Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Gut ; 58(3): 431-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18596195

ABSTRACT

BACKGROUND: Mechanisms of acinar cell death in pancreatitis are poorly understood. Cytochrome c release is a central event in apoptosis in pancreatitis. Here, we assessed the regulation of pancreatic cytochrome c release by Ca(2+), mitochondrial membrane potential (Delta Psi m), and reactive oxygen species (ROS), the signals involved in acute pancreatitis. We used both isolated rat pancreatic mitochondria and intact acinar cells hyperstimulated with cholecystokinin-8 (CCK-8; in vitro model of acute pancreatitis). RESULTS: Micromolar amounts of Ca(2+) depolarised isolated pancreatic mitochondria through a mechanism different from the "classical" (ie, liver) mitochondrial permeability transition pore (mPTP). In contrast with liver, Ca(2+)-induced mPTP opening caused a dramatic decrease in ROS and was not associated with pancreatic mitochondria swelling. Importantly, we found that Ca(2+)-induced depolarisation inhibited cytochrome c release from pancreatic mitochondria, due to blockade of ROS production. As a result, Ca(2+) exerted two opposite effects on cytochrome c release: Ca(2+) per se stimulated the release, whereas Ca(2+)-induced depolarisation inhibited it. This dual effect caused a non-monotonous dose-dependence of cytochrome c release on Ca(2+). In intact acinar cells, cytochrome c release, caspase activation and apoptosis were all stimulated by ROS and Ca(2+), and inhibited by depolarisation, corroborating the findings on isolated pancreatic mitochondria. CONCLUSIONS: These data implicate ROS as a key mediator of CCK-induced apoptotic responses. The results indicate a major role for mitochondria in the effects of Ca(2+ )and ROS on acinar cell death. They suggest that the extent of apoptosis in pancreatitis is regulated by the interplay between ROS, Delta Psi m and Ca(2+). Stabilising mitochondria against loss of Delta Psi m may represent a strategy to mitigate the severity of pancreatitis.


Subject(s)
Cytochromes c/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Pancreas/metabolism , Pancreatitis/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/physiology , Calcium/metabolism , Calcium Signaling , Cell Death/physiology , Membrane Potential, Mitochondrial/physiology , Pancreas/physiology , Pancreatitis/physiopathology , Rats
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Gastroenterology ; 111(4): 1081-91, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8831604

ABSTRACT

BACKGROUND & AIMS: Pancreatitis is characterized by inflammation and death of acinar cells. Death can occur by either necrosis or apoptosis. The initial injury may cause expression of cytokines that mediate activation and infiltration of neutrophils. The aim of this study was to assess the effect of neutrophils and platelet-activating factor (PAF) in cell death responses. METHODS: The effects of neutrophil depletion with antineutrophil serum (ANS) and a PAF antagonist (BN52021) were measured in the cerulein model of pancreatitis. Rats received a 6-hour intravenous infusion of cerulein either alone or after treatment with ANS, BN52021, or both. RESULTS: Cerulein-induced pancreatitis was characterized by neutrophilic infiltration, vacuolization of acinar cells, and foci of necrosis. Treatment with ANS and BN52021 prevented the inflammatory response caused by cerulein and decreased the cell damage. Treatment with ANS increased apoptosis in cerulein-infused animals. When BN52021 was added, apoptosis was abolished. The measurement of PAF in pancreatic tissue showed a ninefold increase with cerulein treatment alone and a 14-fold increase in cerulein-infused, neutrophil-depleted animals. CONCLUSIONS: The results indicate that cerulein stimulates pancreatic production of PAF. PAF mediates both apoptosis and neutrophil chemotaxis in the pancreas. Neutrophils in turn may convert acinar cells undergoing apoptosis into necrotic cells.


Subject(s)
Diterpenes , Neutrophils/physiology , Pancreatitis/etiology , Platelet Activating Factor/physiology , Acute Disease , Amylases/metabolism , Animals , Ceruletide , Ginkgolides , Lactones/pharmacology , Lipase/metabolism , Male , Pancreatitis/metabolism , Pancreatitis/pathology , Rats , Rats, Sprague-Dawley
15.
Gastroenterology ; 110(3): 875-84, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8608898

ABSTRACT

BACKGROUND & AIMS: Mechanisms of cell death in human and experimental pancreatitis remain poorly understood. The aim of this study was to determine the mechanisms of cell death, apoptosis vs. necrosis, in models of pancreatitis induced by pancreatic duct ligation in the opossum and rat. METHODS: Morphological changes were shown by light and electron microscopy, chromatin condensation and DNA breaks were assessed using Hoechst 33258 staining and DNA nickend labeling, and DNA fragmentation was characterized b gel electrophoresis. RESULTS: Cells with morphology of both necrosis and apoptosis were found in pancreata from duct-ligated animals; however, the apoptosis-necrosis ratio was different for the opossum and the rat. Both apoptotic and necrotic cells stained for breaks in DNA, suggesting that both apoptosis and necrosis are associated with DNA fragmentation and that DNA end labeling cannot specifically detect apoptotic cells. No significant oligonucleosomal DNA fragmentation was found in pancreas from duct-ligated animals, although the endonuclease responsible for oligonucleosomal DNA fragmentation was present in pancreatic nuclei. CONCLUSIONS: Duct ligation resulted predominantly in apoptosis in the rat and necrosis in the opossum. The differences in types of cell death in the two models may result from differences in inflammatory cell infiltration.


