Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 7926, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138874

ABSTRACT

In prostate carcinogenesis, expression and/or activation of the Transient Receptor Potential Melastatin 8 channel (TRPM8) was shown to block in vitro Prostate Cancer (PCa) cell migration. Because of their localization at the plasma membrane, ion channels, such as TRPM8 and other membrane receptors, are promising pharmacological targets. The aim of this study was thus to use nanocarriers encapsulating a TRPM8 agonist to efficiently activate the channel and therefore arrest PCa cell migration. To achieve this goal, the most efficient TRPM8 agonist, WS12, was encapsulated into Lipid NanoCapsules (LNC). The effect of the nanocarriers on channel activity and cellular physiological processes, such as cell viability and migration, were evaluated in vitro and in vivo. These results provide a proof-of-concept support for using TRPM8 channel-targeting nanotechnologies based on LNC to develop more effective methods inhibiting PCa cell migration in zebrafish xenograft.


Subject(s)
Anilides/pharmacology , Cell Migration Inhibition/drug effects , Menthol/analogs & derivatives , Prostatic Neoplasms/drug therapy , TRPM Cation Channels/agonists , Anilides/administration & dosage , Humans , Lipids/chemistry , Male , Menthol/administration & dosage , Menthol/pharmacology , Nanocapsules/chemistry , PC-3 Cells , Prostatic Neoplasms/metabolism , TRPM Cation Channels/metabolism
2.
Am J Physiol Cell Physiol ; 310(7): C509-19, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26791491

ABSTRACT

Intracellular Ca(2+) signals play a central role in several cellular processes; therefore it is not surprising that altered Ca(2+) homeostasis regulatory mechanisms lead to a variety of severe pathologies, including cancer. Stromal interaction molecules (STIM) and ORAI proteins have been identified as critical components of Ca(2+) entry in both store-dependent (SOCE mechanism) and independent by intracellular store depletion and have been implicated in several cellular functions. In recent years, both STIMs and ORAIs have emerged as possible molecular targets for cancer therapeutics. In this review we focus on the role of STIM and ORAI proteins in cancer progression. In particular we analyze their role in the different hallmarks of cancer, which represent the organizing principle that describes the complex multistep process of neoplastic diseases.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Neoplasms/metabolism , Animals , Humans
3.
Oncogene ; 34(42): 5383-94, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-25619833

ABSTRACT

In the present study, we have assessed whether a putative calcium channel α2δ2 auxiliary subunit (CACNA2D2 gene) could be involved in prostate cancer (PCA) progression. We therefore carried out experiments to determine whether this protein is expressed in PCA LNCaP cells and in PCA tissues, and whether its expression may be altered during cancer development. In addition, we evaluated the influence on cell proliferation of overexpressing or downregulating this subunit. In vitro experiments show that α2δ2 subunit overexpression is associated with increased cell proliferation, alterations of calcium homeostasis and the recruitment of a nuclear factor of activated T-cells pathway. Furthermore, we carried out in vivo experiments on immuno-deficient nude mice in order to evaluate the tumorigenic potency of the α2δ2 subunit. We show that α2δ2-overexpressing PCA LNCaP cells are more tumorigenic than control LNCaP cells when injected into nude mice. In addition, gabapentin, a ligand of α2δ2, reduces tumor development in LNCaP xenografts. Finally, we show that the action of α2δ2 on tumor development occurs not only through a stimulation of proliferation, but also through a stimulation of angiogenesis, via an increased secretion of vascular endothelial growth factor in cells overexpressing α2δ2.


