Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 2(4): 519-535, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27642624

RESUMO

BACKGROUND AND AIMS: Severe acute pancreatitis is characterized by acinar cell death and inflammation. Necroptosis is an aggressive and pro-inflammatory mode of cell death that can be prevented by necrostatin-1 administration or RIP3 deletion. METHODS: Mouse pancreatic acinar cells were incubated with supramaximally stimulating concentrations of caerulein or sub-micellar concentrations of TLCS and necroptosis was inhibited by either addition of necrostatin or by RIP3 deletion. Cell death was quantitated using either LDH leakage from acini or PI staining of nuclei. Necrosome formation was tracked and quantitated using cell fractionation or immunoprecipitation. Pancreatitis was induced in mice by retrograde intraductal infusion of TLCS or by repetitive supramaximal stimulation with caerulein. RESULTS: Necroptosis was found to be the most prevalent mode of acinar cell in vitro death and little or no apoptosis was observed. Acinar cell death was associated with necrosome formation and prevented by either necrostatin administration or RIP3 deletion. Both of these interventions reduced the severity of TLCS- or caerulein-induced pancreatitis. Delaying necrostatin administration until after pancreatitis had already been established could still reduce the severity of TLCS-induced pancreatitis. CONCLUSIONS: Necroptosis is the predominant mode of acinar cell death in severe experimental mouse pancreatitis. The severity of pancreatitis can be reduced by administration of necrostatin and that necrostatin can still reduce the cell injury of pancreatitis even if it is administered after the disease has already been established. Inhibition of necroptosis may be an effective strategy for the treatment of severe clinical pancreatitis.

3.
J Biol Chem ; 286(15): 13327-35, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21343291

RESUMO

The roles of monocytes/macrophages and their mechanisms of action in the regulation of pancreatitis are poorly understood. To address these issues, we have employed genetically altered mouse strains that either express the human diphtheria toxin receptor (DTR) coupled to the CD11b promoter or have global deletion of TNF-α. Targeted, conditional depletion of monocytes/macrophages was achieved by administration of diphtheria toxin (DT) to CD11b-DTR mice. We show that in the absence of DT administration, pancreatitis is associated with an increase in pancreatic content of Ly-6C(hi) monocytes/macrophages but that this response is prevented by prior administration of DT to CD11b-DTR mice. DT administration also reduces pancreatic edema and acinar cell injury/necrosis in two dissimilar experimental models of acute pancreatitis (a secretagogue-induced model and a model elicited by retrograde pancreatic duct infusion of sodium taurocholate). In the secretagogue-elicited model, the DT-induced decrease in pancreatitis severity is reversed by adoptive transfer of purified Ly-6C(hi) monocytes harvested from non-DT-treated CD11b-DTR mice or by the transfer of purified Ly-6C(hi) monocytes harvested from TNF-α(+/+) donor mice, but it is not reversed by the transfer of Ly-6C(hi) monocytes harvested from TNF-α(-/-) donors. Our studies indicate that the Ly-6C(hi) monocyte subset regulates the severity of pancreatitis by promoting pancreatic edema and acinar cell injury/necrosis and that this phenomenon is dependent upon the expression of TNF-α by those cells. They suggest that therapies targeting Ly-6C(hi) monocytes and/or TNF-α expression by Ly-6C(hi) monocytes might prove beneficial in the prevention or treatment of acute pancreatitis.


Assuntos
Antígenos Ly/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Monócitos/metabolismo , Pâncreas Exócrino/metabolismo , Pancreatite Necrosante Aguda/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Transferência Adotiva , Animais , Antígenos Ly/genética , Antígenos Ly/imunologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Toxina Diftérica/toxicidade , Modelos Animais de Doenças , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/patologia , Monócitos/transplante , Pâncreas Exócrino/imunologia , Pâncreas Exócrino/patologia , Pancreatite Necrosante Aguda/induzido quimicamente , Pancreatite Necrosante Aguda/genética , Pancreatite Necrosante Aguda/imunologia , Pancreatite Necrosante Aguda/patologia , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
4.
Nat Protoc ; 5(2): 335-41, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20134432

