Your activity: 60 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Physiology of gastrin

Physiology of gastrin
Author:
Rodger A Liddle, MD
Section Editor:
J Thomas Lamont, MD
Deputy Editor:
Shilpa Grover, MD, MPH, AGAF
Literature review current through: Dec 2022. | This topic last updated: Jun 24, 2021.

INTRODUCTION — Gastrin is the major hormonal regulator of gastric acid secretion [1]. Its discovery at the turn of the century was based upon its profound effect on meal-stimulated acid secretion, making it one of the first hormones to be described [1]. The study of gastrin accelerated with the isolation and characterization of the peptide in 1964, after which it was found to promote growth of the gastric antrum and have a proliferative effect [2,3]. The cloning and characterization of the gastrin receptor has provided a valuable tool in the study of gastrointestinal hormones [4].

This topic will review the physiology of gastrin. Zollinger-Ellison syndrome and the physiology of gastric acid secretion are discussed in detail elsewhere. (See "Zollinger-Ellison syndrome (gastrinoma): Clinical manifestations and diagnosis" and "Management and prognosis of the Zollinger-Ellison syndrome (gastrinoma)" and "Physiology of gastric acid secretion".)

GASTRIN PHYSIOLOGY

Molecular forms — Human gastrin is the product of a single gene located on chromosome 17. The active hormone is generated from a precursor peptide preprogastrin (figure 1). Human preprogastrin contains 101 amino acids (AA) including a signal peptide (21 AA), spacer sequence (37 AA), gastrin component (34 AA), and a 9 AA extension segment at the carboxyl terminus. The enzymatic processing of preprogastrin produces all of the known physiologically active forms of gastrin.

Preprogastrin is processed into progastrin and gastrin peptide fragments of various sizes by sequential enzymatic cleavage (figure 1). Like other hormones, gastrin is synthesized on rough endoplasmic reticulum, processed in the Golgi apparatus, and packaged in secretory granules, where final modifications occur [5]. In endocrine cells, the glycine residue at the carboxyl terminus is cleaved and the terminus is amidated to form the mature gastrin peptide.

Two major forms of gastrin are secreted (G-34 and G-17), although larger G-71 and smaller G-6 forms exist (table 1). The common feature of all gastrins is an amidated tetrapeptide (Try-Met-Asp-Phe-NH2) at the carboxyl terminus that imparts full biological activity. Modification by sulfation at tyrosine residues produces alternative gastrin forms. The circulating half-life of gastrin is affected by the size of the various molecular forms. The full physiologic response is determined by the presence of the biologically active tetrapeptide and the time available for receptor interaction. (See "Overview of gastrointestinal peptides in health and disease".)

Gastrin can be ectopically expressed in non-endocrine cells, such as tumors. However, because these cells lack secretory vesicles, prohormone convertases, and amidating enzymes, the main forms of gastrin secreted are progastrins including glycine-extended gastrin. These forms of gastrin may have growth-promoting effects on certain tumors. (See 'Unclear clinical implications of hypergastrinemia' below.)

Tissue distribution — The vast majority of gastrin is produced in gastrin-expressing cells (G cells) of the gastric antrum [6]. Much smaller amounts of gastrin are produced in other regions of the gastrointestinal tract including the nonantral stomach, duodenum, jejunum, ileum, and pancreas. Gastrin has also been found outside of the gastrointestinal tract including the brain, adrenal glands, respiratory tract, and reproductive organs, although its biological role in these sites is unknown (table 2).

The G cells are tightly regulated by two counterbalancing hormones, gastrin-releasing peptide and somatostatin, which exert stimulatory and inhibitory effects, respectively.

Gastrin receptors — The receptors for gastrin and cholecystokinin (CCK) are related and constitute the CCK/gastrin receptor family. The CCK1 receptor is abundant in the pancreas and gallbladder and has a 1000-fold higher affinity for CCK than for gastrin. The CCK2/gastrin receptor is found on parietal cells, on enterochromaffin-like (ECL) cells in the stomach, in the brain, and on pancreatic islet cells.  

Gastrin acts via activation of the CCK2 receptor (formerly known as the CCK-B or gastrin receptor), which has equal affinity for CCK and gastrin, which have an identical pentapeptide terminal sequence [4].

Gastrin "receptors" have also been found on somatostatin-secreting D cells. However, this receptor is a CCK1 that has much greater affinity for CCK than for gastrin [7]. This difference in receptor affinity may explain why gastrin is so much more effective as a stimulant of acid secretion, while CCK induces greater release of the inhibitor somatostatin [8]. Knockout mice that have been genetically engineered to be deficient in CCK2 receptors have low acid secretion, while double knockout mice (deficient in both gastrin and CCK receptors) have robust acid secretion in response to vagal stimulation or exogenous histamine [9]. These findings are consistent with the conclusion that CCK1 receptors exert inhibitory effects on acid secretion in vivo, mediated by release of endogenous somatostatin.

The localization of the receptors responsible for trophic actions of gastrin remains uncertain; the primary receptor appears to be on the ECL cell, although receptors may also be present on gastric stem cells.

Biologic actions

Gastric acid secretion — Gastrin is a primary physiologic mediator of gastric acid secretion and is the major endocrine regulator of the secretory response to a protein meal. Gastric acid secretion is influenced by the central, peripheral, and enteric nervous systems and multiple chemical messengers including gastrin, histamine, somatostatin, and acetylcholine.  

The major cellular determinants of acid secretion involve the antral gastrin-secreting G cell, the ECL cell of the stomach that secretes histamine, and the somatostatin-secreting D cell. The acid-secreting parietal cell possesses receptors for acetylcholine, histamine, and gastrin. Although activation of all three of these receptors can stimulate acid secretion, the most important mechanism of acid release occurs via stimulation of the ECL cell to secrete histamine, which in turn stimulates the parietal cell [10,11]. The ECL cell receives stimulatory signals from gastrin and inhibitory signals from somatostatin (figure 2). Somatostatin also provides inhibitory signals to antral G cells and, therefore, inhibits gastric acid secretion.

Gastrin enhances gastric acid secretion from parietal cells primarily by stimulating the synthesis and release of histamine from oxyntic mucosal ECL cells [12-15]. Gastrin also has direct actions on parietal cells [16,17]. Complex mechanisms control gastrin release from the antral G cells.

Gastric distension impacts gastrin secretion, and the effect varies with the degree of distension [18]. Low-grade distention activates vasoactive intestinal peptide neurons, which stimulates somatostatin release and, therefore, inhibits gastrin secretion. Higher grade distention causes cholinergic activation, which reverses the pattern to one of increased gastrin and reduced somatostatin secretion.

Dietary amino acids induce gastrin release; direct actions on the G cell have been demonstrated, but amino acids also activate both cholinergic neurons and bombesin neurons [19]. The release of bombesin (also called gastrin-releasing peptide) from mucosal nerves directly stimulates the G cell [20-22].

Gastrin contributes to the process of inhibiting its own secretion by enhancing the secretion of somatostatin [19].

Cholinergic activation after gastric distention or in response to a meal promotes acid secretion by shifting the balance of stimulatory and inhibitory mechanisms toward the stimulatory side, directly activating the parietal cell and stimulating gastrin release while suppressing somatostatin release [18,19,23].

Other biologic effects — Gastrin is also the best identified trophic regulator of parietal cell mass in humans. This relationship is evidenced by the presence of gastric hypertrophy in gastrinoma patients who have chronic exposure to elevated gastrin levels (picture 1), and atrophy of the parietal cell mass with antrectomy, which decreases gastrin levels.

The CCK2 receptor can be found on pancreatic islet cells, and hypergastrinemia has been associated with islet cell hyperplasia and enhanced insulin secretion [24]. In animals, coadministration of gastrin and glucagon-like peptide-1 can restore normoglycemia in diabetic mice, suggesting that gastrin may have incretin-like effects [24]. Gastrin acting on CCK2 receptors in the proximal tubule exerts natriuretic effects [25]. (See "Glucagon-like peptide 1-based therapies for the treatment of type 2 diabetes mellitus", section on 'Gastrointestinal peptides'.)

Regulation of gastrin — Gastrin release is profoundly influenced by the pH of the stomach. Fasting and increased gastric acid in the stomach inhibit its release, whereas a high gastric pH provides a strong stimulus for its secretion. The G cells are tightly regulated by two counterbalancing hormones, gastrin-releasing peptide and somatostatin, which exert stimulatory and inhibitory effects, respectively.

Physiological regulation of gastrin secretion is highest when nutrients are in direct contact with G cells. Consequently, diversion of food from the antrum of the stomach, as with Roux-en-Y gastric bypass, has been associated with low blood levels of gastrin [26].

HYPERGASTRINEMIA

Causes of hypergastrinemia — Hypergastrinemia is observed in a number of settings, of which use of acid-suppressive medications and chronic atrophic gastritis are by far the most common (table 3). The mechanism of hypergastrinemia in these conditions is related to the stimulation of gastrin production by the alkaline pH environment, explaining the increased plasma gastrin levels often associated with Helicobacter pylori infection [27].

Another important, but far less common, association is the Zollinger-Ellison syndrome, which is caused by a gastrin-producing tumor (picture 1). Most patients with Zollinger-Ellison syndrome have serum gastrin concentrations between 150 and 1000 pg/mL (71 and 475 pmol/L). This degree of gastrin elevation can also be observed in a variety of other settings, which need to be considered when evaluating a patient found to have hypergastrinemia (table 3) [28-30]. (See "Zollinger-Ellison syndrome (gastrinoma): Clinical manifestations and diagnosis".)

The highest gastrin levels (>1000 pg/mL), although rare, are seen with gastrin-secreting tumors or chronic atrophic gastritis accompanying pernicious anemia. Modest elevations in serum gastrin (approximately 200 to 400 pg/mL) are much more common and are often due to acid-suppressive medications (ie, H2 blockers or proton pump inhibitors [PPIs]) or H. pylori-associated gastritis.

Elevated gastrin levels are often seen following small bowel resection due to the loss of enterogastrones (hormones such as secretin that tend to inhibit gastric acid secretion). With loss of enterogastrones, there is an increase in gastric acid secretion and gastric pH is low. 

Other rare causes of achlorhydria-associated hypergastrinemia may occur in the absence of gastritis. For example, impaired potassium secretion into the gastric lumen can reduce hydrogen-potassium proton pump activity on the apical surface of parietal cells, resulting in low acid secretion [31]. Patients with Jervell and Lange-Nielsen syndrome caused by mutations in the potassium channel gene KCNQ1 not only have cardiac arrhythmias due to QT prolongation but have gastric achlorhydria, iron deficiency anemia, and hypergastrinemia as a consequence of impaired potassium ion secretion [32].

Unclear clinical implications of hypergastrinemia — The clinical significance of chronic elevation in serum gastrin levels (as may be seen in patients taking PPIs long-term) is uncertain. Gastrin receptors have been detected on a variety of tumor cells, and gastrin has trophic effects on the stomach and intestine, suggesting that it may have a role in the development of gastrointestinal malignancies [33,34]. The degree to which this might contribute to oncogenesis has not been defined.

Fundic gland polyps — Long-term treatment with PPIs has been associated with the development of gastric fundic polyps and hyperplasia of the oxyntic mucosa. The clinical significance of these observations is uncertain, and their pathogenesis is incompletely understood [35]. Infection with H. pylori may have a role, since gastrin has a growth-promoting effect on the organism [36]. (See "Gastric polyps", section on 'Fundic gland polyps'.)

Gastric carcinoid tumors — Although prolonged hypergastrinemia from potent antisecretory therapy has been associated with the development of enterochromaffin-like (ECL) cell carcinoid tumors in rodents, the incidence of gastric carcinoid tumors is not increased in adults exposed to acid suppression for prolonged periods of time [37-39]. (See "Proton pump inhibitors: Overview of use and adverse effects in the treatment of acid related disorders", section on 'Hypergastrinemia'.)

Studies in patients with gastrinoma and pernicious anemia who develop carcinoids suggest that carcinoid tumor formation requires not only hypergastrinemia but another cofactor, such as loss of the tumor suppressor gene, menin (the genetic defect in multiple endocrine neoplasia type 1 [MEN-1]), or overexpression of a tumor growth promoter such as BCL-2, which occurs in the setting of chronic inflammation and atrophic gastritis [40]. This hypothesis is supported by observation that carcinoid tumors have been reported in up to 30 percent of patients with MEN-1 and 5 percent of patients with atrophic gastritis and pernicious anemia. However, carcinoid tumors occur in only 1 percent of patients with sporadic gastrinomas [41,42]. The role of gastrin appears to be central in the development of ECL carcinoids associated with atrophic gastritis since the gastrin receptor antagonist netazepide has been shown to induce regression of these carcinoids [15].

Colon cancer — It remains controversial whether circulating gastrin has a significant role in the development of human colon cancer. Gastrin has been implicated in colon cancer based upon laboratory and epidemiologic evidence [43-47]. However, an increased rate of colon cancer has not been observed in patients with gastrinoma, pernicious anemia, or in those taking acid-suppressive medications [48]. It is likely that chronic hypergastrinemia by itself is not carcinogenic, but its proliferative effects may expand the pool of cells that could be at risk for carcinogenesis in susceptible patients. One group of patients at risk may be those infected with H. pylori [45,49].

The following observations illustrate some of the laboratory findings:

Gastrin increases the proliferation of colon cancer cell lines and the tumor burden in mice with colon cancer [50].

Gastrin and glycine-extended gastrin have growth-promoting effects on human colon cancer in vivo and in vitro [51].

A specific inhibitor of glycine-extended gastrin inhibited the growth of human colon cancer, suggesting that receptor blockade may be a strategy for treating patients with colon cancer [52].

Progastrin, glycine-extended gastrin, and amidated gastrin have been detected in cell extracts of gastrointestinal malignancies, suggesting that gastrin may be an autocrine or paracrine growth regulator [53-55].

Immunostaining reveals that 80 to 90 percent of colon polyps contain gastrin [56].

Gastrin inhibits apoptosis [57] and enhances angiogenesis [58].

Pancreatic cancer — A role for gastrin in the proliferation of pancreatic cancer cells is supported by the observation that growth of cultured human pancreatic cancer cells can be inhibited by antisense oligonucleotides to gastrin [59]. CCK2/gastrin receptors have been identified on human pancreatic cancer cells, where they can potentiate cancer growth [34]. This effect is believed to be autocrine or paracrine since gastrin and its messenger RNA have been identified in cancers of the human pancreas [60]. Gastrin has also been implicated in a mouse model of pancreatic cancer metastasis [61].

CLINICAL APPLICATIONS OF GASTRIN

Pentagastrin – The gastrin analog, pentagastrin, has been used clinically to stimulate histamine and gastric acid secretion in diagnostic tests of acid secretory capacity [62]. This test has been used to assess the extent of surgical vagotomy. Pentagastrin has been used to screen for thyroid C-cell hyperplasia and medullary carcinomas [63,64]. (See "VIPoma: Clinical manifestations, diagnosis, and management" and "Medullary thyroid cancer: Clinical manifestations, diagnosis, and staging", section on 'Evaluation'.)

Gastrin receptor antagonists – Gastrin receptor antagonists have the theoretical advantage of blocking gastrin-induced gastric acid secretion, while obviating the concern over the trophic effects related to hypergastrinemia. In addition, they could have a role in the treatment and prevention of gastrointestinal malignancies that express gastrin receptors. In one study, a gastrin receptor antagonist reduced tumor size in patients with neuroendocrine tumors associated with autoimmune atrophic gastritis [65]. (See 'Hypergastrinemia' above.)  

SUMMARY

Human gastrin is the product of a single gene located on chromosome 17. The active hormone is generated from a precursor peptide "preprogastrin" (figure 1). (See 'Molecular forms' above.)

The vast majority of gastrin is produced in endocrine cells of the gastric antrum (G cells). Much smaller amounts of gastrin are produced in other regions of the gastrointestinal tract including the nonantral stomach, duodenum, jejunum, ileum, and pancreas. Gastrin has also been found outside of the gastrointestinal tract including the brain, adrenal glands, respiratory tract, and reproductive organs, although its biological role in these sites is unknown (table 2). (See 'Tissue distribution' above.)

The receptors for gastrin and cholecystokinin (CCK) are related and constitute the CCK/gastrin receptor family. The CCK1 receptor is abundant in the pancreas and gallbladder and has a 1000-fold higher affinity for CCK than for gastrin. The CCK2/gastrin receptor is found on parietal cells, on enterochromaffin-like (ECL) cells in the stomach, in the brain, and on pancreatic islet cells.

Gastrin is released from G cells into the circulation in response to a meal or to a high gastric pH. The G cells are tightly regulated by two counterbalancing hormones, gastrin-releasing peptide and somatostatin, which exert stimulatory and inhibitory effects, respectively. (See 'Gastrin physiology' above.)

