int J Mol Sci ; 20(5)2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30813617

ABSTRACT

Increasing evidence implicates the aryl hydrocarbon receptor (AhR) as a possible regulator of mammary carcinogenesis. This study aims to clarify its prognostic impact in breast cancer (BC). Meta-analyses performed at the mRNA level demonstrated that the predictive value of AhR expression in BC depends on the lymph node (LN) status. AhR expression and sub-cellular location were then analyzed by immunohistochemistry in 302 primary BC samples. AhR was expressed in almost 90% of cases with a predominant nuclear location. Nuclear and cytoplasmic AhR levels were significantly correlated and associated with the expression of RIP140 (receptor-interacting protein of 140 kDa), an AhR transcriptional coregulator and target gene. Interestingly, total and nuclear AhR levels were only significantly correlated with short overall survival in node-negative patients. In this sub-group, total and nuclear AhR expression had an even stronger prognostic impact in patients with low RIP140-expressing tumors. Very interestingly, the total AhR prognostic value was also significant in luminal-like BCs and was an independent prognostic marker for LN-negative patients. Altogether, this study suggests that AhR is a marker of poor prognosis for patients with LN-negative luminal-like BCs, which warrants further evaluation.


Subject(s)
Breast Neoplasms/metabolism , Lymph Nodes/pathology , Receptors, Aryl Hydrocarbon/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Nuclear Receptor Interacting Protein 1/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/genetics , Survival Analysis
7.
Int J Mol Sci ; 20(2)2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30669416

ABSTRACT

The aim of this study was to investigate the expression of two nuclear receptor transcriptional coregulators, namely RIP140 (receptor-interacting protein of 140 kDa) and LCoR (ligand-dependent corepressor) in unifocal versus multifocal breast cancers. The expression of these two proteins was analyzed by immunohistochemistry in a matched-pair cohort of 21 unifocal and 21 multifocal breast tumors. The expression of the two estrogen receptors (ERα and ERß) was studied in parallel. RIP140 and LCoR levels appeared lower in unifocal tumors compared to multifocal samples (decreased of immune-reactive scores and reduced number of high expressing cells). In both tumor types, RIP140 and LCoR expression was correlated with each other and with expression of ERß. Very interestingly, the expression of RIP140, LCoR, and ERß was inversely correlated with overall survival only for the unifocal group. The negative correlation with overall and recurrence free survival was more pronounced in patients whose unifocal tumors expressed high levels of both RIP140 and ERß. Altogether, this preliminary report indicates that the ERß/RIP140 signaling is altered in unifocal breast cancers and correlated with patient outcome. Further investigation is needed to decipher the molecular mechanisms and the biological relevance of this deregulation.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , Estrogen Receptor beta/genetics , Gene Expression , Nuclear Receptor Interacting Protein 1/genetics , Adult , Aged , Biomarkers, Tumor , Breast Neoplasms/pathology , Estrogen Receptor beta/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Nuclear Receptor Interacting Protein 1/metabolism , Prognosis , Tumor Burden
8.
Transl Oncol ; 11(5): 1090-1096, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30007204

ABSTRACT

New markers are needed to improve diagnosis and to personalize treatments for patients with breast cancer (BC). Receptor-interacting protein of 140 kDa (RIP140) and ligand-dependent corepressor (LCoR), two transcriptional co-regulators of estrogen receptors, strongly interact in BC cells. Although their role in cancer progression has been outlined in the last few years, their function in BC has not been elucidated yet. In this study, we investigated RIP140 and LCoR localization (cytoplasm vs nucleus) in BC samples from a well-characterized cohort of patients (n = 320). RIP140 and LCoR were expressed in more than 80% of tumors, (predominantly in the cytoplasm), and the two markers were highly correlated. Expression of RIP140 and LCoR in the nucleus was negatively correlated with tumor size. Conversely, RIP140 and LCoR cytoplasmic expression strongly correlated with expression of two tumor aggressiveness markers: N-cadherin and CD133 (epithelial mesenchymal transition and cancer stem cell markers, respectively). Finally, high RIP140 nuclear expression was significantly correlated with longer overall survival, whereas high total or cytoplasmic expression of RIP140 was associated with shorter disease-free survival. Our study strongly suggests that the role of RIP140 and LCoR in BC progression could vary according to their prevalent sub-cellular localization, with opposite prognostic values for nuclear and cytoplasmic expression. The involvement in BC progression/invasiveness of cytoplasmic RIP140 could be balanced by the anti-tumor action of nuclear RIP140, thus explaining the previous contradictory findings about its role in BC.

