Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cells ; 11(23)2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36496976

ABSTRACT

Enhanced invasiveness is one of the defining biological traits of glioblastoma cells, which exhibit an infiltrative nature that severely hinders surgical resection. Among the molecular lesions responsible for GBM aggressiveness, aberrant receptor tyrosine kinase (RTK) signalling is well-characterised. Enhanced RTK signalling directly impacts a myriad of cellular pathways and downstream effectors, which include the Rho GTPase family, key regulators of actin cytoskeletal dynamics. Here, we have analysed the functional crosstalk between oncogenic signals emanating from RTKs and Rho GTPases and focused on the specific contribution of Rnd3 to the invasive phenotype of GBM in this context. We found that RTK inhibition with a panel of RTK inhibitors decreased cell motility and cell invasion and promoted dramatic actin cytoskeleton reorganisation through activation of the RhoA/Rho-associated protein kinase 1 (ROCK) axis. RTK inhibition also significantly decreased Rnd3 expression levels. Consistently, shRNA-mediated Rnd3 silencing revealed that Rnd3 depletion promoted substantial changes in the actin cytoskeleton and reduced cell motility and invasion capacity, recapitulating the effects observed upon RTK inhibition. Our results indicate that Rnd3 is a crucial mediator of RTK oncogenic signalling involved in actin cytoskeletal reorganisation, which contributes to determining the invasive phenotype of GBM cells.


Subject(s)
Actins , Glioblastoma , Humans , Actins/metabolism , Glioblastoma/metabolism , rho GTP-Binding Proteins/metabolism , Signal Transduction , Receptor Protein-Tyrosine Kinases/metabolism
3.
Cancers (Basel) ; 14(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35565200

ABSTRACT

BACKGROUND: Glioma stem cells (GSCs) have self-renewal and tumor-initiating capacities involved in drug resistance and immune evasion mechanisms in glioblastoma (GBM). METHODS: Core-GSCs (c-GSCs) were identified by selecting cells co-expressing high levels of embryonic stem cell (ESC) markers from a single-cell RNA-seq patient-derived GBM dataset (n = 28). Induced c-GSCs (ic-GSCs) were generated by reprogramming GBM-derived cells (GBM-DCs) using induced pluripotent stem cell (iPSC) technology. The characterization of ic-GSCs and GBM-DCs was conducted by immunostaining, transcriptomic, and DNA methylation (DNAm) analysis. RESULTS: We identified a GSC population (4.22% ± 0.59) exhibiting concurrent high expression of ESC markers and downregulation of immune-associated pathways, named c-GSCs. In vitro ic-GSCs presented high expression of ESC markers and downregulation of antigen presentation HLA proteins. Transcriptomic analysis revealed a strong agreement of enriched biological pathways between tumor c-GSCs and in vitro ic-GSCs (κ = 0.71). Integration of our epigenomic profiling with 833 functional ENCODE epigenetic maps identifies increased DNA methylation on HLA genes' regulatory regions associated with polycomb repressive marks in a stem-like phenotype. CONCLUSIONS: This study unravels glioblastoma immune-evasive mechanisms involving a c-GSC population. In addition, it provides a cellular model with paired gene expression, and DNA methylation maps to explore potential therapeutic complements for GBM immunotherapy.

4.
J Med Chem ; 63(3): 1199-1215, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31935092

ABSTRACT

In vitro viability assays against a representative panel of human cancer cell lines revealed that polyamines L1a and L5a displayed remarkable activity with IC50 values in the micromolar range. Preliminary research indicated that both compounds promoted G1 cell cycle arrest followed by cellular senescence and apoptosis. The induction of apoptotic cell death involved loss of mitochondrial outer membrane permeability and activation of caspases 3/7. Interestingly, L1a and L5a failed to activate cellular DNA damage response. The high intracellular zinc-chelating capacity of both compounds, deduced from the metal-specific Zinquin assay and ZnL2+ stability constant values in solution, strongly supports their cytotoxicity. These data along with quantum mechanical studies have enabled to establish a precise structure-activity relationship. Moreover, L1a and L5a showed appropriate drug-likeness by in silico methods. Based on these promising results, L1a and L5a should be considered a new class of zinc-chelating anticancer agents that deserves further development.


