Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
2.
Article in English | MEDLINE | ID: mdl-35571223

ABSTRACT

Pancreatic cancer is one the most lethal cancers. Currently, there are reliable predictive markers to assess cancer development. Widely used CA19-9 molecular marker has been less effective in the diagnosis of early stages of cancer. Objective: To study if the soluble Osteoprotegerin (OPG) and pigment-epithelial derived factor (PEDF) levels in serum will be an indicator of cancer progression. Methods: Soluble OPG and PEDF were measured from human pancreatic cancer patients by ELISA. Results: We show that while OPG has been less predictive features, PEDF is more sensitive than CA19-9 in cancer detection. More importantly, PEDF and CA19-9 as combined markers showed higher sensitivity in stratifying early stages of pancreatic cancer. Conclusion: Results from the pilot studies suggest that PEDF is useful biomarker for pancreatic cancer.

3.
Cell Calcium ; 44(1): 14-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18207570

ABSTRACT

Pancreatitis is an increasingly common disease that carries a significant mortality and which lacks specific therapy. Pathological calcium signalling is an important contributor to the initiating cell injury, caused by or acting through mitochondrial inhibition. A principal effect of disordered cell signalling and impaired mitochondrial function is cell death, either by apoptosis that is primarily protective, or by necrosis that is deleterious, both locally and systemically. Mitochondrial calcium overload is particularly important in necrotic injury, which may include damage mediated by the mitochondrial permeability transition pore. The role of reactive oxygen species remains controversial. Present understanding of the part played by disordered pancreatic acinar calcium signalling and mitochondrial inhibition offers several new potential therapeutic targets.


Subject(s)
Calcium Signaling/physiology , Cell Death/physiology , Mitochondria , Pancreas/physiopathology , Pancreatitis , Animals , Antioxidants/therapeutic use , Caspases/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore , Oxidative Stress/physiology , Pancreas/enzymology , Pancreas/pathology , Pancreas/ultrastructure , Pancreatitis/etiology , Pancreatitis/metabolism , Pancreatitis/physiopathology , Pancreatitis/therapy , Reactive Oxygen Species/adverse effects
4.
Am J Physiol Gastrointest Liver Physiol ; 292(3): G875-86, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158252

ABSTRACT

Bile acids are known to induce Ca(2+) signals in pancreatic acinar cells. We have recently shown that phosphatidylinositol 3-kinase (PI3K) regulates changes in free cytosolic Ca(2+) concentration ([Ca(2+)](i)) elicited by CCK by inhibiting sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA). The present study sought to determine whether PI3K regulates bile acid-induced [Ca(2+)](i) responses. In pancreatic acinar cells, pharmacological inhibition of PI3K with LY-294002 or wortmannin inhibited [Ca(2+)](i) responses to taurolithocholic acid 3-sulfate (TLC-S) and taurochenodeoxycholate (TCDC). Furthermore, genetic deletion of the PI3K gamma-isoform also decreased [Ca(2+)](i) responses to bile acids. Depletion of CCK-sensitive intracellular Ca(2+) pools or application of caffeine inhibited bile acid-induced [Ca(2+)](i) signals, indicating that bile acids release Ca(2+) from agonist-sensitive endoplasmic reticulum (ER) stores via an inositol (1,4,5)-trisphosphate-dependent mechanism. PI3K inhibitors increased the amount of Ca(2+) in intracellular stores during the exposure of acinar cells to bile acids, suggesting that PI3K negatively regulates SERCA-dependent Ca(2+) reloading into the ER. Bile acids inhibited Ca(2+) reloading into ER in permeabilized acinar cells. This effect was augmented by phosphatidylinositol (3,4,5)-trisphosphate (PIP(3)), suggesting that both bile acids and PI3K act synergistically to inhibit SERCA. Furthermore, inhibition of PI3K by LY-294002 completely inhibited trypsinogen activation caused by the bile acid TLC-S. Our results indicate that PI3K and its product, PIP(3), facilitate bile acid-induced [Ca(2+)](i) responses in pancreatic acinar cells through inhibition of SERCA-dependent Ca(2+) reloading into the ER and that bile acid-induced trypsinogen activation is mediated by PI3K. The findings have important implications for the mechanism of acute pancreatitis since [Ca(2+)](i) increases and trypsinogen activation mediate key pathological processes in this disorder.


