Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 85
Filtrer
1.
Gastroenterology ; 160(5): 1755-1770.e17, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33388318

RÉSUMÉ

BACKGROUND & AIMS: Oncogenic KrasG12D induces neoplastic transformation of pancreatic acinar cells through acinar-to-ductal metaplasia (ADM), an actin-based morphogenetic process, and drives pancreatic ductal adenocarcinoma (PDAC). mTOR (mechanistic target of rapamycin kinase) complex 1 (mTORC1) and 2 (mTORC2) contain Rptor and Rictor, respectively, and are activated downstream of KrasG12D, thereby contributing to PDAC. Yet, whether and how mTORC1 and mTORC2 impact on ADM and the identity of the actin nucleator(s) mediating such actin rearrangements remain unknown. METHODS: A mouse model of inflammation-accelerated KrasG12D-driven early pancreatic carcinogenesis was used. Rptor, Rictor, and Arpc4 (actin-related protein 2/3 complex subunit 4) were conditionally ablated in acinar cells to deactivate the function of mTORC1, mTORC2 and the actin-related protein (Arp) 2/3 complex, respectively. RESULTS: We found that mTORC1 and mTORC2 are markedly activated in human and mouse ADM lesions, and cooperate to promote KrasG12D-driven ADM in mice and in vitro. They use the Arp2/3 complex as a common downstream effector to induce the remodeling the actin cytoskeleton leading to ADM. In particular, mTORC1 regulates the translation of Rac1 (Rac family small GTPase 1) and the Arp2/3-complex subunit Arp3, whereas mTORC2 activates the Arp2/3 complex by promoting Akt/Rac1 signaling. Consistently, genetic ablation of the Arp2/3 complex prevents KrasG12D-driven ADM in vivo. In acinar cells, the Arp2/3 complex and its actin-nucleation activity mediated the formation of a basolateral actin cortex, which is indispensable for ADM and pre-neoplastic transformation. CONCLUSIONS: Here, we show that mTORC1 and mTORC2 attain a dual, yet nonredundant regulatory role in ADM and early pancreatic carcinogenesis by promoting Arp2/3 complex function. The role of Arp2/3 complex as a common effector of mTORC1 and mTORC2 fills the gap between oncogenic signals and actin dynamics underlying PDAC initiation.


Sujet(s)
Cellules acineuses/enzymologie , Complexe Arp-2-3/métabolisme , Carcinome du canal pancréatique/enzymologie , Transformation cellulaire néoplasique/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Mutation , Conduits pancréatiques/enzymologie , Tumeurs du pancréas/enzymologie , Protéines proto-oncogènes p21(ras)/génétique , Cellules acineuses/anatomopathologie , Complexe Arp-2-3/génétique , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux , Humains , Complexe-1 cible mécanistique de la rapamycine/génétique , Complexe-2 cible mécanistique de la rapamycine/génétique , Métaplasie , Souris de lignée C57BL , Souris knockout , Conduits pancréatiques/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Compagnon de mTOR insensible à la rapamycine/génétique , Compagnon de mTOR insensible à la rapamycine/métabolisme , Protéine de régulation associée à mTOR/génétique , Protéine de régulation associée à mTOR/métabolisme , Transduction du signal
2.
J Pathol ; 250(1): 42-54, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31531867

RÉSUMÉ

Molecular signalling mediated by the phosphatidylinositol-3-kinase (PI3K)-Akt axis is a key regulator of cellular functions. Importantly, alteration of the PI3K-Akt signalling underlies the development of different human diseases, thus prompting the investigation of the pathway as a molecular target for pharmacologic intervention. In this regard, recent studies showed that small molecule inhibitors of PI3K, the upstream regulator of the pathway, reduced the development of inflammation during acute pancreatitis, a highly debilitating and potentially lethal disease. Here we investigated whether a specific reduction of Akt activity, by using either pharmacologic Akt inhibition, or genetic inactivation of the Akt1 isoform selectively in pancreatic acinar cells, is effective in ameliorating the onset and progression of the disease. We discovered that systemic reduction of Akt activity did not protect the pancreas from initial damage and only transiently delayed leukocyte recruitment. However, reduction of Akt activity decreased acinar proliferation and exacerbated acinar-to-ductal metaplasia (ADM) formation, two critical events in the progression of pancreatitis. These phenotypes were recapitulated upon conditional inactivation of Akt1 in acinar cells, which resulted in reduced expression of 4E-BP1, a multifunctional protein of key importance in cell proliferation and metaplasia formation. Collectively, our results highlight the critical role played by Akt1 during the development of acute pancreatitis in the control of acinar cell proliferation and ADM formation. In addition, these results harbour important translational implications as they raise the concern that inhibitors of PI3K-Akt signalling pathways may negatively affect the regeneration of the pancreas. Finally, this work provides the basis for further investigating the potential of Akt1 activators to boost pancreatic regeneration following inflammatory insults. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Sujet(s)
Cellules acineuses/enzymologie , Prolifération cellulaire , Pancréas exocrine/enzymologie , Conduits pancréatiques/enzymologie , Pancréatite/enzymologie , Protéines proto-oncogènes c-akt/métabolisme , Cellules acineuses/effets des médicaments et des substances chimiques , Cellules acineuses/anatomopathologie , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Céruléine , Modèles animaux de maladie humaine , Mâle , Métaplasie , Souris de lignée C57BL , Souris knockout , Pancréas exocrine/effets des médicaments et des substances chimiques , Pancréas exocrine/anatomopathologie , Conduits pancréatiques/effets des médicaments et des substances chimiques , Conduits pancréatiques/anatomopathologie , Pancréatite/induit chimiquement , Pancréatite/génétique , Pancréatite/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/analyse , Protéines proto-oncogènes c-akt/déficit , Protéines proto-oncogènes c-akt/génétique , Rats , Transduction du signal
3.
Nutrients ; 10(9)2018 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-30213082

