Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nanomaterials (Basel) ; 9(7)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31247939

ABSTRACT

Silver nanoparticles (Ag NPs) exhibit antibacterial activity and are extensively used in numerous applications. The aim of this study was to examine the toxic effect of Ag NPs on the marine microalga, Chlorella vulgaris. The microalgae, at the exponential growth phase, were treated with different concentrations of Ag NPs (50 and 100 nm) for 96 h. X-Ray diffraction (XRD) results indicated that the used NPs are single and pure Ag phase with a mean crystallite size of 21 and 32 nm. Ag NPs were found to have a negative effect on viable cell concentration, a variable effect on chlorophyll a concentration, and increased ROS formation. Transmission electron microscopy (TEM) analysis revealed that Ag NPs were present inside the microalgae cells and formed large aggregates in the culture medium. Ag+ ions, in the form of AgNO3, were also assessed at higher concentrations and found to cause inhibitory effects.

2.
Biochim Biophys Acta Mol Cell Res ; 1865(7): 981-994, 2018 07.
Article in English | MEDLINE | ID: mdl-29678654

ABSTRACT

Calcium (Ca2+) release from the endoplasmic reticulum plays an important role in many cell-fate defining cellular processes. Traditionally, this Ca2+ release was associated with the ER Ca2+ release channels, inositol 1,4,5­triphosphate receptor (IP3R) and ryanodine receptor (RyR). Lately, however, other calcium conductances have been found to be intracellularly localized and to participate in cell fate regulation. Nonetheless, molecular identity and functional properties of the ER Ca2+ release mechanisms associated with multiple diseases, e.g. prostate cancer, remain unknown. Here we identify a new family of transient receptor potential melastatine 8 (TRPM8) channel isoforms as functional ER Ca2+ release channels expressed in mitochondria-associated ER membranes (MAMs). These TRPM8 isoforms exhibit an unconventional structure with 4 transmembrane domains (TMs) instead of 6 TMs characteristic of the TRP channel archetype. We show that these 4TM-TRPM8 isoforms form functional channels in the ER and participate in regulation of the steady-state Ca2+ concentration ([Ca2+]) in mitochondria and the ER. Thus, our study identifies 4TM-TRPM8 isoforms as ER Ca2+ release mechanism distinct from classical Ca2+ release channels.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Aged , Alternative Splicing , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/genetics , Protein Domains , TRPM Cation Channels/chemistry
3.
Sci Rep ; 7(1): 15896, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29162859

ABSTRACT

Despite the tremendous progress in medicine, cancer remains one of the most serious global health problems awaiting new effective therapies. Here we present ferroquine (FQ), the next generation antimalarial drug, as a promising candidate for repositioning as cancer therapeutics. We report that FQ potently inhibits autophagy, perturbs lysosomal function and impairs prostate tumor growth in vivo. We demonstrate that FQ negatively regulates Akt kinase and hypoxia-inducible factor-1α (HIF-1α) and is particularly effective in starved and hypoxic conditions frequently observed in advanced solid cancers. FQ enhances the anticancer activity of several chemotherapeutics suggesting its potential application as an adjuvant to existing anticancer therapy. Alike its parent compound chloroquine (CQ), FQ accumulates within and deacidifies lysosomes. Further, FQ induces lysosomal membrane permeabilization, mitochondrial depolarization and caspase-independent cancer cell death. Overall, our work identifies ferroquine as a promising new drug with a potent anticancer activity.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Ferrous Compounds/pharmacology , Aminoquinolines/chemistry , Animals , Antimalarials/chemistry , Autophagy/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/chemistry , Chloroquine/pharmacology , Female , Ferrous Compounds/chemistry , Hydrogen-Ion Concentration , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Metallocenes , Mice, Nude , Neoplasms/pathology , Permeability , Stress, Physiological , Xenograft Model Antitumor Assays
4.
Sci Rep ; 7(1): 4728, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28680151

