Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochem Soc Trans ; 31(Pt 3): 654-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773176

ABSTRACT

Bacterial and synthetic DNA containing unmethylated 2'-deoxyribo(cytidine-phosphate-guanosine) (CpG) dinucleotides in specific sequence contexts activate the vertebrate innate immune system. A molecular pattern recognition receptor, Toll-like receptor 9 (TLR9), recognizes CpG DNA and initiates the signalling cascade, although a direct interaction between CpG DNA and TLR9 has not been demonstrated yet. TLR9 in different species exhibits sequence specificity. Our extensive structure-immunostimulatory activity relationship studies showed that a number of synthetic pyrimidine (Y) and purine (R) nucleotides are recognized by the receptor as substitutes for the natural nucleotides deoxycytidine and deoxyguanosine in a CpG dinucleotide. These studies permitted development of synthetic YpG, CpR and YpR immunostimulatory motifs, and showed divergent nucleotide motif recognition pattern of the receptor. Surprisingly, we found that synthetic immunostimulatory motifs produce different cytokine induction profiles compared with natural CpG motifs. Importantly, we also found that some of these synthetic immunostimulatory motifs show optimal activity in both mouse and human systems without the need to change sequences, suggesting an overriding of the species-dependent specificity of the receptor by the use of synthetic motifs. In the present paper, we review current understanding of structural recognition and functional modulation of TLR9 receptor by second-generation synthetic CpG DNAs and their potential application as wide-spectrum therapeutic agents.


Subject(s)
DNA-Binding Proteins/genetics , DNA/genetics , Receptors, Cell Surface/genetics , Animals , Base Sequence , Cytosine , DNA/chemical synthesis , DNA-Binding Proteins/physiology , Dinucleoside Phosphates/genetics , Genes, Synthetic , Guanine , Receptors, Cell Surface/physiology , Signal Transduction , Toll-Like Receptor 9 , Vertebrates
2.
Gut ; 50(1): 78-83, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11772971

ABSTRACT

BACKGROUND AND AIM: Recent studies have indicated that prior thermal stress causes upregulation of heat shock protein 70 (HSP70) expression in the pancreas and protects against secretagogue induced pancreatitis. The mechanisms responsible for the protective effect are not known. Similarly, the effects of prior non-thermal stress on HSP70 expression and pancreatitis are not known. The current studies were designed to specifically address these issues. METHODS: In the current studies pancreatitis was induced by administration of a supramaximally stimulating dose of caerulein 12 hours after thermal stress and 24 hours after non-thermal (that is, beta adrenergic stimulation) stress. RESULTS: Both thermal and non-thermal stresses caused pancreatic HSP70 levels to rise and resulted in increased expression of HSP70 in acinar cells. Both forms of stresses protected against caerulein induced pancreatitis and prevented the early intrapancreatic activation of trypsinogen which occurs in this model of pancreatitis. CONCLUSIONS: These results suggest that both thermal and non-thermal stresses protect against pancreatitis by preventing intrapancreatic digestive enzyme activation and that HSP70 may mediate this protective effect.


Subject(s)
Hyperthermia, Induced/methods , Pancreatitis/enzymology , Stress, Physiological/physiopathology , Trypsinogen/physiology , Amylases/physiology , Analysis of Variance , Animals , Blotting, Western , Ceruletide , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , HSP70 Heat-Shock Proteins/physiology , Luminescent Measurements , Male , Pancreatitis/chemically induced , Peroxidase/physiology , Rats , Rats, Wistar
3.
J Clin Invest ; 108(9): 1387-95, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696584