RÉSUMÉ

Pancreatic cancer remains a daunting foe despite a vast number of accumulating molecular analyses regarding the mutation and expression status of a variety of genes. Indeed, most pancreatic cancer cases uniformly present with a mutation in the KRAS allele leading to enhanced RAS activation. Yet our understanding of the many epigenetic/environmental factors contributing to disease incidence and progression is waning. Epidemiologic data suggest that diet may be a key factor in pancreatic cancer development and potentially a means of chemoprevention at earlier stages. While diets high in ω3 fatty acids are typically associated with tumor suppression, diets high in ω6 fatty acids have been linked to increased tumor development. Thus, to better understand the contribution of these polyunsaturated fatty acids to pancreatic carcinogenesis, we modeled early stage disease by targeting mutant KRAS to the exocrine pancreas and administered diets rich in these fatty acids to assess tumor formation and altered cell-signaling pathways. We discovered that, consistent with previous reports, the ω3-enriched diet led to reduced lesion penetrance via repression of proliferation associated with reduced phosphorylated AKT (pAKT), whereas the ω6-enriched diet accelerated tumor formation. These data provide a plausible mechanism underlying previously observed effects of fatty acids and suggest that administration of ω3 fatty acids can reduce the pro-survival, pro-growth functions of pAKT. Indeed, counseling subjects at risk to increase their intake of foods containing higher amounts of ω3 fatty acids could aid in the prevention of pancreatic cancer.


Sujet(s)
Anticarcinogènes/administration et posologie , Transformation cellulaire néoplasique/métabolisme , Régime alimentaire , Acides gras omega-3/administration et posologie , Tumeurs expérimentales/prévention et contrôle , Conduits pancréatiques/enzymologie , Tumeurs du pancréas/prévention et contrôle , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Animaux , Apoptose , Lignée cellulaire , Prolifération cellulaire , Survie cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Régime alimentaire/effets indésirables , Régulation négative , Humains , Souris transgéniques , Mutation , Tumeurs expérimentales/enzymologie , Tumeurs expérimentales/génétique , Tumeurs expérimentales/anatomopathologie , Conduits pancréatiques/anatomopathologie , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phosphorylation , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme
4.
Gastroenterology ; 154(5): 1509-1523.e5, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29273451

RÉSUMÉ

BACKGROUND & AIMS: Intraductal papillary mucinous neoplasias (IPMNs) are precancerous cystic lesions that can develop into pancreatic ductal adenocarcinomas (PDACs). These large macroscopic lesions are frequently detected during medical imaging, but it is unclear how they form or progress to PDAC. We aimed to identify cells that form IPMNs and mutations that promote IPMN development and progression. METHODS: We generated mice with disruption of Pten specifically in ductal cells (Sox9CreERT2;Ptenflox/flox;R26RYFP or PtenΔDuct/ΔDuct mice) and used PtenΔDuct/+ and Pten+/+ mice as controls. We also generated KrasG12D;PtenΔDuct/ΔDuct and KrasG12D;PtenΔDuct/+ mice. Pancreata were collected when mice were 28 weeks to 14.5 months old and analyzed by histology, immunohistochemistry, and electron microscopy. We performed multiplexed droplet digital polymerase chain reaction to detect spontaneous Kras mutations in PtenΔDuct/ΔDuct mice and study the effects of Ras pathway activation on initiation and progression of IPMNs. We obtained 2 pancreatic sections from a patient with an invasive pancreatobiliary IPMN and analyzed the regions with and without the invasive IPMN (control tissue) by immunohistochemistry. RESULTS: Mice with ductal cell-specific disruption of Pten but not control mice developed sporadic, macroscopic, intraductal papillary lesions with histologic and molecular features of human IPMNs. PtenΔDuct/ΔDuct mice developed IPMNs of several subtypes. In PtenΔDuct/ΔDuct mice, 31.5% of IPMNs became invasive; invasion was associated with spontaneous mutations in Kras. KrasG12D;PtenΔDuct/ΔDuct mice all developed invasive IPMNs within 1 month. In KrasG12D;PtenΔDuct/+ mice, 70% developed IPMN, predominately of the pancreatobiliary subtype, and 63.3% developed PDAC. In all models, IPMNs and PDAC expressed the duct-specific lineage tracing marker yellow fluorescent protein. In immunohistochemical analyses, we found that the invasive human pancreatobiliary IPMN tissue had lower levels of PTEN and increased levels of phosphorylated (activated) ERK compared with healthy pancreatic tissue. CONCLUSIONS: In analyses of mice with ductal cell-specific disruption of Pten, with or without activated Kras, we found evidence for a ductal cell origin of IPMNs. We also showed that PTEN loss and activated Kras have synergistic effects in promoting development of IPMN and progression to PDAC.


Sujet(s)
Carcinome du canal pancréatique/enzymologie , Transformation cellulaire néoplasique/métabolisme , Tumeurs kystiques, mucineuses et séreuses/enzymologie , Phosphohydrolase PTEN/déficit , Conduits pancréatiques/enzymologie , Tumeurs du pancréas/enzymologie , Protéines proto-oncogènes p21(ras)/métabolisme , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignage cellulaire , Mouvement cellulaire , Prolifération cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Évolution de la maladie , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Prédisposition génétique à une maladie , Humains , Souris de lignée C57BL , Souris knockout , Mutation , Invasion tumorale , Tumeurs kystiques, mucineuses et séreuses/génétique , Tumeurs kystiques, mucineuses et séreuses/anatomopathologie , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Conduits pancréatiques/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phénotype , Protéines proto-oncogènes p21(ras)/génétique , Transduction du signal , Facteurs temps
5.
J Pathol ; 243(1): 65-77, 2017 09.
Article de Anglais | MEDLINE | ID: mdl-28639695

