Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Ann Oncol ; 28(1): 149-156, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28177473

ABSTRACT

Background: Aneuploidy and chromosomal instability (CIN) are common features of human malignancy that fuel genetic heterogeneity. Although tolerance to tetraploidization, an intermediate state that further exacerbates CIN, is frequently mediated by TP53 dysfunction, we find that some genome-doubled tumours retain wild-type TP53. We sought to understand how tetraploid cells with a functional p53/p21-axis tolerate genome-doubling events. Methods: We performed quantitative proteomics in a diploid/tetraploid pair within a system of multiple independently derived TP53 wild-type tetraploid clones arising spontaneously from a diploid progenitor. We characterized adapted and acute tetraploidization in a variety of flow cytometry and biochemical assays and tested our findings against human tumours through bioinformatics analysis of the TCGA dataset. Results: Cyclin D1 was found to be specifically overexpressed in early but not late passage tetraploid clones, and this overexpression was sufficient to promote tolerance to spontaneous and pharmacologically induced tetraploidy. We provide evidence that this role extends to D-type cyclins and their overexpression confers specific proliferative advantage to tetraploid cells. We demonstrate that tetraploid clones exhibit elevated levels of functional p53 and p21 but override the p53/p21 checkpoint by elevated expression of cyclin D1, via a stoichiometry-dependent and CDK activity-independent mechanism. Tetraploid cells do not exhibit increased sensitivity to abemaciclib, suggesting that cyclin D-overexpressing tumours might not be specifically amenable to treatment with CDK4/6 inhibitors. Conclusions: Our study suggests that D-type cyclin overexpression is an acute event, permissive for rapid adaptation to a genome-doubled state in TP53 wild-type tumours and that its overexpression is dispensable in later stages of tumour progression.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Cyclin C/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Cyclin C/biosynthesis , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytochalasin B/analogs & derivatives , Cytochalasin B/pharmacology , Diploidy , Flow Cytometry , Gene Knockdown Techniques , Genes, p53 , HCT116 Cells , Humans , Protein Kinase Inhibitors/pharmacology , Tetraploidy , Tumor Suppressor Protein p53/metabolism
4.
Oncogene ; 35(30): 4009-19, 2016 07 28.
Article in English | MEDLINE | ID: mdl-26549024

ABSTRACT

The DNA replication machinery invariably encounters obstacles that slow replication fork progression, and threaten to prevent complete replication and faithful segregation of sister chromatids. The resulting replication stress activates ATR, the major kinase involved in resolving impaired DNA replication. In addition, replication stress also activates the related kinase ATM, which is required to prevent mitotic segregation errors. However, the molecular mechanism of ATM activation by replication stress is not defined. Here, we show that monoubiquitinated Proliferating Cell Nuclear Antigen (PCNA), a marker of stalled replication forks, interacts with the ATM cofactor ATMIN via WRN-interacting protein 1 (WRNIP1). ATMIN, WRNIP1 and RAD18, the E3 ligase responsible for PCNA monoubiquitination, are specifically required for ATM signalling and 53BP1 focus formation induced by replication stress, not ionising radiation. Thus, WRNIP1 connects PCNA monoubiquitination with ATMIN/ATM to activate ATM signalling in response to replication stress and contribute to the maintenance of genomic stability.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , Carrier Proteins/physiology , DNA Replication , DNA-Binding Proteins/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Ubiquitin-Protein Ligases/physiology , ATPases Associated with Diverse Cellular Activities , Aphidicolin/pharmacology , DNA Damage , Genomic Instability , Humans , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
5.
Ann Oncol ; 26(7): 1340-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26003169