ABSTRACT

Humans are exposed to multiple exogenous environmental pollutants. Many of these compounds are parts of mixtures that can exacerbate harmful effects of the individual mixture components. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is primarily produced via industrial processes including incineration and the manufacture of herbicides. Both endosulfan and TCDD are persistent organic pollutants which elicit cytotoxic effects by inducing reactive oxygen species generation. Sublethal concentrations of mixtures of TCDD and endosulfan increase oxidative stress, as well as mitochondrial homeostasis disruption, which is preceded by a calcium rise and, in fine, induce cell death. TCDD+Endosulfan elicit a complex signaling sequence involving reticulum endoplasmic destalilization which leads to Ca2+ rise, superoxide anion production, ATP drop and late NADP(H) depletion associated with a mitochondrial induced apoptosis concomitant early autophagic processes. The ROS scavenger, N-acetyl-cysteine, blocks both the mixture-induced autophagy and death. Calcium chelators act similarly and mitochondrially targeted anti-oxidants also abrogate these effects. Inhibition of the autophagic fluxes with 3-methyladenine, increases mixture-induced cell death. These findings show that subchronic doses of pollutants may act synergistically. They also reveal that the onset of autophagy might serve as a protective mechanism against ROS-triggered cytotoxic effects of a cocktail of pollutants in Caco-2 cells and increase their tumorigenicity.


Subject(s)
Endosulfan/toxicity , Environmental Pollutants/toxicity , Mitochondria/drug effects , Polychlorinated Dibenzodioxins/toxicity , Apoptosis , Autophagy , Caco-2 Cells , Calcium/metabolism , Cell Survival/drug effects , Drug Synergism , Endoplasmic Reticulum/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Toxicity Tests, Subchronic
5.
Mol Carcinog ; 56(8): 1851-1867, 2017 08.
Article in English | MEDLINE | ID: mdl-28277613

ABSTRACT

Previous studies showed the effects of resveratrol (RES) on several cancer cells, including prostate cancer (PCa) cell apoptosis without taking into consideration the impact of the tumor microenvironment (TME). The TME is composed of cancer cells, endothelial cells, blood cells, and cancer-associated fibroblasts (CAF), the main source of growth factors. The latter cells might modify in the TME the impact of RES on tumor cells via secreted factors. Recent data clearly show the impact of CAF on cancer cells apoptosis resistance via secreted factors. However, the effects of RES on PCa CAF have not been studied so far. We have investigated here for the first time the effects of RES on the physiology of PCa CAF in the context of TME. Using a prostate cancer CAF cell line and primary cultures of CAF from prostate cancers, we show that RES activates the N-terminal mutated Transient Receptor Potential Ankyrin 1 (TRPA1) channel leading to an increase in intracellular calcium concentration and the expression and secretion of growth factors (HGF and VEGF) without inducing apoptosis in these cells. Interestingly, in the present work, we also show that when the prostate cancer cells were co-cultured with CAF, the RES-induced cancer cell apoptosis was reduced by 40%, an apoptosis reduction canceled in the presence of the TRPA1 channel inhibitors. The present work highlights CAF TRPA1 ion channels as a target for RES and the importance of the channel in the epithelial-stromal crosstalk in the TME leading to resistance to the RES-induced apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , Calcium Channels/metabolism , Cancer-Associated Fibroblasts/drug effects , Nerve Tissue Proteins/metabolism , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Stilbenes/pharmacology , Transient Receptor Potential Channels/metabolism , Amino Acid Sequence , Apoptosis/drug effects , Calcium/metabolism , Calcium Channels/analysis , Calcium Channels/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Humans , Male , Mutation , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Resveratrol , TRPA1 Cation Channel , Transient Receptor Potential Channels/analysis , Transient Receptor Potential Channels/genetics , Tumor Microenvironment/drug effects
6.
Cancer Prev Res (Phila) ; 10(3): 177-187, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28096238