Subject(s)
Calcium Channels/physiology , Cell Proliferation , Cell Transformation, Neoplastic , Neovascularization, Pathologic/etiology , Prostatic Neoplasms/etiology , Animals , Calcium/metabolism , Calcium Channels/genetics , Cell Line, Tumor , Homeostasis , Humans , Male , Mice , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism
4.
Cell Death Dis ; 5: e1193, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24763050

ABSTRACT

The growing number of studies suggested that inhibition of autophagy enhances the efficacy of Akt kinase inhibitors in cancer therapy. Here, we provide evidence that ML-9, a widely used inhibitor of Akt kinase, myosin light-chain kinase (MLCK) and stromal interaction molecule 1 (STIM1), represents the 'two-in-one' compound that stimulates autophagosome formation (by downregulating Akt/mammalian target of rapamycin (mTOR) pathway) and inhibits their degradation (by acting like a lysosomotropic agent and increasing lysosomal pH). We show that ML-9 as a monotherapy effectively induces prostate cancer cell death associated with the accumulation of autophagic vacuoles. Further, ML-9 enhances the anticancer activity of docetaxel, suggesting its potential application as an adjuvant to existing anticancer chemotherapy. Altogether, our results revealed the complex effect of ML-9 on autophagy and indentified ML-9 as an attractive tool for targeting autophagy in cancer therapy through dual inhibition of both the Akt pathway and the autophagy.


Subject(s)
Autophagy/drug effects , Azepines/pharmacology , Lysosomes/drug effects , Prostatic Neoplasms/pathology , Calcium/metabolism , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/metabolism , Down-Regulation/drug effects , Homeostasis/drug effects , Humans , Hydrogen-Ion Concentration/drug effects , Lysosomes/ultrastructure , Male , Models, Biological , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/ultrastructure , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
J Eur Acad Dermatol Venereol ; 25 Suppl 1: 12-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21175869

ABSTRACT

BACKGROUND: Numerous studies have demonstrated the beneficial effect of Avène Thermal Spring Water (TSW) in dermatological diseases but the molecular mechanisms remain unknown. The objective of the present study was to evaluate the effect of Avène TSW on the morphological and molecular features related to the more advanced status of differentiation of human keratinocytes. MATERIAL AND METHODS: Normal human keratinocytes (NHK) were differentiated in medium powder reconstituted with Avène TSW and assessed by RT-PCR and immunohistochemistry. Calcium entry was measured by a Fura-2 AM probe. TRPV6 channel were detected by immunohistochemistry, RT-PCR and western blot. RESULTS: Treatment of NHK with Avène TSW led to an enhanced constitutive calcium entry that resulted in the increased expression of involucrin and cytokeratins 1 and 10. This enhanced constitutive calcium entry in Avène TSW-treated keratinocytes was mediated by the TRPV6 calcium channel. Moreover, Avène TSW-mediated calcium entry was due to the increase in TRPV6 expression as well as the channel abundance at the cell membrane. CONCLUSIONS: An other mechanism of action of Avène TSW is described. Avène TSW treatment induced an enhanced constitutive calcium entry mediated by TRPV6 channel leading to the acceleration of human keratinocytes differentiation.


Subject(s)
Cell Differentiation/drug effects , Keratinocytes/physiology , Mineral Waters/administration & dosage , TRPV Cation Channels/drug effects , Calcium , Calcium Signaling/drug effects , Cells, Cultured , Gene Expression , Humans , Keratin-1/genetics , Keratin-1/metabolism , Keratin-10/genetics , Keratin-10/metabolism , Membrane Transport Proteins/metabolism , Protein Precursors/metabolism , TRPV Cation Channels/metabolism
6.
Oncogene ; 29(32): 4611-6, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20531306

ABSTRACT

Although the transient receptor potential melastatin 8 (TRPM8) cold receptor is highly expressed in prostate cancer (PCa) and constitutes a promising diagnostic and prognostic indicator, the natural agonists of this channel in the prostate, as well as its physiological and pathological functions, remain unknown. In this study, we identified the well-known PCa marker, prostate-specific antigen (PSA), as a physiological TRPM8 agonist. Electrophysiological and Ca(2+) imaging studies demonstrated that PSA activated TRPM8-mediated current by the bradykinin 2 receptor signaling pathway. Further investigation of this mechanism by cell-surface biotinylation revealed that the increase in TRPM8 current induced by PSA was due to an increase in the number of functional TRPM8 channels on the plasma membrane. Importantly, wound-healing and migration assays revealed that TRPM8 activation by PSA reduced motility of the PC3 PCa cell line, suggesting that plasma membrane TRPM8 has a protective role in PCa progression. Consequently, PSA was identified as a natural TRPM8 agonist in the prostate and we propose a putative physiological role for both of these proteins in carcinogenesis, making this pathway a potentially important target for anticancer agent development.