ABSTRACT

Intra-acinar cell activation of digestive enzyme zymogens including trypsinogen is generally believed to be an early and critical event in acute pancreatitis. We have found that the phosphatidylinositol 3-kinase inhibitor wortmannin can reduce the intrapancreatic activation of trypsinogen that occurs during two dissimilar experimental models of rodent acute pancreatitis, secretagogue- and duct injection-induced pancreatitis. The severity of both models was also reduced by wortmannin administration. In contrast, the NF-kappa B activation that occurs during the early stages of secretagogue-induced pancreatitis is not altered by administration of wortmannin. Ex vivo, caerulein-induced trypsinogen activation is inhibited by wortmannin and LY294002. However, the cytoskeletal changes induced by caerulein were not affected by wortmannin. Concentrations of caerulein that induced ex vivo trypsinogen activation do not significantly increase phosphatidylinositol-3,4-bisphosphate or phosphatidylinositol 3,4,5-trisphosphate levels or induce phosphorylation of Akt/PKB, suggesting that class I phosphatidylinositol 3-kinases are not involved. The concentration of wortmannin that inhibits trypsinogen activation causes a 75% decrease in phosphatidylinositol 3-phosphate, which is implicated in vesicle trafficking and fusion. We conclude that a wortmannin-inhibitable phosphatidylinositol 3-kinase is necessary for intrapancreatic activation of trypsinogen and regulating the severity of acute pancreatitis. Our observations suggest that phosphatidylinositol 3-kinase inhibition might be of benefit in preventing acute pancreatitis.


Subject(s)
Pancreatitis/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Trypsinogen/metabolism , Acute Disease , Androstadienes/pharmacology , Animals , Cells, Cultured , Ceruletide/metabolism , Chromones/pharmacology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Lysosomes/metabolism , Male , Mice , Morpholines/pharmacology , NF-kappa B/metabolism , Necrosis , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Rats , Time Factors , Wortmannin
4.
Gastroenterology ; 120(7): 1818-27, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375962

ABSTRACT

BACKGROUND & AIMS: The present study was undertaken to evaluate the role of serine proteases in regulating digestive enzyme secretion in pancreatic acinar cells. METHODS: Isolated acini were stimulated by various secretagogues in the presence or absence of cell-permeant serine protease inhibitors 4-(2-aminoethyl)-benzenesulfonyl fluoride and N(alpha)-p-tosyl-L-phenylalanine chloromethyl ketone. F-actin distribution was studied after staining with rhodamine phalloidin. RESULTS: Both cell-permeant serine protease inhibitors blocked amylase secretion in response to secretagogues that use calcium as a second messenger (e.g., cerulein, carbamylcholine, and bombesin) but not to those that use adenosine 3',5'-cyclic monophosphate (cAMP) as a second messenger (e.g., secretin and vasoactive intestinal polypeptide). Incubation of the acini with these inhibitors also resulted in a dramatic redistribution of the F-actin cytoskeleton. This redistribution was energy dependent. Similar redistribution of F-actin from the apical to the basolateral region was also observed when acini were incubated with a supramaximally stimulating concentration of cerulein, which is known to inhibit secretion. CONCLUSIONS: These results suggest that a serine protease activity is essential for maintaining the normal apical F-actin distribution; its inhibition redistributes F-actin from the apical to the basolateral region and blocks secretion induced by secretagogues that act via calcium. cAMP reverses the F-actin redistribution and hence cAMP-mediated secretion is not affected.


Subject(s)
Actins/metabolism , Calcium/physiology , Pancreas/drug effects , Serine Proteinase Inhibitors/pharmacology , Sulfones/pharmacology , Adenosine Triphosphate/physiology , Animals , Bucladesine/pharmacology , Calcineurin/metabolism , Calpain/metabolism , Ceruletide/pharmacology , Cyclic AMP/physiology , Male , Pancreas/enzymology , Pancreas/ultrastructure , Protein Kinase C/metabolism , Rats , Rats, Wistar
5.
Am J Physiol Gastrointest Liver Physiol ; 280(5): G974-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11292607

ABSTRACT

Complement factor C5a acting via C5a receptors (C5aR) is recognized as an anaphylotoxin and chemoattractant that exerts proinflammatory effects in many pathological states. The effects of C5a and C5aR in acute pancreatitis and in pancreatitis-associated lung injury were evaluated using genetically altered mice that either lack C5aR or do not express C5. Pancreatitis was induced by administration of 12 hourly injections of cerulein (50 microg/kg ip). The severity of pancreatitis was determined by measuring serum amylase, neutrophil sequestration in the pancreas, and acinar cell necrosis. The severity of lung injury was evaluated by measuring neutrophil sequestration in the lung and pulmonary microvascular permeability. In both strains of genetically altered mice, the severity of pancreatitis and pancreatitis-associated lung injury was greater than that noted in the comparison wild-type strains of C5aR- and C5-sufficient animals. This exacerbation of injury in the absence of C5a function indicates that, in pancreatitis, C5a exerts an anti-inflammatory effect. Potentially, C5a and its receptor are capable of both promoting and reducing the extent of acute inflammation.


