Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 761, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909140

ABSTRACT

Replicative senescence is triggered when telomeres reach critically short length and activate permanent DNA damage checkpoint-dependent cell cycle arrest. Mitochondrial dysfunction and increase in oxidative stress are both features of replicative senescence in mammalian cells. However, how reactive oxygen species levels are controlled during senescence is elusive. Here, we show that reactive oxygen species levels increase in the telomerase-negative cells of Saccharomyces cerevisiae during replicative senescence, and that this coincides with the activation of Hog1, a mammalian p38 MAPK ortholog. Hog1 counteracts increased ROS levels during replicative senescence. While Hog1 deletion accelerates replicative senescence, we found this could stem from a reduced cell viability prior to telomerase inactivation. ROS levels also increase upon telomerase inactivation when Mec1, the yeast ortholog of ATR, is mutated, suggesting that oxidative stress is not simply a consequence of DNA damage checkpoint activation in budding yeast. We speculate that oxidative stress is a conserved hallmark of telomerase-negative eukaryote cells, and that its sources and consequences can be dissected in S. cerevisiae.


Subject(s)
Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases , Oxidative Stress , Protein Serine-Threonine Kinases , Reactive Oxygen Species , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Telomerase , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Telomerase/metabolism , Telomerase/genetics , Reactive Oxygen Species/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , DNA Damage
2.
Genes (Basel) ; 14(3)2023 02 28.
Article in English | MEDLINE | ID: mdl-36980890

ABSTRACT

Telomeres are structures made of DNA, proteins and RNA found at the ends of eukaryotic linear chromosomes. These dynamic nucleoprotein structures protect chromosomal tips from end-to-end fusions, degradation, activation of damage checkpoints and erroneous DNA repair events. Telomeres were thought to be transcriptionally silent regions because of their constitutive heterochromatin signature until telomeric long non-coding RNAs (LncRNAs) were discovered. One of them, TERRA (TElomeric Repeat-containing RNA), starts in the subtelomeric regions towards the chromosome ends from different telomeres and has been extensively studied in many evolutionarily distant eukaryotes. Changes in TERRA's expression can lead to telomeric dysfunction, interfere with the replicative machinery and impact telomere length. TERRA also co-localizes in vivo with telomerase, and can form RNA:DNA hybrid structures called R-loops, which have been implicated in the onset of senescence and the alternative lengthening of telomere (ALT) pathway. Yet, the molecular mechanisms involving TERRA, as well as its function, remain elusive. Here, we review the current knowledge of TERRA transcription, structure, expression, regulation and its multiple telomeric and extra-telomeric functions in the budding yeast Saccharomyces cerevisiae.


Subject(s)
RNA, Long Noncoding , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , RNA, Long Noncoding/metabolism , Transcription, Genetic , Telomere/genetics , Telomere/metabolism , Heterochromatin
3.
Cell Death Differ ; 30(5): 1349-1365, 2023 05.
Article in English | MEDLINE | ID: mdl-36869180

ABSTRACT

Cells are inevitably challenged by low-level/endogenous stresses that do not arrest DNA replication. Here, in human primary cells, we discovered and characterized a noncanonical cellular response that is specific to nonblocking replication stress. Although this response generates reactive oxygen species (ROS), it induces a program that prevents the accumulation of premutagenic 8-oxoguanine in an adaptive way. Indeed, replication stress-induced ROS (RIR) activate FOXO1-controlled detoxification genes such as SEPP1, catalase, GPX1, and SOD2. Primary cells tightly control the production of RIR: They are excluded from the nucleus and are produced by the cellular NADPH oxidases DUOX1/DUOX2, whose expression is controlled by NF-κB, which is activated by PARP1 upon replication stress. In parallel, inflammatory cytokine gene expression is induced through the NF-κB-PARP1 axis upon nonblocking replication stress. Increasing replication stress intensity accumulates DNA double-strand breaks and triggers the suppression of RIR by p53 and ATM. These data underline the fine-tuning of the cellular response to stress that protects genome stability maintenance, showing that primary cells adapt their responses to replication stress severity.


