Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(6)2022 Mar 12.
Article in English | MEDLINE | ID: mdl-35326611

ABSTRACT

Radiotherapy (RT) is a key component of cancer treatment. Although improvements have been made over the years, radioresistance remains a challenge. For this reason, a better understanding of cell fates in response to RT could improve therapeutic options to enhance cell death and reduce adverse effects. Here, we showed that combining RT (photons and protons) to noncytotoxic concentration of PARP inhibitor, Olaparib, induced a cell line-dependent senescence-like phenotype. The senescent cells were characterized by morphological changes, an increase in p21 mRNA expression as well as an increase in senescence-associated ß-galactosidase activity. We demonstrated that these senescent cells could be specifically targeted by Navitoclax (ABT-263), a Bcl-2 family inhibitor. This senolytic drug led to significant cell death when combined with RT and Olaparib, while limited cytotoxicity was observed when used alone. These results demonstrate that a combination of RT with PARP inhibition and senolytics could be a promising therapeutic approach for cancer patients.

2.
Trends Cell Biol ; 31(5): 331-344, 2021 05.
Article in English | MEDLINE | ID: mdl-33676803

ABSTRACT

Cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), control the transition between different phases of the cell cycle. CDK/cyclin activity is regulated by CDK inhibitors (CKIs), currently comprising the CDK-interacting protein/kinase inhibitory protein (CIP/KIP) family and the inhibitor of kinase (INK) family. Recent studies have identified a third group of CKIs, called ribosomal protein-inhibiting CDKs (RPICs). RPICs were discovered in the context of cellular senescence, a stable cell cycle arrest with tumor-suppressing abilities. RPICs accumulate in the nonribosomal fraction of senescent cells due to a decrease in rRNA biogenesis. Accordingly, RPICs are often downregulated in human cancers together with other ribosomal proteins, the tumor-suppressor functions of which are still under study. In this review, we discuss unique therapies that have been developed to target CDK activity in the context of cancer treatment or senescence-associated pathologies, providing novel tools for precision medicine.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/chemistry , Humans , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology
3.
Int J Mol Sci ; 21(13)2020 Jul 04.
Article in English | MEDLINE | ID: mdl-32635552

ABSTRACT

Pancreatic cancer is a very aggressive cancer type associated with one of the poorest prognostics. Despite several clinical trials to combine different types of therapies, none of them resulted in significant improvements for patient survival. Pancreatic cancers demonstrate a very broad panel of resistance mechanisms due to their biological properties but also their ability to remodel the tumour microenvironment. Radiotherapy is one of the most widely used treatments against cancer but, up to now, its impact remains limited in the context of pancreatic cancer. The modern era of radiotherapy proposes new approaches with increasing conformation but also more efficient effects on tumours in the case of charged particles. In this review, we highlight the interest in using charged particles in the context of pancreatic cancer therapy and the impact of this alternative to counteract resistance mechanisms.


Subject(s)
Carbon/therapeutic use , Ions/therapeutic use , Pancreatic Neoplasms/radiotherapy , Animals , Drug Resistance, Neoplasm/radiation effects , Humans , Protons , Tumor Microenvironment/radiation effects
4.
Semin Cancer Biol ; 60: 96-106, 2020 02.
Article in English | MEDLINE | ID: mdl-31454669

ABSTRACT

The majority of cancer-associated deaths are related to secondary tumor formation. This multistep process involves the migration of cancer cells to anatomically distant organs. Metastasis formation relies on cancer cell dissemination and survival in the circulatory system, as well as adaptation to the new tissue notably through genetic and/or epigenetic alterations. A large number of proteins are clearly identified to play a role in the metastatic process but the structures and modes of action of these proteins are essentially unknown or poorly described. In this review, we detail the involvement of members of the transmembrane (TMEM) protein family in the formation of metastases or in the mechanisms leading to cancer cell dissemination such as migration and extra-cellular matrix remodelling. While the phenotype associated with TMEM over or down-expression is clear, the mechanisms by which these proteins allow cancer cell spreading remain, for most of them, unclear. In parallel, the 3D structures of these proteins are presented. Moreover, we proposed that TMEM proteins could be used as prognostic markers in different types of cancers and could represent potential targets for cancer treatment. A better understanding of this heterogeneous family of poorly characterized proteins thus opens perspectives for better cancer patient care.


Subject(s)
Multigene Family , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Animals , Biomarkers , Disease Progression , Disease Susceptibility , Humans , Immunomodulation , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Signal Transduction , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/chemistry
5.
Cell Rep ; 29(6): 1469-1481.e9, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693889

ABSTRACT

Constitutive nuclear factor κB (NF-κB) activation is a hallmark of colon tumor growth. Cyclin-dependent kinases (CDKs) are critical cell-cycle regulators, and inhibition of CDK activity has been used successfully as anticancer therapy. Here, we show that the NFE2L3 transcription factor functions as a key regulator in a pathway that links NF-κB signaling to the control of CDK1 activity, thereby driving colon cancer cell proliferation. We found that NFE2L3 expression is regulated by the RELA subunit of NF-κB and that NFE2L3 levels are elevated in patients with colon adenocarcinoma when compared with normal adjacent tissue. Silencing of NFE2L3 significantly decreases colon cancer cell proliferation in vitro and tumor growth in vivo. NFE2L3 knockdown results in increased levels of double homeobox factor 4 (DUX4), which functions as a direct inhibitor of CDK1. The discovered oncogenic pathway governing cell-cycle progression may open up unique avenues for precision cancer therapy.


Subject(s)
Adenocarcinoma/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , Colonic Neoplasms/metabolism , Homeodomain Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin Immunoprecipitation Sequencing , Colonic Neoplasms/genetics , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , Homeodomain Proteins/genetics , Humans , Mass Spectrometry , Mice , Mice, Nude , NF-kappa B/metabolism , RNA, Small Interfering , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transplantation, Heterologous
6.
Biochem Pharmacol ; 162: 224-236, 2019 04.
Article in English | MEDLINE | ID: mdl-30629941

ABSTRACT

Hypoxia is one of the most important biological phenomena that influences cancer agressiveness and chemotherapy resistance. Cancer cells display dysregulated pathways notably resulting from oncogene expression. Tumors also show modifications in extracellular pH, extracellular matrix remodeling, neo-angiogenesis, hypoxia compared to normal tissues. Classically, the conventional anticancer therapies are efficient in cancer cells in normoxic conditions but under hypoxia, chemoresistance may occur. The addition of compounds that potentiate their activity in low oxygen environment could be a strategy to counteract this resistance. To identify new compounds active in hypoxia, we screened one hundred molecules with different chemical structures from an internal chemolibrary. Their potential ability to increase the activity of taxol and etoposide independently of their mechanism of action has been assayed. After a first step of selection, based on biological/pharmacological properties and chemical structure analysis, we identified three potential hits. Two hits are closely related amides/ureas and the third is a thiosemicarbazone. The compounds present no activity in cancer and normal cells when used alone but demonstrate chemosensitizing activity under hypoxia. Finally, by analyzing cell death, the indole thiosemicarbazone was shown to be able to significantly potentiate apoptosis induced by taxol and etoposide in two models of cancer cell lines. This new compound could lead to the development of an original series of chemosensitizers active under hypoxia.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Drug Synergism , Thiosemicarbazones/administration & dosage , Antineoplastic Agents/toxicity , Apoptosis/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , DNA Fingerprinting/methods , Etoposide/administration & dosage , Etoposide/toxicity , Hep G2 Cells , Humans , Paclitaxel/administration & dosage , Paclitaxel/toxicity , Thiosemicarbazones/toxicity
7.
Aging Cell ; 18(2): e12889, 2019 04.
Article in English | MEDLINE | ID: mdl-30614183

ABSTRACT

Most cancers arise in old individuals, which also accumulate senescent cells. Cellular senescence can be experimentally induced by expression of oncogenes or telomere shortening during serial passage in culture. In vivo, precursor lesions of several cancer types accumulate senescent cells, which are thought to represent a barrier to malignant progression and a response to the aberrant activation of growth signaling pathways by oncogenes (oncogene toxicity). Here, we sought to define gene expression changes associated with cells that bypass senescence induced by oncogenic RAS. In the context of pancreatic ductal adenocarcinoma (PDAC), oncogenic KRAS induces benign pancreatic intraepithelial neoplasias (PanINs), which exhibit features of oncogene-induced senescence. We found that the bypass of senescence in PanINs leads to malignant PDAC cells characterized by gene signatures of epithelial-mesenchymal transition, stem cells, and mitochondria. Stem cell properties were similarly acquired in PanIN cells treated with LPS, and in primary fibroblasts and mammary epithelial cells that bypassed Ras-induced senescence after reduction of ERK signaling. Intriguingly, maintenance of cells that circumvented senescence and acquired stem cell properties was blocked by metformin, an inhibitor of complex I of the electron transport chain or depletion of STAT3, a protein required for mitochondrial functions and stemness. Thus, our studies link bypass of senescence in premalignant lesions to loss of differentiation, acquisition of stemness features, and increased reliance on mitochondrial functions.


Subject(s)
Cellular Senescence/drug effects , Metformin/pharmacology , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Stem Cells/cytology , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Aging Cell ; 17(6): e12835, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30216637

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a lethal premature aging that recapitulates many normal aging characteristics. This disorder is caused by mutation in the LMNA gene leading to the production of progerin which induces misshapen nuclei, cellular senescence, and aging. We previously showed that the phospholipase A2 receptor (PLA2R1) promotes senescence induced by replicative, oxidative, and oncogenic stress but its role during progerin-induced senescence and in progeria is currently unknown. Here, we show that knockdown of PLA2R1 prevented senescence induced by progerin expression in human fibroblasts and markedly delayed senescence of HGPS patient-derived fibroblasts. Whole-body knockout of Pla2r1 in a mouse model of progeria decreased some premature aging phenotypes, such as rib fracture and decreased bone content, together with decreased senescence marker. Progerin-expressing human fibroblasts exhibited a high frequency of misshapen nuclei and increased farnesyl diphosphate synthase (FDPS) expression compared to controls; knockdown of PLA2R1 reduced the frequency of misshapen nuclei and normalized FDPS expression. Pamidronate, a FDPS inhibitor, also reduced senescence and misshapen nuclei. Downstream of PLA2R1, we found that p53 mediated the progerin-induced increase in FDPS expression and in misshapen nuclei. These results suggest that PLA2R1 mediates key premature aging phenotypes through a p53/FDPS pathway and might be a new therapeutic target.


Subject(s)
Aging, Premature/metabolism , Aging, Premature/pathology , Receptors, Phospholipase A2/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Nucleus Shape , Cellular Senescence , Disease Models, Animal , Geranyltranstransferase/metabolism , Humans , Lamin Type A/metabolism , Mice, Inbred C57BL , Phenotype , Progeria/metabolism , Progeria/pathology , Tumor Suppressor Protein p53/metabolism
9.
Cell Death Dis ; 9(7): 728, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29950610

ABSTRACT

Tumor-associated macrophages (TAMs) represent potential targets for anticancer treatments as these cells play critical roles in tumor progression and frequently antagonize the response to treatments. TAMs are usually associated to an M2-like phenotype, characterized by anti-inflammatory and protumoral properties. This phenotype contrasts with the M1-like macrophages, which exhibits proinflammatory, phagocytic, and antitumoral functions. As macrophages hold a high plasticity, strategies to orchestrate the reprogramming of M2-like TAMs towards a M1 antitumor phenotype offer potential therapeutic benefits. One of the most used anticancer treatments is the conventional X-ray radiotherapy (RT), but this therapy failed to reprogram TAMs towards an M1 phenotype. While protontherapy is more and more used in clinic to circumvent the side effects of conventional RT, the effects of proton irradiation on macrophages have not been investigated yet. Here we showed that M1 macrophages (THP-1 cell line) were more resistant to proton irradiation than unpolarized (M0) and M2 macrophages, which correlated with differential DNA damage detection. Moreover, proton irradiation-induced macrophage reprogramming from M2 to a mixed M1/M2 phenotype. This reprogramming required the nuclear translocation of NFκB p65 subunit as the inhibition of IκBα phosphorylation completely reverted the macrophage re-education. Altogether, the results suggest that proton irradiation promotes NFκB-mediated macrophage polarization towards M1 and opens new perspectives for macrophage targeting with charged particle therapy.


Subject(s)
Cellular Reprogramming/radiation effects , Macrophages/metabolism , Macrophages/radiation effects , NF-kappa B/metabolism , Protons , Signal Transduction , Cell Nucleus/metabolism , Histones/metabolism , Humans , Protein Transport , Radiation Tolerance/radiation effects , THP-1 Cells , Transcription Factor RelA/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
10.
Nat Cell Biol ; 20(7): 789-799, 2018 07.
Article in English | MEDLINE | ID: mdl-29941930

ABSTRACT

Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.


Subject(s)
Cell Cycle Checkpoints , Cellular Senescence , Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Ribosomes/metabolism , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , HEK293 Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , PC-3 Cells , Phosphorylation , Protein Binding , RNA Precursors/biosynthesis , RNA Precursors/genetics , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , RNA-Binding Proteins , Retinoblastoma Protein/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Signal Transduction , Time Factors
11.
Oncotarget ; 7(22): 32100-12, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27050073

ABSTRACT

Solid tumors often display chemotherapy resistance. Pancreatic ductal adenocarcinoma (PDAC) is the archetype of resistant tumors as current chemotherapies are inefficient. The tumor stroma and extracellular matrix (ECM) are key contributors to PDAC aggressiveness and to limiting the efficacy of chemotherapy. Lysyl oxidase (LOX) family members mediate collagen cross-linking and thus promote ECM stiffening. Our data demonstrate increased LOX, LOXL1, and LOXL2 expression in PDAC, and that the level of fibrillar collagen, which is directly dependent of LOX family activity, is an independent predictive biomarker of adjuvant "Gemcitabine-based chemotherapy" benefit. Experimentally in mice, increased LOX family activity through LOXL2 promotes chemoresistance. This effect of LOX family activity seems to be due to decreased gemcitabine intra-tumoral diffusion. This observation might be explained by increased fibrillar collagen and decreased vessel size observed in tumors with increased LOX family activity. In conclusion, our data support that LOX family activity is both a novel target to improve chemotherapy as well as a novel biomarker to predict gemcitabine benefit in PDAC. Beyond the PDAC, it is possible that targeting LOX family activity might improve efficacy of chemotherapies against different kinds of solid tumors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Pancreatic Neoplasms/drug therapy , Protein-Lysine 6-Oxidase/metabolism , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Diffusion , Female , Fibrillar Collagens/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein-Lysine 6-Oxidase/genetics , Tissue Distribution , Up-Regulation , Xenograft Model Antitumor Assays , Gemcitabine
12.
Nat Commun ; 5: 3792, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24797322

ABSTRACT

Senescence is involved in various pathophysiological conditions. Besides loss of retinoblastoma and p53 pathways, little is known about other pathways involved in senescence. Here we identify two calcium channels; inositol 1,4,5-trisphosphate receptor, type 2 (ITPR2) (also known as inositol 1,4,5-triphosphate receptor 2 (IP3R2)) and mitochondrial calcium uniporter (MCU) as new senescence regulators in a loss-of-function genetic screen. We show that loss of ITPR2, known to mediate endoplasmic reticulum (ER) calcium release, as well as loss of MCU, necessary for mitochondrial calcium uptake, enable escape from oncogene-induced senescence (OIS). During OIS, ITPR2 triggers calcium release from the ER, followed by mitochondrial calcium accumulation through MCU channels. Mitochondrial calcium accumulation leads to a subsequent decrease in mitochondrial membrane potential, reactive oxygen species accumulation and senescence. This ER-mitochondria calcium transport is not restricted to OIS, but is also involved in replicative senescence. Our results show a functional role of calcium release by the ITPR2 channel and its subsequent accumulation in the mitochondria.


Subject(s)
Calcium/metabolism , Cellular Senescence , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism , Humans , Membrane Potential, Mitochondrial , Oncogenes , Oxidative Stress
13.
Oncotarget ; 5(4): 1004-13, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24657971

ABSTRACT

Loss of secreted phospholipase A2 receptor (PLA2R1) has recently been found to render human primary cells more resistant to senescence whereas increased PLA2R1 expression is able to induce cell cycle arrest, cancer cell death or blockage of cancer cell transformation in vitro, suggesting that PLA2R1 displays tumor suppressive activities. Here we report that PLA2R1 expression strongly decreases in samples of human renal cell carcinoma (RCC). Knockdown of PLA2R1 increases renal cancer cell tumorigenicity supporting a role of PLA2R1 loss to promote in vivo RCC growth. Most RCC result from Von Hippel-Lindau (VHL) tumor suppressor loss-of-function and subsequent gain-of-function of the oncogenic HIF-2alpha/c-MYC pathway. Here, by genetically manipulating VHL, HIF-2alpha and c-MYC, we demonstrate that loss of VHL, stabilization of HIF-2alpha and subsequent increased c-MYC activity, binding and transcriptional repression, through induction of PLA2R1 DNA methylation closed to PLA2R1 transcriptional start site, results in decreased PLA2R1 transcription. Our results describe for the first time an oncogenic pathway leading to PLA2R1 transcriptional repression and the importance of this repression for tumor growth.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Phospholipase A2/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Nude , Proto-Oncogene Proteins c-myc/genetics , Receptors, Phospholipase A2/genetics , Signal Transduction , Transfection
14.
Biomed Chromatogr ; 28(7): 1017-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24424787

ABSTRACT

Lysyl oxidase enzymes are reported to be involved in patho-physiological process such as tumorigenesis. ß-Aminopropionitrile (BAPN) is an irreversible inhibitor of lysyl oxidase activity, suggesting a potentially useful therapeutic of interest in oncology. This paper describes the first assay concerning the quantification of BAPN by mass spectrometry. A high-performance liquid chromatography tandem mass spectrometry (LC-MS/MS) assay was developed for the quantification of BAPN in plasma and tumor of mice. This method combines dansyl chloride (Dns) derivatization and extraction using a solid-phase extraction Oasis Max column. Deuterated BAPN was used as internal standard (IS). Separation was achieved using an C18 column HypersylGold, (ThermoElectron), 3.0 µm (100 × 2.1 mm i.d.). Gradient elution with water containing 0.1% acetic acid (A) and acetonitrile containing 0.1% acetic acid (B) was applied. Detection was performed with an electrospray ionization interface operating in negative ion mode. Selected reaction monitoring was used with ion transitions m/z 302 → 249 for BAPN-Dns and m/z 306 → 250 for the IS. The method was fully validated in plasma and was linear and sensitive in the range of 10-500 ng/mL. The lower limit of quantification in plasma was 2.5 ng/mL. This validated assay was successfully applied to a kinetic study of BAPN in mouse plasma and demonstrates that BAPN reaches the tumoral tissue.


Subject(s)
Aminopropionitrile/blood , Chromatography, Liquid/methods , Neoplasms, Experimental/chemistry , Tandem Mass Spectrometry/methods , Aminopropionitrile/analysis , Aminopropionitrile/chemistry , Animals , Drug Stability , Linear Models , Mice , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Reproducibility of Results , Sensitivity and Specificity
15.
Cancer Res ; 73(20): 6334-45, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24008317

ABSTRACT

Little is known about the physiological role of the phospholipase A2 receptor (PLA2R1). PLA2R1 has been described as regulating the replicative senescence, a telomerase-dependent proliferation arrest. The downstream PLA2R1 signaling and its role in cancer are currently unknown. Senescence induction in response to activated oncogenes is a failsafe program of tumor suppression that must be bypassed for tumorigenesis. We now present evidence that PLA2R1 functions in vitro as a tumor suppressor, the depletion of which is sufficient to escape oncogene-induced senescence (OIS), thereby facilitating oncogenic cell transformation. Furthermore, mice that are genetically deficient in PLA2R1 display increased sensitivity to RAS-induced tumorigenesis by facilitating OIS escape, highlighting its physiological role as a tumor suppressor. Unexpectedly, PLA2R1 activated JAK2 and its effector signaling, with PLA2R1-mediated inhibition of cell transformation largely reverted in JAK2-depleted cells. This finding was unexpected as the JAK2 pathway has been associated mainly with protumoral functions and several inhibitors are currently in clinical trials. Taken together, our findings uncover an unanticipated tumor suppressive role for PLA2R1 that is mediated by targeting downstream JAK2 effector signaling.


Subject(s)
Cell Transformation, Neoplastic/genetics , Janus Kinase 2/metabolism , Receptors, Phospholipase A2/metabolism , Skin Neoplasms/genetics , Animals , Cell Culture Techniques , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Cellular Senescence/physiology , Enzyme Activation , Humans , Immunohistochemistry , Janus Kinase 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Receptors, Phospholipase A2/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Transfection
16.
Free Radic Biol Med ; 65: 969-977, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994771

ABSTRACT

Little is known about the biological functions of the phospholipase A2 receptor (PLA2R1) except that it has the ability to bind a few secreted phospholipases A2 (sPLA2's). We have previously shown that PLA2R1 regulates senescence in normal human cells. In this study, we investigated the ability of PLA2R1 to control cancer cell growth. Analysis of expression in cancer cells indicates a marked PLA2R1 decrease in breast cancer cell lines compared to normal or nontransformed human mammary epithelial cells. Accordingly, PLA2R1 ectopic expression in PLA2R1-negative breast cancer cell lines led to apoptosis, whereas a prosenescence response was predominantly triggered in normal cells. PLA2R1 structure-function studies and the use of chemical inhibitors of sPLA2-related signaling pathways suggest that the effect of PLA2R1 is sPLA2-independent. Functional experiments demonstrate that PLA2R1 regulation of cell death is driven by a reactive oxygen species (ROS)-dependent mechanism. While screening for ROS-producing complexes involved in PLA2R1 biological responses, we identified a critical role for the mitochondrial electron transport chain in PLA2R1-induced ROS production and cell death. Taken together, this set of data provides evidence for an important role of PLA2R1 in controlling cancer cell death by influencing mitochondrial biology.


Subject(s)
Apoptosis , Mitochondria/metabolism , Receptors, Phospholipase A2/physiology , Cell Line, Tumor , Cell Proliferation , DNA Fragmentation , Electron Transport Chain Complex Proteins/metabolism , Gene Expression , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism
17.
Cancer Res ; 73(16): 5253-65, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23774215

ABSTRACT

Oncogene-induced senescence (OIS) constitutes a failsafe program that restricts tumor development. However, the mechanisms that link oncogenesis to senescence are not completely understood. We carried out a loss-of-function genetic screen that identified the potassium channel KCNA1 as a determinant of OIS escape that can license tumor growth. Oncogenic stress triggers an increase in KCNA1 expression and its relocation from the cytoplasm to the membrane. Mechanistically, this relocation is due to a loss of protein kinase A (PKA)-induced phosphorylation at residue S446 of KCNA1. Accordingly, sustaining PKA activity or expressing a KCNA1 phosphomimetic mutant maintained KCNA1 in the cytoplasm and caused escape from OIS. KCNA1 relocation to the membrane induced a change in membrane potential that invariably resulted in cellular senescence. Restoring KCNA1 expression in transformation-competent cells triggered variation in membrane potential and blocked RAS-induced transformation, and PKA activation suppressed both effects. Furthermore, KCNA1 expression was reduced in human cancers, and this decrease correlated with an increase in breast cancer aggressiveness. Taken together, our results identify a novel pathway that restricts oncogenesis through a potassium channel-dependent senescence pathway.


Subject(s)
Cell Transformation, Neoplastic/genetics , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Cellular Senescence/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Down-Regulation , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Membrane Potentials/genetics , Mice , NIH 3T3 Cells , Phosphorylation/genetics , Signal Transduction/genetics
18.
Toxicol Appl Pharmacol ; 254(1): 8-17, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21504755

ABSTRACT

The in vitro anticancer activity and toxicity of phyllostictine A, a novel oxazatricycloalkenone recently isolated from a plant-pathogenic fungus (Phyllosticta cirsii) was characterized in six normal and five cancer cell lines. Phyllostictine A displays in vitro growth-inhibitory activity both in normal and cancer cells without actual bioselectivity, while proliferating cells appear significantly more sensitive to phyllostictine A than non-proliferating ones. The main mechanism of action by which phyllostictine displays cytotoxic effects in cancer cells does not seem to relate to a direct activation of apoptosis. In the same manner, phyllostictine A seems not to bind or bond with DNA as part of its mechanism of action. In contrast, phyllostictine A strongly reacts with GSH, which is a bionucleophile. The experimental data from the present study are in favor of a bonding process between GSH and phyllostictine A to form a complex though Michael attack at C=C bond at the acrylamide-like system. Considering the data obtained, two new hemisynthesized phyllostictine A derivatives together with three other natural phyllostictines (B, C and D) were also tested in vitro in five cancer cell lines. Compared to phyllostictine A, the two derivatives displayed a higher, phyllostictines B and D a lower, and phyllostictine C an almost equal, growth-inhibitory activity, respectively. These results led us to propose preliminary conclusions in terms of the structure-activity relationship (SAR) analyses for the anticancer activity of phyllostictine A and its related compounds, at least in vitro.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Ascomycota/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Neoplasms/drug therapy , Alkylation/drug effects , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , DNA/metabolism , Glutathione/metabolism , Heterocyclic Compounds, 3-Ring/toxicity , Humans , Microscopy, Video , Structure-Activity Relationship
19.
J Med Chem ; 54(7): 2012-21, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21388138

ABSTRACT

After the initial discovery of antiproliferative and apoptosis-inducing properties of a camptothecin-inspired pentacycle based on a 1H-indeno[2',1':5,6]dihydropyrido[2,3-d]pyrimidine scaffold, a library of its analogues as well as their oxidized planar counterparts were prepared utilizing a practical multicomponent synthetic protocol. The synthesized compounds exhibited submicromolar to low micromolar antiproliferative potencies toward a panel of human cancer cell lines. Biochemical experiments are consistent with the dihydropyridine library members undergoing intracellular oxidation to the corresponding planar pyridines, which then inhibit topoisomerase II activity, leading to inhibition of proliferation and cell death. Because of facile synthetic preparation and promising antitopoisomerase activity, both the dihydropyridine and planar pyridine-based compounds represent a convenient starting point for anticancer drug discovery.


Subject(s)
Biological Products/chemistry , Biological Products/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , Uracil/chemistry , Uracil/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Products/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Uracil/chemical synthesis
20.
Neoplasia ; 12(9): 727-39, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20824049

ABSTRACT

Glioblastoma (GBM) is the most frequent malignant glioma. Treatment of GBM patients is multimodal with maximum surgical resection, followed by concurrent radiation and chemotherapy with the alkylating drug temozolomide (TMZ). The present study aims to identify genes implicated in the acquired resistance of two human GBM cells of astrocytic origin, T98G and U373, to TMZ. Resistance to TMZ was induced by culturing these cells in vitro for months with incremental TMZ concentrations up to 1 mM. Only partial resistance to TMZ has been achieved and was demonstrated in vivo in immunocompromised mice bearing orthotopic U373 and T98G xenografts. Our data show that long-term treatment of human astroglioma cells with TMZ induces increased expression of facilitative glucose transporter/solute carrier GLUT/SLC2A family members, mainly GLUT-3, and of the AKR1C family of proteins. The latter proteins are phase 1 drug-metabolizing enzymes involved in the maintenance of steroid homeostasis, prostaglandin metabolism, and metabolic activation of polycyclic aromatic hydrocarbons. GLUT-3 has been previously suggested to exert roles in GBM neovascularization processes, and TMZ was found to exert antiangiogenic effects in experimental gliomas. AKR1C1 was previously shown to be associated with oncogenic potential, with proproliferative effects similar to AKR1C3 in the latter case. Both AKR1C1 and AKR1C2 proteins are involved in cancer pro-proliferative cell chemoresistance. Selective targeting of GLUT-3 in GBM and/or AKR1C proteins (by means of jasmonates, for example) could thus delay the acquisition of resistance to TMZ of astroglioma cells in the context of prolonged treatment with this drug.


Subject(s)
Alcohol Oxidoreductases/genetics , Brain Neoplasms/genetics , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Glucose Transport Proteins, Facilitative/genetics , Alcohol Oxidoreductases/metabolism , Aldehyde Reductase , Aldo-Keto Reductases , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Dacarbazine/adverse effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose Transport Proteins, Facilitative/metabolism , HT29 Cells , Humans , Mice , Mice, Nude , Temozolomide , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...