Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncogene ; 32(40): 4825-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23108405

ABSTRACT

Evidence accumulates that the transcription factor nuclear factor E2-related factor 2 (Nrf2) has an essential role in cancer development and chemoresistance, thus pointing to its potential as an anticancer target and undermining its suitability in chemoprevention. Through the induction of cytoprotective and proteasomal genes, Nrf2 confers apoptosis protection in tumor cells, and inhibiting Nrf2 would therefore be an efficient strategy in anticancer therapy. In the present study, pancreatic carcinoma cell lines (Panc1, Colo357 and MiaPaca2) and H6c7 pancreatic duct cells were analyzed for the Nrf2-inhibitory effect of the coffee alkaloid trigonelline (trig), as well as for its impact on Nrf2-dependent proteasome activity and resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and anticancer drug-induced apoptosis. Chemoresistant Panc1 and Colo357 cells exhibit high constitutive Nrf2 activity, whereas chemosensitive MiaPaca2 and H6c7 cells display little basal but strong tert-butylhydroquinone (tBHQ)-inducible Nrf2 activity and drug resistance. Trig efficiently decreased basal and tBHQ-induced Nrf2 activity in all cell lines, an effect relying on a reduced nuclear accumulation of the Nrf2 protein. Along with Nrf2 inhibition, trig blocked the Nrf2-dependent expression of proteasomal genes (for example, s5a/psmd4 and α5/psma5) and reduced proteasome activity in all cell lines tested. These blocking effects were absent after treatment with Nrf2 siRNA, a condition in which proteasomal gene expression and proteasome activity were already decreased, whereas siRNA against the related transcription factor Nrf1 did not affect proteasome activity and the inhibitory effect of trig. Depending on both Nrf2 and proteasomal gene expression, the sensitivity of all cell lines to anticancer drugs and TRAIL-induced apoptosis was enhanced by trig. Moreover, greater antitumor responses toward anticancer drug treatment were observed in tumor-bearing mice when receiving trig. In conclusion, representing an efficient Nrf2 inhibitor capable of blocking Nrf2-dependent proteasome activity and thereby apoptosis protection in pancreatic cancer cells, trig might be beneficial in improving anticancer therapy.


Subject(s)
Alkaloids/pharmacology , Apoptosis , NF-E2-Related Factor 2/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Line, Tumor , Etoposide/pharmacology , Humans , Mice , Mice, SCID , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Proteasome Endopeptidase Complex/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
2.
Int J Cell Biol ; 2011: 816791, 2011.
Article in English | MEDLINE | ID: mdl-22007221

ABSTRACT

Pancreatic stellate cells have been investigated mostly for their activation process, supposed to support the development of pancreatic disease. Few studies have been presented on reversal of the activation process in vitro. Thiazolidinediones (TZDs) have been used as antidiabetics and have now been reported to exert antifibrotic activity. We tested effects of natural and synthetic ligands of peroxisome proliferator-activated receptor gamma (PPARγ) on human pancreatic fibroblastoid cells (hPFCs) in search for specificity of action. Ciglitazone, as a prototype of TZDs, was shown to have reversible growth inhibitory effects on human pancreatic fibroblastoid cells/stellate cells. Cells treated with ciglitazone for three days showed enhanced lipid content and induction of proteins involved in lipid metabolism. Collagen synthesis was reduced in hPFC. Interaction of PPARγ with DNA binding sites upon ligand binding was shown by gel shift analysis. These findings point toward a potential for adipocyte differentiation in human pancreatic fibroblastoid cells.