Subject(s)
Bile Acids and Salts/pharmacology , Calcium/metabolism , Pancreas, Exocrine/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Androstadienes/pharmacology , Animals , Cells, Cultured , Cholecystokinin/pharmacology , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Ionomycin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Pancreas, Exocrine/cytology , Pancreas, Exocrine/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Taurochenodeoxycholic Acid/pharmacology , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology , Thapsigargin/pharmacology , Wortmannin
6.
Am J Physiol Gastrointest Liver Physiol ; 287(6): G1200-12, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15271649

ABSTRACT

Calcium is a key mediator of hormone-induced enzyme secretion in pancreatic acinar cells. At the same time, abnormal Ca(2+) responses are associated with pancreatitis. We have recently shown that inhibition of phosphatidylinositol 3-kinase (PI3-kinase) by LY-294002 and wortmannin, as well as genetic deletion of PI3-kinase-gamma, regulates Ca(2+) responses and the Ca(2+)-sensitive trypsinogen activation in pancreatic acinar cells. The present study sought to determine the mechanisms of PI3-kinase involvement in Ca(2+) responses induced in these cells by CCK and carbachol. The PI3-kinase inhibitors inhibited both Ca(2+) influx and mobilization from intracellular stores induced by stimulation of acini with physiological and pathological concentrations of CCK, as well as with carbachol. PI3-kinase inhibition facilitated the decay of cytosolic free Ca(2+) concentration ([Ca(2+)](i)) oscillations observed in individual acinar cells. The PI3-kinase inhibitors decreased neither CCK-induced inositol 1,4,5-trisphosphate [Ins(1,4,5)P(3)] production nor Ins(1,4,5)P(3)-induced Ca(2+) mobilization, suggesting that the effect of PI3-kinase inhibition is not through Ins(1,4,5)P(3) or Ins(1,4,5)P(3) receptors. PI3-kinase inhibition did not affect Ca(2+) mobilization induced by thapsigargin, a specific inhibitor of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA). Moreover, SERCA blockade with thapsigargin abolished the effects of pharmacological and genetic PI3-kinase inhibition on [Ca(2+)](i) signals, suggesting SERCA as a downstream target of PI3-kinase. Both pharmacological PI3-kinase inhibition and genetic deletion of PI3-kinase-gamma increased the amount of Ca(2+) in intracellular stores during CCK stimulation. Finally, addition of the PI3-kinase product phosphatidylinositol 3,4,5-trisphosphate to permeabilized acini significantly attenuated Ca(2+) reloading into the endoplasmic reticulum. The results indicate that PI3-kinase regulates Ca(2+) signaling in pancreatic acinar cells through its inhibitory effect on SERCA.


Subject(s)
Calcium Signaling/physiology , Calcium-Transporting ATPases/physiology , Pancreas/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Cholecystokinin/pharmacology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Pancreas/cytology , Pancreas/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Thapsigargin/pharmacology
7.
Am J Physiol Gastrointest Liver Physiol ; 280(6): G1197-208, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352813

ABSTRACT

Transcription factor nuclear factor-kappaB (NF-kappaB) is activated in cerulein pancreatitis and mediates cytokine expression. The role of transcription factor activation in other models of pancreatitis has not been established. Here we report upregulation of NF-kappaB and inflammatory molecules, and their correlation with local pancreatic injury, in a model of severe pancreatitis. Rats received intraductal infusion of taurocholate or saline, and the pancreatic head and tail were analyzed separately. NF-kappaB and activator protein-1 (AP-1) activation were assessed by gel shift assay, and mRNA expression of interleukin-6, tumor necrosis factor-alpha, KC, monocyte chemoattractant protein-1, and inducible nitric oxide synthase was assessed by semiquantitative RT-PCR. Morphological damage and trypsin activation were much greater in the pancreatic head than tail, in parallel with a stronger activation of NF-kappaB and cytokine mRNA. Saline infusion mildly affected these parameters. AP-1 was strongly activated in both pancreatic segments after either taurocholate or saline infusion. NF-kappaB inhibition with N-acetylcysteine ameliorated the local inflammatory response. Correlation between localized NF-kappaB activation, cytokine upregulation, and tissue damage suggests a key role for NF-kappaB in the development of the inflammatory response of acute pancreatitis.