ABSTRACT

BACKGROUND: Chromosomal instability (CIN) has been shown to be associated with drug resistance and poor clinical outcome in several cancer types. However, in oestrogen receptor (ER)-negative breast cancer we have previously demonstrated that extreme CIN is associated with improved clinical outcome, consistent with a negative impact of CIN on tumour fitness and growth. The aim of this current study was to validate this finding using previously defined CIN thresholds in a much larger prospective cohort from a randomised, controlled, clinical trial. PATIENTS AND METHODS: As a surrogate measurement of CIN, dual centromeric fluorescence in situ hybridisation was performed for both chromosomes 2 and 15 on 1173 tumours from the breast cancer TACT trial (CRUK01/001). Each tumour was scored manually and the mean percentage of cells deviating from the modal centromere number was used to define four CIN groups (MCD1-4), where tumours in the MCD4 group were defined as having extreme CIN. RESULTS: In a multivariate analysis of disease-free survival, with a median follow-up of 91 months, increasing CIN was associated with improved outcome in patients with ER-negative cancer (P trend = 0.03). A similar pattern was seen in ER-negative/HER2-negative cancers (Ptrend = 0.007). CONCLUSIONS: This prospective validation cohort study further substantiated the association between extreme CIN and improved outcome in ER-negative breast cancers. Identifying such patients with extreme CIN may help distinguish good from poor prognostic groups, and therefore support treatment and risk stratification in this aggressive breast cancer subtype.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Chromosomal Instability , Receptors, Estrogen/metabolism , Adult , Aged , Aged, 80 and over , Anthracyclines/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Prospective Studies , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Survival Rate , Taxoids/administration & dosage , Young Adult
6.
Oncogene ; 34(46): 5699-708, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-25728682

ABSTRACT

Defining mechanisms that generate intratumour heterogeneity and branched evolution may inspire novel therapeutic approaches to limit tumour diversity and adaptation. SETD2 (Su(var), Enhancer of zeste, Trithorax-domain containing 2) trimethylates histone-3 lysine-36 (H3K36me3) at sites of active transcription and is mutated in diverse tumour types, including clear cell renal carcinomas (ccRCCs). Distinct SETD2 mutations have been identified in spatially separated regions in ccRCC, indicative of intratumour heterogeneity. In this study, we have addressed the consequences of SETD2 loss-of-function through an integrated bioinformatics and functional genomics approach. We find that bi-allelic SETD2 aberrations are not associated with microsatellite instability in ccRCC. SETD2 depletion in ccRCC cells revealed aberrant and reduced nucleosome compaction and chromatin association of the key replication proteins minichromosome maintenance complex component (MCM7) and DNA polymerase δ hindering replication fork progression, and failure to load lens epithelium-derived growth factor and the Rad51 homologous recombination repair factor at DNA breaks. Consistent with these data, we observe chromosomal breakpoint locations are biased away from H3K36me3 sites in SETD2 wild-type ccRCCs relative to tumours with bi-allelic SETD2 aberrations and that H3K36me3-negative ccRCCs display elevated DNA damage in vivo. These data suggest a role for SETD2 in maintaining genome integrity through nucleosome stabilization, suppression of replication stress and the coordination of DNA repair.


Subject(s)
Carcinoma, Renal Cell/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Kidney Neoplasms/genetics , Mutation , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , DNA Repair , DNA Replication , Genetic Heterogeneity , Histones/metabolism , Humans , Kidney Neoplasms/metabolism , Microsatellite Instability , Nucleosomes/pathology
7.
Nat Cell Biol ; 2(9): 574-81, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980697

ABSTRACT

Integration of signalling pathways initiated by receptor tyrosine kinases and integrins is essential for growth-factor-mediated biological responses. Here we show that co-stimulation of growth-factor receptors and integrins activates the focal-adhesion kinase (FAK) family to promote outgrowth of neurites in PC12 and SH-SY5Y cells. Pyk2 and FAK associate with adhesion-based complexes that contain epidermal growth factor (EGF) receptors, through their carboxy- and amino-terminal domains. Expression of the C-terminal domain of Pyk2 or of FAK is sufficient to block neurite outgrowth, but not activation of extracellular-signal-regulated kinase (ERK). Moreover, activation and autophosphorylation of Pyk2/FAK, as well as of effectors of their adhesion-targeting domains, such as paxillin, are important for propagation of signals that control neurite formation. Thus, Pyk2/FAK have important functions in signal integration proximal to integrin/growth-factor receptor complexes in neurons.