3.
Leukemia ; 18(10): 1646-55, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15306824

ABSTRACT

In a cell-type- and stimulus-dependent fashion, the early response gene immediate early gene X-1 (IEX-1) is involved in growth control and modulation of apoptosis. The present study demonstrates that, in the two acute promyelocytic leukemia (APL) cell lines NB4 and KG1, exhibiting distinct responsiveness to retinoic acids (RAs), IEX-1 expression is rapidly (30-60 min) induced by all-trans- or cis-RA and independently of other signal transduction mediators, such as TNFalpha, NF-kappaB or MAP kinases. In NB4 cells (expressing PML-RARalpha), this increase is transient and completely reversible, along with a cell cycle arrest, ongoing differentiation and lower sensitivity to anti-cancer-drug-induced apoptosis. In contrast, the RA-induced IEX-1 expression in KG1 cells (expressing PLZF-RARalpha) persists over days, along with continued cell cycle progression and increased apoptotic sensitivity. Furthermore, two functional RA-response elements in the IEX-1 promoter were identified by gel shift and luciferase reporter gene assays. IEX-1 might be a rather unique transcriptional target of the two X-RARalpha fusion receptors exhibiting distinct responsiveness to RAs. Following a different time course of direct transcriptional induction by PML-RARalpha and PLZF-RARalpha in NB4 and KG1 cells, respectively, IEX-1 expression may be involved in the modified actions of these receptors and the distinct phenotypes of APL cells.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic/drug effects , Immediate-Early Proteins/genetics , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/genetics , Tretinoin/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cell Cycle/drug effects , Cell Differentiation/drug effects , Electrophoretic Mobility Shift Assay , Humans , Immediate-Early Proteins/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Luciferases/metabolism , Membrane Proteins , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Phenotype , Promoter Regions, Genetic , Transcription, Genetic , Tumor Cells, Cultured
4.
Regul Pept ; 102(2-3): 101-10, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11730982

ABSTRACT

Gastrin stimulates gastric acid secretion in various species, but the role of the structurally related CCK for the peripheral regulation of acid secretion in humans remains controversial. Moreover, species differences in CCK receptor function and expression have been reported. We therefore sought to identify the cellular targets of CCK and gastrin within the human gastric mucosa in situ. Gastric biopsies were collected from 15 patients without gastric disease. Expression of CCK receptor subtypes was detected in individual cells of the gastric mucosa by reverse transcription (RT)-PCR in situ, immunohistochemistry and confocal laser scanning microscopy, using antisera against the CCK-A or CCK-B/gastrin receptor subtype. Both CCK-A and CCK-B receptors were detected in antral and oxyntic mucosa at the mRNA and protein level. In fundic mucosa, CCK-A receptor mRNA and protein mapped to D cells (37.4+/-7.7). Besides, individual chief cells, mucous neck cells and parietal cells (12.3+/-4.7%) expressed CCK-A receptors. CCK-B/gastrin receptor mRNA and protein were detected in parietal cells (57.4+/-11.1%) and in neuroendocrine cells (33.2+/-4.4%) expressing chromogranin A. Furthermore, epithelial cells within the neck of the gastric gland were found to express the CCK-B/gastrin receptor. We conclude that (i) identification of CCK-A receptors on somatostatin producing D cells in humans provide the anatomical basis for a receptor-mediated mode of action of CCK on somatostatin release and (ii) detection of either CCK receptor subtype in the putative stem cell compartment implies a role of CCK in the maintenance of tissue homeostasis in human gastric mucosa.


Subject(s)
Gastric Mucosa/metabolism , Receptors, Cholecystokinin/metabolism , Adult , Epithelial Cells/metabolism , Female , Gastric Mucosa/anatomy & histology , Gastric Mucosa/cytology , Gene Expression , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Molecular Weight , Neurosecretory Systems/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin-Secreting Cells/metabolism
5.
J Cell Sci ; 114(Pt 15): 2735-46, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11683408

ABSTRACT

In this study we investigated the functional role of FAP-1 as a potential inhibitor of CD95 (Fas, APO-1)-mediated apoptosis in pancreatic cancer cells. Stable transfection of the CD95-sensitive, FAP-1-negative cell line Capan-1 with an FAP-1 cDNA resulted in a strongly decreased sensitivity to CD95-induced apoptosis, as measured by DNA fragmentation and caspase-3 activity. Inhibition of cellular protein tyrosine phosphatases with orthovanadate dose-dependently increased CD95-induced apoptosis in CD95-resistant FAP-1-positive Panc89 and Capan-1-FAP-1 cells almost to the level seen in wild-type Capan-1 cells. Blocking the CD95/FAP-1 interaction in Panc89 cells by cytoplasmic microinjection of a synthetic tripeptide mimicking the C terminus of CD95 resulted in a mean 5.5-fold increase in apoptosis compared to cells that received a control peptide. Using confocal laser scanning microscopy we show that in Panc89 cells FAP-1 is mainly associated with the Golgi complex and with peripheral vesicles. FAP-1 displayed enhanced colocalization with CD95 upon CD95 stimulation in the Golgi complex but not in surface-associated vesicles. This correlated with a decrease in plasma membrane staining for CD95 as determined by FACS analysis. Inhibition of Golgi anterograde transport by brefeldin A abolished the anti-CD95-induced colocalization of FAP-1 and CD95 as well as the decrease in cell-surface-associated CD95. Finally, we demonstrate by immunohistochemistry that FAP-1 is strongly expressed in tumor cells from pancreatic carcinoma tissues. Taken together, these results show that FAP-1 can protect pancreatic carcinoma cells from CD95-mediated apoptosis, probably by preventing anti-CD95-induced translocation of CD95 from intracellular stores to the cell surface.


Subject(s)
Adenocarcinoma , Apoptosis/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Pancreatic Neoplasms , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , fas Receptor/metabolism , Brefeldin A/pharmacology , Carrier Proteins/analysis , Cytoplasmic Vesicles/chemistry , Cytoplasmic Vesicles/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Humans , Jurkat Cells , Peptide Fragments/pharmacology , Protein Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/antagonists & inhibitors , fas Receptor/analysis
6.
Neurosci Lett ; 313(1-2): 21-4, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11684330

ABSTRACT

This study investigates whether potassium ion (K+) channels are involved in the nitric oxide (NO)-induced relaxation in segments of the isolated rat basilar artery, mounted onto a wire myograph. A high extracellular K+ concentration partly inhibited the relaxant effects of the NO donors DEA/NO and SIN-1 (3-morpholino-sydnonimine). Whereas single applications of the K+ channel inhibitors tetraethyl-ammonium (10(-3) M), glibenclamide (10(-6) M), 4-aminopyridine (10(-3) M), or BaCl(2) (5 x 10(-5) M) did not affect the responses to DEA/NO, a combination of these inhibitors reduced the effects of DEA/NO. These data suggest, that the relaxant effects of NO donors are partly mediated via activation of K+channels. Different K+ channel types seem to be involved that function in a redundant manner and compensate for each other.


Subject(s)
Basilar Artery/physiology , Hydrazines/pharmacology , Molsidomine/analogs & derivatives , Nitric Oxide Donors/pharmacology , Potassium Channels/physiology , Vasodilation/physiology , 4-Aminopyridine/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Basilar Artery/drug effects , Glyburide/pharmacology , In Vitro Techniques , Male , Molsidomine/pharmacology , Nitrogen Oxides , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Tetraethylammonium/pharmacology , Vasodilation/drug effects
8.
Pancreas ; 23(1): 49-54, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11451147

ABSTRACT

Investigation of pancreatic interstitial fibroblasts has proven difficult in situ. We have established a method for the isolation of pancreatic fibroblastoid/stellate cells by outgrowth from pancreatic tissue explanted into culture dishes. This technique gives a high yield of viable cells from small tissue samples. Outgrown fibroblastoid cells were established as a primary cell line and characterized during long-term culture. We investigated the development of stellate cell markers, i.e. fat storage, expression of desmin, and alpha-smooth muscle actin (alphaSMA), over weeks in culture. AlphaSMA, investigated by indirect immunofluorescence staining and Western blot analysis, revealed a constant rise in expression during routine culture. After 13 passages. approximately 100% of cells were positive for alphaSMA expression, indicating a myofibroblast type of differentiation in vitro.


Subject(s)
Cell Culture Techniques/methods , Fibroblasts/cytology , Pancreas/cytology , Actins/analysis , Animals , Azo Compounds , Biomarkers , Blotting, Western , Cell Differentiation , Cell Line , Cell Separation , Ceruletide/toxicity , Coloring Agents , Contact Inhibition , Culture Media , Desmin/analysis , Fluorescent Antibody Technique, Indirect , Lipids/analysis , Male , Muscle, Smooth/chemistry , Muscle, Smooth/ultrastructure , Pancreas/drug effects , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology , Rats , Rats, Wistar , Staining and Labeling
9.
Oncogene ; 20(1): 69-76, 2001 Jan 04.
Article in English | MEDLINE | ID: mdl-11244505

ABSTRACT

P22PRG1/IEX-1 is a putative NF-kappaB target gene implicated in the regulation of cellular viability. Here, we show that in HeLa cells TNFalpha induces expression of p22PRG1/IEX-1 in an NF-kappaB dependent fashion. Blockade of NF-kappaB activation by various NF-kappaB inhibitors abolished TNFalpha-induced p22PRG1/IEX-1 expression and increased the sensitivity to apoptosis induced by TNFalpha, an activating Fas-antibody or the anti-cancer drug etoposide. Surprisingly, ectopic expression of p22PRG1/IEX-1 in HeLa cells transfected with an inducible p22PRG1/IEX-1-expression vector augments the susceptibility to apoptosis initiated by death-receptor ligands or by etoposide. In addition, p22PRG1/IEX-1 expressing HeLa cells exhibit an accelerated progression through the cell cycle. Transfection of an antisense hammerhead ribozyme targeted to p22PRG1/IEX-1 reduced the speed in cell cycle progression and decreased the apoptotic response to death ligands. Our data demonstrate that p22PRG1/IEX-1 is specifically induced during NF-kappaB activation, but this seems not to be related to the anti-apoptotic actions of NF-kappaB. Instead, NF-kappaB dependent recruitment of p22PRG1/IEX-1 might be related to a modulation in the cell cycle, and hereby, p22PRG1/IEX-1 may accelerate cell growth on the one hand, but may trigger apoptosis on the other. Oncogene (2001) 20, 69 - 76.


Subject(s)
Apoptosis/genetics , HeLa Cells/cytology , HeLa Cells/metabolism , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Leucine/analogs & derivatives , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , NF-kappa B/physiology , Neoplasm Proteins , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Death/genetics , Genetic Vectors/metabolism , Gliotoxin/pharmacology , HeLa Cells/drug effects , Humans , Hydrolysis , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/physiology , Immunosuppressive Agents/pharmacology , Leucine/pharmacology , Leupeptins/pharmacology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Membrane Proteins , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , RNA, Catalytic/metabolism , Sulfasalazine/pharmacology , Tetracycline/pharmacology , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
10.
J Cell Biol ; 152(5): 911-22, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238448

ABSTRACT

Epimorphin was recently described as a mesenchymal factor modulating morphogenesis of murine mammary ducts, skin, liver, and lung in vitro. In this study epimorphin was analyzed in a human, pancreatic adenocarcinoma cell line (A818-6) which develops single layer epithelial hollow spheres resembling normal pancreatic ductal structures in vitro. Soluble 34- and 31-kD isoforms of epimorphin were found in the culture supernatant of A818-6 cells. In lysates of A818-6 cells we detected the 34-and 31-kD isoforms and the dimers, and in lysates of fibroblasts the 150-kD tetramers of epimorphin additionally. A neutralizing monoclonal antibody against epimorphin (MC-1) efficiently blocked the development of hollow sphere structures from A818-6 cells. Coculture of A818-6 cells with fibroblasts stimulated the development of hollow sphere structures in general and increased differentiation in 5-6-d-old hollow spheres. A818-6 hollow sphere development in the presence of fibroblasts was also blocked by MC-1. In this novel system for human duct-like differentiation of pancreatic epithelial cells, we provide evidence for an autocrine and paracrine function of epimorphin as a major mediator for morphogenesis.


Subject(s)
Autocrine Communication , Membrane Glycoproteins/metabolism , Pancreatic Ducts/cytology , Adenocarcinoma , Aged , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Autocrine Communication/drug effects , Biomarkers/analysis , Cell Adhesion , Cell Aggregation/drug effects , Cell Differentiation , Cell Division/drug effects , Cell Line , Coculture Techniques , Culture Media, Conditioned/metabolism , Dimerization , Female , Fibroblasts , Fluorescent Antibody Technique , Growth Substances/pharmacology , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Morphogenesis/drug effects , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism , Pancreatic Ducts/ultrastructure , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Isoforms/metabolism , Solubility , Syntaxin 1 , Tumor Cells, Cultured
11.
Regul Pept ; 90(1-3): 47-52, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10828492

ABSTRACT

TGFbeta1 is a multifunctional factor, controlling cellular growth and extracellular matrix production. Deletion of the TGFbeta1 gene in mice results in multiple inflammatory reactions. Targeted overexpression of TGFbeta1 in pancreatic islet cells leads to fibrosis of the exocrine pancreas in transgenic mice. In pancreatic fibrosis interstitial fibroblasts are primary candidates for production and deposition of extracellular matrix. Still, little is known about regulation of these cells during development of pancreatic disease. We established primary cell lines of pancreatic fibroblastoid/stellate cells (PFC) from rat pancreas. Investigation of rPFCs in vitro shows TGFbeta1 expression by RT-PCR analysis. Mature TGFbeta1 was detected in culture supernatants by immunoassay. Rat PFCs in culture possess both receptors TGFbeta receptor type I, and type II, necessary for TGFbeta1 signal transduction. Inhibition of TGFbeta1 activity by means of neutralizing antibodies interferes with an autocrine loop and results in a 2-fold stimulation of cell growth. So far, pancreatic fibroblastoid/stellate cells in vitro were known as a target of TGFbeta1 action, but not as a source of TGFbeta1. Our data indicate TGFbeta1 activity in rat pancreas extends beyond regulation of matrix production, but appears to be important in growth control of pancreatic fibroblastoid cells.


Subject(s)
Fibroblasts/cytology , Pancreas/cytology , Transforming Growth Factor beta/physiology , Animals , Cell Division , Cells, Cultured , Fibrosis , Rats , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
12.
Biochem Biophys Res Commun ; 260(3): 820-8, 1999 Jul 14.
Article in English | MEDLINE | ID: mdl-10403848

ABSTRACT

We have evaluated the presence and localization of the CCK(A) receptor in rat, mouse, pig and human fetal pancreas by Northern, Western blots and immunofluorescence techniques. In the rat, parallelism exists between development of the CCK(A) receptor mRNA and protein with maximal peaks of expression during the suckling period. In the course of pancreatitis induction, CCK(A) receptor mRNA were maximally expressed and sustained during the gland's regeneration. In the rat and mouse pancreas, the CCK(A) receptor protein is localized around the acinar cells and beta cells of the islets of Langerhans. In the adult pig and fetal human pancreas, the CCK(A) receptor proteins were detected by Western blot. By immunofluorescence, its detection was possible only in the islet of Langerhans of the pig pancreas. These new findings support the views that CCK plays important and various roles in specific physiological systems of the pancreas of different species.


Subject(s)
Gene Expression Regulation, Developmental , Islets of Langerhans/metabolism , Pancreas/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Cholecystokinin/genetics , Female , Humans , Intestinal Mucosa/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Islets of Langerhans/growth & development , Male , Mice , Pancreas/cytology , Pancreas/embryology , Pancreas/growth & development , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/analysis , Receptors, Cholecystokinin/genetics , Regeneration , Species Specificity , Swine
13.
J Biol Chem ; 274(15): 10203-12, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10187805

ABSTRACT

The 55-kDa receptor for tumor necrosis factor (TR55) triggers multiple signaling cascades initiated by adapter proteins like TRADD and FAN. By use of the primary amine monodansylcadaverine (MDC), we addressed the functional role of tumor necrosis factor (TNF) receptor internalization for intracellular signal distribution. We show that MDC does not prevent the interaction of the p55 TNF receptor (TR55) with FAN and TRADD. Furthermore, the activation of plasmamembrane-associated neutral sphingomyelinase activation as well as the stimulation of proline-directed protein kinases were not affected in MDC-treated cells. In contrast, activation of signaling enzymes that are linked to the "death domain" of TR55, like acid sphingomyelinase and c-Jun-N-terminal protein kinase as well as TNF signaling of apoptosis in U937 and L929 cells, are blocked in the presence of MDC. The results of our study suggest a role of TR55 internalization for the activation of select TR55 death domain signaling pathways including those leading to apoptosis.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Cadaverine/analogs & derivatives , Mitogen-Activated Protein Kinases , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Cadaverine/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Ceramides/metabolism , Endocytosis , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Potassium/metabolism , Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I , Sphingomyelin Phosphodiesterase/metabolism , TNF Receptor-Associated Factor 1 , U937 Cells , fas Receptor/immunology , fas Receptor/metabolism
14.
Eur J Pharmacol ; 352(2-3): 343-50, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9716372