ABSTRACT

Accruing evidence indicates that exposure to environmental compounds may adversely affect human health and promote carcinogenesis. Triclosan (TCS), an antimicrobial agent widely used as a preservative in personal care products, has been shown to act as an endocrine disruptor in hormone-dependent tissues. Here, we demonstrate a new molecular mechanism by which TCS stimulates the secretion by human prostate cancer stromal cells of vascular endothelial growth factor (VEGF), a factor known to promote tumor growth. This mechanism involves an increase in intracellular calcium levels due to the direct activation of a membrane ion channel. Using calcium imaging and electrophysiology techniques, we show for the first time that environmentally relevant concentrations of TCS activate a cation channel of the TRP family, TRPA1 (Transient Receptor Potential Ankirin 1), in primary cultured human prostate cancer stromal cells. The TCS-induced TRPA1 activation increased basal calcium in stromal cells and stimulated the secretion of VEGF and epithelial cells proliferation. Interestingly, immunofluorescence labeling performed on formalin-fixed paraffin-embedded prostate tissues showed an exclusive expression of the TRPA1 channel in prostate cancer stromal cells. Our data demonstrate an impact of the environmental factor TCS on the tumor microenvironment interactions, by activating a tumor stroma-specific TRPA1 ion channel. Cancer Prev Res; 10(3); 177-87. ©2017 AACR.


Subject(s)
Anti-Infective Agents, Local/toxicity , Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/metabolism , Stromal Cells/drug effects , Transient Receptor Potential Channels/metabolism , Triclosan/toxicity , Vascular Endothelial Growth Factor A/metabolism , Carcinogens, Environmental/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Stromal Cells/metabolism , TRPA1 Cation Channel , Tumor Microenvironment/drug effects
7.
Environ Sci Pollut Res Int ; 24(4): 4144-4152, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27933501

ABSTRACT

Graphene oxide (GO), a recently discovered material, has been investigated for many applications in various fields. Thus, an immense awareness should be paid on the potential effects of the material on the environment as huge quantities of GO may get to the environment. Aquatic organisms, marine algae as an example, are exposed to such material when disposed to the environment. Accordingly, it is significant to assess the probable interactions of GO with algae in evaluating its possible environmental risks. In this study, we have examined the effect of different concentrations of GO on Picochlorum sp. during the different growth phases. The results showed that the toxicity of GO increases with increasing its concentration. The lowest concentration (0.5 mg L-1) was found to improve the algae growth and pigment content of Picochlorum sp. In contrast, higher GO concentrations had a negative consequence on the growth of algae and photosynthetic pigment concentration.


Subject(s)
Chlorophyta/drug effects , Graphite/pharmacology , Photosynthesis/drug effects
8.
FASEB J ; 30(9): 3155-70, 2016 09.
Article in English | MEDLINE | ID: mdl-27317670

ABSTRACT

Testes of most male mammals present the particularity of being externalized from the body and are consequently slightly cooler than core body temperature (4-8°C below). Although, hypothermia of the testis is known to increase germ cells apoptosis, little is known about the underlying molecular mechanisms, including cold sensors, transduction pathways, and apoptosis triggers. In this study, using a functional knockout mouse model of the cold and menthol receptors, dubbed transient receptor potential melastatine 8 (TRPM8) channels, we found that TRPM8 initiated the cold-shock response by differentially modulating cold- and heat-shock proteins. Besides, apoptosis of germ cells increased in proportion to the cooling level in control mice but was independent of temperature in knockout mice. We also observed that the rate of germ cell death correlated positively with the reactive oxygen species level and negatively with the expression of the detoxifying enzymes. This result suggests that the TRPM8 sensor is a key determinant of germ cell fate under hypothermic stimulation.-Borowiec, A.-S., Sion, B., Chalmel, F., Rolland, A. D., Lemonnier, L., De Clerck, T., Bokhobza, A., Derouiche, S., Dewailly, E., Slomianny, C., Mauduit, C., Benahmed, M., Roudbaraki, M., Jégou, B., Prevarskaya, N., Bidaux, G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation.