Subject(s)
NF-kappa B/physiology , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Taurocholic Acid , Amylases/blood , Animals , Cell Movement , Chemokines/genetics , Cytokines/genetics , Gene Expression , Lipase/blood , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , Neutrophils/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Pancreas/pathology , Pancreatitis/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , Transcription Factor AP-1/physiology , Trypsin/physiology
8.
Am J Physiol Cell Physiol ; 279(6): C1993-2003, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11078716

ABSTRACT

Cytokines produced by pancreatic acinar cells may mediate cell death and recruitment of inflammatory cells into pancreas in pancreatitis and other disorders. Here, we demonstrate mRNA expression for a number of cytokines in acini isolated from rat pancreas. Using RNA from microscopically selected individual cells, we confirmed the acinar cell as a source for cytokine expression. Competitive RT-PCR, Western blot analysis, and immunocytochemistry showed large amounts of monocyte chemotactic protein-1 and interleukin-6 compared with other cytokines. Cytokine expression was inhibited by either inhibitors of p38 mitogen-activated protein kinase (MAPK), SB-202190 and SB-203580, or (less strongly) by the transcription factor nuclear factor (NF)-kappaB inhibitor MG-132. A combination of SB-203580 and MG-132 inhibited mRNA expression of all cytokines by >90%. The results suggest a major role for p38 MAPK and involvement of NF-kappaB in cytokine expression in pancreatic acinar cells. In contrast to isolated acini, we detected no or very low cytokine expression in normal rat pancreas. Our results indicate that activation of p38 MAPK, transcription factors, and cytokines occurs during removal of the pancreas from the animal and isolation of acini.


Subject(s)
Interleukin-6/metabolism , Mitogen-Activated Protein Kinases/metabolism , Pancreas/cytology , Pancreatitis/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines/genetics , Chemokines/metabolism , Chemokines, CXC , Cytokines/genetics , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression/immunology , Imidazoles/pharmacology , In Vitro Techniques , Interleukin-6/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Pancreas/enzymology , Pancreas/immunology , Pancreatitis/immunology , Pyridines/pharmacology , RNA, Messenger/analysis , Rats , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
9.
Am J Physiol Gastrointest Liver Physiol ; 279(4): G666-76, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11005752

ABSTRACT

Neutrophil infiltration into the pancreas is a key event in pancreatitis. Here we show that intercellular adhesion molecule-1 (ICAM-1), which regulates neutrophil adhesion, is present on rat pancreatic acinar cells, is upregulated by a hormone (cerulein) and mediates direct binding of neutrophils to acinar cells. ICAM-1 was upregulated in pancreas of rats with experimental pancreatitis induced by supramaximal doses of cerulein. Furthermore, cerulein time and dose dependently stimulated expression of ICAM-1 mRNA and protein in isolated pancreatic acinar cells. Inhibitory analysis showed that activation of transcription factor nuclear factor-kappaB (NF-kappaB) was involved in ICAM-1 upregulation by cerulein, but NF-kappaB did not mediate basal expression of ICAM-1 mRNA in acinar cells. With an adhesion assay, we found that neutrophils bind to isolated pancreatic acinar cells and that cerulein upregulates the extent of adhesion. Neutralizing ICAM-1 antibody blocked neutrophil binding to both control and cerulein-stimulated acinar cells, suggesting ICAM-1 involvement in this adhesion. Thus the acinar cell is capable of targeting neutrophils to its surface, a process that may be important for inflammatory and cell death responses in pancreatitis and other pancreatic disorders.


Subject(s)
Cell Adhesion/physiology , Ceruletide/pharmacology , Gene Expression Regulation/drug effects , Intercellular Adhesion Molecule-1/genetics , Neutrophils/physiology , Pancreas/physiology , Animals , Cell Adhesion/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Intercellular Adhesion Molecule-1/physiology , Kinetics , Leupeptins/pharmacology , Male , Pancreas/cytology , Pancreas/drug effects , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/physiopathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects
10.
Biochem J ; 351(Pt 1): 265-71, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10998370