Subject(s)
Antineoplastic Agents/pharmacology , Chelating Agents/pharmacology , Polyamines/pharmacology , Zinc/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/pharmacokinetics , Drug Design , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Models, Chemical , Molecular Structure , Polyamines/chemical synthesis , Polyamines/pharmacokinetics , Quantum Theory , Structure-Activity Relationship , Zinc/chemistry
5.
Cell Oncol (Dordr) ; 42(4): 521-536, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30980364

ABSTRACT

PURPOSE: Although EGFR activation is a hallmark of glioblastoma (GBM), anti-EGFR therapy has so far not yielded the desired effects. Targeting PI3K/Akt has been proposed as a strategy to increase the cellular sensitivity to EGFR inhibitors. Here we evaluated the contribution of FOXO3a, a key Akt target, in the response of GBM cells to EGFR inhibition. METHODS: FOXO3a activation was assessed by immunofluorescence and gene reporter assays, and by evaluating target gene expression using Western blotting and qRT-PCR. Cellular effects were evaluated using cell viability and apoptosis assays, i.e., Annexin V/PI staining and caspase 3/7 activity measurements. Drug synergism was evaluated by performing isobolographic analyses. Gene silencing experiments were performed using stable shRNA transfections. RESULTS: We found that EGFR inhibition in GBM cells led to FOXO3a activation and to transcriptional modulation of its key targets, including repression of the oncogene FOXM1. In addition, we found that specific FOXO3a activation recapitulated the molecular effects of EGFR inhibition, and that the FOXO3a activator trifluoperazine, a FDA-approved antipsychotic agent, reduced GBM cell growth. Subsequent isobolographic analyses of combination experiments indicated that trifluoperazine and erlotinib cooperated synergistically and that their concomitant treatment induced a robust activation of FOXO3a, leading to apoptosis in GBM cells. Using gene silencing, we found that FOXO3a is essential for the response of GBM cells to EGFR inhibition. CONCLUSIONS: Our data indicate that FOXO3a activation is a crucial event in the response of GBM cells to EGFR inhibition, suggesting that FOXO3a may serve as an actionable therapeutic target that can be modulated using FDA-approved drugs.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , Forkhead Box Protein O3/metabolism , Glioblastoma/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Transcriptional Activation/genetics , Trifluoperazine/pharmacology , Trifluoperazine/therapeutic use
6.
Biochem Pharmacol ; 115: 28-42, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27349984

ABSTRACT

A series of compounds containing the sulfonamide scaffold were synthesized and screened for their in vitro anticancer activity against a representative panel of human cancer cell lines, leading to the identification of N-(2-methyl-1H-indol-5-yl)-1-naphthalenesulfonamide (8e) as a compound showing a remarkable activity across the panel, with IC50 values in the nanomolar-to-low micromolar range. Cell cycle distribution analysis revealed that 8e promoted a severe G2/M arrest, which was followed by cellular senescence as indicated by the detection of senescence-associated ß-galactosidase (SA-ß-gal) in 8e-treated cells. Prolonged 8e treatment also led to the onset of apoptosis, in correlation with the detection of increased Caspase 3/7 activities. Despite increasing γ-H2A.X levels, a well-established readout for DNA double-strand breaks, in vitro DNA binding studies with 8e did not support interaction with DNA. In agreement with this, 8e failed to activate the cellular DNA damage checkpoint. Importantly, tubulin staining showed that 8e promoted a severe disorganization of microtubules and mitotic spindle formation was not detected in 8e-treated cells. Accordingly, 8e inhibited tubulin polymerization in vitro in a dose-dependent manner and was also able to robustly inhibit cancer cell motility. Docking analysis revealed a compatible interaction with the colchicine-binding site of tubulin. Remarkably, these cellular effects were reversible since disruption of treatment resulted in the reorganization of microtubules, cell cycle re-entry and loss of senescent markers. Collectively, our data suggest that this compound may be a promising new anticancer agent capable of both reducing cancer cell growth and motility.