Subject(s)
Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , Testis/physiology , Animals , Cold Temperature , Gene Expression Regulation , HEK293 Cells , Humans , Male , Meiosis , Mice , Mice, Knockout , Oxidation-Reduction , TRPM Cation Channels/genetics
9.
Oncotarget ; 7(20): 29063-80, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27074561

ABSTRACT

Since its cloning a decade ago, TRPM8 channel has emerged as a promising prognostic marker and a putative therapeutic target in prostate cancer (PCa). However, recent studies have brought to light the complexity of TRPM8 isoforms in PCa. Consequently, the respective role of each TRPM8 isoform needs to be deciphered prior to considering TRPM8 as an attractive therapeutic target. Full-length (6 transmembrane (TM)-domain) TRPM8 channel is overexpressed in early PCa and repressed in advanced prostate tumors whereas the localization of the truncated, 4TM-TRPM8 channel (4 transmembrane (TM)-domain), in the membranes of endoplasmic reticulum (ER) is independent of the pathogenic status of epithelial cells. In the same line, expression of non-channel cytoplasmic small TRPM8 isoforms (namely sM8) is conserved in cancer cells. In this study, we identify sM8s as putative regulator of PCa cell death. Indeed, suppression of sM8 isoforms was found to induce concomitantly ER stress, oxidative stress, p21 expression and apoptosis in human epithelial prostate cancer cells. We furthermore demonstrate that induction of such mechanisms required the activity of 4TM-TRPM8 channels at the ER-mitochondria junction. Our study thus suggests that targeting sM8 could be an appropriate strategy to fight prostate cancer.


Subject(s)
Prostatic Neoplasms/pathology , TRPM Cation Channels/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Protein Isoforms/metabolism
10.
Proc Natl Acad Sci U S A ; 112(26): E3345-54, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26080404

ABSTRACT

Deviation of the ambient temperature is one of the most ubiquitous stimuli that continuously affect mammals' skin. Although the role of the warmth receptors in epidermal homeostasis (EH) was elucidated in recent years, the mystery of the keratinocyte mild-cold sensor remains unsolved. Here we report the cloning and characterization of a new functional epidermal isoform of the transient receptor potential M8 (TRPM8) mild-cold receptor, dubbed epidermal TRPM8 (eTRPM8), which is localized in the keratinocyte endoplasmic reticulum membrane and controls mitochondrial Ca(2+) concentration ([Ca(2+)]m). In turn, [Ca(2+)]m modulates ATP and superoxide (O2(·-)) synthesis in a cold-dependent manner. We report that this fine tuning of ATP and O2(·-) levels by cooling controls the balance between keratinocyte proliferation and differentiation. Finally, to ascertain eTRPM8's role in EH in vivo we developed a new functional knockout mouse strain by deleting the pore domain of TRPM8 and demonstrated that eTRPM8 knockout impairs adaptation of the epidermis to low temperatures.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cold Temperature , Epidermis/metabolism , Keratinocytes/cytology , Protein Isoforms/physiology , TRPM Cation Channels/physiology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Superoxides/metabolism
11.
Biofouling ; 30(7): 845-58, 2014.
Article in English | MEDLINE | ID: mdl-25115519

ABSTRACT

This study was designed to establish the presence and function of the mucous layer surrounding spores of Bacillus subtilis. First, an external layer of variable thickness and regularity was often observed on B. subtilis spores. Further analyses were performed on B. subtilis 98/7 spores surrounded by a thick layer. The mechanical removal of the layer did not affect their resistance to heat or their ability to germinate but rendered the spore less hydrophilic, more adherent to stainless steel, and more resistant to cleaning. This layer was mainly composed of 6-deoxyhexoses, ie rhamnose, 3-O-methyl-rhamnose and quinovose, but also of glucosamine and muramic lactam, known also to be a part of the bacterial peptidoglycan. The specific hydrolysis of the peptidoglycan using lysozyme altered the structure of the required mucous layer and affected the physico-chemical properties of the spores. Such an outermost mucous layer has also been seen on spores of B. licheniformis and B. clausii isolated from food environments.