ABSTRACT

Regulation of cytosolic Ca(2+) is important for a variety of cell functions. The ryanodine receptor (RyR) is a Ca(2+) channel that conducts Ca(2+) from internal pools to the cytoplasm. To demonstrate the presence of the RyR in the pancreatic acinar cell, we performed reverse transcriptase (RT)-PCR, Western blot, immunocytochemistry and microscopic Ca(2+)-release measurements on these cells. RT-PCR showed the presence of mRNA for RyR isoforms 1, 2 and 3 in both rat pancreas and dispersed pancreatic acini. Furthermore, mRNA expression for RyR isoforms 1 and 2 was demonstrated by RT-PCR in individual pancreatic acinar cells selected under the microscope. Western-blot analysis of acinar cell immunoprecipitates, using antibodies against RyR1 and RyR2, showed a high-molecular-mass (>250 kDa) protein band that was much less intense when immunoprecipitated in the presence of RyR peptide. Functionally, permeablized acinar cells stimulated with the RyR activator, palmitoyl-CoA, released Ca(2+) from both basolateral and apical regions. These data show that pancreatic acinar cells express multiple isoforms of the RyR and that there are functional receptors throughout the cell.


Subject(s)
Pancreas/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Membrane Permeability , Cell Polarity , Cells, Cultured , Immunohistochemistry , Molecular Weight , Palmitoyl Coenzyme A/pharmacology , Pancreas/cytology , Pancreas/drug effects , Precipitin Tests , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics
11.
Am J Physiol Cell Physiol ; 278(2): C363-71, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666032

ABSTRACT

In this study, we show that particulate guanylate cyclase (GC) is present in rat pancreatic acinar cells and is located both on plasma membrane and membranes of endoplasmic reticulum (ER). Western blot analysis indicates that the enzyme isoform GC-A is present in the acinar cell membranes. The specific inhibitors of ER Ca(2+)-ATPase thapsigargin, 2,5-di-(t-butyl)-1,4-hydroquinone (BHQ), and cyclopiazonic acid all activated particulate GC in pancreatic acini, both in membrane fractions and intact cells. These inhibitors also induced dephosphorylation of GC. Dose dependencies of Ca(2+)-ATPase inhibition and GC activation by BHQ are very similar, and those for thapsigargin partially overlap. ER Ca(2+)-ATPase and GC are coimmunoprecipitated both by antisera against membrane GC and by antisera against ER Ca(2+)-ATPase, suggesting a physical association between the two enzymes. The results suggest that thapsigargin and the other inhibitors act through ER Ca(2+)-ATPase to activate membrane GC in pancreatic acinar cells, although their direct effect on GC cannot be excluded.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Endoplasmic Reticulum/enzymology , Guanylate Cyclase/metabolism , Pancreas/enzymology , Animals , Calcium/metabolism , Calcium-Transporting ATPases/analysis , Cell Fractionation , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/analysis , Hydroquinones/pharmacology , Indoles/pharmacology , Pancreas/cytology , Phosphorylation , Rats , Thapsigargin/pharmacology
12.
Gastroenterology ; 117(3): 706-16, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10464148

ABSTRACT

BACKGROUND & AIMS: Although alcoholism is a major cause of pancreatitis, the pathogenesis of this disorder remains obscure. Failure to produce experimental alcoholic pancreatitis suggests that ethanol may only increase predisposition to pancreatitis. This study sought to develop a model of ethanol pancreatitis by determining if an ethanol diet sensitizes rats to pancreatitis caused by cholecystokinin octapeptide (CCK-8). METHODS: Rats were fed intragastrically either control or ethanol diet for 2 or 6 weeks. The animals were then infused for 6 hours with either saline or CCK-8 at a dose of 3000 pmol. kg(-1). h(-1), which by itself did not induce pancreatitis. The following parameters were measured: serum amylase and lipase levels, pancreatic weight, inflammatory infiltration, number of apoptotic acinar cells, pancreatic messenger RNA (mRNA) expression of cytokines and chemokines, and nuclear factor (NF)-kappaB activity. RESULTS: All measures of pancreatitis, as well as NF-kappaB activity and mRNA expression for tumor necrosis factor alpha, interleukin 6, monocyte chemotactic protein 1, macrophage inflammatory protein 2, and inducible nitric oxide synthase, were significantly increased only in rats treated with ethanol plus CCK-8. CONCLUSIONS: An ethanol diet sensitizes rats to pancreatitis caused by CCK-8. The combined action of ethanol and CCK-8 results in NF-kappaB activation and up-regulation of proinflammatory cytokines and chemokines in the pancreas. These mechanisms may contribute to the development of alcoholic pancreatitis.


