Organ to organ communication is important in the maintenance of fluid and electrolyte balance and blood pressure (BP) homeostasis. The gastrointestinal tract and the kidney are major organs involved in this process. The natriuresis following the ingestion of a certain amount of sodium, with or without food, may be due to an enterokine, gastrin, secreted by G-cells in the stomach and duodenum and released into the circulation. Abnormalities in stomach sodium sensing and impaired generation of gastrin, gastrin-mediated increase in renal dopamine production, gastrin-mediated natriuresis, or interaction with D1-like receptors cause hypertension. The sodium sensor in the stomach is not known but new data show that sodium sensor in the stomach, specifically in G-cells, is Scn7a. Scn7a colocalizes with gastrin in mouse stomach. Stomach/ duodenum-selective silencing of gastrin impairs the renal response to a salt load and increases BP. Stomach /duodenum selective silencing of Scn7a prevents the increase in serum gastrin following ingestion of sodium (independent of chloride or osmolality) that is also associated with impaired sodium excretion and increased BP in salt-resistant BALB/c mice fed a high sodium diet. However, BP is not increased in patients who have had gastric bypass. Indeed, the high BP can be normalized by gastric bypass. Sleeve gastrectomy actually enhances the increase in plasma gastrin following a mixed meal. By contrast, Roux-en-Y gastric bypass surgery prevents the increase in plasma gastrin following a mixed meal but either type of bypass surgery increases plasma levels of natriuretic enterokines, such as GLP-1. GLP- 1 from L-cells that is secreted via gastrin-dependent and -independent (e.g. sodium) mechanisms may also participate in this gastro-renal communication. Therefore, this competitive renewal will test the overall hypothesis that the sodium sensor that is responsible for increasing the secretion of gastrin from stomach G-cells and GLP-1 from intestinal L-cells, in response to an oral sodium load, is the sodium channel, Scn7a. Gastrin, acting on renal CCKBR, GLP-1, acting on its receptor GLP-1R, also in the kidney, and dopamine produced in the kidney, acting on renal D1R, interact to negatively regulate renal sodium transport and keep the BP in the normal range.

Public Health Relevance

The gastrointestinal tract and the kidney are major organs involved in fluid and electrolyte balance and blood pressure (BP) homeostasis and the natriuresis following the ingestion of a certain amount of sodium, with or without food, may be due to an enterokine, gastrin, secreted by G-cells in the stomach and duodenum. The sodium sensor in the stomach is not known but new data show that sodium sensor in G-cells is Scn7a, however, BP is not increased in patients who have had Roux-en-Y gastric bypass surgery because plasma levels of natriuretic enterokines, such as GLP-1 are increased. Therefore, this competitive renewal will test the overall hypothesis that Scn7a is responsible for increasing the secretion of gastrin from stomach/duodenum G-cells and GLP-1 from intestinal L-cells, in response to an oral sodium load and that Gastrin, GLP-1, and dopamine (produced) in the kidney, acting on their receptors in the kidney, negatively regulate renal sodium transport and keep the BP in the normal range.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK039308-33
Application #
9844462
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Ketchum, Christian J
Project Start
1991-07-25
Project End
2020-12-31
Budget Start
2020-01-01
Budget End
2020-12-31
Support Year
33
Fiscal Year
2020
Total Cost
Indirect Cost
Name
George Washington University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
043990498
City
Washington
State
DC
Country
United States
Zip Code
20052
Li, Fengmin; Yang, Jian; Villar, Van Anthony M et al. (2018) Loss of renal SNX5 results in impaired IDE activity and insulin resistance in mice. Diabetologia 61:727-737
Luo, Hao; Chen, Caiyu; Guo, Li et al. (2018) Exposure to Maternal Diabetes Mellitus Causes Renal Dopamine D1 Receptor Dysfunction and Hypertension in Adult Rat Offspring. Hypertension 72:962-970
Wu, Gengze; Jose, Pedro A; Zeng, Chunyu (2018) Noncoding RNAs in the Regulatory Network of Hypertension. Hypertension 72:1047-1059
Asico, Laureano D; Cuevas, Santiago; Ma, Xiaobo et al. (2018) Nephron segment-specific gene expression using AAV vectors. Biochem Biophys Res Commun 497:19-24
Jiang, Xiaoliang; Zhang, Yanrong; Yang, Yu et al. (2017) Gastrin stimulates renal dopamine production by increasing the renal tubular uptake of l-DOPA. Am J Physiol Endocrinol Metab 312:E1-E10
Tiu, Andrew C; Bishop, Michael D; Asico, Laureano D et al. (2017) Primary Pediatric Hypertension: Current Understanding and Emerging Concepts. Curr Hypertens Rep 19:70
Diao, Zhenyu; Asico, Laureano D; Villar, Van Anthony M et al. (2017) Increased renal oxidative stress in salt-sensitive human GRK4?486V transgenic mice. Free Radic Biol Med 106:80-90
Yatabe, Midori Sasaki; Iwahori, Toshiyuki; Watanabe, Ami et al. (2017) Urinary Sodium-to-Potassium Ratio Tracks the Changes in Salt Intake during an Experimental Feeding Study Using Standardized Low-Salt and High-Salt Meals among Healthy Japanese Volunteers. Nutrients 9:
Yang, Jian; Jose, Pedro A; Zeng, Chunyu (2017) Gastrointestinal-Renal Axis: Role in the Regulation of Blood Pressure. J Am Heart Assoc 6:
Barati, Michelle T; Ketchem, Corey J; Merchant, Michael L et al. (2017) Loss of NHERF-1 expression prevents dopamine-mediated Na-K-ATPase regulation in renal proximal tubule cells from rat models of hypertension: aged F344 rats and spontaneously hypertensive rats. Am J Physiol Cell Physiol 313:C197-C206

Showing the most recent 10 out of 181 publications