Subject(s)
Pancreatic Ducts/surgery , Pancreatitis/pathology , Animals , Apoptosis , Cell Death , DNA/analysis , DNA Damage , Disease Models, Animal , Ligation , Microscopy, Electron , Necrosis , Opossums , Pancreas/chemistry , Pancreas/pathology , Pancreas/ultrastructure , Pancreatitis/etiology , Pancreatitis/metabolism , Rats , Rats, Sprague-Dawley
16.
Am J Physiol ; 268(6 Pt 1): G900-7, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7541946

ABSTRACT

Regulation of guanosine 3',5'-cyclic monophosphate (cGMP) formation by calcium and calcium-binding proteins was studied at the levels of nitric oxide synthase (NOS) and guanylyl cyclase (GC) in dispersed pancreatic acini isolated from guinea pig. In intact cells, in the cytosol, and on diethylaminoethyl fractions from cytosolic proteins, GC activity was negatively regulated by Ca2+. An increase in Ca2+ concentration ([Ca2+]) from 25 to 950 nM suppressed cGMP formation by 85%. On the other hand, NOS was stimulated by agents increasing cytosolic [Ca2+] and inhibited by intracellular Ca2+ chelators. Thus Ca2+ regulates cGMP production in opposite directions by activating NOS and inhibiting GC. Calmodulin antagonists W-7, trifluoperazine, and R-24571 inhibited NOS, suggesting that the enzyme is regulated by calmodulin as in other cell types. Calmodulin antagonists appeared to inhibit GC. In particular, 200 microM W-7 completely abolished the cGMP rise evoked by the nitric oxide donor, nitroprusside. The effect was not reversed by addition of excess calmodulin. The findings suggest that the negative regulation of GC by Ca2+ is due to factors other than calmodulin but affected by calmodulin antagonists.


Subject(s)
Calcium/pharmacology , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Pancreas/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Alkaloids/pharmacology , Amino Acid Oxidoreductases/metabolism , Animals , Calcium-Binding Proteins/metabolism , Calmodulin/antagonists & inhibitors , Cytosol/metabolism , Guinea Pigs , Homeostasis , Imidazoles/pharmacology , In Vitro Techniques , Isoquinolines/pharmacology , Kinetics , Nitric Oxide Synthase , Pancreas/cytology , Pancreas/drug effects , Piperazines/pharmacology , Protein Kinase C/antagonists & inhibitors , Staurosporine , Sulfonamides/pharmacology , Trifluoperazine/pharmacology
17.
Biochim Biophys Acta ; 1195(1): 45-50, 1994 Oct 12.
Article in English | MEDLINE | ID: mdl-7918564

ABSTRACT

The purpose of present study was to examine the effects of sphingosine on cellular Ca2+ transports using dispersed rat pancreatic acini. The results demonstrated that sphingosine had a specific effect to inhibit Ca2+ uptake into the cell's agonist-sensitive pool as well as inhibiting microsomal Ca(2+)-ATPase. The ability of sphingosine to inhibit Ca2+ uptake resulted in both augmentation of Ca2+ release from the pool by inositol 1,4,5-trisphosphate (IP3) and conversion of the Ca2+ release by inositol 1,4,5-trisphosphate from a transient response to a sustained response. Furthermore, by preventing Ca2+ pool refilling sphingosine mimicked the effect of the agonist, carbachol, to maintain an increased [Ca2+]i during sustained stimulation. These results suggest that regulation of Ca(2+)-ATPase by sphingosine or a sphingosine-like agent mediates some of the effects of agonist on cell Ca2+ transports.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Pancreas/drug effects , Sphingosine/pharmacology , Animals , Calcium-Transporting ATPases/antagonists & inhibitors , Carbachol/pharmacology , Cell Membrane Permeability , Cells, Cultured , Dose-Response Relationship, Drug , Fura-2 , Inositol 1,4,5-Trisphosphate/pharmacology , Microsomes/drug effects , Microsomes/enzymology , Pancreas/metabolism , Rats , Sincalide/pharmacology
18.
Ann N Y Acad Sci ; 713: 41-8, 1994 Mar 23.
Article in English | MEDLINE | ID: mdl-8185204

ABSTRACT

Figure 1 summarizes our current concept of a signaling mechanism to explain agonist-induced Ca2+ entry in the pancreatic acinar cell. We propose that cGMP can modulate Ca2+ entry under conditions of internal Ca2+ store depletion and that the NO signaling system may be involved in coupling Ca2+ depletion to cGMP formation. The finding that Ca2+ entry after Ca2+ store depletion can occur with no elevation in [Ca2+]i37 raises the possibility that alternative signaling pathways may converge to stimulate cGMP formation or that additional messengers may activate plasmalemmal Ca2+ entry mechanisms in parallel.


Subject(s)
Calcium/metabolism , Cholecystokinin/pharmacology , Cyclic GMP/physiology , Pancreas/physiology , Acetylcholine/pharmacology , Animals , Cholecystokinin/physiology , Models, Biological , Pancreas/cytology , Pancreas/drug effects , Second Messenger Systems , Signal Transduction
19.
Am J Physiol ; 266(3 Pt 1): G350-6, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8166275

ABSTRACT

Guanosine 3',5'-cyclic monophosphate (cGMP) rise is one of the early events in neurotransmitter or hormone-induced cascade of reactions in pancreatic acinar cells. The mechanism of agonist-stimulated guanylyl cyclase activation in these cells remains, however, unknown. In the present work, mechanisms of cGMP rise, as well as of Ca2+ influx, induced by carbachol were studied on acinar cells isolated from rat and guinea pig pancreas. In both types of acinar cells, blocking nitric oxide (NO) production by inhibitors of NO synthase, NG-monomethyl-L-arginine (L-NMMA) or NG-nitro-L-arginine, abolished carbachol-induced cGMP rise in a dose-dependent manner. The inhibition was reversed by addition of excess L-arginine. L-NMMA also caused inhibition of the basal cGMP level, suggesting a role for NO in cGMP homeostasis in resting cells. Carbachol was found to increase [3H]arginine conversion to [3H]citrulline. This conversion was inhibited by L-NMMA. By contrast, inhibition of carbon monoxide production by Zn-protoporphyrin did not affect carbachol-stimulated cellular cGMP levels. There was no increase in cellular cGMP levels in response to exogenous arachidonic acid (AA). Blocking of lipoxygenase oxidation of AA by nordihydroguaiaretic acid did not produce any changes in carbachol-induced cGMP rise. Indomethacin, a cyclooxygenase inhibitor, increased basal cGMP level through L-NMMA-sensitive mechanism. Blockade of NO production inhibited carbachol-induced increase in 45Ca2+ uptake in both guinea pig and rat acinar cells. The concentration-response curves for inhibition by L-NMMA of 45Ca2+ uptake and cGMP formation were superimposable. L-NMMA also suppressed stimulation of Mn2+ quenching by carbachol in fura 2-loaded acini.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Calcium/metabolism , Cyclic GMP/biosynthesis , Nitric Oxide/biosynthesis , Pancreas/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Carbachol/pharmacology , Guinea Pigs , Indomethacin/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitroarginine , Pancreas/cytology , Rats , omega-N-Methylarginine
20.
FEBS Lett ; 324(3): 274-6, 1993 Jun 21.
Article in English | MEDLINE | ID: mdl-8405365

ABSTRACT

The role of intracellular Ca2+ and H+ concentrations in radiation-induced interphase death of rat thymocytes has been studied. In response to concanavalin A treatment in the Ca(2+)-containing medium, or to the CaCl2 treatment in the Ca(2+)-free medium, the [Ca2+]i rise in irradiated cells was as in the non-treated cells. No changes in the level of [Ca2+]i and pHi were found within 1 h after irradiation of thymocytes with a dose of 6 Gy. 15 microM 5-(N-ethyl-N-isopropyl)-amiloride, an inhibitor of Na+/H+ exchange, did not affect the DNA fragmentation. The fragmentation was prevented by 2-4 microM (1-[bis(4-chlorophenyl)methyl]-3-[2-(2,4-dichlorophenyl)]-2-[(2,4- dichlorophenyl)-methoxy]-ethyl)-1-H-imidazolium chloride, an inhibitor of calmodulin. The above data indicate that triggering of interphase death in irradiated thymocytes is not mediated by changes in either [Ca2+]i or pHi. Such changes seem to be involved in intermediate steps of the interphase death process.


Subject(s)
Antiporters/metabolism , Calcium/metabolism , Hydrogen-Ion Concentration , Thymus Gland/radiation effects , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Antiporters/antagonists & inhibitors , Biological Transport/drug effects , Calmodulin/antagonists & inhibitors , Cell Death , Concanavalin A/pharmacology , DNA Damage/drug effects , DNA Damage/radiation effects , Imidazoles/pharmacology , Male , Radiation Injuries, Experimental/metabolism , Rats , Rats, Wistar , Thymus Gland/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...