Subject(s)
Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Prostate-Specific Antigen/pharmacology , Prostatic Neoplasms/pathology , TRPM Cation Channels/metabolism , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Kinase C/metabolism , Receptor, Bradykinin B2/metabolism , Signal Transduction/drug effects , TRPM Cation Channels/agonists
7.
Cell Death Dis ; 1: e75, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-21364678

ABSTRACT

The molecular nature of calcium (Ca(2+))-dependent mechanisms and the ion channels having a major role in the apoptosis of cancer cells remain a subject of debate. Here, we show that the recently identified Orai1 protein represents the major molecular component of endogenous store-operated Ca(2+) entry (SOCE) in human prostate cancer (PCa) cells, and constitutes the principal source of Ca(2+) influx used by the cell to trigger apoptosis. The downregulation of Orai1, and consequently SOCE, protects the cells from diverse apoptosis-inducing pathways, such as those induced by thapsigargin (Tg), tumor necrosis factor α, and cisplatin/oxaliplatin. The transfection of functional Orai1 mutants, such as R91W, a selectivity mutant, and L273S, a coiled-coil mutant, into the cells significantly decreased both SOCE and the rate of Tg-induced apoptosis. This suggests that the functional coupling of STIM1 to Orai1, as well as Orai1 Ca(2+)-selectivity as a channel, is required for its pro-apoptotic effects. We have also shown that the apoptosis resistance of androgen-independent PCa cells is associated with the downregulation of Orai1 expression as well as SOCE. Orai1 rescue, following Orai1 transfection of steroid-deprived cells, re-established the store-operated channel current and restored the normal rate of apoptosis. Thus, Orai1 has a pivotal role in the triggering of apoptosis, irrespective of apoptosis-inducing stimuli, and in the establishment of an apoptosis-resistant phenotype in PCa cells.


Subject(s)
Apoptosis , Calcium Channels/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Substitution , Antineoplastic Agents/therapeutic use , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/physiology , Cell Line, Tumor , Cisplatin/therapeutic use , Humans , Male , Membrane Proteins/metabolism , Mutation , Neoplasm Proteins/metabolism , ORAI1 Protein , Phenotype , Prostatic Neoplasms/drug therapy , Stromal Interaction Molecule 1 , Thapsigargin/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use
8.
Oncogene ; 28(15): 1792-806, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19270724

ABSTRACT

Accumulating data point to K(+) channels as relevant players in controlling cell cycle progression and proliferation of human cancer cells, including prostate cancer (PCa) cells. However, the mechanism(s) by which K(+) channels control PCa cell proliferation remain illusive. In this study, using the techniques of molecular biology, biochemistry, electrophysiology and calcium imaging, we studied the expression and functionality of intermediate-conductance calcium-activated potassium channels (IK(Ca1)) in human PCa as well as their involvement in cell proliferation. We showed that IK(Ca1) mRNA and protein were preferentially expressed in human PCa tissues, and inhibition of the IK(Ca1) potassium channel suppressed PCa cell proliferation. The activation of IK(Ca1) hyperpolarizes membrane potential and, by promoting the driving force for calcium, induces calcium entry through TRPV6, a cation channel of the TRP (Transient Receptor Potential) family. Thus, the overexpression of the IK(Ca1) channel is likely to promote carcinogenesis in human prostate tissue.


