Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Cancer Cell Int ; 24(1): 132, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38594745

ABSTRACT

BACKGROUND: The metabolism of cancer cells generally differs from that of normal cells. Indeed, most cancer cells have a high rate of glycolysis, even at normal oxygen concentrations. These metabolic properties can potentially be exploited for therapeutic intervention. In this context, we have developed troglitazone derivatives to treat hormone-sensitive and triple-negative breast cancers, which currently lack therapeutic targets, have an aggressive phenotype, and often have a worse prognosis than other subtypes. Here, we studied the metabolic impact of the EP13 compound, a desulfured derivative of Δ2-troglitazone that we synthetized and is more potent than its parent compounds. METHODS: EP13 was tested on two triple-negative breast cancer cell lines, MDA-MB-231 and Hs578T, and on the luminal cell line MCF-7. The oxygen consumption rate (OCR) of the treated cell lines, Hs578T mammospheres and isolated mitochondria was measured using the XFe24 Seahorse analyser. ROS production was quantified using the MitoSOX fluorescent probe. Glycolytic activity was evaluated through measurement of the extracellular acidification rate (ECAR), glucose consumption and lactate production in extracellular medium. The synergistic effect of EP13 with glycolysis inhibitors (oxamate and 2-deoxyglucose) on cell cytotoxicity was established using the Chou-Talalay method. RESULTS: After exposure to EP13, we observed a decrease in the mitochondrial oxygen consumption rate in MCF7, MDA-MB-231 and Hs578T cells. EP13 also modified the maximal OCR of Hs578T spheroids. EP13 reduced the OCR through inhibition of respiratory chain complex I. After 24 h, ATP levels in EP13-treated cells were not altered compared with those in untreated cells, suggesting compensation by glycolysis activity, as shown by the increase in ECAR, the glucose consumption and lactate production. Finally, we performed co-treatments with EP13 and glycolysis inhibitors (oxamate and 2-DG) and observed that EP13 potentiated their cytotoxic effects. CONCLUSION: This study demonstrates that EP13 inhibits OXPHOS in breast cancer cells and potentiates the effect of glycolysis inhibitors.

2.
ACS Biomater Sci Eng ; 9(2): 1020-1029, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36720461

ABSTRACT

We present the design, fabrication, and characterization of an implantable neural interface based on anisotropic magnetoresistive (AMR) magnetic-field sensors that combine reduced size and high performance at body temperature. The sensors are based on La0.67Sr0.33MnO3 (LSMO) as a ferromagnetic material, whose epitaxial growth has been suitably engineered to get uniaxial anisotropy and large AMR output together with low noise even at low frequencies. The performance of LSMO sensors of different film thickness and at different temperatures close to 37 °C has to be explored to find an optimum sensitivity of ∼400%/T (with typical detectivity values of 2 nT·Hz-1/2 at a frequency of 1 Hz and 0.3 nT·Hz-1/2 at 1 kHz), fitted for the detection of low magnetic signals coming from neural activity. Biocompatibility tests of devices consisting of submillimeter-size LSMO sensors coated by a thin poly(dimethyl siloxane) polymeric layer, both in vitro and in vivo, support their high suitability as implantable detectors of low-frequency biological magnetic signals emerging from heterogeneous electrically active tissues.


Subject(s)
Magnetic Fields , Prostheses and Implants , Anisotropy , Polymers
3.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743305

ABSTRACT

Breast cancer is one of the leading causes of cancer-related death among females worldwide. A major challenge is to develop innovative therapy in order to treat breast cancer subtypes resistant to current treatment. In the present study, we examined the effects of two Troglitazone derivatives Δ2-TGZ and AB186. Previous studies showed that both compounds induce apoptosis, nevertheless AB186 was a more potent agent. The kinetic of cellular events was investigated by real-time cell analysis system (RTCA) in MCF-7 (hormone dependent) and MDA-MB-231 (triple negative) breast cancer (TNBC) cells, followed by cell morphology analysis by immuno-localization. Both compounds induced a rapid modification of both impedance-based signals and cellular morphology. This process was associated with an inhibition of cell migration measured by wound healing and transwell assays in TNBC MDA-MB-231 and Hs578T cells. In order to identify cytoplasmic targets of AB186, we performed surface plasmon resonance (SPR) and pull-down analyses. Subsequently, 6 cytoskeleton components were identified as potential targets. We further validated α-tubulin as one of the direct targets of AB186. In conclusion, our results suggested that AB186 could be promising to develop novel therapeutic strategies to treat aggressive forms of breast cancer such as TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Triple Negative Breast Neoplasms/metabolism , Tubulin
5.
Eur J Med Chem ; 187: 111939, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31838327