Subject(s)
Growth Substances/metabolism , Integrins/metabolism , Neurites/physiology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , Cytoskeletal Proteins/metabolism , Enzyme Activation , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Growth Substances/pharmacology , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Paxillin , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Tumor Cells, Cultured
8.
J Biol Chem ; 274(21): 14893-901, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329689

ABSTRACT

The protein tyrosine kinase Pyk2 acts as an upstream regulator of mitogen-activated protein (MAP) kinase cascades in response to numerous extracellular signals. The precise molecular mechanisms by which Pyk2 activates distinct MAP kinase pathways are not yet fully understood. In this report, we provide evidence that the protein tyrosine kinase Src and adaptor proteins Grb2, Crk, and p130Cas act as downstream mediators of Pyk2 leading to the activation of extracellular signal-regulated kinase (ERK) and c-Jun amino-terminal kinase (JNK). Pyk2-induced activation of Src is necessary for phosphorylation of Shc and p130Cas and their association with Grb2 and Crk, respectively, and for the activation of ERK and JNK cascades. Expression of a Grb2 mutant with a deletion of the amino-terminal Src homology 3 domain or the carboxyl-terminal tail of Sos strongly reduced Pyk2-induced ERK activation, with no apparent effect on JNK activity. Grb2 with a deleted carboxyl-terminal Src homology 3 domain partially blocked Pyk2-induced ERK and JNK pathways, whereas expression of dominant interfering mutants of p130Cas or Crk specifically inhibited JNK but not ERK activation by Pyk2. Taken together, our data reveal specific pathways that couple Pyk2 with MAP kinases: the Grb2/Sos complex connects Pyk2 to the activation of ERK, whereas adaptor proteins p130Cas and Crk link Pyk2 with the JNK pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Proto-Oncogene Proteins/metabolism , Signal Transduction , Focal Adhesion Kinase 2 , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-crk , Retinoblastoma-Like Protein p130 , src Homology Domains
9.
Cell Calcium ; 24(1): 9-16, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9793684

ABSTRACT

Although the LTD4-induced Ca2+ influx in human epithelial cells has been shown to be regulated by a pertussis toxin-sensitive heterotrimeric G-protein, most likely a G alpha i3 protein [Adolfsson J.L.P., Ohd J.F., Sjölander A. Leukotriene D4-induced activation and translocation of the G-protein alpha i3-subunit in human epithelial cells. Biochem Biophys Res Commun 1996; 226: 413-419], the signalling pathway further downstream is still unclear. In the present study, we investigated the possible involvement of cAMP and protein kinase A activity in the LTD4-induced Ca2+ influx in the epithelial cell line Int 407. Stimulation with LTD4, but not with the calcium ionophore ionomycin, triggered a rapid increase (peak at 7 s) in the cellular cAMP level, an effect that was totally abolished by pertussis toxin. Furthermore, the LTD4-induced Ca2+ signal was reduced by 60% when cells that had been pre-incubated with the protein kinase A inhibitor Rp-cAMPS (50 microM for 30 min) were stimulated in a calcium containing medium. In contrast, Rp-cAMPS had no apparent effect on the LTD4-induced Ca2+ signal when the cells were stimulated in a calcium-depleted medium. The immediate LTD4-induced protein tyrosine phosphorylation (15 s), previously shown to be necessary for the subsequent Ca2+ influx, was abolished not only by pretreatment with pertussis toxin but also by exposure to Rp-cAMPS. Furthermore, direct activation of the cellular adenylyl cyclase activity by treatment with forskolin alone induced a prompt Ca2+ signal in the presence, but not in the absence, of extracellular Ca2+, identical results were obtained with the cell permeable cAMP analogue 8-bromo-cAMP. In addition, forskolin induced protein tyrosine phosphorylation similar to that seen with LTD4. These results suggest that protein kinase A activity participates in the regulation of the LTD4-induced Ca2+ influx at a site that is downstream of the activation of the pertussis toxin-sensitive G-protein but upstream of a LTD4-stimulated tyrosine kinase(s).


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Leukotriene D4/physiology , Adenylyl Cyclases/metabolism , Calcium Signaling , Cell Line , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Epithelial Cells , GTP-Binding Proteins/metabolism , Humans , Isoenzymes/metabolism , Phosphorylation , Thionucleotides/pharmacology , Tyrosine/metabolism
10.
Biochem J ; 316 ( Pt 1): 239-45, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8645211

ABSTRACT

We have previously shown that the leukotriene D4 (LTD4)-induced mobilization of intracellular Ca2+ in epithelial cells is mediated by a G-protein that is distinctly different from the pertussis toxin-sensitive G-protein that regulates the subsequent influx of Ca2+. In the present study, we attempted to gain further knowledge about the mechanisms involved in the LTD4-induced mobilization of intracellular Ca2+ in epithelial cells by investigating the effects of compactin, an inhibitor of the isoprenylation pathway, on this signalling event. In cells preincubated with 10 microM compactin for 48 h, the LTD4-induced mobilization of intracellular Ca2+ was reduced by 75% in comparison with control cells. This reduction was reversed by co-administration of mevalonate (1 mM). The effect of compactin occurred regardless of whether or not Ca2+ was present in the extracellular medium, suggesting that isoprenylation must occur before Ca2+ is released from intracellular stores. In accordance with this, we also found that both the LTD4-induced formation of inositol 1,4,5-trisphosphate and the LTD4-induced phosphorylation of phospholipase C gamma 1 (PLC gamma 1) on tyrosine residues were significantly reduced in compactin-pretreated cells. These results open up the possibility that the activation of PLC gamma 1 is related to a molecule that is sensitive to impaired activity of the isoprenylation pathway, such as a small monomeric G-protein. This idea was supported by the observation that Clostridium botulinum C3 exoenzyme-induced inhibition of Rho proteins abolished the LTD4-induced intracellular mobilization of Ca2+. A regulatory role of Rho proteins in the LTD4-induced activation of PLC gamma 1 is unlikely to be indirectly mediated via an effect on the cytoskeleton, since cytochalasin D had no major effect on the LTD4-induced mobilization of Ca2+. Although the mechanism of interaction remains to be elucidated, the present findings indicate an important role of an isoprenylated protein such as Rho in the LTD4-induced Ca2+ signal.


Subject(s)
Botulinum Toxins , Calcium/metabolism , GTP-Binding Proteins/metabolism , Leukotriene D4/pharmacology , Lovastatin/analogs & derivatives , Membrane Proteins/metabolism , Signal Transduction/drug effects , ADP Ribose Transferases/pharmacology , Cell Line , Cytochalasin D/pharmacology , Cytosol/drug effects , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Intestines , Isoenzymes/metabolism , Kinetics , Lovastatin/pharmacology , Mevalonic Acid/pharmacology , Protein Prenylation , Type C Phospholipases/metabolism , rhoB GTP-Binding Protein
11.
Cell Calcium ; 17(3): 177-86, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7621531

ABSTRACT

Leukotriene D4 (LTD4) has been found to induce calcium signalling in the intestinal epithelial cell line Int 407, and this action involves the activation of both different GTP-binding proteins (G-proteins) and phospholipase C of the gamma-subtype (PLC-gamma). With this knowledge as the incentive, we investigated the possible regulatory role of protein tyrosine kinase activities in the calcium signalling system of the LTD4 receptor. The tyrosine kinase inhibitors genistein and herbimycin. A both reduced the LTD4-induced calcium signal by 70% when Int 407 cells were stimulated in the presence of extracellular calcium, but had no effect on the signal when the cells were stimulated in a calcium-free medium. In accordance with these findings, pretreatment with a tyrosine kinase inhibitor also blocked thapsigargin-induced cellular influx of calcium. These inhibitors had no effect on the intracellular mobilisation of calcium, which was supported by the findings that LTD4 was able to induce an increase in the tyrosine phosphorylation of PLC-gamma even when one of the tyrosine kinase inhibitors was present. Of possible interest regarding the effect of genistein on LTD4-induced calcium influx is that two major tyrosine phosphorylated protein bands were detected in immunoprecipitates obtained with PLC-gamma antibodies from LTD4-stimulated cells. These proteins, which associate with PLC-gamma, have estimated molecular weights of 84 and 97 kD. Preincubation with genistein completely abolished the LTD4-induced increase in tyrosine phosphorylation of the major 97 kD band, whereas the 84 kD protein band, like the PLC-gamma band, still exhibited an increased phosphorylation of tyrosine residues in response to LTD4. Neither this effect nor any of the other effects of genistein were induced when cells were preincubated with daidzein, an inactive analogue of genistein. The present results suggest that LTD4-induced calcium signalling in epithelial cells involves not only tyrosine phosphorylation of PLC-gamma, but also a tyrosine kinase-dependent step which occurs downstream of PLC-gamma activation and is directly implicated in the regulation of agonist-mediated calcium influx.