Subject(s)
Calcium/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Prostatic Neoplasms/pathology , Benzimidazoles/pharmacology , Calcium Channels/physiology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27 , G1 Phase , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/analysis , Intracellular Signaling Peptides and Proteins/analysis , Male , Membrane Potentials , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , S100 Proteins/analysis , TRPV Cation Channels/physiology , Tumor Suppressor Protein p53/physiology
9.
Oncogene ; 26(52): 7380-5, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17533368

ABSTRACT

The transient receptor potential channel, subfamily V, member 6 (TRPV6), is strongly expressed in advanced prostate cancer and significantly correlates with the Gleason >7 grading, being undetectable in healthy and benign prostate tissues. However, the role of TRPV6 as a highly Ca(2+)-selective channel in prostate carcinogenesis remains poorly understood. Here, we report that TRPV6 is directly involved in the control of prostate cancer cell (LNCaP cell line) proliferation by decreasing: (i) proliferation rate; (ii) cell accumulation in the S-phase of cell cycle and (iii) proliferating cell nuclear antigen (PCNA) expression. We demonstrate that the Ca(2+) uptake into LNCaP cells is mediated by TRPV6, with the subsequent downstream activation of the nuclear factor of activated T-cell transcription factor (NFAT). TRPV6-mediated Ca(2+) entry is also involved in apoptosis resistance of LNCaP cells. Our results suggest that TRPV6 expression in LNCaP cells is regulated by androgen receptor, however, in a ligand-independent manner. We conclude that the upregulation of TRPV6 Ca(2+) channel in prostate cancer cells may represent a mechanism for maintaining a higher proliferation rate, increasing cell survival and apoptosis resistance as well.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cell Proliferation , NFATC Transcription Factors/metabolism , Prostatic Neoplasms/metabolism , TRPV Cation Channels/metabolism , Apoptosis , Calcium Channels/genetics , Calcium Signaling , Enzyme Inhibitors/pharmacology , Humans , Male , Prostatic Neoplasms/genetics , S Phase/physiology , Signal Transduction , TRPV Cation Channels/genetics , Thapsigargin/pharmacology , Tumor Cells, Cultured
10.
Cell Death Differ ; 14(7): 1295-304, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17479110

ABSTRACT

Plasma membrane ion channels contribute to virtually all basic cellular processes, including such crucial ones for maintaining tissue homeostasis as proliferation, differentiation, and apoptosis. Enhanced proliferation, aberrant differentiation, and impaired ability to die are the prime reasons for abnormal tissue growth, which can eventually turn into uncontrolled expansion and invasion, characteristic of cancer. Prostate cancer (PCa) cells express a variety of plasma membrane ion channels. By providing the influx of essential signaling ions, perturbing intracellular ion concentrations, regulating cell volume, and maintaining membrane potential, PCa cells are critically involved in proliferation, differentiation, and apoptosis. PCa cells of varying metastatic ability can be distinguished by their ion channel characteristics. Increased malignancy and invasiveness of androgen-independent PCa cells is generally associated with the shift to a 'more excitable' phenotype of their plasma membrane. This shift is manifested by the appearance of voltage-gated Na(+) and Ca(2+) channels which contribute to their enhanced apoptotic resistance together with downregulated store-operated Ca(2+) influx, altered expression of different K(+) channels and members of the Transient Receptor Potential (TRP) channel family, and strengthened capability for maintaining volume constancy. The present review examines channel types expressed by PCa cells and their involvement in metastatic behaviors.


Subject(s)
Carcinoma/metabolism , Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Ion Channels/metabolism , Prostatic Neoplasms/metabolism , Animals , Carcinoma/physiopathology , Cell Death/physiology , Cell Survival/physiology , Humans , Ions/metabolism , Male , Neoplasm Invasiveness/physiopathology , Prostatic Neoplasms/physiopathology
11.
Biochem Soc Trans ; 35(Pt 1): 133-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17233619

ABSTRACT

A major clinical problem with PC (prostate cancer) is the cell's ability to survive and proliferate upon androgen withdrawal. Indeed, deregulated cell differentiation and proliferation, together with the suppression of apoptosis, provides the condition for abnormal tissue growth. Here, we examine the differential role of TRP (transient receptor potential) channels in the control of Ca(2+) homoeostasis and growth of PC cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Transient Receptor Potential Channels/physiology , Cell Differentiation , Cell Proliferation , Endoplasmic Reticulum/metabolism , Humans , Male , Models, Biological , RNA, Small Interfering/metabolism , TRPM Cation Channels/metabolism , Transient Receptor Potential Channels/chemistry
12.
Subcell Biochem ; 45: 405-27, 2007.
Article in English | MEDLINE | ID: mdl-18193646