Subject(s)
Antimitotic Agents/pharmacology , Cell Movement/drug effects , Indoles/pharmacology , Sulfonamides/pharmacology , Antimitotic Agents/chemical synthesis , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Humans , Indoles/chemical synthesis , Jurkat Cells , MCF-7 Cells , Microtubules/drug effects , Molecular Structure , Sulfonamides/chemical synthesis , Tubulin/drug effects
7.
Br J Haematol ; 174(6): 899-910, 2016 09.
Article in English | MEDLINE | ID: mdl-27220900

ABSTRACT

Mantle Cell Lymphoma (MCL) is an aggressive lymphoma subtype that accounts for 6-8% of non-Hodgkin lymphomas. The disease is mostly incurable and characterized by a continuous pattern of relapse. Major changes have recently been implemented in the management of MCL, but continuous relapses still mark this disease as a challenge for clinicians. We previously reported the efficacy of GemOx-R (Gemcitabine, Oxaliplatin and Rituximab) in patients with refractory and relapsing MCL. We present results for a larger series with longer follow-up and including high-risk frontline patients, showing an overall response rate of 83%. The efficacy of each component of GemOx-R was evaluated in a panel of MCL cell lines. Also, patient-derived primary cells were used in ex vivo experiments. The results show that oxaliplatin has a profound effect on cellular viability and is the most effective drug within this regimen. We further present synergistic efficacy of oxaliplatin combined with cytarabine in MCL cells. Interestingly, this synergistic effect was not seen when cisplatin and cytarabine were combined, indicating that among the platinum-derived agents oxaliplatin may be the preferred approach. Taken together our findings suggest that oxaliplatin alone or combined with cytarabine could constitute an alternative backbone for MCL regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Lymphoma, Mantle-Cell/drug therapy , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Lymphoma, Mantle-Cell/diagnosis , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Recurrence , Retreatment , Retrospective Studies , Rituximab/administration & dosage , Salvage Therapy , Treatment Outcome
8.
PLoS One ; 10(9): e0137800, 2015.
Article in English | MEDLINE | ID: mdl-26368127

ABSTRACT

Differential redox homeostasis in normal and malignant cells suggests that pro-oxidant-induced upregulation of cellular reactive oxygen species (ROS) should selectively target cancer cells without compromising the viability of untransformed cells. Consequently, a pro-oxidant deviation well-tolerated by nonmalignant cells might rapidly reach a cell-death threshold in malignant cells already at a high setpoint of constitutive oxidative stress. To test this hypothesis, we took advantage of a selected number of amine-pyridine-based Fe(II) complexes that operate as efficient and robust oxidation catalysts of organic substrates upon reaction with peroxides. Five of these Fe(II)-complexes and the corresponding aminopyridine ligands were selected to evaluate their anticancer properties. We found that the iron complexes failed to display any relevant activity, while the corresponding ligands exhibited significant antiproliferative activity. Among the ligands, none of which were hemolytic, compounds 1, 2 and 5 were cytotoxic in the low micromolar range against a panel of molecularly diverse human cancer cell lines. Importantly, the cytotoxic activity profile of some compounds remained unaltered in epithelial-to-mesenchymal (EMT)-induced stable populations of cancer stem-like cells, which acquired resistance to the well-known ROS inducer doxorubicin. Compounds 1, 2 and 5 inhibited the clonogenicity of cancer cells and induced apoptotic cell death accompanied by caspase 3/7 activation. Flow cytometry analyses indicated that ligands were strong inducers of oxidative stress, leading to a 7-fold increase in intracellular ROS levels. ROS induction was associated with their ability to bind intracellular iron and generate active coordination complexes inside of cells. In contrast, extracellular complexation of iron inhibited the activity of the ligands. Iron complexes showed a high proficiency to cleave DNA through oxidative-dependent mechanisms, suggesting a likely mechanism of cytotoxicity. In summary, we report that, upon chelation of intracellular iron, the pro-oxidant activity of amine-pyrimidine-based iron complexes efficiently kills cancer and cancer stem-like cells, thus providing functional evidence for an efficient family of redox-directed anti-cancer metallodrugs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Amines/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Humans , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oxidants/chemistry , Oxidants/pharmacology , Oxidative Stress/drug effects , Pyridines/chemistry , Reactive Oxygen Species/metabolism
9.
Bioconjug Chem ; 25(8): 1537-46, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25036647

