Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Cell Physiol ; 227(12): 3768-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22392896

ABSTRACT

The major effect of T3 on mitochondrial activity has been partly explained by the discovery of p43, a T3-dependent transcription factor of the mitochondrial genome. P43 is imported into mitochondria in an atypical manner which is not yet fully understood. Our aim was to characterize the p43 sequences inducing its mitochondrial import, using in organello import experiments with wild-type or mutated proteins and validation in CV1 cells. We find that several sequences define the mitochondrial addressing. Two alpha helices in the C-terminal part of p43 are actual mitochondrial import sequences as fusion to a cytosolic protein induces its mitochondrial translocation. Helix 5 drives the atypical mitochondrial import process, whereas helices 10/11 induce a classical import process. However, despite its inability to drive a mitochondrial import, the N-terminal region of p43 also plays a permissive role as in the presence of the C-terminal import sequences different N-terminal regions determine whether the protein is imported or not. These results can be extrapolated to other mitochondrial proteins related to the nuclear receptor superfamily, devoid of classical mitochondrial import sequences.


Subject(s)
Mitochondria, Liver/metabolism , Thyroid Hormone Receptors alpha/chemistry , Thyroid Hormone Receptors alpha/metabolism , Triiodothyronine/metabolism , Animals , Binding Sites , Cell Line , Male , Mutation , Plasmids , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Protein Transport/physiology , Rats , Rats, Wistar
2.
Exp Cell Res ; 317(14): 2059-71, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21664352

ABSTRACT

We have previously shown that mitochondrial protein synthesis regulates myoblast differentiation, partly through the control of c-Myc expression, a cellular oncogene regulating myogenin expression and myoblast withdrawal from the cell cycle. In this study we provide evidence of the involvement of Calcineurin in this regulation. In C2C12 myoblasts, inhibition of mitochondrial protein synthesis by chloramphenicol decreases Calcineurin expression. Conversely, stimulation of this process by overexpressing the T3 mitochondrial receptor (p43) increases Calcineurin expression. Moreover, expression of a constitutively active Calcineurin (ΔCN) stimulates myoblast differentiation, whereas a Calcineurin antisense has the opposite effect. Lastly, ΔCN expression or stimulation of mitochondrial protein synthesis specifically increases slow myosin heavy chain expression. In conclusion, these data clearly suggest that, partly via Calcineurin expression, mitochondrial protein synthesis is involved in muscle development through the control of myoblast differentiation and probably the acquisition of the contractile and metabolic phenotype of muscle fibres.


Subject(s)
Calcineurin/genetics , Cell Differentiation , Cytokines/metabolism , Gene Expression Regulation , Mitochondria, Muscle/metabolism , Myoblasts/cytology , Myosins/biosynthesis , Animals , Birds , Calcineurin/metabolism , Cells, Cultured , Cytokines/genetics , Humans , Mice , Myoblasts/metabolism , Myosins/metabolism , Protein Isoforms/biosynthesis , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Cardiovasc Res ; 116(3): 633-644, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31147690

ABSTRACT

AIMS: Regulated cell death is a main contributor of myocardial ischaemia-reperfusion (IR) injury during acute myocardial infarction. In this context, targeting apoptosis could be a potent therapeutical strategy. In a previous study, we showed that DAXX (death-associated protein) was essential for transducing the FAS-dependent apoptotic signal during IR injury. The present study aims at evaluating the cardioprotective effects of a synthetic peptide inhibiting FAS:DAXX interaction. METHODS AND RESULTS: An interfering peptide was engineered and then coupled to the Tat cell penetrating peptide (Tat-DAXXp). Its internalization and anti-apoptotic properties were demonstrated in primary cardiomyocytes. Importantly, an intravenous bolus injection of Tat-DAXXp (1 mg/kg) 5 min before reperfusion in a murine myocardial IR model decreased infarct size by 48% after 24 h of reperfusion. In addition, Tat-DAXXp was still efficient after a 30-min delayed administration, and was completely degraded and eliminated within 24 h thereby reducing risks of potential side effects. Importantly, Tat-DAXXp reduced mouse early post-infarction mortality by 67%. Mechanistically, cardioprotection was supported by both anti-apoptotic and pro-survival effects, and an improvement of myocardial functional recovery as evidenced in ex vivo experiments. CONCLUSIONS: Our study demonstrates that a single dose of Tat-DAXXp injected intravenously at the onset of reperfusion leads to a strong cardioprotection in vivo by inhibiting IR injury validating Tat-DAXXp as a promising candidate for therapeutic application.