ABSTRACT

Cancer is caused by defects in the mechanisms underlying cell proliferation and cell death. Calcium ions are central to both phenomena, serving as major signalling agents with spatial localization, magnitude and temporal characteristics of calcium signals ultimately determining cell's fate. There are four primary compartments: extracellular space, cytoplasm, endoplasmic reticulum and mitochondria that participate in the cellular Ca2+ circulation. They are separated by own membranes incorporating divers Ca2(+)-handling proteins whose concerted action provides for Ca2+ signals with the spatial and temporal characteristics necessary to account for specific cellular response. The transformation of a normal cell into a cancer cell is associated with a major re-arrangement of Ca2+ pumps, Na/Ca exchangers and Ca2+ channels, which leads to the enhanced proliferation and impaired ability to die. In the present chapter we examine what changes in Ca+ signalling and the mechanisms that support it underlie the passage from normal to pathological cell growth and death control. Understanding this changes and identifying molecular players involved provides new prospects for cancers treatment.


Subject(s)
Calcium Signaling/physiology , Cell Proliferation , Neoplasms/pathology , Animals , Apoptosis/physiology , Calcium-Transporting ATPases/physiology , Cell Cycle/drug effects , Cytosol/physiology , Endoplasmic Reticulum/physiology , Humans , Mitochondria/physiology , Neoplasms/physiopathology
13.
Endocr Relat Cancer ; 13(1): 181-95, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16601287

ABSTRACT

Neuroendocrine differentiation (NED) has been implicated in prostate cancer progression and hormone-therapy failure. Neuroendocrine cells are non-proliferating and escape apoptotic cell death, although their origin and the causes of their apoptotic resistance have as yet been poorly elucidated. This study demonstrates a new mechanism involved in controlling NED. We report that epidermal growth factor (5-50 ng/ml) promotes neuroendocrine-like differentiation of androgen-independent DU145 prostate cancer cells. This differentiation is associated with an increase in the expression of Neuron Specific Enolase (NSE) and a reduction in cell proliferation and is blocked by inhibiting tyrosine kinase activity with genistein and with compound 56 (C56). An increase in the cAMP level, using dibutryl cAMP (db-cAMP) (1 mM) and isobutylmethylxanthine (100 microM), does not promote NED by itself, but does increase the effect of EGF on NED. In addition, EGF-induced NED protects cells from apoptosis induced with thapsigargin (1 microM) by reducing the thapsigargin-induced cytosolic calcium overload. In order to describe how EGF-induced NED protects cells against thapigargin-induced calcium overload we investigated the spatiotemporal calcium signalling linked to apoptosis. By using thapsigargin in various conditions on DU145 cells and using micro-fluorimetric calcium measurements, we show that depletion of intracellular calcium store induces apoptosis and that the amplitude and duration of the capacitive calcium entry are two apoptosis-modulating parameters. We show that protection against thapsigargin-induced apoptosis conferred by NED is achieved by reducing the amount and the speed of calcium that can be released from calcium pools, as well as modulating the amplitude of the subsequent calcium entry.


Subject(s)
Androgens/metabolism , Apoptosis/drug effects , Cell Differentiation/drug effects , Epidermal Growth Factor/pharmacology , Neoplasms, Hormone-Dependent/pathology , Neurosecretory Systems/drug effects , Prostatic Neoplasms/pathology , Antineoplastic Agents/pharmacology , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Humans , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Phosphopyruvate Hydratase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Thapsigargin/pharmacology
14.
Endocr Relat Cancer ; 12(2): 335-49, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15947107

ABSTRACT

Neuroendocrine (NE) differentiation of prostate epithelial/basal cells is a hallmark of advanced, androgen-independent prostate cancer, for which there is no successful therapy. Here we report for the first time on alterations in regulatory volume decrease (RVD) and its key determinant, swelling-activated Cl- current (I(Cl,swell)), associated with NE differentiation of androgen-dependent LNCaP prostate cancer epithelial cells. NE-differentiating regimens, namely, chronic cAMP elevation or androgen deprivation, resulted in generally augmented I(Cl,swell) and enhanced RVD. This occurred as a result of both the increased endogenous expression of ClC-3, which is a volume-sensitive Cl- channel involved, as we show, in I(Cl,swell) in LNCaP (lymph-node carcinoma of the prostate) cells and the weaker negative I(Cl,swell) control from Ca2+ entering via store-dependent pathways. The changes in the RVD of NE-differentiated cells generally mimicked those reported for Bcl-2-conferred apoptotic resistance. Our results suggest that strengthening the mechanism that helps to maintain volume constancy may contribute to better survival rates of apoptosis-resistant NE cells.


Subject(s)
Androgens/physiology , Chloride Channels/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Apoptosis , Calcium/metabolism , Cell Differentiation , Cell Size , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Patch-Clamp Techniques , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation
15.
Endocr Relat Cancer ; 12(2): 367-82, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15947109

ABSTRACT

TRPM8 (melastatine-related transient receptor potential member 8), a member of the transient receptor potential (TRP) superfamily of cation channels, has been shown to be a calcium-channel protein. TRPM8 mRNA has also been shown to be overexpressed in prostate cancer and is considered to play an important role in prostate physiology. This study was designed to determine the androgen-regulation mechanisms for TRPM8 mRNA expression and to identify the phenotype of TRPM8-expressing cells in the human prostate. Our findings show that trpm8 gene expression requires a functional androgen receptor. Furthermore, this article argues strongly in favour of the fact that the trpm8 gene is a primary androgen-responsive gene. Single-cell reverse transcriptase PCR and immunohistochemical experiments also showed that the trpm8 gene was mainly expressed in the apical secretory epithelial cells of the human prostate and trpm8 down-regulation occurred during the loss of the apical differentiated phenotype of the primary cultured human prostate epithelial cells. The androgen-regulated trpm8 expression mechanisms are important in understanding the progression of prostate cancer to androgen-independence. These findings may contribute to design a strategy to predict prostate cancer status from the TRPM8 mRNA level. Furthermore, as the TRPM8 channel is localized in human prostate cells, it will be interesting to understand its physiological function in the normal prostate and its potential role in prostate cancer development.


Subject(s)
Gene Expression Regulation, Neoplastic , Ion Channels/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/physiology , 5-alpha-Dihydroprogesterone/metabolism , 5-alpha-Dihydroprogesterone/pharmacology , Androgens/metabolism , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Humans , Ion Channels/metabolism , Male , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Response Elements , TRPM Cation Channels , Tumor Cells, Cultured
16.
FEBS Lett ; 556(1-3): 121-6, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14706838

ABSTRACT

It has previously been suggested that volume-regulated anion channels (VRACs) and store-operated channels (SOCs) interact with each other according to their expected colocalization in the plasma membrane of LNCaP cells. In order to study interactions between these two channels, we used 2-aminoethoxydiphenyl borate (2-APB) as a regular SOC inhibitor. Surprisingly 2-APB reduced VRAC activity in a dose-dependent manner (IC(50)=122.8 microM), but not 2,2-diphenyltetrahydrofuran (a structural analog of 2-APB). This effect was also present in keratinocytes. We conclude that 2-APB is an inhibitor of the VRAC family, and is also a potent tool to study the SOC-VRAC interaction in LNCaP cells.