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a high-affinity ligand for at least two types of G-protein coupled receptors, the PACAP type 1 and type 2 receptor. In this study it is demonstrated that the C-terminal PACAP-fragment PACAP(6-27) stimulates serotonin release from rat peritoneal mast cells with higher potency (EC50: 0.2 vs. 2.0 microM) than the PACAP receptor ligand PACAP(1-27). PACAP-induced degranulation of rat peritoneal mast cells was abolished by pertussis toxin and by benzalkonium chloride (IC50: 9.1 microg/ml) indicating the involvement of heterotrimeric G-proteins of the Gi-type. The PACAP effect was also reduced by inhibitors of the phosphatidylinositol specific phospholipase C ((U73122), IC50: 4 microM; (ET-18-O-CH3), IC50: 18 microM), by D609, a specific inhibitor of the phosphatidylcholine specific phospholipase C (IC50: 41 microM), by the protein kinase C-inhibitor staurosporine (IC50: 0.6 microM) and by the lipoxygenase inhibitor nordihydroguaiaretic acid (NGDA) but not by indomethacin. It is concluded that PACAP peptides stimulate secretion in rat peritoneal mast cells in a PACAP receptor-independent manner, probably via direct activation of heterotrimeric G-proteins of the Gi-type; these G-proteins may lead to a sequential activation of different signaling cascades (see above), which may converge at the level of one or more staurosporine-sensitive protein kinase.


Subject(s)
Mast Cells/drug effects , Neuropeptides/pharmacology , Peritoneal Cavity/cytology , Signal Transduction/drug effects , Adenylate Cyclase Toxin , Animals , Bridged-Ring Compounds/pharmacology , Estrenes/pharmacology , Indomethacin/pharmacology , Masoprocol/pharmacology , Mast Cells/metabolism , Norbornanes , Pertussis Toxin , Phospholipid Ethers/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Pyrrolidinones/pharmacology , Rats , Staurosporine/pharmacology , Thiocarbamates , Thiones/pharmacology , Virulence Factors, Bordetella/pharmacology , p-Methoxy-N-methylphenethylamine/pharmacology
15.
Ann N Y Acad Sci ; 865: 141-6, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9928006

ABSTRACT

In this study, the secretory effects of PACAP and PACAP analogues on [3H]serotonin-loaded purified rat peritoneal mast cells (RPMCs) were investigated. PACAP(1-27) and PACAP(6-27) stimulated [3H]serotonin release with low potency (ED50: 2 x 10(-6) M) but high efficacy. The N-terminally truncated PACAP form, PACAP(6-27), stimulated tracer release with an ED50 of 0.2 x 10(-6) M, indicating a high-affinity PACAP receptor-independent mechanism of action. The secretory response to PACAP(1-27) could be inhibited by 60-min preincubation with pertussis toxin (ptx), which inhibits G proteins. U73122, a cell-permeable phospholipase C inhibitor, dose-dependently inhibited the secretory effect of 5 microM PACAP(1-27) with an IC50 value of 4 microM (N = 4; p < 0.006). We conclude that PACAP exerts a secretory effect in RPMCs by high-affinity PACAP receptor-independent direct activation of one or more G proteins, which may then activate the PLC-dependent signal-transduction pathway.


Subject(s)
Cell Degranulation/physiology , GTP-Binding Proteins/physiology , Mast Cells/physiology , Neuropeptides/pharmacology , Receptors, Pituitary Hormone/physiology , Adenylate Cyclase Toxin , Animals , Cell Degranulation/drug effects , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Estrenes/pharmacology , In Vitro Techniques , Mast Cells/drug effects , Peptide Fragments/pharmacology , Peritoneal Cavity , Pertussis Toxin , Pituitary Adenylate Cyclase-Activating Polypeptide , Pyrrolidinones/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Sequence Deletion , Serotonin/metabolism , Signal Transduction , Thionucleotides/pharmacology , Type C Phospholipases/antagonists & inhibitors , Virulence Factors, Bordetella/pharmacology , p-Methoxy-N-methylphenethylamine/pharmacology
16.
Am J Physiol ; 270(6 Pt 1): G939-47, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8764200