Subject(s)
Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , Co-Repressor Proteins/antagonists & inhibitors , Molecular Chaperones/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Animals , Cell Line , Cell Survival/drug effects , Co-Repressor Proteins/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function/drug effects , Signal Transduction , fas Receptor/metabolism
4.
Mol Endocrinol ; 22(2): 317-30, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962381

ABSTRACT

The ubiquitin-proteasome pathway has been recognized as an important regulator in the hormonal response by estrogen receptor (ER) alpha, but its impact on ERbeta function is poorly characterized. In the current study, we investigated the role of the ubiquitin-proteasome pathway in regulating ERbeta activity and identified regulatory sites within the activation function (AF)-1 domain that modulate ERbeta ubiquitination and nuclear dynamics in a hormone-independent manner. Although both ERalpha and ERbeta were dependent on proteasome function for their maximal response to estrogen, they were regulated differently by proteasome inhibition in the absence of hormone, an effect shown to be dependent on their respective AF-1 domain. Given the role of AF-1 phosphorylation to regulate ER activity, we found that sequential substitutions of specific serine residues contained in MAPK consensus sites conferred transcriptional activation of ERbeta in a proteasome-dependent manner through reduced ubiquitination and enhanced accumulation of mutant receptors. Specifically, serines 94 and 106 within ERbeta AF-1 domain were found to modulate subnuclear mobility of the receptor to transit between inactive clusters and a more mobile state in a proteasome-dependent manner. In addition, cellular levels of ERbeta were regulated through these sites by facilitating the recruitment of the ubiquitin ligase E6-associated protein in a phosphorylation-dependent manner. These findings suggest a role for ERbeta AF-1 in contributing to the activation-degradation cycling of the receptor through a functional clustering of phosphorylated serine residues that cooperate in generating signals to the ubiquitin-proteasome pathway.


Subject(s)
Cell Nucleus/metabolism , Estrogen Receptor beta/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites/genetics , Cell Line , Consensus Sequence , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Fluorescence Recovery After Photobleaching , Humans , Immunoblotting , Immunoprecipitation , Leupeptins/pharmacology , Mice , Models, Biological , Mutation , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors , Serine/genetics , Serine/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
Mol Cancer Ther ; 18(8): 1386-1395, 2019 08.
Article in English | MEDLINE | ID: mdl-31092563

ABSTRACT

Differently from cytotoxic chemotherapies, targeted therapies do not necessarily drive cancer cells toward death, but reduce cell proliferation, angiogenesis, and/or prevent metastasis without affecting healthy cells. Oncogenic proteins that are hyperactivated and/or overexpressed in cancer cells are prime targets for such therapies. On the other hand, the activity of tumor suppressor proteins is more difficult to harness. Here, we identified a short SOX9 sequence (S9pep) located at the hinge between the HMG DNA-binding domain and the SOX-E central conserved domain that mimics SOX9 tumor-suppressive properties. Doxycycline-induced S9pep expression in DLD-1 colorectal cancer cells inhibited the growth potential of these cells, including colorectal cancer stem cells, restored cell-cell contact inhibition, and inhibited the activity of the oncogenic Wnt/ß-catenin signaling pathway. It also significantly decreased tumor growth in BALB/cAnNCrl mice grafted with mouse doxycycline-inducible CT26 colorectal cancer cells in which S9pep was induced by treating them with doxycycline. As the Wnt/ß-catenin signaling pathway is constitutively activated in 80% of colorectal cancer and SOX9-inactivating mutations are present in up to 11% of colorectal cancer, S9pep could be a promising starting point for the development of a peptide-based therapeutic approach to restore a SOX9-like tumor suppressor function in colorectal cancer.


Subject(s)
Biological Mimicry , Peptides/pharmacology , SOX9 Transcription Factor/chemistry , SOX9 Transcription Factor/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Peptides/chemistry , Proto-Oncogene Proteins c-myc , Spheroids, Cellular , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
MAbs ; 11(3): 593-605, 2019 04.
Article in English | MEDLINE | ID: mdl-30604643

