Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Environ Health Perspect ; 124(4): 452-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26359731

ABSTRACT

BACKGROUND: There are concerns that diminished prostaglandin action in fetal life could increase the risk of congenital malformations. Many endocrine-disrupting chemicals have been found to suppress prostaglandin synthesis, but to our knowledge, pesticides have never been tested for these effects. OBJECTIVES: We assessed the ability of pesticides that are commonly used in the European Union to suppress prostaglandin D2 (PGD2) synthesis. METHODS: Changes in PGD2 secretion in juvenile mouse Sertoli cells (SC5 cells) were measured using an ELISA. Coincubation with arachidonic acid (AA) was conducted to determine the site of action in the PGD2 synthetic pathway. Molecular modeling studies were performed to assess whether pesticides identified as PGD2-active could serve as ligands of the cyclooxygenase-2 (COX-2) binding pocket. RESULTS: The pesticides boscalid, chlorpropham, cypermethrin, cyprodinil, fenhexamid, fludioxonil, imazalil (enilconazole), imidacloprid, iprodione, linuron, methiocarb, o-phenylphenol, pirimiphos-methyl, pyrimethanil, and tebuconazole suppressed PGD2 production. Strikingly, some of these substances-o-phenylphenol, cypermethrin, cyprodinil, linuron, and imazalil (enilconazole)-showed potencies (IC50) in the range between 175 and 1,500 nM, similar to those of analgesics intended to block COX enzymes. Supplementation with AA failed to reverse this effect, suggesting that the sites of action of these pesticides are COX enzymes. The molecular modeling studies revealed that the COX-2 binding pocket can accommodate most of the pesticides shown to suppress PGD2 synthesis. Some of these pesticides are also capable of antagonizing the androgen receptor. CONCLUSIONS: Chemicals with structural features more varied than previously thought can suppress PGD2 synthesis. Our findings signal a need for in vivo studies to establish the extent of endocrine-disrupting effects that might arise from simultaneous interference with PGD2 signaling and androgen action. CITATION: Kugathas S, Audouze K, Ermler S, Orton F, Rosivatz E, Scholze M, Kortenkamp A. 2016. Effects of common pesticides on prostaglandin D2 (PGD2) inhibition in SC5 mouse Sertoli cells, evidence of binding at the COX-2 active site, and implications for endocrine disruption. Environ Health Perspect 124:452-459; http://dx.doi.org/10.1289/ehp.1409544.


Subject(s)
Cyclooxygenase 2/metabolism , Endocrine Disruptors/toxicity , Pesticides/toxicity , Prostaglandin D2/antagonists & inhibitors , Sertoli Cells/drug effects , Androgen Receptor Antagonists , Animals , Arachidonic Acid/metabolism , Catalytic Domain , Male , Mice , Models, Molecular , Prostaglandin D2/metabolism , Protein Binding , Sertoli Cells/metabolism
2.
Toxicol Appl Pharmacol ; 278(3): 201-8, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24055644

ABSTRACT

Many xenobiotics have been identified as in vitro androgen receptor (AR) antagonists, but information about their ability to produce combined effects at low concentrations is missing. Such data can reveal whether joint effects at the receptor are induced at low levels and may support the prioritisation of in vivo evaluations and provide orientations for the grouping of anti-androgens in cumulative risk assessment. Combinations of 30 AR antagonists from a wide range of sources and exposure routes (pesticides, antioxidants, parabens, UV-filters, synthetic musks, bisphenol-A, benzo(a)pyrene, perfluorooctane sulfonate and pentabromodiphenyl ether) were tested using a reporter gene assay (MDA-kb2). Chemicals were combined at three mixture ratios, equivalent to single components' effect concentrations that inhibit the action of dihydrotesterone by 1%, 10% or 20%. Concentration addition (CA) and independent action were used to calculate additivity expectations. We observed complete suppression of dihydrotestosterone effects when chemicals were combined at individual concentrations eliciting 1%, 10% or 20% AR antagonistic effect. Due to the large number of mixture components, the combined AR antagonistic effects occurred at very low concentrations of individual mixture components. CA slightly underestimated the combined effects at all mixture ratios. In conclusion, large numbers of AR antagonists from a wide variety of sources and exposure routes have the ability of acting together at the receptor to produce joint effects at very low concentrations. Significant mixture effects are observed when chemicals are combined at concentrations that individually do not induce observable AR antagonistic effects. Cumulative risk assessment for AR antagonists should apply grouping criteria based on effects where data are available, rather than on criteria of chemical similarity.