ABSTRACT

Cell internalization is a major issue in drug design. Although squaramide-based compounds are receiving much attention because of their interesting bioactivity, cell uptake and trafficking within cells of this type of compounds are still unknown. In order to monitor the cell internalization process of cyclosquaramide compounds we have prepared two fluorescent probes by covalently linking a fluorescent dye (BODIPY derivative or fluorescein) to a noncytotoxic cyclosquaramide framework. These two probes (C2-BDP and C2-FITC) rapidly internalize across live cell membranes through endocytic receptor-mediated mechanisms. Due to its higher fluorescence and photochemical stability, C2-BDP is a superior dye than C2-FITC. C2-BDP remains sequestered in late endosomes allowing their fast and selective imaging in various live cell types. Cyclosquaramide-cell membrane interactions facilitate cell uptake and have been investigated by binding studies in solution as well as in live cells. Cyclosquaramide 1 (C2-BDP) can be used as a highly fluorescent probe for the rapid and selective imaging of late endosomes in live cells.


Subject(s)
Amides/chemistry , Cyclobutanes/chemistry , Cyclobutanes/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Animals , Biological Transport , Boron Compounds/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Models, Molecular , Molecular Conformation , Molecular Weight , NIH 3T3 Cells , Photochemical Processes
10.
BMC Complement Altern Med ; 14: 38, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24460687

ABSTRACT

BACKGROUND: Retama monosperma L. (Boiss.) or Genista monosperma L. (Lam.), locally named as "R'tam", is an annual and spontaneous plant belonging to the Fabaceae family. In Morocco, Retama genus is located in desert regions and across the Middle Atlas and it has been widely used in traditional medicine in many countries. In this study, we show that Retama monosperma hexane extract presents significant anti-leukemic effects against human Jurkat cells. METHODS: Human Jurkat cells, together with other cell lines were screened with different concentrations of Retama monosperma hexane extract at different time intervals. Growth inhibition was determined using luminescent-based viability assays. Cell cycle arrest and apoptosis were measured by flow cytometry analysis. Combined caspase 3 and 7 activities were measured using luminometric caspase assays and immunoblots were performed to analyze expression of relevant pro- and anti-apoptotic proteins. GC-MS were used to determine the chemical constituents of the active extract. RESULTS: Retama monosperma hexane extract (Rm-HE) showed significant cytotoxicity against Jurkat cells, whereas it proved to be essentially ineffective against both normal mouse fibroblasts (NIH3T3) and normal lymphocytes (TK-6). Cytometric analysis indicated that Rm-HE promoted cell cycle arrest and apoptosis induction accompanied by DNA damage induction indicated by an increase in p-H2A.X levels. Rm-HE induced apoptosis was partially JNK-dependent and characterized by an increase in Fas-L levels together with activation of caspases 8, 3, 7 and 9, whereas neither the pro-apoptotic nor anti-apoptotic mitochondrial membrane proteins analyzed were significantly altered. Chemical identification analysis indicated that α-linolenic acid, campesterol, stigmasterol and sitosterol were the major bioactive components within the extract. CONCLUSIONS: Our data suggest that bioactive compounds present in Rm-HE show significant anti leukemic activity inducing cell cycle arrest and cell death that operates, at least partially, through the extrinsic apoptosis pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Fabaceae/chemistry , Leukemia, T-Cell/drug therapy , Phytotherapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Cholesterol/therapeutic use , Fas Ligand Protein/metabolism , Humans , Jurkat Cells , Leukemia, T-Cell/metabolism , MAP Kinase Kinase 4/metabolism , Mice , NIH 3T3 Cells , Phytosterols/pharmacology , Phytosterols/therapeutic use , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Signal Transduction/drug effects , Sitosterols/pharmacology , Sitosterols/therapeutic use , Stigmasterol/pharmacology , Stigmasterol/therapeutic use , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use
11.
ChemMedChem ; 7(8): 1472-80, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22777958

