Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Oncotarget ; 7(35): 56699-56712, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27527859

ABSTRACT

Our aim was to decipher the role and clinical relevance of the YAP/TAZ transcriptional coactivators in the regulation of the proliferation/quiescence balance in human colon cancer cells (CCC) and survival after 5FU-based chemotherapy. The prognostic value of YAP/TAZ on tumor relapse and overall survival was assessed in a five-year follow-up study using specimens of liver metastases (n = 70) from colon cancer patients. In 5FU-chemoresistant HT29-5F31 and -chemosensitive HCT116 and RKO CCC, a reversible G0 quiescent state mediated by Cyclin E1 down-regulation was induced by 5FU in 5F31 cells and recapitulated in CCC by either YAP/TAZ or Cyclin E1 siRNAs or the YAP inhibitor Verteporfin. Conversely, the constitutive active YAPdc-S127A mutant restricted cellular quiescence in 5FU-treated 5F31 cells and sustained high Cyclin E1 levels through CREB Ser-133 phosphorylation and activation. In colon cancer patients, high YAP/TAZ level in residual liver metastases correlated with the proliferation marker Ki-67 (p < 0.0001), high level of the YAP target CTGF (p = 0.01), shorter disease-free and overall survival (p = 0.008 and 0.04, respectively). By multivariate analysis and Cox regression model, the YAP/TAZ level was an independent factor of overall (Hazard ratio [CI 95%] 2.06 (1.02-4.16) p = 0.045) and disease-free survival (Hazard ratio [CI 95%] 1.98 (1.01-3.86) p = 0.045). Thus, YAP/ TAZ pathways contribute to the proliferation/quiescence switch during 5FU treatment according to the concerted regulation of Cyclin E1 and CREB. These findings provide a rationale for therapeutic interventions targeting these transcriptional regulators in patients with residual chemoresistant liver metastases expressing high YAP/TAZ levels.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin E/metabolism , Drug Resistance, Neoplasm , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Recurrence, Local , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Aged , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Disease-Free Survival , Fluorouracil/chemistry , Follow-Up Studies , HCT116 Cells , HT29 Cells , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Middle Aged , Mutation , Neoplasm Metastasis , Porphyrins/pharmacology , Prognosis , Proportional Hazards Models , Trans-Activators , Transcription Factors/metabolism , Transcriptional Activation , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Verteporfin , YAP-Signaling Proteins
2.
Sci Rep ; 6: 30405, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27465291

ABSTRACT

Metastatic cancer relapses following the reactivation of dormant, disseminated tumour cells; however, the cells and factors involved in this reactivation are just beginning to be identified. Using an immunotherapy-based syngeneic model of melanoma dormancy and GFP-labelled dormant cell-derived cell lines, we determined that vaccination against melanoma prevented tumour growth but did not prevent tumour cell dissemination or eliminate all tumour cells. The persistent disseminated melanoma tumour cells were quiescent and asymptomatic for one year. The quiescence/activation of these cells in vitro and the dormancy of melanoma in vivo appeared to be regulated by glucocorticoid-induced leucine zipper (GILZ)-mediated immunosuppression. GILZ expression was low in dormant cell-derived cultures, and re-expression of GILZ inactivated FOXO3A and its downstream target, p21CIP1. The ability of dormancy-competent cells to re-enter the cell cycle increased after a second round of cellular dormancy in vivo in association with shortened tumour dormancy period and faster and more aggressive melanoma relapse. Our data indicate that future cancer treatments should be adjusted according to the stage of disease progression.


Subject(s)
Forkhead Box Protein O3/genetics , Melanoma/genetics , Neoplastic Stem Cells/metabolism , Resting Phase, Cell Cycle/genetics , Transcription Factors/genetics , Animals , Biomarkers, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Gene Expression , Genes, Reporter , Humans , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Melanoma, Experimental , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Prognosis , Signal Transduction
4.
Cell Commun Signal ; 12: 52, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25223735