Subject(s)
Antioxidants/toxicity , Drug Interactions , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Models, Biological , Nonsteroidal Anti-Androgens/toxicity , Pesticides/toxicity , Androgens/chemistry , Androgens/pharmacology , Cell Line, Tumor , Consumer Product Safety , Dihydrotestosterone/antagonists & inhibitors , Dihydrotestosterone/pharmacology , Genes, Reporter/drug effects , Humans , Industrial Waste/adverse effects , Osmolar Concentration , Promoter Regions, Genetic/drug effects , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Recombinant Fusion Proteins/metabolism , Response Elements/drug effects , Risk Assessment/methods
3.
Environ Health Perspect ; 120(11): 1578-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23008280

ABSTRACT

BACKGROUND: Many pesticides in current use have recently been revealed as in vitro androgen receptor (AR) antagonists, but information about their combined effects is lacking. OBJECTIVE: We investigated the combined effects and the competitive AR antagonism of pesticide mixtures. METHODS: We used the MDA-kb2 assay to test a combination of eight AR antagonists that did not also possess AR agonist properties ("pure" antagonists; 8 mix: fludioxonil, fenhexamid, ortho-phenylphenol, imazalil, tebuconazole, dimethomorph, methiocarb, pirimiphos-methyl), a combination of five AR antagonists that also showed agonist activity (5 mix: cyprodinil, pyrimethanil, vinclozolin, chlorpropham, linuron), and all pesticides combined (13 mix). We used concentration addition (CA) and independent action (IA) to formulate additivity expectations, and Schild plot analyses to investigate competitive AR antagonism. RESULTS: A good agreement between the effects of the mixture of eight "pure" AR antagonists and the responses predicted by CA was observed. Schild plot analysis revealed that the 8 mix acted by competitive AR antagonism. However, the observed responses of the 5 mix and the 13 mix fell within the "prediction window" boundaries defined by the predicted regression curves of CA and IA. Schild plot analysis with these mixtures yielded anomalous responses incompatible with competitive receptor antagonism. CONCLUSIONS: A mixture of widely used pesticides can, in a predictable manner, produce combined AR antagonist effects that exceed the responses elicited by the most potent component alone. Inasmuch as large populations are regularly exposed to mixtures of antiandrogenic pesticides, our results underline the need for considering combination effects for these substances in regulatory practice.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Dihydrotestosterone/antagonists & inhibitors , Pesticides/pharmacology , Receptors, Androgen/metabolism , Risk Assessment/methods , Androgen Receptor Antagonists/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Pesticides/chemistry
5.
Environ Health Perspect ; 119(6): 794-800, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21310686

