50 results on '"Ségal-Bendirdjian E"'
Search Results
2. Epigenetic plasticity of hTERT gene promoter determines retinoid capacity to repress telomerase in maturation-resistant acute promyelocytic leukemia cells
- Author
-
Azouz, A, Wu, Y-L, Hillion, J, Tarkanyi, I, Karniguian, A, Aradi, J, Lanotte, M, Chen, G-Q, Chehna, M, and Ségal-Bendirdjian, E
- Published
- 2010
- Full Text
- View/download PDF
3. Telomerase targeting by retinoids in cells from patients with myeloid leukemias of various subtypes, not only APL
- Author
-
Pendino, F, Hillion, J, Dudognon, C, Delaunay, J, Mourah, S, Podgorniak, M-P, Lafon, I, Chomienne, C, Lanotte, M, Dombret, H, Rousselot, P, and Ségal-Bendirdjian, E
- Published
- 2006
- Full Text
- View/download PDF
4. Retinoid/arsenic combination therapy of promyelocytic leukemia: induction of telomerase-dependent cell death
- Author
-
Tarkanyi, I, Dudognon, C, Hillion, J, Pendino, F, Lanotte, M, Aradi, J, and Ségal-Bendirdjian, E
- Published
- 2005
- Full Text
- View/download PDF
5. A novel mechanism of retinoic acid resistance in acute promyelocytic leukemia cells through a defective pathway in telomerase regulation
- Author
-
Pendino, F, Sahraoui, T, Lanotte, M, and Ségal-Bendirdjian, E
- Published
- 2002
- Full Text
- View/download PDF
6. Autonomous Rexinoid Death Signaling Is Suppressed by Converging Signaling Pathways in Immature Leukemia Cells
- Author
-
Benoit, G R, Flexor, M, Besançon, F, Altucci, L, Rossin, A, Hillion, J, Balajthy, Z, Legres, L, Ségal-Bendirdjian, E, Gronemeyer, H, and Lanotte, M
- Published
- 2001
7. p62/SQSTM1 upregulation constitutes a survival mechanism that occurs during granulocytic differentiation of acute myeloid leukemia cells
- Author
-
Trocoli, A, primary, Bensadoun, P, additional, Richard, E, additional, Labrunie, G, additional, Merhi, F, additional, Schläfli, A M, additional, Brigger, D, additional, Souquere, S, additional, Pierron, G, additional, Pasquet, J-M, additional, Soubeyran, P, additional, Reiffers, J, additional, Ségal-Bendirdjian, E, additional, Tschan, M P, additional, and Djavaheri-Mergny, M, additional
- Published
- 2014
- Full Text
- View/download PDF
8. Epigenetic plasticity of hTERT gene promoter determines retinoid capacity to repress telomerase in maturation-resistant acute promyelocytic leukemia cells.
- Author
-
Azouz, Abdulkader, Wu, Y-L, Hillion, J, Tarkanyi, I, Karniguian, A, Aradi, J, Lanotte, Michele, Chen, G-Q, Chehna, M, Ségal-Bendirdjian, E, Azouz, Abdulkader, Wu, Y-L, Hillion, J, Tarkanyi, I, Karniguian, A, Aradi, J, Lanotte, Michele, Chen, G-Q, Chehna, M, and Ségal-Bendirdjian, E
- Abstract
The expression of hTERT gene, encoding the catalytic subunit of telomerase, is a feature of most cancer cells. Changes in the chromatin environment of its promoter and binding of transcriptional factors have been reported in differentiating cells when its transcription is repressed. However, it is not clear whether these changes are directly involved in this repression or only linked to differentiation. In a maturation-resistant acute promyelocytic leukemia (APL) cell line (NB4-LR1), we have previously identified a new pathway of retinoid-induced hTERT repression independent of differentiation. Using a variant of this cell line (NB4-LR1(SFD)), which resists to this repression, we show that although distinct patterns of histone modifications and transcription factor binding at the proximal domain of hTERT gene promoter could concur to modulate its expression, this region is not sufficient to the on/off switch of hTERT by retinoids. DNA methylation analysis of the hTERT promoter led to the identification of two distinct functional domains, a proximal one, fully unmethylated in both cell lines, and a distal one, significantly methylated in NB4-LR1(SFD) cells, whose methylation was further re-enforced by retinoid treatment. Interestingly, we showed that the binding to this distal domain of a known hTERT repressor, WT1, was defective only in NB4-LR1(SFD) cells. We propose that epigenetic modifications targeting this distal region could modulate the binding of hTERT repressors and account either for hTERT reactivation and resistance to retinoid-induced hTERT downregulation., info:eu-repo/semantics/published
- Published
- 2010
9. Cyclic AMP can promote APL progression and protect myeloid leukemia cells against anthracycline-induced apoptosis
- Author
-
Gausdal, G, primary, Wergeland, A, additional, Skavland, J, additional, Nguyen, E, additional, Pendino, F, additional, Rouhee, N, additional, McCormack, E, additional, Herfindal, L, additional, Kleppe, R, additional, Havemann, U, additional, Schwede, F, additional, Bruserud, Ø, additional, Gjertsen, B T, additional, Lanotte, M, additional, Ségal-Bendirdjian, E, additional, and Døskeland, S O, additional
- Published
- 2013
- Full Text
- View/download PDF
10. Ectopic expression of Bcl-2 switches over nuclear signalling for cAMP-induced apoptosis to granulocytic differentiation
- Author
-
Séité, P, primary, Ruchaud, S, additional, Hillion, J, additional, Gendron, M-C, additional, Bruland, O, additional, Ségal-Bendirdjian, E, additional, Doskeland, S O, additional, Lillehaug, J R, additional, and Lanotte, M, additional
- Published
- 2000
- Full Text
- View/download PDF
11. Inhibition of DNA topoisomerases I and II and induction of apoptosis by erbstatin and tyrphostin derivatives
- Author
-
Markovits, J., primary, Larsen, A.K., additional, Ségal-Bendirdjian, E., additional, Fossé, P., additional, Saucier, J.-M., additional, Gazit, A., additional, Levitzki, A., additional, Umezawa, K., additional, and Jacquemin-Sablon, A., additional
- Published
- 1994
- Full Text
- View/download PDF
12. hTERT DNA Methylation Analysis Identifies a Biomarker for Retinoic Acid-Induced hTERT Repression in Breast Cancer Cell Lines.
- Author
-
Nguyen E, Richerolle A, Sánchez-Bellver J, Varennes J, and Ségal-Bendirdjian E
- Abstract
Telomerase reactivation is responsible for telomere preservation in about 90% of cancers, providing cancer cells an indefinite proliferating potential. Telomerase consists of at least two main subunits: a catalytic reverse transcriptase protein ( hTERT ) and an RNA template subunit. Strategies to inhibit hTERT expression seem promising for cancer treatment. Previous works showed that all- trans retinoic acid (ATRA) induces hTERT repression in acute promyelocytic leukemia cells, resulting in their death. Here, we investigated the effects of ATRA in a subset of breast cancer cell lines. The mutational status of hTERT promoter and the methylation patterns at a single CpG resolution were assessed. We observed an inverse relationship between hTERT expression after ATRA treatment and the methylation level of a specific CpG at chr5: 1,300,438 in a region of hTERT gene at -5 kb of the transcription initiation site. This observation highlighted the significance of this region, whose methylation profile could represent a promising biomarker to predict the sensitivity to ATRA-induced hTERT repression in specific breast cancer subtypes. As hTERT repression promotes drug-induced cell death, checking the methylation status of this unique region and the specific CpG included can help in decision-making to include ATRA in combination therapy and contributes to a better clinical outcome.
- Published
- 2022
- Full Text
- View/download PDF
13. The epigenetic regulator RINF (CXXC5) maintains SMAD7 expression in human immature erythroid cells and sustains red blood cells expansion.
- Author
-
Astori A, Matherat G, Munoz I, Gautier EF, Surdez D, Zermati Y, Verdier F, Zaidi S, Feuillet V, Kadi A, Lauret E, Delattre O, Lefèvre C, Fontenay M, Ségal-Bendirdjian E, Dusanter-Fourt I, Bouscary D, Hermine O, Mayeux P, and Pendino F
- Subjects
- Adult, Animals, Cell Cycle, Epigenesis, Genetic, Humans, Mice, RNA, Messenger, DNA-Binding Proteins, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Smad7 Protein genetics, Transcription Factors
- Abstract
The gene CXXC5, encoding a Retinoid-Inducible Nuclear Factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome (MDS) and adult acute myeloid leukemia (AML). RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknow. Here, we evaluated the consequences of RINF silencing on cytokineinduced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells (RBCs), without affecting cell viability. The phenotype induced by RINF-silencing was TGFβ-dependent and mediated by SMAD7, a TGFβ- signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and MDS patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdowndependent erythroid phenotype. Finally, RINF silencing affects 5’-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a Tet2- anchoring platform in mouse. Altogether, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFβ superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis.
- Published
- 2022
- Full Text
- View/download PDF
14. hMZF-2 , the Elusive Transcription Factor.
- Author
-
Chebly A, Peloponese JM, Ségal-Bendirdjian E, Merlio JP, Tomb R, and Chevret E
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
15. Complex context relationships between DNA methylation and accessibility, histone marks, and hTERT gene expression in acute promyelocytic leukemia cells: perspectives for all-trans retinoic acid in cancer therapy.
- Author
-
Garsuault D, Bouyer C, Nguyen E, Kandhari R, Prochazkova-Carlotti M, Chevret E, Forgez P, and Ségal-Bendirdjian E
- Subjects
- Cell Line, Tumor, Chromatin metabolism, Cluster Analysis, CpG Islands genetics, Epigenesis, Genetic drug effects, Genetic Loci, Genome, Human, Humans, Nucleosomes drug effects, Nucleosomes metabolism, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, Telomerase metabolism, Tretinoin pharmacology, DNA Methylation genetics, Gene Expression Regulation, Leukemic, Histone Code genetics, Leukemia, Promyelocytic, Acute drug therapy, Leukemia, Promyelocytic, Acute genetics, Telomerase genetics, Tretinoin therapeutic use
- Abstract
Telomerase (hTERT) reactivation and sustained expression is a key event in the process of cellular transformation. Therefore, the identification of the mechanisms regulating hTERT expression is of great interest for the development of new anticancer therapies. Although the epigenetic state of hTERT gene promoter is important, we still lack a clear understanding of the mechanisms by which epigenetic changes affect hTERT expression. Retinoids are well-known inducers of granulocytic maturation in acute promyelocytic leukemia (APL). We have previously shown that retinoids repressed hTERT expression in the absence of maturation leading to growth arrest and cell death. Exploring the mechanisms of this repression, we showed that transcription factor binding was dependent on the epigenetic status of hTERT promoter. In the present study, we used APL cells lines and publicly available datasets from APL patients to further investigate the integrated epigenetic events that promote hTERT promoter transition from its silent to its active state, and inversely. We showed, in APL patients, that the methylation of the distal domain of hTERT core promoter was altered and correlated with the outcome of the disease. Further studies combining complementary approaches carried out on APL cell lines highlighted the significance of a domain outside the minimal promoter, localized around 5 kb upstream from the transcription start site, in activating hTERT. This domain is characterized by DNA hypomethylation and H3K4Me3 deposition. Our findings suggest a cooperative interplay between hTERT promoter methylation, chromatin accessibility, and histone modifications that force the revisiting of previously proposed concepts regarding hTERT epigenetic regulation. They represent, therefore, a major advance in predicting sensitivity to retinoid-induced hTERT repression and, more generally, in the potential development of therapies targeting hTERT expression in cancers., (© 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
16. Non-canonical Roles of Telomerase: Unraveling the Imbroglio.
- Author
-
Ségal-Bendirdjian E and Geli V
- Abstract
Telomerase plays a critical role in stem cell function and tissue regeneration that depends on its ability to elongate telomeres. For nearly two decades, it turned out that TERT regulates a broad spectrum of functions including signal transduction, gene expression regulation, and protection against oxidative damage that are independent of its telomere elongation activity. These conclusions that were mainly obtained in cell lines overexpressing telomerase were further strengthened by in vivo models of ectopic expression of telomerase or models of G1 TERT knockout mice without detectable telomere dysfunction. However, the later models were questioned due to the presence of aberrantly shortened telomere in the germline of the parents TERT
+/- that were used to create the G1 TERT-/- mice. The physiological relevance of the functions associated with overexpressed telomerase raised also some concerns due to artifactual situations and localizations and complications to quantify the level of TERT. Another concern with non-canonical functions of TERT was the difficulty to separate a direct TERT-related function from secondary effects. Despite these concerns, more and more evidence accumulates for non-canonical roles of telomerase that are non-obligatory extra-telomeric. Here, we review these non-canonical roles of the TERT subunit of telomerase. Also, we emphasize recent results that link TERT to mitochondria and protection to reactive oxygen species suggesting a protective role of TERT in neurons. Throughout this review, we dissect some controversies regarding the non-canonical functions of telomerase and provide some insights to explain these discrepancies. Finally, we discuss the importance of understanding these alternative functions of telomerase for the development of anticancer strategies., (Copyright © 2019 Ségal-Bendirdjian and Geli.)- Published
- 2019
- Full Text
- View/download PDF
17. Cisplatin increases PD-L1 expression and optimizes immune check-point blockade in non-small cell lung cancer.
- Author
-
Fournel L, Wu Z, Stadler N, Damotte D, Lococo F, Boulle G, Ségal-Bendirdjian E, Bobbio A, Icard P, Trédaniel J, Alifano M, and Forgez P
- Subjects
- A549 Cells, Aged, Animals, Antineoplastic Agents, Immunological pharmacology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor, Cisplatin pharmacology, Drug Synergism, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Immunotherapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lymphatic Metastasis, Male, Mice, Middle Aged, Neoadjuvant Therapy, Up-Regulation drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents, Immunological administration & dosage, B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Cisplatin administration & dosage, Lung Neoplasms drug therapy
- Abstract
The number of clinical protocols testing combined therapies including immune check-point inhibitors and platinum salts is currently increasing in lung cancer treatment, however preclinical studies and rationale are often lacking. Here, we evaluated the impact of cisplatin treatment on PD-L1 expression analyzing the clinicopathological characteristics of patients who received cisplatin-based neoadjuvant chemotherapy followed by surgery and showed that cisplatin-based induction treatment significantly increased PD-L1 staining in both tumor and immune cells from the microenvironment. Twenty-two patients exhibited positive PD-L1 staining variation after neoadjuvant chemotherapy; including 9 (23.1%) patients switching from <50% to ≥50% of stained tumor-cells. We also confirmed the up-regulation of PD-L1 by cisplatin, at both RNA and protein levels, in nude and immunocompetent mice bearing tumors grafted with A549, LNM-R, or LLC1 lung cancer cell lines. The combined administration of anti-PD-L1 antibodies (3 mg/kg) and cisplatin (1 mg/kg) to mice harboring lung carcinoma significantly reduced tumor growth compared to single agent treatments and controls. Overall, these results suggest that cisplatin treatment could synergize with PD-1/PD-L1 blockade to increase the clinical response, in particular through early and sustainable enhancement of PD-L1 expression., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
18. Modulation of lung cancer cell plasticity and heterogeneity with the restoration of cisplatin sensitivity by neurotensin antibody.
- Author
-
Wu Z, Fournel L, Stadler N, Liu J, Boullier A, Hoyeau N, Fléjou JF, Duchatelle V, Djebrani-Oussedik N, Agopiantz M, Ségal-Bendirdjian E, Gompel A, Alifano M, Melander O, Trédaniel J, and Forgez P
- Subjects
- Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung metabolism, Adenocarcinoma of Lung pathology, Animals, Antineoplastic Agents pharmacology, Apoptosis, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Proliferation, Female, Follow-Up Studies, Humans, Lung Neoplasms drug therapy, Lung Neoplasms immunology, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Mice, Mice, Inbred C57BL, Mice, Nude, Neurotensin immunology, Neurotensin metabolism, Prognosis, Receptors, Neurotensin immunology, Receptors, Neurotensin metabolism, Retrospective Studies, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Adenocarcinoma of Lung drug therapy, Antibodies, Monoclonal pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Cisplatin pharmacology, Drug Resistance, Neoplasm, Neurotensin antagonists & inhibitors, Receptors, Neurotensin antagonists & inhibitors
- Abstract
Overall survival of patients with metastatic non-small cell lung cancer (NSCLC) has significantly improved with platinum-based salt treatments and recently with targeted therapies and immunotherapies. However, treatment failure occurs due to acquired or emerging tumor resistance. We developed a monoclonal antibody against the proform of neurotensin (LF-NTS mAb) that alters the homeostasis of tumors overexpressing NTSR1. Neurotensin is frequently overexpressed along with its high affinity receptor (NTSR1) in tumors from epithelial origins. This ligand/receptor complex contributes to the progression of many tumor types by activation of the cellular effects involved in tumor progression (proliferation, survival, migration, and invasion). We demonstrate that LF-NTS mAb operates on the plasticity of tumor cells overexpressing NTSR1 and lowers their aggressiveness. The mAb enables the restoration of platinum-based therapies responsiveness, while also decreasing metastatic processes. Efficacy dosage with long-term treatment showed no obvious adverse events, while demonstrating improvement in the performance status. Our data suggests that LF-NTS mAb is an ideal candidate to be safely added to the conventional standard of care in order to improve its efficacy., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
19. Platinum Complexes Can Bind to Telomeres by Coordination.
- Author
-
Saker L, Ali S, Masserot C, Kellermann G, Poupon J, Teulade-Fichou MP, Ségal-Bendirdjian E, and Bombard S
- Subjects
- Molecular Structure, Telomere chemistry, Cisplatin chemistry, G-Quadruplexes, Platinum chemistry
- Abstract
It is suggested that several compounds, including G-quadruplex ligands, can target telomeres, inducing their uncapping and, ultimately, cell death. However, it has never been demonstrated whether such ligands can bind directly and quantitatively to telomeres. Here, we employed the property of platinum and platinum-G-quadruplex complexes to target G-rich sequences to investigate and quantify their covalent binding to telomeres. Using inductively coupled plasma mass spectrometry, surprisingly, we found that, in cellulo, in the presence of cisplatin, a di-functional platinum complex, telomeric DNA was platinated 13-times less than genomic DNA in cellulo, as compared to in vitro data. On the contrary, the amount of mono-functional platinum complexes (Pt-ttpy and Pt-tpy) bound either to telomeric or to genomic DNA was similar and occurred in a G-quadruplex independent-manner. Importantly, the quantification revealed that the low level of cisplatin bound to telomeric DNA could not be the direct physical cause of TRF2 displacement from telomeres. Altogether, our data suggest that platinum complexes can affect telomeres both directly and indirectly.
- Published
- 2018
- Full Text
- View/download PDF
20. Telomerase regulation by the long non-coding RNA H19 in human acute promyelocytic leukemia cells.
- Author
-
El Hajj J, Nguyen E, Liu Q, Bouyer C, Adriaenssens E, Hilal G, and Ségal-Bendirdjian E
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Down-Regulation, Gene Expression Regulation, Neoplastic drug effects, Humans, Leukemia, Promyelocytic, Acute metabolism, MicroRNAs genetics, RNA, Long Noncoding metabolism, Telomerase metabolism, Leukemia, Promyelocytic, Acute genetics, RNA, Long Noncoding genetics, Telomerase genetics, Tretinoin pharmacology
- Abstract
Background: Since tumor growth requires reactivation of telomerase (hTERT), this enzyme is a challenging target for drug development. Therefore, it is of great interest to identify telomerase expression and activity regulators. Retinoids are well-known inducers of granulocytic maturation associated with hTERT repression in acute promyelocytic leukemia (APL) blasts. In a maturation-resistant APL cell line, we have previously identified a new pathway of retinoid-induced hTERT transcriptional repression independent of differentiation. Furthermore, we reported the isolation of a cell variant resistant to this repression. Those cell lines could serve as unique tools to identify new telomerase regulators., Methods: Using a microarray approach we identified the long non-coding RNA, H19 as a potential candidate playing a role in telomerase regulation. Expression of H19, hTERT, and hTR were examined by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Telomerase activity was quantified by quantitative telomeric repeats amplification protocol (qTRAP). In vitro and in vivo assays were performed to investigate H19 function on telomerase expression and activity., Results: We showed both in retinoid-treated cell lines and in APL patient cells an inverse relationship between the expression of H19 and the expression and activity of hTERT. Exploring the mechanistic link between H19 and hTERT regulation, we showed that H19 is able to impede telomerase function by disruption of the hTERT-hTR interaction., Conclusions: This study identifies a new way of telomerase regulation through H19's involvement and thereby reveals a new function for this long non-coding RNA that can be targeted for therapeutic purpose.
- Published
- 2018
- Full Text
- View/download PDF
21. Neurotensin Receptor 1 Antagonist SR48692 Improves Response to Carboplatin by Enhancing Apoptosis and Inhibiting Drug Efflux in Ovarian Cancer.
- Author
-
Liu J, Agopiantz M, Poupon J, Wu Z, Just PA, Borghese B, Ségal-Bendirdjian E, Gauchotte G, Gompel A, and Forgez P
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Female, Humans, Middle Aged, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Pyrazoles adverse effects, Quinolines adverse effects, Receptors, Neurotensin antagonists & inhibitors, Xenograft Model Antitumor Assays, Carboplatin administration & dosage, Ovarian Neoplasms drug therapy, Pyrazoles administration & dosage, Quinolines administration & dosage, Receptors, Neurotensin genetics
- Abstract
Purpose: The high affinity receptor 1 (NTSR1) and its agonist, neurotensin (NTS), are correlated with tumor cell aggressiveness in most solid tumors. As chemoresistance and tumor aggressiveness are often related, we decided to study the role of the NTSR1 complex within platinum-based chemotherapy responses. In an ovarian model, we studied carboplatin because it is the main standard of care for ovarian cancer. Experimental Design: Experimental tumors and in vitro studies were performed using SKOV3 and A2780 cells treated with carboplatin, with or without a very specific NTSR1 antagonist, SR48692. We measured the effects of these treatments on cell apoptosis and apoptosis-related proteins, platinum accumulation in the cell and nucleus, and the expression and localization of platinum transporters. NTS and NTSR1 labeling was measured in patients with ovarian cancer. Results: SR48692 enhanced the response to carboplatin in ovarian cancer cells and experimental tumors. When SR48692 is combined with carboplatin, we noted a major improvement of platinum-induced DNA damage and cell death, as well as a decrease in tumor growth. The relationship of these results to clinical studies was made by the detection of NTS and NTSR1 in 72% and 74% of ovarian cancer, respectively. Furthermore, in a large series of high-grade ovarian cancer, NTSR1 mRNA was shown to correlate with higher stages and platinum resistance. Conclusions: This study strongly suggests that the addition of NTSR1 inhibitor in combination with platinum salt-based therapy will improve the response to the drug. Clin Cancer Res; 23(21); 6516-28. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
22. Association of a Platinum Complex to a G-Quadruplex Ligand Enhances Telomere Disruption.
- Author
-
Charif R, Granotier-Beckers C, Bertrand HC, Poupon J, Ségal-Bendirdjian E, Teulade-Fichou MP, Boussin FD, and Bombard S
- Subjects
- Acridines toxicity, Cell Cycle Checkpoints drug effects, Cell Line, Cell Proliferation drug effects, Cisplatin toxicity, DNA Damage drug effects, Humans, Ligands, Microscopy, Fluorescence, Organoplatinum Compounds toxicity, Telomere metabolism, Telomere Shortening drug effects, Telomeric Repeat Binding Protein 2 metabolism, Coordination Complexes toxicity, G-Quadruplexes drug effects, Platinum chemistry, Telomere drug effects
- Abstract
Telomeres protect the ends of chromosomes against illegitimate recombination and repair. They can be targets for G-quadruplex ligands and platinum complexes due to their repeated G-rich sequences. Protection of telomeres is ensured by a complex of six proteins, including TRF2, which inhibits the DNA damage response pathway. We analyzed telomere modifications induced in cancer cells by the experimental hybrid platinum complex, Pt-MPQ, comprising both an ethylene diamine monofunctional platinum complex and a G-quadruplex recognition moiety (MPQ). Pt-MPQ promotes the displacement of two telomeric proteins (TRF2 and TRF1) from telomeres, as well as the formation of telomere damage and telomere sister losses, whereas the control compound MPQ does not. This suggests that the platinum moiety potentiates the targeting of the G-quadruplex ligand to telomeres, opening a new perspective for telomere biology and anticancer therapy. Interestingly, the chemotherapy drug cisplatin, which has no specific affinity for G-quadruplex structures, partially induces the TRF2 delocalization from telomeres but produces less telomeric DNA damage, suggesting that this TRF2 displacement could be independent of G-quadruplex recognition.
- Published
- 2017
- Full Text
- View/download PDF
23. Exploring the mechanism of inhibition of human telomerase by cysteine-reactive compounds.
- Author
-
Kellermann G, Dingli F, Masson V, Dauzonne D, Ségal-Bendirdjian E, Teulade-Fichou MP, Loew D, and Bombard S
- Subjects
- Amino Acid Sequence, Binding Sites genetics, Cysteine chemistry, Cysteine genetics, Humans, Mass Spectrometry methods, Models, Molecular, Molecular Structure, Mutagenesis, Protein Binding, Protein Domains, Recombinant Proteins chemistry, Reverse Transcriptase Inhibitors chemistry, Reverse Transcriptase Inhibitors pharmacology, Sequence Homology, Amino Acid, Structure-Activity Relationship, Telomerase antagonists & inhibitors, Telomerase genetics, Cysteine metabolism, Recombinant Proteins metabolism, Reverse Transcriptase Inhibitors metabolism, Telomerase metabolism
- Abstract
Telomerase is an almost universal cancer target that consists minimally of a core protein human telomerase reverse transcriptase (hTERT) and a RNA component human telomerase RNA (hTR). Some inhibitors of this enzyme are thought to function by the covalent binding to one or several cysteine residues; however, this inhibition mechanism has never been investigated because of the difficulty in producing telomerase. In this study, we use a recent method to produce recombinant hTERT to analyze the effect of cysteine-reactive inhibitors on telomerase. Using mass spectrometry and mutagenesis analysis, we identify several targeted residues in separated domains of the hTERT protein and show that cysteine-reactive reagents abolish the interaction with the CR4/5 region of hTR., (© 2017 Federation of European Biochemical Societies.)
- Published
- 2017
- Full Text
- View/download PDF
24. Heparan Sulfate Proteoglycans Promote Telomerase Internalization and MHC Class II Presentation on Dendritic Cells.
- Author
-
Galaine J, Kellermann G, Guillaume Y, Boidot R, Picard E, Loyon R, Queiroz L, Boullerot L, Beziaud L, Jary M, Mansi L, André C, Lethier L, Ségal-Bendirdjian E, Borg C, Godet Y, and Adotévi O
- Subjects
- CD4-Positive T-Lymphocytes immunology, Cell Line, Tumor, Epitopes, T-Lymphocyte immunology, Humans, Immunotherapy, Lymphocyte Activation, Monocytes, Peptides metabolism, Telomerase immunology, Antigen Presentation, Dendritic Cells immunology, HLA-DR Antigens immunology, Heparan Sulfate Proteoglycans metabolism, Telomerase metabolism
- Abstract
Telomerase is a prototype-shared tumor Ag and represents an attractive target for anticancer immunotherapy. We have previously described promiscuous and immunogenic HLA-DR-restricted peptides derived from human telomerase reverse transcriptase (hTERT) and referred as universal cancer peptide (UCP). In nonsmall cell lung cancer, the presence of spontaneous UCP-specific CD4 T cell responses increases the survival of chemotherapy-responding patients. However, the precise mechanisms of hTERT's uptake, processing, and presentation on MHC-II molecules to stimulate CD4 T cells are poorly understood. In this work, by using well-characterized UCP-specific CD4 T cell clones, we showed that hTERT processing and presentation on MHC-II involve both classical endolysosomal and nonclassical cytosolic pathways. Furthermore, to our knowledge, we demonstrated for the first time that hTERT's internalization by dendritic cells requires its interaction with surface heparan sulfate proteoglycans. Altogether, our findings provide a novel mechanism of tumor-specific CD4 T cell activation and will be useful for the development of novel cancer immunotherapies that harness CD4 T cells., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
25. WT1 expression is inversely correlated with MYCN amplification or expression and associated with poor survival in non-MYCN-amplified neuroblastoma.
- Author
-
Masserot C, Liu Q, Nguyen E, Gattolliat CH, Valteau-Couanet D, Bénard J, Huber C, and Ségal-Bendirdjian E
- Subjects
- Adolescent, Cell Line, Tumor, Child, Child, Preschool, Disease-Free Survival, Gene Expression Regulation, Neoplastic, Genetic Heterogeneity, Humans, Infant, Infant, Newborn, N-Myc Proto-Oncogene Protein, Biomarkers, Tumor metabolism, Gene Amplification, Neuroblastoma genetics, Neuroblastoma mortality, Nuclear Proteins genetics, Oncogene Proteins genetics, WT1 Proteins metabolism
- Abstract
Neuroblastoma (NB) is the most common extra cranial solid tumor in childhood and the most frequently diagnosed neoplasm during infancy. A striking feature of this tumor is its clinical heterogeneity. Several tumor progression markers have been delineated so far, among which MYCN amplification, which occurs in about 25% of total NB cases, with the percentage increasing to 30% in advanced stage NB. Although MYCN amplification is strongly correlated with NB of poor outcome, the MYCN status cannot alone predict all cases of poor survival in NB. Indeed NB without MYCN amplification (about 70-80% of NB) are not always favorable. WT1 was initially identified as a tumor suppressor gene involved in the development of a pediatric renal tumor (Wilms' tumor). Here, we describe an inverse correlation between WT1 expression and MYCN amplification and expression. However and most notably, our results show that WT1 gene expression is associated with a poor outcome for patients showing non-MYCN-amplified tumors. Thus WT1 expression is clinically significant in NB and may be a prognostic marker for better risk stratification and for an optimized therapeutic management of NB., (Copyright © 2015 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Identification of human telomerase assembly inhibitors enabled by a novel method to produce hTERT.
- Author
-
Kellermann G, Kaiser M, Dingli F, Lahuna O, Naud-Martin D, Mahuteau-Betzer F, Loew D, Ségal-Bendirdjian E, Teulade-Fichou MP, and Bombard S
- Subjects
- Acridines chemistry, Acridines pharmacology, Genetic Techniques, HEK293 Cells, Humans, Niacinamide analogs & derivatives, RNA chemistry, RNA metabolism, RNA Folding, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins isolation & purification, Recombinant Fusion Proteins metabolism, Small Molecule Libraries, Structure-Activity Relationship, Telomerase antagonists & inhibitors, Telomerase chemistry, Telomerase genetics, Telomerase metabolism, Thiazoles, Trisaccharides chemistry, Trisaccharides pharmacology, Telomerase biosynthesis
- Abstract
Telomerase is the enzyme that maintains the length of telomeres. It is minimally constituted of two components: a core reverse transcriptase protein (hTERT) and an RNA (hTR). Despite its significance as an almost universal cancer target, the understanding of the structure of telomerase and the optimization of specific inhibitors have been hampered by the limited amount of enzyme available. Here, we present a breakthrough method to produce unprecedented amounts of recombinant hTERT and to reconstitute human telomerase with purified components. This system provides a decisive tool to identify regulators of the assembly of this ribonucleoprotein complex. It also enables the large-scale screening of small-molecules capable to interfere with telomerase assembly. Indeed, it has allowed us to identify a compound that inhibits telomerase activity when added prior to the assembly of the enzyme, while it has no effect on an already assembled telomerase. Therefore, the novel system presented here may accelerate the understanding of human telomerase assembly and facilitate the discovery of potent and mechanistically unique inhibitors., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
27. A preclinical mouse model of glioma with an alternative mechanism of telomere maintenance (ALT).
- Author
-
Jeitany M, Pineda JR, Liu Q, Porreca RM, Hoffschir F, Desmaze C, Silvestre DC, Mailliet P, Junier MP, Londoño-Vallejo A, Ségal-Bendirdjian E, Chneiweiss H, and Boussin FD
- Subjects
- Adult, Animals, Cell Line, Tumor, DNA Methylation, Disease Models, Animal, G-Quadruplexes, Gene Expression Regulation, Glioma metabolism, Heterografts, Humans, Interleukin Receptor Common gamma Subunit genetics, Ligands, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Phenotype, Sister Chromatid Exchange, Telomerase genetics, Telomerase metabolism, Telomere metabolism, Telomere Homeostasis, Glioma genetics, Telomere genetics
- Abstract
Glioblastoma multiforme is the most aggressive primary tumor of the central nervous system. Glioma stem cells (GSCs), a small population of tumor cells with stem-like properties, are supposedly responsible for glioblastoma multiforme relapse after current therapies. In approximately thirty percent of glioblastoma multiforme tumors, telomeres are not maintained by telomerase but through an alternative mechanism, termed alternative lengthening of telomere (ALT), suggesting potential interest in developing specific therapeutic strategies. However, no preclinical model of ALT glioma was available until the isolation of TG20 cells from a human ALT glioma. Herein, we show that TG20 cells exhibit a high level of telomeric recombination but a stable karyotype, indicating that their telomeres retain their protective function against chromosomal instability. TG20 cells possess all of the characteristic features of GSCs: the expression of neural stem cell markers, the generation of intracerebral tumors in NOD-SCID-IL2Rγ (NSG) mice as well as in nude mice, and the ability to sustain serial intracerebral transplantations without expressing telomerase, demonstrating the stability of the ALT phenotype in vivo. Furthermore, we also demonstrate that 360B, a G-quadruplex ligand of the pyridine derivative series that impairs telomere replication and mitotic progression in cancer cells, prevents the development of TG20 tumors. Together, our results show that intracerebral grafts of TG20 cells in immunodeficient mice constitute an efficient preclinical model of ALT glioblastoma multiforme and that G-quadruplex ligands are a potential therapy for this specific type of tumor., (© 2014 The Authors. Published by Wiley Periodicals, Inc. on behalf of UICC.)
- Published
- 2015
- Full Text
- View/download PDF
28. Pro-survival role of p62 during granulocytic differentiation of acute myeloid leukemia cells.
- Author
-
Ségal-Bendirdjian E, Tschan MP, Reiffers J, and Djavaheri-Mergny M
- Abstract
p62 regulates key signaling pathways including those that control cell death and autophagy. Recently, we reported that p62 is upregulated during all-trans retinoic acid (ATRA)-induced terminal differentiation of acute myeloid leukemia (AML) cells. This response reduces levels of ubiquitinated protein aggregates in mature cells and protects these cells against ATRA treatment. Thus, p62 confers a survival advantage to mature AML cells.
- Published
- 2014
- Full Text
- View/download PDF
29. Activation of both protein kinase A (PKA) type I and PKA type II isozymes is required for retinoid-induced maturation of acute promyelocytic leukemia cells.
- Author
-
Nguyen E, Gausdal G, Varennes J, Pendino F, Lanotte M, Døskeland SO, and Ségal-Bendirdjian E
- Subjects
- Cell Differentiation drug effects, Cell Line, Tumor, Cyclic AMP metabolism, Cytoplasm drug effects, Cytoplasm metabolism, Humans, Isoenzymes metabolism, Leukemia, Promyelocytic, Acute enzymology, Antineoplastic Agents pharmacology, Cyclic AMP-Dependent Protein Kinase Type I metabolism, Cyclic AMP-Dependent Protein Kinase Type II metabolism, Leukemia, Promyelocytic, Acute drug therapy, Leukemia, Promyelocytic, Acute pathology, Tretinoin pharmacology
- Abstract
Acute promyelocytic leukemia (APL) is characterized by granulopoietic differentiation arrest at the promyelocytic stage. In most cases, this defect can be overcome by treatment with all-trans-retinoic acid (ATRA), leading to complete clinical remission. Cyclic AMP signaling has a key role in retinoid treatment efficacy: it enhances ATRA-induced maturation in ATRA-sensitive APL cells (including NB4 cells) and restores it in some ATRA-resistant cells (including NB4-LR1 cells). We show that the two cell types express identical levels of the Cα catalytic subunit and comparable global cAMP-dependent protein kinase A (PKA) enzyme activity. However, the maturation-resistant NB4-LR1 cells have a PKA isozyme switch: compared with the NB4 cells, they have decreased content of the juxtanuclearly located PKA regulatory subunit IIα and PKA regulatory subunit IIβ, and a compensatory increase of the generally cytoplasmically distributed PKA-RIα. Furthermore, the PKA regulatory subunit II exists mainly in the less cAMP-responsive nonautophosphorylated state in the NB4-LR1 cells. By the use of isozyme-specific cAMP analog pairs, we show that both PKA-I and PKA-II must be activated to achieve maturation in NB4-LR1 as well as NB4 cells. Therefore, special attention should be paid to activating not only PKA-I but also PKA-II in attempts to enhance ATRA-induced APL maturation in a clinical setting.
- Published
- 2013
- Full Text
- View/download PDF
30. Antitumor trans-N-heterocyclic carbene-amine-Pt(II) complexes: synthesis of dinuclear species and exploratory investigations of DNA binding and cytotoxicity mechanisms.
- Author
-
Chtchigrovsky M, Eloy L, Jullien H, Saker L, Ségal-Bendirdjian E, Poupon J, Bombard S, Cresteil T, Retailleau P, and Marinetti A
- Subjects
- Antineoplastic Agents chemical synthesis, Apoptosis drug effects, Apoptosis Inducing Factor metabolism, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cisplatin pharmacology, DNA Fragmentation, Diamines chemical synthesis, Diamines pharmacology, Humans, Inhibitory Concentration 50, Organoplatinum Compounds chemical synthesis, Antineoplastic Agents pharmacology, DNA metabolism, Organoplatinum Compounds pharmacology
- Abstract
A series of bimetallic [(NHC)PtX2]2(diamine) complexes have been prepared as a new chemotype for potential anticancer agents. These complexes display an uncommon set of structural features as far as they combine two bifunctional, trans-configured platinum centers. They display cytotoxic activities in the micromolar range on many cancerous cell lines and do not cross-react with cisplatin in A2780/DDP cell lines. They bind slowly to double-stranded DNAs, giving monoadducts as the major products. Pathways for cellular toxicity have been investigated for both mono- and bimetallic trans-(NHC)PtX2(amine) complexes. It has been highlighted that, unlike cisplatin, these complexes do not induce cell cycle arrest. They trigger apoptosis in A2780 cells by a pathway involving translocation of apoptosis-inducing factor and caspase 12 to the nucleus. Moreover, bimetallic complexes may induce necrosis.
- Published
- 2013
- Full Text
- View/download PDF
31. Loss of the malignant phenotype of human neuroblastoma cells by a catalytically inactive dominant-negative hTERT mutant.
- Author
-
Samy M, Gattolliat CH, Pendino F, Hillion J, Nguyen E, Bombard S, Douc-Rasy S, Bénard J, and Ségal-Bendirdjian E
- Subjects
- Animals, Apoptosis, Caspase 8 metabolism, Cell Line, Tumor, Cell Shape, Cell Transformation, Neoplastic pathology, Child, Genome, Human genetics, Humans, Male, Mice, Mice, Nude, N-Myc Proto-Oncogene Protein, Neuroblastoma genetics, Nuclear Proteins, Oncogene Proteins, Phenotype, Telomere Homeostasis, Transduction, Genetic, Tumor Suppressor Protein p53 metabolism, Biocatalysis, Genes, Dominant genetics, Mutant Proteins metabolism, Neuroblastoma enzymology, Neuroblastoma pathology, Telomerase metabolism
- Abstract
Telomerase, a ribonucleoprotein complex mainly composed of the reverse transcriptase catalytic subunit (human telomerase reverse transcriptase, hTERT) and the RNA component (hTR), is a key enzyme of cancer progression. That aggressive stage 4-neuroblastoma expressed high levels of telomerase activity, whereas favorable tumors had no or little telomerase expression and activity, prompted us to investigate the role of this enzyme in this tumor model of altered proliferation, neuronal differentiation, and apoptosis. A human MYCN-amplified neuroblastoma cell line (IGR-N-91) was engineered to stably express either the normal hTERT protein (WT-hTERT) or a catalytically inactive dominant-negative mutant of this protein (DN-hTERT). We showed that DN-hTERT expression inhibited the endogenous hTERT in the malignant neuroblasts without telomere shortening nor loss of in vitro proliferative capacity. Importantly, DN-hTERT expression induced major changes in cell morphology of neuroblasts that switched them from a neuronal to a substrate adherent phenotype, which was more prone to apoptosis and lost their tumorigenic properties in nude mice. These biologic effects arose from modifications in the expression of genes involved in both apoptosis and neuroblastoma biology. Taken together these results highlighted the functional relevance of noncanonical functions of hTERT in the determination of neuroblast cell fate. Therefore, our results envision new therapeutic strategies for metastatic neuroblastoma therapeutic management., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
32. cFos mediates cAMP-dependent generation of ROS and rescue of maturation program in retinoid-resistant acute promyelocytic leukemia cell line NB4-LR1.
- Author
-
Carrier JL, Javadi P, Bourrier E, Camus C, Ségal-Bendirdjian E, and Karniguian A
- Subjects
- Alternative Splicing, Cell Line, Tumor, Cell Membrane metabolism, DNA Primers genetics, Drug Resistance, Neoplasm, Epigenesis, Genetic, Exons, Genetic Variation, Humans, Hyaluronan Receptors biosynthesis, Neutrophils metabolism, RNA Interference, RNA, Messenger metabolism, Reactive Oxygen Species, Time Factors, Cyclic AMP metabolism, Proto-Oncogene Proteins c-fos metabolism, Retinoids pharmacology
- Abstract
A determining role has been assigned to cAMP in the signaling pathways that relieve resistance to anti-leukemia differentiation therapy. However, the underlying mechanisms have not been elucidated yet. Here, we identify cFos as a critical cAMP effector, able to regulate the re-expression and splicing of epigenetically silenced genes associated with maturation (CD44) in retinoid-resistant NB4-LR1 leukemia cells. Furthermore, using RNA interference approach, we show that cFos mediates cAMP-induced ROS generation, a critical mediator of neutrophil maturation, and in fine differentiation. This study highlights some of the mechanisms by which cAMP acts to overcome resistance, and reveals a new alternative cFos-dependent pathway which, though nonexistent in retinoid-sensitive NB4 cells, is essential to rescue the maturation program of resistant cells.
- Published
- 2012
- Full Text
- View/download PDF
33. hTERT promotes imatinib resistance in chronic myeloid leukemia cells: therapeutic implications.
- Author
-
Deville L, Hillion J, Pendino F, Samy M, Nguyen E, and Ségal-Bendirdjian E
- Subjects
- Apoptosis drug effects, Benzamides, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, Enzyme Activation drug effects, Gene Expression Regulation, Enzymologic drug effects, HEK293 Cells, Humans, Imatinib Mesylate, K562 Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Piperazines chemistry, Pyrimidines chemistry, Telomerase genetics, Transcription, Genetic drug effects, Tretinoin chemistry, Tretinoin pharmacology, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Piperazines pharmacology, Pyrimidines pharmacology, Telomerase metabolism
- Abstract
Imatinib mesylate has shown remarkable efficacy in the treatment of patients in the chronic phase of chronic myeloid leukemia. However, despite an overall significant hematological and cytogenetic response, imatinib therapy may favor the emergence of drug-resistant clones, ultimately leading to relapse. Some imatinib resistance mechanisms had not been fully elucidated yet. In this study we used sensitive and resistant sublines from a Bcr-Abl positive cell line to investigate the putative involvement of telomerase in the promotion of imatinib resistance. We showed that sensitivity to imatinib can be partly restored in imatinib-resistant cells by targeting telomerase expression, either by the introduction of a dominant-negative form of the catalytic protein subunit of the telomerase (hTERT) or by the treatment with all-trans-retinoic acid, a clinically used drug. Furthermore, we showed that hTERT overexpression favors the development of imatinib resistance through both its antiapoptotic and telomere maintenance functions. Therefore, combining antitelomerase strategies to imatinib treatment at the beginning of the treatment should be promoted to reduce the risk of imatinib resistance development and increase the probability of eradicating the disease.
- Published
- 2011
- Full Text
- View/download PDF
34. The telomere story or the triumph of an open-minded research.
- Author
-
Gilson E and Ségal-Bendirdjian E
- Subjects
- Animals, Cell Biology history, Chromosomes metabolism, History, 19th Century, History, 20th Century, Humans, Nobel Prize, Cellular Senescence physiology, Telomerase metabolism, Telomere metabolism
- Abstract
The Nobel Assembly at Karolinska Institute has decided to award The Nobel Prize in Physiology or Medicine 2009 jointly to Elizabeth H. Blackburn, Carol W. Greider and Jack W. Szostak for the discovery of "how chromosomes are protected by telomeres and the enzyme telomerase". This discovery had major impacts within the scientific community and led to intense research in this field. All the studies performed are now the bases for future investigations and stimulate the development of potential new therapies., (Copyright (c) 2010 Elsevier Masson SAS. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
35. Functional involvement of RINF, retinoid-inducible nuclear factor (CXXC5), in normal and tumoral human myelopoiesis.
- Author
-
Pendino F, Nguyen E, Jonassen I, Dysvik B, Azouz A, Lanotte M, Ségal-Bendirdjian E, and Lillehaug JR
- Subjects
- Amino Acid Sequence, Carrier Proteins genetics, DNA-Binding Proteins, Gene Expression Profiling, Gene Expression Regulation drug effects, Granulocyte Precursor Cells drug effects, Granulocyte Precursor Cells physiology, HL-60 Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, K562 Cells, Models, Biological, Molecular Sequence Data, Myelopoiesis drug effects, Oligonucleotide Array Sequence Analysis, Sequence Homology, Amino Acid, Transcription Factors, Tretinoin pharmacology, Tumor Cells, Cultured, Carrier Proteins physiology, Hematologic Neoplasms genetics, Intracellular Signaling Peptides and Proteins physiology, Myelopoiesis genetics
- Abstract
Retinoids triggers differentiation of acute promyelocytic leukemia (APL) blasts by transcriptional regulation of myeloid regulatory genes. Using a microarray approach, we have identified a novel retinoid-responsive gene (CXXC5) encoding a nuclear factor, retinoid-inducible nuclear factor (RINF), that contains a CXXC-type zinc-finger motif. RINF expression correlates with retinoid-induced differentiation of leukemic cells and with cytokine-induced myelopoiesis of normal CD34(+) progenitors. Furthermore, short hairpin RNA (shRNA) interference suggests for this gene a regulatory function in both normal and tumoral myelopoiesis. Interestingly, RINF localizes to 5q31.3, a small region often deleted in myeloid leukemia (acute myeloid leukemia [AML]/myelodysplasia [MDS]) and suspected to harbor one or several tumor suppressor gene.
- Published
- 2009
- Full Text
- View/download PDF
36. Telomerase regulation in hematological cancers: a matter of stemness?
- Author
-
Deville L, Hillion J, and Ségal-Bendirdjian E
- Subjects
- Animals, DNA, Neoplasm metabolism, Hematologic Neoplasms drug therapy, Humans, Neoplasm Proteins antagonists & inhibitors, Telomerase antagonists & inhibitors, Telomere metabolism, Cell Transformation, Neoplastic metabolism, Gene Expression Regulation, Leukemic, Gene Expression Regulation, Neoplastic, Hematologic Neoplasms enzymology, Leukemia enzymology, Neoplasm Proteins biosynthesis, Telomerase biosynthesis
- Abstract
Human telomerase is a nuclear ribonucleoprotein enzyme complex that catalyzes the synthesis and extension of telomeric DNA. This enzyme is highly expressed and active in most malignant tumors while it is usually not or transiently detectable in normal somatic cells, suggesting that it plays an important role in cellular immortalization and tumorigenesis. As most leukemic cells are generally telomerase-positive and have often shortened telomeres, our understanding of how telomerase is deregulated in these diseases could help to define novel therapies targeting the telomere/telomerase complex. Nonetheless, considering that normal hematopoietic stem cells and some of their progeny do express a functional telomerase, it is tempting to consider such an activity in leukemias as a sustained stemness feature and important to understand how telomere length and telomerase activity are regulated in the various forms of leukemias.
- Published
- 2009
- Full Text
- View/download PDF
37. Telomeres and telomerase: from basic research to clinical applications.
- Author
-
Ségal-Bendirdjian E and Gilson E
- Subjects
- Animals, Cellular Senescence, Chromatin physiology, Humans, Telomerase genetics, Telomere genetics, Telomerase metabolism, Telomere physiology
- Published
- 2008
- Full Text
- View/download PDF
38. Immunodetection of human telomerase reverse-transcriptase (hTERT) re-appraised: nucleolin and telomerase cross paths.
- Author
-
Wu YL, Dudognon C, Nguyen E, Hillion J, Pendino F, Tarkanyi I, Aradi J, Lanotte M, Tong JH, Chen GQ, and Ségal-Bendirdjian E
- Subjects
- Antibodies metabolism, Antibody Specificity, Cell Differentiation, Cells, Cultured, Cross Reactions, DNA-Binding Proteins deficiency, Electrophoresis, Gel, Two-Dimensional, Electrophoresis, Polyacrylamide Gel, Fluorescent Antibody Technique, Gene Expression Profiling, HeLa Cells, Humans, Immunoprecipitation, Mass Spectrometry, Peptide Mapping, RNA, Messenger metabolism, Telomerase deficiency, Nucleolin, DNA-Binding Proteins analysis, Phosphoproteins analysis, RNA-Binding Proteins analysis, Telomerase analysis
- Abstract
The involvement of telomerase in cellular immortalization and senescence has often been assessed by means of telomerase expression at the RNA level and quantification of telomerase activity by the telomeric repeat amplification protocol assay. However, these methods either neglected the existence of various telomerase splice variants, or ignored the nonconventional functions of telomerase independent of its ability to elongate and maintain telomere length. Immunodetection of telomerase is now being recognized as a necessary approach to precisely elucidate its roles in oncogenesis and senescence. A few antibodies directed against the catalytic subunit of the human telomerase (hTERT) are currently used but their specificity is not always demonstrated. A survey of the literature showed inconsistencies and led us to comparatively re-evaluate the most frequently used antibodies. Surprisingly, mass spectrometry, two-dimensional gel analysis and immunofluorescent experiments revealed that the most frequently used hTERT immunoprobe, a mouse monoclonal antibody that was claimed to be directed against an hTERT protein epitope, in fact recognizes nucleolin rather than telomerase. Our findings have interesting implications regarding the biology of nucleolin and telomerase in the context of pathophysiological investigations recently carried out.
- Published
- 2006
- Full Text
- View/download PDF
39. Diagnostics, prognostic and therapeutic exploitation of telomeres and telomerase in leukemias.
- Author
-
Deville L, Hillion J, Lanotte M, Rousselot P, and Ségal-Bendirdjian E
- Subjects
- Humans, Prognosis, Leukemia diagnosis, Leukemia enzymology, Leukemia genetics, Leukemia therapy, Telomerase metabolism, Telomere metabolism
- Abstract
Telomeres are specialized structures at the end of human chromosomes. Telomere length decreases with each cell division, thus, reflecting the mitotic history of somatic cells. Telomerase, the ribonucleoprotein enzyme which maintains telomeres of eukaryotic chromosomes, is up-regulated in the vast majority of human neoplasia but not in normal somatic tissues. In contrast to other somatic cells, normal primitive human hematopoietic cells and some peripheral blood cells expressed low levels of telomerase activity. This activity is thought to play an important role in self-renewal of hematopoietic stem cells. In malignant disorders, telomere lengths are generally shortened and telomerase expression and activity enhanced with high differences in the levels. Although it is necessary to be cautious in interpreting these data, there are indications that telomere length and telomerase expression and activity can serve as a molecular marker of the clinical progression and prognosis of most leukemias. Approaches that directly target telomerase, telomeres or telomerase regulatory mechanisms have been developed. Some of these anti-telomerase strategies in combination with conventional drugs proved to be promising in some types of leukemias.
- Published
- 2006
- Full Text
- View/download PDF
40. Telomeres and telomerase: Pharmacological targets for new anticancer strategies?
- Author
-
Pendino F, Tarkanyi I, Dudognon C, Hillion J, Lanotte M, Aradi J, and Ségal-Bendirdjian E
- Subjects
- Animals, Humans, Neoplasms pathology, Telomere chemistry, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Neoplasms drug therapy, Telomerase antagonists & inhibitors, Telomere drug effects
- Abstract
Telomeres are located at the ends of eukaryotic chromosomes. Human telomerase, a cellular reverse transcriptase, is a ribonucleoprotein enzyme that catalyzes the synthesis and extension of telomeric DNA. It is composed of at least, a template RNA component (hTR; human Telomerase RNA) and a catalytic subunit, the telomerase reverse transcriptase (hTERT). The absence of telomerase is associated with telomere shortening and aging of somatic cells, while high telomerase activity is observed in over 85% of human cancer cells, strongly indicating its key role during tumorigenesis. Several details regarding telomere structure and telomerase regulation have already been elucidated, providing new targets for therapeutic exploitation. Further support for anti-telomerase approaches comes from recent studies indicating that telomerase is endowed of additional functions in the control of growth and survival of tumor cells that do not depend only on the ability of this enzyme to maintain telomere length. This observation suggests that inhibiting telomerase or its synthesis may have additional anti-proliferative and apoptosis inducing effect, independently of the reduction of telomere length during cell divisions. This article reviews the basic information about the biology of telomeres and telomerase and attempts to present various approaches that are currently under investigation to inhibit its expression and its activity. We summarize herein distinct anti-telomerase approaches like antisense strategies, reverse transcriptase inhibitors, and G-quadruplex interacting agents, and also review molecules targeting hTERT expression, such as retinoids and evaluate them for their therapeutic potential. "They conceive a certain theory, and everything has to fit into that theory. If one little fact will not fit it, they throw it aside. But it is always the facts that will not fit in that are significant". "Death on the Nile". Agatha Christie.
- Published
- 2006
- Full Text
- View/download PDF
41. Inhibition of human telomerase by oligonucleotide chimeras, composed of an antisense moiety and a chemically modified homo-oligonucleotide.
- Author
-
Tarkanyi I, Horváth A, Szatmari I, Eizert H, Vámosi G, Damjanovich S, Ségal-Bendirdjian E, and Aradi J
- Subjects
- Base Sequence, Cell Line, Tumor, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, HIV enzymology, Humans, Inhibitory Concentration 50, Molecular Structure, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense metabolism, RNA-Directed DNA Polymerase metabolism, Telomerase metabolism, Time Factors, Oligonucleotides, Antisense chemistry, Oligonucleotides, Antisense pharmacology, Telomerase antagonists & inhibitors
- Abstract
Most tumor cells attain their immortality by reactivating telomerase. We report here the telomerase inhibitory potential of chimeric oligonucleotides composed of a 13mer antisense sequence targeting the telomerase RNA template region and a (s4dU)n moiety at its 3' or 5'-end. The increase of the thiolated chain length enhances the telomerase inhibitory potential, but decreases specificity, indicated by HIV reverse transcriptase inhibition. Chimeras with 5' (s4dU)(n)s were more potent inhibitors than the antisense alone or the 3' modified ones. Cy5-labeled (s4dU)4AS and (s4dU)8AS proved the internalization of the oligonucleotides, raising the possibility to be tested as cellular anti-telomerase agents.
- Published
- 2005
- Full Text
- View/download PDF
42. [Cell death signalling: recent advances and therapeutic application].
- Author
-
Ségal-Bendirdjian E, Dudognon C, Mathieu J, Hillion J, and Besançon F
- Subjects
- Autophagy physiology, Caspases physiology, Endoplasmic Reticulum physiology, Enzyme Activation, NF-kappa B physiology, Neoplasms therapy, Nucleoside-Phosphate Kinase physiology, Proto-Oncogene Proteins c-bcl-2 physiology, Receptors, Cell Surface physiology, Receptors, Tumor Necrosis Factor physiology, Apoptosis physiology, Signal Transduction physiology
- Abstract
Apoptosis is presently one of the most obvious targets for cancer treatment as its frequent inactivation in tumors contributes to carcinogenesis as well as resistance to chemotherapy. As knowledge of the apoptotic pathways and their regulation increases, it becomes obvious that this regulation is more complex than previously expected. Furthermore, there is growing evidence that alternative signalling pathways for cell death have to be considered. Understanding all the molecular events that regulate cell death may provide new opportunities for pathway-based rational therapy and for drug development. This review will focus on the emerging knowledge about these pathways and how this knowledge may be translated into more effective treatments for cancer.
- Published
- 2005
43. Death receptor signaling regulatory function for telomerase: hTERT abolishes TRAIL-induced apoptosis, independently of telomere maintenance.
- Author
-
Dudognon C, Pendino F, Hillion J, Saumet A, Lanotte M, and Ségal-Bendirdjian E
- Subjects
- Apoptosis Regulatory Proteins, DNA-Binding Proteins, Green Fluorescent Proteins, Humans, Luminescent Proteins metabolism, Membrane Glycoproteins physiology, Receptors, Tumor Necrosis Factor metabolism, TNF-Related Apoptosis-Inducing Ligand, Tretinoin pharmacology, Tumor Necrosis Factor-alpha physiology, Apoptosis physiology, Membrane Glycoproteins antagonists & inhibitors, Receptors, Tumor Necrosis Factor physiology, Signal Transduction, Telomerase metabolism, Telomerase physiology, Telomere, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Human telomerase has been implicated in cell immortalization and cancer. Recent works suggest that telomerase confers additional function required for tumorigenesis that does not depend on its ability to maintain telomeres. This new action may influence tumor therapy outcomes by yet unraveled mechanisms. Here, we show that overexpression of the catalytic subunit of telomerase (hTERT) protects a maturation-resistant acute promyelocytic leukemia (APL) cell line from apoptosis induced by the tumor necrosis factor (TNF) or TNF-related apoptosis-inducing ligand (TRAIL) and not from apoptosis induced by chemotherapeutic drugs such as etoposide or cisplatin. Conversely, in these cells, TRAIL-induced cell death is magnified by all-trans retinoic acid (ATRA) treatment, independently of telomerase activity on telomeres. Of note, this response is subordinated neither to maturation nor to telomere shortening. This work underlines that retinoids and death receptor signaling cross-talks offer new perspectives for antitumor therapy.
- Published
- 2004
- Full Text
- View/download PDF
44. Retinoic acid receptor alpha and retinoid-X receptor-specific agonists synergistically target telomerase expression and induce tumor cell death.
- Author
-
Pendino F, Dudognon C, Delhommeau F, Sahraoui T, Flexor M, Bennaceur-Griscelli A, Lanotte M, and Ségal-Bendirdjian E
- Subjects
- Cell Division drug effects, DNA-Binding Proteins, Drug Synergism, Genetic Vectors, Humans, Kinetics, Leukemia, Promyelocytic, Acute, Plasmids, Receptors, Retinoic Acid genetics, Recombinant Fusion Proteins, Retinoic Acid Receptor alpha, Retinoid X Receptors, Reverse Transcriptase Polymerase Chain Reaction, Telomerase genetics, Telomerase metabolism, Telomere ultrastructure, Transcription Factors genetics, Transfection, Tumor Cells, Cultured, Cell Death drug effects, Receptors, Retinoic Acid agonists, Telomerase antagonists & inhibitors, Transcription Factors agonists, Tretinoin pharmacology
- Abstract
Retinoids modulate growth and differentiation of cancer cells through activation of gene transcription via the nuclear retinoic-acid receptors (RAR) and retinoid-X receptors (RXR). Their use in differentiation therapy of acute promyelocytic leukemia (APL) represents a model concept for reprogramming cancer cells. However, they also regulate antiproliferative genes whose functions do not mechanistically concur to this program. Recently, we have shown that, independently of maturation, a long-term all-trans retinoic acid (ATRA) treatment of the maturation-resistant APL cell line (NB4-LR1) represses telomerase (hTERT), leading to telomere shortening and death. Using retinoid-receptor-specific agonists, we demonstrate herein that cross-talk between RARalpha and RXR dual-liganded to their respective agonists resulted in strong synergistic downregulation of hTERT and subsequent cell death. Importantly, unlike ATRA, this synergy was obtained at very low agonist concentrations and occurred in other ATRA maturation-resistant APL cells. These findings provide the first demonstration that dual-liganded RXR and RARalpha signaling should allow efficient targeting of telomerase in differentiation-resistant tumor cells. Such a combination therapy might hold promise in clinic to avoid side effects of ATRA whose administration can indiscriminately activate all RARs. Given the tissue-specific expression of RARs, a tissue-selective therapy targeting telomerase in tumor cells by synthetic agonists can be envisioned.
- Published
- 2003
- Full Text
- View/download PDF
45. Apoptosome-independent pathway for apoptosis. Biochemical analysis of APAF-1 defects and biological outcomes.
- Author
-
Belmokhtar CA, Hillion J, Dudognon C, Fiorentino S, Flexor M, Lanotte M, and Ségal-Bendirdjian E
- Subjects
- Animals, Apoptotic Protease-Activating Factor 1, Blotting, Southern, Blotting, Western, Caspase 3, Caspase 9, Caspases metabolism, Cell Line, Cell Survival, Cell-Free System, Cytochrome c Group metabolism, Enzyme Activation, Exons, Flow Cytometry, Gene Deletion, Granzymes, Membrane Potentials, Mice, Mitochondria metabolism, Models, Genetic, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Serine Endopeptidases pharmacology, Signal Transduction, Staurosporine pharmacology, Time Factors, Tumor Cells, Cultured, Apoptosis, Proteins genetics, Proteins physiology
- Abstract
Induction and execution of apoptosis programs are generally believed to be mediated through a hierarchy of caspase activation. By using two cellular variants obtained from the L1210 cell line (L1210/S and L1210/0), we have shown previously that staurosporine induces apoptotic cell death through both caspase-dependent and caspase-independent pathways. Both pathways normally coexisted in L1210/S cells, whereas L1210/0 cells lacked the ability to activate caspases despite the confirmed presence of both procaspase-3 and -9. Here we show that this defect in caspase activation is not due to mechanisms such as an absence of cytochrome c release, the expression of non-functional caspases, or the presence of an endogenous inhibitor but results from the loss of apoptosis protease activator protein-1 (APAF-1) expression. This absence of APAF-1 protein results from multiple alterations at both genomic and transcriptional levels. However, although this lack of APAF-1 delays the apoptotic program, it does not hamper its execution. Importantly, in these cells, apoptosis develops not only in an APAF-1-independent way but also in the absence of caspase-3 and -9 activation. Altogether these findings provide evidence that apoptosis may occur through alternative signaling pathways independent of APAF-1 expression and totally dissociated from any caspase processing. Therefore, the L1210/0 variant sub-line provides a valuable tool for the elucidation of these pathways.
- Published
- 2003
- Full Text
- View/download PDF
46. [Current concepts on apoptotic signalling pathways: new targets for anticancer strategies].
- Author
-
Ségal-Bendirdjian E, Hillion J, and Belmokhtar CA
- Subjects
- Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis Regulatory Proteins, Apoptotic Protease-Activating Factor 1, Caspases metabolism, Enzyme Activation, Ligands, Membrane Glycoproteins physiology, Mitochondria drug effects, Mitochondria physiology, Neoplasms drug therapy, Proteins physiology, Proto-Oncogene Proteins c-bcl-2 physiology, Receptors, Tumor Necrosis Factor physiology, Signal Transduction drug effects, TNF-Related Apoptosis-Inducing Ligand, Tumor Necrosis Factor-alpha physiology, Apoptosis physiology, Signal Transduction physiology
- Abstract
Apoptosis is an essential physiological process that plays a critical role in development and cellular homeoastasis. This process is tightly regulated through multiple independent signalling pathways. Defects in apoptosis may contribute both to tumorigenesis and drug resistance. Understanding the molecular events that contribute to apoptosis enable a more rational approach to anticancer strategy development. These strategies will allow not only the development of new molecules targeting recently elucidated apoptotic signalling pathways, but also a better use of already kown drugs through new associations in so far as these target distinct signalling pathways.
- Published
- 2003
47. Orchestration of multiple arrays of signal cross-talk and combinatorial interactions for maturation and cell death: another vision of t(15;17) preleukemic blast and APL-cell maturation.
- Author
-
Benoit G, Roussel M, Pendino F, Ségal-Bendirdjian E, and Lanotte M
- Subjects
- Apoptosis, Bone Marrow Cells metabolism, Bone Marrow Cells pathology, Disease Progression, Hematopoiesis, Humans, Leukemia, Promyelocytic, Acute etiology, Leukemia, Promyelocytic, Acute pathology, Neoplasm Proteins metabolism, Oncogene Proteins, Fusion metabolism, Receptors, Retinoic Acid metabolism, Retinoid X Receptors, Stem Cells metabolism, Stem Cells pathology, Transcription Factors metabolism, Cell Differentiation, Cell Transformation, Neoplastic pathology, Leukemia, Promyelocytic, Acute physiopathology, Receptor Cross-Talk, Signal Transduction, Translocation, Genetic
- Abstract
Despite intensive molecular biology investigations over the past 10 years, and an important breakthrough on how PML-RARalpha, the fusion protein resulting from t(15;17), can alter RARalpha and PML functions, no definitive views on how leukemia is generated and by what mechanism(s) the normal phenotype is restored, are yet available. 'Resistances' to pharmacological levels of all-trans-retinoic acid (ATRA) have been observed in experimental in vivo and in vitro models. In this review, we emphasize the key role played by signal cross-talk for both normal and neoplastic hemopoiesis. After an overview of reported experimental data on APL-cell maturation and apoptosis, we apply our current knowledge on signaling pathways to underline those which might generate signal cross-talks. The design of biological models suitable to decipher the integration of signal cross-talks at the transcriptional level should be our first priority today, to generate some realistic therapeutic approaches After 'Ten Years of Molecular APL', we still know very little about how the disease develops and how effective medicines work.
- Published
- 2001
- Full Text
- View/download PDF
48. Staurosporine induces apoptosis through both caspase-dependent and caspase-independent mechanisms.
- Author
-
Belmokhtar CA, Hillion J, and Ségal-Bendirdjian E
- Subjects
- Amino Acid Chloromethyl Ketones pharmacology, Animals, Apoptosis physiology, Caspases biosynthesis, Caspases genetics, Cysteine Proteinase Inhibitors pharmacology, DNA Fingerprinting, DNA, Neoplasm analysis, Enzyme Activation drug effects, Enzyme Induction drug effects, Leukemia L1210 pathology, Mice, Mice, Inbred DBA, Models, Biological, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Tumor Cells, Cultured, Apoptosis drug effects, Caspases physiology, Staurosporine pharmacology
- Abstract
Sensitivity of tumor cells to anticancer therapy depends on the ability of the drug to induce apoptosis. However, multiple signaling pathways control this induction and thus determine this sensitivity. We report here that staurosporine, a well known inducer of apoptosis in a wide range of cell lines, displays distinct ability to trigger apoptosis in two different L1210 sublines (termed L1210/S and L1210/0). Staurosporine treatment resulted in an early cell death (within 3 h) in L1210/S cells, while in L1210/0 cells, death occurred only after 12 h. In both instances, death occurred by apoptosis. A broad spectrum caspase inhibitor, Z-VAD-fmk, blocked early apoptosis in L1210/S cells but did not confer any protection on late apoptosis in L1210/0 cells. Protection by Z-VAD-fmk observed in L1210/S cells was not lasting and unmasked a secondary process of cell death that also exhibited characteristics of apoptosis. Thus, staurosporine induces apoptotic cell death through at least two redundant parallel pathways. These two pathways normally coexist in L1210/S cells. However, the early cell death mechanism depending on caspase activation disguises the late caspase-independent apoptotic process. Staurosporine-induced apoptosis in L1210/0 cells develops only by the caspase-independent mechanism due to a general defect in caspase activation.
- Published
- 2001
- Full Text
- View/download PDF
49. Nuclear translocation of a leukocyte elastase Inhibitor/Elastase complex during staurosporine-induced apoptosis: role in the generation of nuclear L-DNase II activity.
- Author
-
Belmokhtar CA, Torriglia A, Counis MF, Courtois Y, Jacquemin-Sablon A, and Ségal-Bendirdjian E
- Subjects
- Ammonium Sulfate, Animals, Biological Transport, Chemical Precipitation, Cytoplasm enzymology, DNA Fragmentation, Endodeoxyribonucleases chemistry, Endodeoxyribonucleases isolation & purification, Enzyme Inhibitors pharmacology, Immunoblotting, Mice, Nuclear Proteins metabolism, Staurosporine pharmacology, Time Factors, Tumor Cells, Cultured, Apoptosis, Cell Nucleus metabolism, Endodeoxyribonucleases metabolism, Leukocyte Elastase metabolism, Serpins metabolism
- Abstract
Using L1210 murine leukemia cells, we have previously shown that in response to treatment with drugs having different targets, apoptotic cell death occurs through at least two different signaling pathways. Here, we present evidence that nuclear extracts from staurosporine-treated cells elicit DNase II activity that is not detected in nuclear extracts from cisplatin-treated cells. This activity correlates with the accumulation of two nuclear proteins (70 and 30 kDa) which are detected by an anti-L-DNase II antibody. Partial purification of this DNase II activity suggests that the 30-kDa protein could be the nuclease responsible for staurosporine-induced DNA fragmentation. The 70-kDa protein is also recognized by an anti-elastase antibody, suggesting that it carries residues belonging to both L-DNase II and elastase. Since previous findings showed that L-DNase II was generated from the leukocyte inhibitor of elastase, we propose that the 70-kDa protein results from an SDS-stable association between these two proteins and is translocated from the cytoplasm to the nucleus during staurosporine-induced apoptosis., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
50. Alteration in p53 pathway and defect in apoptosis contribute independently to cisplatin-resistance.
- Author
-
Ségal-Bendirdjian E, Mannone L, and Jacquemin-Sablon A
- Subjects
- Animals, Annexin A5, Cell Cycle drug effects, Cyclin B metabolism, Cyclin B1, Cyclin-Dependent Kinase Inhibitor p21, Cyclins metabolism, DNA Fragmentation drug effects, Endonucleases metabolism, Enzyme Inhibitors pharmacology, Etoposide pharmacology, Fluorescein-5-isothiocyanate, Gene Expression Regulation, Neoplastic, Mice, Nuclear Proteins metabolism, Staurosporine pharmacology, Tumor Cells, Cultured, Apoptosis drug effects, Cisplatin pharmacology, Drug Resistance genetics, Tumor Suppressor Protein p53 genetics
- Abstract
The accumulation of molecular genetic defects selected during the adaptation process in the development of cisplatin-resistance was studied using progressive cisplatin-resistant variants (L1210/DDP2, L1210/DDP5, L1210/DDP10) derived from a murine leukemia cell line (L1210/0). Of these cell lines, only the most resistant L1210/DDP10 was cross-resistant to etoposide and deficient in apoptosis induced by these two drugs, indicating that resistance to DNA-damaging agents correlates with a defect in apoptosis. This defect was tightly associated with the loss of a Ca2+/Mg2+-dependent nuclear endonuclease activity present in the less cisplatin-resistant cells. Evidence is presented that p53-dependent function (a) is lost not only in the apoptosis defective L1210/DDP10 cells, but also in the apoptosis susceptible L1210/DDP5 cells; (b) is unrelated to drug-induced cell cycle perturbations. These results suggest that deficiency in the p53 pathway and resistance to DNA-damaging agents due to a defect in apoptosis are independent events.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.