RESUMO

Mechanistic studies of acute pancreatitis require animal models because clinical material is generally not available during the early phases of the disease. Here we describe a protocol to induce biliary pancreatitis by retrogradely infusing bile acids into the pancreatic duct of anesthetized mice. The resulting model replicates events believed to be responsible for the onset of clinical biliary (i.e., gallstone) pancreatitis and creates highly reproducible pancreatitis with a severity that depends on the concentration of infused bile acid. Pancreatitis reaches its maximal level of severity within 24 h of induction, and it resolves over the subsequent week. This protocol enables the investigator to use genetically modified strains of mice, and it requires only relatively simple and easily learned techniques of small animal surgery. With practice and gentle technique, the surgery (from induction of anesthesia to completion of the infusion) can be completed within 25 min per animal.


Assuntos
Ácidos e Sais Biliares/toxicidade , Ductos Pancreáticos/patologia , Pancreatite/induzido quimicamente , Animais , Ácidos e Sais Biliares/administração & dosagem , Modelos Animais de Doenças , Infusões Parenterais , Camundongos , Ductos Pancreáticos/efeitos dos fármacos , Pancreatite/patologia , Pancreatite/cirurgia , Valores de Referência , Ácido Taurocólico/toxicidade , Ácido Taurolitocólico/análogos & derivados , Ácido Taurolitocólico/toxicidade
6.
Gastroenterology ; 138(2): 715-25, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19900448

RESUMO

BACKGROUND & AIMS: The mechanisms by which reflux of bile acids into the pancreas induces pancreatitis are unknown. We reasoned that key events responsible for this phenomenon might be mediated by Gpbar1, a recently identified and widely expressed G-protein-coupled, cell surface bile acid receptor. METHODS: Acute pancreatitis was induced in wild-type and Gpbar1(-/-) mice by either retrograde ductal infusion of taurolithocholic acid-3-sulfate (TLCS) or supramaximal secretagogue stimulation with caerulein. In vitro experiments were performed in which acini obtained from wild-type and Gpbar1(-/-) mice were exposed to either submicellar concentrations of TLCS (200-500 microM) or a supramaximally stimulating concentration of caerulein (10 nM). RESULTS: Gpbar1 is expressed at the apical pole of acinar cells and its genetic deletion is associated with reduced hyperamylasemia, edema, inflammation, and acinar cell injury in TLCS-induced, but not caerulein-induced, pancreatitis. In vitro, genetic deletion of Gpbar1 is associated with markedly reduced generation of pathological calcium transients, intracellular activation of digestive zymogens, and cell injury when these responses are induced by exposure to TLCS, but not when they are induced by exposure to caerulein. CONCLUSIONS: Gpbar1 may play a critical role in the evolution of bile-acid-induced pancreatitis by coupling exposure to bile acids with generation of pathological intracellular calcium transients, intra-acinar cell zymogen activation, and acinar cell injury. Acute biliary pancreatitis may be a "receptor-mediated" disease and interventions that interfere with Gpbar1 function might prove beneficial in the treatment and/or prevention of biliary acute pancreatitis.


Assuntos
Ácidos e Sais Biliares/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Pancreatite/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Doença Aguda , Amilases/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Ceruletídeo/efeitos adversos , Modelos Animais de Doenças , Precursores Enzimáticos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/induzido quimicamente , Receptores Acoplados a Proteínas G/genética , Índice de Gravidade de Doença , Ácido Taurolitocólico/efeitos adversos , Ácido Taurolitocólico/análogos & derivados
7.
J Biol Chem ; 283(30): 20703-12, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18511423