ABSTRACT

BACKGROUND: Evidence suggests that there is widespread decline in male reproductive health and that antiandrogenic pollutants may play a significant role. There is also a clear disparity between pesticide exposure and data on endocrine disruption, with most of the published literature focused on pesticides that are no longer registered for use in developed countries. OBJECTIVE: We used estimated human exposure data to select pesticides to test for antiandrogenic activity, focusing on highest use pesticides. METHODS: We used European databases to select 134 candidate pesticides based on highest exposure, followed by a filtering step according to known or predicted receptor-mediated antiandrogenic potency, based on a previously published quantitative structure-activity relationship (QSAR) model. In total, 37 pesticides were tested for in vitro androgen receptor (AR) antagonism. Of these, 14 were previously reported to be AR antagonists ("active"), 4 were predicted AR antagonists using the QSAR, 6 were predicted to not be AR antagonists ("inactive"), and 13 had unknown activity, which were "out of domain" and therefore could not be classified with the QSAR ("unknown"). RESULTS: All 14 pesticides with previous evidence of AR antagonism were confirmed as antiandrogenic in our assay, and 9 previously untested pesticides were identified as antiandrogenic (dimethomorph, fenhexamid, quinoxyfen, cyprodinil, λ-cyhalothrin, pyrimethanil, fludioxonil, azinphos-methyl, pirimiphos-methyl). In addition, we classified 7 compounds as androgenic. CONCLUSIONS: Due to estimated antiandrogenic potency, current use, estimated exposure, and lack of previous data, we strongly recommend that dimethomorph, fludioxonil, fenhexamid, imazalil, ortho-phenylphenol, and pirimiphos-methyl be tested for antiandrogenic effects in vivo. The lack of human biomonitoring data for environmentally relevant pesticides presents a barrier to current risk assessment of pesticides on humans.


Subject(s)
Androgen Antagonists/pharmacology , Pesticides/pharmacology , Androgen Antagonists/classification , Androgens/classification , Androgens/pharmacology , Cell Line, Tumor , Environmental Monitoring , Europe , Humans , Male , Pesticides/classification , Quantitative Structure-Activity Relationship , Tumor Stem Cell Assay , Yeasts
6.
Cell Signal ; 23(2): 478-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21044681

ABSTRACT

Mitochondria are central players in programmed cell death and autophagy. While phosphoinositides are well established regulators of membrane traffic, cellular signalling and the destiny of certain organelles, their presence and role for mitochondria remain elusive. In this study we show that removal of PtdIns(4,5)P2 by phosphatases or masking the lipid with PH domains leads to fission of mitochondria and increased autophagy. Induction of general autophagy by amino acid starvation also coincides with the loss of mitochondrial PtdIns(4,5)P2, suggesting an important role for this lipid in the processes that govern mitophagy. Our findings reveal that PKCα can rescue the removal or masking of PtdIns(4,5)P2, indicating that the inositol lipid is upstream of PKC.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Phosphatidylinositol 4,5-Diphosphate/physiology , 3T3 Cells , Animals , Apoptosis , Autophagy , Glucagon/pharmacology , Hep G2 Cells , Humans , Mice , Mitochondria/ultrastructure , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/physiology , Tumor Cells, Cultured
7.
J Clin Invest ; 120(3): 681-93, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20197621

ABSTRACT

Irreversible cell growth arrest, a process termed cellular senescence, is emerging as an intrinsic tumor suppressive mechanism. Oncogene-induced senescence is thought to be invariably preceded by hyperproliferation, aberrant replication, and activation of a DNA damage checkpoint response (DDR), rendering therapeutic enhancement of this process unsuitable for cancer treatment. We previously demonstrated in a mouse model of prostate cancer that inactivation of the tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (Pten) elicits a senescence response that opposes tumorigenesis. Here, we show that Pten-loss-induced cellular senescence (PICS) represents a senescence response that is distinct from oncogene-induced senescence and can be targeted for cancer therapy. Using mouse embryonic fibroblasts, we determined that PICS occurs rapidly after Pten inactivation, in the absence of cellular proliferation and DDR. Further, we found that PICS is associated with enhanced p53 translation. Consistent with these data, we showed that in mice p53-stabilizing drugs potentiated PICS and its tumor suppressive potential. Importantly, we demonstrated that pharmacological inhibition of PTEN drives senescence and inhibits tumorigenesis in vivo in a human xenograft model of prostate cancer. Taken together, our data identify a type of cellular senescence that can be triggered in nonproliferating cells in the absence of DNA damage, which we believe will be useful for developing a "pro-senescence" approach for cancer prevention and therapy.