Subject(s)
Bacillus subtilis/physiology , Biofilms , Mucus/physiology , Bacillus/physiology , Biofouling , Spores/physiology , Surface Properties
12.
Environ Sci Pollut Res Int ; 21(5): 3515-23, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24271730

ABSTRACT

In screening indigenous soil filamentous fungi for polycyclic aromatic hydrocarbons (PAHs) degradation, an isolate of the Fusarium solani was found to incorporate benzo[a]pyrene (BaP) into fungal hyphae before degradation and mineralization. The mechanisms involved in BaP uptake and intracellular transport remain unresolved. To address this, the incorporation of two PAHs, BaP, and phenanthrene (PHE) were studied in this fungus. The fungus incorporated more BaP into cells than PHE, despite the 400-fold higher aqueous solubility of PHE compared with BaP, indicating that PAH incorporation is not based on a simple diffusion mechanism. To identify the mechanism of BaP incorporation and transport, microscopic studies were undertaken with the fluorescence probes Congo Red, BODIPY®493/503, and FM®4-64, targeting different cell compartments respectively fungal cell walls, lipids, and endocytosis. The metabolic inhibitor sodium azide at 100 mM totally blocked BaP incorporation into fungal cells indicating an energy-requirement for PAH uptake into the mycelium. Cytochalasins also inhibited BaP uptake by the fungus and probably its intracellular transport into fungal hyphae. The perfect co-localization of BaP and BODIPY reveals that lipid bodies constitute the intracellular storage sites of BaP in F. solani. Our results demonstrate an energy-dependent uptake of BaP and its cytoskeleton-dependent intracellular transport by F. solani.


Subject(s)
Benzo(a)pyrene/metabolism , Fusarium/metabolism , Phenanthrenes/metabolism , Soil Pollutants/metabolism , Biodegradation, Environmental , Biological Transport , Cytochalasin B/pharmacology , Cytoskeleton/metabolism , Fluorescent Dyes/pharmacology , Sodium Azide/pharmacology
13.
Biol Open ; 2(9): 941-51, 2013.
Article in English | MEDLINE | ID: mdl-24143281

ABSTRACT

It is strongly suspected that potassium (K(+)) channels are involved in various aspects of prostate cancer development, such as cell growth. However, the molecular nature of those K(+) channels implicated in prostate cancer cell proliferation and the mechanisms through which they control proliferation are still unknown. This study uses pharmacological, biophysical and molecular approaches to show that the main voltage-dependent K(+) current in prostate cancer LNCaP cells is carried by large-conductance BK channels. Indeed, most of the voltage-dependent current was inhibited by inhibitors of BK channels (paxillin and iberiotoxin) and by siRNA targeting BK channels. In addition, we reveal that BK channels constitute the main K(+) channel family involved in setting the resting membrane potential in LNCaP cells at around -40 mV. This consequently promotes a constitutive calcium entry through T-type Cav3.2 calcium channels. We demonstrate, using single-channel recording, confocal imaging and co-immunoprecipitation approaches, that both channels form macromolecular complexes. Finally, using flow cytometry cell cycle measurements, cell survival assays and Ki67 immunofluorescent staining, we show that both BK and Cav3.2 channels participate in the proliferation of prostate cancer cells.