ABSTRACT

Targeting transferrin receptor 1 (TfR1) with monoclonal antibodies is a promising therapeutic strategy in cancer as tumor cells often overexpress TfR1 and show increased iron needs. We have re-engineered six anti-human TfR1 single-chain variable fragment (scFv) antibodies into fully human scFv2-Fcγ1 and IgG1 antibodies. We selected the more promising candidate (H7), based on its ability to inhibit TfR1-mediated iron-loaded transferrin internalization in Raji cells (B-cell lymphoma). The H7 antibody displayed nanomolar affinity for its target in both formats (scFv2-Fcγ1 and IgG1), but cross-reacted with mouse TfR1 only in the scFv2-Fc format. H7 reduced the intracellular labile iron pool and, contrary to what has been observed with previously described anti-TfR1 antibodies, upregulated TfR1 level in Raji cells. H7 scFv2-Fc format elimination half-life was similar in FcRn knock-out and wild type mice, suggesting that TfR1 recycling contributes to prevent H7 elimination in vivo. In vitro, H7 inhibited the growth of erythroleukemia and B-cell lymphoma cell lines (IC50 0.1 µg/mL) and induced their apoptosis. Moreover, the Im9 B-cell lymphoma cell line, which is resistant to apoptosis induced by rituximab (anti-CD20 antibody), was sensitive to H7. In vivo, tumor regression was observed in nude mice bearing ERY-1 erythroleukemia cell xenografts treated with H7 through a mechanism that involved iron deprivation and antibody-dependent cytotoxic effector functions. Therefore, targeting TfR1 using the fully human anti-TfR1 H7 is a promising tool for the treatment of leukemia and lymphoma.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Antigens, CD/immunology , Antineoplastic Agents, Immunological , Leukemia, Erythroblastic, Acute , Receptors, Transferrin/immunology , Single-Chain Antibodies , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , CHO Cells , Cricetulus , HEK293 Cells , Humans , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Knockout , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology
7.
Clin Cancer Res ; 25(15): 4775-4790, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31061069

ABSTRACT

PURPOSE: For the development of new anticancer therapeutic radiopharmaceuticals, including alpha particle emitters, it is important to determine the contribution of targeted effects in irradiated cells, and also of nontargeted effects in nonirradiated neighboring cells, because they may affect the therapeutic efficacy and contribute to side effects. EXPERIMENTAL DESIGN: Here, we investigated the contribution of nontargeted cytotoxic and genotoxic effects in vitro and in vivo (in xenografted mice) during alpha (212Pb/212Bi, 213Bi) and Auger (125I) radioimmunotherapy (RIT). RESULTS: Between 67% and 94% (alpha RIT) and 8% and 15% (Auger RIT) of cancer cells were killed by targeted effects, whereas 7% to 36% (alpha RIT) and 27% to 29% (Auger RIT) of cells were killed by nontargeted effects. We then demonstrated that the nontargeted cell response to alpha and Auger RIT was partly driven by lipid raft-mediated activation of p38 kinase and JNK. Reactive oxygen species also played a significant role in these nontargeted effects, as demonstrated by NF-κB activation and the inhibitory effects of antioxidant enzymes and radical scavengers. Compared with RIT alone, the use of RIT with ASMase inhibitor (imipramine) or with a lipid raft disruptor (e.g., methyl-beta-cyclodextrin or filipin) led to an increase in clonogenic cell survival in vitro and to larger tumors and less tissue DNA damage in vivo. These results were supported by an inhibitory effect of pravastatin on Auger RIT. CONCLUSIONS: Cell membrane-mediated nontargeted effects play a significant role during Auger and alpha RIT, and drugs that modulate cholesterol level, such as statins, could interfere with RIT efficacy.


Subject(s)
Cholesterol/metabolism , Imipramine/pharmacology , MAP Kinase Kinase 4/metabolism , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radiopharmaceuticals/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bismuth/pharmacology , Cell Line, Tumor , Cell Survival , Female , Filipin/pharmacology , Humans , Iodine Radioisotopes/pharmacology , Lead Radioisotopes/pharmacology , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Radioisotopes/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , beta-Cyclodextrins/pharmacology
8.
EMBO Mol Med ; 10(4)2018 04.
Article in English | MEDLINE | ID: mdl-29438985