Subject(s)
Cell Proliferation , Cellular Senescence , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Animals , Cell Line, Tumor , DNA Damage/genetics , Disease Models, Animal , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Knockout , Neoplasm Transplantation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Protein Biosynthesis/genetics , Transplantation, Heterologous
8.
FEBS J ; 276(15): 4037-50, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19549188

ABSTRACT

Screening a compound library of compound 48/80 analogues, we identified 2-[5-(2-chloroethyl)-2-acetoxy-benzyl]-4-(2-chloroethyl)-phenyl acetate (E1) as a novel inhibitor of the phosphoinositide 3-kinase/Akt pathway. In order to determine the mechanism of action of E1, we analysed the effect of E1 on components of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway. E1 demonstrated dose-dependent and time-dependent repression of Akt and mTOR activity in prostate and breast cancer cell lines, PC-3 and MCF-7, respectively. Inhibition of Akt and mTOR activity by E1 also coincided with increased c-Jun NH2-terminal kinase (JNK) phosphorylation. However, the mode of action of E1 is different from that of the mTOR inhibitor rapamycin. Proliferation and cell cycle analysis revealed that E1 induced cell cycle arrest and cell death in PC-3 and MCF-7 cells. Moreover, pretreatment of cancer cells with the JNK inhibitor SP600125 abolished the repression of Akt and mTOR activity by E1, indicating that the inhibition of Akt and mTOR by E1 is mediated through JNK activation. Consistently, E1 repressed Akt and mTOR activity in wild-type and p38-null mouse embryonic fibroblasts (MEFs), but not in MEFs lacking JNK1/2, and JNK-null MEFs were less sensitive to the antiproliferative effects of E1. We further showed that E1 can function cooperatively with suboptimal concentrations of paclitaxel to induce cell death in PC-3 and MCF-7 cells. Taken together, these data suggest that E1 induces cancer cell death through the JNK-dependent repression of Akt and mTOR activity and may provide a valuable compound for further development and research.


Subject(s)
MAP Kinase Kinase 4/metabolism , Phenylacetates/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Vinyl Chloride/pharmacology , Animals , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/physiology , Humans , Mice , Paclitaxel/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
9.
Cell Signal ; 21(5): 745-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19168130

ABSTRACT

The protein kinase C (PKC) family is the most prominent target of tumor-promoting phorbol esters. For the PKCepsilon isozyme, different intracellular localizations and oncogenic potential in several but not all experimental systems have been reported. To obtain information about PKCepsilon-signaling, we investigated the effects of constitutively active rat PKCepsilon (PKCepsilonA/E, alanine 159 is replaced by glutamic acid) in HeLa cells in a doxycycline-inducible vector. Upon induction of PKCepsilonA/E expression by doxycycline, the major part of PKCepsilonA/E was localized to the Golgi. This led (i) to phosphorylations of PKCepsilon(S729), Elk-1(S383), PDK1(S241) and Rb(S807/S811), (ii) to elevated expression of receptor of activated C kinase 2 (RACK2) after 12 h, and (iii) increased colony formation in soft agar, increased cell migration and invasion, but not to decreased doubling time. Following induction of PKCepsilonA/E-expression by doxycycline for 24 h and additional short-term treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA), PKCepsilonA/E translocated to the plasma membrane and increased phosphorylation of MARCKS(S152/156). Treatment with doxycycline/TPA or TPA alone increased phosphorylations of Elk-1(S383), PDK1(S241), Rb(S807/S811), PKCdelta(T505), p38MAPK(T180/Y182), MEK1/2(S217/S221) and ERK2(T185/T187). MARCKS was not phosphorylated after treatment with TPA alone, demonstrating that in this system it is phosphorylated only by PKCepsilon localized to the plasma membrane but not by PKCalpha or delta, the other TPA-responsive PKC isozymes in HeLa cells. These results demonstrate that PKCepsilon can induce distinctly different signaling from the Golgi and from the plasma membrane.


