Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Nature ; 384(6607): 327-33, 1996 Nov 28.
Article in English | MEDLINE | ID: mdl-8934515

ABSTRACT

The gene apparently responsible for a heritable form of murine pituitary-dependent dwarfism (Ames dwarf, df) has been positionally cloned, identifying a novel, tissue-specific, paired-like homeodomain transcription factor, termed Prophet of Pit-1 (Prop-1). The df phenotype results from an apparent failure of initial determination of the Pit-1 lineage required for production of growth hormone, prolactin or thyroid-stimulating hormone, resulting in dysmorphogenesis and failure to activate Pit-1 gene expression. These results imply that a cascade of tissue-specific regulators is responsible for the determination and differentiation of specific cell lineages in pituitary organogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Dwarfism, Pituitary/genetics , Homeodomain Proteins/genetics , Pituitary Gland, Anterior/embryology , Transcription Factors/genetics , Alleles , Amino Acid Sequence , Animals , Cell Lineage , Dwarfism, Pituitary/embryology , Female , Gene Expression , Homeodomain Proteins/physiology , Hypothalamus/physiology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pituitary Gland, Anterior/abnormalities , Pituitary Gland, Anterior/physiology , Point Mutation , Sequence Homology, Amino Acid , Signal Transduction , Transcription Factor Pit-1
13.
Nature ; 364(6434): 208-13, 1993 Jul 15.
Article in English | MEDLINE | ID: mdl-8391647

ABSTRACT

The molecular basis for the little (lit) mouse phenotype, characterized by a hypoplastic anterior pituitary gland, is the mutation of a single nucleotide that alters Asp 60 to Gly in the growth hormone releasing factor receptor. Detailed analysis of the lit mouse anterior pituitary reveals spatially distinct proliferative zones of growth hormone-producing stem cells and mature somatotrophs, each regulated by a different trophic factor. This sequential growth factor requirement for a specific cell type may exemplify a common strategy for regulating cellular proliferation in other mammalian organs.


Subject(s)
Dwarfism, Pituitary/genetics , Growth Hormone-Releasing Hormone/physiology , Mutation , Pituitary Gland, Anterior/cytology , Receptors, Neuropeptide , Receptors, Neurotransmitter/genetics , Receptors, Pituitary Hormone-Regulating Hormone , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Differentiation/genetics , Cell Division/genetics , Chromosome Mapping , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Oligodeoxyribonucleotides , Phenotype , Pituitary Gland, Anterior/pathology , Promoter Regions, Genetic , Receptors, Neurotransmitter/physiology , Signal Transduction/genetics , Transcription Factor Pit-1 , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...