14.
PLoS One ; 8(10): e77507, 2013.
Article in English | MEDLINE | ID: mdl-24116231

ABSTRACT

For almost 30 years, keratinocyte differentiation has been studied in numerous cell models including keratinocyte primary culture with various supplemented culture media. In this respect, it has become quite difficult to draw comparisons between studies using such a variety of culture conditions. Serum-free condition with low calcium has been used to culture basal proliferating cells, though differentiation is induced by various procedures. These latter include the addition of calcium at mM concentration and a concomitant addition of serum and calcium. Lowering the incubation temperature of cells has also been reported to induce a premature differentiation of keratinocytes in organotypic skin culture. This effect of temperature on keratinocyte differentiation has been poorly depicted, although average human skin temperature has been shown to be about 32 °C. However, studying differentiation and quantifying shifts in the differentiation rate of a cell population implies to precisely know i) the proportion of differentiated cells in the whole population, and ii) to which extent and to which level of expression, the induction of a gene or a protein might be considered as a marker of differentiation. This lack has rarely been taken into consideration and has surely led to over-interpretations of single protein induction and to consequent extrapolations to real differentiation processes. By means of paralleled analyses with immunocytofluorescence, flow cytometry, and with multiple differentiation markers quantify by qPCR and western-blot, we studied the paradoxical connection between calcium, serum, multilayer culture and incubation temperature on the differentiation of in vitro keratinocytes. Conversely to previous reports, we have shown that calcium switch is indeed a potent model for inducing calcium-dependent genes, but is not an efficient procedure when one wishes to assess the keratinocyte differentiation rate. Moreover, we have demonstrated that a synergic stimulation by calcium, serum, confluence and lower incubation temperature amplified the differentiation rate.


Subject(s)
Cell Differentiation , Keratinocytes/cytology , Animals , Calcium/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Keratinocytes/metabolism , Mice , Skin/cytology , Temperature
15.
FASEB J ; 27(4): 1600-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23322163

ABSTRACT

The endoplasmic reticulum (ER) is involved in many cellular functions, including protein folding and Ca(2+) homeostasis. The ability of cells to respond to the ER stress is critical for cell survival, and disruption in such regulation can lead to apoptosis. ER stress is accompanied by alterations in Ca(2+) homeostasis, and the ER Ca(2+) store depletion by itself can induce ER stress and apoptosis. Despite that, the ER Ca(2+) leak channels activated in response to the ER stress remain poorly characterized. Here we demonstrate that ER Ca(2+) depletion during the ER stress occurs via translocon, the ER protein complex involved in translation. Numerous ER stress inducers stimulate the ER Ca(2+) leak that can be prevented by translocon inhibitor, anisomycin. Expression of GRP78, an ER stress marker, increased following treatment with puromycin (a translocon opener) and was suppressed by anisomycin, confirming a primary role of translocon in ER stress induction. Inhibition of ER store depletion by anisomycin significantly reduces apoptosis stimulated by the ER stress inducers. We suggest that translocon opening is physiologically modulated by GRP78, particularly during the ER stress. The ability to modulate the ER Ca(2+) permeability and subsequent ER stress can lead to development of a novel therapeutic approach.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Unfolded Protein Response , Anisomycin/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Homeostasis/physiology , Humans , Puromycin/pharmacology , Unfolded Protein Response/physiology
16.
Cell Calcium ; 47(4): 350-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20172609

ABSTRACT

Orai1, together with STIM1 and STIM2, constitutes the molecular basis for store-operated calcium entry (SOCE) and we have investigated their role in cell proliferation and cell cycle progression in HEK293 cells. 48-h serum deprival, and a 24-h treatment with 1 mM hydroxyurea or with 10 microM RO-3306--a cyclin-dependent kinase 1 inhibitor--induced cell cycle block in G1, S and G2/M, respectively. SOCE amplitude, monitored in whole-cell voltage clamped cells, was markedly reduced (60-70%) in all conditions, with full reversibility within 4h. Silencing of Orai and STIM1 using siRNA resulted in a large inhibition of SOCE (70-80%) whereas siSTIM2 had a smaller but significant effect (30%). However, the cell population doubling time was not affected in siSTIM1 cells (18 h, the same as in control cells) but was increased in both siOrai1 cells (29 h) and in siSTIM2 (23 h) even when combined with siSTIM1. This suggests that STIM1 plays no role in cell proliferation in HEK293 cells while STIM2 is involved in both SOCE and cell proliferation in these cells. Finally, the cell cycle block induced SOCE inhibition was associated with reduced Orai1 expression with full recovery within 4h, whereas the expression of STIM1 and STIM2 remained unaltered. These observations reveal a tight relation between cell proliferation, calcium entry and Orai1 expression in HEK293 cells.


Subject(s)
Calcium Channels/metabolism , Cell Adhesion Molecules/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , TRPC Cation Channels/metabolism , CDC2 Protein Kinase/pharmacology , Calcium Channels/genetics , Cell Adhesion Molecules/genetics , Cell Line , Cell Proliferation/drug effects , Humans , Hydroxyurea/pharmacology , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , ORAI1 Protein , Patch-Clamp Techniques , Quinolines/pharmacology , RNA, Small Interfering/genetics , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , Thiazoles/pharmacology , Transfection
17.
PLoS One ; 4(9): e7068, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19763272

ABSTRACT

BACKGROUND: During androgen ablation prostate cancer cells' growth and survival become independent of normal regulatory mechanisms. These androgen-independent cells acquire the remarkable ability to adapt to the surrounding microenvironment whose factors, such as neurotransmitters, influence their survival. Although findings are becoming evident about the expression of alpha(1A)-adrenoceptors in prostate cancer epithelial cells, their exact functional role in androgen-independent cells has yet to be established. Previous work has demonstrated that membrane lipid rafts associated with key signalling proteins mediate growth and survival signalling pathways in prostate cancer cells. METHODOLOGY/PRINCIPAL FINDINGS: In order to analyze the membrane topology of the alpha(1A)-adrenoceptor we explored its presence by a biochemical approach in purified detergent resistant membrane fractions of the androgen-independent prostate cancer cell line DU145. Electron microscopy observations demonstrated the colocalization of the alpha(1A)-adrenoceptor with caveolin-1, the major protein component of caveolae. In addition, we showed that agonist stimulation of the alpha(1A)-adrenoceptor induced resistance to thapsigargin-induced apoptosis and that caveolin-1 was necessary for this process. Further, immunohistofluorescence revealed the relation between high levels of alpha(1A)-adrenoceptor and caveolin-1 expression with advanced stage prostate cancer. We also show by immunoblotting that the TG-induced apoptosis resistance described in DU145 cells is mediated by extracellular signal-regulated kinases (ERK). CONCLUSIONS/SIGNIFICANCE: In conclusion, we propose that alpha(1A)-adrenoceptor stimulation in androgen-independent prostate cancer cells via caveolae constitutes one of the mechanisms contributing to their protection from TG-induced apoptosis.


Subject(s)
Apoptosis , Caveolae/metabolism , Drug Resistance, Neoplasm , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Cholesterol/metabolism , Humans , Male , Membrane Microdomains , Models, Biological , Neurotransmitter Agents , Prostatic Neoplasms/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Sphingomyelins/metabolism , Thapsigargin/pharmacology
18.
J Biol Chem ; 283(15): 10162-73, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18230611

ABSTRACT

Because prostate cancer is, in its early stages, an androgen-dependent pathology, treatments aiming at decreasing testosterone plasma concentration have been developed for many years now. However, a significant proportion of patients suffer a relapse after a few years of hormone therapy. The androgen-independent stage of prostate cancer has been shown to be associated with the development of neuroendocrine differentiation. We previously demonstrated that neuroendocrine prostate cancer cells derived from LNCaP cells overexpress CaV3.2 T-type voltage-dependent calcium channels. We demonstrate here using prostatic acid phosphatase as a marker of prostate secretion and FM1-43 fluorescence imaging of membrane trafficking that neuroendocrine differentiation is associated with an increase in calcium-dependent secretion which critically relies on CaV3.2 T-type calcium channel activity. In addition, we show that these channels are expressed by neuroendocrine cells in prostate cancer tissues obtained from patients after surgery. We propose that CaV3.2 T-type calcium channel up-regulation may account for the alteration of secretion during prostate cancer development and that these channels, by promoting the secretion of potential mitogenic factors, could participate in the progression of the disease toward an androgen-independent stage.


Subject(s)
Biomarkers, Tumor/metabolism , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Carcinoma, Neuroendocrine/metabolism , Growth Substances/metabolism , Prostatic Neoplasms/metabolism , Acid Phosphatase , Androgens/blood , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/therapy , Cell Differentiation , Cell Line, Tumor , Hormone Replacement Therapy , Humans , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Tyrosine Phosphatases/metabolism , Testosterone/blood , Up-Regulation
19.
Biochem J ; 401(1): 49-55, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16965263

ABSTRACT

Prolactin (PRL) has been shown to be involved in the differentiation and proliferation of numerous tissues, including the prostate gland. Moreover, variations in [Ca2+]ER (calcium concentration within the endoplasmic reticulum) may play a role in cell growth. However, few studies have focused on the regulation of calcium homoeostasis by prolactin. The present study evaluates the regulation of calcium pools as well as the possible role of [Ca2+]ER variations as a signal for growth modulation by PRL. We show that PRL stimulates the proliferation of normal SV40 immortalized epithelial prostate (PNT1A) cells with a maximum effect at a dose of 100 ng/ml. We also show that 100 ng/ml PRL increases the [Ca2+]ER when measured either by indirect quantification with Fura-2AM after application of 1 mM thapsigargin or by direct quantification with Mag-Fura-2AM within the endoplas-mic reticulum. Western blot analysis shows that the SERCA 2b (sarcoendoplasmic calcium ATPase 2b) is over-expressed in PNT1A cells treated with 100 ng/ml PRL for 24 h. A small inter-fering RNA SERCA 2a/b, used to down-regulate endogenous SERCA 2b expression, reduced both PNT1A cell proliferation and [Ca2+]ER. We thus identify [Ca2+]ER and SERCA 2b as protagonists in PRL-induced proliferation.


Subject(s)
Cell Division/drug effects , Endoplasmic Reticulum/enzymology , Prolactin/pharmacology , Prostate/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Calcium/metabolism , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Prostate/cytology , Prostate/enzymology , RNA, Small Interfering/genetics , Transfection
20.
Biochimie ; 86(9-10): 633-42, 2004.
Article in English | MEDLINE | ID: mdl-15556273

ABSTRACT

We showed that the metabolism of arachidonic acid (AA) in HepG2 cells generates reactive oxygen species (ROS), which activate the p38 mitogen-activated protein kinase (MAPK) pathway and the redox-sensitive transcription factors AP-1 and NF-kappaB, leading to the induction of the antioxidant manganese superoxide dismutase gene. The present study reports that AA decreases the HepG2 cell growth by 40% and 55% after a treatment for 24 and 48 h, respectively. This effect was blocked by an inhibitor of lipoxygenase/cytochrome P450 monooxygenase pathways and by the antioxidants. In addition, AA induced an oxidative stress, as an accumulation of malondialdehyde (MDA)-modified proteins, resulting to a generation of MDA and H(2)O(2) was observed after 24 h. This AA-induced oxidative stress was associated with the lack of an increase in the H(2)O(2)-degrading enzyme level. In contrast, 5,8,11,14-eicosatetraynoic acid, a nonmetabolizable analog of AA, had not effect. The peroxisome proliferator-activated receptor gamma (PPARgamma) with AA metabolites as ligands was upregulated by the fatty acid but was not involved in the AA effect because its transcriptional activity estimated by reporter gene assays was negatively controlled by p38 MAPK pathway. These findings suggest that the effect of AA on human hepatoma cell growth by inducing an oxidative stress may present a clinical interest in the treatment of the liver cancer.


Subject(s)
Arachidonic Acid/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Lipid Peroxidation/drug effects , Liver Neoplasms/metabolism , Oxidative Stress/drug effects , Arachidonic Acid/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Humans , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...