Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
Life Sci Alliance ; 6(10)2023 10.
Article in English | MEDLINE | ID: mdl-37536978

ABSTRACT

Triple-negative breast cancers (TNBCs) are the most aggressive breast cancers, and therapeutic options mainly rely on chemotherapy and immunotherapy. Although synthetic glucocorticoids (GCs) are given to alleviate the side effects of these treatments, GCs and their receptor, the glucocorticoid receptor (GR), were recently associated with detrimental effects, albeit the mechanisms involved remain elusive. Here, we identified the arginine methyltransferase PRMT5 as a master coregulator of GR, serving as a scaffold protein to recruit phospho-HP1γ and subsequently RNA polymerase II, independently of its methyltransferase activity. Moreover, the GR/PRMT5/HP1γ complex regulated the transcription of GC-target genes involved in cell motility and triggering cell migration of human TNBC cells in vitro and in a zebrafish model. Of note, we observed that GR/PRMT5 interaction was low in primary tumors but significantly increased in residual tumors treated with chemotherapy and GCs in neoadjuvant setting. These data suggest that the routine premedication prescription of GCs for early TNBC patients should be further assessed and that this complex could potentially be modulated to specifically target deleterious GR effects.


Subject(s)
Cell Movement , Glucocorticoids , Protein-Arginine N-Methyltransferases , Triple Negative Breast Neoplasms , Cell Movement/drug effects , Triple Negative Breast Neoplasms/pathology , Glucocorticoids/adverse effects , Humans , Animals , Zebrafish , Receptors, Glucocorticoid/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Cell Line, Tumor
2.
EMBO Mol Med ; 15(8): e17248, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37458145

ABSTRACT

Endocrine therapies targeting estrogen signaling, such as tamoxifen, have significantly improved management of estrogen receptor alpha (ERα)-positive breast cancers. However, their efficacy is limited by intrinsic and acquired resistance to treatment, and there is currently no predictive marker of response to these anti-estrogens to guide treatment decision. Here, using two independent cohorts of breast cancer patients, we identified nuclear PRMT5 expression as an independent predictive marker of sensitivity to tamoxifen. Mechanistically, we discovered that tamoxifen stimulates ERα methylation by PRMT5, a key event for its binding to corepressors such as SMRT and HDAC1, participating in the inhibition of the transcriptional activity of ERα. Although PRMT5 is mainly localized in the cytoplasm of tumor cells, our analyses show that tamoxifen triggers its nuclear translocation in tamoxifen-sensitive tumors but not in resistant ones. Hence, we unveil a biomarker of sensitivity to tamoxifen in ERα-positive breast tumors that could be used to enhance the response of breast cancer patients to endocrine therapy, by fostering its nuclear expression.


Subject(s)
Breast Neoplasms , Tamoxifen , Humans , Female , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Signal Transduction , Biomarkers , Drug Resistance, Neoplasm , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/pharmacology , Protein-Arginine N-Methyltransferases/therapeutic use
3.
Int J Mol Sci ; 23(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36076907

ABSTRACT

The progesterone receptor (PR) is a key player in major physiological and pathological responses in women, and the signaling pathways triggered following hormone binding have been extensively studied, particularly with respect to breast cancer development and progression. Interestingly, growing evidence suggests a fundamental role for PR on breast cancer cell homeostasis in hormone-depleted conditions, with hormone-free or unliganded PR (uPR) involved in the silencing of relevant genes prior to hormonal stimulation. We herein identify the protein arginine methyltransferase PRMT1 as a novel actor in uPR signaling. In unstimulated T47D breast cancer cells, PRMT1 interacts and functions alongside uPR and its partners to target endogenous progesterone-responsive promoters. PRMT1 helps to finely tune the silencing of responsive genes, likely by promoting a proper BRCA1-mediated degradation and turnover of unliganded PR. As such, PRMT1 emerges as a key transcriptional coregulator of PR for a subset of relevant progestin-dependent genes before hormonal treatment. Since women experience periods of hormonal fluctuation throughout their lifetime, understanding how steroid receptor pathways in breast cancer cells are regulated when hormones decline may help to determine how to override treatment failure to hormonal therapy and improve patient outcome.