ABSTRACT

Breast cancer is a major medical threat which cannot be sufficiently addressed by current therapies because of spontaneous or acquired treatment resistance. Besides, triple-negative breast cancer (TNBC) tumors do not respond to targeted therapies, thus new therapeutic strategies are needed. In this context, we designed and prepared new desulfured troglitazone (TGZ)-derived molecules and evaluated them in vitro for their anti-proliferative activity, with a special focus on triple-negative breast cancer cell lines. Optimization of the synthetic strategies and deracemization of the lead compound were performed to give highly active compound 10 with low-micromolar potency. Further studies revealed that this compound triggers apoptosis rather than cell cycle arrest as observed with TGZ.


Subject(s)
Antineoplastic Agents/pharmacology , Troglitazone/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Troglitazone/chemical synthesis , Troglitazone/chemistry
6.
Eur J Med Chem ; 158: 334-352, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30223121

ABSTRACT

Because of the complex biological networks, many pathologic disorders fail to be treated with a molecule directed towards a single target. Thus, combination therapies are often necessary, but they have many drawbacks. An alternative consists in building molecules intended to interact with multiple targets, called designed multiple ligands. We followed such a strategy in order to treat metabolic syndrome, by setting up molecules directed towards both type 1 angiotensin II (AT1) receptor and peroxisome proliferator-activated receptor-γ (PPAR-γ). For this purpose, many molecules were prepared by merging both pharmacophores following three different strategies. Their ability to activate PPAR-γ and to block AT1 receptors were evaluated in vitro. This strategy led to the preparation of many new PPAR-γ activating and AT1 blocking molecules. Among them, some exhibited both activities, highlighting the convenience of this approach.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Drug Design , PPAR gamma/agonists , Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Animals , Chromans/chemical synthesis , Chromans/chemistry , Chromans/pharmacology , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Ligands , MCF-7 Cells , Male , Molecular Docking Simulation , PPAR gamma/metabolism , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology
7.
Breast Cancer Res Treat ; 171(3): 581-591, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29971627

ABSTRACT

PURPOSE: Cancer cells often elicit a higher glycolytic rate than normal cells, supporting the development of glycolysis inhibitors as therapeutic agents. 2-Deoxyglucose (2-DG) is used in this context due to its ability to compete with glucose. However, many studies do not take into account that 2-DG inhibits not only glycolysis but also N-glycosylation. Since there are limited publications on 2-DG mechanism of action in breast cancer, we studied its effects in breast cancer cell lines to determine the part played by glycolysis inhibition and N-linked glycosylation interference. METHODS AND RESULTS: 2-Deoxyglucose behaved as an anticancer agent with a similar efficiency on cell number decrease between the hormone-dependent MCF-7 and hormone-independent MDA-MB-231 breast cancer cells. It also interfered with the N-linked glycosylation process in both cell lines as illustrated by the migration profile of the lysosomal-associated membrane protein 2 and calumenin. These results are reinforced by the appearance of an abnormal Man7GlcNAc2 structure both on lipid-linked oligosaccharides and N-linked glycoproteins of 2-DG incubated MDA-MB-231 cells. Besides, 2-DG-induced a transient endoplasmic reticulum stress that was more sustained in MDA-MB-231 cells. Both changes were abrogated by mannose. 2-DG, even in the presence of mannose, decreased glycolysis in both cell lines. Mannose partially reversed the effects of 2-DG on cell numbers with N-linked glycosylation interference accounting for 37 and 47% of 2-DG anti-cancerous effects in MDA-MB-231 and MCF-7 cells, respectively. CONCLUSION: N-linked glycosylation interference and glycolysis disruption both contribute to the anticancer properties of 2-DG in breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Deoxyglucose/pharmacology , Glycolysis/drug effects , Glycosylation/drug effects , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Glucose/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/chemistry , Humans , MCF-7 Cells
8.
Int J Oncol ; 52(6): 1991-2000, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620161