Subject(s)
NADPH Oxidases , NF-kappa B , Humans , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Cytokines/genetics , Genomic Instability
4.
Sci Adv ; 7(35)2021 08.
Article in English | MEDLINE | ID: mdl-34452908

ABSTRACT

Double-strand breaks (DSBs) are harmful lesions and a major cause of genome instability. Studies have suggested a link between the nuclear envelope and the DNA damage response. Here, we show that lamin B1, a major component of the nuclear envelope, interacts directly with 53BP1 protein, which plays a pivotal role in the DSB repair. This interaction is dissociated after DNA damage. Lamin B1 overexpression impedes 53BP1 recruitment to DNA damage sites and leads to a persistence of DNA damage, a defect in nonhomologous end joining and an increased sensitivity to DSBs. The identification of interactions domains between lamin B1 and 53BP1 allows us to demonstrate that the defect of 53BP1 recruitment and the DSB persistence upon lamin B1 overexpression are due to sequestration of 53BP1 by lamin B1. This study highlights lamin B1 as a factor controlling the recruitment of 53BP1 to DNA damage sites upon injury.


Subject(s)
DNA Breaks, Double-Stranded , Lamin Type B , DNA Damage , DNA End-Joining Repair , Lamin Type B/genetics , Lamin Type B/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
5.
Biol Chem ; 402(10): 1257-1268, 2021 09 27.
Article in English | MEDLINE | ID: mdl-33977679

ABSTRACT

The protease activities are tightly regulated by inhibitors and dysregulation contribute to pathological processes such as cancer and inflammatory disorders. Tissue factor pathway inhibitor 2 (TFPI-2) is a serine proteases inhibitor, that mainly inhibits plasmin. This protease activated matrix metalloproteases (MMPs) and degraded extracellular matrix. Other serine proteases are implicated in these mechanisms like kallikreins (KLKs). In this study, we identified for the first time that TFPI-2 is a potent inhibitor of KLK5 and 12. Computer modeling showed that the first Kunitz domain of TFPI-2 could interact with residues of KLK12 near the catalytic triad. Furthermore, like plasmin, KLK12 was able to activate proMMP-1 and -3, with no effect on proMMP-9. Thus, the inhibition of KLK12 by TFPI-2 greatly reduced the cascade activation of these MMPs and the cleavage of cysteine-rich 61, a matrix signaling protein. Moreover, when TFPI-2 bound to extracellular matrix, its classical localisation, the KLK12 inhibition was retained. Finally, TFPI-2 was downregulated in human non-small-cell lung tumour tissue as compared with non-affected lung tissue. These data suggest that TFPI-2 is a potent inhibitor of KLK12 and could regulate matrix remodeling and cancer progression mediated by KLK12.


Subject(s)
Glycoproteins , Kallikreins , Carcinoma, Non-Small-Cell Lung , Humans , Lipoproteins , Lung Neoplasms
6.
Nucleic Acids Res ; 44(2): 648-56, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26446986

ABSTRACT

To rescue collapsed replication forks cells utilize homologous recombination (HR)-mediated mechanisms to avoid the induction of gross chromosomal abnormalities that would be generated by non-homologous end joining (NHEJ). Using DNA interstrand crosslinks as a replication barrier, we investigated how the Fanconi anemia (FA) pathway promotes HR at stalled replication forks. FA pathway inactivation results in Fanconi anemia, which is associated with a predisposition to cancer. FANCD2 monoubiquitination and assembly in subnuclear foci appear to be involved in TIP60 relocalization to the chromatin to acetylates histone H4K16 and prevents the binding of 53BP1 to its docking site, H4K20Me2. Thus, FA pathway loss-of-function results in accumulation of 53BP1, RIF1 and RAP80 at damaged chromatin, which impair DNA resection at stalled replication fork-associated DNA breaks and impede HR. Consequently, DNA repair in FA cells proceeds through the NHEJ pathway, which is likely responsible for the accumulation of chromosome abnormalities. We demonstrate that the inhibition of NHEJ or deacetylase activity rescue HR in FA cells.


