"Medicine, Faculty of"@en . "Cellular and Physiological Sciences, Department of"@en . "DSpace"@en . "UBCV"@en . "Yip, Linda"@en . "2010-01-16T18:42:26Z"@en . "2004"@en . "Doctor of Philosophy - PhD"@en . "University of British Columbia"@en . "Adenosine has been shown to inhibit gastric acid secretion in the rat stomach, likely by regulating gastrin and somatostatin release. The current study demonstrates that A\u00E2\u0082\u0081 and A\u00E2\u0082\u0082[sub A] receptors may play significant roles in this action. Specifically, in the isolated vascularly perfused rat stomach, the effect of various selective adenosine agonists and antagonists suggests that the inhibition of immunoreactive gastrin (IRG) and somatostatin-like immunoreactivity (SLI) release is mediated by A\u00E2\u0082\u0081 receptors, while the stimulation of SLI release is mediated by A\u00E2\u0082\u0082[sub A] receptors. To determine the site at which adenosine acts to elicit these actions, immunohistochemistry was performed. Both A\u00E2\u0082\u0081 receptor immunoreactivity (A\u00E2\u0082\u0081R-IR) and A\u00E2\u0082\u0082[sub A] receptor immunoreactivity (A\u00E2\u0082\u0082[sub A]R-IR) were observed on mucosal somatostatin-containing D-cells. A\u00E2\u0082\u0081R-IR was also observed on gastrin-containing G-cells, while neither A\u00E2\u0082\u0081R-IR nor A\u00E2\u0082\u0082[sub A]R-IR was observed on parietal cells. These results suggest that adenosine does not act directly on the parietal cells to inhibit gastric acid secretion. Instead, adenosine may act on Ai receptors of G-cells and D-cells to inhibit IRG and SLI release, respectively, and may act on A\u00E2\u0082\u0082[sub A] receptors of D-cells to stimulate SLI release. The localization of A\u00E2\u0082\u0081R-IR and A\u00E2\u0082\u0082[sub A]R-IR in the gastric plexi suggests that adenosine may also act indirectly on the gastric plexi by altering enteric neural transmission to regulate IRG and SLI release. The coding regions of the gastric A\u00E2\u0082\u0081 and A\u00E2\u0082\u0082[sub A] receptors were also examined and found to be structurally identical to those in the rat brain, but expressed in extremely low levels. Gene expression was measured using Real-Time reverse-transcriptase polymerase chain reaction (RT-PCR) assays developed in the current study. Using these assays, A\u00E2\u0082\u0081 and A\u00E2\u0082\u0082[sub A] receptor gene expression was found to be altered by fasting and omeprazole treatment. These treatments produced reciprocal changes in A\u00E2\u0082\u0081 receptor and gastrin gene expression, and similar changes in A\u00E2\u0082\u0082[sub A] receptor and somatostatin gene expression. Further studies demonstrate that omeprazole treatment also altered adenosine agonist-induced changes in SLI release, suggesting that changes in adenosine receptor gene expression may result in actual changes in receptor expression. These results suggest that changes in the gastric state may alter A\u00E2\u0082\u0081 and A\u00E2\u0082\u0082[sub A] receptor expression, which in turn regulates the synthesis and release of gastrin and somatostatin, and consequently gastric acid secretion."@en . "https://circle.library.ubc.ca/rest/handle/2429/18326?expand=metadata"@en . "ADENOSINE A , A N D A 2 A RECEPTORS IN THE R A T STOMACH: BIOLOGICAL ACTIONS, C E L L U L A R LOCALIZATION, STRUCTURE A N D GENE EXPRESSION by LINDA YIP B.Sc , The University of British Columbia, 1997 A THESIS SUBMITTED IN PARTIAL F U L F I L M E N T OF THE REQUIREMENTS FOR THE DEGREE OF THE F A C U L T Y OF G R A D U A T E STUDIES (Department of Physiology) We accept this thesis as conforming to the required standard THE UNIVERSITY OF BRITISH C O L U M B I A April 2004 DOCTOR OF PHILOSOPHY in \u00C2\u00A9 Linda Yip, 2004 Abstract Adenosine has been shown to inhibit gastric acid secretion in the rat stomach, likely by regulating gastrin and somatostatin release. The current study demonstrates that A i and A2A receptors may play significant roles in this action. Specifically, in the isolated vascularly perfused rat stomach, the effect of various selective adenosine agonists and antagonists suggests that the inhibition of immunoreactive gastrin (IRG) and somatostatin-like immunoreactivity (SLI) release is mediated by A i receptors, while the stimulation of SLI release is mediated by A2A receptors. To determine the site at which adenosine acts to elicit these actions, immunohistochemistry was performed. Both A i receptor immunoreactivity (AiR-IR) and A2A receptor immunoreactivity (A 2AR-IR) were observed on mucosal somatostatin-containing D-cells. A iR - IR was also observed on gastrin-containing G-cells, while neither A i R - I R nor A 2 A R - I R was observed on parietal cells. These results suggest that adenosine does not act directly on the parietal cells to inhibit gastric acid secretion. Instead, adenosine may act on A i receptors of G-cells and D-cells to inhibit IRG and SLI release, respectively, and may act on A 2 A receptors of D-cells to stimulate SLI release. The localization of A iR - IR and A 2 A R - I R in the gastric plexi suggests that adenosine may also act indirectly on the gastric plexi by altering enteric neural transmission to regulate IRG and SLI release. The coding regions of the gastric A i and A 2 A receptors were also examined and found to be structurally identical to those in the rat brain, but expressed in extremely low levels. Gene expression was measured using Real-Time reverse-transcriptase polymerase chain reaction (RT-PCR) assays developed in the current study. Using these assays, A i and A 2 A receptor gene expression was found to be altered by fasting and omeprazole treatment. Ill These treatments produced reciprocal changes in A i receptor and gastrin gene expression, and similar changes in A2A receptor and somatostatin gene expression. Further studies demonstrate that omeprazole treatment also altered adenosine agonist-induced changes in SLI release, suggesting that changes in adenosine receptor gene expression may result in actual changes in receptor expression. These results suggest that changes in the gastric state may alter A i and A 2 A receptor expression, which in turn regulates the synthesis and release of gastrin and somatostatin, and consequently gastric acid secretion. Table of contents Abstract ii Table of contents iv List of tables viii List of figures ix Acknowledgements xiii Chapter 1: Introduction 1 I. Background 2 1. Regulation of endogenous adenosine levels 2 1.1 The de novo synthesis pathway 3 1.2 Intracellular and extracellular salvage 3 1.3 Presynaptically and postsynaptically released ATP as a source of adenosine 8 1.4 Cellular release and reuptake of adenosine by adenosine transporters 9 1.5 Metabolism of adenosine 10 1.6 Endogenous adenosine levels 12 2. The adenosine receptors 15 2.1 Classification of the adenosine receptors 15 2.2 Pharmacological characterization of the adenosine receptors 16 2.3 Molecular characterization of the adenosine receptors 22 2.4 Regulation of adenosine receptor gene and protein expression 25 2.5 Structural characterization of the adenosine receptors 29 2.6 Signal transduction mechanism 31 3. Biological actions of adenosine in the gastrointestinal tract 33 3.1 Gastroprotective actions of adenosine 35 3.2 Effect of adenosine on gastric acid secretion 37 II. Hypothesis 50 III. Rationale 50 IV. Objectives and specific aims 51 Chapter 2: Methods 52 I. Effect of adenosine analogs on gastric SLI and IRG release 52 1. Isolated vascularly perfused rat stomach preparation 52 2. Drug administration and sample collection 53 3. Radioimmunoassay (RIA) for the measurement of SLI release 54 4. Radioimmunoassay for the measurement of IRG release 57 5. Data analysis 60 II. Adenosine receptor mRNA: distribution, structure, and quantification 61 1. Distribution of gastric adenosine receptor mRNA 61 1.1 Primer design and synthesis 61 1.2 Optimization of RT-PCR conditions 63 1.3 Tissue extraction 63 1.4 Total R N A extraction and quantification 64 1.5 Formaldehyde gel electrophoresis 66 1.6 DNase I treatment 66 1.7 First strand cDNA synthesis 67 1.8 PCR 67 1.9 Agarose gel electrophoresis 68 1.10 Restriction enzyme digestion 68 2. Structure of the adenosine receptors 69 2.1 Detection of A i and A 2 A receptor coding regions by RT-PCR 70 2.2 Cloning and D N A sequence analysis of the adenosine receptors 70 3. Quantification of gastric Aj and A 2 A receptor gene expression 73 3.1 Quantification by competitive RT-PCR 73 3.1.1 RNA competitor design and synthesis 73 3.1.2 Competitive RT-PCR assay 77 3.1.3 Competitive RT-PCR data analysis 78 3.2 Quantification by Real-Time RT-PCR 80 3.2.1 Design and synthesis of primers and probes 82 3.2.2 Synthesis of A i and A 2 A receptor R N A standards 82 3.2.3 Two step Real-Time RT-PCR assay 86 3.2.4 Real-Time RT-PCR data collection and analysis 87 3.2.5 Optimization of Real-Time RT-PCR 88 III. Regulation of adenosine receptor gene expression and function in altered physiological states of the stomach 89 1. Quantification of A i and A 2 A receptor, somatostatin and gastrin gene expression 92 1.1 Animal treatment 92 1.2 Tissue extraction 93 1.3 Sample preparation 93 1.4 Real-Time RT-PCR 94 1.5 Data collection and analysis 95 2. Effect of omeprazole on A i and A 2 A receptor-induced changes in SLI and IRG release 97 IV. Cellular localization of the gastric Ai and A 2 A receptors 98 1. Tissue preparation for immunohistochemistry (IHC) 98 2. Adenosine receptor antibodies and control peptides 99 3. Antibodies used for double-staining 101 4. Confocal microscopy 102 5. Quantification of co-localization 103 6. Control studies 103 Chapter 3: Results 105 I. Effect of adenosine on gastric IRG and SLI release 105 1. Effect of adenosine analogs on gastric IRG release 105 2. Effect of adenosine receptor antagonists on gastric IRG release 108 3. Effect of adenosine receptor agonists on gastric SLI release 108 4. Effect of adenosine receptor antagonists on gastric SLI release 115 5. Involvement of endogenous adenosine on SLI release 117 II. Distribution and sequence analysis of adenosine receptor mRNA 121 1. Distribution of adenosine receptor mRNA in various regions of the stomach 121 2. A i and A 2 A receptor coding region and A 2 B and A3 receptor partial cDNA sequence 129 III. Quantification of gastric Adenosine Ai and A 2 A receptor mRNA expression 133 1. Competitive RT-PCR 133 2. Real-Time RT-PCR 135 IV. Effect of fasting on adenosine receptor, somatostatin and gastrin gene expression 144 1. Effect of fasting on body weight and Total R N A concentration 144 2. Effect of fasting on 18S rRNA and G A P D H gene expression 144 3. Effect of fasting on adenosine A i and A 2 A receptor expression 146 4. Effect of fasting on gastrin and somatostatin gene expression 149 V. Omeprazole treatment on adenosine receptor and gastric peptide gene expression 149 1. Effect of omeprazole treatment on 18S rRNA and G A P D H gene expression 152 2. Effect of omeprazole treatment on adenosine A i and A 2 A receptor expression 152 3. Effect of omeprazole treatment on gastrin and somatostatin gene expression 155 VI. Omeprazole on adenosine agonist-induced changes in IRG and SLI release 155 1. Effect of omeprazole on CPA-induced changes in IRG release 157 2. Effect of omeprazole on agonist-induced changes in SLI release 157 VII. Cellular localization of the gastric adenosine A\ and A 2 A receptors 163 1. Cellular localization and distribution of AiR-IR 163 2. Double staining with the A i receptor antibody 167 3. Cellular localization and distribution of A2AR-IR 171 4. Double staining with the A 2 A receptor antibody 178 Chapter 4: Discussion 188 I. Alteration of IRG and SLI release by selective adenosine analogs 189 1. Effect of adenosine receptor-selective analogs on IRG release 189 2. Effect of adenosine receptor-selective analogs on gastric SLI release 191 3. Regulation of gastric SLI and IRG release by adenosine 196 II. Distribution and structure of gastric adenosine receptor mRNA 199 III. Quantification of Aj and A 2 A receptor gene expression 202 1. Competitive RT-PCR and Real-Time RT-PCR assay development 202 2. Changes of gastric A i and A 2 A receptor gene expression and function by altered states of the stomach 208 IV. Cellular localization of the adenosine Ai and A 2 A receptor 220 1. Adenosine A i and A 2 A receptors of the gastric mucosa 222 1.1 Adenosine A i receptors on gastrin-secreting G-cells 224 1.2 Adenosine A i and A 2 A receptors on somatostatin secreting D-cells 225 2. Adenosine A i and A 2 A receptors of the enteric plexi 226 3. Adenosine A i and A 2 A receptors of the gastric vasculature 230 V. Conclusion 232 References 234 Appendix 1: Table of abbreviations 280 List of tables Table 1: Adenosine receptor G protein coupling, signal transduction, and regulation of expression 32 Table 2: Effect of adenosine and adenosine agonists on gastric acid secretion in the rat 41 Table 3: Adenosine receptor RT-PCR primers and PCR conditions 62 Table 4: A i and A 2 A receptor Real-Time RT-PCR assay: primers and probes 83 Table 5: Optimization of primer and probe concentrations for Real-Time RT-PCR 90 Table 6: Optimization of M g C l 2 concentrations for Real-Time RT-PCR 91 Table 7: Gastrin and somatostatin Real-Time RT-PCR assay: primers and probes 96 Table 8: Effect of fasting and omeprazole treatment on body weight, tissue weight and total R N A concentration 145 List of figures Fig. 1. The de novo synthesis and metabolism of adenosine 4 Fig. 2. The molecular structure of adenosine 5 Fig. 3. The intracellular and extracellular synthesis and metabolism of adenosine 6 Fig. 4. Adenosine agonists and antagonists 18 Fig. 5. Schematic representation of the human adenosine A i , A 2 A , and A3 receptors 30 Fig. 6. Typical elution profile for the separation of labeled gastrin 58 Fig. 7. Design and synthesis of the adenosine A i and A 2 A receptor R N A competitors 75 Fig. 8. Analysis of competitive RT-PCR data 79 Fig. 9. Cleavage of the fluorogenic probe during Real-Time RT-PCR 81 Fig. 10. Orientation of A i and A 2 A receptor transcript insertion into pGEM-T vector 85 Fig. 11. Effect of CPA on gastric IRG release 106 Fig. 12. Effect of various concentrations of CPA, and 0.1 uM of CGS 21680 and IB-MECA on IRG release 107 Fig. 13. Effect of DPCPX and D M P X on gastric IRG release 109 Fig. 14. Effect of DPCPX and D M P X on adenosine-induced changes in IRG release 110 Fig. 15. Effect of 0.1 uM and 1 uM CPA on gastric SLI release I l l Fig. 16. Effect of 0.1 uM CGS 21680 and IB-MECA on gastric SLI release 112 Fig. 17. Effect of various concentrations of adenosine agonists on gastric SLI release 114 Fig. 18. Effect of D M P X on basal, adenosine, and adenosine agonist-stimulated SLI release 116 Fig. 19. Effect of Z M 241385 on basal and stimulated SLI release 118 Fig. 20. Effect of Z M 241385 and DPCPX on basal and adenosine and agonist-stimulated SLI release 119 Fig. 21. Effect of E H N A on basal and adenosine-induced SLI release 120 Fig. 22. Results of A i , A 2 A , A 2 B and A3 RT-PCR performed using rat gastric tissues 122 Fig. 23. Amplification of the A i and A 2 A receptor coding region by RT-PCR 123 Fig. 24. Restriction enzyme digestion of the adenosine A] receptor PCR amplicon 124 Fig. 25. Restriction enzyme digestion of the adenosine A 2 A receptor PCR amplicon 125 Fig. 26. Restriction enzyme digestion of the adenosine A 2 B receptor PCR amplicon 126 Fig. 27. Restriction enzyme digestion of the adenosine A3 receptor PCR amplicon 127 Fig. 28. Restriction enzyme digestion of the full length adenosine A 2 A receptor amplicon 128 Fig. 29. Alignment of the rat mucosal and brain A i receptor coding regions 130 Fig. 30. Alignment of the rat mucosal and brain A 2 A receptor coding regions 131 Fig. 31. Alignment of the rat mucosal and brain A 2 B and A3 cDNA sequences 132 Fig. 32. Striatal A i and A 2 A receptor gene expression measured by competitive RT-PCR. 134 Fig. 33. Effect of template concentrations on quantification of A i and A 2 A receptor mRNA by competitive RT-PCR 136 Fig. 34. Quantification of gastric A i receptor gene expression by competitive RT-PCR 137 Fig. 35. Quantification of gastric A 2 A receptor gene expression by competitive RT-PCR 138 Fig. 36. Competitive RT-PCR quantification of A i and A 2 A receptor gene expression in the rat stomach 139 Fig. 37. Results of a representative A i Real-Time RT-PCR assay 140 Fig. 38. Results of a representative A 2 A Real-Time RT-PCR assay 142 Fig. 39. Real-Time RT-PCR quantification of gastric and striatal adenosine A | and A 2 A receptor gene expression 143 Fig. 40. Effect of fasting on gastric housekeeping gene expression 147 Fig. 41. Effect of fasting on adenosine receptor gene expression 148 Fig. 42. Effect of fasting on gastric peptide gene expression 150 Fig. 43. Comparison between fasting-induced changes in somatostatin and adenosine A 2 A receptor gene expression 151 Fig. 44. Effect of omeprazole treatment on gastric housekeeping gene expression 153 Fig. 45. Effect of omeprazole treatment on adenosine A i and A 2 A receptor gene expression 154 Fig. 46. Effect of omeprazole treatment on gastric peptide gene expression 156 Fig. 47. Effect of CPA on gastric IRG release from perfused stomachs of 3 day omeprazole-treated and control animals 158 Fig. 48. Effect of omeprazole treatment on CPA-induced changes in IRG release 159 Fig. 49. Effect of CGS 21680 on gastric SLI release in 3 day omeprazole-treated and control animals 160 Fig. 50. Effect of omeprazole treatment on CGS 21680-induced changes in SLI release 161 Fig. 51. Effect of omeprazole treatment on CPA-induced changes in SLI release 162 Fig. 52. Representative AiR-IR and A 2 A R - I R in various regions of the rat stomach 164 Fig. 53. Confocal images of AjR-IR in the corpus of the rat stomach 165 Fig. 54. Confocal images of AiR-IR in the antrum of the rat stomach 166 Fig. 55. Double staining of AiR-IR with gastrin-IR 168 Fig. 56. Double staining of AjR-IR with somatostatin-IR in the gastric mucosa 169 Fig. 57. Double staining of AiR-IR and somatostatin-IR in the gastric muscle 170 Fig. 58. Double-staining of AiR-IR with H + K + -ATPase P-IR 172 Fig. 59. Double staining of AiR-IR with VWF-IR 173 Fig. 60. Localization of AiR-IR relative to VWF-IR in the corporeal muscle 174 Fig. 61. Double staining of AiR-IR with PGP 9.5-IR 175 Fig. 62. Confocal images of A 2 A R - I R in the corpus of the rat stomach 176 Fig. 63. Confocal images of A 2 A R - I R in the antrum of the rat stomach 177 Fig. 64. Double staining of A 2 A R - I R with somatostatin-IR in the gastric mucosa 180 Fig. 65. Double staining of A 2 A R - I R with somatostatin-IR in the gastric muscle of the corpus 181 Fig. 66. Double staining of A 2 A R - I R with somatostatin-IR in the gastric muscle of the antrum 182 Fig. 67. Double staining of A 2 A R - I R with gastrin-IR 183 Fig. 68. Double-staining of A 2 A R - I R with H + K + -ATPase p-IR 184 Fig. 69. Double staining of A 2 A R - I R with VWF-IR 185 Fig. 70. Localization of A 2 A R - I R relative to VWF - IR in the corporeal muscle 186 Fig. 71. Double staining of A 2 A R - I R with PGP 9.5-IR 187 Acknowledgements I am most grateful to my supervisor, Dr. Kenny Y . N . Kwok for his many years of guidance and support, and for his extraordinary kindness and generosity. The academic skills I've acquired from him will certainly serve me well throughout my career. M y sincerest gratitude to Dr. Ross MacGillivray for his support and encouragement throughout my Ph.D. Many thanks also to Jeff Hewitt for teaching me the techniques of molecular biology that have figured so prominently in my work, and for useful technical discussion. I would also like to acknowledge Dr. Alison Buchan for her patience and guidance during our collaboration, through which I acquired my knowledge of immunohistochemistry. Thanks also to Dr. Ray Pederson for carefully reading over this thesis and making graduate studies in this department a more social experience. I am also grateful to other members of my supervisory committee, Dr. Steve Vincent and Dr. Chris Mcintosh, for direction in preparation for my comprehensive exam. In addition, I would like to express my appreciation for the students who have worked in my laboratory, particularly Henry Leung, who contributed to the immunohistochemistry and perfusion studies. Thanks also to Zaira Khan for her administrative support, and finally to Stephan Gudmundson, my family, and friends, who supported me throughout this experience. Financial support from The University of British Columbia, in the form of several University Graduate Fellowship awards, from the Cordula and Gunter Paetzold Fellowship, and from the Wah-Sheung Prize was greatly appreciated. Chapter 1: Introduction Adenosine is found ubiquitously throughout the body, and has been shown to act as a neurotransmitter, neuromodulator, and local mediator in various biological systems (Ralevic and Burnstock 1998). Adenosine exerts its effect by activating purinergic receptors belonging to the PI receptor family. These receptors are classified into the adenosine A i , A 2 A , A2B, and A3 receptor subtypes (Fredholm et al., 2001). In the stomach, adenosine has been shown to protect against stress-, indomethacin-, and ethanol-induced ulcerative formations (Glavin et al., 1987; Cho and Ogle, 1990; Cho et al., 1991, 1995; Bozkurt et al., 1998). This gastroprotective action is likely due to the inhibitory effect of adenosine on gastric acid secretion. Although adenosine has been shown to exert its acid-inhibitory effect in dogs and guinea pigs by acting directly on parietal cells (Gerber et al., 1985; Heldsinger et al., 1986), this does not occur in the rat (Puurunen et al., 1987). Instead, adenosine inhibits the release of gastrin, a major secretagogue of gastric acid, and stimulates the release of somatostatin, a potent inhibitor of acid secretion (Kwok et al., 1990). Adenosine elicits these actions by activating adenosine receptors, but the receptor subtypes involved have not been determined. This thesis examines the adenosine receptor subtypes involved in regulating gastrin and somatostatin secretion in the rat stomach, and characterizes the cellular localization, abundance, and structure of these receptors. Various physiological states of the stomach have been shown to alter gastric acid secretion by regulating the release and synthesis of gastrin and somatostatin. Another objective of this thesis is to examine whether adenosine may be involved in this regulatory process. Thus, changes in adenosine receptor gene expression were examined under altered physiological states of the stomach that are known to alter gastrin and somatostatin synthesis and release. This chapter provides background information pertinent to the current study, and is organized into three sections. The first section discusses the regulation of endogenous adenosine levels by processes controlling the synthesis, release, reuptake and metabolism of adenosine. The second section describes the classification and pharmacological and molecular characterization of each adenosine receptor subtype. This section also reviews the regulation of adenosine receptor gene and protein expression. Since this thesis focuses on the Aj and A 2 A receptors, emphasis is placed on background literature related to these two subtypes. The third section discusses the gastric actions of adenosine, with particular attention given to the role of adenosine in regulating gastric secretion. This section also describes key experiments that this study is based upon. This chapter concludes by presenting the experimental hypothesis, rationale and specific aims of the thesis research. I. Background 1. Regulation of endogenous adenosine levels Since adenosine is the endogenous ligand for all four adenosine receptor subtypes, it is essential to understand how the level of this nucleoside is regulated. Several processes are involved in the production, release, reuptake and metabolism of adenosine. In the stomach, adenosine is produced by intracellular and extracellular salvage pathways that produce adenosine from sources already containing an intact purine ring. De novo synthesis occurs mainly in cells of the liver, and is responsible for the complete formation of adenosine from sources that do not contain a purine ring. Diet is not a source of significant amounts of adenosine since most purines absorbed by the intestines are catabolized by xanthine oxidase in the esophageal, gastric, small intestinal and large intestinal epithelium (Moriwaki et al., 1996). When adenosine is present in the extracellular fluid, it is rapidly removed by cellular reuptake through specialized nucleoside transporters (discussed in section 1.4), degraded to inosine by adenosine deaminase (ADA) or converted to A M P by adenosine kinase (AK). The next section reviews the regulation of endogenous adenosine levels by the synthesis, release, reuptake and metabolism of adenosine. 1.1 The de novo synthesis pathway Although de novo synthesis of adenosine has not been shown to occur in the stomach, this pathway is included in this overview to illustrate the biosynfhetic origin of the purine ring. A simplified pathway for the de novo synthesis and metabolism of adenosine is described in Fig. 1. Components of the purine ring are donated by glutamine, glycine, formyl-tetrahydrofolic acid, and aspartate during this process (see Fig. 2). De novo synthesis occurs mainly in the liver. In other tissues, including the stomach, adenosine is produced through intracellular and extracellular salvage pathways. 1.2 Intracellular and extracellular salvage Under normoxic conditions, intracellular adenosine is formed mainly by the conversion of S-adenosyl homocysteine (SAH) to adenosine (Lloyd et al., 1988) (See Fig. 3). S A H is formed when S-adenosyl methionine (SAM) donates a methyl group to methylate DNA, mRNA, RNA, and rRNA. S A H is hydrolyzed by the enzyme S-adenosyl 10-step de novo purine ring formation Glutamine, Glycine, \u00E2\u0080\u00A24 i Aspartate t RibOSe 5-PhOSphate + A T P \u00E2\u0080\u00A2 P R P P formyl-tetrahydrofolic acid ||\/|p 1 2 Aspartate 5 4 i Inosine -4 Adenosine A A M P -4\u00E2\u0080\u0094- A M P S i hypoxanthine \u00E2\u0080\u0094 - \u00E2\u0080\u0094 \u00E2\u0080\u00A2 xanthine \u00E2\u0080\u0094 - \u00E2\u0080\u0094 \u00E2\u0080\u00A2 H 2 0 2 + U r a t e Fig. 1. The de novo synthesis and metabolism of adenosine. The de novo synthesis of adenosine occurs intracellularly through the actions of the following enzymes: 1) PRPP synthase; 2) adenylosuccinate synthetase ; 3) adenylosuccinate lyase; and 4) endo-5' nucleotidase. Intracellular metabolism of adenosine utilize the following enzymes: 5) adenosine deaminase; 6) phosphorlase; and 7) xanthine oxidase. In the initial step, two phosphates from ATP are transferred to ribose 5-phosphate to form PRPP. The purine ring is formed by a 10-step reaction which converts PRPP to IMP. IMP is converted to AMPS by the addition of aspartate. AMP is hydrolyzed by 5'endonucleotidase to form adenosine (Stone and Simmonds, 1991). [Abbreviations: 5'phospho-a-D-riobsyl 1 pyrophosphate (PRPP), inosine monophosphate (IMP), adenylosuccinate (AMPS), and adenosine monophosphate (AMP)]. Adenosine Source Glutamine [ Glycine 1 Formyl-tetrahydrofolic acid J Aspartate j Ribose 5-phosphate Fig. 2. The molecular structure of adenosine. The ribose moiety is donated by ribose 5-phosphate. The purine ring is formed by de novo synthesis and consists of components donated by glutamine, glycine, formyl-tetrahydrofolic acid and aspartate. extracellular A T P E-NTPase E-NPP AP CAMP A D P E-NTPase E-NPP AP A M P Ecto-5' NT Inosine I ADA - \u00E2\u0080\u00A2 Adenosine Adenosine Transporter T ATP ATPase ^ . M l K \u00E2\u0080\u00A2 A D P ATPas V A M P ^ Encfo 5-WT 4K intracellular S A M -> S A H > Adenosine SAHH Inosine Fig. 3. The intracellular and extracellular synthesis and metabolism of adenosine. Adenosine is produced intracellularly and extracellularly through salvage pathways using sources which already contain an intact purine ring. Enzymes responsible for the extracellular synthesis (blue), intracellular synthesis (red), and metabolism (green) of adenosine are shown. Cellular release and reuptake of adenosine occur through the adenosine transporters. [Abbreviations: ecto-nucleoside triphosphate diphoshohydrolase (E-NTPase), ecto-nucleotide pyrophosphatase/phosphodiesterase (E-NPP), alkaline phosphatase (AP), ecto-5' nucleotidase (ecto-5' NT), adenosine deaminase (ADA), adenosine kinase (AK), cyclic AMP (cAMP), nicotinamide adenine dinucleotide (NAD), S-adenosyl methionine (SAM), S-adenosyl homocysteine (SAH), S-adenosyl homocysteine hydrolase (SAHH)] homocysteine hydrolase (SAHH) to form adenosine (Lloyd et al., 1988). Under hypoxic or ischemic conditions, the contribution of S A H to adenosine levels is negligible (Deussen et al., 1989). Instead, intracellular adenosine is formed primarily by the dephosphorylation of adenosine triphosphate (ATP) (Geiger et al., 1997). ATP is converted to adenosine diphosphate (ADP) and then adenosine monophosphate (AMP) by ATPase, and A M P is then dephosphorylated by either cytosolic AMP-specific or IMP-specific 5' nucleotidase to produce adenosine (Sala-Newby et al., 1999). In the stomach, extracellular adenosine may be synthesized though the hydrolysis of A M P by 5' nucleotidase. The membrane-bound form of 5' nucleotidase has been localized in the corpus and antrum of the stomach (Sakai et al., 1981; Grover et al., 1983, 1985; Borkje et al., 1986). The stomach is divided into three morphological and functionally distinct regions, the fundus, corpus and antrum, as shown in Fig. 36. The activity of 5' nucleotidase was found on plasma membrane enriched fractions extracted from canine antral and corporeal smooth muscle cells (Sakai et al., 1981; Grover et al., 1983), and rat corporeal smooth muscle cells (Grover et al., 1985). The activity of 5' nucleotidase has also been measured in biopsy samples extracted from human antral and corporeal gastric mucosa (Borkje et al., 1986). Thus, the activity of this enzyme may regulate gastric adenosine levels. The state of the stomach has been shown to alter 5' nucleotidase activity. The activity of this enzyme is reduced in the gastric juice of patients suffering from gastric cancer, gastric ulcers, and gastritis (Durak et al., 1994). This may be due to changes in gastric acidity since 5' nucleotidase activity has been shown to be altered by changes in acidity in the rat skeletal muscle. In particular, increased acidity was shown to increase extracellular adenosine levels by stimulation of 5' nucleotidase activity (Cheng et al., 2000). Extracellular adenosine is synthesized from A M P as shown in Fig. 3. A M P levels are regulated by a number of enzymes which hydrolyze ATP, ADP, nicotinamide adenine dinucleotide (NAD + ) and cyclic A M P (cAMP). These enzymes are classified into the ecto-nucleoside triphosphate diphoshohydrolase (E-NTPase) family, the ecto-nucleotide pyrophosphatase/phosphodiesterase (E-NPP) family, and the alkaline phosphatase family. The characteristics of these enzymes have been reviewed by Zimmerman (Zimmermann, 2000). The enzymes are not as important as 5' nucleotidase in regulating extracellular adenosine levels since the dephosphorylation of A M P to adenosine is the rate-limiting step in the production of extracellular adenosine (Dunwiddie et al., 1997). Therefore, 5' nucleotidase activity may be an important regulator of extracellular adenosine production in the stomach. 1.3 Presynaptically and postsynaptically released ATP as a source of adenosine Extracellular adenosine can also be derived from the degradation of ATP released from purinergic nerve terminals (Burnstock et al., 1970; Burnstock, 1975). Purinergic nerves were proposed by Burnstock based on studies performed in the guinea pig and toad stomach (Burnstock et al., 1970). In the stomach, the cell bodies of purinergic nerves are found in the myenteric plexus, and the axons of these nerves project to the circular and longitudinal muscles (Burnstock, 1975). ATP is released from these nerves and acts as a neurotransmitter (Su et al., 1971; Burnstock, 1975). ATP is stored in synaptic vesicles with other neurotransmitters, and is released by nerve depolarization (Su et al., 1971). In the brain, ATP is released presynaptically from a number of neurons, including cholinergic, glutamatergic and catecholaminergic neurons, and from glial cells (Ralevic and Burnstock, 1998). In the gut, ATP was also suggested to be co-released with nitric oxide (Selemidis et al., 1997), pituitary adenylate cyclase-activating polypeptide (Furness et al., 1995; Imoto et a l , 1998), and vasoactive intestinal peptide (VIP) (Furness et al., 1995). Viz i and colleagues have also suggested that acetylcholine and norepinephrine may elicit ATP release from postsynaptic sites through a process that they referred to as cascade transmission (Vizi et al., 1992). This type of release has been observed in the gut. Specifically, acetylcholine was shown to induce ATP release from smooth muscle cells of the guinea pig ileum via activation of postsynaptic M3 receptors (Nitahara et al., 1995). Thus, in - the stomach, adenosine may be derived from the hydrolysis of presynaptically or postsynaptically released ATP. Although cAMP may also be released from neurons and can be metabolized to form adenosine (see Fig. 3), cAMP was not shown to significantly alter extracellular adenosine levels unless there is continuous release over an extended period of time (Brundege et al., 1997). 1.4 Cellular release and reuptake of adenosine by adenosine transporters Adenosine transporters are responsible for the cellular release and reuptake of intracellular and extracellular adenosine, respectively (Geiger et al., 1997). Adenosine transporters are classified into two groups, the equilibrative or facilitated-diffusion carriers and the concentrative or sodium-dependent nucleoside transporters (Thorn and Jarvis, 1996). The equilibrative nucleoside transporters are found in most cell types and are divided into two groups, the es (equilibrative, sensitive) or ei (equilibrative, insensitive) carriers, based on whether they are sensitive to blockade by nitrobenzylthioinosine, a synthetic nucleoside analogue (Lee and Jarvis, 1988). Both classes translocate adenosine bidirectionally across the plasma membrane, and are blocked by inhibitors, such as dipyridamole, dilazep and lidoflazine (Thorn and Jarvis, 1996). Our laboratory has suggested that these transporters may be involved in regulating the endogenous level of extracellular adenosine in the rat stomach (Kwok et al., 1990). The administration of dipyridamole was shown to significantly enhance basal and adenosine-induced augmentation of gastric somatostatin-like immunoreactivity (SLI) release in the isolated perfused stomach preparation (Kwok et al., 1990). Other studies have also suggested that the ei transporters function as an adenosine-scavenging system to maintain the extracellular adenosine level at the basolateral membrane of the intestinal epithelia (Tally et al., 1996). Concentrative nucleoside transporters have not been studied extensively in the stomach, but have been detected on epithelial cells of the intestines (Ritzel et al., 2001). Currently, four concentrative nucleoside transporters (N1-N4) have been identified and characterized (Thorn and Jarvis, 1996). A l l four can transport adenosine bidirectionally across the membrane, but are generally responsible for the influx of adenosine. Currently, no selective inhibitors exist for the concentrative nucleoside transporters (Geiger et al., 1997). Like 5' nucleotidase, the expression of equilibrative adenosine transporters may be altered to regulate endogenous adenosine concentrations (Lorbar et al., 1999). Although this has not been examined in the stomach, alterations in adenosine transporter activity have been observed in the hearts of aged rats. A decrease in adenosine transporter expression was observed with increased levels of extracellular adenosine in these animals (Lorbar et al., 1999). 1.5 Metabolism of adenosine Extracellular adenosine can be regulated by the action of A D A , an enzyme that converts adenosine to inosine (See Fig. 3). A D A is expressed in the cytoplasm and on cell membranes of all tissues (Franco et al., 1997), and can be inhibited by erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride (EHNA) (Mendelson et al., 1983). A D A is highly expressed in the thymus and spleen in humans (Moriwaki et al., 1999), and the lung, spleen, and intestines in rats (Centelles et al., 1987). The expression of the A D A gene has also been detected in the epithelia of the mouse esophagus, forestomach and duodenum (Witte et al., 1991). The high expression of A D A throughout the gastrointestinal tract suggests that much of the adenosine that is absorbed through the gut is metabolized. In ADA-deficient mice, a significantly elevated level of adenosine was observed in various regions of the gastrointestinal tract, including the forestomach, hindstomach, esophagus, duodenum, jejunum and ileum (Xu and Kellems, 2000). Thus, A D A plays a significant regulatory role in modulating endogenous adenosine levels in the gastrointestinal tract. In humans, changes in gastric A D A activity have been shown to occur with changes in gastric acidity (Namiot et al., 1990, 1991, 1993). In particular, hypersecretors of acid were shown to exhibit increased A D A activity, while patients suffering from achlorhydria, a condition characterized by a lack of gastric acid secretion, were shown to exhibit lowered A D A activity (Namiot et al., 1990). Thus, A D A may play a role in regulating gastric acid secretion by modulating endogenous adenosine levels. Mucosal A D A activity has also been shown to decrease in the vicinity of a healed ulcer (Namiot et al., 1993) and in the gastric mucosa after treatment with ranitidine, a gastric acid inhibitor used for treatment of ulcers (Namiot et al., 1991). These findings suggest that increased adenosine levels may be involved in ulcer healing. As shown on Fig. 3, the cytosolic enzyme adenosine kinase (AK) is responsible for catalyzing the phosphorylation of adenosine to A M P (Geiger et al., 1997), and is found in various gastrointestinal tissues, including the stomach, duodenum, ileum, and large intestine (Andres and Fox, 1979). This enzyme has been shown to regulate endogenous adenosine levels. For example, inhibition of A K activity in the hippocampus has been shown to increase intracellular adenosine levels (Pak et al., 1994). During oxidative and metabolic stress, decreased A K activity has also been shown to significantly increase adenosine levels in DDT1 MF-2 smooth muscle cells (Sinclair et al., 2000). 1.6 Endogenous adenosine levels As described in the previous sections, the endogenous adenosine level is tightly regulated by processes controlling its synthesis, release, reuptake and metabolism. Although adenosine can be synthesized intracellularly and can be produced extracellularly, under well oxygenated conditions, the majority of adenosine (92%) is produced intracellularly and is released into the extracellular space, as illustrated in the isolated guinea pig heart (Deussen, 2000). In plasma, adenosine has a short half-life of only a few seconds because it is rapidly degraded by A D A or removed by cellular reuptake (Geiger et al., 1997). Under normoxic conditions, basal adenosine levels are consistently found in the nanomolar range (see below). Although the adenosine concentration has not been measured in the stomach, the venous adenosine concentration in the rat jejunum has been shown to be 62 nM (Sawmiller and Chou, 1990). This concentration is consistent with levels measured elsewhere, such as in the rat cerebrospinal fluid (160 nM) (Meno et al., 1991), cerebral cortex (30-50 nM) (Phillis et al., 1987), and striatum (41- 210 nM) (Ballarin et al., 1991; Pazzagli et al., 1995; Melani et al., 1999). In whole blood and plasma of rats, adenosine concentrations were found to be 79 nM (Phillis et al., 1992) and 320 nM (Conlay et al., 1997), respectively. Arterial adenosine concentrations have also been measured in the rat and found to be 59 n M (Sawmiller and Chou, 1990). In human blood, the concentration was shown to be 35 nM (Geiger et al., 1997). During periods of stress, adenosine levels can rise to micromolar concentrations. In rats, ischemia induced by medial cerebral artery occlusion was shown to increase adenosine concentrations in the striatum and hippocampus to 3 \iM (Melani et al., 1999) and 30 uM (Latini et al., 1999), respectively. The increase in adenosine concentration is due to enhanced adenosine production caused by the degradation of ATP. The adenosine production rate in the rat heart has been shown to increase by approximately 70-fold during ischemia, from a normoxic production rate of 6.4-7.7 nmol min\"1 g\"1 (Lorbar et al., 1999) to an ischemic production rate of 410 nmol min\"1 g\"1 (Meghji et al., 1988). The plasma adenosine level has also been shown to increase to micromolar concentration after chronic blockade of adenosine receptors (Conlay et al., 1997). In rats treated with caffeine for two weeks, the plasma adenosine concentration was found to increase from 320 nM to 3.2 uM (Conlay et al., 1997). Thus, endogenous adenosine levels are clearly elevated by oxidative stress and by treatment with adenosine antagonists. The concentration of adenosine exposed to target tissues may be important because adenosine has been shown to elicit concentration-dependent physiological effects. For example, our laboratory has shown that gastric somatostatin release is inhibited by low concentrations of adenosine (0.01 [xM), but is stimulated by higher concentrations (>0.6 uM) (Kwok et al., 1990). The physiological actions of adenosine are determined, in part, by the adenosine receptor subtype stimulated. These subtypes are described in detail in the next section. However, it is worth mentioning here that the activation of each adenosine receptor subtype depends on the local extracellular adenosine concentration. The high affinity adenosine A i and A 2 A receptor subtypes may be stimulated by relatively low concentrations of adenosine, while the low affinity A 2 B and A3 receptors may be activated by extremely high concentrations of adenosine, such as those present during oxidative stress (Ralevic and Burnstock, 1998). This concentration-dependent activation of adenosine receptors is suggested by studies showing that adenosine elicits A p , A 2 A - , A 2 B - , and A3- mediated effects with E C 5 0 values of 73, 150, 5100, and 6500 nM, respectively (Daly and Padgett, 1992; Zhou et al., 1992; Peakman and Hil l , 1994). In certain tissues, adenosine has been shown to elicit distinct effects depending on the concentration in which it is administered, and the adenosine receptor subtype that is activated. For example, in the renal vasculature, the administration of low micromolar concentrations of adenosine was shown to vasoconstrict renal afferent arterioles via activation of A i receptors, while higher concentrations of adenosine (>10 uM) resulted in vasodilation of these vessels via activation of A 2 A receptors (Tang et al., 1999; Tabrizchi and Bedi, 2001). In the lung, low adenosine concentrations mediate anti-inflammatory actions by activating the A 2 A receptors, while high adenosine concentrations mediate pro-inflammatory actions by activation of the A3 receptors (Blackburn, 2003). These observations raise the possibility that the concentration-dependent effect of adenosine on somatostatin release in the isolated rat stomach is mediated by the activation of different adenosine receptor subtypes. 2. The adenosine receptors 2.1 Classification of the adenosine receptors The concept of purinergic receptors was introduced in the late 1970s by Burnstock (Burnstock, 1978). These receptors were classified into the Pi and P 2 purinoceptors, based mainly on their binding preference for adenosine or ATP, respectively. The PI receptors (or adenosine receptors) were initially characterized by the rank order of binding preference (adenosine > A M P > ADP > ATP), by their sensitivity to blockade by methylxanthines, and by their ability to alter cAMP levels through the modulation of adenylate cyclase activity (Burnstock, 1978). The A i and A 2 receptor subtypes were proposed in 1979 by van Calker to describe the two distinct plasma membrane receptors on cultured brain cells that were capable of inhibiting and stimulating adenylate cyclase activity, respectively (van Calker et al., 1979). Two A 2 receptor subtypes were later identified: a high-affinity A 2 receptor (A 2 A ) on rat striatal membranes, and a low affinity A 2 receptor (A 2B) on rat cerebral cortical membranes (Daly, 1983). After binding studies demonstrated that these receptors represented distinct receptors subtypes (Bruns et al., 1986), the A 2 A and A 2 B nomenclature was proposed by Bruns to refer to these high and low affinity A 2 receptors, respectively. While the A i , A 2 A and A 2 B receptors were initially identified and named based on pharmacological studies, the fourth adenosine receptor subtype, the A3 receptor, was cloned and its molecular structure was determined before it was identified as a distinct subtype using pharmacological means (Zhou et al., 1992). Currently, the nomenclature and classification of the adenosine receptors adhere to the principles of receptor nomenclature adopted by the International Union of Pharmacology (IUPHAR). Each distinct receptor subtype is identified based on both molecular and pharmacological evidence. The existence of the four adenosine receptor subtypes is now well established (Ralevic and Burnstock, 1998; Fredholm et al., 2001), and the gene expression of all four subtypes has been demonstrated in various tissues, including the rat stomach (Dixon et al., 1996). Although additional adenosine receptor subtypes have been proposed, the structural and pharmacological evidence required by the IUPHAR to support their existence is lacking (Cornfield et al., 1992). 2.2 Pharmacological characterization of the adenosine receptors The adenosine receptor subtypes can be characterized pharmacologically by the rank order of potency of various adenosine analogs in eliciting certain responses. In the past two decades, numerous adenosine agonists and antagonists have been developed. Nearly all known agonists are based on the structure of adenosine, while antagonists are based on both xanthine and non-xanthine compounds (Fredholm et al., 2001). Due to the large number of ligands available for these receptors, the following section focuses on ligands that are commonly used to characterize the four adenosine receptors subtypes, and on the ligands which are used in the present study. Adenosine agonists are based on the structure of adenosine, and require an intact ribose moiety (Fredholm et al., 2001; Jacobson et al., 2001). Certain structural modifications have been shown to increase the metabolic stability of adenosine, resulting in longer half lives and/or resistance to degradation and cellular reuptake (Pavan 1998). Adenosine can be modified at three sites to increase metabolic stability and confer receptor subtype selectivity (Jacobson, 1996; Klotz, 2000): the 5' position of the ribose ring, the C2 position and the N 6 position of the purine ring (see Fig. 4). Substitutions at the N 6 position generally increase A i and A 3 receptor selectivity, while substitutions at the C2 position generally increase A 2 A receptor selectivity (Bruns et al., 1986; Klotz, 2000). Agonists containing modifications at more than one site are generally more potent (Jacobson, 1996). The binding affinity and the structure of some of the most common adenosine receptor agonists are shown in Fig. 4. Analogs containing bulky substitutions at the N 6 position, such as N 6 -cyclopentyladenosine (CPA), 5'-N6-cyclohexyladenosine (CHA), R(-)-N6-(2-phenylisopropyl)adenosine (R-PIA), and (S)-N6-(2-phenylisopropyl)adenosine (S-PIA), preferentially bind to the A i receptor (Bruns et al., 1986; Klotz et a l , 1998). CPA is one of the most potent and selective A i receptor agonists (see Fig. 4), and has been used extensively to characterize A i receptor-mediated responses in the gut. In the gastric antrum of the guinea pig, CPA was used to demonstrate the involvement of the A i receptor in the inhibition of fast excitatory postsynaptic potentials (Christofi et al., 1992). CPA has also been used to demonstrate the presence of A i receptors in the rat duodenum (Nicholls et al., 1992; Brownhill et al., 1996; Peachey et al., 1996), jejunum (Hancock and Coupar, 1995), and colon (Peachey et al., 1999). A 2 A receptors prefer agonists with bulky substitutions at the C2 position, such as 2-phenylaminoadenosine (CV1808) and the potent A 2 A receptor agonist 2-p-(2-carboxyethyl)phenethylamino-5'N-ethylcarboxamidoadenosine (CGS 21680) (Hutchison et al., 1989; Lupica et al., 1990). CGS 21680 is highly selective for A 2 A receptors, and exhibits a 140-fold selectivity for A 2 A receptors over A i receptors (Hutchison et al., 1989). CGS 21680 has low affinity for the A 2 B and A 3 receptors (Klotz et al., 1998) (see Fig. 4), and was used to characterize the first A 2 A receptors cloned from the rat striatum (Fink et al., 1992). In the rat isolated perfused superior mesenteric arterial bed, CGS 21680 was used to demonstrate A 2 A receptor-mediated vasodilation (Hiley et al., 1995). In our laboratory, CGS NH, u OH OH Adenosine N* position (purine) C2 position (purine) 5' position (ribose) PIA R-PIAfL-PIAI A,: 2 V 860 A \u00E2\u0080\u009E : 3800-33700 A,:16 S-PIA (D-PIA1 A,: 75 V7800 A \u00E2\u0080\u009E : 62,800 A,: 45 00 CPA a . 00 A,: 2.3 OH OH Mm ^21,000-34,400 A,: 43 CGS 21680 A,: 290 M* A , : 38,100 A,: 87 NECA A,: 14 A^: 330-360 A,: 6 NH, 00 NH2 IB-MECA A,: 3.7 V 2,500 A \u00E2\u0080\u009E : 54,000 A,: 1.2 9' NH 0 f*7 I R 3 Xanthine Caffeine A,: 29,100 A ^ ; 48,100 Theophylline i 1 A,: 6.800 V 1 7 0 0 Aj,: 7.900 A , 86,000 8-PT x^3 A,: 86 A\u00C2\u00BB: 848 DPCPX r - 0 r1 A,: 3 M M A 129 51 3960 DMPX x n A,: 12,000 A,,: 9.000 ZM 241385 (non-xanthine) IC A,: 260 A\u00C2\u00BB: 0.8 A _ : 32 A,: 10.000 Fig. 4. Adenosine agonists (A) and antagonists (B). Adenosine agonists are based on the structure of adenosine. These compounds contain an intact ribose moiety, and may be modified at the N 6 and C2 position of the purine ring or at the 5' position of the ribose moiety. The K i values (nM), which indicate the relative binding affinities at different receptor subtypes, are shown (Bruns et al., 1986; Klotz et al., 1998;Ralevic and Burnstock, 1998; Fredholm et al., 2001). Many adenosine antagonists are xanthine-based, containing possible modifications at the 1,3, 7, or 8 position of the xanthine ring. Caffeine and theophylline are naturally occurring xanthines. 