Subject(s)
Disease Models, Animal , Pancreatitis, Alcoholic , Amylases/blood , Animals , Apoptosis , Chemokines/biosynthesis , Cytokines/biosynthesis , Ethanol/administration & dosage , Ethanol/pharmacology , Lipase/blood , Male , NF-kappa B/metabolism , Nuclear Proteins/analysis , Pancreas/drug effects , Pancreatitis, Alcoholic/blood , Pancreatitis, Alcoholic/metabolism , Pancreatitis, Alcoholic/pathology , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sincalide/administration & dosage , Sincalide/blood , Sincalide/pharmacology
13.
Pancreas ; 18(3): 231-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10206480

ABSTRACT

Increased activity of various proteases is observed in both human and experimental pancreatitis; however, the information on the effects of specific protease inhibitors on the disease is limited. In this study we show that a novel elastase inhibitor, guamerin-derived synthetic peptide (GDSP), improves the parameters of cerulein-induced acute pancreatitis in the rat. The effects of GDSP on pancreatic weight, serum amylase and lipase, morphologic changes in the pancreas, neutrophil infiltration, and nuclear factor KB (NF-KB) activation were measured in rats infused with supramaximal dose of cerulein (5 (g/kg/h) for 6 h. The effects of GDSP were also measured on superoxide formation by activated human neutrophils. The effects of GDSP were compared with those of another elastase inhibitor, elastatinal. GDSP significantly inhibited edema formation, neutrophil infiltration, acinar cell damage, and plasma lipase and amylase increases caused by cerulein. GDSP also completely inhibited superoxide formation in the human neutrophils stimulated by N-formyl-methionine-leucine-phenyl-alanine (fMLP) or 12-O-tetradecanoylphorbol-13-acetate (TPA). Elastatinal had some of the same effects as GDSP but was less potent and effective. These results demonstrate a beneficial effect of GDSP, a novel specific elastase inhibitor, on the development of rat cerulein pancreatitis.


Subject(s)
Ceruletide , Enzyme Inhibitors/therapeutic use , Invertebrate Hormones/chemistry , Pancreatic Elastase/antagonists & inhibitors , Pancreatitis/drug therapy , Peptides/therapeutic use , Animals , Calcium/metabolism , Enzyme Inhibitors/chemistry , Humans , Male , NF-kappa B/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Oligopeptides/therapeutic use , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/pathology , Peptides/chemistry , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
14.
Am J Physiol ; 275(6): G1402-14, 1998 12.
Article in English | MEDLINE | ID: mdl-9843778

ABSTRACT

Inflammation and cell death are critical to pathogenesis of acute pancreatitis. Here we show that transcription factor nuclear factor-kappaB (NF-kappaB), which regulates these processes, is activated and plays a role in rat cerulein pancreatitis. NF-kappaB was strongly activated in the pancreas within 30 min of cerulein infusion; a second phase of NF-kappaB activation was prominent at 3-6 h. This biphasic kinetics could result from observed transient degradation of the inhibitory protein IkappaBalpha and slower but sustained degradation of IkappaBbeta. The hormone also caused NF-kappaB translocation and IkappaB degradation in vitro in dispersed pancreatic acini. Both p65/p50 and p50/p50, but not c-Rel, NF-kappaB complexes were manifest in pancreatitis and in isolated acini. Coinfusion of CCK JMV-180, which abolishes pancreatitis, prevented cerulein-induced NF-kappaB activation. The second but not early phase of NF-kappaB activation was inhibited by a neutralizing tumor necrosis factor-alpha antibody. Antioxidant N-acetylcysteine (NAC) blocked NF-kappaB activation and significantly improved parameters of pancreatitis. In particular, NAC inhibited intrapancreatic trypsin activation and mRNA expression of cytokines interleukin-6 and KC, which were dramatically induced by cerulein. The results suggest that NF-kappaB activation is an important early event that may contribute to inflammatory and cell death responses in acute pancreatitis.


Subject(s)
Ceruletide , NF-kappa B/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Ceruletide/pharmacology , Chemokines , Cytokines/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , I-kappa B Proteins , Interleukin-6/genetics , Isomerism , Kinetics , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Pancreas/drug effects , Rats , Rats, Sprague-Dawley , Sincalide/analogs & derivatives , Sincalide/pharmacology , Tumor Necrosis Factor-alpha/physiology
15.
J Cell Sci ; 111 ( Pt 2): 283-94, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9405319

ABSTRACT

To understand how phosphatidylinositol 3-kinase (PI3K) modulates cell structure and function, we examined the molecular and cellular defects of a Dictyostelium mutant strain (pik1(Delta)2(Delta)) missing two (DdPIK1 and 2) of three PI3K genes, which are homologues of the mammalian p110 subunit. Levels of [32P]phosphatidylinositol 3, 4 bisphosphate (PI(3,4)P2) and [32P]phosphatidylinositol trisphosphate (PIP3) were reduced in pik1(Delta)2(Delta), which had major defects in morphological and functional correlates of macropinocytosis. This was accompanied by dramatic deficits in a subset of F-actin-enriched structures such as circular ruffles, actin crowns and pseudopodia. Although pik1(Delta)2(Delta) were mobile, they failed to aggregate into streams. Therefore we conclude that PIK1 and 2, possibly through modulation of the levels of PIP3 and PI(3,4)P2, regulate the organization of actin filaments necessary for circular ruffling during macropinocytosis, the extension of pseudopodia and the aggregation of cells into streams, but not the regulation of cell motility.


Subject(s)
Actins/metabolism , Dictyostelium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Pinocytosis/physiology , Animals , Cell Movement , Dictyostelium/genetics , Genetic Complementation Test , Mutagenesis , Phagocytosis , Phosphatidylinositol 3-Kinases/genetics , Phosphorus Radioisotopes , Vacuoles
16.
Cell Calcium ; 22(3): 217-28, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9330792

ABSTRACT

Regulation of agonist-activated Ca2+ influx by the NOS pathway through generation of cGMP is being found in an increasing number of cell types. In the present work, we examined the role of the NOS pathway in agonist-evoked [Ca2+]i oscillations and attempted to identify the NOS isoform most likely to regulate Ca2+ influx. For this, we first show that two Ca(2+)-mobilizing agonists acting on pancreatic acinar cells, bombesin (BS) and the cholecystokinin analog CCK-JMV-180 (CCKJ), evokes different type of [Ca2+]i oscillations. The BS-evoked [Ca2+]i oscillations rapidly became acutely dependent on the presence of extracellular Ca2+, whereas the CCKJ-evoked oscillations continue for long periods of time in the absence of Ca2+ influx. This differential behavior allowed us to isolate Ca2+ influx and study its regulation while controlling for non specific effects on all other Ca2+ transporting events involved in generating [Ca2+]i oscillations. Inhibitors of selective steps in the NOS pathway inhibited agonist-induced cGMP production. The inhibitors were then used to show that scavenging NO with reduced hemoglobin, inhibition of guanylyl cyclase with 1H-[1,2,4] oxadiazolo[4,3-a] quinoxaline-1-one (ODQ) and inhibition of protein kinase G with Rp-8-pCPT-cGMPS inhibited [Ca2+]i oscillations evoked by BS but not those evoked by CCKJ. These findings were extended to duct and acinar cells of the SMG. In these cells, Ca(2+)-mobilizing agonists stimulate large Ca2+ influx, which was inhibited by all inhibitors of the NOS pathway. Western blot analysis and immunolocalization revealed that the cells did not express iNOS, eNOS was expressed only in blood vessels and capillaries whereas nNOS was expressed at high levels next to the plasma membrane of all cells. Accordingly, the nNOS inhibitor 7-nitroindazole (7-NI) inhibited BS- but not CCKJ-evoked [Ca2+]i oscillations and Ca2+ influx into SMG acinar and duct cells. Thus, together, our findings favor nNOS as the isoform activated by the Ca2+ released from internal stores to generate cGMP and regulate Ca2+ influx.