RÉSUMÉ

Acinar-to-ductal metaplasia (ADM) is a reversible epithelial transdifferentiation process that occurs in the pancreas in response to acute inflammation. ADM can rapidly progress towards pre-malignant pancreatic intraepithelial neoplasia (PanIN) lesions in the presence of mutant KRas and ultimately pancreatic adenocarcinoma (PDAC). In the present work, we elucidate the role and related mechanism of glycogen synthase kinase-3beta (GSK-3ß) in ADM development using in vitro 3D cultures and genetically engineered mouse models. We show that GSK-3ß promotes TGF-α-induced ADM in 3D cultured primary acinar cells, whereas deletion of GSK-3ß attenuates caerulein-induced ADM formation and PanIN progression in KrasG12D transgenic mice. Furthermore, we demonstrate that GSK-3ß ablation influences ADM formation and PanIN progression by suppressing oncogenic KRas-driven cell proliferation. Mechanistically, we show that GSK-3ß regulates proliferation by increasing the activation of S6 kinase. Taken together, these results indicate that GSK-3ß participates in early pancreatitis-induced ADM and thus could be a target for the treatment of chronic pancreatitis and the prevention of PDAC progression. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Sujet(s)
Cellules acineuses/enzymologie , Épithélioma in situ/prévention et contrôle , Transdifférenciation cellulaire , Glycogen synthase kinase 3 beta/déficit , Pancréas exocrine/enzymologie , Conduits pancréatiques/enzymologie , Tumeurs du pancréas/prévention et contrôle , Pancréatite/enzymologie , Cellules acineuses/effets des médicaments et des substances chimiques , Cellules acineuses/anatomopathologie , Animaux , Épithélioma in situ/enzymologie , Épithélioma in situ/génétique , Épithélioma in situ/anatomopathologie , Prolifération cellulaire , Transdifférenciation cellulaire/effets des médicaments et des substances chimiques , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Cellules cultivées , Céruléine , Modèles animaux de maladie humaine , Évolution de la maladie , Prédisposition génétique à une maladie , Glycogen synthase kinase 3 beta/génétique , Protéines à homéodomaine/génétique , Mâle , Métaplasie , Souris knockout , Pancréas exocrine/effets des médicaments et des substances chimiques , Pancréas exocrine/anatomopathologie , Conduits pancréatiques/effets des médicaments et des substances chimiques , Conduits pancréatiques/anatomopathologie , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Pancréatite/induit chimiquement , Pancréatite/génétique , Pancréatite/anatomopathologie , Phénotype , Protéines proto-oncogènes p21(ras)/génétique , Ribosomal Protein S6 Kinases/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Facteurs temps , Transactivateurs/génétique , Facteur de nécrose tumorale alpha/pharmacologie
6.
PLoS One ; 10(5): e0126432, 2015.
Article de Anglais | MEDLINE | ID: mdl-25993003

RÉSUMÉ

The mechanism by which pancreas secretes high HCO3- has not been fully resolved. This alkaline secretion, formed in pancreatic ducts, can be achieved by transporting HCO3- from serosa to mucosa or by moving H+ in the opposite direction. The aim of the present study was to determine whether H+/K+-ATPases are expressed and functional in human pancreatic ducts and whether proton pump inhibitors (PPIs) have effect on those. Here we show that the gastric HKα1 and HKß subunits (ATP4A; ATP4B) and non-gastric HKα2 subunits (ATP12A) of H+/K+-ATPases are expressed in human pancreatic cells. Pumps have similar localizations in duct cell monolayers (Capan-1) and human pancreas, and notably the gastric pumps are localized on the luminal membranes. In Capan-1 cells, PPIs inhibited recovery of intracellular pH from acidosis. Furthermore, in rats treated with PPIs, pancreatic secretion was inhibited but concentrations of major ions in secretion follow similar excretory curves in control and PPI treated animals. In addition to HCO3-, pancreas also secretes K+. In conclusion, this study calls for a revision of the basic model for HCO3- secretion. We propose that proton transport is driving secretion, and that in addition it may provide a protective pH buffer zone and K+ recirculation. Furthermore, it seems relevant to re-evaluate whether PPIs should be used in treatment therapies where pancreatic functions are already compromised.


Sujet(s)
H(+)-K(+)-Exchanging ATPase/métabolisme , Pancréas/effets des médicaments et des substances chimiques , Pancréas/métabolisme , Inhibiteurs de la pompe à protons/pharmacologie , Animaux , Lignée cellulaire , Muqueuse gastrique/effets des médicaments et des substances chimiques , Muqueuse gastrique/enzymologie , H(+)-K(+)-Exchanging ATPase/génétique , Humains , Imidazoles/pharmacologie , Mâle , Oméprazole/pharmacologie , Conduits pancréatiques/effets des médicaments et des substances chimiques , Conduits pancréatiques/enzymologie , Suc pancréatique/métabolisme , Rats , Rat Wistar
7.
Hepatogastroenterology ; 61(131): 842-8, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-26176084

RÉSUMÉ

BACKGROUND/AIMS: Main pancreatic duct dilatation raises concerns about the possibility of pancreatobiliary malignancy. We evaluated the etiologic yield of endosonography (EUS) for main pancreatic duct dilatation without definite pathology on Ultrasonography (US). METHODOLOGY: A retrospective review was conducted in 54 consecutive patients referred for EUS. RESULTS: No pathological finding (37.0%, 20/54), followed by periampullary cancer (35.2%, 19/54), was the most common finding. Elevated alkaline phosphatase (ALK-P) and marked common bile duct (CBD) dilatation (≥ 12 mm) were the predictors of malignancy (p < 0.05). Among the 37 subjects with available ALK-P and CBD diameter, the probability of malignancy was 84.6% (11/13) for both elevated ALK-P and marked CBD dilatation, 16.7% (1/6) for isolated elevated ALK-P, 18.2% (2/11) for isolated marked CBD dilatation, and none (0/7) was for subjects with neither elevated ALK-P nor marked CBD dilatation, respectively. The overall accuracy of EUS for periampullary carcinomas was 94.7% (18/19) and for choledocholithiasis was 100% (7/7), respectively. EUS had a 100.0% (20/20) sensitivity and a 97.1% (33/34) specificity in the diagnosis of no pathological obstruction. CONCLUSIONS: EUS is accurate for main pancreatic duct dilatation without definite pathology on US, and the presence of concomitant elevated ALK-P and CBD dilatation highly suggests malignancy.