8-PT, DPCPX, and DMPX are xanthine-based derivatives, and Z M 241385 is a non-xanthine A 2 A receptor-selective antagonist 21680 was also shown to decrease perfusion pressure in the isolated vascularly perfused rat stomach in a concentration-dependent manner (Yip and Kwok, 2004). Selective agonists have not been developed for the A 2 B receptor, despite intense efforts (Cristalli et al., 1998; de Zwart et al., 1998). Currently, one of the most potent agonists for this receptor is the non-selective 5' modified agonist N-ethylcarboxamidoadenosine (NECA; see Fig. 4) (Ralevic and Burnstock, 1998). N E C A and CGS 21680 bind to A 2 A receptors with similar affinity, but N E C A binds to the A 2 B receptors with higher affinity than CGS 21680 (Brackett and Daly, 1994; Klotz et al., 1998). Thus, in various tissues, including the gut, A 2 A and A 2 B receptor-mediated effects are often distinguished by the relative potency of N E C A and CGS 21680. In the rat duodenum (Brownhill et al., 1996; Nicholls et a l , 1996) and distal colon (Peachey et al., 1999), A 2 B receptor-mediated actions were demonstrated by the potent effect of N E C A and by the lack of an effect by CGS 21680. Actions mediated by A 2 A and A 2 B receptors can also be distinguished by the EC50 values of N E C A and CGS 21680 (Brackett and Daly, 1994). In particular, in PC12 cells that express A 2 A receptors, activation of adenylyl cyclase elicited by CGS 21680 and N E C A were characterized by EC50 values of 0.07 and 0.13 p.M, respectively (Brackett and Daly, 1994). In fibroblast 3T3 cells expressing A 2 B receptors, the activation of adenylyl cyclase activity elicited by N E C A was characterized by an EC50 of 1.9 \xM, and CGS 21680 was unable to elicit a maximal response (Brackett and Daly, 1994). Although CPA, CGS 21680, and N E C A can also bind to the A 3 receptors, N 6 -substituted adenosine-5'-uronamides, such as l-deoxy-l-[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-B-D-ribofuranuronamide (IB-MECA), are more selective for this receptor subtype (Gallo-Rodriguez et al., 1994) (see Fig. 4). In the gut, I B - M E C A has been used to examine the involvement of A3 receptors in the relaxation of the guinea pig taenia caeci (Prentice and Hourani, 1997). This agonist has also been used to demonstrate the involvement of A3 receptors in protecting against colitis in the mouse (Mabley et al., 2003). The adenosine receptor involved in eliciting a specific effect can be determined pharmacologically by comparing the relative potency of various adenosine agonists. Based on the selectivity and binding affinity of various agonists at each adenosine receptor subtype (see Fig. 4), the following rank order of potency can be used to characterize A i , A 2 A , A 2 B and A 3 receptor mediated actions: A i : CPA > C H A > R-PIA > N E C A > IB-MECA > CGS 21680; A 2 A : CGS 21680 = N E C A = CV1808> CPA > IB-MECA; A 2 B : N E C A > CGS 21680 = CV1808 > CPA > IB-MECA; A 3 : IB-MECA > N E C A > CPA > CGS 21680. Since adenosine agonists are not specific, and are only selective for a given adenosine receptor subtype, the use of selective antagonists is also necessary to characterize the adenosine receptor subtype involved in eliciting various actions. Adenosine receptors, with the exception of the A 3 receptor in some species, are blocked by methylxanthines such as caffeine and theophylline (Zhou et al., 1992; Klotz, 2000). A number of selective xanthine-based antagonists have been developed by modification of the 1,3, 7, and/or 8 positions of the xanthine ring. Unlike human A 3 receptors, rodent A 3 receptors are not blocked by xanthine derivatives (Zhou et al., 1992; Salvatore et al., 1993; van Galen et al., 1994). Several classes of non-xanthine derivatives have also been developed as A i , A 2 A , A 2 B and A3 receptor-selective antagonists. The characteristics of each class have been reviewed by Baraldi (Baraldi et al., 2000). The structure and binding affinities of several common xanthine and non-xanthine antagonists are shown in Fig. 4. One of the most potent and widely used A i receptor antagonists, 8-cyclopentyl-l,3-dipropylxanthine (DPCPX) (Lohse et a l , 1987; Klotz, 2000; Muller, 2001), is produced by the substitution of a cycloalkyl group at position 8 of the xanthine ring (See Fig. 4). This antagonist has previously been used to demonstrate the involvement of A i receptors in various actions in the gastrointestinal tract. For example, DPCPX was shown to abolish adenosine-induced inhibition of gastrin release from canine antral G-cells (Schepp et al., 1990), inhibit CPA-induced contraction in the rat colonic muscularis mucosae (Bailey et al., 1992; Reeves et al., 1993), and inhibit CPA-induced relaxation in the rat duodenal longitudinal muscle (Nicholls et al., 1996). The first A 2 receptor antagonist, 3,7-dimethyl-l-propargylxanthine (DMPX), was generated by substitutions at the 1, 3, and 7 positions of the xanthine ring (Seale et al., 1988). Although this compound does not exhibit extremely high selectivity or affinity for the A 2 receptors, it remains one of the most widely used A 2 receptor antagonists due to its high solubility in water (Muller et al., 1997). More recently, a potent and highly selective non-xanthine A 2 A receptor antagonist, 4-(2-[7-Amino-2-(2-furyl)[l,2,4]triazolo[2,3-a][l,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385), was developed (Palmer et al., 1995b; Poucher et al., 1995, 1996). This antagonist, shown in Fig. 4, binds with high affinity to membranes expressing the A 2 A receptor (Palmer et al., 1995b; Poucher et al., 1995), but binds with low affinity to membranes expressing the A i and A3 receptors (Poucher et al., 1995). Z M 241385 was also shown not to antagonize A 2 B receptor-mediated actions in the guinea pig aorta (Poucher et al., 1995). While selective A 2 B receptor agonists are not available, several selective A 2 B antagonists have been developed. These include 1,3-dialkylxanthine derivatives, and 8-phenylxanthine carboxylic congener derivatives (Kim et al., 2000). Some A 2 B receptor antagonists demonstrate species-dependent differences in ligand binding affinity (Fozard et al., 2003). However, these differences are not as apparent as in the A 3 receptor. In humans, A3 receptors are blocked by high concentrations of xanthine derivatives such as DPCPX (Salvatore et al., 1993). In rats, these receptors were not blocked by xanthine derivatives, including DPCPX (Zhou et al., 1992), but instead were blocked by a number of non-xanthine derivatives such as 9-chloro-2(2-furyl)[l,2, 4]triazolo[l,5-c]quinazolin-5-amine (CGS 15943) (Baraldi and Borea, 2000). Since a number of selective adenosine agonists and antagonists are available, a pharmacological approach may be used to characterize the adenosine receptor subtype(s) involved in mediating various physiological actions. 2.3 Molecular characterization of the adenosine receptors The four adenosine receptors subtypes have been cloned and characterized in various tissues of different species, including the human brain (Furlong et al., 1992; Libert et al., 1992; Pierce et al., 1992; Salvatore et al., 1993) and rat brain (Reppert et al., 1991; Fink et al., 1992; Stehle et al., 1992; Zhou et al., 1992). In this section, the cloning and characterization of the human and rat adenosine receptor genes are examined. In addition, the regulation of adenosine receptor gene and receptor expression is also reviewed. Emphasis is placed on studies related to the A i and A 2 A receptors since these two receptors are the focus of the current study. Both the A i and A 2 A receptors were first cloned from a canine thyroid library (Libert et al., 1989), and thought to represent new members of the G protein-coupled family before they were recognized as clones of the A i and A 2 A receptor (Furlong et al., 1992; Libert et al., 1992). The human A i receptor is localized on chromosome lq32.1 (Townsend-Nicholson et al., 1995), and contains six exons and five introns (Libert et al., 1992). A l l introns are present in the 5' untranslated region (UTR) except for one, which interrupts the coding region at a position corresponding to the second intracellular loop. The coding region of the human A i receptor consists of 987 bp (Libert et al., 1992). The coding region of the rat brain receptor was similar and consists of 981 bp (Mahan et al., 1991; Reppert et al., 1991). Two distinct A i receptor transcripts, a 3.5 kb and a 5.5 kb transcript, were revealed by Northern blot analysis in rat tissues (Reppert et al., 1991). Both transcripts are expressed throughout the body in various tissues, including the brain and stomach. Another study by Mahan and co-workers has also identified two distinct transcripts, 3.1 kb and 5.6 kb in size, which have been detected in various tissues, including the rat brain and stomach (Mahan et al., 1991). The 3.5 and 3.1 kb species were found to be the predominant forms in the stomach (Mahan et al., 1991; Reppert et al., 1991). Two human A i receptor transcripts have also been demonstrated in the brain (Ren and Stiles, 1994a). These transcripts differ in the 5' untranslated region (UTR) by 54 bp, and were initially thought to occur through alternative splicing (Ren and Stiles, 1994a). However, two different promoters, promoters A and B (Ren and Stiles, 1995), were later found to control the synthesis of the two transcripts. For the rat A\ receptor, Reppert has suggested that the two transcripts are generated by posttranscriptional modifications (Reppert et al., 1991). However, the promoter regions and 5' UTR of the rat A i receptor gene have not been identified. Thus, the origin of the two rat A i receptor transcripts remains unclear. The human and rat A 2 A receptor genes exhibit some similarities with each other (Chern et al., 1992; Fink et al., 1992; Furlong et al., 1992). The human A 2 A receptor gene is 9-10 kb in size, localized on chromosome 22ql 1.2 (Le et al., 1996), and contains 2 exons separated by a 6-6.4 kb intron, localized at a position corresponding to the second intracellular loop (Le et al., 1996; Peterfreund et al., 1996). Northern blot analysis reveals the expression of only one human A 2 A transcript that is 2.6 kb in size (Peterfreund et al., 1996). The rat A 2 A receptor gene was also shown to contain 2 exons, separated by a 7.2 kb intron (Chu et al., 1996). The 5' UTR of the rat A 2 A receptor has also been examined, and contains two independent promoters situated 338 bp apart (Chu et al., 1996). These two promoters were shown to drive the synthesis of two distinct transcripts that differ in the 5' UTR but encode for the same polypeptide sequence. The rat A 2 B receptor was first isolated from a rat brain cDNA library (Stehle et al., 1992). Extremely high levels of two distinct A 2 B receptor transcripts, a 1.8 kb and a 2.2 kb transcript, were found by Northern blot analysis and were expressed in the large intestine and caecum in rats (Stehle et al., 1992). In humans, two A 2 B receptor transcripts, a 1.8 kb and a 2.4 kb transcript, were also shown to be expressed in V A 13 fibroblast cells (Rivkees and Reppert, 1992). The origin of the two human and rat A 2 B receptor transcripts is unclear since the promoter regions and 5' UTR of these genes have yet to be determined. The human A 2 B receptor has been shown to contain a single intron at a position corresponding to the second intracellular loop (Pierce et al., 1992; Jacobson et al., 1995), and is localized on chromosome 17p 12 (Jacobson et al., 1995). The A3 receptor was first discovered by its cloning and characterization in the rat brain (Zhou et al., 1992). This receptor also contains a single intron at a position corresponding to the second intracellular loop. A variant of the rat A 3 receptor, the A3i, has also been cloned and characterized (Sajjadi et al., 1996). This transcript is identical to the A3 receptor cloned by Zhou, except for a 57 bp insertion in the coding region, which occurs from alternative splicing of the intron. This variant was detected in a number of tissues, including the heart, brain, liver and testis (Sajjadi et al., 1996). However, it is unclear whether this variant is also present in the stomach. Evidence suggests that alternative splicing does not occur in the human A 3 receptor gene (Murrison et al., 1996) and only one transcriptional start site was identified when the 5' UTR of this gene was analyzed (Murrison et al., 1996). The human A3 receptor gene is localized on chromosome lpl3.3 (Atkinson et al., 1997), and like the other adenosine receptor genes, it contains an intron (~2.2 kb) in the second intracellular loop. Of the four adenosine receptor subtypes, only the rat A3 receptor has been shown to be alternatively spliced within its coding region. The presence of a variant A3 receptor may have functional implications. 2.4 Regulation of adenosine receptor gene and protein expression The synthesis and expression of adenosine receptors can be regulated at a transcriptional or post-transcriptional level, and may be altered by changes in the physiological environment such as during periods of stress (see Table 1). The genomic structure of the A i , A 2 A , and A3 receptor has been examined and the promoter regions of these genes contain putative binding sites for various transcription factors. A number of binding sites have been identified in the two promoters of the human A i receptor gene (Ren and Stiles, 1995, 1998; Rivkees et al., 1999). Some of these sites were shown to interact with glucocorticoids in the regulation of A i receptor gene transcription (Ren and Stiles, 1999). The promoter region of the rat A i receptor gene has not been defined. However, as in humans, the transcription of this gene is regulated by glucocorticoids (Svenningsson and Fredholm, 1997), suggesting that the promoter region of the rat A i receptor may also contain binding sites for glucocorticoids. The A 2 A receptors also respond to glucocorticoid treatment. Dexamethasone, a synthetic glucocorticoid, has been shown to attenuate the action of N E C A in stimulating cAMP production (Gerwins and Fredholm, 1991), suggesting that glucocorticoids decrease A 2 A receptor expression. However, it is unclear i f glucocorticoids act at a transcriptional or post-transcriptional level to regulate A 2 A receptor expression. A transcriptional repressor of the A 2 A receptor gene has recently been identified in the rat and has been shown to bind to specific nuclear factor-1 proteins to repress gene transcription (Lee et al., 2003). The A 2 A receptor has also been shown to regulate the expression of its own mRNA (Saitoh et al., 1994). Activation of the A 2 A receptor has been shown to transiently increase and then decrease A 2 A receptor gene and protein expression in PC 12 cells (Saitoh et al., 1994). The translation of Aj and A 2 A receptor mRNA can also be regulated by the presence of out-of-frame A U G codons in the 5' UTR (Ren and Stiles, 1994a; Chu et al., 1996; Lee et al., 1999). Translational regulation of the A 2 A receptor may occur in other species as well, since the 5' UTR of the rat A 2 A receptor gene exhibits high interspecies homology (Chu et al., 1996). Stress-induced changes in A i and A 2 A receptor gene and protein expression have also been demonstrated. During periods of oxidative stress, A i and A 2 A receptor gene and/or protein expression is upregulated (Nie et al., 1998; Kobayashi and Millhorn, 1999). A i receptor mRNA and receptor expression were increased in DDT MF-2 cells during oxidative stress. This was mediated by enhanced transcription of the A i receptor by the transcription factor nuclear factor KB (NFKB) (Nie et al., 1998). In the gut, oxidative stress induced by chronic ileitis has also been shown to increase A i receptor gene expression (Sundaram et al., 2003). In PC 12 cells, stress induced by hypoxia was shown to increase A 2 A receptor mRNA and to increase A 2 A receptor protein expression through the synthesis of new A 2 A receptors (Kobayashi and Millhorn, 1999). These changes were mediated by P K C and may result from an increase in A 2 A receptor transcription. This is consistent with another study showing that the transcription of A 2 A receptors was enhanced in human neuroblastoma SH-SY5Y cells by the activation of P K C (Peterfreund et al., 1997). Other forms of stress have also been shown to alter A \ receptor expression in the rat. Stress induced by sustained performance, such as repeated restraint stress, was shown to exhibit increased A i receptor agonist binding in the rat hypothalamus (Anderson et al., 1988). Rats suffering from acute renal failure were also shown to exhibit increased A i receptor binding in renal membranes and increased A i receptor mRNA expression (Gould et al., 1997). Gould has suggested that the increased A i receptor expression was due to enhanced A i receptor transcription. Stress induced by sleep deprivation has also been shown to increase A i receptor mRNA expression in rats (Basheer et al., 2000). Prolonged wakefulness was shown to increase extracellular adenosine levels, which increased A i receptor activation. A i receptor activation was then shown to enhance the activity of the transcription factor N F K B , which initiates transcription of the A i receptor (Basheer et al., 2001). Changes in A i and A 2 A receptor expression can also be induced pharmacologically. Treatment of DDT 1 MF-2 cells with R-PIA was shown to decrease A i receptor expression in membrane preparations within hours (Ramkumar et al., 1991). Chronic treatment with R-PIA has also been shown to decrease A i receptor expression in isolated adipocytes (Green et al., 1992). In the presence of caffeine, an adenosine antagonist, both A i and A 2 A receptor expression are increased. Chronic daily intraperitoneal treatments with caffeine also increase A] receptor expression (Fredholm, 1982), although long-term oral treatment with caffeine did not alter A i receptor mRNA or receptor expression (Johansson et al., 1993). Similarly, in the mouse brain, striatal A 2 A receptor expression was upregulated by chronic intraperitoneal caffeine treatment (Traversa et al., 1994), while long-term oral caffeine treatment did not alter A 2 A receptor mRNA or receptor expression (Johansson et al., 1993). These contradictory findings may result from the different route of drug administration and the preferential exposure of these compounds to different sites of the body. The regulation of A 2 B receptor gene and protein expression is not as well understood. A study in chicken melanocytes has shown that A 2 B receptor mRNA expression can be upregulated by c-MyB (Worpenberg et al., 1997), a transcription factor that is encoded by the c-myb proto-oncogene. In human melanocytes, cell damage induced by 4-tertiary butylphenol, a phenolic agent, was shown to upregulate A 2 B receptor expression (Le Poole et al., 1999). In mouse bone marrow-derived macrophages, A 2 B mRNA expression can also be upregulated by IFN-y, a cytokine involved in activating macrophages (Xaus et al., 1999). The genomic structure of the A3 receptor has been examined, and a number of binding motifs have been identified in the 5' UTR of the human A3 gene (Murrison et al., 1996; Atkinson et al., 1997). These transcriptional binding sites may be important in regulating the promoter activity of the human A3 receptor. Possible binding sites have also been identified in the promoter of the mouse A3 receptor gene (Zhao et al., 1999; Yaar et al., 2002). In particular, the GATA-6 and CRE binding sites are involved in enhancing and inhibiting A3 receptor promoter activity, respectively (Yaar et al., 2002). The regulation of A 3 receptor gene expression has some similarities to that of the A i receptor. As with the A i receptor, glucocorticoids and oxidative stress upregulate A3 receptor mRNA expression (Ramkumar et al., 1995; Sundaram et al., 2003). A3 receptor mRNA expression was significantly increased by dexamethasone treatment (Ramkumar et al., 1995) and by oxidative stress during chronic ileitis (Sundaram et al., 2003). Alterations in A3 receptor expression can also be induced pharmacologically. For example, the exposure of CHO cells to N E C A for 24 h has been shown to significantly decrease A3 receptor expression (Palmer et al., 1995a). The studies presented in this section demonstrate that adenosine receptor gene and protein expression may be regulated through transcriptional and post-transcriptional mechanisms. In addition, the expression of these receptors can be altered by stress-induced changes and by treatment with various adenosine analogs. 2.5 Structural characterization of the adenosine receptors A l l four adenosine receptor subtypes belong to the G protein-coupled receptor family, which signal by activating various heterotrimeric G proteins (Olah and Stiles, 1995). The adenosine receptors are characterized by seven hydrophobic transmembrane domains, an extracellular N-terminal, an intracellular C-terminal, and three intracellular and three extracellular loops (Ralevic and Burnstock, 1998). In the rat, the A j , A 2 A , A 2 b and A3 receptors are comprised of 326-7, 410, 332 and 320 amino acids, respectively (Mahan et al., 1991; Reppert et a l , 1991; Stehle et al., 1992; Zhou et al., 1992; Chu et al., 1996). The large size of the A 2 A receptor results from its extended carboxyl tail (see Fig. 5). Each transmembrane domain consists of approximately 21-28 amino acids and all four receptors are glycosylated by asparagine (N)-linked glycoproteins at the second extracellular loop. Presently, the precise three-dimensional conformation of the adenosine receptors is unknown. Structural analysis identified receptor domains that participate in ligand recognition and binding. Most of these studies concentrate on the A i , A 2 A , and A 3 receptors, due to the lack of highly selective A 2 B receptor analogs. Molecular modeling of the A] I s \u00C2\u00AB\u00E2\u0080\u00A2** I V v v _ - \u00E2\u0080\u00A2 \u00C2\u00BB f i N P S L Q T s v v i M V i r L V A t g V 1 \u00C2\u00BBY r - 1 v D f l R V K L M S '/A \u00E2\u0080\u00A2 M 1 v \" c \ 6 B V p V L l \"e i M M \u00C2\u00BB, 1 V ' V R Y K U l L S I S ^JT\" Y r i m I E T Y L I. I L c v v c n B P S D S L D T IIIIII B Fig. 5. Schematic representation of the human adenosine A, (A), A 2 A (B), and A 3 (C) receptors. Transmembrane domains are numbered from 1 to 7, and amino acids shaded in black have been demonstrated, by site-directed mutagenesis, to participate in ligand binding, (figures are adapted from Olah and Stiles, 2000, and Gao et al., 2002). g (IJzerman et al., 1992), A 2 A (IJzerman et al., 1994; Kim et al., 1995) and A 3 (van Galen et al., 1994) receptors has also been performed based on the structure of bacteriorhodopsin and/or rhodopsin, two structurally well characterized 7 transmembrane-spanning receptors. Based on these modeling studies and additional site-directed mutation studies, several amino acids were found to be involved in ligand binding at the A i , A 2 A and A 3 receptors (See Fig. 5). 2.6 Signal transduction mechanism Table 1 shows that multiple signal transduction mechanisms and second messengers are involved in adenosine receptor signaling (Ralevic and Burnstock, 1998; Fredholm et al., 2001). The A i receptor couples to the G;/G 0 family of G proteins (Freissmuth et al., 1991; Jockers et al., 1994). Stimulation of A i receptors inhibits adenylate cyclase activity through the release of G a i , which decreases cAMP production and modulates cAMP-dependent protein kinase activity (Londos et al., 1980; Ralevic and Burnstock, 1998). This receptor also signals through a number of other messengers, such as phospholipase C (PLC), PKC, C a 2 + channels, K + channels, and mitogen activated protein kinases (MAPK), as described in Table 1. The A 2 A receptor is coupled to the G s protein in most tissues (Olah, 1997), and to G0if, a member of the G s subfamily of G proteins, in the striatum (Kull et al., 2000). Activation of the A 2 A receptor can increase adenylate cyclase activity and cAMP production (Ralevic and Burnstock, 1998). However, these receptors can also mediate changes in the activity of other second messengers such as C a 2 + channels, K A T p channels, and M A P K (Table 1). Like the A 2 A receptors, the A 2 B receptors also stimulate adenylate cyclase activity and increase cAMP production (Ralevic and Burnstock, 1998) through activation of the G s protein (Pierce et al., 1992). The A 2 B receptors can also couple to G q /G i i to regulate PLC Table 1: Adenosine receptor G protein coupling, signal transduction, and regulation of expression Receptor Subtype G protein Coupling Effectors Second Messengers Receptor Expression A, Gj, G 0 4-AC T P L C I P L C 1 cAMP (van Calker et al., 1979) T IP3, DAG, PKC (Gerwins and Fredholm, 1995) 1IP3, DAG, PKC (Delahunty et al., 1988) T PLD (Gerwins and Fredholm, 1995) T K + channels (Belardinelli et al., 1995) iCa2+ channels (Dolphin et al., 1986) t MAPK (ERK1/2, P42/p44, p38) (Dickenson et al., 1998; Schulte and Fredholm, 2000; Robinson and Dickenson, 2001) upregulated by glucocorticoid treatment (Svenningsson and Fredholm, 1997), oxidative stress (Nie et al., 1998), nuclear factor KB binding (Nie et al., 1998), caffeine treatment (i.p.) (Fredholm, 1982), and stress [sustained performance stress (Anderson et al., 1988), acute renal failure (Gould et al., 1997), sleep deprivation (Basheer et al., 2000)] suppressed by agonist treatment (PIA) (Green et al., 1992), and out of frame AUG codons (Ren and Stiles, 1994) A 2 A G s , Goif t AC t cAMP (Furlong et al., 1992) t Ca 2 + channels (Gubitz et al., 1996) f K + channels (Akatsuka et al., 1994) t MAPK (ERK1/2) (Sexl et al., 1997; Schulte and Fredholm, 2000) upregulated by oxidative stress (Kobayashi et al., 1998), PKC activation (Peterfreund et al., 1997), and treatment with caffeine (i.p.) (Traversa et al., 1994). suppressed by glucocorticoid treatment (Gerwins and Fredholm, 1991), binding of nuclear factor-1 proteins (Lee et al., 2003), A 2 A receptor activation (Saitoh et al., 1994), and out of frame AUG codons (Chu et al., 1996). A 2 B G s , Gq/Gn TAC tPLC tcAMP (Feoktistov and Biaggioni, 1995) t IP3, DAG, PKC (Feoktistov and Biaggioni, 1995) TMAPK (ERK 1/2, JNK, p38) (Feoktistov et al., 1999; Schulte and Fredholm, 2000) upregulated by c-MyB transcription factor (Worpenberg et al., 1997), cell damage (Le Poole et al., 1999), and IFN-y (Xaus et al., 1999). A 3 Gj, Gq/Gn iAC TPLC lcAMP(Zhou et al., 1992) t IP3, DAG, PKC (Abbracchio et al., 1995) TMAPK (ERK 1/2) (Schulte and Fredholm, 2000) upregulated by GATA-6 binding (Yaar et al., 2002), oxidative stress (Sundaram et al., 2003), and glucocorticoid treatment (Ramkumar et al., 1995). suppressed by CRE binding (Yaar et al., 2002), and treatment with agonist (NECA)(Palmer et al., 1995) and M A P K activity (Yakel et al., 1993; Feoktistov and Biaggioni, 1995; Schulte and Fredholm, 2000) (Table 1). The A3 receptor is linked to the inhibition of adenylate cyclase activity (Zhou et al., 1992) through activation of the Gj protein (Palmer et al., 1995a), and can alter PLC and M A P K activity via activation of the G q /Gn protein (Abbracchio et al., 1995; Schulte and Fredholm, 2000), as shown in Table 1. Since each adenosine receptor subtype is coupled to specific signal transduction mechanisms, adenosine can mediate specific actions throughout the body by binding to different adenosine receptor subtypes. 3. Biological actions of adenosine in the gastrointestinal tract In the gastrointestinal tract, adenosine has been shown to modulate neurotransmitter release (Christofi, 2001), to alter gastric mucosal blood flow (Nagata et al., 1996; Burnstock, 2001), and to regulate gastric endocrine and exocrine secretion (Burnstock, 2001). The objectives of this thesis were to examine the role of the adenosine receptors in modulating gastrin and somatostatin secretion and to examine adenosine receptor gene expression under different physiological states of the stomach. A detailed review of the gastrointestinal actions of adenosine is presented in this section with emphasis on the role of adenosine in regulating gastric hormone secretion. Adenosine modulates gastrointestinal motility, neurotransmission, and secretion in various species by acting on different adenosine receptors (Burnstock, 2001). Although all four adenosine receptors have been localized in the enteric nervous system of humans (Christofi et al., 2001), few biological studies have studied the gastrointestinal effects of adenosine in humans. The majority of biological studies have been performed in animal models such as the guinea pig and rat (Burnstock, 2001; Christofi, 2001), where adenosine modulates gastrointestinal neurotransmitter release and motility. In the guinea pig ileum, adenosine has been shown to inhibit peristalsis (Shinozuka et al., 1985) and to inhibit cholinergic (Wiklund and Gustafsson, 1987) and adrenergic neurotransmission (Barajas-Lopez et al., 1991) via activation of presynaptic A i receptors (Barajas-Lopez et al., 1991; Nitahara et al., 1995). Adenosine also depolarizes submucosal neurons of the ileum through activation of A 2 A receptors (Barajas-Lopez et al., 1991). In the guinea pig colon, adenosine was shown to inhibit contraction through activation of A 2 B receptors on smooth muscle cells (Kadowaki et al., 2000). Thus, adenosine can act on different adenosine receptor subtypes to exert distinct actions on various regions of the guinea pig gastrointestinal tract. In the rat, adenosine also modulates gastrointestinal neurotransmission, motility, and secretion. Adenosine was shown to induce relaxation of the gastric fundus (Matharu and Hollingsworth, 1992), enhance lower gastrointestinal propulsion (Suzuki et al., 1995), and inhibit peristalsis in the jejunum (Coupar and Hancock, 1994). These actions may be mediated by the activation of various adenosine receptor subtypes. In the rat ileum, the A i subtype was shown to modulate the myenteric reflex response by eliciting ascending contraction and descending relaxation (Storr et al., 2002). In the rat duodenum, A i and A 2 B receptors were found to inhibit contraction of the longitudinal muscle and stimulate contraction of the muscularis mucosae, respectively (Nicholls et al., 1996). In the rat stomach, adenosine has been shown to alter gastric mucosal blood flow (Cho and Ogle, 1990; Cho et al., 1991; Cho et al., 1995), and to regulate ethanol-induced gastric dilation in submucosal arterioles (Nagata et al., 1996). Our laboratory demonstrated that the administration of the A 2 A receptor agonist, CGS 21680, significantly decreases perfusion pressure in the isolated vascularly perfused rat stomach (Yip and Kwok, 2004), suggesting that the A 2 A receptor subtype may be involved in mediating gastric vasodilation. Adenosine also regulates gastric endocrine and exocrine secretion. In particular, adenosine and its analogs can inhibit (Gerber et al., 1984; Glavin et al., 1987; Scarpignato et al., 1987), stimulate (Puurunen et al., 1986), or have no effect (Puurunen et al., 1987; Puurunen and Huttunen, 1988) on gastric acid secretion, depending on the route of drug administration. This nucleoside can also inhibit gastrin release (DeSchryver-Kecskemeti et al., 1981; Harty and Franklin, 1984; Kwok et al., 1990) and stimulate or inhibit gastric somatostatin release, depending on the concentration administered (Kwok et al., 1990). The gastric secretory actions of adenosine are discussed in section 3.2. 3.1 Gastroprotective actions of adenosine In humans, caffeine has been implicated in promoting gastric ulcer formation (Cooke, 1976). Although the mechanism remains unclear, adenosine antagonists such as caffeine and theophylline were shown to stimulate gastric acid secretion (Krasnow and Grossman, 1949), possibly by inhibiting the action of adenosine. Studies have suggested that the gastric juice of ulcer patients contains decreased levels of adenosine (Durak et al., 1994) since xanthine oxidase activity and 5'nucleotidase activity in these patients were shown to be increased and decreased, respectively. As reviewed in Section 1,5' nucleotidase is responsible for the synthesis of adenosine (see Fig. 3), while xanthine oxidase is involved in the metabolism of adenosine (see Fig. 1). Ulcer patients also exhibit decreased A D A activity in the vicinity of a healed ulcer after ranitidine treatment (Namiot et al., 1993) and increased A D A activity in the area surrounding an unhealed ulcer (Namiot et al., 1993). A D A is a metabolic enzyme of adenosine (see Fig. 3). Thus, it is likely that increased and decreased adenosine concentrations are also found surrounding a healed and unhealed ulcer, respectively. These observations suggest that adenosine may be involved in the healing process and may be important in protecting the stomach against ulcer formation. In rats, adenosine can act as a gastroprotective agent. Adenosine protects against indomethacin- (Bozkurt et al., 1998), ethanol- (Cho and Ogle, 1990; Cho et al., 1991), cold restraint stress- (Geiger and Glavin, 1985; Westerberg and Geiger, 1987), and ischemia/reperfusion- (Konturek et al., 2001; Pajdo et al., 2001) induced gastric ulcer formation. The ability of the non-selective adenosine antagonists 8-phenyltheophylline (8-PT) or 8-(p-sulfophenyl)theophylline (8-SPT) to block the protective action of adenosine in some of these studies confirms that adenosine receptors are involved (Geiger and Glavin, 1985; Westerberg and Geiger, 1987; Konturek et al., 2001; Pajdo et al., 2001). In indomethacin- and ethanol-treated rats, adenosine was also shown to inhibit indomethacin-induced gastric acid secretion (Bozkurt et al., 1998) and basal acid secretion (Cho and Ogle, 1990), respectively. In ethanol- and ischemia-induced ulcer studies, an increase in gastric mucosal blood flow was also observed (Cho et al., 1991; Konturek et al., 2001; Pajdo et al., 2001). Thus, the ability of adenosine to inhibit gastric acid secretion and to increase gastric mucosal blood flow may play an important role in the gastroprotective action of adenosine. Although the action of adenosine is mainly protective, studies by Ushijima have shown that adenosine may also exacerbate gastric mucosa damage (Ushijima et al., 1985, 1992). Contrary to Westerberg (Westerberg and Geiger, 1987), the administration of adenosine analogs increased restraint stress-induced gastric ulcer formation in rats. The cause of this functional difference is currently undetermined. 3.2 Effect of adenosine on gastric acid secretion In humans, indirect evidence suggests that adenosine is involved in regulating gastric acid secretion. Studies have shown that fundic A D A activity increases in parallel with gastric acid output (Namiot et al., 1990). Since A D A is a metabolic enzyme of adenosine, an increase or decrease in the activity of this enzyme should decrease or increase adenosine levels, respectively. Patients suffering from hypersecretion of acid exhibited higher fundic A D A activities (Namiot et al., 1990), while those suffering from the absence of gastric acid secretion, a condition known as achlorhydria, exhibited lower A D A activities. The inhibition of gastric acid secretion by ranitidine also decreased A D A activity (Namiot et al., 1991). Thus, in humans, it is possible that gastric acid secretion is regulated, in part, by A D A activity, and consequently by the adenosine level. The role of adenosine in regulating gastric acid secretion is also supported by the gastric action of caffeine and theophylline, two non-selective adenosine receptor antagonists. Both of these compounds stimulate gastric acid secretion (Krasnow and Grossman, 1949; Cohen and Booth, 1975), and may mediate this action by blocking the inhibitory effect of endogenously released adenosine. Studies using experimental animal models indicate that the effect of adenosine on gastric acid secretion is likely to be species-specific, and may depend on the site(s) at which adenosine acts. Gastric acid is secreted by parietal cells which reside in the corporeal mucosa of the stomach (Sachs et al., 1997). The regulation of gastric acid secretion is complex and is controlled by neural, paracrine, and endocrine pathways that modulate the release of stimulatory mediators such as gastrin, acetylcholine, and histamine, and the release of inhibitory compounds such as somatostatin and prostaglandins (Hersey and Sachs, 1995). Adenosine has been shown to inhibit gastric acid secretion in the dog, guinea pig, and rat, but in rabbits, adenosine stimulates gastric acid secretion. In isolated gastric glands (Gil-Rodrigo et al., 1990) and enriched parietal cells (Ota et al., 1989; Ainz et al., 1993) of the rabbit, adenosine and its analogs were shown to significantly stimulate basal and potentiate histamine-stimulated aminopyrine uptake. [14C]aminopyrine uptake was used as an index of gastric acid secretion since it can become protonated and trapped in the acidic canaliculi of the parietal cell (Berglindh et al., 1976). The rank order of potency of different agonists in stimulating acid secretion was shown to be N E C A > 2-chloroadenosine (2-CA) > adenosine, which suggests the involvement of adenosine A 2 receptor (Ota et al., 1989). The stimulatory action of adenosine on isolated gastric glands (Gil-Rodrigo et al., 1990) and N E C A on enriched parietal cells (Ainz et al., 1993) were also inhibited by theophylline. Since activation of A 2 receptors increases cAMP production, this proposal is consistent with findings demonstrating the ability of adenosine to increase cAMP levels in the enriched rabbit parietal cell preparation (Ota et al., 1989). In these experiments, the effect of adenosine was not altered by the endogenous release of the acid secretagogue histamine (Ainz et al., 1993) or by the production of the acid inhibitor prostaglandin E 2 (Ota et al., 1992). Thus, in the rabbit, adenosine may act directly on the A 2 receptors of parietal cells to stimulate gastric acid secretion. In this species, ATP was shown to inhibit histamine-stimulated aminopyrine uptake in the isolated gastric gland (Gil-Rodrigo et al., 1990). Further experiments demonstrated that this is likely due to the activation of ATP receptors, of the P 2y subtype, on the parietal cells (Gil-Rodrigo et al., 1996). The inhibitory action of ATP is consistent with the localization of P 2 y receptors on gastric glands of the rabbit fundus (Vallejo et al., 1996). Thus, in rabbits, purinergic compounds are able to directly alter gastric acid secretion. In particular, adenosine can act on A 2 receptors and ATP can act on P 2 y receptors to stimulate and inhibit gastric acid secretion, respectively, from parietal cells. In other species, such as the dog, adenosine has been shown to inhibit histamine- and methacholine-stimulated gastric acid secretion in vivo (Gerber et al., 1984). This inhibitory effect may be due to the direct action of adenosine on the parietal cells since 2-chloroadenosine (2-CA) and R-PIA have been shown to inhibit histamine-stimulated aminopyrine uptake from isolated canine parietal cells (Gerber et al., 1985). Activation of A i receptors is likely involved in this action since the A i selective analog R-PIA was more potent than 2-CA in inhibiting histamine-stimulated cAMP accumulation. Additional studies have also shown that the inhibitory effect of adenosine is unlikely to be due to changes in the release of the acid secretagogue histamine (Payne and Gerber, 1997), or the activity of phosphodiesterase (Gerber et al., 1984, 1985). Phosphodiesterase metabolizes cAMP, and inhibitors of this enzyme, such as high concentrations of theophylline and caffeine, may stimulate gastric acid secretion (Barnette et al., 1995). Furthermore, adenosine does not mediate its effect by altering the synthesis of prostaglandin E 2 , an inhibitor of gastric acid secretion (Gerber et al., 1984; Geiger and Glavin, 1985). These findings suggest that, in the dog, adenosine may act directly on A i receptors of the parietal cells to inhibit gastric acid secretion. In dogs, adenosine has also been shown to modulate the release of gastrin (Schepp et al., 1990), a potent gastric acid secretagogue (Hersey and Sachs, 1995). In isolated canine antral cells, R-PIA was shown to significantly inhibit forskolin-stimulated gastrin release, while C V 1808 had no effect on this action (Schepp et al., 1990). CV1808, however, slightly stimulated basal gastrin release and potentiated bombesin-stimulated gastrin release (Schepp et al., 1990). Since R-PIA and C V 1808 preferentially bind to the A i and A 2 receptors, respectively, these results suggest that A\ receptors may be involved in inhibiting gastrin release, while the A 2 receptors are involved in stimulating gastrin release. In guinea pigs, adenosine was shown to inhibit histamine and gastrin-stimulated acid secretion from oxyntic cells (Heldsinger et al., 1986). Adenosine and its analogs inhibited histamine-stimulated hydroxyl ion production with the following rank order of potency: R-PIA > NEC A > adenosine. The preferential action of R-PIA on A i receptors suggests that this subtype is involved in the inhibitory action of adenosine on gastric acid secretion (Heldsinger et al., 1986). This adenosine A i receptor-mediated inhibitory action is similar to that in the dog (Gerber et al., 1985). The gastric effects of adenosine have been studied extensively in the rat. Various studies have shown that adenosine inhibits, stimulates, or does not alter gastric acid secretion (see Table 2), depending on the route of drug administration, the state of anesthesia, and whether or not the vagus is intact. Intraperitoneal (i.p.), subcutaneous (s.c), intraduodenal (i.d.), oral, intragastric, and intracerebroventricular (i.c.v.) administration of adenosine and its analogs inhibited gastric acid secretion (Puurunen et al., 1986; Scarpignato et al., 1987; Puurunen and Huttunen, 1988; Westerberg and Geiger, 1989; Cho and Ogle, 1990), while intravenous (i.v.) administration of these drugs stimulated gastric acid secretion (Puurunen et al., 1986). The localization of the adenosine receptors and their preferential exposure to adenosine through different routes of administration may account for these functional differences. The various effects of adenosine suggest that this nucleoside may regulate gastric acid secretion through more than one site of action. Possible sites of action are addressed in the following discussion. Table 2: Adenosine and adenosine agonists on gastric acid secretion in the rat Administration Drug State Effect Reference i.p. R-PIA, S-PIA, 2-CA, NECA conscious \u00E2\u0080\u00A21 basal acid secretion i i.d adenosine anaesthetized no effect 2 i.d. R-PIA anaesthetized \u00E2\u0080\u00A2I basal acid secretion 2 oral R-PIA anaesthetized i basal gastric secretion 2 intragastric R-PIA anaesthetized I basal gastric secretion 2 s.c. R-PIA anaesthetized 1 basal acid secretion 2 s.c. adenosine anaesthetized \u00E2\u0080\u00A2I basal gastric secretion 2-5 s.c. R-PIA conscious 4- basal gastric secretion 6 s.c. S-PIA conscious no effect 6 s.c. adenosine conscious I indomethacin-induced acid secretion 7 i.v. R-PIA vagotomized & anaesthetized no effect 8 RPIA & NECA isolated parietal cells no effect 8 i.v. adenosine, NECA, R-PIA, CHA, S-PIA, 2-CA anaesthetized T basal acid secretion 8 i.v. R-PIA vagotomized & anaesthetized no effect 9 i.c.v. R-PIA anaesthetized no effect 9 i.c.v. R-PIA conscious I basal acid secretion 9 i.c.v. adenosine, NECA, R-PIA, S-PIA, conscious I basal acid secretion 10 i.c.v. NECA vagotomized & conscious no effect 10 i.c.v. NECA anaesthetized no effect 10 1 Westerberg and Geiger, 1989;2 Scarpignato et al., 1987;3-5 Cho and Ogle, 1990; Cho et al., 1991; Cho et al , 1995; 6Glavin et al., 1987; 7Bozkurt et al , 1998;8 Puurunen et a l , 1987;9 Puurunen et a l , 1986; 1 0 Puurunen and Huttunen, 1988. In conscious rats, the i.e.v. administration of adenosine has been shown to inhibit gastric acid secretion. This effect may be mediated by the action of adenosine on the central nervous system. The regulation of gastric acid secretion by the central nervous system involves the dorsomotor nucleus of the vagus (DMNV), the hypothalamus and the nucleus tractus solitarius (NTS) (Hersey and Sachs, 1995). The hypothalamus and NTS receive sensory input from the periphery and regulate the activity of the D M N V . Activation of the D M N V can stimulate gastric acid secretion. Central stimuli from the hypothalamus and the NTS are integrated at the D M N V , and efferent fibers originating from the D M N V travel to the stomach via the vagus nerve. The vagus also contains fibers originating from the nucleus ambiguus, but these nerves are mainly involved in controlling gastric motility (Hersey and Sachs, 1995). The i.e.v. administration of adenosine analogs to conscious rats with intact vagi inhibited gastric acid secretion with the following rank order of potency: N E C A > R-PIA > S-PIA (Puurunen and Huttunen, 1988). The higher potency of N E C A suggests that the A 2 receptor is involved in this action. The i.c.v. administration of N E C A , however, did not alter gastric acid secretion in vagotomized rats (Puurunen and Huttunen, 1988) or anaesthetized rats with intact vagi (Puurunen and Huttunen, 1988). Anesthetics can suppress vagal tone (Murthy et al., 1982; Yamamura et al., 1983). Thus, the central A 2 receptors may inhibit gastric acid secretion by suppressing central stimulatory vagal impulses to the stomach. Furthermore, the inhibitory effect of N E C A was attenuated by the removal of brain monoamines such as noradrenaline, dopamine and serotonin, but was not affected by opioid or prostaglandin inhibitors (Puurunen and Huttunen, 1988). Thus, in the brain, the A 2 receptors may mediate the inhibitory effect on acid secretion by altering the release of biogenic monoamines. Gastric acid secretion was also inhibited by s.c, i.p., i.d., orally and intragastrically administered adenosine agonists (see Table 2). However, it is unclear whether this action is mediated centrally or peripherally. In one study, the s.c. administration of R-PIA was able to inhibit gastric acid secretion, while the s.c. administration of S-PIA had no effect (Glavin et al., 1987). In another study, the potency of N E C A in inhibiting acid secretion was equal to that of R-PIA when administered i.p. (Westerberg and Geiger, 1989). Thus, both the A i and A 2 receptors may be involved in the inhibition of gastric acid secretion. Gastric acid secretion is regulated peripherally through several mechanisms. Acetylcholine, gastrin, and histamine stimulate gastric acid secretion by acting on the M3 receptor, C C K B receptor, and H 2 receptor, respectively, which are expressed on parietal cells (Hersey and Sachs, 1995). Somatostatin and prostaglandins can also act directly on parietal cells to inhibit acid secretion (Schubert and Shamburek, 1990). Thus, adenosine may modulate gastric acid secretion by regulating the release of these substances. Unlike in other species, adenosine does not act directly on the parietal cells to alter gastric acid secretion in the rat. N E C A and R-PIA were shown to have no effect on basal, histamine-stimulated, or carbachol-stimulated aminopyrine accumulation in the rat isolated enriched parietal cell preparation (Puurunen et al., 1987). Histamine and carbachol were shown to stimulate acid secretion by increasing cAMP and inositol phosphate levels, respectively, but neither N E C A nor R-PIA elicited this effect (Puurunen et al., 1987). Histamine is released from enterochromaffm-like (ECL) cells of the stomach (Sachs et a l , 1997). Its release is stimulated by gastrin, Ach, and P-adrenergic agonists, and is inhibited by somatostatin (Hersey and Sachs, 1995). Although the effect of adenosine on histamine has not been examined in the rat, adenosine was shown not to alter histamine release in both the dog and rabbit (Ainz et al., 1993; Payne and Gerber, 1997). Acetylcholine is released from the enteric nervous system and can stimulate gastric acid secretion either by acting directly on the parietal cells or indirectly by stimulating gastrin secretion or inhibiting somatostatin secretion (Hersey and Sachs, 1995). Studies on gastrointestinal motility have demonstrated that adenosine inhibits acetylcholine release from the myenteric plexus of several species. Adenosine was found to inhibit nicotinic neurotransmission in the antrum (Christofi et al., 1992) and ileum (Gustafsson, 1981). In the submucosal plexus, adenosine inhibits the presynaptic release of acetylcholine through activation of the A i receptor (Barajas-Lopez et al., 1991). Although adenosine can regulate acetylcholine release, this action has only been implicated in the modulation of gastric motility. It is unclear whether alterations in acetylcholine release are involved in the adenosine-induced inhibition of gastric acid secretion. Adenosine may inhibit gastric acid secretion by stimulating the release of the gastric acid inhibitor, somatostatin. Somatostatin is released from D-cells of the gastric mucosa and from enteric nerves (Mcintosh, 1985). The stomach contains two distinct populations of D-cells, one residing in the corpus and the other in the antrum. In the present study, the term \"corpus\" refers to the acid-secreting portion of the stomach (see Fig. 36). However, a number of studies also refer to this region as the \"fundus\". Thus, in this chapter, both \"corpus\" and \"fundus\" are used to describe the acid-secreting region of the stomach. In Chapter 3 (Results), the term \"fundus\" will be used exclusively to describe the non-glandular region of the stomach. Somatostatin-containing D-cells of the fundus are localized at the basal mucosa and are characterized as small rounded or pyramid-shaped cells with long processes (Keast et al., 1985). In the antrum, D-cell density is higher than in the fundus (Keast et al., 1985). Antral D-cells are also localized at the base of the gastric gland (Buchan et al., 1985a) and are characterized as flask-shaped cells with long processes (Keast et a l , 1985). The fundic and antral D-cells of the rat are distinct and respond differently to gastric acidity and the presence of food (Schwarting et al., 1986). Antral D-cells are open to the luminal environment (Hersey and Sachs, 1995). These cells detect changes in the intraluminal pH, and respond to decreases in gastric pH by releasing somatostatin (Schusdziarra et al., 1983). Increases in gastric pH, such as those induced by omeprazole treatment, have been shown to decrease somatostatin gene expression (Brand and Stone, 1988; Wu et al., 1990a; Wu et al., 1990b; Sandvik et al., 1995), plasma somatostatin concentration, antral somatostatin concentration, and antral D-cell density (Allen et al., 1986; Lee et al., 1992; Pawlikowski et al., 1992). Omeprazole is a drug that inhibits gastric acid secretion by binding irreversibly to the proton pump, H + K + ATPase, on parietal cells (Hirschowitz et al., 1995). The prandial state of the stomach also alters somatostatin synthesis and release. After prolonged fasting (96 h), antral D-cell density has also been shown to decrease (Schwarting et al., 1986). Fundic D-cells are closed cells that do not interact with the luminal environment. Unlike antral D-cells, changes to the intraluminal pH and prolonged fasting do not alter fundic somatostatin release (Schusdziarra et al., 1983) and D-cell density (Schwarting et al., 1986), respectively. Instead, somatostatin release from fundic D-cells may be stimulated by intrinsic (Schubert et al., 1991) and extrinsic (Sandvik et al., 1993) nerve stimulation. The distribution of gastric somatostatin-containing nerve fibers differs among species. In the rat stomach, these fibers are not present in the mucosa, external muscle or myenteric plexus, but a few single somatostatin-containing fibers have been observed in the muscularis mucosae (Keast et al., 1985). In another study, somatostatin-containing fibers were absent from the mucosa and submucosal plexus; however, a few somatostatin-containing fibers were demonstrated in the smooth muscle and in the myenteric ganglia (Ekblad et al., 1985). Somatostatin is released from the long processes extending out from the D-cells, and has been shown to act in a paracrine manner to inhibit gastrin release from G-cells, acid secretion from parietal cells (Park et al., 1987; Kusumoto et al., 1995) and histamine release from E C L cells (Prinz et al., 1993). Fundic D-cells have also been shown to be in close proximity to the vasculature (Kusumoto et al., 1995), and antral D-cell processes have been shown to project to the blood capillaries (Kusumoto and Grube, 1987). Thus, somatostatin may be released into the blood and act in an endocrine manner. Somatostatin release is inhibited by cholinergic agents (Koop et al., 1982), enkephalins (Mcintosh et al., 1983), substance P (Kwok et al., 1985; Mcintosh et al., 1987a), neurokinins and tachykinins (Kwok et al., 1988) and is stimulated by p-adrenergic agents (Koop et al., 1982; Koop et al., 1983), histamine (Mcintosh et al., 1987a), cholecystokinin (CCK) (DelValle et al., 1996), secretin/VIP (Chiba et al., 1980; Saffouri et al., 1984), calcitonin gene related peptide (CGRP) (Manela et al., 1995; Rasmussen et al., 2001), glucagon (Chiba et al., 1980) and bombesin/gastrin releasing peptide (GRP) (Martindale et al., 1982). Our laboratory has demonstrated that basal somatostatin release can also be stimulated by adenosine in a concentration-dependent manner (Kwok et al., 1990). The stimulatory effect of adenosine on SLI was not altered by atropine and hexamethonium, cholinergic antagonists, or by propranolol, a P-adrenergic antagonist. Therefore, adenosine-induced attenuation of gastric SLI release does not involve changes in cholinergic or adrenergic neurotransmission (Kwok et al., 1990). Adenosine was also shown not to mediate its stimulatory action on SLI through its vasodilatory effects since nitroprusside, a potent vasodilator, did not alter gastric SLI release (Yip and Kwok, 2004). It is possible that adenosine acts directly on A 2 A receptors on D-cells to stimulate somatostatin release. Activation of A 2 A receptors has been shown to stimulate adenylate cyclase activity, cAMP production, PKC activity, and to activate L-type C a 2 + channels (see Table 1). The same second