Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Commun Biol ; 5(1): 1068, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36207615

ABSTRACT

TGF-ß signaling is involved in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis, representing one of the four major pathways genetically altered in 100% of PDAC cases. TGF-ß exerts complex and pleiotropic effects in cancers, notably via the activation of SMAD pathways, predominantly SMAD2/3/4. Though SMAD2 and 3 are rarely mutated in cancers, SMAD4 is lost in about 50% of PDAC, and the role of SMAD2/3 in a SMAD4-null context remains understudied. We herein provide evidence of a SMAD2/3 oncogenic effect in response to TGF-ß1 in SMAD4-null human PDAC cancer cells. We report that inactivation of SMAD2/3 in SMAD4-negative PDAC cells compromises TGF-ß-driven collective migration mediated by FAK and Rho/Rac signaling. Moreover, RNA-sequencing analyses highlight a TGF-ß gene signature related to aggressiveness mediated by SMAD2/3 in the absence of SMAD4. Using a PDAC patient cohort, we reveal that SMAD4-negative tumors with high levels of phospho-SMAD2 are more aggressive and have a poorer prognosis. Thus, loss of SMAD4 tumor suppressive activity in PDAC leads to an oncogenic gain-of-function of SMAD2/3, and to the onset of associated deleterious effects.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Smad3 Protein/metabolism , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Humans , Pancreatic Neoplasms/metabolism , RNA , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1/metabolism , Pancreatic Neoplasms
2.
Sci Rep ; 10(1): 3880, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127548

ABSTRACT

Transforming growth factor (TGFß) is a secreted factor, which accumulates in tissues during many physio- and pathological processes such as embryonic development, wound healing, fibrosis and cancer. In order to analyze the effects of increased microenvironmental TGFß concentration in vivo, we developed a conditional transgenic mouse model (Flpo/Frt system) expressing bioactive TGFß in fibroblasts, a cell population present in the microenvironment of almost all tissues. To achieve this, we created the genetically-engineered [Fsp1-Flpo; FSFTGFßCA] mouse model. The Fsp1-Flpo allele consists in the Flpo recombinase under the control of the Fsp1 (fibroblast-specific promoter 1) promoter. The FSFTGFßCA allele consists in a transgene encoding a constitutively active mutant form of TGFß (TGFßCA) under the control of a Frt-STOP-Frt (FSF) cassette. The FSFTGFßCA allele was created to generate this model, and functionally validated by in vitro, ex vivo and in vivo techniques. [Fsp1-Flpo; FSFTGFßCA] animals do not present any obvious phenotype despite the correct expression of TGFßCA transgene in fibroblasts. This [Fsp1-Flpo; FSFTGFßCA] model is highly pertinent for future studies on the effect of increased microenvironmental bioactive TGFß concentrations in mice bearing Cre-dependent genetic alterations in other compartments (epithelial or immune compartments for instance). These dual recombinase system (DRS) approaches will enable scientists to study uncoupled spatiotemporal regulation of different genetic alterations within the same mouse, thus better replicating the complexity of human diseases.


Subject(s)
Fibroblasts/metabolism , Transforming Growth Factor beta/genetics , Animals , Gene Expression , Genetic Engineering , Hep G2 Cells , Humans , Mice , Mice, Transgenic , Models, Animal
3.
Genesis ; 58(5): e23359, 2020 05.
Article in English | MEDLINE | ID: mdl-32191380

ABSTRACT

Recombination systems represent a major breakthrough in the field of genetic model engineering. The Flp recombinases (Flp, Flpe, and Flpo) bind and cleave DNA Frt sites. We created a transgenic mouse strain ([Fsp1-Flpo]) expressing the Flpo recombinase in fibroblasts. This strain was obtained by random insertion inside mouse zygotes after pronuclear injection. Flpo expression was placed under the control of the promoter of Fsp1 (fibroblast-specific protein 1) gene, whose expression starts after gastrulation at Day 8.5 in cells of mesenchymal origin. We verified the correct expression and function of the Flpo enzyme by several ex vivo and in vivo approaches. The [Fsp1-Flpo] strain represents a genuine tool to further target the recombination of transgenes with Frt sites specifically in cells of mesenchymal origin or with a fibroblastic phenotype.