Subject(s)
Calcium/metabolism , Nitric Oxide Synthase/metabolism , Pancreatic Ducts/metabolism , Submandibular Gland/metabolism , Animals , Blotting, Western , Bombesin/pharmacology , Cells, Cultured , Cholecystokinin/analogs & derivatives , Cholecystokinin/pharmacology , Cyclic GMP/metabolism , Immunohistochemistry , Indazoles/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Pancreatic Ducts/cytology , Rats , Signal Transduction , Submandibular Gland/cytology
17.
J Clin Invest ; 100(7): 1853-62, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9312187

ABSTRACT

The aim of this study was to determine whether tumor necrosis factor-alpha (TNFalpha) and receptors for TNFalpha are expressed in the exocrine pancreas, and whether pancreatic acinar cells release and respond to TNFalpha. Reverse transcription PCR, immunoprecipitation, and Western blot analysis demonstrated the presence of TNFalpha and 55- and 75-kD TNFalpha receptors in pancreas from control rats, rats with experimental pancreatitis induced by supramaximal doses of cerulein, and in isolated pancreatic acini. Immunohistochemistry showed TNFalpha presence in pancreatic acinar cells. ELISA and bioassay measurements of TNFalpha indicated its release from pancreatic acinar cells during incubation in primary culture. Acinar cells responded to TNFalpha. TNFalpha potentiated NF-kappaB translocation into the nucleus and stimulated apoptosis in isolated acini while not affecting LDH release. In vivo studies demonstrated that neutralization of TNFalpha with an antibody produced a mild improvement in the parameters of cerulein-induced pancreatitis. However, TNFalpha neutralization greatly inhibited apoptosis in a modification of the cerulein model of pancreatitis which is associated with a high percentage of apoptotic cell death. The results indicate that pancreatic acinar cells produce, release, and respond to TNFalpha. This cytokine regulates apoptosis in both isolated pancreatic acini and experimental pancreatitis.


Subject(s)
Apoptosis/drug effects , Pancreas/metabolism , Pancreatitis/etiology , Receptors, Tumor Necrosis Factor/analysis , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies/pharmacology , Apoptosis/genetics , Biological Assay , Cell Compartmentation/drug effects , Cell Separation , Cells, Cultured , Ceruletide/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , NF-kappa B/metabolism , Neutralization Tests , Nuclear Proteins/metabolism , Pancreas/cytology , Pancreas/drug effects , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Precipitin Tests , Protein Binding , RNA, Messenger/analysis , Rats , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
18.
Gastroenterology ; 113(2): 649-58, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9247487

ABSTRACT

BACKGROUND & AIMS: The mechanisms whereby intracellular messengers mediate zymogen granule transport and exocytosis in the pancreatic acinar cell are not well defined. Electron microscopy has shown a periluminal network of actin in the acinar cell, suggesting a role for actin and myosin in the transport process. The possible involvement of two types of myosin in the secretory process was investigated, and their distribution in acinar cells was determined. METHODS: Antibodies specific to myosin I or to myosin II were used for immunocytochemistry and Western blot analysis. Ultrastructural studies were also performed. RESULTS: Western blot analysis showed that myosin I and myosin II were present in total pancreatic homogenate but that only myosin I was present on isolated zymogen granules and their membranes. By immunocytochemistry, myosin I was shown in the apical aspect of acinar cells colocalized with glycoprotein 2, a marker for zymogen granules, and actin. By immunocytochemistry, myosin I was also localized on isolated zymogen granules. CONCLUSIONS: The immunolocalization of myosin I to zymogen granule membranes and its close association with periluminal actin suggest that myosin I plays a direct role in the process of transport and exocytosis of zymogen granules in the pancreatic acinar cell.


Subject(s)
Cytoplasmic Granules/chemistry , Enzyme Precursors/analysis , Myosins/analysis , Pancreas/cytology , Actins/analysis , Actins/physiology , Amino Acid Sequence , Animals , Antibodies/analysis , Antibodies/immunology , Blotting, Western , Cytoplasmic Granules/ultrastructure , Electrophoresis, Polyacrylamide Gel , Exocytosis/physiology , Immunohistochemistry , Membrane Proteins/analysis , Membrane Proteins/physiology , Microscopy, Confocal , Microscopy, Electron , Myosins/immunology , Myosins/physiology , Pancreas/chemistry , Pancreas/metabolism , Rats , Rats, Sprague-Dawley
19.
Pancreas ; 14(2): 133-41, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9057185