messengers were shown to be involved in mediating somatostatin release from D-cells. In human cultured D-cells, somatostatin release was stimulated by activation of PKC, production of cAMP, and increases in intracellular C a 2 + concentration (Buchan et a l , 1990). In rat fundic D-cells, bombesin-induced release of somatostatin was also found to be mediated through a calcium-phospholipid-dependent mechanism (Schaffer et a l , 1997). Furthermore, in canine fundic D-cells, CCK-stimulated somatostatin release was mediated by an increase in intracellular C a 2 + induced by activation of L-type C a 2 + channels (DelValle et a l , 1996). Since the A 2 A receptors are linked to second messengers which are involved in stimulating somatostatin release, it is possible that activation of A 2 A receptors on D-cells stimulates somatostatin release. Experiments have shown that somatostatin gene transcription is regulated by cAMP (Montminy et a l , 1996a, 1996b). A cAMP response element (CRE), which confers cAMP inducibility, has been identified in the somatostatin gene (Comb et a l , 1986) and can mediate the transcriptional induction of somatostatin. Activation of the A 2 A receptors has been shown to increase the expression of genes that contain a cAMP response element (CRE), such as the tyrosine hydroxylase gene (Chae and Kim, 1997). Adenosine may therefore regulate the synthesis and release of somatostatin by activating A 2 A receptors on D-cells. The cellular localization of these receptors, however, has not been determined in the stomach. Gastrin is a major hormonal stimulant of gastric acid secretion (Edkins, 1905; Kovacs et al., 1989). In the stomach, it is synthesized in G-cells of the antral mucosa. These cells are open to the luminal environment, and their activity is partially regulated by the gastric contents. Gastrin secretion is enhanced by a number of chemical and physical factors such as acetylcholine (Matsuno et al., 1997), P-adrenergic agonists (DeSchryver-Kecskemeti et al., 1981; Koop et al., 1983), bombesin/GRP (Martindale et al., 1982; Guo et al., 1988; Campos et al., 1990), vagotomy (Stening and Grossman, 1970; Pederson et al., 1984), proteins and amino acids (Guo et al., 1988; DelValle and Yamada, 1990), gastric distension (Lloyd et al., 1992), and intraluminal pH (Hoist et al., 1983). Gastric pH serves as a negative feedback mechanism since luminal acidification (pH < 3.5) has been shown to inhibit the further release of gastrin (Magee, 1996). Increased gastric pH induced by omeprezole treatment, conversely, has been shown to increase gastrin mRNA, antral gastrin and plasma gastrin levels (Brand and Stone, 1988; Inauen et al., 1988; Larsson et al., 1988; Dockray et al., 1991). Gastrin release is inhibited by the antral somatostatin-secreting D-cells (Saffouri et al., 1980). D-cells may act as chemoreceptors for intraluminal pH and modulate somatostatin secretion to control gastrin secretion (Hersey and Sachs, 1995). An inhibition of basal somatostatin release may stimulate gastrin secretion since basal gastrin secretion was shown to be tonically restrained by somatostatin (Saffouri et al., 1980). Somatostatin acts on somatostatin 2 receptors located on the G-cells (Zaki et al., 1996), and inhibits gastrin secretion by inhibiting adenylate cyclase activity (DelValle et al., 1990). Our laboratory has shown that adenosine can inhibit immunoreactive gastrin (IRG) release from the isolated vascularly perfused rat stomach in a concentration-dependent manner (Kwok et a l , 1990). Adenosine was also found to inhibit carbachol- and norepinephrine-stimulated gastrin secretion in rat antral mucosa cells (DeSchryver-Kecskemeti et a l , 1981), and to inhibit basal and carbachol-stimulated gastrin secretion in the rat antral mucosal fragments (Harty and Franklin, 1984). The effect of adenosine on gastrin secretion is probably not centrally mediated since the administration of N E C A (i.c.v.) did not alter serum gastrin concentrations (Puurunen and Huttunen, 1988). The adenosine receptor subtype involved in inhibiting gastrin release in the rat has not been examined. However, in the dog, adenosine was suggested to inhibit gastrin secretion by activating A i receptors localized on G-cells (Schepp et a l , 1990). Activation of A i receptors inhibits adenylate cyclase activity, cAMP production, and C a 2 + channel activity, and has also been shown to alter P K C activity (See Table 1) (Ralevic and Burnstock, 1998). The same second messengers are involved in regulating gastrin secretion. In isolated rat and pig parietal cells, cAMP is required to elicit aminopyrine accumulation (Cabero et a l , 1993; L i et a l , 1995). Cyclic A M P has also been shown to potentiate bombesin-evoked gastrin release from isolated perfused rat stomachs (Guo et a l , 1988) and to stimulate gastrin release from isolated human G-cells in culture (Campos et a l , 1990). Activation of P K C (Campos et a l , 1990) and an elevation in intracellular C a 2 + concentration have also been shown to stimulate gastrin release (Campos et a l , 1990; Buchan and Meloche, 1994; Seensalu et a l , 1997). The release of gastrin from isolated G-cells was also shown to be mediated by the cytosolic Ca concentration (Campos et a l , 1990; Buchan and Meloche, 1994; Seensalu et a l , 1997). Thus, it is possible that activation of A i receptors on G-cells leads to an inhibition of gastrin release through an inhibition of cAMP production and C a 2 + channel activity. The A i receptor may also be involved in regulating gastrin gene expression since the expression of this gene has been shown to be cAMP-dependent (Shiotani and Merchant, 1995). II. Hypothesis I hypothesize that activation of gastric adenosine receptors plays a role in the regulation of gastric exocrine and endocrine secretion. In particular, adenosine may inhibit gastric acid secretion by inhibiting gastrin release and stimulating somatostatin release. I propose that adenosine acts on adenosine A i receptors on G-cells and A 2 A receptors on D-cells to inhibit gastrin and stimulate somatostatin release, respectively. I also hypothesize that changes to the gastric state, such as those induced by fasting and omeprazole treatment, may alter adenosine receptor gene and protein expression to regulate gastrin and somatostatin release. III. Rationale Gastric acid secretion may be inhibited by adenosine through the regulation of gastrin and somatostatin release since adenosine has previously been shown to alter IRG and SLI release in the isolated vascularly perfused stomach. Adenosine was shown to inhibit basal and carbachol-stimulated gastrin release from isolated G-cell preparations (Schepp et al., 1990). Thus, this compound may act directly on A i receptors localized on G-cells to inhibit gastrin release. Adenosine may also stimulate somatostatin release by acting on A 2 A receptors localized on the gastric D-cells since previous studies performed in our laboratory have shown that the effect of adenosine was not altered by cholinergic and adrenergic agonists. In addition, gastric adenosine receptor expression may be altered under different physiological conditions since changes to the prandial and acid secretory state of the stomach have been shown to regulate the synthesis and release of gastrin and somatostatin (Brand and Stone, 1988; Wu et a l , 1991; Sandvik et a l , 1993). IV. Objectives and specific aims The objectives of this thesis were to determine the adenosine receptor subtypes that are involved in regulating SLI and IRG release, to provide information about the distribution, localization, and structure of these receptors, and to study how adenosine receptors are regulated under different physiological states of the stomach. The specific aims of this study are outlined below: 1. Determine the adenosine receptor subtypes involved in regulating IRG and SLI release in the isolated vascularly perfused rat stomach, using selective adenosine receptor agonists and antagonists. 2. Determine the distribution of adenosine receptor gene expression in morphologically and functionally distinct regions of the rat stomach using RT-PCR, and determine the structure of the gastric A i , A 2 A , and A3 receptor by cloning and sequencing. 3. Develop a sensitive (competitive RT-PCR and Real-Time RT-PCR) assay to measure adenosine A i and A 2 A receptor gene expression in various regions of the rat stomach. 4. Examine whether adenosine receptor gene expression and function are regulated by different physiological states of the stomach, in particular, fasting and omeprazole treatment. 5. Investigate the cellular localization and distribution of gastric A\ and A 2 A receptors in the stomach by immunohistochemistry. Chapter 2: Methods Animals were treated in accordance with the guidelines of the University of British Columbia Committee on Animal Care. I. Effect of adenosine analogs on gastric SLI and IRG release 1. Isolated vascularly perfused rat stomach preparation Male Wistar rats (250 to 325 g) were housed in light and temperature controlled rooms with free access to food and water. Animals were deprived of food, but had free access to water for at least 14 h prior to experiments. Rats were anesthetized with an i.p. injection (60 mg/kg) of sodium pentobarbital (Somnotol\u00C2\u00AE, M T C Pharmaceuticals, Cambridge, ON, Canada). The surgical procedures used to isolate the stomach for perfusion have previously been described (Kwok et al., 1990; Kwok and Mcintosh, 1990). The stomach was exposed by an abdominal midline incision. The superior mesenteric artery and vasculature supplying the left and right adrenal glands and kidneys were tied or cut between double ligatures. The pancreas and spleen were carefully dissected out from the stomach without damaging the gastroepiploic artery. To empty the content of the stomach, a drainage cannula was inserted into the stomach through the gastroduodenal junction. The pancreas, spleen, small and large intestines were then removed. Arterial perfusion was achieved by inserting a cannula into the aorta with the tip lying adjacent to the celiac artery. Perfusate was introduced into the stomach via this arterial cannula. Six hundred units of heparin (Sigma-Aldrich, St. Louis, MO) in 2 ml saline solution were immediately injected. The portion of the body above the diaphragm was removed. The vena cava was tied, and the portal vein was cannulated to allow for the collection of venous effluent. The stomach was perfused at a rate of 3 ml/min using a peristaltic pump (Cole-Parmer Instrument Co. Chicago, IL). The perfusate was composed of Krebs' solution (120 mM NaCl, 4.4 m M KC1, 2.5 m M CaCl 2 , 1.2 m M MgS0 4 -7H 2 0, 1.5 m M K H 2 P 0 4 , 25 mM NaHC03, and 5.1 m M dextrose) containing 0.2% bovine serum albumin (BSA, RIA grade; Sigma-Aldrich) and 3% dextran (Clinical grade; Sigma-Aldrich). The perfusate was continuously gassed with a mixture of 95% 0 2 and 5% C 0 2 to maintain a pH of 7.4. Both 0 the perfusate and the stomach preparation were kept at 37 C by thermostatically-controlled heating units throughout the experiment. 2. Drug administration and sample collection Venous effluent was collected via the portal cannula. After a 30 min equilibration period, samples were collected at 5 min intervals. Three basal samples were obtained before drugs were introduced. Drugs were administered into the perfusate via side-arm infusion at a rate calculated to give the final perfusion concentrations. The following drugs were purchased from Sigma-Aldrich: adenosine hemisulphate salt, N6-cyclopentyladenosine (CPA), 2-p-(2-carboxyethyl) phenethylamino-5'N-ethylcarboxamidoadenosine HC1 (CGS 21680), 1 -deoxy-1 - [6- [[(3 -iodophenyl)methyl] amino] -9H-purin-9-yl] -N-methyl-B-D-ribofuranuronamide (IB-MECA), 8-cyclopentyl-l,3-dipropylxanthine (DPCPX), and 3,7-dimethyl-l-propargylxanthine(DMPX). 4-(2-[7-amino-2-(2-furyl)[l,2,4]triazolo[2,3-a][l,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385) was procured from Tocris Cookson Inc. (Ellisville, MO). Adenosine analogs were first dissolved in a small volume of dimethylsulfoxide (DMSO) (BDH, Toronto, ON) and subsequently diluted with saline or perfusate to 0.03 or 0.5% before perfusing into the stomach. The perfusion of DMSO alone, at these concentrations, did not alter basal IRG or SLI release. Five min samples were collected into ice-cold polyethylene scintillation vials containing 0.3 ml of Trasylol\u00C2\u00AE (aprotinin, 10,000 KIU/ml; Miles Labs., Etobicoke, ON). For the measurement of SLI release, 0.5 ml aliquots of each sample were transferred into ice-cold test-tubes containing 0.05 ml Trasylol. For the measurement of IRG release, 1 ml aliquots of each sample were transferred into ice-cold test tubes. Aliquots of samples were frozen and stored at -20\u00C2\u00B0C until assayed. 3. Radioimmunoassay (RIA) for the measurement of SLI release The RIA used to measure SLI has previously been described (Mcintosh et al;, 1978; Mcintosh et al., 1987a; Mcintosh et al., 1987b), and has been used routinely in our laboratory (Kwok et al., 1988; Kwok et a l , 1990). Iodination of somatostatin: Somatostatin was iodinated using chloramine-T. In neutral buffer, chloramine-T oxidizes iodide and allows it to bind, by electrophilic substitution, to residues containing an aromatic group, such as tyrosine. Five pg of !Tyr-somatostatin (Penninsula Laboratories Inc., Belmont, CA) was dissolved in 10 jul of distilled water, followed by the addition of 10 pi 0.5 M phosphate buffer (pH 7.5). Then 10 pi of chloramine-T (2 pg/pl 0.05 M phosphate buffer, pH 7.5, E M Sciences, Darmstadt, Germany) and 1 mCi of Na[ 1 2 5I] (in 10 p.1 of NaOH) were added to the peptide solution. The mixture was incubated for 30 sec, and the reaction was terminated by the addition of 10 pi sodium metabisulphite (5 pg/pl 0.05 M phosphate buffer, pH 7.5, E M Sciences). One milliliter of hormone free plasma and 20 mg of QUSO G32 (Philadelphia Quartz Co., Valley Forge, PA) was then added. The mixture was vortexed, and centrifuged for 2-3 min. The supernatant (fraction A) was removed and kept. The pellet was resuspended in 1 ml of water. The mixture was again vortexed and centrifuged. The supernatant (fraction B) was removed, and the pellet was resuspended in 1 ml distilled water. The procedure was repeated. The supernatant (fraction C) was removed, and the pellet was resuspended in 1 ml of solution containing glacial acetic acid/acetone/water (1:39:40). This solution is used to elute the labeled peptide. The mixture was centrifuged for 20 min, and the supernatant (fraction D) was removed. The pellet was re-suspended in 1 ml of water (fraction E). Fractions A - E were counted, and fraction D, containing the labeled protein, was diluted to 1 x 106 cpm/10 pi in 0.1 M acetic acid containing 0.5% BSA (Miles, Kankake, IL). Aliquots of 100 pi were lyophilized, and stored at -20\u00C2\u00B0C. The somatostatin label was purified on the day of the assay. The lyophilized label was re-dissolved in 2 m M ammonium acetate buffer (pH 4.6) and then applied to a column (12 cm x 1 cm) containing CM-cellulose C M 52 (Whatman Biosystem Ltd., Kent, England) which was previously equilibrated with the same buffer. The column was washed with approximately 50 ml of this buffer, before the label was eluted with 200 m M ammonium acetate buffer (pH 4.6). Buffer was introduced into the column at a rate of ~1 ml/min using a peristaltic pump. Two min fractions were collected during elution. Aliquots (10 pi) from each fraction were counted. The peak fraction was neutralized with 5 M NaOH, diluted to 3,500 \u00C2\u00B1 500 cpm/100 pi, and used for the assays. Standards: To prepare the standards, somatostatin-14 (Bachem California, Torrance, CA) was dissolved in 0.1 M acetic acid containing 0.05% BSA (Miles, Kankake, IL) to a final concentration of 5 p.g/50 (0.1. These 50 p.1 aliquots were lyophilized and stored at -20\u00C2\u00B0C. On the day of the assay, the lyophilized standard was re-dissolved in 100 pi of distilled water and 400 JU.1 of assay buffer to yield a stock of 10 ug/ml. This stock was then serially diluted with assay buffer to obtain standard concentrations of: 500, 250, 125, 62.5, 31.25, 15.625, 7.8, and3.9pg/ml. Assay: Each assay tube contained 100 ul of standard or sample, 100 p.1 of label, and 200 p.1 of somatostatin antibody (ascites SOMA 03 monoclonal antibody, final dilution of 1:4,000,000). The specificity of this antibody has previously been described (Mcintosh et a l , 1987b). The somatostatin antibody and label were diluted in assay buffer [23.77 m M Sodium barbital, 3.9 m M sodium acetate, 43.6 m M sodium chloride, and 0.247 m M ethylmercurithiosalicylic acid sodium salt (Eastman Kodak C o , Rochester, NY)]. The concentration of the drugs used in the present experiments did not cross-react with the antibody. Non-specific binding (NSB) tubes for standards and samples were prepared and contained buffer in place of the antibody, while zero standard tubes contained buffer in place of the standard. Total count (TC) tubes contained only 100 p.1 of labeled somatostatin. Triplicates were prepared for total count, standard, NSB standard, and zero tubes, while duplicates were prepared for the NSB sample and each unknown sample. Assay tubes were incubated for 72 h at 7\u00C2\u00B0C. Free and bound labeled somatostatin were separated by adding 1 ml of dextran-coated activated charcoal [1.25% activated charcoal (Fischer Scientific, Fairlawn, NJ), 0.25% dextran T-70 (Pharmacia, Uppsala, Sweden), and 0.1% hormone free plasma in 0.05 M phosphate buffer, pH 7.5]. Tubes were incubated for 10 min, and centrifuged at 3,000 rpm for 30 min at 10\u00C2\u00B0C. The supernatant was discarded and the charcoal pellet was air dried for >6 h, before the radioactivity was counted. The percentage bound of sample was calculated using the following equation: % Bound = [(TC-standard)/TC - (TC-NSB)/TC)]cpm x 100. The standard curve of the % bound vs. Log(standard concentration) was plotted using GraphPad Prism (v. 3.0, GraphPad software, San Diego, CA), and unknown somatostatin concentrations were determined by interpolation using the standard curve. Inter- and intra-assay variations were found to be <3% and <6.5%, respectively. 4. Radioimmunoassay for the measurement of IRG release IRG was measured by specific RIA as previously described (Jaffe and Walsh, 1978; Fujimiya and Kwok, 1997). Iodination of Gastrin: Gastrin was iodinated using chloramine-T. Five pg of synthetic human gastrin-1 (Penninsula Laboratories, Merseyside, England) dissolved in 10 pi 0.4 M phosphate buffer (pH 7.4) was combined with 10 pi choloramine-T, (5 pg/pl in 0.04 M phosphate buffer, pH 7.4) and 0.3 mCi Na [ l 2 5I]. After a 30 sec incubation, the reaction was terminated with the addition of 10 pi sodium metabisulphite (5 pg/pl in 0.04 M phosphate buffer, pH 7.4). The iodinated gastrin was purified using a DEAE-Sephadex A25 (Amersham Biosciences, Uppsala, Sweden) column (12 cm x 0.5 cm) equilibrated with 0.05 M imidazole buffer (pH 7.5). The label was eluted with 1 M NaCl in a 0.05 M imidazole buffer (pH 7.5) gradient at a flow rate of 1.0 ml/min. A typical elution profile for the labeled gastrin is shown in Figure 6. The four fractions, collected after the second peak, were screened by RIA. The fraction of labeled gastrin that provided the best standard curve, as indicated by measuring control samples of 50 and 100 pg/ml gastrin was further diluted and used. 1500-1 1 1 1 1 1 1 1 1 1 1 1 l Sample Fig. 6. Typical elution profile for the separation of labeled gastrin. 125I-conjugated synthetic human gastrin-I was purified on a DEAE-Sephadex A25 column. The label was eluted with 1 M NaCl in a 0.05 M imidazole buffer gradient (pH 7.5) at a flow rate of 1.0 ml/min. Fractions in the second peak have been shown to exhibit higher specific activity (Jaffe and Walsh, 1978). Thus, the four fractions collected after peak II (# 61-64) were selected for screening by radioimmunoassay. Standards: Human synthetic gastrin-1(2000 pg/ml; Penninsula Laboratories, San Carlos, CA) was serially diluted with assay buffer to obtain 1,000, 500, 250, 125, 62.5, 31.25, 15.625, 7.8, and 3.9 pg/ml standard samples. Assay: The assay buffer was composed of 0.02 M sodium barbital (pH 8.4) containing 0.5% B S A (RIA grade, Sigma-Aldrich). Each assay tube contained 700 pi assay buffer, 100 pi standard (0 to 1000 pg/ml) or sample, 100 pi gastrin antibody (PM1, rabbit polyclonal, final dilution of 1:90,000) and 100 pi labeled gastrin (-2000 counts/tube). The gastrin antibody (PM-1) was kindly provided by Dr. R. Pederson (Physiology, UBC). NSB samples contained buffer in place of the antibody, while zero standard tubes contained buffer in place of the sample, and total count tubes contained only labeled somatostatin. Triplicates were prepared for total count, standard, NSB standard, and zero tubes, while duplicates were prepared for the NSB sample, and each unknown sample. Assay tubes were incubated for 48 h at 7\u00C2\u00B0C. Free and bound labeled gastrin were separated by the addition of 200 pi dextran-coated activated charcoal (1.25% activated charcoal, 0.25% dextran T-70, and 6.5% charcoal extracted plasma in 0.04 M phosphate buffer, pH 6.5). Samples were incubated for 10 min, and centrifuged at 3,000 rpm for 30 min at 10\u00C2\u00B0C. The supernatant was discarded and the charcoal pellet was dried for >6 h, before the radioactivity was counted. The % bound was calculated as described for the SLI RIA. The IRG of unknown samples were determined by interpolation using the standard curve. The inter- and intra-assay variations of this assay were found to be <5.9% and <3.2%, respectively. 5. Data analysis SLI measurements: Although the basal release rate of SLI varied among animals, previous experiments have demonstrated that basal SLI release is well maintained during a 50 min perfusion period (Saffouri et a l , 1980; Kwok et a l , 1988, 1990). Therefore, results are expressed as mean \u00C2\u00B1 SEM of SLI release (%), and calculated as follows: [SLI release (pg/min) during a 5 min period + SLI release (pg/min) during period 1] x 100. To compare the effect of various analogs, results are also expressed as percent change (SLI release), which is calculated as follows: [mean SLI release in the presence of drug - mean basal SLI release (periods 1-3)] pg/min + [mean basal SLI release (periods 1-3)] pg/min x 100. IRG measurements: Basal IRG release is also maintained in the perfused stomach preparation (Saffouri et a l , 1980; Pederson et a l , 1984; Kwok et a l , 1990). The mean \u00C2\u00B1 S E M of IRG release (%) was also calculated as described above. The percentage inhibition of IRG release by various analogs was calculated as follows: [mean basal IRG release (periods 1-3) - mean IRG release in the presence of drug] pg/min + [mean basal IRG release (periods 1-3)] pg/min x 100. Statistics: Statistical significance (P < 0.05) was determined using one-way A N O V A followed by Dunnett's multiple comparison test and the paired or unpaired Student's t-test when appropriate. Statistics were performed using GraphPad Prism, and the EC50 for different drugs were also estimated using the same software. II. Adenosine receptor mRNA: distribution, structure, and quantification 1. Distribution of gastric adenosine receptor mRNA RT-PCR was performed to determine whether the mRNA of all four adenosine receptor subtypes are expressed in various regions of the rat stomach, including the fundus, corpus, antrum, and the mucosa. 1.1 Primer design and synthesis Adenosine A i , A 2 A , A 2 B , and A3 receptor PCR primers were designed based on previously published cDNA sequences of these receptors in the rat brain (See Table 3). Primers were designed with the assistance of Mr. Jeff Hewitt (Department of Biochemistry and Molecular Biology, UBC) using the software program PCGene. Upper and lower primer regions were selected based on several criteria. Regions with similar melting temperature (Tm) were selected. Primers had a GC content of 40-60%. Complementarity between the upper and lower primers, runs of >3 Gs or Cs at the 3' end, and the occurrence of a T at the 3' end were avoided. Primer sets, designated as short length (SL) primers, were used to amplify only a portion of the adenosine receptor cDNA. These were designed for all four adenosine receptors. Full length (FL) primers, which amplify the entire coding region of the receptors, were designed for the A i and A 2 A receptors. The sequences of these primers are listed in Table 3. The primers were synthesized by the Nucleic Acids Protein Services (NAPS) Unit at U B C and were diluted in autoclaved distilled water. Since single stranded D N A at a concentration of 33 Table 3: Adenosine receptor RT-PCR primers and PCR conditions Primer Set' Accession No. 2 Position (brain sequences) Primer Sequence Amplicon Length Annealing Temp. (\u00C2\u00B0C) 3 [MgCl2] m M 4 A,SL M64299 373-392 bp 636-655 bp Upper: 5'TTC C A G GCT GCC TAC ATT GG3' Lower: 5 'CAA TGG CGA GCA GAG CCA GA3 ' 283 bp 64 1.5 A i F L M64299 322-341 bp 1388-1407 bp Upper: 5' GTC TGC TGA TGT GCC C A G CT3' Lower: 5' AC A GGG TGG GAC AGG GAG A A 3' 1086 bp 58 1.5 A 2 A S L S47609 99-118 bp 520-539 bp Upper: 5' GTG G A G CTG GCC ATC GCT GT 3' Lower: 5' GCC G C A GGT CTT CGT GGA GT 3' 441 bp 60 1.5 A 2 A F L S47609 41-61 bp 1355-1377 Upper: 5'CTG CTG AGC CTG CCC A A G TGT3' Lower: 5'CCC TTC TCT TTG GGT TAC CCG3' 1335 bp 58 4.5 A 2 B S L M91466 223-242 bp 583-602 bp Upper: 5 ' A G A CCC CCA CCA ACT ACT TT 3' Lower: 5' GCT CTT ATT CGT GAT GCC AT 3' 380 bp 53 1.5 A 3 S L M94152 319-338 bp 788-807 bp Upper: 5' A A A GCC A A C AAT ACC A C G A C 3' Lower: 5' GGA GCT GTT TTG A G A GAG CT 3' 489 bp 58 3.0 Primer sets designated as \" S L \" (short length) span only part of the coding region of the receptor. length) span the entire coding region of the receptor. Accession number of adenosine receptor sequences in the rat brain Optimal annealing temperatures used for PCR Optimal MgCh concentration used for PCR hose designated as \" F L \" (full-(j,g/ml has an A260 reading of 1, concentrations were determined spectrophotometrically using the following equation: Concentration (ug/ml) = A260 x 33 pg/ml x 100 (dilution factor). 1.2 Optimization of RT-PCR conditions Several variables were examined to determine the optimal conditions for the A i , A 2 A , A 2 B , and A 3 PCR reactions, including different M g C l 2 concentration (0.5 mM-7 mM) and annealing temperature during PCR (50-70\u00C2\u00B0C). The optimum conditions for all RT-PCR reactions performed in this study are listed in Table 3. 1.3 Tissue extraction Male Wistar rats (200-250g) were housed in light and temperature controlled rooms, with free access to food and water. Rats were anaesthetized with Somnotol\u00C2\u00AE (60 mg/kg, i.p.). The fundus, corpus, and antrum were dissected out, rinsed in sterile ice-cold saline, flash frozen in liquid nitrogen, and stored at -80\u00C2\u00B0C until total R N A was extracted. The total R N A from the gastric mucosa (containing the gastrin secreting G-cells and somatostatin secreting D-cells) was obtained as follows: The whole stomach was rinsed in sterile ice-cold saline and pinned onto a cork board with the luminal surface facing up. The mucosa was gently scraped off using a sterile glass slide, and total R N A was extracted immediately, as described in the next section. The striatum was also dissected from the brain and used as a positive control since this tissue has been shown to express the mRNA of all four adenosine receptors (Dixon et a l , 1996). 1.4 Total RNA extraction and quantification Total R N A was initially extracted using the acid guanidinium thiocyanate-phenol-chloroform extraction method, previously described by Chomczynski and Sacchi (Chomczynski and Sacchi, 1987). Since this method is fairly time consuming, total RNA extractions were subsequently performed using Trizol\u00C2\u00AE reagent (Invitrogen, Carlsbad, CA). This reagent produced higher yields of total RNA at greater speed. The choice of extraction method did not affect RT-PCR results, and both methods are described in the next section. A l l materials and reagents used in the preparation of R N A and first strand cDNA were DNase- and RNase-free. Solutions were prepared using filtered distilled water which has been treated with diethylpyrocarbonate (DEPC) (Sigma-Aldrich), as previously described (Sambrooket al., 1989a). For the guanidinium fhiocyanate-phenol-chloroform method (Chomczynski and Sacchi, 1987), tissues were ground to fine particles in a sterile mortar containing liquid nitrogen. Samples were transferred to sterile tubes, and homogenized on ice in Solution D [4 M guanidinium thiocyanate, 25 m M sodium citrate (pH 7), 0.5% sarcosyl and 0.1 M 2-mercaptoethanol] (1 ml/100 mg tissue) using the Tissumizer\u00C2\u00AE tissue homogenizer (Tekmar Co. Cincinnati, OH). The following reagents were then added, 2 M sodium acetate (0.1 ml/100 mg), water-saturated phenol (1 ml/100 mg), and chloroform-isoamyl alcohol [49:1] (0.2 ml/100 mg). The mixture was vortexed for 10 sec and incubated on ice for 15 min. The homogenate was centrifuged at 10,000 g for 20 min at 4\u00C2\u00B0C. The aqueous phase was removed and R N A was precipitated from this phase by the addition of isopropanol (1 ml/100 mg). Samples were incubated for 1 h at -20\u00C2\u00B0C, and then centrifuged at 10,000 g for 20 min at 4\u00C2\u00B0C. The resulting pellet was dissolved in Solution D (0.3 ml/100 mg) at 37\u00C2\u00B0C. An equal volume of isopropanol was added and the mixture was allowed to reprecipitate at -20\u00C2\u00B0C for 1 h. The mixture was centrifuged at 10,000 g for 10 min at 4\u00C2\u00B0C. Isopropanol was then removed and the R N A pellet was resuspended in 75% ethanol (1 ml/100 mg). After removal of the ethanol, the R N A pellet was vacuum-dried for 15 min, dissolved in DEPC-treated water (100 pl/100 mg), aliquoted, and stored at -80\u00C2\u00B0C. To extract total R N A using Trizol\u00C2\u00AE reagent, tissues were homogenized on ice in Trizol\u00C2\u00AE (1 ml/100 mg tissue) using the Tissumizer\u00C2\u00AE tissue homogenizer. Samples were incubated at room temperature for 5 min, followed by the addition of chloroform (0.2 ml/ml Trizol\u00C2\u00AE). Samples were shaken vigorously by hand for 15 sec, incubated in room temperature for 3 min and then centrifuged at 12,000 g for 15 min at 4\u00C2\u00B0C. The aqueous phase was removed, and the total R N A was precipitated from this phase by the addition of isopropanol (0.5 ml/ml Trizol\u00C2\u00AE). Samples were incubated at room temperature for 10 min and then centrifuged at 12,000 g for 10 min at 4\u00C2\u00B0C. The resulting R N A pellet was washed in 75% ethanol (lml/ml Trizol\u00C2\u00AE), and centrifuged at 7,500 g for 5 min at 4\u00C2\u00B0C. The ethanol was removed and the R N A pellet was air-dried for 10 min at room temperature. The total RNA was finally rediluted in DEPC-treated water (100 pl/ml Trizol\u00C2\u00AE), aliquoted, and stored at -80\u00C2\u00B0C. RNA purity and concentration: The total R N A purity was determined by measuring the absorbance at 260 nm (A260) and 280 nm (A280), while the concentration was estimated by measuring the A260- The spectrophotometer was calibrated with DEPC-treated water, and total R N A samples were diluted 100-fold in DEPC-treated water prior to measurement. Pure R N A gives an A26o/A28o ratio of 2 (Chomczynski and Sacchi, 1987). The A260/A280 ratio for all our samples exceeded 1.8. Since total R N A at a concentration of 40 pg/ml has an A260 of 1, total RNA concentrations were determined using the following equation: concentration (pg/ml) = A260 x 40 pg/ml x 100 (dilution factor). 1.5 Formaldehyde gel electrophoresis The integrity of the R N A samples was examined by formaldehyde gel electrophoresis to ensure that the samples were not degraded during total R N A extraction. Formaldehyde gel electrophoresis was performed using a method modified from the Molecular Cloning Laboratory Manual (Sambrook et a l , 1989a). R N A samples were prepared on ice by mixing 10 pg of total R N A (in a volume of 4.5 pi) with 10 pi deionized formamide and 2 pi 5x formaldehyde gel running buffer [0.1 M MOPS, 40 m M sodium acetate, and 5 m M EDTA]. The mixture was incubated for 15 min at 55\u00C2\u00B0C. This was followed by the addition of 1 pi ethidium bromide (1 mg/ml) and 2 pi formaldehyde gel loading buffer [50% glycerol, 1 mM EDTA (pH 8.0), 0.25 % bromphenol blue, 0.25% xylene cyanol]. Samples were electrophoresed through a 1% (w/v) agarose gel containing 5x formaldehyde gel running buffer and formaldehyde in a final concentration of lx and 2.2 M , respectively. The gel was submerged in lx gel loading buffer. The samples were loaded along with a 0.24-9.5 kb R N A ladder (Invitrogen), and run at 100 V for 1 h. Gels were viewed under U V light using the Stratagene Eagle Eye II system (La Jolla, CA). Images were acquired and saved using the Eagle Sight image analysis software (version 3.1, 1997, Stratagene). 1.6 DNase I treatment R N A samples were treated with DNase I (Amersham Pharmacia, Piscataway, NJ) to remove any residual D N A contamination that may remain in samples after total R N A extraction. DNase I treatment was performed at room temperature in lx first strand buffer [50 m M Tris-HCl (pH 8.3 at 25\u00C2\u00B0C), 75 m M KC1, 3 m M MgCl 2 ] containing 1 U DNase I/pg total RNA. The reaction was allowed to proceed for 15 min, before 1 pi of 25 m M EDTA was added to stop the reaction (Sanyal et al., 1997). Samples were then incubated for an additional 10minat65\u00C2\u00B0C. 1.7 First strand cDNA synthesis First strand cDNA was synthesized from 5 pg of DNase I treated total R N A using Superscript II RNase H - Reverse Transcriptase (Invitrogen). Total R N A (5 pg) was reverse transcribed in lx first strand buffer [50 mM Tris-HCl (pH 8.3 at 25\u00C2\u00B0C), 75 m M KC1, 3 mM MgCl 2 ] containing 200 ng random hexamers, 10 mM DTT, 0.5 m M dNTP mix, and 200 U Superscript II RNase H - Reverse Transcriptase, in a total volume of 20 pi. As a negative control for PCR, a sample was prepared using autoclaved distilled water in place of total RNA. 1.8 PCR The PCR reaction mixture for all PCR reactions consisted of 2 pi cDNA in 1 x PCR buffer [20 m M Tris-HCl (pH 8.4), 50 m M KC1], containing 0.2 m M dNTP mix, 1.5-4.5 mM M g C l 2 (see Table 3 for M g C l 2 concentrations), 100 ng forward primer, 100 ng reverse primer (see Table 3 for primer sequences), and 1 U Platinum Taq D N A polymerase (Invitrogen), in a total volume of 50 pi. For each experiment, a positive control sample containing striatal cDNA, and a negative control sample were included. The PCR was performed as follows using the Robocycler Temperature cycler (Stratagene, La Jolla, CA): cDNA was completely denatured (2 min at 94\u00C2\u00B0C) during the first step of PCR. Samples were then subjected to 30 cycles of amplification. Each cycle consisted of a 45 sec denaturation period at 94\u00C2\u00B0C, a 1 min annealing period at 53\u00C2\u00B0C - 60\u00C2\u00B0C (See Table 3), and a 1 min extension period at 72\u00C2\u00B0C. The sample was then subjected to a final extension period for 5 min at 72\u00C2\u00B0C. PCR products were visualized by gel electrophoresis as described below. 1.9 Agarose gel electrophoresis To determine the size of the amplicons produced by PCR, agarose gel electrophoresis was performed as previously described (Sambrook et a l , 1989b). The PCR products (10 pi) were mixed with 3 pi gel loading dye [0.25% bromphenol blue in 50% glycerol, 10 m M Tris, 1 mM EDTA, pH 8], and loaded on a 2.5% agarose gel (w/v) containing l x Tris acetate EDTA (TAE) buffer [40 m M Tris acetate and 1 m M EDTA (pH 8.5)] and ethidium bromide (0.5 pg/ml). The gel was placed in lx T A E buffer and run at 100 V for 45 min. Bands were visualized and photographed under U V light using the Stratagene Eagle Eye II System and the Eagle Sight Image Analysis software program. 1.10 Restriction enzyme digestion To confirm the identities of the PCR amplicons, restriction enzyme digestion was performed. PCR products generated by the A i , A 2 A , A 2 B and A3 RT-PCR reactions were concentrated, and cleaved with restrictive enzymes that were selected based on their cleavage sites. Cleavage sites within the PCR amplicons were determined using PCGENE. The following restriction enzymes were used to cleave the following PCR amplicons: Nar I and Sac I for the A , receptor; EcoRI, Mbo I, Pst I, and Rsa I for the A 2 A receptor; Ava II and Hpa II for the A 2 B receptor; Ava II and Hinf I for the A3 receptor. A l l restriction enzymes were purchased from Amersham Pharmacia (Uppsala, Sweden). To concentrate the DNA, PCR products (300 pi) were mixed with 150 pi of 7.5 M ammonium acetate and 600 pi 95% ethanol. Samples were incubated for 1 h at -20\u00C2\u00B0C and then centrifuged at 10,000 g for 20 min at 4\u00C2\u00B0C. The resulting pellet was washed in 1 ml 70% ethanol, and centrifuged at 10,000 g for 5 min at 4\u00C2\u00B0C. The ethanol was removed and the pellet was re-dissolved in 50 pi of autoclaved distilled water. The restriction enzyme digestion was performed using the buffer provided with the enzyme, according to the manufacturer's instructions. For these reactions, 5 pi of the concentrated PCR product was digested in a final volume of 10 pi. Samples were electrophoresed through a 1.5-2 % agarose gel along with a 1 kb ladder (Invitrogen), as described earlier. The Eagle Sight Image analysis software was used to determine the size of the fragments. This measurement is based on the rate of D N A migration through the agarose gel. The software produces a standard curve of migration distance vs. Log[fragment size (bp)] using the fragments of the 1 kb ladder. The size of the fragments generated by restriction enzyme digestion was interpolated by comparing the migration distance of the fragment against the standard curve. 2. Structure of the adenosine receptors To determine if the gastric A i and A 2 A receptors undergo alternative splicing and if the structure of these receptors are the same as those in the brain, the entire coding region of the gastric A i and A 2 A receptors were amplified by RT-PCR, cloned, and sequenced. In addition, part of the gastric A 2 B and A3 receptor gene sequence was also determined. These A 2 B and A3 gene sequences are located between the short length upper and lower primers listed in Table 3. This portion of the A3 receptor gene is of particular interest since its sequence can reveal whether the A 3 i variant of the A3 receptor is expressed in the stomach. 2.1 Detection of Ai and A 2 A receptor coding regions by RT-PCR Primers: The primers used for this experiment are listed in Table 3. The A i F L and A 2 A F L primers span the entire coding region of the A i and A 2 A receptor, respectively. RT-PCR: To examine whether alternative splicing occurs for the A i and A 2 A receptors, RT-PCR was performed in all gastric tissues, including the fundus, corpus, antrum and mucosa using the A i F L and A 2 A F L primers. RT-PCR was performed as previously described using the PCR conditions listed in Table 3. Fundus, corpus, antrum and mucosa R N A was used as the template for these reactions. A sample containing striatal R N A and a sample containing no template was also included as positive and negative controls, respectively. 2.2 Cloning and DNA sequence analysis of the adenosine receptors PCR products and purification: RT-PCR was performed for all four receptors using rat gastric mucosal R N A as the template and the A i F L , A 2 A F L , A 2 B S L , and A3SL primers. The PCR products were concentrated using the QIAquick PCR purification kit (Qiagen, Mississauga, ON). According to manufacturer's instructions, 1.5 ml Buffer PB was mixed with 300 pi of the PCR product. The D N A was bound to a QIAquick column by centrifugation and washed with PE buffer. The D N A was finally eluted with 50 pi of autoclaved distilled water. The purified D N A was electrophoresed through a 2% agarose gel, as described earlier, and extracted from the gel using the QIAquick gel extraction kit (Qiagen), according to the manufacturer's instructions. The fragment was excised from the gel using a sterile razor blade and weighed. Buffer QX1 (300 jal/100 mg gel) was added, and the mixture was vortexed for 30 sec. QIAEX II buffer (5 pi) was then added and the sample was incubated at 50\u00C2\u00B0C for 10 min, and vortexed every 2 min, and the sample was then centrifuged at 10,000 g for 30 sec. The pellet was washed with Buffer QX1 (500 pi), followed by two washes in Buffer PE (500 pi). The pellet was air dried for 10-15 min, and rediluted in autoclaved distilled water. Ligation and transformation: The purified adenosine receptor transcripts were ligated into the pGEM-T vector (Promega, Madison, WI) by combining 5 pi of the purified A , F L , A 2 A F L , A 2 B S L or A 3 S L PCR product with 1 pi lOx T4 D N A Ligase Buffer, 1 pi pGEM-T vector (50 ng), 1 pi T4 D N A ligase and 2 pi autoclaved distilled water. The mixture was incubated overnight at 4\u00C2\u00B0C, and then heated to 70\u00C2\u00B0C for 10 min, and cooled to room temperature. DHot-competent E. coli cells (Invitrogen) were transformed with the ligated PCR product/pGEM-T vector by combining 2 pi of this sample with 50 pi of cells. The mixture was incubated on ice for 20 min, heat shocked at 37\u00C2\u00B0C for 50 sec, and placed on ice. The volume of Luria-Bertani (LB) broth [0.1% NaCl containing tryptone (lOg/L) and yeast extract (5g/L)] added to cells depended on the size of the PCR products, which were transformed into the cells. Cells containing the A[FL, A 2 A F L , A 2 B S L , and A 3 S L transcript were combined with 900 pi, 1 ml, 1.5 ml, and 1.5 ml of L B , respectively. Cells were incubated for 1 h at 37\u00C2\u00B0C, and then plated on L B plates containing ampicillin (100 pg/ml), isopropyl P-D-thiogalactopyranoside (0.5 mM) and 5-bromo-4-chloro-3-indolyl-B-D-galactoside (80 pg/ml). Cells were grown overnight at 37\u00C2\u00B0C. Colonies containing inserts appear white since the inserts interrupts the lacZ gene in the pGEM-T vector. White colonies were screened by RT-PCR. Colonies containing the correct insert were grown in 5 ml of L B broth containing ampicillin (100 pg/ml). Plasmid D N A was purified from these cells using the QIAprep Miniprep kit (Qiagen). The sample was centrifuged at 3,000 rpm for 5 min at 4\u00C2\u00B0C to pellet the cells. Cells were resuspended in 250 pi Buffer PI , followed by the addition of 250 pi of Buffer P2. Cells were allowed to lyse for 5 min prior to the addition of 350 pi N3 Buffer. The sample was then centrifuged at 10,000 g for 10 min at 4\u00C2\u00B0C. The supernatant was applied to a QIAprep column and centrifuged for 30 sec. The column was washed with 750 pi PE buffer, and plasmid D N A was eluted from the column with 50 pi of Buffer EB [10 m M Tris-Cl (pH 8.5)] DNA sequence analysis: The D N A sequences were determined at the NAPS Unit (UBC) using both the T7 and SP6 primers. This procedure was successful in obtaining the A i F l , A2BSL and A3SL sequences. However, due to the length of the A 2 A F L sequence, 2 additional primers located within the A 2 A F L transcript were required to complete the sequence analysis. These primers were based on the rat brain A 2 A receptor gene (accession #S47609). Their sequences are as follows: 5' TTG TCC TGG TCC T C A CGC 3' (position 313-330 bp) and 5' A G G GCC GGG T G A CCT GTC 3' (position 541-558 bp). The sequence of the rat brain A i and A 2 A receptor coding regions, and the partial cDNA sequence of the A 2 B and A3 receptors were aligned with the published sequences of these receptors in the rat brain using the \"University of Southern California (USC) Sequence Alignment Server\" (www-hto.usc.edu/software). These sequences were submitted to GenBank, and assigned accession numbers (see Results). 3. Quantification of gastric Ai and A 2 A receptor gene expression 3.1 Quantification by competitive RT-PCR Competitive RT-PCR was performed to quantify the level of adenosine A\ and A 2 A receptor gene expression in various regions of the stomach, including the fundus, corpus, antrum, and mucosa. The general steps for measuring A i and A 2 A receptor gene expression using competitive RT-PCR are as follows: 1) synthesize, purify, and quantify the A i and A 2 A receptor R N A competitors; 2) perform the competitive RT-PCR assay by preparing samples containing a fixed amount of tissue total R N A and various amounts of competitor R N A as the template; 3) separate and visualize the PCR products by agarose gel electrophoresis; 4) determine the relative intensities of the tissue template and competitor bands; 5) calculate the tissue template level based on the relative intensity of the competitor vs. the template band. The intensities of both bands are approximately equal when the concentration of the template is equal to the concentration of the competitor. Each step is described in detail below. 3.1.1 RNA competitor design and synthesis Previous experiments have shown that the reverse transcription step can vary considerably between assays (Ferre, 1992). Thus, to control for the efficiency of the reverse transcription step, R N A competitors were synthesized for this experiment instead of D N A competitors. The RNA competitors were synthesized by deleting a small portion (< 15%) of the adenosine receptor transcripts. The deletion allows for differentiation between the template and competitor bands when PCR products are visualized by electrophoresis. The small deletion also controls for differences in PCR amplification efficiency that may occur due to differences in R N A length and secondary structure. The adenosine receptor RNA competitors were synthesized as described in the following section and in Figure 7. The general method has previously been described by Tang et al. (1996). Vectors containing the A i and A 2 A competitor: To synthesize vectors containing the A] receptor competitor, RT-PCR was performed using the A jFL primer set (see Table 3). The RT-PCR products were concentrated using the QIAquick PCR purification kit, and gel purified using the QIAquick gel extraction kit, as described earlier. The A i competitor was synthesized by deleting a 136 bp fragment from the A i receptor template to produce a competitor containing 950 bp. To produce this deletion, the A i template was digested with Bpm I (New England BioLabs, Beverly, MA) , as directed by the manufacturer. Three fragments (515 bp, 435 bp and 136 bp) were formed. The sample was electrophoresed through a 2% agarose gel as previously described. The two fragments containing the 5' and 3' primer sites (515 bp and 435 bp) were excised from the gel, purified using the QIAquick gel extraction kit, and religated with the pGEM-T vector using T4 ligase. The A 2 A competitor was synthesized in a similar manner. RT-PCR was performed using the A 2 A S L primers (see Table 3). The A 2 A competitor was synthesized by deleting a 57 bp fragment from the A 2 A template to produce a 384 bp competitor. The A 2 A template was digested with Tsp509 I (New England BioLabs), according to manufacturer's instructions, to produce 3 fragments (286 bp, 98 bp, and 57 bp). The two fragments containing the primer sites (286 bp and 98 bp) were gel-purified and religated with the pGEM-T vector. Transformation and in vitro Transcription: Vectors containing the modified A i , and A 2 A inserts were transformed into D H a competent cells, as described earlier. Bacterial colonies were screened by PCR to ensure that the colonies that were chosen contain the Cleavage * / sites ^ 5' primer 3' primer 1. Digest with enzyme which cleaves -100 bp apart / 1 \ 5' primer A discard B 2. separate fragments and 3 primer purity by gel electrophoresis 5' primer A B 3' primer 3. Re-ligate A+B 1 4. Clone modified adenosine receptor mRNA 5. Linearize and purify plasmid containing the modified adenosine receptor transcript 6. Synthesize RNA competitors by in vitro transcription, purify, and quantify Fig. 7. Design and synthesis of the adenosine A, and A 2 A receptor RNA competitors. A) The general protocol used for the design and synthesis of A , and A 2 A competitors. B) Agarose gels showing PCR amplicons generated using corpus tissue total RNA as the template (lanes 2; A , , 1086 bp; A 2 A , 441 bp) or A, and A 2 A RNA competitor as the template (lanes 3; A , , 950 bp; A 2 A , 384 bp). A 100 kb ladder is shown on lanes 1. The competitor and tissue template bands can be differentiated easily by their size. modified adenosine receptor transcripts. The modified A i and A 2 A receptor transcripts generated PCR amplicons which were 950 and 382 bp in length, respectively (see Fig. 7B). Plasmids from the chosen colonies were isolated using the QIAprep Miniprep kit and linearized by Not I (Amersham Pharmacia) digestion, according to manufacturer's instructions. Complete digestion was confirmed by agarose gel electrophoresis. The linearized plasmids were then purified and concentrated as described earlier. In vitro transcription was performed using the Riboprobe in vitro Transcription System - T7 (Promega). To synthesize the competitor RNA, a reaction mixture (20 pi) containing lx Transcription Optimized buffer [40 m M Tris-HCl (pH 7.9), 6 m M M g C l 2 , 2 mM spermidine, 10 m M NaCl], 10 m M DTT, 20 U RNasin ribonuclease inhibitor, 0.5 m M rNTPs, 0.2 pg linearized template, and 20 U T7 R N A polymerase, was incubated at 37\u00C2\u00B0C for 1 h. The template D N A was digested with 0.2 U of RQ1 RNase-free DNase. The RNA competitors were purified using the RNeasy mini kit (Qiagen). The volume of the sample was increased to 100 pi with DEPC-treated water. Buffer RLT (350 pi) and ethanol (250 pi) was then added. The sample was applied to a RNeasy mini spin column and centrifuged for 15 sec at 8,000 g. The column was washed twice with 500 pi of Buffer RPE, and the RNA competitors were eluted from the spin column with 30 pi of DEPC-treated water. The purity and concentration of the R N A competitors were determined spectrophotometrically, as described earlier. The integrity of the competitors was examined by gel electrophoresis through a formaldehyde gel. R N A competitors were then serially diluted from 0.1 pmol to 0.01 amol, aliquoted, and stored at -80\u00C2\u00B0C. R N A competitors were thawed only once prior to use to avoid damage caused by multiple freeze-thaw cycles. 3.1.2 Competitive RT-PCR assay Total RNA extraction: Tissues were dissected from various regions of the stomach, including the fundus, corpus, antrum and mucosa. As a positive control, the level of A i and A 2 A receptor gene expression was also examined in the striatum. This tissue has been shown to express extremely high levels of A 2 A receptor mRNA and relatively high levels of A t receptor mRNA (Dixon et a l , 1996). Total RNA was extracted using Trizol\u00C2\u00AE Reagent, and the purity and concentration of the RNA were determined by measuring the absorbance of A260/A280, and A 2 6o, as described earlier. The A260/A280 ratio for all samples exceeded 1.8. First strand cDNA synthesis: First strand cDNA was synthesized using Superscript II as described earlier with the following modification. Various amounts of A t or A 2 A receptor competitor R N A (from 0.01 amol to 1 pmol) and a fixed amount of total RNA (2.5 pg) were used as the template for the reaction. The cDNA that was synthesized was then used as the template for PCR. PCR: PCR was performed using the A i F L and A2ASL primer sets listed in Table 3. The A i and A2A receptor competitive PCR reactions mixture (50 pi total) contained 0.5 pi and 2.0 pi of cDNA, respectively, in lx PCR buffer [20 m M Tris-HCl (pH 8.4), 50 mM KC1] with 0.2 mM dNTP mix, 1.5-4.5 m M M g C l 2 (see Table 3), 100 ng forward primer, 100 ng reverse primer, and 1 U Platinum Taq D N A Polymerase. PCR was performed using the Robocycler Temperature cycler as described previously. However, the cycles of amplification were limited to 30 cycles to avoid reaching the plateau phase. Reaching this phase of the reaction has previously been shown to introduce errors into the R N A measurements (Wang et a l , 1989; Freeman et a l , 1999). The PCR products, containing the competitor and template amplicons, were separated by electrophoresis through a 2.5% agarose gel containing ethidium bromide, as described earlier. Bands were visualized and photographed under U V light using the Eagle Eye II System. To determine whether altering the initial amount of cDNA in the PCR mixture affects the final A\ and A 2 A mRNA measurements, an equal volume of undiluted, 2x , 4x, or 8x diluted cDNA was used as the template for these PCR reactions. 