1
|
Zhai H, Shi Y, Chen X, Wang J, Lu Y, Zhang F, Liu Z, Lei T, Fan D. CacyBP/SIP promotes the proliferation of colon cancer cells. PLoS One 2017; 12:e0169959. [PMID: 28196083 PMCID: PMC5308830 DOI: 10.1371/journal.pone.0169959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/25/2016] [Indexed: 01/30/2023] Open
Abstract
CacyBP/SIP is a component of the ubiquitin pathway and is overexpressed in several transformed tumor tissues, including colon cancer, which is one of the most common cancers worldwide. It is unknown whether CacyBP/SIP promotes the proliferation of colon cancer cells. This study examined the expression level, subcellular localization, and binding activity of CacyBP/SIP in human colon cancer cells in the presence and absence of the hormone gastrin. We found that CacyBP/SIP was expressed in a high percentage of colon cancer cells, but not in normal colonic surface epithelium. CacyBP/SIP promoted the cell proliferation of colon cancer cells under both basal and gastrin stimulated conditions as shown by knockdown studies. Gastrin stimulation triggered the translocation of CacyBP/SIP to the nucleus, and enhanced interaction between CacyBP/SIP and SKP1, a key component of ubiquitination pathway which further mediated the proteasome-dependent degradation of p27kip1 protein. The gastrin induced reduction in p27kip1 was prevented when cells were treated with the proteasome inhibitor MG132. These results suggest that CacyBP/SIP may be promoting growth of colon cancer cells by enhancing ubiquitin-mediated degradation of p27kip1.
Collapse
Affiliation(s)
- Huihong Zhai
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiong Chen
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jun Wang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Faming Zhang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhengxiong Liu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ting Lei
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- * E-mail:
| |
Collapse
|
2
|
Selvik LKM, Rao S, Steigedal TS, Haltbakk I, Misund K, Bruland T, Prestvik WS, Lægreid A, Thommesen L. Salt-inducible kinase 1 (SIK1) is induced by gastrin and inhibits migration of gastric adenocarcinoma cells. PLoS One 2014; 9:e112485. [PMID: 25384047 PMCID: PMC4226541 DOI: 10.1371/journal.pone.0112485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022] Open
Abstract
Salt-inducible kinase 1 (SIK1/Snf1lk) belongs to the AMP-activated protein kinase (AMPK) family of kinases, all of which play major roles in regulating metabolism and cell growth. Recent studies have shown that reduced levels of SIK1 are associated with poor outcome in cancers, and that this involves an invasive cellular phenotype with increased metastatic potential. However, the molecular mechanism(s) regulated by SIK1 in cancer cells is not well explored. The peptide hormone gastrin regulates cellular processes involved in oncogenesis, including proliferation, apoptosis, migration and invasion. The aim of this study was to examine the role of SIK1 in gastrin responsive adenocarcinoma cell lines AR42J, AGS-GR and MKN45. We show that gastrin, known to signal through the Gq/G11-coupled CCK2 receptor, induces SIK1 expression in adenocarcinoma cells, and that transcriptional activation of SIK1 is negatively regulated by the Inducible cAMP early repressor (ICER). We demonstrate that gastrin-mediated signalling induces phosphorylation of Liver Kinase 1B (LKB1) Ser-428 and SIK1 Thr-182. Ectopic expression of SIK1 increases gastrin-induced phosphorylation of histone deacetylase 4 (HDAC4) and enhances gastrin-induced transcription of c-fos and CRE-, SRE-, AP1- and NF-κB-driven luciferase reporter plasmids. We also show that gastrin induces phosphorylation and nuclear export of HDACs. Next we find that siRNA mediated knockdown of SIK1 increases migration of the gastric adenocarcinoma cell line AGS-GR. Evidence provided here demonstrates that SIK1 is regulated by gastrin and influences gastrin elicited signalling in gastric adenocarcinoma cells. The results from the present study are relevant for the understanding of molecular mechanisms involved in gastric adenocarcinomas.
Collapse
Affiliation(s)
- Linn-Karina M. Selvik
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Technology, Sør-Trøndelag University College, Trondheim, Norway
| | - Shalini Rao
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Technology, Sør-Trøndelag University College, Trondheim, Norway
| | - Tonje S. Steigedal
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ildri Haltbakk
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristine Misund
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Torunn Bruland
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Wenche S. Prestvik
- Department of Technology, Sør-Trøndelag University College, Trondheim, Norway
| | - Astrid Lægreid
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Liv Thommesen
- Department of Technology, Sør-Trøndelag University College, Trondheim, Norway
- * E-mail:
| |
Collapse
|
3
|
Zhai HH, Meng J, Wang JB, Liu ZX, Li YF, Feng SS. CacyBP/SIP nuclear translocation induced by gastrin promotes gastric cancer cell proliferation. World J Gastroenterol 2014; 20:10062-10070. [PMID: 25110433 PMCID: PMC4123335 DOI: 10.3748/wjg.v20.i29.10062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 03/12/2014] [Accepted: 05/05/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of nuclear translocation of calcyclin binding protein, also called Siah-1 interacting protein (CacyBP/SIP), in gastric carcinogenesis.
METHODS: The expression of CacyBP/SIP protein in gastric cancer cell lines was detected by Western blot. Immunofluorescence experiments were performed on gastric cancer cell lines that had been either unstimulated or stimulated with gastrin. To confirm the immunofluorescence findings, the relative abundance of CacyBP/SIP in nuclear and cytoplasmic compartments was assessed by Western blot. The effect of nuclear translocation of CacyBP/SIP on cell proliferation was examined using MTT assay. The colony formation assay was used to measure clonogenic cell survival. The effect of CacyBP/SIP nuclear translocation on cell cycle progression was investigated. Two CacyBP/SIP-specific siRNA vectors were designed and constructed to inhibit CacyBP/SIP expression in order to reduce the nuclear translocation of CacyBP/SIP, and the expression of CacyBP/SIP in stably transfected cells was determined by Western blot. The effect of inhibiting CacyBP/SIP nuclear translocation on cell proliferation was then assessed.
RESULTS: CacyBP/SIP protein was present in most of gastric cancer cell lines. In unstimulated cells, CacyBP/SIP was distributed throughout the cytoplasm; while in stimulated cells, CacyBP/SIP was found mainly in the perinuclear region. CacyBP/SIP nuclear translocation generated a growth-stimulatory effect on cells. The number of colonies in the CacyBP/SIP nuclear translocation group was significantly higher than that in the control group. The percentage of stimulated cells in G1 phase was significantly lower than that of control cells (69.70% ± 0.46% and 65.80% ± 0.60%, control cells and gastrin-treated SGC7901 cells, P = 0.008; 72.99% ± 0.46% and 69.36% ± 0.51%, control cells and gastrin-treated MKN45 cells, P = 0.022). CacyBP/SIPsi1 effectively down-regulated the expression of CacyBP/SIP, and cells stably transfected by CacyBP/SIPsi1 were then chosen for further cellular assays. In CacyBP/SIPsi1 stably transfected cells, CacyBP/SIP was shown to be distributed throughout the cytoplasm, irregardless of whether they were stimulated or not. After CacyBP/SIP nuclear translocation was reduced, there had no major effect on cell proliferation, as shown by MTT assay. There had no enhanced anchorage-dependent growth upon stimulation, as indicated by colony formation in flat plates. No changes appeared in the percentage of cells in G0-G1 phase in either cell line (71.09% ± 0.16% and 70.86% ± 0.25%, control cells and gastrin-treated SGC7901-CacyBP/SIPsi1 cells, P = 0.101; 74.17% ± 1.04% and 73.07% ± 1.00%, control cells and gastrin-treated MKN45-CacyBP/SIPsi1 cells, P = 0.225).