ABSTRACT

BACKGROUND: It is well established that inflammation promotes cancer, including melanoma, although the exact mechanisms involved are less known. In this study, we tested the hypothesis that inflammatory factors affect the cancer stem cell (CSC) compartment responsible for tumor development and relapse. RESULTS: Using an inducible histone 2B-GFP fusion protein as a tracer of cell divisional history, we determined that tumor necrosis factor (TNF), which is a classical pro-inflammatory cytokine, enlarged the CSC pool of GFP-positive label-retaining cells (LRCs) in tumor-like melanospheres. Although these cells acquired melanoma stem cell markers, including ABCB5 and CD271, and self-renewal ability, they lost their capacity to differentiate, as evidenced by the diminished MelanA expression in melanosphere cells and the loss of pigmentation in a skin equivalent model of human melanoma. The undifferentiated cell phenotype could be reversed by LY294002, which is an inhibitor of the PI3K/AKT signaling pathway, and this reversal was accompanied by a significant reduction in CSC phenotypic markers and functional properties. Importantly, the changes induced by a transient exposure to TNF were long-lasting and observed for many generations after TNF withdrawal. CONCLUSIONS: We conclude that pro-inflammatory TNF targets the quiescent/slow-cycling melanoma SC compartment and promotes PI3K/AKT-driven expansion of melanoma SCs most likely by preventing their asymmetrical self-renewal. This TNF effect is maintained and transferred to descendants of LRC CSCs and is manifested in the absence of TNF, suggesting that a transient exposure to inflammatory factors imprints long-lasting molecular and/or cellular changes with functional consequences long after inflammatory signal suppression. Clinically, these results may translate into an inflammation-triggered accumulation of quiescent/slow-cycling CSCs and a post-inflammatory onset of an aggressive tumor.


Subject(s)
Melanoma/metabolism , Neoplastic Stem Cells/metabolism , Skin Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Cell Line, Tumor , Cells, Cultured , Female , Fibroblasts , Humans , Keratinocytes , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin/metabolism
5.
Mar Drugs ; 12(2): 779-98, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24473175

ABSTRACT

Lamellarin D (LamD) is a marine alkaloid with broad spectrum antitumor activities. Multiple intracellular targets of LamD, which affect cancer cell growth and induce apoptosis, have been identified. These include nuclear topoisomerase I, relevant kinases (such as cyclin-dependent kinase 2) and the mitochondrial electron transport chain. While we have previously demonstrated that LamD at micromolar range deploys strong cytotoxicity by inducing mitochondrial apoptosis, mechanisms of its cytostatic effect have not yet been characterized. Here, we demonstrated that induction of cellular senescence (depicted by cell cycle arrest in G2 associated with ß-galactosidase activity) is a common response to subtoxic concentrations of LamD. Cellular senescence is observed in a large panel of cancer cells following in vitro or in vivo exposure to LamD. The onset of cellular senescence is dependent on the presence of intact topoisomerase I since topoisomerase I-mutated cells are resistant to senescence induced by LamD. LamD-induced senescence occurs without important loss of telomere integrity. Instead, incubation with LamD results in the production of intracellular reactive oxygen species (ROS), which are critical for senescence as demonstrated by the inhibitory effect of antioxidants. In addition, cancer cells lacking mitochondrial DNA also exhibit cellular senescence upon LamD exposure indicating that LamD can trigger senescence, unlike apoptosis, in the absence of functional mitochondria. Overall, our results identify senescence-associated growth arrest as a powerful effect of LamD and add compelling evidence for the pharmacological interest of lamellarins as potential anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Coumarins/pharmacology , DNA Topoisomerases, Type I/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , DNA, Mitochondrial/metabolism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice, SCID , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Telomere/metabolism , Topoisomerase Inhibitors/pharmacology
6.
Stem Cells Dev ; 23(8): 839-51, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24266654

ABSTRACT

Dental pulp stem cells (DPSCs) remain quiescent until activated in response to severe dental pulp damage. Once activated, they exit quiescence and enter regenerative odontogenesis, producing reparative dentin. The factors and signaling molecules that control the quiescence/activation and commitment to differentiation of human DPSCs are not known. In this study, we determined that the inhibition of insulin-like growth factor 1 receptor (IGF-1R) and p38 mitogen-activated protein kinase (p38 MAPK) signaling commonly activates DPSCs and promotes their exit from the G0 phase of the cell cycle as well as from the pyronin Y(low) stem cell compartment. The inhibition of these two pathways, however, inversely determines DPSC fate. In contrast to p38 MAPK inhibitors, IGF-1R inhibitors enhance dental pulp cell sphere-forming capacity and reduce the cells' colony-forming capacity without inducing cell death. The inverse cellular changes initiated by IGF-1R and p38 MAPK inhibitors were accompanied by inverse changes in the levels of active signal transducer and activator of transcription 3 (STAT3) factor, inactive glycogen synthase kinase 3, and matrix extracellular phosphoglycoprotein, a marker of early odontoblast differentiation. Our data suggest that there is cross talk between the IGF-1R and p38 MAPK signaling pathways in DPSCs and that the signals provided by these pathways converge at STAT3 and inversely regulate its activity to maintain quiescence or to promote self-renewal and differentiation of the cells. We propose a working model that explains the possible interactions between IGF-1R and p38 MAPK at the molecular level and describes the cellular consequences of these interactions. This model may inspire further fundamental study and stimulate research on the clinical applications of DPSC in cellular therapy and tissue regeneration.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation , Dental Pulp/cytology , MAP Kinase Signaling System , Receptor, IGF Type 1/metabolism , STAT3 Transcription Factor/metabolism , Calcification, Physiologic , Cell Proliferation , Cells, Cultured , Humans , Imidazoles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Resting Phase, Cell Cycle , Young Adult , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Mol Pharmacol ; 85(2): 226-36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24225022