Subject(s)
DNA Nucleotidyltransferases/genetics , S100 Calcium-Binding Protein A4/genetics , Animals , Cells, Cultured , DNA Nucleotidyltransferases/metabolism , Fibroblasts/metabolism , Gastrula/metabolism , Gene Targeting/methods , HaCaT Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Zygote/metabolism
5.
Cell Death Dis ; 10(12): 886, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31767842

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the solid tumors with the poorest prognosis. The stroma of this tumor is abundant and composed of extracellular matrix and stromal cells (including cancer-associated fibroblasts and immune cells). Nerve fibers invading this stroma represent a hallmark of PDAC, involved in neural remodeling, which participates in neuropathic pain, cancer cell dissemination and tumor relapse after surgery. Pancreatic cancer-associated neural remodeling is regulated through functional interplays mediated by physical and molecular interactions between cancer cells, nerve cells and surrounding Schwann cells, and other stromal cells. In the present study, we show that Schwann cells (glial cells supporting peripheral neurons) can enhance aggressiveness (migration, invasion, tumorigenicity) of pancreatic cancer cells in a transforming growth factor beta (TGFß)-dependent manner. Indeed, we reveal that conditioned medium from Schwann cells contains high amounts of TGFß able to activate the TGFß-SMAD signaling pathway in cancer cells. We also observed in human PDAC samples that high levels of TGFß signaling activation were positively correlated with perineural invasion. Secretome analyses by mass spectrometry of Schwann cells and pancreatic cancer cells cultured alone or in combination highlighted the central role of TGFß in neuro-epithelial interactions, as illustrated by proteomic signatures related to cell adhesion and motility. Altogether, these results demonstrate that Schwann cells are a meaningful source of TGFß in PDAC, which plays a crucial role in the acquisition of aggressive properties by pancreatic cancer cells.

6.
Cell Mol Gastroenterol Hepatol ; 4(2): 263-282, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28752115

ABSTRACT

BACKGROUND & AIMS: Transforming growth factor beta (TGFß) acts either as a tumor suppressor or as an oncogene, depending on the cellular context and time of activation. TGFß activates the canonical SMAD pathway through its interaction with the serine/threonine kinase type I and II heterotetrameric receptors. Previous studies investigating TGFß-mediated signaling in the pancreas relied either on loss-of-function approaches or on ligand overexpression, and its effects on acinar cells have so far remained elusive. METHODS: We developed a transgenic mouse model allowing tamoxifen-inducible and Cre-mediated conditional activation of a constitutively active type I TGFß receptor (TßRICA) in the pancreatic acinar compartment. RESULTS: We observed that TßRICA expression induced acinar-to-ductal metaplasia (ADM) reprogramming, eventually facilitating the onset of KRASG12D-induced pre-cancerous pancreatic intraepithelial neoplasia. This phenotype was characterized by the cellular activation of apoptosis and dedifferentiation, two hallmarks of ADM, whereas at the molecular level, we evidenced a modulation in the expression of transcription factors such as Hnf1ß, Sox9, and Hes1. CONCLUSIONS: We demonstrate that TGFß pathway activation plays a crucial role in pancreatic tumor initiation through its capacity to induce ADM, providing a favorable environment for KRASG12D-dependent carcinogenesis. Such findings are highly relevant for the development of early detection markers and of potentially novel treatments for pancreatic cancer patients.

7.
Cancer Res ; 75(20): 4335-50, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26282171

ABSTRACT

The transcription accessory factor TIF1γ/TRIM33/RFG7/PTC7/Ectodermin functions as a tumor suppressor that promotes development and cellular differentiation. However, its precise function in cancer has been elusive. In the present study, we report that TIF1γ inactivation causes cells to accumulate chromosomal defects, a hallmark of cancer, due to attenuations in the spindle assembly checkpoint and the post-mitotic checkpoint. TIF1γ deficiency also caused a loss of contact growth inhibition and increased anchorage-independent growth in vitro and in vivo. Clinically, reduced TIF1γ expression in human tumors correlated with an increased rate of genomic rearrangements. Overall, our work indicates that TIF1γ exerts its tumor-suppressive functions in part by promoting chromosomal stability.


Subject(s)
Cell Cycle Checkpoints/genetics , Chromosomal Instability , Gene Expression Regulation, Neoplastic , Mitosis/genetics , Neoplasms/genetics , Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Carcinoma in Situ , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Gene Silencing , Humans , Mice , Mice, Knockout , Neoplasms/pathology , Ploidies , Spindle Apparatus/metabolism
8.
J Cell Biol ; 205(3): 409-28, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24821840

ABSTRACT

Transforming growth factor ß (TGF-ß) isoforms are secreted as inactive complexes formed through noncovalent interactions between the bioactive TGF-ß entity and its N-terminal latency-associated peptide prodomain. Extracellular activation of the latent TGF-ß complex is a crucial step in the regulation of TGF-ß function for tissue homeostasis. We show that the fibrinogen-like (FBG) domain of the matrix glycoprotein tenascin-X (TNX) interacts physically with the small latent TGF-ß complex in vitro and in vivo, thus regulating the bioavailability of mature TGF-ß to cells by activating the latent cytokine into an active molecule. Activation by the FBG domain most likely occurs through a conformational change in the latent complex and involves a novel cell adhesion-dependent mechanism. We identify α11ß1 integrin as a cell surface receptor for TNX and show that this integrin is crucial to elicit FBG-mediated activation of latent TGF-ß and subsequent epithelial-to-mesenchymal transition in mammary epithelial cells.


Subject(s)
Epithelial-Mesenchymal Transition , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Protein Precursors/metabolism , Tenascin/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cattle , Cell Adhesion , Cell Line, Tumor , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , Integrins/genetics , Integrins/metabolism , Mice , Phosphorylation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Precursors/genetics , RNA Interference , Receptors, Collagen/genetics , Receptors, Collagen/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Tenascin/genetics , Transfection , Transforming Growth Factor beta1/genetics
9.
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
10.
EMBO J ; 32(5): 688-700, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23386060

ABSTRACT

Stringent regulation of the interferon (IFN) signalling pathway is essential for maintaining the immune response to pathogens and tumours. The transcription factor STAT1 is a crucial mediator of this response. Here, we show that hCAF1/CNOT7 regulates class I and II IFN pathways at different crucial steps. In resting cells, hCAF1 can control STAT1 trafficking by interacting with the latent form of STAT1 in the cytoplasm. IFN treatment induces STAT1 release, suggesting that hCAF1 may shield cytoplasmic STAT1 from undesirable stimulation. Consistently, hCAF1 silencing enhances STAT1 basal promoter occupancy associated with increased expression of a subset of STAT1-regulated genes. Consequently, hCAF1 knockdown cells exhibit an increased protection against viral infection and reduced viral replication. Furthermore, hCAF1 participates in the extinction of the IFN signal, through its deadenylase activity, by speeding up the degradation of some STAT1-regulated mRNAs. Since abnormal and unbalanced JAK/STAT activation is associated with immune disorders and cancer, hCAF1 could play a major role in innate immunity and oncogenesis, contributing to tumour escape.


Subject(s)
Breast Neoplasms/metabolism , Interferons/pharmacology , STAT1 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factors/metabolism , Virus Replication/drug effects , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Cytoplasm/drug effects , Cytoplasm/metabolism , Exoribonucleases , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Immunoprecipitation , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
11.
Endocr Rev ; 32(5): 597-622, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21680538

ABSTRACT

Estrogen signaling pathways, because of their central role in regulating the growth and survival of breast tumor cells, have been identified as suitable and efficient targets for cancer therapies. Agents blocking estrogen activity are already widely used clinically, and many new molecules have entered clinical trials, but intrinsic or acquired resistance to treatment limits their efficacy. The basic molecular studies underlying estrogen signaling have defined the critical role of estrogen receptors (ER) in many aspects of breast tumorigenesis. However, important knowledge gaps remain about the role of posttranslational modifications (PTM) of ER in initiation and progression of breast carcinogenesis. Whereas major attention has been focused on the phosphorylation of ER, many other PTM (such as acetylation, ubiquitination, sumoylation, methylation, and palmitoylation) have been identified as events modifying ER expression and stability, subcellular localization, and sensitivity to hormonal response. This article will provide an overview of the current and emerging knowledge on ER PTM, with a particular focus on their deregulation in breast cancer. We also discuss their clinical relevance and the functional relationship between PTM. A thorough understanding of the complete picture of these modifications in ER carcinogenesis might not only open new avenues for identifying new markers for prognosis or prediction of response to endocrine therapy but also could promote the development of novel therapeutic strategies.


Subject(s)
Breast Neoplasms , Protein Processing, Post-Translational , Receptors, Estrogen , Acetylation , Animals , Antineoplastic Agents, Hormonal , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Estrogen Receptor Modulators/therapeutic use , Estrogen Receptor alpha/analysis , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Estrogens/physiology , Female , Humans , Methylation , Mutation , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Signal Transduction , Ubiquitination
12.
Med Sci (Paris) ; 26(6-7): 636-40, 2010.
Article in French | MEDLINE | ID: mdl-20619167

ABSTRACT

Regulation of the proteome by post-translational modifications (PTM) emerges as a major contributing factor to the functional diversity in biology regulating cellular processes. Because PTM are key to the physiologic functions of the proteins involved, it is imperative that we understand the << coding >> that these modifications impart to regulate diverse activities. As estrogen signalling mediates a plethora of PTM not only on the receptors themselves but also on their coregulators, we investigate to << crack >> the ER code. Besides the long-known phosphorylation, other covalent additions such as acetylation, ubiquitination, sumoylation and methylation have been described for estrogen receptors in recent years. These modifications affect receptor stability and activity, and provide potential mechanisms for cell- or-gene-specific regulation. A better understanding of the impact of these PTMs on estrogen receptor should help in the identification of new drugs for breast cancer treatments.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Protein Processing, Post-Translational , Breast Neoplasms/enzymology , Enzyme Activation , Estrogen Receptor alpha/genetics , Female , Histone Acetyltransferases/metabolism , Humans , Phosphorylation , Protein Kinases/metabolism
13.
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
14.
Mol Cell ; 31(2): 212-21, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18657504

ABSTRACT

Evidence is emerging that estrogen receptor alpha (ERalpha) is central to the rapid transduction of estrogen signaling to the downstream kinase cascades; however, the mechanisms underlying this nongenomic function are not fully understood. Here we report a paradigm of ERalpha regulation through arginine methylation by PRMT1, which transiently methylates arginine 260 within the ERalpha DNA-binding domain. This methylation event is required for mediating the extranuclear function of the receptor by triggering its interaction with the p85 subunit of PI3K and Src. Furthermore, we find that the focal adhesion kinase (FAK), a Src substrate involved in the migration process, is also recruited in this complex. Our data indicate that the methylation of ERalpha is a physiological process occurring in the cytoplasm of normal and malignant epithelial breast cells and that ERalpha is hypermethylated in a subset of breast cancers.


Subject(s)
Arginine/metabolism , Estrogens/pharmacology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme Activation/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Genome, Human/genetics , Humans , Methylation/drug effects , Mice , Models, Biological , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor Cross-Talk/drug effects , Substrate Specificity/drug effects
15.
J Cell Sci ; 120(Pt 4): 638-47, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17264152

ABSTRACT

Protein arginine methylation is an emergent post-translational modification involved in a growing number of cellular processes, including transcriptional regulation, cell signaling, RNA processing and DNA repair. Although protein arginine methyltransferase 1 (PRMT1) is the major arginine methyltransferase in mammals, little is known about the regulation of its activity, except for the regulation induced by interaction with the antiproliferative protein BTG1 (B-cell translocation gene 1). Since the protein hCAF1 (CCR4-associated factor 1) was described to interact with BTG1, we investigated a functional link between hCAF1 and PRMT1. By co-immunoprecipitation and immunofluorescence experiments we demonstrated that endogenous hCAF1 and PRMT1 interact in vivo and colocalize in nuclear speckles, a sub-nuclear compartment enriched in small nuclear ribonucleoproteins and splicing factors. In vitro methylation assays indicated that hCAF1 is not a substrate for PRMT1-mediated methylation, but it regulates PRMT1 activity in a substrate-dependent manner. Moreover, small interfering RNA (siRNA)-mediated silencing of hCAF1 in MCF-7 cells significantly modulates the methylation of endogenous PRMT1 substrates. Finally, we demonstrated that in vitro and in the cellular context, hCAF1 regulates the methylation of Sam68 and histone H4, two PRMT1 substrates. Since hCAF1 and PRMT1 have been involved in the regulation of transcription and RNA metabolism, we speculate that hCAF1 and PRMT1 could contribute to the crosstalk between transcription and RNA processing.


Subject(s)
Arginine/metabolism , Gene Expression Regulation , Methylation , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , HeLa Cells , Histones/metabolism , Humans , Protein-Arginine N-Methyltransferases/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics
16.
Mol Endocrinol ; 19(11): 2671-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15961505

ABSTRACT

The steroid hormone 17beta-estradiol (estrogen) plays a significant role in the normal physiology of the mammary gland and breast cancer development primarily through binding to its receptor, the estrogen receptor alpha (ERalpha). ERalpha is a nuclear transcription factor undergoing different types of posttranslational modifications, i.e. phosphorylation, acetylation, and ubiquitination, which regulate its transcriptional activation and/or stability. Here we identify ERalpha as a new target for small ubiquitin-like modifier (SUMO)-1 modification in intact cells and in vitro. Moreover, ERalpha sumoylation occurs strictly in the presence of hormone. SUMO-1 appears to regulate ERalpha-dependent transcription. Using a series of mutants, we demonstrated that ERalpha is sumoylated at conserved lysine residues within the hinge region. Mutations that prevented SUMO modification impaired ERalpha-induced transcription without influencing ERalpha cellular localization. In addition to identifying protein inhibitor of activated signal transducer and activator of transcription (PIAS)1 and PIAS3 as E3 ligases for ERalpha, we also found that PIAS1 and PIAS3, as well as Ubc9, modulated ERalpha-dependent transcription independently from their SUMO-1 conjugation activity. These findings identify sumoylation as a new mechanism modulating ERalpha-dependent cellular response and provide a link between the SUMO and estrogen pathways.


Subject(s)
Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Molecular Chaperones/metabolism , Protein Inhibitors of Activated STAT/metabolism , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Estradiol/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Humans , Molecular Chaperones/genetics , Mutation , Protein Inhibitors of Activated STAT/genetics , Sequence Deletion , Small Ubiquitin-Related Modifier Proteins/genetics , Transcription, Genetic , Transcriptional Activation , Ubiquitin-Conjugating Enzymes/metabolism
17.
RNA ; 11(4): 487-94, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15769875

ABSTRACT

The yeast Pop2 protein, belonging to the eukaryotic Caf1 family, is required for mRNA deadenylation in vivo. It also catalyzes poly(A) degradation in vitro, even though this property has been questioned. Caf1 proteins are related to RNase D, a feature supported by the recently published structure of Pop2. Yeast Pop2 contains, however, a divergent active site while its human homologs harbor consensus catalytic residues. Given these differences, we tested whether its deadenylase activity is conserved in the human homologs Caf1 and Pop2. Our data demonstrate that both human factors degrade poly(A) tails indicating their involvement in mRNA metabolism.


Subject(s)
Conserved Sequence , Poly A/metabolism , Proteins/metabolism , RNA Stability , RNA, Messenger/metabolism , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Molecular Sequence Data , Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleases/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription Factors
18.
J Cell Sci ; 116(Pt 14): 2929-36, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12771185

ABSTRACT

The yeast CCR4-NOT complex exists in two forms (1.0 and 1.9 MDa) that share several common subunits, including yCCR4, yCAF1 and five NOT proteins (NOT1-5). Here, we report that different complexes containing mammalian homologs of CCR4-NOT subunits exist in mammalian cells, with estimated sizes of approximately 1.9 MDa, approximately 1 MDa and approximately 650 kDa, and that BTG2, a member of a protein family with antiproliferative functions, can associate with these complexes. Immunoprecipitation and gel filtration experiments established that BTG2 interacts in vivo with hCCR4 protein via hCAF1 and hPOP2. Moreover, we show that hCCR4, as well as hCAF1 and BTG2, modulate the transcription regulation mediated by ERalpha. Finally, we demonstrate that the cellular localization of hCAF1 and the cell content in hCAF1-containing complexes change as cells progress from quiescence to S phase. These findings suggest that the different regulatory pathways in which hCAF1 is involved, notably transcription regulation and mRNA turnover, may occur through distinct CCR4 complexes in the course of cell-cycle progression.


Subject(s)
Genes, Tumor Suppressor/physiology , Immediate-Early Proteins/physiology , Proteins , Transcription Factors/metabolism , Cell Cycle , Cell Separation , Chromatography, Gel , Cytoplasm/metabolism , Estrogen Receptor alpha , Flow Cytometry , Genes, Reporter , Genetic Vectors , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immediate-Early Proteins/metabolism , Immunoblotting , Microscopy, Fluorescence , Precipitin Tests , Protein Binding , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , Recombinant Proteins/metabolism , S Phase , Transcription, Genetic , Transfection , Tumor Suppressor Proteins , Two-Hybrid System Techniques
19.
J Gen Virol ; 82(Pt 3): 507-512, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11172091

ABSTRACT

The K8 protein of Kaposi's sarcoma-associated herpesvirus (KSHV)/human herpesvirus-8 is a member of the bZIP family of transcription factors, which has homology with the Epstein-Barr virus transcription and replication factor, EB1. In this report, we have analysed the subcellular localization of the K8 protein and characterized a 12 amino acid sequence rich in basic residues which is responsible for targeting the protein to the cell nucleus. Furthermore, we show that a K8 mutant lacking the nuclear localization sequence can be directed to the nucleus by co-expression with an intact K8 protein, suggesting that K8 homodimerizes in the cytoplasm of the cell in vivo.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/virology , Herpesvirus 8, Human/metabolism , Nuclear Localization Signals/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Amino Acids , Animals , Basic-Leucine Zipper Transcription Factors , Binding Sites , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Nucleus/metabolism , HeLa Cells , Herpesvirus 8, Human/genetics , Humans , Leucine Zippers/genetics , Mutagenesis , Nuclear Localization Signals/biosynthesis , Nuclear Localization Signals/genetics , Peptide Fragments , Rabbits , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins , Sarcoma, Kaposi/virology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...