ABSTRACT

The present study was designed to examine the role of Ca2+ in the regulation of digestive enzyme synthesis, to determine whether changes in intracellular Ca2+ stores or cytosolic Ca2+ caused the observed effects, and to establish the steps in the pathway of protein synthesis where the regulation occurs. Protein synthesis, polysome size, and the ratio of completed to nascent polypeptides were measured as a function of Ca2+ in the intracellular stores and the cytoplasm of pancreatic acinar cells. Rat acini and rabbit pancreatic lobules were incubated in media containing 1 mM CaCl2 with the following additives: cholecystokinin (CCK) octapeptide; the inhibitors of microsomal Ca2+ ATPase, thapsigargin (THP) and 2,5-di(tertbutyl)-hydroquinone (BHQ); the intracellular Ca2+ chelator, 1,2-bis(O-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA); an inhibitor of translational initiation, 7-methylguanosine 5'-triphosphate; and an inhibitor of translation elongation, cyclohexamide. THP and BHQ depleted intracellular pools of Ca2+ and caused a sustained elevation in cytosolic [Ca2+]. Under these conditions, the polysome size diminished, and the ratio of completed proteins increased twofold relative to nascent polypeptides despite an overall decrease in net protein synthesis (55.3 +/- 2.7% of control). These effects paralleled those caused by incubation with 1 nM CCK. Incubation of pancreatic acini with BAPTA plus THP or BHQ depleted the pool [Ca2+] without changing the cytosolic [Ca2+]. In addition, these agents decreased the net protein synthesis (30.1 +/- 3.6% compared to control) and polysome size and increased the ratio of completed to nascent polypeptides to 2:1. These results suggest that depletion of intracellular stores of Ca2+ without changes in cytosolic [Ca2+] decreases protein synthesis at translational initiation.


Subject(s)
Calcium/physiology , Pancreas/enzymology , Protein Biosynthesis , Animals , Calcium Chloride/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Chelating Agents , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Hydroquinones/pharmacology , Kinetics , Pancreas/ultrastructure , Polyribosomes/ultrastructure , Rabbits , Rats , Sincalide/pharmacology , Thapsigargin/pharmacology
20.
J Biol Chem ; 272(50): 31435-40, 1997 Dec 12.
Article in English | MEDLINE | ID: mdl-9395476

ABSTRACT

The regulation of cytosolic Ca2+ is important for a variety of cell functions. One non-inositol 1,4,5-trisphosphate (IP3) compound that may regulate Ca2+ is palmitoyl-coenzyme A (CoA), a fatty acid-CoA that is reported to cause Ca2+ release from intracellular stores of oocytes, myocytes, and hepatocytes. To study the role of palmitoyl-CoA in the pancreatic acinar cell, rat pancreatic acini were isolated by collagenase digestion, permeablized with streptolysin O, and the release of Ca2+ from internal stores was measured with fura-2. Palmitoyl-CoA released Ca2+ from internal stores (EC50 = 14 microM). The palmitoyl-CoA-sensitive pool was distinct from, and overlapping with the IP3-sensitive Ca2+ pool. The effects of submaximal doses of IP3 or cyclic ADP-ribose plus palmitoyl-CoA were additive. Fatty acid-CoA derivatives with carbon chain lengths of 16-18 were the most potent and efficacious. Ryanodine and caffeine or elevated resting [Ca2+] sensitized the Ca2+ pool to the actions of palmitoyl-CoA. Fatty acid-CoA levels in pancreatic acini were measured by extraction with 2-propanol/acetonitrile, followed by separation and quantification using reverse phase high performance liquid chromatography, and were found to be 10.17 +/- 0.93 nmol/mg protein. These data suggest the presence of an IP3-insensitive palmitoyl-CoA-sensitive Ca2+ store in pancreatic acinar cells and suggest that palmitoyl-CoA may be needed for Ca2+-induced Ca2+ release.


Subject(s)
Acyl Coenzyme A/metabolism , Calcium/metabolism , Pancreas/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Caffeine/pharmacology , Calcium-Transporting ATPases/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Palmitoyl Coenzyme A/administration & dosage , Palmitoyl Coenzyme A/metabolism , Palmitoyl Coenzyme A/pharmacology , Rats , Ryanodine/pharmacology , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...