Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 977
Filter
1.
Arq Bras Cir Dig ; 33(2): e1506, 2020.
Article in Portuguese, English | MEDLINE | ID: mdl-32844883

ABSTRACT

BACKGROUND: Acid inhibition from chronic proton pump inhibitor use and a possible increase in gastrin can lead to changes in the regulation of hydrochloric acid production. However, it has not known whether such chronic use changes the presence of gastrin, delta, and enterochromaffin-like cells in the stomach or the relationship between gastrin and delta cells. AIM: To analyze the number of gastrin-producing gastrin cells, somatostatin-producing cells, and histamine-producing cells in patients who were chronic users of proton pump inhibitor, with or without related Helicobacter pylori infection. METHODS: Biopsies from 105 patients, including 81 chronic proton pump inhibitor users (experimental group) and 24 controls, were processed immunohistochemically and subjected to counting of gastrin, delta, and enterochromaffin-like cells in high-magnification microscopic fields and in 10 glands. RESULTS: Gastrin cell, delta cell, and enterochromaffin-like cells counts were similar across the groups and appeared to be unaffected by Helicobacter pylori infection. The ratio between gastrin cells and delta cells was higher in the chronic users of proton pump inhibitor group than in controls. CONCLUSION: Chronic users of proton pump inhibitor does not affect gastrin cell, delta cell, and enterochromaffin-like cell counts significantly, but may alter the ratio between gastrin cells and delta cells.


Subject(s)
Enterochromaffin-like Cells/metabolism , Gastrins/blood , Helicobacter Infections/therapy , Helicobacter pylori/isolation & purification , Proton Pump Inhibitors/therapeutic use , Proton Pumps/metabolism , Stomach Diseases/chemically induced , Case-Control Studies , Enterochromaffin-like Cells/drug effects , Gastrins/physiology , Helicobacter Infections/diagnosis , Humans , Proton Pump Inhibitors/adverse effects , Stomach , Stomach Diseases/blood
2.
Article in English | MEDLINE | ID: mdl-32210918

ABSTRACT

The structurally-related peptides, gastrin and cholecystokinin (CCK), were originally discovered as humoral stimulants of gastric acid secretion and pancreatic enzyme release, respectively. With the aid of methodological advances in biochemistry, immunochemistry, and molecular biology in the past several decades, our concept of gastrin and CCK as simple gastrointestinal hormones has changed considerably. Extensive in vitro and in vivo studies have shown that gastrin and CCK play important roles in several cellular processes including maintenance of gastric mucosa and pancreatic islet integrity, neurogenesis, and neoplastic transformation. Indeed, gastrin and CCK, as well as their receptors, are expressed in a variety of tumor cell lines, animal models, and human samples, and might contribute to certain carcinogenesis. In this review, we will briefly introduce the gastrin and CCK system and highlight the effects of gastrin and CCK in the regulation of cell proliferation and apoptosis in both normal and abnormal conditions. The potential imaging and therapeutic use of these peptides and their derivatives are also summarized.


Subject(s)
Cell Physiological Phenomena , Cholecystokinin/physiology , Gastrins/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Physiological Phenomena/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cholecystokinin/pharmacology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/physiology , Gastrins/pharmacology , Humans , Pancreas/metabolism , Pancreas/pathology , Pancreas/physiology , Signal Transduction/drug effects
3.
ABCD (São Paulo, Impr.) ; 33(2): e1506, 2020. tab, graf
Article in English | LILACS | ID: biblio-1130535

ABSTRACT

ABSTRACT Background: Acid inhibition from chronic proton pump inhibitor use and a possible increase in gastrin can lead to changes in the regulation of hydrochloric acid production. However, it has not known whether such chronic use changes the presence of gastrin, delta, and enterochromaffin-like cells in the stomach or the relationship between gastrin and delta cells. Aim: To analyze the number of gastrin-producing gastrin cells, somatostatin-producing cells, and histamine-producing cells in patients who were chronic users of proton pump inhibitor, with or without related Helicobacter pylori infection. Methods: Biopsies from 105 patients, including 81 chronic proton pump inhibitor users (experimental group) and 24 controls, were processed immunohistochemically and subjected to counting of gastrin, delta, and enterochromaffin-like cells in high-magnification microscopic fields and in 10 glands. Results: Gastrin cell, delta cell, and enterochromaffin-like cells counts were similar across the groups and appeared to be unaffected by Helicobacter pylori infection. The ratio between gastrin cells and delta cells was higher in the chronic users of proton pump inhibitor group than in controls. Conclusion: Chronic users of proton pump inhibitor does not affect gastrin cell, delta cell, and enterochromaffin-like cell counts significantly, but may alter the ratio between gastrin cells and delta cells.


RESUMO Racional: A inibição ácida pelo uso crônico de inibidores de bomba de prótons e o possível aumento da gastrina podem ser seguidos de alterações na regulação da produção do ácido clorídrico. Ainda não está definido se o uso crônico altera a quantidade de células G, D e ECL no estômago ou a razão células G/D. Objetivo: Avaliar o número de células G - produtoras de gastrina -, células D - produtoras de somatostatina - e células ECL - produtoras de histamina -, em pacientes com uso crônico de inibidores de bomba de prótons, com ou sem infecção pelo Helicobacter pylori. Método: Trata-se de estudo retrospectivo avaliando 105 pacientes, 81 usadores crônicos de inibidores de bomba de prótons e 24 controles, através de biópsias com contagem das células G, D e ECL por estudo imunoistoquímico, de forma quantitativa onde havia maior número de células positivas por campo microscópico de grande aumento e em 10 glândulas. Resultados: Não houve diferença estatística comparando-se o número de células G, D e ECL. A razão entre as células G e D foi maior nos pacientes usadores crônicos de inibidores de bomba de prótons. Conclusão: O uso crônico de inibidores de prótons parece não interferir na contagem das células G, D e ECL, porém, interfere na razão entre as células G e D.


Subject(s)
Humans , Stomach Diseases/chemically induced , Gastrins/blood , Helicobacter pylori/isolation & purification , Helicobacter Infections/therapy , Proton Pumps/metabolism , Enterochromaffin-like Cells/metabolism , Proton Pump Inhibitors/therapeutic use , Stomach , Stomach Diseases/blood , Gastrins/physiology , Case-Control Studies , Helicobacter Infections/diagnosis , Enterochromaffin-like Cells/drug effects , Proton Pump Inhibitors/adverse effects
4.
Endocrinology ; 160(9): 2093-2100, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31294787

ABSTRACT

Regulated endocrine-specific protein-18 (RESP18), a novel 18-kDa protein, was first identified in neuroendocrine tissue. Subsequent studies showed that Resp18 is expressed in the adrenal medulla, brain, pancreas, pituitary, retina, stomach, superior cervical ganglion, testis, and thyroid and also circulates in the plasma. Resp18 has partial homology with the islet cell antigen 512, also known as protein tyrosine phosphatase, receptor type N (PTPRN), but does not have phosphatase activity. Resp18 might serve as an intracellular signal; however, its function is unclear. It is regulated by dopamine, glucocorticoids, and insulin. We recently reported that the targeted disruption of the Resp18 locus in Dahl salt-sensitive rats increased their blood pressure and caused renal injury. The aim of the present review was to provide a comprehensive summary of the reported data currently available, especially the expression and proposed organ-specific function of Resp18.


Subject(s)
Nerve Tissue Proteins/physiology , Animals , Cardiovascular Diseases/prevention & control , Dopamine/physiology , Gastrins/physiology , Glucocorticoids/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Insulin/physiology , Nerve Tissue Proteins/chemistry , Parkinson Disease/etiology
5.
Gene ; 704: 42-48, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30980943

ABSTRACT

Angiogenesis is recognized as a sign of cancer and facilitates cancer progression and metastasis. Suppression of angiogenesis is a desirable strategy for gastric cancer (GC) management. In this study, we showed a novel role of gastrin in angiogenesis of GC. We observed that treatment with gastrin 17 (G17) increased the proliferation of AGS cells and enhanced tube formation during normoxia and hypoxia. The expression level of VEGF were increased by G17 treatment as well. Experiments on the mechanism showed that G17 promoted HIF-1α expression, which subsequently enhanced ß-catenin nuclear localization and activation of TCF3 and LEF1 and finally resulted in angiogenesis by upregulating VEGF. An in vivo experiment confirmed that G17 enhanced GC cell proliferation and angiogenesis in the resultant tumor. In conclusion, our findings indicate that gastrin promotes angiogenesis via activating HIF-1α/ß-catenin/VEGF axis in GC.


Subject(s)
Gastrins/pharmacology , Neovascularization, Pathologic/chemically induced , Stomach Neoplasms/blood supply , Stomach Neoplasms/pathology , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Gastrins/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oxygen/pharmacology , Oxygen/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Stomach Neoplasms/genetics , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
Digestion ; 97(4): 340-344, 2018.
Article in English | MEDLINE | ID: mdl-29587283

ABSTRACT

Gastrin acts physiologically as a gut hormone to stimulate acid secretion after meal and as a cell-growth factor of oxyntic mucosa. Increase in serum gastrin level happens under various conditions including Zollinger-Ellison syndrome, antral G cell hyperplasia, autoimmune gastritis, atrophic gastritis, renal failure, vagotomy, Helicobacter pylori infection and acid suppressive therapy. As acid suppressive therapy causes hypergastrinemia, the association between acid suppressive therapy and gastric neuroendocrine cell tumor (NET) has been discussed during the past 30 years. In this review article, the definition of hypergastrinemia and the related disorders including acid suppressive therapy and gastric NET are discussed.


Subject(s)
Gastrins/physiology , Gastritis/drug therapy , Neuroendocrine Tumors/blood , Proton Pump Inhibitors/adverse effects , Stomach Neoplasms/blood , Carcinogenesis/chemically induced , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastrins/blood , Gastritis/blood , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/blood , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/drug effects , Helicobacter pylori/metabolism , Humans , Neuroendocrine Tumors/pathology , Stomach Neoplasms/pathology
7.
Endocr Dev ; 32: 1-7, 2017.
Article in English | MEDLINE | ID: mdl-28873390

ABSTRACT

The physiological studies by British physiologists William Maddock Bayliss and Ernest Henry Starling, at the beginning of the last century, demonstrated the existence of specific messenger molecules (hormones) circulating in the blood that regulate the organ function and physiological mechanisms. These findings led to the concept of endocrinology. The first 2 hormones were secretin, discovered in 1902, and gastrin, discovered in 1905. Both hormones that have been described are produced in the gut. This chapter summarizes the history around the discovery of these 2 hormones, which is perceived as the birth of endocrinology. It is noteworthy that after the discovery of these 2 gastrointestinal hormones, many other hormones were detected outside the gut, and thereafter gut hormones faded from both the clinical and scientific spotlight. Only recently, the clinical importance of the gut as the body's largest endocrine organ producing a large variety of hormones has been realized. Gastrointestinal hormones are essential regulators of metabolism, growth, development and behavior and are therefore the focus of a modern pediatric endocrinologist.


Subject(s)
Endocrinology/history , Gastrointestinal Hormones/history , Animals , France , Gastrins/history , Gastrins/physiology , Gastrointestinal Hormones/physiology , Gastrointestinal Tract , Germany , History, 20th Century , History, 21st Century , Hormones/history , Humans , London , Pancreas , Secretin/history , Secretin/physiology
8.
PLoS One ; 12(9): e0184514, 2017.
Article in English | MEDLINE | ID: mdl-28902909

ABSTRACT

The cytoprotective protein clusterin is often dysregulated during tumorigenesis, and in the stomach, upregulation of clusterin marks emergence of the oxyntic atrophy (loss of acid-producing parietal cells)-associated spasmolytic polypeptide-expressing metaplasia (SPEM). The hormone gastrin is important for normal function and maturation of the gastric oxyntic mucosa and hypergastrinemia might be involved in gastric carcinogenesis. Gastrin induces expression of clusterin in adenocarcinoma cells. In the present study, we examined the expression patterns and gastrin-mediated regulation of clusterin in gastric tissue from: humans; rats treated with proton pump (H+/K+-ATPase) inhibitors and/or a gastrin receptor (CCK2R) antagonist; H+/K+-ATPase ß-subunit knockout (H/K-ß KO) mice; and Mongolian gerbils infected with Helicobacter pylori and given a CCK2R antagonist. Biological function of secretory clusterin was studied in human gastric cancer cells. Clusterin was highly expressed in neuroendocrine cells in normal oxyntic mucosa of humans and rodents. In response to hypergastrinemia, expression of clusterin increased significantly and its localization shifted to basal groups of proliferative cells in the mucous neck cell-chief cell lineage in all animal models. That shift was partially inhibited by antagonizing the CCK2R in rats and gerbils. The oxyntic mucosa of H/K-ß KO mice contained areas with clusterin-positive mucous cells resembling SPEM. In gastric adenocarcinomas, clusterin mRNA expression was higher in diffuse tumors containing signet ring cells compared with diffuse tumors without signet ring cells, and clusterin seemed to be secreted by tumor cells. In gastric cancer cell lines, gastrin increased secretion of clusterin, and both gastrin and secretory clusterin promoted survival after starvation- and chemotherapy-induced stress. Overall, our results indicate that clusterin is overexpressed in hypergastrinemic rodent models of oxyntic preneoplasia and stimulates gastric cancer cell survival.


Subject(s)
Clusterin/physiology , Gene Expression Regulation, Neoplastic , Parietal Cells, Gastric/pathology , Stomach Neoplasms/pathology , Aged , Aged, 80 and over , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Clusterin/genetics , Clusterin/metabolism , Female , Gastrins/metabolism , Gastrins/physiology , Gene Expression Profiling , Gerbillinae , Humans , Male , Mice, Knockout , Middle Aged , Parietal Cells, Gastric/metabolism , Proton Pump Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/antagonists & inhibitors , Stomach Neoplasms/metabolism
9.
Curr Opin Gastroenterol ; 33(6): 430-438, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28787289

ABSTRACT

PURPOSE OF REVIEW: The present review summarizes the past year's literature, both clinical and basic science, regarding physiologic and pharmacologic regulation of gastric acid secretion in health and disease. RECENT FINDINGS: Gastric acid kills microorganisms, assists digestion, and facilitates absorption of iron, calcium, and vitamin B12. The main stimulants of acid secretion are the hormone gastrin, released from antral G cells; paracrine agent histamine, released from oxyntic enterochromaffin-like cells; and neuropeptide acetylcholine, released from antral and oxyntic intramural neurons. Gastrin is also a trophic hormone that participates in carcinogenesis. Helicobacter pylori may increase or decrease acid secretion depending upon the acuity and predominant anatomic focus of infection; most patients manifest hypochlorhydria. Despite the fact that proton pump inhibitors (PPIs) are amongst the most widely prescribed drugs, they are underutilized in patients at high risk for UGI bleeding. Although generally considered well tolerated, concerns have been raised regarding associations between PPI use and dementia, kidney disease, myocardial infarction, pneumonia, osteoporosis, dysbiosis, small bowel injury, micronutrient deficiency, and fundic gland polyps. SUMMARY: Our understanding of the physiologic, pathophysiologic, and pharmacologic regulation of gastric secretion continues to advance. Such knowledge is crucial for improved and safe management of acid-peptic disorders.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Acetylcholine/physiology , Gastric Acid/physiology , Gastrins/physiology , Helicobacter Infections/metabolism , Helicobacter pylori , Histamine/physiology , Humans , Proton Pump Inhibitors/pharmacology
10.
Curr Gastroenterol Rep ; 19(7): 32, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28608155

ABSTRACT

PURPOSE OF REVIEW: Neuroendocrine tumors (NETs) were initially identified as a separate entity in the early 1900s as a unique malignancy that secretes bioactive amines. GI-NETs are the most frequent type and represent a unique subset of NETs, because at least 75% of these tumors represent gastrin stimulation of the enterochromaffin-like cell located in the body of the stomach. The purpose of this review is to understand the specific role of gastrin in the generation of Gastric NETs (G-NETs). RECENT FINDINGS: We review here the origin of enterochromaffin cells gut and the role of hypergastrinemia in gastric enteroendocrine tumorigenesis. We describe generation of the first genetically engineered mouse model of gastrin-driven G-NETs that mimics the human phenotype. The common mechanism observed in both the hypergastrinemic mouse model and human carcinoids is translocation of the cyclin-dependent inhibitor p27kip to the cytoplasm and its subsequent degradation by the proteasome. Therapies that block degradation of p27kip, the CCKBR2 gastrin receptor, or gastrin peptide are likely to facilitate treatment.


Subject(s)
Gastrins/physiology , Neuroendocrine Tumors/etiology , Stomach Neoplasms/etiology , Animals , Carcinoid Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoplasm/metabolism , Disease Models, Animal , Enterochromaffin-like Cells/physiology , Gastrins/blood , Humans , Mice , Neuroendocrine Tumors/metabolism , Phenotype , Receptor, Cholecystokinin B/metabolism , Stomach Neoplasms/metabolism
11.
Curr Opin Gastroenterol ; 32(6): 452-460, 2016 11.
Article in English | MEDLINE | ID: mdl-27607343

ABSTRACT

PURPOSE OF REVIEW: The present review summarizes the past year's literature, both clinical and basic science, regarding neuroendocrine and intracellular regulation of gastric acid secretion and proper use of antisecretory medications. RECENT FINDINGS: Gastric acid kills microorganisms, modulates the gut microbiome, assists in digestion of protein, and facilitates absorption of iron, calcium, and vitamin B12. The main stimulants of acid secretion are gastrin, released from antral G cells; histamine, released from oxyntic enterochromaffin-like cells; and acetylcholine, released from antral and oxyntic intramural neurons. Other stimulants include ghrelin, motilin, and hydrogen sulfide. The main inhibitor of acid secretion is somatostatin, released from oxyntic and antral D cells. Glucagon-like peptide-1 also inhibits acid secretion. Proton pump inhibitors (PPIs) reduce acid secretion and, as a result, decrease somatostatin and thus stimulate gastrin secretion. Although considered well tolerated drugs, concerns have been raised this past year regarding associations between PPI use and kidney disease, dementia, and myocardial infarction; the quality of evidence, however, is very low. SUMMARY: Our understanding of the physiology of gastric secretion and proper use of PPIs continues to advance. Such knowledge is crucial for improved management of acid-peptic disorders.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Chloride Channels/physiology , Gastrins/physiology , Gastrointestinal Microbiome/drug effects , Histamine/physiology , Humans , Neurosecretory Systems/physiology , Potassium Channels/physiology , Proton Pump Inhibitors/adverse effects , Somatostatin/physiology
12.
Cancer Res ; 76(12): 3618-28, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197176

ABSTRACT

Subpopulations of cancer stem-like cells (CSC) are thought to drive tumor progression and posttreatment recurrence in multiple solid tumors. However, the mechanisms that maintain stable proportions of self-renewing CSC within heterogeneous tumors under homeostatic conditions remain poorly understood. Progastrin is a secreted peptide that exhibits tumor-forming potential in colorectal cancer, where it regulates pathways known to modulate colon CSC behaviors. In this study, we investigated the role of progastrin in regulating CSC phenotype in advanced colorectal cancer. Progastrin expression and secretion were highly enriched in colon CSC isolated from human colorectal cancer cell lines and colon tumor biopsies. Progastrin expression promoted CSC self-renewal and survival, whereas its depletion by RNA interference-mediated or antibody-mediated strategies altered the homeostatic proportions of CSC cells within heterogeneous colorectal cancer tumors. Progastrin downregulation also decreased the frequency of ALDH(high) cells, impairing their tumor-initiating potential, and inhibited the high glycolytic activity of ALDH(high) CSC to limit their self-renewal capability. Taken together, our results show how colorectal CSC maintain their tumor-initiating and self-renewal capabilities by secreting progastrin, thereby contributing to the tumor microenvironment to support malignancy. Cancer Res; 76(12); 3618-28. ©2016 AACR.


Subject(s)
Colonic Neoplasms/pathology , Gastrins/physiology , Neoplastic Stem Cells/physiology , Protein Precursors/physiology , Aldehyde Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Cell Survival , Humans , Mice , Tumor Microenvironment
15.
Surg Today ; 46(6): 633-40, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26148809

ABSTRACT

Pancreatic regeneration (PR) is an interesting phenomenon that could provide clues as to how the control of diabetes mellitus might be achieved. Due to the different regenerative abilities of the pancreas and liver, the molecular mechanism responsible for PR is largely unknown. In this review, we describe five representative murine models of PR and thirteen humoral mitogens that stimulate ß-cell proliferation. We also describe pancreatic ontogenesis, including the molecular transcriptional differences between α-cells and ß-cells. Furthermore, we review 14 murine models which carry defects in genes related to key transcription factors for pancreatic ontogenesis to gain further insight into pancreatic development.


Subject(s)
Epidermal Growth Factor/physiology , Incretins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Pancreas/physiology , Regeneration/genetics , Regeneration/physiology , Transcription Factors/physiology , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Gastrins/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Glucagon-Secreting Cells , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Mice , Mice, Knockout , Models, Animal , Pancreas/cytology , Transcription Factors/genetics
16.
Curr Opin Gastroenterol ; 31(6): 479-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26376477

ABSTRACT

PURPOSE OF REVIEW: This review summarizes the past year's literature regarding the neuroendocrine and intracellular regulation of gastric acid secretion, discussing both basic and clinical aspects. RECENT FINDINGS: Gastric acid facilitates the digestion of protein as well as the absorption of iron, calcium, vitamin B12, and certain medications. High acidity kills ingested microorganisms and limits bacterial overgrowth, enteric infection, and possibly spontaneous bacterial peritonitis. The main stimulants of acid secretion are gastrin, released from antral gastrin cells; histamine, released from oxyntic enterochromaffin-like cells; and acetylcholine, released from antral and oxyntic intramural neurons. Ghrelin and coffee also stimulate acid secretion whereas somatostatin, cholecystokinin, glucagon-like peptide-1, and atrial natriuretic peptide inhibit acid secretion. Although 95% of parietal cells are contained within the oxyntic mucosa (fundus and body), 50% of human antral glands contain parietal cells. Proton pump inhibitors are considered well tolerated drugs, but concerns have been raised regarding dysbiosis, atrophic gastritis, hypergastrinemia, hypomagnesemia, and enteritis/colitis. SUMMARY: Our understanding of the functional anatomy and physiology of gastric secretion continues to advance. Such knowledge is crucial for improved management of acid-peptic disorders, prevention and management of neoplasia, and the development of novel medications.