ABSTRACT

We report the synthesis and biological evaluation of a new series of oligosquaramide-based macrocycles as anticancer agents. Compound 7, considered as representative of this series, exhibited significant antiproliferative activity against the NCI-60 human tumor cell line panel, with IC(50) values ranging from 1 to 10 µM. The results show that sensitivity to cyclosquaramides is clearly dependent on cell type, underscoring a degree of biological selectivity. The observed antiproliferative effects appear to be related to deregulation of protein phosphorylation, as compounds 7 and 8 are effective inhibitors of several important kinases such as ABL1, CDK4, CHK1, PKC, c-MET, and FGFR, among others. The corresponding acyclic oligosquaramides and smaller cyclosquaramides did not show antitumor activity, suggesting that a macrocyclic structure with minimal molecular size plays a key role in the observed antitumor activity.


Subject(s)
Amides/chemistry , Antineoplastic Agents/chemistry , Protein Kinase Inhibitors/chemistry , Amides/chemical synthesis , Amides/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Macrocyclic Compounds/chemistry , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , Protein Kinases/chemistry , Protein Kinases/metabolism
12.
PLoS One ; 7(6): e38770, 2012.
Article in English | MEDLINE | ID: mdl-22701710

ABSTRACT

Enforced EGFR activation upon gene amplification and/or mutation is a common hallmark of malignant glioma. Small molecule EGFR tyrosine kinase inhibitors, such as erlotinib (Tarceva), have shown some activity in a subset of glioma patients in recent trials, although the reported data on the cellular basis of glioma cell responsiveness to these compounds have been contradictory. Here we have used a panel of human glioma cell lines, including cells with amplified or mutant EGFR, to further characterize the cellular effects of EGFR inhibition with erlotinib. Dose-response and cellular growth assays indicate that erlotinib reduces cell proliferation in all tested cell lines without inducing cytotoxic effects. Flow cytometric analyses confirm that EGFR inhibition does not induce apoptosis in glioma cells, leading to cell cycle arrest in G(1). Interestingly, erlotinib also prevents spontaneous multicellular tumour spheroid growth in U87MG cells and cooperates with sub-optimal doses of temozolomide (TMZ) to reduce multicellular tumour spheroid growth. This cooperation appears to be schedule-dependent, since pre-treatment with erlotinib protects against TMZ-induced cytotoxicity whereas concomitant treatment results in a cooperative effect. Cell cycle arrest in erlotinib-treated cells is associated with an inhibition of ERK and Akt signaling, resulting in cyclin D1 downregulation, an increase in p27(kip1) levels and pRB hypophosphorylation. Interestingly, EGFR inhibition also perturbs Rho GTPase signaling and cellular morphology, leading to Rho/ROCK-dependent formation of actin stress fibres and the inhibition of glioma cell motility and invasion.


Subject(s)
Cell Movement/drug effects , Dacarbazine/analogs & derivatives , ErbB Receptors/pharmacology , Glioma/physiopathology , Signal Transduction/drug effects , rho GTP-Binding Proteins/metabolism , Cell Proliferation/drug effects , Colony-Forming Units Assay , Dacarbazine/pharmacology , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Glioma/metabolism , Humans , Immunoblotting , Signal Transduction/physiology , Temozolomide
13.
Br J Haematol ; 156(3): 334-45, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22107151