ABSTRACT

There is a therapeutic need for glucocorticoid receptor (GR) ligands that distinguish between the transrepression and transactivation activity of the GR, the later thought to be responsible for side effects. These ligands are known as "dissociated glucocorticoids" (dGCs). The first published dGCs, RU24782 (9α-fluoro-11ß-hydroxy-16α-methylpregna-21-thiomethyl-1,4-diene-3,20-dione) and RU24858 (9α-fluoro-11ß-hydroxy-16α-methylpregna-21-cyanide-1,4-diene-3,20-dione), do not have the 17α-hydroxyl group that characterizes dexamethasone (Dex; 9α-fluoro-11ß,17α,21-trihydroxy-16α-methylpregna-1,4-diene-3,20-dione), and they differ from one another by having C21-thiomethyl and C21-cyanide moieties, respectively. Our aim was therefore to establish the structural basis of their activity. Both RU24782 and RU24858 induced a transactivation activity highly dependent on the GR expression level but always lower than dexamethasone. They also display less ability than dexamethasone to trigger steroid receptor coactivator 1 (SRC-1) recruitment and histone H3 acetylation. Docking studies, validated by mutagenesis experiments, revealed that dGCs are not anchored by Gln642, in contrast to Dex, which is hydrogen bonded to this residue via its 17α-hydroxyl group. This contact is essential for SRC-1 recruitment and subsequent dexamethasone-induced GR transactivation, but not transrepression. The ability of dGCs to make contacts with Ile747, for both RU24858 and RU24782 and with Asn564 for RU24858 are not strong enough to maintain GR in a conformation able to efficiently recruit SRC-1, unless SRC-1 is overexpressed. Overall, our findings provide some structural guidelines for the synthesis of potential new dissociated glucocorticoids with a better therapeutic ratio.


Subject(s)
Glucocorticoids/pharmacology , Receptors, Glucocorticoid/genetics , Transcriptional Activation/drug effects , Active Transport, Cell Nucleus , Animals , Binding Sites , COS Cells , Cells, Cultured , Chlorocebus aethiops , Dexamethasone/pharmacology , Glucocorticoids/chemistry , Glucocorticoids/metabolism , Humans , Nuclear Receptor Coactivator 1/physiology , Promoter Regions, Genetic , Protein Conformation , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Transcription Factors/genetics
8.
Clin Cancer Res ; 20(4): 837-46, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24323901

ABSTRACT

PURPOSE: Metastasis and drug resistance are the major limitations in the survival and management of patients with cancer. This study aimed to identify the mechanisms underlying HT29 colon cancer cell chemoresistance acquired after sequential exposure to 5-fluorouracil (5FU), a classical anticancer drug for treatment of epithelial solid tumors. We examined its clinical relevance in a cohort of patients with colon cancer with liver metastases after 5FU-based neoadjuvant chemotherapy and surgery. RESULTS: We show that a clonal 5F31 cell population, resistant to 1 µmol/L 5FU, express a typical cancer stem cell-like phenotype and enter into a reversible quiescent G0 state upon reexposure to higher 5FU concentrations. These quiescent cells overexpressed the tyrosine kinase c-Yes that became activated and membrane-associated upon 5FU exposure. This enhanced signaling pathway induced the dissociation of the Yes/YAP (Yes-associated protein) molecular complex and depleted nuclear YAP levels. Consistently, YES1 silencing decreased nuclear YAP accumulation and induced cellular quiescence in 5F31 cells cultured in 5FU-free medium. Importantly, YES1 and YAP transcript levels were higher in liver metastases of patients with colon cancer after 5FU-based neoadjuvant chemotherapy. Moreover, the YES1 and YAP transcript levels positively correlated with colon cancer relapse and shorter patient survival (P < 0.05 and P < 0.025, respectively). CONCLUSIONS: We identified c-Yes and YAP as potential molecular targets to eradicate quiescent cancer cells and dormant micrometastases during 5FU chemotherapy and resistance and as predictive survival markers for colon cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/metabolism , Fluorouracil/pharmacology , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Transcription Factors/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation , Checkpoint Kinase 2/metabolism , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm , Fluorouracil/therapeutic use , Gene Expression , HT29 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Neoadjuvant Therapy , Neoplasm Micrometastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Proportional Hazards Models , Protein Transport , Proto-Oncogene Proteins c-yes/genetics , Transcription Factors/genetics
9.
Oncotarget ; 4(11): 1986-98, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161908

ABSTRACT

Vemurafenib/PLX4032, a selective inhibitor of mutant BRAFV600E, constitutes a paradigm shift in melanoma therapy. Unfortunately, acquired resistance, which unavoidably occurs, represents one major limitation to clinical responses. Recent studies have highlighted that vemurafenib activated oxidative metabolism in BRAFV600E melanomas expressing PGC1α. However, the oxidative state of melanoma resistant to BRAF inhibitors is unknown. We established representative in vitro and in vivo models of human melanoma resistant to vemurafenib including primary specimens derived from melanoma patients. Firstly, our study reveals that vemurafenib increased mitochondrial respiration and ROS production in BRAFV600E melanoma cell lines regardless the expression of PGC1α. Secondly, melanoma cells that have acquired resistance to vemurafenib displayed intrinsically high rates of mitochondrial respiration associated with elevated mitochondrial oxidative stress irrespective of the presence of vemurafenib. Thirdly, the elevated ROS level rendered vemurafenib-resistant melanoma cells prone to cell death induced by pro-oxidants including the clinical trial drug, elesclomol. Based on these observations, we propose that the mitochondrial oxidative signature of resistant melanoma constitutes a novel opportunity to overcome resistance to BRAF inhibition.


Subject(s)
Indoles/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Melanoma/enzymology , Melanoma/genetics , Mice , Mice, SCID , Mitochondria/genetics , Oxidative Phosphorylation , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Reactive Oxygen Species/metabolism , Vemurafenib , Xenograft Model Antitumor Assays
10.
Melanoma Res ; 23(5): 373-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23852164

ABSTRACT

Recently, the BRAF V600 inhibitor, vemurafenib, has revolutionized the therapeutic management of metastatic melanoma. However, adverse effects and the onset of resistance are frequently observed, limiting the efficacy of this treatment. Patient-derived tumor xenografts (PDTX) engrafted in immunocompromised mice have been proposed as valuable preclinical models that can predict clinical response to treatments. Here, we established a PDTX model of BRAF V600E melanoma useful for testing the efficacy of vemurafenib. First, we validated the stability of the model that was similar to the original tumor with respect to histology, immunohistochemistry, mutational status, and fluorine-18 fluorodeoxyglucose ([(18)F]FDG)-PET/computed tomography (CT). Next, the sensitivity of the xenografts to vemurafenib was determined by tumor growth inhibition and decreased in standardized uptake value on [(18)F]FDG-PET/CT. Finally, this result, using personalized PDTX, allowed successful rechallenge with vemurafenib in a patient who was administered a lower dose of vemurafenib because of the onset of adverse events. Overall, we found that PDTX provides 'real-time' results in an animal that phenocopies the biology and expected vemurafenib responses of the tumor in a patient with BRAF V600E melanoma. Thus, this 'coclinical' trial using PDTX can help guide vemurafenib treatment for metastatic melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Adult , Animals , Antineoplastic Agents/adverse effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Fluorodeoxyglucose F18 , Humans , Indoles/adverse effects , Melanoma/enzymology , Melanoma/genetics , Melanoma/secondary , Mice, SCID , Multimodal Imaging , Mutation , Positron-Emission Tomography , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Radiopharmaceuticals , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sulfonamides/adverse effects , Time Factors , Tomography, X-Ray Computed , Tumor Burden/drug effects , Vemurafenib , Xenograft Model Antitumor Assays
11.
Stem Cells ; 31(4): 641-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355370

ABSTRACT

Melanoma is one of the most aggressive and extremely resistant to conventional therapies neoplasms. Recently, cellular resistance was linked to the cancer stem cell phenotype, still controversial and not well-defined. In this study, we used a Rhodamine 123 (Rh123) exclusion assay to functionally identify stem-like cells in metastatic human melanomas and melanoma cell lines. We demonstrate that a small subset of Rh123-low-retention (Rh123(low)) cells is enriched for stem cell-like activities, including the ability to self-renew and produce nonstem Rh123(high) progeny and to form melanospheres, recapitulating the phenotypic profile of the parental tumor. Rh123(low) cells are relatively quiescent and chemoresistant. At the molecular level, we show that melanoma Rh123(low) cells overexpress HIF1α, pluripotency factor OCT4, and the ABCB5 marker of melanoma stem cells and downregulate the expression of Cyclin D1 and CDK4. Interestingly, a short treatment with LY294002, an inhibitor of the PI3K/AKT pathway, specifically reverts a subset of Rh123(high) cells to the Rh123(low) phenotype, whereas treatment with inhibitors of mammalian target of rapamycin, phosphatase and tensin homolog or mitogen-activated protein kinase signaling does not. This phenotypic switching was associated with reduced levels of the HIF1α transcript and an increase in the level of phosphorylated nuclear FOXO3a preferentially in Rh123(low) cells. Moreover, the Rh123(low) cells became less quiescent and displayed a significant increase in their melanosphere-forming ability. All the above indicates that the Rh123(low) melanoma stem cell pool is composed of cycling and quiescent cells and that the PI3K/AKT signaling while maintaining the quiescence of Rh123(low) G0 cells promotes the exit of cycling cells from the stem cell compartment.


Subject(s)
Melanoma/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rhodamine 123/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Chromones/pharmacology , Cyclin D1/genetics , Cyclin D1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Morpholines/pharmacology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured
12.
Cancer Res ; 72(19): 5035-47, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22865452

ABSTRACT

Cancer cells can undergo a metabolic reprogramming from oxidative phosphorylation to glycolysis that allows them to adapt to nutrient-poor microenvironments, thereby imposing a selection for aggressive variants. However, the mechanisms underlying this reprogramming are not fully understood. Using complementary approaches in validated cell lines and freshly obtained human specimens, we report here that mitochondrial respiration and oxidative phosphorylation are slowed in metastatic melanomas, even under normoxic conditions due to the persistence of a high nuclear expression of hypoxia-inducible factor-1α (HIF-1α). Pharmacologic or genetic blockades of the HIF-1α pathway decreased glycolysis and promoted mitochondrial respiration via specific reduction in the expression of pyruvate dehydrogenase kinase-3 (PDK3). Inhibiting PDK3 activity by dichloroacetate (DCA) or siRNA-mediated attenuation was sufficient to increase pyruvate dehydrogenase activity, oxidative phosphorylation, and mitochondrial reactive oxygen species generation. Notably, DCA potentiated the antitumor effects of elesclomol, a pro-oxidative drug currently in clinical development, both by limiting cell proliferation and promoting cell death. Interestingly, this combination was also effective against BRAF V600E-mutant melanoma cells that were resistant to the BRAF inhibitor vemurafenib. Cotreatment of melanomas with DCA and elesclomol in vivo achieved a more durable response than single agent alone. Our findings offer a preclinical validation of the HIF-1/PDK3 bioenergetic pathway as a new target for therapeutic intervention in metastatic melanoma, opening the door to innovative combinations that might eradicate this disease.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/metabolism , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Dichloroacetic Acid/administration & dosage , Dichloroacetic Acid/pharmacology , Female , HL-60 Cells , Humans , Hydrazines/administration & dosage , Hydrazines/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, SCID , Middle Aged , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Apoptosis ; 17(4): 364-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22127645

ABSTRACT

F14512, an epipodophyllotoxin derivative equipped with a spermine moiety, is selectively taken up by the polyamine transport system over-active in tumor cells. F14512 was identified as a selective anticancer agent with a broad spectrum of antitumor activities and is currently undergoing phase I clinical trial in onco-hematology. However, the mechanism by which F14512 exerts its selective effects on neoplastic cells remains poorly understood. In this study, using mainly P388 leukemia cells, we showed that activation of the DNA damage response by F14512 did not induce immediate apoptosis but resulted in an early growth arrest. F14512-induced G2 arrest was accompanied by the appearance of a senescence-like phenotype (characterized by an increased ß-galactosidase staining) with up-regulation of the cyclin-dependent kinase inhibitor p16, and cyclin D1. The early senescence-based cell cycle block was characterized by a marked increase of the level of the IAP protein survivin, but not cIAP2, in P388 cells as well as in three other leukemia and melanoma cell types. The Thr(34)-phosphorylated form of survivin was observed within 4 h after F14512 exposure. Inhibition of survivin by siRNA resulted in a switch from senescence-like growth arrest to apoptosis. Compared with the parental drug etoposide, F14512-induced DNA damage signaling pathway resulted in greater senescence like-growth arrest and delayed apoptosis. Collectively, our data show that senescence arrest and subsequent apoptosis are powerful mechanisms mediating the chemotherapeutic effects of F14512 and identify survivin as the molecular determinant responsible for a qualitative shift in cell fate from senescence to apoptosis upon treatment with F14512.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Neoplasms/metabolism , Podophyllotoxin/analogs & derivatives , Repressor Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/physiopathology , Podophyllotoxin/pharmacology , Repressor Proteins/genetics , Survivin
14.
PLoS One ; 6(7): e21924, 2011.
Article in English | MEDLINE | ID: mdl-21789194

ABSTRACT

Challenges today concern chronic myeloid leukemia (CML) patients resistant to imatinib. There is growing evidence that imatinib-resistant leukemic cells present abnormal glucose metabolism but the impact on mitochondria has been neglected. Our work aimed to better understand and exploit the metabolic alterations of imatinib-resistant leukemic cells. Imatinib-resistant cells presented high glycolysis as compared to sensitive cells. Consistently, expression of key glycolytic enzymes, at least partly mediated by HIF-1α, was modified in imatinib-resistant cells suggesting that imatinib-resistant cells uncouple glycolytic flux from pyruvate oxidation. Interestingly, mitochondria of imatinib-resistant cells exhibited accumulation of TCA cycle intermediates, increased NADH and low oxygen consumption. These mitochondrial alterations due to the partial failure of ETC were further confirmed in leukemic cells isolated from some imatinib-resistant CML patients. As a consequence, mitochondria generated more ROS than those of imatinib-sensitive cells. This, in turn, resulted in increased death of imatinib-resistant leukemic cells following in vitro or in vivo treatment with the pro-oxidants, PEITC and Trisenox, in a syngeneic mouse tumor model. Conversely, inhibition of glycolysis caused derepression of respiration leading to lower cellular ROS. In conclusion, these findings indicate that imatinib-resistant leukemic cells have an unexpected mitochondrial dysfunction that could be exploited for selective therapeutic intervention.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Leukemia/pathology , Leukemia/physiopathology , Mitochondria/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Arsenic Trioxide , Arsenicals/pharmacology , Benzamides , Cell Death/drug effects , Cell Line, Tumor , Cell Respiration/drug effects , Electron Transport/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Imatinib Mesylate , Isothiocyanates/pharmacology , Leukemia/metabolism , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Models, Biological , Oxidative Stress/drug effects , Oxides/pharmacology , Reactive Oxygen Species/metabolism
15.
J Steroid Biochem Mol Biol ; 126(3-5): 104-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21569845

ABSTRACT

The ability of inhibitors of transcription and translation to prevent glucocorticoid-induced apoptosis has been interpreted to indicate that the cell death machinery requires de novo protein synthesis. The transcriptional inhibitors actinomycin D (Act D) and DRB as well as the translational inhibitors CHX and puromycin inhibited early loss of mitochondrial membrane integrity in a dose-dependent manner. This effect was not observed with the transcriptional inhibitor α-amanitin suggesting they may have additional effects. Their role in the glucocorticoid receptor (GR) intracellular trafficking was therefore investigated. Here, we show that Act D and CHX reduced glucocorticoid binding, GR turnover and impaired GR nuclear translocation. We performed the same experiments in different thymocyte subpopulations of Balb/c mice. At the highest dose tested, actinomycin D and cycloheximide abolished glucocorticoid-induced cell death of CD4+CD8+ and CD4+CD8-. In all subsets, Act D, DRB, as well as CHX and puromycin prevented receptor nuclear translocation, indicating a general alteration of GR trafficking. Overall, our data support a direct effect of macromolecular inhibitors on GR activation and trafficking. Finally, direct alterations of the functional properties of the glucocorticoid receptor might be responsible for cell death prevention by actinomycin D, DRB, cycloheximide and puromycin.


Subject(s)
Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Receptors, Glucocorticoid/metabolism , Alpha-Amanitin/pharmacology , Animals , Cells, Cultured , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Glucocorticoids/metabolism , HeLa Cells , Humans , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/metabolism , Mice , Mice, Inbred BALB C , Protein Binding , Protein Transport/drug effects , Puromycin/pharmacology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism
16.
Exp Dermatol ; 19(10): 929-30, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20849536

ABSTRACT

Recently, human embryonic stem cells have been differentiated in vitro into functional epidermal keratinocytes. Here, we demonstrated that these cells can be generated also from non-embryonic, human umbilical cord blood (hUCB) cells that have the potential to differentiate into cells of non-hematopoietic lineage. Human UCB mono-nucleated cells were cultivated in monolayer and in three-dimensional skin equivalent cultures and assayed for the presence of phenotype-specific markers. Our results determined that after one month of culturing in serum containing medium, the hUCB cells produced morphologically homogeneous colonies of epithelial cells expressing keratinocyte-specific markers. They also formed stratified epidermis in organ cultures that contained sporadic CD1a-positive cells within the accurate strata. We concluded that hUCB cells have the capacity to differentiate into functional epidermal keratinocytes and may serve as a source of high-quality keratinocytes for clinical applications.


Subject(s)
Epidermal Cells , Fetal Blood/cytology , Keratinocytes/cytology , Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Humans , Organ Culture Techniques
17.
Apoptosis ; 15(7): 769-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20151196

ABSTRACT

Lamellarin D (Lam D), a marine alkaloid, exhibits a potent cytotoxicity against many different tumors. The pro-apoptotic function of Lam D has been attributed to its direct induction of mitochondrial permeability transition (MPT). This study was undertaken to explore the mechanisms through which Lam D promotes changes in mitochondrial function and as a result apoptosis. The use of eight Lam derivatives provides useful structure-apoptosis relationships. We demonstrate that Lam D and structural analogues induce apoptosis of cancer cells by acting directly on mitochondria inducing reduction of mitochondrial membrane potential, swelling and cytochrome c release. Cyclosporin A, a well-known inhibitor of MPT, completely prevents mitochondrial signs of apoptosis. The drug decreases calcium uptake by mitochondria but not by microsomes indicating that Lam D-dependent permeability is specific to mitochondrial membranes. In addition, upon Lam D exposure, a rapid decline of mitochondrial respiration and ATP synthesis occurs in isolated mitochondria as well as in intact cells. Evaluation of the site of action of Lam D on the electron-transport chain revealed that the activity of respiratory chain complex III is reduced by a half. To determine whether Lam D could induce MPT-dependent apoptosis by inhibiting mitochondrial respiration, we generated respiration-deficient cells (rho0) derived from human melanoma cells. In comparison to parental cells, rho0 cells are totally resistant to the induction of MPT-dependent apoptosis by Lam D. Our results indicate that functional mitochondria are required for Lam D-induced apoptosis. Inhibition of mitochondrial respiration is responsible for MPT-dependent apoptosis of cancer cells induced by Lam-D.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis , Coumarins/toxicity , Heterocyclic Compounds, 4 or More Rings/toxicity , Isoquinolines/toxicity , Mitochondria/drug effects , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Respiration/drug effects , Coumarins/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Isoquinolines/chemistry , Jurkat Cells , Mice , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Oxygen Consumption/drug effects , Rats
18.
Stem Cells Dev ; 19(2): 209-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19799519

ABSTRACT

Cancer cells are unequal in a tumor mass and in established cultures. This is attributable to cancer stem cells with the unique ability to self-renew and to generate differentiating progeny. This ability is controlled at the level of asymmetric division by mechanisms that are yet not well defined. We found that normal and cancer keratinocyte fate was linked to the asymmetric distribution of epidermal growth factor receptor (EGFR) during mitosis. Although essential for epithelial cell proliferation, differentiation, and survival, this receptor was not present on the surface of cells satisfying criteria for stem cells such as quiescence, competence to produce functionally distinct daughters, high proliferative and clonogenic potential, sphere formation ability, and expression of stem cell markers. In contrast, keratinocytes displaying EGFR acquired a more differentiated phenotype, suggesting that EGFR may be involved in a switch from stem to transient amplifying cell fate. This switch was associated with changes in the expression profile of cell cycle, survival, and mitochondria controlling proteins that varied between normal and cancer cells. In conclusion, it appears that an unequal distribution of EGFR at mitosis controls keratinocyte fate by balancing quiescence and cycling of EGFR(-) cells, clearly malfunctioning in cancer. We believe that our findings provide mechanistic insights into the development of resistance to anti-EGFR therapies.


Subject(s)
Carcinoma, Basal Cell/metabolism , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Keratinocytes/metabolism , Skin Neoplasms/metabolism , Blotting, Western , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Fluorescent Antibody Technique , Humans , Hyaluronan Receptors/metabolism , Keratinocytes/cytology , Skin Neoplasms/pathology , Tumor Cells, Cultured , fas Receptor/metabolism
19.
Mol Cancer Ther ; 8(12): 3307-17, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952118

ABSTRACT

Lamellarin D, a potent cytotoxic marine alkaloid, exerts its antitumor action through two complementary pathways: a nuclear route via topoisomerase I inhibition and a mitochondrial targeting. The present study was designed to investigate the contribution of these two pathways for apoptosis in cancer cells. Lamellarin D promoted nuclear apoptosis in leukemia cells without prominent cell cycle arrest. Signals transmitted by lamellarin D initiated apoptosis via the intrinsic apoptotic pathway. The drug induced conformational activation of Bax and decreased the expression levels of antiapoptotic proteins Bcl-2 and cIAP2 in association with activation of caspase-9 and caspase-3. Upon lamellarin D exposure, Fas and Fas-L expression was not modified in leukemia cells. Moreover, leukemia cells deficient in caspase-8 or Fas-associated protein with death domain underwent apoptosis through the typical mitochondrial apoptotic cascade, indicating that cell death induced by lamellarin D was independent of the extrinsic apoptotic pathway. Lamellarin D also exerted a topoisomerase I-mediated DNA damage response resulting in H2AX phosphorylation, and the upregulation of the DNA repair protein Rad51 and of p53, as well as the phosphorylation of p53 at serine 15. However, lamellarin D killed efficiently mutated p53 or p53 null cancer cells, and sensitivity to lamellarin D was abrogated neither by cycloheximide nor in enucleated cells. Lamellarin D-induced cytochrome c release occurs independently of nuclear factors in a cell-free system. These results suggest that lamellarin D exerts its cytotoxic effects primarily by inducing mitochondrial apoptosis independently of nuclear signaling. Thus, lamellarin D constitutes a new proapoptotic agent that may bypass certain forms of apoptosis resistance that occur in tumor cells.


Subject(s)
Apoptosis/drug effects , Coumarins/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Mitochondria/metabolism , Animals , Apoptosis/physiology , Baculoviral IAP Repeat-Containing 3 Protein , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , DNA Damage , DNA Topoisomerases, Type I/metabolism , Flow Cytometry , HCT116 Cells , Histones/metabolism , Humans , Immunoblotting , Inhibitor of Apoptosis Proteins/metabolism , Jurkat Cells , Microscopy, Fluorescence , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/physiopathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , bcl-2-Associated X Protein/metabolism
20.
J Cell Mol Med ; 13(7): 1321-35, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18466357

ABSTRACT

The death of dendritic cells (DCs) can potentially influence immune responses by affecting the duration of DC stimulation of lymphocytes. Here, we report that cultured mature monocyte-derived DCs manifest early mitochondrial damage (i.e. within 24 hrs), characterized by mitochondrial membrane potential (psi Delta m) disruption and mitochondrial release of pro-apoptotic factors, followed by reactive oxygen species (ROS) production and activation of caspases. Afterwards, DCs with mitochondrial alterations are condemned to undergo apoptosis and necrosis. Macroarray analysis results (validated by real time quantitative-PCR (QRT-PCR) and immunoblotting), showed up-regulation of the pro-apoptotic member of the Bcl-2 family, Bim, while expression of several anti-apoptotic molecules was down-regulated. Importantly, pre-apoptotic DCs (characterized by a low Delta psi m) showed a modified phenotype, with down-regulation of HLA-DR and of the co-stimulatory molecules CD80 and CD86. Moreover, sorted viable low psi Delta m DCs were unable to activate allogeneic T cells, indicating that pre-apoptotic DCs have already lost some of their immuno-stimulatory capabilities long before any detectable signs of death occur. Perturbations to mitochondrial respiration with rotenone identified the same modifications to DC immune functions. These data indicate a strong requirement for mitochondrial integrity for the immuno-stimulatory capacities of DC. Determining Delta psi m could be a useful parameter to select 'fully' functional DCs for anti-tumour vaccines.


Subject(s)
Apoptosis , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunization , Mitochondria/pathology , Monocytes/cytology , Monocytes/immunology , Apoptosis/drug effects , Apoptosis/genetics , Caspases/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Enzyme Activation/drug effects , Gene Expression Profiling , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Monocytes/drug effects , Phenotype , Rotenone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...