Subject(s)
Gastric Acid/metabolism , Gastrins/physiology , H(+)-K(+)-Exchanging ATPase/physiology , Histamine/physiology , Humans , Neurosecretory Systems/anatomy & histology , Neurosecretory Systems/pathology , Neurosecretory Systems/physiology , Proton Pump Inhibitors/adverse effects , Signal Transduction/physiology
17.
J Biol Chem ; 290(24): 15197-209, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25922072

ABSTRACT

The guanine nucleotide exchange factor Rgnef (also known as ArhGEF28 or p190RhoGEF) promotes colon carcinoma cell motility and tumor progression via interaction with focal adhesion kinase (FAK). Mechanisms of Rgnef activation downstream of integrin or G protein-coupled receptors remain undefined. In the absence of a recognized G protein signaling homology domain in Rgnef, no proximal linkage to G proteins was known. Utilizing multiple methods, we have identified Rgnef as a new effector for Gα13 downstream of gastrin and the type 2 cholecystokinin receptor. In DLD-1 colon carcinoma cells depleted of Gα13, gastrin-induced FAK Tyr(P)-397 and paxillin Tyr(P)-31 phosphorylation were reduced. RhoA GTP binding and promoter activity were increased by Rgnef in combination with active Gα13. Rgnef co-immunoprecipitated with activated Gα13Q226L but not Gα12Q229L. The Rgnef C-terminal (CT, 1279-1582) region was sufficient for co-immunoprecipitation, and Rgnef-CT exogenous expression prevented Gα13-stimulated SRE activity. A domain at the C terminus of the protein close to the FAK binding domain is necessary to bind to Gα13. Point mutations of Rgnef-CT residues disrupt association with active Gα13 but not Gαq. These results show that Rgnef functions as an effector of Gα13 signaling and that this linkage may mediate FAK activation in DLD-1 colon carcinoma cells.


Subject(s)
Colonic Neoplasms/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Gastrins/physiology , Guanine Nucleotide Exchange Factors/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , Humans , Paxillin/chemistry , Paxillin/metabolism , Phosphorylation , Receptor, Cholecystokinin B/metabolism , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/metabolism , Tyrosine/metabolism
18.
Oncogene ; 34(24): 3120-30, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25109333

ABSTRACT

Angiogenesis is essential in tumor progression and metastatic process, and increased angiogenesis has been associated with poor prognosis and relapse of colorectal cancer (CRC). VEGF has become the main target of anti-angiogenic therapy. However, most patients relapse after an initial response or present a resistance to the treatment. Identification of new pro-angiogenic factors may help to improve anti-angiogenic therapy. In this study, we demonstrated that the pro-hormone progastrin (PG), over-expressed in CRC, recognized as a growth factor, is a potent pro-angiogenic factor. In transgenic mice and human colorectal HPs producing high levels of PG, we correlated PG overexpression with an increased vascularization. In vitro, exogenous PG and conditioned media (CM) from CRC cells producing PG increased endothelial cell proliferation and migration. We also showed that treatment with exogenous PG can increase the ability of endothelial cells to form capillary-like structures. Moreover, we demonstrated that PG enhanced endothelial permeability. The finding that PG stimulated the phosphorylation of vascular endothelial (VE)-cadherin, p125-FAK, paxillin and induced actin remodelling was consistent with a role of these components in PG-stimulated endothelial cell migration and permeability. The pro-angiogenic effects observed with CM were significantly inhibited when CRC cells expressed a PG shRNA. In vivo, we found an important decrease in tumor growth and neovascularization when the CRC cells expressing the PG shRNA were xenografted in mice or in the chick chorioallantoic membrane model. We also observed an increase in the coverage of blood vessels by pericytes and a decrease in endothelial permeability when PG expression was blocked. Our results demonstrate that PG is a new pro-angiogenic factor in CRC and an attractive therapeutic target.


Subject(s)
Colorectal Neoplasms/blood supply , Gastrins/physiology , Neovascularization, Pathologic/genetics , Protein Precursors/physiology , Animals , Cells, Cultured , Chick Embryo , Colorectal Neoplasms/pathology , Gastrins/genetics , Gastrins/pharmacology , HCT116 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, SCID , Mice, Transgenic , Protein Precursors/genetics , Protein Precursors/pharmacology , RNA, Small Interfering/pharmacology
19.
Dig Dis Sci ; 60(6): 1522-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25480404

ABSTRACT

Gastric cancer occurs almost exclusively in patients with gastritis. Since Helicobacter pylori (Hp) was proved to cause gastritis, Hp was also expected to play a role in gastric carcinogenesis. Despite extensive studies, the mechanisms by which Hp cause gastric cancer are still poorly understood. However, there is evidence that the anatomical site of Hp infection is of major importance. Infection confined to the antral mucosa protects against gastric cancer but predisposes to duodenal ulcer, whereas Hp infection of the oxyntic mucosa increases the risk of gastric cancer. Hp infection does not predispose to cancers in the gastric cardia. In patients with atrophic gastritis of the oxyntic mucosa, the intragastric pH is elevated and the concentration of microorganisms in the stomach is increased. This does not lead to increased risk of gastric cancer at all anatomical sites. The site specificity of Hp infection in relation to cancer risk indicates that neither Hp nor the changes in gastric microflora due to gastric hypoacidity are carcinogenic per se. However, reduced gastric acidity also leads to hypergastrinemia, which stimulates the function and proliferation of enterochromaffin-like (ECL) cells located in the oxyntic mucosa. The ECL cell may be more important in human gastric carcinogenesis than previously realized, as every condition causing long-term hypergastrinemia in animals results in the development of neoplasia in the oxyntic mucosa. Patients with hypergastrinemia will far more often develop carcinomas in the gastric corpus. In conclusion, hypergastrinemia may explain the carcinogenic effect of Hp.


Subject(s)
Gastrins/physiology , Gastritis/microbiology , Helicobacter Infections/pathology , Stomach Neoplasms/pathology , Animals , Gastric Mucosa/pathology , Helicobacter pylori/pathogenicity , Humans , Hydrogen-Ion Concentration , Risk
20.
Gut ; 64(4): 544-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24951258

ABSTRACT

OBJECTIVE: Progastrin is the incompletely cleaved precursor of gastrin that is secreted by G-cells in the gastric antrum. Both gastrin and progastrin bind to the CCK2 receptor (Cckbr or CCK2R) expressed on a subset of gastric epithelial cells. Little is known about how gastrin peptides and CCK2R regulate gastric stem cells and carcinogenesis. Interconversion among progenitors in the intestine is documented, but the mechanisms by which this occurs are poorly defined. DESIGN: We generated CCK2R-CreERT mice and performed inducible lineage tracing experiments. CCK2R+ antral cells and Lgr5+ antral stem cells were cultured in a three-dimensional in vitro system. We crossed progastrin-overexpressing mice with Lgr5-GFP-CreERT mice and examined the role of progastrin and CCK2R in Lgr5+ stem cells during MNU-induced carcinogenesis. RESULTS: Through lineage tracing experiments, we found that CCK2R defines antral stem cells at position +4, which overlapped with an Lgr5(neg or low) cell population but was distinct from typical antral Lgr5(high) stem cells. Treatment with progastrin interconverts Lgr5(neg or low) CCK2R+ cells into Lgr5(high) cells, increases CCK2R+ cell numbers and promotes gland fission and carcinogenesis in response to the chemical carcinogen MNU. Pharmacological inhibition or genetic ablation of CCK2R attenuated progastrin-dependent stem cell expansion and carcinogenesis. CONCLUSIONS: CCK2R labels +4 antral stem cells that can be activated and expanded by progastrin, thus identifying one hormonal trigger for gastric stem cell interconversion and a potential target for gastric cancer chemoprevention and therapy.


Subject(s)
Carcinogenesis , Pyloric Antrum/cytology , Receptor, Cholecystokinin B/physiology , Stem Cells/physiology , Animals , Cells, Cultured , Gastrins/physiology , Mice , Protein Precursors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...