Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(22): 3149-3158, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27991926

ABSTRACT

Chronic inflammation (CI) is a risk factor for pancreatic cancer (PC) including the most common type, ductal adenocarcinoma (PDAC), but its role and the mechanisms involved are unclear. To investigate the role of CI in PC, we generated genetic mouse models with pancreatic specific CI in the presence or absence of TP53. Mice were engineered to express either cyclooxygenase-2 (COX-2) or IκB kinase-2 (IKK2), and TP53+/+ or TP53f/f specifically in adult pancreatic acinar cells by using a full-length pancreatic elastase promoter-driven Cre. Animals were followed for >80 weeks and pancreatic lesions were evaluated histologically and immunohistochemically. The presence of K-ras mutations was assessed by direct sequencing, locked nuclei acid (LNA)-based PCR, and immunohistochemistry. We observed that sustained COX-2/IKK2 expression caused histological abnormalities of pancreas, including increased immune cell infiltration, proliferation rate and DNA damage. A minority of animals with CI developed pre-neoplastic lesions, but cancer was not observed in any TP53+/+ animals within 84 weeks. In contrast, all animals with CI-lacking TP53 developed various subtypes of PC, including acinar cell carcinoma, ductal adenocarcinoma, sarcomatoid carcinoma and neuroendocrine tumors, and all died within 65 weeks. No evidence of K-ras mutations was observed. Variations in the activity of the Hippo, pERK and c-Myc pathways were found in the diverse cancer subtypes. In summary, chronic inflammation is extremely inefficient at inducing PC in the presence of TP53. However, in the absence of TP53, CI leads to the development of several rare K-ras-independent forms of PC, with infrequent PDAC. This may help explain the rarity of PDAC in persons with chronic inflammatory conditions.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Inflammation/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Chronic Disease , Disease Models, Animal , Genes, ras , Inflammation/genetics , Inflammation/metabolism , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
2.
Oncogene ; 33(4): 532-5, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-23334325

ABSTRACT

Oncogenic Ras mutations are widely considered to be locked in a permanent 'On' state and 'constitutively active'. Yet, many healthy people have cells possessing mutant Ras without apparent harm, and in animal models mutant Ras causes transformation only after upregulation of Ras activity. Here, we demonstrate that oncogenic K-Ras is not constitutively active but can be readily activated by upstream stimulants to lead to prolonged strong Ras activity. These data indicate that in addition to targeting K-Ras downstream effectors, interventions to reduce K-Ras activation may have important cancer-preventive value, especially in patients with oncogenic Ras mutations. As other small G proteins are regulated in a similar manner, this concept is likely to apply broadly to the entire Ras family of molecules.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Electrophoresis, Polyacrylamide Gel , Genes, ras/genetics , Humans , Mice , Mice, Mutant Strains , Mutation , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
3.
Gastroenterology ; 121(6): 1380-90, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729117

ABSTRACT

BACKGROUND & AIMS: Pancreatic acinar cells from various species express cholecystokinin (CCK) A, CCK-B, or a combination of these CCK receptor subtypes. The presence and functional roles of CCK receptors on human acinar cells remain unclear. METHODS: Acini isolated from human pancreas were treated with CCK receptor agonists, CCK-8 and gastrin, and an agonist for m3 muscarinic acetylcholine receptors (m3 AchR), carbachol. Functional parameters measured included intracellular [Ca(2+)], amylase secretion, and ERK phosphorylation. Binding studies were performed using (125)I-CCK-8. Expression of messenger RNAs (mRNAs) was determined using real-time quantitative reverse-transcription polymerase chain reaction (RT-PCR) and localized by in situ hybridization. RESULTS: Human acini did not respond to CCK agonists. In contrast, they responded to carbachol with robust increases in each of the functional parameters. Moreover, the cells responded to CCK agonists after adenoviral-mediated gene transfer of CCK-A or CCK-B receptors. A low level of specific and a high level of nonspecific binding of (125)I-CCK-8 were observed. Quantitative RT-PCR indicated that the message levels for CCK-A receptors were approximately 30-fold lower than those of CCK-B receptors, which were approximately 10-fold lower than those of m3 Ach receptors. In situ hybridization indicated the presence of m3 Ach receptor and insulin mRNA but not CCK-A or CCK-B receptor mRNAs in adult human pancreas. CONCLUSIONS: These data indicate that human pancreatic acinar cells do not respond to CCK receptor agonists in terms of expected functional parameters and show that this is due to an insufficient level of receptor expression.


Subject(s)
Gastrins/pharmacology , Pancreas/drug effects , Pancreas/physiology , Sincalide/pharmacology , Humans , In Situ Hybridization , In Vitro Techniques , Insulin/genetics , Pancreas/cytology , Protein Isoforms/genetics , RNA, Messenger/metabolism , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/metabolism , Receptors, Cholinergic/genetics
4.
Am J Physiol Cell Physiol ; 281(1): C311-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11401854

ABSTRACT

Transforming growth factor-beta (TGF-beta) inhibits pancreatic acinar cell growth. In many cell types, TGF-beta mediates its growth inhibitory effects by activation of Smad proteins. Recently, it has been reported that Smad proteins may interact with the mitogen-activated protein (MAP) kinase signaling pathways. In this study, we report on the interactions between the TGF-beta and MAP kinase signaling pathways in isolated rat pancreatic acinar cells. TGF-beta activated the MAP kinases extracellular signal-related kinases (ERKs) and p38 in pancreatic acinar cells, but had no effect on c-jun NH2-terminal kinase activity. Activation of MAP kinase by TGF-beta was maximal 4 h after treatment. The ability of TGF-beta to activate ERKs was concentration dependent and dependent on protein synthesis. TGF-beta's stimulation of ERK activation was blocked by PD-98059, an inhibitor of MAP kinase kinase 1, and by adenoviral transfer of dominant negative RasN17. Furthermore, adenoviral-mediated expression of dominant negative Smad4 blocked the ability of TGF-beta to activate acinar cell MAP kinase, demonstrating that this activation is downstream of Smads. The biological relevance of ERK activation by TGF-beta was indicated by demonstrating that inhibition of ERK signaling by PD-98059 blocked the ability of TGF-beta to activate the transcription factor activator protein-1. These studies provide new insight into the signaling mechanisms by which TGF-beta mediates biological actions in pancreatic acinar cells.


Subject(s)
DNA-Binding Proteins/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Pancreas/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Adenoviridae/genetics , Animals , Cholecystokinin/pharmacology , Cycloheximide/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, Reporter , Immunoblotting , In Vitro Techniques , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pancreas/cytology , Pancreas/enzymology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Wistar , Smad4 Protein , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases , ras Proteins/genetics , ras Proteins/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 280(6): G1247-53, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352818

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a potent inhibitor of pancreatic acinar cell growth. Smad4 is a central mediator in the TGF-beta signaling pathway. To study the effect of Smad4 on pancreatic growth, cell cycle protein expression, and the expression of a TGF-beta-responsive promoter in vitro, we constructed an adenovirus containing dominant-negative COOH terminal truncated Smad4 (AddnSmad4) downstream of the rat elastase promoter. Acinar cells expressed dominant-negative Smad4 within 8 h after infection, and expression persisted for 72 h. Mouse pancreatic acini were infected with either AddnSmad4 or control adenovirus expressing green fluorescent protein, and TGF-beta was added 8 h after infection. Acinar cells were then incubated for 1, 2, or 3 days, and [(3)H]thymidine incorporation was determined. AddnSmad4 significantly reduced TGF-beta inhibition of [(3)H]thymidine incorporation, with maximal effects on day 3. AddnSmad4 also completely blocked TGF-beta-mediated growth inhibition in the presence of basic fibroblast growth factor. We next examined the effects of AddnSmad4 on TGF-beta-induced expression of the cell cycle regulatory proteins p21(Cip1) and p27(Kip1). TGF-beta induced upregulation of p21(Cip1), which was completely blocked by AddnSmad4. AddnSmad4 also inhibited TGF-beta-induced expression of the TGF-beta-responsive luciferase reporter 3TP-Lux. These results show that Smad4 is essential in TGF-beta-mediated signaling in pancreatic acinar cells.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/pharmacology , Pancreas/physiology , Signal Transduction/drug effects , Trans-Activators/pharmacology , Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins , Adenoviridae/genetics , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/antagonists & inhibitors , Cyclins/metabolism , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Gene Transfer Techniques , Genes, Dominant , Genes, Reporter/physiology , Luciferases/genetics , Male , Mice , Microtubule-Associated Proteins/metabolism , Pancreas/cytology , Smad4 Protein , Thymidine/metabolism , Trans-Activators/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects
6.
Am J Physiol Cell Physiol ; 280(3): C465-72, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171565

ABSTRACT

In the cholecystokinin (CCK) hyperstimulation model of acute pancreatitis, two early intracellular events, activation of trypsinogen and activation of nuclear factor-kappaB (NF-kappaB), are thought to be important in the development of the disease. In this study, the relationship between these two events was investigated. NF-kappaB activity was monitored by using a DNA binding assay and mob-1 chemokine gene expression. Intracellular trypsin activity was measured by using a fluorogenic substrate. Protease inhibitors including FUT-175, Pefabloc, and E-64d prevented CCK stimulation of intracellular trypsinogen and NF-kappaB activation. Likewise, the NF-kappaB inhibitors pyrrolidine dithiocarbamate and N-acetyl-L-cysteine inhibited CCK stimulation of NF-kappaB and intracellular trypsinogen activation. These results suggested a possible codependency of these two events. However, CCK stimulated NF-kappaB activation in Chinese hamster ovary-CCK(A) cells, which do not express trypsinogen, indicating that trypsin is not necessary for CCK activation of NF-kappaB. Furthermore, adenovirus-mediated expression in acinar cells of active p65 subunits to stimulate NF-kappaB, or of inhibitory kappaB-alpha molecules to inhibit NF-kappaB, did not affect either basal or CCK-mediated trypsinogen activation. Thus trypsinogen and NF-kappaB activation are independent events stimulated by CCK.


Subject(s)
Cholecystokinin/pharmacology , Intracellular Membranes/metabolism , NF-kappa B/physiology , Pancreas/metabolism , Trypsinogen/physiology , Acetylcysteine/pharmacology , Animals , CHO Cells , Cricetinae , Enzyme Inhibitors/pharmacology , Male , NF-kappa B/antagonists & inhibitors , Pancreas/cytology , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Thiocarbamates/pharmacology , Trypsin/metabolism
7.
Ann Surg ; 232(1): 73-80, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10862198

ABSTRACT

OBJECTIVE: To investigate whether transforming growth factor beta (TGFbeta) signaling is disrupted in human pancreatic cancer cells, and to study the role of TGFbeta receptors and Smad genes. SUMMARY BACKGROUND DATA: TGFbeta is a known inhibitor of pancreatic growth. Disruption of the TGFbeta signaling pathway may play a role in pancreatic cancer development. METHODS: The effect of TGFbeta on the BxPC-3, MiaPaCa-2, and PANC-1 pancreatic cancer cell lines was evaluated by [3H]thymidine incorporation and a TGFbeta-responsive reporter assay. Expression of TGFbeta receptors and Smads 2 and 3 was assessed by cross-linking assays and reverse transcriptase-polymerase chain reaction (RT-PCR). The ability to restore TGFbeta responsiveness was evaluated by transfection of TGFbeta signaling components. RESULTS: TGFbeta produced little inhibition of DNA synthesis and did not activate a TGFbeta-responsive reporter in pancreatic cancer cell lines. 125TGFbeta cross-linking and RT-PCR confirmed the presence of TGFbeta receptors and Smad2 and Smad3 transcripts. Transfection of TGFbeta receptors or Smads 2 and 3 did not restore responsiveness. However, transfection of Smad4 into the BxPC-3 pancreatic cancer cell line restored TGFbeta responsiveness. CONCLUSIONS: Pancreatic cancer cells show loss of TGFbeta responsiveness. Smads 2 and 3 and TGFbeta receptors are not defective in the cell lines studied. Transfection of Smad4 into one of the cell lines restored TGFbeta responsiveness, suggesting an important role for Smad4 in pancreatic cancer. It is likely that other, as yet unidentified genes are important in TGFbeta resistance in pancreatic cancer cells.


Subject(s)
DNA-Binding Proteins/physiology , Pancreatic Neoplasms/physiopathology , Signal Transduction/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Animals , Humans , Pancreatic Neoplasms/genetics , Rats , Rats, Wistar , Signal Transduction/genetics , Smad2 Protein , Smad3 Protein , Smad4 Protein , Transfection , Tumor Cells, Cultured
8.
J Biol Chem ; 275(25): 19115-20, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10748042

ABSTRACT

The cholecystokinin (CCK) analog JMV-180 acts as a partial agonist in rats and a full agonist in mice. Whether this functional variability is due to species differences in CCK receptor structure or to alterations in the cellular environment is unknown. To address this question, an adenoviral construct encoding the rat CCK(A) receptor (AdCCK(A)R) was used to express the rat receptor in acini from CCK(A) receptor-deficient mice (CCK(A)R -/-). Infection of CCK(A)R -/- acini in vitro with pAdCCK(A)R led to a time-dependent increase in (125)I-CCK(8) binding. The affinity for JMV-180 of the adenovirally transferred rat and the endogenous mouse CCK(A) receptors was not different. In native mouse acini, JMV-180 acted as a full agonist (both stimulation and inhibition of amylase release). In contrast, in mouse acini expressing pAdCCK(A)R JMV-180 acted as a partial agonist (only stimulation of amylase release). In addition, the pattern of protein synthesis induced by JMV-180 in CCK(A)R -/- mouse acini infected with AdCCK(A)R resembled the pattern observed in wild-type rats (lack of inhibition) rather than the respective pattern in wild-type mice (inhibition). These data suggest that species differences in the CCK(A) receptor of rats and mice account for the observed divergence in the acinar cell response to JMV-180.


Subject(s)
Receptors, Cholecystokinin/agonists , Sincalide/analogs & derivatives , Adenoviridae/genetics , Amino Acid Sequence , Animals , Male , Mice , Molecular Sequence Data , Protein Binding , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Sincalide/pharmacology , Species Specificity
9.
Am J Physiol Cell Physiol ; 278(2): C344-51, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666030

ABSTRACT

Supraphysiological concentrations of cholecystokinin (CCK) induce chemokine expression in rat pancreatic acini through the activation of the transcription factor NF-kappaB. In the current study, the intracellular signals involved in these pathophysiological effects of CCK were investigated. CCK induction of mob-1 expression in isolated rat pancreatic acini was blocked by the protein kinase C (PKC) inhibitors GF-109203X and Ro-32-0432 and by the intracellular Ca(2+) chelator BAPTA. CCK induced NF-kappaB nuclear translocation, and DNA binding was also blocked by GF-109203X and BAPTA. Direct activation of PKC with TPA induced mob-1 chemokine expression and activated NF-kappaB DNA binding to a similar extent as did CCK. Increasing intracellular Ca(2+) using ionomycin had no effect on mob-1 mRNA levels or NF-kappaB activity. Both CCK and TPA treatments decreased inhibitory kappaB-alpha (IkappaB-alpha) levels, whereas ionomycin had no effect. However, the effects of TPA on IkappaB-alpha degradation were less complete than for CCK. In combination, TPA and ionomycin degraded IkappaB-alpha to a similar extent as CCK. Therefore, activation of NF-kappaB and mob-1 expression by supraphysiological CCK is likely mediated by both PKC activation and elevated intracellular Ca(2+).


Subject(s)
Calcium/metabolism , Cholecystokinin/pharmacology , Cytokines/genetics , NF-kappa B/metabolism , Protein Kinase C/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Carcinogens/pharmacology , Cell Nucleus/metabolism , Chelating Agents/pharmacology , Chemokine CXCL10 , Chemokines, CXC , DNA-Binding Proteins/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , In Vitro Techniques , Indoles/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Male , Maleimides/pharmacology , Pancreas/cytology , Pancreas/enzymology , Phosphorylation , RNA, Messenger/analysis , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology
10.
Curr Opin Gastroenterol ; 16(5): 404-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-17031110

ABSTRACT

The pancreatic acinar cell is a valuable cell model for understanding how activation of plasma membrane receptors generates signals that propagate, amplify, diversify, and integrate to control cellular function. A primary signaling system involves the activation of heterotrimeric G proteins that stimulate phospholipases, leading to the generation of phospholipid messengers. A major action of the phospholipid messengers is the control of cytoplasmic Ca(2+) levels. The complex mechanisms involved in controlling the initiation, form, and spatial pattern of Ca(2+) release are being revealed in increasing detail and complexity. The connections between the signaling networks and the final events of secretion are beginning to be revealed. Advances have also been made in understanding the processes that underlie the pathologic effects of receptor overactivation.

11.
Am J Physiol ; 277(1): C74-82, 1999 07.
Article in English | MEDLINE | ID: mdl-10409110

ABSTRACT

Inflammatory mediators are involved in the early phase of acute pancreatitis, but the cellular mechanisms responsible for their generation within pancreatic cells are unknown. We examined the role of nuclear factor-kappaB (NF-kappaB) in cholecystokinin octapeptide (CCK-8)-induced mob-1 chemokine expression in pancreatic acinar cells in vitro. Supraphysiological, but not physiological, concentrations of CCK-8 increased inhibitory kappaB (IkappaB-alpha) degradation, NF-kappaB activation, and mob-1 gene expression in isolated pancreatic acinar cells. CCK-8-induced IkappaB-alpha degradation was maximal within 1 h. Expression of mob-1 was maximal within 2 h. Neither bombesin nor carbachol significantly increased mob-1 mRNA or induced IkappaB-alpha degradation. Thus the concentration, time, and secretagogue dependence of mob-1 gene expression and IkappaB-alpha degradation were similar. Inhibition of NF-kappaB with pharmacological agents or by adenovirus-mediated expression of the inhibitory protein IkappaB-alpha also inhibited mob-1 gene expression. These data indicate that the NF-kappaB signaling pathway is required for CCK-8-mediated induction of mob-1 chemokine expression in pancreatic acinar cells. This supports the hypothesis that NF-kappaB signaling is of central importance in the initiation of acute pancreatitis.


Subject(s)
Cytokines/metabolism , NF-kappa B/physiology , Pancreas/metabolism , Sincalide/pharmacology , Adenoviridae/genetics , Animals , Bombesin/pharmacology , Carbachol/pharmacology , Chemokine CXCL10 , Chemokines/genetics , Chemokines, CXC , Cytokines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Transfer Techniques , I-kappa B Proteins , Male , NF-kappa B/drug effects , Pancreas/cytology , Protein Isoforms/metabolism , Pyrrolidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Thiocarbamates/pharmacology , Time Factors
12.
J Biol Chem ; 274(31): 21701-6, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419481

ABSTRACT

Activation of G(q) protein-coupled receptors can either stimulate or inhibit cell growth. Previously, these opposite effects were explained by differences in the cell models. Here we show that activation of m3 muscarinic acetylcholine receptors ectopically expressed in NIH3T3 cells can cause stimulation and inhibition of growth in the same cell. A clonal cell line was selected from cells that formed foci agonist dependently (3T3/m3 cells). In quiescent 3T3/m3 cells, carbachol stimulated DNA synthesis. In contrast, when 3T3/m3 cells were growing, either due to the presence of serum or after transformation with oncogenic v-src, carbachol inhibited growth. This inhibition was not due to reduction of extracellular signal-regulated kinase activity because carbachol induced extracellular signal-regulated kinase phosphorylation in both quiescent and growing 3T3/m3 cells. Investigating the cell cycle mechanisms involved in growth inhibition, we found that carbachol treatment decreased cyclin D1 levels, increased p21(cip1) expression, and led to hypophosphorylation of the retinoblastoma gene product (Rb). Proteasome inhibitors blocked the carbachol-induced degradation of cyclin D1. Effects on p21(cip1) were blocked by a protein kinase C inhibitor. Thus, m3 muscarinic acetylcholine receptors couple to both growth-stimulatory and -inhibitory signaling pathways in NIH3T3 cells, and the observed effects of receptor activation depend on the context of cellular growth.


Subject(s)
Carbachol/pharmacology , Cell Division/physiology , Mitogen-Activated Protein Kinases , Receptors, Muscarinic/physiology , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Transformation, Neoplastic , Clone Cells , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA/biosynthesis , Genes, src , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , N-Methylscopolamine/pharmacokinetics , Phosphorylation , Receptor, Muscarinic M3 , Receptors, Muscarinic/drug effects , Retinoblastoma Protein/metabolism
13.
Life Sci ; 64(6-7): 369-74, 1999.
Article in English | MEDLINE | ID: mdl-10069498

ABSTRACT

The cell model studied has a fundamental influence on the function and regulation of G protein linked receptors. These cell-dependent effects are illustrated in the current communication focusing on M3 muscarinic, CCK and GRP receptors. Receptors interact with multiple cellular mechanisms. The most obvious are those involved in coupling to signaling mechanisms such as G proteins. Receptors are themselves phosphorylated and dephosphorylated by cellular kinases and phosphatases. Receptors may sequester, internalize, down-regulate and recycle via interactions with a number of separate cellular mechanisms. When the number and complexity of interactions between the cell and the receptor are taken into account it is not surprising that the cell model has a primary influence on receptor function and regulation. The implications of the importance of the cell model in receptor function for studies aimed at answering physiologic questions are discussed.


Subject(s)
Receptors, Bombesin/physiology , Receptors, Cholecystokinin/physiology , Receptors, Muscarinic/physiology , Signal Transduction , Animals , Cell Line , Down-Regulation , GTP-Binding Proteins/metabolism , Pancreas/cytology , Pancreas/metabolism , Phosphorylation , Protein Binding , Receptor, Cholecystokinin A , Receptor, Muscarinic M3 , Receptors, Bombesin/metabolism , Receptors, Cholecystokinin/metabolism , Receptors, Muscarinic/metabolism
14.
Am J Physiol ; 276(2): G499-506, 1999 02.
Article in English | MEDLINE | ID: mdl-9950825

ABSTRACT

CCK stimulates pleiotrophic responses in pancreatic acinar cells; however, the intracellular signaling pathways involved are not well understood. To evaluate the role of the ras gene product in CCK actions, a strategy involving in vitro adenoviral-mediated gene delivery of a dominant-negative mutant Ras (RasN17) was utilized. Isolated acini were infected with various titers of either a control adenovirus or an adenoviral construct expressing RasN17 for 24 h before being treated with CCK. Titer-dependent expression of RasN17 in the acini was confirmed by Western blotting. Infection with control adenovirus [10(6)-10(9) plaque-forming units/mg acinar protein (multiplicity of infection of approximately 1-1,000)] had no effect on CCK stimulation of acinar cell amylase release, extracellular-regulated kinase (ERK) or c-Jun kinase (JNK) kinases, or DNA synthesis. In contrast, infection with adenovirus bearing rasN17 increased basal amylase release, inhibited CCK-mediated JNK activation, had no effect on CCK activation of ERK, and inhibited DNA synthesis. These data demonstrate important roles for Ras in specific actions of CCK on pancreatic acinar function.


Subject(s)
Gene Transfer Techniques , Mutation/genetics , Pancreas/physiology , Sincalide/antagonists & inhibitors , Sincalide/pharmacology , ras Proteins/genetics , Adenoviridae/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Culture Techniques , DNA/biosynthesis , Gene Expression Regulation/physiology , Genes, Dominant/physiology , Male , Pancreas/cytology , Pancreas/drug effects , Rats , Rats, Wistar
15.
Gastroenterology ; 113(6): 1966-75, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9394737

ABSTRACT

BACKGROUND & AIMS: The molecular mechanisms underlying pancreatitis are largely unknown. The goal of this study was to identify an early genetic event that correlated with pancreatitis. METHODS: Differential display of messenger RNAs (mRNAs) was conducted on normal pancreas vs. those of animals with secretagogue-induced pancreatitis. Northern blots from normal animals and animals with experimental acute pancreatitis were probed with cloned complementary DNAs for chemokines. Pancreatitis was induced with cerulein and by retrograde injection of bile salts. Immunocytochemistry was used to identify the source of chemokine expression. Pyrrolidine dithiocarbamate was tested for effects on chemokine expression and pancreatitis. RESULTS: A differentially amplified band was consistently observed early after cerulein hyperstimulation. This band was identified as a portion of the mob-1 gene, an alpha-chemokine. Northern analysis indicated that mRNAs for mob-1 and another chemokine, mcp-1, were induced after cerulein hyperstimulation in vivo. mob-1 mRNA was also induced by retrograde injection of bile salts and by cerulein in acinar cells in vitro. mob-1 protein was localized to exocrine cells in pancreata of diseased animals. Pyrrolidine dithiocarbamate inhibited both chemokine gene expression and early inflammatory characteristics of pancreatitis. CONCLUSIONS: Chemokines are induced in acinar cells by treatments that induce pancreatitis and may play an important role in the early stages of the disease.


Subject(s)
Chemokines/genetics , Gene Expression/physiology , Pancreas/physiopathology , Pancreatitis/genetics , Acute Disease , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL10 , Chemokines/antagonists & inhibitors , Chemokines, CXC , Cytokines/genetics , Cytokines/metabolism , Fluorescent Antibody Technique , NF-kappa B/antagonists & inhibitors , Pancreas/pathology , Pancreatitis/pathology , Pyrrolidines/pharmacology , RNA, Messenger/metabolism , Rats , Thiocarbamates/pharmacology , Time Factors
16.
Am J Physiol ; 273(5): C1449-57, 1997 11.
Article in English | MEDLINE | ID: mdl-9374628

ABSTRACT

Rat cholecystokinin-B (CCK-B) receptors were transfected into Chinese hamster ovary (CHO)-K1 (CHO-CCK-B) and Swiss 3T3 (Swiss 3T3-CCK-B) cells, and the effects of receptor activation on cell proliferation and intracellular signaling were investigated. CCK octapeptide (CCK-8) treatment had no effect on cell growth in quiescent CHO-CCK-B cells but inhibited DNA synthesis, proliferation, and colony formation when the cells were grown in fetal bovine serum (FBS). In contrast, CCK-8 stimulated DNA synthesis in quiescent Swiss 3T3-CCK-B cells and had no effect when the cells were grown in FBS. These differences in growth responses were not due to differences in the level of receptor expression, as similar numbers of receptors were present in both cell types. To determine whether the different growth effects were due to differences in receptor coupling to common second messenger pathways, we investigated the effects of CCK-8 on several known intracellular signals. In both cell types, CCK-8 stimulated increases in intracellular Ca2+ concentration and polyphosphoinositide hydrolysis with similar potencies and efficacies. CCK-8 also stimulated arachidonate release from both cell types, although the potency was higher in the CHO cells. Adenosine 3',5'-cyclic monophosphate generation was observed at high agonist concentrations in both cell types and was much greater in cells with higher receptor density. In summary, receptor activation had opposite effects on growth parameters in CHO and Swiss 3T3 cells, but only minor differences were observed in the characteristics of CCK-B receptor coupling to specific second messengers in the two cell types. Thus cellular context is a principal determinant of the biological effects of CCK-B receptor activation, and differences in biological responses may occur independently of major differences in receptor coupling.


Subject(s)
Receptors, Cholecystokinin/physiology , Sincalide/pharmacology , 3T3 Cells , Animals , Arachidonic Acid/metabolism , CHO Cells , Calcium/metabolism , Cattle , Cell Division/drug effects , Cricetinae , Cyclic AMP/metabolism , DNA/biosynthesis , Kinetics , Mice , Phosphatidylinositols/metabolism , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/biosynthesis , Recombinant Proteins/biosynthesis , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection
17.
Surgery ; 122(2): 428-33; discussion 433-4, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9288150

ABSTRACT

BACKGROUND: The development of pancreatic cancer involves an accumulation of genetic changes, including oncogene activation and mutations in tumor suppressor genes important in cell cycle regulation. As a step to developing gene therapies to restore cell cycle control, we hypothesized that adenoviral-mediated gene transfer of a constitutively active, nonphosphorylatable form of the retinoblastoma gene (AdRb) would inhibit human pancreatic tumor cell proliferation. METHODS: Transfection efficiency was assessed by beta-gal staining with an adenovirus expressing the beta-galactosidase gene (AdLacZ). The effect of AdRb on DNA synthesis in pancreatic cancer cell lines was determined by tritiated thymidine incorporation. Western blotting with an antihemagglutinin antibody directed to the hemagglutinin-tagged AdRb construct was performed to confirm transfection of pancreatic cancer cells. Apoptosis was evaluated with a TUNEL assay. RESULTS: Efficient transfection of human pancreatic cancer cell lines was achieved with AdLacZ. AdRb inhibited tritiated thymidine uptake in the cancer cell lines BxPC-3, MIA PaCa-2, and PANC-1. Western blotting confirmed transfection of cancer cells with AdRb. AdRb did not inhibit growth by apoptosis. CONCLUSIONS: Adenoviral-mediated gene delivery of constitutively active Rb produces significant growth inhibition in human pancreatic cancer cell lines and is not a result of apoptosis. Further studies examining the role of Rb in pancreatic cancer are warranted.


Subject(s)
Cell Division , Genes, Retinoblastoma , Pancreatic Neoplasms/pathology , Retinoblastoma Protein/biosynthesis , Adenoviridae , DNA, Neoplasm/biosynthesis , Genetic Vectors , Humans , Kinetics , Recombinant Fusion Proteins/biosynthesis , Thymidine/metabolism , Time Factors , Transfection , Tumor Cells, Cultured , beta-Galactosidase/biosynthesis
18.
Gastroenterology ; 112(4): 1354-61, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9098021

ABSTRACT

BACKGROUND & AIMS: Mammalian heat shock protein 27 (hsp27) is believed to function under normal physiological conditions and during cellular stress. Recent studies indicate a role for hsp27 in regulating actin-cytoskeletal dynamics. In the present study, secretagogue-regulated phosphorylation of hsp27 in rat exocrine pancreas was investigated both in vivo and in isolated acinar cells. METHODS: Western analysis after two-dimensional electrophoresis was used to measure the phosphorylation of hsp27 after treatment of rats or acinar cells with secretagogues. Cholecystokinin-stimulated mitogen-activated protein kinase-activated protein (MAPKAP) kinase 2 activity was measured after immunoprecipitation of the kinase. RESULTS: hsp27 exists as three isoforms in acini: one nonphosphorylated (pI 6.2) and two phosphorylated (pIs 5.9 and 5.7) forms. Infusion of rats with a secretory or supermaximal dose of cerulein produced an acidic shift in hsp27, indicating an increase in its phosphorylation; the higher dose, known to cause pancreatitis, had a twofold greater effect. In isolated acini, increases in hsp27 phosphorylation were evident at 10 pmol/L and maximal at 1 nmol/L cholecystokinin. The hsp27-specific kinase MAPKAP kinase 2 was activated 2.4-fold with 1 nmol/L cholecystokinin treatment. CONCLUSIONS: hsp27 phosphorylation was stimulated by low and high concentrations of cholecystokinin, both in vivo and in vitro. Phosphorylation was potentially mediated via the MAPKAP kinase 2 intracellular signaling pathway.


Subject(s)
Cholecystokinin/pharmacology , Heat-Shock Proteins/metabolism , Pancreas/metabolism , Animals , Blotting, Western , Enzyme Activation , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Pancreas/cytology , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley
19.
Gastroenterology ; 112(3): 952-9, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9041258

ABSTRACT

BACKGROUND & AIMS: Cholecystokinin (CCK) acting via CCK(A) receptors and gastrin acting via CCK(B) receptors exert trophic effects on a variety of nontransformed tissues. However, their role as hormonal regulators of pancreatic cancer is controversial. The aim of this study was to determine the effects of activation of CCK(A) and CCK(B) receptors on the growth of human pancreatic cancer cells in vitro. METHODS: Two human pancreatic cell lines MiaPaca-2 and Panc-1 were transfected stably with both CCK receptor subtypes. Effects of CCK on various growth parameters including DNA synthesis, nuclear labeling, and colony formation were evaluated. RESULTS: Cells expressing either receptor subtype, but not untransfected cells, bound ligand and mobilized Ca2+ in response to CCK. CCK treatment caused a sustained pronounced inhibition of anchorage-independent growth. Similarly, CCK treatment inhibited anchorage-dependent growth. Receptor activation caused a concentration and time-dependent reduction in [3H]thymidine incorporation and nuclear labeling in cells cultured anchored to a plastic substrate. However, these effects on anchorage-dependent growth were transient, suggesting cellular desensitization. CONCLUSIONS: These data indicate that both CCK receptor subtypes can mediate growth inhibitory responses in pancreatic cancer cell lines and raise the possibility that CCK exerts a predominant growth inhibitory action on human pancreatic cancer cells.


Subject(s)
Pancreatic Neoplasms/pathology , Receptors, Cholecystokinin/physiology , Cell Division/drug effects , DNA/biosynthesis , Dose-Response Relationship, Drug , Humans , Receptors, Cholecystokinin/analysis , Receptors, Cholecystokinin/genetics , Sincalide/pharmacology , Tumor Cells, Cultured
20.
Digestion ; 58(4): 361-7, 1997.
Article in English | MEDLINE | ID: mdl-9324163

ABSTRACT

It was recently found that cholecystokinin (CCK) activates mitogen-activated protein kinases (MAPK) in isolated rat pancreatic acini. The present study evaluates whether one or both types of CCK receptors are capable of MAPK activation in pancreatic AR42J acinar cells as well as CHO cells transfected with CCK-A or CCK-B receptors. CCK significantly increased p44 MAPK and p42 MAPK activities in AR42J cells. Minimal, half-maximal, and maximal responses were observed at 30 and 500 pM and 10 nM, respectively, after CCK-8 stimulation and at 100 pM and 1.5 and 30 nM, respectively, after gastrin stimulation. Glycine-extended gastrin had no effect at 100 nM and a small but significant effect at 1 microM. The CCK-B receptor antagonist L365,260 almost totally blocked MAPK activation in AR42J cells after stimulation with gastrin and glycine-extended gastrin and substantially reduced the activation of both kinases by CCK-8, while the CCK-A receptor antagonist L364,718 was much less effective. The CCK-A-selective agonist A71376, however, was an effective stimulant of MAPK activity. In an alternative approach, stably transfected CHO cells bearing either CCK-A or CCK-B receptors were stimulated with CCK-8. Each receptor induced a time-dependent increase in activity of both MAPKs by five- to sixfold in CCK-A- and CCK-B-bearing cells. In conclusion, both CCK-A and CCK-B receptors activate MAPK in AR42J cells and in transfected CHO cells.


Subject(s)
MAP Kinase Kinase Kinases , Phenylurea Compounds , Protein Kinases/metabolism , Receptors, Cholecystokinin/physiology , Animals , Benzodiazepinones/pharmacology , Blotting, Western , CHO Cells/drug effects , CHO Cells/metabolism , Carcinoma, Acinar Cell/physiopathology , Cells, Cultured , Cricetinae , Devazepide , Dose-Response Relationship, Drug , Gastrins/pharmacology , MAP Kinase Kinase Kinase 4 , Oligopeptides/pharmacology , Pancreatic Neoplasms/physiopathology , Protein Kinases/drug effects , Rats , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/drug effects , Reference Values , Sincalide/pharmacology , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...