ABSTRACT

The clinical management of metastatic colorectal cancer (mCRC) faces major challenges. Here, we show that nilotinib, a clinically approved drug for chronic myeloid leukaemia, strongly inhibits human CRC cell invasion in vitro and reduces their metastatic potential in intrasplenic tumour mouse models. Nilotinib acts by inhibiting the kinase activity of DDR1, a receptor tyrosine kinase for collagens, which we identified as a RAS-independent inducer of CRC metastasis. Using quantitative phosphoproteomics, we identified BCR as a new DDR1 substrate and demonstrated that nilotinib prevents DDR1-mediated BCR phosphorylation on Tyr177, which is important for maintaining ß-catenin transcriptional activity necessary for tumour cell invasion. DDR1 kinase inhibition also reduced the invasion of patient-derived metastatic and circulating CRC cell lines. Collectively, our results indicate that the targeting DDR1 kinase activity with nilotinib may be beneficial for patients with mCRC.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Discoidin Domain Receptor 1/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Receptors, Collagen/metabolism , Animals , Discoidin Domain Receptor 1/genetics , HCT116 Cells , HEK293 Cells , Humans , Mice , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-bcr/genetics , Pyrimidines/pharmacology , Receptors, Collagen/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
9.
Endocr Relat Cancer ; 14(4): 1039-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045955

ABSTRACT

Recent data suggest that chemokines could be essential players in breast carcinogenesis. We previously showed that the CXC chemokine CXCL8 (interleukin-8) was overexpressed in estrogen receptor alpha (ERalpha)-negative breast cell lines. Analysis of CXCL8 chromosomal location showed that several CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL4V1, CXCL5, CXCL6, CXCL7, and CXCL8) were localized in the same narrow region (360 kb in size) of chromosome 4. We thus hypothesized that they could belong to the same cluster. Quantification of these chemokines in breast tumors showed that samples expressing high CXCL8 also produced elevated levels of CXCL1, CXCL3, and CXCL5, and displayed low content of ERalpha. CXCL1, CXCL2, CXCL3, CXCL5, and CXCL8 were co-regulated both in tumors and in breast cancer cell lines. CXCL5 and CXCL8 were mainly produced by epithelial cells, whereas CXCL1, CXCL2, and CXCL3 had a high expression in blood cells. The overexpression of these chemokines in tumor cells was not the result of gene amplification, but rather of an enhanced gene transcription. Our data suggest that high CXCL8 expression in tumors is mainly correlated to activating protein-1 (AP-1) pathway and to a minor extent to NF-kappaB pathway. Interestingly, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, and CXCL8 chemokines were present at higher levels in metastases when compared with grade I and III biopsies. High levels of CXCL8, CXCL1, and CXCL3 accounted for a shorter relapse-free survival of ERalpha-positive patients treated with tamoxifen. In summary, we present evidences that multiple CXC chemokines are co-expressed in CXCL8-positive breast tumors. In addition, these chemokines could account for the higher aggressiveness of these types of tumors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemokines, CXC/genetics , Chromosomes, Human, Pair 4 , Gene Expression Regulation, Neoplastic , Up-Regulation , Aged , Breast Neoplasms/surgery , Cells, Cultured , Chromosome Mapping , Disease-Free Survival , Endothelium, Vascular , Female , Humans , Lymphatic Metastasis , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins
10.
Mol Endocrinol ; 20(4): 749-63, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16322094

ABSTRACT

The regulation of gene expression by thyroid hormone (T3) involves binding of the hormone to nuclear receptors [thyroid hormone receptor (TR)] acting as T3-dependent transcription factors encoded by TRalpha (NR1A1) and TRbeta (NR1A2) genes. Several TRalpha variants have already been characterized, but only some of them display T3 binding activity. In this study, we have identified another transcript, TRalpha-DeltaE6, produced by alternative splicing with microexon 6b instead of exon 6. This splicing leads to the synthesis of a protein devoid of a hinge domain. The TRalpha-DeltaE6 transcript is detected in all mouse tissues tested. Although TRalpha-DeltaE6 did not bind DNA, its expression induced a TRalpha1 sequestration in the cytoplasm. Functional studies demonstrated that TRalpha-DeltaE6 inhibits the transcriptional activity of TRalpha1 and retinoic X receptor-alpha, but not of retinoic acid receptor-alpha. We also found that TRalpha-DeltaE6 efficiently decreased the ability of TRalpha to inhibit MyoD transcriptional activity during myoblast proliferation. Consequently, when overexpressed in myoblasts, it stimulated terminal differentiation. We suggest that this novel TRalpha variant may act as down regulator of overall T3 receptor activity, including its ability to repress MyoD transcriptional activity during myoblast proliferation.


Subject(s)
Myoblasts/cytology , Myoblasts/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cell Line , Cell Proliferation , Chickens , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression Regulation , Genetic Variation , In Vitro Techniques , Mice , Molecular Sequence Data , Quail , Rabbits , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Tissue Distribution
11.
Cancer Res ; 65(10): 4282-91, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899820

ABSTRACT

Mitochondrial dysfunctions are frequently reported in cancer cells, but their direct involvement in tumorigenesis remains unclear. To understand this relation, we stimulated mitochondrial activity by overexpression of the mitochondrial triiodothyronine receptor (p43) in human dermal fibroblasts. In all clones, this stimulation induced morphologic changes and cell fusion in myotube-like structures associated with the expression of several muscle-specific genes (Myf5, desmin, connectin, myosin, AchRalpha). In addition, these clones displayed all the in vivo and in vitro features of cell transformation. This phenotype was related to an increase in c-Jun and c-Fos expression and extinction of tumor suppressor gene expression (p53, p21WAF1, Rb3). Lastly, reactive oxygen species (ROS) production was increased in positive correlation to the stimulation of mitochondrial activity. The direct involvement of mitochondrial activity in this cell behavior was studied by adding chloramphenicol, an inhibitor of mitochondrial protein synthesis, to the culture medium. This inhibition resulted in partial restoration of the normal phenotype, with the loss of the ability to fuse, a strong decrease in muscle-specific gene expression, and potent inhibition of the transformed phenotype. However, expression of tumor suppressor genes was not restored. Similar results were obtained by using N-acetylcysteine, an inhibitor of ROS production. These data indicate that stimulation of mitochondrial activity in human dermal fibroblasts induces cell transformation through events involving ROS production.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cell Transformation, Neoplastic/pathology , Mitochondria/physiology , Peptide Elongation Factor Tu/biosynthesis , Skin/pathology , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Chloramphenicol/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/physiology , Humans , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins , Protein Synthesis Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
12.
Clin Cancer Res ; 23(11): 2806-2816, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27923843

ABSTRACT

Purpose: AXL receptor tyrosine kinase has been described as a relevant molecular marker and a key player in invasiveness, especially in triple-negative breast cancer (TNBC).Experimental Design: We evaluate the antitumor efficacy of the anti-AXL monoclonal antibody 20G7-D9 in several TNBC cell xenografts or patient-derived xenograft (PDX) models and decipher the underlying mechanisms. In a dataset of 254 basal-like breast cancer samples, genes correlated with AXL expression are enriched in EMT, migration, and invasion signaling pathways.Results: Treatment with 20G7-D9 inhibited tumor growth and bone metastasis formation in AXL-positive TNBC cell xenografts or PDX, but not in AXL-negative PDX, highlighting AXL role in cancer growth and invasion. In vitro stimulation of AXL-positive cancer cells by its ligand GAS6 induced the expression of several EMT-associated genes (SNAIL, SLUG, and VIM) through an intracellular signaling implicating the transcription factor FRA-1, important in cell invasion and plasticity, and increased their migration/invasion capacity. 20G7-D9 induced AXL degradation and inhibited all AXL/GAS6-dependent cell signaling implicated in EMT and in cell migration/invasion.Conclusions: The anti-AXL antibody 20G7-D9 represents a promising therapeutic strategy in TNBC with mesenchymal features by inhibiting AXL-dependent EMT, tumor growth, and metastasis formation. Clin Cancer Res; 23(11); 2806-16. ©2016 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Cell Proliferation/drug effects , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Triple Negative Breast Neoplasms/therapy , Animals , Antibodies, Anti-Idiotypic/immunology , Cell Movement/drug effects , Cell Movement/immunology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
13.
Oncogene ; 24(10): 1698-710, 2005 Mar 03.
Article in English | MEDLINE | ID: mdl-15674337

ABSTRACT

The btg1 (B-cell translocation gene 1) gene coding sequence was isolated from a translocation break point in a case of B-cell chronic lymphocytic leukaemia. We have already shown that BTG1, considered as an antiproliferative protein, strongly stimulates myoblast differentiation. However, the mechanisms involved in this influence remained unknown. In cultured myoblasts, we found that BTG1 stimulates the transcriptional activity of nuclear receptors (T3 and all-trans retinoic acid receptors but not RXRalpha and PPARgamma), c-Jun and myogenic factors (CMD1, Myf5, myogenin). Immunoprecipitation experiments performed in cells or using in vitro-synthesized proteins and GST pull-down assays established that BTG1 directly interacts with T3 and all-trans retinoic acid receptors and with avian MyoD (CMD1). These interactions are mediated by the transactivation domain of each transcription factor and the A box and C-terminal part of BTG1. NCoR presence induces the ligand dependency of the interaction with nuclear receptors. Lastly, deletion of BTG1 interacting domains abrogates its ability to stimulate nuclear receptors and CMD1 activity, and its myogenic influence. In conclusion, BTG1 is a novel important coactivator involved in the regulation of myoblast differentiation. It not only stimulates the activity of myogenic factors, but also of nuclear receptors already known as positive myogenic regulators.