RESUMO

Protease-activated receptor-2 (PAR2) is a 7-transmembrane G-protein-coupled tethered ligand receptor that is expressed by pancreatic acinar and ductal cells. It can be physiologically activated by trypsin. Previously reported studies (Namkung, W., Han, W., Luo, X., Muallem, S., Cho, K. H., Kim, K. H., and Lee, M. G. (2004) Gastroenterology 126, 1844-1859; Sharma, A., Tao, X., Gopal, A., Ligon, B., Andrade-Gordon, P., Steer, M. L., and Perides, G. (2005) Am. J. Physiol. 288, G388-G395) have shown that PAR2 activation exerts a protective effect on the experimental model of pancreatitis induced by supramaximal secretagogue (caerulein) stimulation. We now show that PAR2 exerts a worsening effect on a different model of experimental pancreatitis, i.e. one induced by retrograde pancreatic ductal infusion of bile salts. In vitro studies using freshly prepared pancreatic acini show that genetic deletion of PAR2 reduces bile salt-induced pathological calcium transients, acinar cell injury, and activation of c-Jun N-terminal kinase, whereas genetic deletion of PAR2 has the opposite or no effect on these pancreatitis-related events when they are elicited, in vitro, by caerulein stimulation. Studies employing a combination of trypsin inhibition and activation of PAR2 with the activating peptide SLIGRL show that all these differences indeed depend on the activation of PAR2. These studies are the first to report that a single perturbation can have model-specific and opposite effects on pancreatitis, and they underscore the importance of performing mechanistic pancreatitis studies using two dissimilar models of the disease to detect idiosyncratic, model-specific events. We suggest PAR2 activation exerts a worsening effect on the severity of clinical pancreatitis and that interventions interfering with PAR2 activation may be of benefit in the treatment of patients with severe pancreatitis.


Assuntos
Pancreatite/enzimologia , Pancreatite/metabolismo , Receptor PAR-2/fisiologia , Doença Aguda , Animais , Ácidos e Sais Biliares/farmacologia , Ceruletídeo/farmacologia , Ativação Enzimática , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Pâncreas/citologia , Peptídeos/química , Estrutura Terciária de Proteína , Receptor PAR-2/metabolismo
8.
Lancet ; 371(9607): 143-52, 2008 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-18191686

RESUMO

Acute pancreatitis is an inflammatory disease of the pancreas. Acute abdominal pain is the most common symptom, and increased concentrations of serum amylase and lipase confirm the diagnosis. Pancreatic injury is mild in 80% of patients, who recover without complications. The remaining patients have a severe disease with local and systemic complications. Gallstone migration into the common bile duct and alcohol abuse are the most frequent causes of pancreatitis in adults. About 15-25% of pancreatitis episodes are of unknown origin. Treatment of mild disease is supportive, but severe episodes need management by a multidisciplinary team including gastroenterologists, interventional radiologists, intensivists, and surgeons. Improved understanding of pathophysiology and better assessments of disease severity should ameliorate the management and outcome of this complex disease.


Assuntos
Amilases/sangue , Infecções por Birnaviridae/fisiopatologia , Vírus da Necrose Pancreática Infecciosa/patogenicidade , Lipase/sangue , Pancreatite , Doença Aguda , Antibacterianos/uso terapêutico , Infecções por Birnaviridae/classificação , Infecções por Birnaviridae/metabolismo , Colecistectomia , Cálculos Biliares/complicações , Cálculos Biliares/tratamento farmacológico , Humanos , Incidência , Pancreatite/diagnóstico , Pancreatite/fisiopatologia , Pancreatite/terapia , Índice de Gravidade de Doença
9.
J Biol Chem ; 282(30): 22140-9, 2007 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-17537724