ABSTRACT

The FOXO3 (Forkhead/winged helix box class O 3) transcription factor is a crucial regulator of haematopoietic cell fate that controls proliferation and apoptosis, among other processes. Despite the central role of FOXO3 as a tumour suppressor and phosphatidylinositol 3-kinase (PI3K)/AKT effector, little is known about its involvement in mantle cell lymphoma (MCL) biology. This study investigated the expression and activity of FOXO3 in MCL cell lines and in primary cultures. We analysed the expression of key FOXO regulators and targets, and studied the effect of modulators of FOXO function on cell viability and apoptosis. FOXO3 was constitutively inactivated in MCL cell lines, and showed cytoplasmic localization in patient-derived cells. PI3K and AKT, but not mammalian target of rapamycin (mTOR), inhibitors induced FOXO3 nuclear translocation and activation in correlation with their impact on MCL proliferation and survival. Moreover, FOXO3-defective cells were resistant to PI3K/AKT inhibitors. Reactivation of FOXO function with a nuclear export inhibitor had a profound effect on cell viability, consistent with FOXO3 nuclear accumulation. Interestingly, inhibition of FOXO3 nuclear export enhanced the effect of doxorubicin. Taken together, our results confirm that FOXO3 is a relevant regulator of proliferation and apoptosis in MCL, and suggest that reactivation of FOXO3 function might be a useful therapeutic strategy in MCL patients.


Subject(s)
Forkhead Transcription Factors/physiology , Genes, Tumor Suppressor , Lymphoma, Mantle-Cell/pathology , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Doxorubicin/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Mantle-Cell/genetics , Neoplasm Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Transcription, Genetic , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
14.
Eur J Haematol ; 85(5): 371-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20727006

ABSTRACT

Mantle cell lymphoma has been considered an incurable disease with current chemotherapy regimens. Recent intense chemoimmunotherapy induction regimens with or without consolidation with autologous stem cell transplantation procedures are showing a potential for cure in a sizable fraction of patients. Similarly, in the salvage setting, preliminary experience with non-myeloablative allogeneic transplant may cure some patients even after multiple therapeutic failures. However, the recent knowledge of the three basic biologic derangements that are integrated in the disease may change the therapeutic approach of the disease in the near future. In fact, new drugs that target more specifically the major molecular alterations of the disease are being progressively incorporated into the therapeutic armamentarium of the disease. In the near future, more individualized approaches that will take into account not only risk factors present at diagnosis but also biomarkers representative of the molecular alterations present in the disease are foreseen. In this review, we are going to discuss the current therapeutic approaches and the main new drugs that target more specifically the major molecular pathways alterations of the disease.


Subject(s)
Drug Delivery Systems/methods , Lymphoma, Mantle-Cell/drug therapy , Biomarkers , Humans , Lymphoma, Mantle-Cell/therapy , Risk Factors
15.
Cell Physiol Biochem ; 24(5-6): 379-90, 2009.
Article in English | MEDLINE | ID: mdl-19910678

ABSTRACT

Uncoupling protein-2 (UCP2) and uncoupling protein-5 (UCP5) are ion carriers located in the inner mitochondrial membrane that mediate a regulated discharge of the proton gradient generated by the respiratory chain and are possibly involved in the protection against free radical production. Mitochondrial dysfunction is involved in the pathogenesis of colon cancer for which UCP2 has been proposed as a potential therapeutic target. The object of this study was to investigate the possible effects of mitochondrial dysfunction and oxidative stress on the mitochondrial content of UCP2 and UCP5 in colonic cells. Proliferation rate/index, antioxidant systems, reactive oxygen species, oxidative damage, mitochondrial markers and protein expression of UCP2 and UCP5 were analysed in non-metastatic HT-29 cells, metastatic SW-620 cells, and in a HT-29 differentiated derivative (Glu-R). In this study, we found a clear relationship between mitochondrial dysfunction in colonic epithelial cells and the levels of UCP2 and UCP5 proteins. Furthermore, hydrogen peroxide was responsible for the coordinated activation of the antioxidant response and the increase of UCP5 protein levels. Our findings suggest that uncoupling activity could act as a local feedback mechanism to limit the enhanced steady-state H(2)O(2) production in dysfunctional mitochondria of colonic cells.