Sujet(s)
Lithiase cholédocienne/imagerie diagnostique , Endosonographie , Conduits pancréatiques/imagerie diagnostique , Tumeurs du pancréas/imagerie diagnostique , Sujet âgé , Phosphatase alcaline/sang , Marqueurs biologiques tumoraux/sang , Lithiase cholédocienne/sang , Lithiase cholédocienne/anatomopathologie , Dilatation pathologique , Femelle , Humains , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Conduits pancréatiques/enzymologie , Conduits pancréatiques/anatomopathologie , Tumeurs du pancréas/sang , Tumeurs du pancréas/anatomopathologie , Valeur prédictive des tests , Pronostic , Études rétrospectives , Tomodensitométrie , Régulation positive
8.
Pancreas ; 42(4): 655-62, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23271397

RÉSUMÉ

OBJECTIVES: Cigarette smoking is a major risk factor for pancreatic cancer (PaCa). However, the mechanisms of smoking-induced PaCa remain unknown. Here we investigated the effect of smoking compounds on cell death pathways in pancreatic ductal cells, precursors of PaCa. METHODS: Human pancreatic ductal cells (HPDE6-c7) were cultured with cigarette smoke extract (CSE) or smoking compound 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Apoptosis and autophagy were assessed by DNA fragmentation and immunofluorescence, respectively. RESULTS: Exposure to CSE or NNK decreased DNA fragmentation and up-regulated BclXL. Akt kinase was activated by smoking compounds through reactive oxygen species-dependent mechanism. Specifically, Akt activation was prevented by inhibition of nicotinamide adenine dinucleotide oxidase. Molecular or pharmacologic inhibitions of Akt prevented the antiapoptotic effect of smoking compounds. Smoking compounds stimulated rapid (1 hour) and transient activation of 5'-adenosine monophosphate-activated protein kinase and formation of autophagic vacuoles, indicating stimulation of autophagy. Repeated exposure to CSE/NNK (48 hours or longer) abolished the early activation of autophagic markers. Inhibition of Akt prevented the antiautophagic effect of long exposure to smoking compounds, indicating that smoking-induced late activation of Akt prevents autophagy. CONCLUSIONS: Long exposure of pancreatic ductal cells to smoking compounds inhibited apoptosis and autophagy. The results revealed a central role for Akt kinase in mediating key procarcinogenic effects of smoking compounds.


Sujet(s)
Conduits pancréatiques/enzymologie , Conduits pancréatiques/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Fumer/métabolisme , Fumer/anatomopathologie , Adenylate kinase/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/physiologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/physiologie , Cancérogènes/toxicité , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Cellules cultivées , Fragmentation de l'ADN/effets des médicaments et des substances chimiques , Humains , Souris , Nitrosamines/toxicité , Conduits pancréatiques/effets des médicaments et des substances chimiques , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/étiologie , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Espèces réactives de l'oxygène/métabolisme , Facteurs de risque , Fumée/effets indésirables , Fumer/effets indésirables , Nicotiana/toxicité , Protéine bcl-X/métabolisme
9.
J Biol Chem ; 286(1): 280-9, 2011 Jan 07.
Article de Anglais | MEDLINE | ID: mdl-20978133

RÉSUMÉ

Pancreas secretes fluid rich in digestive enzymes and bicarbonate. The alkaline secretion is important in buffering of acid chyme entering duodenum and for activation of enzymes. This secretion is formed in pancreatic ducts, and studies to date show that plasma membranes of duct epithelium express H(+)/HCO(3)(-) transporters, which depend on gradients created by the Na(+)/K(+)-ATPase. However, the model cannot fully account for high-bicarbonate concentrations, and other active transporters, i.e. pumps, have not been explored. Here we show that pancreatic ducts express functional gastric and non-gastric H(+)-K(+)-ATPases. We measured intracellular pH and secretion in small ducts isolated from rat pancreas and showed their sensitivity to H(+)-K(+) pump inhibitors and ion substitutions. Gastric and non-gastric H(+)-K(+) pumps were demonstrated on RNA and protein levels, and pumps were localized to the plasma membranes of pancreatic ducts. Quantitative analysis of H(+)/HCO(3)(-) and fluid transport shows that the H(+)-K(+) pumps can contribute to pancreatic secretion in several species. Our results call for revision of the bicarbonate transport physiology in pancreas, and most likely other epithelia. Furthermore, because pancreatic ducts play a central role in several pancreatic diseases, it is of high relevance to understand the role of H(+)-K(+) pumps in pathophysiology.


Sujet(s)
Hydrogénocarbonates/métabolisme , H(+)-K(+)-Exchanging ATPase/métabolisme , Conduits pancréatiques/enzymologie , Conduits pancréatiques/métabolisme , Sodium-Potassium-Exchanging ATPase/métabolisme , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Régulation de l'expression des gènes codant pour des enzymes , H(+)-K(+)-Exchanging ATPase/génétique , Concentration en ions d'hydrogène , Conduits pancréatiques/effets des médicaments et des substances chimiques , Inhibiteurs de la pompe à protons , Protons , Rats , Rat Wistar , Sodium-Potassium-Exchanging ATPase/antagonistes et inhibiteurs , Sodium-Potassium-Exchanging ATPase/génétique , Estomac/enzymologie
10.
J Neurosci Res ; 88(14): 3198-205, 2010 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-20806405

RÉSUMÉ

Noxious stimuli cause prompt phosphorylation of extracellular signal-regulated kinase (ERK) in the spinal dorsal horn that contributes to facilitation of pain sensation and is often used as an immediate marker for excitation of spinal neurons following somatic and colonic nociception. Here we asked whether two distinct pronociceptive stimuli with proteinase-activated receptor-2 (PAR2) agonists and hydrogen sulfide (H(2)S) in the pancreas cause phosphorylation of ERK in the spinal dorsal horn and also examined involvement of their possible downstream signaling molecules, transient receptor potential vanilloid-1 (TRPV1) and T-type Ca(2+) channels, respectively. Capsaicin (a TRPV1 agonist), trypsin (an endogenous PAR2 agonist), SLIGRL-NH(2) (a PAR2-activating peptide), and NaHS (an H(2)S donor) were infused into the pancreatic duct in anesthetized rats, and phosphorylated ERK in the spinal cord was detected by immunohistochemistry. Intraductal administration of capsaicin and trypsin caused prompt phosphorylation of ERK in the superficial layers of T9, but not T5 or T12, spinal dorsal horn. SLIGRL-NH(2) and NaHS, administered in the same manner, also produced ERK phosphorylation in the corresponding spinal regions. Mibefradil, a T-type Ca(2+) channel blocker, abolished the phosphorylation of ERK caused by intraductal NaHS but not SLIGRL-NH(2). In contrast, capsazepine, an inhibitor of TRPV1, suppressed the phosphorylation of ERK caused by intraductal SLIGRL-NH(2) but not NaHS. Our data thus demonstrate that pancreatic pronociceptive stimuli with PAR2 agonists and H(2)S cause ERK phosphorylation in the spinal dorsal horn, through activation of TRPV1 and T-type Ca(2+) channels, respectively, and that those two pronociceptive pathways are independent of each other.