Subject(s)
Calcium/metabolism , Intestinal Mucosa/drug effects , Leukotriene D4/pharmacology , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Benzoquinones , Cell Line , Cytosol/metabolism , Enzyme Activation/drug effects , Epithelium , GTP-Binding Proteins/physiology , Genistein , Humans , Ileum/cytology , Ileum/embryology , Intestinal Mucosa/metabolism , Isoenzymes/physiology , Isoflavones/pharmacology , Jejunum/cytology , Jejunum/embryology , Lactams, Macrocyclic , Phospholipase C gamma , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinones/pharmacology , Rifabutin/analogs & derivatives , Terpenes/pharmacology , Thapsigargin , Type C Phospholipases/physiology
12.
Ann N Y Acad Sci ; 744: 155-60, 1994 Nov 15.
Article in English | MEDLINE | ID: mdl-7825836

ABSTRACT

In a human epithelial cell line LTD4 induces a calcium signal that is dependent on both intracellular mobilization and influx of calcium. This calcium signal is generated via the activation of dual G protein pathways. Whereas the intracellular mobilization of calcium is regulated by a pertussis toxin-insensitive G protein, the subsequent influx of calcium is regulated by a pertussis toxin-sensitive G protein. Furthermore, a LTD4-induced cellular elevation of cAMP also participates in the regulation of this calcium signal. The increase in cAMP is directly related to the LTD4-induced influx of calcium, perhaps by an activation of protein kinase A and a subsequent phosphorylation of a plasma membrane channel. This model of the LTD4-induced signaling pathway in epithelial cells is outlined in Figure 2.


Subject(s)
Leukotriene D4/metabolism , Signal Transduction , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Epithelial Cells , GTP-Binding Proteins/metabolism , Humans
13.
Immunology ; 79(4): 574-9, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8104888

ABSTRACT

Subsets of neurons in the thymic cortex, Peyer's patches and lymphoid tissues of the respiratory system deliver vasoactive intestinal peptide (VIP) at nanomolar concentrations. The possible effects of VIP on B-cell adhesiveness in these tissues were examined in studies of the homotypic aggregation (HA) of human B-lymphoblastoid cells of the Raji line, which express a mean of 27,950 VIP receptors/cell with a mean Kd of 0.8 nM. Mean HA, assessed microscopically, attained a maximum of 54% after 8 hr with 0.1 microgram/ml of phorbol 12-myristate 13-acetate (PMA) (P < 0.01) and 31% after 24 hr with 10(-8) M VIP (P < 0.05), as contrasted with 13% and 20% at the respective times in medium alone, and both stimuli also increased the mean size of aggregates. The presence of the phosphodiesterase inhibitor Ro 20-1724 permitted 10(-9) M VIP, which had no effect alone, to raise the mean cyclic AMP content of Raji cells by more than 10-fold and concurrently to elevate mean HA from 55% in medium alone at 48 hr to 70% and from 55% at 72 hr to 68% (P < 0.05 for both). Monoclonal antibodies to lymphocyte function-associated (LFA-1) adhesive protein and to intercellular adherence molecule-1 (ICAM-1) suppressed significantly the HA of Raji cells induced by VIP and PMA. The effects of VIP on compartmental immunity in the lungs and intestines thus may be mediated in part by increases in lymphocyte adhesiveness, which could contribute to the regional accumulation of specifically immunocompetent cells.


Subject(s)
B-Lymphocytes/immunology , Vasoactive Intestinal Peptide/immunology , Antigens, CD/immunology , Cell Adhesion , Cell Adhesion Molecules/immunology , Cell Aggregation , Cell Line , Cyclic AMP/analysis , Humans , Intercellular Adhesion Molecule-1 , Lymphocyte Function-Associated Antigen-1/immunology , Tetradecanoylphorbol Acetate
14.
J Biol Chem ; 265(34): 20976-81, 1990 Dec 05.
Article in English | MEDLINE | ID: mdl-2174431

ABSTRACT

Leukotrienes are recognized as important mediators of the inflammatory process. Recently, increasing attention has been paid to the role of noninflammatory cells in the regulation of the inflammatory process. To further increase our knowledge of this matter we have, in the present study, investigated leukotriene-induced Ca2+ signaling, using a single cell technique in a human epithelial cell line, Intestine 407. It was evident that both LTD4 and LTE4, at physiological concentrations (10 nM), triggered rapid and pronounced cytosolic free Ca2+ transients, due to both influx across the plasma membrane and intracellular mobilization. Preincubation with pertussis toxin (1200 ng/ml) decreased the level of agonist-induced Ca2+ transients to an extent similar to that caused by depletion of extracellular Ca2+, suggesting that the toxin affected the influx but not the intracellular mobilization of Ca2+. Indeed, by using the Mn2+ quenching technique, it could be shown that pertussis toxin totally inhibited the influx of Ca2+. The fact that, even after pertussis toxin treatment, direct G-protein activation by AIF4- was still able to trigger a cytosolic free Ca2+ transient, indicates that, in these cells, G-proteins (GTP-binding proteins) that are insensitive to pertussis toxin are capable of mediating a Ca2+ signal. In order to test the idea that such G-proteins regulate mobilization of intracellular Ca2+ induced by LTD4 and LTE4, we electropermeabilized and preincubated the Intestine 407 cells with guanosine-5'-O-(2-thiodiphosphate) (GDP beta S), let them reseal, and, after loading with fura2, investigated the effects on agonist-stimulated Ca2+ signaling. Electropermeabiization and resealing alone did not significantly affect the Ca2+ responses triggered by LTD4 or LTE4. Addition of GDP beta S, in the presence of extracellular Ca2+, reduced the Ca2+ responses by approximately 60-70%. In Ca2(+)-depleted medium, GDP beta S also impaired the LTD4-induced response by 65%, however, it had no effect on the Ca2+ response induced by LTE4. In conclusion, LTD4 and LTE4 trigger cytosolic free Ca2+ signaling in a human epithelial cell line by causing both an influx of Ca2+ and mobilization of intracellular Ca2+. The Ca2(+)-signaling mechanism appears to consist of dual pathways, since the influx is regulated by a pertussis toxin-sensitive G-protein, but, the mobilization of Ca2+ is not. Furthermore, our data suggest that the LTD4-induced mobilization is regulated by a pertussis toxin-insensitive G-protein whereas the LTE4-induced mobilization is relatively insensitive to both pertussis toxin and GDP beta S.


Subject(s)
Aluminum Compounds , Calcium/metabolism , Fluorides , GTP-Binding Proteins/metabolism , SRS-A/analogs & derivatives , SRS-A/pharmacology , Signal Transduction/drug effects , Aluminum/pharmacology , Biological Transport, Active/drug effects , Cell Line , Cell Membrane Permeability , Cytosol/metabolism , Epithelium/drug effects , Epithelium/metabolism , Epithelium/physiology , Fluorine/pharmacology , Fura-2 , Humans , Kinetics , Leukotriene E4 , Pertussis Toxin , Sodium Fluoride/pharmacology , Spectrometry, Fluorescence , Time Factors , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...