Subject(s)
Colonic Neoplasms/metabolism , Hydrogen Peroxide/pharmacology , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Antioxidants/metabolism , Biomarkers/metabolism , Cell Line, Tumor , HT29 Cells , Humans , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Uncoupling Proteins , Oxidative Stress , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
16.
J Biol Chem ; 284(51): 35287-96, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19850923

ABSTRACT

The Rho GTPase family member RhoE inhibits RhoA/ROCK signaling to promote actin stress fiber and focal adhesion disassembly. We have previously reported that RhoE also inhibits cell cycle progression and Ras-induced transformation, specifically preventing cyclin D1 translation. Here we investigate the molecular mechanisms underlying those observations. RhoE inhibits the phosphorylation of the translational repressor 4E-BP1 in response to extracellular stimuli. However, RhoE does not affect the activation of mTOR, the major kinase regulating 4E-BP1 phosphorylation, as indicated by the phosphorylation levels of the mTOR substrate S6K, the dynamics of mTOR/Raptor association, and the observation that RhoE, as opposed to rapamycin, does not impair cellular growth. Interestingly, RhoE prevents the release of the eukaryotic initiation factor eIF4E from 4E-BP1, inhibiting cap-dependent translation. Accordingly, RhoE also inhibits the expression and the transcriptional activity of the eIF4E target c-Myc. Consistent with its crucial role in cell proliferation, we show that eIF4E can rescue both cell cycle progression and Ras-induced transformation in RhoE-expressing cells, indicating that the inhibition of eIF4E function is critical to mediate the anti-proliferative effects of RhoE.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , RNA Caps/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Carrier Proteins/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chlorocebus aethiops , Cyclin D1/genetics , Cyclin D1/metabolism , Enzyme Activation/physiology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factors , Focal Adhesions/genetics , Focal Adhesions/metabolism , Gene Expression Regulation/physiology , Humans , Mice , NIH 3T3 Cells , Phosphoproteins/genetics , Phosphorylation/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Caps/genetics , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/physiology , Stress Fibers/genetics , Stress Fibers/metabolism , TOR Serine-Threonine Kinases , ras Proteins/genetics , ras Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Blood Rev ; 23(5): 205-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19362399

ABSTRACT

Mantle cell lymphoma (MCL) is a well-defined lymphoid malignancy characterized by a rapid clinical evolution and poor response to current therapeutic protocols. The hallmark genetic alteration of MCL is the t(11;14)(q13;32) chromosomal translocation that leads to the overexpression of cyclin D1. Recently, new molecular alterations of major importance in the pathogenic mechanisms of this disease have been discovered, and have revealed the biological heterogeneity of MCL. The first section of our review discusses our current understanding of the molecular biology of this entity according to recent information from comparative genomic hybridization (CGH) and expression profiling studies, which are leading to the identification of several druggable targets. In the second section we revise new therapeutic strategies based on new drug families that target key molecular pathways of major relevance in this malignancy. We analyze emerging agents that are already producing significant results in different models of human cancers, including MCL. Based on the current knowledge and recent studies, we suggest that the encouraging results described here should provide a rationale platform for the design of new treatments that may overcome the resistance of this aggressive lymphoma to conventional therapy and improve patient prognosis.


Subject(s)
Apoptosis/physiology , Cyclin D1/metabolism , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , DNA Damage/physiology , Enzyme Inhibitors/therapeutic use , Gene Expression Profiling , Humans , Immunologic Factors/therapeutic use , Protein Kinase Inhibitors/therapeutic use
18.
Mol Cancer Ther ; 7(10): 3237-46, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18852127

ABSTRACT

Cisplatin is a conventional chemotherapeutic agent that binds covalently to purine DNA bases and mediates cellular apoptosis. A better understanding of the downstream cellular targets of cisplatin will provide information on its mechanism of action and help to understand the mechanism of drug resistance. In this study, we have investigated the effects of cisplatin in a panel of colon carcinoma cell lines and the involvement of the phosphoinositide-3-kinase/forkhead/winged helix box class O (FOXO) pathway in cisplatin action and resistance. Cisplatin-sensitive and cisplatin-resistant cell lines have been characterized in cell viability, flow cytometry, and clonogenic assays. The main components of the phosphoinositide-3-kinase/protein kinase B pathway, particularly FOXO3a, have been analyzed in sensitive and resistant cells on cisplatin treatment. Interestingly, in sensitive cells, cisplatin induces FOXO3a dephosphorylation and nuclear translocation, and expression of its target genes, whereas in resistant cells the effect of cisplatin on FOXO3a is incomplete. Consistent with this, protein kinase B/FOXO signaling axis modulators triciribine and psammaplysene A sensitize the resistant HT29 cells to cisplatin treatment. Critically, knockdown of FOXO3a expression using small interfering RNA rescues sensitive SW620 cells from cisplatin-induced short- and long-term cell death. Together, our findings suggest that FOXO3a is a relevant mediator of the cytotoxic effects of cisplatin in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/enzymology , Drug Screening Assays, Antitumor , Forkhead Box Protein O3 , Gene Silencing/drug effects , Humans , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Stem Cell Assay
19.
Mol Cell Biol ; 28(19): 5886-98, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18644865

ABSTRACT

The phosphoinositide-3 kinase (PI3K)/Akt signal pathway plays a key role in the tumorigenesis of many cancers and in the subsequent development of drug resistance. Using the K562 chronic myelogenous leukemia (CML) cell line and the doxorubicin-resistant derivative lines KD30 and KD225 as models, we observed that enhanced PI3K/Akt activity and the acquisition of chemoresistance correlated unexpectedly with the increased expression and nuclear accumulation of FOXO3a. Moreover, we found that the induction of FOXO3a activity in naïve K562 cells was sufficient to enhance PI3K/Akt activity and to confer resistance to the cytotoxic effects of doxorubicin. Conversely, the knockdown of endogenous FOXO3a expression reduced PI3K/Akt activity and sensitized these cells to doxorubicin. Further chromatin immunoprecipitation and promoter mutation analyses demonstrated that FOXO3a regulates the expression of the PI3K catalytic subunit p110alpha through the activation of a promoter region proximal to a novel untranslated exon upstream from the reported transcription start site of the p110alpha gene PIK3CA. As was the case for FOXO3a, the expression or knockdown of p110alpha was sufficient to amplify or reduce PI3K/Akt activity, respectively. Thus, our results suggest that the chronic activation of FOXO3a by doxorubicin in CML cells can enhance survival through a feedback mechanism that involves enhanced p110alpha expression and hyperactivation of the PI3K/Akt pathway.


Subject(s)
Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Exons , Forkhead Box Protein O3 , Humans , K562 Cells , Mice , Oncogene Protein v-akt/metabolism , Promoter Regions, Genetic , Signal Transduction
20.
Biochim Biophys Acta ; 1783(1): 144-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17900713

ABSTRACT

Mitogenic response to growth factors is concomitant with the modulation they exert on the levels of Fructose 2,6-bisphosphate (Fru-2,6-P2), an essential activator of the glycolytic flux. In mammalian cells, decreased Fru-2,6-P2 concentration causes cell cycle delay, whereas high levels of Fru-2,6-P2 sensitize cells to apoptosis. In order to analyze the cell cycle consequences due to changes in Fru-2,6-P2 levels, the bisphosphatase-dead mutant (H258A) of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase enzyme was over-expressed in Schizosaccharomyces pombe cells and the variation in cell phenotype was studied. The results obtained demonstrate that the increase in Fru-2,6-P2 levels results in a defective division of S. pombe, as revealed by an altered multisepted phenotype. The H258A-expressing cells showed impairment of cytokinesis, but normal nuclear division. In order to identify cellular mediators responsible for this effect, we transformed different S. pombe strains and observed that the cytokinetic defect was absent in cells defective for Wee1 kinase function. Therefore, in S. pombe, Wee1 integrates the metabolic signal emerging from changes in Fru-2,6-P2 content, thus coupling metabolism with cell proliferation. As the key regulators of the cell cycle checkpoints are conserved throughout evolution, these results may help to understand the experimental evidences obtained by manipulation of Fru-2,6-P2 levels in mammalian cells.


Subject(s)
Fructosediphosphates/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Cell Division , Gene Expression Regulation, Fungal , Mutation/genetics , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...