Subject(s)
Antigens, CD/physiology , Complement C5a/physiology , Lung/physiopathology , Pancreatitis/physiopathology , Receptors, Complement/physiology , Acute Disease , Animals , Anti-Inflammatory Agents , Antigens, CD/genetics , Capillaries/pathology , Capillaries/physiopathology , Ceruletide , Complement C5a/deficiency , Complement C5a/genetics , Crosses, Genetic , Inflammation , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced , Pancreatitis/pathology , Peroxidase/analysis , Receptor, Anaphylatoxin C5a , Receptors, Complement/deficiency , Receptors, Complement/genetics
6.
J Biol Chem ; 276(22): 18742-7, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278554

ABSTRACT

Prior stress ameliorates caerulein-induced pancreatitis in rats. NF-kappaB is a proinflammatory transcription factor activated during caerulein pancreatitis. However, the effects of prior stress on pancreatic NF-kappaB activation are unknown. In the current study, the effect of prior water immersion stress on caerulein and tumor necrosis factor-alpha (TNF-alpha)-induced NF-kappaB activation in the pancreas was evaluated. Water immersion of rats for up to 6 h prevents supramaximal caerulein-induced pancreatic IkappaB-alpha degradation and NF-kappaB activation in vivo. NF-kappaB activity is also inhibited in vitro in pancreatic acini prepared from water-immersed animals. TNF-alpha-induced NF-kappaB activation in pancreas or in pancreatic acini is unaffected by prior water immersion. Chelation of intracellular Ca(2+) by 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetate/acetoxymethyl ester has similar effects to water immersion in preventing caerulein but not TNF-alpha-induced NF-kappaB activation in pancreas. Both the spike response and the sustained rise in [Ca(2+)](i) in response to supramaximal caerulein stimulation are reduced markedly in acini prepared from water-immersed animals as compared with normal animals. Our findings indicate that, in addition to Ca(2+)-dependent mechanisms, Ca(2+)-independent signaling events also may lead to NF-kappaB activation in pancreatic acinar cells. Water immersion stress prevents supramaximal caerulein-induced NF-kappaB activation in pancreas in vivo and in vitro by affecting intracellular Ca(2+) homeostasis.


Subject(s)
Calcium/metabolism , Ceruletide/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , Pancreas/metabolism , Water/metabolism , Animals , Blotting, Western , Chelating Agents/pharmacology , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Male , NF-KappaB Inhibitor alpha , Rats , Rats, Wistar , Signal Transduction , Stress, Physiological , Time Factors , Tumor Necrosis Factor-alpha/metabolism
7.
Biochem Biophys Res Commun ; 280(1): 388-95, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11162528

ABSTRACT

Intra-acinar cell nuclear factor-kappaB (NF-kappaB) and trypsinogen activation are early events in secretagogue-induced acute pancreatitis. We have studied the relationship between NF-kappaB and trypsinogen activation in rat pancreas. CCK analogue caerulein induces early (within 15 min) parallel activation of both NF-kappaB and trypsinogen in pancreas in vivo as well as in pancreatic acini in vitro. However, NF-kappaB activation can be induced without trypsinogen activation by lipopolysaccharide in pancreas in vivo and by phorbol ester in pancreatic acini in vitro. Stimulation of acini with caerulein after 6 h of culture results in NF-kappaB but not trypsinogen activation. Protease inhibitors (AEBSF, TLCK, and E64d) inhibit both intracellular trypsin activity and NF-kappaB activation in caerulein stimulated acini. A chymotrypsin inhibitor (TPCK) inhibits NF-kappaB activation but not trypsin activity. The proteasome inhibitor MG-132 prevents caerulein-induced NF-kappaB activation but does not prevent trypsinogen activation. These findings indicate that although caerulein-induced NF-kappaB and trypsinogen activation are temporally closely related, they are independent events in pancreatic acinar cells. NF-kappaB activation per se is not required for the development of early acinar cell injury by supramaximal secretagogue stimulation.


Subject(s)
Ceruletide/pharmacology , NF-kappa B/metabolism , Pancreas/metabolism , Trypsinogen/metabolism , Animals , Cells, Cultured , Cysteine Endopeptidases/metabolism , DNA/metabolism , Enzyme Activation , Kinetics , Leupeptins/pharmacology , Male , Multienzyme Complexes/metabolism , Pancreas/drug effects , Proteasome Endopeptidase Complex , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Trypsin/metabolism
8.
J Clin Invest ; 106(1): 81-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10880051

ABSTRACT

Rodents given a supramaximally stimulating dose of cholecystokinin or its analogue cerulein develop acute pancreatitis with acinar cell injury, pancreatic inflammation, and intrapancreatic digestive enzyme (i.e., trypsinogen) activation. Prior thermal stress is associated with heat shock protein 70 (HSP70) expression and protection against cerulein-induced pancreatitis. However, thermal stress can also induce expression of other HSPs. The current studies were performed using an in vitro system to determine whether HSP70 can actually mediate protection against pancreatitis and, if so, to define the mechanism underlying that protection. We show that in vitro exposure of freshly prepared rat pancreas fragments to a supramaximally stimulating dose of cerulein results in changes similar to those noted in cerulein-induced pancreatitis, i.e., intra-acinar cell trypsinogen activation and acinar cell injury. Short-term culture of the fragments results in HSP70 expression and loss of the pancreatitis-like changes noted after addition of cerulein. The culture-induced enhanced HSP70 expression can be prevented by addition of either the flavonoid antioxidant quercetin or an antisense oligonucleotide to HSP70. Under these latter conditions, addition of a supramaximally stimulating concentration of cerulein results in trypsinogen activation and acinar cell injury. These findings indicate that the protection against cerulein-induced pancreatitis that follows culture-induced (and possibly thermal) stress is mediated by HSP70. They suggest that the HSP acts by preventing trypsinogen activation within acinar cells.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Pancreas/pathology , Trypsinogen/metabolism , Animals , Ceruletide/pharmacology , Enzyme Activation , HSP70 Heat-Shock Proteins/analysis , Oligonucleotides, Antisense/pharmacology , Oligopeptides/analysis , Organ Culture Techniques , Quercetin/pharmacology , Rats , Rats, Wistar
9.
Gastroenterology ; 119(1): 220-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10889172

ABSTRACT

BACKGROUND & AIMS: Heat shock proteins (Hsps), induced by cell stress, are known to protect against cellular injury. Recent studies have indicated that Hsp60 expression, induced by exposure to water immersion stress, protects against pancreatitis induced by administration of supramaximal doses of cerulein in rats. However, the mechanisms responsible for this protection are not known. METHODS: Rats were water-immersed for 3-12 hours. Pancreatitis was induced by cerulein administration. RESULTS: The results confirm that prior induction of Hsp60 expression by water-immersion stress significantly ameliorates the severity of cerulein-induced pancreatitis as judged by the markedly reduced degree of hyperamylasemia, pancreatic edema, and acinar cell necrosis. Water immersion also prevents the subcellular redistribution of cathepsin B from a lysosome-enriched fraction to a heavier, zymogen granule-enriched fraction that is known to occur in this model of pancreatitis. Intra-acinar cell activation of trypsinogen that occurs shortly after exposure to a supramaximally stimulating dose of cerulein both in vivo and in vitro is prevented by prior water-immersion stress and Hsp60 expression. The protection against pancreatitis that follows water-immersion stress is not caused by alterations of cholecystokinin receptors, because water immersion does not alter the typical biphasic amylase secretory response to stimulation with cerulein. CONCLUSIONS: Water-immersion stress induces Hsp60 expression, ameliorates cerulein-induced pancreatitis, and prevents intra-acinar cell activation of trypsinogen. We suggest that Hsp60 protects against cerulein-induced pancreatitis by preventing trypsinogen activation within acinar cells.


Subject(s)
Chaperonin 60/metabolism , Immersion , Pancreatitis/prevention & control , Stress, Physiological/physiopathology , Amylases/metabolism , Animals , Ceruletide/pharmacology , Immunohistochemistry , Male , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Rats , Rats, Wistar , Stress, Physiological/metabolism , Time Factors , Tissue Distribution , Water
10.
Am J Physiol ; 276(4): G835-42, 1999 04.
Article in English | MEDLINE | ID: mdl-10198325

ABSTRACT

The mechanisms responsible for intrapancreatic digestive enzyme activation as well as the relationship between that activation and cell injury during pancreatitis are not understood. We have employed an in vitro system in which freshly prepared pancreatic acini are exposed to a supramaximally stimulating concentration of the CCK analog caerulein to explore these issues. We find that in vitro trypsinogen activation depends on the continued presence of Ca2+ in the suspending medium and that it is half-maximal in the presence of 0.3 mM Ca2+. Caerulein-induced trypsinogen activation can be halted by removal of Ca2+ from the suspending medium or by chelation of intracellular Ca2+. Increasing intracellular Ca2+ with either ionomycin or thapsigargin does not induce trypsinogen activation. We have monitored cell injury by measuring the leakage of lactate dehydrogenase (LDH) from acini and by quantitating intercalation of propidium iodide (PI) into DNA. Leakage of LDH and intercalation of PI in response to supramaximal stimulation with caerulein can be detected only after caerulein-induced trypsinogen activation has already occurred, and these indications of cell injury can be prevented by addition of a cell-permeant protease inhibitor. Our findings indicate that caerulein-induced intra-acinar cell activation of trypsinogen depends on a rise in intracellular Ca2+, which reflects entry of Ca2+ from the suspending medium. Intra-acinar cell activation of trypsinogen is an early as well as a critical event in pancreatitis. The subsequent cell injury in this model is mediated by activated proteases.


Subject(s)
Calcium/metabolism , Pancreas/physiology , Trypsin/metabolism , Trypsinogen/metabolism , Animals , Calcium/pharmacology , Cells, Cultured , Ceruletide/toxicity , Enzyme Activation , Female , Ionomycin/pharmacology , Kinetics , Male , Pancreas/cytology , Pancreas/injuries , Rats , Rats, Wistar , Secretin/pharmacology , Thapsigargin/pharmacology
11.
Gastroenterology ; 116(3): 694-701, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10029629

ABSTRACT

BACKGROUND & AIMS: Intercellular adhesion molecule 1 (ICAM-1) and neutrophils play important roles in many inflammatory processes, but their importance in both acute pancreatitis and pancreatitis-associated lung injury has not been defined. METHODS: To address this issue, mice that do not express ICAM-1 were used and depleted of neutrophils by administration of antineutrophil serum. Pancreatitis was induced by administering either supramaximal doses of the secretagogue cerulein or feeding a choline-deficient, ethionine-supplemented diet. The severity of pancreatitis was evaluated by quantitating serum amylase, pancreatic edema, acinar cell necrosis, and pancreas myeloperoxidase activity (i.e., neutrophil content). Lung injury was evaluated by quantitating lung myeloperoxidase activity and pulmonary microvascular permeability. ICAM-1 was quantitated by enzyme-linked immunosorbent assay and was localized by light-microscopic immunohistochemistry. RESULTS: It was found that serum, pancreas, and lung ICAM-1 levels increase during pancreatitis. Both pancreatitis and the associated lung injury are blunted, but not completely prevented, in mice deficient in ICAM-1. Neutrophil depletion also reduces the severity of both pancreatitis and lung injury. However, the combination of neutrophil depletion with ICAM-1 deficiency does not reduce the severity of pancreatitis or lung injury to a greater extent than either neutrophil depletion or ICAM-1 deficiency alone. Neither pancreatitis nor pancreatitis-associated lung injury are completely prevented by ICAM-1 deficiency, neutrophil depletion, or combined ICAM-1 deficiency plus neutrophil depletion. CONCLUSIONS: The observations indicate that ICAM-1 plays an important, neutrophil-mediated, proinflammatory role in pancreatitis and pancreatitis-associated lung injury. The studies also indicate that ICAM-1 and neutrophil-independent events also contribute to the evolution of pancreatitis and lung injury in these models.


Subject(s)
Intercellular Adhesion Molecule-1/physiology , Lung/physiopathology , Microcirculation/physiology , Neutrophils/physiology , Pancreatitis/physiopathology , Pulmonary Circulation/physiology , Acute Disease , Animals , Capillary Permeability , Ceruletide , Choline Deficiency , Death , Ethionine/pharmacology , Female , Intercellular Adhesion Molecule-1/genetics , Lung/blood supply , Lung/pathology , Male , Mice , Mice, Knockout , Pancreatitis/blood , Pancreatitis/complications , Peroxidase/analysis
12.
Gastroenterology ; 115(5): 1238-47, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9797380

ABSTRACT

BACKGROUND & AIMS: Recent reports suggest that platelet-activating factor (PAF) plays a role in pancreatitis and pancreatitis-associated lung injury. In this study, the effects on these processes of termination of PAF action by recombinant PAF-acetylhydrolase (rPAF-AH) were investigated. METHODS: Rats were given rPAF-AH and then infused with a supramaximally stimulating dose of cerulein to induce mild pancreatitis. Opossums underwent biliopancreatic duct ligation to induce severe pancreatitis, and rPAF-AH administration was begun 2 days later. RESULTS: In mild, secretagogue-induced pancreatitis, rPAF-AH given before the cerulein reduced hyperamylasemia, acinar cell vacuolization, and pancreatic inflammation but did not alter pancreatic edema or pulmonary microvascular permeability. In severe, biliopancreatic duct ligation-induced pancreatitis, rPAF-AH delayed and reduced the extent of inflammation and acinar cell injury/necrosis and completely prevented lung injury even though the rPAF-AH administration was begun after the onset of pancreatitis. CONCLUSIONS: PAF plays an important role in the regulation of pancreatic injury but not pancreatic edema or increased pulmonary microvascular permeability in mild, secretagogue-induced pancreatitis. PAF plays a critical role in the regulation of progression of pancreatic injury and mediation of pancreatitis-associated lung injury in severe biliary pancreatitis. Amelioration of pancreatitis and prevention of pancreatitis-associated lung injury can be achieved with rPAF-AH even if treatment is begun after pancreatitis is established.


Subject(s)
Pancreatitis/drug therapy , Phospholipases A/therapeutic use , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Acute Disease , Animals , Bile Ducts , Ceruletide , Disease Models, Animal , Gastrointestinal Agents , Ligation , Lung/drug effects , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/etiology , Lung Diseases/pathology , Male , Opossums , Pancreas/drug effects , Pancreas/pathology , Pancreatic Ducts , Pancreatitis/chemically induced , Pancreatitis/complications , Pancreatitis/etiology , Platelet Activating Factor/analysis , Rats , Rats, Wistar , Recombinant Proteins
13.
Am J Physiol ; 275(2): G352-62, 1998 08.
Article in English | MEDLINE | ID: mdl-9688663

ABSTRACT

Supramaximal stimulation of the pancreas with the CCK analog caerulein causes acute edematous pancreatitis. In this model, active trypsin can be detected in the pancreas shortly after the start of supramaximal stimulation. Incubation of pancreatic acini in vitro with a supramaximally stimulating caerulein concentration also results in rapid activation of trypsinogen. In the current study, we have used the techniques of subcellular fractionation and both light and electron microscopy immunolocalization to identify the site of trypsinogen activation and the subsequent fate of trypsin during caerulein-induced pancreatitis. We report that trypsin activity and trypsinogen-activation peptide (TAP), which is released on activation of trypsinogen, are first detectable in a heavy subcellular fraction. This fraction is enriched in digestive enzyme zymogens and lysosomal hydrolases. Subsequent to trypsinogen activation, both trypsin activity and TAP move to a soluble compartment. Immunolocalization studies indicate that trypsinogen activation occurs in cytoplasmic vacuoles that contain the lysosomal hydrolase cathepsin B. These observations suggest that, during the early stages of pancreatitis, trypsinogen is activated in subcellular organelles containing colocalized digestive enzyme zymogens and lysosomal hydrolases and that, subsequent to its activation, trypsin is released into the cytosol.


Subject(s)
Pancreas/enzymology , Pancreatitis/enzymology , Trypsin/metabolism , Trypsinogen/metabolism , Acute Disease , Animals , Cathepsin B/metabolism , Cell Fractionation , Ceruletide , Edema , Enzyme Activation , Immunohistochemistry , Kinetics , Lysosomes/enzymology , Male , Oligopeptides/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/pathology , Rats , Rats, Wistar , Subcellular Fractions/enzymology , Time Factors , Vacuoles/enzymology
14.
J Pineal Res ; 22(1): 20-5, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9062866

ABSTRACT

Animal welfare concerns have led to increased demands for use of post-operative analgesia in association with experimental surgery. Such treatment is appropriate provided it has no effect on experimental parameters. In order to assess any effects of analgesics on endogenous melatonin production, groups of six ewes were bled at 30 min intervals for the first 4 hr of darkness a) without analgesic treatment and b) immediately after analgesic administration. Analgesics tested were ketoprofen, phenylbutazone (both non-steroidal anti-inflammatory drugs or NSAIDs) and buprenorphine (an opioid). Plasma melatonin was measured by radioimmunoassay and 4 hr secretion profiles computed for each animal. Ketoprofen and buprenorphine treatment reduced mean four hour melatonin secretion profiles by 50.6% (from 1,347.3 to 665.1 pg/ml.hr) and 38.6% (from 287.8 to 171.3 pg/ml.hr), respectively. Only in the case of phenylbutazone was the response not statistically significant; hence this drug has been selected as a post-operative analgesic for future experimental studies involving measurement of melatonin levels.


Subject(s)
Analgesics/pharmacology , Melatonin/metabolism , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Buprenorphine/pharmacology , Darkness , Female , Ketoprofen/pharmacology , Melatonin/blood , Phenylbutazone/pharmacology , Sheep
15.
J Pineal Res ; 16(4): 193-7, 1994 May.
Article in English | MEDLINE | ID: mdl-7807374

ABSTRACT

Both melatonin and pineal antigonadotropic peptides have the same end effect, i.e., prevention of the hypertrophic response when tested in the conventional compensatory ovarian hypertrophy (COH) model. The present work was undertaken to study the effect of melatonin and a melatonin- and steroid-free inhibin-like ovine pineal antigonadotropin (PI) on serum follicle stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) following hemoiovariectomy in adult Holtzman rats and also to ascertain if any similarity exists in their mode of action during COH. While melatonin prevented the transient rise in FSH at 12 hr after unilateral ovariectomy (ULO), thus retaining the basal preoperative level, PI depressed basal levels of FSH too. In addition, melatonin suppressed PRL and LH levels at 12 hr and 120 hr after ULO, respectively. PI, on the other hand, had no effect on serum LH and PRL levels. In light of our earlier in vitro results, which showed a direct inhibitory effect of PI and not of melatonin on pituitary FSH synthesis and release, the present results indicate a dichotomy in the mode of action of PI and melatonin. PI acts directly at the level of the pituitary while melatonin may act at the level of the hypothalamus or higher brain centers to suppress the FSH surge and the ensuing compensatory response.


Subject(s)
Inhibins/pharmacology , Melatonin/pharmacology , Ovary/drug effects , Ovary/pathology , Animals , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone/blood , Hypertrophy , Luteinizing Hormone/blood , Pineal Gland/drug effects , Prolactin/blood , Radioimmunoassay , Rats , Rats, Sprague-Dawley
16.
J Pineal Res ; 13(2): 72-9, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1453311

ABSTRACT

In an attempt to define the antigonadotropic activity associated with the protein/peptide extracts of ovine pineals, melatonin- and steroid-free fractions obtained after subjecting crude ovine pineal acetone powder to soft gel chromatography were tested in three bioassay systems: (1) the conventional compensatory ovarian hypertrophy inhibition assay in rats, (2) the coupled bioassay in immature mice that enables one to distinguish factors acting at the level of the pituitary from those acting at the gonadal level, and (3) the classical in vitro pituitary cell culture assay for inhibin. The large molecular weight fraction, referred to as PI, behaved like a classical antigonadotropin as it suppressed compensatory ovarian hypertrophy following unilateral ovariectomy. Further, it not only lowered the basal and gonadotropin releasing hormone (GnRH)-stimulated release of follicle stimulating hormone (FSH) by the cultured pituitary cells, but also inhibited the human chorionic gonadotropin (hCG)-induced uterine weight gain in the coupled assay, thus acting like inhibin in these two assay systems. In addition, it showed immunological cross-reactivity with ovine testicular inhibin. The present results strongly support the view that inhibin-like activity may be the major player in what has so far been referred to as antigonadotropic activity.


Subject(s)
Inhibins/physiology , Pineal Gland/chemistry , Sheep/metabolism , Animals , Biological Assay , Cells, Cultured , Chromatography, Gel , Cross Reactions , Female , Follicle Stimulating Hormone/metabolism , Inhibins/isolation & purification , Male , Organ Size/drug effects , Ovariectomy , Ovary/drug effects , Pineal Gland/physiology , Pituitary Gland, Anterior/metabolism , Rats , Testis/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...