1
|
Li X, Wang X, Hu X, Tang P, Chen C, He L, Chen M, Bello ST, Chen T, Wang X, Wong YT, Sun W, Chen X, Qu J, He J. Cortical HFS-Induced Neo-Hebbian Local Plasticity Enhances Efferent Output Signal and Strengthens Afferent Input Connectivity. eNeuro 2025; 12:ENEURO.0045-24.2024. [PMID: 39809536 PMCID: PMC11810566 DOI: 10.1523/eneuro.0045-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 12/17/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
High-frequency stimulation (HFS)-induced long-term potentiation (LTP) is generally regarded as a homosynaptic Hebbian-type LTP, where synaptic changes are thought to occur at the synapses that project from the stimulation site and terminate onto the neurons at the recording site. In this study, we first investigated HFS-induced LTP on urethane-anesthetized rats and found that cortical HFS enhances neural responses at the recording site through the strengthening of local connectivity with nearby neurons at the stimulation site rather than through synaptic strengthening at the recording site. This enhanced local connectivity at the stimulation site leads to increased output propagation, resulting in signal potentiation at the recording site. Additionally, we discovered that HFS can also nonspecifically strengthen distant afferent synapses at the HFS site, thereby expanding its impact beyond local neural connections. This form of plasticity exhibits a neo-Hebbian characteristic as it exclusively manifests in the presence of cholecystokinin release, induced by HFS. The cortical HFS-induced local LTP was further supported by a behavioral task, providing additional evidence. Our results unveil a previously overlooked mechanism underlying cortical plasticity: synaptic plasticity is more likely to occur around the soma site of strongly activated cortical neurons rather than solely at their projection terminals.
Collapse
Affiliation(s)
- Xiao Li
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xue Wang
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
| | - Xiaohan Hu
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
| | - Peng Tang
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
- Center of Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Shatin, Hong Kong
| | - Congping Chen
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Ling He
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Center of Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Shatin, Hong Kong
| | - Mengying Chen
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
| | - Stephen Temitayo Bello
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
| | - Tao Chen
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
- Center of Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Shatin, Hong Kong
| | - Xiaoyu Wang
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Yin Ting Wong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Wenjian Sun
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Xi Chen
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
| | - Jianan Qu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Jufang He
- Departments of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
- Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon, Hong Kong
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Center of Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Shatin, Hong Kong
| |
Collapse
|
2
|
Zeng Q, Ou L, Wang W, Guo DY. Gastrin, Cholecystokinin, Signaling, and Biological Activities in Cellular Processes. Front Endocrinol (Lausanne) 2020; 11:112. [PMID: 32210918 PMCID: PMC7067705 DOI: 10.3389/fendo.2020.00112] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022] Open
Abstract
The structurally-related peptides, gastrin and cholecystokinin (CCK), were originally discovered as humoral stimulants of gastric acid secretion and pancreatic enzyme release, respectively. With the aid of methodological advances in biochemistry, immunochemistry, and molecular biology in the past several decades, our concept of gastrin and CCK as simple gastrointestinal hormones has changed considerably. Extensive in vitro and in vivo studies have shown that gastrin and CCK play important roles in several cellular processes including maintenance of gastric mucosa and pancreatic islet integrity, neurogenesis, and neoplastic transformation. Indeed, gastrin and CCK, as well as their receptors, are expressed in a variety of tumor cell lines, animal models, and human samples, and might contribute to certain carcinogenesis. In this review, we will briefly introduce the gastrin and CCK system and highlight the effects of gastrin and CCK in the regulation of cell proliferation and apoptosis in both normal and abnormal conditions. The potential imaging and therapeutic use of these peptides and their derivatives are also summarized.
Collapse
Affiliation(s)
- Qiang Zeng
- Health Management Institute, People's Liberation Army General Hospital, Beijing, China
| | - Lei Ou
- Health Management Institute, People's Liberation Army General Hospital, Beijing, China
| | - Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- *Correspondence: Wei Wang
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- Dong-Yu Guo
| |
Collapse
|
3
|
Gastrin-induced proliferation involves MEK partner 1 (MP1). In Vitro Cell Dev Biol Anim 2013; 49:162-9. [PMID: 23408059 PMCID: PMC3611038 DOI: 10.1007/s11626-013-9588-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 01/30/2013] [Indexed: 12/20/2022]
Abstract
The peptide hormone gastrin is an important factor for the maintenance and homeostasis of the gastric mucosa. We show that gastrin stimulates proliferation in a dose-dependent manner in the human gastric adenocarcinoma cell line AGS-GR. Furthermore, we demonstrate that the MAPK scaffold protein MEK partner 1 (MP1) is important for gastrin-induced phosphorylation of ERK1 and ERK2 and that MP1 promotes gastrin-induced proliferation of AGS-GR cells. Our results suggest a role of MP1 in gastrin-induced cellular responses involved in proliferation and homeostasis of the gastric mucosa.
Collapse
|
4
|
El-Kouhen K, Morisset J. Cholecystokinin and Somatostatin Negatively Affect Growth of the Somatostatin-RIN-14B Cells. Int J Endocrinol 2009; 2009:875167. [PMID: 20011057 PMCID: PMC2778184 DOI: 10.1155/2009/875167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 09/03/2008] [Accepted: 09/29/2008] [Indexed: 12/03/2022] Open
Abstract
With the exclusive presence of the pancreatic CCK-2 receptors on the pancreatic delta cells of six different species, this study was undertaken to determine the role of cholecystokinin and gastrin on growth of these somatostatin (SS) cells. For this study, the SS-RIN-14B cells were used in culture and their growth was evaluated by cell counting. Results. To our surprise, we established by Western blot that these RIN cells possess the two CCK receptor subtypes, CCK-1 and CCK-2. Occupation of the CCK-1 receptors by caerulein, a CCK analog, led to inhibition of cell proliferation, an effect prevented by a specific CCK-1 receptor antagonist. Occupation of the CCK-2 receptors by the gastrin agonist pentagastrin had no effect on cell growth. Proliferation was not affected by SS released from these cells but was inhibited by exogenous SS. Conclusions. Growth of the SS-RIN-14B cells can be negatively affected by occupation of their CCK-1 receptors and by exogenous somatostatin.
Collapse
Affiliation(s)
- Karim El-Kouhen
- Service de Gastroentréologie, Département de Médecine, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | - Jean Morisset
- Service de Gastroentréologie, Département de Médecine, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| |
Collapse
|
5
|
Harper EA, Mitchell EA, Griffin EP, Kalindjian SB. Thermodynamic analysis does not allow discrimination of agonists and antagonists at human CCK2S-receptors. Eur J Pharmacol 2008; 581:1-12. [DOI: 10.1016/j.ejphar.2007.11.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 11/15/2007] [Accepted: 11/21/2007] [Indexed: 11/16/2022]
|
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
|
Ma KT, Si JQ, Zhang ZQ, Zhao L, Fan P, Jin JL, Li XZ, Zhu L. Modulatory effect of CCK-8S on GABA-induced depolarization from rat dorsal root ganglion. Brain Res 2006; 1121:66-75. [PMID: 17055464 DOI: 10.1016/j.brainres.2006.08.094] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2006] [Revised: 07/30/2006] [Accepted: 08/25/2006] [Indexed: 11/25/2022]
Abstract
CCK is a brain-gut peptide that is abundantly distributed in both gastrointestinal tract and mammalian brain. The sulfated octapeptide fragment of cholecystokinin (CCK-8S) has been shown to be involved in numerous physiological functions such as behavior, anxiety, learning/memory processes and neuropathic pain. CCK-8S is one of the strongest endogenous anti-opioid substances and suppresses opioid peptides-mediated 'pre-synaptic inhibition' of gamma-aminobutyric acid (GABA) release. Here we provide evidence that CCK-8S modulates GABA-evoked membrane depolarization in rat dorsal root ganglion (DRG) neurons using intracellular recording technique. Bath application CCK-8S-induced membrane depolarization in most of the rat DRG neurons. The depolarization was blocked by prolumide but not LY225910. Pretreatment with CCK-8S suppressed the GABA-evoked depolarization in a concentration-dependent manner. The CCK-8S inhibition was also time-dependent and reached the peak at about 2 min. The inhibitory effect of CCK-8S was strongly suppressed by pre-incubation of CCK-B receptor antagonist LY225910, phospholipase C inhibitor U73122, protein kinase C inhibitor chelerythrine and calcium chelator BAPTA-AM, respectively. The protein kinase A inhibitor H-89 did not affect CCK-8S effect. The results suggest that CCK-8S inhibits GABA-A receptor function by activation of CCK-B receptor followed by activation of intracellular PLC-Ca(2+)-PKC cascade. Thus, CCK-8S might enhance nociceptive information transmission through inhibition of the "pre-synaptic inhibition" evoked by GABA, which may explain its role in modulation of primary sensory information (especially pain).
Collapse
Affiliation(s)
- Ke-Tao Ma
- The Fundamental Medical School of Wuhan University, Hubei 430071, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Kombian SB, Ananthalakshmi KVV, Parvathy SS, Matowe WC. Cholecystokinin-2 receptors couple to cAMP–protein kinase A to depress excitatory synaptic currents in rat nucleus accumbens in vitro. Can J Physiol Pharmacol 2006; 84:203-11. [PMID: 16900946 DOI: 10.1139/y05-119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently reported that the activation of cholecystokinin-2 receptors depress evoked excitatory postsynaptic currents (EPSCs) in nucleus accumbens (NAc) indirectly through γ-aminobutyric acid (GABA) acting on γ-aminobutyric acid-B (GABAB) receptors. Here, we determined the second messenger system that couples cholecystokinin-2 receptors to the observed synaptic depression. Using in vitro forebrain slices of rats and whole-cell patch recording, we tested the hypothesis that cholecystokinin-2 receptors are coupled to cAMP and protein kinase A signaling pathway. Cholecystokinin-8S induced inward currents and depressed evoked EPSCs. Forskolin, an activator of adenylyl cyclase and rolipram that is an inhibitor of phosphodiesterase type IV, independently increased EPSC amplitude and blocked the inward current and synaptic depression induced by cholecystokinin-8S. Furthermore, the membrane-permeable cAMP analog, 8-bromo-cAMP, blocked the cholecystokinin-8S effects. H89, a protein kinase A inhibitor, also blocked cholecystokinin-8S effects. However, depression of the evoked EPSC by baclofen, a GABABreceptor agonist, was not blocked by H89 or forskolin. These findings indicate that cholecystokinin-2, but not GABAB, receptors are coupled to the adenylyl cyclase – cAMP – protein kinase A signaling pathway in the NAc to induce inward currents and cause synaptic depression.
Collapse
Affiliation(s)
- Samuel B Kombian
- Department of Applied Therapeutics, Kuwait University, Box 24923, Safat 13110, Kuwait.
| | | | | | | |
Collapse
|
9
|
Müerköster S, Isberner A, Arlt A, Witt M, Reimann B, Blaszczuk E, Werbing V, Fölsch UR, Schmitz F, Schäfer H. Gastrin suppresses growth of CCK2 receptor expressing colon cancer cells by inducing apoptosis in vitro and in vivo. Gastroenterology 2005; 129:952-68. [PMID: 16143134 DOI: 10.1053/j.gastro.2005.06.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 05/26/2005] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The role of amidated gastrin17 (G17) and the gastrin/CCKB/CCK2 receptor in colorectal carcinogenesis is still a controversial issue. Here, we investigated the effect of G17 on proliferation and apoptosis of CCK2 receptor-expressing human colon cancer cell lines in vitro and in vivo. METHODS Proliferation was determined by cell counting and cell cycle analysis. Apoptosis was analyzed by annexin V staining, TUNEL staining, caspase-3/7 assay, and JC1 (delta psi) assay. Signal-transduction pathways were analyzed by Western blotting and gel-shift and luciferase assays. An in vivo tumor model with subcutaneously inoculated colon cancer cells in SCID mice was used, and systemic hypergastrinemia was induced by omeprazole. RESULTS In Colo320 cells stably transfected with the wild-type CCK2 receptor (Colo320wt) or in Lovo cells endogenously expressing CCK2 receptors, G17 treatment inhibited proliferation along with a G2/M cell cycle arrest. Furthermore, the administration of G17 significantly augmented apoptosis of CCK2 receptor-expressing cells. In contrast, G17 had no effect on proliferation and apoptosis in Colo320 cells stably transfected with a tumor-derived CCK2 receptor mutant (Colo320mut) or in cells lacking CCK2 receptor expression. Systemic hypergastrinemia in severe combined immunodeficiency (SCID) mice suppressed the growth of Colo320wt tumors accompanied by enhanced apoptosis as compared with untreated tumors. In contrast, omeprazole did not affect Colo320mut tumors reflecting a loss-of-function state of the CCK2(mut) receptor. This is supported by the observation that, in Colo320wt cells, but not in Colo320mut cells, G17 treatment induced the MAPK/ERK/AP-1 pathway and inhibited the activity of NF-kappaB. CONCLUSIONS G17 exerts an antiproliferative and proapoptotic effect on human colon cancer cells expressing the wild-type CCK2 receptor. This supports the view that amidated gastrin prevents rather than promotes colorectal carcinogenesis.
Collapse
Affiliation(s)
- Susanne Müerköster
- Division of Molecular Gastroenterology and Hepatology, First Department of Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cheng ZJ, Harikumar KG, Ding WQ, Holicky EL, Miller LJ. Analysis of the cellular and molecular mechanisms of trophic action of a misspliced form of the type B cholecystokinin receptor present in colon and pancreatic cancer. Cancer Lett 2005; 222:95-105. [PMID: 15837546 DOI: 10.1016/j.canlet.2004.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2004] [Revised: 08/05/2004] [Accepted: 09/02/2004] [Indexed: 11/20/2022]
Abstract
Gastrin and cholecystokinin (CCK) have trophic action on cells expressing wild type A or B CCK receptors. Potential relevance to pancreatic and colonic cancers was raised by the demonstration of a misspliced type B CCK receptor that, when expressed in Balb3T3 cells, had constitutive activity to stimulate intracellular calcium. We attempted to confirm and extend this observation in CHO cells by establishing lines expressing similar densities of variant or wild type B CCK receptor. While both were capable of normal binding and agonist-induced signaling, neither expressed constitutive signaling and both had similar basal growth. Agonist stimulation of cells expressing misspliced receptor had greater increases in calcium and greater growth rates than control cells despite similar MAP kinase phosphorylation responses. Thus, this variant receptor can potentiate peptide-stimulated signaling and trophic action and may contribute to the proliferation of neoplasms expressing it.
Collapse
Affiliation(s)
- Zhi-Jie Cheng
- Mayo Clinic Cancer Center, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic in Scottsdale, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | | | | | | | | |
Collapse
|
11
|
Kombian SB, Ananthalakshmi KVV, Parvathy SS, Matowe WC. Cholecystokinin inhibits evoked inhibitory postsynaptic currents in the rat nucleus accumbens indirectly through gamma-aminobutyric acid and gamma-aminobutyric acid type B receptors. J Neurosci Res 2005; 79:412-20. [PMID: 15605383 DOI: 10.1002/jnr.20349] [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: 11/08/2022]
Abstract
We recently reported that cholecystokinin (CCK) excited nucleus accumbens (NAc) cells and depressed excitatory synaptic transmission indirectly through gamma-aminobutyric acid (GABA), acting on presynaptic GABAB receptors (Kombian et al. [2004] J. Physiol. 555:71-84). The present study tested the hypothesis that CCK modulates inhibitory synaptic transmission in the NAc. Using in vitro forebrain slices containing the NAc and whole-cell patch recording, we examined the effects of CCK on evoked inhibitory postsynaptic currents (IPSCs) recorded at a holding potential of -80 mV throughout CCK-8S caused a reversible inward current accompanied by a concentration-dependent decrease in evoked IPSC amplitude. Maximum IPSC depression was approximately 25% at 10 microM, with an estimated EC50 of 0.1 microM. At 1 microM, CCK-8S induced an inward current of 28.3 +/- 4.8 pA (n=6) accompanied by an IPSC depression of -18.8% +/- 1.6% (n=6). This CCK-induced IPSC depression was blocked by pretreatment with proglumide (100 microM; -3.7% +/- 6.9%; n=4) and by LY225910 (100 nM), a selective CCKB receptor antagonist (4.4% +/- 2.6%; n=4). It was not blocked by SCH23390 (10 microM; -23.5% +/- 1.3%; P < 0.05; n=7) or sulpiride (10 microM; -21.8% +/- 5.1%; P <0.05; n=4), dopamine receptor antagonists. By contrast, it was blocked by CGP55845 (1 microM; -0.4% +/- 3.4%; n=5) a potent GABAB receptor antagonist, and by forskolin (50 microM; 9.9% +/- 5.2%; n=4), an adenylyl cyclase activator, and H-89 (1 microM; 6.9% +/- 3.9%; n=4), a protein kinase A (PKA) inhibitor. These results indicate that CCK acts on CCKB receptors to increase extracellular levels of GABA, which then acts on GABAB receptors to decrease IPSC amplitude.
Collapse
Affiliation(s)
- Samuel B Kombian
- Department of Applied Therapeutics, Faculty of Pharmacy, Kuwait University, Safat, Kuwait.
| | | | | | | |
Collapse
|
12
|
Liu PQ, Morton MF, Reik A, de la Rosa R, Mendel MC, Li XY, Case CC, Pabo CO, Moreno V, Kempf A, Pyati J, Shankley NP. Cell lines for drug discovery: elevating target-protein levels using engineered transcription factors. ACTA ACUST UNITED AC 2004; 9:44-51. [PMID: 15006148 DOI: 10.1177/1087057103260115] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Drug discovery requires high-quality, high-throughput bioassays for lead identification and optimization. These assays are usually based on immortalized cell lines, which express the selected drug target either naturally or as a consequence of transfection with the cDNA encoding the target. Natural untransfected cell lines often fail to achieve the levels of expression required to provide assays of sufficient quality with a high enough signal-to-noise ratio. Unfortunately, the use of cDNA is increasingly restricted, as the sequences for more and more genes become subject to patent restrictions. To overcome these limitations, the authors demonstrate that engineered transcription factors with Cys2-His2 zinc finger DNA-binding domains can be used to effectively activate an endogenous gene of interest without the use of isolated cDNA of the target gene. Using this approach, the authors have generated a cell line that provides a high-quality and pharmacologically validated G-protein-coupled receptor bioassay. In principle, this technology is applicable to any gene of pharmaceutical importance in any cell type.
Collapse
Affiliation(s)
- Pei-Qi Liu
- Sangamo BioSciences, Inc., Point Richmond Technology Center, Richmond, CA 94804, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Noble PJM, Wilde G, White MRH, Pennington SR, Dockray GJ, Varro A. Stimulation of gastrin-CCKB receptor promotes migration of gastric AGS cells via multiple paracrine pathways. Am J Physiol Gastrointest Liver Physiol 2003; 284:G75-84. [PMID: 12488236 DOI: 10.1152/ajpgi.00300.2002] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Responses to G protein-coupled receptor stimulation may be mediated by paracrine factors. We have developed a coculture system to study paracrine regulation of migration of gastric epithelial (AGS) cells after stimulation of gastrin-CCK(B) receptors. In cells expressing this receptor, G-17 stimulated migration by activation of protein kinase C. However, G-17 also stimulated the migration of cells expressing green fluorescent protein, but not the receptor, when they were cocultured with receptor-expressing cells consistent with activation of paracrine signals. The use of various pharmacological inhibitors indicated that gastrin stimulated migration via activation of the EGF receptor (EGR-R), the erbB-2 receptor tyrosine kinase, and the MAP kinase pathway. However, gastrin also released fibroblast growth factor (FGF)-1, and migration was inhibited by the FGF receptor tyrosine kinase inhibitor SU-5402. Flow cytometry indicated that in both cell types, gastrin increased MAP kinase via activation of EGF-R but not FGF-R1 or erbB-2. We conclude that gastrin-CCK(B) receptors stimulate epithelial cell migration partly via paracrine mechanisms; transactivation of EGF-R is only one component of the paracrine pathway.
Collapse
Affiliation(s)
- Peter J M Noble
- Physiological Laboratory, University of Liverpool, United Kingdom
| | | | | | | | | | | |
Collapse
|
14
|
Zhang W, Segura BJ, Mulholland MW. Cholecystokinin-8 induces intracellular calcium signaling in cultured myenteric neurons from neonatal guinea pigs. Peptides 2002; 23:1793-1801. [PMID: 12383867 DOI: 10.1016/s0196-9781(02)00136-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The responsiveness of cultured myenteric neurons to cholecystokinin (CCK-8) was examined using fura-2-based digital microfluorimetric measurement of intracellular calcium ([Ca(2+)](i)). CCK-8 (10(-10)-10(-6)M) evoked concentration-dependent increases in percentage of neurons responding (8-52%) and delta[Ca(2+)](i) (76-169 nM). Gastrin (1 microM) also induced an increase in [Ca(2+)](i) in 29+/-6% of neurons (delta[Ca(2+)](i): 71+/-3 nM). L-364,718, an antagonist for the CCK-A receptor, blocked [Ca(2+)](i) response to CCK-8. Removal of extracellular calcium eliminated CCK-induced [Ca(2+)](i) increments, as did the addition of the calcium channel inhibitors nickel (1mM) and lanthanum (5mM). Nifedipine (1-50 microM) dose-dependently attenuated CCK-caused [Ca(2+)](i) responses. CCK evokes [Ca(2+)](i) signaling in myenteric neurons by the influx of extracellular calcium, likely through L-type calcium channels.
Collapse
Affiliation(s)
- Weizhen Zhang
- Department of Surgery, Michigan Gastrointestinal Peptide Center, University of Michigan, Ann Arbor, MI 48109-0331, USA
| | | | | |
Collapse
|
15
|
Ji B, Bi Y, Simeone D, Mortensen RM, Logsdon CD. Human pancreatic acinar cells lack functional responses to cholecystokinin and gastrin. Gastroenterology 2001; 121:1380-90. [PMID: 11729117 DOI: 10.1053/gast.2001.29557] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- B Ji
- Department of Physiology, University of Michigan, 7710 Medical Sciences Building II, Ann Arbor, MI 48109-0622, USA
| | | | | | | | | |
Collapse
|
16
|
Zhukova E, Sinnett-Smith J, Wong H, Chiu T, Rozengurt E. CCK(B)/gastrin receptor mediates synergistic stimulation of DNA synthesis and cyclin D1, D3, and E expression in Swiss 3T3 cells. J Cell Physiol 2001; 189:291-305. [PMID: 11748587 DOI: 10.1002/jcp.10018] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In order to develop a model system for identifying signaling pathways and cell cycle events involved in gastrin-mediated mitogenesis, we have used high efficiency retroviral-mediated transfection of cholecystokinin (CCK)(B)/gastrin receptor into Swiss 3T3 cells. The retrovirally-transfected CCK(B)/gastrin receptor binds 125I-CCK-8 with high affinity (Kd = 1.1 nM) and is functionally coupled to intracellular signaling pathways including rapid and transient increase in Ca2+ fluxes, protein kinase C-dependent protein kinase D activation, and MEK-dependent ERK1/2 activation. In the presence of insulin, CCK-8 or gastrin induced a 66.5 +/- 8.8-fold (mean +/- SEM, n = 24 in eight independent experiments) increase in cellular DNA synthesis, reaching a level similar to that achieved by stimulation with a saturating concentration of fresh serum, and much greater than the response to each agonist added alone. CCK-8 also induced a striking increase in the expression of cyclins D1, D3, and E and hyperphosphorylation of Rb acting synergistically with insulin. Similar effects were observed when CCK(B)/gastrin receptor was activated in the presence of EGF or bombesin. Our results demonstrate that activation of CCK(B)/gastrin receptor retrovirally-transfected into Swiss 3T3 induces a potent synergistic effect on DNA synthesis, accumulation of cyclins D1, D3, and E and hyperphosphorylation of Rb in combination with insulin, EGF, or bombesin. Thus, the CCK(B)/gastrin receptor transfected into Swiss 3T3 cells provides a novel model system to elucidate mitogenic signal transduction pathways and cell cycle events activated via this receptor.
Collapse
Affiliation(s)
- E Zhukova
- Department of Medicine, School of Medicine and Molecular Biology Institute, University of California, Los Angeles, CA 90095-1786, USA
| | | | | | | | | |
Collapse
|
17
|
Abstract
Gastrin, produced by G cells in the gastric antrum, has been identified as the circulating hormone responsible for stimulation of acid secretion from the parietal cell. Gastrin also acts as a potent cell-growth factor that has been implicated in a variety of normal and abnormal biological processes including maintenance of the gastric mucosa, proliferation of enterochromaffin-like cells, and neoplastic transformation. Here, we review the models used to study the effects of gastrin on cell proliferation in vivo and in vitro with respect to mechanisms by which this hormone might influence normal and cancerous cell growth. Specifically, human and animal models of hypergastrinemia and hypogastrinemia have been described in vivo, and several cells that express cholecystokinin (CCK)B/gastrin receptors have been used for analysis of intracellular signaling pathways initiated by biologically active amidated gastrins. The binding of gastrin or CCK to their common cognate receptor triggers the activation of multiple signal transduction pathways that relay the mitogenic signal to the nucleus and promote cell proliferation. A rapid increase in the synthesis of lipid-derived second messengers with subsequent activation of protein phosphorylation cascades, including mitogen-activated protein kinase, is an important early response to these signaling peptides. Gastrin and CCK also induce rapid Rho-dependent actin remodeling and coordinate tyrosine phosphorylation of cellular proteins including the non-receptor tyrosine kinases p125fak and Src and the adaptor proteins p130cas and paxillin. This article reviews recent advances in defining the role of gastrin and CCK in the control of cell proliferation in normal and cancer cells and in dissecting the signal transduction pathways that mediate the proliferative responses induced by these hormonal GI peptides in a variety of normal and cancer cell model systems.
Collapse
Affiliation(s)
- E Rozengurt
- Department of Medicine, School of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, University of California, Los Angeles, California 90095, USA.
| | | |
Collapse
|
18
|
Abstract
Gastric epithelial organization and function are controlled and maintained by a variety of endocrine and paracrine mediators. Peptides encoded by the gastrin gene are an important part of this system because targeted deletion of the gene, or of the gastrin-CCKB receptor gene, leads to decreased numbers of parietal cells and decreased gastric acid secretion. Recent studies indicate that the gastrin precursor, preprogastrin, gives rise to a variety of products, each with a distinctive spectrum of biological activity. The conversion of progastrin to smaller peptides is regulated by multiple mechanisms including prohormone phosphorylation and secretory vesicle pH. Progastrin itself stimulates colonic epithelial proliferation; biosynthetic intermediates (Gly-gastrins) stimulate colonic epithelial proliferation and gastric epithelial differentiation; and C-terminally amidated gastrins stimulate colonic proliferation, gastric epithelial proliferation and differentiation, and acid secretion. The effects of progastrin-derived peptides on gastric epithelial function are mediated in part by release of paracrine factors that include histamine, epidermal growth factor (EGF)-receptor ligands, and Reg. The importance of the appropriate regulation of this system is shown by the observation that prolonged moderate hypergastrinemia in transgenic mice leads to remodelling of the gastric epithelium, and in the presence of Helicobacter, to gastric cancer.
Collapse
Affiliation(s)
- G J Dockray
- Physiological Laboratory, University of Liverpool, Liverpool, L69 3BX, United Kingdom.
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Akagi K, Nagao T, Urushidani T. Correlation between Ca(2+) oscillation and cell proliferation via CCK(B)/gastrin receptor. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1452:243-53. [PMID: 10590313 DOI: 10.1016/s0167-4889(99)00137-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gastrin stimulates cell proliferation through the CCK(B) receptor coupled to Gq-protein, whereas the m3 muscarinic receptor, which also couples to Gq, has no trophic effects. In order to elucidate the cause of the difference, we stably transfected CHO cells with human CCK(B) and m3 receptors. Stimulation of the CCK(B), but not the m3 receptor increased cell growth. Activation of MAP kinase via the m3 receptor was to the same extent as that via CCK(B), indicating that there is an initial signaling common to both receptors. Stimulation of either receptor induced a transient increase in [Ca(2+)](i) followed by a sustained plateau phase. After 2 h of stimulation, the [Ca(2+)](i) response to the m3 receptor disappeared, whereas that to the CCK(B) receptor remained as a [Ca(2+)](i) oscillation. Removal of extracellular Ca(2+), which abolished [Ca(2+)](i) oscillation, completely inhibited DNA synthesis via CCK(B). When the C-terminal part of the CCK(B) receptor was truncated, the trophic effect as well as the [Ca(2+)](i) response after 2 h of stimulation disappeared, whereas the chimeric CCK(B) receptor with the C-terminal region of the m3 receptor preserved its ability to elicit both DNA synthesis and [Ca(2+)](i) oscillation. These results suggest that desensitization might be a principal determinant of cell proliferation, and the persistence of the [Ca(2+)](i) response as [Ca(2+)](i) oscillation could be essential for this type of signal transduction.
Collapse
Affiliation(s)
- K Akagi
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Hongo, Bunkyo-Ku, Tokyo, Japan
| | | | | |
Collapse
|
21
|
Pommier B, Da Nascimento S, Dumont S, Bellier B, Million E, Garbay C, Roques BP, Noble F. The cholecystokininB receptor is coupled to two effector pathways through pertussis toxin-sensitive and -insensitive G proteins. J Neurochem 1999; 73:281-8. [PMID: 10386981 DOI: 10.1046/j.1471-4159.1999.0730281.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Previous binding studies have suggested the existence of two affinity states for type B cholecystokinin receptors (CCK(B)R), which could correspond to different coupling states of the receptor to G proteins. To test this hypothesis, we have further investigated signal transduction pathways coupled to rat CCK(B)R stably transfected in Chinese hamster ovary cells. We show that CCK(B)R are coupled to two distinct transduction pathways involving two different G proteins, a pertussis toxin-insensitive/phospholipase C pathway leading to the production of inositol phosphate and arachidonic acid, and a pertussis toxin-sensitive/phospholipase A2 pathway leading to the release of arachidonic acid. We further demonstrate that the relative degree of activation of each effector pathway by different specific CCK(B)R agonists is the same, and that a specific CCK(B)R antagonist, RB213, can differentially antagonize the two signal transduction pathways elicited by these agonists. Taken all together, these data could be explained by the recently proposed theory assuming that the receptor can exist in a three-state model in which two active conformations corresponding to the complex formed by the receptor with two different G proteins coexist. According to this model, agonists or antagonists could recognize preferentially either conformation of the activated receptor, leading to variable behavior in a system containing a single receptor type.
Collapse
Affiliation(s)
- B Pommier
- Département de Pharmacochimie Moléculaire et Structurale, INSERM U266, CNRS UMR 8600, UFR des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Stepan VM, Dickinson CJ, del Valle J, Matsushima M, Todisco A. Cell type-specific requirement of the MAPK pathway for the growth factor action of gastrin. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G1363-72. [PMID: 10362639 DOI: 10.1152/ajpgi.1999.276.6.g1363] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Gastrin (G17) has a CCKB receptor-mediated growth-promoting effect on the AR42J rat acinar cell line that is linked to induction of both mitogen-activated protein kinase (MAPK) and c-fos gene expression. We investigated the mechanisms that regulate the growth factor action of G17 on the rat pituitary adenoma cell line GH3. Both AR42J and GH3 cells displayed equal levels of CCKB receptor expression and similar binding kinetics of 125I-labeled G17. G17 stimulation of cell proliferation was identical in both cell lines. G17 stimulation of GH3 cell proliferation was completely blocked by the CCKB receptor antagonist D2 but not by the MEK inhibitor PD-98059 or the protein kinase C inhibitor GF-109203X, which completely inhibited G17 induction of AR42J cell proliferation. G17 induced a c-fos SRE-luciferase reporter gene plasmid more than fourfold in the AR42J cells, whereas it had no effect in the GH3 cells. In contrast to what we observed in the AR42J cells, G17 failed to stimulate MAPK activation and Shc tyrosyl phosphorylation and association with the adapter protein Grb2. Epidermal growth factor induced the MAPK pathway in the GH3 cells, demonstrating the integrity of this signaling system. G17 induced Ca2+ mobilization in both the GH3 and AR42J cells. The calmodulin inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide inhibited AR42J cell proliferation by 20%, whereas it completely blocked G17 induction of GH3 cell growth. The Ca2+ ionophore ionomycin stimulated GH3 cell proliferation to a level similar to that observed in response to G17, but it had no effect on AR42J cell proliferation. Thus there are cell type specific differences in the requirement of the MAPK pathway for the growth factor action of G17. Whereas in the AR42J cells G17 stimulates cell growth through activation of MAPK and c-fos gene expression, in the GH3 cells, G17 fails to activate MAPK, and it induces cell proliferation through Ca2+-dependent signaling pathways. Furthermore, induction of Ca2+ mobilization in the AR42J cells appears not to be sufficient to sustain cell proliferation.
Collapse
Affiliation(s)
- V M Stepan
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
23
|
Logsdon CD. The influence of the cellular context on receptor function: a necessary consideration for physiologic interpretations of receptor expression studies. Life Sci 1999; 64:369-74. [PMID: 10069498 DOI: 10.1016/s0024-3205(98)00576-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
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.
Collapse
Affiliation(s)
- C D Logsdon
- Department of Physiology, University of Michigan, Ann Arbor 48109, USA
| |
Collapse
|
24
|
Yule DI, Baker CW, Williams JA. Calcium signaling in rat pancreatic acinar cells: a role for Galphaq, Galpha11, and Galpha14. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G271-9. [PMID: 9887004 DOI: 10.1152/ajpgi.1999.276.1.g271] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Stimulus-secretion coupling in the pancreatic acinar cell is initiated by the secretagogues CCK and ACh and results in the secretion by exocytosis of the contents of zymogen granules. A key event in this pathway is the G protein-activated production of second messengers and the subsequent elevation of cytosolic-free Ca2+. The aim of this study was therefore to define the heterotrimeric G protein alpha-subunits present and participating in this pathway in rat pancreatic acinar cells. RT-PCR products were amplified from pancreatic acinar cell mRNA with primers specific for Galphaq, Galpha11, and Galpha14 but were not amplified with primers specific for Galpha15. The sequences of these PCR products confirmed them to be portions of the rat homologues of Galphaq, Galpha11, and Galpha14. The pancreatic-derived cell line AR42J similarly expressed Galphaq, Galpha11, and Galpha14; however, the Chinese hamster ovary (CHO) cell line only expressed Galpha11 and Galphaq. These data indicate that caution should be exercised when comparing signal transduction pathways between different cell types. The expression of these proteins in acinar cells was confirmed by immunoblotting samples of acinar membrane protein using specific antisera to the individual G protein alpha-subunits. The role of these proteins in Ca2+ signaling events was investigated by microinjecting a neutralizing antibody directed against a homologous sequence in Galphaq, Galpha11, and Galpha14 into acinar cells and CHO cells. Ca2+ signaling was inhibited in acinar cells and receptor-bearing CHO cells in response to both physiological and supermaximal concentrations of agonists. The inhibition was >75% in both cell types. These data indicate a role for Galphaq and/or Galpha11 in intracellular Ca2+ concentration signaling in CHO cells, and in addition to Galphaq and Galpha11, Galpha14 may also fulfill this role in rat pancreatic acinar cells.
Collapse
Affiliation(s)
- D I Yule
- Department of Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48105, USA
| | | | | |
Collapse
|