ABSTRACT

Okadaic acid, a serine/threonine phosphatase inhibitor, has been shown to inhibit rat pancreatic enzyme secretion by interference with late processes in stimulus-secretion coupling. To further characterize its action, we studied the effect of okadaic acid on secretion of newly synthesized proteins, protein synthesis, and cellular ultrastructure in pancreatic lobules derived from rats stimulated in vivo by feeding the synthetic proteinase inhibitor FOY-305. Okadaic acid completely blocked protein secretion at concentrations that inhibit the Ca2+/calmodulin-dependent protein phosphatase 2b, calcineurin. Protein synthesis was abolished at 10(-6) mol/l and reduced by 60% at 5 x 10(-7) mol/l okadaic acid. Pancreatic lobules exposed to 5 x 10(-7) mol/l okadaic acid for 20 min fully restored their secretory capacity on removal of the drug; whereas, after a preincubation with okadaic acid for > 40 min, protein secretion remained impaired during the recovery period. Electron microscopic examination of pancreatic acinar cells treated with 5 x 10(-7) mol/l okadaic acid revealed a dilated Golgi complex after 15 and 30 min and a subsequent fragmentation of Golgi cisternae into clouds of small uniform vesicles after 60 min. Reassembly of Golgi stacks occurred after a 60-min recovery without okadaic acid. These data indicate that serine/threonine phosphatases play an important role not only in the regulation of pancreatic enzyme synthesis and exocytosis but also are crucial for the maintenance of normal Golgi architecture and function in the exocrine rat pancreas. These effects are probably not exclusively mediated via type 2b calcineurin-like protein phosphatases.


Subject(s)
Ethers, Cyclic/pharmacology , Gabexate/analogs & derivatives , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Pancreas/drug effects , Pancreas/enzymology , Animals , Cholecystokinin/physiology , Enzymes/biosynthesis , Enzymes/metabolism , Esters , Guanidines/pharmacology , Male , Microscopy, Electron , Okadaic Acid , Pancreas/cytology , Protease Inhibitors/pharmacology , Protein Biosynthesis , Rats , Rats, Wistar
17.
EMBO J ; 14(11): 2424-35, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7781597

ABSTRACT

Furin, a subtilisin-like eukaryotic endoprotease, is responsible for proteolytic cleavage of cellular and viral proteins transported via the constitutive secretory pathway. Cleavage occurs at the C-terminus of basic amino acid sequences, such as R-X-K/R-R and R-X-X-R. Furin was found predominantly in the trans-Golgi network (TGN), but also in clathrin-coated vesicles dispatched from the TGN, on the plasma membrane as an integral membrane protein and in the medium as an anchorless enzyme. When furin was vectorially expressed in normal rat kidney (NRK) cells it accumulated in the TGN similarly to the endogenous glycoprotein TGN38, often used as a TGN marker protein. The signals determining TGN targeting of furin were investigated by mutational analysis of the cytoplasmic tail of furin and by using the hemagglutinin (HA) of fowl plague virus, a protein with cell surface destination, as a reporter molecule, in which membrane anchor and cytoplasmic tail were replaced by the respective domains of furin. The membrane-spanning domain of furin grafted to HA does not localize the chimeric molecule to the TGN, whereas the cytoplasmic domain does. Results obtained on furin mutants with substitutions and deletions of amino acids in the cytoplasmic tail indicate that wild-type furin is concentrated in the TGN by a mechanism involving two independent targeting signals, which consist of the acidic peptide CPSDSEEDEG783 and the tetrapeptide YKGL765. The acidic signal in the cytoplasmic domain of a HA-furin chimera is necessary and sufficient to localize the reporter molecule to the TGN, whereas YKGL is a determinant for targeting to the endosomes. The data support the concept that the acidic signal, which is the dominant one, retains furin in the TGN, whereas the YKGL motif acts as a retrieval signal for furin that has escaped to the cell surface.


Subject(s)
Golgi Apparatus/enzymology , Subtilisins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Line , Cytoplasm/enzymology , DNA, Recombinant/genetics , Endosomes/enzymology , Furin , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Microscopy, Immunoelectron , Molecular Sequence Data , Mutation , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Subcellular Fractions/enzymology , Subtilisins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...