ABSTRACT

15-Deoxy-∆12,14-prostaglandin J2 (15d­PGJ2) is a natural agonist of peroxisome proliferator-activated receptor Î³ (PPARγ) that displays anticancer activity. Various studies have indicated that the effects of 15d­PGJ2 are due to both PPARγ-dependent and -independent mechanisms. In the present study, we examined the effects of a biotinylated form of 15d­PGJ2 (b­15d­PGJ2) on hormone-dependent MCF­7 and triple­negative MDA­MB­231 breast cancer cell lines. b­15d­PGJ2 inhibited cell proliferation more efficiently than 15d­PGJ2 or the synthetic PPARγ agonist, efatutazone. b­15d­PGJ2 was also more potent than its non-biotinylated counterpart in inducing apoptosis. We then analyzed the mechanisms underlying this improved efficiency. It was found not to be the result of biotin receptor-mediated increased incorporation, since free biotin in the culture medium did not decrease the anti-proliferative activity of b­15d­PGJ2 in competition assays. Of note, b­15d­PGJ2 displayed an improved PPARγ agonist activity, as measured by transactivation experiments. Molecular docking analyses revealed a similar insertion of b­15d­PGJ2 and 15d­PGJ2 into the ligand binding domain of PPARγ via a covalent bond with Cys285. Finally, PPARγ silencing markedly decreased the cleavage of the apoptotic markers, poly(ADP-ribose) polymerase 1 (PARP­1) and caspase­7, that usually occurs following b­15d­PGJ2 treatment. Taken together, our data indicate that biotinylation enhances the anti-proliferative and pro-apoptotic activity of 15d­PGJ2, and that this effect is partly mediated via a PPARγ-dependent pathway. These results may aid in the development of novel therapeutic strategies for breast cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , PPAR gamma/chemistry , Prostaglandin D2/analogs & derivatives , Binding Sites/genetics , Biotinylation/methods , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Models, Molecular , Molecular Docking Simulation , PPAR gamma/agonists , PPAR gamma/genetics , Prostaglandin D2/chemistry , Prostaglandin D2/pharmacology , Thiazolidinediones/pharmacology
10.
Breast Cancer Res Treat ; 165(3): 517-527, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28681173

ABSTRACT

PURPOSE: 40% of triple-negative breast cancer (TNBC) do not express claudin-1, a major constituent of tight junction. Patients with these "claudin-1-low" tumors present a higher relapse incidence. A major challenge in oncology is the development of innovative therapies for such poor prognosis tumors. In this context, we study the anticancer effects of ∆2-TGZ, a compound derived from troglitazone (TGZ), on cell models of these tumors. METHODS AND RESULTS: In MDA-MB-231 and Hs578T "claudin-1-low" TNBC cells, Δ2-TGZ treatment induced claudin-1 protein expression and triggered apoptosis as measured by FACS analysis (annexin V/PI co-staining). Interestingly, in the non-tumorigenic human breast epithelial cell line MCF-10A, the basal level of claudin-1 was not modified following Δ2-TGZ treatment, which did not induce apoptosis. Furthermore, claudin-1-transfected MDA-MB-231 and Hs578T cells displayed a significant increase of cleaved PARP-1 and caspase 7, caspase 3/7 activities, and TUNEL staining. RNA interference was performed in order to inhibit Δ2-TGZ-induced claudin-1 expression in both the cells. In absence of claudin-1, a decrease of cleaved PARP-1 and caspase 7 and caspase 3/7 activities were observed in MDA-MB-231 but not in Hs578T cells. CONCLUSION: Claudin-1 overexpression and Δ2-TGZ treatment are associated to apoptosis in MDA-MB-231 and Hs578T "claudin-1-low" TNBC. Moreover, in MDA-MB-231 cells, claudin-1 is involved in the pro-apoptotic effect of Δ2-TGZ. Our results suggest that claudin-1 re-expression could be an interesting therapeutic strategy for "claudin-1-low" TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Claudin-1/metabolism , Sulfuric Acid Esters/pharmacology , Thiazolidinediones/pharmacology , Triple Negative Breast Neoplasms/metabolism , Apoptosis/genetics , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Claudin-1/genetics , Female , Gene Expression Regulation , Humans , Protein Transport , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Troglitazone
11.
Cell Cycle ; 15(24): 3402-3412, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27753533

ABSTRACT

We have previously shown that Δ2-Troglitazone (Δ2-TGZ) displayed anticancer effects on breast cancer cell lines grown in low serum conditions (1% fetal calf serum (FCS)). The present study was performed in order to characterize the effects of Δ2-TGZ in high serum containing medium and to determine if starvation could influence the response of breast cancer cells to this compound, keeping in mind the potential interest for breast cancer therapy. We observed that in high serum conditions (10% FCS), a 48 h treatment with Δ2-TGZ induced a decrease in cell numbers in MDA-MB-231 and MCF-7 breast cancer cell lines. The IC50 values were higher than in low serum conditions. Furthermore, in contrast to our previous results obtained in 1% FCS conditions, we observed that in 10% FCS-containing medium, MCF-7 cells were more sensitive to Δ2-TGZ than MDA-MB-231 cells. Δ2-TGZ also induced endoplasmic reticulum (ER) stress mainly in MDA-MB-231 cells. Besides, in high serum conditions, Δ2-TGZ induced a G0/G1 cell cycle arrest, an inhibition of BrdU incorporation and a reduced level of cyclin D1. We observed a limited cleavage of PARP and a limited proportion of cells in sub-G1 phase. Thus, in high serum conditions, Δ2-TGZ displayed cytostatic effects rather than apoptosis as previously reported in 1% FCS-containing medium. Our results are in accordance with studies suggesting that serum starvation could potentiate the action of diverse anti-cancer agents.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Chromans/pharmacology , Serum/metabolism , Thiazolidinediones/pharmacology , Cell Count , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Transcription Factor CHOP/metabolism , Troglitazone , Tumor Cells, Cultured
12.
Sex Dev ; 10(5-6): 267-278, 2016.
Article in English | MEDLINE | ID: mdl-27648840

ABSTRACT

Amphibians have been widely used to study developmental biology due to the fact that embryo development takes place independently of the maternal organism and that observations and experimental approaches are easy. Some amphibians like Xenopus became model organisms in this field. In the first part of this article, the differentiation of the gonads in amphibians and the mechanisms governing this process are reviewed. In the second part, the state of the art about sex reversal, which can be induced by steroid hormones in general and by temperature in some species, is presented. Also information about pollutants found in the environment that could interfere with the development of the amphibian reproductive apparatus or with their reproductive physiology is given. Such compounds could play a part in the amphibian decline, since in the wild, many amphibians are endangered species.


Subject(s)
Amphibians/genetics , Amphibians/physiology , Animals , Disorders of Sex Development/genetics , Gonads/metabolism , Gonads/physiology , Reproduction/genetics , Reproduction/physiology , Sex Differentiation/genetics , Sex Differentiation/physiology
13.
Curr Top Med Chem ; 16(19): 2115-24, 2016.
Article in English | MEDLINE | ID: mdl-26881718

ABSTRACT

The existence of unresponsive tumors and the appearance of resistant tumors during the course of treatments both justify that we increase urgently the panel of pharmacological molecules able to fight cancer. An interesting strategy is drug reprofiling (also known as drug repositioning, drug repurposing or drug retasking) that consists of identifying and developing new uses for existing drugs. This review illustrates drug reprofiling with troglitazone (TGZ), a synthetic PPARγ agonist initially used for the treatment of type II diabetes. The fact that TGZ also displays anticancer effects is known since the end of the nineties but its development as an anticancer agent was slowed down due to hepatotoxic side effects. Part of the knowledge available for TGZ, mainly the molecular basis for PPARγ activation, its metabolization pathways and the side effects on hepatocytes, were taken into account to elaborate new molecules. Key findings were that unsaturated TGZ derivatives, when compared to TGZ, do not activate PPARγ, exhibit a higher efficiency on cancer cells and a lower toxicity towards hepatocytes. However, a weakness is that the mechanisms involved in the anticancer effects are still not completely understood and that the efficiency of such derivatives has not yet been completely studied in vivo. Data about this point should become available very soon from animal models and this will be a prerequisite to initiate clinical trials with these potential new anticancer drugs developed from a drug repurposing strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Chromans/pharmacology , Drug Repositioning/methods , Thiazolidinediones/pharmacology , Animals , Antineoplastic Agents/chemistry , Chromans/adverse effects , Chromans/chemistry , Diabetes Mellitus, Type 2/drug therapy , Hepatocytes/drug effects , Humans , Liver/drug effects , PPAR gamma/metabolism , Structure-Activity Relationship , Thiazolidinediones/adverse effects , Thiazolidinediones/chemistry , Troglitazone
14.
BMC Syst Biol ; 9: 28, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26080803

ABSTRACT

BACKGROUND: Estrogen receptor alpha36 (ERalpha36), a variant of estrogen receptor alpha (ER) is expressed in about half of breast tumors, independently of the [ER+]/[ER-] status. In vitro, ERalpha36 triggers mitogenic non-genomic signaling and migration ability in response to 17beta-estradiol and tamoxifen. In vivo, highly ERalpha36 expressing tumors are of poor outcome especially as [ER+] tumors are submitted to tamoxifen treatment which, in turn, enhances ERalpha36 expression. RESULTS: Our study aimed to validate ERalpha36 expression as a reliable prognostic factor for cancer progression from an estrogen dependent proliferative tumor toward an estrogen dispensable metastatic disease. In a retrospective study, we tried to decipher underlying mechanisms of cancer progression by using an original modeling of the relationships between ERalpha36, other estrogen and growth factor receptors and metastatic marker expression. Nonlinear correlation analyses and mutual information computations led to characterize a complex network connecting ERalpha36 to either non-genomic estrogen signaling or to metastatic process. CONCLUSIONS: This study identifies ERalpha36 expression level as a relevant classifier which should be taken into account for breast tumors clinical characterization and [ER+] tumor treatment orientation, using a generic approach for the rapid, cheap and relevant evaluation of any candidate gene expression as a predictor of a complex biological process.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Computational Biology/methods , Disease Progression , Estrogen Receptor alpha/genetics , Genetic Variation , Breast Neoplasms/classification , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Neoplasm Metastasis , Prognosis
15.
Mol Carcinog ; 54(5): 393-404, 2015 May.
Article in English | MEDLINE | ID: mdl-24293218

ABSTRACT

Our aim was to better understand peroxisome proliferator-activated receptor gamma (PPARγ)-independent pathways involved in anti-cancer effects of thiazolidinediones (TZDs). We focused on Δ2-troglitazone (Δ2-TGZ), a PPARγ inactive TZD that affects breast cancer cell viability. Appearance of TUNEL positive cells, changes in mitochondrial membrane potential, cleavage of poly(ADP-ribose) polymerase (PARP)-1 and caspase-7 revealed that apoptosis occurred in both hormone-dependent MCF7 and hormone-independent MDA-MB-231 breast cancer cells after 24 and 48 h of treatment. A microarray study identified endoplasmic reticulum (ER) stress as an essential cellular function since many genes involved in ER stress were upregulated in MCF7 cells following Δ2-TGZ treatment. Δ2-TGZ-induced ER stress was further confirmed in MCF7 cells by phosphorylation of pancreatic endoplasmic reticulum kinase-like endoplasmic reticulum kinase (PERK) and its target eIF2α after 1.5 h, rapid increase in activating transcription factor (ATF) 3 mRNA levels, splicing of X-box binding protein 1 (XBP1) after 3 h, accumulation of binding immunogloblulin protein (BiP) and CCAAT-enhancer-binding protein homologous protein (CHOP) after 6 h. Immunofluorescence microscopy indicated that CHOP was relocalized to the nucleus of treated cells. Similarly, in MDA-MB-231 cells, overexpression of ATF3, splicing of XBP1, and accumulation of BiP and CHOP were observed following Δ2-TGZ treatment. In MCF7 cells, knock-down of CHOP or the inhibition of c-Jun N-terminal kinase (JNK) did not impair cleavage of PARP-1 and caspase-7. Altogether, our results show that ER stress is an early response of major types of breast cancer cells to Δ2-TGZ, prior to, but not causative of apoptosis.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Chromans/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/drug effects , Hypoglycemic Agents/pharmacology , PPAR gamma/antagonists & inhibitors , Thiazolidinediones/pharmacology , Biomarkers, Tumor , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chromans/chemistry , Endoplasmic Reticulum/metabolism , Female , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thiazolidinediones/chemistry , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Troglitazone , Tumor Cells, Cultured
16.
Biochem Pharmacol ; 92(1): 102-11, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25083915

ABSTRACT

The use of energy restriction mimetic agents (ERMAs) to selectively target cancer cells addicted to glycolysis could be a promising therapeutic approach. Thiazolidinediones (TZDs) are synthetic agonists of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)γ that were developed to treat type II diabetes. These compounds also display anticancer effects which appear mainly to be independent of their PPARγ agonist activity but the molecular mechanisms involved in the anticancer action are not yet well understood. Results obtained on ciglitazone derivatives, mainly in prostate cancer cell models, suggest that these compounds could act as ERMAs. In the present paper, we introduce how compounds like 2-deoxyglucose target the Warburg effect and then we discuss the possibility that the PPARγ-independent effects of various TZD could result from their action as ERMAs.


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxyglucose/pharmacology , Energy Metabolism/drug effects , Neoplasms/drug therapy , Thiazolidinediones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Deoxyglucose/therapeutic use , Humans , Neoplasms/pathology , Thiazolidinediones/therapeutic use
17.
Eur J Med Chem ; 83: 129-40, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-24953030

ABSTRACT

Δ2-Troglitazone derivatives were shown to exhibit anti-proliferative activity in a PPARγ-independent manner. We prepared various compounds in order to increase their potency and decrease their toxicity towards non-malignant primary cultured hepatocytes. Many compounds induced viabilities less than 20% at 10 µM on various cancer cell lines. Furthermore, five of them showed hepatocyte viability of 80% or more at 200 µM. In addition, compounds 17 and 18 exhibited promising maximum tolerated doses on a murine model, enabling future investigations.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Chromans/pharmacology , Chromans/toxicity , Drug Design , Thiazolidinediones/pharmacology , Thiazolidinediones/toxicity , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromans/chemistry , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Thiazolidinediones/chemistry , Troglitazone
18.
Endocrinology ; 154(10): 3931-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24025226

ABSTRACT

Müllerian inhibiting substance (MIS, also known as anti-Müllerian hormone), is a key factor of male sex differentiation in vertebrates. In amniotes, it is responsible for Müllerian duct regression in male embryos. In fish, despite the absence of Müllerian ducts, MIS is produced and controls germ cell proliferation during gonad differentiation. Here we show for the first time the presence of MIS in an amphibian species, Pleurodeles waltl. This is very astonishing because in caudate amphibians, Müllerian ducts do not regress in males. Phylogenetic analysis of MIS P. waltl ortholog revealed that the deduced protein segregates with MIS from other vertebrates and is clearly separated from other TGF-ß family members. In larvae, MIS mRNA was expressed at higher levels in the developing testes than in the ovaries. In the testis, MIS mRNA expression was located within the lobules that contain Sertoli cells. Besides, expression of MIS was modified in the case of sex reversal: it increased after masculinizing heat treatment and decreased after estradiol feminizing exposure. In addition to the data obtained recently in the fish medaka, our results suggest that the role of MIS on Müllerian ducts occurred secondarily during the course of evolution.


Subject(s)
Amphibian Proteins/metabolism , Anti-Mullerian Hormone/metabolism , Gene Expression Regulation, Developmental , Ovary/metabolism , Pleurodeles/physiology , Testis/metabolism , Amphibian Proteins/biosynthesis , Amphibian Proteins/chemistry , Amphibian Proteins/genetics , Animals , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/chemistry , Anti-Mullerian Hormone/genetics , Female , In Situ Hybridization , Larva/growth & development , Larva/metabolism , Liver/growth & development , Liver/metabolism , Male , Metamorphosis, Biological , Mullerian Ducts/growth & development , Mullerian Ducts/metabolism , Organ Culture Techniques , Ovary/growth & development , Phylogeny , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sex Differentiation , Testis/cytology , Testis/growth & development
19.
PLoS One ; 8(4): e61758, 2013.
Article in English | MEDLINE | ID: mdl-23626723

ABSTRACT

Long chain alkylphenols are man-made compounds still present in industrial and agricultural processes. Their main use is domestic and they are widespread in household products, cleansers and cosmetics, leading to a global environmental and human contamination. These molecules are known to exert estrogen-like activities through binding to classical estrogen receptors. In vitro, they can also interact with the G-protein coupled estrogen receptor. Testicular germ cell tumor etiology and progression are proposed to be stimulated by lifelong estrogeno-mimetic exposure. We studied the transduction signaling pathways through which an alkyphenol mixture triggers testicular cancer cell proliferation in vitro and in vivo. Proliferation assays were monitored after exposure to a realistic mixture of 4-tert-octylphenol and 4-nonylphenol of either TCam-2 seminoma derived cells, NT2/D1 embryonal carcinoma cells or testis tumor in xenografted nude mice. Specific pharmacological inhibitors and gene-silencing strategies were used in TCam-2 cells in order to demonstrate that the alkylphenol mix triggers CREB-phosphorylation through a rapid, ERα36-PI3kinase non genomic pathway. Microarray analysis of the mixture target genes revealed that this pathway can modulate the expression of the DNA-methyltransferase-3 (Dnmt3) gene family which is involved in DNA methylation control. Our results highlight a key role for ERα36 in alkylphenol non genomic signaling in testicular germ cell tumors. Hence, ERα36-dependent control of the epigenetic status opens the way for the understanding of the link between endocrine disruptor exposure and the burden of hormone sensitive cancers.


Subject(s)
Carcinogens, Environmental/pharmacology , Carcinoma/genetics , Estrogen Receptor alpha/genetics , Phenols/pharmacology , Seminoma/genetics , Testicular Neoplasms/genetics , Androstadienes/pharmacology , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Epigenesis, Genetic/drug effects , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Seminoma/metabolism , Seminoma/pathology , Signal Transduction , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Wortmannin
20.
Eur J Med Chem ; 51: 206-15, 2012 May.
Article in English | MEDLINE | ID: mdl-22409968

ABSTRACT

Breast cancer is the most prevalent cancer in women. The development of resistances to therapeutic agents and the absence of targeted therapy for triple negative breast cancer motivate the search for alternative treatments. With this aim in mind, we synthesised new derivatives of troglitazone, a compound which was formerly used as an anti-diabetic agent and which exhibits anti-proliferative activity on various cancer cell lines. Among the compounds prepared, some displayed micromolar activity against hormone-dependent and hormone-independent breast cancer cells. Furthermore, the influence of the compounds on the viability of primary cultures of human hepatocytes was evaluated. This enabled us to obtain for the first time interesting structure-toxicity relationships in this family of compounds, resulting in 6b and 8b, which show good anti-proliferative activities and poor toxicity towards hepatocytes, compared to troglitazone.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Breast Neoplasms/pathology , Chemistry Techniques, Synthetic , Chromans/pharmacology , Chromans/toxicity , Thiazolidinediones/pharmacology , Thiazolidinediones/toxicity , Toxicity Tests , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromans/chemical synthesis , Chromans/chemistry , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry , Troglitazone
SELECTION OF CITATIONS
SEARCH DETAIL
...