3.1.3 Competitive RT-PCR data analysis During the acquisition of gel images, special care was taken not to allow any bands to reach saturation density. The saturation of the bands can be detected by the Eagle Sight Image analysis software program. The images were saved, and band densities were determined by densiometric analysis using the Eagle Sight image analysis software (see Figure 8A). Analysis of competitive RT-PCR images has previously been described (Piatak et al., 1993; Auboeuf and Vidal, 1997). The competitor band densities were corrected for inherent variation in band density resulting from the difference between template and competitor PCR product length. The correction factor is determined by dividing the size of the tissue template amplicon by the size of the competitor amplicon. Competitor band densities, therefore, were multiplied by the following correction factors: 1.14 (1086 bp/950 bp) for the A i competitor and 1.15 (441 bp/383 bp) for the A 2 A receptor competitor. Levels of A i and A 2 A receptor mRNA in each tissue sample were determined by plotting the Log [density ratio (corrected competitor band/template band)] against the Log (initial competitor concentration). Since the amount of A i and A 2 A template mRNA is equal to the initial Template band density -A, Template band \" A: Competitor band-Competitor -band density Initial competitor concentration (amol) 25 10 5 2.5 1 42544 65255 69654 61349 71169 111215 62938 27463 19595 5594 2.99 1.10 0.45 0.36 0.09 Corrected density ratio: competitor/template B 0) *\u00E2\u0080\u00A2> ro a. E CD o. E o o CO i-'35 c \u00E2\u0080\u00A2a o 0.6 0.4 0.2 0 -0 .2 -0.4 -0 .6 -0 .8 -1 -1 .2 - y = 1.04x -1 > ^ 0 0 .25 0 .5 0.75/^ m >r Log [tis \u00E2\u0080\u00A2 1 1.25 1 sue A 1 concentration] R2 = 0.98 Log [initial A 1 competitor concentration (amol)] Fig. 8. Analysis of competitive RT-PCR data. A) Results of an A, receptor competitive RT-PCR assay performed using various amounts of Kx competitor and a fixed amount (2.5 pg) of corpus tissue total RNA. Band densities were acquired using the Eagle Sight software. The competitor band density was multiplied by a correction factor to account for the difference in band density resulting from the smaller size of the competitor (see Methods). B) The Log density ratio (competitor band/ template band) was plotted against the Log (initial competitor concentration). The Log density ratio of the (competitor band/template band) is equal to zero when the template and corrected competitor band densities are equivalent. Thus, the A t receptor mRNA concentration in the tissue is determined using the x-intercept of this graph. amount of competitor R N A when the template and corrected competitor band are of equal density, the initial amount of tissue A i and A 2 A template was determined at the x-intercept (See Figure 8B). Data were expressed in units of amol of Aj or A 2 A receptor mRNA/pg total RNA. These values were then converted to the number of copies of A i or A 2 A receptor mRNA transcript/pg total RNA. Statistical significance was determined using Student's unpaired t-test, and performed using GraphPad Prism; P < 0.05 was considered significant. 3.2 Quantification by Real-Time RT-PCR The gene expression of the A i and A 2 A receptor were also quantified using Real-Time RT-PCR when the ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, CA) became available. Absolute quantification of gastric A i and A 2 A receptor gene expression was measured using a standard curve generated by known amounts of A i and A 2 A receptor mRNA. Standard curve samples were prepared using in vitro transcribed A i or A 2 A receptor RNA. A two-step Real-Time PCR assay was performed. This technique is described in Fig. 9, and is based on the 5' nuclease activity of Taq D N A polymerase (Heid et al., 1996; Lockey et al., 1998). A double-labeled fluorogenic probe containing a reporter dye such as 6-carboxyfluorescein (FAM) and a quencher dye such as 6-carboxytetramethylrhodamine (TAMRA) is included in each PCR reaction. The probe hybridizes to the template at a position flanked by the forward and reverse primers. During PCR amplification, the hybridized probe is cleaved by the 5' nuclease activity of Taq D N A polymerase. When the Fig. 9. Cleavage of the fluorogenic probe during Real-Time RT-PCR. During the extension phase of the Real-Time RT-PCR reaction, the primers and probes are bound to the template DNA. Taq polymerase synthesizes the new DNA strand (A), and encounters the probe (B). As Taq polymerase continues to synthesize the new strand, the 5' to 3' exonuclease activity of this enzyme degrades the probe. When the probe is intact, the energy emitted by the reporter dye, F A M is absorbed by the quencher dye, TAMRA, due to their close proximity. After the probe is degraded, F A M and TAMRA separate and the energy emitted by F A M is no longer quenched by TAMRA (C) and is detected by the ABI prism 7700 Sequence Detection System. probe is intact, the quencher dye will suppress reporter fluorescence due to Forster type energy transfer. Upon cleavage of the probe, reporter emissions are no longer quenched and are detected by the ABI Prism Sequence Detector 7700. The methods used to synthesize the A i and A 2 A receptor primers and probes are described in detail below. 3.2.1 Design and synthesis of primers and probes Primers and probes were designed using the software program Primer Express (version 1.0, Applied Biosystems) according to the following guidelines. The guanine (G) and cytosine (C) content of the primers and probes were kept between 30-80% and > 4 runs of an identical nucleotide were avoided. The melting temperatures (Tra) of the primers were kept between 58-60\u00C2\u00B0C, and each primer contained < 2 G/C bases at the 3' end. The Tm of the probes was 10\u00C2\u00B0C higher than that of the primers. Probe sequences contained more C than G, and no G at the 5' end. For both probes, the reporter dye, F A M was linked to the 5' end and the quencher dye, T A M R A was linked to the 3' end. The fluorogenic probes were synthesized by Synthegen, L L C (Houston, Texas) and the primers were synthesized by the NAPS Unit at UBC. The sequences of the A i and A 2 A primers and probes are listed in Table 4. 3.2.2 Synthesis of A ! and A 2 A receptor R N A standards R N A transcripts expressing the entire coding region of the adenosine Aj and A 2 A receptors were used as the standards for quantification during Real-Time RT-PCR. To synthesize these standards, plasmids containing the A i F L and A 2 A F L transcripts were used. Table 4: A i and A 2 A receptor Real-Time RT-PCR assay: primers and probes Rat A i Adenosine Receptor Position Length Tm %GC Sequence Forward 963 22 59 59 5' C G G T G A C C C C C A G A A G T A C T A C 3' Probe 989 26 70 58 5' C A G C G A C T T G G C G A T C T T C A G C T C C T 3' Reverse 1039 21 59 52 5' G G G C A A A G A G G A A G A G G A T G A 3 ' Amplicon 962-1039 77 84 60 Rat A 2 A Adenosine Receptor2 Position Length Tm %GC Sequence Forward 910 22 58 55 5' A C C C C T T C A T C T A C G C C T A C A G 3' Probe 936 22 69 68 5' C G G G A G T T C C G C C A G A C C T T C C 3' Reverse 978 20 59 55 5' CGTGGGTTCGGATGATCTTC 3' Amplicon 910-978 69 84 59 Based on A i receptor sequence in the rat brain; Accession number: M64299 Based on A 2 A receptor sequence in the rat brain; Accession number: S47609 Plasmids were generated by our previous cloning experiments, described in section 2.2. The orientation of the adenosine A i and A 2 A receptor insertions in the plasmid was determined to examine whether the 5' or 3' strand of the insertion would be synthesized by T7 polymerase during in vitro transcription. To determine the orientation of the A\ and A 2 A insertions, a Sac I digestion and Nco I digestion were performed, respectively. These restriction enzymes were chosen for the position of their cleavage sites, which allows the differentiation between the two orientations. Only the plasmids with insertions in Orientation I (see Fig. 10) were selected. In this orientation, T7 R N A polymerase synthesizes the 5' strand of the cloned inserts during in vitro transcription. The selected plasmids were linearized by Not I digestion. This enzyme does not cleave the A i or A 2 A insert, but cleaves at a site located after the cloned insert. The linearized plasmids were purified using the Qiagen miniprep kit, as described earlier. Run-off transcripts were synthesized using the Riboprobe in vitro transcription kit and T7 R N A polymerase, as described previously in section III. RNA standards were treated with DNase I, and then purified using the Qiagen RNeasy mini Kit. The quality of the RNA standards was examined by gel electrophoresis, as described earlier. The concentrations of the R N A standards were estimated spectrophotometrically, and then accurately determined using the RiboGreen Reagent Quantitation kit (Molecular Probes, Eugene, OR). Serial dilutions of the A i and A 2 A R N A standards were made. R N A concentrations were determined in lx TE buffer [10 m M Tris-HCl, 1 m M EDTA (pH 7.5) in DEPC-treated water] containing RiboGreen R N A quantitation reagent (0.025 pi /ml), a sensitive fluorescent nucleic acid stain. When bound to RNA, RiboGreen reagent has an Sac I (Pos. 63 of A , insert in Orientation I) A, Insertion (1086 bp) Sac I (Pos. 63 of A , insert in Orientation II) Sac I (Pos. 94 of pGEM-T vector) pGEM-T Vector (3003 bp) B . Orientation I Orientation II Strand synthesized by T7 Polymerase 5' strand 3' strand Molecular Weight of Strand synthesized 368.11 kD 373.44 kD S a c I Cleavage Sites 1 & 3 2 & 3 Fragments generated (bp) 3270 & 818 3737 & 351 Nco I (Pos. 73 of A M insert in Orientation I) Au Insertion (1335 bp) Nco I (Pos. 73 of A ^ insert in Orientation II) Nco I (Pos. 37 of pGEM-T vector) pGEM-T Vector (3003 bp) Orientation I Orientation II Strand synthesized by T7 Polymerase 5' strand 3' strand Molecular Weight of Strand synthesized 450.16 kD 453.10 kD Nco I Cleavage Sites 1 & 3 2 & 3 Fragments generated (bp) 4291 & 47 3019 & 1319 Fig. 10. Orientation of A , and A 2 A receptor transcript insertion into pGEM-T vector. Plasmids containing the A, (A) and A 2 A (B) transcripts were cleaved with restriction enzymes, Sac I and Nco I, respectively. When the transcript is inserted in one orientation (orientation I), plasmids were cleaved at sites 1 and 3, and when inserted in the other orientation (orientation II), plasmids were cleaved at sites 2 and 3. The size of the fragments generated by restriction enzyme digestion are shown in the tables. excitation wavelength of 480 nm and emission wavelength of 520 nm. The A\ and A 2 A R N A standards were quantified by measuring the fluorescence against a standard curve composed of samples containing known amounts of ribosomal R N A standard (16S and 23S rRNA from E. coli cells; provided by the manufacturer). The fluorescence measurements were performed using the FL600 Microplate Fluorescence reader (Biotek Inc., Winooski, VT), and R N A concentrations (ng/ml) were determined using the KC4 Kineticalc Software (version 2.6, Biotek Inc.). R N A concentrations were converted from ng/ml to copies/ml using the molecular weight of the standards: A i R N A standard: 368.11 kD; A 2 A R N A standard: 450.16 kD. R N A standards were serially diluted to 103 - 10 1 2 copies/pl in RNase-free water, aliquoted, and stored at -80\u00C2\u00B0C. Standards were thawed only once prior to use. 3.2.3 Two step Real-Time RT-PCR assay Real-Time RT-PCR was performed to measure the level of adenosine A i and A 2 A receptor mRNA in the fundus, corpus, antrum, mucosa and striatum. Total R N A was extracted using Trizol\u00C2\u00AE reagent, as described in section I, and quantified using the RiboGreen R N A Quantitation Kit. Total R N A was treated with DNase I. Two-step Real-Time PCR was performed using Superscript II RNase H-Reverse transcriptase, and the TaqMan PCR Core Reagents Kit (Applied Biosystems, Foster City, CA) as described below. Step 1 - Reverse transcription: According to the manufacturer's instructions, 1 pg of DNase I-treated tissue R N A was reverse transcribed in a total volume of 10 pi containing 200 ng random hexamers, 20 U RNAguard RNase inhibitor, lx first strand buffer, 10 mM DTT, 0.5 mM dNTP mix, and 100 U Superscript II RNase H - Reverse Transcriptase. Various amounts of R N A standard and a negative control sample containing DNase I-treated RNase-free water in place of template, were reverse transcribed simultaneously. The latter was subsequently used as the template for the negative control during Real-Time PCR. Step 2 - PCR: The standard curve for each Real-Time PCR assay was composed of at least 6 concentrations of either the A i or A 2 A receptor standard. Each assay also contained a negative control sample containing no template. A l l reactions were performed in triplicate. The optimization of the A i and A 2 A Real-Time RT-PCR assay conditions are described in the following section (3.2.5.). The PCR reaction consisted of a final volume of 25 pi reaction mixture containing lx TaqMan Buffer A , 200 p M of each dATP, dCTP, and dGTP, 400 p M dUTP, 0.01 U/pl AmpErase uracil-N-glycosylase (UNG) and 0.025 U/pl AmpliTaq Gold D N A Polymerase from the TaqMan PCR Core Kit. This reaction mixture also contained 0.5 pi of tissue or standard cDNA, or 0.5 pi of the control sample (which does not contain template), 100 nM probe, various concentrations of forward and reverse primer (Ai receptor assay contains 300 nM of each, A 2 A receptor assay contains 100 nM of each) and M g C l 2 (Ai receptor assay contains 7.5 mM, A 2 A receptor assay contains 4.5 mM). The reaction was performed using the A B I Prism 7700 Sequence Detector (Applied Biosystems) with the following cycling parameters: a 2 min hold at 50\u00C2\u00B0C for U N G incubation, a 10 min hold at 95\u00C2\u00B0C for AmpliTaq Gold activation, followed by 40 cycles of amplification consisting of a 15 s denaturation step at 95\u00C2\u00B0C and a 1 min anneal/extend period at 60\u00C2\u00B0C. 3.2.4 Real-Time RT-PCR data collection and analysis The data was collected by the Sequence Detection Software (version 1.6.3, Applied Biosystems) during each cycle of PCR amplification. At the end of the PCR, the data was analyzed by the software program, and an amplification plot was generated. The amplification plot shows the normalized reporter emissions (Rn) vs. cycle number. Reporter emissions were normalized with the passive reference dye, ROX, which was present in the TaqMan Buffer A. The threshold cycle (Cr), where the software detects an increase in fluorescence associated with exponential growth, was determined using the first 15 cycles to represent the baseline. The threshold cycle (Cr) of each reaction occurred where the Rn exceeded the baseline Rn by the value of the threshold. The average Rn and standard deviation of Rn in the first 15 cycles were determined. To ensure that the change in Rn is associated with exponential growth of the amplicon during quantification, the threshold was determined by multiplying the standard deviation of the first 15 cycles by a factor of 10. To quantify the levels of adenosine A i and A 2 A receptor mRNA, standard curves of CT VS. Log (initial A i or A 2 A receptor RNA standard concentrations) were generated. The initial concentration of each unknown sample was determined by interpolation using the C T value determined by the assay. The correlation coefficient of each standard curve was >0.95. The threshold cycle of the control samples containing no template exceeded 40 cycles in every assay, indicating the absence of D N A contamination. For the absolute quantification of adenosine A i and A 2 A receptor mRNA levels, measurements were not normalized using the housekeeping genes, and the results were expressed as copies of mRNA per pg total RNA. Statistical significance was determined using GraphPad Prism and the two-tailed unpaired Student's t-test; P < 0.05 was considered significant. 3.2.5 Optimization of Real-Time RT-PCR Real-Time RT-PCR assays were optimized by adjusting the primer, probe, and M g C l 2 concentrations. RT-PCR samples containing 100, 200, or 300 nM of probe and either 100, 200, or 300 nM of primer were prepared. A i or A 2 A R N A (1x10 s copies) was used as the template for these reactions. The conditions resulting in a fairly low CT were selected for Real-Time RT-PCR. Due to the expense of the probe, the lowest acceptable concentration of this component was selected (see Table 5). Various M g C l 2 concentrations (3.5, 4.5, 5.5, 6.5, and 7.5 nM) were then subsequently tested using these primer and probe concentrations (see Table 6). The following optimized A i and A 2 A Real-Time RT-PCR assay conditions were established. The A i receptor assay contained 100 nM probe, 300 nM primers, and 7.5 nM M g C l 2 , while the A 2 A receptor assay contained 100 nM probe, 100 nM primers, and 4.5 nM M g C l 2 . III. Regulation of adenosine receptor gene expression and function in altered physiological states of the stomach The effect of fasting and omeprazole treatment on A i and A 2 A receptor mRNA levels was examined to determine if changes in the physiological state of the stomach alter adenosine receptor gene expression. The gene expression of gastrin and somatostatin was also measured to determine if changes in adenosine receptor gene expression occurred with changes in gastric peptide expression. The effect of omeprazole on adenosine agonist-induced changes in IRG and SLI release was also examined to further determine if changes in adenosine receptor gene expression may result in changes in adenosine receptor function. Table 5: Optimization of primer and probe concentrations for Real-Time RT-PCR Probe Concentration (nM) Primer Concentration (nM) A i Assay: Cj A 2 A Assay: C T ' 100 40.0 29.1 100 200 40.0 29.4 300 35.5 30.1 100 40.0 28.9 200 200 35.1 29.1 300 40.0 29.9 100 40.0 29.5 300 200 40.0 29.2 300 40.0 29.1 1 The threshold cycle (Cj) was obtained using 1x10s copies of A i or A 2 A R N A standard as the template for Real Time RT-PCR. Real-Time RT-PCR was performed as described in the methods section. Table 6: Optimization of MgCl 2 concentrations for Real-Time RT-PCR MgCh Concentration (nM) A i Assay: C j 1 A 2 A Assay: Cr 1 3.5 40.0 28.6 4.5 40.0 27.6 5.5 40.0 28.2 6.5 40.0 27.6 7.5 30.3 27.6 1 The threshold cycle (Cj) was obtained using 1x10s copies of Aj or A 2 A R N A standard as the template for Real Time RT-PCR. Real-Time RT-PCR was performed as described in the methods section. 1. Quantification of Ai and A 2 A receptor, somatostatin and gastrin gene expression 1.1 Animal treatment Fasting: Male Wistar rats (250-300g) housed in light and temperature-controled rooms with free access to water were deprived of food for 24 or 36 h. Control animals were allowed free access to both food and water. Rats were anaesthetized with Somnotol\u00C2\u00AE (60 mg/kg i.p.), and tissues were extracted between 11 a.m. to 12 p.m. for all groups. Omeprazole treatment: Male Wistar rats (250-270g) housed in light and temperature-controlled rooms with free access to food and water were used for this experiment. Omeprazole (AstraZeneca, Molndal, Sweden) was prepared for administration according to the manufacturer's instructions. Omeprazole (50 pmol/ml) was thoroughly dispersed in vehicle using the Tissumizer\u00C2\u00AE tissue homogenizer. The vehicle consisted of 0.25% Methocel\u00C2\u00AE (Dow, Midland, Michigan) containing 2mg/ml N a H C 0 3 (pH 9.0). The drug was aliquoted, frozen, and stored at -20\u00C2\u00B0C. The drug was thawed overnight at 4\u00C2\u00B0C and brought to room temperature prior to administration. A new aliquot of drug was used daily to prevent degradation due to repeated freezing and thawing. Test groups were treated by gavage with omeprazole at a dose of 400 pmol/kg once daily between 9 and 10 a.m. for either 1 or 3 days. This omeprazole concentration and length of treatment were selected since gastric acid secretion is decreased by up to 80% after 1 day of treatment at 400 pmol/kg (Larsson et al., 1988; Lee et al., 1992), and steady state gastric acid inhibition is achieved after 3 days of treatment with the same dose (Carlsson et al., 1986). Control groups were treated similarly with vehicle. Animals were sacrificed 24 h after the last treatment of omeprazole. Rats were anaesthetized with Somnotol\u00C2\u00AE(60 mg/kg, i.p.), and tissue extraction occurred between 10 a.m. to 11 a.m. 1.2 Tissue extraction The corporeal mucosa, corporeal muscle, and antrum, were dissected. These regions were selected because the corporeal mucosa contains somatostatin-secreting D-cells, the corporeal muscle contains somatostatin-releasing nerve fibers, and the antrum contains D-cells, gastrin-secreting G-cells, and somatostatin-releasing nerve fibers. Care was taken to avoid inclusion of tissue at the antro-corpus border. The corpus tissue was rinsed in ice-cold saline, and the mucosa was removed from the corpus with gentle scraping using a sterile glass slide. The mucosa was then rinsed off the slide with Trizol\u00C2\u00AE reagent. Total R N A was extracted immediately from the corporeal mucosa as described in previous section. The corporeal muscle was flash frozen in liquid nitrogen and stored at -80\u00C2\u00B0C until RNA extraction. Since the antrum is a relatively small tissue, total R N A was extracted from the whole antrum to ensure consistency between samples. The antral tissue was rinsed in sterile ice-cold saline, flash frozen in liquid nitrogen, and stored at -80\u00C2\u00B0C until R N A extraction. 1.3 Sample preparation Total R N A was extracted using Trizol Reagent\u00C2\u00AE, and concentrations were determined using the RiboGreen Quantitation Kit, as described earlier. Total R N A samples were then treated with DNase I and reverse transcribed as described in section II (3.2). 1.4 Real-Time RT-PCR The Real-Time RT-PCR procedure established in the present study was employed to measure adenosine A i and A 2 A receptor mRNA levels. For these experiments, glyceraldehyde-3'-phosphate dehydrogenase (GAPDH), 18S ribosomal R N A (rRNA), somatostatin and gastrin gene expression levels were also measured. GAPDH and 18S rRNA gene expression: G A P D H mRNA and 18S rRNA levels were used as endogenous controls, and were measured using the Rodent G A P D H Control Reagents Kit (Applied Biosystems) and the TaqMan ribosomal R N A control reagents kit (Applied Biosystems), respectively. G A P D H mRNA and 18S rRNA levels were determined using the protocol described in the PE Applied Biosystems User Bulletin #2. A relative standard curve was constructed'using the Rodent Control R N A Standard (50 ng/pl) provided with the Rodent G A P D H Control Reagents Kit. Rodent Control R N A (100 ng) was reverse transcribed into 10 pi of cDNA using Superscript II RNase H - Reverse Transcriptase. This sample was designated as the 10 ng/pl sample since 10 ng of Rodent control total RNA was used to produce 1 pi of cDNA. Reverse transcription was performed as described in section II (3.2). The 10 ng/pl sample was diluted to 7.5, 5, 2.5, 1, 0.5, and 0.1 ng/pl, and these samples were used to construct the relative standard curve for the Real-Time RT-PCR assay. The 18S and G A P D H Real-Time RT-PCR assays were performed according to the manufacturer's instruction. Data were collected and analyzed as described in section II (3.2.4). Since fasting did not affect 18S rRNA levels, this value was subsequently used to normalize A\ receptor, A 2 A receptor, somatostatin and gastrin gene expression for possible differences arising from variations in total RNA extraction efficiency, first strand cDNA synthesis efficiency, and total RNA and cDNA degradation. Gastrin and somatostatin gene expression: Relative gastrin and somatostatin gene expression levels were measured using a sample of rat antrum total R N A to produce the standard curve. Antral total RNA (1 pg) was reverse transcribed into 10 pi of cDNA using Superscript II RNase H - Reverse Transcriptase. This sample was designated as the 100 ng/pl sample since 100 ng total R N A was reverse transcribed to produce 1 pi of cDNA. This sample was diluted to various concentrations to produce a relative standard curve. Standard sample concentrations ranged from 0.1 ng/pl to 100 ng/pl The primers and probes used to measure somatostatin and gastrin gene expression were designed using Primer Express and are listed in Table 7. The same guidelines which were used to design the A i and A 2 A receptor primers and probes were used here (see Section 3.2.1). The primers and probes were synthesized by the NAPS Unit at U B C , and by Synthegen, respectively. Gastrin and somatostatin mRNA levels were measured by performing a two-step Real-Time RT-PCR assay as described in section II (3.2.3). The primer, probe and MgCl2 concentrations used for these assays were optimized according to the method described in section II (3.2.5). For both assays, the primer and probe concentrations were found to be 100 nM, and the M g C l 2 concentration was found to be 7.5 mM. 1.5 Data collection and analysis Data were collected as described in section 3.2.4. To compare gene expression levels between the test and control animals, measurements were first normalized using the endogenous housekeeping-gene (18S rRNA) expression levels. This was performed according to the procedures described in the Applied Biosystems User bulletin #2: The Table 7: Gastrin and somatostatin Real-Time RT-PCR assay: primers and probes Rat Gastrin1 Position Length Tm %GC Sequence Forward 6 22 61 50 5 ' T G C C A C A A C A G T T A A C C G T T C C 3 ' Probe 80 28 69 57 5 T G C T G G C T C T A G C T A C C T T C T C G G A A G C 3 ' Reverse 129 20 61 60 5' C T G G G A G C G A G G T T T C C A A G 3 ' Amplicon 6-129 124 82 54 Rat Somatosl tatin2 Position Length Tm %GC Sequence Forward 234 21 59 52 5' TG A G C C C A A C C A G A C A G A G A A 3 ' Probe 260 23 69 65 5 ' C C C T G G A G C C T G A G G A T T T G C C C 3 ' Reverse 308 21 58 57 5' CTC ATCTCGTCCTGCTC A G C T 3 ' Amplicon 234-308 75 84 60 Based on rat gastrin mRNA sequence; Accession number: M38653 2 Based on rat somatostatin mRNA sequence; Accession number: M25890 OS adenosine A i or A 2 A receptor, gastrin or somatostatin gene expression levels for each individual sample were divided by the housekeeping-gene expression level to obtain a normalized value of measurement (MN). The mean \u00C2\u00B1 S.E.M. of M N for the fed control and fasted test group were calculated. The mean of each control group was then expressed as 100%, while the gene expression levels of the test groups were expressed as a percentage of controls. Statistical significance was determined using the two-tailed unpaired Student's t-test, where P < 0.05 is considered significant. 2. Effect of omeprazole on A] and A 2 A receptor-induced changes in S L I and I R G release Male Wistar rats (250-275g) were treated with omeprazole as described in the previous section. Rats were deprived of food for 12 h but had free access to water prior to stomach perfusion. Stomach perfusion was performed 24 h after the last drug treatment. Animals were anaesthetized with Somnotol (60mg/kg), and stomachs were prepared for perfusion as decribed in Section I (1). The A i receptor agonist CPA and A 2 A receptor agonist CGS 21680 were administered by side arm infusion at a rate to give a final concentration of 0.1 or 1 pM. These concentrations were selected because CPA has been shown to significantly inhibit and stimulate gastric SLI release when it is administered at a concentration of 0.1 and 1 pM, respectively. Both concentrations of CPA have also been shown to inhibit IRG release. The administration of CGS 21680 has been shown to significantly stimulate gastric SLI release at a concentration of 0.1 p M and to maximally stimulate SLI release at a concentration of 1 p M (see results). The effect of omeprazole treatment on CPA induced changes in SLI and IRG release and CGS-induced changes in SLI release was examined. Samples were collected and stored as described in section I (2). Radioimmunoassay was performed to measure SLI and IRG release as described on section I (3 & 4). The release of SLI and IRG were both expressed as the mean \u00C2\u00B1 S E M for each 5 min sample. SLI and IRG release (%), and the % change in release was performed as described in Section I, 5. Statistical significance was determined using the paired Student's t-test, where P < 0.05 is considered significant. Single-factor A N O V A for repeated measures followed by Dunnett's t-test was also performed using GraphPad Prism. IV. Cellular localization of the gastric Ai and A 2 A receptors 1. Tissue preparation for immunohistochemistry (IHC) The methods used for IHC have previously been described (Yip et al., 2003). Four male Wistar rats (225-250 g) were anaesthetized with Somnotol (60 mg/kg i.p.). Stomachs were dissected out, cut along the greater curvature, and thoroughly rinsed with ice cold saline. Tissue specimens were immersion fixed in freshly prepared 0.1 M phosphate buffer (pH 7.4) containing 4% paraformaldehyde for 30 min (A 2 A receptor IHC) or overnight (Ai receptor IHC) at 4\u00C2\u00B0C. This was followed by cryoprotection in 0.1 M phosphate buffer containing 20% sucrose for 12 h. The corpus (excluding the region adjacent to the fundus and antrum) and the antrum were mounted on corks, embedded in O.C.T. compound (Miles, Etobicoke, ON) and frozen in isopentane (-60\u00C2\u00B0C) for 1 min. Cryostat sections of 30 pm thickness were cut at -25\u00C2\u00B0C and placed in 0.1M phosphate buffered saline (PBS) containing 0.1% sodium azide. Random tissue sections were placed in 0.1 M PBS (pH 7.4) and stained as floating sections to enhance the penetration of antisera into these sections. Prior to the application of primary antibody, sections were sequentially incubated in 0.1 M PBS containing 50 m M NH 4C1 for 30 min, 0.1 M PBS containing 100 mM glycine for 10 min, and 0.1 M PBS containing 1% BSA and 0.3% Triton X-100 for 1 h. Primary and secondary antibodies were diluted in 0.1 M PBS containing 1% BSA, 0.3% Triton X-100, and 0.1% sodium azide. Sections were incubated in adenosine A i or A 2 A receptor primary antibody for 72 h at 4\u00C2\u00B0C, washed in 0.1 M PBS (3x15 min), and incubated with cyanine Cy3-conjugated secondary antibody overnight at 4\u00C2\u00B0C. Sections were again washed in 0.1 M PBS (3x15 min) and either double stained or mounted onto glass slides. For double staining, tissue sections were incubated in one of the other primary antibodies for 72 h at 4\u00C2\u00B0C, washed in 0.1 M PBS (3x15 min) and incubated with Alexa Fluor\u00C2\u00AE 488-conjugated secondary antibody overnight at 4\u00C2\u00B0C. Sections were again washed in 0.1 M PBS (3x15 min) and mounted onto glass slides. Coverslips were placed on sections using a mixture of 0.1 M PBS in glycerin (1:9) and sealed with nail polish. 2. Adenosine receptor antibodies and control peptides The rabbit anti-Ai adenosine receptor antibody was purchased from Sigma-Aldrich and was developed using amino acids 309-326 of the rat A i adenosine receptor C-domain as the immunogen. This antibody has been shown to cross-react strongly with the rat A i receptor (Nakata, 1993; Ciruela et al., 1995; Rivkees et al., 1995). The specificity of this antibody in rat tissues has been examined by Western blotting (Carruthers et al., 2001). When samples were deglycosylated, this antibody detected only one band of the expected size (38-40k Da). In addition, pre-absorption of this antibody with the A i receptor peptide resulted in a lack of immunoreactivity (Caruthers, 2001). The A i receptor antibody was used at a dilution of 1:500 and immunostaining was detected using Donkey anti-rabbit IgG conjugated to Cy3 (Jackson ImmunoReasearch, West Grove, PA) at a concentration of 1:2000. Since an anti-rat A 2 A receptor antibody was unavailable, a rabbit anti-canine A 2 A receptor antibody was used. This antibody was purchased from Alpha Diagnostic International (San Antonio, TX) and was generated using an immunogen corresponding to a 30 amino acid sequence in the 4 t h intracellular domain of the canine A 2 A receptor. This region has 43% homology with the rat A 2 A receptor and has been shown to cross-react with the rat adenosine A 2 A receptors (Diniz et a l , 2003). Its specificity has previously been established (Nie et a l , 1999; Christofi et a l , 2001). This antibody was used at a dilution of 1:200 and immunostaining was detected using donkey anti-rabbit IgG conjugated to Cy3 at a concentration of 1:2000. Since the blocking peptide for the A i receptor antibody was not available from Sigma-Aldrich, one was custom made by the NAPS Unit at U B C . This synthetic peptide was based on the following immunogen sequence provided by Sigma-Aldrich: Cys-Gln-Pro-Lys-Pro-Pro-Ile-Asp-Glu-Asp-Leu-Pro-Glu-Glu-Lys-Ala-Glu-Asp. The blocking peptide was re-dissolved in 0.1 M PBS (pH 7.4), and the concentration was determined using the B C A assay (Pierce, Rockford, IL). The blocking peptide for the A 2 A receptor antibody was obtained from Alpha Diagnostic International. Preabsorption of the A 2 A receptor primary antibody with this control peptide has been shown to abolish A 2 A receptor labeling (Christofi et a l , 2001). 3. Antibodies used for double-staining To examine whether the adenosine A i and A 2 A receptors are localized on specific cell types, double staining was performed to identify somatostatin, gastrin, H + K + -ATPase p, von Willebrand's factor (VWF), and protein gene product (PGP) 9.5. The secondary antibody, donkey anti-mouse IgG conjugated to Alexa Fluor\u00C2\u00AE 488 (1:2000, Molecular Probes, Eugene, OR) was used to detect this staining. Somatostatin-IR was used to identify D-cells of the corporeal and antral mucosa and somatostatin-IR nerves within the muscle layers and enteric plexi. Somatostatin was stained using the somatostatin antibody (SOMA 8; 1:500) provided by the M R C regulatory peptide group (UBC). G-cells of the antral mucosa were identified by gastrin staining using the gastrin antibody (109-21; 1: 30,000) provided by the late Dr. John Walsh. Both these antibodies have been used to identify G-cells (Pederson et al., 1984) and D-cells (Buchan et al., 1985b) in the rat stomach. The parietal cells were identified by staining the P-subunit of the proton pump (H +K +-ATPase). The H + K + -ATPase P antibody was purchased from Affinity Bioreagents Inc. (Golden, CO) and used at a concentration of 1:2000, as suggested by the manufacturer. To identify vascular endothelial cells lining blood vessels in the stomach, V W F was stained. This protein is synthesized by endothelial cells and is stored in the Weibel-Palade bodies of the endothelial cells (Schmugge et al., 2003). The V W F antibody was used at a dilution of 1:100 and was purchased from Serotec (Oxford, England). PGP 9.5 staining was used as a neuronal marker. This protein is expressed in the cytoplasm of central and peripheral neurons (Schofield et al., 1995), and has previously been used to study the innervation of the gastrointestinal tract (Yip et al., 2003). The PGP 9.5 antibody was used at a dilution of 1:200 and was purchased from Abeam Limited (Cambridge, UK). The specificity of these antibodies have previously been characterized (Wall et a l , 1980; Buchan et al., 1985b; Vincent et al., 1985; Wilson et al., 1988; Chow and Forte, 1993). 4. Confocal microscopy A i receptor immunoreactivity (AiR-IR) and A 2 A receptor immunoreactivity ( A 2 A R -IR) were viewed using the Biorad Radiance 2000 confocal scanning laser system mounted on a Nikon Eclipse TE300 inverted microscope. The system utilizes a krypton gas laser with an excitation wavelength of 568 nm and emission filter 575-625 nm (for visualization of Cy3), and an excitation wavelength of 488 and emission filter 500-530 nm (for visualization of Alexa 488). Lens magnification of x40 was used with a zoom factor of 1.0 and z-step of 0.5 to 1.0 pm, while lens magnification of x60 was used with a zoom factor of 1.0-1.6 and a z-step of 0.3 to 0.5 pm. The software program Lasersharp 2000 (version 4.1, Biorad) was used to scan tissues sequentially using the red and green collection channels and the Kalman collection filter (n=2). The Kalman collection filter averages the images collected to increase the signal to noise ratio. Images of 512x512 pixels were obtained and analyzed using NIH image (National Institutes of Health, Bethesda, M D , USA) and Adobe Photoshop (version 7.0, Adobe Systems Inc.). To determine whether co-localization occurs, a single optical image collected from the red channel (AiR-IR or A 2 A R-IR) was overlaid on the corresponding single optical image collected from the green channel (somatostatin, gastrin, V W F , PGP, H + K + -ATPase, VIP or CGRP-IR) using Adobe Photoshop. 5. Quantification of co-localization Co-localization of AiR-IR and A 2 A R - I R with somatostatin-IR and gastrin-IR was quantified in the antral and corporeal mucosa by examining at least 3 tissue sections from 4 different animals. For quantification of AjR-IR with gastrin-IR, at least 12 fields of view were analyzed. AiR-IR was significantly more abundant than A 2 A R - I R . The corporeal and antral mucosa contained only 1.1\u00C2\u00B10.2 and 1.3\u00C2\u00B10.2 A 2 A R - I R cells per view, respectively. Thus, a total of at least 55 fields of view were analyzed for quantification of A 2 A R - I R with somatostatin-IR. 6. Control studies Immunoneutralization experiments were performed to confirm antibody specificity. The A]R antibody was neutralized by incubating l p l of the stock antibody (Sigma-Aldrich) with 0.1 pg of the blocking peptide in 0.5 ml 0.1 M PBS containing 1% BSA, 0.3% Triton X -100, and 0.1% sodium azide for 2 nights at 4\u00C2\u00B0C. As suggested by the manufacturer, the A 2 A R antibody was neutralized by incubating 1 pg of antibody with 40 pg of control peptide in 0.2 ml of 0.1 M PBS containing 1% BSA, 0.3% Triton X-100, and 0.1% sodium azide for 2 nights at 4\u00C2\u00B0C. A positive control lacking the control peptide was also prepared for both antibodies. Tissues stained with the neutralized antibody did not demonstrate AiR-IR or A 2 A R - I R , while tissues stained with the positive control demonstrated extensive AiR-IR and A 2 A R-IR . Additional control experiments were also performed to ensure that non-specific binding did not occur. These included sections which were processed as follows: 1) incubation in 1% B S A in place of the primary antibody, 2) incubation in adenosine A i or A 2 A receptor primary antibody without secondary antibody, and 3) incubation in adenosine A i or A 2 A primary antibody followed by Alexa 488-conjugated anti-mouse IgG secondary antibody. No immunostaining was observed following these procedures. To ensure that bleed-through did not occur, single-stained sections were examined under the microscope using both filters. Bleed-through was not detected for any of the antibodies used. Chapter 3: Results I. Effect of adenosine on gastric IRG and SLI release 1. Effect of adenosine analogs on gastric IRG release The effect of CPA (Ai-selective agonist), CGS 21680 (A2A-selective agonist), and IB-M E C A (A3-selective agonist) on the inhibition of IRG release from the isolated vascularly perfused stomach was examined. The basal release of IRG during periods 1-3 remained relatively constant for all experiments. The basal IRG release (periods 1 to 3) ranged from 114\u00C2\u00B119 to 117\u00C2\u00B119pg/min (Fig. 11). When 1 p M CPA was administered, gastric IRG release was inhibited immediately upon drug perfusion (Fig. 11). The inhibition was apparent throughout the drug perfusion period, and the release of IRG returned to basal levels within 5 min after drug removal. The effect of various concentrations of CPA on IRG release was also examined. Results are expressed in % inhibition and are summarized in Fig. 12. CPA caused a concentration-dependent inhibition of IRG release. Maximal inhibition was achieved at a concentration of 1 pM. The administration of 10 p M CPA did not further suppress IRG release. The empirical EC50 of CPA in inhibiting IRG release was estimated to be 0.067 pM, with a confidence interval between 0.014 and 0.325 pM. The administration of CGS 21680 (0.1 pM) and IB-MECA (0.1 pM) did not alter basal IRG release. 200 - i o 1 5 0 H (0 | 100-5 50H CPA (1uM) I X 1 2 3 4 5 6 7 8 9 10 5 min periods Fig. 11. Effect of CPA on gastric IRG release. Results are expressed as IRG release (%) as described in the Methods section. Each column represents the mean \u00C2\u00B1 SEM of 5 experiments. *P < 0.05 and ***p < 0.001 when compared with period 3 using repeated measures A N O V A followed by Dunnett's multiple comparison test. 1 0 0 n 90-80-70-60-50-40-30-20-10-0--10-*** *** 0.001 0.01 0.1 1 10 0.1 CPA CGS 21680 (|iM) (|iM) 0.1 IB-MECA (ixM) Fig. 12. Effect of various concentrations of CPA, and 0.1 uM of CGS 21680 and IB-MECA on IRG release. Results are expressed as the % inhibition of IRG release, and calculated as described in the Methods section. Each column represents the mean \u00C2\u00B1 SEM of at least 5 experiments; *P < 0.05 and ***P < 0.001 when the mean IRG release (pg/min) during drug perfusion periods 4-7) is compared to the mean basal IRG release (periods 1-3). 2. Effect of adenosine receptor antagonists on gastric IRG release Adenosine has been shown to inhibit gastric IRG release (Kwok et a l , 1990). To determine the receptor subtype involved in this adenosine-mediated action, the effect of the A i receptor-selective antagonist, DPCPX, and the A 2 receptor-selective antagonists, D M P X , on adenosine-induced inhibition of IRG release were examined. Neither antagonists altered basal IRG release (Fig. 13). Fig. 14 shows the effect of DPCPX and D M P X on adenosine-induced changes in IRG release. After a 15 min basal period, 1 p M of the antagonist was administered alone for 5 min prior to the concomitant perfusion with adenosine (10 pM) for 15 min. In the presence of 1 p M DPCPX, the adenosine-induced inhibition of IRG release was completely abolished. D M P X (1 pM), however, had no effect on adenosine-induced changes in IRG release. 3. Effect of adenosine receptor agonists on gastric SLI release To determine which adenosine receptor subtype is involved in regulating gastric SLI release, the effect of CPA (Apselective agonist), CGS 21680 (A2A-selective agonist), and IB-M E C A (A3-selective agonist) were examined. The basal release of SLI during periods 1-3 remained relatively constant for all experiments. In experiments examining the effect of agonists (Fig. 15 and 16), the rates of basal SLI release (period 1-3) were 174\u00C2\u00B124 tol78\u00C2\u00B127 pg/min (CPA 0.1 pM), 173\u00C2\u00B119 to 180\u00C2\u00B121 pg/min (CPA 1 pM), 92\u00C2\u00B118 to 95\u00C2\u00B118 pg/min (CGS 21680) and 191\u00C2\u00B139 to 195\u00C2\u00B141 pg/min (IB-MECA). When administered at a concentration of 0.1 pM, CPA immediately inhibited gastric SLI release (Fig. 15A). The inhibition continued throughout the drug perfusion period, and SLI release returned to basal (A 10 | 100-o CC 50-DPCPX(VM) 2 3 4 5 6 7 8 9 10 5 min periods B 200-i 9 1 5 ( H CO | 100-0\u00C2\u00A3 O 0\u00C2\u00A3 50-DMRX(1 nM) 2 3 4 5 6 7 8 9 10 5 min periods Fig. 13. Effect of DPCPX and DMPX on gastric IRG release. Each column represents the mean \u00C2\u00B1 SEM of at least 5 experiments. Statistics were performed using repeated measures ANOVA followed by Dunnett's multiple comparison test. Neither drug altered basal IRG release. (A <0 cc o ft ADO(10nM) 50-*** *** 1 2 3 4 5 6 7 8 9 10 5 min periods B 200 : v 1 5 \u00C2\u00B0 : tn to | 100-1 K O K DPCFX(1 nM) ADO(10iiM) > 50-0 : 1 2 3 4 5 6 7 8 9 10 5 min periods 4) in a a> IB-MECA > CPA. 4. Effect of adenosine receptor antagonists on gastric SLI release The effect of D M P X , an A 2 receptor-selective antagonist was studied to examine the role of A 2 receptors in adenosine and adenosine agonist-induced gastric SLI release. When perfused through the gastric vasculature for 20 min, 1 p M D M P X did not alter basal SLI release, while 10 p M D M P X significantly suppressed SLI release. Experiments similar to those described for Fig. 14 were then performed to test the effect of D M P X on adenosine and adenosine receptor agonist-induced release of SLI; 1 or 10 p M D M P X was administered 5 min prior to the concomitant perfusion with the agonist for 15 min. Results are summarized in Fig. 18. In the presence of 1 p M D M P X , adenosine induced-SLI release was attenuated. D M P X (1 pM) also inhibited the response elicited by CGS 21680 (0.01 and 0.1 pM) and IB-M E C A (lpM), and completely abolished the effects of CPA (1 pM) on gastric SLI release. CGS 21680-induced release of SLI was significantly suppressed by D M P X in a concentration-dependent manner. The % change of SLI release induced by 0.01 p M CGS 21680 in the presence of 10 p M D M P X was not significantly different from the basal SLI release. 800- I 600-* 400- X 200-X X o--200-DMPX(nM) 1 10 0 1 0 1 10 0 1 10 0 1 0 1 + ADO + CGS + CGS + CPA + IB-MECA (1 uty) (0.01 uM) (0.1 nM) (1 H-M) (1(xM) Fig. 18. Effect of DMPX on basal, adenosine, and adenosine agonist-stimulated SLI release. The effect of DMPX on CPA-, CGS 21680- (CGS), and IB-MECA-induced SLI release was examined. Results are expressed as % changes. Each column represents the mean \u00C2\u00B1 SEM of at least 5 experiments; tp < 0.05 when the mean SLI release (pg/min) in the presence of DMPX (periods 4-7) is compared to the mean basal SLI release (periods 1-3), *P < 0.05 and **P < 0.01 when the mean release of SLI (periods 4-7) in the presence of agonists was compared with the mean release of SLI in the presence of both DMPX and agonists. To test i f the stimulatory effect on SLI release is mediated by the A 2 A receptor, the effect of the potent selective A 2 A receptor antagonist, Z M 241385, was examined. When perfused alone, Z M 241385 (1 pM) significantly inhibited basal SLI release during period 5 and 7 (Fig. 19A). However, when the % change of SLI release was calculated, SLI release in the presence of 1 p M Z M 241385 did not differ significantly from basal SLI release (Fig. 20). Z M 241385 abolished the stimulatory effect of adenosine; in the presence of Z M 241385 and adenosine, the release of SLI was inhibited (Fig. 20). When perfused concomitantly with CGS 21680 (0.1 pM), Z M 241385 (1 pM) also abolished the stimulatory effect of CGS 21680, and a significant inhibition of SLI release was apparent (Fig. 19). The effect of 10 p M Z M 241385 on CGS 21680 (0.1 pM) were similar to that of 1 p M Z M 241385. The stimulatory effect of CPA ( lpM) and IB-MECA (1 pM) on SLI release was also completely abolished by Z M 241385. In the presence of both the agonist and antagonist, an inhibition of SLI was apparent (Fig. 20A). The effect of the A i selective antagonist DPCPX was also examined. DPCPX (1 pM) significantly inhibited basal SLI release, but had no effect on CGS 21680- (0.1 pM) stimulated SLI release (Fig. 20). 5. Involvement of endogenous adenosine on SLI release To examine the role of endogenous adenosine in modulating SLI release, experiments were also performed to test the effect of EHNA, an A D A inhibitor, on gastric SLI release using the same experimental protocol described in Fig. 13. E H N A (1-10 pM) caused a concentration-dependent increase in SLI release, while 0.1 p M E H N A had no effect (Fig. 21). The stimulatory effect of adenosine on SLI release was potentiated in the presence of EHNA. gj 200-(0 aj CD DC \u00E2\u0080\u0094 100-_l (/) Z M 241385 (1|xM) X 1 2 3 4 5 6 7 8 9 10 5 min periods B 2000-1 1500-Q) W CO CD 0) 0\u00C2\u00A3 1000 500 -OGS (0.1 nM) I ELTZL 1 2 3 4 5 6 7 8 9 10 5 min periods 300-1 0) (A CO aj \u00E2\u0080\u0094 100-(0 ZM 241385 (1 (iM) 2 0 0 - CGS (0.1 nM) * * * * * 1 2 3 4 5 6 7 8 9 10 5 min periods Fig. 19. Effect of Z M 241385 on basal and stimulated SLI release. A: The effect of Z M 241385 on basal SLI release; B: The effect of CGS 21680 (CGS) on basal SLI release; C: The effect of Z M 241385 on CGS-stimulated SLI release. Each column represents the mean \u00C2\u00B1 SEM of at least 4 experiments; *P < 0.05 when compared with period 3 of respective experiments using repeated measures A N O V A followed by Dunnett's multiple comparison test. 800 |) 600 1 u gT 400 CO 200 -200 t 1 10 ZM 241385 (M.M) X I ZM 241385 (nM) + ADO (1 nM) 0 1 10 ZM 241385 (nM) + C G S (0.1 nM) \" a 0 1 0 1 ZM241385 (nM) ZM241385((iM) + CPA (1 (iM) + IB-MECA (1 ^M) B 1000-800-v at c 10 600-400-200-0--200J I DPCPX (UM) 0 1 D P C P X (nM) f C G S (0.1 M-M) Fig. 20. Effect of Z M 241385 (A) and DPCPX (B) on basal and adenosine (ADO) and agonist-stimulated SLI release. The effect of Z M 241385 on CPA-, CGS 21680- (CGS), and IB-MECA-induced SLI release was examined. Results are expressed as % changes. Each column represents the mean \u00C2\u00B1 SEM of at least 4 experiments; ^P < 0.05 and ^P < 0.01 when the mean SLI release (pg/min) in the presence of Z M 241385 or DPCPX (periods 4-7) was compared to the mean basal SLI release (periods 1-3), **P < 0.01 when the mean release of SLI (periods 4-7) in the presence of agonists was compared with the mean release of SLI in the presence of both Z M 241385 and agonists. 600-1 0> O) 500-(0 c O 400-T 1000 bp-W 500 bp . J 400 bp-W 300 b p * Fig. 22. Results of A, , A 2 A , A 2 B and A 3 RT-PCR performed using rat gastric tissues. RT-PCR was performed using tissues from the rat fundus (A), corpus (B), antrum (C) and mucosa (D) and primers designed to detect the A, (1086 bp), A 2 A (441 bp), A 2 B (380 bp), and A 3 (489 bp) receptor mRNA (see Table 3). Al l four adenosine receptor subtypes are present in the gastric tissues examined. 1337 bp Fig. 23. Amplification of the A, (A) and A 2 A (B) receptor coding region by RT-PCR. RT-PCR was performed using tissues obtained from the striatum (positive control; lane 2), fundus (lane 3), corpus (lane 4), antrum (lane 5) and mucosa (lane 6). The A[FL and A 2 A F L primer sets (see Table 3) used span the entire coding region of the rat adenosine A, and A 2 A receptor and generated only one amplicon, of the expected length, for each tissue examined. 1000 bp 800 bp 300 bp \u00E2\u0080\u00A2 200 bp \u00E2\u0080\u00A2 \u00E2\u0080\u00A2303 bp 208 bp Uncleaved size & region amplified* Enzyme Recognition site Cleavage site Expected fragments (bp) Observed fragments (bp) 1086 bp (322-1407 bp) Nar l G G / C G C C 527 880; 206 907; 208 Sac I GAGCT/C 631 776; 310 791; 303 *Positions based on A, receptor sequence in the rat brain (Accession No. M64299) Fig. 24. Restriction enzyme digestion of the adenosine A, receptor PCR amplicon. RT-PCR products synthesized using the A[FL primer set were concentrated, purified, and cleaved with restriction enzymes, Nar I and Sac I, and separated on a 2% agarose gel. Expected fragment sizes are listed in the table. The size of the observed fragments were estimated using the Stratagene Eagle Sight Image Analysis Software, as described in the Methods. EcoRI Mbo I Pst l Rsa I Uncleaved size & region amplified* Enzyme Recognition site Cleavage site Expected fragments (bp) Observed fragments (bp) 441 bp from 99-449 bp EcoRI G/AATTC 385 286; 155 300;165 Mbo I /GATC 160 380; 61 390 Ps t l CTGCA/G 178 362; 79 361 Rsa I GT/AC 399 300; 141 319;160 *Positions based on A 2 A receptor sequence in the rat brain (Accession No. S47609) Fig. 25. Restriction enzyme digestion of the adenosine A 2 A receptor PCR amplicon. RT-PCR products synthesized using the A 2 A S L primer set were concentrated, purified, and cleaved with restriction enzymes, EcoR I, Mbo, I, Pst I, and Rsa I. Fragments were separated on a 2% agarose gel. Expected fragment sizes are listed in the table. The sizes of the observed fragments were estimated using the Stratagene Eagle Sight Image Analysis Software, as described in the Methods. Ava II Hpa II Uncleaved size & region amplified* Enzyme Recognition site Cleavage site Expected fragments (bp) Observed fragments (bp) 380 bp from 223-603 bp Ava II G/G(A or T)CC 479 256;124 278 Hpa II C / C G G 413 190; 190 190 *Positions based on A 2 B receptor sequence in the rat brain (Accession No. M91466) Fig. 26. Restriction enzyme digestion of the adenosine A 2 B receptor PCR amplicon. RT-PCR products synthesized using the A 2 B S L primer set were concentrated, purified, and cleaved with restriction enzymes, Ava II and Hpa II. Fragments were separated on a 2% agarose gel. Expected fragment sizes are listed in the table. The sizes of the observed fragments were estimated using the Stratagene Eagle Sight Image Analysis Software, as described in the Methods. Ava II Hinf I 360 bp Uncleaved size & region amplified* Enzyme Recognition site Cleavage site Expected fragments (bp) Observed fragments (bp) 489 bp from 319-788 bp Ava II G/G(A or T)CC 452; 546 133; 262; 94 288 Hinf I G/A(ACG or T)TC 653 334;155 360 *Positions based on A 3 receptor sequence in the rat brain (Accession No. M94152) Fig. 27. Restriction enzyme digestion of the adenosine A 3 receptor PCR amplicon. RT-PCR products synthesized using the A 3 SL primer set were concentrated, purified, and cleaved with restriction enzymes, Ava II and Hinf I. Fragments were separated on a 1% agarose gel. Expected fragment sizes are listed in the table. The sizes of the observed fragments were estimated using the Stratagene Eagle Sight Image Analysis Software, as described in the Methods. EcoR I Hinf I Uncleaved size & region amplified* Enzyme Recognition site Cleavage site Expected fragments (bp) Observed fragments (bp) 1337 bp from 41-1377 bp EcoR I G/AATTC 385 992; 345 1015; 356 Hinf I G/A(ACG or T)TC 719 658; 679 -690 *Positions based on A 2 A receptor sequence in the rat brain (Accession No. S47609) Fig. 28. Restriction enzyme digestion of the full length adenosine A 2 A receptor amplicon. RT-PCR products synthesized using the A 2 A F L primer set were concentrated, purified, and cleaved with restriction enzymes, EcoR I and Hinf I. Fragments were separated on a 2% agarose gel. Expected fragment sizes are listed in the table. The sizes of the observed fragments were estimated using the Stratagene Eagle Sight Image Analysis Software, as described in the Methods. 2. Ai and A 2 A receptor coding region and A 2 B and A3 receptor partial cDNA sequence Experiments were performed to determine the sequences of the A i and A 2 A receptor coding regions, and the sequences of the partial A 2 B and A3 receptor cDNAs. The mucosal A i and A 2 A receptor sequences cloned using the full length A i and A 2 A receptor primers (Table 3) were submitted to Genbank and assigned accession numbers AF042079 and AF228684, respectively. The A 2 B and A3 receptor sequences cloned using the short length A 2 B and A 3 receptor primers (Table 3) were assigned accession numbers AF084241 and AF102804, respectively. Results of the sequencing experiments demonstrate that the coding region of the gastric mucosal A i receptor (accession # AF042079) was identical to the published sequence in the rat brain (Mahan et a l , 1991). The sequence of the gastric mucosal A 2 A receptor coding region (accession # AF228684) was also found to be identical to that in the brain (Fink et a l , 1992). Fig. 29 and 30 show the alignments of the rat mucosal and rat brain A i and A 2 A receptor coding regions. D N A sequence analysis was also performed on the PCR amplicons generated using the A 2 B S L and A3SL primer sets listed in Table 3. An alignment of these regions with corresponding regions of the rat brain receptors is shown in Fig. 31. The sequence of the mucosal A 2 B receptor PCR amplicon is identical to that of the rat brain. However, analysis of the A3 receptor PCR amplicon reveals a silent mutation at position 492. At the third position of the codon for alanine, adenine (A) is replaced by thymine (T). The position of this mutation corresponds to amino acid #59 of the A3 receptor protein, which is situated in the second transmembrane domain (Salvatore et a l , 1993). 1 tgtctgctgatgtgcccagctcctgcccaccatgccgccctacatctcggccttccaggc 60 541 gtttgagaaggttatcagcatggagtacatggtctacttcaacttcttcgtctgggtgct 600 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11111111111 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 1 1 1 1 1 11111111111 1 1 1 1 1 1 1 1 1 1 II 1111 1111111111111111111111111111111 1 1 1 1 1 1 1 1 1 1 1 276 tgtctgctgatgtgcccagctcctgcccaccatgccgccctacatctcggccttccaggc 335 816 gtttgagaaggttatcagcatggagtacatggtctacttcaacttcttcgtctgggtgct 875 61 tgcctacattggcatcgaggtgctcattgccttggtctctgtgcccggaaatgtactggt 120 601 gccgccactgctcctcatggtcctcatctacctggaggtcttctacctgatccgtaagca 660 1II111111II111 1 1 111111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11111111 1 1 1 1 1 1 1 II 1 1 1 11111111111111111111111111111111111 1 1 1 1 1 1 1 1 1 1 1 336 tgcctacattggcatcgaggtgctcattgccttggtctctgtgcccggaaatgtactggt 395 876 gccgccactgctcctcatggtcctcatctacctggaggtcttctacctgatccgtaagca 935 121 gatttgggctgtgaaggtgaaccaggcacttcgcgatgccaccttctgcttcatcgtgtc 180 661 gctcaacaaaaaggtgtcagcctcctccggtgacccccagaagtactacgggaaggagct 720 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 396 gatttgggctgtgaaggtgaaccaggcacttcgcgatgccaccttctgcttcatcgtgtc 455 936 gctcaacaaaaaggtgtcagcctcctccggtgacccccagaagtactacgggaaggagct 995 181 actggcggtagctgatgtggccgttggcgccctggtcatcccactggccatccttatcaa 240 721 gaagatcgccaagtcgctggccctcatcctcttcctctttgccctcagctggctgccgct 780 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 456 actggcggtagctgatgtggccgttggcgccctggtcatcccactggccatccttatcaa 515 996 gaagatcgccaagtcgctggccctcatcctcttcctctttgccctcagctggctgccgct 1055 241 cattgggccacagacctacttccacacctgcctcatggtggcctgccctgtcctcatcct 300 781 gcatatcttgaactgtatcaccctcttctgccccacctgccagaaacccagcattctgat 840 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 516 cattgggccacagacctacttccacacctgcctcatggtggcctgccctgtcctcatcct 575 1056 gcatatcttgaactgtatcaccctcttctgccccacctgccagaaacccagcattctgat 1115 301 cacccagagctccattctggctctgctcgccattgctgtggatcgatacctccgagtcaa 360 841 ctacatcgccatcttcctcacacacggcaactccgccatgaaccccatcgtctatgcctt 900 1 11 1 1 1 11 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 1 11 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 II1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 576 cacccagagctccattctggctctgctcgccattgctgtggatcgatacctccgagtcaa 635 1116 ctacatcgccatcttcctcacacacggcaactccgccatgaaccccatcgtctatgcctt 1175 361 gatccctctccggtacaagacagtggtgacccagcggcgggcggcagtggccatagctgg 420 901 ccggatccacaagttccgggtcacctttctgaagatttggaatgaccacttccgatgcca 960 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 111 1 111 1 1 II 1 1 1 11 1 1 11 1 1 1 1 1 1 1 1 1 1 1 636 gatccctctccggtacaagacagtggtgacccagcggcgggcggcagtggccatagctgg 695 1176 ccggatccacaagttccgggtcacctttctgaagatttggaatgaccacttccgatgcca 1235 421 ctgctggattctctcccttgtggtaggcctgacacccatgtttggctggaacaacctgag 480 961 gcctaagcctcccatcgatgaagacctcccagaggagaaagctgaggactagactctgcc 1020 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 696 ctgctggattctctcccttgtggtaggcctgacacccatgtttggctggaacaacctgag 755 1236 gcctaagcctcccatcgatgaagacctcccagaggagaaagctgaggactagactctgcc 1295 481 tgtggtagagcaagactggagagccaacggcagtgttggggagcccgtgatcaagtgtga 540 1021 ttgctccgtctagcccatgcccagcggctctctgttcaactcccacgttctccctgtccc 1080 1 1 1 1 1 1 1 1 1 1 1 1 1 111111 111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1111 11111 II 1 1 1 111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 756 tgtggtagagcaagactggagagccaacggcagtgttggggagcccgtgatcaagtgtga 815 1296 ttgctccgtctagcccatgcccagcggctctctgttcaactcccacgttctccctgtccc 1355 1081 accctgtc 1088 1 1 1 1 1 1 1 1 1356 accctgtc 1363 Fig. 29. Alignment of the rat mucosal and brain A! receptor coding regions. The rat mucosal A, receptor cDNA sequence (top strand; accession number: AF042079) is aligned with the corresponding region the rat brain receptor (bottom strand; accession number: M64299) o 1 ctgctgagcctgcccaagtgtggctgctcccaccatgggctcctcggtgtacatcacggt 60 661 gcccctgccaggggagcggactcggtccacgctgcagaaggaggtccacgctgccaagtc 720 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 M 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 I 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 41 ctgctgagcctgcccaagtgtggctgctcccaccatgggctcctcggtgtacatcacggt 100 701 gcccctgccaggggagcggactcggtccacgctgcagaaggaggtccacgctgccaagtc 760 61 ggagctggccatcgctgtgctggccatcctgggcaacgtgctcgtgtgctgggccgtgtg 120 721 cctggccatcatcgtcgggctctttgctctgtgctggttgccgctgcacatcatcaactg 780 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1111 1 1 1111 1 1 11II1 1 1 111II 1 1 1 1 1 1 1 1 1 1 1 1 101 ggagctggccatcgctgtgctggccatcctgggcaacgtgctcgtgtgctgggccgtgtg 160 761 cctggccatcatcgtcgggctctttgctctgtgctggttgccgctgcacatcatcaactg 820 121 gatcaacagtaacctgcagaacgtcaccaacttctttgtggtatcgctggcggcggctga 180 781 tttcaccttcttctgctccacgtgccggcacgcccctccgtggctcatgtacctggccat 840 11111111111111II1111111111111111111111II 1 1 M 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 161 gatcaacagtaacctgcagaacgtcaccaacttctttgtggtatcgctggcggcggctga 220 821 tttcaccttcttctgctccacgtgccggcacgcccctccgtggctcatgtacctggccat 880 181 cattgcagtgggtgtgctcgccatccccttcgctatcaccatcagcaccggcttctgcgc 240 841 catcctctcccacagcaactccgtcgtcaaccccttcatctacgcctacaggatccggga 900 11111111 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 1 1111111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1111 1 1 1 1 1 1 1 1 1 1 1 1 111111111111111111111111111111111II11111 1 1 1 1 1 221 cattgcagtgggtgtgctcgccatccccttcgctatcaccatcagcaccggcttctgcgc 280 881 catcctctcccacagcaactccgtcgtcaaccccttcatctacgcctacaggatccggga 940 241 cgcctgccacggctgcctcttcttcgcctgttttgtcctggtcctcacgcagagttccat 300 901 gttccgccagaccttccggaagatcatccgaacccacgtcctgaggcggcaggaaccctt 960 1 1 11 111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II11111111 1 1 1 1 1 1111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 1 1 1 1 1 1 111 1 1 1 1 1 1 1 1111 1 II 1 1 1 1 1 111 1 1 1 1 1 1 1111 1 1 281 cgcctgccacggctgcctcttcttcgcctgttttgtcctggtcctcacgcagagttccat 340 941 gttccgccagaccttccggaagatcatccgaacccacgtcctgaggcggcaggaaccctt 1000 301 ctttagcctcttggctatcgccatcgaccgctacatcgccatccgaattccactccggta 360 961 ccaggcagggggttccagtgcctgggccctggcagctcacagcactgagggagagcaggt 1020 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 11 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 11 1 11 1 11 1 1 11 1111 1 1 1 1 1 1 1 1 1 II 1 1 341 ctttagcctcttggctatcgccatcgaccgctacatcgccatccgaattccactccggta 400 1001 ccaggcagggggttccagtgcctgggccctggcagctcacagcactgagggagagcaggt 1060 361 caatggcttggtgacaggtgtgagggcgaagggcatcattgcaatttgctgggtgctgtc 420 1021 tagcccccgccttaatggccaccccctgggggtatgggccaacggcagtgccacccattc 1080 11 1 1 1 1 1 1 1 1 1 II 1 II 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 1 11 11 1 1 11 1 1 1 1 1 1 1 1 1 1 1 1 1 11 1 1 11 11 1111 11111 1 1111111111 1 1 1 1 II 1 1 1 1 1 1 401 caatggcttggtgacaggtgtgagggcgaagggcatcattgcaatttgctgggtgctgtc 460 1061 tagcctccgccttaatggccaccccctgggggtatgggccaacggcagtgccacccattc 1120 421 gtttgccattggcctgacccccatgctgggctggaacaactgcagtcagaaagacgggaa 480 1081 cggacggcggcccaatggctacactctggggctggggggtggagggagtgcccaaggctc 1140 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II II 1 1 1 II11II1111111II11II111111 1 1 1 1 II II 1 1 II 1 1 1 II 1 461 gtttgccattggcctgacccccatgctgggctggaacaactgcagtcagaaagacgggaa 520 1121 cggacggcggcccaatggctacactctggggctggggggtggagggagtgcccaaggctc 1180 481 ctccacgaagacctgcggcgagggccgggtgacctgtctgttcgaggacgtggtgcccat 540 1141 tcctcgggatgtggagcttcctacccaggagcgccaggaaggccaagagcaccctggcct 1200 1 II 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1111111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 11111111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 111II1111111111 1 1 1111111II1 1 1 1 1 1 1 1 1 1 1 1 521 ctccacgaagacctgcggcgagggccgggtgacctgtctgttcgaggacgtggtgcccat 580 1181 tcctcgggatgtggagcttcctacccaggagcgccaggaaggccaagagcaccctggcct 1240 541 gaattacatggtttactacaacttctttgcgttcgtgttactgccccttctgctcatgct 600 1201 aaggggtcatctggtccaggctagagtaggagcttcctcatggtcttcagagtttgcccc 1260 1 1 1 1 1 1 1111111111111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1II1111II1 II 1 1 1 1 1 1 1 1 111 II 1 11 111 1 111 1 11 1 II1111111 1 1 111 1 1 1 1 1 II 1 1 1 581 gaattacatggtttactacaacttctttgcgttcgtgttactgccccttctgctcatgct 640 1241 aaggggtcatctggtccaggctagagtaggagcttcctcatggtcttcagagtttgcccc 1300 601 ggccatctacctacggatttttctggcggcccggagacagctgaagcagatggagagcca 660 1261 ttcctgagggaaagacattttaatatttttggttggctggaccaatctcactaagggaag 1320 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1111 1 1II11 1 1111 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 II 1 1 1 1 1 641 ggccatctacctacggatttttctggcggcccggagacagctgaagcagatggagagcca 700 1301 ttcctgagggaaagacattttaatatttttggttggctggaccaatctcactaagggaag 1360 1321 agaaacccaatgggc 1335 1 1 1 1 1 1 1 1 1 1 1 1 M 1 1361 I I I I I I I I I I I I I I I agaaacccaatgggc 1375 Fig. 30. Alignment of the rat mucosal and brain A 2 A receptor coding regions. The rat mucosal A 2 A receptor cDNA sequence (top strand; accession number: AF228684) is aligned with the corresponding region the rat brain receptor (bottom strand; accession number: S47609) 1 a c t a c t t t c t g g t g t c c c t g g c g a c g g c g g a c g t g g c t g t g g g a c t c t t c g c c a t c c c c t I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I 2 3 5 a c t a c t t t c t g g t g t c c c t g g c g a c g g c g g a c g t g g c t g t g g g a c t c t t c g c c a t c c c c t 61 t t g c c a t c a c c a t c a g c c t g g g c t t c t g c a c g g a c t t t c a c a g c t g c c t c t t c c t c g c c t I I I I I I I I I I I I I I I I I I I I I I i I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I 2 9 5 t t g c c a t c a c c a t c a g c c t g g g c t t c t g c a c g g a c t t t c a c a g c t g c c t c t t c c t c g c c t 1 2 1 g c t t c g t g c t g g t g c t c a c a c a g a g c t c c a t c t t t a g c c t c t t g g c g g t g g c t g t c g a c c I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I 3 5 5 g c t t c g t g c t g g t g c t c a c a c a g a g c t c c a t c t t t a g c c t c t t g g c g g t g g c t g t c g a c c 1 8 1 g g t a t c t g g c c a t t c g c g t c c c g c t c a g g t a t a a a g g t t t g g t c a c t g g a a c a c g a g c a a I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I II I I I I I I I I I I I I 4 1 5 g g t a t c t g g c c a t t c g c g t c c c g c t c a g g t a t a a a g g t t t g g t c a c t g g a a c a c g a g c a a 2 4 1 g a g g g a t c a t c g c t g t c c t c t g g g t c c t t g c c t t t g g c a t t g g a c t g a c t c c t t t c c t g g I I I I I I I I II I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I 4 7 5 g a g g g a t c a t c g c t g t c c t c t g g g t c c t t g c c t t t g g c a t t g g a c t g a c t c c t t t c c t g g 3 0 1 g t t g g a a c a g t a a a g a c c g t g c c a c c a g c a a c t g c a c a g a a c c t g g g g a t g g c a t c 3 5 6 I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I 5 3 5 g t t g g a a c a g t a a a g a c c g t g c c a c c a g c a a c t g c a c a g a a c c t g g g g a t g g c a t c 5 9 0 60 2 9 4 1 2 0 354 180 414 2 4 0 474 3 0 0 534 B 3 1 9 61 3 7 9 1 2 1 4 3 9 1 8 1 4 9 9 2 4 1 5 5 9 3 0 1 6 1 9 3 6 1 6 7 9 4 2 1 7 3 9 4 8 1 7 9 9 a a a g c c a a c a a t a c c a c g a c g a g t g c c t t g t g g t t g c a a a t c a c c t a c g t c a c c a t g g a g I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I a a a g c c a a c a a t a c c a c g a c g a g t g c c t t g t g g t t g c a a a t c a c c t a c g t c a c c a t g g a g g c t g c c a t t g g t c t c t g t g c t g t a g t g g g c a a c a t g c t g g t c a t c t g g g t g g t c a a g c t g I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I g c t g c c a t t g g t c t c t g t g c t g t a g t g g g c a a c a t g c t g g t c a t c t g g g t g g t c a a g c t g a a c c g c a c t c t g a g g a c c a c c a c c t t c t a t t t c a t c g t c t c c c t a g c a c t g g c t j a c a t t I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I a a c c g c a c t c t g a g g a c c a c c a c c t t c t a t t t c a t c g t c t c c c t a g c a c t g g c a j a c a t t g c t g t t g g g g t g c t g g t c a t a c c c t t g g c c a t t g c c g t c a g c c t g g a g g t c c a g a t g c a c I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I g c t g t t g g g g t g c t g g t c a t a c c c t t g g c c a t t g c c g t c a g c c t g g a g g t c c a g a t g c a c t t c t a t g c o t g c c t t t t c a t g t c c t g t g t g c t t c t g g t c t t c a c c c a t g c t t c c a t c a t g I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I II t t c t a t g c c t g c c t t t t c a t g t c c t g t g t g c t t c t g g t c t t c a c c c a t g c t t c c a t c a t g t c c t t g c t g g c c a t t g c t g t a g a c c g a t a c c t g c g a g t c a a g c t g a c a g t c a g a t a t a g a I I I I I II I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I t c c t t g c t g g c c a t t g c t g t a g a c c g a t a c c t g c g a g t c a a g c t g a c a g t c a g a t a t a g a a c g g t t a c c a c t c a a a g a a g a a t a t g g c t a t t c c t g g g c c t c t g c t g g c t a g t g t c c t t t I I I II I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I II I I I a c g g t t a c c a c t c a a a g a a g a a t a t g g c t a t t c c t g g g c c t c t g c t g g c t a g t g t c c t t t c t g g t g g g a c t g a c c c c c a t g t t t g g c t g g a a t a g a a a a g t g a c c t t a g a g c t c t c t c a a I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I II I II I I I I I I I I I I I I I I I c t g g t g g g a c t g a c c c c c a t g t t t g g c t g g a a t a g a a a a g t g a o c t t a g a g c t c t c t c a a a a c a g c t c c 4 8 9 I I I I I I II I a a c a g c t c c 8 0 7 60 3 7 8 1 2 0 438 1 8 0 4 9 8 2 4 0 5 5 8 3 0 0 618 3 6 0 6 7 8 4 2 0 7 3 8 4 8 0 7 9 8 Fig. 31. Alignment of the rat mucosal and brain A 2 B and A 3 cDNA sequences. The partial cDNA sequence of the rat mucosal A 2 B (A) and A 3 (B) receptor was aligned with corresponding regions of the rat brain receptors. A: Top strand: gastric mucosal A 2 B receptor sequence (accession number: AF084241); Bottom strand: brain A 2 B receptor sequence (accession number: M91466). B: Top strand: gastric mucosal A 3 receptor sequence (accession number: AF102804) ; Bottom strand: brain A 3 receptor sequence (accession number: M94152). A silent mutation at position 174 bp of the mucosal A 3 receptor cDNA sequence is detected. III. Quantification of gastric adenosine Ai and A 2 A receptor mRNA expression To study the changes in adenosine A\ and A 2 A receptor gene expression induced by various states of the stomach, competitive RT-PCR and Real-Time RT-PCR assays were developed. 1. Competitive RT-PCR Fig. 32 shows the results of a representative A i and A 2 A receptor competitive RT-PCR assay which was performed to quantify A i and A 2 A receptor mRNA levels in the rat striatum. This tissue has been shown to contain a moderate level of A i receptor mRNA and an extremely high level of A 2 A receptor mRNA. In agreement with previous studies, the level of striatal A 2 A receptor mRNA (6.6xl0 7 copies/pg total RNA) was significantly greater than the level of A i receptor mRNA (3.9xl0 6 copies/pg total RNA). See Fig. 32C. When the competitive RT-PCR samples are electrophoresed through an agarose gel, the density of the template band was shown to increase as the initial amount of competitor added was decreased. The Log [density ratio (corrected competitor band/template band)] plotted against the Log (initial competitor concentration) displayed a linear relationship with a R value of > 0.95, and a slope of approximately 1.0. The graphs generated by the A i and A 2 A competitive RT-PCR assays were shown to have slopes of 1.0510.04, and 1.07+0.05, respectively, for all assays performed. An ideal slope of 1.0 indicates that the amplification efficiency of the template is equal to that of the competitor (Freeman et al., 1999). The R 2 values for all assays performed in these studies were > 0.95. B E j\u00C2\u00A3 0.40 0 00 q 8 2- -0.20 'ui c: 2. -0.40 \"[competitor] -template \"competitor y = 0.8516x- 1.0522 R ! = 0.98 5 0.7 0.9 1.1/ 1.3 1.5 1.7 1.9 2.1 2 Log [intial competitor concentration (amol)] =0 0.75 -pH Log [intial competitor concentration (amol)] 100 < K o 4 a B S t r i a t u m Fig. 32. Striatal A , and A 2 A receptor gene expression measured by competitive RT-PCR. Results of a representative Aj (A) and A 2 A (B) receptor competitive RT-PCR assay performed using 2.5 pg of striatum total RNA and various amounts of A, and A 2 A RNA competitor as the template. See inset for competitor concentrations (amol) used. C: A, (filled bar) and A 2 A (empty bar) receptor mRNA expression in the striatum (* P < 0.05, n = 4) Similar mRNA measurements were also obtained regardless of the amount of PCR template used. A i and A 2 A receptor mRNA measurements were not significantly altered by using equal volumes of undiluted, 2x, 4x, or 8x diluted cDNA as the template for PCR (Fig. 33). The results of representative A i and A 2 A competitive RT-PCR assays performed using fundus, corpus, antrum and mucosa total RNA are shown in Fig. 34 and 35. A i and A 2 A receptor mRNA levels in all gastric regions were lower than those measured in the striatum (Fig. 32C). The level of A i receptor mRNA found in the corpus, antrum and mucosa were approximately 3-fold higher than in the fundus (Fig. 36). Among the gastric tissues, the highest level of A 2 A receptor mRNA was found in the corpus. The fundus, antrum and mucosa contained similar levels of A 2 A receptor mRNA, which were approximately 2 fold less than in the corpus. In the antrum and mucosa, the level of A i receptor mRNA was significantly higher than the level of A 2 A receptor mRNA, but in the fundus and corpus these levels were not significantly different. 2. Real-Time RT-PCR Fig. 37A shows a representative amplification plot generated by a Real-Time RT-PCR assay for measuring adenosine A i receptor mRNA. The corresponding standard curve is also shown in Fig. 37B. Results show that this assay was able to amplify and measure adenosine A i receptor mRNA within a 7 log range of A i receptor R N A standard concentrations. When the highest concentration of standard was used as the template ( l x l O 9 copies/pl), the reaction was affected by limiting reagents at cycle 22, and the plateau phase of the reaction was reached by cycle 26. However, this does not affect quantification since measurements were made during the exponential phase at the threshold cycle. The standard 0.75-, o n Q. 0.50-E S 0.25 a E o S \u00C2\u00A3 -0.25-1 (A C 0) \u00C2\u00A7> -0.50 u. 0.00--0.75-1 0.75 1.00 Log (initial A 1 competitor concentration) Sample A^amol/ng total RNA) undiluted 0.82 2 x diluted 0.88 4 x diluted 0.79 8 x diluted 0.79 0.75-, \u00E2\u0080\u00A22 0.50-5 m a. \u00C2\u00A3 a> *^ I 0.25-a> a E 8, o.oo-\"o\" \u00C2\u00A3 -0.25 '55 c \u00C2\u00A9 w -0.50 o 00 1.25 _o 75J Log (initial A 2 A competitor concentration) Sample (amol/(ig total RNA) undiluted 0.57 2 x diluted 0.50 4 x diluted 0.50 8 x diluted 0.54 Fig. 33. Effect of template concentrations on quantification of A, and A 2 A receptor mRNA by competitive RT-PCR. A[ (A) and A 2 A (B) receptor competitive RT-PCR experiments were performed using undiluted (\u00E2\u0080\u00A2), 2x (A), 4x (\u00E2\u0080\u00A2), or 8x (\u00E2\u0080\u00A2) diluted cDNA as the template. The cDNA samples were synthesized using a fixed amount (2.5 pg) of corpus total RNA and various amounts of A, or A 2 A receptor RNA competitor. 0.38 amol/|ig total RNA Log [intia! competitor concentration (amol)] B I 0.5 -0.25 2.0 amol/|ig total RNA Log [intial competitor concentration (amol)] 5 0.25 tj 0.00 & -Ol \u00C2\u00A3 -0.50 c M 2, -0.75 00 o -1 -1.00 y= 1.15X-0.56 RJ = 0.99 1.2 amol/ng total RNA 1.6 amol/ng total RNA Log [intial competitor concentration (amol)] Log [intial competitor concentration (amol)] Fig. 34. Quantification of gastric A[ receptor gene expression by competitive RT-PCR. Results of representative competitive RT-PCR assays for the fundus (A), corpus (B), antrum (C) and mucosa (D) are presented. The A, receptor gene expression levels measured in these tissues were 0.38, 2.0, 1.2, and 1.6 amol/pg total RNA, respectively. Fig. 35. Quantification of gastric A 2 A receptor gene expression by competitive RT-PCR. Results of representative competitive RT-PCR assays for the fundus (A), corpus (B), antrum (C) and mucosa (D) are presented. The A 2 A gene expression levels measured in these tissues were 0.36, 0.68, 0.59 and 0.29 amol/pg total RNA, respectively. \u00C2\u00A3 oo Fundus Mucosa: Antral/Corporeal 714 \u00C2\u00B177* A , A : 288 \u00C2\u00B1 63 Fig. 36. Competitive RT-PCR quantification of A, and A 2 A receptor gene expression in the rat stomach. Adenosine receptor mRNA levels (in thousand copies/pg total RNA) are shown for each region. *P < 0.05 when A, receptor mRNA levels are compared to A 2 A receptor mRNA levels, n > 4. 1.600 0 .000 - 0 . 2 0 0 0 2 4 6 8 10 12 14 16 18 2 0 2 2 2 4 2 6 2 8 3 0 3 2 34 3 6 3 8 4 0 Cycle Number B i i i n u r n i i i i i n n i i n m n i 111iiHI i i l i u m i i i i u i i i I IHUIII i i i i i m i i m i n i ) 1 0 * 2 1 0 * 3 1 0 * 4 1 0 * 5 10 *6 1 0 * 7 1 0 * 8 1 0 * 9 10*10 A 1 Receptor RNA Standard Concentration Fig. 37. Results of a representative A, receptor Real-Time RT-PCR assay. A: An amplification plot showing changes in normalized reporter emission after each cycle of PCR. Samples contained lx lO 9 (\u00E2\u0080\u00A2), l x lO 8 ( ), l x lO 7 ( ), l x lO 6 (\u00E2\u0080\u00A2), l x lO 5 (\u00E2\u0080\u00A2), l x l O 4 (\u00E2\u0080\u00A2), l x lO 3 (\u00E2\u0080\u00A2) initial copies of A t receptor RNA. B: The corresponding standard curve generated by the amplification plot. The threshold cycle (CT) represents the cycle where the change in normalized reporter emission exceed the threshold. curve generated by plotting the threshold cycle against the Log(Ai R N A standard concentration) was found to be linear with a R value of 0.99. Fig. 38 shows the amplification plot and standard curve obtained for a representative A 2 A receptor Real-Time PCR assay. The standard curve was also linear within a 7 log range of A 2 A R N A standard concentrations, with a Rvalue of 0.98. Fig. 39 shows the absolute levels of adenosine A i and A 2 A receptor mRNA measured in the striatum and in various regions of the stomach, including the fundus, corpus, antrum, mucosa, corporeal muscle and corporeal mucosa. In agreement with previous studies, adenosine A i and A 2 A receptor levels were highest in the striatum (Dixon et a l , 1996). Striatal A i and A 2 A receptor mRNA levels were approximately 2-fold and 70-fold higher than the highest gastric A i and A 2 A mRNA levels in the corporeal muscle region, respectively. In all gastric tissues, A i receptor mRNA levels were significantly higher than A 2 A receptor mRNA levels. In the fundus, corpus, antrum, mucosa, corporeal muscle and corporeal mucosa, the A i receptor mRNA levels were 3.4, 7.8, 8.3, 28.4, 14.4, and 14.6 times higher than A 2 A receptor mRNA levels, respectively. The highest and lowest levels of A i receptor mRNA were found in the corporeal muscle and corporeal mucosa, respectively. No significant difference in the A i receptor mRNA levels was observed among the corpus, antrum and the whole stomach mucosa. A 2 A receptor mRNA levels were not significantly different among the fundus, corpus and antrum, but were lowest in the whole gastric mucosa and in the corporeal mucosa. i i i i i i i i i i 2 4 6 8 I I I I I I I I I I I I I I I 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 Cycle Number B i i ilium i i II nm i MI nm iiiiiim iiiiiim iiniiiii MIIIIUI i in inn iiiimfi 1 0 * 2 1 0 * 3 10*4 1 0 * 5 1 0 * 6 1 0 * 7 1 0 * 8 1 0 * 9 10*10 A 2 A Receptor RNA Standard Concentration Fig. 38. Results of a representative A 2 A Real-Time RT-PCR assay. A: An amplification plot showing changes in normalized reporter emission after each cycle of PCR. Samples contained lx lO 9 (\u00E2\u0080\u00A2), l x lO 8 ( ), l x lO 7 ( ), l x lO 6 (\u00E2\u0080\u00A2), l x lO 5 (\u00E2\u0080\u00A2), l x lO 4 (\u00E2\u0080\u00A2), lx lO 3 (\u00E2\u0080\u00A2) initial copies of A 2 A receptor RNA. B: The corresponding standard curve generated by the amplification plot. The threshold cycle (CT) represents the cycle where the change in normalized reporter emission exceed the threshold. < z or 5 o E 3000 2500 2000 1500 D00 Striatum F u n d u s C o r p u s A n t r u m M u c o s a C o r p o r e a l C o r p o r e a l m u s c l e m u c o s a Striatum Fundus Corpus Antrum M u c o s a Corporeal Corporeal muscle mucosa Fig. 39. Real-Time RT-PCR quantification of gastric and striatal adenosine A 1 (A) and A 2 A (B) receptor gene expression. The level of A , and A 2 A receptor gene expression was highest in the striatum. A ! receptor gene expression was also significantly higher than A2A receptor gene expression in all gastric tissues examined (n > 4). Note the difference in the scale of the ordinate. IV. Effect of fasting on adenosine receptor, somatostatin and gastrin gene expression To examine whether the prandial state of the stomach affects adenosine receptor gene expression, rats were fasted for either 24 or 36 h, and gene expression was quantified using Real-Time RT-PCR. Real-Time RT-PCR was utilized for this study since this technique is more sensitive and specific than competitive RT-PCR (Heid et al., 1996). In these experiments, gastrin and somatostatin gene expression were also measured by Real-Time RT-PCR to determine if changes in adenosine receptor gene expression occur with changes in gastric peptide gene expression. 1. Effect of fasting on body weight and Total RNA concentration Rats subjected to 24 and 36 h of fasting were shown to exhibit significant weight loss compared to control animals (see Table 8). Fasting for 36 h, but not 24 h, was also shown to significantly decrease the mass of the antral tissue. Corporeal mucosa and corporeal muscle mass were not affected by either 24 or 36 h of fasting. The total R N A concentration (pg total RNA/mg tissue) was also altered by fasting in some regions (see Table 8). Fasting for 24 and 36 h significantly decreased the total RNA concentration in the antrum, while fasting for 24 h significantly decreased total R N A concentration in the corporeal muscle. 2. Effect of fasting on 18S rRNA and GAPDH gene expression The expression levels of housekeeping genes were measured and used as the endogenous control to standardize the amount of sample added to each reaction. Two Table 8: Effect of fasting and omeprazole treatment on body weight, tissue weight and total RNA concentration Fasting Omeprazole 24 h 36 h 1 day 3 day Change in weight (g) Test -28\u00C2\u00B12* -35\u00C2\u00B11* 5\u00C2\u00B12 17\u00C2\u00B13 Control 6\u00C2\u00B11 13\u00C2\u00B11 6\u00C2\u00B12 18\u00C2\u00B12 Tissue weight (mg) Antrum Test 155\u00C2\u00B110 117\u00C2\u00B110* 121\u00C2\u00B110 163=1=17 Control 163\u00C2\u00B116 182\u00C2\u00B120 115=1=11 153\u00C2\u00B126 Corporeal Mucosa Test 55=1=4 69\u00C2\u00B111 63\u00C2\u00B14 74\u00C2\u00B16 Control 63\u00C2\u00B16 65\u00C2\u00B110 65\u00C2\u00B18 83\u00C2\u00B19 Corporeal Muscle Test 792\u00C2\u00B123 815\u00C2\u00B141 689\u00C2\u00B168 658\u00C2\u00B153 Control 804\u00C2\u00B141 804\u00C2\u00B140 634\u00C2\u00B117 665\u00C2\u00B156 Total RNA (M-g/mg tissue) Antrum Test 1.1\u00C2\u00B10.1* l .OiO. l* 2.2\u00C2\u00B10.2 1.7\u00C2\u00B10.3 Control 1.4\u00C2\u00B10.1 1.7\u00C2\u00B10.2 2.1\u00C2\u00B10.2 2.0\u00C2\u00B10.3 Corporeal Mucosa Test 2.7\u00C2\u00B10.3 2.4=1=0.5 2.3\u00C2\u00B10.6 2.7=1=0.3 Control 2.8\u00C2\u00B10.3 3.9\u00C2\u00B10.6 2.2\u00C2\u00B10.2 2.1\u00C2\u00B10.2 Corporeal Muscle Test 4.7\u00C2\u00B10.4* 4.7\u00C2\u00B10.2 5.9=1=0.3 4.1\u00C2\u00B10.3 Control 5.9\u00C2\u00B10.3 4.6\u00C2\u00B10.2 6.6\u00C2\u00B10.6 4.5=1=0.4 *P < 0.05 when test animals were compared to controls using the unpaired Student's t-test, n > 8 per group. commonly used housekeeping genes were measured, G A P D H and 18S rRNA. Results show that G A P D H mRNA levels were not significantly altered after 24 h of fasting in all gastric tissues examined. However, after 36 h of fasting, significant decreases in G A P D H mRNA levels were observed in the antrum (53% of control), corporeal mucosa (56% of control) and corporeal muscle (70% of control) (Fig. 40). The levels of 18S rRNA were not altered by 24 or 36 h of fasting in all tissues studied (See Fig. 40B). Therefore, 18S rRNA levels were used as the endogenous standard to control for differences in total R N A concentration and R N A integrity in the following studies. 3. Effect of fasting on adenosine A i and A 2 A receptor expression Following a 24 h fast, A i receptor mRNA levels in the corporeal mucosa and antrum were significantly increased to 134 \u00C2\u00B1 11% and 157 + 19% of control levels, respectively (Fig. 41A). No significant change in A i receptor gene expression was observed in these tissues when the fasting period was extended to 36 h. In addition, fasting for 24 and 36 h did not alter A i receptor mRNA expression in the corporeal muscle. The effect of fasting on A 2 A receptor mRNA expression differed between the corporeal mucosa and the antrum. In the corporeal mucosa, adenosine A 2 A receptor expression was not altered by 24 h of fasting. However, following 36 h of fasting, A 2 A receptor expression was significantly reduced to 65 \u00C2\u00B1 6 % of the control levels (Fig. 41B). In the antrum, the gene expression of adenosine A 2 A receptor was significantly increased to 173 \u00C2\u00B1 2 1 % of control levels after a 24 h fast, but unchanged following a 36 h fast. Similar to A i receptor gene expression, fasting did not alter A 2 A receptor gene expression in the corporeal muscle. Antrum Corporeal Mucosa Corporeal Muscle 200 c 150 o Antrum Corporeal Mucosa Corporeal Muscle Fig. 40. Effect of fasting on gastric housekeeping gene expression. G A P D H mRNA (A) and 18S rRNA (B) levels were measured in rats fasted for 24 h (grey) or 36 h (black). Results are expressed as a percentage of the control (white) as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05; n > 7. Antrum Corporeal Mucosa Corporeal Muscle T T T T * 1 Antrum Corporeal Mucosa Corporeal Muscle Fig. 41. Effect of fasting on adenosine receptor gene expression. A[ receptor mRNA (A) and A 2 A receptor mRNA (B) levels were measured in rats fasted for 24 h (grey) or 36 h (black). Results are expressed as a percentage of the control (white) as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05; n > 7. 4. Effect of fasting on gastrin and somatostatin gene expression Antral gastrin gene expression was significantly reduced to 50 \u00C2\u00B1 9% and 54 + 10% of control levels after 24 and 36 h of fasting, respectively (Fig. 42A). Following fasting, changes in somatostatin gene expression (Fig. 42B) resembled changes in A 2 A receptor gene expression (Fig. 41B). In the corporeal mucosa, somatostatin mRNA levels were not affected by 24 h of fasting, but significantly decreased (53 \u00C2\u00B1 5 % of controls) after 36 h of fasting. In the antrum, somatostatin gene expression was significantly augmented (196 \u00C2\u00B1 27% of controls) after 24 h of fasting, but unaltered after 36 h of fasting. Similar to A 2 A receptor gene expression, fasting did not affect somatostatin gene expression in the corporeal muscle. When the changes in somatostatin gene expression were compared to changes in adenosine A 2 A receptor gene expression, a linear relationship with a correlation coefficient of 0.96 was observed (Fig. 43). V. Omeprazole treatment on adenosine receptor and gastric peptide gene expression To examine if the inhibition of gastric acid secretion alters adenosine receptor gene expression, rats were treated with 400 pmol/kg omeprazole daily for 1 or 3 days. Omeprazole inhibits gastric acid secretion by binding irreversibly to the proton pump, H + K + ATPase, on parietal cells (Hirschowitz et al., 1995). Gastrin and somatostatin gene expression were also measured to examine i f their expression is also altered with adenosine receptor gene expression. Antrum Corporeal Mucosa Corporeal Muscle Fig. 42. Effect of fasting on gastric peptide gene expression. Gastrin mRNA (A) and somatostatin mRNA (B) levels were measured in rats fasted for 24 h (grey) or 36 h (black). Results are expressed as a percentage of the control (white) as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05; n > 7. Fig. 43. Comparison between fasting-induced changes in somatostatin and adenosine A 2 A receptor gene expression. Changes in gene expression were measured after 24 or 36 h of fasting in various gastric tissues: corporeal mucosa (36 h; blue), corporeal mucosa (24 h; yellow), corporeal muscle (24 h, green); corporeal muscle (36 h; red), antrum (36 h; purple), antrum (24 h; orange). Each point represents the mean \u00C2\u00B1 SEM of at least 7 animals. 1. Effect of omeprazole treatment on 18S rRNA and GAPDH gene expression Omeprazole treatment for 1 or 3 days did not alter the weight of the rats, the mass of the gastric tissues, or the total RNA concentration of these tissues (see Table 8). Housekeeping gene expression levels were measured and used as the endogenous control. Similar to the effects of fasting, omeprazole treatment also altered G A P D H mRNA levels in some regions. G A P D H gene expression was significantly decreased to 65\u00C2\u00B15% of control levels in the corporeal mucosa after 3 days of treatment, and significantly increased to 130\u00C2\u00B19% in the corporeal muscle after 1 day of treatment (Fig. 44A). However, levels of 18S rRNA were not altered by 1 or 3 days of omeprazole treatment (Fig. 44B). Therefore, this housekeeping gene was used as the endogenous control for the Real-Time RT-PCR quantification of A i and A 2 A receptor, somatostatin and gastrin mRNA levels. 2. Effect of omeprazole treatment on adenosine Ai and A 2 A receptor expression Omeprazole significantly decreased A i receptor gene expression in the antrum to 66\u00C2\u00B112% and 47\u00C2\u00B18% of control levels after 1 day and 3 days of treatment, respectively (see Fig. 45A). In the corporeal mucosa, A i receptor gene expression was significantly decreased to 62\u00C2\u00B16% of control levels after 1 day of omeprazole treatment, but changes were not apparent after 3 days of treatment. A 2 A receptor gene expression was significantly decreased to 57\u00C2\u00B17% of control levels in the antrum after 3 days of treatment (Fig. 45B). In the corporeal mucosa, A 2 A receptor gene expression was also decreased to 67\u00C2\u00B114% of control levels after 1 day of omeprazole treatment, but changes were not apparent 3 days after treatment. Both A i and A 2 A receptor A 2 0 0 -\"5 175 -C Antrum Corporeal Mucosa Corporeal Muscle Antrum Corporeal Mucosa Corporeal Muscle Fig. 44.: Effect of omeprazole treatment on gastric housekeeping gene expression. G A P D H mRNA (A) and 18S rRNA (B) levels were measured in rats treated with omeprazole for 1 (grey) or 3 days (black). Results are expressed as a percentage of the control (white), as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05; n>7. Antrum Corporeal Mucosa Corporeal Muscle 200 i B _ 2 175-c o Antrum Corporeal Mucosa Corporeal Muscle Fig. 45. Effect of omeprazole treatment on adenosine A , and A 2 A receptor gene expression. Aj receptor (A) and A 2 A (B) receptor mRNA levels were measured in rats treated with omeprazole for 1 (grey) or 3 (black) days. Results are expressed as a percentage of the control (white) as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05, ** P <0.01, ***P < 0.001 ; n > 7. gene expressions were not altered by either 1 or 3 days of omeprazole treatment in the corporeal muscle. 3. Effect of omeprazole treatment on gastrin and somatostatin gene expression Antral gastrin gene expression was significantly increased to 201 \u00C2\u00B1 18% and 268 \u00C2\u00B142% of control levels after 1 and 3 days of omeprazole treatment, respectively (Fig. 46A). In contrast, antral somatostatin gene expression was significantly reduced to 71\u00C2\u00B16% and 58\u00C2\u00B14% of controls levels, and corporeal mucosal somatostatin gene expression was reduced to 54\u00C2\u00B113% and 54\u00C2\u00B110% of control levels after 1 day and 3 days of treatment, respectively (Fig. 46B). In the corporeal muscle, somatostatin gene expression was not altered by 1 day of treatment. However, after 3 days of treatment, somatostatin gene expression was significantly reduced to 45\u00C2\u00B16% of control levels. VI. Omeprazole on adenosine agonist-induced changes in IRG and SLI release The effect of omeprazole treatment on adenosine-agonist induced changes in IRG and SLI release was examined. After 1 or 3 days of omeprazole treatment, stomachs were perfused with either CPA (Ai-selective agonist) or CGS 21680 (A2A-selective agonist) at a concentration of 0.1 p M or 1 pM. The lower concentration of 0.1 p M was chosen based on the estimated E C 5 0 value of CPA (0.067 pM) and CGS 21680 (0.06 pM) in inhibiting IRG and stimulating SLI release, respectively. The effect of 1.0 p M concentration was examined since this concentration of CPA and CGS 21680 elicited Antrum 200 Antrum Corporeal Mucosa Corporeal Muscle Fig. 46. Effect of omeprazole treatment on gastric peptide gene expression. Gastrin (A) and somatostatin (B) mRNA levels were measured in rats treated with omeprazole for 1 (grey) or 3 (black) days. Results are expressed as a percentage of the control (white), as described in the Methods section. Statistics were performed using the Student's unpaired t-test. * P < 0.05, ** P O . 0 1 , ***p<0.001 ;n>7. maximal inhibition and stimulation of IRG and SLI release, respectively (see Section I of this chapter). The effect of CGS 21680 on IRG release was not examined since this compound did not alter IRG release (see section 1.1). 1. Effect of omeprazole on CPA-induced changes in IRG release Fig. 47 shows the effect of 1 p M CPA on IRG release in animals treated with omeprazole for 3 days. The administration of CPA resulted in a significant inhibition of IRG release in both control and treated animals. However, there was no difference in the release of IRG between these two groups. Results of CPA-induced IRG release in omeprazole-treated and control animals are summarized in Fig. 48. There were no changes in CPA-induced IRG release between treated and control animals. 2. Effect of omeprazole on agonist-induced changes in SLI release Fig. 49 shows the effect of 1 p M CGS 21680 on SLI release in control and 3 day omeprazole-treated animals. In these experiments, SLI release was enhanced by CGS 21680 in both treated and control animals. However, the CGS 21680-induced SLI release was significantly attenuated by 3 day omeprazole treatment. Results of CGS 21680-induced SLI release in control and 1 or 3 day omeprazole-treated animals are summarized in Fig. 50. As described earlier, the administration of 0.1 p M CPA significantly inhibited gastric SLI release, while the administration of 1 p M CPA significantly stimulated SLI release (see Section I). These observations were also apparent in the current experiments (Fig. 51). The inhibition of SLI release induced by 0.1 p M CPA was not altered by either 1 or 3 days of 1 2 3 4 5 6 7 8 9 10 5 min periods B 200-\u00E2\u0080\u0094 150-01 y V - * . v Fig. 64. Double staining of A 2 A R-IR with somatostatin-IR in the gastric mucosa. Confocal images showing the co-localization of A 2 A R-IR (red) with somatostatin-IR (green) in the rat corporeal (A-C) and antral (D-F) mucosa. Cells expressing both A 2 A R-IR and somatostatin-IR are indicated by arrows and appear yellow (C & F). Somatostatin-IR cells not expressing A 2 A R-IR are indicated by the arrowheads, (z-step = 0.5 pm, scale bars = 25 pm for all images). o A \ B S i C \ \u00E2\u0080\u00A2 % r P D E V F fk. - \u00E2\u0080\u00A2 \u00E2\u0080\u00A2 \" \u00E2\u0080\u00A2\u00E2\u0080\u00A2 \u00E2\u0080\u00A2- A If\" * * CM \" i Fig. 65. Double staining of A 2 A R-IR with somatostatin-IR in the gastric muscle of the corpus. Confocal images show co-localization of A 2 A R-IR (red) with somatostatin-IR (green) in the myenteric plexus (A-C) and circular muscle (D-F) of the rat corpus. Co-localization does occur (yellow; arrows); however, this was only observed occasionally. C M = circular muscle, (z-step = 0.5 pm, scale bars = 25 pm). A ^ ( u B c L M ^ D E \ F \ C M Fig. 66. Double staining of A 2 A R-IR with somatostatin-IR in the gastric muscle of the antrum. Confocal images show co-localization of A 2 A R-IR (red) with somatostatin-IR (green) in a cell body of the myenteric plexus (A-C) and nerve fibers in the circular muscle (D-F) of the rat antrum. Co-localization was shown to occur (yellow), however this was not frequent. L M = longitudinal muscle, CM = circular muscle, (z-step = 0.5 pm, scale bars = 25 pm). to A 1 \ V * 1 c I / i r * 1 Fig. 67. Double staining of A 2 A R-IR with gastrin-IR. Confocal images show the lack of co-localization of A 2 A R-IR (A) with gastrin-IR (B) in the antrum. Cells containing A 2 A R-IR (arrows) do not express gastrin-IR (arrowheads), (z-step = 0.5 pm; scale bars = 25 pm). Fig. 68. Double-staining of A 2 A R-IR (red) with H+K+-ATPase pMR (green). A 2 A R-IR (A; arrow) is shown not to co-localize with H +K +-ATPase (3-IR (B; arrowhead) in the corporeal mucosa, (z-step = 0.5 pm; scale bar = 25 pm). Fig. 69. Double staining of A 2 A R-IR (red) with VWF-IR (green). Confocal images show the co-localization of A 2 A R-IR with VWF-IR in the myenteric plexus region of the corpus (A-C; arrows) and in the circular muscle of the antrum (D-F; arrows). Regions expressing both immunoreactivities appear yellow, (z-step = 1 pm; scale bars = 25 pm). Fig. 70. Localization of A 2 A R-IR relative to VWF-IR in the corporeal muscle. Images shown in A-C represent the immunoreactivity of 16 stacked sections. Images shown on D-F and G-I represent the immunoreactivity taken from a single section (z-step =1.0 pm). A 2 A R-IR (red) is localized with VWF-IR (green), and is also found to be expressed in close proximity to VWF-IR. The A 2 A R-IR indicated by the arrows on panel D and G may represent A 2 A receptors localized on on vascular smooth muscle cells, (scale bar s= 50 pm). A \ \u00C2\u00B01 % C . , L M C M \ V , Y , \u00C2\u00A7 $ is E > | L M > v j C M Fig. 71. Double staining of A 2 A R-IR (red) with PGP 9.5-IR (green). Confocal images show the co-localization of A 2 A R-IR with PGP 9.5-IR in nerve fibers of the myenteric plexus and circular muscle of the corpus (A-C) and antrum (D-F) (arrows). Cells expressing both immunoreactivities appear yellow (C and F). (CM = circular muscle; LM= longitudinal muscle; z-step = 0.5 pm; scale bars = 25 pm.). or. - J Chapter 4: Discussion This study is the first to establish the significant roles of adenosine A i and A 2 A receptors in modulating gastric somatostatin and gastrin release. Biological and immunohistochemical results suggest that adenosine may inhibit IRG release, at least in part, by activating Aj receptors on the G-cells and may alter SLI release by activating both A i and A 2 A receptors on the D-cells. Results of the molecular biological experiments further demonstrate that the adenosine A i and A 2 A receptors are structurally identical to those in the brain, and are expressed in all functionally and morphologically distinct regions of the stomach. The expression and function of these receptors can be altered by different physiological states of the stomach, suggesting that adenosine receptor expression may be changed under different gastric secretory states. This discussion is organized into the following sections: Section I, the adenosine receptor subtypes involved in regulating gastric IRG and SLI release; Section II, the distribution of adenosine receptor mRNA in functionally distinct regions of the stomach and the structure of the gastric adenosine receptors; Section III, the quantitative RT-PCR techniques developed to measure gastric adenosine receptor gene expression; Section TV, the changes in A i and A 2 A receptor gene expression and function by alterations in the physiological states of the stomach; Section V , the significant implications of the cellular localization and distribution of gastric A i and A 2 A receptor, as illustrated by immunohistochemistry experiments. I. Alteration of IRG and SLI release by selective adenosine analogs While the effect of adenosine on inhibiting IRG release (DeSchryver-Kecskemeti et a l , 1981; Harty and Franklin, 1984; Kwok et al., 1990) and stimulating SLI release (Kwok et al., 1990) has been described, the adenosine receptor subtypes involved have not been identified with certainty. The present study, therefore, was performed to elucidate the adenosine receptor(s) involved in these actions. Using adenosine receptor subtype-selective analogs, the present study demonstrates that the inhibition of IRG release was mediated by the activation of adenosine A i receptors, while the release of SLI was inhibited and stimulated by the activation of the A i and A 2 A receptors, respectively. 1. Effect of adenosine receptor-selective analogs on IRG release To determine the adenosine receptor subtype involved in the inhibition of IRG release, the effect of various adenosine analogs was examined in the isolated vascularly perfused rat stomach preparation. This preparation has been used in our earlier investigation of the effect of adenosine on IRG release (Kwok et al., 1990). Adenosine was found to suppress IRG release concentration-dependently. In the present study, only the administration of CPA significantly inhibited IRG release. CPA is a potent A i receptor agonist that exhibits a greater binding affinity for the A\ receptor than the A 2 and A3 receptors (Klotz et al., 1998; Ji and Jacobson, 1999). The administration of CGS 21680 and IB-MECA did not significantly alter basal IRG release. CGS21680 is a potent A 2 A receptor agonist, which exhibits a 140-fold selectivity for A 2 A receptors over A i receptors (Hutchison et al., 1989), and IB-MECA is a potent A 3 receptor agonist (Gallo-Rodriguez et al., 1994) that exhibits a greater affinity for A3 receptors than A i and A 2 receptors. CPA significantly inhibits basal IRG release starting at a concentration of 1 nM. However, CGS 21680 and IB-M E C A were unable to elicit an effect even at a concentration of 100 nM. At this concentration, both analogs have been shown to potently elicit various A 2 A and A3 receptor-mediated actions in the rat (Hutchison et al., 1989; Zucchi et al., 2001). The potent inhibitory effect of CPA and the lack of effect of CGS 21680 and IB-MECA suggest that the A i receptor is likely the subtype involved in modulating IRG release. To further test this hypothesis, the effect of selective A i and A 2 receptor antagonists on adenosine-induced inhibition of IRG release was examined. DPCPX, a selective A i receptor antagonist that exhibits a 700-fold preference for the A i receptor over the A 2 receptor (Bruns et al., 1987), completely abolished the inhibition of IRG release induced by adenosine. D M P X , an A 2 receptor-selective antagonist (Seale et al., 1988), however, did not alter either basal or adenosine-induced IRG release. The complete blockade of the inhibitory effect of adenosine by DPCPX and the lack of an effect by D M P X further support the suggestion that the A i receptor subtype is responsible for the inhibition of IRG release. Results of these experiments do not identify the site at which adenosine acts to produce its inhibitory effect, but several sites are suggested. Using immunohistochemistry, the present study demonstrates that A i receptors were localized on nerve fibers throughout the enteric plexi and on gastrin-containing G-cells. In the enteric plexi, Christofi and co-workers have suggested that these receptors reside in presynaptic nerve terminals and function to modulate neurotransmitter release (Christofi et al., 1992). In addition, adenosine has been shown to inhibit the release of acetylcholine and noradrenaline in the guinea pig enteric plexi (Barajas-Lopez et al., 1991; Christofi et al., 1992), most likely through activation of A i receptors at the presynaptic nerve terminal (Nitahara et al., 1995). In the isolated vascularly perfused rat stomach, IRG release is regulated by cholinergic and adrenergic agents (Koop et al., 1982, 1983). Thus, adenosine may regulate IRG release indirectly by modulating neurotransmitter release in the enteric plexi. Evidence also suggests that adenosine may regulate IRG release by acting directly on the G-cells. In rat antral mucosal fragments, adenosine inhibits basal and carbachol-stimulated gastrin release (Harty and Franklin, 1984). Adenosine has also been shown to inhibit forskolin-stimulated gastrin release in isolated canine G-cells (Schepp et al., 1990). Furthermore, the use of subtype-selective analogs specifically implicated the A i receptors in mediating this inhibitory effect (Schepp et al., 1990). Additional support for a direct action on G-cells is provided by the present immunohistochemistry experiments which demonstrate intense A i receptor staining on all antral G-cells of the rat stomach. The presence of adenosine A 2 A receptors on G-cells was not detected. This observation lends support to the biological experiments, which demonstrated the lack of effect of CGS 21680 on IRG release. In summary, these results clearly show that the adenosine A i receptor subtype is involved in suppressing IRG release in the rat. In addition, I propose that adenosine may regulate IRG release directly by acting on the G-cells, and/or indirectly by modulating neurotransmitter release in the enteric plexi. 2. Effect of adenosine receptor-selective analogs on gastric SLI release The adenosine receptor subtype(s) involved in regulating SLI release was also examined by studying the effect of various adenosine analogs. In the isolated vascularly perfused rat stomach, our laboratory has demonstrated that adenosine may inhibit or augment gastric SLI release depending on the concentration used (Kwok et al., 1990). Results of the present study suggest that the stimulatory effect of adenosine is most likely mediated by activation of the A 2 A receptor. The rank order of potency of adenosine analogs in augmenting SLI release was shown to be: CGS 21680 > IB-MECA > CPA. At 1 \M, CGS 21680, an A 2 A receptor-selective agonist, was shown to elicit an increase in SLI release (% change) that was approximately 26-fold and 7-fold greater than the increase elicited by the same concentration of CPA and IB-MECA, respectively. CGS 21680 potently stimulated gastric SLI release with an EC 5o of 0.06 \xM. Although CGS 21680 is selective for the A 2 A receptor, it can act on both A 2 A and A 2 B receptors. The stimulatory effect of CGS 21680 on SLI release, however, is not likely due to the activation of A 2 B receptors since this drug has a much lower affinity for the A 2 B receptors (Brackett and Daly, 1994). In the present study, the EC50 of CGS 21680 in augmenting SLI release was found to be in the sub-micromolar range (0.06 uM), as suggested for A 2 A receptor-mediated actions (Brackett and Daly, 1994). In PC 12 membranes expressing A 2 A receptors, the stimulation of adenylate cyclase activity by CGS 21680 was characterized by an E C 5 0 of 0.07 p M (Brackett and Daly, 1994). In NIH 3T3 membranes expressing A 2 B receptors, 100 \xM and 3 \xM CGS 21680 was shown have little effect on adenylate cyclase activity and cAMP production, respectively (Brackett and Daly, 1994; Klotz et al., 1998). Thus, actions mediated by the A 2 A and A 2 B receptors can be differentiated by the potency of CGS 21680 in eliciting those effects. The sub-micromolar EC50 value of CGS 21680 in augmenting SLI release suggests the involvement of A 2 A receptors, but not A 2 B receptors. The involvement of A 2 A receptors is also supported by the effect of various selective adenosine antagonists on adenosine and adenosine agonist-induced SLI release. When administered at 1 \xM, D M P X , an A 2 receptor antagonist, completely abolished the stimulatory effect elicited by CPA (1 \xM), and significantly attenuated IB-MECA- ( lpM) and CGS 21680- (0.01 and 0.1 uM) stimulated SLI release. The inhibitory effect of D M P X on CGS 21680- (0.01 and 0.1 pM) induced SLI release was concentration-dependent. To ascertain whether the A 2 A receptors are involved in the augmentation of SLI release, the effect of the potent non-xanthine A 2 A receptor antagonist Z M 241385 (Poucher et al., 1995) on SLI release was tested. This A 2 A receptor-selective antagonist was able to completely abolish the stimulatory effect of CGS 21680 and adenosine on SLI release when administered at a concentration of 1 uM. This compound is 6,700-fold more selective for A 2 A receptors than A i receptors (Poucher et al., 1995), while D M P X is only 4-fold more selective for A 2 receptors than A i receptors (Seale et al., 1988). These results support the proposal that A 2 A receptors are involved in stimulating SLI release. The A i receptors are not likely involved in the augmentation of SLI release since D M P X and Z M 241385 completely abolished CPA-induced SLI release. Furthermore, the A i receptor-selective antagonist, DPCPX, did not alter the effect of CGS 21680 on SLI release. The involvement of the A3 receptor in augmenting SLI release is also unlikely since the stimulatory effect induced by IB-MECA was sensitive to blockade by D M P X and was completely abolished by Z M 241385. In addition, studies have shown that adenosine-induced SLI release can be abolished by the administration of the non-selective antagonist 8-PT (Kwok et al., 1990). Since the rat A3 receptors were shown to be resistant to blockade by xanthine derivatives (Zhou et al., 1992) and A3 receptor-mediated effects in the rat were resistant to blockade by 8-PT (van Galen et al., 1994; Peachey et al., 1996), the involvement of A 3 receptors in augmenting SLI release is unlikely. The stimulatory action of CPA and IB-MECA on SLI release, thus, may be due to their non-specific effect on A 2 A receptors when administered at high concentrations. Potent A 2 B receptor-selective antagonists were not available, so it is not possible to thoroughly examine the role of the A 2 B receptor in augmenting SLI release. However, the EC50 of CGS 21680 in eliciting this effect strongly suggests that the A 2 B receptors are not involved. Thus, the augmentation of gastric SLI release by adenosine is most likely mediated solely by the A 2 A receptor subtype. In addition to the stimulatory effect of adenosine on SLI release, studies have shown that at low concentrations (10 nM), adenosine can also significantly inhibit SLI release (Kwok et al., 1990). Similar results were obtained in the present experiments, in which a low concentration of CPA (0.1 pM) was shown to significantly inhibit SLI release. It is possible that CPA acts specifically on A] receptors to inhibit SLI release at low concentrations, while this compound acts non-specifically on A 2 A receptors to stimulate SLI release at higher concentrations. This proposal agrees well with studies demonstrating that, in tissues containing several adenosine receptor subtypes, A i receptors are preferentially activated by lower levels of adenosine. In the rat, studies have shown that adenosine elicits A j - , A 2 A - , A 2 B - , and A 3 - mediated effects with EC50 values of 73, 150, 5100, and 6500 nM, respectively (Daly and Padgett, 1992; Zhou et al., 1992; Peakman and Hil l , 1994). The present proposal that the inhibition of SLI release is mediated by A i receptors also explains the observation that basal SLI release was lower in the presence of Z M 241385 and adenosine than in the presence of Z M 241385 alone. When administered alone, Z M 241385 may inhibit SLI release by blocking the effect of endogenous adenosine on the A 2 A receptors, thereby unmasking the A i receptor-mediated inhibition of SLI release. When Z M 241385 is administered together with adenosine, adenosine will only act on A i receptors to inhibit SLI release. Consequently, the inhibitory effect of adenosine is enhanced in the presence of Z M 241385. These results demonstrate that adenosine may play a dual role in the regulation of SLI release in the rat. Activation of A i and A 2 A receptors may result in inhibition and stimulation of SLI release, respectively. Results of the perfusion experiments do not clearly identify where adenosine acts to regulate SLI release, but results from the immunohistochemistry experiments suggest several possibilities. These studies demonstrate that A i and A 2 A receptors were expressed in the gastric vasculature, the enteric plexi, and D-cells. Previous studies performed in our laboratory have shown that the stimulatory effect of adenosine on SLI release is not likely due to its vasodilatory action in the stomach. Like CGS 21680, nitroprusside, a vasodilator, was found to decrease perfusion pressure in the isolated vascularly perfused rat stomach. However, nitroprusside was shown not to alter SLI release (Yip and Kwok, 2004). The presence of A 2 A receptors on enteric nerves suggests that adenosine may alter SLI release by regulating neurotransmitter release since cholinergic and p-adrenergic agents are capable of modulating gastric SLI release (Saffouri et al., 1980; Koop et al., 1983). Earlier work performed in our laboratory has demonstrated that the cholinergic blockers, atropine and hexamethonium, and the P-adrenergic blocker, propranolol, did not alter adenosine-induced changes in SLI release (Kwok et al., 1990). Thus, it is unlikely that adenosine stimulates SLI release through A 2 A receptor-mediated changes in acetylcholine and noradrenaline release. The possibility that adenosine may alter gastric SLI release through non-cholinergic and non-adrenergic neurotransmission cannot be eliminated since both A i and A 2 A receptors were localized on nerve fibers throughout the enteric plexi and muscle layers. The enteric peptides CGRP (Ren et al., 1993; Manela et al., 1995) and VIP (Chiba et al., 1980) can stimulate the release of gastric SLI. Thus, the possibility that adenosine modulates the release of these peptides to subsequently enhance SLI release remains to be examined. However, the localization of both A i and A 2 A receptors on the corporeal and antral somatostatin-secreting D-cells strongly suggests that adenosine can regulate SLI release, at least in part, by acting directly on the gastric D-cells. The presence of both receptors on the D-cells further lends support to our proposal that adenosine may inhibit and stimulate SLI release by acting on the A i and A 2 A receptors, respectively. 3. Regulation of gastric SLI and IRG release by adenosine Results from the present study suggest that endogenous adenosine may be involved in regulating basal SLI release. The administration of D M P X and Z M 241385 significantly inhibited basal SLI release, suggesting that basal SLI release may be maintained by activation of A 2 A receptors. When EHNA, an A D A inhibitor (Mendelson et al., 1983), was administered, both basal and adenosine-stimulated SLI release were enhanced. A D A is expressed in the gastric epithelia (Witte et al., 1991) and is responsible for metabolizing adenosine to inosine (Geiger et al., 1997). The enhancement of basal and adenosine-stimulated SLI release by E H N A suggests that A D A may play a role in regulating endogenous adenosine levels in the rat stomach. The significant regulatory role of A D A in maintaining adenosine concentrations has been demonstrated in ADA-deficient mice, in which a lack of A D A activity has been shown to significantly increase the endogenous adenosine level in the stomach (Xu and Kellems, 2000). Previous studies performed in our laboratory have also demonstrated that the adenosine uptake inhibitor dipyridamole can enhance basal and adenosine-induced SLI release (Kwok et al., 1990), indicating that endogenous adenosine is also regulated by the cellular reuptake of this nucleoside through equilibrative adenosine transporters. The extracellular adenosine level in the isolated vascularly perfused rat stomach may, therefore, be regulated by the cellular reuptake and metabolic degradation of adenosine. Since E H N A and dipyridamole can alter basal and adenosine-induced SLI release, endogenously released adenosine may be involved in maintaining the release of SLI and IRG under normal physiological states of the stomach. Although the origin of endogenous adenosine in the present study has not been determined, extracellular adenosine in the stomach may originate from several sources. In the perfused stomach preparation, endogenous adenosine may be derived from the metabolism of ATP released into the stomach (see Chapter 1, Section I, 1.3). ATP may be released by enteric nerves or smooth muscle cells of the gastrointestinal tract (Burnstock, 1975; Nitahara et al., 1995). In the stomach, purinergic nerves of the myenteric plexus project to the circular and longitudinal muscles (Burnstock, 1975). ATP was demonstrated to be co-released with other neurotransmitters such as nitric oxide (Selemidis et al., 1997), VIP (Furness et al., 1995) and pituitary adenylate cyclase-activating polypeptide (Furness et al., 1995; Imoto et al., 1998), and may be released by nerve depolarization (Su et al., 1971). In guinea pigs, ATP was also been shown to be released from smooth muscle cells of the ileum (Nitahara et al., 1995). Thus, ATP released from gastric neurons and smooth muscle cells may be an important source of endogenous adenosine in the rat stomach. Adenosine derived from the metabolism of ATP, thus, may depend on neural activity of the enteric plexi. The endogenous adenosine level can change under various conditions. During normoxic conditions, endogenous adenosine concentrations are in the nanomolar range. In the whole blood and plasma of rats, adenosine concentrations have been found to be 79 nM (Phillis et al., 1992) and 320 nM (Conlay et al., 1997), respectively, while in the jejunum, the venous adenosine concentration was found to be 62 nM (Sawmiller and Chou, 1990). Significant elevations in adenosine concentrations can occur during periods of stress, such as during hypoxia, ischemia, and hypoglycemia. The production of ATP is reduced when low glucose or oxygen levels produce an elevated level of A M P (Milusheva et al., 1996), which stimulates 5' nucleotidase activity to promote adenosine formation (Worku and Newby, 1983). The level of adenosine, thus, may increase to micromolar concentrations during ischemia (Latini et al., 1999). For example, ischemia has been shown to increase the adenosine concentration in the striatum from a basal level of 41- 210 nM (Ballarin et al., 1991; Pazzagli et al., 1995; Melani et al., 1999) to an ischemic level of 3 \xM (Melani et al., 1999). The physiological state of the stomach may also alter the endogenous level of adenosine. In humans, the acidity of the stomach may alter adenosine concentrations by altering A D A and 5' nucleotidase activity. In particular, in the fundic mucosa, A D A activity and gastric acid output have been observed to change in parallel (Namiot et al., 1990). Hypersecretors of acid exhibited increased A D A activity, while patients suffering from achlorhydria exhibited lowered A D A activity (Namiot et al., 1990). The activity of 5' nucleotidase was found to be reduced in the gastric juice of patients suffering from gastric cancer, gastric ulcers, and gastritis (Durak et al., 1994). The activity of A D A and 5' nucleotidase, therefore, may be regulated by the acidity of the stomach. This observation is consistent with studies, in the rat skeletal muscle, showing that increased acidity can stimulate 5' nucleotidase activity (Cheng et al., 2000). The prandial state of the stomach may also alter the adenosine level. In the canine jejunum, the venous adenosine level was shown to be increased by the presence of food in the intestinal lumen (Sawmiller and Chou, 1990). Changes to the prandial state or acidity of the stomach may, therefore, alter gastric adenosine concentrations and subsequently influence gastric actions mediated by adenosine, such as the regulation of SLI and IRG release. As discussed earlier, activation of A i receptors has been shown to inhibit LRG and SLI release, while activation of A 2 A receptors has been shown to stimulate SLI release. A i receptors are preferentially activated by lower concentrations of adenosine, while A 2 A receptors are preferentially activated by higher concentrations of adenosine. Therefore, during conditions that lead to an increase in the endogenous adenosine concentration, A 2 A receptors may be preferentially activated to stimulate SLI release, while during conditions that would lead to a decrease in the endogenous adenosine concentration, A i receptors may be preferentially activated to inhibit SLI and IRG release. II. Distribution and structure of gastric adenosine receptor mRNA The mRNAs of all four adenosine receptors are expressed in extremely low levels within the whole rat stomach (Dixon et al., 1996). The localization of adenosine receptors on functionally and morphologically distinct regions of the stomach is unknown. However, functional studies suggest that these receptors may be localized on specific regions of the stomach. For example, studies demonstrating the protective effect of adenosine on gastric mucosal damage (Geiger and Glavin, 1985; Westerberg and Geiger, 1987; Cho and Ogle, 1990; Cho et al., 1991; Bozkurt et al., 1998) and studies showing the inhibitory and stimulatory effect of adenosine on gastrin and somatostatin release, respectively (Kwok et al., 1990), suggest that the adenosine receptors may be expressed on the gastric mucosa. The current study is the first to demonstrate the localization of all four adenosine receptors in the gastric corpus, antrum, and mucosa of the rat using RT-PCR. The specificity of each RT-PCR reaction was confirmed by restriction enzyme digestion using multiple restriction enzymes. Small fragments generated by restriction enzyme digestion (< 150 bp) were often not detected by agarose gel electrophoresis due to their low intensity fluorescence. The larger fragments (>150 bp) were easily detected, and their sizes compared well with the expected sizes, differing by < 9%. The A i , A 2 A , A 2 B and A3 receptor RT-PCR reactions established for this study, therefore, are highly specific. The presence of adenosine receptors in the corpus, antrum and mucosa may have important functional implications since these regions contain cells which secrete gastric acid, somatostatin and/or gastrin. The parietal cells, G-cells and D-cells reside in the gastric mucosa of the stomach (Dockray et al., 1996; Sachs et al., 1997). The corpus contains parietal cells and D-cells, while the antrum contains G-cells and a population of D-cells that are distinct from those in the corpus. In addition to their presence in these tissues, all four adenosine receptors were also expressed in the gastric fundus, the non-glandular region of the stomach that is responsible for the accommodation of food. The presence of A i and A 2 A receptor mRNA throughout the stomach suggests the possibility that adenosine receptors may be expressed on various sites of the stomach, including the parietal cells, G-cells and D-cells, nerve fibers and vasculature. Indeed, the localization and distribution of adenosine receptors were confirmed on some of these structures by the present immunohistochemistry experiments. The structure of the A i and A 2 A receptors was also examined in the present study by RT-PCR and by cloning and sequencing of the RT-PCR amplicons. Only one A i and A 2 A receptor amplicon is produced when RT-PCR was performed using primers that span the entire coding region of the receptor. Northern blot analysis has previously demonstrated the existence of two A i receptor transcripts in various tissues of the rat, including the brain and stomach (Mahan et al., 1991; Reppert et al., 1991). Two distinct A 2 A receptor transcripts have also been demonstrated in rat PC12 cells (Chu et al., 1996). The A i and A 2 A receptor genes both contain 2 exons, which are separated by one intron (Ralevic and Burnstock, 1998). The synthesis of only one RT-PCR amplicon in the present study demonstrates that the intron is not alternatively spliced to generate variant forms of the A i and A 2 A receptor in the rat stomach. Cloning and sequencing of the mucosal A i and A 2 A receptor amplicons further demonstrate that the coding region of these receptors is identical to published sequences in the rat brain (Mahan et al., 1991; Fink et al., 1992). Thus, only one form of the gastric A i and A 2 A receptors exists in the stomach. The multiple A i and A 2 A receptor mRNA transcripts, detected by previous Northern blot analysis studies (Mahan et al., 1991; Reppert et a l , 1991; Chu et al., 1996) may arise from differences in the non-coding regions. In rat PC 12 cells, the A 2 A receptor gene was shown to express two different promoters (Chu et al., 1996). These two promoters drive the synthesis of two distinct A 2 A receptor transcripts that differ in the 5'UTR. Similarly, in humans, two separate promoters were found in the A i receptor gene (Ren and Stiles, 1995). These promoters drive the synthesis of two distinct A i receptor transcripts which were also shown to differ in the 5'UTR. It is unclear whether this also occurs in the rat stomach since the promoter regions of the rat A i receptor gene have not yet been identified. Unlike the A i and A 2 A receptors, two forms of the A3 receptor exist in various rat tissues (Zhou et al., 1992; Sajjadi et al., 1996). The predominant form was cloned by Zhou and coworkers (1992), and was shown to contain 320 amino acids. The alternate form, the A 3 i receptor, was cloned by Sajjadi and coworkers (1996) and was found to be expressed in the rat brain and spleen (Zhou et al., 1992; Sajjadi et al., 1996). This form contains a 17 amino acid insertion which is produced by alternative splicing of the intron. The RT-PCR primers used in the present study were able to detect both the A3 and A 3 i receptor transcripts. Results of our study demonstrate that alternative splicing was not shown to occur in any region of the rat stomach since only one A3 receptor transcript was detected. Cloning and sequencing of the PCR amplicon revealed that the A 3 receptor, but not the A3i receptor, is expressed in the rat stomach. A silent mutation was also shown to occur at position 492 (second transmembrane domain). This mutation has previously been observed in the A3i receptor transcript of the rat brain (Sajjadi et al., 1996). Other point mutations reported in the A 3 i receptor, however, were not observed in this region of the rat mucosal A 3 receptor gene. Results of these studies demonstrate that all four adenosine receptors are expressed in functionally distinct regions of the rat stomach, and that each subtype exists in only one structural form. Furthermore, the gastric A i and A 2 A receptors are structurally identical to the A i and A 2 A receptors of the brain in the rat. III. Quantification of Ai and A 2 A receptor gene expression 1. Competitive RT-PCR and Real-Time RT-PCR assay development In the rat stomach, the level of adenosine A i and A 2 A receptor mRNA is extremely low (Dixon et al., 1996). Quantification of gastric adenosine receptor expression using conventional techniques such as Northern blot and slot blot is not possible since moderate levels of mRNA expression and relatively large sample sizes are required. In the present study, competitive RT-PCR and Real-Time RT-PCR techniques were developed to measure absolute A i and A 2 A receptor gene expression in different regions of the rat stomach. Both methods are widely accepted techniques for quantifying low levels of gene expression in small tissue samples, such as the gastric mucosa (Raeymaekers, 1995; Heid et al., 1996). Quantitative RT-PCR has been used to measure A i and A 2 A receptor gene expression in the rat brain (Rose'Meyer et al., 2003). The relative changes in A i receptor mRNA expression have been determined in the rat brain using competitive RT-PCR (Othman et al., 2002) and in the rat heart using Real-Time PCR (Ashton et al., 2003). The present study is the first to quantify the absolute level of A i and A 2 A receptor gene expression in the rat stomach, where the expression of these receptors are extremely low (Dixon et al., 1996). Both competitive RT-PCR and Real-Time RT-PCR assays were developed to measure this gene expression. Competitive RT-PCR is based on the competitive co-amplification of a fixed quantity of target template with known amounts of internal standard or R N A competitor. The A i and A 2 A receptor competitive RT-PCR assays developed in this study were able to measure gene expression ranging from 105 tolO 7 copies/pg total RNA. Each reaction tube contained a fixed amount of tissue RNA; 0.125 pg and 0.5 pg were used in the A , and A 2 A receptor assays, respectively. The amount of template used was optimized so that the tissue template and competitor band were both easily visualized, and so that both the tissue template and competitor cDNA amplified with equal efficiency during PCR. Using the competitive RT-PCR assay, striatal A i and A 2 A receptor mRNA levels were found to be at least 4.7 times and 116 times greater than in any region of the stomach, respectively. This is consistent with studies demonstrating moderate A i and abundant A 2 A receptor gene expression in the striatum and extremely low Aj and A 2 A receptor gene expression in the stomach (Dixon et al., 1996). A i receptor gene expression was shown to be significantly higher than A 2 A receptor gene expression in the antrum and mucosa, but not in the fundus and the corpus. These results do not agree well with results from the Real-Time RT-PCR experiments. Real-Time RT-PCR results demonstrate that A i receptor gene expression is significantly higher than A 2 A receptor gene expression in all gastric regions examined. The discrepancy between the competitive RT-PCR and Real-Time RT-PCR results is addressed later in this section. Real-Time RT-PCR is based on the cleavage of a fluorogenic probe that hybridizes to the template at a position located between the forward and reverse primers (See Fig. 9). The Real-Time RT-PCR assays used to measure A i and A 2 A receptor gene expression were shown to be considerably more sensitive than the corresponding competitive RT-PCR assay. The Real-Time RT-PCR assays were able to measure a range of concentrations exceeding a 7 log scale, and as few as 500 copies of A i or A 2 A receptor RNA/u.g total tissue RNA. The latter is equivalent to 25 copies of A i or A 2 A receptor transcript per reaction since each reaction mixture only contained 0.05 [ig of total RNA as the template. Using Real-Time RT-PCR, A i receptor gene expression was lowest in the corporeal mucosa, while A 2 A receptor gene expression was lowest in the whole stomach and corporeal mucosa. These results correspond well with the results of the immunohistochemistry studies. AiR-IR and A 2 A R - I R were found on some mucosal cells, but the majority of AiR-IR and A 2 A R - I R was localized on the nerve fibers and vasculature of the muscle and myenteric plexus of the corpus and antrum. In the antral mucosa, AiR-IR was localized on all G-cells and some D-cells, while A 2 A R - I R was only localized on some D-cells. Thus, the higher level of A i compared to A 2 A receptor mRNA in the antrum and whole stomach mucosa may reflect the higher number of G-cells compared to D-cells in these regions (Lehy et al., 1979). Striatal A i and A 2 A receptor mRNA levels measured by Real-Time RT-PCR were shown to be at least 2- and 70-fold higher, respectively, than levels measured in any region of the stomach. The mRNA levels of the A i and A 2 A receptor measured using this technique were consistently lower than those measured using competitive RT-PCR; A i and A 2 A receptor mRNA levels were found to be 2.4\u00C2\u00B10.2 and 21\u00C2\u00B13 times lower, respectively. The absolute level of A\ receptor gene expression has not previously been quantified in the rat stomach or striatum. Only the relative level of A i receptor gene expression in various tissues have been determined using RT-PCR (Dixon et al., 1996). The striatal A 2 A receptor mRNA levels measured by Real-Time RT-PCR (2.2 x 106 copies/pg total RNA), however, compared well with levels measured in a previous study (Lee et al., 1999). The striatal A 2 A receptor gene expression measured by Lee and co-workers (1999) was approximately 1.4 x 106 copies/pg total RNA, using a similar experimental protocol. The sequence of the primers and fluorogenic probe, and the total R N A extraction protocol used, however, were different, and may account for the small difference between their striatal A 2 A mRNA level and that found in the present study. The striatal A 2 A receptor level determined by competitive RT-PCR (6.6x 107 copies/pg total RNA) was not in the same range as that measured by Real-Time RT-PCR. Real-Time RT-PCR is likely the more accurate and reliable technique. The added component of the fluorogenic probe significantly increases the specificity of the Real-Time RT-PCR assay. This technique is more sensitive since the fluorogenic signal emitted by the reporter dye is detected by a highly sensitive CCD camera. Studies have shown that Real-Time RT-PCR can reliably detect a single specific D N A molecule in a high background of non-specific D N A (Lockey et al., 1998). Our assays were able to consistently measure as few as 25 copies of A i and A 2 A receptor RNA per reaction. Changes in reporter fluorescence are also monitored in a Real-Time manner. Thus, quantification is made at the start of the exponential phase and the errors that may be introduced by reaching the plateau phase are avoided (Raeymaekers, 1995; Schmittgen, 2001). The difference between the A i and A 2 A receptor mRNA levels measured by competitive RT-PCR and Real-Time RT-PCR is unclear. Several factors can introduce errors to competitive RT-PCR measurements, but in the present study, care was taken to avoid these. For example, during competitive RT-PCR, the number of cycles of amplification was limited to 30 to avoid reaching the plateau phase. As the reaction approaches the plateau phase, there is an increased likelihood of heteroduplex formation. These complexes are formed when the competitor and tissue template PCR products anneal. These formations can significantly decrease the sensitivity and accuracy of competitive RT-PCR quantification (Henley et al., 1996). These complexes would appear as an additional band above the competitor and tissue template bands. However, in the present study, heteroduplex formations were not observed for either the A i or A 2 A receptor competitive RT-PCR assays. Accurate quantification with competitive RT-PCR also requires that the template and the competitor behave in a similar manner during the reverse transcription and PCR step (Schmittgen, 2001). In this study, R N A competitors were used instead of D N A competitors to control for variations in the reverse transcription step (Orlando et al., 1998). In addition, homologous competitors, which have a sequence similar to that of the tissue template, were also used to minimize differences in the amplification efficiency during PCR. An equal amplification efficiency of the template and competitor is required for reproducible and accurate quantification (Connolly et al., 1995). The amplification efficiencies were determined in this study by plotting the log [density ratio (competitor/template) band] against the log (initial competitor concentration) for each assay. When the slope of the competitive RT-PCR relationship is equal to 1, both the template and competitor cDNA should amplify with equal efficiency (Connolly et a l , 1995). The slopes obtained for the A i and A 2 A receptor competitive RT-PCR assays were found to be 1.05\u00C2\u00B10.04 and 1.07\u00C2\u00B10.05, respectively. Thus, a significant difference in amplification efficiency between the tissue template and competitor cDNA was not apparent. Some studies have shown that, even when these slopes are ideal (= 1.0), competitive RT-PCR can yield results that differ significantly from actual levels. For example, Hayward et al. have shown that when a known quantity of Na +K +-ATPase mRNA was measured by competitive RT-PCR, the value reported was almost 4-fold higher although the slope of the competitive RT-PCR relationship remained at 1.01 (Hayward et al., 1998). These investigators further demonstrated, using mathematical modeling, that small differences in the amplification efficiency between the competitor and unknown template can drastically alter competitive RT-PCR measurements, while still producing a relationship with a slope of 1.0. A 20% difference in amplification efficiency between the competitor and unknown template was found to result in a 9 to 20-fold difference between the actual mRNA level and the value estimated by competitive RT-PCR (Hayward et al., 1998). In our studies, A i and A 2 A receptor mRNA levels measured by competitive RT-PCR were found to be 2.4 and 21-fold higher, respectively, than levels measured by Real-Time RT-PCR. Thus, a difference in the amplification efficiency between the competitor and unknown template may exist in our assays, despite the precautions taken. The A i and A 2 A competitors contain a 136 and 57 bp deletion. Decreasing the size of the deletion has been shown to increase the accuracy of competitive RT-PCR (Hayward et al., 1998). However, the resulting similarity between the size of the competitor and unknown template make it difficult to distinguish between the two bands during gel electrophoresis. In view of the limitations of competitive RT-PCR, Real-Time RT-PCR was used in our subsequent experiments to measure changes in A i and A 2 A receptor gene expression after various treatments, including food deprivation and omeprazole treatment. This technique is highly specific and is not subject to the limitations of competitive RT-PCR. In addition, this technique was chosen over competitive RT-PCR since striatal A 2 A receptor mRNA levels measured by Real-Time RT-PCR compared well with previously published results (Lee et a l , 1999). The extremely high sensitivity of this method also allows for quantification of A i and A 2 A receptor gene expression in small gastric tissue samples such as the mucosa, where the expression levels of these genes are extremely low. 2. Changes of gastric A i and A 2 A receptor gene expression and function by altered states of the stomach The present study shows that the A i and A 2 A receptors participate in the regulation of IRG and SLI release in the rat stomach. However, it is unclear i f the expression of these receptors is altered by different secretory states of the stomach. Changes to adenosine receptor gene expression may subsequently alter the modulatory effect of adenosine on IRG and SLI release. Previous studies have shown that oxidative stress (Nie et al., 1998; Kobayashi and Millhorn, 1999) or stress induced by acute renal failure (Gould et al., 1997) and sleep deprivation (Basheer et al., 2000) can alter adenosine receptor gene and/or protein expression. The present study demonstrates that stress induced by fasting and omeprazole treatment may also alter adenosine receptor gene expression. Real-Time RT-PCR was performed to measure changes in A] and A 2 A receptor, gastrin and somatostatin gene expression. A l l measurements were standardized with an endogenous control to account for differences resulting from variations in total R N A extraction efficiency, R N A integrity and R N A concentration. Two common house-keeping genes, G A P D H and 18S rRNA, were considered for use as the endogenous control. These genes can only be used as the endogenous control i f their expression is not altered by the experimental treatment. In the current study, G A P D H gene expression was significantly decreased in the corpus mucosa after 24 and 36 h of fasting, and in the antrum after 36 h of fasting. Similar results have been reported in animals which were subjected to 48 h of fasting (Yamada et a l , 1997a). G A P D H mRNA levels were also significantly altered in the corporeal muscle and mucosa after 1 and 3 days of omeprazole treatment, respectively. These findings are consistent with another study, which demonstrated that a single dose of omeprazole was capable of increasing G A P D H mRNA levels in the corpus (Sandvik et al., 1995). Furthermore, several studies have cautioned against the use of G A P D H mRNA as an endogenous control since its expression can be altered by a variety of conditions and agents such as hypoxia, oxidative stress, heat shock, glucose and insulin (Schmittgen and Zakrajsek, 2000; Suzuki et al., 2000). In the current study, 18S rRNA levels were not affected by fasting or omeprazole treatment. Thus, this gene was used as the endogenous control for the measurement of all gene expression. Previous experiments have also demonstrated that 18S rRNA expression was not altered by 48 h of fasting (Yamada et al., 1997a). Using 18S rRNA expression as the endogenous control, both fasting and omeprazole treatment were shown to alter adenosine A , and A 2 A receptor gene expression in the corporeal mucosa and antrum, but not in the corporeal muscle layer. In the fasting experiments, an increase in A i receptor gene expression was observed in both the corporeal mucosa and antrum after 24 h, but not after 36 h, of fasting. Similarly, fasting was shown to increase A 2 A receptor gene expression in the antrum after 24, but not after 36 h, of fasting. Although adenosine receptor protein expression may change in parallel with its mRNA level, the return of adenosine receptor gene expression to control levels after 36 h of fasting does not necessarily imply that adenosine receptor protein expression also returns to control levels. It is possible that the elevated adenosine receptor mRNA levels observed after 24 h of fasting resulted in increased adenosine receptor expression, which is maintained even after 36 h of fasting. The effect of fasting on A2A receptor gene expression differed in the antrum and corporeal mucosa. In the antrum, A2A receptor gene expression was shown to increase after 24 h of fasting. In the corporeal mucosa, its gene expression was shown to gradually decrease and become significant after 36 h of fasting. The different effect of fasting on A2A receptor gene expression in the antrum and corporeal mucosa suggests that the gene expression of this receptor may be differentially regulated in these two gastric regions. The lack of change in A i and A2A receptor gene expression in the corporeal muscle suggests that neither gene is altered by fasting in this tissue. It is currently unclear how adenosine receptor gene expression is altered by fasting. The absence of food in the stomach has been shown to increase gastric acidity (Matsumoto et al., 1989; Jiang et al., 2002). In the rat, Jiang and colleagues (2002) demonstrated that fasting for 24 h significantly decreased the gastric intraluminal pH to 2.0. Similarly, Matsumoto and co-workers (1989) demonstrated that fasting significantly decreased gastric intraluminal pH from a non-fasting level of 3.5 to 2.5, 2.0, and 1.6 after 1, 2, and 3 days of fasting, respectively, and that the intraluminal pH did not return to control levels until after 8 days of fasting. These studies suggest that changes in gastric acidity during fasting may alter gastric adenosine receptor gene expression. In agreement with this proposal, omeprazole-induced achlorhydria was also shown to alter A i and A2A receptor gene expression. Omeprazole (400 fj,mol/kg) was found to completely inhibit gastric acid secretion within 2 h of administration (Lee et al., 1992). Gastric acid secretion was decreased by up to 80% 24 h after the first treatment (Larsson et al., 1988; Lee et al., 1992), and continuous inhibition of acid secretion was achieved after 3 days of treatment (Carlsson et al., 1986). Results show that omeprazole treatment inhibited both A i and A2A receptor gene expression in the antrum, which is contrary to what occurs during fasting. Since fasting increases gastric acidity and omeprazole decreases gastric acidity, intraluminal acidity may play a role in altering adenosine receptor gene expression. In support of this proposal, the gene expression of gastric A i receptors was observed to be increased after fasting but decreased after omeprazole treatment. Results of our immunohistochemistry experiments demonstrate that A i receptors were expressed on D-cells and G-cells and A2A receptors were expressed on D-cells of the gastric mucosa. In the antrum, both of these cell types are open to the luminal environment and, thus, can respond to changes in gastric acidity, while the D-cells of the corpus are closed and do not readily respond to changes in gastric acidity (Sachs et al., 1997). Therefore, it is possible that gene expression of adenosine receptors in the antrum is readily altered by changes in gastric acidity, while adenosine receptor gene expression in D-cells of the corpus mucosa may not be directly affected by gastric acidity. This suggestion may explain why A2A receptor gene expression in the corporeal mucosa did not alter according to the changes in gastric acidity. In particular, A2A receptor gene expression was decreased by both fasting and omeprazole treatment. The possible mechanisms involved in the regulation of adenosine receptor gene expression by gastric acidity have not been examined in the present study. However, there is evidence to suggest that alterations in the activities of various mitogen-activated protein kinases (MAPK) may be involved. Malek and colleagues (1999) have shown that activation of ERK1, ERK2 and I N K can decrease adenosine receptor A2A mRNA levels in rat PC 12 cells. The activities of various M A P K respond to changes in both intracellular and extracellular pH (Feifel et al., 2002). Exposure of human epidermoid carcinoma cell line A431 to low extracellular pH was shown to increase the activity of M A P K (ERK2, JNK, and p38 M A P kinase), and exposure of mouse Swiss 3T3 cells to low extracellular pH was shown to stimulate the activity of M A P K (p42, p44 M A P kinase, ERKs, p38 M A P kinase and JNK) (Xue and Lucocq, 1997). In the Barrett's esophagus adenocarcinoma cell line (SEG-1), increased exposure to acidic media increased ERK, p38 M A P kinase, and JNK activities (Souza et al., 2002). The activity of p38 M A P kinase was also shown to be increased in esophageal mucosa cells of patients who suffer from Barrett's esophagus (Souza et al., 2002). Barrett's esophagus is associated with gastroesophageal reflux disease. Thus, the increased p38 M A P kinase activity in these cells is likely due to the increased exposure to acid. The studies mentioned above have demonstrated that changes in acidity may alter M A P K activity. Increases in M A P K activity may subsequently alter adenosine receptor gene expression. The effect of specific M A P K on A i receptor gene expression has not been determined. However, in the rat pituitary-derived cell line, A i receptor transcription is stimulated by epidermal growth factor (EGF) (Navarro et al., 1999). EGF has also been shown to activate M A P K pathways (Yamada et al., 1997b). Although M A P K activity may alter adenosine receptor gene expression in some cell lines (Malek et al., 1999), this has not been examined in the stomach. Adenosine receptor gene expression may also be regulated by changes in intracellular pH. During periods of oxidative stress, such as hypoxia and ischemia, intracellular pH is decreased (Kintner et al., 2000). Previous studies have shown that oxidative stress can induce A i receptor gene expression in DDT1-MF2 cell line (Nie et al., 1998), and that hypoxia can stimulate A 2 A receptor gene expression (Kobayashi and Millhorn, 1999). During fasting, it is possible that increased intraluminal acidity leads to subsequent increases in intracellular acidity in the gastric epithelium. In the guinea pig gastric gland, decreasing the luminal pH from 3.0 to 1.9 was shown to decrease intracellular pH in cells of the gastric gland (Schreiber et al., 2000). Similar results have also been found in the isolated Necturus antral mucosa, where acidification of the luminal pH to 2.0 was shown to significantly decrease intracellular pH of the antral mucosal cells (Kivilaakso and Kiviluoto, 1988; Kiviluoto et al., 1990). Since the intraluminal pH level has been shown to decrease from 3.5 to 2.0 after 24 h and 48 h of fasting (Matsumoto et a l , 1989; Jiang et al., 2002), it is possible that the increase in A i and A 2 A receptor mRNA levels observed in the antrum after 24 h of fasting result from intracellular acidification. Although fasting and omeprazole treatment may regulate adenosine A i and A 2 A receptor gene expression by altering the intraluminal acidity, it is unclear whether adenosine receptor gene expression may also respond to other factors. For example, during fasting, adenosine receptor gene expression may be altered by the absence of specific nutritional components of food in the stomach. The presence of peptones, phenylalanine and olive oil, in the gastric lumen has been shown to regulate the gene expression of gastrin and somatostatin (Wu et al., 1990b; Wu et al., 1991). Omeprazole treatment has also been shown to increase plasma levels of GRP (Takehara et al., 1996), which has been shown to regulate the activity of D-cells and G-cells. D-cells of the antrum and corpus can be regulated directly (Schaffer et al., 1997) and indirectly (Schubert et al., 1991) by GRP, respectively, while G-cells can be directly modulated by GRP (Campos et al., 1990). The G-cells and D-cells were shown to express adenosine receptors. Thus, fasting and omeprazole-treatment may also induce changes in adenosine receptor gene expression by preventing the exposure of the gastric lumen to nutritional components of food and by enhanced GRP release, respectively. Since activation of A i and A 2 A receptors inhibited gastrin and stimulated somatostatin release, respectively, changes in A\ and A 2 A receptor expression may also regulate gastrin and somatostatin synthesis and release. Therefore, the effect of fasting and omeprazole treatment on gastrin and somatostatin gene expression was also examined. Results show that gastrin gene expression was significantly inhibited by 24 and 36 h of fasting. These findings are consistent with previous studies demonstrating a significant reduction in gastrin gene expression after 12, 24, 48 and 72 h of fasting (Dimaline et al., 1991; Wuet al., 1991; Larsson and Hougaard, 1993; Sandvik et al., 1993; Yamada et al., 1997a). These results are also consistent with the decrease in antral gastrin concentration, G-cell number and serum gastrin content observed after fasting (Lichtenberger et al., 1975; Track et al., 1978; Shulkes et al., 1983; Dimaline et al., 1991; Larsson and Hougaard, 1993). Omeprazole treatment, on the other hand, was shown to increase gastrin mRNA levels after 1 and 3 days of treatment. Increased gastrin gene expression has also been demonstrated after 4 and 14 days of omeprazole treatment (Wu et al., 1990a; Dockray et al., 1991). The increased gastrin mRNA levels are most likely responsible for the enhanced levels of antral gastrin content and plasma gastrin concentrations (Brand and Stone, 1988; Inauen et al., 1988; Larsson et al., 1988; Dockray et al., 1991), which have been observed following short term (5 days or less) omeprazole treatment. Both fasting and omeprazole treatment were shown to induce reciprocal changes in A i receptor and gastrin gene expression. Our immunohistochemistry results demonstrate that A i receptors are expressed on G-cells, which suggests that increased expression and activation of A i receptors may directly affect gastrin gene expression. Adenosine has been shown to modulate the expression of various genes in the murine heart (Asakura et al., 2002), and may modulate gene expression by activating A i receptors to inhibit cAMP production. The transcription of certain genes is cAMP-dependent (Dragon et al., 2002). In particular, gastrin gene expression was regulated by cAMP levels (Shiotani and Merchant, 1995). Since activation of A i receptors inhibits adenylate cyclase activity and cAMP formation (Ralevic and Burnstock, 1998), it is possible that activation of A i receptor on G-cells may inhibit gastrin gene expression by inhibiting cAMP accumulation. In addition, somatostatin has been shown to inhibit gastrin synthesis by suppressing adenylate cyclase activity and subsequent cAMP production (DelValle and Yamada, 1990). Somatostatin gene expression was also altered by fasting and omeprazole treatment. The antral somatostatin mRNA level was shown to be increased following 24 h of fasting, but returned to control levels after 36 h of fasting. Previous experiments have demonstrated a significant increase in antral somatostatin gene expression following both 24 h (Wu et al., 1991) and 36 h of starvation (Sandvik et al., 1995). The cause of this discrepancy is unclear. However, it may be due to the use of a different housekeeping gene as the endogenous control. Sandvik and co-workers utilized G A P D H mRNA as the endogenous control. Sandvik reported no significant change in G A P D H mRNA expression after 36 h of fasting (Sandvik et al., 1995), contrary to this study and another report (Yamada et al., 1997a). In the corporeal mucosa, the somatostatin mRNA level was reduced following 36 h of fasting. This decrease is consistent with previous studies demonstrating a significant decrease in the somatostatin mRNA level in the oxyntic region following a 48 h fasting period (Sandvik et al., 1993). Results of the present experiments show that antral somatostatin gene expression was significantly decreased after 1 and 3 days of omeprazole treatment (400pmol/kg). These findings are similar to those reported in previous studies (Brand and Stone, 1988; Wu et al., 1990a; Wu et al., 1990b; Sandvik et al., 1995). The lowered antral somatostatin mRNA levels are consistent with the decreased plasma concentration of somatostatin, antral somatostatin concentration, and antral D-cell density that have been shown to occur in the rat stomach during achlorhydria (Allen et al., 1986; Lee et al., 1992; Pawlikowski et al., 1992). In the corpus, the present study shows that somatostatin mRNA levels were significantly decreased after omeprazole treatment. These observations are in agreement with several studies demonstrating reduced corporeal somatostatin mRNA expression during achlorhydria (Tari et al., 1991; Sandvik et al., 1995). Similar to our results, Tari and co-workers reported that the somatostatin mRNA level in the corpus gradually decreased over a period of 3 days during drug treatment (Tari et al., 1991). Other studies have shown that omeprazole can also cause an increase, rather than a decrease, in corporeal somatostatin gene expression (Kapuscinski and Shulkes, 1995; Bolkent et al., 2001). However, these observations were made 8 h after a single treatment of omeprazole (400 pmol/kg) (Kapuscinski and Shulkes, 1995) or after long-term (2 months) omeprazole treatment (Bolkent et al., 2001). The present study demonstrates that somatostatin gene expression was inhibited by achlorhydria in the corporeal muscle. Our immunohistochemistry experiments demonstrate that, in this region, somatostatin was expressed on very few nerve fibers of the circular muscle and myenteric plexus. Achlorhydria has previously been shown to alter neuropeptide mRNA expression in the enteric plexi (Dimaline et al., 1992). In particular, Dimaline and colleagues have shown that omeprazole treatment can increase the antral GRP mRNA level. The increase in somatostatin gene expression in this tissue may be secondary to changes in GRP levels, since GRP has been shown to stimulate somatostatin release (Martindale et al., 1982). Both fasting and omeprazole treatment induced similar changes in A2A receptor and somatostatin gene expression. Results of perfusion experiments performed in the present study demonstrate that activation of A2A receptors stimulated somatostatin release. Furthermore, results of the immunohistochemistry study demonstrate the expression of A 2 A receptors on D-cells. Thus, it is possible that changes in A2A receptor expression regulate somatostatin synthesis and release. Previous studies have shown that the rat somatostatin gene contains a cAMP response element (Montminy et al., 1986) and that its expression is regulated by cAMP levels (Montminy et al., 1996b). The A2A receptor couples to the G s protein and can stimulate adenylate cyclase activity to increase cAMP accumulation (Dalziel and Westfall, 1994; Fredholm et al., 2001). A number of studies have demonstrated that activation of A2A receptors induces the expression of genes regulated by the cAMP response element (CRE). Asher et al. have demonstrated that activation of A2 receptors can induce cAMP-dependent gene expression through the phosphorylation of CRE binding protein (CREB) (Asher et al., 2002). In vascular smooth muscle cells, activation of the A2 receptor was suggested to upregulate lysyl oxidase gene expression (Ravid et al., 1999). In addition, CGS 21680 (A2A-selective agonist) has been shown to induce tyrosine hydroxylase gene expression in rat PC12 cells (Chae and Kim, 1997). Like the somatostatin gene, both the lysyl oxidase and tyrosine hydroxylase genes also contain a CRE in the 5' upstream promoter region. Therefore, it is possible that activation of adenosine A2A receptors also increases somatostatin gene expression through activation of the CRE. Although the direct modulation of the gastrin and somatostatin gene expression by activation of the A i and A2A receptors remains a possibility, specific experiments have not been performed in the present study to confirm this proposal. The regulation of gastrin and somatostatin mRNA expression is complex and may also depend on a number of other factors. To examine whether changes in adenosine receptor mRNA levels may result in actual changes in adenosine receptor function, the effect of omeprazole treatment on adenosine agonist-induced changes in IRG and SLI release was also examined. The level of gene expression often changes in parallel with protein synthesis and receptor function. However, the results of the present study suggest that changes in gastric adenosine receptor gene expression do not always result in changes in receptor function. CPA, an A i receptor agonist, was demonstrated to be a potent inhibitor of IRG release in the isolated vascularly perfused rat stomach. When administered at low (<0.1 uM) and high (>1 uM) concentrations, CPA suppressed and stimulated SLI release, respectively. A similar concentration-dependent effect has been demonstrated using adenosine (Kwok et al., 1990). After treatment with omeprazole, A i receptor gene expression was reduced in the corporeal mucosa and antrum. Following 1 or 3 days of omeprazole treatment, CPA-induced inhibition of IRG and SLI release was not altered. However, the stimulatory effect on SLI release induced by the higher concentration (1 uM) of CPA was significantly decreased following 3 day treatment with omeprazole. As discussed earlier, the inhibition of IRG and SLI release induced by CPA is likely mediated by activation of A i receptors, while the augmentation of SLI release induced by high concentrations of CPA is likely mediated by the non-selective activation of A 2 A receptors. The attenuation of CPA-stimulated SLI release after omeprazole treatment, therefore suggests that A 2 A receptor expression is reduced after 3 days of omeprazole treatment. In agreement with this proposal, the stimulatory action of the A 2 A receptor agonist CGS 21680 on SLI release was also reduced after 3 days of omeprazole treatment. Both 0.1 and 1 p M CGS 21680 potently stimulated the release of SLI in earlier experiments. Although the stimulatory effect of 0.1 p M CGS 21680 on SLI release was suppressed after 3 days of treatment, this effect was not statistically significant. The release of SLI induced by 1 p M CGS 21680, however, was significantly attenuated after 3 day omeprazole treatment. This attenuation of A 2 A receptor function is consistent with the reduced level of antral A 2 A receptor gene expression observed after 3 days of omeprazole treatment. Taken together, results from the gene expression and perfusion experiments suggest that omeprazole treatment decreases the number of functional A 2 A receptors, but does not always produce parallel changes in adenosine receptor expression. In the present study, A 2 A receptor gene expression was also reduced in the corporeal mucosa after 1 day of omeprazole treatment, but significant changes in CGS 21680-induced SLI release were not observed at this time. Thus, alterations in A 2 A receptor mRNA levels may not necessarily produce changes in A 2 A receptor function. It is possible that, although A 2 A receptor mRNA levels may be altered after 24 h of achlorhydria, actual changes in A 2 A receptor expression may require more than 24 h to occur. The protein synthesis and expression of the adenosine receptors may also be controlled by other factors. The synthesis of both the A\ and A 2 A receptors can be regulated at the translational level (Ren and Stiles, 1994b; Chu et al., 1996; Lee et al., 1999). The translation of the human A i receptor was shown to be inhibited by two upstream A U G codons in exon 4 (Ren and Stiles, 1994b), while translation of the human A 2 A receptor is inhibited by the presence of several upstream open-reading frames and regions of high GC content (Chu et al., 1996). The expression of the rat A2A receptor is suppressed by the presence of an upstream out-of-frame A U G codon in the 5' untranslated region (Lee et al., 1999). Thus, in addition to the regulation of A\ and A 2 A receptor expression by its mRNA levels, receptor expression can also be controlled by translational mechanisms. It is uncertain whether fasting and omeprazole treatment also alter the translation of the A i and A2A receptor genes. Since A i and A 2 A receptor gene expression was examined in whole tissue extracts, it is difficult to assess whether changes in A ( and A2A receptor mRNA expression only occur in anatomical structures that are involved in the direct control of IRG and SLI release, such as the G-cells and D-cells. Our immunohistochemistry experiments have demonstrated that, in the stomach, the adenosine A i and A2A receptors are not expressed solely on these cells. The significant functional implications of the widespread localization of A i and A2A receptors are discussed in the following section. IV. Cellular localization of the adenosine Ai and A 2 A receptor From the discussion above, it is clear that both adenosine A i and A2A receptors play a role in regulating gastric secretions. However, studies examining the cellular localization of gastric adenosine receptors are lacking. Autoradiography studies have not been performed in this tissue and in situ hybridization experiments are not sensitive enough to detect the low levels of adenosine receptor mRNA in the stomach (Dixon et al., 1996). The present study uses the technique of immunohistochemistry to determine the cellular localization of adenosine A i and A 2 A receptors in the rat stomach. This study is the first to provide the anatomical basis for various gastric actions of adenosine. In the present study, AiR-IR and A2AR-ER- were found to be widely distributed throughout the stomach. Both were present on cells of the mucosa, cell bodies of the myenteric plexus, nerve fibers of the muscle layers and enteric plexi, and the gastric vasculature. The regional localization and relative density of AjR-IR and A2AR-IR in the gastric tissues agree well with results of the Real-Time RT-PCR experiments. Overall, A i R -IR appeared to be more abundant than A 2 AR-IR . Although some mucosal cells were shown to contain intense adenosine receptor staining, the majority of AiR-IR and A 2 A R - I R was localized in the enteric plexi. The localization of the gastric A i and A 2 A receptors in various regions of the stomach also agrees well with their distribution in other tissues of the gastrointestinal tract. Although the anatomical distribution of adenosine receptors in the rat intestines has not been determined, immunohistochemical localization of the A i and A 2 A receptors has been demonstrated in the human intestines (Christofi et al., 2001). Some anatomical structures, which were labeled in the present study, were also labeled in the human intestines. AiR-IR was demonstrated in both the myenteric and submucosal plexi of the human jejunum and colon, respectively. Neurons of the circular muscle of these tissues contained AiR-IR. Using the same A 2 A receptor antibody as the present study, Christofi and co-workers (2001) also demonstrated the expression of A 2 A R - I R in myenteric and submucosal neurons of the human jejunum and colon. In the jejunum, A 2 A R - I R was found to be localized in epithelial cells, while, in the colon, staining of this receptor was observed in myenteric neurons. A 2 A R - I R was also detected on some nerve fibers in the intestines, in particular, in the longitudinal muscle of the jejunum and circular muscle of the colon. The neuronal localization of these receptors in the stomach and intestine agrees with the role of adenosine in the regulation of motility. The cellular localization of these receptors in the mucosa and vasculature provide the anatomical basis for the known effect of adenosine on somatostatin and gastrin release and vascular actions in the stomach, respectively. 1. Adenosine Ai and A 2 A receptors of the gastric mucosa Results show that cells containing distinct AiR-IR and A 2 A R - I R were distributed throughout the corporeal and antral mucosa. These findings support the role of adenosine in regulating various gastric secretions. Tissue sections were double stained to determine whether these receptors are localized on specific cells of the mucosa, in particular, the acid-secreting parietal cells, gastrin-secreting G-cells, and somatostatin-secreting D-cells. Although E C L cells are known to be present in the mucosa, the localization of adenosine receptors on these cells was not examined. E C L cells are difficult to identify by immunohistochemistry since the preservation of histamine and histidine decarboxylase, E C L cell markers, require special fixation techniques (Bordi et al., 2000). E C L cells are commonly identified by chromogranin A immunostaining. However, this product is not exclusive to the E C L cells, and is also found in other endocrine cells such as G-cells. Results of our studies suggest that the A j R and A 2 A R are unlikely to be expressed on E C L cells of the rat. E C L cells make up a large proportion of the endocrine cell population, and reside at the base of the oxyntic mucosa (Dockray et al., 1996; Bordi et al., 2000). In the corporeal mucosa, the majority of A i R - I R was localized on D-cells. The A i receptors that were not on D-cells were scattered throughout the mucosa and were not confined to the basal region. A 2 A R - I R was not abundant in the corporeal mucosa, and was also scattered throughout the oxyntic mucosa. Since the E C L cells make up a large proportion of the endocrine cell population and are localized only at the base of the oxyntic mucosa, any appreciable A i or A 2 A receptor expression on these cells should be apparent. The effect of adenosine on histamine release has not been studied in rats. However, previous studies have shown that adenosine does not alter histamine release in either dog or rabbit (Ainz et al., 1993; Payne and Gerber, 1997). These studies, in combination with our immunohistochemistry results, suggest that adenosine is unlikely to alter histamine release in the rat. Adenosine has been shown to stimulate or inhibit gastric acid secretion in the rat stomach, depending on the route of drug administration. When administered orally or intragastrically, adenosine and its analogs have been shown to decrease basal gastric acid secretion (see Table 2). These routes of administration expose adenosine to the gastric mucosa. Thus, adenosine may mediate its gastric acid inhibitory effect by acting on adenosine receptors in mucosal cells. Although radioligand binding studies have shown extremely low specific binding of H-PIA, a selective A i receptor agonist, on isolated rat parietal cell preparations (Puurunen et al., 1987), results of the present experiments rule out the possible existence of A i receptors on parietal cells. Results suggest that adenosine does not act directly on the parietal cells since neither A i nor A 2 A receptors were found on these cells. In the present study, parietal cells were identified by the staining of the H + K + -ATPase p subunit of the proton pump. Abundant H + K + -ATPase (3-IR was localized throughout the isthmus, neck, and base of the oxyntic glands, as previously shown (Jiang et al., 2002). Double-staining experiments demonstrate that H + K + -ATPase P-IR was not co-localized with either A i R - I R or A 2 AR-IR - The absence of adenosine receptors on parietal cells also agrees with previous studies showing that R-PIA (Ai agonist) and N E C A (A 2 agonist) do not alter carbachol- and histamine-stimulated 14C-aminopyrine accumulation in isolated rat parietal cells (Puurunen et al., 1987). Adenosine analogs also did not alter basal or histamine-stimulated adenylate cyclase activity or carbachol-stimulated inositol phosphate formation in these cells (Puurunen et al., 1987). 1.1 Adenosine Ai receptors on gastrin-secreting G-cells Adenosine has been shown to inhibit gastrin release from the rat stomach (Kwok et al., 1990). The present experiments further demonstrate that this action involves A i receptors, but not A 2 or A3 receptors. In agreement with these observations, our immunohistochemistry experiments demonstrate that A i R - I R was present on all mucosal cells expressing gastrin-IR, while A 2 A R - I R was not observed on any of these cells. The presence of A i receptors on G-cells suggests that adenosine may act directly on the G-cells to regulate gastrin release. This direct action may also occur in dogs since adenosine has been shown to inhibit forskolin-stimulated gastrin release through the activation of A i receptors in primary cultures of canine G-cells (Schepp et al., 1990). Several possible mechanisms may be involved in the A i receptor-mediated inhibition of gastrin release. The release of gastrin is stimulated by the activation of multiple second messenger pathways. In isolated G-cells, activation of adenylyl cyclase, PLC and PKC are involved in stimulating gastrin release (Campos et al., 1990; Buchan and Meloche, 1994; Seensalu et al., 1997). Activation of A i receptors has been shown to inhibit adenylate cyclase activity, the most common signaling pathway recognized for the A i receptor (Fredholm et al., 2001). In G-cells, this second messenger pathway may be activated to inhibit basal gastrin release. Activation of the A i receptor may also oppose the actions of agents, such as forskolin, that act by stimulating adenylate cyclase. Activation of A i receptors has also been shown to alter PLC and P K C activity in some tissues (see Table 1). In the stomach, these pathways may also be involved in the inhibitory action of adenosine on gastrin release. The release of gastrin is induced by the influx of extracellular calcium (Buchan and Meloche, 1994). High local concentrations of calcium are required for the exocytosis of gastrin (Oomori et al., 1997), and previous studies showed that gastrin secretion from G-cells requires the activation of L-type C a 2 + channels (Ray et al., 1997). Activation of A\ receptors inhibits L-type channel activity in atrial myocytes (Belardinelli et al., 1995). Thus, these receptors may also inhibit gastrin secretion through the inactivation of these channels. 1.2 Adenosine A] and A 2 A receptors on somatostatin secreting D-cells The present study demonstrates that both A i and A 2 A receptors were involved in modulating somatostatin release. Results of double staining immunohistochemistry experiments further demonstrates that both receptor subtypes were expressed on somatostatin-secreting D-cells of the corpus and antrum, suggesting that adenosine may act directly on D-cells to regulate somatostatin release. In isolated rat oxyntic D-cells, somatostatin release has been shown to involve both phospholipid-dependent and cAMP-dependent signaling pathways (Schaffer et al., 1997). In human oxyntic D-cells, somatostatin release was also stimulated by the activation of adenylate cyclase and PKC pathways (Buchan et al., 1990, 1993). The adenosine A 2 A receptor is coupled to the G s protein and has been shown to stimulate adenylate cyclase activity (see Table 1). In striatal neurons, stimulation of this receptor has also been shown to activate the P K C pathway. Thus, A 2 A receptors may stimulate somatostatin release by activating adenylate cyclase and P K C pathways. Conversely, A i receptors are coupled to the pertussis toxin-sensitive G protein, G;, and activation of this receptor may inhibit somatostatin release through the inhibition of adenylate cyclase activity. The activation of a pertussis-toxin sensitive G protein has previously been implicated in mediating the inhibitory effect of carbachol on pentagastrin-stimulated somatostatin release in canine oxyntic D-cells (Chiba et al., 1989). Somatostatin is released from D-cell processes by an exocytotic process requiring high local concentrations of Ca 2 + . In isolated oxyntic D-cells, CCK-stimulated somatostatin release was shown to involve the influx of extracellular Ca through the L-type Ca channels (DelValle et al., 1996). The release of somatostatin from D-cells is also stimulated by membrane depolarization (Patel, 1999). In the present study, confocal microscopy performed at high magnification reveals that most AiR-IR appeared to be concentrated at the end of the D-cell processes. These receptors have previously been localized on presynaptic nerve terminals, and have been shown to inhibit the exocytosis of synaptic vesicles through the inactivation of N-type and L-type C a 2 + channels (Ralevic and Burnstock, 1998). Activation of A i receptors has also been shown to hyperpolarize neurons by increasing K + conductance (Belardinelli et al., 1995). Thus, in D-cells, A i receptors may inhibit somatostatin release by regulating exocytosis or by modulating the membrane potential. 2. Adenosine A i and A 2 A receptors of the enteric plexi The localization of gastric A i and A 2 A receptors in nerve fibers of the myenteric and submucosal plexi is not surprising since studies have demonstrated adenosine-mediated actions in the intestinal enteric plexi (Burnstock, 2001; Christofi, 2001). Adenosine receptors in this region are likely involved in regulating gastric motility by modulating neurotransmitter release. Numerous studies examining the purinergic control of gastrointestinal motility have been performed in guinea pigs (Christofi, 2001). In general, activation of A i and A 2 A receptors has been shown to inhibit and stimulate gastrointestinal motility, respectively. In the rat jejunum, activation of the A i receptors has been shown to inhibit peristalsis (Hancock and Coupar, 1995), and in the ileum, these receptor have been shown to enhance descending relaxation (Storr et al., 2002). In the rat stomach, activation of A i receptors in the enteric plexi has been shown to mediate gastric relaxation (Matharu and Hollingsworth, 1992). Conversely, activation of A 2 receptor has been suggested to potentiate vagally-stimulated gastric contractions (Cho et al., 1993). Thus, adenosine A i and A 2 A receptors are involved in the inhibitory (relaxation) and stimulatory (contraction) control of gastrointestinal motility, respectively. The effect of adenosine on gastrointestinal motility is likely mediated by its action on presynaptic and postsynaptic neurotransmission in the enteric plexi. Most studies that examine the neuromodulatory role of adenosine in the intestines have been performed in the guinea pig ileum, in particular, the guinea pig myenteric plexus-longitudinal muscle preparations. In the submucosal plexus of the guinea pig ileum, electrophysiological studies have shown that adenosine inhibits enteric neurotransmission by suppressing the excitability of myenteric neurons (Zafirov et al., 1985; Palmer et al., 1987). This inhibitory effect was mediated by presynaptic A i receptors on myenteric neurons (Christofi and Wood, 1993). Similarly, activation of presynaptic A i receptors were shown to inhibit neurotransmitter release from submucosal neurons (Barajas-Lopez et al., 1991). In the guinea pig ileum, adenosine inhibits acetylcholine release from enteric nerves (Sawynok and Jhamandas, 1976; Hayashi et al., 1978; Gustafsson et al., 1985). This inhibition was presynaptic in nature (Gustafsson et al., 1985; Christofi and Cook, 1986). A i receptor-mediated presynaptic inhibition of noradrenaline (Barajas-Lopez et al., 1991), neurokinin A (Broad et al., 1992) and substance P (Broad et al., 1993) release from enteric nerves endings has also been demonstrated. In addition to its inhibitory effect on neurotransmitter release, adenosine may stimulate neurotransmitter release by acting directly on enteric neurons. This postsynaptic excitatory effect was shown to be mediated by A 2 A receptors (Barajas-Lopez et a l , 1991). Although the effect of adenosine on the intestines has been studied extensively, the effect of this nucleoside is not well-studied in the stomach. In the gastric antrum of the guinea pig, adenosine and its analogs have been shown to inhibit nicotinic transmission in the myenteric plexus by acting on presynaptic adenosine receptors (Christofi et al., 1992). Furthermore, adenosine and its analogs have been shown to inhibit fast EPSPs in approximately 60% of gastric neurons (Christofi et al., 1992). Thus, the relatively high level of AiR-IR and A2AR-IR staining observed in the myenteric plexus of the rat stomach in the present study fits well with the known modulatory effects of adenosine on neurotransmission. In the rat stomach, adenosine has been demonstrated to protect against ethanol-induced gastric lesion formation (see Chapter 1, Section 3), but the mechanism involved has not been determined. Recent studies have shown that inhibition of gastric motility by anti-muscarinic drugs can protect the stomach against lesion formation induced by ethanol (Schicho et al., 2003). Thus, the A i receptor-mediated inhibition of acetylcholine release may be involved in protecting the stomach against ethanol injury. Adenosine has also been shown to regulate gastric IRG and SLI release. Similar to its action on motility, the effect of adenosine on the release of these gastric peptides may be secondary to its effect on adrenergic and cholinergic neurotransmission. Adrenergic and cholinergic agents have been shown to alter the release of IRG and SLI (Saffouri et a l , 1980; Koop et al., 1982, 1983; Schubert and Makhlouf, 1992). Previous experiments performed in our laboratory have shown that the adenosine-induced release of SLI was not altered in the presence of the cholinergic blockers, atropine and hexamethonium, or the P-adrenergic blocker, propranolol (Kwok et al., 1990). Thus, adenosine is unlikely to modulate SLI release by acting on adrenergic or cholinergic nerves. The present study shows that adenosine receptors were expressed in the enteric plexi. Since various non-adrenergic, non-cholinergic nerve fibers are present in the plexi, it is possible that adenosine stimulates SLI release indirectly by modulating non-adrenergic, non-cholinergic neurotransmission. CGRP and VIP have been demonstrated to stimulate gastric somatostatin secretion (Chiba et al., 1980; Manela et a l , 1995; Rasmussen et al., 2001). While the effect of adenosine on CGRP and VIP release has not been examined in the stomach, endogenous adenosine has been shown to tonically control CGRP and VIP release in the CNS (Sebastiao and Ribeiro, 2000). The co-localization of A 2 A R - I R with somatostatin-IR in some nerves of the myenteric plexus and circular muscle observed in the present study suggests that activation of A 2 A receptors may elicit somatostatin release from nerve fibers. However, most of the somatostatin-IR was shown to reside in the D-cells, as previously reported (Vinik et al., 1981; Ekblad et al., 1985; Keast et al., 1985). Only sparse somatostatin staining was observed in the myenteric plexus and on nerve fibers. These somatostatin-IR fibers do not appear to project to the mucosa, and most of these fibers do not express A 2 A R - I R . Thus, adenosine is more likely to act directly on mucosal D-cells to elicit SLI release. The possibility that adenosine mediates its inhibitory effect on IRG release through adrenergic or cholinergic mechanisms has not been examined. It is also possible that adenosine mediates its inhibitory actions on IRG release by modulating the release of CGRP and VIP. These neurotransmitters have been shown to inhibit IRG release (Chiba et al., 1980; Manela et al., 1995). However, the localization of A i R on all gastrin-secreting G-cells of the mucosa demonstrated by the present study suggests that adenosine may inhibit gastrin secretion, at least in part, by acting on A i receptors on the G-cells. This proposal is supported further by the ability of adenosine to inhibit gastrin release from canine G-cells (Schepp et al., 1990) and rat antral mucosal cells in culture (DeSchryver-Kecskemeti et al., 1981). 3. Adenosine A i and A 2 A receptors of the gastric vasculature The vascular localization of the gastric adenosine receptors has been suggested by a number of studies demonstrating the action of adenosine on gastric mucosal blood flow (Cho and Ogle, 1990; Cho et al., 1991; Cho et al., 1995; Nagata et al., 1996). In the pulmonary artery (Biaggioni et al., 1989) and the renal vasculature (Holz and Steinhausen, 1987), activation of A i receptors has been shown to mediate vasoconstriction. Conversely, activation of A 2 A receptors has been shown to mediate relaxation in coronary and pulmonary arteries, renal vasculature and the aorta (Tabrizchi and Bedi, 2001). In the rat stomach, subcutaneous administration of adenosine has been shown to increase gastric mucosal blood flow (Cho et al., 1991; Cho et al., 1995). Adenosine has been shown to induce vasodilation in submucosal microvessels of the stomach by acting on A 2 receptors (Gerber and Guth, 1989; Nagata et al., 1996). This vasodilatory action may be involved in protecting the stomach against ethanol-induced gastric injury since the administration of ethanol has previously been shown to vasoconstrict submucosal blood vessels (Yonei and Guth, 1991). The perfusion of CGS 21680 into the isolated vascularly perfused rat stomach has also been demonstrated to decrease the gastric perfusion pressure (Yip and Kwok, 2004), suggesting that the A 2 A receptors are involved in mediating gastric vasodilation. Adenosine has been proposed to act on A 2 A receptors localized on vascular smooth muscle cells and endothelial cells (Tabrizchi and Bedi, 2001). In the rat mesenteric arterial bed, adenosine-induced vasodilation was shown to involve A 2 A receptors localized on both of these cell types (Hiley et al., 1995). The immunohistochemical localization of the A i and A 2 A receptors has previously been demonstrated in the vascular smooth muscle cells and endothelial cells of the human skeletal muscle (Lynge and Hellsten, 2000). Results of the current study clearly demonstrate that, in the stomach, both A i and A 2 A receptors are localized on vascular endothelial cells and are also likely expressed on vascular smooth muscle cells. v The vasodilatory actions of adenosine in the stomach are mainly attributed to activation of A 2 A receptors. An A i receptor-mediated action in the gastric vasculature has not yet been demonstrated. In the renal vasculature, the effect of adenosine was concentration-dependent. At low micromolar concentrations, adenosine elicited vasoconstriction via activation of A i receptors, while at higher concentrations (>10 | J M ) , adenosine elicited vasodilation via activation of A 2 receptors (Tabrizchi and Bedi, 2001). In the rat gastric submucosal vessels, adenosine-induced vasodilation was demonstrated using >10 uM adenosine (Nagata et al., 1996). The effect of lower concentrations of adenosine has not been examined. The regulation of vascular tone by the A i receptor remains to be confirmed in the stomach. In summary, the results of the immunohistochemistry study provide several new and significant insights into the distribution and localization of adenosine receptors. They confirm the widespread distribution of A i and A 2 A receptor expression in the enteric plexi and vasculature, and further support the suggestion that adenosine acts on these anatomical structures. The localization of A i and A 2 A receptors on the G-cells and D-cells support the proposal that adenosine may act directly on these cells to modulate gastrin and somatostatin release, respectively. The lack of adenosine receptors on the parietal cells provides further evidence that adenosine does not regulate gastric acid secretion by acting directly on the parietal cells. V. Conclusion This study provides fundamental information on the functional role of A i and A 2 A receptors in the rat stomach. Results suggest that adenosine may be an important regulator of gastric acid secretion. Unlike in other species, adenosine does not alter gastric acid secretion by acting directly on parietal cells in the rat. This lack of effect is confirmed in the present study by demonstrating the absence of AiR-IR and A 2 A R - I R on rat parietal cells. Instead, this study suggests that adenosine likely regulates gastric acid secretion by acting on A i receptors to inhibit gastrin and A 2 A receptors to stimulate somatostatin release. The localization of A\ and A 2 A receptors on the G-cells and D-cells suggests that adenosine may act directly on these cells to modulate gastrin and somatostatin release. Since these receptors were also present on nerve fibers throughout the enteric plexi, the possibility that adenosine may modulate gastrin and somatostatin indirectly by altering neurotransmission cannot be ruled out. The localization of A] and A 2 A receptors in the vasculature and enteric plexi also agrees with the known actions of adenosine in regulating gastric mucosal blood flow and motility. The mucosal adenosine A i and A 2 A receptors were shown to be structurally identical to those in the brain, and expressed in extremely low levels, using Real-Time RT-PCR and competitive RT-PCR. Real-Time RT-PCR was also used to examine the relationship between altered physiological states of the stomach and gastric A i and A 2 A receptor gene expression. Like gastrin and somatostatin, the gene expression of A i and A 2 A receptors is regulated by the gastric luminal environment. Fasting and omeprazole treatment were shown to alter adenosine receptor gene expression, most likely by changing the intraluminal acidity. Since these receptors are localized on G-cells and D-cells, changes in their expression may alter gastrin and somatostatin synthesis and secretion. Changes in adenosine receptor gene expression can be accompanied by corresponding changes in receptor function, suggesting that adenosine receptor expression may be determined, in part, by its gene expression. The present experiments provide fundamental information regarding the function, distribution, and cellular localization of the gastric A i and A 2 A receptors. Furthermore, this study suggests that under different physiological states of the stomach adenosine receptor expression may be altered to modulate gastrin and somatostatin synthesis and release, and, therefore, gastric acid secretion. Understanding the processes involved in the purinergic control of gastric functions is critical in understanding the role adenosine receptors may play in protecting the stomach in pathophysiological states. This understanding is vital to the future development of adenosine-based therapeutic agents. References Abbracchio MP, Brambilla R, Ceruti S, Kim HO, von Lubitz DK, Jacobson K A and Cattabeni F (1995) G protein-dependent activation of phospholipase C by adenosine A3 receptors in rat brain. Mol Pharmacol 48:1038-1045. Ainz LF, Salgado C, Gandarias JM, Gomez R, Vallejo A and Gil-Rodrigo CE (1993) Pl(A2/Ra)-purinoceptors may mediate the stimulatory effect of adenosine and adenosine analogs on acid formation in isolated rabbit parietal cells. Pharmacol Res 27:319-334. Akatsuka Y , Egashira K, Katsuda Y , Narishige T, Ueno H, Shimokawa H and Takeshita A (1994) ATP sensitive potassium channels are involved in adenosine A2 receptor mediated coronary vasodilatation in the dog. Cardiovasc Res 28:906-911. Allen JM, Bishop A E , Daly MJ , Larsson H, Carlsson E, Polak J M and Bloom SR (1986) Effect of inhibition of acid secretion on the regulatory peptides in the rat stomach. Gastroenterology. 90:970-977'. Anderson SM, Leu JR and Kant GJ (1988) Chronic stress increases the binding of the A l adenosine receptor agonist, [3H]cyclohexyladenosine, to rat hypothalamus. Pharmacol Biochem Behav 30:169-175. Andres C M and Fox IH (1979) Purification and properties of human placental adenosine kinase. J Biol Chem 254:11388-11393. Asakura M , Kitakaze M , Sakata Y , Asanuma H, Sanada S, Kim J, Ogida H , Liao Y , Node K, Takashima S, Tada M and Hori M (2002) Adenosine-induced cardiac gene expression of ischemic murine hearts revealed by cDNA array hybridization. Ore J 66:93-96. Asher O, Cunningham TD, Yao L, Gordon AS and Diamond I (2002) Ethanol stimulates cAMP-responsive element (CRE)-mediated transcription via CRE-binding protein and cAMP-dependent protein kinase. J Pharmacol Exp Ther 301:66-70. Ashton K J , Holmgren K, Peart J, Lankford AR, Paul Matherne G, Grimmond S and Headrick JP (2003) Effects of A l adenosine receptor overexpression on normoxic and post-ischemic gene expression. Cardiovasc Res 57:715-726. Atkinson MR, Townsend-Nicholson A, Nicholl JK, Sutherland GR and Schofield PR (1997) Cloning, characterisation and chromosomal assignment of the human adenosine A3 receptor (ADORA3) gene. Neurosci Res 29:73-79. Auboeuf D and Vidal H (1997) The use of the reverse transcription-competitive polymerase chain reaction to investigate the in vivo regulation of gene expression in small tissue samples. Anal Biochem 245:141-148. Bailey SJ, Hickman D and Hourani S M (1992) Characterization of the Pl-purinoceptors mediating contraction of the rat colon muscularis mucosae. Br J Pharmacol 105:400-404. Ballarin M , Fredholm B B , Ambrosio S and Mahy N (1991) Extracellular levels of adenosine and its metabolites in the striatum of awake rats: inhibition of uptake and metabolism. Acta Physiol Scand 142:97-103. Barajas-Lopez C, Surprenant A and North R A (1991) Adenosine A l and A2 receptors mediate presynaptic inhibition and postsynaptic excitation in guinea pig submucosal neurons. J Pharmacol Exp Ther 258:490-495. Baraldi PG and Borea PA (2000) New potent and selective human adenosine A(3) receptor antagonists. Trends Pharmacol Sci 21:456-459. Baraldi PG, Cacciari B, Romagnoli R, Merighi S, Varani K , Borea PA and Spalluto G (2000) A(3) adenosine receptor ligands: history and perspectives. Med Res Rev 20:103-128. Barnette MS, Grous M , Cieslinski L B , Burman M , Christensen SB and Torphy TJ (1995) Inhibitors of phosphodiesterase IV (PDE IV) increase acid secretion in rabbit isolated gastric glands: correlation between function and interaction with a high-affinity rolipram binding site. J Pharmacol Exp Ther 273:1396-1402. Basheer R, Porkka-Heiskanen T, Strecker RE, Thakkar M M and McCarley RW (2000) Adenosine as a biological signal mediating sleepiness following prolonged wakefulness. Biol Signal Receptors 9:319-327. Basheer R, Rainnie DG, Porkka-Heiskanen T, Ramesh V and McCarley RW (2001) Adenosine, prolonged wakefulness, and A1-activated NF-kappaB D N A binding in the basal forebrain of the rat. Neuroscience 104:731-739. Belardinelli L, Shryock JC, Song Y , Wang D and Srinivas M (1995) Ionic basis of the electrophysiological actions of adenosine on cardiomyocytes. FASEB J 9:359-365. Berglindh T, Helander HF and Obrink K J (1976) Effects of secretagogues on oxygen consumption, aminopyrine accumulation and morphology in isolated gastric glands. Acta Physiol Scand 97:401-414. Biaggioni I, King LS, Enayat N , Robertson D and Newman JH (1989) Adenosine produces pulmonary vasoconstriction in sheep. Evidence for thromboxane A2/prostaglandin endoperoxide-receptor activation. Circ Res 65:1516-1525. Blackburn M R (2003) Too much of a good thing: Adenosine overload in adenosine-deaminase-deficient mice. Trends Pharmacol Sci 24:66-70. Bolkent S, Yilmazer S, Kaya F and Ozturk M (2001) Effects of acid inhibition on somatostatin-producing cells in the rat gastric fundus. Acta Histochem 103:413-422. Bordi C, D'Adda T, Azzoni C and Ferraro G (2000) Classification of gastric endocrine cells at the light and electron microscopical levels. Microsc Res Tech 48:258-271. Borkje B, Odegaard S, Skagen DW, Andersen K J and Schrumpf E (1986) Enzyme activities in biopsy specimens from human gastric mucosa. Scand J Gastroenterol 21:1051-1057. Bozkurt A , Yuksel M , Haklar G, Kurtel H, Yegen BC and Alican I (1998) Adenosine protects against indomethacin-induced gastric damage in rats. DigDis Sci 43:1258-1263. Brackett L E and Daly JW (1994) Functional characterization of the A2b adenosine receptor in NIH 3T3 fibroblasts. Biochem Pharmacol 47:801-814. Brand SJ and Stone D (1988) Reciprocal regulation of antral gastrin and somatostatin gene expression by omeprazole-induced achlorhydria. J Clin Invest 82:1059-1066. Broad R M , McDonald TJ, Brodin E and Cook M A (1992) Adenosine A l receptors mediate inhibition of tachykinin release from perifused enteric nerve endings. Am J Physiol 262:G525-531. Broad R M , McDonald TJ and Cook M A (1993) Adenosine and 5-HT inhibit substance P release from nerve endings in myenteric ganglia by distinct mechanisms. Am J Physiol 264:G454-461. Brownhill VR, Hourani S M and Kitchen I (1996) Differential ontogeny of adenosine receptors in the longitudinal muscle and muscularis mucosae of the rat isolated duodenum. Eur J Pharmacol 317:321-328. Brundege JM, Diao L, Proctor WR and Dunwiddie T V (1997) The role of cyclic A M P as a precursor of extracellular adenosine in the rat hippocampus. Neuropharmacology 36:1201-1210. Bruns RF, Fergus JH, Badger EW, Bristol JA, Santay L A , Hartman JD, Hays SJ and Huang CC (1987) Binding of the Al-selective adenosine antagonist 8-cyclopentyl-l,3-dipropylxanthine to rat brain membranes. Naunyn Schmiedebergs Arch Pharmacol 335:59-63. Bruns RF, Lu G H and Pugsley T A (1986) Characterization of the A2 adenosine receptor labeled by [3HJNECA in rat striatal membranes. Mol Pharmacol 29:331-346. Buchan A M , Curtis SB and Meloche R M (1990) Release of somatostatin immunoreactivity from human antral D cells in culture. Gastroenterology 99:690-696. Buchan A M and Meloche R M (1994) Signal transduction events involved in bombesin-stimulated gastrin release from human G cells in culture. Can J Physiol Pharmacol 72:1060-1065. Buchan A M , Meloche R M , Kwok Y N and Kofod H (1993) Effect of cholecystokinin and secretin on somatostatin release from cultured antral cells. Gastroenterology 104:1414-1419. Buchan A M , Seal A M and Debas HT (1985a) Increased antral G-cell number and gastrin content in dogs after massive small bowel resection. Dig Dis Sci 30:236-239. Buchan A M , Sikora L K , Levy JG, Mcintosh CH, Dyck I and Brown JC (1985b) An immunocytochemical investigation with monoclonal antibodies to somatostatin. Histochemistry. 83:175-180. Burnstock G (1975) Comparative studies of purinergic nerves. J Exp Zool 194:103-133. Burnstock G (1978) A basis for distinguishing two types of purinergic receptor, in Cell membrane receptors for drugs and hormones: a multidisiplinary approach (Straub RW ed) pp 107-118, Raven press, New York. Burnstock G (2001) Purinergic signalling in the gut, in Purinergic andpyrimidinergic signalling I (Abbracchio MP and Williams M eds) pp 141-238, Springer, Berlin. Burnstock G, Campbell G, Satchell D and Smythe A (1970) Evidence that adenosine triphosphate or a related nucleotide is the transmitter substance released by non-adrenergic inhibitory nerves in the gut. Br J Pharmacol 40:668-688. Cabero JL, L i ZQ and Mardh S (1993) Gastrin action on aminopyrine accumulation in isolated pig parietal cells requires cAMP. Biochim Biophys Acta 1177:245-252. Campos RV, Buchan A M , Meloche R M , Pederson RA, Kwok Y N and Coy D H (1990) Gastrin secretion from human antral G cells in culture. Gastroenterology 99:36-44. Carlsson E, Larsson H, Mattsson H, Ryberg B and Sundell G (1986) Pharmacology and toxicology of omeprazole-with special reference to the effects on the gastric mucosa. Scand J Gastroenterol Suppl 118:31-38. Carruthers A M , Sellers L A , Jenkins DW, Jarvie E M , Feniuk W and Humphrey PP (2001) Adenosine A( l ) receptor-mediated inhibition of protein kinase A-induced calcitonin gene-related peptide release from rat trigeminal neurons. Mol Pharmacol 59:1533-1541. Centelles JJ, Franco R and Bozal J (1987) Distribution of adenosine deaminase in some rat tissues. Inhibition by ethanol and dimethyl sulfoxide. Comp Biochem Physiol 86:95-98. Chae HD and Kim K T (1997) Stimulation of the A2A adenosine receptor increases expression of the tyrosine hydroxylase gene. Brain Res Mol Brain Res 44:31-38. Cheng B, Essackjee HC and Ballard HJ (2000) Evidence for control of adenosine metabolism in rat oxidative skeletal muscle by changes in pH. J Physiol 522 Pt 3:467-477. Chern Y , King K , Lai H L and Lai HT (1992) Molecular cloning of a novel adenosine receptor gene from rat brain. Biochem Biophys Res Commun 185:304-309. Chiba T, Fujita T and Yamada T (1989) Carbachol inhibits stimulant-induced increases in fundic D-cell cytosolic Ca2+ concentration. Am J Physiol 257:G308-312. Chiba T, Taminato T, Kadowaki S, Abe H, Chihara K, Seino Y , Matsukura S and Fujita T (1980) Effects of glucagon, secretin, and vasoactive intestinal polypeptide on gastric somatostatin and gastrin release from isolated perfused rat stomach. Gastroenterology 79:67-71. Cho C H , Kaan SK and Wang H H (1995) Different pharmacological actions of adenosine on gastric function and mucosal damage in normotensive and spontaneously hypertensive rats. Pharmacology 50:261-265. Cho C H , Liao N X , Liu X G , Hui W M , Luk CT and Lam SK (1991) Effects of adenosine on gastric and cardiovascular systems, and ethanol ulceration in male and female rats. Eur J Pharmacol 201:157-162. Cho C H and Ogle CW (1990) Modulatory action of adenosine on gastric function and ethanol-induced mucosal damage in rats. DigDis Sci 35:1334-1339. Cho C H , Qiu BS, Ballard HJ and Ogle CW (1993) Adenosine and cholinergic-induced gastric contraction in rats. Digestion 54:98-104. Chomczynski P and Sacchi N (1987) Single-step method of R N A isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156-159. Chow DC and Forte JG (1993) Characterization of the beta-subunit of the H(+)-K(+)-ATPase using an inhibitory monoclonal antibody. Am J Physiol 265 :0 562-1570. Christofi FL (2001) Unlocking mysteries of gut sensory transmission: is adenosine the key? News Physiol Sci 16:201 -207. Christofi F L and Cook M A (1986) Affinity of various purine nucleosides for adenosine receptors on purified myenteric varicosities compared to their efficacy as presynaptic inhibitors of acetylcholine release. J Pharmacol Exp Ther 237:305-311. Christofi FL, Tack J and Wood JD (1992) Suppression of nicotinic synaptic transmission by adenosine in myenteric ganglia of the guinea-pig gastric antrum. Eur J Pharmacol 216:17-22. Christofi FL and Wood JD (1993) Presynaptic inhibition by adenosine A l receptors on guinea pig small intestinal myenteric neurons. Gastroenterology 104:1420-1429. Christofi FL, Zhang H, Y u JG, Guzman J, Xue J, Kim M , Wang Y Z and Cooke HJ (2001) Differential gene expression of adenosine A l , A2a, A2b, and A3 receptors in the human enteric nervous system. J Comp Neurol 439:46-64. Chu Y Y , Tu K H , Lee Y C , Kuo ZJ, Lai H L and Chern Y (1996) Characterization of the rat A2a adenosine receptor gene. DNA Cell Biol 15:329-337. Ciruela F, Casado V, Mallol J, Canela EI, Lluis C and Franco R (1995) Immunological identification of A l adenosine receptors in brain cortex. JNeurosci Res 42:818-828. Cohen S and Booth GH, Jr. (1975) Gastric acid secretion and lower-esophageal-sphincter pressure in response to coffee and caffeine. N Engl J Med 293:897-899. Comb M , Birnberg NC, Seasholtz A, Herbert E and Goodman H M (1986) A cyclic A M P -and phorbol ester-inducible D N A element. Nature 323:353-356. Conlay L A , Conant JA, deBros F and Wurtman R (1997) Caffeine alters plasma adenosine levels. Nature 389:136. Connolly AR, Cleland L G and Kirkham BW (1995) Mathematical considerations of competitive polymerase chain reaction. J Immunol Methods 187:201-211. Cooke A R (1976) Gastric damage by drugs and the role of the mucosal barrier. AustNZJ Med 6 Suppl LSuppl 1:26-32. Cornfield LJ , Hu S, Hurt SD and Sills M A (1992) [3H]2-phenylaminoadenosine ([3H]CV 1808) labels a novel adenosine receptor in rat brain. J Pharmacol Exp Ther 263:552-561. Coupar IM and Hancock D L (1994) The adenosine agonist N E C A inhibits intestinal secretion and peristalsis. JPharm Pharmacol 46:801-804. Cristalli G, Camaioni E, Costanzi S, Vittori S, Volpini R and Klotz K N (1998) Characterization of potent ligands at human recombinant adenosine receptors. DrugDev flas 45:176-181. Daly JW and Padgett W L (1992) Agonist activity of 2- and 5'-substituted adenosine analogs and their N6-cycloalkyl derivatives at A l - and A2-adenosine receptors coupled to adenylate cyclase. Biochem Pharmacol 43:1089-1093. Dalziel H H and Westfall DP (1994) Receptors for adenine nucleotides and nucleosides: subclassification, distribution, and molecular characterization. Pharmacol Rev 46:449-466. de Zwart M , Link R, von Frijtag Drabbe Kunzel JK, Cristalli G, Jacobson K A , Townsend-Nicholson A and AP IJ (1998) A functional screening of adenosine analogues at the adenosine A2B receptor: a search for potent agonists. Nucleosides Nucleotides 17:969-985. Delahunty T M , Cronin M J and Linden J (1988) Regulation of GH3-cell function via adenosine A l receptors. Inhibition of prolactin release, cyclic A M P production and inositol phosphate generation. Biochem J255:69-11. DelValle J, Park J, Chiba T and Yamada T (1990) Cellular mechanisms of somatostatin action in the gut. Metab Clin Exp 39:134-137. DelValle J, Wakasugi J, Takeda H and Yamada T (1996) Linkage of [Ca2+]i in single isolated D cells to somatostatin secretion induced by cholecystokinin. Am J Physiol 270:G897-901. DelValle J and Yamada T (1990) Amino acids and amines stimulate gastrin release from canine antral G-cells via different pathways. J Clin Invest 85:139-143. DeSchryver-Kecskemeti K, Greider M H , Rieders ER, Komyati SE and McGuigan JE (1981) In vitro gastrin secretion by rat antrum: effects of neurotransmitter agonists, antagonists, and modulators of secretion. Lab Invest 44:158-163. Deussen A (2000) Metabolic flux rates of adenosine in the heart. Naunyn Schmiedebergs Arch Pharmacol 362:351-363. Deussen A, Lloyd H G and Schrader J (1989) Contribution of S-adenosylhomocysteine to cardiac adenosine formation. JMol Cell Cardiol 21:773-782. Dickenson JM, Blank JL and Hil l SJ (1998) Human adenosine A l receptor and P2Y2-purinoceptor-mediated activation of the mitogen-activated protein kinase cascade in transfected CHO cells. Br J Pharmacol 124:1491-1499. Dimaline R, Evans D, Varro A and Dockray GJ (1991) Reversal by omeprazole of the depression of gastrin cell function by fasting in the rat. J Physiol 433:483-493. Dimaline R, Sandvik A K , Barclay G and Dockray GJ (1992) Functional control of gastrin releasing peptide (GRP) mRNA in rat stomach. FEBSLett 301:291-293. Diniz C, Leal S and Goncalves J (2003) Regional differences in the adenosine A(2) receptor-mediated modulation of contractions in rat vas deferens. Eur J Pharmacol 460:191-199. Dixon A K , Gubitz A K , Sirinathsinghji DJ, Richardson PJ and Freeman TC (1996) Tissue distribution of adenosine receptor mRNAs in the rat. Br J Pharmacol 118:1461-1468. Dockray GJ, Hamer C, Evans D, Varro A and Dimaline R (1991) The secretory kinetics of the G cell in omeprazole-treated rats. Gastroenterology 100:1187-1194. Dockray GJ, Varro A and Dimaline R (1996) Gastric endocrine cells: gene expression, processing, and targeting of active products. Physiol Rev 76:767-798. Dolphin A C , Forda SR and Scott R H (1986) Calcium-dependent currents in cultured rat dorsal root ganglion neurones are inhibited by an adenosine analogue. J Physiol 2>13mAl-61. Dragon S, Offenhauser N and Baumann R (2002) cAMP and in vivo hypoxia induce tob, ifr l , and fos expression in erythroid cells of the chick embryo. Am J Physiol 282. Dunwiddie TV, Diao L and Proctor WR (1997) Adenine nucleotides undergo rapid, quantitative conversion to adenosine in the extracellular space in rat hippocampus. / Neurosci 17:7673-7682. Durak I, Ormeci N , Akyol O, Canbolat O, Kavutcu M and Bulbul M (1994) Adenosine deaminase, 5'-nucleotidase, xanthine oxidase, superoxide dismutase, and catalase activities in gastric juices from patients with gastric cancer, ulcer, and atrophic gastritis. DigDis Sci 39:721-728. Edkins JS (1905) The chemical mechanism of gastric secretion. J Physiol 34:133. Ekblad E, Ekelund M , Graffner H, Hakanson R and Sundler F (1985) Peptide-containing nerve fibers in the stomach wall of rat and mouse. Gastroenterology 89:73-85. Feifel E, Obexer P, Andratsch M , Euler S, Taylor L, Tang A , Wei Y , Schramek H, Curthoys NP and Gstraunthaler G (2002) p38 M A P K mediates acid-induced transcription of PEPCK in LLC-PK(l)-FBPase(+) cells. Am J Physiol Renal Physiol 283:F678-688. Feoktistov I and Biaggioni I (1995) Adenosine A2b receptors evoke interleukin-8 secretion in human mast cells. An enprofylline-sensitive mechanism with implications for asthma. J Clin Invest 96:1979-1986. Feoktistov I, Goldstein A E and Biaggioni I (1999) Role of p38 mitogen-activated protein kinase and extracellular signal-regulated protein kinase kinase in adenosine A2B receptor-mediated interleukin-8 production in human mast cells. Mol Pharmacol 55:726-734. Ferre F (1992) Quantitative or semi-quantitative PCR: reality versus myth. PCR Meth Appl 2:1-9. Fink JS, Weaver DR, Rivkees SA, Peterfreund RA, Pollack A E , Adler E M and Reppert S M (1992) Molecular cloning of the rat A2 adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Brain Res Mol Brain Res 14:186-195. Fozard JR, Baur F and Wolber C (2003) Antagonist pharmacology of adenosine A2B receptors from rat, guinea pig and dog. Eur J Pharmacol 475:79-84. Franco R, Casado V, Ciruela F, Saura C, Mallol J, Canela EI and Lluis C (1997) Cell surface adenosine deaminase: much more than an ectoenzyme. Prog Neurobiol 52:283-294. Fredholm BB (1982) Adenosine actions and adenosine receptors after 1 week treatment with caffeine. Acta Physiol Scand 115:283-286. Fredholm BB, AP IJ, Jacobson K A , Klotz K N and Linden J (2001) International Union of Pharmacology. X X V . Nomenclature and classification of adenosine receptors. Pharmacol Rev 53:527-552. Freeman W M , Walker SJ and Vrana K E (1999) Quantitative RT-PCR: pitfalls and potential. Biotechniques 26:112-122, 124-115. Freissmuth M , Selzer E and Schutz W (1991) Interactions of purified bovine brain A l -adenosine receptors with G-proteins. Reciprocal modulation of agonist and antagonist binding. Biochem J 275 ( Pt 3):651-656. Fujimiya M and Kwok Y N (1997) Effect of carbachol on vascular and luminal release of immunoreactive gastrin from isolated perfused rat duodenum. DigDis Sci 42:634-639. Furlong TJ, Pierce K D , Selbie L A and Shine J (1992) Molecular characterization of a human brain adenosine A2 receptor. Brain Res Mol Brain Res 15:62-66. Furness JB, Young H M , Pompolo S, Bornstein JC, Kunze W A and McConalogue K (1995) Plurichemical transmission and chemical coding of neurons in the digestive tract. Gastroenterology 108:554-563. Gallo-Rodriguez C, Ji X D , Melman N , Siegman BD, Sanders L H , Orlina J, Fischer B, Pu Q, Olah M E , van Galen PJ and et al. (1994) Structure-activity relationships of N6-benzyladenosine-5'-uronamides as A3-selective adenosine agonists. J Med Chem 37:636-646. Gao ZG, Chen A , Barak D, Kim SK, Muller CE and Jacobson K A (2002) Identification by site-directed mutagenesis of residues involved in ligand recognition and activation of the human A3 adenosine receptor. J Biol Chem 277:19056-19063. Geiger JD and Glavin GB (1985) Adenosine receptor activation in brain reduces stress-induced ulcer formation. Eur J Pharmacol 115:185-190. Geiger JD, Parkinson PE and Kowaluk E A (1997) Regulators of endogenous adenosine levels as therapeutic agents, in Purinertgic Approaches in Experimental Therapeutics (Jacobson K A and Jarvis MF eds) pp 55-84, Wiley-Liss, Inc., New York. Gerber JG, Fadul S, Payne N A and Nies AS (1984) Adenosine: a modulator of gastric acid secretion in vivo. J Pharmacol Exp Ther 231:109-113. Gerber JG and Guth PH (1989) Role of adenosine in the gastric blood flow response to pentagastrin in the rat. J Pharmacol Exp Ther 251:550-556. Gerber JG, Nies AS and Payne N A (1985) Adenosine receptors on canine parietal cells modulate gastric acid secretion to histamine. J Pharmacol Exp Ther 233:623-627. Gerwins P and Fredholm BB (1991) Glucocorticoid receptor activation leads to up-regulation of adenosine A l receptors and down-regulation of adenosine A2 responses in DDT1 MF-2 smooth muscle cells. Mol Pharmacol 40:149-155. Gerwins P and Fredholm BB (1995) Activation of phospholipase C and phospholipase D by stimulation of adenosine A l , bradykinin or P2U receptors does not correlate well with protein kinase C activation. Naunyn Schmiedebergs Arch Pharmacol 351:194-201. Gil-Rodrigo CE, Bergaretxe I, Carou M , Galdiz B, Salgado C and Ainz LF (1996) Inhibitory action of extracellular adenosine 5'-triphosphate on parietal cells isolated from rabbit gastric mucosa. Gen Physiol Biophys 15:251-264. Gil-Rodrigo CE, Galdiz B, Gandarias JM, Gomez R and Ainz LF (1990) Characterization of the effects of adenosine, adenosine 5'-triphosphate and related purines on acid secretion in isolated rabbit gastric glands. Pharmacol Res 22:103-113. Glavin GB, Westerberg VS and Geiger JD (1987) Modulation of gastric acid secretion by adenosine in conscious rats. Can J Physiol Pharmacol 65:1182-1185. Gould J, Morton MJ, Sivaprasadarao A, Bowmer CJ and Yates MS (1997) Renal adenosine A l receptor binding characteristics and mRNA levels during the development of acute renal failure in the rat. Br J Pharmacol 120:947-953. Green A , Milligan G and Dobias SB (1992) Gi down-regulation as a mechanism for heterologous desensitization in adipocytes. J Biol Chem 267:3223-3229. Grover A K , Agrawal DK, Ahmad S, Daniel EE, Kwan C Y , Oakes PJ, Sipos SN, Berenski C and Jung C (1985) Target size of 5'-nucleotidase in smooth muscle. JBiochem (Tokyo) 98:573-575. Grover A K , Oakes P, Sipos SN, Kwan C Y and Garfield RE (1983) Studies on canine gastric antrum smooth muscle: preparation and characterization of a plasma membrane enriched fraction. Can J Physiol Pharmacol 61:927-940. Gubitz A K , Widdowson L, Kurokawa M , Kirkpatrick K A and Richardson PJ (1996) Dual signalling by the adenosine A2a receptor involves activation of both N - and P-type calcium channels by different G proteins and protein kinases in the same striatal nerve terminals. JNeurochem 67:374-381. Guo Y S , Bhalla V K , Thompson JC and Singh P (1988) Effect of cyclic nucleotides on bombesin-evoked gastrin release from isolated perfused rat stomach. Regal Pept 22:361-369. Gustafsson L (1981) Influence of adenosine on responses to vagal nerve stimulation in the anesthetized rabbit. Acta Physiol Scand 111 :263-268. Gustafsson L E , Wiklund NP, Lundin J and Hedqvist P (1985) Characterization of pre- and post-junctional adenosine receptors in guinea-pig ileum. Acta Physiol Scand 123:195-203. Hancock D L and Coupar IM (1995) Functional characterization of the adenosine receptor mediating inhibition of peristalsis in the rat jejunum. Br J Pharmacol 115:739-744. Harty RE and Franklin PA (1984) Effects of exogenous and endogenous adenosine on gastrin release from rat antral mucosa. Gastroenterology 86:1107. Hayashi E, Mori M , Yamada S and Kumitomo M (1978) Effects of purine compounds on cholinergic nerves. Specificity of adenosine and related compounds on acetylcholine release in electircally stimulated guinea pig ileum. Eur J Pharmacol 48:297-307. Hayward A L , Oefner PJ, Sabatini S, Kainer DB, Hinojos C A and Doris PA (1998) Modeling and analysis of competitive RT-PCR. Nucleic Acids Res 26:2511-2518. Heid CA, Stevens J, Livak K J and Williams P M (1996) Real time quantitative PCR. Genome Res 6:986-994. Heldsinger A A , Vinik AI and Fox IH (1986) Inhibition of guinea-pig oxyntic cell function by adenosine and prostaglandins. J Pharmacol Exp Ther 237:351-356. Henley W N , Schuebel K E and Nielsen D A (1996) Limitations imposed by heteroduplex formation on quantitative RT-PCR. Biochem Biophys Res Commun 226:113-117. Hersey SJ and Sachs G (1995) Gastric acid secretion. Physiol Rev 75:155-189. Hiley CR, Bottrill FE, Warnock J and Richardson PJ (1995) Effects of pH on responses to adenosine, CGS 21680, carbachol and nitroprusside in the isolated perfused superior mesenteric arterial bed of the rat. Br J Pharmacol 116:2641-2646. Hirschowitz BI, Keeling D, Lewin M , Okabe S, Parsons M , Sewing K , Wallmark B and Sachs G (1995) Pharmacological aspects of acid secretion. DigDis Sci 40:3S-23S. Hoist JJ, Jensen SL, Knuhtsen S, Nielsen OV and Rehfeld JF (1983) Effect of vagus, gastric inhibitory polypeptide, and HC1 on gastrin and somatostatin release from perfused pig antrum. Am J Physiol 244:G515-522. Holz FG and Steinhausen M (1987) Renovascular effects of adenosine receptor agonists. Ren Physiol 10:272-282. Hutchison AJ , Webb RL, Oei HH, Ghai GR, Zimmerman M B and Williams M (1989) CGS 21680C, an A2 selective adenosine receptor agonist with preferential hypotensive activity. J Pharmacol Exp Ther 251:47-55. IJzerman A , Van Galen PJ and Jacobson K A (1992) Molecular modeling of adenosine receptors. I. The ligand binding site on the A l receptor. DrugDes Discov 9:49-67. IJzerman AP, van der Wenden E M , van Galen PJ and Jacobson K A (1994) Molecular modeling of adenosine receptors. The ligand binding site on the rat adenosine A2A receptor. Eur J Pharmacol 268:95-104. Imoto A , Inoue R, Tanaka M and Ito Y (1998) Inhibitory N A N C neurotransmission in choledocho-duodenal junction of rabbits~a possible role of P A C A P . JAuton Nerv Syst 70:189-199. Inauen W, Wyss PA, Kayser S, Baumgartner A, Schurer-Maly CC, Koelz HR and Halter F (1988) Influence of prostaglandins, omeprazole, and indomethacin on healing of experimental gastric ulcers in the rat. Gastroenterology. 95:636-641. Jacobson K A (1996) Specific Ligands for the Adenosine Receptor Family. Neurotransmissions 12:1-6. Jacobson K A , Ravi RG, Nandanan E, Kim HS, Moro S, Kim Y C , Lee K, Barak D, Marquez V E and Ji X D (2001) Ribose modified nucleosides and nucleotides as ligands for purine receptors. Nucleosides, Nucleotides & Nucleic Acids 20:333-341. Jacobson M A , Johnson RG, Luneau CJ and Salvatore C A (1995) Cloning and chromosomal localization of the human A2b adenosine receptor gene (ADORA2B) and its pseudogene. Genomics 27:374-376. Jaffe B and Walsh JH (1978) Gastrin and related peptides, in Methods of Hormone Radioimmunoassay (Jaffe b M and Behrman HR eds) pp 455-477, Academic Press, New York. Ji X D and Jacobson K A (1999) Use of the triazolotriazine [3H]ZM 241385 as a radioligand at recombinant human A2B adenosine receptors. Drug Des Discov 16:217-226. Jiang X , Suzaki E and Kataoka K (2002) Immunofluorescence detection of gastric H(+)/K(+)-ATPase and its alterations as related to acid secretion. Histochem Cell Biol 117:21-27. Jockers R, Linder M E , Hohenegger M , Nanoff C, Bertin B, Strosberg A D , Marullo S and Freissmuth M (1994) Species difference in the G protein selectivity of the human and bovine Al-adenosine receptor. J Biol Chem 269:32077-32084. Johansson B, Ahlberg S, van der Ploeg I, Brene S, Lindefors N , Persson H and Fredholm BB (1993) Effect of long term caffeine treatment on A l and A2 adenosine receptor binding and on mRNA levels in rat brain. Naunyn Schmiedebergs Arch Pharmacol 347:407-414. Kadowaki M , Takeda M , Tokita K, Hanaoka K and Tomoi M (2000) Molecular identification and pharmacological characterization of adenosine receptors in the guinea-pig colon. Br J Pharmacol 129:871-876. Kapuscinski M and Shulkes A (1995) Secretory and biosynthetic responses of gastrin and somatostatin to acute changes in gastric acidity. J Gastroenterol Hepatol 10:405-412. Keast JR, Furness JB and Costa M (1985) Distribution of certain peptide-containing nerve fibres and endocrine cells in the gastrointestinal mucosa in five mammalian species. J Comp Neurol 236:403-422. Kim J, Wess J, van Rhee A M , Schoneberg T and Jacobson K A (1995) Site-directed mutagenesis identifies residues involved in ligand recognition in the human A2a adenosine receptor. J Biol Chem 270:13987-13997. Kim Y C , Ji X , Melman N , Linden J and Jacobson K A (2000) Anilide derivatives of an 8-phenylxanthine carboxylic congener are highly potent and selective antagonists at human A(2B) adenosine receptors. J Med Chem 43:1165-1172. Kintner DB, Anderson M K , Fitzpatrick JH, Jr., Sailor K A and Gilboe DD (2000) 31P-MRS-based determination of brain intracellular and interstitial pH: its application to in vivo H+ compartmentation and cellular regulation during hypoxic/ischemic conditions. Neurochem Res 25:1385-1396. Kivilaakso E and Kiviluoto T (1988) Intracellular pH in isolated Necturus antral mucosa in simulated ulcerogenic conditions. Gastroenterology 95:1198-1205. Kiviluoto T, Paimela H, Mustonen H and Kivilaakso E (1990) Intracellular pH in isolated Necturus antral mucosa exposed to luminal acid. Gastroenterology 98:901-908. Klotz K N (2000) Adenosine receptors and their ligands. Naunyn Schmiedebergs Arch Pharmacol 362:382-391. Klotz K N , Hessling J, Hegler J, Owman C, Kull B, Fredholm BB and Lohse M J (1998) Comparative pharmacology of human adenosine receptor subtypes - characterization of stably transfected receptors in CHO cells. Naunyn Schmiedebergs Arch Pharmacol 357:1-9. Kobayashi S and Millhorn DE (1999) Stimulation of expression for the adenosine A 2 A receptor gene by hypoxia in PC 12 cells. A potential role in cell protection. J Biol Chem 274:20358-20365. Konturek SJ, Brzozowski T, Pajdo R, Konturek PC, Kwiecien S, Sliwowski Z, Pawlik M , Ptak A , Drozdowicz D and Hahn E G (2001) Gastric preconditioning induced by short ischemia: the role of prostaglandins, nitric oxide and adenosine. Med Sci Monit 7:610-621. Koop H, Behrens I, Bothe E, Koschwitz H, Mcintosh C H , Pederson RA, Arnold R and Creutzfeldt W (1983) Adrenergic control of rat gastric somatostatin and gastrin release. Scand J Gastroenterol 18:65-71. Koop H , Behrens I, Bothe E, Mcintosh CH, Pederson RA, Arnold R and Creutzfeldt W (1982) Adrenergic and cholinergic interactions in rat gastric somatostatin and gastrin release. Digestion 25:96-102. Kovacs TO, Walsh JH, Maxwell V , Wong HC, Azuma T and Katt E (1989) Gastrin is a major mediator of the gastric phase of acid secretion in dogs: proof by monoclonal antibody neutralization. Gastroenterology 97:1406-1413. Krasnow S and Grossman MI (1949) Stimulation of gastric secretion in man by theophylline ethylenediamine. Proc Soc Exp Biol Med 71:335-336. Kull B, Svenningsson P and Fredholm BB (2000) Adenosine A(2A) receptors are colocalized with and activate g(olf) in rat striatum. Mol Pharmacol 58:771-777. Kusumoto Y and Grube D (1987) Somatotostatin (D-) cells in the rat pyloric antrum, with special reference to the destination of their cytoplasmic processes. Biomed Res 8:145-151. Kusumoto Y , Sato A G , Kaneda K and Nakamae E (1995) Cytology and microenvironment of somatostatin (D) cells in the gastric fundic mucosa of rodents. Hiroshima J Med Sci 44:105-112. Kwok Y N , Mcintosh C and Brown J (1990) Augmentation of release of gastric somatostatin-like immunoreactivity by adenosine, adenosine triphosphate and their analogs. J Pharmacol Exp Ther 255:781-788. Kwok Y N and Mcintosh C H (1990) Release of substance P-like immunoreactivity from the vascularly perfused rat stomach. Eur J Pharmacol 180:201-207. Kwok Y N , Mcintosh C H , Pederson R A and Brown JC (1985) Effect of substance P on somatostatin release from the isolated perfused rat stomach. Gastroenterology 88:90-95. Kwok Y N , Mcintosh C H , Sy H and Brown JC (1988) Inhibitory actions of tachykinins and neurokinins on release of somatostatin-like immunoreactivity from the isolated perfused rat stomach. J Pharmacol Exp Ther 246:726-731. Larsson H, Carlsson E, Ryberg B, Fryklund J and Wallmark B (1988) Rat parietal cell function after prolonged inhibition of gastric acid secretion. Am J Physiol 254:G33-39. Larsson LI and Hougaard D M (1993) Sensitive detection of rat gastrin mRNA by in situ hybridization with chemically biotinylated oligodeoxynucleotides: validation, quantitation, and double-staining studies. JHistochem Cytochem 41:157-163. Latini S, Bordoni F, Pedata F and Corradetti R (1999) Extracellular adenosine concentrations during in vitro ischaemia in rat hippocampal slices. Br J Pharmacol 127:729-739. Le F, Townsend-Nicholson A, Baker E, Sutherland GR and Schofield PR (1996) Characterization and chromosomal localization of the human A2a adenosine receptor gene: ADORA2A. Biochem Biophys Res Commun 223:461-467'. Le Poole IC, Yang F, Brown TL, Cornelius J, Babcock GF, Das P K and Boissy RE (1999) Altered gene expression in melanocytes exposed to 4-tertiary butyl phenol (4-TBP): upregulation of the A2b adenosine receptor 1. J Invest Dermatol 113:725-731. Lee CW and Jarvis S M (1988) Nucleoside transport in rat cerebral-cortical synaptosomes. Evidence for two types of nucleoside transporters. Biochem J 249:557-564. Lee H, Hakanson R, Karlsson A, Mattsson H and Sundler F (1992) Lansoprazole and omeprazole have similar effects on plasma gastrin levels, enterochromaffm-like cells, gastrin cells and somatostatin cells in the rat stomach. Digestion 51:125-132. Lee Y C , Chang CW, Su CW, Lin TN, Sun SH, Lai H L and Chern Y (1999) The 5' untranslated regions of the rat A 2 A adenosine receptor gene function as negative translational regulators. JNeurochem 73:1790-1798. Lee Y C , Lai HL, Sun C N , Chien C L and Chern Y (2003) Identification of nuclear factor 1 (NF1) as a transcriptional modulator of rat A(2A) adenosine receptor. Brain Res Mol Brain Res 111:61-73. Lehy T, Gres L and Ferreira de Castro E (1979) Quantitation of gastrin and somatostatin cell populations in the antral mucosa of the rat. Comparative distribution and evolution through different life stages. Cell Tissue Res 198:325-333. L i ZQ, Cabero JL and Mardh S (1995) Gastrin and carbachol require cAMP to elicit aminopyrine accumulation in isolated pig and rat parietal cells. Am J Physiol 268:G82-Libert F, Parmentier M , Lefort A , Dinsart C, Van Sande J, Maenhaut C, Simons MJ, Dumont JE and Vassart G (1989) Selective amplification and cloning of four new members of the G protein-coupled receptor family. Science 244:569-572. Libert F, Van Sande J, Lefort A , Czernilofsky A , Dumont JE, Vassart G, Ensinger H A and Mendla K D (1992) Cloning and functional characterization of a human A l adenosine receptor. Biochem Biophys Res Commun 187:919-926. Lichtenberger L M , Lechago J and Johnson L R (1975) Depression of antral and serum gastrin concentration by food deprivation in the rat. Gastroenterology 68:1473-1479. Lloyd HG, Deussen A, Wuppermann H and Schrader J (1988) The transmethylation pathway as a source for adenosine in the isolated guinea-pig heart. Biochem J 252:489-494. Lloyd K C , Raybould HE, Tache Y and Walsh JH (1992) Role of gastrin, histamine, and acetylcholine in the gastric phase of acid secretion in anesthetized rats. Am J Physiol 262:G747-755. Lockey C, Otto E and Long Z (1998) Real-time fluorescence detection of a single D N A molecule. Biotechniques 24:744-746. Lohse MJ , Klotz K N , Lindenborn-Fotinos J, Reddington M , Schwabe U and Olsson R A (1987) 8-Cyclopentyl-l,3-dipropylxanthine (DPCPX)--a selective high affinity antagonist radioligand for A l adenosine receptors. Naunyn Schmiedebergs Arch Pharmacol 336:204-210. Londos C, Cooper D M and Wolff J (1980) Subclasses of external adenosine receptors. Proc Natl Acad Sci USA 77:2551-2554. Lorbar M , Fenton RA, Duffy A J, Graybill C A and Dobson JG, Jr. (1999) Effect of aging on myocardial adenosine production, adenosine uptake and adenosine kinase activity in rats. JMol Cell Cardiol 31:401-412. Lupica CR, Cass W A , Zahniser NR and Dunwiddie TV (1990) Effects of the selective adenosine A2 receptor agonist CGS 21680 on in vitro electrophysiology, cAMP formation and dopamine release in rat hippocampus and striatum. J Pharmacol Exp Ther 252:1134-1141. Lynge J and Hellsten Y (2000) Distribution of adenosine A l , A 2 A and A2B receptors in human skeletal muscle. Acta Physiol Scand 169:283-290. Mabley J, Soriano F, Pacher P, Hasko G, Marton A, Wallace R, Salzman A and Szabo C (2003) The adenosine A3 receptor agonist, N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide, is protective in two murine models of colitis. Eur J Pharmacol 466:323-329. Magee DF (1996) Pyloric antral inhibition of gastrin release. J Gastroenterol 31:75 8-763. Mahan L C , McVittie L D , Smyk-Randall E M , Nakata H , Monsma FJ, Jr., Gerfen CR and Sibley DR (1991) Cloning and expression of an A1 adenosine receptor from rat brain. Mol Pharmacol 40:1-7. Malek RL, Nie Z, Ramkumar V and Lee N H (1999) Adenosine A(2A) receptor mRNA regulation by nerve growth factor is TrkA-, Src-, and Ras-dependent via extracellular regulated kinase and stress-activated protein kinase/c-Jun NH(2)-terminal kinase. J Biol Chem 274:35499-35504. Manela FD, Ren J, Gao J, McGuigan JE and Harty RF (1995) Calcitonin gene-related peptide modulates acid-mediated regulation of somatostatin and gastrin release from rat antrum. Gastroenterology 109:701-706. Martindale R, Kauffman GL, Levin S, Walsh JH and Yamada T (1982) Differential regulation of gastrin and somatostatin secretion from isolated perfused rat stomachs. Gastroenterology 83:240-244. Matharu MS and Hollingsworth M (1992) Purinoceptors mediating relaxation and spasm in the rat gastric fundus. Br J Pharmacol 106:395-403. Matsumoto A, Asada S, Saitoh O, Tei H, Okumura Y , Hirata I and Ohshiba S (1989) A study on gastric ulcers induced by long-term fasting in rats. Scand J Gastroenterol Suppl 162:75-78. Matsuno M , Matsui T, Iwasaki A and Arakawa Y (1997) Role of acetylcholine and gastrin-releasing peptide (GRP) in gastrin secretion. J Gastroenterol 32:579-586. Mcintosh C, Arnold R, Bothe E, Becker H, Kobberling J and Creutzfeldt W (1978) Gastrointestinal somatostatin: extraction and radioimmunoassay in different species. Gut. 19:655-663. Mcintosh C H (1985) Gastrointestinal somatostatin: distribution, secretion and physiological significance. Life Sci 37:2043-2058. Mcintosh C H , Bakich V , Kwok Y N , Wong J and Brown JC (1987a) The effects of substance P, histamine and histamine antagonists on somatostatin and gastrin release from the isolated perfused rat stomach. Regul Pept 19:253-263. Mcintosh C H , Kwok Y N , Mordhorst T, Nishimura E, Pederson R A and Brown JC (1983) Enkephalinergic control of somatostatin secretion from the perfused rat stomach. Can J Physiol Pharmacol 61:657-663. Mcintosh CHS, Kwok Y N , Tang C and Brown JC (1987b) The use of monoclonal antibodies in RIA for gastrointestinal hormones. J Clin Immunoassay 10:79-84. Meghji P, Middleton K M and Newby A C (1988) Absolute rates of adenosine formation during ischaemia in rat and pigeon hearts. Biochem 7249:695-703. Melani A , Pantoni L, Corsi C, Bianchi L, Monopoli A , Bertorelli R, Pepeu G and Pedata F (1999) Striatal outflow of adenosine, excitatory amino acids, gamma-aminobutyric acid, and taurine in awake freely moving rats after middle cerebral artery occlusion: correlations with neurological deficit and histopathological damage. Stroke 30:2448-2454; discussion 2455. Mendelson WB, Kuruvilla A , Watlington T, Goehl K, Paul S M and Skolnick P (1983) Sedative and electroencephalographic actions of erythro-9-(2-hydroxy-3-nonyl)-adenine (EHNA): relationship to inhibition of brain adenosine deaminase. Psychopharmacology 79:126-129. Meno JR, Ngai A C , Ibayashi S and Winn HR (1991) Adenosine release and changes in pial arteriolar diameter during transient cerebral ischemia and reperfusion. J Cereb Blood FlowMetab 11:986-993. Milusheva EA, Doda M , Baranyi M and Viz i ES (1996) Effect of hypoxia and glucose deprivation on ATP level, adenylate energy charge and [Ca2+]o-dependent and independent release of [3H]dopamine in rat striatal slices. Neurochem Int 28:501-507. Montminy M , Brindle P, Arias J, Ferreri K and Armstrong R (1996a) Regulation of somatostatin gene transcription by cAMP. Adv Pharmacol 36:1-13. Montminy M , Brindle P, Arias J, Ferreri K and Armstrong R (1996b) Regulation of somatostatin gene transcription by cyclic adenosine monophosphate. Metab Clin Exp 45:4-7. Montminy MR, Sevarino K A , Wagner JA, Mandel G and Goodman R H (1986) Identification of a cyclic-AMP-responsive element within the rat somatostatin gene. Proc Natl Acad Sci USA 83:6682-6686. Moriwaki Y , Yamamoto T and Higashino K (1999) Enzymes involved in purine metabolism--a review of histochemical localization and functional implications. Histol Histopathol 14:1321-1340. Moriwaki Y , Yamamoto T, Yamaguchi K, Takahashi S and Higashino K (1996) Immunohistochemical localization of aldehyde and xanthine oxidase in rat tissues using polyclonal antibodies. Histochem Cell Biol 105:71-79. Muller CE (2001) A l adenosine receptors and their ligands: overview and recent developments. Farmaco 56:77-80. Muller CE, Geis U , Hipp J, Schobert U , Frobenius W, Pawlowski M , Suzuki F and Sandoval-Ramirez J (1997) Synthesis and structure-activity relationships of 3,7-dimethyl-l-propargylxanthine derivatives, A2A-selective adenosine receptor antagonists. J Med Chem 40:4396-4405. Murrison E M , Goodson SJ, Edbrooke M R and Harris C A (1996) Cloning and characterisation of the human adenosine A3 receptor gene. FEBS Lett 384:243-246. Murthy VS, Zagar M E , Vollmer RR and Schmidt D H (1982) Pentobarbital-induced changes in vagal tone and reflex vagal activity in rabbits. Eur J Pharmacol 84:41-50. Nagata H, Sekizuka E, Morishita T, Tatemichi M , Kurokawa T, Mizuki A and Ishii H (1996) Adenosine A2-receptor mediates ethanol-induced arteriolar dilation in rat stomach. Am J Physiol 271 :G1028-1033. Nakata H (1993) Development of an antiserum to rat-brain A l adenosine receptor: application for immunological and structural comparison of A l adenosine receptors from various tissues and species. Biochim Biophys Acta 1177:93-98. Namiot Z, Marcinkiewicz M , Jaroszewicz W, Stasiewicz J and Gorski J (1993) Mucosal adenosine deaminase activity and gastric ulcer healing. Eur J Pharmacol 243:301-303. Namiot Z, Rutkiewicz J, Stasiewicz J, Baranczuk E and Marcinkiewicz M (1991) Adenosine deaminase activity in the gastric mucosa in patients with gastric ulcer. Effects of ranitidine and sucralfate. Eur J Pharmacol 205:101-103. Namiot Z, Rutkiewicz J, Stasiewicz J and Gorski J (1990) Adenosine deaminase activity in the human gastric mucosa in relation to acid secretion. Digestion 45:172-175. Navarro A, Zapata R, Canela EI, Mallol J, Lluis C and Franco R (1999) Epidermal growth factor (EGF)-induced up-regulation and agonist- and antagonist-induced desensitization and internalization of A l adenosine receptors in a pituitary-derived cell line. Brain Res 816:47-57. Nicholls J, Brownhill V R and Hourani S M (1996) Characterization of Pl-purinoceptors on rat isolated duodenum longitudinal muscle and muscularis mucosae. Br J Pharmacol 117:170-174. Nicholls J, Howard S M and Kitchen I (1992) Characterization of Pl-purinoceptors on rat duodenum and urinary bladder. Br J Pharmacol 105:639-642. Nie Z, Mei Y , Ford M , Rybak L, Marcuzzi A , Ren H, Stiles G L and Ramkumar V (1998) Oxidative stress increases A l adenosine receptor expression by activating nuclear factor kappa B. Mol Pharmacol 53:663-669. Nie Z, Mei Y , Malek RL , Marcuzzi A , Lee N H and Ramkumar V (1999) A role of p75 in NGF-mediated down-regulation of the A(2A) adenosine receptors in PC 12 cells. Mol Pharmacol 56:947-954. Nitahara K , Kittel A , Liang SD and Viz i ES (1995) A1-receptor-mediated effect of adenosine on the release of acetylcholine from the myenteric plexus: role and localization of ecto-ATPase and 5'-nucleotidase. Neuroscience 67:159-168. Olah M E (1997) Identification of A2a adenosine receptor domains involved in selective coupling to Gs. Analysis of chimeric A l / A 2 a adenosine receptors. J Biol Chem 272:337-344. Olah M E and Stiles G L (1995) Adenosine receptor subtypes: characterization and therapeutic regulation. Ann Rev Pharmacol Toxicol 35:581-606. Olah M E and Stiles G L (2000) The role of receptor structure in determining adenosine receptor activity. Pharmacol Ther 85:55-75. Oomori Y , Satoh Y , Ishikawa K and Gesase AP (1997) Exocytosis in the antral gastrin cells of mouse, rat, and guinea pig after stimulation by carbamylcholine. Cell Tissue Res 289:463-472. Orlando C, Pinzani P and Pazzagli M (1998) Developments in quantitative PCR. Clin Chem Lab Med 36:255-269. Ota S, Hata Y , Hiraishi H, Mutoh H, Terano A and Sugimoto T (1992) The effects of acid secretagogues on protective agents of gastric cells from adult rabbits in vitro. J Clin Gastroenterol 14:S156-161. Ota S, Hiraishi H , Terano A, Mutoh H, Kurachi Y , Shimada T, Ivey K J and Sugimoto T (1989) Effect of adenosine and adenosine analogs on [14C]aminopyrine accumulation by rabbit parietal cells. DigDis Sci 34:1882-1889. Othman T, Legare D, Sadri P, Lautt WW and Parkinson FE (2002) A preliminary investigation of the effects of maternal ethanol intake during gestation and lactation on brain adenosine A( l ) receptor expression in rat offspring. Neurotoxicol Teratol 24:275-279. Pajdo R, Brzozowski T, Konturek PC, Kwiecien S, Konturek SJ, Sliwowski Z, Pawlik M , Ptak A , Drozdowicz D and Hahn E G (2001) Ischemic preconditioning, the most effective gastroprotective intervention: involvement of prostaglandins, nitric oxide, adenosine and sensory nerves. Eur J Pharmacol 427:263-276. Pak M A , Haas HL, Decking U K and Schrader J (1994) Inhibition of adenosine kinase increases endogenous adenosine and depresses neuronal activity in hippocampal slices. Neuropharmacology 33:1049-1053. Palmer JM, Wood JD and Zafirov D H (1987) Purinergic inhibition in the small intestinal myenteric plexus of the guinea-pig. J Physiol 387:357-369. Palmer T M , Gettys TW and Stiles G L (1995a) Differential interaction with and regulation of multiple G-proteins by the rat A3 adenosine receptor. J Biol Chem 270:16895-16902. Palmer T M , Poucher SM, Jacobson K A and Stiles G L (1995b) 125I-4-(2-[7-amino-2-[2-furyl][l,2,4]triazolo[2,3-a][l,3,5] triazin-5-yl-amino]ethyl)phenol, a high affinity antagonist radioligand selective for the A2a adenosine receptor. Mol Pharmacol 48:970-974. Park J, Chiba T and Yamada T (1987) Mechanisms for direct inhibition of canine gastric parietal cells by somatostatin. J Biol Chem 262:14190-14196. Patel Y C (1999) Somatostatin and its receptor family. Front Neuroendocrinol 20:157-198. Pawlikowski M , Karbownik M , Lewinski A , Pisarek H, Wajs E and Szkudlinski M (1992) Effects of omeprazole on the number of immunoreactive gastrin- and somatostatin-cells in the rat gastric mucosa. Histol Histopathol 7:153-156. Payne N A and Gerber JG (1997) Effect of adenosine and histamine receptor stimulation on canine histamine release to pentagastrin. Digestion 58:352-360. Pazzagli M , Corsi C, Fratti S, Pedata F and Pepeu G (1995) Regulation of extracellular adenosine levels in the striatum of aging rats. Brain Res 684:103-106. Peachey JA, Hourani S M and Kitchen I (1996) Differential development of adenosine A l and A2b receptors in the rat duodenum. Br J Pharmacol 119:949-958. Peachey JA, Hourani S M and Kitchen I (1999) Ontogeny of adenosine receptors in the longitudinal muscle and muscularis mucosae of the rat distal colon. Naunyn Schmiedebergs Arch Pharmacol 359:140-146. Peakman M C and Hi l l SJ (1994) Adenosine A2B-receptor-mediated cyclic A M P accumulation in primary rat astrocytes. Br J Pharmacol 111:191-198. Pederson RA, Kwok Y N , Buchan A M , Mcintosh C H and Brown JC (1984) Gastrin release from isolated perfused rat stomach after vagotomy. Am J Physiol 247:G248-252. Peterfreund RA, Gies E K and Fink JS (1997) Protein kinase C regulates adenosine A2a receptor mRNA expression in SH-SY5Y cells. Eur J Pharmacol 336:71-80. Peterfreund RA, MacCollin M , Gusella J and Fink JS (1996) Characterization and expression of the human A2a adenosine receptor gene. JNeurochem 66:362-368. Phillis JW, O'Regan M H and Perkins L M (1992) Measurement of rat plasma adenosine levels during normoxia and hypoxia. Life Sci 51:PL149-152. Phillis JW, Walter GA, O'Regan M H and Stair RE (1987) Increases in cerebral cortical perfusate adenosine and inosine concentrations during hypoxia and ischemia. J Cereb Blood Flow Metab 7:679-686. Piatak M , Jr., Luk K C , Williams B and Lifson JD (1993) Quantitative competitive polymerase chain reaction for accurate quantitation of HIV D N A and R N A species. Biotechniques 14:70-81. Pierce K D , Furlong TJ, Selbie L A and Shine J (1992) Molecular cloning and expression of an adenosine A2b receptor from human brain. Biochem Biophys Res Commun 187:86-93. Poucher SM, Keddie JR, Brooks R, Shaw GR and McKillop D (1996) Pharmacodynamics of Z M 241385, a potent A2a adenosine receptor antagonist, after enteric administration in rat, cat and dog. JPharm Pharmacol 48:601-606. Poucher SM, Keddie JR, Singh P, Stoggall SM, Caulkett PW, Jones G and Coll M G (1995) The in vitro pharmacology of Z M 241385, a potent, non-xanthine A2a selective adenosine receptor antagonist. Br J Pharmacol 115:1096-1102. Prentice DJ and Hourani S M (1997) Adenosine analogues relax guinea-pig taenia caeci via an adenosine A2B receptor and a xanthine-resistant site. Eur J Pharmacol 323:103-106. Prinz C, Kajimura M , Scott DR, Mercier F, Helander HF and Sachs G (1993) Histamine secretion from rat enterochromaffinlike cells. Gastroenterology 105:449-461. Puurunen J, Aittakumpu R and Tanskanen T (1986) Vagally mediated stimulation of gastric acid secretion by intravenously administered adenosine derivatives in anaesthetized rats. Acta Pharmacol Toxicol 58:265-271. Puurunen J and Huttunen P (1988) Central gastric antisecretory action of adenosine in the rat. Eur J Pharmacol 147:59-66. Puurunen J, Ruoff HJ and Schwabe U (1987) Lack of direct effect of adenosine on the parietal cell function in the rat. Pharmacol Toxicol 60:315-317. Raeymaekers L (1995) A commentary on the practical applications of competitive PCR. Genome Res 5:91-94. Ralevic V and Burnstock G (1998) Receptors for purines and pyrimidines. Pharmacol Rev 50:413-492. Ramkumar V , Olah M E , Jacobson K A and Stiles G L (1991) Distinct pathways of desensitization of A l - and A2-adenosine receptors in DDT1 MF-2 cells. Mol Pharmacol 40:639-647. Ramkumar V , Wilson M , Dhanraj DN, Gettys TW and A l i H (1995) Dexamethasone up-regulates A3 adenosine receptors in rat basophilic leukemia (RBL-2H3) cells. J Immunol 154:5436-5443. Rasmussen TN, Schmidt P, Poulsen SS and Hoist JJ (2001) Effect of calcitonin gene-related peptide (CGRP) on motility and on the release of substance P, neurokinin A , somatostatin and gastrin in the isolated perfused porcine antrum. Neurogastroenterol Motil 13:353-359. Ravid K, Smith-Mungo LI, Zhao Z, Thomas K M and Kagan H M (1999) Upregulation of lysyl oxidase in vascular smooth muscle cells by cAMP: role for adenosine receptor activation. J Cell Biochem 75:177-185. Ray JM, Squires PE, Meloche R M , Nelson DW, Snutch TP and Buchan A M (1997) L-type calcium channels regulate gastrin release from human antral G cells. Am J Physiol 273:G281-288. Reeves JJ, Coates J, Jarvis JE, Sheehan M J and Strong P (1993) Characterization of the adenosine receptor mediating contraction in rat colonic muscularis mucosae. Br J Pharmacol 110:1255-1259. Ren H and Stiles G L (1994a) Characterization of the human A l adenosine receptor gene. Evidence for alternative splicing. J Biol Chem 269:3104-3110. Ren H and Stiles G L (1994b) Posttranscriptional mRNA processing as a mechanism for regulation of human A l adenosine receptor expression. Proc Natl Acad Sci USA 91:4864-4866. Ren H and Stiles G L (1995) Separate promoters in the human A l adenosine receptor gene direct the synthesis of distinct messenger RNAs that regulate receptor abundance. Mol Pharmacol 48:975-980. Ren H and Stiles G L (1998) A single-stranded D N A binding site in the human A l adenosine receptor gene promoter. Mol Pharmacol 53:43-51. Ren H and Stiles G L (1999) Dexamethasone stimulates human A l adenosine receptor ( A l AR) gene expression through multiple regulatory sites in promoter B. Mol Pharmacol 55:309-316. Ren J, Young RL, Lassiter DC and Harty RF (1993) Calcitonin gene-related peptide mediates capsaicin-induced neuroendocrine responses in rat antrum. Gastroenterology 104:485-491. Reppert SM, Weaver DR, Stehle JH and Rivkees SA (1991) Molecular cloning and characterization of a rat A1-adenosine receptor that is widely expressed in brain and spinal cord. Mol Endocrinol 5:1037-1048. Ritzel M W , Ng A M , Yao SY, Graham K, Loewen SK, Smith K M , Hyde RJ, Karpinski E, Cass CE, Baldwin SA and Young JD (2001) Recent molecular advances in studies of the concentrative Na+-dependent nucleoside transporter (CNT) family: identification and characterization of novel human and mouse proteins (hCNT3 and mCNT3) broadly selective for purine and pyrimidine nucleosides (system cib). Mol Membr Biol 18:65-72. Rivkees SA, Chen M , Kulkarni J, Browne J and Zhao Z (1999) Characterization of the murine A l adenosine receptor promoter, potent regulation by GATA-4 and Nkx2.5. J Biol Chem 274:14204-14209. Rivkees SA, Price SL and Zhou FC (1995) Immunohistochemical detection of A l adenosine receptors in rat brain with emphasis on localization in the hippocampal formation, cerebral cortex, cerebellum, and basal ganglia. Brain Res 677:193-203. Rivkees SA and Reppert S M (1992) RFL9 encodes an A2b-adenosine receptor. Mol Endocrinol 6:1598-1604. Robinson A J and Dickenson J M (2001) Regulation of p42/p44 M A P K and p38 M A P K by the adenosine A l receptor in DDT1MF-2 cells. Eur J Pharmacol 413:151-161. Rose'Meyer RB, Mellick AS, Garnham BG, Harrison GJ, Massa H M and Griffiths L R (2003) The measurement of adenosine and estrogen receptor expression in rat brains following ovariectomy using quantitative PCR analysis. Brain Res Brain Res Protoc 11:9-18. Sachs G, Zeng N and Prinz C (1997) Physiology of isolated gastric endocrine cells. Ann Rev Physiol 59:243-256. Saffouri B, DuVal JW, Arimura A and Makhlouf G M (1984) Effects of vasoactive intestinal peptide and secretin on gastrin and somatostatin secretion in the perfused rat stomach. Gastroenterology 86:839-842. Saffouri B, Weir GC, Bitar K N and Makhlouf G M (1980) Gastrin and somatostatin secretion by perfused rat stomach: functional linkage of antral peptides. Am J Physiol 238:G495-501. Saitoh O, Saitoh Y and Nakata H (1994) Regulation of A2a adenosine receptor mRNA expression by agonists and forskolin in PC12 cells. Neuroreport 5:1317-1320. Sajjadi FG, Boyle DL, Domingo RC and Firestein GS (1996) cDNA cloning and characterization of A3i , an alternatively spliced rat A3 adenosine receptor variant. FEBS Lett 382:125-129. Sakai Y , McLean J, Grover A K , Garfield RE, Fox JE and Daniel EE (1981) Isolation and characterization of subcellular membranes from canine stomach smooth muscle. Can J Physiol Pharmacol 59:1260-1267. Sala-Newby GB, Skladanowski A C and Newby A C (1999) The mechanism of adenosine formation in cells. Cloning of cytosolic 5'-nucleotidase-I. J Biol Chem 274:17789-17793. Salvatore CA, Jacobson M A , Taylor HE, Linden J and Johnson R G (1993) Molecular cloning and characterization of the human A3 adenosine receptor. Proc Natl Acad Sci U SA 90:10365-10369. Sambrook J, Fritsch EF and Maniatis T (1989a) Extraction, purification, and analysis of messenger of R N A from eukaryotic cells, in Molecular cloning: a laboratory manual (Nolan C ed) pp 7.1-7.87, Cold Spring Harbor Laboratory Press, Plainview. Sambrook J, Fritsch EF and Maniatis T (1989b) Gel electrophoresis of DNA, in Molecular cloning: a laboratory manual (Nolan C ed) pp 6.1-6.62, Cold Spring Harbor Laboratory Press, Plainview. Sandvik A K , Dimaline R, Brenna E and Waldum H L (1995) Differential expression and regulation of SSTR2 messenger RNA in rat gastric antrum and corpus. Am J Physiol 269:G542-547. Sandvik A K , Dimaline R, Forster ER, Evans D and Dockray GJ (1993) Differential control of somatostatin messenger RNA in rat gastric corpus and antrum. Role of acid, food, and capsaicin-sensitive afferent neurons. J Clin Invest 91:244-250. Sanyal A , O'Driscoll SW, Bolander M E and Sarkar G (1997) An effective method of completely removing contaminating genomic D N A from an R N A sample to be used for PCR. Mol Biotechnol 8:135-137. Sawmiller DR and Chou CC (1990) Jejunal adenosine increases during food-induced jejunal hyperemia. Am J Physiol 258:G370-376. Sawynok J and Jhamandas K H (1976) Inhibition of acetylcholine release from cholinergic nerves by adenosine, adenine nucleotides and morphine: antagonism by theophylline. J Pharmacol Exp Ther 197:379-390. Scarpignato C, Tramacere R, Zappia L and Del Soldato P (1987) Inhibition of gastric acid secretion by adenosine receptor stimulation in the rat. Pharmacology 34:264-268. Schaffer K, Herrmuth H, Mueller J, Coy D H , Wong HC, Walsh JH, Classen M , Schusdziarra V and Schepp W (1997) Bombesin-like peptides stimulate somatostatin release from rat fundic D cells in primary culture. Am J Physiol 273:G686-695. Schepp W, Soil A H and Walsh JH (1990) Dual modulation by adenosine of gastrin release from canine G-cells in primary culture. Am J Physiol 259:G556-563. Schicho R, Schemann M , Pabst M A , Holzer P and Lippe IT (2003) Capsaicin-sensitive extrinsic afferents are involved in acid-induced activation of distinct myenteric neurons in the rat stomach. Neurogastroenterol Motil 15:33-44. Schmittgen TD (2001) Real-time quantitative PCR. Methods 25:383-385. Schmittgen TD and Zakrajsek B A (2000) Effect of experimental treatment on housekeeping gene expression: validation by real-time, quantitative RT-PCR. JBiochem Biophys Methods 46:69-81. Schmugge M , Rand M L and Freedman J (2003) Platelets and von Willebrand factor. Transfus Apheresis Sci 28:269-277. Schofield IN, Day IN, Thompson RJ and Edwards Y H (1995) PGP9.5, a ubiquitin C-terminal hydrolase; pattern of mRNA and protein expression during neural development in the mouse. Brain Res Dev Brain Res 85:229-238. Schreiber S, Nguyen TH, Stuben M and Scheid P (2000) Demonstration of a pH gradient in the gastric gland of the acid-secreting guinea pig mucosa. Am J Physiol Gastrointest Liver Physiol 279:G597-604. Schubert M L , Jong M J and Makhlouf G M (1991) Bombesin/GRP-stimulated somatostatin secretion is mediated by gastrin in the antrum and intrinsic neurons in the fundus. Am J Physiol 26LG885-889. Schubert M L and Makhlouf G M (1992) Neural, hormonal, and paracrine regulation of gastrin and acid secretion. Yale J Biol Med 65:553-560. Schubert M L and Shamburek RD (1990) Control of acid secretion. Gastroenterol Clin North Am 19:1-25. Schulte G and Fredholm BB (2000) Human adenosine A( l ) , A(2A), A(2B), and A(3) receptors expressed in Chinese hamster ovary cells all mediate the phosphorylation of extracellular-regulated kinase 1/2. Mol Pharmacol 58:477-482. Schusdziarra V , de la Fuente A , Swobodnik W, Fussganger R and Pfeiffer EF (1983) Somatostatin release from perifused rat and human gastric mucosa. Clin Physiol Biochem 1:12-16. Schwarting H, Koop H, Gellert G and Arnold R (1986) Effect of starvation on endocrine cells in the rat stomach. Regul Pept 14:33-39. Seale TW, Abla K A , Shamim MT, Carney J M and Daly JW (1988) 3,7-Dimethyl-l-propargylxanthine: a potent and selective in vivo antagonist of adenosine analogs. Life 5ci 43:1671-1684. Sebastiao A M and Ribeiro JA (2000) Fine-tuning neuromodulation by adenosine. Trends Pharmacol Sci 21:341-346. Seensalu R, Avedian D, Barbuti R, Song M , Slice L and Walsh JH (1997) Bombesin-induced gastrin release from canine G cells is stimulated by Ca2+ but not by protein kinase C, and is enhanced by disruption of rho/cytoskeletal pathways. J Clin Invest 100:1037-1046. Selemidis S, Satchell D G and Cocks T M (1997) Evidence that NO acts as a redundant N A N C inhibitory neurotransmitter in the guinea-pig isolated taenia coli. Br J Pharmacol 121:604-611. Sexl V , Mancusi G, Holler C, Gloria-Maercker E, Schutz W and Freissmuth M (1997) Stimulation of the mitogen-activated protein kinase via the A2A-adenosine receptor in primary human endothelial cells. J Biol Chem 272:5792-5799. Shinozuka K, Maeda T and Hayashi E (1985) Possibilities for adenosine modulation of peristaltic reflex in guinea pig isolated ileum. JPharmacobiodyn 8:877-884. Shiotani A and Merchant JL (1995) cAMP regulates gastrin gene expression. Am J Physiol 269:G458-464. Shulkes A , Caussignac Y , Lamers CB, Solomon TE, Yamada T and Walsh JH (1983) Starvation in the rat: effect on peptides of the gut and brain. AustJ Exp Biol Med Sci 61 ( Pt 5):581-587. Sinclair CJ, LaRiviere CG, Young JD, Cass CE, Baldwin SA and Parkinson FE (2000) Purine uptake and release in rat C6 glioma cells: nucleoside transport and purine metabolism under ATP-depleting conditions. JNeurochem 75:1528-1538. Souza RF, Shewmake K, Terada LS and Spechler SJ (2002) Acid exposure activates the mitogen-activated protein kinase pathways in Barrett's esophagus. Gastroenterology. 122:299-307. Stehle JH, Rivkees SA, Lee J J, Weaver DR, Deeds JD and Reppert S M (1992) Molecular cloning and expression of the cDNA for a novel A2-adenosine receptor subtype. Mol Endocrinol 6:384-393. Sterling GF and Grossman MI (1970) Gastric acid response to pentagastrin and histamine after extragastric vagotomy in dogs. Gastroenterology 59:364-371. Stone TW and Simmonds H A (1991) Purines: basic and clinical aspects. Kluwer academic publishers, Dordrecht. Storr M , Thammer J, Dunkel R, Schusdziarra V and Allescher HD (2002) Modulatory effect of adenosine receptors on the ascending and descending neural reflex responses of rat ileum. BMC Neurosci 3:21. Su C, Bevan JA and Burnstock G (1971) [3H]adenosine triphosphate: release during stimulation of enteric nerves. Science 173:336-338. Sundaram U , Hassanain H, Suntres Z, Y u JG, Cooke HJ, Guzman J and Christofi F L (2003) Rabbit chronic ileitis leads to up-regulation of adenosine A1/A3 gene products, oxidative stress, and immune modulation. Biochem Pharmacol 65:1529-1538. Suzuki M , Tomaru A , Kishibayashi N and Karasawa A (1995) Effects of the adenosine A l -receptor antagonist on defecation, small intstinal propulsion and gastric emptying in rats. Jpn J Pharmacol 68:119-123. Suzuki T, Higgins PJ and Crawford DR (2000) Control selection for R N A quantitation. Biotechniques 29:332-337. Svenningsson P and Fredholm BB (1997) Glucocorticoids regulate the expression of adenosine A l but not A(2A) receptors in rat brain. J Pharmacol Exp Ther 280:1094-1101. Tabrizchi R and Bedi S (2001) Pharmacology of adenosine receptors in the vasculature. Pharmacol Ther 91:133-147. Takehara Y , Sumii K, Tari A , Yoshihara M , Sumii M , Haruma K , Kajiyama G, Wu SV and Walsh JH (1996) Evidence that endogenous GRP in rat stomach mediates omeprazole-induced hypergastrinemia. Am J Physiol 27LG799-G804. Tally KJ , Hrnjez BJ, Smith JA, Mun EC and Matthews JB (1996) Adenosine scavenging: a novel mechanism of chloride secretory control in intestinal epithelial cells. Surgery 120:248-254. Tang L, Parker M , Fei Q and Loutzenhiser R (1999) Afferent arteriolar adenosine A2a receptors are coupled to K A T P in in vitro perfused hydronephrotic rat kidney. Am J Physiol 277:F926-933. Tari A , Wu V, Sumii M , Sachs G and Walsh JH (1991) Regulation of rat gastric H+/K(+)-ATPase alpha-subunit mRNA by omeprazole. Biochim Biophys Acta 1129:49-56. Thorn JA and Jarvis S M (1996) Adenosine transporters. Gen Pharmacol 27:613-620. Townsend-Nicholson A, Baker E, Schofield PR and Sutherland GR (1995) Localization of the adenosine A l receptor subtype gene (ADORA1) to chromosome lq32.1. Genomics 26:423-425. Track NS, Creutzfeldt C, Arnold R and Creutzfeldt W (1978) The antral gastrin-producing G-cell: biochemical and ultrastructural responses to feeding. Cell Tiss Res 194:131-139. Traversa U , Rosati A M , Florio C and Vertua R (1994) Effects of chronic administration of adenosine antagonists on adenosine A l and A2a receptors in mouse brain. In Vivo 8:1073-1078. Ushijima I, Mizuki Y , Hara T, Obara N , Minematsu N and Yamada M (1992) Effects of lithium and purinergic compounds on the behavioral and physiological aspects of restraint stress in rats. Pharmacol Biochem Behav 42:431-435. Ushijima I, Mizuki Y and Yamada M (1985) Development of stress-induced gastric lesions involves central adenosine A1-receptor stimulation. Brain Res 339:351-355. Vallejo AI, Bo X and Burnstock G (1996) P2Y purinoceptors in gastric gland plasma membranes. Eur J Pharmacol 312:209-214. van Calker D, Muller M and Hamprecht B (1979) Adenosine regulates via two different types of receptors, the accumulation of cyclic A M P in cultured brain cells. J Neurochem 33:999-1005. van Galen PJ, van Bergen A H , Gallo-Rodriguez C, Melman N , Olah M E , AP IJ, Stiles G L and Jacobson K A (1994) A binding site model and structure-activity relationships for the rat A3 adenosine receptor. Mol Pharmacol 45:1101-1111. Vincent SR, Mcintosh C H , Buchan A M and Brown JC (1985) Central somatostatin systems revealed with monoclonal antibodies. J Comp Neurol 238:169-186. Vinik AI, Gaginella TS, O'Dorisio T M , Shapiro B and Wagner L (1981) The distribution and characterization of somatostatin-like immunoreactivity in epithelial cells, submucosa, and muscle of the rat stomach and intestine. Endocrinology 109:1921-1926. Viz i ES, Sperlagh B and Baranyi M (1992) Evidence that ATP released from the postsynaptic site by noradrenaline, is involved in mechanical responses of guinea-pig vas deferens: cascade transmission. Neuroscience 50:455-465. Wall RT, Counts RB, Harker L A and Striker GE (1980) Binding and release of factor VHI/von Willebrand's factor by human endothelial cells. Br J Haematol 46:287-298. Wang A M , Doyle M V and Mark DF (1989) Quantitation of mRNA by the polymerase chain reaction. Proc Natl Acad Sci USA 86:9717-9721. Westerberg VS and Geiger JD (1987) Central effects of adenosine analogs on stress-induced gastric ulcer formation. Life Sci 41:2201-2205. Westerberg VS and Geiger JD (1989) Adenosine analogs inhibit gastric acid secretion. Eur J Pharmacol 160:275-281. Wiklund NP and Gustafsson L E (1987) On the nature of endogenous purines modulating cholinergic neurotransmission in the guinea-pig ileum. Acta Physiol Scand 131:11-18. Wilson PO, Barber PC, Hamid QA, Power BF, Dhillon AP, Rode J, Day IN, Thompson RJ and Polak J M (1988) The immunolocalization of protein gene product 9.5 using rabbit polyclonal and mouse monoclonal antibodies. Br J Exp Pathol 69:91-104. Witte DP, Wiginton DA, Hutton J J and Aronow BJ (1991) Coordinate developmental regulation of purine catabolic enzyme expression in gastrointestinal and postimplantation reproductive tracts. J Cell Biol 115:179-190. Worku Y and Newby A C (1983) The mechanism of adenosine production in rat polymorphonuclear leucocytes. Biochem 7214:325-330. Worpenberg S, Burk O and Klempnauer K H (1997) The chicken adenosine receptor 2B gene is regulated by v-myb. Oncogene 15:213-221. Wu SV, Giraud A , Mogard M , Sumii K and Walsh JH (1990a) Effects of inhibition of gastric secretion on antral gastrin and somatostatin gene expression in rats. Am J Physiol 258.-G788-793. Wu SV, Sumii K, Tari A , Mogard M and Walsh JH (1990b) Regulation of gastric somatostatin gene expression. Metab Clin Exp 39:125-130. Wu V , Sumii K, Tari A , Sumii M and Walsh JH (1991) Regulation of rat antral gastrin and somatostatin gene expression during starvation and after refeeding. Gastroenterology 101:1552-1558. Xaus J, Mirabet M , Lloberas J, Soler C, Lluis C, Franco R and Celada A (1999) IFN-gamma up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J Immunol 162:3607-3614. Xu PA and Kellems RE (2000) Function of murine adenosine deaminase in the gastrointestinal tract. Biochem Biophys Res Commun 269:749-757'. Xue L and Lucocq J M (1997) Low extracellular pH induces activation of E R K 2, JNK, and p38 in A431 and Swiss 3T3 cells. Biochem Biophys Res Commun 241:236-242. Yaar R, Cataldo L M , Tzatsos A , Francis CE, Zhao Z and Ravid K (2002) Regulation of the A3 adenosine receptor gene in vascular smooth muscle cells: role of a cAMP and G A T A element. Mol Pharmacol 62:1167-1176. Yakel JL, Warren RA, Reppert S M and North R A (1993) Functional expression of adenosine A2b receptor in Xenopus oocytes. Mol Pharmacol 43:277-280. Yamada Ff, Chen D, Monstein HJ and Hakanson R (1997a) Effects of fasting on the expression of gastrin, cholecystokinin, and somatostatin genes and of various housekeeping genes in the pancreas and upper digestive tract of rats. Biochem Biophys Res Commun 231:835-838. Yamada M , Ikeuchi T and Hatanaka H (1997b) The neurotrophic action and signalling of epidermal growth factor. Prog Neurobiol 51:19-37. Yamamura T, Kimura T and Furukawa K (1983) Effects of halothane, thiamylal, and ketamine on central sympathetic and vagal tone. Anesth Analg 62:129-134. Yip L and Kwok Y N (2004) Role of adenosine A 2 A receptor in the regulation of gastric somatostatin release. J Pharmacol Exp Ther (in press). Yip L, Kwok Y N and Buchan A M (2003) Cellular localization and distribution of neurokinin-1 receptors in the rat stomach. Auton Neurosci 104:95-108. Yonei Y and Guth PH (1991) Ethanol-induced gastric injury. Role of submucosal venoconstriction and leukotrienes. DigDis Sci 36:601-608. Zafirov D H , Palmer J M and Wood JD (1985) Adenosine inhibits forskolin-induced excitation in myenteric neurons. Eur J Pharmacol 113:143-144. Zaki M , Harrington L, McCuen R, Coy DH, Arimura A and Schubert M L (1996) Somatostatin receptor subtype 2 mediates inhibition of gastrin and histamine secretion from human, dog, and rat antrum. Gastroenterology 111 :919-924. Zhao Z, Francis C and Ravid K (1999) Characterization of the mouse A3 adenosine receptor gene: exon/intron organization and promoter activity. Genomics 57:152-155. Zhou QY, L i C, Olah M E , Johnson RA, Stiles GL and Civelli O (1992) Molecular cloning and characterization of an adenosine receptor: the A3 adenosine receptor. Proc Natl Acad Sci USA 89:7432-7436. Zimmermann H (2000) Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedebergs Arch Pharmacol 362:299-309. Zucchi R, Y u G, Ghelardoni S, Ronca F and Ronca-Testoni S (2001) A3 adenosine receptor stimulation modulates sarcoplasmic reticulum Ca(2+) release in rat heart. Cardiovasc Res 50:56-64. Appendix 1: Table of abbreviations 2-CA 2-chloroadenosine 8-PT 8-phenyltheophylline 8-SPT 8-(p-sulfophenyl)theophyline A,R-IR A, receptor immunoreactivity A 2 AR-IR A 2 A receptor immunoreactivity ADA adenosine deaminase ADP adenosine diphosphate AK adenosine kinase AMP adenosine monophosphate ATP adenosine triphosphate BSA bovine serum albumin cAMP cyclic AMP CCK cholecystokinin CGRP calcitonin gene related peptide CGS 15943 9-chloro-2(2-furyl)[l,2, 4]triazolo [ 1,5-c]quinazolin-5-amine CGS 21680 2-p-(2-carboxyethyl)phenethylamino-5 'N-ethylcarboxamidoadenosine CHA 5'-N -cyclohexyladenosine CPA N6-cyclopentyladenosine CRE cAMP response element CREB CRE binding protein C T threshold cycle CV1808 2-phenylaminoadenosine DEPC diethlypyrocarbonate DMNV dorsomotor nucleus of the vagus DMPX 3,7-dimethyl-1 -propargylxanthine DMSO dimethylsulfoxide DPCPX 8-cyclopentyl-l,3-dipropylxanthine ECL enterochro maffin-like EGF epidermal growth factor EHNA erythro-9-(2-hydroxy-3 -nonyl)adenine hydrochloride E-NPP ecto-nucleotide pyrophosphatase E-NTPase ecto-nucleoside triphosphate diphoshohydrolase EPSP excitatory postsynaptic potential FAM 6-carboxyfluorescein FL full length GAPDH glyceraldehyde-3 '-phosphate dehydrogenase GRP gatrin releasing peptide i.c.v. intracerebroventricular i.d. intraduodenal i.p. intraperitoneal i.v. intravenous IB-MECA l-deoxy-l-[6-[[(3-iodophenyl)methyl] amino] -9H-purin-9-yl]-N-methyl-B-D-ribofuranuronamide IHC immunohistochemistry IR immunoreactivity IRG immunoreactive gastrin IUPHAR International Union of Pharmacology LB Luria-Bertani MAPK mitogen activated protein kinases NAD + nicotinamide adenine dinucleotide NECA N-ethylcarboxamidoadenosine N F K B nuclear factor KB NSB non-specific binding NTS nucleus tractus solitarius PCR polymerase chain reaction PGP protein gene product PIA Nb-(2-phenylisopropyl)adenosine PKC protein kinase C PLC phospholipase C RIA radioimmunoassay Rn reporter emissions R-PIA R(-)-N6-(2-phenylisopropyl)adenosine RT-PCR reverse-transcriptase PCR s.c. subcutaneous SAH S-adenosyl homocysteine SAHH S-adenosyl homocysteine hydrolase S-adenosyl methionine SL short length SLI somatostatin-like immunoreactivity S-PIA (S)-N6-(2-phenylisopropyl) adenosine TAMRA 6-carboxytetramethylrhodamine TC total count Tm melting temperature UTR untranslated region VIP vasoactive intestinal peptide VWF von Willebrand's factor ZM 241385 4-(2-[7-Amino-2 (2furyl)[l,2,4]triazolo [2,3-a][l,3,5]triazin-5ylamino]ethyl)phenol "@en . "Thesis/Dissertation"@en . "10.14288/1.0092880"@en . "eng"@en . "Physiology"@en . "Vancouver : University of British Columbia Library"@en . "University of British Columbia"@en . "For non-commercial purposes only, such as research, private study and education. Additional conditions apply, see Terms of Use https://open.library.ubc.ca/terms_of_use."@en . "Graduate"@en . "Adenosine A\u00E2\u0082\u0081 and A\u00E2\u0082\u0082\u00E2\u0082\u0084 receptors in the rat stomach : biological actions, cellular localization, structure and gene expression"@en . "Text"@en . "http://hdl.handle.net/2429/18326"@en .