CONCLUSION: CacyBP/SIP nuclear translocation promotes the proliferation and cell cycle progression of gastric cancer cells.
Collapse
|
4
|
Copps J, Murphy RF, Lovas S. The production and role of gastrin-17 and gastrin-17-gly in gastrointestinal cancers. Protein Pept Lett 2010; 16:1504-18. [PMID: 20001914 DOI: 10.2174/092986609789839269] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gastrointestinal peptide hormone gastrin is responsible for initiating the release of gastric acid in the stomach in response to the presence of food and/or humoral factors such as gastrin releasing peptide. However, it has a role in the growth and maintenance of the gastric epithelium, and has been implicated in the formation and growth of gastric cancers. Hypergastrinemia resulting from atrophic gastritis and pernicious anemia leads to hyperplasia and carcinoid formation in rats, and contributes to tumor formation in humans. Additionally, gastrin has been suspected to play a role in the formation and growth of cancers of the colon, but recent studies have instead implicated gastrin processing intermediates, such as gastrin-17-Gly, acting upon a putative, non-cholecystokinin receptor. This review summarizes the production and chemical structures of gastrin and of the processing intermediate gastrin-17-Gly, as well as their activities in the gastrointestinal tract, particularly the promotion of colon cancers.
Collapse
Affiliation(s)
- Jeffrey Copps
- Department of Biomedical Sciences, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA
| | | | | |
Collapse
|
5
|
Zhai HH, Chen X, Lu YY, Wang X, Fan DM. Expression and nucleus translocation of calcyclin binding protein in colon cancer cells. Shijie Huaren Xiaohua Zazhi 2008; 16:3953-3957. [DOI: 10.11569/wcjd.v16.i35.3953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To verify expression and location of CacyBP/SIP in colon cancer cells, so to explore the role of CacyBP/SIP in colon cancer.
METHODS: The expression of CacyBP/SIP in the three colon cancer cell lines was detected using Western blot. The location of CacyBP/SIP in the colon cancer cell and location after stimulation by KCl, gastrin(10-8 mol/L) were determined separately by indirect immunofluorescence. The change of Ca2+ concentration in the colon cancer cells stimulated by gastrin was detected by laser copolymerization focal.
RESULTS: CacyBP/SIP was detected in the HT29 and SW480 colon cancer cells and undetected in the Lovo colon cancer cells. CacyBP/SIP was located in the cytoplasm of colon cancer cells. After stimulation by KCl, CacyBP/SIP translocated into nucleus; after KCl was washed off, CacyBP/SIP was relocated in the cytoplasm. Upon stimulation by gastrin, CacyBP/SIP also translocated into the nucleus. Compared with the colon cancer cell without gastrin, intra-cellular Ca2+ concentration with gastrin was increased (25.33 ± 0.57 vs 21 ± 1, P < 0.05).
CONCLUSION: CacyBP/SIP is expressed in the colon cancer cells and located in the cytoplasm. CacyBP/SIP could translocate into the nucleus after increased intra-cellular Ca2+ concentration. These results suggest that CacyBP/SIP may be involved in the progression of colon cancer through nuclear translocation.
Collapse
|
6
|
Harper EA, Roberts SP, Kalindjian SB. Thermodynamic analysis of ligands at cholecystokinin CCK2 receptors in rat cerebral cortex. Br J Pharmacol 2007; 151:1352-67. [PMID: 17592503 PMCID: PMC2189820 DOI: 10.1038/sj.bjp.0707355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 05/01/2007] [Accepted: 05/22/2007] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Several studies using radioligand binding assays, have shown that measurement of thermodynamic parameters can allow discrimination of agonists and antagonists (Weiland et al., 1979; Borea et al., 1996a). Here we investigate whether agonists and antagonists can be thermodynamically discriminated at CCK(2) receptors in rat cerebral cortex. EXPERIMENTAL APPROACH The pK(L) of [(3)H]-JB93182 in rat cerebral cortex membranes was determined at 4, 12, 21 and 37 degrees C in 50 mM Tris-HCl buffer (buffer B pH 6.96; containing 0.089 mM bacitracin). pK(I) values of ligands of diverse chemical structure and with differing intrinsic activity (alpha), as defined by the lumen-perfused rat and mouse stomach bioassays, were determined in buffer B at 4, 12, 21 and 37 degrees C. KEY RESULTS [(3)H]-JB93182 labelled a homogeneous population of receptors in rat cerebral cortex at 4, 12, 21 and 37 degrees C and the pK(L) and B(max) were not altered by incubation temperature. [(3)H]-JB93182 binding reached equilibrium after 10, 50, 90 and 220 min at 37, 21, 12 and 4 degrees C, respectively. pK(I) values for R-L-365,260, R-L-740,093, YM220, PD134,308 and JB95008 were higher at 4 degrees C than at 37 degrees C. There was no effect of temperature on pK(I) values for pentagastrin, CCK-8S, S-L-365,260, YM022, PD140,376 and JB93242. CONCLUSIONS AND IMPLICATIONS CCK(2) receptor agonists and antagonists at rat CCK(2) receptors cannot be discriminated by thermodynamic analysis using [(3)H]-JB93182 as the radioligand.
Collapse
Affiliation(s)
- E A Harper
- James Black Foundation, 68 Half Moon Lane, Dulwich, London, UK.
| | | | | |
Collapse
|
7
|
Yang YM, Chung JM, Rhim H. Cholecystokinin-8S-Induced Intracellular Calcium Signaling in Acutely Isolated Periaqueductal Gray Neurons of the Rat. Biol Pharm Bull 2007; 30:297-302. [PMID: 17268069 DOI: 10.1248/bpb.30.297] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many behavior studies indicate that cholecystokinin (CCK) is related to nociception and anxiety/panic actions in the midbrain periaqueductal gray (PAG). We previously reported that a sulfated form of CCK octapeptide (CCK-8S) produced excitatory effects at both pre- and postsynaptic loci in PAG neurons using slice preparations and whole-cell patch-clamp recordings. Here, we further examined the detailed mechanism of CCK-8S in acutely isolated PAG neurons of the rat using fura-2-based imaging of intracellular Ca2+ concentration ([Ca2+]i) and whole-cell patch-clamp recordings. Application of 1 microM CCK-8S produced an increase of [Ca2+]i, and its effect did not desensitize. This CCK-8S-induced [Ca2+]i increase was inhibited by the CCK2 receptor antagonist L-365260 but not by the CCK1 receptor antagonist L-364718. In addition, the effect of CCK-8S was eliminated by removing extracellular Ca2+, but not by an addition of the intracellular Ca2+ reuptake inhibitor thapsigargin. When simultaneous recordings of [Ca2+]i imaging and whole-cell patch-clamp were performed, CCK-8S-induced [Ca2+]i increase was significantly reduced at a membrane holding potential of -60 mV while CCK-8S-induced inward current was still observed. Current-voltage plots revealed that CCK-8S-induced inward current reversed near the equilibrium potential for K+ ions with a decreased membrane conductance. However, CCK-8S produced a significant inhibition on high-voltage-activated Ca2+ channel currents. These results suggest that CCK-8S can excite PAG neurons by inhibiting K+ channels, and CCK-8S-induced [Ca2+]i increase occurs secondary to depolarization. The evidence presented here expands our understanding of cellular mechanisms for CCK-mediated anti-analgesic and anxiogenic actions in the PAG.
Collapse
Affiliation(s)
- Yu-Mi Yang
- Biomedical Research Center, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | | | | |
Collapse
|
8
|
Yu HG, Tong SL, Ding YM, Ding J, Fang XM, Zhang XF, Liu ZJ, Zhou YH, Liu QS, Luo HS, Yu JP. Enhanced expression of cholecystokinin-2 receptor promotes the progression of colon cancer through activation of focal adhesion kinase. Int J Cancer 2006; 119:2724-2732. [PMID: 16998832 DOI: 10.1002/ijc.22207] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Focal adhesion kinase (FAK) is suggested to be intimately involved in the progression of malignancies. Our previous research has demonstrated that activation of cholecystokinin-2 receptor (CCK2R) by gastrin stimulates a rapid activation of FAK pathway in human colon cancer cells. The purpose of this study is to determine the role of CCK2R and FAK in the progression of colon cancer. In this study, matched tissue samples of primary colon cancer and adjacent normal colon mucosa from the same patient were collected from 45 patients with colon cancer undergoing surgical resection. The gastrin expression was detected using reverse transcription polymerase chain reaction (RT-PCR). The CCK2R expression was examined by in situ hybridization and RT-PCR. The expression of FAK and phosphorylated FAK at tyrosine 397 (phospho-FAK) were detected using immunohistochemistry and immunoblotting. Colo320 and SW787, 2 colon cancer cell lines with or without CCK2R expression, were recruited in this study. Antisense oligonucleotide of FAK was used to block the expression of FAK. Invasiveness and motility of colon cancer cells were detected by Boyden chamber. In this series, enhanced expression of gastrin, CCK2R, FAK and phospho-FAK were observed in colon cancer tissues. CCK2R expression correlated with expression of phospho-FAK. Coexpression of CCK2R and phospho-FAK associated with invasion and lymph node metastasis. Increased invasion and motility was induced by gastrin in Colo320 cells. Overexpression of CCK2R by stable transfection of CCK2R plasmid amplified this increase and incubation with 1 microM L-365,260, a specific CCK2R antagonist, completely inhibited the effect of gastrin. FAK antisense largely blocked the increase of invasion and motility in Colo320 cells. Our data represent the evidence for the CCK2R regulating invasion and motility of colon cancer cells, and support a role of CCK2R in the progression of colon cancer. FAK play a critical role in this CCK2R-mediated effect.
Collapse
Affiliation(s)
- Hong-Gang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
N/A, 李 茹, 陈 蔚. N/A. Shijie Huaren Xiaohua Zazhi 2006; 14:979-984. [DOI: 10.11569/wcjd.v14.i10.979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
10
|
Yu HG, Schrader H, Otte JM, Schmidt WE, Schmitz F. Rapid tyrosine phosphorylation of focal adhesion kinase, paxillin, and p130Cas by gastrin in human colon cancer cells. Biochem Pharmacol 2004; 67:135-146. [PMID: 14667936 DOI: 10.1016/j.bcp.2003.08.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the expression of CCK(2) receptors is widely reported in human colorectal cancers, little is known on its role in mediating the proliferative effects of mature amidated gastrin (G17 amide) on colorectal cancers. The purpose of the present study was to determine the effects of G17 amide on tyrosine phosphorylation of focal adhesion kinase (FAK), paxillin, and p130 Crk-associated substrate (p130(Cas)) in Colo 320 cells, a human colorectal cancer cell line which expresses CCK(2) receptors. By immunoprecipitation and immunoblotting, an increase in tyrosine phosphorylation of FAK (tyrosine-397), paxillin (tyrosine-31), and p130(Cas) was detected in a time- and dose-dependent manner. Overexpression of CCK(2) receptors in Colo 320 cells (Colo 320 WT) by stable transfection with the human CCK(2) receptor cDNA resulted in an increased tyrosine phosphorylation of FAK, paxillin, and p130(Cas). After incubation with 1 microM L-365,260, a specific CCK(2) receptor antagonist, this increase was completely inhibited. Our results demonstrate that in human colon cancer cells, gastrin caused a rapid tyrosine phosphorylation of FAK, paxillin, and p130(Cas) by activation of CCK(2) receptor. The phosphorylation of these proteins might be important in mediating gastrin effects on proliferation, apoptosis, and metastasis.
Collapse
Affiliation(s)
- Hong-Gang Yu
- Laboratory for Molecular Gastroenterology, Department of Medicine I, St. Josef-Hospital, Ruhr-University of Bochum, Gudrunstr. 56, D-44791, Bochum, Germany
| | | | | | | | | |
Collapse
|
11
|
Günther R, Carstens OC, Schmidt WE, Fölsch UR. Transient agonist-induced regulation of the cholecystokinin-A and cholecystokinin-B receptor mRNA levels in rat pancreatic acinar AR42J cells. Pancreatology 2003; 3:47-54. [PMID: 12649564 DOI: 10.1159/000069142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2001] [Accepted: 07/25/2002] [Indexed: 12/11/2022]
Abstract
BACKGROUND CCK-8 and gastrin exert multiple effects in the gastrointestinal tract and the nervous system. Their actions are mediated via the G-protein coupled CCK-A and CCK-B receptors. METHODS Rat pancreatic acinar tumor AR42J cells express both CCK receptor subtypes. This cell line was used to characterize the agonist-dependent regulation of CCK-A and CCK-B receptor gene expression. RESULTS CCK-8 (10 nM) or gastrin (10 nM) reduced CCK-A receptor mRNA expression to 56% and 53%, respectively 2 h after hormonal exposure. In contrast, the level of CCK-B receptor gene expression was upregulated to 157% and 153%, respectively. These effects are most probably linked to the CCK-B receptor in AR42J cells. The phorbolester PMA (100 nM), a protein kinase C activator, downregulated CCK-A receptor expression but did not affect CCK-B receptor gene transcription. Activation of protein kinase A by forskolin (10 microM) or Bt(2)cAMP (100 microM) is not involved in the transient regulation of CCK receptor mRNA expression. Both elevated CCK-B and decreased CCK-A receptor mRNA expression returned to basal levels 6 h after continuous stimulation. CONCLUSION These results demonstrate that CCK-A and CCK-B receptor mRNA levels are differentially regulated by their agonists via distinct signal transduction mechanisms in AR42J cells.
Collapse
Affiliation(s)
- Rainer Günther
- Laboratory of Molecular Gastroenterology, 1st Department of Medicine, Christian-Albrechts-University of Kiel, Germany.
| | | | | | | |
Collapse
|
12
|
McCormack K. A New Perspective on Signal Transduction in Neuropathic Pain The Emerging Role of the G Protein By Dimer in Transducing and Modulating Opioid Signaling. Pain 2003. [DOI: 10.1201/9780203911259.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Elberg G, Hipkin RW, Schonbrunn A. Homologous and heterologous regulation of somatostatin receptor 2. Mol Endocrinol 2002; 16:2502-14. [PMID: 12403839 DOI: 10.1210/me.2002-0207] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously demonstrated that phosphorylation of somatostatin receptor 2A (sst2A) is rapidly increased in transfected cells both by agonist and by the protein kinase C (PKC) activator phorbol myristate acetate (PMA). Here, we investigate whether PKC-mediated receptor phosphorylation is involved in the homologous or heterologous regulation of endogenous sst2 receptors in AR42J pancreatic acinar cells upon stimulation by agonist or by cholecystokinin (CCK) or bombesin (BBS). Somatostatin, PMA, CCK, and BBS all increased sst2A receptor phosphorylation 5- to 10-fold within minutes. Somatostatin binding also caused rapid internalization of the ligand-receptor complex, and PMA, CCK, and BBS all stimulated this internalization further. Additionally, sst2 receptor-mediated inhibition of adenylyl cyclase was desensitized by all treatments. Somatostatin, as well as peptidic (SMS201-995) and nonpeptidic (L-779,976) sst2 receptor agonists increased the EC(50) for somatostatin inhibition 20-fold. In contrast, pretreatment with BBS, CCK, or PMA caused a modest 2-fold increase in the EC(50) for cyclase inhibition. Whereas the PKC inhibitor GF109203X abolished sst2A receptor phosphorylation by CCK, BBS, and PMA, it did not alter the effect of somatostatin, demonstrating that these reactions were catalyzed by different kinases. Consistent with a functional role for PKC-mediated receptor phosphorylation, GF109203X prevented PMA stimulation of sst2 receptor internalization. Surprisingly, however, GF109203X did not inhibit BBS and CCK stimulation of sst2A receptor endocytosis. These results demonstrate that homologous and heterologous hormones induce sst2A receptor phosphorylation by PKC-independent and -dependent mechanisms, respectively, and produce distinct effects on receptor signaling and internalization. In addition, the heterologous hormones also modulate sst2 receptor internalization by a novel mechanism that is independent of receptor phosphorylation.
Collapse
Affiliation(s)
- Gerard Elberg
- Department of Integrative Biology and Pharmacology, University of Texas Health Sciences Center Houston, Houston, Texas 77225, USA
| | | | | |
Collapse
|
14
|
Lindström E, Eliasson L, Björkqvist M, Håkanson R. Gastrin and the neuropeptide PACAP evoke secretion from rat stomach histamine-containing (ECL) cells by stimulating influx of Ca2+ through different Ca2+ channels. J Physiol 2001; 535:663-77. [PMID: 11559765 PMCID: PMC2278808 DOI: 10.1111/j.1469-7793.2001.00663.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
1. Gastrin and PACAP stimulate secretion of histamine and pancreastatin from isolated rat stomach ECL cells. We have examined whether or not secretion depends on the free cytosolic Ca2+ concentration ([Ca2+]i) and the pathways by which gastrin and PACAP elevate [Ca2+]i. Secretion was monitored by radioimmunoassay of pancreastatin and changes in [Ca2+]i by video imaging. The patch clamp technique was used to record whole-cell currents and membrane capacitance (reflecting exocytosis). 2. In the presence of 2 mM extracellular Ca2+, gastrin and PACAP induced secretion and raised [Ca2+]i. Without extracellular Ca2+ (or in the presence of La3+) no secretion occurred. The extracellular Ca2+ concentration required to stimulate secretion was 10 times higher for gastrin than for PACAP. Depletion of intracellular Ca2+ pools by thapsigargin had no effect on the capacity of gastrin and PACAP to stimulate secretion. 3. Gastrin-evoked secretion was inhibited 60-80 % by L-type channel blockers and 40 % by the N-type channel blocker omega-conotoxin GVIA. Combining L-type and N-type channel blockers did not result in greater inhibition than L-type channel blockers alone. Whole-cell patch clamp measurements confirmed that the ECL cells are equipped with voltage-dependent inward Ca2+ currents. A 500 ms depolarising pulse from -60 mV to +10 mV which maximally opened these channels resulted in an increase in membrane capacitance of 100 fF reflecting exocytosis of secretory vesicles. 4. PACAP-evoked secretion was reduced 40 % by L-type channel blockers but was not influenced by inhibition of N-type channels. SKF 96365, a blocker of both L-type and receptor-operated Ca2+ channels, inhibited PACAP-evoked secretion by 85 %. Combining L-type channel blockade with SKF 96365 abolished PACAP-evoked secretion. 5. The results indicate that gastrin- and PACAP-evoked secretion depends on Ca2+ entry and not on mobilisation of intracellular Ca2+. While gastrin stimulates secretion via voltage-dependent L-type and N-type Ca2+ channels, PACAP acts via L-type and receptor-operated Ca2+ channels.
Collapse
Affiliation(s)
- E Lindström
- Institute of Physiological Sciences, Department of Pharmacology, University of Lund, Lund, Sweden.
| | | | | | | |
Collapse
|
15
|
Thommesen L, Nørsett K, Sandvik AK, Hofsli E, Laegreid A. Regulation of inducible cAMP early repressor expression by gastrin and cholecystokinin in the pancreatic cell line AR42J. J Biol Chem 2000; 275:4244-50. [PMID: 10660591 DOI: 10.1074/jbc.275.6.4244] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The CREM gene encodes both activators and repressors of cAMP-induced transcription. Inducible cAMP early repressor (ICER) isoforms are generated upon activation of an alternative, intronic promoter within the CREM gene. ICER is proposed to down-regulate both its own expression and the expression of other genes that contain cAMP-responsive elements such as a number of growth factors. Thus, ICER has been postulated to play a role in proliferation and differentiation. Here we show that ICER gene expression is induced by gastrin, cholecystokinin (CCK), and epidermal growth factor in AR42J cells. The time course of gastrin- and CCK-mediated ICER induction is rapid and transient, similar to forskolin- and phorbol 12-myristate 13-acetate-induced ICER expression. The specific CCK-B receptor antagonist L740,093 blocks the gastrin but not the CCK response, indicating that both the CCK-B and the CCK-A receptor can mediate ICER gene activation. Noteworthy, CREB is constitutively phosphorylated at Ser-133 in AR42J cells, and ICER induction proceeds in the absence of increased CREB Ser(P)-133. Gastrin-mediated ICER induction was not reduced in the presence of the protein kinase A inhibitor H-89, indicating a protein kinase A-independent mechanism. This is the first report on ICER inducibility via G(q)/G(11) protein-coupled receptors.
Collapse
Affiliation(s)
- L Thommesen
- Department of Physiology and Biomedical Engineering, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
| | | | | | | | | |
Collapse
|
16
|
Bestervelt L, Barr B, Dethloff L. Divergent proliferative responses to a gastrin receptor ligand in synchronized and unsynchronized rat pancreatic AR42J tumour cells. Cell Signal 2000; 12:53-61. [PMID: 10676848 DOI: 10.1016/s0898-6568(99)00067-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Depending upon experimental model, the CCK-B/gastrin receptor ligand CI-988 exhibits either agonist or antagonist activity. To confirm that CI-988 behaves as an antagonist toward gastrin-stimulated growth, its effects on cell proliferation were investigated in unsynchronized and synchronized AR42J rat pancreatic tumour cells. In unsynchronized cultures CI-988 alone had no effect, but inhibited gastrin-stimulated cell proliferation. In contrast, in synchronized cultures, CI-988 stimulated cell proliferation. Similarly, CI-988 inhibited gastrin-stimulated cAMP production in unsynchronized cells, but stimulated cAMP formation in synchronized cultures. Therefore, CI-988 stimulation of cAMP production and proliferation in AR42J cell cultures appears to be cell cycle-dependent. CI-988 inhibited gastrin-stimulated intracellular calcium ([Ca2+]i) mobilization in both populations and thus acted as an antagonist toward this pathway. Because CCK receptor densities and affinities were similar in both cell populations, the data suggest that CI-988's divergent effects on cell proliferation are governed by postreceptor signalling events which vary with cell cycle.
Collapse
Affiliation(s)
- L Bestervelt
- Department of Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Research, Ann Arbor, MI 48105, USA
| | | | | |
Collapse
|
17
|
Lambert M, Bui ND. Dexamethasone-induced decrease in HMG-CoA reductase and protein-farnesyl transferase activities does not impair ras processing in AR 4-2J cells. Mol Cell Biochem 1999; 202:101-8. [PMID: 10706000 DOI: 10.1023/a:1007016403736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Rat pancreatic acinar cells AR 4-2J respond to dexamethasone by differentiation and a decreased proliferation rate. Protein labelling by [3H]-mevalonolactone, used as a precursor of farnesyl and geranylgeranyl isoprenoid groups, was increased in the presence of dexamethasone. In these same conditions, dexamethasone decreased HMG-CoA reductase activity, leading to a diminished isotopic dilution of the mevalonate precursor. As ras proteins, known to be involved in the regulation of proliferation and differentiation, need to be farnesylated for full biological function, we also measured the level of farnesyl transferase activity and found a dose-dependent decrease in dexamethasone treated cells. Despite these negative effects of dexamethasone on mevalonate pathway, there was no appearance of non-isoprenylated forms of ras, indicating that the level of isoprenoid precursors and farnesyl transferase activity were not limiting in this model.
Collapse
Affiliation(s)
- M Lambert
- Department of Biochemistry and Nutrition, Medical School, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
18
|
Raychowdhury R, Zhang Z, Höcker M, Wang TC. Activation of human histidine decarboxylase gene promoter activity by gastrin is mediated by two distinct nuclear factors. J Biol Chem 1999; 274:20961-9. [PMID: 10409643 DOI: 10.1074/jbc.274.30.20961] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human histidine decarboxylase gene is regulated by gastrin through a cis-acting element known as the gastrin response element (GAS-RE) that was initially localized to a site (+2 to +24) downstream of the transcriptional start site. Electrophoretic mobility shift assays using sequentially deleted DNA probes and nuclear extracts from AGS-B gastric cancer cells showed that the GAS-RE is actually composed of two overlapping binding sites (GAS-RE1, +1 to +19; and GAS-RE2, +11 to +27) that bind distinct nuclear factors. Reporter gene assays demonstrated that each element alone could confer gastrin responsiveness, but the presence of both elements was required for complete gastrin response. Stimulation of AGS-B cells with gastrin for 10-20 min resulted in a >2-fold increase in factor binding. The binding was inhibited by pretreatment of AGS-B cells with cycloheximide and the MEK1 inhibitor PD98059, indicating a requirement for protein synthesis and also indicating that activation occurs through the MEK/mitogen-activated protein kinase pathway. UV cross-linking and Southwestern blot analysis showed that GAS-RE1 bound a 52-kDa protein, whereas GAS-RE2 bound a 35-kDa protein. Hence, activation of histidine decarboxylase gene promoter activity by gastrin is most likely mediated by two separate nuclear factors.
Collapse
Affiliation(s)
- R Raychowdhury
- Gastrointestinal Unit and the Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
19
|
Rodríguez-Martín E, Boyano-Adánez MC, Bodega G, Martín M, Hernández C, Quin Y, Vadillo M, Arilla-Ferreiro E. Redistribution of protein kinase C isoforms in rat pancreatic acini during lactation and weaning. FEBS Lett 1999; 445:356-60. [PMID: 10094489 DOI: 10.1016/s0014-5793(99)00133-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Freshly enzymatically isolated pancreatic acini from lactating and weaning Wistar rats were used to investigate the role of protein kinase C (PKC) isoforms during these physiologically relevant pancreatic secretory and growth processes. The combination of immunoblot and immunohistochemical analysis shows that the PKC isoforms alpha, delta, and epsilon are present in pancreatic acini from control, lactating and weaning rats. A vesicular distribution of PKC-alpha, -delta, and -epsilon was detected by immunohistochemical analysis in the pancreatic acini from all the experimental groups. PKC-delta showed the strongest PKC immunoreactivity (PKC-IR). In this vesicular distribution, PKC-IR was located at the apical region of the acinar cells. No differences were observed between control, lactating and weaning rats. However, the immunoblot analysis of pancreatic PKC isoforms during lactation and weaning showed a significant translocation of PKC-delta from the cytosol to the membrane fraction when compared with control animals. Translocation of PKC isoforms (alpha, delta and epsilon) in response to 12-O-tetradecanoyl phorbol 13-acetate (TPA) 1 microM (15 min, 37 degrees C) was comparable in pancreatic acini from control, lactating and weaning rats. In the control group, a significant translocation of all the isoforms (alpha, delta and epsilon) from the cytosol to the membrane was observed. The PKC isoform most translocated by TPA was PKC-delta. In contrast, no statistically significant increase in PKC-delta translocation was detected in pancreatic acini isolated from lactating or weaning rats. These results suggest that the PKC isoforms are already translocated to the surface of the acinar cells from lactating or weaning rats. In addition, they suggest that isoform specific spatial PKC distribution and translocation occur in association with the growth response previously described in the rat exocrine pancreas during lactation and weaning.
Collapse
Affiliation(s)
- E Rodríguez-Martín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
In addition to its fundamental role in stimulating gastric acid secretion, the peptide hormone gastrin induces growth-promoting effects on diversity of target cells. Various mechanisms, including endocrine, paracrine, and autocrine, have been proposed for gastrin's growth-promoting actions. The mitogenic effects of gastrin are mediated by specific cell surface receptors activated after gastrin binding. The functionally defined receptors for gastrin include cholecystokinin A (CCKA) receptor, which is discriminating for sulfated CCK8; cholecystokinin B (CCKB)/gastrin receptor, which binds gastrin17 sulfated, and nonsulfated CCK8 with nearly equal affinities; cholecystokinin C (CCKC), which is a low-affinity gastrin binding protein; and novel, high-affinity receptors selective for amidated gastrin, processing intermediates of gastrin, or both. The signaling pathways mediating gastrin's stimulation of the CCKB/gastrin receptor have been progressively outlined, and the pathways mediating other receptors have been slowly emerging. Engagement of the gastrin receptor initiates various biochemical and molecular events, including recruitment and activation of tyrosine kinases, activation of the phospholipase C signaling pathway leading to phosphoinositide breakdown, intracellular calcium mobilization and protein kinase C stimulation, activation of the mitogen-activated protein kinase pathway, and induction of early response genes. Current emphasis is on understanding the functional significance of processing intermediate forms of gastrin, and the receptor subtypes and pathways that promote the trophic/mitogenic effects of the different molecular forms of gastrin.
Collapse
Affiliation(s)
- R R Yassin
- Department of Medicine, MCP Hahnemann University, Philadelphia, PA 19102-1192, USA
| |
Collapse
|
21
|
Kidd M, Tang LH, Schmid SW, Miu K, Modlin IM. A polyamine pathway-mediated mitogenic mechanism in enterochromaffin-like cells of Mastomys. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:G370-6. [PMID: 9688665 DOI: 10.1152/ajpgi.1998.275.2.g370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We have previously demonstrated that in Mastomys species proliferation of gastric enterochromaffin-like (ECL) cells is predominantly regulated by gastrin and by transforming growth factor-alpha (TGF-alpha) in the naive and neoplastic state, respectively. In this study we examined whether these intracellular mitogenic responses are mediated by polyamines and ornithine decarboxylase (ODC), the rate-limiting enzyme for polyamine biosynthesis. An ECL cell preparation of high purity was used to measure the effect of the polyamine derivatives putrescine, spermidine, and spermine on DNA synthesis by bromodeoxyuridine uptake. Both putrescine and spermidine augmented gastrin-stimulated, but not basal, DNA synthesis in naive cells. This proliferative response correlated with an increase in ODC activity that was partially inhibited (20%) by difluoromethylornithine (DFMO), an inhibitor of ODC (IC50, 30 pM). In contrast, all polyamines increased both basal and TGF-alpha-stimulated DNA synthesis as well as ODC activity in tumor ECL cells. DFMO completely inhibited the proliferative response of TGF-alpha (IC50, 3 pM). Thus polyamine biosynthesis is involved in proliferation of ECL cells and in particular the mitogenesis of tumor cells, suggesting a role for this pathway in the regulation of ECL cell transformation.
Collapse
Affiliation(s)
- M Kidd
- Gastrointestinal Surgical Pathobiology Research Group, Yale University School of Medicine and the West Haven Veterans Affairs Medical Center, New Haven, Connecticut 06520-8062, USA
| | | | | | | | | |
Collapse
|
22
|
Ghrib F, Pyronnet S, Bastié MJ, Fagot-Revurat P, Pradayrol L, Vaysse N. Arachidonic-acid-selective cytosolic phospholipase A2 is involved in gastrin-induced AR4-2J-cell proliferation. Int J Cancer 1998; 75:239-45. [PMID: 9462714 DOI: 10.1002/(sici)1097-0215(19980119)75:2<239::aid-ijc12>3.0.co;2-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastrin/CCK(B) G protein-coupled receptors have been shown to mediate proliferation stimulated by their endogenous ligands. The present study demonstrates the proliferative effect of arachidonic acid on AR4-2J cells. Gastrin induces an [3H]arachidonic-acid release in a dose-dependent manner. The use of a specific inhibitor of cPLA2, AACOCF3 established the involvement of a cPLA2 in the proliferative effect of gastrin. The results also demonstrate that a cytosolic high-molecular-weight PLA2 is activated by gastrin in AR4-2J cells.
Collapse
Affiliation(s)
- F Ghrib
- INSERM U.151, Groupe de Recherche de Biologie et Pathologie Digestive, Institut Louis Bugnard, CHU Rangueil, Toulouse, France.
| | | | | | | | | | | |
Collapse
|
23
|
Kato Y, Yu D, Schwartz MZ. Growth factor enhancement of intestinal function: dramatic response but lack of synergism. J Pediatr Surg 1997; 32:1598-600. [PMID: 9396534 DOI: 10.1016/s0022-3468(97)90461-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND/PURPOSE The authors have shown that gastrin and epidermal growth factor (EGF) exert a trophic effect on intestinal epithelial cells. Because these peptides may have different mechanisms by which they stimulate these cells, this study was designed to determine the effect of gastrin and EGF on the intestinal epithelial cell and to evaluate their potential synergistic effect. METHODS Twenty young adult male Sprague-Dawley rats underwent placement of jugular venous catheters that were connected to subcutaneously placed osmotic minipumps. The rats were divided into four groups based on the content of the osmotic pump: Group 1, saline (control, n = 5); Group 2, EGF at 150 microg/kg/d (n = 5); Group 3, gastrin at 13.5 nmol/kg/d (n = 5); and Group 4, EGF at 150 microg/kg/d plus gastrin at 13.5 nmol/kg/d (n = 5). After a 14-day intravenous infusion, [C14] galactose and [C14] glycine absorption (pmol/cm2 intestine), mucosal DNA content (microg/mg mucosa), and protein content (microg/mg mucosa) were measured in the small intestine of each rat. RESULTS The galactose absorption, glycine absorption, DNA content, and protein content were significantly increased by EGF (69%, 28%, 64%, and 55%, respectively) and gastrin (72%, 60%, 93%, and 48%, respectively) when compared with control. Combining EGF and gastrin also significantly increased these parameters (61%, 44%, 96%, and 70%, respectively) when compared with control. However, these data demonstrate no further enhancement than the effect of each peptide alone. CONCLUSION EGF and gastrin individually may be useful for patients who have inadequate intestinal function, but when combined did not exert a synergistic benefit.
Collapse
Affiliation(s)
- Y Kato
- Dupont Hospital for Children, Wilmington, DE 19899, USA
| | | | | |
Collapse
|
24
|
Höcker M, Henihan RJ, Rosewicz S, Riecken EO, Zhang Z, Koh TJ, Wang TC. Gastrin and phorbol 12-myristate 13-acetate regulate the human histidine decarboxylase promoter through Raf-dependent activation of extracellular signal-regulated kinase-related signaling pathways in gastric cancer cells. J Biol Chem 1997; 272:27015-24. [PMID: 9341140 DOI: 10.1074/jbc.272.43.27015] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gastrin stimulates transcription of the human histidine decarboxylase (HDC) gene through binding to the G-protein-coupled cholecystokinin-B/gastrin receptor. We have explored the possibility that mitogen-activated protein kinase cascades play a role in mediating the effects of gastrin on transcription in a gastric cancer (AGS-B) cell line. Gastrin and phorbol 12-myristate 13-acetate (PMA) treatment of AGS-B cells was found to increase the phosphorylation of tyrosine residues of extracellular signal-regulated kinases (ERKs) 1 and 2 and increase ERK activity as determined by the in vitro phosphorylation of myelin basic protein. Reporter gene assays also demonstrated that gastrin and PMA stimulated Elk-1- and c-Myc-dependent transactivation, consistent with gastrin- and PMA-induced activation of ERKs. Overexpression of wild type ERK-1 and ERK-2 or activation of endogenous ERKs using activated MEK-1 (mitogen-activated protein kinase kinase or ERK kinase) overexpression stimulated HDC promoter activity in a dose-dependent fashion. Interruption of the ERK-related pathway using expression vectors for kinase-deficient ERKs or an ERK-specific phosphatase (PAC-1) blocked gastrin- and PMA-stimulated HDC promoter activity. In contrast, inhibition of the Jun kinase pathway using an interfering dominant negative SEK-1 (stress-activated protein kinase/ERK-1) mutant did not inhibit HDC promoter activity. Furthermore, whereas gastrin stimulated phosphorylation of Shc proteins and association with Grb2, activation of the HDC promoter was not influenced by expression of dominant negative Ras (N15 or N17) proteins. However, gastrin stimulated Raf-1 kinase activity, and activation of the HDC promoter was blocked by coexpression of a dominant negative Raf-1 construct. Overall, these data demonstrate that gastrin regulates HDC transcription in a Rafdependent, Ras-independent fashion predominantly through activation of the ERK-related pathway.
Collapse
Affiliation(s)
- M Höcker
- Gastrointestinal Unit and Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Daulhac L, Kowalski-Chauvel A, Pradayrol L, Vaysse N, Seva C. Ca2+ and protein kinase C-dependent mechanisms involved in gastrin-induced Shc/Grb2 complex formation and P44-mitogen-activated protein kinase activation. Biochem J 1997; 325 ( Pt 2):383-9. [PMID: 9230117 PMCID: PMC1218571 DOI: 10.1042/bj3250383] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The proliferative effects of gastrin on normal and neoplastic gastro-intestinal tissues have been shown to be mediated by the gastrin/CCKB (G/CCKB) G-protein-coupled receptors. We have recently reported that gastrin stimulates the tyrosine phosphorylation of Shc proteins and their subsequent association with the Grb2/Sos complex, leading to mitogen-activated protein kinase (MAPK) activation, a pathway known to play an important role in cell proliferation. We undertook the present study to characterize the signalling pathways used by this receptor to mediate the activation of the Shc/Grb2 complex. Since G/CCKB receptor occupancy leads to the activation of the phospholipase C (PLC)/protein kinase C (PKC) pathway, we examined whether PKC stimulation and Ca2+ mobilization contribute to the phosphorylation of Shc proteins and their association with Grb2 in response to gastrin. Our results indicate that Shc proteins are tyrosine phosphorylated and associate with Grb2 in response to phorbol esters, suggesting that activation of PKC is a potential signalling pathway leading to activation of the Shc/Grb2 complex. Inhibition of PKC by GF109203X completely blocked the effect of PMA on Shc tyrosine phosphorylation and its subsequent association with Grb2, but had a partial inhibitory effect on the response to gastrin. Depletion of the intracellular Ca2+ pools by treatment with thapsigargin blocked the increase in intracellular free calcium concentration induced by gastrin and diminished the ability of the peptide to stimulate Shc phosphorylation and recruitment of Grb2. In addition, removal of extracellular Ca2+ partially inhibited the effect of gastrin on Shc phosphorylation as well as its association with Grb2, indicating that the effects of gastrin are also mediated by Ca2+-dependent mechanisms. Furthermore, we show that blockage of the two major early signals generated by activation of PLC, which induced the activation of the Shc/Grb2 complex, also blocked gastrin-induced MAPK activation.
Collapse
Affiliation(s)
- L Daulhac
- INSERM U.151, Groupe de Recherche de Biologie et Pathologie Digestive, Institut Louis Bugnard, CHU Rangueil, 31054 Toulouse, France
| | | | | | | | | |
Collapse
|
26
|
Abstract
The study of gastrin continues to serve as an excellent model for gastrointestinal regulatory processes. This review highlights some recent advances in the field by outlining gastrin biosynthesis, summarizing current understanding of gastrin receptors, describing the regulation of gastrin release, and discussing the clinical implications of gastrin in the pathogenesis of peptic ulcer disease. Emphasis is on three emerging areas of gastrin research: the novel finding that one of gastrin's posttranslational processing intermediates has biological activity distinct from that of the mature peptide; elucidation of gastrin's signal transduction mechanisms that mediate the trophic effects of the peptide; and the role of gastrin in peptic ulcer disease pathogenesis secondary to Helicobacter pylori infection.
Collapse
Affiliation(s)
- M Sawada
- University of Michigan Medical Center, Ann Arbor 48109-0658, USA
| | | |
Collapse
|
27
|
Kowalski-Chauvel A, Pradayrol L, Vaysse N, Seva C. Gastrin stimulates tyrosine phosphorylation of insulin receptor substrate 1 and its association with Grb2 and the phosphatidylinositol 3-kinase. J Biol Chem 1996; 271:26356-61. [PMID: 8824290 DOI: 10.1074/jbc.271.42.26356] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The growth-promoting effects of gastrin on normal and neoplastic gastrointestinal tissues have been shown to be mediated by the gastrin/CCKB receptor, which belongs to the family of G protein-coupled receptors. However, the downstream signaling pathways activated by gastrin are not well characterized. In the present study, we demonstrate that gastrin stimulates tyrosine phosphorylation of insulin receptor substrate 1 (IRS-1), the major cytoplasmic substrate of the insulin receptor. The gastrin-induced phosphorylation of IRS-1 was rapid and transient, occurring within 30 s of treatment and diminishing thereafter. IRS-1 binds several proteins containing Src homology 2 domains through its multiple tyrosine phosphorylation sites. Following gastrin stimulation, we observed a time- and dose-dependent association of IRS-1 with the p85 regulatory subunit of phosphatidylinositol 3-kinase (PI 3-kinase). In addition, activation of PI 3-kinase was detected in anti-IRS-1 immunoprecipitates from gastrin-treated cells, suggesting that tyrosine phosphorylation of IRS-1, which leads to the rapid recruitment of p85, might be one mechanism used by gastrin to activate PI 3-kinase. We have previously reported that tyrosine phosphorylation of Shc and its association with the Grb2-Sos complex may contribute to the activation of the mitogen-activated protein kinase pathway by gastrin. We report here that Grb2 also interacts with tyrosine-phosphorylated IRS-1 in response to gastrin. Taken together, our results suggest that IRS-1 may serve as a converging target in the signaling pathways stimulated by receptors that belong to different families, such as the gastrin/CCKB G protein-coupled receptor and the insulin receptor.
Collapse
Affiliation(s)
- A Kowalski-Chauvel
- INSERM U.151, Groupe de Recherche de Biologie et Pathologie digestive, Institut Louis Bugnard, CHU Rangueil, 31054 Toulouse, France
| | | | | | | |
Collapse
|
28
|
Smith AJ, Freedman SB. CCK-B receptor-mediated stimulation of polyphosphoinositide turnover in GH3 pituitary cells in response to cholecystokinin and pentagastrin. Life Sci 1996; 58:883-95. [PMID: 8786694 DOI: 10.1016/0024-3205(96)00031-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
CCK-B receptor-mediated polyphosphoinositide (PPI) turnover in GH3 pituitary cells has been examined and comparisons are made with Ca2+ mobilisation and receptor binding data, previously described. Sulphated cholecystokinin octapeptide (CCK-8s) and the CCK-B-selective agonist pentagastrin dose-dependently stimulated PPI turnover in GH3 cells with similar maximal increases of 3.0 fold and 3.3 fold, respectively, in production of [3H]inositol phosphates over control. Responses, measured over 30min periods in the presence of 10mM LiCl, were generally maximal for both agonists at 100nM. Consistent with their [125]Bolton Hunter CCK-8s (BHCCK) binding affinities and with effects on Ca2+ mobilisation, CCK-8s was slightly more potent than pentagastrin in stimulating PPI turnover (EC50s 1.3nM and 3.9nM respectively). Both peptides showed higher potency in the PPI assay than in Ca2+ studies. 100nM pentagastrin-induced PPI turnover was dose-dependently inhibited by the CCK-B receptor-selective antagonist L-365,260 (IC50 470nM) whilst the CCK-A receptor antagonist, devazepide, only produced weak partial inhibition (18% at 10,000nM). Antagonists alone were observed to depress control activity in PPI turnover but not in Ca2+ mobilisation assays. The selectivity of L-365,260 compared to devazepide was similar in binding studies to that for both 100nM pentagastrin-induced functional responses. Schild analysis of antagonism of PPI turnover by L-365,260 yielded a line with slope close to unity (1.07) and a pKB of 8.27+/-0.05.
Collapse
Affiliation(s)
- A J Smith
- Merck, Sharp and Dohme Research Laboratories, Neuroscience Research Centre, Essex, UK
| | | |
Collapse
|
29
|
Seva C, Kowalski-Chauvel A, Blanchet JS, Vaysse N, Pradayrol L. Gastrin induces tyrosine phosphorylation of Shc proteins and their association with the Grb2/Sos complex. FEBS Lett 1996; 378:74-8. [PMID: 8549807 DOI: 10.1016/0014-5793(95)01414-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrin/CCKB G protein-coupled receptors have been shown to mediate proliferative effects of their endogenous ligands. In the present study, we examined the signal transduction mechanisms linked to the G/CCKB receptor occupancy. We report here that gastrin stimulates MAP kinase activation in a dose- and time-dependent manner, a pathway known to play a key role in cell proliferation. We also characterized the molecular events, upstream of p21-Ras, that may link the MAP kinase pathway to G/CCKB receptors. Gastrin induced a rapid and transient increase in tyrosine phosphorylation of several proteins including the 2 isoforms (46 and 52 kDa) of the adaptor protein Shc. Phosphorylated Shc subsequently associated with a complex that includes Grb2 and the p21-Ras activator, Sos. Our results also indicate that Sos becomes phosphorylated in response to gastrin as shown by a reduction in electrophoretic mobility of the protein. Tyrosine phosphorylation of Shc and subsequent complex formation with Grb2 and Sos appear to be a common mechanism by which tyrosine kinase receptors and the G/CCKB G protein-coupled receptor stimulate the Ras-dependent MAP kinase pathway.
Collapse
Affiliation(s)
- C Seva
- INSERM U.151., Institut Louis Bugnard, CHU Rangueil, Toulouse, France
| | | | | | | | | |
Collapse
|
30
|
Wojcikiewicz RJ. Type I, II, and III inositol 1,4,5-trisphosphate receptors are unequally susceptible to down-regulation and are expressed in markedly different proportions in different cell types. J Biol Chem 1995; 270:11678-83. [PMID: 7744807 DOI: 10.1074/jbc.270.19.11678] [Citation(s) in RCA: 342] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The type I inositol 1,4,5-trisphosphate (InsP3) receptor can be rapidly depleted from cells during stimulation of phosphoinositide hydrolysis because its degradation is accelerated (Wojcikiewicz, R. J. H., Furuichi, T., Nakade, S., Mikoshiba, K., and Nahorski, S. R. (1994) J. Biol. Chem. 269, 7963-7969). The present study examines the regulatory properties of type II and III InsP3 receptors. Initially, the relative abundance of InsP3 receptors was defined in a range of cell types by quantitative immunoblotting. These studies showed that the proportions in which type I, II, and III InsP3 receptors are expressed differs greatly and that some cells (for example, AR4-2J rat pancreatoma cells) express all three receptors. Analysis of the effects of cholecystokinin and bombesin on AR4-2J cells showed that each of the InsP3 receptors could be down-regulated during activation of phosphoinositide hydrolysis, but that depletion of the type II receptor was limited. Such a discrepancy was also seen in rat cerebellar granule cells and was found to result from the type II receptor being relatively resistant to degradation. In conclusion, type I, II, and III receptors can all be down-regulated, but with different characteristics. As the relative abundance of InsP3 receptors is extremely variable, the extent to which activation of the down-regulatory process alters intracellular signaling will vary depending on which InsP3 receptors are expressed.
Collapse
Affiliation(s)
- R J Wojcikiewicz
- Department of Pharmacology, College of Medicine, State University of New York Health Science Center, Syracuse 13210-2339, USA
| |
Collapse
|