Subject(s)
Boron Compounds/pharmacology , Calcium Signaling/drug effects , Chloride Channels/antagonists & inhibitors , Egtazic Acid/analogs & derivatives , Calcium/metabolism , Calcium Signaling/physiology , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Furans/chemistry , Furans/pharmacology , Humans , Hypotonic Solutions/pharmacology , Inhibitory Concentration 50 , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Prostatic Neoplasms/metabolism
17.
Cell Death Differ ; 11(3): 321-30, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14685164

ABSTRACT

Neuroendocrine (NE) differentiation is a hallmark of advanced, androgen-independent prostate cancer, for which there is no successful therapy. NE tumor cells are nonproliferating and escape apoptotic cell death; therefore, an understanding of the apoptotic status of the NE phenotype is imperative for the development of new therapies for prostate cancer. Here, we report for the first time on alterations in intracellular Ca(2+) homeostasis, which is a key factor in apoptosis, caused by NE differentiation of androgen-dependent prostate cancer epithelial cells. NE-differentiating regimens, either cAMP elevation or androgen deprivation, resulted in a reduced endoplasmic reticulum Ca(2+)-store content due to both SERCA 2b Ca(2+) ATPase and luminal Ca(2+) binding/storage chaperone calreticulin underexpression, and to a downregulated store-operated Ca(2+) current. NE-differentiated cells showed enhanced resistance to thapsigargin- and TNF-alpha-induced apoptosis, unrelated to antiapoptotic Bcl-2 protein overexpression. Our results suggest that targeting the key players determining Ca(2+) homeostasis in an attempt to enhance the proapoptotic potential of malignant cells may prove to be a useful strategy in the treatment of advanced prostate cancer.


Subject(s)
Apoptosis/drug effects , Calcium/metabolism , Cell Differentiation , Homeostasis , Neurosecretory Systems , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Blotting, Western , Calcium Channels/metabolism , Calcium-Transporting ATPases/metabolism , Calreticulin/metabolism , Cell Line, Tumor , Electric Capacitance , Electric Impedance , Electrophysiology , Endoplasmic Reticulum/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fluorescent Dyes , Fura-2 , Humans , Kinetics , Male , Models, Biological , Patch-Clamp Techniques , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Thapsigargin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
18.
Cell Calcium ; 33(5-6): 357-73, 2003.
Article in English | MEDLINE | ID: mdl-12765682

ABSTRACT

Ca2+ homeostasis mechanisms, in which the Ca2+ entry pathways play a key role, are critically involved in both normal function and cancerous transformation of prostate epithelial cells. Here, using the lymph node carcinoma of the prostate (LNCaP) cell line as a major experimental model, we characterize prostate-specific store-operated Ca2+ channels (SOCs)--a primary Ca2+ entry pathway for non-excitable cells--for the first time. We show that prostate-specific SOCs share major store-dependent, kinetic, permeation, inwardly rectifying, and pharmacological (including dual, potentiation/inhibition concentration-dependent sensitivity to 2-APB) properties with "classical" Ca2+ release-activated Ca2+ channels (CRAC), but have a higher single channel conductance (3.2 and 12pS in Ca2+- and Na+-permeable modes, respectively). They are subject to feedback inhibition via Ca2+-dependent PKC, CaMK-II and CaM regulatory pathways and are functionally dependent on caveolae integrity. Caveolae also provide a scaffold for spatial co-localization of SOCs with volume-regulated anion channels (VRAC) and their Ca2+-mediated interaction. The TRPC1 and TRPV6 members of the transient receptor potential (TRP) channel family are the most likely molecular candidates for the formation of prostate-specific endogenous SOCs. Differentiation of LNCaP cells to an androgen-insensitive, apoptotic-resistant neuroendocrine phenotype downregulates SOC current. We conclude that prostate-specific SOCs are important determinants in the transition to androgen-independent prostate cancer.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Epithelial Cells/metabolism , Prostatic Neoplasms/metabolism , Biomarkers , Calcium Channels/genetics , Electrophysiology , Endoplasmic Reticulum/metabolism , Epithelial Cells/pathology , Humans , Kinetics , Male , Oligonucleotides, Antisense/pharmacology , Prostatic Neoplasms/pathology , RNA, Messenger/drug effects , TRPC Cation Channels , TRPV Cation Channels , Tumor Cells, Cultured
19.
Cell Calcium ; 34(1): 75-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12767895