Sujet(s)
Extracellular Signal-Regulated MAP Kinases/métabolisme , Sulfure d'hydrogène/toxicité , Système de signalisation des MAP kinases/physiologie , Nocicepteurs/métabolisme , Conduits pancréatiques/métabolisme , Cellules de la corne dorsale/métabolisme , Récepteur de type PAR-2/agonistes , Récepteur de type PAR-2/physiologie , Animaux , Inhibiteurs des canaux calciques/toxicité , Capsaïcine/analogues et dérivés , Capsaïcine/toxicité , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/physiologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mâle , Nocicepteurs/effets des médicaments et des substances chimiques , Nocicepteurs/enzymologie , Douleur rebelle/induit chimiquement , Douleur rebelle/traitement médicamenteux , Douleur rebelle/métabolisme , Conduits pancréatiques/effets des médicaments et des substances chimiques , Conduits pancréatiques/enzymologie , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/physiologie , Cellules de la corne dorsale/effets des médicaments et des substances chimiques , Cellules de la corne dorsale/enzymologie , Rats , Rat Wistar
11.
Biochem Biophys Res Commun ; 398(3): 389-94, 2010 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-20599723

RÉSUMÉ

Pancreatic ductal epithelial cells (PDECs) were induced to differentiate into insulin-producing cells by hepatocyte growth factor (HGF) in our previous study, but the mechanism through which this induction occurs is still unknown. HGF is a ligand that activates a tyrosine kinase encoded by the c-Met proto-oncogene. This activation is followed by indirect activation of multiple downstream signal transduction pathways (including MAPKs and the PI3K/AKT signaling pathways) that initiate various biological effects. Therefore, we speculated that the differentiation of PDECs is through either the MAPK signaling pathway or the PI3K/AKT signaling pathway. To test this hypothesis, isolated PDECs from adult rats were stimulated by adding HGF to their medium for 28days. Then, the expression levels of several protein kinases, including MAPKs (ERK1/2, p38, and JNK) and AKT, were determined by Western blotting to determine if specific protein kinases are activated in these pathways. Subsequently, re-isolated from adult rats and cultured PDECs were pre-treated with specific inhibitors of proteins shown to be activated in these signaling pathways; these cells were then induced to differentiate by the addition of HGF. The expression levels of protein kinases were determined by Western blotting, and the differentiation rate of insulin-positive cells was determined by flow cytometry. The change of PDEC differentiation rates were compared between the groups in which cells with or without inhibitors pretreatment to determine the specific signaling pathway(s) that may be involved in HGF-induced differentiation of PDECs. After isolating PDECs and stimulating them with HGF for 28days, the expression levels of phosphorylated ERK1/2 as well as total and phosphorylated AKT of cultured cells were significantly increased compared to the normal control group (p<0.05), suggesting that the signaling pathways involving ERK1/2 and Akt (MEK-ERK and PI3K-AKT) are activated during HGF-induced PDEC differentiation. MEK1/2 or PI3K inhibitors were separately added to the culture medium of PDECs pre-treated with HGF. These results show that compared to the HGF-treated group, the differentiation rate of insulin-positive cells was significantly decreased in the HGF/LY294002 (PI3K inhibitor) group (13.47+/-1.57% vs. 33.47+/-1.34%, p<0.05); however, the differentiation rate of insulin-positive cells was not significantly different in the HGF/PD98059 (MEK1/2 inhibitor) group. These data suggest that HGF induces PDECs to differentiate into insulin-producing cells through the PI3K/AKT signaling pathway.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance des hépatocytes/pharmacologie , Cellules à insuline/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Conduits pancréatiques/effets des médicaments et des substances chimiques , Animaux , Cellules cultivées , Cellules épithéliales/cytologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/enzymologie , Flavonoïdes/pharmacologie , Cellules à insuline/cytologie , Cellules à insuline/enzymologie , Mitogen-Activated Protein Kinase 1/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase 3/antagonistes et inhibiteurs , Conduits pancréatiques/cytologie , Conduits pancréatiques/enzymologie , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Rats , Transduction du signal
12.
J Cell Physiol ; 225(3): 720-33, 2010 Nov.
Article de Anglais | MEDLINE | ID: mdl-20533305

RÉSUMÉ

Tricellulin (TRIC) is a tight junction protein at tricellular contacts where three epithelial cells meet, and it is required for the maintenance of the epithelial barrier. To investigate whether TRIC is regulated via a c-Jun N-terminal kinase (JNK) pathway, human pancreatic HPAC cells, highly expressed at tricellular contacts, were exposed to various stimuli such as the JNK activators anisomycin and 12-O-tetradecanoylphorbol 13-acetate (TPA), and the proinflammatory cytokines IL-1ß, TNFα, and IL-1α. TRIC expression and the barrier function were moderated by treatment with the JNK activator anisomycin, and suppressed not only by inhibitors of JNK and PKC but also by siRNAs of TRIC. TRIC expression was induced by treatment with the PKC activator TPA and proinflammatory cytokines IL-1ß, TNFα, and IL-1α, whereas the changes were inhibited by a JNK inhibitor. Furthermore, in normal human pancreatic duct epithelial cells using hTERT-transfected primary cultured cells, the responses of TRIC expression to the various stimuli were similar to those in HPAC cells. TRIC expression in tricellular tight junctions is strongly regulated together with the barrier function via the JNK transduction pathway. These findings suggest that JNK may be involved in the regulation of tricellular tight junctions including TRIC expression and the barrier function during normal remodeling of epithelial cells, and prevent disruption of the epithelial barrier in inflammation and other disorders in pancreatic duct epithelial cells.


Sujet(s)
Cellules épithéliales/enzymologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Protéines membranaires/métabolisme , Conduits pancréatiques/enzymologie , Jonctions serrées/enzymologie , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Activation enzymatique , Activateurs d'enzymes/pharmacologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Humains , Médiateurs de l'inflammation/métabolisme , Interleukine-1 alpha/métabolisme , Interleukine-1 bêta/métabolisme , JNK Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Protéine-2 à domaine MARVEL , Protéines membranaires/génétique , Facteur de transcription NF-kappa B/métabolisme , Occludine , Conduits pancréatiques/effets des médicaments et des substances chimiques , Perméabilité , Protéine kinase C/antagonistes et inhibiteurs , Protéine kinase C/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Transport des protéines , Interférence par ARN , Telomerase/génétique , Telomerase/métabolisme , Jonctions serrées/effets des médicaments et des substances chimiques , Facteurs temps , Transfection , Facteur de nécrose tumorale alpha/métabolisme
13.
World J Gastroenterol ; 16(15): 1879-84, 2010 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-20397266

RÉSUMÉ

AIM: To perform a comparative analysis of clinicopathological correlations of cyclooxygenase-2 (COX-2) expression in pancreatic cancer, examined by monoclonal and polyclonal antibodies. METHODS: The COX-2 expression in 85 resection specimens of pancreatic ductal adenocarcinoma was immunohistochemically examined using both monoclonal and polyclonal antibodies. The final immunoscores were obtained by multiplying the percentage of positive cells with the numeric score reflecting the staining intensity. COX-2 expression levels were classified into three categories (0, 1+, and 2+) and the clinicopathological correlations were statistically evaluated and analyzed. RESULTS: The positive tumor expression rates of COX-2 were 80.5% using monoclonal antibody and 69.4% using polyclonal antibody. In the Kaplan-Meier analysis, no significant correlations were found between levels of COX-2 expression and overall survival (OS), but trends to longer OS were found in COX-2 negative cases using monoclonal antibody. Significantly longer disease free survival was revealed in COX-2 negative cases using monoclonal antibody (P = 0.019). No correlations between COX-2 expression levels and grade (G), tumor (T) status and nodal (N) status were demonstrated. Low histological grade showed a strong association with a longer OS (P < 0.001). Correlation of survival and T status revealed a shorter OS in T3 tumors, but the results reached only marginal statistical significance (P = 0.070). In the multivariate Cox proportional hazards regression model, histological grade, T and N status remained valuable predictors of a worse survival with borderline significance for T [hazards ratio (HR) = 4.18 for G (if G = 3, P < 0.001); HR = 1.64 for T (if T = 3, P = 0.065); HR = 2.53 for N (if N = 1, P = 0.006)]. Higher grade, T or N status was associated with a worse OS. CONCLUSION: The immunohistochemically assessed level of COX-2 expression does not seem to represent a valuable independent prognostic factor and is not superior to the conventional prognostic factors.


Sujet(s)
Cyclooxygenase 2/biosynthèse , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas/enzymologie , Tumeurs du pancréas/chirurgie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps monoclonaux/métabolisme , Femelle , Humains , Immunohistochimie/méthodes , Mâle , Adulte d'âge moyen , Conduits pancréatiques/enzymologie , Conduits pancréatiques/anatomopathologie , Pronostic , Modèles des risques proportionnels
14.
J Surg Res ; 159(1): 443-50, 2010 Mar.
Article de Anglais | MEDLINE | ID: mdl-20031158

RÉSUMÉ

The gelatinases B (MMP9) and A (MMP2) are two members of the matrix metalloproteinase (MMPs) family that are expressed in human cancer, and play a critical role in tumor cell invasion and metastasis. Caveolin-1 (Cav1) has recently been identified as a tumor metastasis modifier gene. However, the effect and mechanism of Cav1 in pancreatic carcinoma cell invasion remain unknown. In this study, we investigated the expression of Cav1, MMP2, and MMP9 in several different pancreatic carcinoma cell lines. We transfected pcDNA3.0-Cav1 plasmid and Cav1 siRNA into SW1990 and Bxpc3 cells, respectively. Using cell invasion assay, we found that overexpression of Cav1 inhibited cell invasion, whereas the knockdown of Cav1 in Bxpc3 cells promoted cell invasion. Moreover, to explore the mechanisms underlying these observations, we further investigated the expression of MMP2, MMP9, phospho-Akt, and phospho-Erk by Western blot, and the activities of MMP2 and MMP9 by gelatin zymography. The results indicated that Cav1 gene could inhibit pancreatic carcinoma cell invasion, at least in part, probably through Erk-MMP signal pathway, suggesting that the endogenous expression or re-expression of Cav1 might help therapeutically reduce their invasive potential in pancreatic carcinoma cells.


Sujet(s)
Carcinomes/enzymologie , Cavéoline-1/métabolisme , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/métabolisme , Tumeurs du pancréas/enzymologie , Cavéoline-1/génétique , Lignée cellulaire tumorale , 4H-1-Benzopyran-4-ones , Cellules épithéliales/enzymologie , Extracellular Signal-Regulated MAP Kinases/métabolisme , Flavonoïdes , Techniques de knock-down de gènes , Humains , Système de signalisation des MAP kinases , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Morpholines , Invasion tumorale , Conduits pancréatiques/enzymologie , Inhibiteurs des phosphoinositide-3 kinases , Protéines proto-oncogènes c-akt/métabolisme , Interférence par ARN , ARN messager/métabolisme , Petit ARN interférent/métabolisme
15.
Int J Oncol ; 35(2): 347-57, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19578749

RÉSUMÉ

Matriptase, also known as MT-SP1, is a type II transmembrane serine protease strongly implicated in both the development and progression of a variety of epithelial cancers. Evidence comes from studies of its expression in human cancers and from mouse models of spontaneous cancer. Matriptase is considered to be a major activator of two key stimulators of invasive growth, namely hepatocyte growth factor/scatter factor and urokinase-type plasminogen activator. The aim of this study was to examine the role of matriptase in pancreatic ductal adenocarcinoma by expression analysis and functional assays in vitro. Immunohistochemical analysis of matriptase performed on microtissue arrays and large samples of 55 pancreatic ductal adenocarcinomas and on 31 samples of normal pancreatic ducts revealed that although matriptase expression differed greatly in both malignant and normal ductal pancreatic tissue, matriptase scores were significantly (p=0.02) elevated in pancreatic ductal adenocarcinoma compared to normal pancreatic ducts. To evaluate the role of matriptase during development of pancreatic cancer, we studied the effects of newly designed matriptase inhibitors on the processing of the zymogen of urokinase-type plasminogen activator in the human adenocarcinoma cell lines AsPC-1 and BxPC-3. In both cell lines, at 1 microM, all matriptase inhibitors completely prevented zymogen activation. At lower inhibitor concentrations, the degree of inhibition of zymogen processing correlated with the affinities of the inhibitors towards matriptase indicating that this is a specific result of matriptase inhibition. Furthermore, matriptase inhibitors reduced the phosphorylation of the HGF receptor/cMet and the overall cellular invasiveness of the human pancreatic adenocarcinoma cell line AsPC-1. Our findings demonstrate for the first time that matriptase may be involved in the progression of pancreatic ductal adenocarcinoma and that matriptase inhibition may contribute to preventing the progression of this devastating disease.


Sujet(s)
Adénocarcinome/enzymologie , Carcinome du canal pancréatique/enzymologie , Tumeurs du pancréas/enzymologie , Serine endopeptidases/physiologie , Inhibiteurs de la sérine protéinase/pharmacologie , Adénocarcinome/anatomopathologie , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Humains , Immunohistochimie , Conduits pancréatiques/enzymologie , Tumeurs du pancréas/anatomopathologie , Phosphorylation , Protéines proto-oncogènes c-met/métabolisme , Serine endopeptidases/analyse
16.
Proc Natl Acad Sci U S A ; 105(50): 19915-9, 2008 Dec 16.
Article de Anglais | MEDLINE | ID: mdl-19052237

RÉSUMÉ

The regenerative process in the pancreas is of particular interest because diabetes results from an inadequate number of insulin-producing beta cells and pancreatic cancer may arise from the uncontrolled growth of progenitor/stem cells. Continued and substantial growth of islet tissue occurs after birth in rodents and humans, with additional compensatory growth in response to increased demand. In rodents there is clear evidence of pancreatic regeneration after some types of injury, with proliferation of preexisting differentiated cell types accounting for some replacement. Additionally, neogenesis or the budding of new islet cells from pancreatic ducts has been reported, but the existence and identity of a progenitor cell have been debated. We hypothesized that the progenitor cells are duct epithelial cells that after replication undergo a regression to a less differentiated state and then can form new endocrine and exocrine pancreas. To directly test whether ductal cells serve as pancreatic progenitors after birth and give rise to new islets, we generated transgenic mice expressing human carbonic anhydrase II (CAII) promoter: Cre recombinase (Cre) or inducible CreER(TM) to cross with ROSA26 loxP-Stop-loxP LacZ reporter mice. We show that CAII-expressing cells within the pancreas act as progenitors that give rise to both new islets and acini normally after birth and after injury (ductal ligation). This identification of a differentiated pancreatic cell type as an in vivo progenitor of all differentiated pancreatic cell types has implications for a potential expandable source for new islets for replenishment therapy for diabetes.


Sujet(s)
Carbonic anhydrase II/biosynthèse , Ilots pancréatiques/physiologie , Pancréas exocrine/physiologie , Conduits pancréatiques/cytologie , Régénération , Cellules souches/enzymologie , Animaux , Carbonic anhydrase II/génétique , Cellules épithéliales/cytologie , Cellules épithéliales/enzymologie , Gènes rapporteurs , Humains , Souris , Souris transgéniques , Conduits pancréatiques/enzymologie , Transplantation de cellules souches , Cellules souches/cytologie , Transgènes , beta-Galactosidase/génétique
17.
Pancreas ; 37(3): e39-44, 2008 Oct.
Article de Anglais | MEDLINE | ID: mdl-18815537

RÉSUMÉ

OBJECTIVES: To further explore the oncogenic activity of Aurora A kinase while attempting to develop a useful mouse model for pancreatic cancer, Aurora A kinase was targeted to pancreatic duodenal homeobox gene-1 (Pdx-1)-positive cells. METHODS: Aurora A kinase overexpression was targeted to mouse pancreas tissues using the Pdx-1 promoter in a transgenic model. The pancreas tissues of 7- to 11-month-old transgenic animals were evaluated for metastatic adenocarcinomas, preinvasive ductal neoplasia, or other histological anomalies. RESULTS: Examination of pancreatic tissue from Pdx-1-Aurora A transgenic mice revealed abnormalities, such as mild islet cell hyperplasia, lymphocytic infiltration, and general dysplasia between ductal/islet cell interfaces. However, most tissues from these transgenic mice were normal. CONCLUSIONS: The overexpression of Aurora A can potentially initiate the development of mild abnormalities in pancreatic tissue; however, neither preinvasive ductal neoplasia nor fully metastatic adenocarcinomas were observed. Combining the Pdx-1-Aurora A transgenic model with other genetic alterations may provide additional insight.


