Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Asian Pac J Cancer Prev ; 23(12): 4205-4212, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36580003

ABSTRACT

BACKGROUND: Plants play an important role in cancer therapy. They are source of natural molecules which can induce apoptosis in cancer cells by affecting molecular mechanisms implicated in cancer progression. The MAP Kinase/ERK1/2 and PI3K/AKT signaling pathways are two classical signaling pathways implicated in cancer progression and constitute therapeutic targets against cancer. This study aimed to evaluate the effect of euphol on MAP Kinase/ERK1/2 and PI3K/AKT signaling pathways in glioblastoma and prostate cancer cells. Euphol is a tetracyclique triterpene alcohol isolated from Tapinanthus sp. which is a hemi parasitic plant belonging to Loranthaceae family. METHODS: Plant powder was extracted by maceration and euphol was isolated and described using respectively column chromatography separation on silica gel and spectroscopic data. Cytotoxic effect of euphol was evaluated using XTT assay and its effect on MAP Kinase/ERK1/2 and PI3K/AKT protein expression was investigated by Western immunoblot analysis. Apotosis was analyzed by evaluating caspase-3/7 activity. RESULTS: Our investigations demonstrated that this compound has an important cytotoxic effect on C6 and U87 MG glioblastoma (GBM) cells and PC-3 prostate cancer cells. Furthermore, euphol-induced apoptosis revealed by elevated caspase 3/7 activity, was correlated with a significant inhibition of MAP kinase/Erk 1/2 and PI3K/Akt signaling pathway in glioblastoma U87 MG cells. The reverse effect was observed in C6 glioblastoma cells, where apoptosis was correlated with a long-lasting activation of Erk 1/2.  In PC-3 cells, euphol had no or limited effect on Erk 1/2 and Akt activity. CONCLUSION: These results indicate that euphol induces cell death in glioblastoma and prostate cancer cells and regulates significantly Erk1/2 and Akt activity in glioblastoma cells.


Subject(s)
Glioblastoma , Loranthaceae , Prostatic Neoplasms , Male , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Loranthaceae/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , MAP Kinase Signaling System , Cell Proliferation
2.
Asian Pac J Cancer Prev ; 22(4): 1087-1093, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33906300

ABSTRACT

BACKGROUND: Cancer incidence has been growing in an alarming rate worldwide and new therapeutics are needed, particularly for intractable and chemoresistant cases. We evaluated the cytotoxic effects of Combretum fragrans F. Hoffm (Combretaceae) on glioblastoma (U87MG and C6) and prostate (PC-3) cancer cell lines. METHODS: The cytotoxic effect of the methanolic extract of the stem bark of Combretum fragrans was assessed using XTT (2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino) carbonyl]-2H-tetrazolium hydroxide) test. Expressions of Akt and ERK1/2 were determined using Western blot technique, while Caspase-3/7 kits were used to evaluate caspase-3/7 activity. RESULTS: C. fragrans extract inhibited the proliferation of U87 (IC50 = 20.13 µg/mL), C6 (IC50 = 12.17 µg/mL), and PC-3 (IC50 = 11.50 µg/mL) cells. Treatment with the extract resulted in lower levels (p < 0.001) of phospho-ERK1/2 and phospho-Akt in U87 cells, and instead, higher levels of phospho-ERK1/2 (p < 0.001) in C6 and PC-3 cells. An increase in caspase-3/7 activity was observed, mainly after 24 hours of treatment, indicating the activation of apoptotic processes. CONCLUSION: Altogether, these results suggest that C. fragrans have potent anticancer properties. This plant should be further investigated for developing new anticancer drugs.


Subject(s)
Combretum , Glioblastoma/drug therapy , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Plant Stems
3.
PLoS One ; 11(5): e0155950, 2016.
Article in English | MEDLINE | ID: mdl-27191720

ABSTRACT

The EGF-family of tyrosine-kinase receptors activates cytoplasmic pathways involved in cell proliferation, migration and differentiation in response to specific extracellular ligands. Beside these canonical pathways, the nuclear localization of the ErbB receptors in primary tumours and cancer cell lines led to investigate their role as transcriptional regulators of cancer genes. The nuclear localization of ErbB3 has been reported in various cancer tissues and cell lines but the nuclear functions and the putative correlation with tumour progression and resistance to therapy remain unclear. We first assessed ErbB3 expression in normal and tumour prostate tissues. The nuclear staining was mainly due to an isoform matching the C-terminus domain of the full length ErbB3185kDa receptor. Nuclear staining was also restricted to cancer cells and was increased in advanced castration-resistant prostate cancer when compared to localized tumours, suggesting it could be involved in the progression of prostate cancer up to the terminal castration-resistant stage. ChIP-on-chip experiments were performed on immortalized and tumour cell lines selected upon characterization of endogenous nuclear expression of an ErbB380kDa isoform. Among the 1840 target promoters identified, 26 were selected before ErbB380kDa-dependent gene expression was evaluated by real-time quantitative RT-PCR, providing evidence that ErbB380kDa exerted transcriptional control on those genes. Some targets are already known to be involved in prostate cancer progression even though no link was previously established with ErbB3 membrane and/or nuclear signalling. Many others, not yet associated with prostate cancer, could provide new therapeutic possibilities for patients expressing ErbB380kDa. Detecting ErbB380kDa could thus constitute a useful marker of prognosis and response to therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Nucleus/metabolism , Gene Regulatory Networks , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptor, ErbB-3/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, ErbB-3/genetics
4.
Biochem Biophys Res Commun ; 454(4): 524-30, 2014 11 28.
Article in English | MEDLINE | ID: mdl-25450687

ABSTRACT

An increasing number of G protein-coupled receptors, like receptors for vasoactive intestinal peptide (VIP), are found in cell nucleus. As VIP receptors are involved in the regulation of glioma cell proliferation and migration, we investigated the expression and the nuclear localization of the VIP receptors VPAC1 and VPAC2 in this cancer. First, by applying Western blot and immunofluorescence detection in three human glioblastoma (GBM) cell lines, we observed a strong nuclear staining for the VPAC1 receptor and a weak nuclear VPAC2 receptor staining. Second, immunohistochemical staining of VPAC1 and VPAC2 on tissue microarrays (TMA) showed that the two receptors were expressed in normal brain and glioma tissues. Expression in the non-nuclear compartment of the two receptors significantly increased with the grade of the tumors. Analysis of nuclear staining revealed a significant increase of VPAC1 staining with glioma grade, with up to 50% of GBM displaying strong VPAC1 nuclear staining, whereas nuclear VPAC2 staining remained marginal. The increase in VPAC receptor expression with glioma grades and the enhanced nuclear localization of the VPAC1 receptors in GBM might be of importance for glioma progression.


Subject(s)
Cell Nucleus/metabolism , Glioma/pathology , Receptors, Vasoactive Intestinal Peptide, Type II/analysis , Receptors, Vasoactive Intestinal Polypeptide, Type I/analysis , Active Transport, Cell Nucleus , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Tissue Array Analysis , Tumor Cells, Cultured , Young Adult
5.
PLoS One ; 9(5): e96136, 2014.
Article in English | MEDLINE | ID: mdl-24798431

ABSTRACT

High-risk human papillomavirus oncoproteins E6 and E7 play a major role in HPV-related cancers. One of the main functions of E7 is the degradation of pRb, while E6 promotes the degradation of p53, inactivating the p14ARF-p53 pathway. pRb and p14ARF can repress ribosomal DNA (rDNA) transcription in part by targeting the Upstream Binding Factor 1 (UBF1), a key factor in the activation of RNA polymerase I machinery. We showed, through ectopic expression and siRNA silencing of p14ARF and/or E7, that E7 stimulates UBF1-mediated rDNA gene transcription, partly because of increased levels of phosphorylated UBF1, preventing the inhibitory function of p14ARF. Unexpectedly, activation of rDNA gene transcription was higher in cells co-expressing p14ARF and E7, compared to cells expressing E7 alone. We did not find a difference in P-UBF1 levels that could explain this data. However, p14ARF expression induced E7 to accumulate into the nucleolus, where rDNA transcription takes place, providing an opportunity for E7 to interact with nucleolar proteins involved in this process. GST-pull down and co-immunoprecipitation assays showed interactions between p14ARF, UBF1 and E7, although p14ARF and E7 are not able to directly interact. Co-expression of a pRb-binding-deficient mutant (E7C24G) and p14ARF resulted in EC24G nucleolar accumulation, but not in a significant higher activation of rDNA transcription, suggesting that the inactivation of pRb is involved in this phenomenon. Thus, p14ARF fails to prevent E7-mediated UBF1 phosphorylation, but could facilitate nucleolar pRb inactivation by targeting E7 to the nucleolus. While others have reported that p19ARF, the mouse homologue of p14ARF, inhibits some functions of E7, we showed that E7 inhibits a p53-independent function of p14ARF. These results point to a mutually functional interaction between p14ARF and E7 that might partly explain why the sustained p14ARF expression observed in most cervical pre-malignant lesions and malignancies may be ineffective.


Subject(s)
Cell Transformation, Viral , DNA, Ribosomal/metabolism , Human papillomavirus 16/metabolism , Papillomavirus E7 Proteins/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Protein p14ARF/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Nucleolus , DNA, Ribosomal/genetics , Female , Human papillomavirus 16/genetics , Humans , Mice , Mutation, Missense , Papillomavirus E7 Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Tumor Suppressor Protein p14ARF/genetics
6.
Cell Signal ; 24(5): 1074-85, 2012 May.
Article in English | MEDLINE | ID: mdl-22261253

ABSTRACT

EGFR family members are tyrosine kinase transmembrane receptors that, in response to specific extracellular ligands, activate cytoplasmic pathways involved in cell proliferation, migration and differentiation. More recently, a pivotal role for EGF receptors has emerged, through the description of their nuclear localization.We report here the characterization of a nuclear variant of the kinase-defective ErbB3 receptor, ErbB3(80 kDa), spanning the intracytoplasmic domain of the receptor. We assessed the putative transcriptional functions of ErbB3(80 KDa) in cancer cells, through the regulation of the proliferative Cyclin D1 gene, an already known target of the ErbB3 cytoplasmic signaling. We demonstrate here that the binding of ErbB3(80 KDa) on the promoter activates Cyclin D1 transcription and subsequent protein expression, leading to an increased cell proliferation. This mechanism can be balanced in response to the ectopic expression of the tumor suppressor p14ARF that physically interacts with ErbB3(100 kDa) and sequesters it into nucleoli. Our data also show that ErbB3(80 kDa) increases the transcription of proliferative genes even though the cytoplasmic pathways are not activated. This nuclear ErbB3 pathway and the target genes concerned need to be further studied. Indeed, such mechanism could explain the tumor relapse observed in response to treatments aimed at blocking the receptor activation in response to ligand binding.


Subject(s)
Cell Proliferation , Cyclin D1/genetics , Promoter Regions, Genetic , Receptor, ErbB-3/metabolism , Tumor Suppressor Protein p14ARF/physiology , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , Cyclin D1/metabolism , Down-Regulation , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Molecular Sequence Data , Protein Binding , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Transcriptional Activation , Tumor Suppressor Protein p14ARF/metabolism , p21-Activated Kinases/metabolism
7.
Med Sci (Paris) ; 22(5): 519-24, 2006 May.
Article in French | MEDLINE | ID: mdl-16687121

ABSTRACT

The nucleolar Arf protein has initially been shown to regulate cell cycle through the so-called Arf-mdm2-p53 pathway. In addition to this well characterized pathway, convergent data published since 2000 indicate that Arf can inhibit cell proliferation in absence of p53, suggesting the existence of a p53-independent pathway. Several partners have recently been described that could participate in an alternative regulatory process. Recent results show that : (1) Arf binds the rDNA promoter to inhibit the transcription of the 47S rRNA precursor and (2) Arf interacts with the nucleophosmin/B23 protein to negatively regulate rRNA maturation, it is assumed that the tumour suppressor may downregulate the cell cycle progression through the control of ribosome biogenesis, thus resulting in completion of cell cycle arrest.


Subject(s)
Cell Proliferation , Ribosomes/physiology , Tumor Suppressor Protein p14ARF/physiology , Humans , RNA, Ribosomal/biosynthesis
8.
Leuk Res ; 30(10): 1273-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16533530

ABSTRACT

Expression of p14(ARF) and p16(INK4a) tumor suppressor genes was investigated in 109 patients with chronic myeloid leukemia (CML). The p14(ARF) and p16(INK4a) mRNA levels were significantly low in patients in chronic phase (CP) at presentation and high in patients treated with interferon-alpha (IFN-alpha), especially in non-responders. A moderate overexpression of p14(ARF) with a normal expression of p16(INK4a) was observed in imatinib-resistant patients. Although protein expression did not consistently match mRNA levels, a role for the two cell cycle regulators in the IFN-alpha signaling pathway is suggested as well as a relation with the resistance to IFN-alpha or imatinib therapy.


Subject(s)
Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Tumor Suppressor Protein p14ARF/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , RNA, Messenger/genetics , Reference Values , Retrospective Studies , Signal Transduction
9.
Exp Cell Res ; 312(7): 1185-93, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16510138

ABSTRACT

p16INK4a-pRB-E2F and ARF-MDM2-p53 are two major tumor suppressor networks involved in cell proliferation control. The nucleolar ARF protein binds to MDM2 to activate the growth suppressive functions of p53, but can also exert its tumor suppressor activity independently of p53, through mechanisms involving other regulators: in that manner, p14ARF has been shown to inhibit the transcriptional activity of E2F1 in vitro, suggesting that the two pathways intersect with one another. More recently, ARF has been shown to inhibit ribosomal RNA processing, and to specifically interact with the rRNA promoter, suggesting a role in the regulation of both maturation and transcription processes. We show here that E2F1 can bind the rRNA promoter and modulate its activity through the interaction with two E2F1-binding sequences we have identified. The regulation of ribosome biogenesis appears as a major p53-independent process, which involves both ARF and E2F1 to control cell proliferation.


Subject(s)
E2F1 Transcription Factor/physiology , Gene Expression Regulation/physiology , Promoter Regions, Genetic , RNA, Ribosomal/biosynthesis , Base Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , RNA, Ribosomal/genetics , Up-Regulation
10.
Oncogene ; 24(15): 2580-4, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15735732

ABSTRACT

Both human and murine ARF proteins have been recently reported to impair rRNA maturation and ribosomes biogenesis through a p53-independent pathway. A specific interaction has been established between 5.8S rRNA and the murine p19ARF specie. We report here, by use of both in vitro and ChIP-RNA assays, the absence of any interaction between the human p14ARF and the homologous 5.8S rRNA. Our data are not consistent with the involvement of a 5.8S-p14ARF complex in ribosome biogenesis in man. Rather they suggest that the human protein does not require such an interaction to achieve a similar function. This result is a new argument in favour of functional differences between human and murine ARF proteins.


Subject(s)
RNA, Ribosomal, 5.8S/metabolism , Tumor Suppressor Protein p14ARF/pharmacology , Animals , Cyclin-Dependent Kinase Inhibitor p16 , Humans , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
11.
Oncogene ; 23(49): 8097-104, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15361825

ABSTRACT

The tumor suppressor Arf (Alternative Reading Frame) protein (p14ARF in human and p19ARF in mouse) is mainly located in the nucleolus consistent with its subcellular localization, the protein has been shown to specifically interact with 5.8S rRNA and with B23/Nucleophosmin and to regulate ribosome biogenesis. Here, we show that the p14ARF protein interacts with chromatin and is recovered by chromatin immunoprecipitation (ChIP) in a fraction that contains a DNA sequence of the rRNA gene promoter. In addition, topoisomerase I (Topo I) that has been shown to interact with p14ARF coprecipitates with p14ARF containing chromatin. These data, in view of the function for Topo I in rRNA transcription, are consistent with a role for the p14ARF-Topo I complex in rRNA transcription and/or maturation.


Subject(s)
Chromatin/metabolism , Genes, rRNA , Promoter Regions, Genetic , Tumor Suppressor Protein p14ARF/physiology , DNA Topoisomerases, Type I/metabolism , Humans
12.
Oncogene ; 22(13): 1945-54, 2003 Apr 03.
Article in English | MEDLINE | ID: mdl-12673200

ABSTRACT

We recently reported an interaction between the p14(ARF) protein and human topoisomerase I (Topo I) resulting in the stimulation of the relaxation activity of Topo I. Our data showed that the complex between the two proteins was located within the nucleolus. In the present work, we have investigated the regions of p14(ARF) involved in this interaction by using targeted point mutagenesis and deletion mutants. A region encompassing exon 2-encoded sequence was required for physical binding of p14(ARF) to Topo I as well as for stimulatory activity of the enzyme. Exon 1 beta-encoded segment was not implicated in the interaction. Moreover, among p14(ARF) point mutants selected for their high conservation among different mammalian species, mutant p14(ARF) (RR87, 88AA) did not stimulate Topo I in spite of its association with the enzyme, suggesting its direct implication in the functional activity of ARF. In contrast, one mutant, p14(ARF) (R71A), was more efficient than wild-type protein to activate Topo I, suggesting that this residue is a key element to modulate Topo I activity. Finally, only ARF-Topo I complexes containing p14(ARF) exon 2 segment were found to be localized in the nucleolus, suggesting that this subnuclear location is linked to the biological function of the ARF-Topo I complex.


Subject(s)
DNA Topoisomerases, Type I/chemistry , Tumor Suppressor Protein p14ARF/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Bone Neoplasms/pathology , Cell Nucleolus/metabolism , Cells, Cultured , DNA Topoisomerases, Type I/metabolism , Enzyme Activation , Genes, p16 , Humans , Molecular Sequence Data , Mutation, Missense , Osteosarcoma/pathology , Point Mutation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Species Specificity , Spodoptera/cytology , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...