Subject(s)
Protein Kinase C-epsilon/metabolism , Signal Transduction , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Movement/drug effects , Doxycycline/pharmacology , HeLa Cells , Humans , Phosphorylation , Rats , Tetradecanoylphorbol Acetate/pharmacology
10.
Methods Mol Biol ; 462: 135-44, 2009.
Article in English | MEDLINE | ID: mdl-19160665

ABSTRACT

This chapter describes a method for the preparation of giant unilamellar vesicles containing phosphatidylinositol 4,5-bisphosphate that are larger than 20 microm in size. The phospholipids composition of the vesicular membrane is such that fluid lamellar and liquid-ordered or gel phases are formed and separate within the confines of one vesicle. It outlines the preparation of a protein fluorescent label, pleckstrin homology domain from phospholipase C-delta 1, that binds specifically to phosphatidylinositol 4,5-bisphosphate. Using fluorescence microscopy, the presence and spatial position of this phosphorylated phosphatidylinositol lipid on the lipid membrane have been located with the pleckstrin homology domain. We show that phosphatidylinositol 4,5-bisphosphate and the phospholipase C-delta 1 pleckstrin homology domain are located to the fluid phase of the vesicle membrane. This approach can therefore show how membrane physical properties can affect enzyme binding to phosphatidylinositol 4,5-bisphosphate and thus further the understanding of important membrane processes such as endocytosis.


Subject(s)
Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Phosphatidylinositol 4,5-Diphosphate/analysis , Phospholipase C delta/chemistry , Phospholipase C delta/metabolism , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/isolation & purification , Animals , Fluorescence , Micromanipulation , Microscopy, Fluorescence , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Structure, Tertiary
11.
Methods Mol Biol ; 462: 213-22, 2009.
Article in English | MEDLINE | ID: mdl-19160672

ABSTRACT

This chapter describes an indirect approach to measure PTEN's lipid phosphatase activity in vivo. PTEN counteracts phosphatidylinositol 3-kinase action in dephosphorylating 3-phosphorylated phosphoinositides. Therefore, PtdIns(3,4,5)P3-dependent activation and phosphorylation of the survival kinase Akt can be used as readout for cellular PTEN activity. Here we have outlined a detailed procedure employing a phosphoserine-specific anti-Akt antibody to examine the content of phosphorylated Akt by immunofluorescence and its dependence on PTEN activity.


Subject(s)
PTEN Phosphohydrolase/analysis , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/metabolism , 3T3 Cells , Animals , Antibodies/immunology , Antibody Specificity , Mice , Microscopy, Fluorescence , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/immunology , Staining and Labeling
13.
J Biol Chem ; 283(9): 5477-85, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18077453

ABSTRACT

In addition to the well characterized phosphoinositide second messengers derived from the plasma membrane, increasing evidence supports the existence of a nuclear phosphoinositide signaling network. The aim of this investigation was to dissect the role played by nuclear phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) in cell cycle progression and to determine the cell cycle regulatory component(s) that are involved. A number of cytosolic/nuclear PtdIns(4,5)P2-deficient Swiss 3T3 cell lines were established, and their G 0/G 1/S cell cycle phase transitions induced by defined mitogens were examined. Our results demonstrate that nuclear PtdIns(4,5)P2 down-regulation caused a delay in phorbol ester-induced S phase entry and that this was at least in part channeled through cyclin A2 at the transcriptional level. In summary, these data identify cyclin A2 as a downstream effector of the nuclear PtdIns(4,5)P2 signaling network and highlight the importance of nuclear PtdIns(4,5)P2 in the regulation of mammalian mitogenesis.


