Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Cell ; 28(15): 2146-2154, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28539403

ABSTRACT

Mice with germline absence of SEC23B die perinatally, exhibiting massive pancreatic degeneration. We generated mice with tamoxifen-inducible, pancreatic acinar cell-specific Sec23b deletion. Inactivation of Sec23b exclusively in the pancreatic acinar cells of adult mice results in decreased overall pancreatic weights from pancreatic cell loss (decreased pancreatic DNA, RNA, and total protein content), as well as degeneration of exocrine cells, decreased zymogen granules, and alterations in the endoplasmic reticulum (ER), ranging from vesicular ER to markedly expanded cisternae with accumulation of moderate-density content or intracisternal granules. Acinar Sec23b deletion results in induction of ER stress and increased apoptosis in the pancreas, potentially explaining the loss of pancreatic cells and decreased pancreatic weight. These findings demonstrate that SEC23B is required for normal function of pancreatic acinar cells in adult mice.


Subject(s)
Acinar Cells/physiology , Pancreas/physiology , Vesicular Transport Proteins/metabolism , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Mice , Mice, Transgenic , Pancreas/cytology , Pancreas/metabolism , Vesicular Transport Proteins/genetics
2.
Am J Physiol Gastrointest Liver Physiol ; 299(5): G1154-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20798356

ABSTRACT

Dietary protein can stimulate pancreatic growth in the absence of CCK release, but there is little data on the regulation of CCK-independent growth. To identify mechanisms whereby protein stimulates pancreatic growth in the absence of CCK release, C57BL/6 control and CCK-null male mice were fed normal-protein (14% casein) or high-protein (75% casein) chow for 7 days. The weight of the pancreas increased by 32% in C57BL/6 mice and 26% in CCK-null mice fed high-protein chow. Changes in pancreatic weight in control mice were due to both cell hypertrophy and hyperplasia since there was an increase in protein-to-DNA ratio, total DNA content, and DNA synthesis. In CCK-null mice pancreatic growth was almost entirely due to hypertrophy with both protein-to-DNA ratio and cell size increasing without significant increases in DNA content or DNA synthesis. ERK, calcineurin, and mammalian target of rapamycin complex 1 (mTORC1) are activated in models of CCK-induced growth, but there were no differences in ERK or calcineurin activation between fasted and fed CCK-null mice. In contrast, mTORC1 activation was increased after feeding and the duration of activation was prolonged in mice fed high-protein chow compared with normal-protein chow. Changes in pancreatic weight and RNA content were completely inhibited, and changes in protein content were partially abated, when the mTORC1 inhibitor rapamycin was administered during high-protein chow feeding. Prolonged mTORC1 activation is thus required for dietary protein-induced pancreatic growth in the absence of CCK.


Subject(s)
Cholecystokinin/metabolism , Dietary Proteins/metabolism , Pancreas, Exocrine/growth & development , Transcription Factors/metabolism , Analysis of Variance , Animals , Blotting, Western , Calcineurin/metabolism , Cholecystokinin/genetics , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Organ Size , Pancreas, Exocrine/metabolism , Phosphorylation , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
3.
J Physiol ; 573(Pt 3): 775-86, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16613881

ABSTRACT

Cholecystokinin (CCK)-induced pancreatic growth in mice involves parallel increases in DNA and protein. The mammalian target of rapamycin (mTOR) signalling pathway regulates mRNA translation and its activation is implicated in growth of various tissues. The aim of this study was to elucidate whether mTOR activation is required for pancreatic growth in a mouse model of increased endogenous CCK release. In mice fed chow containing the synthetic protease inhibitor camostat, protein synthetic rates and phosphorylation of two downstream targets of mTOR, eukaryotic initiation factor 4E binding protein 1 (4E-BP1) and the ribosomal protein S6 (S6), increased in comparison with fasted controls. The camostat-induced increases in protein synthesis and 4E-BP1 and S6 phosphorylation were almost totally abolished by administration of the mTOR inhibitor rapamycin 1 h prior to camostat feeding. In contrast, the phosphorylation of ERK1/2 and JNK and the expression of the early response genes c-jun, c-fos, ATF3 and egr-1 induced by camostat feeding were not affected by rapamycin. In mice fed camostat for 7 days, the ratio of pancreatic to body weight increased by 143%, but when rapamycin was administered daily this was reduced to a 22% increase. Changes in pancreatic mass were paralleled by protein and DNA content following camostat feeding and rapamycin administration. Moreover, while BrdU incorporation, an indicator of DNA synthesis, was increased to 448% of control values after 2 days of camostat feeding, rapamycin administration completely inhibited this increase. We conclude that the mTOR signalling pathway is required for CCK-induced cell division and pancreatic growth.


Subject(s)
Gabexate/analogs & derivatives , Pancreas/drug effects , Protease Inhibitors/pharmacology , Protein Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , DNA/biosynthesis , Diet , Esters , Eukaryotic Initiation Factors , Gabexate/administration & dosage , Gabexate/pharmacology , Guanidines , Male , Mice , Mice, Inbred ICR , Organ Size , Pancreas/enzymology , Pancreas/growth & development , Phosphoproteins/metabolism , Phosphorylation , Protease Inhibitors/administration & dosage , Protein Biosynthesis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6/metabolism , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
4.
Pharmacol Toxicol ; 91(6): 297-303, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12688372

ABSTRACT

Cholecystokinin (CCK) acting through its G protein-coupled receptor is now known to activate a variety of intracellular signaling mechanisms and thereby regulate a complex array of cellular functions in pancreatic acinar cells. The best studied mechanism is the coupling through heterotrimeric G proteins of the Gq family to activate a phospholipase C leading to an increase in inositol trisphosphate and release of intracellular Ca2+. This pathway along with protein kinase C activation in response to the increase in diacylglycerol stimulates the secretion of digestive enzymes by the process of exocytosis. CCK also activates signaling pathways in acini more related to other processes. The three mitogen activated protein kinase cascades leading to ERKs, JNKs and p38 MAPK are all activated by CCK. CCK activates the ERK cascade by PKC activation of Raf which in turn activates MEK and ERKs. JNKs are activated by a distinct mechanism which requires higher concentrations of CCK. Both ERKs and JNKs are presumed to regulate gene expression. CCK activation of p38 MAPK also plays a role in regulating the actin cytoskeleton through phosphorylation of the small heat shock protein HSP27. The PI3K-PKB-mTOR pathway is activated by CCK and plays a major role in regulating protein synthesis at the translational level. This includes both activation of p70 S6K leading to phosphorylation of ribosomal protein S6 and the phosphorylation of the binding protein for initiation factor 4E leading to formation of the mRNA cap binding complex. Other signaling pathways activated by CCK receptors include NF-kappaB and a variety of tyrosine kinases. Further work is needed to understand how CCK receptors activate most of the above pathways and to better understand the biological events regulated by these diverse signaling pathways.


Subject(s)
Cholecystokinin/physiology , GTP-Binding Proteins/metabolism , Pancreas , Signal Transduction/physiology , Animals , Calcium/metabolism , Cholecystokinin/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Pancreas/enzymology , Pancreas/metabolism , Pancreas/physiology , Receptors, Cholecystokinin/metabolism , Receptors, Cholecystokinin/physiology , Rodentia
SELECTION OF CITATIONS
SEARCH DETAIL