9.
Oncotarget ; 7(15): 19693-708, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26930713

ABSTRACT

Histone lysine acetylation is an epigenetic mark regulated by histone acetyltransferases and histone deacetylases (HDAC) which plays an important role in tumorigenesis. In this study, we observed a strong overexpression of class IIa HDAC9, at the mRNA and protein levels, in the most aggressive human breast cancer cell lines (i.e. in basal breast cancer cells vs luminal ones or in malignant vs begnin MCF10A breast epithelial cell lines). HDAC9 overexpression was associated with higher rates of gene transcription and increased epigenetic marks on the HDAC9 promoter. Ectopic expression of HDAC9 in MCF7 luminal breast cancer cells led to an increase in cell proliferation and to a decrease in apoptosis. These effects were associated with a deregulated expression of several genes controlled by HDAC inhibitors such as CDKN1A, BAX and TNFRSF10A. Inversely, knock-down of HDAC9 expression in MDA-MB436 basal breast cancer cells reduced cell proliferation. Moreover, high HDAC9 expression decreased the efficacy of HDAC inhibitors to reduce cell proliferation and to regulate CDKN1A gene expression. Interestingly, the gene encoding the transcription factor SOX9 was identified by a global transcriptomic approach as an HDAC9 target gene. In stably transfected MCF7 cells, SOX9 silencing significantly decreased HDAC9 mitogenic activity. Finally, in a large panel of breast cancer biopsies, HDAC9 expression was significantly increased in tumors of the basal subtype, correlated with SOX9 expression and associated with poor prognosis. Altogether, these results indicate that HDAC9 is a key factor involved in mammary carcinogenesis and in the response to HDAC inhibitors.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Repressor Proteins/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Microscopy, Fluorescence , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
10.
Biochim Biophys Acta ; 1856(1): 144-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26116758

ABSTRACT

RIP140 is a transcriptional coregulator (also known as NRIP1) which plays very important physiological roles by finely tuning the activity of a large number of transcription factors. Noticeably, the RIP140 gene has been shown to be involved in the regulation of energy expenditure, in mammary gland development and intestinal homeostasis as well as in behavior and cognition. RIP140 is also involved in the regulation of various oncogenic signaling pathways and participates in the development and progression of solid tumors. This short review aims to summarize the role of this transcription factor on nuclear estrogen receptors, E2F and Wnt signaling pathways based on recent observations focusing on breast, ovary, liver and colon tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neoplasms/metabolism , Nuclear Proteins/physiology , Transcription, Genetic , Estrogens/metabolism , Female , Humans , Male , Neoplasms/classification , Nuclear Receptor Interacting Protein 1 , Signal Transduction , Wnt Proteins/metabolism
11.
J Hematol Oncol ; 8: 20, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25879677

ABSTRACT

RIP140 is a transcriptional coregulator, (also known as NRIP1), which finely tunes the activity of various transcription factors and plays very important physiological roles. Noticeably, the RIP140 gene has been implicated in the control of energy expenditure, behavior, cognition, mammary gland development and intestinal homeostasis. RIP140 is also involved in the regulation of various oncogenic signaling pathways and participates in the development and progression of solid tumors. During the past years, several papers have reported evidences linking RIP140 to hematologic malignancies. Among them, two recent studies with correlative data suggested that gene expression signatures including RIP140 can predict survival in chronic lymphocytic leukemia (CLL). This review aims to summarize the literature dealing with the expression of RIP140 in CLL and to explore the potential impact of this factor on transcription pathways which play key roles in this pathology.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Nuclear Proteins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Humans , Nuclear Proteins/genetics , Nuclear Receptor Interacting Protein 1
12.
J Clin Invest ; 124(5): 1899-913, 2014 May.
Article in English | MEDLINE | ID: mdl-24667635

ABSTRACT

Deregulation of the Wnt/APC/ß-catenin signaling pathway is an important consequence of tumor suppressor APC dysfunction. Genetic and molecular data have established that disruption of this pathway contributes to the development of colorectal cancer. Here, we demonstrate that the transcriptional coregulator RIP140 regulates intestinal homeostasis and tumorigenesis. Using Rip140-null mice and mice overexpressing human RIP140, we found that RIP140 inhibited intestinal epithelial cell proliferation and apoptosis. Interestingly, following whole-body irradiation, mice lacking RIP140 exhibited improved regenerative capacity in the intestine, while mice overexpressing RIP140 displayed reduced recovery. Enhanced RIP140 expression strongly repressed human colon cancer cell proliferation in vitro and after grafting onto nude mice. Moreover, in murine tissues and human cancer cells, RIP140 stimulated APC transcription and inhibited ß-catenin activation and target gene expression. Finally, RIP140 mRNA and RIP140 protein levels were decreased in human colon cancers compared with those in normal mucosal tissue, and low levels of RIP140 expression in adenocarcinomas from patients correlated with poor prognosis. Together, these results support a tumor suppressor role for RIP140 in colon cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/biosynthesis , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Homeostasis , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/pathology , Epithelial Cells/pathology , Female , Heterografts , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins/genetics , Nuclear Receptor Interacting Protein 1
13.
Mol Endocrinol ; 28(2): 183-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24422630

ABSTRACT

Estrogen receptors (ERs) are ligand-activated transcription factors involved in many physiological and pathological processes, including breast cancer. Their activity is fine-tuned by posttranslational modifications, notably sumoylation. In the present study, we investigated the role of the small ubiquitin-related modifier (SUMO) protease, SUMO1/sentrin/suppressor of Mif 2-specific peptidase 2 (SENP2), in the regulation of ERα activity. We first found SENP2 to significantly repress estradiol-induced transcriptional activity in breast cancer cells (MCF7 and T47D). This effect was observed with a reporter plasmid and on endogenous genes such as TFF1 and CTSD, which were shown to recruit SENP2 in chromatin immunoprecipitation experiments. Using glutathione S-transferase pull-down, coimmunoprecipitation and proximity ligation assays, SENP2 was found to interact with ERα and this interaction to be mediated by the amino-terminal region of the protease and the hinge region of the receptor. Interestingly, we demonstrated that ERα repression by SENP2 is independent of its SUMO protease activity and requires a transcriptional repressive domain located in the amino-terminal end of the protease. Using small interfering RNA assays, we evidenced that this domain recruits the histone deacetylase 3 (HDAC3), to be fully active. Furthermore, using both overexpression and knockdown strategies, we showed that SENP2 robustly represses estrogen-dependent and independent proliferation of MCF7 cells. We provided evidence that this effect requires both the proteolytic and transcriptional activities of SENP2. Altogether, our study unravels a new property for a SUMO protease and identifies SENP2 as a classical transcription coregulator.


Subject(s)
Cysteine Endopeptidases/physiology , Estrogen Receptor alpha/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing , Breast Neoplasms , Cell Proliferation , Estradiol/physiology , Female , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Transcription, Genetic
14.
Med Sci (Paris) ; 29(11): 1026-33, 2013 Nov.
Article in French | MEDLINE | ID: mdl-24280507

ABSTRACT

Cancer cell metabolism described by Otto Warburg in the thirties became a cancer specific hallmark, also called "Warburg effect". Cancer cells use essentially glucose as fuel, through glycolysis, in order to meet their energy and biomass needs to insure their cell proliferation. Recent advances describe Warburg effect regulation by oncogenes and tumor suppressor genes. Moreover, mutations in some glycolysis enzymes are found in various cancers, highlighting the role of cell metabolism in cancer. In this review, we describe the mechanisms responsible for the Warburg effect at the molecular and cellular level, the role of cell signalling along with the implication of different transcription factors. As a cause or a consequence of tumorigenesis, the Warburg effect is now considered as a promising therapeutic target in the fight against cancer.


Subject(s)
Models, Theoretical , Neoplasms/history , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cells/metabolism , Energy Metabolism , Germany , Glucose/metabolism , Glycolysis/genetics , History, 19th Century , History, 20th Century , Humans , Mutation , Neoplasms/etiology , Neoplasms/therapy , Signal Transduction
15.
Mol Endocrinol ; 27(9): 1429-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23885094

ABSTRACT

In hormone-dependent tissues such as breast and ovary, tumorigenesis is associated with an altered expression ratio between the two estrogen receptor (ER) subtypes. In this study, we investigated the effects of ERß ectopic expression on 17ß-estradiol (E2)-induced transactivation and cell proliferation in ERα-positive BG1 ovarian cancer cells. As expected, ERß expression strongly decreased the mitogenic effect of E2, significantly reduced E2-dependent transcriptional responses (both on a stably integrated estrogen response element [ERE] reporter gene and on E2-induced mRNAs), and strongly enhanced the formation of ER heterodimers as evidenced by chromatin immunoprecipitation analysis. Inhibition by the ERα-selective ligand propyl pyrazole triol was less marked than with the pan-agonist (E2) or the ERß-selective (8ß-vinyl-estradiol) ligands, indicating that ERß activation reinforced the inhibitory effects of ERß. Interestingly, in E2-stimulated BG1 cells, ERß was more efficient than ERα to regulate the expression of receptor-interacting protein 140 (RIP140), a major ERα transcriptional corepressor. In addition, we found that the RIP140 protein interacted better with ERß than with ERα (both in vitro and in intact cells by fluorescence cross-correlation spectroscopy). Moreover, RIP140 recruitment on the stably integrated reporter ERE was increased upon ERß overexpression, and ERß activity was more sensitive to repression by RIP140. Finally, small interfering RNA-mediated knockdown of RIP140 expression abolished the repressive effect exerted by activated ERß on the regulation of ERE-controlled transcription by estrogens. Altogether, these data demonstrate the inhibitory effects of ERß on estrogen signaling in ovarian cancer cells and the key role that RIP140 plays in this phenomenon.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Animals , COS Cells , Cell Line, Tumor , Cell Proliferation , Chlorocebus aethiops , DNA, Neoplasm/metabolism , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Ligands , Nuclear Receptor Interacting Protein 1 , Ovarian Neoplasms/genetics , Protein Binding/genetics , Transcriptional Activation/genetics
16.
FEBS Lett ; 584(11): 2218-24, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20398657