RESUMO

Pancreatic and lung inflammation during acute pancreatitis is a poorly understood, but clinically important, phenomenon. The proto-oncogene Tpl2 (tumor progression locus-2) has recently been shown to have important immunomodulatory effects on some inflammatory processes, but its importance to pancreatitis has not been previously examined. Our studies were designed to (a) define the effects of Tpl2 on pancreatic and lung inflammation during pancreatitis and (b) identify mechanisms and cell types responsible for those effects. We examined pancreatitis-associated Tpl2 effects in wild type and Tpl2(-/-) mice subjected to either secretagogue-induced or bile salt-induced pancreatitis. To determine the myeloid or non-myeloid lineage of cells responsible for the Tpl2 effects, we used Tpl2(-/-) chimeric mice generated by lethal irradiation followed by bone marrow transplantation. Mechanisms responsible for the effects of Tpl2 ablation on caerulein-induced proinflammatory events were evaluated under in vivo and in vitro conditions using the techniques of electrophoretic mobility shift assay, immunoblot analysis, and quantitative reverse transcription-PCR. We found that Tpl2 ablation markedly reduced pancreatic and lung inflammation in these two dissimilar models of pancreatitis, but it did not alter pancreatic injury/necrosis in either model. The reduction in caerulein-induced pancreatic inflammation is dependent upon Tpl2 ablation in non-myeloid cells and is associated with both in vivo and in vitro inhibition of MEK, JNK, and AP-1 activation and the expression of MCP-1, MIP-2, and interleukin-6. Non-myeloid cell expression of Tpl2 regulates pancreatic inflammation during pancreatitis by mediating proinflammatory signals and the generation of neutrophil chemoattracting factors.


Assuntos
Inflamação/fisiopatologia , Pulmão/fisiopatologia , MAP Quinase Quinase Quinases/fisiologia , Pâncreas/fisiopatologia , Pancreatite/fisiopatologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Medula Óssea/fisiopatologia , Modelos Animais de Doenças , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Knockout , Neutrófilos/fisiologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética
10.
Gut ; 56(11): 1590-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17591621

RESUMO

OBJECTIVE: Most mechanistic studies of pancreatitis in mice employ the secretagogue-induced model. The currently reported studies were designed to develop an alternative, and possibly more clinically relevant, mouse model of pancreatitis. DESIGN: Na-taurocholate (10-50 microl, 1-5%) in saline, or saline alone, was retrogradely infused into the mouse pancreatic duct. The animals were killed 6-24 hours later and the severity of pancreatitis in the pancreatic head and tail was examined by quantitating hyperamylasemia, pancreatic edema, acinar cell necrosis, and pancreatic inflammation. In addition, intrapancreatic activation of trypsinogen, generation of IL-6, intrapulmonary sequestration of neutrophils, and alterations in lung compliance were evaluated. The effects of Na-taurocholate on in-vitro acinar cell calcium transients, viability, and trypsinogen activation were examined. RESULTS: Little or no evidence of pancreatitis was observed in mice infused with saline alone or in the tail of pancreata removed from animals infused with Na-taurocholate. In the head of the pancreas, evidence of pancreatitis was observed 12-24 hours after infusion of 20-50 microl 2-5% Na-taurocholate and the earliest morphological changes involved terminal duct and acinar cells. Intrapancreatic trypsin activity was transiently elevated within 5 minutes of Na-taurocholate infusion and pancreatic IL-6 levels were elevated 24 hours later. Under in-vitro conditions, Na-taurocholate triggered pathological acinar cell calcium transients, cell death, and calcium-dependent trypsinogen activation. CONCLUSION: This clinically relevant model of acute biliary pancreatitis yields reproducible results and its severity can be easily manipulated. It is ideally suited for use in mechanistic studies employing genetically modified mouse strains.


Assuntos
Doenças Biliares/induzido quimicamente , Colagogos e Coleréticos , Modelos Animais de Doenças , Pancreatite/induzido quimicamente , Ácido Taurocólico , Doença Aguda , Animais , Feminino , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ductos Pancreáticos , Pneumonia/induzido quimicamente , Reprodutibilidade dos Testes
11.
Am J Physiol Gastrointest Liver Physiol ; 292(6): G1738-46, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17332471