Subject(s)
Cell Differentiation , Myoblasts/cytology , Neoplasm Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cells, Cultured , Humans , Hydroxamic Acids/pharmacology , Neoplasm Proteins/chemistry , Protein-Arginine N-Methyltransferases/physiology , Proto-Oncogene Proteins c-jun/physiology , Transcription, Genetic
14.
Endocrinology ; 147(7): 3408-18, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16556763

ABSTRACT

Although physical interactions with other receptors have been reported, heterodimeric complexes of T(3) nuclear receptors (TR) with retinoid X receptors (RXRs) are considered as major regulators of T(3) target gene expression. However, despite the potent T(3) influence in proliferating myoblasts, RXR isoforms are not expressed during proliferation, raising the question of the nature of the complex involved in TRalpha transcriptional activity. We have previously established that c-Jun induces TRalpha1 transcriptional activity in proliferating myoblasts not expressing RXR. This regulation is specific to the muscle lineage, suggesting the involvement of a muscle-specific factor. In this study, we found that MyoD expression in HeLa cells stimulates TRalpha1 activity, an influence potentiated by c-Jun coexpression. Similarly, in the absence of RXR, MyoD or c-Jun overexpression in myoblasts induces TRalpha1 transcriptional activity through a direct repeat 4 or an inverted palindrome 6 thyroid hormone response element. The highest rate of activity was recorded when c-Jun and MyoD were coexpressed. Using c-Jun-negative dominants, we established that MyoD influence on TRalpha1 activity needs c-Jun functionality. Furthermore, we demonstrated that TRalpha1 and MyoD physically interact in the hinge region of the receptor and the transactivation and basic helix loop helix domains of MyoD. RXR expression (spontaneously occurring at the onset of myoblast differentiation) in proliferating myoblasts abrogates these interactions. These data suggest that in the absence of RXR, TRalpha1 transcriptional activity in myoblasts is mediated through a complex including MyoD and c-Jun.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , MyoD Protein/physiology , Myoblasts/cytology , Proto-Oncogene Proteins c-jun/physiology , Receptors, Thyroid Hormone/metabolism , Thyroid Hormone Receptors alpha/biosynthesis , Transcription, Genetic , Triiodothyronine, Reverse/metabolism , Animals , Cell Proliferation , HeLa Cells , Humans , MyoD Protein/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Quail , Retinoid X Receptor alpha/metabolism
15.
Oncotarget ; 7(50): 82228-82243, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27429045

ABSTRACT

SOX9 inactivation is frequent in colorectal cancer (CRC) due to SOX9 gene mutations and/or to ectopic expression of MiniSOX9, a dominant negative inhibitor of SOX9. In the present study, we report a heterozygous L142P inactivating mutation of SOX9 in the DLD-1 CRC cell line and we demonstrate that the conditional expression of a wild type SOX9 in this cell line inhibits cell growth, clonal capacity and colonosphere formation while decreasing both the activity of the oncogenic Wnt/ß-catenin signaling pathway and the expression of the c-myc oncogene. This activity does not require SOX9 transcriptional function but, rather, involves an interaction of SOX9 with nuclear ß-catenin. Furthermore, we report that SOX9 inhibits tumor development when conditionally expressed in CRC cells injected either subcutaneous or intraperitoneous in BALB/c mice as an abdominal metastasis model. These observations argue in favor of a tumor suppressor activity for SOX9. As an siRNA targeting SOX9 paradoxically also inhibits DLD-1 and HCT116 CRC cell growth, we conclude that there is a critical level of endogenous active SOX9 needed to maintain CRC cell growth.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , SOX9 Transcription Factor/metabolism , Wnt Signaling Pathway , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterozygote , Humans , Mice, Inbred BALB C , Mutation , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , SOX9 Transcription Factor/genetics , Time Factors , Transfection , Tumor Burden , beta Catenin/genetics , beta Catenin/metabolism
16.
FASEB J ; 17(3): 426-36, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631582