Subject(s)
Breast Neoplasms , Protein-Arginine N-Methyltransferases , Receptors, Progesterone , Breast Neoplasms/metabolism , Female , Humans , Progesterone/metabolism , Progestins , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Progesterone/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology
4.
J Vis Exp ; (185)2022 07 20.
Article in English | MEDLINE | ID: mdl-35938827

ABSTRACT

Arginine methylation is emerging as a key post-translational modification involved in a large range of biological processes. Its study in tissue is often limited by the lack of a specific antibody recognizing the target arginine residue. Proximity ligation assay (PLA) was originally developed to study protein/protein interactions. Here, we describe in detail a PLA protocol dedicated to the detection of arginine methylation that we applied to the glucocorticoid receptor (GR). Having previously shown that PRMT5 dimethylates GRs in cells, we used PLA with a pan symmetrical dimethyl antibody and an anti-GR antibody to measure GR methylation in breast tumors. We demonstrate that PLA offers a unique approach to measure arginine methylation of a target protein, even when the site of methylation has not been identified. This technique could be extended to other post-translational modifications where effective pan antibodies are available. Hence, we detail the PLA technology used to detect arginine methylation in fixed tissue using GR as an example.


Subject(s)
Arginine , Biological Phenomena , Antibodies/metabolism , Arginine/metabolism , Methylation , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Proteins/metabolism
5.
Breast Cancer Res ; 23(1): 57, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34020697

ABSTRACT

BACKGROUND: Endocrine therapies targeting estrogen signaling have significantly improved breast cancer (BC) patient survival, although 40% of ERα-positive BCs do not respond to those therapies. Aside from genomic signaling, estrogen triggers non-genomic pathways by forming a complex containing methylERα/Src/PI3K, a hallmark of aggressiveness and resistance to tamoxifen. We aimed to confirm the prognostic value of this complex and investigated whether its targeting could improve tumor response in vivo. METHODS: The interaction of ERα/Src and ERα/PI3K was studied by proximity ligation assay (PLA) in a cohort of 440 BC patients. We then treated patient-derived BC xenografts (PDXs) with fulvestrant or the PI3K inhibitor alpelisib (BYL719) alone or in combination. We analyzed their anti-proliferative effects on 6 ERα+ and 3 ERα- PDX models. Genomic and non-genomic estrogen signaling were assessed by measuring ERα/PI3K interaction by PLA and the expression of estrogen target genes by RT-QPCR, respectively. RESULTS: We confirmed that ERα/Src and ERα/PI3K interactions were associated with a trend to poorer survival, the latter displaying the most significant effects. In ERα+ tumors, the combination of BYL719 and fulvestrant was more effective than fulvestrant alone in 3 models, irrespective of PI3K, PTEN status, or ERα/PI3K targeting. Remarkably, resistance to fulvestrant was associated with non-genomic ERα signaling, since genomic degradation of ERα was unaltered in these tumors, whereas the treatment did not diminish the level of ERα/PI3K interaction. Interestingly, in 2 ERα- models, fulvestrant alone impacted tumor growth, and this was associated with a decrease in ERα/PI3K interaction. CONCLUSIONS: Our results demonstrate that ERα/PI3K may constitute a new prognostic marker, as well as a new target in BC. Indeed, resistance to fulvestrant in ERα+ tumors was associated with a lack of impairment of ERα/PI3K interaction in the cytoplasm. In addition, an efficient targeting of ERα/PI3K in ERα- tumors could constitute a promising therapeutic option.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Fulvestrant/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Receptors, Estrogen/metabolism , Thiazoles/therapeutic use , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Female , Genomics , Humans , Mice , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Cancers (Basel) ; 12(11)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187149

ABSTRACT

RAS proteins (KRAS, NRAS and HRAS) are frequently activated in different cancer types (e.g., non-small cell lung cancer, colorectal cancer, melanoma and bladder cancer). For many years, their activities were considered redundant due to their high degree of sequence homology (80% identity) and their shared upstream and downstream protein partners. However, the high conservation of the Hyper-Variable-Region across mammalian species, the preferential activation of different RAS proteins in specific tumor types and the specific post-translational modifications and plasma membrane-localization of each paralog suggest they could ensure discrete functions. To gain insights into RAS proteins specificities, we explored their proximal protein-protein interaction landscapes using the proximity-dependent biotin identification technology (BioID) in Flp-In T-REx 293 cell lines stably transfected and inducibly expressing wild type KRAS4B, NRAS or HRAS. We identified more than 800 high-confidence proximal interactors, allowing us to propose an unprecedented comparative analysis of wild type RAS paralogs protein networks. These data bring novel information on poorly characterized RAS functions, e.g., its putative involvement in metabolic pathways, and on shared as well as paralog-specific protein networks that could partially explain the complexity of RAS functions. These networks of protein interactions open numerous avenues to better understand RAS paralogs biological activities.

7.
Breast Cancer Res ; 22(1): 50, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32429997

ABSTRACT

BACKGROUND: Alterations in estrogen and progesterone signaling, via their respective receptors, estrogen receptor alpha (ERα) and progesterone receptor (PR), respectively, are largely involved in the development of breast cancer (BC). The recent identification of ERα-36, a splice variant of ERα, has uncovered a new facet of this pathology. Although ERα-36 expression is associated with poor prognosis, metastasis development, and resistance to treatment, its predictive value has so far not been associated with a BC subtype and its mechanisms of action remain understudied. METHODS: To study ERα-36 expression in BC specimens, we performed immunochemical experiments. Next, the role of ERα-36 in progesterone signaling was investigated by generating KO clones using the CRISPR/CAS9 technology. PR signaling was also assessed by proximity ligation assay, Western blotting, RT-QPCR, and ChIP experiments. Finally, proliferation assays were performed with the IncuCyte technology and migration experiments using scratch assays. RESULTS: Here, we demonstrate that ERα-36 expression at the plasma membrane is correlated with a reduced disease-free survival in a cohort of 160 BC patients, particularly in PR-positive tumors, suggesting a crosstalk between ERα-36 and PR. Indeed, we show that ERα-36 interacts constitutively with PR in the nucleus of tumor cells. Moreover, it regulates PR expression and phosphorylation on key residues, impacting the biological effects of progesterone. CONCLUSIONS: ERα-36 is thus a regulator of PR signaling, interfering with its transcriptional activity and progesterone-induced anti-proliferative effects as well as migratory capacity. Hence, ERα-36 may constitute a new prognostic marker as well as a potential target in PR-positive BC.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Protein Isoforms , Receptor, ErbB-2/genetics , Receptors, Progesterone/genetics , Retrospective Studies , Survival Rate
8.
Methods ; 175: 66-71, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31499160

ABSTRACT

Arginine methylation is now recognized as a major contributor to proteome diversity and is, as such, involved in a large range of cellular processes. There is a growing need for assessing endogenous protein arginine methylation in cells. Besides the classical immunoprecipitation, in situ proximity ligation assay (PLA) is a useful technique allowing at the same time the detection, localization and quantification of arginine methylation of a given protein within a cellular context. Here, we described in depth a standard PLA protocol applied to the detection of arginine methylation in combination with RNA interference and specific methyltransferase inhibitors. We demonstrated that the glucocorticoid receptor is methylated by the arginine methyltransferase PRMT5 inside the nucleus of MCF-7 cells. In addition, the automated quantification of protein arginine methylation performed using Image J is reported. Hence, we demonstrated that PLA offers a novel approach to study protein arginine methylation and could be extended to other post-translational modifications when specific antibodies are available.


Subject(s)
Arginine/metabolism , Enzyme Assays/methods , Epigenomics/methods , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Glucocorticoid/metabolism , Cell Nucleus/metabolism , DNA Ligases/chemistry , Enzyme Inhibitors/chemistry , Humans , Immunoprecipitation , MCF-7 Cells , Methylation , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , RNA Interference , Sensitivity and Specificity , Software
9.
Int J Mol Sci ; 20(11)2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31195751

ABSTRACT

Endocrine therapies targeting oestrogen signalling have significantly improved breast cancer management. However, their efficacy is limited by intrinsic and acquired resistance to treatment, which remains a major challenge for oestrogen receptor α (ERα)-positive tumours. Though many studies using in vitro models of endocrine resistance have identified putative actors of resistance, no consensus has been reached. We demonstrated previously that oestrogen non-genomic signalling, characterized by the formation of the ERα/Src/PI3K complex, is activated in aggressive breast cancers (BC). We wondered herein whether the activation of this pathway is also involved in resistance to endocrine therapies. We studied the interactions between ERα and Src or PI3K by proximity ligation assay (PLA) in in-vitro and in-vivo endocrine therapy-resistant breast cancer models. We reveal an increase in ERα/Src and ERα/PI3K interactions in patient-derived xenografts (PDXs) with acquired resistance to tamoxifen, as well as in tamoxifen-resistant MCF-7 cells compared to parental counterparts. Moreover, no interactions were observed in breast cancer cells resistant to other endocrine therapies. Finally, the use of a peptide inhibiting the ERα-Src interaction partially restored tamoxifen sensitivity in resistant cells, suggesting that such components could constitute promising targets to circumvent resistance to tamoxifen in BC.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Estrogens/pharmacology , Signal Transduction , Tamoxifen/pharmacology , Animals , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism
10.
Oncogene ; 38(21): 4015-4027, 2019 05.
Article in English | MEDLINE | ID: mdl-30692633

ABSTRACT

Aside from its well-known nuclear routes of signaling, estrogen also mediates its effects through cytoplasmic signaling. Estrogen signaling involves numerous posttranslational modifications of its receptor ERα, the best known being phosphorylation. Our research group previously showed that upon estrogen stimulation, ERα is methylated on residue R260 and forms the mERα/Src/PI3K complex, central to the rapid transduction of nongenomic estrogen signals. Regulation of ERα signaling via its phosphorylation by growth factors is well recognized, and we wondered whether they could also trigger ERα methylation (mERα). Here, we found that IGF-1 treatment of MCF-7 cells induced rapid ERα methylation by the arginine methyltransferase PRMT1 and triggered the binding of mERα to IGF-1R. Mechanistically, we showed that PRMT1 bound constitutively to IGF-1R and that PRMT1 became activated upon IGF-1 stimulation. Moreover, we found that expression or pharmacological inhibition of PRMT1 impaired mERα and IGF-1 signaling. Our findings were substantiated in a cohort of breast tumors in which IGF-1R expression was positively correlated with ERα/Src and ERα/PI3K expression, hallmarks of nongenomic estrogen signaling, reinforcing the link between IGF-1R and mERα. Altogether, these results provide a new insight into ERα and IGF-1R interference, and open novel perspectives for combining endocrine therapies with PRMT1 inhibitors in ERα-positive tumors.


Subject(s)
Breast Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Genes, src/genetics , Humans , MCF-7 Cells , Methylation , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/physiology , Receptor, IGF Type 1/metabolism
11.
Int J Cancer ; 144(3): 595-606, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30289978

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is the main enzyme responsible for the symmetrical dimethylation of arginine residues on target proteins in both the cytoplasm and the nucleus. Though its activity has been associated with tumor progression in various cancers, the expression pattern of this oncoprotein has been scarcely studied in breast cancer. In the current work, we analyzed its expression in a large cohort of breast cancer patients, revealing higher nuclear PRMT5 levels in ERα-positive tumors and an association with prolonged disease free and overall survival. Interestingly, high PRMT5 nuclear expression was also associated with higher nuclear liver kinase B1 (LKB1), suggesting that a functional relationship may occur. Consistently, several approaches provided evidence that PRMT5 and LKB1 interact directly in the cytoplasm of mammary epithelial cells. Moreover, although PRMT5 is not able to methylate LKB1, we found that PRMT5 is a bona fade substrate for LKB1. We identified T132, 139 and 144 residues, located in the TIM-Barrel domain of PRMT5, as target sites for LKB1 phosphorylation. The point mutation of PRMT5 T139/144 to A139/144 drastically decreased its methyltransferase activity, due probably to the loss of its interaction with regulatory proteins such as MEP50, pICln and RiOK1. In addition, modulation of LKB1 expression modified PRMT5 activity, highlighting a new regulatory mechanism that could have clinical implications.


Subject(s)
Breast Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , AMP-Activated Protein Kinase Kinases , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , MCF-7 Cells , Middle Aged , Phosphorylation
12.
J Mol Cell Biol ; 8(2): 129-43, 2016 04.
Article in English | MEDLINE | ID: mdl-26892023

ABSTRACT

Mitochondria-associated endoplasmic reticulum membranes (MAM) play a key role in mitochondrial dynamics and function and in hepatic insulin action. Whereas mitochondria are important regulators of energy metabolism, the nutritional regulation of MAM in the liver and its role in the adaptation of mitochondria physiology to nutrient availability are unknown. In this study, we found that the fasted to postprandial transition reduced the number of endoplasmic reticulum-mitochondria contact points in mouse liver. Screening of potential hormonal/metabolic signals revealed glucose as the main nutritional regulator of hepatic MAM integrity both in vitro and in vivo Glucose reduced organelle interactions through the pentose phosphate-protein phosphatase 2A (PP-PP2A) pathway, induced mitochondria fission, and impaired respiration. Blocking MAM reduction counteracted glucose-induced mitochondrial alterations. Furthermore, disruption of MAM integrity mimicked effects of glucose on mitochondria dynamics and function. This glucose-sensing system is deficient in the liver of insulin-resistant ob/ob and cyclophilin D-KO mice, both characterized by chronic disruption of MAM integrity, mitochondrial fission, and altered mitochondrial respiration. These data indicate that MAM contribute to the hepatic glucose-sensing system, allowing regulation of mitochondria dynamics and function during nutritional transition. Chronic disruption of MAM may participate in hepatic mitochondrial dysfunction associated with insulin resistance.


Subject(s)
Endoplasmic Reticulum/metabolism , Glucose/pharmacology , Intracellular Membranes/metabolism , Liver/metabolism , Mitochondria/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Endoplasmic Reticulum/ultrastructure , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intracellular Membranes/drug effects , Liver/drug effects , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/ultrastructure , Nutritional Status/drug effects , Phosphoprotein Phosphatases/metabolism , Postprandial Period/drug effects , Signal Transduction/drug effects , Voltage-Dependent Anion Channel 1/metabolism
13.
PLoS One ; 10(5): e0126181, 2015.
Article in English | MEDLINE | ID: mdl-25951181

ABSTRACT

BACKGROUND: Protein arginine methylation is a common post translational modification that regulates protein properties. This modification is carried out by a family of nine arginine methyltransferases (PRMTs). Arginine methylation has already been linked to tumourigenesis as overexpression of these enzymes was associated with various cancers, notably in breast cancers. Since the Jumonji Domain Containing 6 protein (JMJD6) possesses an arginine demethylase activity able to remove the methyl mark, we wanted to assess its potential role in breast tumourigenesis. METHODS: The expression of the protein by tissue microarray immunohistochemical staining was performed on a cohort of 133 breast tumours. Using cell lines stably overexpressing or knocked down for JMJD6, we evaluated its role on cell proliferation, cell migration, colony formation and mice tumour xenografts. RESULTS: The analysis of JMJD6 expression in a cohort of breast tumour samples indicates that JMJD6 was highly expressed in aggressive breast tumours. Moreover, high expression of JMJD6 was associated with poor disease-free survival of patients in this cohort. JMJD6 silencing in breast tumoural cells promotes certain characteristics of tumourigenesis including proliferation, migration in vitro, and tumour growth in vivo. These effects are dependent on its demethylase activity as an enzymatic dead mutant lost these properties. CONCLUSIONS: Although JMJD6 displays anti-tumoral properties in cell lines, its expression in breast tumours may be a marker of poor prognosis, suggesting that its function could be altered in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Jumonji Domain-Containing Histone Demethylases/physiology , Animals , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Nude , Middle Aged , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL