Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cancer Med ; 8(11): 5173-5182, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31350815

ABSTRACT

PURPOSE: To assess the incidence of BCR-ABL kinase domain (KD) mutation detection and its prognostic significance in chronic phase chronic myeloid leukemia (CP-CML) patients treated with tyrosine kinase inhibitors (TKIs). PATIENTS AND METHODS: We analyzed characteristics and outcome of 253 CP-CML patients who had at least one mutation analysis performed using direct sequencing. Of them, 187 patients were early CP (ECP) and 66 were late CP late chronic phase (LCP) and 88% were treated with Imatinib as first-line TKI. RESULTS: Overall, 80 (32%) patients harbored BCR-ABL KD mutations. A BCR-ABL KD mutation was identified in 57% of patients, who progressed to accelerated or blastic phases (AP-BP), and 47%, 29%, 35%, 16% and 26% in patients in CP-CML at the time of mutation analysis who lost a complete hematologic response, failed to achieve or loss of a prior complete cytogenetic and major molecular response, respectively. Overall survival and cumulative incidence of CML-related death were significantly correlated with the disease phase whatever the absence or presence of a mutation was and for the latter the mutation subgroup (T315I vs P-loop vs non-T315I non-P-loop) (P<.001). Considering patients who were in CP at the time of mutation analysis, LCP mutated patients had a significantly worse outcome than ECP-mutated patients despite a lower incidence of T315I and P-loop mutations (P<.001). With a median follow-up from mutation analysis to last follow-up of 5 years, T315I and P-loop mutations were not associated with a worse outcome in ECP patients (P = .817). CONCLUSION: Our results suggest that early mutation detection together with accessibility to 2nd and 3rd generation TKIs have reversed the worst outcome associated with BCR-ABL KD mutations whatever the mutation subgroup in CP-CML patients.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Mutation , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Mutational Analysis , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Mutation Rate , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Survival Analysis , Young Adult
2.
Hemasphere ; 2(3): e41, 2018 Jun.
Article in English | MEDLINE | ID: mdl-31723769

ABSTRACT

Dasatinib is an ABL1 tyrosine kinase inhibitor (TKI) with a short in vivo plasmatic half-life but with good efficiency, which is not fully understood. We investigated the possibility that circulating erythrocytes store and then provide dasatinib to target cells. In vitro coincubation of dasatinib-treated cells with naïve leukemic cells followed by analysis of kinase inhibition, apoptosis induction, fluorescent molecule exchanges, and dasatinib dosage were performed. Cells incubated with clinically relevant concentrations of dasatinib for a short time retained, after a washout procedure, an intracellular pool of dasatinib which was transferable to naïve BCR-ABL1 expressing cells and induced their apoptosis. This was verified in total blood where the huge cellular volume of erythrocytes constituted a large reservoir of dasatinib able to induce apoptosis in naïve BCR-ABL1 cell lines and primitive chronic myeloid leukemia (CML) CD34+ cells. This dasatinib transfer necessitated a contact between donor and acceptor cells. A component exchange occurred during this contact, carrying dasatinib and other TKIs such as nilotinib or the fluorescent sunitinib. An active pool of dasatinib could be buried inside the circulating erythrocytes, out of reach of detoxifying mechanisms, but still available for target cells and thus extending the acute effect of the plasmatic pool of the drug.

3.
Leuk Res ; 61: 44-52, 2017 10.
Article in English | MEDLINE | ID: mdl-28888102

ABSTRACT

Tyrosine kinase inhibitors (TKI) constitute the frontline treatment for chronic myeloid leukemia patients. Dasatinib, a second-generation TKI, was developed to overcome TKI resistances. However, dasatinib resistances are also described but remain less characterized. To mimic in vivo acquired dasatinib resistance, the BCR-ABL1-positive cell line K562 was transiently treated with a pharmacological concentration of dasatinib, for a short time in the presence of stem cell factor. A dasatinib resistant counterpart (K562 RES) was developed. Investigation of resistance mechanisms using kinase substrate arrays revealed that FYN was overactivated in K562 RES. The FYN inhibitor KX2-391 cooperated with dasatinib to block K562 RES proliferation. Cell tracking experiments showed that activated FYN support cell proliferation independently of BCR-ABL1 in K562 RES cells. Moreover, the MEK-ERK pathway was found hyper-phosphorylated in K562 RES cells even in the presence of dasatinib. Actually, ERK1/2 activity supported viability in K562 RES only in the absence of BCR-ABL1 activity. Finally, BCR-ABL1 and MEK inhibitor combination was sufficient to induce cell death even in non-proliferating resistant cells. Considering the conditions used to generate this dasatinib resistant cell line, such a resistance mechanism could be found in dasatinib treated patients. Consequently, it is valuable to know that inhibition of the MEK-ERK1/2 axis can overcome this resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Dasatinib/pharmacology , Drug Resistance, Neoplasm/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Apoptosis/drug effects , Blotting, Western , Fusion Proteins, bcr-abl/genetics , Humans , In Situ Hybridization, Fluorescence , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Polymerase Chain Reaction , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/drug effects , Proto-Oncogene Proteins c-fyn/metabolism
4.
Oncotarget ; 7(1): 845-59, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26625317

ABSTRACT

In spite of intensive research to improve treatment of acute myeloid leukemia (AML) more than half of all patients continue to develop a refractory disease. Therefore there is need to improve AML treatment. The overexpression of the BCL-2 family anti-apoptotic members, like BCL-2 or BCL-xL has been largely reported in lymphoid tumors but also in AML and other tumors. To counteract the anti-apoptotic effect of BCL-2, BH3 mimetics have been developed to target cancer cells. An increase in activity of ERK1/2 mitogen activated protein (MAP) kinase has also been reported in AML and might be targeted by MEK1/2 inhibitors. Hence, in the current work, we investigated whether the association of a BH3 mimetic such ABT-263 and the MEK1/2 inhibitor pimasertib (MEKI), was efficient to target AML cells. A synergistic increasing of apoptosis was observed in AML cell lines and in primary cells without affecting normal bone marrow cells. Such cooperation was confirmed on tumor growth in a mouse xenograft model of AML. In addition we demonstrated that MEKI sensitized the cells to apoptosis through its ability to promote a G1 cell cycle arrest. So, this combination of a MAP Kinase pathway inhibitor and a BH3 mimetic could be a promising strategy to improve the treatment of AML.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Niacinamide/analogs & derivatives , Sulfonamides/pharmacology , Acute Disease , Aniline Compounds/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , G1 Phase Cell Cycle Checkpoints/drug effects , HL-60 Cells , Humans , Immunohistochemistry , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Niacinamide/administration & dosage , Niacinamide/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Sulfonamides/administration & dosage , Tumor Burden/drug effects , U937 Cells , Xenograft Model Antitumor Assays
5.
Blood ; 123(12): 1850-9, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24470590

ABSTRACT

Telomere erosion may be counteracted by telomerase. Here we explored telomere length (TL) and telomerase activity (TA) in primary cutaneous T-cell lymphoma (CTCL) by using quantitative polymerase chain reaction and interphase quantitative fluorescence in situ hybridization assays. Samples from patients with Sézary syndrome (SS), transformed mycosis fungoides (T-MF), and cutaneous anaplastic large cell lymphoma were studied in parallel with corresponding cell lines to evaluate the relevance of TL and TA as target candidates for diagnostic and therapeutic purposes. Compared with controls, short telomeres were observed in aggressive CTCL subtypes such as SS and T-MF and were restricted to neoplastic cells in SS. While no genomic alteration of the hTERT (human telomerase catalytic subunit) locus was observed in patients' tumor cells, TA was detected. To understand the role of telomerase in CTCL, we manipulated its expression in CTCL cell lines. Telomerase inhibition rapidly impeded in vitro cell proliferation and led to cell death, while telomerase overexpression stimulated in vitro proliferation and clonogenicity properties and favored tumor development in immunodeficient mice. Our data indicate that, besides maintenance of TL, telomerase exerts additional functions in CTCL. Therefore, targeting these functions might represent an attractive therapeutic strategy, especially in aggressive CTCL.


Subject(s)
Lymphoma, T-Cell, Cutaneous/enzymology , Skin Neoplasms/enzymology , Telomerase/physiology , Telomere Homeostasis/physiology , Animals , Case-Control Studies , Cell Death , Cell Line, Tumor , Cell Proliferation , Female , Heterografts , Humans , Lymphoma, Large-Cell, Anaplastic/enzymology , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Lymphoma, T-Cell, Cutaneous/pathology , Male , Mice , Mice, SCID , Mycosis Fungoides/enzymology , Mycosis Fungoides/genetics , Neoplasm Transplantation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Sezary Syndrome/enzymology , Sezary Syndrome/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Telomerase/antagonists & inhibitors , Telomerase/genetics , Telomere Homeostasis/genetics
6.
PLoS One ; 8(11): e78582, 2013.
Article in English | MEDLINE | ID: mdl-24223824

ABSTRACT

PURPOSE: BIM is essential for the response to tyrosine-kinase inhibitors (TKI) in chronic myeloid leukaemia (CML) patients. Recently, a deletion polymorphism in intron 2 of the BIM gene was demonstrated to confer an intrinsic TKI resistance in Asian patients. The present study aimed at identifying mutations in the BIM sequence that could lead to imatinib resistance independently of BCR-ABL mutations. EXPERIMENTAL DESIGN: BIM coding sequence analysis was performed in 72 imatinib-treated CML patients from a French population of our centre and in 29 healthy controls (reference population) as a case-control study. Real-time quantitative PCR (RT qPCR) was performed to assess Bim expression in our reference population. RESULTS: No mutation with amino-acid change was found in the BIM coding sequence. However, we observed a silent single nucleotide polymorphism (SNP) c465C>T (rs724710). A strong statistical link was found between the presence of the T allele and the high Sokal risk group (p = 0.0065). T allele frequency was higher in non responsive patients than in the reference population (p = 0.0049). Similarly, this T allele was associated with the mutation frequency on the tyrosine kinase domain of BCR-ABL (p<0.001) and the presence of the T allele significantly lengthened the time to achieve a major molecular response (MMR). Finally, the presence of the T allele was related to a decreased basal expression of the Bim mRNA in the circulating mononuclear cells of healthy controls. CONCLUSION: These results suggest that the analysis of the c465C>T SNP of BIM could be useful for predicting the outcome of imatinib-treated CML patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , Benzamides/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/genetics , Piperazines/therapeutic use , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics , Pyrimidines/therapeutic use , RNA, Messenger/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Biomarkers, Pharmacological/metabolism , Case-Control Studies , Child , Child, Preschool , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression , Gene Frequency , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Mutation Rate , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism
7.
Neuro Oncol ; 14(12): 1441-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104476

ABSTRACT

Putative cancer stem cells have been identified in glioblastoma (GBM), associated with resistance to conventional therapies. Overcoming this resistance is a major challenge to manage this deadly brain tumor. Epidermal growth factor receptor (EGFR) is commonly amplified, over-expressed, and/or mutated in GBM, making it a compelling target for therapy. This study investigates the behavior of 3 primary neurosphere (NS) cell lines and their adherent counterparts originated from human GBM resections, when treated with EGFR-tyrosine kinase inhibitor erlotinib, associated or not with cyclopamine, a hedgehog pathway inhibitor. Adherent cells cultured in the presence of serum expressed the glial fibrillary acidic protein, whereas NS-forming cells cultured in serum-free medium expressed CD133, nestin, and Oct-4, markers of neural stem and progenitor cells. For the 3 adherent cell lines, erlotinib has a moderate effect (50% inhibitory concentration [IC50], >10 µM). Conversely, erlotinib induced a strong cell growth inhibition (IC50, <1 µM) on NS-forming cells, related to the EGFR gene amplification and EGFR protein expression. A short exposure to erlotinib reduced nestin-positive cell proliferation, but NS-initiating activity and self-renewal were not altered. EGFR pathway seems essential for GBM progenitor cell proliferation but dispensable for cancer stem-like cell self-renewal. Inhibition of hedgehog pathway with cyclopamine was evaluated in association with erlotinib on NS growth. Although each drug separately had no effect on sphere initiation, their combination significantly decreased the sphere number (P < .001). Our findings show synergic efficiency for erlotinib-cyclopamine association and provide a suitable in vitro model to explore drug combinations on GBM cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/pathology , Cell Differentiation/drug effects , Glioblastoma/pathology , Neoplastic Stem Cells/drug effects , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Comparative Genomic Hybridization , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence , Neoplastic Stem Cells/pathology , Quinazolines/administration & dosage , Veratrum Alkaloids/administration & dosage
8.
Exp Hematol ; 40(5): 367-78.e2, 2012 May.
Article in English | MEDLINE | ID: mdl-22240609

ABSTRACT

Chronic myeloid leukemia (CML) tumorigenicity is driven by the oncogenic BCR-ABL tyrosine kinase. Specific tyrosine kinase inhibitors (TKI) have been designed and are now used for the treatment of CML. These TKI induce apoptosis in leukemic cells in a BIM-dependent mechanism. We hypothesized that an increase in BIM activity could sensitize CML cells to TKI. We blocked the anti-apoptotic proteins of the Bcl-2 family by using ABT-737, a Bcl-2 and Bcl-XL inhibitor. ABT-737 modified Bcl-2 protein interactions toward a pro-apoptotic phenotype. Its combination with TKI resulted in a strong synergism in CML cell lines. The association also induced a large decrease in X-linked inhibitor of apoptosis (XIAP), followed by caspase-3 activation. This XIAP decrease was due to post-translational events. The mitochondrial serine protease HtrA2/Omi was identified as being responsible for this off-target effect. Then, ABT-737 and TKI cooperate at several levels to induce apoptosis of CML cells lines, and the benefit of this association was also observed in CML hematopoietic progenitors. Interestingly, a lethal effect was also observed in the more immature CD34(+)CD38(-) TKI-insensitive population. Combination therapy might by an interesting strategy for the treatment of CML patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/drug effects , Nitrophenols/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Sulfonamides/pharmacology , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , Antigens, CD34/analysis , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , Benzamides , Cell Line, Tumor/drug effects , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Hematopoietic Stem Cells/enzymology , High-Temperature Requirement A Serine Peptidase 2 , Humans , Imatinib Mesylate , K562 Cells/drug effects , Membrane Proteins/physiology , Mitochondrial Proteins/physiology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/enzymology , Proto-Oncogene Proteins/physiology , Serine Endopeptidases/physiology , X-Linked Inhibitor of Apoptosis Protein/genetics
9.
Cancer Biol Ther ; 11(12): 1017-27, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21508666

ABSTRACT

Gliomas are the most common malignant primary brain tumors in adults. The median survival never exceeds 12 months, owing to inherent resistance to both radio and chemotherapies. Epidermal Growth Factor Receptor (EGFR) is amplified, overexpressed, and/or mutated in glioblastomas (GBM), making it a rational for therapy. Erlotinib, an EGFR kinase inhibitor is strongly associated with clinical response in several cancers. Inhibition of cell proliferation and induction of apoptosis by erlotinib were investigated in U87-MG and DBTRG-05MG, two human glioblastoma cell lines. The expression of several apoptosis-related proteins was investigated in these cell lines and in tumoral tissue from glioblastomas. Both cell lines expressed wild-type EGFR but were deficient for PTEN. Erlotinib induced a marked accumulation of the BIM protein, but the activation of caspase-3 machinery was missing, regardless of the decrease in XIAP. Moreover, in U87-MG, erlotinib promoted accumulation of αB-crystallin a small heat shock protein capable to impair caspase activation. DBTRG-05MG was found deficient for procaspase 3 and constitutively overexpressed αB-crystallin. Similarly, deficiencies in PTEN and procaspase 3 were constantly found in samples from glioblastoma samples, while αB-crystallin expression was inconsistent. In cell lines, high concentrations of erlotinib induced cell death through a caspase independent process and an autophagic process was evidenced in U87-MG. Inhibition of autophagy induced a marked increase in the death-inducing activity of erlotinib. These results confirm that glioblastoma cell lines exhibit several anti-apoptotic mechanisms, and underline that EGFR targeted therapy must be associated to other inhibitors to achieve an antitumoral effect.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Glioblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspases/metabolism , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Glioblastoma/ultrastructure , HeLa Cells , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects
11.
Anal Chem ; 81(23): 9590-8, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19873978

ABSTRACT

Recombinant fluorescent probes allow the detection of molecular events inside living cells. Many of them exploit the intracellular space to provide positional signals and, thus, require detection by single cell imaging. We describe here a novel strategy based on probes capable of encoding the spatial dimension of intracellular signals into "all-or-none" fluorescence intensity changes (differential anchorage probes, DAPs). The resulting signals can be acquired in single cells at high throughput by automated flow cytometry, (i) bypassing image acquisition and analysis, (ii) providing a direct quantitative readout, and (iii) allowing the exploration of large experimental series. We illustrate our purpose with DAPs for Bax and the effector caspases 3 and 7, which are keys players in apoptotic cell death, and show applications in basic research, high content multiplexed library screening, compound characterization, and drug profiling.


Subject(s)
Fluorescent Dyes/metabolism , Intracellular Space/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle , Cell Death , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation , Drug Discovery , Flow Cytometry , High-Throughput Screening Assays , Humans , Molecular Imaging , Molecular Sequence Data , Peptide Hydrolases/metabolism , Recombinant Proteins/chemistry , bcl-2-Associated X Protein/metabolism
12.
Cancer Res ; 68(23): 9809-16, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047160

ABSTRACT

Targeting the tyrosine kinase activity of Bcr-Abl is an attractive therapeutic strategy in chronic myeloid leukemia (CML) and in Bcr-Abl-positive acute lymphoblastic leukemia. Whereas imatinib, a selective inhibitor of Bcr-Abl tyrosine kinase, is now used in frontline therapy for CML, second-generation inhibitors of Bcr-Abl tyrosine kinase such as nilotinib or dasatinib have been developed for the treatment of imatinib-resistant or imatinib-intolerant disease. In the current study, we generated nilotinib-resistant cell lines and investigated their mechanism of resistance. Overexpression of BCR-ABL and multidrug resistance gene (MDR-1) were found among the investigated mechanisms. We showed that nilotinib is a substrate of the multidrug resistance gene product, P-glycoprotein, using verapamil or PSC833 to block binding. Up-regulated expression of p53/56 Lyn kinase, both at the mRNA and protein level, was found in one of the resistant cell lines and Lyn silencing by small interfering RNA restored sensitivity to nilotinib. Moreover, failure of nilotinib treatment was accompanied by an increase of Lyn mRNA expression in patients with resistant CML. Two Src kinase inhibitors (PP1 and PP2) partially removed resistance but did not significantly inhibit Bcr-Abl tyrosine kinase activity. In contrast, dasatinib, a dual Bcr-Abl and Src kinase inhibitor, inhibited the phosphorylation of both BCR-ABL and Lyn, and induced apoptosis of the Bcr-Abl cell line overexpressing p53/56 Lyn. Such mechanisms of resistance are close to those observed in imatinib-resistant cell lines and emphasize the critical role of Lyn in nilotinib resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Pyrimidines/pharmacology , src-Family Kinases/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents , Dasatinib , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Thiazoles/pharmacology , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
13.
Exp Hematol ; 36(12): 1648-59, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922616

ABSTRACT

OBJECTIVE: Triptolide has shown antitumor activity in a broad range of solid tumors and on leukemic cells in vitro. MATERIALS AND METHODS: The THP1 cell line and primary acute myeloid leukemia (AML) cells were cultured with triptolide alone or in association with AraC or idarubicin in increasing concentrations. Apoptosis was measured by flow cytometry using DiOC6(3) for the cell line and fluorescein isothiocyanateAnnexin-V and CD45 labeling for fresh blast cells. Protein expression was measured by Western blot. Cell cycle distribution of apoptotic cells was measured by flow cytometry. RESULTS: A synergistic effect was observed when triptolide was added to idarubicin or to AraC to induce apoptosis of THP-1 leukemic cells. The triptolide/AraC association was also investigated in vitro on primary blast cells from 25 AML patients. This combination induced significantly higher percentages of apoptosis vs treatment with each drug separately (p<0.005). The IkappaB and X-linked inhibitor of apoptosis protein contents, which were altered by triptolide in idarubicin-treated cells, were not modified in AraC-treated cells. The association of AraC with triptolide increased the number of cells blocked in the S phase and most underwent apoptosis. CONCLUSION: These results suggest that, by modifying the cell cycle kinetics, AraC sensitizes AML cells to apoptosis induced by low concentration triptolide. The in vitro proapoptotic effect of triptolide associated with the antiproliferative activity of AraC warrants further clinical investigation for treatment of AML patients, especially elderly patients for whom low-dose AraC treatment could be improved by the addition of triptolide.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cytarabine/pharmacology , Diterpenes/pharmacology , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Phenanthrenes/pharmacology , Annexin A5/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cytarabine/agonists , Cytarabine/therapeutic use , Diterpenes/agonists , Diterpenes/therapeutic use , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Epoxy Compounds/agonists , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Humans , I-kappa B Proteins/metabolism , Idarubicin/agonists , Idarubicin/therapeutic use , Leukemia, Myeloid, Acute/metabolism , Leukocyte Common Antigens/metabolism , Phenanthrenes/agonists , Phenanthrenes/therapeutic use , X-Linked Inhibitor of Apoptosis Protein/metabolism
14.
Exp Cell Res ; 314(20): 3701-11, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-18930044

ABSTRACT

In order to clarify the role of HMW FGF-2 in glioma development and angiogenesis, we over-expressed different human FGF-2 isoforms in C6 rat glioma cell line using a tetracycline-regulated expression system. Phenotypic modifications were analyzed in vitro and compared to untransfected cells or to cells over-expressing 18 kDa FGF-2 or all FGF-2 isoforms. In particular, we demonstrate that HMW FGF-2 has unique features in inhibiting glioma cell proliferation. HMW FGF-2 expressing cells showed a cell-cycle arrest at the G2M, demonstrating a role of HMW FGF-2 in controlling the entry in mitosis. Moreover, hydroxyurea was ineffective in blocking cells at the G1S boundary when HMW FGF-2 was expressed. We also show that the HMW FGF-2 isoforms inhibit 4E-BP1 phosphorylation at critical sites restoring the translation inhibitory activity of 4E-BP1. In vivo, inhibition of tumor growth was observed when cells expressed HMW FGF-2. This indicates that HMW FGF-2 inhibits tumor growth in glioma cells by acting on cell-cycle progression and protein translation.


Subject(s)
Cell Cycle/genetics , Cell Proliferation , Fibroblast Growth Factor 2/genetics , Glioma/genetics , Protein Biosynthesis/genetics , Animals , Cell Cycle/physiology , Cell Line, Tumor , Down-Regulation , Glioma/metabolism , Glioma/pathology , Humans , Mice , Mice, SCID , Molecular Weight , Protein Isoforms/genetics , Rats , Transfection , Tumor Burden/genetics , Xenograft Model Antitumor Assays
15.
Cancer Biol Ther ; 6(6): 912-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17538248

ABSTRACT

It is an important challenge to better understand the mechanisms of tyrosine kinase inhibitors-induced apoptosis in CML cells. Thus, we have investigated how this apoptosis can be modulated by extracellular factors. Apoptosis induced by imatinib and nilotinib was determined in BCR-ABL expressing cell lines and primary CML CD34+ cells. Both molecules induced apoptosis of BCR-ABL expressing cells. This apoptosis was inhibited by protein synthesis inhibition in both K562 and CML CD34+ cells. In K562, 80% inhibition of the BCR-ABL auto-phosphorylation by either imatinib or nilotinib induced a two fold increase in Bim-EL expression and induction of apoptosis in 48 h. Bim accumulation preceded apoptosis induction which was completely abolished by depletion in Bim using shRNA. However, the anti-proliferative effect of imatinib was preserved in Bim-depleted cells. When K562 cells were cultured in a cytokine containing medium, the pro-apoptotic effect of nilotinib was decreased by 68% and this was related to a decrease in Bim-EL dephosphorylation and accumulation. Similarly, the presence of a combination of cytokines inhibited 88% of NIL- and 39% of IMA-induced apoptosis in primary CML CD34+ cells. In conclusion, both nilotinib and imatinib induce apoptosis through Bim accumulation independently of cell cycle arrest. However, the pro-apoptotic effect of both molecules can be attenuated by the presence of cytokines and growth factors, particularly concerning nilotinib. Thus BCR-ABL inhibition restores the cytokine dependence but is not sufficient to induce apoptosis when other signaling pathways are activated.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cytokines/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Membrane Proteins/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , Animals , Antigens, CD34/biosynthesis , Bcl-2-Like Protein 11 , Benzamides , Cell Line, Tumor , Humans , Imatinib Mesylate , K562 Cells , Mice , Models, Biological
16.
J Biol Chem ; 282(22): 16413-22, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17400550

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a major transcription factor sensitive to oxygen levels, which responds to stress factors under both hypoxic and nonhypoxic conditions. UV irradiation being a common stressor of skin, we looked at the effect of UVB on HIF-1alpha expression in keratinocytes. We found that UVB induces a biphasic HIF-1alpha variation through reactive oxygen species (ROS) generation. Whereas rapid production of cytoplasmic ROS down-regulates HIF-1alpha expression, delayed mitochondrial ROS generation results in its up-regulation. Indeed, activation of p38 MAPK and JNK1 mediated by mitochondrial ROS were required for HIF-1alpha phosphorylation and accumulation after UVB irradiation. Our experiments also revealed a key role of HIF-1alpha in mediating UVB-induced apoptosis. We conclude that the broad impact of the HIF-1 transcription factor on gene expression could make it a key regulator of UV-responsive genes and photocarcinogenesis.


Subject(s)
Apoptosis/radiation effects , Cell Transformation, Neoplastic/radiation effects , Cytoplasm/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Keratinocytes/metabolism , Ultraviolet Rays , Up-Regulation/radiation effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cytoplasm/pathology , Humans , Keratinocytes/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitogen-Activated Protein Kinase 8/metabolism , Phosphorylation/radiation effects , Protein Processing, Post-Translational/radiation effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cancer Biol Ther ; 6(4): 603-11, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17374988

ABSTRACT

Proteasome inhibitors are a novel class of compounds that might increase sensitivity to chemotherapy for acute myeloid leukemia (AML). We quantified apoptosis in THP-1 cells incubated with idarubicin (IDA) alone or together with a low concentration of MG132 or bortezomib. The combination of both drugs yielded a percentage of apoptotic cells that was significantly higher than the additive effect of both drugs administered separately (p < 0.01). Isobologram analysis showed that both MG132 and bortezomib interacted synergistically with IDA to induce apoptosis of THP1 cells. Western blot analysis of Bax and Bim show an acumulation of these pro-apoptotic proteins in THP1 treated cells. This increase in Bim preceded the induction of apoptosis and participated in idarubicin-induced apoptosis. Proteasome inhibition also potentiated IDA-induced apoptosis in primary blast cells from 22 AML patients while no such effect was found on normal lymphocytes, PHA-stimulated lymphocytes, normal cord blood CD34+ cells or bone marrow normal myeloid cells. These data show that MG132 and bortezomib specifically sensitize leukemic cells to IDA through an increase in BIM and Bax pro-apoptotic Bcl-2 family proteins.


Subject(s)
Anthracyclines/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/metabolism , Proteasome Inhibitors , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Bcl-2-Like Protein 11 , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Female , Humans , Idarubicin/pharmacology , Leupeptins/pharmacology , Male , Middle Aged , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Tumor Cells, Cultured
18.
Stem Cells ; 24(1): 65-73, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16123391

ABSTRACT

Physiological bone marrow oxygen concentrations are everywhere lower than 4% and almost null in some areas. We compared the effects of 20%, 3%, and 0.1% O2 concentrations on cord blood CD34+ cell survival, cycle, and functionality in serum-free cultures for 72 hours with or without interleukin-3 (IL-3). As from 24 hours, IL-3 improved cell survival and proliferation in all conditions. After 72 hours, cells were 1.5 and 2.5 times more in quiescence (G0) at 3% and 0.1% O2, respectively, than at 20%; transforming growth factor-beta signaling seemed not to be involved. To explore cell cycle further, fresh CD34+ cells were stained with PKH26 and cultured for 72 hours, and then undivided and divided cells were sorted. At 0.1% O2, 46.5%+/-19.1% of divided cells returned to G0 compared with 7.9%+/-0.3% at 20%. Colony formation and nonobese diabetic/severe combined immunodeficient mice engraftment efficiency were similar after 3 days at 20% and 0.1% O2 concentrations but lower than at T0. In conclusion, a low O2 concentration, close to those found in bone marrow stem cell niches, induces the G0 return of CD34+ cells without impairing their functional capacity.


Subject(s)
Antigens, CD34/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Interleukin-3/pharmacology , Oxygen/metabolism , Resting Phase, Cell Cycle , Animals , Cell Cycle , Cell Survival , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oxygen/chemistry , Stem Cell Factor/pharmacology
19.
J Cell Biochem ; 97(1): 135-44, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16173076

ABSTRACT

CD34 is a cell surface glycoprotein expressed on hematopoietic stem and progenitor cells that disappears with their maturation. This gene is transcribed in two alternatively spliced mRNAs that encode full length and truncated form of CD34 cell surface antigen. Some publications suggested that CD34 full length plays a role in the maintenance of their self renewal capacity. An examination of CD34 regulation by a low O2 concentration that ensures a better maintenance of stem cells may provide important insights into the molecular control of hematopoiesis. Using human cord blood CD34+ cells, we first compared the effect of short term (24 h) culture in hypoxia (1% O2) and normoxia (20% O2) on the expression of full length and truncated form of cd34 transcripts and on the expression of the CD34 antigen. Hypoxia maintained a larger quantity of cd34 full length transcripts and a higher cd34 full length/cd34 truncated form ratio than normoxia. After 72 h of culture at 1% and 20% O2, sorted CD34low sub-population from 1% O2 primary culture still contained more cd34 full length mRNAs than those from 20% O2, maintained better CD34 antigen expression during secondary culture at 20% O2 and contained more undifferentiated cells. This work provides the first evidence of the regulation of the cd34 gene by hypoxia resulting in a delayed higher and longer antigen expression by cord blood cells. We suggest that this phenomenon is related to the better maintenance of primitive stem cells in hypoxia.


Subject(s)
Antigens, CD34/genetics , Gene Expression Regulation , Oxygen/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Antigens, CD34/metabolism , Base Sequence , Cell Differentiation , Cell Line , DNA Primers , Humans , Immunophenotyping , Polymerase Chain Reaction
20.
Kidney Int ; 68(5): 2068-78, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16221206

ABSTRACT

BACKGROUND: Platelets are potential contributors to glomerular injury via the release of chemotactic and/or mitogenic mediators upon activation or through direct CD154/CD40-dependent interaction with cell components of the glomerulus. We examined whether platelets could activate mesangial cells and the potential role of the platelet-associated CD154. METHODS: Thrombin-activated platelets from systemic lupus erythematosus (SLE) patients or from disease or healthy controls were grown with human mesangial cells in the presence or not of a neutralizing anti-CD154 antibody either in contact or in a noncontact setting, the platelets and mesangial cells being separated by a pore size semipermeable membrane. The induction of mesangial cell surface antigens was assayed by flow cytometry. The quantification of mesangial cell proliferation was performed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and the production of transforming growth factor-beta1 (TGF-beta1), platelet-derived growth factor (PDGF) and soluble CD40 were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: Activated platelets from patients with SLE could induce an up-regulation of the expression of CD40 on mesangial cells with a concomitant release of soluble CD40. This induction required a direct contact between platelets and mesangial cells and was dependent upon the platelet-associated CD154. Pathologic consequences of the up-regulation of CD40 were a CD40-dependent stimulation of the proliferation of mesangial cells and a CD40-dependent increased production of TGF-beta1 by these cells. CONCLUSION: Platelets from patients with SLE can activate mesangial cells through CD40/CD154 interactions, leading to an induction of proliferation of the mesangial cells and an enhanced production of TGF-beta1, a profibrotic cytokine.


Subject(s)
Blood Platelets/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mesangial Cells/metabolism , Adult , Aged , Blood Platelets/cytology , Cell Communication/physiology , Cells, Cultured , Coculture Techniques , Female , Humans , Male , Mesangial Cells/cytology , Middle Aged , Platelet Activation , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...