RESUMO

We have hypothesized that the colocalization of digestive zymogens with lysosomal hydrolases, which occurs during the early stages of every experimental pancreatitis model, facilitates activation of those zymogens by lysosomal hydrolases such as cathepsin B and that this activation triggers acute pancreatitis by leading to acinar cell injury. Some, however, have argued that the colocalization phenomenon may be the result, rather than the cause, of zymogen activation during pancreatitis. To resolve this controversy and explore the causal relationships between zymogen activation and other early pancreatitis events, we induced pancreatitis in mice by repeated supramaximal secretagogue stimulation with caerulein. Some animals were pretreated with the cathepsin B inhibitor CA-074 me to inhibit cathepsin B, prevent intrapancreatic activation of digestive zymogens, and reduce the severity of pancreatitis. We show that inhibition of cathepsin B by pretreatment with CA-074 me prevents intrapancreatic zymogen activation and reduces organellar fragility, but it does not alter the caerulein-induced colocalization phenomenon or subcellular F-actin redistribution or prevent caerulein-induced activation of NF-kappaB, ERK1/2, and JNK or upregulated expression of cytochemokines. We conclude 1) that the colocalization phenomenon, F-actin redistribution, activation of proinflammatory transcription factors, and upregulated expression of cytochemokines are not the results of zymogen activation, and 2) that these early events in pancreatitis are not dependent on cathepsin B activity. In contrast, zymogen activation and increased subcellular organellar fragility during caerulein-induced pancreatitis are dependent on cathepsin B activity.


Assuntos
Catepsina B/metabolismo , Pâncreas/metabolismo , Pancreatite/metabolismo , Tripsina/metabolismo , Tripsinogênio/metabolismo , Actinas/metabolismo , Doença Aguda , Amilases/metabolismo , Animais , Arilsulfatases/metabolismo , Catepsina B/antagonistas & inibidores , Ceruletídeo , Quimiocina CCL2/metabolismo , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lisossomos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/enzimologia , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/enzimologia , Pancreatite/patologia , Pancreatite/prevenção & controle , Transporte Proteico , Vesículas Secretórias/enzimologia , Índice de Gravidade de Doença , Fatores de Tempo
12.
Pancreatology ; 6(6): 507-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17035695

RESUMO

In this interview, Dr. Michael Steer shares his life experience in pancreatic research, discusses the importance of mentorship and gives advice to young investigators starting in this field. Dr. Michael Steer is a world-renown investigator who has made an extraordinary contribution to the understanding of pancreatic physiology. His achievements in the field of acute pancreatitis were the foundation for the characterization of the cellular basis for this disease.


Assuntos
Gastroenterologia/educação , Mentores , Pâncreas/fisiologia , Ensino , Humanos , Jornalismo Médico , Projetos de Pesquisa
13.
World J Gastroenterol ; 12(13): 1985-90, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16610045

RESUMO

Acute pancreatitis is generally believed to be a disease in which the pancreas is injured by digestive enzymes that it normally produces. Most of the potentially harmful digestive enzymes produced by pancreatic acinar cells are synthesized and secreted as inactive zymogens which are normally activated only upon entry into the duodenum but, during the early stages of acute pancreatitis, those zymogens become prematurely activated within the pancreas and, presumably, that activation occurs within pancreatic acinar cells. The mechanisms responsible for intracellular activation of digestive enzyme zymogens have not been elucidated with certainty but, according to one widely recognized theory (the "co-localization hypothesis"), digestive enzyme zymogens are activated by lysosomal hydrolases when the two types of enzymes become co-localized within the same intracellular compartment. This review focuses on the evidence supporting the validity of the co-localization hypothesis as an explanation for digestive enzyme activation during the early stages of pancreatitis. The findings, summarized in this review, support the conclusion that co-localization of lysosomal hydrolases with digestive enzyme zymogens plays a critical role in permitting the intracellular activation of digestive enzymes that leads to acinar cell injury and pancreatitis.


Assuntos
Precursores Enzimáticos/metabolismo , Pâncreas/enzimologia , Pancreatite/enzimologia , Doença Aguda , Amilases/metabolismo , Animais , Catepsina B/metabolismo , Ativação Enzimática , Humanos , Hidrolases/metabolismo , Lisossomos/enzimologia , Transporte Proteico , Tripsinogênio/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 289(4): G686-95, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15976386