Subject(s)
Carrier Proteins/metabolism , Chromatin/metabolism , Fanconi Anemia/metabolism , Histone Acetyltransferases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Acetylation , Carrier Proteins/genetics , Chromatin/drug effects , Chromatin/genetics , DNA End-Joining Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Histone Acetyltransferases/genetics , Histone Chaperones , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysine Acetyltransferase 5 , MRE11 Homologue Protein , Mitomycin/pharmacology , Nuclear Proteins/genetics , Tumor Suppressor p53-Binding Protein 1 , Ubiquitination
7.
Tumour Biol ; 36(7): 4979-86, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25677900

ABSTRACT

The KLK13 gene is dysregulated in several carcinomas, and its expression levels seem to be associated with disease prognosis. The aim of our study was to investigate the prognostic potential of KLK13 mRNA expression for patients with nonsmall cell lung cancer (NSCLC). Total RNA was isolated from cancerous and normal tissues from a cohort of 128 NSCLC patients. The KLK13 mRNA transcription levels were measured using a sensitive quantitative RT-PCR method. The results were normalized by dividing the KLK13 mRNA values with the geometric mean of mRNA expression from four reference genes: beta-actin, TATA-binding protein, hypoxanthine phosphoribosyltransferase 1, and acidic ribosomal phosphoprotein P0. The malignant tissues from the majority of patients (59.3 %) contained significantly more KLK13 mRNA transcripts than did the paired nonmalignant tissues (median difference 11.1-fold, P = 0.008). KLK13 was expressed at higher levels in females than that in males (P = 0.021). No other statistically significant association with clinicopathological data was observed. Kaplan-Meier survival analyses demonstrated that patients with KLK13-positive tumors survived significantly longer than those with KLK13-negative ones (P = 0.009). KLK13 expression was also shown to be able to stratify high-risk individuals among patients with early disease stages (P = 0.030). Multivariate Cox regression analysis showed that KLK13 expression is a favorable, independent prognostic indicator of overall survival (OS) (P = 0.024). Our results suggest that KLK13 mRNA expression constitutes a novel biomarker for the prediction of overall survival in NSCLC and that its quantitative assessment in tumor tissues can aid in treatment decision making.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Kallikreins/genetics , Prognosis , Adult , Aged , Biomarkers, Tumor/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kallikreins/biosynthesis , Kaplan-Meier Estimate , Male , Middle Aged , RNA, Messenger/biosynthesis
8.
Proc Natl Acad Sci U S A ; 111(2): 763-8, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24347643

ABSTRACT

Homologous recombination deficient (HR(-)) mammalian cells spontaneously display reduced replication fork (RF) movement and mitotic extra centrosomes. We show here that these cells present a complex mitotic phenotype, including prolonged metaphase arrest, anaphase bridges, and multipolar segregations. We then asked whether the replication and the mitotic phenotypes are interdependent. First, we determined low doses of hydroxyurea that did not affect the cell cycle distribution or activate CHK1 phosphorylation but did slow the replication fork movement of wild-type cells to the same level than in HR(-) cells. Remarkably, these low hydroxyurea doses generated the same mitotic defects (and to the same extent) in wild-type cells as observed in unchallenged HR(-) cells. Reciprocally, supplying nucleotide precursors to HR(-) cells suppressed both their replication deceleration and mitotic extra centrosome phenotypes. Therefore, subtle replication stress that escapes to surveillance pathways and, thus, fails to prevent cells from entering mitosis alters metaphase progression and centrosome number, resulting in multipolar mitosis. Importantly, multipolar mitosis results in global unbalanced chromosome segregation involving the whole genome, even fully replicated chromosomes. These data highlight the cross-talk between chromosome replication and segregation, and the importance of HR at the interface of these two processes for protection against general genome instability.


Subject(s)
Chromosomal Instability/physiology , DNA Replication/physiology , Homologous Recombination/physiology , Mitosis/physiology , Animals , Aphidicolin , Cell Line , Centrosome/physiology , Chromosome Segregation/physiology , Cricetinae , Cricetulus , Flow Cytometry , Hydroxyurea/metabolism , Microscopy, Video , Statistics, Nonparametric , Time-Lapse Imaging
9.
Cell Rep ; 5(1): 21-8, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24095737

ABSTRACT

The choice of the appropriate double-strand break (DSB) repair pathway is essential for the maintenance of genomic stability. Here, we show that the Bloom syndrome gene product, BLM, counteracts CtIP/MRE11-dependent long-range deletions (>200 bp) generated by alternative end-joining (A-EJ). BLM represses A-EJ in an epistatic manner with 53BP1 and RIF1 and is required for ionizing-radiation-induced 53BP1 focus assembly. Conversely, in the absence of 53BP1 or RIF1, BLM promotes formation of A-EJ long deletions, consistent with a role for BLM in DSB end resection. These data highlight a dual role for BLM that influences the DSB repair pathway choice: (1) protection against CtIP/MRE11 long-range deletions associated with A-EJ and (2) promotion of DNA resection. These antagonist roles can be regulated, according to cell-cycle stage, by interacting partners such as 53BP1 and TopIII, to avoid unscheduled resection that might jeopardize genome integrity.


Subject(s)
Carrier Proteins/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Amino Acid Sequence , Carrier Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases , Humans , MRE11 Homologue Protein , Nuclear Proteins/metabolism , Transfection
10.
Nucleus ; 3(5): 411-7, 2012.
Article in English | MEDLINE | ID: mdl-22895091

ABSTRACT

Progeroid phenotypes are mainly encountered in 2 types of syndromes: in laminopathies, which are characterized by nuclear shape abnormalities due to lamin A alteration, and in DNA damage response defect syndromes. Because lamin A dysregulation leads to DNA damages, it has been proposed that senescence occurs in both types of syndromes through the accumulation of damages. We recently showed that elevated oxidative stress is responsible for lamin B1 accumulation, nuclear shape alteration and senescence in the DDR syndrome, ataxia telangiectasia (A-T). Interestingly, overexpression of lamin B1 in wild type cells is sufficient to induce senescence without the induction of DNA damages. Here, we will discuss the importance of controlling the lamins level in order for maintenance nuclear architecture and we will comment the relationships of lamins with other senescence mechanisms. Finally, we will describe emerging data reporting redox control by lamins, leading us to propose a general mechanism by which reactive oxygen species can induce senescence through lamin dysregulation and NSA.


Subject(s)
Cell Nucleus/metabolism , Cellular Senescence , Lamin Type A/metabolism , Lamin Type B/metabolism , Oxidative Stress , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Nucleus Shape , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Mice , Oxidation-Reduction , Progeria/metabolism , Progeria/pathology , Protein Serine-Threonine Kinases/metabolism , Telomere/metabolism , Tumor Suppressor Proteins/metabolism
11.
Am J Cancer Res ; 2(3): 249-68, 2012.
Article in English | MEDLINE | ID: mdl-22679557

ABSTRACT

A DNA double strand break (DSB) is a highly toxic lesion, which can generate genetic instability and profound genome rearrangements. However, DSBs are required to generate diversity during physiological processes such as meiosis or the establishment of the immune repertoire. Thus, the precise regulation of a complex network of processes is necessary for the maintenance of genomic stability, allowing genetic diversity but protecting against genetic instability and its consequences on oncogenesis. Two main strategies are employed for DSB repair: homologous recombination (HR) and non-homologous end-joining (NHEJ). HR is initiated by single-stranded DNA (ssDNA) resection and requires sequence homology with an intact partner, while NHEJ requires neither resection at initiation nor a homologous partner. Thus, resection is an pivotal step at DSB repair initiation, driving the choice of the DSB repair pathway employed. However, an alternative end-joining (A-EJ) pathway, which is highly mutagenic, has recently been described; A-EJ is initiated by ssDNA resection but does not require a homologous partner. The choice of the appropriate DSB repair system, for instance according the cell cycle stage, is essential for genome stability maintenance. In this context, controlling the initial events of DSB repair is thus an essential step that may be irreversible, and the wrong decision should lead to dramatic consequences. Here, we first present the main DSB repair mechanisms and then discuss the importance of the choice of the appropriate DSB repair pathway according to the cell cycle phase. In a third section, we present the early steps of DSB repair i.e., DSB signaling, chromatin remodeling, and the regulation of ssDNA resection. In the last part, we discuss the competition between the different DSB repair mechanisms. Finally, we conclude with the importance of the fine tuning of this network for genome stability maintenance and for tumor protection in fine.

12.
EMBO J ; 31(5): 1080-94, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22246186

ABSTRACT

We report crosstalk between three senescence-inducing conditions, DNA damage response (DDR) defects, oxidative stress (OS) and nuclear shape alterations. The recessive autosomal genetic disorder Ataxia telangiectasia (A-T) is associated with DDR defects, endogenous OS and premature ageing. Here, we find frequent nuclear shape alterations in A-T cells, as well as accumulation of the key nuclear architecture component lamin B1. Lamin B1 overexpression is sufficient to induce nuclear shape alterations and senescence in wild-type cells, and normalizing lamin B1 levels in A-T cells reciprocally reduces both nuclear shape alterations and senescence. We further show that OS increases lamin B1 levels through p38 Mitogen Activated Protein kinase activation. Lamin B1 accumulation and nuclear shape alterations also occur during stress-induced senescence and oncogene-induced senescence (OIS), two canonical senescence situations. These data reveal lamin B1 as a general molecular mediator that controls OS-induced senescence, independent of established Ataxia Telangiectasia Mutated (ATM) roles in OIS.


Subject(s)
Aging , Lamin Type B/metabolism , Oxidative Stress , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Humans
13.
Cancer Res ; 71(10): 3590-602, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21444675

ABSTRACT

Genetic stability requires coordination of a network of pathways including DNA repair/recombination and apoptosis. In addition to its canonical anti-apoptotic role, Bcl-2 negatively impacts genome stability. In this study, we identified the breast cancer tumor suppressor BRCA1, which plays an essential role in homologous recombination (HR), as a target for Bcl-2 in the repression of HR. Indeed, ionizing radiation-induced BRCA1 foci assembly was repressed when Bcl-2 was expressed ectopically, in human SV40 fibroblasts, or spontaneously, in lymphoma t(14:18) cells and in HeLa and H460 cancer cell lines. Moreover, we showed that the transmembrane (TM) domain of Bcl-2 was required for both inhibition of BRCA1 foci assembly and the inhibition of HR induced by a double-strand break targeted into an intrachromosomal HR substrate by the meganuclease I-SceI. Fluorescence confocal microscopy, proximity ligation assay, and electron microscopy analyses as well as Western blot analysis of subcellular fractions showed that Bcl-2 and BRCA1 colocalized to mitochondria and endoplasmic reticulum in a process requiring the TM domain of Bcl-2. Targeting BRCA1 to the endomembranes depletes BRCA1 from the nucleus and, thus, accounts for the inhibition of HR. Furthermore, our findings support an apoptosis-stimulatory role for the cytosolic form of BRCA1, suggesting a new tumor suppressor function of BRCA1. Together, our results reveal a new mode of BRCA1 regulation and for HR in the maintenance of genome stability.


Subject(s)
Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombination, Genetic , Cell Line, Tumor , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Mitochondria/metabolism , Models, Genetic , Subcellular Fractions/metabolism
14.
Oncol Rep ; 17(4): 713-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17342305

ABSTRACT

Any factor affecting BRCA gene regulation may be of interest in the prevention of breast tumourigenesis. We studied the influence of dietary docosahexaenoic acid (DHA), a major omega-3 fatty acid present in marine products, on rat autochthonous mammary tumourigenesis. DHA-supplementation significantly reduced the incidence of tumours (30%, P=0.007) and led to a 60% increase (P=0.02) in BRCA1 protein level. Since DHA influences the product of a major tumour suppressor gene, this finding may contribute to the observation that high-fish consumption reduces the risk of breast cancer.


Subject(s)
BRCA1 Protein/metabolism , Dietary Fats, Unsaturated/administration & dosage , Docosahexaenoic Acids/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/prevention & control , Animals , BRCA1 Protein/analysis , BRCA1 Protein/genetics , Female , Mammary Neoplasms, Animal/chemistry , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
15.
Int J Biochem Cell Biol ; 39(4): 774-86, 2007.
Article in English | MEDLINE | ID: mdl-17307016

ABSTRACT

Ionic channel activity is involved in fundamental cellular behaviour and participates in cancerous features such as proliferation, migration and invasion which in turn contribute to the metastatic process. In this study, we investigated the expression and role of voltage-gated sodium channels in non-small-cell lung cancer cell lines. Functional voltage-gated sodium channels expression was investigated in normal and non-small-cell lung cancer cell lines. The measurement, in patch-clamp conditions, of tetrodotoxin-inhibitable sodium currents indicated that the strongly metastatic cancerous cell lines H23, H460 and Calu-1 possess functional sodium channels while normal and weakly metastatic cell lines do not. While all the cell lines expressed mRNA for numerous sodium channel isoforms, only H23, H460 and Calu-1 cells had a 250 kDa protein corresponding to the functional channel. The other cell lines also had another protein of 230 kDa which is not addressed to the membrane and might act as a dominant negative isoform to prevent channel activation. At the membrane potential of these cells, channels are partially open. This leads to a continuous entry of sodium, disrupting sodium homeostasis and down-stream signaling pathways. Inhibition of the channels by tetrodotoxin was responsible for a 40-50% reduction of in vitro invasion. These experiments suggest that the functional expression of voltage-gated sodium channels might be an integral component of the metastatic process in non-small-cell lung cancer cells probably through its involvement in the regulation of intracellular sodium homeostasis. These channels could serve both as novel markers of the metastatic phenotype and as potential new therapeutic targets.


Subject(s)
Cell Movement/physiology , Sodium Channels/physiology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Profiling , Homeostasis/drug effects , Humans , Intracellular Fluid/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Membrane Potentials/drug effects , Microscopy, Confocal , Neoplasm Invasiveness , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Tetrodotoxin/pharmacology
16.
Int J Biochem Cell Biol ; 38(2): 196-208, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16194618

ABSTRACT

Long-chain omega-3 polyunsaturated fatty acids are thought to inhibit the development of breast cancer. We investigated the effects of docosahexaenoic and eicosapentaenoic acids on the proliferation of MDA-MB-231 human mammary epithelial cells. Both docosahexaenoic and eicosapentaenoic acids decreased cell growth with a higher efficiency for docosahexaenoic acid (87% at 100 microM versus 74% for eicosapentaenoic acid). The effect on specific cell cycle phases was studied. G2/M duration was markedly increased by docosahexaenoic and by eicosapentaenoic acids (respectively by more than seven- and six-fold at 50 microM) when cells were synchronized at the G1/S boundary and released in the cell cycle. In contrast, there was no alteration of G1 or S phases. The expression of cyclin A, cyclin B1 and cyclin-dependent kinase 1, the regulators required for the progression from G2 to mitosis, were all decreased by these fatty acids (western blot). Since omega-3 fatty acids had no effect on the S phase, thus ruling out an involvement of cyclin A in their anti-proliferative effect, we examined whether the regulation of the cyclin-dependent kinase 1-cyclin B1 complex was altered. Upon omega-3 fatty acids treatment, cyclin B1 phosphorylation was inhibited and the expression of the cell division cycle 25C phosphatase, which dephosphorylates cyclin-dependent kinase 1, was decreased. We conclude that the anti-proliferative effect of omega-3 fatty acids occurs via the regulation of the cyclin-dependent kinase 1-cyclin B1 complex.


Subject(s)
Antineoplastic Agents/metabolism , Breast Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin B/metabolism , Fatty Acids, Omega-3/pharmacology , Apoptosis/physiology , CDC2 Protein Kinase/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cyclin B/genetics , Cyclin B1 , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/metabolism , Female , Humans , Phospholipids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...