Subject(s)
Cell Nucleus/metabolism , Cyclin A/biosynthesis , Phosphatidylinositol 4,5-Diphosphate/metabolism , S Phase/physiology , Second Messenger Systems/physiology , Transcription, Genetic/physiology , 3T3 Cells , Animals , Cyclin A/metabolism , Cyclin A2 , Down-Regulation/drug effects , Down-Regulation/physiology , Mice , S Phase/drug effects , Second Messenger Systems/drug effects , Transcription, Genetic/drug effects
14.
J Chem Biol ; 1(1-4): 3-15, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19568794

ABSTRACT

Recently, light microscopy moved back into the spotlight, which is mainly due to the development of revolutionary technologies for imaging real-time events in living cells. It is truly fascinating to see enzymes "at work" and optically acquired images certainly help us to understand biological processes better than any abstract measurements. This review aims to point out elegant examples of recent cell-biological imaging applications that have been developed with a chemical approach. The discussed technologies include nanoscale fluorescence microscopy, imaging of model membranes, automated high-throughput microscopy control and analysis, and fluorescent probes with a special focus on visualizing enzyme activity, free radicals, and protein-protein interaction designed for use in living cells.

15.
Cell Signal ; 19(2): 321-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16942862

ABSTRACT

The secretagogue compound 48/80 (c48/80) is a well known activator of calcium mediated processes and PKCs, and is a potent inducer of mast cell degranulation. As the latter process is a phosphoinositide 3-kinase (PI 3-kinase) mediated event, we wished to address whether or not c48/80 was an activator of PI 3-kinases. The data presented here reveal that c48/80 is an effective activator of PI 3-kinases as judged by the increased phosphorylation of PKB and p70(S6K) in fibroblasts in a PI 3-kinase dependent fashion. Compound 48/80 effectively translocates PKB to the plasma membrane and induces phosphorylation at serine 473 (S473), detected by fluorescence imaging of fixed cells. At higher concentrations the secretagogue is inhibitory towards PKB phosphorylation on S473. Conversely, p70(S6K) phosphorylation on T389 is unaffected at high doses. We provide evidence that the differential effect on the two PI 3-kinase effectors is due to activation of PKCalpha by c48/80, itself a PI 3-kinase dependent process. We conclude that compound 48/80 is an effective activator of PI 3-kinase dependent pathways, leading to the activation of effectors including PKB/Akt, p70(S6K) and PKCalpha. The latter is only activated by higher doses of c48/80 resulting in an inhibition of the c48/80 induced PKB phosphorylation, thus explaining the observed biphasic activation profile for PKB in response to this secretagogue.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , p-Methoxy-N-methylphenethylamine/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Calcium/metabolism , Catalysis , Cell Line , Cell Membrane/metabolism , Enzyme Activation , Humans , Mice , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Signal Transduction
16.
Virchows Arch ; 448(3): 277-87, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16328348

ABSTRACT

Transcriptional E-cadherin down-regulation can be mediated by Snail, a zinc finger transcription factor. To be able to examine nuclear Snail immunoreactivity in archival human cancers, we established a monoclonal antibody against the purified human Snail protein. The specificity of the selected rat antibody Sn9H2 was demonstrated by Western blot analysis using extracts from different cell lines and by immunofluorescence and immunohistochemistry of primary tissues. Subsequently, a series of 340 adenocarcinomas of the upper gastrointestinal tract, including tumours from the oesophagus (n=154), cardia (n=102) and stomach (n=84), arranged in tissue microarrays, were examined for Snail expression and were correlated to E-cadherin expression and clinico-pathological parameters. Nuclear Snail immunoreactivity was seen in 27 tumours (7.9%) and tended to be more frequent in oesophageal adenocarcinomas (11.1%) than in cardiac (6.9%) or gastric (3.6%) carcinomas (p=0.0428). In 35% of the Snail-positive cases, E-cadherin immunoreactivity was lost. No correlation was found for nuclear Snail expression and tumour grade, Lauren's classification, WHO classification, tumour stage and tumour size. The pattern of Snail expression observed with our new hybridoma, Sn9H2, which is currently the only antibody that reacts with endogenous nuclear (active) Snail, suggests only a minor role of Snail in tumours of the upper gastrointestinal tract.


Subject(s)
Adenocarcinoma/metabolism , Cell Nucleus/metabolism , Gastrointestinal Neoplasms/metabolism , Transcription Factors/metabolism , Upper Gastrointestinal Tract/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/pathology , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Hybridomas/immunology , Hybridomas/metabolism , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/metabolism , Snail Family Transcription Factors , Tissue Array Analysis , Transcription Factors/genetics , Upper Gastrointestinal Tract/pathology
17.
ACS Chem Biol ; 1(12): 780-90, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17240976

ABSTRACT

Phosphatase and tensin homologue deleted on chromosome 10 (PTEN), a phosphoinositide 3-phosphatase, is an important regulator of insulin-dependent signaling. The loss or impairment of PTEN results in an antidiabetic impact, which led to the suggestion that PTEN could be an important target for drugs against type II diabetes. Here we report the design and validation of a small- molecule inhibitor of PTEN. Compared with other cysteine-based phosphatases, PTEN has a much wider active site cleft enabling it to bind the PtdIns(3,4,5)P3 substrate. We have exploited this feature in the design of vanadate scaffolds complexed to a range of different organic ligands, some of which show potent inhibitory activity. A vanadyl complexed to hydroxypicolinic acid was found to be a highly potent and specific inhibitor of PTEN that increases cellular PtdIns(3,4,5)P3 levels, phosphorylation of Akt, and glucose uptake in adipocytes at nanomolar concentrations. The findings presented here demonstrate the applicability of a novel and specific chemical inhibitor against PTEN in research and drug development.


Subject(s)
Hypoglycemic Agents/chemistry , Organometallic Compounds/chemistry , PTEN Phosphohydrolase/antagonists & inhibitors , Vanadium/chemistry , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Dose-Response Relationship, Drug , Drug Design , Fibroblasts/drug effects , Fibroblasts/enzymology , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Ligands , Mice , Molecular Sequence Data , Organometallic Compounds/pharmacology , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
18.
J Cell Physiol ; 202(3): 805-13, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15389640

ABSTRACT

Tumor progression is characterized by loss of cell adhesion and increase of invasion and metastasis. The cell adhesion molecule E-cadherin is frequently down-regulated or mutated in tumors. In addition to down-regulation of cell adhesion, degradation of the extracellular matrix by matrix metalloproteinases is necessary for tumor cell spread. To investigate a possible link between E-cadherin and matrix metalloproteinase 3 (MMP-3), we examined expression of MMP-3 in human MDA-MB-435S cells transfected with wild-type (wt) or three different tumor-associated mutant E-cadherin variants with alterations in exons 8 or 9, originally identified in gastric carcinoma patients. In the presence of wt E-cadherin, the MMP-3 protein level was decreased in cellular lysates and in the supernatant where a secreted form of the protein is detectable. Down-regulation of MMP-3 was not found in MDA-MB-435S transfectants expressing mutant E-cadherin variants which indicates that E-cadherin mutations interfere with the MMP-3 suppressing function of E-cadherin. The mechanism of regulation of MMP-3 by E-cadherin is presently not clear. We have previously found that cell motility is enhanced by expression of the mutant E-cadherin variants used in this study. Here, we found that application of the synthetic inhibitor of MMP-3 NNGH and small interfering RNA (siRNA) directed against MMP-3 reduce mutant E-cadherin-enhanced cell motility. Taken together, our results point to a functional link between MMP-3 and E-cadherin. MMP-3 is differentially regulated by expression of wt or mutant E-cadherin. On the other hand, MMP-3 plays a role in the enhancement of cell motility by mutant E-cadherin. Both observations may be highly relevant for tumor progression since they concern degradation of the extracellular matrix and tumor cell spread.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Exons , Matrix Metalloproteinase 3/metabolism , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Biological Assay , Cell Line, Tumor , Cell Movement , Cell Shape , Gene Expression Profiling , Humans , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase Inhibitors , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis
19.
Int J Cancer ; 111(5): 711-9, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15252840

ABSTRACT

In our study, we aimed to investigate the expression of N-cadherin and E-cadherin and their dependency on epithelial-mesenchymal transition regulators SNAI1, SIP1 and TWIST in human colon cancer. Expression of E-cadherin and N-cadherin was examined by immunohistochemistry in 80 colon carcinomas by using paraffin embedded and formalin fixed tissues. Those cases were partly analyzed for mRNA expression of N-cadherin (42 cases), TWIST (18 cases), SNAI1 (25 cases) and SIP1 (25 cases) by real-time quantitative RT-PCR. Additionally, colon carcinomas that showed amplification of 20q13, the localization of the human SNAI1 gene, were examined. We found cytoplasmic and/or membrane-associated immunoreactivity of N-cadherin in 35/80 (44%) of the cases. However, there was no correlation to upregulated TWIST mRNA levels, as we have shown previously for diffuse-type gastric cancers with abnormal N-cadherin expression. Reduced and/or cytoplasmic E-cadherin immunoreactivity was detected in 19% (15/80) of the cases. Expression of SNAI1 or SIP1 mRNA was not seen in any of the 25 cases analyzed. There was no correlation between amplification of 20q13 and SNAI1 mRNA expression. Remarkably, N-cadherin was almost exclusively expressed in those cases showing normal E-cadherin immunoreactivity, suggesting a mutual exclusion between abnormal E-cadherin reduction and upregulation of N-cadherin. For the first time, we postulate a role for N-cadherin in primary colon cancer progression, which may be similar to the effect discovered by others in breast cancer cell lines, where coexpressed N-cadherin can exert a dominant function over E-cadherin's adhesive function and thus promote tumor invasiveness.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/physiopathology , Cadherins/biosynthesis , Colonic Neoplasms/genetics , Colonic Neoplasms/physiopathology , Gene Expression Profiling , Neoplasm Invasiveness/physiopathology , Cadherins/genetics , Cadherins/pharmacology , Cell Transformation, Neoplastic , Cytoplasm , Disease Progression , Humans , Nuclear Proteins/pharmacology , RNA/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/pharmacology , Twist-Related Protein 1
20.
Carcinogenesis ; 24(12): 1879-86, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12949051

ABSTRACT

In this study, we investigated whether tumor-associated E-cadherin mutations impair the tumor-suppressive function of the cell adhesion molecule and influence metastasis formation in a severe combined immunodeficiency mouse model. The investigated E-cadherin mutations were in frame deletions of exons 8 (del 8) or 9 (del 9) and a point mutation in exon 8 (p8). Transfected human MDA-MB-435S carcinoma cells stably expressing wild-type (wt) or mutant E-cadherin were injected into the mouse mammary fat pad. Mice transplanted with wt E-cadherin transfectants developed significantly smaller tumors than animals transplanted with the E-cadherin-negative parental cell line. Animals transplanted with del 9 or p8 E-cadherin transfectants produced medium size tumors, indicating that these mutations impair the tumor-suppressive function of E-cadherin. In contrast, mice transplanted with del 8 E-cadherin transfectants developed tumors of approximately the same sizes as animals transplanted with wt E-cadherin expressing cells. Lung metastases were induced by all cell lines without significant differences. Immunohistochemical analysis of E-cadherin expression in the tumors revealed a heterogeneous staining pattern, indicating loss or down-regulation of E-cadherin in some tumor cells. Metastases were completely negative for E-cadherin. Our data suggest that the type of mutation determines whether the tumor-suppressive function of E-cadherin is impaired.


Subject(s)
Cadherins/genetics , Mutation , Animals , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Down-Regulation , Exons , Flow Cytometry , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...