RESUMO

Pancreatic acini secrete digestive enzymes in response to a variety of secretagogues including CCK and agonists acting via proteinase-activated receptor-2 (PAR2). We employed the CCK analog caerulein and the PAR2-activating peptide SLIGRL-NH(2) to compare and contrast Ca(2+) changes and amylase secretion triggered by CCK receptor and PAR2 stimulation. We found that secretion stimulated by both agonists is dependent on a rise in cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) and that this rise in [Ca(2+)](i) reflects both the release of Ca(2+) from intracellular stores and accelerated Ca(2+) influx. Both agonists, at low concentrations, elicit oscillatory [Ca(2+)](i) changes, and both trigger a peak plateau [Ca(2+)](i) change at high concentrations. Although the two agonists elicit similar rates of amylase secretion, the rise in [Ca(2+)](i) elicited by caerulein is greater than that elicited by SLIGRL-NH(2). In Ca(2+)-free medium, the rise in [Ca(2+)](i) elicited by SLIGRL-NH(2) is prevented by the prior addition of a supramaximally stimulating concentration of caerulein, but the reverse is not true; the rise elicited by caerulein is neither prevented nor reduced by prior addition of SLIGRL-NH(2). Both the oscillatory and the peak plateau [Ca(2+)](i) changes that follow PAR2 stimulation are prevented by the phospholipase C (PLC) inhibitor U73122, but U73122 prevents only the oscillatory [Ca(2+)](i) changes triggered by caerulein. We conclude that 1) both PAR2 and CCK stimulation trigger amylase secretion that is dependent on a rise in [Ca(2+)](i) and that [Ca(2+)](i) rise reflects release of calcium from intracellular stores as well as accelerated influx of extracellular calcium; 2) PLC mediates both the oscillatory and the peak plateau rise in [Ca(2+)](i) elicited by PAR2 but only the oscillatory rise in [Ca(2+)](i) elicited by CCK stimulation; and 3) the rate of amylase secretion elicited by agonists acting via different types of receptors may not correlate with the magnitude of the [Ca(2+)](i) rise triggered by those different types of secretagogue.


Assuntos
Amilases/metabolismo , Cálcio/fisiologia , Colecistocinina/fisiologia , Pâncreas/metabolismo , Receptor PAR-2/fisiologia , Animais , Ceruletídeo/farmacologia , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligopeptídeos/farmacologia , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Pirrolidinonas/farmacologia , Receptores da Colecistocinina/efeitos dos fármacos , Fosfolipases Tipo C/antagonistas & inibidores
15.
Am J Physiol Gastrointest Liver Physiol ; 288(2): G388-95, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15458925

RESUMO

Protease-activated receptor-2 (PAR-2) is a widely expressed tethered ligand receptor that can be activated by trypsin and other trypsin-like serine proteases. In the exocrine pancreas, PAR-2 activation modulates acinar cell secretion of digestive enzymes and duct cell ion channel function. During acute pancreatitis, digestive enzyme zymogens, including trypsinogen, are activated within the pancreas. We hypothesized that trypsin, acting via PAR-2, might regulate the severity of that disease, and to test this hypothesis, we examined the effect of either genetically deleting or pharmacologically activating PAR-2 on the severity of secretagogue-induced experimental pancreatitis. We found that experimental acute pancreatitis is more severe in PAR-2(-/-) than in wild-type mice and that in vivo activation of PAR-2, achieved by parenteral administration of the PAR-2-activating peptide SLIGRL-NH2, reduces the severity of pancreatitis. In the pancreas during the early stages of pancreatitis, the MAPK ERK1/2 is activated and translocated to the nucleus, but nuclear translocation is reduced by activation of PAR-2. Our findings indicate that PAR-2 exerts a protective effect on pancreatitis and that activation of PAR-2 ameliorates pancreatitis, possibly by inhibiting ERK1/2 translocation to the nucleus. Our observations suggest that PAR-2 activation may be of therapeutic value in the treatment and/or prevention of severe clinical pancreatitis, and they lead us to speculate that, from a teleological standpoint, PAR-2 may have evolved in the pancreas as a protective mechanism designed to dampen the injurious effects of intrapancreatic trypsinogen activation.