ABSTRACT

Products of the steroid receptor RNA activator (SRA1) gene have the unusual property to function both at the RNA and the protein levels. SRA-RNA has long been known to increase the activity of multiple nuclear receptors. It has more recently been proposed than steroid receptor RNA activator protein (SRAP) also modulates steroid receptors activity. Herein, we show for the first time that SRAP physically interacts with multiple transcription factors and is recruited to specific promoter regions. Artificially recruiting SRAP to the promoter of a luciferase reporter gene under the control of the strong transcriptional activator VP16 leads to a decrease in transcription. Altogether we propose that SRAP could be a new transcriptional regulator, able to function as a repressor through direct association with promoters.


Subject(s)
Proteins/metabolism , RNA, Untranslated/metabolism , Transcription Factors/metabolism , Carrier Proteins/metabolism , Humans , RNA/metabolism , RNA, Long Noncoding , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism
17.
Biochemistry ; 49(4): 772-81, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20039662

ABSTRACT

Quantitative characterization of protein interactions in live cells remains one of the most important challenges in modern biology. In the present work we have used two-photon, two-color, fluorescence cross-correlation spectroscopy (FCCS) in transiently transfected COS-7 cells to measure the concentrations and interactions of estrogen receptor (ER) subtypes alpha and beta with one of their transcriptional coactivator proteins, TIF2, as well as heterodimerization between the two ER subtypes. Using this approach in a systematic fashion, we observed a strong ligand-dependent modulation of receptor-coactivator complexation, as well as strong protein concentration dependence for complex formation in the absence of ligand. These quantitative values for protein and complex concentrations provide the first estimates for the ER-TIF2 K(d) for the full-length proteins and in a cellular context (agonist, < approximately 6 nM; antagonist, > approximately 3 microM; unliganded, approximately 200 nM). Coexpression of the two ER subtypes revealed substantial receptor heterodimer formation. They also provide, for the first time, estimated homo- and heterodimerization constants found to be similar and in the low nanomolar range. These results underscore the importance of receptor and coregulator expression levels and stability in the tissue-dependent modulation of receptor function under normal and pathological conditions.


Subject(s)
Receptors, Estrogen/metabolism , Spectrometry, Fluorescence/methods , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Estrogen/chemistry
18.
Trends Endocrinol Metab ; 21(3): 181-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005732

ABSTRACT

Estrogen signaling pathways regulate multiple cellular processes including proliferation and differentiation, and dysregulation of these pathways underlies several human pathologies. Post-translational modifications (PTMs) play an important role in estrogen signaling. This review focuses on recent findings pertinent to arginine methylation of non-histone proteins and their implications in estrogen signaling. We describe protein arginine methyltransferases and demethylases, the role of methylarginine proteins in estrogen action and crosstalk with other PTMs such as phosphorylation and lysine methylation. The relationships between various PTMs form a specific code that is likely to play an important role in hormone signaling. In addition, dysregulation of arginine methylation or of enzymes responsible for these modifications could be key events in estrogen-dependent cancers such as breast cancer.


Subject(s)
Arginine/metabolism , Estrogens/metabolism , Neoplasms/metabolism , Signal Transduction/physiology , Animals , Breast Neoplasms/metabolism , Female , Humans , Male , Methylation , Models, Biological , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Estrogen/metabolism
20.
Nucl Recept Signal ; 4: e024, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17088940

ABSTRACT

The nuclear receptor superfamily comprises ligand-regulated transcription factors that control various developmental and physiological pathways. These receptors share a common modular structure and regulate gene expression through the recruitment of a large set of coregulatory proteins. These transcription cofactors regulate, either positively or negatively, chromatin structure and transcription initiation. One of the first proteins to be identified as a hormone-recruited cofactor was RIP140. Despite its recruitment by agonist-liganded receptors, RIP140 exhibits a strong transcriptional repressive activity which involves several inhibitory domains and different effectors. Interestingly, the RIP140 gene, located on chromosome 21 in humans, is finely regulated at the transcriptional level by various nuclear receptors. In addition, the protein undergoes several post-translational modifications which control its repressive activity. Finally, experiments performed in mice devoid of the RIP140 gene indicate that this transcriptional cofactor is essential for female fertility and energy homeostasis. RIP140 therefore appears to be an important modulator of nuclear receptor activity which could play major roles in physiological processes and hormone-dependent diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...