ABSTRACT

The importance of mitochondrial activity has recently been extended to the regulation of developmental processes. Numerous pathologies associated with organelle's dysfunctions emphasize their physiological importance. However, regulation of mitochondrial genome transcription, a key element for organelle's function, remains poorly understood. After characterization in the organelle of a truncated form of the triiodothyronine nuclear receptor (p43), a T3-dependent transcription factor of the mitochondrial genome, our purpose was to search for other mitochondrial receptors involved in the regulation of organelle transcription. We show that a 44 kDa protein related to RXRalpha (mt-RXR), another nuclear receptor, is located in the mitochondrial matrix. We found that mt-RXR is produced after cytosolic or intramitochondrial enzymatic cleavage of the RXRalpha nuclear receptor. After mitochondrial import and binding to specific sequences of the organelle genome, mt-RXR induces a ligand-dependent increase in mitochondrial RNA levels. mt-RXR physically interacts with p43 and acts alone or through a heterodimerical complex activated by 9-cis-retinoic acid and T3 to increase RNA levels. These data indicate that hormonal regulation of mitochondrial transcription occurs through pathways similar to those that take place in the nucleus and open a new way to better understand hormone and vitamin action at the cellular level.


Subject(s)
Mitochondria/genetics , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/physiology , Thyroid Hormone Receptors alpha/chemistry , Thyroid Hormone Receptors alpha/physiology , Transcription Factors/chemistry , Transcription Factors/physiology , Alitretinoin , Animals , Binding Sites , Calpain/metabolism , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Genome , Ligands , Male , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/ultrastructure , Models, Biological , Protein Transport , RNA/biosynthesis , RNA, Mitochondrial , Rats , Rats, Wistar , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Transcription, Genetic , Tretinoin/pharmacology , Triiodothyronine/pharmacology
17.
Nat Commun ; 6: 6218, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645278

ABSTRACT

Osteoporosis is caused by excessive activity of bone-degrading osteoclasts over bone-forming osteoblast. Standard antiosteolytic treatments inhibit bone resorption by inducing osteoclast loss, with the adverse effect of hindering also bone formation. Formation of the osteoclast sealing zone requires Dock5, a guanine nucleotide exchange factor for the small GTPase Rac, and C21, a chemical inhibitor of Dock5, decreases bone resorption by cultured osteoclasts. Here we show that C21 directly inhibits the exchange activity of Dock5 and disrupts osteoclast podosome organization. Remarkably, C21 administration protects mice against bone degradation in models recapitulating major osteolytic diseases: menopause, rheumatoid arthritis and bone metastasis. Furthermore, C21 administration does not affect bone formation and is not toxic. Our results validate the pharmacological inhibition of Dock5 as a novel therapeutic route for fighting osteolytic diseases while preserving bone formation.


Subject(s)
Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteolysis/drug therapy , Sulfonamides/therapeutic use , Animals , Arthritis/chemically induced , Arthritis/drug therapy , Female , Male , Mice , Mice, Inbred C57BL , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Sulfonamides/chemistry , Sulfonamides/pharmacology , Benzenesulfonamides
18.
Stem Cell Reports ; 4(4): 531-40, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25754205

ABSTRACT

Cancer stem cells (CSC) are responsible for cancer chemoresistance and metastasis formation. Here we report that Δ133p53ß, a TP53 splice variant, enhanced cancer cell stemness in MCF-7 breast cancer cells, while its depletion reduced it. Δ133p53ß stimulated the expression of the key pluripotency factors SOX2, OCT3/4, and NANOG. Similarly, in highly metastatic breast cancer cells, aggressiveness was coupled with enhanced CSC potential and Δ133p53ß expression. Like in MCF-7 cells, SOX2, OCT3/4, and NANOG expression were positively regulated by Δ133p53ß in these cells. Finally, treatment of MCF-7 cells with etoposide, a cytotoxic anti-cancer drug, increased CSC formation and SOX2, OCT3/4, and NANOG expression via Δ133p53, thus potentially increasing the risk of cancer recurrence. Our findings show that Δ133p53ß supports CSC potential. Moreover, they indicate that the TP53 gene, which is considered a major tumor suppressor gene, also acts as an oncogene via the Δ133p53ß isoform.


Subject(s)
Alternative Splicing , Cell Self Renewal/genetics , Neoplastic Stem Cells/metabolism , RNA Isoforms , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Spheroids, Cellular , Transcription Factors/genetics , Tumor Cells, Cultured
19.
MAbs ; 6(5): 1314-26, 2014.
Article in English | MEDLINE | ID: mdl-25517316

ABSTRACT

Ovarian cancer has the highest mortality rate among gynecologic malignancies. The monoclonal antibody 12G4 specifically recognizes the human Müllerian inhibiting substance type II receptor (MISRII) that is strongly expressed in human granulosa cell tumors (GCT) and in the majority of human epithelial ovarian cancers (EOC). To determine whether MISRII represents an attractive target for antibody-based tumor therapy, we first confirmed by immunohistochemistry with 12G4 its expression in all tested GCT samples (4/4) and all, but one, EOC human tissue specimens (13/14). We then demonstrated in vitro the internalization of 12G4 in MISRII(high)COV434 cells after binding to MISRII and its ability to increase the apoptosis rate (FACS, DNA fragmentation) in MISRII(high)COV434 (GCT) and MISRII(medium)NIH-OVCAR-3 (EOC) cells that express different levels of MISRII. A standard (51)Cr release assay showed that 12G4 mediates antibody-dependent cell-meditated cytotoxicity. Finally, in vivo assessment of 12G4 anti-tumor effects showed a significant reduction of tumor growth and an increase of the median survival time in mice xenografted with MISRII(high)COV434 or MISRII(medium)NIH-OVCAR-3 cells and treated with 12G4 in comparison to controls treated with an irrelevant antibody. Altogether, our data indicate that MISRII is a new promising target for the control of ovarian GCTs and EOCs. A humanized version of the 12G4 antibody, named 3C23K, is in development for the targeted therapy of MISRII-positive gynecologic cancers.


Subject(s)
Antibodies, Monoclonal/immunology , Ovarian Neoplasms/immunology , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Apoptosis/drug effects , Apoptosis/immunology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Granulosa Cell Tumor/immunology , Granulosa Cell Tumor/metabolism , Granulosa Cell Tumor/therapy , Humans , Immunohistochemistry , Immunotherapy/methods , Mice, Nude , Microscopy, Fluorescence , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
20.
PLoS One ; 8(7): e69613, 2013.
Article in English | MEDLINE | ID: mdl-23922757

ABSTRACT

BACKGROUND AND PURPOSE: We assessed the contribution of antibody internalization in the efficacy and toxicity of intraperitoneal α-radioimmunotherapy (RIT) of small volume carcinomatosis using (212)Pb-labeled monoclonal antibodies (mAbs) that target HER2 (internalizing) or CEA (non-internalizing) receptors. MATERIALS AND METHODS: Athymic nude mice bearing 2-3 mm intraperitoneal tumor xenografts were intraperitoneally injected with similar activities (370, 740 and 1480 kBq; 37 MBq/mg) of (212)Pb-labeled 35A7 (anti-CEA), trastuzumab (anti-HER2) or PX (non-specific) mAbs, or with equivalent amounts of unlabeled mAbs, or with NaCl. Tumor volume was monitored by bioluminescence and survival was reported. Hematologic toxicity and body weight were assessed. Biodistribution of (212)Pb-labeled mAbs and absorbed dose-effect relationships using MIRD formalism were established. RESULTS: Transient hematological toxicity, as revealed by white blood cells and platelets numbering, was reported in mice treated with the highest activities of (212)Pb-labeled mAbs. The median survival (MS) was significantly higher in mice injected with 1.48 MBq of (212)Pb-35A7 (non-internalizing mAbs) (MS = 94 days) than in animals treated with the same activity of (212)Pb-PX mAbs or with NaCl (MS = 18 days). MS was even not reached after 130 days when follow-up was discontinued in mice treated with 1.48 MBq of (212)Pb-trastuzumab. The later efficacy was unexpected since final absorbed dose resulting from injection of 1.48 MBq, was higher for (212)Pb-35A7 (35.5 Gy) than for (212)Pb-trastuzumab (27.6 Gy). These results also highlight the lack of absorbed dose-effect relationship when mean absorbed dose was calculated using MIRD formalism and the requirement to perform small-scale dosimetry. CONCLUSIONS: These data indicate that it might be an advantage of using internalizing anti-HER2 compared with non-internalizing anti-CEA (212)Pb-labeled mAbs in the therapy of small volume xenograft tumors. They support clinical investigations of (212)Pb-mAbs RIT as an adjuvant treatment after cytoreductive surgery in patients with peritoneal carcinomatosis.


Subject(s)
Antibodies, Monoclonal/immunology , Lead Radioisotopes , Peritoneal Neoplasms/diagnosis , Radioimmunotherapy/methods , Receptor, ErbB-2/immunology , Receptors, Cell Surface/immunology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Receptor, ErbB-2/metabolism , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL