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










Publication year range
1.
PLoS One ; 15(10): e0240676, 2020.
Article in English | MEDLINE | ID: mdl-33052979

ABSTRACT

The impact of cancer on lifespan is significantly increasing worldwide. Enhanced activity of drug efflux pumps and the incidences of the tumor microenvironment such as the apparition of a hypoxic gradient inside of the bulk tumor, are the major causes of chemotherapy failure. For instance, expression of Hypoxia-inducible factor (HIF-1α) has been associated with metastasis, resistance to chemotherapy and reduced survival rate. One of the current challenges to fight against cancer is therefore to find new molecules with therapeutic potential that could overcome this chemoresistance. In the present study, we focused on the bioactive plant flavonoid quercetin, which has strong antioxidant and anti-proliferative properties. We examined the efficacy of combined treatments of quercetin and the anti-cancer drugs gemcitabine and doxorubicin, known to specifically act on human pancreatic and hepatic cancer cells, respectively. Moreover, our study aimed to investigate more in-depth the implication of the multidrug transporter MDR1 and HIF-1α n chemoresistance and if quercetin could act on the activity of the drug efflux pumps and the hypoxia-associated effects. We observed that the anti-cancer drugs, were more effective when administered in combination with quercetin, as shown by an increased percentage of dead cells up to 60% in both 2D and 3D cultures. In addition, our results indicated that the combination of anti-cancer drugs and quercetin down-regulated the expression of HIF-1α and increased the expression levels of the regulator of apoptosis p53. Moreover, we observed that quercetin could inhibit the efflux activity of MDR1. Finally, our in vitro study suggests that the efficiency of the chemotherapeutic activity of known anti-cancer drugs might be significantly increased upon combination with quercetin. This flavonoid may therefore be a promising pharmacological agent for novel combination therapy since it potentializes the cytotoxic activity of gemcitabine and doxorubicin on by targeting the chemoresistance developed by the pancreatic and liver cancer cells respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antioxidants/pharmacology , Deoxycytidine/analogs & derivatives , Doxorubicin/pharmacology , Quercetin/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenocarcinoma/drug therapy , Cell Hypoxia/drug effects , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Gemcitabine
2.
Sci Rep ; 10(1): 13750, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792639

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive types of cancer, which begins within the brain. It is the most invasive type of glioma developed from astrocytes. Until today, Temozolomide (TMZ) is the only standard chemotherapy for patients with GBM. Even though chemotherapy extends the survival of patients, there are many undesirable side effects, and most cases show resistance to TMZ. FL3 is a synthetic flavagline which displays potent anticancer activities, and is known to inhibit cell proliferation, by provoking cell cycle arrest, and leads to apoptosis in a lot of cancer cell lines. However, the effect of FL3 in glioblastoma cancer cells has not yet been examined. Hypoxia is a major problem for patients with GBM, resulting in tumor resistance and aggressiveness. In this study, we explore the effect of FL3 in glioblastoma cells under normoxia and hypoxia conditions. Our results clearly indicate that this synthetic flavagline inhibits cell proliferation and induced senescence in glioblastoma cells cultured under both conditions. In addition, FL3 treatment had no effect on human brain astrocytes. These findings support the notion that the FL3 molecule could be used in combination with other chemotherapeutic agents or other therapies in glioblastoma treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytes/drug effects , Benzofurans/pharmacology , Brain Neoplasms/drug therapy , Cellular Senescence/drug effects , Glioblastoma/drug therapy , Aglaia/chemistry , Anaerobiosis/physiology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/pathology , Humans , Plant Preparations/pharmacology
3.
Toxicol In Vitro ; 60: 27-35, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31028861

ABSTRACT

The molecular pathways by which flavagline derivatives exert their cytotoxicity against various cancer cell types are well documented, while the mechanisms that prevent their cytotoxic effects on normal cells still have to be clarified. Here we provide the underlying molecular events by which normal skin cells remain unaffected after exposure to the synthetic flavagline FL3. Indeed, the anticancer agent fails to trigger apoptosis of healthy cells and is unable to induce the depolarization of their mitochondrial membrane and the cytosolic release of cytochrome C, in contrast to what is observed for cancer cells. Most importantly, FL3 specifically induces in normal cells, but not in malignant cells, an activation of Bad, without significant mitochondrial and cytosolic redistribution of Bax or Bcl-2. Moreover, gene knockdown of Bad sensitizes the normal fibroblastic cells to FL3 and induces a caspase-3 dependent apoptosis. Bad activation, known to promote survival and block apoptosis, explains therefore the lack of cytotoxicity of FL3 on normal skin cells. Finally, these findings provide new insights into the molecular mechanisms of resistance of healthy cells against FL3 cytotoxicity and identify it as a promising anticancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Skin/cytology , Apoptosis/drug effects , Cell Line , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Keratinocytes/drug effects , Keratinocytes/physiology , Membrane Potential, Mitochondrial/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , bcl-Associated Death Protein/metabolism
4.
Exp Cell Res ; 360(2): 138-145, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28867479

ABSTRACT

Angiogenesis is now well known for being involved in tumor progression, aggressiveness, emergence of metastases, and also resistance to cancer therapies. In this study, to better mimic tumor angiogenesis encountered in vivo, we used 3D culture of osteosarcoma cells (MG-63) that we deposited on 2D endothelial cells (HUVEC) grown in monolayer. We report that endothelial cells combined with tumor cells were able to form a well-organized network, and that tubule-like structures corresponding to new vessels infiltrate tumor spheroids. These vessels presented a lumen and expressed specific markers as CD31 and collagen IV. The combination of 2D endothelial cells and 3D microtissues of tumor cells also increased expression of angiogenic factors as VEGF, CXCR4 and ICAM1. The cell environment is the key point to develop tumor vascularization in vitro and to be closer to tumor encountered in vivo.


Subject(s)
Bone Neoplasms/pathology , Cell Culture Techniques/methods , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/physiology , Neovascularization, Pathologic/pathology , Osteosarcoma/pathology , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Neovascularization, Pathologic/genetics , Osteosarcoma/blood supply , Osteosarcoma/genetics , Tissue Scaffolds/chemistry
5.
Cell Adh Migr ; 11(4): 316-326, 2017 07 04.
Article in English | MEDLINE | ID: mdl-27463962

ABSTRACT

The low-density lipoprotein receptor-related protein-1 (LRP-1) is a member of Low Density Lipoprotein Receptor (LDLR) family, which is ubiquitously expressed and which is described as a multifunctional endocytic receptor which mediates the clearance of various extracellular matrix molecules including serine proteinases, proteinase-inhibitor complexes, and matricellular proteins. Several studies showed that high LRP-1 expression promotes breast cancer cell invasiveness, and LRP-1 invalidation leads to cell motility abrogation in both tumor and non-tumor cells. Furthermore, our group has reported that LRP-1 silencing prevents the invasion of a follicular thyroid carcinoma despite increased pericellular proteolytic activities from MMP2 and uPA using a 2D-cell culture model. As the use of 3D culture systems is becoming more and more popular due to their promise as enhanced models of tissue physiology, the aim of the present work is to characterize for the first time how the 3D collagen type I matrix may impact the ability of LRP-1 to regulate the migratory properties of thyroid carcinoma using as a model FTC-133 cells. Our results show that inhibition of LRP-1 activity or expression leads to morphological changes affecting cell-matrix interactions, reorganizations of the actin-cytoskeleton especially by inhibiting FAK activation and increasing RhoA activity and MLC-2 phosphorylation, thus preventing cell migration. Taken together, our results suggest that LRP-1 silencing leads to a decrease of cell migratory capacity in a 3D configuration.


Subject(s)
Cell Movement , Collagen/pharmacology , Extracellular Matrix/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape/drug effects , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Silencing , Humans , LDL-Receptor Related Protein-Associated Protein/pharmacology , Myosin Light Chains/metabolism , Rats , Recombinant Proteins/pharmacology , Reproducibility of Results
6.
Cell Biol Toxicol ; 33(3): 295-306, 2017 06.
Article in English | MEDLINE | ID: mdl-27981389

ABSTRACT

As initiators of the carcinogenic process, cancer stem cells (CSCs) are considered as new targets for anti-cancer therapies. However, these cells are hidden in the cancer bulk and remain relatively insensitive to chemotherapy, which targets their proliferative capacities. Alternatively, growing evidences have pointed out that a differentiation therapy could adversely affect these cells, which consequently should lose their self-renewal properties and become less aggressive. In order to evaluate the differentiation potential of an emerging class of anti-cancer drugs, we used the poorly differentiated teratocarcinomal cell as a model of Oct4-expressing CSC and determined the molecular mechanisms induced by the highly active flavagline FL3. The drug, administrated at sublethal concentration and for long period, was able to downregulate the expression levels of the stemness factors Oct4 and Nanog at both transcriptional and translational levels, concomitantly with a decrease of clonogenicity. The appearance of specific neural markers further demonstrated the differentiation properties of FL3. Interestingly, an expression of active caspase-3 and an upregulation of the expression of the germ cell nuclear factor were observed in treated cells; this suggests that the suppression of Oct4 expression required for the induction of differentiation involves overlapping mechanisms of protein degradation and gene repression. Finally, this study shows that FL3, like all-trans retinoic acid (ATRA), acts as a differentiation inducer of teratocarcinomal cells. Thus, FL3 offers an alternative possibility for cancer treatment since it could target the carcinogenic process by inducing the differentiation of ATRA-resistant and Oct4-expressing CSCs, without toxic side effects on normal cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/drug effects , Cell Differentiation/drug effects , Neoplastic Stem Cells/drug effects , Carcinogenesis/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3 , Tretinoin/metabolism , Up-Regulation/drug effects
7.
Invest New Drugs ; 33(1): 64-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25404486

ABSTRACT

Cancer stem cells (CSCs) are potential targets for innovative anticancer therapies that involve natural products with potential chemopreventive effects. We therefore analyzed the antineoplastic activity of rooperol, the aglycone of the plant-derived compound hypoxoside, on a model of Oct4-expressing cancer stem-like cell, i.e. the human embryonal carcinoma (EC) cell NT2/D1. Rooperol selectively inhibited the proliferation of NT2/D1 cells in a concentration-dependent manner and had no effect on either normal embryonic fibroblasts which are more restrictive pluripotent stem cells or on NCCIT p53-mutant EC cells. Accordingly, rooperol only eliminates colon carcinoma cells expressing p53. Rooperol treatment triggered cell death on NT2/D1 cells through the alteration of mitochondrial membrane potential and production of reactive oxygen species (ROS). Rooperol-induced apoptosis was associated with activation of p53 and concentration-dependent changes of the expression levels of both caspase 3 and poly ADP ribose polymerase type 1 cleaved subunits. These modifications were accompanied by a downregulation of Oct4 and its two partners involved in the maintenance of cell pluripotency and self-renewal, Nanog and Sox2.Treatment with intracellular membrane permeant O2 (-) scavengers prevented rooperol-induced apoptosis and upregulation of the expression of p53 and active caspase-3. Our findings indicate that rooperol mediates its growth inhibitory effects on CSCs via a mitochondrial redox-sensitive mechanism. We propose that abrogating the expression of the stemness regulators is a prerequisite for rooperol to fully exert its pro-apoptotic properties on wild-type p53-bearing CSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Catechols/pharmacology , Embryonal Carcinoma Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Alkynes , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/physiology , Glucosides , Humans , Membrane Potential, Mitochondrial/drug effects , Teratocarcinoma
8.
Biochem Pharmacol ; 89(2): 185-96, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24607276

ABSTRACT

Cancer stem cells (CSCs) are considered as the initiators of the carcinogenic process and are therefore emerging targets for innovative anticancer therapies. In order to evaluate the anticancer chemopreventive activity of flavagline derivatives, we used the pluripotent teratocarcinomal cell as a model of Oct4-expressing cancer stem-like cell and determined the underlying cellular and molecular mechanisms induced by a synthetic flavagline. We precisely investigated the effects of the flavagline derivative FL3 on the human embryonal carcinoma (EC) cell line NT2/D1 and compared the responses to those of a normal more restrictive pluripotent stem cell line (i.e. BJ fibroblast cell line). FL3 selectively inhibited the proliferation of NT2/D1 cells by inducing G1 phase cell cycle arrest in a dose-dependent manner. Moreover, FL3 treatment specifically triggered apoptosis in association with an induction of the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and caspase-3 activation followed by a drastic downregulation of the master regulator of stemness Oct4. Forced inhibition of p38 MAPK activity by the specific pharmacological inhibitor SB203580 or by p38 MAPK gene knockdown using small-interfering RNA (siRNA) counteracted the effects of FL3, demonstrating that its chemopreventive action is related to growth inhibition and a p38-dependent caspase-3-dependent induction of apoptosis in Oct4-expressing CSCs. This study also shows that FL3 selectively kills poorly differentiated and highly aggressive carcinomal cells, but has little effect on normal stem-like cells. Thus FL3 offers great promise for cancer treatment since it is able to target the carcinogenic process without affecting normal cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Caspase 3/metabolism , MAP Kinase Signaling System/physiology , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , HCT116 Cells , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
PLoS One ; 7(3): e32526, 2012.
Article in English | MEDLINE | ID: mdl-22412883

ABSTRACT

Polyphenols are natural compounds widely present in fruits and vegetables, which have antimutagenic and anticancer properties. The aim of the present study was to determine the anticancer effect of a polyphenol-rich Aronia melanocarpa juice (AMJ) containing 7.15 g/L of polyphenols in the acute lymphoblastic leukemia Jurkat cell line, and, if so, to clarify the underlying mechanism and to identify the active polyphenols involved. AMJ inhibited cell proliferation, which was associated with cell cycle arrest in G(2)/M phase, and caused the induction of apoptosis. These effects were associated with an upregulation of the expression of tumor suppressor p73 and active caspase 3, and a downregulation of the expression of cyclin B1 and the epigenetic integrator UHRF1. AMJ significantly increased the formation of reactive oxygen species (ROS), decreased the mitochondrial membrane potential and caused the release of cytochrome c into the cytoplasm. Treatment with intracellular ROS scavengers prevented the AMJ-induced apoptosis and upregulation of the expression of p73 and active caspase 3. The fractionation of the AMJ and the use of identified isolated compounds indicated that the anticancer activity was associated predominantly with chlorogenic acids, some cyanidin glycosides, and derivatives of quercetin. AMJ treatment also induced apoptosis of different human lymphoblastic leukemia cells (HSB-2, Molt-4 and CCRF-CEM). In addition, AMJ exerted a strong pro-apoptotic effect in human primary lymphoblastic leukemia cells but not in human normal primary T-lymphocytes. Thus, the present findings indicate that AMJ exhibits strong anticancer activity through a redox-sensitive mechanism in the p53-deficient Jurkat cells and that this effect involves several types of polyphenols. They further suggest that AMJ has chemotherapeutic properties against acute lymphoblastic leukemia by selectively targeting lymphoblast-derived tumor cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , DNA-Binding Proteins/metabolism , Leukemia/metabolism , Nuclear Proteins/metabolism , Photinia/chemistry , Plant Extracts/pharmacology , Tumor Suppressor Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , DNA-Binding Proteins/genetics , Humans , Jurkat Cells , Lymphocytes/drug effects , Lymphocytes/immunology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nuclear Proteins/genetics , Oxidation-Reduction , Polyphenols/pharmacology , Reactive Oxygen Species/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases
10.
J Exp Clin Cancer Res ; 30: 41, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21496237

ABSTRACT

Over-expressed in numerous cancers, Ubiquitin-like containing PHD Ring Finger 1 (UHRF1, also known as ICBP90 or Np95) is characterized by a SRA domain (Set and Ring Associated) which is found only in the UHRF family. UHRF1 constitutes a complex with histone deacetylase 1 (HDAC1) and DNA methyltransferase 1 (DNMT1) via its SRA domain and represses the expression of several tumour suppressor genes (TSGs) including p16INK4A, hMLH1, BRCA1 and RB1. Conversely, UHRF1 is regulated by other TSGs such as p53 and p73. UHRF1 is hypothetically involved in a macro-molecular protein complex called "ECREM" for "Epigenetic Code Replication Machinery". This complex would be able to duplicate the epigenetic code by acting at the DNA replication fork and by activating the right enzymatic activity at the right moment. There are increasing evidence that UHRF1 is the conductor of this replication process by ensuring the crosstalk between DNA methylation and histone modifications via the SRA and Tandem Tudor Domains, respectively. This cross-talk allows cancer cells to maintain the repression of TSGs during cell proliferation. Several studies showed that down-regulation of UHRF1 expression in cancer cells by natural pharmacological active compounds, favors enhanced expression or re-expression of TSGs, suppresses cell growth and induces apoptosis. This suggests that hindering UHRF1 to exert its role in the duplication of the methylation patterns (DNA + histones) is responsible for inducing apoptosis. In this review, we present UHRF1 expression as a target of several natural products and we discuss their underlying molecular mechanisms and benefits for chemoprevention and chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , Down-Regulation , Genes, Tumor Suppressor , Neoplasms/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , DNA/metabolism , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Humans , Neoplasms/drug therapy , Ubiquitin-Protein Ligases
11.
Cell Signal ; 23(1): 152-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20807569

ABSTRACT

Thymoquinone (TQ), the active principle of Nigella sativa black seeds, has anti-proliferative properties on numerous cancer cell types. Others and we have previously reported that TQ acts as agent that triggers cell cycle arrest and apoptosis through either a p53- or p73-dependent pathway. However, the immediate targets recruited upon TQ-induced cytotoxicity have not yet been clearly identified. We therefore asked whether cyclic nucleotide phosphodiesterases (PDEs) could be involved in TQ-triggered pro-apoptotic reactivity; PDEs are regulators of intracellular levels of cyclic nucleotides and therefore can modulate cAMP and cGMP-dependent cell death pathways. Our results showed that TQ specifically repressed PDE1A expression in the acute lymphoblastic leukemia Jurkat cell line. This effect is concomitant with the previously described sequential deregulation of the expression of the tumor suppressor protein p73 and the epigenetic integrator UHRF1 (Ubiquitin-like, PHD Ring Finger 1). Interestingly, RNA-interference knock-down of PDE1A expression as well as decreased PDE1A expression induced growth inhibition of Jurkat cells, cell cycle arrest and apoptosis through an activation of p73 and a repression of UHRF1. Conversely, PDE1A re-expression counteracted the cellular pro-apoptotic effects of TQ in association with a p73 repression and UHRF1 re-expression. Altogether, our results show that TQ induced an initial down-regulation of PDE1A with a subsequent down-regulation of UHRF1 via a p73-dependent mechanism. This study further proposes that PDE1A might be involved in the epigenetic code inheritance by regulating, via p73, the epigenetic integrator UHRF1. Our findings also suggest that a forced inhibition of PDE1A expression might be a new therapeutic strategy for the management of acute lymphoblastic leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Benzoquinones/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Down-Regulation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Jurkat Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , RNA Interference , Tumor Protein p73 , Ubiquitin-Protein Ligases
12.
Invest New Drugs ; 29(2): 239-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-19943082

ABSTRACT

Cancer stem cells are expected to be responsible for tumor initiation and metastasis. These cells are therefore potential targets for innovative anticancer therapies. However, the absence of bona fide cancer stem cell lines is a real problem for the development of such approaches. Since teratocarcinoma cells are totipotent stem cells with a high degree of malignancy, we used them as a model of cancer stem cells in order to evaluate the anticancer chemopreventive activity of red wine polyphenols (RWPs) and to determine the underlying cellular and molecular mechanisms. We therefore investigated the effects of RWPs on the embryonal carcinoma (EC) cell line P19 which was grown in the same culture conditions as the most appropriate normal cell line counterpart, the pluripotent embryonic fibroblast cell line NIH/3T3. The present study indicates that RWPs selectively inhibited the proliferation of P19 EC cells and induced G1 cell cycle arrest in a dose-dependent manner. Moreover, RWPs treatment specifically triggered apoptosis of P19 EC cells in association with a dramatic upregulation of the tumor suppressor gene p53 and caspase-3 activation. Our findings suggest that the chemopreventive activity of RWPs on tumor initiation and development is related to a growth inhibition and a p53-dependent induction of apoptosis in teratocarcinoma cells. In addition, this study also shows that the EC cell line is a convenient source for studying the responses of cancer stem cells to new potential anticancer agents.


Subject(s)
Apoptosis/drug effects , Flavonoids/pharmacology , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phenols/pharmacology , Teratocarcinoma/pathology , Wine , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Flavonoids/chemistry , G1 Phase/drug effects , Mice , NIH 3T3 Cells , Phenols/chemistry , Polyphenols , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
13.
Eur J Cancer ; 46(5): 983-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20074931

ABSTRACT

Several epidemiological studies suggest that a diet rich in fruits and vegetables, which contain high levels of polyphenols, is associated with a reduced risk of cancer. The aim of the present study was to determine whether a red wine polyphenolic extract (RWPs, a rich source of polyphenols; 2.9g/L) affects the proliferation of human lymphoblastic leukaemia cells (Jurkat cells) and, if so, to determine the underlying mechanism. Cell proliferation and viability were determined by the MTS and trypan blue exclusion assays, respectively. Cell cycle analysis, apoptosis activity and oxidative stress levels were assessed by flow cytometry, and the expression of p73, UHRF1 and active caspase-3 by Western blot analysis. RWPs inhibited the proliferation of Jurkat cells and induced G0/G1 cell cycle arrest in a concentration-dependent manner. Moreover, RWPs triggered apoptosis, which is associated with an increased expression level of the pro-apoptotic protein p73 and the active caspase-3. RWPs induced apoptosis confirmed by DNA fragmentation analysis, and this effect was associated with down-regulation of the antiapoptotic protein UHRF1. Furthermore RWPs significantly increased the formation of reactive oxygen species (ROS). Intracellular scavengers of superoxide anions (MnTMPyP, MnTBAP, PEG-SOD) prevented the RWPs-induced formation of ROS and apoptosis, while native extracellular superoxide dismutase (SOD) was without effect. In addition, the effect of RWPs on the expression levels of p73, active caspase-3 and UHRF1 was also prevented by MnTMPyP. Thus, these findings indicate that RWPs induce apoptosis in Jurkat cells by a redox-sensitive mechanism involving the intracellular formation of superoxide anions and consequently the up-regulation of p73 and down-regulation of UHRF1.


Subject(s)
Apoptosis , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Flavonoids/pharmacology , Nuclear Proteins/metabolism , Phenols/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Proteins/metabolism , Wine , Caspase 3/biosynthesis , Cell Cycle/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Jurkat Cells/drug effects , Neoplasm Proteins/metabolism , Oxidation-Reduction , Polyphenols , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Tumor Protein p73 , Ubiquitin-Protein Ligases , Up-Regulation/drug effects
14.
Genet Epigenet ; 2009(2): 29-36, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-21643543

ABSTRACT

Epigenetics is the study of the transmission of cell memory through mitosis or meiosis that is not based on the DNA sequence. At the molecular level the epigenetic memory of a cell is embedded in DNA methylation, histone post-translational modifications, RNA interference and histone isoform variation. There is a tight link between histone post-translational modifications (the histone code) and DNA methylation, as modifications of histones contribute to the establishment of DNA methylation patterns and vice versa. Interestingly, proteins have recently been identified that can simultaneously read both methylated DNA and the histone code. UHRF1 ful-fills these requirements by having unique structural domains that allow concurrent recognition of histone modifications and methylated DNA. Herein, we review our current knowledge of UHRF1 and discuss how this protein ensures the link between histone marks and DNA methylation. Understanding the molecular functions of this protein may reveal the physiological relevance of the linkage between these layers of epigenetic marks.

15.
Biochem Pharmacol ; 79(9): 1251-60, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20026309

ABSTRACT

The salvage anti-tumoral pathway which implicates the p53-related p73 gene is not yet fully characterized. We therefore attempted to identify the up- and down-stream events involved in the activation of the p73-dependent pro-apoptotic pathway, by focusing on the anti-apoptotic and epigenetic integrator UHRF1 which is essential for cell cycle progression. For this purpose, we analyzed the effects of a known anti-neoplastic drug, thymoquinone (TQ), on the p53-deficient acute lymphoblastic leukemia (ALL) Jurkat cell line. Our results showed that TQ inhibits the proliferation of Jurkat cells and induces G1 cell cycle arrest in a dose-dependent manner. Moreover, TQ treatment triggers programmed cell death, production of reactive oxygen species (ROS) and alteration of the mitochondrial membrane potential (DeltaPsim). TQ-induced apoptosis, confirmed by the presence of hypodiploid G0/G1 cells, is associated with a rapid and sharp re-expression of p73 and dose-dependent changes of the levels of caspase-3 cleaved subunits. These modifications are accompanied by a dramatic down-regulation of UHRF1 and two of its main partners, namely DNMT1 and HDAC1, which are all involved in the epigenetic code regulation. Knockdown of p73 expression restores UHRF1 expression, reactivates cell cycle progression and inhibits TQ-induced apoptosis. Altogether our results showed that TQ mediates its growth inhibitory effects on ALL p53-mutated cells via the activation of a p73-dependent mitochondrial and cell cycle checkpoint signaling pathway which subsequently targets UHRF1.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzoquinones/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Tumor Suppressor Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Humans , Jurkat Cells , Nuclear Proteins/genetics , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases
16.
Biochem Biophys Res Commun ; 390(3): 523-8, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19800870

ABSTRACT

Tat-interactive protein, 60kDa (Tip60) is a histone acetyltransferase with specificity toward lysine 5 of histone H2A (H2AK5) and plays multiple roles in chromatin remodeling processes. Co-immunoprecipitation experiments performed on Jurkat cells, showed that Tip60 is present in the same macro-molecular complex as UHRF1 (Ubiquitin-like containing PHD and RING domain 1), DNMT1 (DNA methyltransferase 1), and HDAC1 (histone deacetylase 1). Furthermore, immunocytochemistry experiments confirmed that Tip60 co-localizes with the UHRF1/DNMT1 complex. Although down-regulation of UHRF1 by RNA interference enhanced Tip60 expression, a significant decrease of the level of acetylated H2AK5 was observed. Consistently, we have observed that down-regulation of Tip60 and DNMT1 by RNA interference, dramatically reduced the levels of acetylated H2AK5. Altogether, these results suggest that Tip60 is a novel partner of the epigenetic integration platform interplayed by UHRF1, DNMT1 and HDAC1 involved in the epigenetic code replication.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Epigenesis, Genetic , Histone Acetyltransferases/metabolism , Acetylation , CCAAT-Enhancer-Binding Proteins/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Knockdown Techniques , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Immunoprecipitation , Jurkat Cells , Lysine/metabolism , Lysine Acetyltransferase 5 , Ubiquitin-Protein Ligases
17.
Stem Cells ; 26(3): 734-44, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18096721

ABSTRACT

Although the properties of embryonic stem (ES) cells make these cells very attractive in the field of replacement therapy, the molecular mechanisms involved in the maintenance of their pluripotency are not fully characterized. Starting from the observation that most pluripotent markers are also expressed by spermatogonia stem cells, we identified Tex19 as a new potential pluripotency marker. We show that Tex19 is a mammalian-specific protein duplicated in mouse and rat, renamed Tex19.1 and Tex19.2, whereas only one form is found in human. In mouse, both forms are localized on chromosome 11 and transcribed in opposite directions. Tex19 proteins are well conserved, showing two highly conserved domains that do not present any similarity with any other known domains. We show that Tex19.2 is specifically detected in the male somatic gonad lineage, whereas Tex19.1 expression is very similar to that of Oct4. Transcripts are maternally inherited, and expression starts as soon as the early embryo and later is limited to the germ line. Tex19.1 transcripts were also detected in mouse pluripotent stem cells, and expression of Tex19.1, like that of Oct4, decreases after murine embryonic stem and germ cell differentiation. Human TEX19 was more closely related to murine Tex19.1 and was also detected in adult testis and in undifferentiated ES cells. By immunofluorescence, we found that Tex19.1 protein localizes to the nucleus of mouse ES and inner cell mass cells. All these results suggest that Tex19.1, as well as human TEX19, could be a new factor involved in the maintenance of self-renewal or pluripotency of stem cells.


Subject(s)
Germ Cells/metabolism , Nuclear Proteins/metabolism , Pluripotent Stem Cells/metabolism , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Line , Cell Lineage , Conserved Sequence , Female , Gene Expression Regulation, Developmental , Genome , Germ Cells/cytology , Humans , Male , Mice , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Pluripotent Stem Cells/cytology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Rats , Sequence Homology, Amino Acid , Species Specificity , Synteny , Testis/cytology , Testis/metabolism
18.
Cancer Res ; 66(12): 6002-7, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778170

ABSTRACT

The potential role of alpha5beta1 integrins in cancer has recently attracted much interest. However, few alpha5beta1-selective antagonists have been developed compared with other integrins. The most specific nonpeptidic alpha5beta1 antagonist described thus far, SJ749, inhibits angiogenesis by affecting adhesion and migration of endothelial cells. We investigated the effects of SJ749 in two human astrocytoma cell lines, A172 and U87, which express different levels of alpha5beta1. SJ749 dose-dependently inhibited adhesion of both cell types on fibronectin. Application of SJ749 to spread cells led to formation of nonadherent spheroids for A172 cells but had no effect on U87 cell morphology. SJ749 also reduced proliferation of A172 cells due to a long lasting G0-G1 arrest, whereas U87 cells were only slightly affected. However, under nonadherent culture conditions (soft agar), SJ749 significantly reduced the number of colonies formed only by U87 cells. As U87 cells express more alpha5beta1 than A172 cells, we specifically examined the effect of SJ749 on A172 cells overexpressing alpha5. Treatment of alpha5-A172 cells with SJ749 decreased colony formation similarly to that observed in U87 cells. Therefore, in nonadherent conditions, the effect of SJ749 on tumor cell growth characteristics depends on the level of alpha5beta1 expression. Our study highlights the importance of alpha5beta1 as an anticancer target and shows for the first time that a small nonpeptidic alpha5beta1-specific antagonist affects proliferation of tumor cells.


Subject(s)
Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Integrin alpha5beta1/antagonists & inhibitors , Propionates/pharmacology , Pyridines/pharmacology , Spiro Compounds/pharmacology , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/metabolism , Spheroids, Cellular , Substrate Specificity , Tumor Stem Cell Assay
19.
Med Sci (Paris) ; 20(12): 1091-5, 2004 Dec.
Article in French | MEDLINE | ID: mdl-15581461

ABSTRACT

In sexually reproducing animals all gametes of either sex arise from primordial germ cells (PGC). PGC represent a small cell population, appearing early during embryo development. They represent a key cell population responsible for the survival and the evolution of a species. Indeed, the production of gametes will assure fertilisation and therefore the establishment of the next generation. Until recently only few laboratories were working on PGC biology. A new interest emerged since these cells have the ability to function as pluripotent stem cells when established as cell lines. Indeed, like embryonic stem cells (ESC), embryonic germ cells (EGC) are able to differentiate in a wide variety of tissues. In vivo, EGC are able, after injection into a host blastocyst cavity to colonise the inner cell mass and to participate in embryonic development. In vitro studies in human and mouse have also shown their capacity to differentiate into a large variety of cell types allowing the study of processes involved in cardiomyocyte, haematopoietic, neuronal and myogenic differentiation pathways. We present here the last updates of PGC ontogeny focusing mainly on the murine model.


Subject(s)
Germ Cells/growth & development , Animals , Cell Differentiation , Cell Movement , Mice
20.
Science ; 300(5623): 1251-6, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12730498

ABSTRACT

Continuation of mammalian species requires the formation and development of the sexually dimorphic germ cells. Cultured embryonic stem cells are generally considered pluripotent rather than totipotent because of the failure to detect germline cells under differentiating conditions. Here we show that mouse embryonic stem cells in culture can develop into oogonia that enter meiosis, recruit adjacent cells to form follicle-like structures, and later develop into blastocysts. Oogenesis in culture should contribute to various areas, including nuclear transfer and manipulation of the germ line, and advance studies on fertility treatment and germ and somatic cell interaction and differentiation.


Subject(s)
Blastocyst/physiology , Cell Differentiation , Embryo, Mammalian/cytology , Oocytes/physiology , Oogenesis , Totipotent Stem Cells/physiology , Transcription Factors , Animals , Biomarkers/analysis , Blastocyst/cytology , Cell Adhesion , Cell Aggregation , Cell Lineage , Cell Separation , Cells, Cultured , DNA-Binding Proteins/genetics , Estradiol/metabolism , Female , Gene Expression , Genes, Reporter , Meiosis , Mice , Mice, Transgenic , Octamer Transcription Factor-3 , Oocytes/cytology , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Recombinant Fusion Proteins , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...