Sujet(s)
Protéines à homéodomaine/génétique , Ilots pancréatiques/enzymologie , Conduits pancréatiques/enzymologie , Régions promotrices (génétique) , Protein-Serine-Threonine Kinases/métabolisme , Transactivateurs/génétique , Animaux , Aurora kinase A , Aurora kinases , Humains , Hyperplasie , Ilots pancréatiques/anatomopathologie , Lymphocytes/anatomopathologie , Souris , Souris de lignée C3H , Souris de lignée C57BL , Souris transgéniques , Conduits pancréatiques/anatomopathologie , Protein-Serine-Threonine Kinases/génétique
18.
Mol Cell Biol ; 28(20): 6373-83, 2008 Oct.
Article de Anglais | MEDLINE | ID: mdl-18710955

RÉSUMÉ

During pancreas development, transcription factors play critical roles in exocrine and endocrine differentiation. Transcriptional regulation in eukaryotes occurs within chromatin and is influenced by posttranslational histone modifications (e.g., acetylation) involving histone deacetylases (HDACs). Here, we show that HDAC expression and activity are developmentally regulated in the embryonic rat pancreas. We discovered that pancreatic treatment with different HDAC inhibitors (HDACi) modified the timing and determination of pancreatic cell fate. HDACi modified the exocrine lineage via abolition and enhancement of acinar and ductal differentiation, respectively. Importantly, HDACi treatment promoted the NGN3 proendocrine lineage, leading to an increased pool of endocrine progenitors and modified endocrine subtype lineage choices. Interestingly, treatments with trichostatin A and sodium butyrate, two inhibitors of both class I and class II HDACs, enhanced the pool of beta cells. These results highlight the roles of HDACs at key points in exocrine and endocrine differentiation. They show the powerful use of HDACi to switch pancreatic cell determination and amplify specific cellular subtypes, with potential applications in cell replacement therapies in diabetes.


Sujet(s)
Lignage cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Inhibiteurs de désacétylase d'histone , Ilots pancréatiques/cytologie , Pancréas/effets des médicaments et des substances chimiques , Pancréas/enzymologie , Cellules souches/cytologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Femelle , Acides hydroxamiques/pharmacologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/enzymologie , Protéines de tissu nerveux/métabolisme , Pancréas/cytologie , Pancréas/embryologie , Pancréas exocrine/cytologie , Pancréas exocrine/effets des médicaments et des substances chimiques , Pancréas exocrine/enzymologie , Conduits pancréatiques/cytologie , Conduits pancréatiques/effets des médicaments et des substances chimiques , Conduits pancréatiques/enzymologie , Cellules sécrétant le polypeptide pancréatique/cytologie , Cellules sécrétant le polypeptide pancréatique/effets des médicaments et des substances chimiques , Cellules sécrétant le polypeptide pancréatique/enzymologie , Rats , Rat Wistar , Cellules souches/effets des médicaments et des substances chimiques , Acide valproïque/pharmacologie
19.
J Anim Physiol Anim Nutr (Berl) ; 92(3): 399-404, 2008 Jun.
Article de Anglais | MEDLINE | ID: mdl-18477323

RÉSUMÉ

The NBT-PABA test is an established method for diagnosis of pancreatic exocrine insufficiency. In the present study the NBT-PABA test was used to test and compare the efficacy of two multienzyme preparations (product A and B) differing in galenic preparation in minipigs in which pancreatic exocrine insufficiency (PEI) was induced by pancreatic duct ligation. Without enzyme substitution no distinct increase in PABA was found in blood after oral administration of NBT-PABA. Administration of both enzyme preparations led to a clear dose dependent rise in PABA-concentrations in blood. Interestingly, the two preparations showed different time curves of serum PABA concentration, indicating differences in the kinetic of proteolytic enzyme action. It is concluded that the NBT-PABA test can be a very useful test for indirectly evaluating proteolytic enzyme efficacy in vivo, and also gives information about the kinetics of enzyme action, not only the end-result of enzyme action (like digestibility trials which were used traditionally). A single test is performed in a few hours and there is no need for fistulated animals.


Sujet(s)
Acide 4-amino-benzoïque/pharmacocinétique , Insuffisance pancréatique exocrine/médecine vétérinaire , Conduits pancréatiques/enzymologie , Porc miniature , Complexe vitaminique B/pharmacocinétique , Acide 4-amino-benzoïque/sang , Administration par voie orale , Animaux , Aire sous la courbe , Relation dose-effet des médicaments , Insuffisance pancréatique exocrine/diagnostic , Ligature/médecine vétérinaire , Conduits pancréatiques/chirurgie , Tests de la fonction pancréatique/méthodes , Tests de la fonction pancréatique/médecine vétérinaire , Suidae/métabolisme , Porc miniature/métabolisme , Complexe vitaminique B/sang
20.
Pancreas ; 36(2): 153-9, 2008 Mar.
Article de Anglais | MEDLINE | ID: mdl-18376306

RÉSUMÉ

OBJECTIVE: The purpose of our study was to determine whether the phosphatidylinositol 3-kinase (PI3K)/Akt pathway contributes to expression of pancreatic duodenal homeobox-1 (PDX-1) in duct cells and the cell differentiation during pancreatic regeneration. METHODS: The role of PI3K in PDX-1 expression and duct cell differentiation with pancreatic regeneration in mice after partial pancreatectomy (Px) was examined using either wortmannin, a pharmacological PI3K inhibitor, or small-interfering RNA directed to the p85alpha regulatory subunit of PI3K. Akt phosphorylation, a marker of PI3K activation, and PDX-1 expression were assessed by Western blot analysis and immunohistochemistry. RESULTS: Both PDX-1 levels and Akt phosphorylation were concomitantly increased in pancreatic ducts after partial Px and, conversely, blocked by treatment with wortmannin or p85alpha small-interfering RNA. Pancreatic duct cell differentiation, as assessed by appearance of insulin-positive cells 3 days after partial Px, was effectively reduced by wortmannin. CONCLUSIONS: The PI3K/Akt activation plays a critical role for both PDX-1 expression and pancreatic duct cell differentiation into insulin-producing cells during pancreatic regeneration.


Sujet(s)
Protéines à homéodomaine/métabolisme , Pancréas/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Régénération , Transduction du signal , Transactivateurs/métabolisme , Androstadiènes/pharmacologie , Animaux , Technique de Western , Différenciation cellulaire , Prolifération cellulaire , Activation enzymatique , Immunohistochimie , Insuline/métabolisme , Mâle , Souris , Souris de lignée C57BL , Pancréas/cytologie , Pancréas/effets des médicaments et des substances chimiques , Pancréas/enzymologie , Pancréas/chirurgie , Pancréatectomie , Conduits pancréatiques/enzymologie , Conduits pancréatiques/métabolisme , Phosphatidylinositol 3-kinases/génétique , Inhibiteurs des phosphoinositide-3 kinases , Phosphorylation , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Interférence par ARN , Petit ARN interférent/métabolisme , Régénération/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps , Wortmannine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...