Gastrin is a primary physiologic mediator of gastric acid secretion. Gastrin stimulates ECL cells, which then secrete histamine, which in turn stimulates parietal cells to secrete acid (figure 2). (See 'Gastrin physiology' above.)

The clinical significance of chronic elevation in serum gastrin levels (as may be seen in patients taking proton pump inhibitors long-term) is uncertain. Gastrin receptors have been detected on a variety of tumor cells, and gastrin has trophic effects on the stomach and intestine, suggesting that it may have a role in the development of gastrointestinal malignancies. However, the degree to which this might contribute to oncogenesis has not been defined. (See 'Unclear clinical implications of hypergastrinemia' above.)

The gastrin analog, pentagastrin, has been used clinically to stimulate histamine and gastric acid secretion in diagnostic tests of acid secretory capacity. This test has been used to assess the extent of surgical vagotomy and to screen for thyroid C-cell hyperplasia and medullary carcinomas. (See "VIPoma: Clinical manifestations, diagnosis, and management" and "Medullary thyroid cancer: Clinical manifestations, diagnosis, and staging", section on 'Evaluation'.)

  1. Edkins JS. On the chemical mechanism of gastric secretion. Proc R Soc Lond B Biol Sci 1905; 76:376.
  2. GREGORY RA, TRACY HJ. THE CONSTITUTION AND PROPERTIES OF TWO GASTRINS EXTRACTED FROM HOG ANTRAL MUCOSA. Gut 1964; 5:103.
  3. Joshi SN, Gardner JD. Gastrin and colon cancer: a unifying hypothesis. Dig Dis 1996; 14:334.
  4. Kopin AS, Lee YM, McBride EW, et al. Expression cloning and characterization of the canine parietal cell gastrin receptor. Proc Natl Acad Sci U S A 1992; 89:3605.
  5. Rehfeld JF. Processing of precursors of gastroenteropancreatic hormones: diagnostic significance. J Mol Med (Berl) 1998; 76:338.
  6. Rehfeld JF. The new biology of gastrointestinal hormones. Physiol Rev 1998; 78:1087.
  7. Soll AH, Amirian DA, Park J, et al. Cholecystokinin potently releases somatostatin from canine fundic mucosal cells in short-term culture. Am J Physiol 1985; 248:G569.
  8. Schmidt WE, Schmitz F. Cellular localization of cholecystokinin receptors as the molecular basis of the periperal regulation of acid secretion. Pharmacol Toxicol 2002; 91:351.
  9. Chen D, Friis-Hansen L, Håkanson R, Zhao CM. Genetic dissection of the signaling pathways that control gastric acid secretion. Inflammopharmacology 2005; 13:201.
  10. Hersey SJ, Sachs G. Gastric acid secretion. Physiol Rev 1995; 75:155.
  11. Sachs G, Prinz P. Gastric enterochromaffin-like cells and the regulation of acid secretion. News in Physiological Sci 1996; 11:57.
  12. Sandvik AK, Waldum HL. Aspects of the regulation of gastric histamine release. Scand J Gastroenterol Suppl 1991; 180:108.
  13. Kidd M, Modlin IM, Tang LH. Gastrin and the enterochromaffin-like cell: an acid update. Dig Surg 1998; 15:209.
  14. Chen D, Monstein HJ, Nylander AG, et al. Acute responses of rat stomach enterochromaffinlike cells to gastrin: secretory activation and adaptation. Gastroenterology 1994; 107:18.
  15. Waldum HL, Hauso Ø, Fossmark R. The regulation of gastric acid secretion - clinical perspectives. Acta Physiol (Oxf) 2014; 210:239.
  16. Soll AH, Amirian DA, Thomas LP, et al. Gastrin receptors on isolated canine parietal cells. J Clin Invest 1984; 73:1434.
  17. Bitziou E, Patel BA. Simultaneous detection of gastric acid and histamine release to unravel the regulation of acid secretion from the guinea pig stomach. Am J Physiol Gastrointest Liver Physiol 2012; 303:G396.
  18. Schubert ML, Makhlouf GM. Gastrin secretion induced by distention is mediated by gastric cholinergic and vasoactive intestinal peptide neurons in rats. Gastroenterology 1993; 104:834.
  19. Schubert ML, Makhlouf GM. Neural, hormonal, and paracrine regulation of gastrin and acid secretion. Yale J Biol Med 1992; 65:553.
  20. Sugano K, Park J, Soll AH, Yamada T. Stimulation of gastrin release by bombesin and canine gastrin-releasing peptides. Studies with isolated canine G cells in primary culture. J Clin Invest 1987; 79:935.
  21. Campos RV, Buchan AM, Meloche RM, et al. Gastrin secretion from human antral G cells in culture. Gastroenterology 1990; 99:36.
  22. Seensalu R, Avedian D, Barbuti R, et al. 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 1997; 100:1037.
  23. Schubert ML, Peura DA. Control of gastric acid secretion in health and disease. Gastroenterology 2008; 134:1842.
  24. Rehfeld JF. Incretin physiology beyond glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide: cholecystokinin and gastrin peptides. Acta Physiol (Oxf) 2011; 201:405.
  25. Chen Y, Asico LD, Zheng S, et al. Gastrin and D1 dopamine receptor interact to induce natriuresis and diuresis. Hypertension 2013; 62:927.
  26. Meek CL, Lewis HB, Reimann F, et al. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides 2016; 77:28.
  27. Calam J, Gibbons A, Healey ZV, et al. How does Helicobacter pylori cause mucosal damage? Its effect on acid and gastrin physiology. Gastroenterology 1997; 113:S43.
  28. Okosdinossian ET, Munshid HA, Wasfi AI, et al. Fasting plasma-gastrin in vitiligo. Lancet 1978; 1:997.
  29. Netter P, Faure G, Brassine A, et al. Hypergastrinemia in rheumatoid arthritis is related to Sjögren's syndrome. J Rheumatol 1985; 12:651.
  30. Korman MG, Laver MC, Hansky J. Hypergastrinaemia in chronic renal failure. Br Med J 1972; 1:209.
  31. Song P, Groos S, Riederer B, et al. KCNQ1 is the luminal K+ recycling channel during stimulation of gastric acid secretion. J Physiol 2009; 587:3955.
  32. Winbo A, Sandström O, Palmqvist R, Rydberg A. Iron-deficiency anaemia, gastric hyperplasia, and elevated gastrin levels due to potassium channel dysfunction in the Jervell and Lange-Nielsen Syndrome. Cardiol Young 2013; 23:325.
  33. Clerc P, Dufresne M, Saillan C, et al. Differential expression of the CCK-A and CCK-B/gastrin receptor genes in human cancers of the esophagus, stomach and colon. Int J Cancer 1997; 72:931.
  34. de Weerth A, von Schrenck T, Löhr M, et al. Human pancreatic cancer cell lines express the CCKB receptor. Hepatogastroenterology 1999; 46:472.
  35. el-Zimaity HM, Jackson FW, Graham DY. Fundic gland polyps developing during omeprazole therapy. Am J Gastroenterol 1997; 92:1858.
  36. Chowers MY, Keller N, Tal R, et al. Human gastrin: a Helicobacter pylori--specific growth factor. Gastroenterology 1999; 117:1113.
  37. Jensen RT. Involvement of cholecystokinin/gastrin-related peptides and their receptors in clinical gastrointestinal disorders. Pharmacol Toxicol 2002; 91:333.
  38. Havu N. Enterochromaffin-like cell carcinoids of gastric mucosa in rats after life-long inhibition of gastric secretion. Digestion 1986; 35 Suppl 1:42.
  39. McCarthy DM. Proton Pump Inhibitor Use, Hypergastrinemia, and Gastric Carcinoids-What Is the Relationship? Int J Mol Sci 2020; 21.
  40. Cadiot G, Laurent-Puig P, Thuille B, et al. Is the multiple endocrine neoplasia type 1 gene a suppressor for fundic argyrophil tumors in the Zollinger-Ellison syndrome? Gastroenterology 1993; 105:579.
  41. Bordi C, D'Adda T, Azzoni C, et al. Hypergastrinemia and gastric enterochromaffin-like cells. Am J Surg Pathol 1995; 19 Suppl 1:S8.
  42. Gibril F, Schumann M, Pace A, Jensen RT. Multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome: a prospective study of 107 cases and comparison with 1009 cases from the literature. Medicine (Baltimore) 2004; 83:43.
  43. Nakata H, Wang SL, Chung DC, et al. Oncogenic ras induces gastrin gene expression in colon cancer. Gastroenterology 1998; 115:1144.
  44. Renga M, Brandi G, Paganelli GM, et al. Rectal cell proliferation and colon cancer risk in patients with hypergastrinaemia. Gut 1997; 41:330.
  45. Thorburn CM, Friedman GD, Dickinson CJ, et al. Gastrin and colorectal cancer: a prospective study. Gastroenterology 1998; 115:275.
  46. Grabowska AM, Watson SA. Role of gastrin peptides in carcinogenesis. Cancer Lett 2007; 257:1.
  47. Bombski G, Gasiorowska A, Orszulak-Michalak D, et al. Elevated plasma gastrin, CEA, and CA 19-9 levels decrease after colorectal cancer resection. Int J Colorectal Dis 2003; 18:148.
  48. Orlando LA, Lenard L, Orlando RC. Chronic hypergastrinemia: causes and consequences. Dig Dis Sci 2007; 52:2482.
  49. Zuo Y, Jing Z, Bie M, et al. Association between Helicobacter pylori infection and the risk of colorectal cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e21832.
  50. Bold RJ, Ishizuka J, Townsend CM Jr. Progress toward hormonal therapy of gastrointestinal cancer. Ann Surg 1996; 223:4.
  51. Stepan VM, Sawada M, Todisco A, Dickinson CJ. Glycine-extended gastrin exerts growth-promoting effects on human colon cancer cells. Mol Med 1999; 5:147.
  52. Litvak DA, Hellmich MR, Iwase K, et al. JMV1155: a novel inhibitor of glycine-extended progastrin-mediated growth of a human colon cancer in vivo. Anticancer Res 1999; 19:45.
  53. Kelly A, Hollande F, Shulkes A, Baldwin GS. Expression of progastrin-derived peptides and gastrin receptors in a panel of gastrointestinal carcinoma cell lines. J Gastroenterol Hepatol 1998; 13:208.
  54. Van Solinge WW, Nielsen FC, Friis-Hansen L, et al. Expression but incomplete maturation of progastrin in colorectal carcinomas. Gastroenterology 1993; 104:1099.
  55. Ciccotosto GD, McLeish A, Hardy KJ, Shulkes A. Expression, processing, and secretion of gastrin in patients with colorectal carcinoma. Gastroenterology 1995; 109:1142.
  56. Smith AM, Watson SA. Gastrin and gastrin receptor activation: an early event in the adenoma-carcinoma sequence. Gut 2000; 47:820.
  57. Ramamoorthy S, Stepan V, Todisco A. Intracellular mechanisms mediating the anti-apoptotic action of gastrin. Biochem Biophys Res Commun 2004; 323:44.
  58. Clarke PA, Dickson JH, Harris JC, et al. Gastrin enhances the angiogenic potential of endothelial cells via modulation of heparin-binding epidermal-like growth factor. Cancer Res 2006; 66:3504.
  59. Smith JP, Verderame MF, Zagon IS. Antisense oligonucleotides to gastrin inhibit growth of human pancreatic cancer. Cancer Lett 1999; 135:107.
  60. Smith JP, Shih A, Wu Y, et al. Gastrin regulates growth of human pancreatic cancer in a tonic and autocrine fashion. Am J Physiol 1996; 270:R1078.
  61. Mu G, Ding Q, Li H, et al. Gastrin stimulates pancreatic cancer cell directional migration by activating the Gα12/13-RhoA-ROCK signaling pathway. Exp Mol Med 2018; 50:1.
  62. Payne NA, Gerber JG. Differential effects of somatostatin and prostaglandins on gastric histamine release to pentagastrin. J Pharmacol Exp Ther 1992; 263:520.
  63. Care AD, Bates RF, Swaminathan R, Ganguli PC. The role of gastrin as a calcitonin secretagogue. J Endocrinol 1971; 51:735.
  64. Parthemore JG, Deftos LJ. Calcitonin secretion in normal human subjects. J Clin Endocrinol Metab 1978; 47:184.
  65. Boyce M, Moore AR, Sagatun L, et al. Netazepide, a gastrin/cholecystokinin-2 receptor antagonist, can eradicate gastric neuroendocrine tumours in patients with autoimmune chronic atrophic gastritis. Br J Clin Pharmacol 2017; 83:466.
Topic 2551 Version 15.0

References