ABSTRACT

This study investigates the calcium mechanisms involved in growth arrest induced by extracellular ATP in DU-145 androgen-independent human prostate cancer cells. Exposure of DU-145 cells to 100 microM ATP produced an increase in cytoplasmic calcium concentration ([Ca(2+)](i)), due to a mobilization of calcium from the endoplasmic reticulum stores and to subsequent capacitative calcium entry (CCE). We have shown that this [Ca(2+)](i) increase occurs after stimulation by ATP of the phospholipase C (PLC) pathway. For the first time, we have identified the inositol 1,4,5-trisphosphate receptor (IP(3)R) isoforms expressed in this cell line and have demonstrated a participation of protein kinase C in CCE. Using fluorescence imaging, we have shown that a long-term treatment with ATP leads to a decrease in the intraluminal endoplasmic reticulum calcium concentration as well as in the amount of releasable Ca(2+). Modulating extracellular free calcium concentrations indicated that variations in [Ca(2+)](i) did not affect the ATP-induced growth arrest of DU-145 cells. However, treating cells with 1 nM thapsigargin (TG) to deplete intracellular calcium pools prevented the growth arrest induced by ATP. Altogether, these results indicate that growth arrest induced in DU-145 cells by extracellular ATP is not correlated with an increase in [Ca(2+)](i) but rather with a decrease in intracellular calcium pool content.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Carcinoma/metabolism , Prostatic Neoplasms/metabolism , Adenosine Triphosphate/pharmacology , Calcium/pharmacology , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Carcinoma/physiopathology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor , Cytoplasm/drug effects , Cytoplasm/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Male , Prostatic Neoplasms/physiopathology , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Thapsigargin/pharmacology , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism
20.
J Physiol ; 548(Pt 3): 823-36, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12724346

ABSTRACT

Although the prostate gland is a rich source of alpha1-adreno- (alpha1-AR) and m1-cholino receptors (m1-AChR), the membrane processes associated with their activation in glandular epithelial cells is poorly understood. We used the whole-cell patch-clamp technique to show that the agonists of the respective receptors, phenylephrine (PHE) and carbachol (CCh), activate cationic membrane currents in lymph node carcinoma of the prostate (LNCaP) human prostate cancer epithelial cells, which are not dependent on the filling status of intracellular IP3-sensitive Ca2+ stores, but directly gated by diacylglycerol (DAG), as evidenced by the ability of its membrane permeable analogue, OAG, to mimic the effects of the agonists. The underlying cationic channels are characterized by the weak field-strength Eisenman IV permeability sequence for monovalent cations (PK(25) > PCs(4.6) > PLi(1.4) > PNa(1.0)), and the following permeability sequence for divalent cations: PCa(1.0) > PMg(0.74) > PBa(0.6) > PSr(0.36) > PMn(0.3). They are 4.3 times more permeable to Ca2+ than Na+ and more sensitive to the inhibitor 2-APB than SK&F 96365. RT-PCR analysis shows that DAG-gated members of the transient receptor potential (TRP) channel family, including TRPC1 and TRPC3, are present in LNCaP cells. We conclude that, in prostate cancer epithelial cells, alpha1-ARs and m1-AChRs are functionally coupled to Ca2+-permeable DAG-gated cationic channels, for which TRPC1 and TRPC3 are the most likely candidates.


Subject(s)
Carbachol/pharmacology , Ion Channels/physiology , Phenylephrine/pharmacology , Base Sequence , Calcium Channels/physiology , DNA Primers , Electrophysiology/methods , Evoked Potentials/drug effects , Evoked Potentials/physiology , Humans , Ion Channels/genetics , Male , Potassium Channels/drug effects , Potassium Channels/physiology , Prostatic Neoplasms , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/physiology , Receptors, Muscarinic/physiology , Reverse Transcriptase Polymerase Chain Reaction , Ruthenium Red/pharmacology , TRPC Cation Channels , Tetraethylammonium/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...