Assuntos
Pancreatite/fisiopatologia , Receptor PAR-2/fisiologia , Animais , Ceruletídeo , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligopeptídeos/farmacologia , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Peptídeos/farmacologia , Receptor PAR-2/genética , Receptor PAR-2/metabolismo
16.
Am J Physiol Gastrointest Liver Physiol ; 283(5): G1166-74, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12381531

RESUMO

Cyclooxygenase-2 (COX-2), a widely distributed enzyme, plays an important role in inflammation. We have studied the role of COX-2 in acute pancreatitis and pancreatitis-associated lung injury using both the pharmacological inhibition of COX-2 and genetic deletion of COX-2. Pancreatitis was induced in mice by 12 hourly injections of cerulein. The severity of pancreatitis was assessed by measuring serum amylase, pancreatic trypsin activity, intrapancreatic sequestration of neutrophils, and acinar cell necrosis. The severity of lung injury was evaluated by measuring lactate dehydrogenase levels in the bronchoalveolar lavage fluid and by quantitating neutrophil sequestration in the lung. In both the pharmacologically inhibited and genetically altered mice, the severity of pancreatitis and pancreatitis-associated lung injury was reduced compared with the noninhibited strains of COX-2-sufficient mice. This reduction in injury indicates that COX-2 plays an important proinflammatory role in pancreatitis and its associated lung injury. Our findings support the concept that COX-2 inhibitors may play a beneficial role in the prevention of acute pancreatitis or in the reduction of its severity.


Assuntos
Isoenzimas/metabolismo , Pneumopatias/complicações , Pneumopatias/patologia , Pancreatite/complicações , Pancreatite/patologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Celecoxib , Ceruletídeo , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/deficiência , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , NF-kappa B/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Nitrobenzenos/farmacologia , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Prostaglandina-Endoperóxido Sintases/deficiência , Prostaglandina-Endoperóxido Sintases/genética , Pirazóis , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença , Sulfonamidas/farmacologia , Tripsinogênio/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 283(3): G794-800, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12181196

RESUMO

Intrapancreatic activation of trypsinogen is believed to play a critical role in the initiation of acute pancreatitis, but mechanisms responsible for intrapancreatic trypsinogen activation during pancreatitis have not been clearly defined. In previous in vitro studies, we have shown that intra-acinar cell activation of trypsinogen and acinar cell injury in response to supramaximal secretagogue stimulation could be prevented by the cell permeant cathepsin B inhibitor E64d (Saluja A, Donovan EA, Yamanaka K, Yamaguchi Y, Hofbauer B, and Steer ML. Gastroenterology 113: 304-310, 1997). The present studies evaluated the role of intrapancreatic trypsinogen activation, this time under in vivo conditions, in two models of pancreatitis by using another highly soluble cell permeant cathepsin B inhibitor, L-3-trans-(propylcarbamoyl)oxirane-2-carbonyl-L-isoleucyl-L-proline methyl ester (CA-074me). Intravenous administration of CA-074me (10 mg/kg) before induction of either secretagogue-elicited pancreatitis in mice or duct infusion-elicited pancreatitis in rats markedly reduced the extent of intrapancreatic trypsinogen activation and substantially reduced the severity of both pancreatitis models. These observations support the hypothesis that, during the early stages of pancreatitis, trypsinogen activation in the pancreas is mediated by the lysosomal enzyme cathepsin B. Our findings also suggest that pharmacological interventions that inhibit cathepsin B may prove useful in preventing acute pancreatitis or reducing its severity.


Assuntos
Catepsina B/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Pancreatite/fisiopatologia , Tripsinogênio/metabolismo , Animais , Catepsina B/antagonistas & inibidores , Ceruletídeo/farmacologia , Infusões Parenterais , Masculino , Camundongos , Camundongos Endogâmicos , Ductos Pancreáticos , Pancreatite/induzido quimicamente , Pancreatite/patologia , Ratos , Ratos Wistar , Índice de Gravidade de Doença , Ácido Taurocólico/administração & dosagem
18.
Am J Physiol Lung Cell Mol Physiol ; 283(3): L541-8, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12169573

RESUMO

Severe pancreatitis is frequently associated with acute lung injury (ALI) and the respiratory distress syndrome. The role of granulocyte-macrophage colony-stimulating factor (GM-CSF) in mediating the ALI associated with secretagogue-induced experimental pancreatitis was evaluated with GM-CSF knockout mice (GM-CSF -/-). Pancreatitis was induced by hourly (12x) intraperitoneal injection of a supramaximally stimulating dose of the cholecystokinin analog caerulein. The resulting pancreatitis was similar in GM-CSF-sufficient (GM-CSF +/+) control animals and GM-CSF -/- mice. Lung injury, quantitated by measuring lung myeloperoxidase activity (an indicator of neutrophil sequestration), alveolar-capillary permeability, and alveolar membrane thickness was less severe in GM-CSF -/- than in GM-CSF +/+ mice. In GM-CSF +/+ mice, pancreas, lung and serum GM-CSF levels increase during pancreatitis. Lung levels of macrophage inflammatory protein (MIP)-2 are also increased during pancreatitis, but, in this case, the rise is less profound in GM-CSF -/- mice than in GM-CSF +/+ controls. Administration of anti-MIP-2 antibodies was found to reduce the severity of pancreatitis-associated ALI. Our findings indicate that GM-CSF plays a critical role in coupling pancreatitis to ALI and suggest that GM-CSF may act indirectly by regulating the release of other proinflammatory factors including MIP-2.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Pneumopatias/etiologia , Pneumopatias/fisiopatologia , Pancreatite/complicações , Doença Aguda , Animais , Anticorpos/farmacologia , Ceruletídeo , Quimiocina CXCL2 , Quimiocinas/imunologia , Quimiocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Contagem de Leucócitos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Camundongos Knockout/genética , Pâncreas/metabolismo , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Pancreatite/fisiopatologia , Valores de Referência , Índice de Gravidade de Doença
19.
Gastroenterology ; 122(1): 156-65, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11781290

RESUMO

BACKGROUND & AIMS: Prior thermal stress induces heat shock protein 70 (HSP70) expression in the pancreas and protects against secretagogue-induced pancreatitis, but it is not clear that this thermal stress-induced protection is actually mediated by HSP70 since thermal stress may have other, non-HSP related, effects. METHODS: In the present study, we have administered antisense (AS) oligonucleotides, which prevent pancreatic expression of HSP70 to rats, in vivo, to evaluate this issue. In a separate series of experiments, designed to examine the role of pancreatitis-induced HSP70 expression in modulating the severity of pancreatitis, rats not subjected to prior thermal stress were given AS-HSP70 before cerulein administration, and trypsinogen activation as well as the severity of pancreatitis were evaluated. RESULTS: Hyperthermia induced HSP70 expression, prevented intrapancreatic trypsinogen activation, and protected against cerulein-induced pancreatitis. Administration of AS-HSP70 but not sense-HSP70 reduced the thermal stress-induced HSP70 expression, restored the ability of supramaximal cerulein stimulation to cause intrapancreatic trypsinogen activation, and abolished the protective effect of prior thermal stress against pancreatitis. In non-thermally stressed animals, pretreatment with AS-HSP70 before the induction of pancreatitis exacerbated all the parameters associated with pancreatitis. CONCLUSIONS: These findings lead us to conclude that HSP70 induction, rather than some other thermal stress-related phenomenon, mediates the thermal stress-induced protection against pancreatitis and that it protects against pancreatitis by preventing intrapancreatic activation of trypsinogen. The worsening of pancreatitis, which occurs when non-thermally stressed animals are given AS-HSP70 before cerulein, suggests that cerulein-induced HSP70 expression in nontreated animals acts to limit the severity of pancreatitis.


Assuntos
Proteínas de Choque Térmico HSP70/genética , Transtornos de Estresse por Calor/metabolismo , Pancreatite/metabolismo , Tripsinogênio/metabolismo , Doença Aguda , Animais , Ceruletídeo , Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Pancreatite/induzido quimicamente , Pancreatite/patologia , Ratos , Ratos Wistar , Estresse Fisiológico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA