1
|
Chrysant SG. The Interaction of Kidneys and Gut in Development of Salt-Sensitive Hypertension. Cardiol Rev 2024; 32:356-361. [PMID: 37273192 DOI: 10.1097/crd.0000000000000518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The incidence of salt-sensitive hypertension is quite common and varies between 30-60% in hypertensive patients. Regarding the causal role of high salt intake in the development of salt-sensitive hypertension, recent evidence has demonstrated that the gut through its microbiota plays a significant role in its genesis. Besides the gut, the kidneys also play important role in salt-sensitive hypertension and there is clinical and experimental evidence of an interrelationship between the gut and the kidneys in the development of salt-sensitive hypertension through the so-called "gastro-renal axis." The gut besides being an absorptive organ, it is also a hormonal secretory organ involving the secretion of gastrin, dopamine, norepinephrine, angiotensin, and aldosterone which through their action with the kidneys are involved in the development of salt-sensitive hypertension. In addition, the kidneys exert a protective role against the development of hypertension through the secretion of prostaglandins and their vasodilatory action. To assess the current evidence on the role of high salt intake and the interplay of the gut and kidneys in its development, a Medline search of the English literature was contacted between 2012 and 2022, and 46 pertinent papers were selected. These papers together with collateral literature will be discussed in this review.
Collapse
Affiliation(s)
- Steven G Chrysant
- From the University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
2
|
Hinrichs GR, Hovind P, Asmar A. The GLP-1-mediated gut-kidney cross talk in humans: mechanistic insight. Am J Physiol Cell Physiol 2024; 326:C567-C572. [PMID: 38105752 PMCID: PMC11193450 DOI: 10.1152/ajpcell.00476.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Incretin-based therapy is an antidiabetic and antiobesity approach mimicking glucagon-like peptide-1 (GLP-1) with additional end-organ protection. This review solely focuses on randomized, controlled mechanistic human studies, investigating the renal effects of GLP-1. There is no consensus about the localization of GLP-1 receptors (GLP-1Rs) in human kidneys. Rodent and primate data suggest GLP-1R distribution in smooth muscle cells in the preglomerular vasculature. Native GLP-1 and GLP-1R agonists elicit renal effects. Independently of renal plasma flow and glomerular filtration rate, GLP-1 has a natriuretic effect but only during volume expansion. This is associated with high renal extraction of GLP-1, suppression of angiotensin II, and increased medullary as well as cortical perfusion. These observations may potentially indicate that impaired GLP-1 sensing could establish a connection between salt sensitivity and insulin resistance. It is concluded that a functional GLP-1 kidney axis exists in humans, which may play a role in renoprotection.
Collapse
Affiliation(s)
- Gitte R Hinrichs
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Peter Hovind
- Department of Clinical Physiology & Nuclear Medicine, Bispebjerg-Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ali Asmar
- Department of Clinical Physiology & Nuclear Medicine, Bispebjerg-Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Physiology & Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Han Y, Bagchi P, Yun CC. Regulation of the intestinal Na +/H + exchanger NHE3 by AMP-activated kinase is dependent on phosphorylation of NHE3 at S555 and S563. Am J Physiol Cell Physiol 2024; 326:C50-C59. [PMID: 38047302 PMCID: PMC11192475 DOI: 10.1152/ajpcell.00540.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/05/2023]
Abstract
Electroneutral NaCl transport by Na+/H+ exchanger 3 (NHE3, SLC9A3) is the major Na+ absorptive mechanism in the intestine and decreased NHE3 activity contributes to diarrhea. Patients with diabetes often experience gastrointestinal adverse effects and medications are often a culprit for chronic diarrhea in type 2 diabetes (T2D). We have shown previously that metformin, the most widely prescribed drug for the treatment of T2D, induces diarrhea by inhibition of Na+/H+ exchanger 3 (NHE3) in rodent models of T2D. Metformin was shown to activate AMP-activated protein kinase (AMPK), but AMPK-independent glycemic effects of metformin are also known. The current study is undertaken to determine whether metformin inhibits NHE3 by activation of AMPK and the mechanism by which NHE3 is inhibited by AMPK. Inhibition of NHE3 by metformin was abolished by knockdown of AMPK-α1 or AMPK-α2. AMPK activation by 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) phosphorylated NHE3 at S555. S555 is the primary site of phosphorylation by protein kinase A (PKA), but AMPK phosphorylated S555 independently of PKA. Using Mass spectrometry, we found S563 as a newly recognized phosphorylation site in NHE3. Altering either S555 or S563 to Ala was sufficient to block the inhibition of NHE3 activity by AMPK. NHE3 inhibition is dependent on ubiquitination by the E3 ubiquitin ligase Nedd4-2 and metformin was shown to induce NHE3 internalization via Nedd4-2-mediated ubiquitination. AICAR did not increase NHE3 ubiquitination when S555 or S563 was mutated. We conclude that AMPK activation inhibits NHE3 activity and NHE3 inhibition is associated with phosphorylation of NHE3 at S555 and S563.NEW & NOTEWORTHY We show that AMP-activated protein kinase (AMPK) phosphorylates NHE3 at S555 and S563 to inhibit NHE3 activity in intestinal epithelial cells. Phosphorylation of NHE3 by AMPK is necessary for ubiquitination of NHE3.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University, Atlanta, Georgia, United States
| | - C Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
4
|
Han Y, Srinivasan S, Yun CC. Inhibition of protein kinase C-α and activation of ezrin by Lactobacillus acidophilus restore Na +/H + exchange activity and fluid absorption in db/db mice. Am J Physiol Endocrinol Metab 2023; 325:E214-E226. [PMID: 37467022 PMCID: PMC10511175 DOI: 10.1152/ajpendo.00145.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023]
Abstract
Gastrointestinal (GI) complications, including diarrhea, constipation, and gastroparesis, are common in patients with diabetes. Dysregulation of the Na+/H+ exchanger NHE3 in the intestine is linked to diarrhea and constipation, and recent studies showed that NHE3 expression is reduced in type 1 diabetes and metformin causes diarrhea in the db/db mouse model of type 2 diabetes (T2D) via inhibition of NHE3. In this study, we investigated whether NHE3 expression is altered in type 2 diabetic intestine and the underlying mechanism that dysregulates NHE3. NHE3 expression in the brush border membrane (BBM) of the intestine of diabetic mice and humans was decreased. Protein kinase C (PKC) activation is associated with pathologies of diabetes, and immunofluorescence (IF) analysis revealed increased BBM PKCα abundance. Inhibition of PKCα increased NHE3 BBM abundance and NHE3-mediated intestinal fluid absorption in db/db mice. Previous studies have shown that Lactobacillus acidophilus (LA) stimulates intestinal ion transporters. LA increased NHE3 BBM expression and mitigated metformin-mediated inhibition of NHE3 in vitro and in vivo. To understand the underlying mechanism of LA-mediated stimulation of NHE3, we used Caco-2bbe cells overexpressing PKCα that mimic the elevated state of PKCα in T2D. LA diminished PKCα BBM expression, increased phosphorylation of ezrin, and the interaction of NHE3 with NHE regulatory factor 2 (NHERF2). In addition, inhibition of PKCι blocked phosphorylation of ezrin and activation of NHE3 by LA. These findings demonstrate that NHE3 is downregulated in T2D, and LA restores NHE3 expression via regulation of PKCα, PKCι, and ezrin.NEW & NOTEWORTHY We used mouse models of type 2 diabetes (T2D) and human patient-derived samples to show that Na+/H+ exchanger 3 (NHE3) expression is decreased in T2D. We show that protein kinase C-α (PKCα) is activated in diabetes and inhibition of PKCα increased NHE3 expression and mitigates diarrhea. We show that Lactobacillus acidophilus (LA) stimulates NHE3 via inhibition of PKCα and phosphorylation of ezrin.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Shanthi Srinivasan
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - C Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
5
|
Jiang X, Liu Y, Zhang XY, Liu X, Liu X, Wu X, Jose PA, Duan S, Xu FJ, Yang Z. Intestinal Gastrin/CCKBR (Cholecystokinin B Receptor) Ameliorates Salt-Sensitive Hypertension by Inhibiting Intestinal Na +/H + Exchanger 3 Activity Through a PKC (Protein Kinase C)-Mediated NHERF1 and NHERF2 Pathway. Hypertension 2022; 79:1668-1679. [PMID: 35674015 PMCID: PMC9278716 DOI: 10.1161/hypertensionaha.121.18791] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The present study directly tested the crucial role of intestinal gastrin/CCKBR (cholecystokinin B receptor) in the treatment of salt-sensitive hypertension. Methods: Adult intestine-specific Cckbr-knockout mice (Cckbrfl/flvillin-Cre) and Dahl salt-sensitive rats were studied on the effect of high salt intake (8% NaCl, 6–7 weeks) on intestinal Na+/H+ exchanger 3 expression, urine sodium concentration, and blood pressure. High-salt diet increased urine sodium concentration and systolic blood pressure to a greater extent in Cckbrfl/flvillin-Cre mice and Dahl salt-sensitive rats than their respective controls, Cckbrfl/flvillin mice and SS13BN rats. We constructed gastrin-SiO2 microspheres to enable gastrin to stimulate specifically and selectively intestinal CCKBR without its absorption into the circulation. Results: Gastrin-SiO2 microspheres treatment prevented the high salt-induced hypertension and increase in urine Na concentration by inhibiting intestinal Na+/H+ exchanger 3 trafficking and activity, increasing stool sodium without inducing diarrhea. Gastrin-mediated inhibition of intestinal Na+/H+ exchanger 3 activity, related to a PKC (protein kinase C)-mediated activation of NHERF1 and NHERF2. Conclusions: These results support a crucial role of intestinal gastrin/CCKBR in decreasing intestinal sodium absorption and keeping the blood pressure in the normal range. The gastrointestinal administration of gastrin-SiO2 microspheres is a promising and safe strategy to treat salt-sensitive hypertension without side effects.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| | - Yunpeng Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| | - Xin-Yang Zhang
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, P.R. China (X.-Y.Z., S.D., F.-J.X.)
| | - Xue Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| | - Xing Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| | - Xianxian Wu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| | - Pedro A Jose
- Department of Pharmacology and Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC.,Division of Kidney Diseases and Hypertension, Department of Medicine (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Shun Duan
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, P.R. China (X.-Y.Z., S.D., F.-J.X.)
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, P.R. China (X.-Y.Z., S.D., F.-J.X.)
| | - Zhiwei Yang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, P.R. China (X.J., Y.L., Xue Liu, Xing Liu, X.W., Z.Y.)
| |
Collapse
|
6
|
Han Y, Yun CC. Metformin Inhibits Na +/H + Exchanger NHE3 Resulting in Intestinal Water Loss. Front Physiol 2022; 13:867244. [PMID: 35444557 PMCID: PMC9014215 DOI: 10.3389/fphys.2022.867244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
Glycemic control is the key to the management of type 2 diabetes. Metformin is an effective, widely used drug for controlling plasma glucose levels in diabetes, but it is often the culprit of gastrointestinal adverse effects such as abdominal pain, nausea, indigestion, vomiting, and diarrhea. Diarrhea is a complex disease and altered intestinal transport of electrolytes and fluid is a common cause of diarrhea. Na+/H+ exchanger 3 (NHE3, SLC9A3) is the major Na+ absorptive mechanism in the intestine and our previous study has demonstrated that decreased NHE3 contributes to diarrhea associated with type 1 diabetes. The goal of this study is to investigate whether metformin regulates NHE3 and inhibition of NHE3 contributes to metformin-induced diarrhea. We first determined whether metformin alters intestinal water loss, the hallmark of diarrhea, in type 2 diabetic db/db mice. We found that metformin decreased intestinal water absorption mediated by NHE3. Metformin increased fecal water content although mice did not develop watery diarrhea. To determine the mechanism of metformin-mediated regulation of NHE3, we used intestinal epithelial cells. Metformin inhibited NHE3 activity and the effect of metformin on NHE3 was mimicked by a 5'-AMP-activated protein kinase (AMPK) activator and blocked by pharmacological inhibition of AMPK. Metformin increased phosphorylation and ubiquitination of NHE3, resulting in retrieval of NHE3 from the plasma membrane. Previous studies have demonstrated the role of neural precursor cell expressed, developmentally down-regulated 4-2 (Nedd4-2) in regulation of human NHE3. Silencing of Nedd4-2 mitigated NHE3 inhibition and ubiquitination by metformin. Our findings suggest that metformin-induced diarrhea in type 2 diabetes is in part caused by reduced Na+ and water absorption that is associated with NHE3 inhibition, probably by AMPK.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - C. Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
7
|
Jenkin KA, Han Y, Lin S, He P, Yun CC. Nedd4-2-dependent Ubiquitination Potentiates the Inhibition of Human NHE3 by Cholera Toxin and Enteropathogenic Escherichia coli. Cell Mol Gastroenterol Hepatol 2021; 13:695-716. [PMID: 34823064 PMCID: PMC8789535 DOI: 10.1016/j.jcmgh.2021.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Diarrhea is one of the most common illnesses and is often caused by bacterial infection. Recently, we have shown that human Na+/H+ exchanger NHE3 (hNHE3), but not non-human NHE3s, interacts with the E3 ubiquitin ligase Nedd4-2. We hypothesize that this property of hNHE3 contributes to the increased severity of diarrhea in humans. METHODS We used humanized mice expressing hNHE3 in the intestine (hNHE3int) to compare the contribution of hNHE3 and mouse NHE3 to diarrhea induced by cholera toxin (CTX) and enteropathogenic Escherichia coli (EPEC). We measured Na+/H+ exchange activity and fluid absorption. The role of Nedd4-2 on hNHE3 activity and ubiquitination was determined by knockdown in Caco-2bbe cells. The effects of protein kinase A (PKA), the primary mediator of CTX-induced diarrhea, on Nedd4-2 and hNHE3 phosphorylation and their interaction were determined. RESULTS The effects of CTX and EPEC were greater in hNHE3int mice than in control wild-type (WT) mice, resulting in greater inhibition of NHE3 activity and increased fluid accumulation in the intestine, the hallmark of diarrhea. Activation of PKA increased ubiquitination of hNHE3 and enhanced interaction of Nedd4-2 with hNHE3 via phosphorylation of Nedd4-2 at S342. S342A mutation mitigated the Nedd4-2-hNHE3 interaction and blocked PKA-induced inhibition of hNHE3. Unlike non-human NHE3s, inhibition of hNHE3 by PKA is independent of NHE3 phosphorylation, suggesting a distinct mechanism of hNHE3 regulation. CONCLUSIONS The effects of CTX and EPEC on hNHE3 are amplified, and the unique properties of hNHE3 may contribute to diarrheal symptoms occurring in humans.
Collapse
Affiliation(s)
- Kayte A. Jenkin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Songbai Lin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Chris Yun, PhD, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia 30324. fax: (404) 727-5767.
| |
Collapse
|
8
|
NFAT5 Is Involved in GRP-Enhanced Secretion of GLP-1 by Sodium. Int J Mol Sci 2021; 22:ijms22083951. [PMID: 33921209 PMCID: PMC8069329 DOI: 10.3390/ijms22083951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
Gastrin, secreted by G-cells, and glucagon-like peptide-1 (GLP-1), secreted by L-cells, may participate in the regulation of sodium balance. We studied the effect of sodium in mice in vivo and mouse ileum and human L-cells, on GLP-1 secretion, and the role of NFAT5 and gastrin-releasing peptide receptor (GRPR) in this process. A high-sodium diet increases serum GLP-1 levels in mice. Increasing sodium concentration stimulates GLP-1 secretion from mouse ileum and L-cells. GRP enhances the high sodium-induced increase in GLP-1 secretion. High sodium increases cellular GLP-1 expression, while low and high sodium concentrations increase NFAT5 and GRPR expression. Silencing NFAT5 in L-cells abrogates the stimulatory effect of GRP on the high sodium-induced GLP-1 secretion and protein expression, and the sodium-induced increase in GRPR expression. GLP-1 and gastrin decrease the expression of Na+-K+/ATPase and increase the phosphorylation of sodium/hydrogen exchanger type 3 (NHE3) in human renal proximal tubule cells (hRPTCs). This study gives a new perspective on the mechanisms of GLP-1 secretion, especially that engendered by ingested sodium, and the ability of GLP-1, with gastrin, to decrease Na+-K+/ATPase expression and NHE3 function in hRPTCs. These results may contribute to the better utilization of current and future GLP-1-based drugs in the treatment of hypertension.
Collapse
|
9
|
Abraham T, McGovern CO, Linton SS, Wilczynski Z, Adair JH, Matters GL. Aptamer-Targeted Calcium Phosphosilicate Nanoparticles for Effective Imaging of Pancreatic and Prostate Cancer. Int J Nanomedicine 2021; 16:2297-2309. [PMID: 33776434 PMCID: PMC7989532 DOI: 10.2147/ijn.s295740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/06/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose Accurate tumor identification and staging can be difficult. Aptamer-targeted indocyanine green (ICG)-nanoparticles can enhance near-infrared fluorescent imaging of pancreatic and prostate tumors and could improve early cancer detection. This project explored whether calcium-phosphosilicate nanoparticles, also known as NanoJackets (NJs), that were bioconjugated with a tumor-specific targeting DNA aptamer could improve the non-invasive detection of pancreatic and prostate tumors. Methods Using in vivo near-infrared optical imaging and ex vivo fluorescence analysis, DNA aptamer-targeted ICG-loaded NJs were compared to untargeted NJs for detection of tumors. Results Nanoparticles were bioconjugated with the DNA aptamer AP1153, which binds to the CCK-B receptor (CCKBR). Aptamer bioconjugated NJs were not significantly increased in size compared with unconjugated nanoparticles. AP1153-ICG-NJ accumulation in orthotopic pancreatic tumors peaked at 18 h post-injection and the ICG signal was cleared by 36 h with no evidence on uptake by non-tumor tissues. Ex vivo tumor imaging confirmed the aptamer-targeted NJs accumulated to higher levels than untargeted NJs, were not taken up by normal pancreas, exited from the tumor vasculature, and were well-dispersed throughout pancreatic and prostate tumors despite extensive fibrosis. Specificity for AP1153-NJ binding to the CCK-B receptor on pancreatic tumor cells was confirmed by pre-treating tumor-bearing mice with the CCK receptor antagonist proglumide. Proglumide pre-treatment reduced the in vivo tumoral accumulation of AP1153-NJs to levels comparable to that of untargeted NJs. Conclusion Through specific interactions with CCK-B receptors, tumor-targeted nanoparticles containing either ICG or rhodamine WT were well distributed throughout the matrix of both pancreatic and prostate tumors. Tumor-targeted NJs carrying various imaging agents can enhance tumor detection.
Collapse
Affiliation(s)
- Thomas Abraham
- Departments of Neural and Behavioral Sciences and the Microscopy Imaging Core Facility, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Christopher O McGovern
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Samuel S Linton
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Zachary Wilczynski
- Departments of Materials Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - James H Adair
- Departments of Materials Science, The Pennsylvania State University, University Park, PA, 16802, USA.,Department of Biomedical Engineering and Pharmacology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Gail L Matters
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
10
|
Liu C, Chen K, Wang H, Zhang Y, Duan X, Xue Y, He H, Huang Y, Chen Z, Ren H, Wang H, Zeng C. Gastrin Attenuates Renal Ischemia/Reperfusion Injury by a PI3K/Akt/Bad-Mediated Anti-apoptosis Signaling. Front Pharmacol 2020; 11:540479. [PMID: 33343341 PMCID: PMC7740972 DOI: 10.3389/fphar.2020.540479] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/24/2020] [Indexed: 12/25/2022] Open
Abstract
Ischemic/reperfusion (I/R) injury is the primary cause of acute kidney injury (AKI). Gastrin, a gastrointestinal hormone, is involved in the regulation of kidney function of sodium excretion. However, whether gastrin has an effect on kidney I/R injury is unknown. Here we show that cholecystokinin B receptor (CCKBR), the gastrin receptor, was significantly up-regulated in I/R-injured mouse kidneys. While pre-administration of gastrin ameliorated I/R-induced renal pathological damage, as reflected by the levels of serum creatinine and blood urea nitrogen, hematoxylin and eosin staining and periodic acid-Schiff staining. The protective effect could be ascribed to the reduced apoptosis for gastrin reduced tubular cell apoptosis both in vivo and in vitro. In vitro studies also showed gastrin preserved the viability of hypoxia/reoxygenation (H/R)-treated human kidney 2 (HK-2) cells and reduced the lactate dehydrogenase release, which were blocked by CI-988, a specific CCKBR antagonist. Mechanistically, the PI3K/Akt/Bad pathway participates in the pathological process, because gastrin treatment increased phosphorylation of PI3K, Akt and Bad. While in the presence of wortmannin (1 μM), a PI3K inhibitor, the gastrin-induced phosphorylation of Akt after H/R treatment was blocked. Additionally, wortmannin and Akt inhibitor VIII blocked the protective effect of gastrin on viability of HK-2 cells subjected to H/R treatment. These studies reveals that gastrin attenuates kidney I/R injury via a PI3K/Akt/Bad-mediated anti-apoptosis signaling. Thus, gastrin can be considered as a promising drug candidate to prevent AKI.
Collapse
Affiliation(s)
- Chao Liu
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Ken Chen
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Huaixiang Wang
- Department of Lishilu Outpatient, General Hospital of the PLA Rocket Force, Beijing, China
| | - Ye Zhang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Xudong Duan
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yuanzheng Xue
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongye He
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Yu Huang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Zhi Chen
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Hongyong Wang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Institute of Cardiology & Chongqing Key Laboratory of Hypertension Research, Chongqing, China.,Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Asmar A, Cramon PK, Asmar M, Simonsen L, Sorensen CM, Madsbad S, Moro C, Hartmann B, Rehfeld JF, Holst JJ, Hovind P, Jensen BL, Bülow J. Increased oral sodium chloride intake in humans amplifies selectively postprandial GLP-1 but not GIP, CCK, and gastrin in plasma. Physiol Rep 2020; 8:e14519. [PMID: 32770661 PMCID: PMC7413881 DOI: 10.14814/phy2.14519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022] Open
Abstract
Human studies have demonstrated that physiologically relevant changes in circulating glucagon-like peptide-1 (GLP-1) elicit a rapid increase in renal sodium excretion when combined with expansion of the extracellular fluid volume. Other studies support the involvement of various gastrointestinal hormones, e.g., gastrin and cholecystokinin (CCK) in a gut-kidney axis, responsible for a rapid-acting feed-forward natriuretic mechanism. This study was designed to investigate the hypothesis that the postprandial GLP-1 plasma concentration is sensitive to the sodium content in the meal. Under fixed sodium intake for 4 days prior to each experimental day, 10 lean healthy male participants were examined twice in random order after a 12-hr fasting period. Arterial blood samples were collected at 10-20-min intervals for 140 min after 75 grams of oral glucose + 6 grams of oral sodium chloride (NaCl) load versus 75 grams of glucose alone. Twenty-four-hour baseline urinary sodium excretions were similar between study days. Arterial GLP-1 levels increased during both oral glucose loads and were significantly higher at the 40-80 min period during glucose + NaCl compared to glucose alone. The postprandial arterial responses of CCK, gastrin, and glucose-dependent insulinotropic polypeptide as well as glucose, insulin, and C-peptide did not differ between the two study days. Arterial renin, aldosterone, and natriuretic peptides levels did not change within subjects or between study days. Angiotensin II levels were significantly lower at the time GLP-1 was higher (60-80 min) during glucose + NaCl. Sodium intake in addition to a glucose load selectively amplifies the postprandial GLP-1 plasma concentration. Thus, GLP-1 may be part of an acute feed-forward mechanism for natriuresis.
Collapse
Affiliation(s)
- Ali Asmar
- Department of Clinical Physiology, Nuclear Medicine and PET, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
| | - Per K. Cramon
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
| | - Meena Asmar
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
- Department of EndocrinologyOdense University HospitalOdenseDenmark
| | - Lene Simonsen
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
| | | | - Sten Madsbad
- Department of EndocrinologyHvidovre HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
| | - Cedric Moro
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMR 1048Institute of Metabolic and Cardiovascular DiseasesPaul Sabatier UniversityToulouseFrance
| | - Bolette Hartmann
- Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Jens F. Rehfeld
- Department of Clinical Biochemistry, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Jens J. Holst
- Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Peter Hovind
- Department of Clinical Physiology, Nuclear Medicine and PET, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
| | - Boye L. Jensen
- Department of Cardiovascular and Renal ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Jens Bülow
- Department of Clinical Physiology and Nuclear MedicineBispebjerg and Frederiksberg HospitalUniversity Hospital of CopenhagenCopenhagenDenmark
- Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
12
|
Xu P, Gildea JJ, Zhang C, Konkalmatt P, Cuevas S, Bigler Wang D, Tran HT, Jose PA, Felder RA. Stomach gastrin is regulated by sodium via PPAR-α and dopamine D1 receptor. J Mol Endocrinol 2020; 64:53-65. [PMID: 31794424 PMCID: PMC7654719 DOI: 10.1530/jme-19-0053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022]
Abstract
Gastrin, secreted by stomach G cells in response to ingested sodium, stimulates the renal cholecystokinin B receptor (CCKBR) to increase renal sodium excretion. It is not known how dietary sodium, independent of food, can increase gastrin secretion in human G cells. However, fenofibrate (FFB), a peroxisome proliferator-activated receptor-α (PPAR-α) agonist, increases gastrin secretion in rodents and several human gastrin-secreting cells, via a gastrin transcriptional promoter. We tested the following hypotheses: (1.) the sodium sensor in G cells plays a critical role in the sodium-mediated increase in gastrin expression/secretion, and (2.) dopamine, via the D1R and PPAR-α, is involved. Intact human stomach antrum and G cells were compared with human gastrin-secreting gastric and ovarian adenocarcinoma cells. When extra- or intracellular sodium was increased in human antrum, human G cells, and adenocarcinoma cells, gastrin mRNA and protein expression/secretion were increased. In human G cells, the PPAR-α agonist FFB increased gastrin protein expression that was blocked by GW6471, a PPAR-α antagonist, and LE300, a D1-like receptor antagonist. LE300 prevented the ability of FFB to increase gastrin protein expression in human G cells via the D1R, because the D5R, the other D1-like receptor, is not expressed in human G cells. Human G cells also express tyrosine hydroxylase and DOPA decarboxylase, enzymes needed to synthesize dopamine. G cells in the stomach may be the sodium sensor that stimulates gastrin secretion, which enables the kidney to eliminate acutely an oral sodium load. Dopamine, via the D1R, by interacting with PPAR-α, is involved in this process.
Collapse
Affiliation(s)
- Peng Xu
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| | - John J Gildea
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| | - Chi Zhang
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| | - Prasad Konkalmatt
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Santiago Cuevas
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Dora Bigler Wang
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| | - Hanh T Tran
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, School of Medicine & Health Sciences, Washington, District of Columbia, USA
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Robin A Felder
- Department of Pathology, The University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
13
|
Abstract
Purpose of Review Hypertension is related to impaired metabolic homeostasis and can be regarded as a metabolic disorder. This review presents possible mechanisms by which metabolic disorders increase blood pressure (BP) and discusses the importance of the gut as a novel modulator of BP. Recent Findings Obesity and high salt intake are major risk factors for hypertension. There is a hypothesis of “salt-induced obesity”; i.e., high salt intake may tie to obesity. Heightened sympathetic nervous system (SNS) activity, especially in the kidney and brain, increases BP in obese patients. Adipokines, including adiponectin and leptin, and renin-angiotensin-aldosterone system (RAAS) contribute to hypertension. Adiponectin induced by a high-salt diet may decrease sodium/glucose cotransporter (SGLT) 2 expression in the kidney, which results in reducing BP. High salt can change secretions of adipokines and RAAS-related components. Evidence has been accumulating linking the gastrointestinal tract to BP. Glucagon-like peptide-1 (GLP-1) and ghrelin decrease BP in both rodents and humans. The sweet taste receptor in enteroendocrine cells increases SGLT1 expression and stimulates sodium/glucose absorption. Roux-en-Y gastric bypass improves glycemic and BP control due to reducing the activity of SGLT1. Na/H exchanger isoform 3 (NHE3) increases BP by stimulating the intestinal absorption of sodium. Gastrin functions as an intestinal sodium taste sensor and inhibits NHE3 activity. Intestinal mineralocorticoid receptors also regulate sodium absorption and BP due to changing ENaC activity. Gastric sensing of sodium induces natriuresis, and gastric distension increases BP. Changes in the composition and function of gut microbiota contribute to hypertension. A high-salt/fat diet may disrupt the gut barrier, which results in systemic inflammation, insulin resistance, and increased BP. Gut microbiota regulates BP by secreting vasoactive hormones and short-chain fatty acids. BP-lowering effects of probiotics and antibiotics have been reported. Bariatric surgery improves metabolic disorders and hypertension due to increasing GLP-1 secretion, decreasing leptin secretion and SNS activity, and changing gut microbiome composition. Strategies targeting the gastrointestinal system may be therapeutic options for improving metabolic abnormalities and reducing BP in humans. Summary SNS, brain, adipocytes, RAAS, the kidney, the gastrointestinal tract, and microbiota play important roles in regulating BP. Most notably, the gut could be a novel target for treatment of hypertension as a metabolic disorder.
Collapse
|
14
|
Luo H, Chen C, Guo L, Xu Z, Peng X, Wang X, Wang J, Wang N, Li C, Luo X, Wang H, Jose PA, Fu C, Huang Y, Shi W, Zeng C. Exposure to Maternal Diabetes Mellitus Causes Renal Dopamine D 1 Receptor Dysfunction and Hypertension in Adult Rat Offspring. Hypertension 2018; 72:962-970. [PMID: 30354705 PMCID: PMC6207228 DOI: 10.1161/hypertensionaha.118.10908] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/05/2018] [Indexed: 01/01/2023]
Abstract
Epidemiological and experimental studies suggest that maternal diabetes mellitus programs hypertension that is associated with impaired sodium excretion in the adult offspring. However, the underlying mechanisms are not clear. Because dopamine receptor function is involved in the pathogenesis of hypertension, we hypothesized that impaired renal dopamine D1 receptor function is also involved in the hypertension in offspring of maternal diabetes mellitus. Maternal diabetes mellitus was induced by a single intraperitoneal injection of streptozotocin (35 mg/kg) to pregnant Sprague-Dawley rats at day 0 of gestation. Compared with the offspring of mothers injected with citrate buffer (control mother offspring), the diabetic mother offspring (DMO) had increased systolic blood pressure and impaired D1 receptor-mediated diuresis and natriuresis, accompanied by increased renal PKC (protein kinase C) expression and activity, GRK-2 (G protein-coupled receptor kinase-2) expression, D1 receptor phosphorylation, D1 receptor/Gαs uncoupling, and loss of D1 receptor-mediated inhibition of Na+-K+-ATPase activity in renal proximal tubule cells from DMO. Inhibition of PKC reduced the increased GRK-2 expression and normalized D1 receptor function in primary cultures of renal proximal tubule cells from DMO. In addition, DMO, relative to control mother offspring, in vivo, had increased oxidative stress, indicated by decreased renal glutathione and increased renal malondialdehyde and urine 8-isoprostane. Normalization of oxidative stress with tempol also normalized the renal D1 receptor phosphorylation, D1 receptor-mediated diuresis and natriuresis, and blood pressure in DMO. Our present study indicates that maternal diabetes mellitus-programed hypertension in the offspring is caused by impaired renal D1 receptor function because of oxidative stress that is mediated by increased PKC-GRK-2 activity.
Collapse
Affiliation(s)
- Hao Luo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Caiyu Chen
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Li Guo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Zaicheng Xu
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoyu Peng
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xinquan Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Jialiang Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Na Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chuanwei Li
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoli Luo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hongyong Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Pedro A. Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Chunjiang Fu
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu Huang
- Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Sha Tin, Hong Kong, China
| | - Weibin Shi
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
15
|
Takahashi-Iwanaga H, Kimura S, Konno K, Watanabe M, Iwanaga T. Intrarenal signaling mediated by CCK plays a role in salt intake-induced natriuresis. Am J Physiol Renal Physiol 2017; 313:F20-F29. [DOI: 10.1152/ajprenal.00539.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 11/22/2022] Open
Abstract
The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.
Collapse
Affiliation(s)
| | - Shunsuke Kimura
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihiko Iwanaga
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
16
|
The Effect of Salt Intake and Potassium Supplementation on Serum Gastrin Levels in Chinese Adults: A Randomized Trial. Nutrients 2017; 9:nu9040389. [PMID: 28420122 PMCID: PMC5409728 DOI: 10.3390/nu9040389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/31/2017] [Accepted: 04/13/2017] [Indexed: 01/01/2023] Open
Abstract
Excess dietary salt is strongly correlated with cardiovascular disease, morbidity, and mortality. Conversely, potassium likely elicits favorable effects against cardiovascular disorders. Gastrin, which is produced by the G-cells of the stomach and duodenum, can increase renal sodium excretion and regulate blood pressure by acting on the cholecystokinin B receptor. The aim of our study was to assess the effects of altered salt and potassium supplementation on serum gastrin levels in humans. A total of 44 subjects (38–65 years old) were selected from a rural community in northern China. All subjects were sequentially maintained on a relatively low-salt diet for 7 days (3.0 g/day of NaCl), a high-salt diet for 7 days (18.0 g/day of NaCl), and then a high-salt diet supplemented with potassium for another 7 days (18.0 g/day of NaCl + 4.5 g/day of KCl). The high-salt intake significantly increased serum gastrin levels (15.3 ± 0.3 vs. 17.6 ± 0.3 pmol/L). This phenomenon was alleviated through potassium supplementation (17.6 ± 0.3 vs. 16.5 ± 0.4 pmol/L). Further analyses revealed that serum gastrin was positively correlated with 24 h urinary sodium excretion (r = 0.476, p < 0.001). By contrast, gastrin level was negatively correlated with blood pressure in all dietary interventions (r = −0.188, p = 0.031). The present study indicated that variations in dietary salt and potassium supplementation affected the serum gastrin concentrations in the Chinese subjects.
Collapse
|
17
|
Affiliation(s)
- Jian Yang
- Department of Nutrition, Daping Hospital, The Third Military Medical University, Chongqing, China.,Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Pedro A Jose
- Division of Renal Disease & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Jiang X, Zhang Y, Yang Y, Yang J, Asico LD, Chen W, Felder RA, Armando I, Jose PA, Yang Z. Gastrin stimulates renal dopamine production by increasing the renal tubular uptake of l-DOPA. Am J Physiol Endocrinol Metab 2017; 312:E1-E10. [PMID: 27780818 PMCID: PMC5283882 DOI: 10.1152/ajpendo.00116.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 10/18/2016] [Accepted: 10/23/2016] [Indexed: 01/02/2023]
Abstract
Gastrin is a peptide hormone that is involved in the regulation of sodium balance and blood pressure. Dopamine, which is also involved in the regulation of sodium balance and blood pressure, directly or indirectly interacts with other blood pressure-regulating hormones, including gastrin. This study aimed to determine the mechanisms of the interaction between gastrin and dopamine and tested the hypothesis that gastrin produced in the kidney increases renal dopamine production to keep blood pressure within the normal range. We show that in human and mouse renal proximal tubule cells (hRPTCs and mRPTCs, respectively), gastrin stimulates renal dopamine production by increasing the cellular uptake of l-DOPA via the l-type amino acid transporter (LAT) at the plasma membrane. The uptake of l-DOPA in RPTCs from C57Bl/6J mice is lower than in RPTCs from normotensive humans. l-DOPA uptake in renal cortical slices is also lower in salt-sensitive C57Bl/6J than in salt-resistant BALB/c mice. The deficient renal cortical uptake of l-DOPA in C57Bl/6J mice may be due to decreased LAT-1 activity that is related to its decreased expression at the plasma membrane, relative to BALB/c mice. We also show that renal-selective silencing of Gast by the renal subcapsular injection of Gast siRNA in BALB/c mice decreases renal dopamine production and increases blood pressure. These results highlight the importance of renal gastrin in stimulating renal dopamine production, which may give a new perspective in the prevention and treatment of hypertension.
Collapse
MESH Headings
- Amino Acid Transport System y+L/drug effects
- Amino Acid Transport System y+L/metabolism
- Animals
- Blood Pressure/drug effects
- Blood Pressure/physiology
- Cells, Cultured
- Dopamine/biosynthesis
- Dopamine/urine
- Down-Regulation
- Gastrins/genetics
- Gastrins/metabolism
- Gastrins/pharmacology
- Gene Silencing
- Humans
- Immunoblotting
- Kidney/drug effects
- Kidney/metabolism
- Kidney Cortex/drug effects
- Kidney Cortex/metabolism
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Levodopa/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- RNA, Small Interfering
- Real-Time Polymerase Chain Reaction
- Receptor, Cholecystokinin B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical, Beijing, China
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yanrong Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical, Beijing, China
| | - Yu Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
- Department of Cardiology, Daping Hospital, The Third Military Medical University and Chongqing Institute of Cardiology, Chongqing, China
| | - Laureano D Asico
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei Chen
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical, Beijing, China
| | - Robin A Felder
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Medicine, Division of Kidney Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC; and
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical, Beijing, China;
| |
Collapse
|
19
|
Chen Y, Wu S, Tian Y, Kong J. Phosphorylation and subcellular localization of Na+/H+ exchanger isoform 3 (NHE3) are associated with altered gallbladder absorptive function after formation of cholesterol gallstones. J Physiol Biochem 2016; 73:133-139. [DOI: 10.1007/s13105-016-0533-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 10/21/2016] [Indexed: 01/14/2023]
|
20
|
Afsar B, Vaziri ND, Aslan G, Tarim K, Kanbay M. Gut hormones and gut microbiota: implications for kidney function and hypertension. ACTA ACUST UNITED AC 2016; 10:954-961. [PMID: 27865823 DOI: 10.1016/j.jash.2016.10.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Increased blood pressure (BP) and chronic kidney disease are two leading risk factors for cardiovascular disease. Increased sodium intake is one of the most important risk factors for development of hypertension. Recent data have shown that gut influences kidney function and BP by variety of mechanisms. Various hormones and peptides secreted from gut such as gastrin, glucocorticoids, Glucagon-like peptide-1 impact on kidney function and BP especially influencing sodium absorption from gut. These findings stimulate scientist to find new therapeutic options such as tenapanor for treatment of hypertension by blocking sodium absorption from gut. The gastrointestinal tract is also occupied by a huge community of microbes (microbiome) that under normal condition has a symbiotic relationship with the host. Alterations in the structure and function of the gut microbiota have been shown to play a key role in the pathogenesis and complications of numerous diseases including hypertension. Based on these data, in this review, we provide a summary of the available data on the role of gut and gut microbiota in regulation of BP and kidney function.
Collapse
Affiliation(s)
- Baris Afsar
- Division of Nephrology, Department of Medicine, Konya Numune State Hospital, Konya, Turkey
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Department of Medicine, Schools of Medicine and Biological Science, University of California, Irvine, CA, USA
| | - Gamze Aslan
- Department of Cardiology, Koc University School of Medicine, Istanbul, Turkey
| | - Kayhan Tarim
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
21
|
Sohn M, Kim K, Uddin MJ, Lee G, Hwang I, Kang H, Kim H, Lee JH, Ha H. Delayed treatment with fenofibrate protects against high-fat diet-induced kidney injury in mice: the possible role of AMPK autophagy. Am J Physiol Renal Physiol 2016; 312:F323-F334. [PMID: 27465995 DOI: 10.1152/ajprenal.00596.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/22/2016] [Indexed: 11/22/2022] Open
Abstract
Fenofibrate activates not only peroxisome proliferator-activated receptor-α (PPARα) but also adenosine monophosphate-activated protein kinase (AMPK). AMPK-mediated cellular responses protect kidney from high-fat diet (HFD)-induced injury, and autophagy resulting from AMPK activation has been regarded as a stress-response mechanism. Thus the present study examined the role of AMPK and autophagy in the renotherapeutic effects of fenofibrate. C57BL/6J mice were divided into three groups: normal diet (ND), HFD, and HFD + fenofibrate (HFD + FF). Fenofibrate was administered 4 wk after the initiation of the HFD when renal injury was initiated. Mouse proximal tubule cells (mProx24) were used to clarify the role of AMPK. Feeding mice with HFD for 12 wk induced insulin resistance and kidney injury such as albuminuria, glomerulosclerosis, tubular injury, and inflammation, which were effectively inhibited by fenofibrate. In addition, fenofibrate treatment resulted in the activation of renal AMPK, upregulation of fatty acid oxidation (FAO) enzymes and antioxidants, and induction of autophagy in the HFD mice. In mProx24 cells, fenofibrate activated AMPK in a concentration-dependent manner, upregulated FAO enzymes and antioxidants, and induced autophagy, all of which were inhibited by treatment of compound C, an AMPK inhibitor. Fenofibrate-induced autophagy was also significantly blocked by AMPKα1 siRNA but not by PPARα siRNA. Collectively, these results demonstrate that delayed treatment with fenofibrate has a therapeutic effect on HFD-induced kidney injury, at least in part, through the activation of AMPK and induction of subsequent downstream effectors: autophagy, FAO enzymes, and antioxidants.
Collapse
Affiliation(s)
- Minji Sohn
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Keumji Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Gayoung Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Inah Hwang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hyeji Kang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hyunji Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Jung Hwa Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
22
|
Affiliation(s)
- Pedro A Jose
- From the Departments of Medicine and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC (P.A.J.); Department of Pathology, The University of Virginia, Charlottesville (R.A.F.); Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Centre, Peking Union Medical College, Beijing, P.R. China (Z.Y.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing, P.R. China (C.Z.); and Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.).
| | - Robin A Felder
- From the Departments of Medicine and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC (P.A.J.); Department of Pathology, The University of Virginia, Charlottesville (R.A.F.); Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Centre, Peking Union Medical College, Beijing, P.R. China (Z.Y.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing, P.R. China (C.Z.); and Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.)
| | - Zhiwei Yang
- From the Departments of Medicine and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC (P.A.J.); Department of Pathology, The University of Virginia, Charlottesville (R.A.F.); Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Centre, Peking Union Medical College, Beijing, P.R. China (Z.Y.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing, P.R. China (C.Z.); and Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.)
| | - Chunyu Zeng
- From the Departments of Medicine and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC (P.A.J.); Department of Pathology, The University of Virginia, Charlottesville (R.A.F.); Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Centre, Peking Union Medical College, Beijing, P.R. China (Z.Y.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing, P.R. China (C.Z.); and Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.)
| | - Gilbert M Eisner
- From the Departments of Medicine and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC (P.A.J.); Department of Pathology, The University of Virginia, Charlottesville (R.A.F.); Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Centre, Peking Union Medical College, Beijing, P.R. China (Z.Y.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing, P.R. China (C.Z.); and Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.)
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Hypertension, which is present in about one quarter of the world's population, is responsible for about 41% of the number one cause of death - cardiovascular disease. Not included in these statistics is the effect of sodium intake on blood pressure, even though an increase or a marked decrease in sodium intake can increase blood pressure. This review deals with the interaction of gut microbiota and the kidney with genetics and epigenetics in the regulation of blood pressure and salt sensitivity. RECENT FINDINGS The abundance of the gut microbes, Firmicutes and Bacteroidetes, is associated with increased blood pressure in several models of hypertension, including the spontaneously hypertensive and Dahl salt-sensitive rats. Decreasing gut microbiota by antibiotics can increase or decrease blood pressure that is influenced by genotype. The biological function of probiotics may also be a consequence of epigenetic modification, related, in part, to microRNA. Products of the fermentation of nutrients by gut microbiota can influence blood pressure by regulating expenditure of energy, intestinal metabolism of catecholamines, and gastrointestinal and renal ion transport, and thus, salt sensitivity. SUMMARY The beneficial or deleterious effect of gut microbiota on blood pressure is a consequence of several variables, including genetics, epigenetics, lifestyle, and intake of antibiotics. These variables may influence the ultimate level of blood pressure and control of hypertension.
Collapse
|
24
|
Liu T, Konkalmatt PR, Yang Y, Jose PA. Gastrin decreases Na+,K+-ATPase activity via a PI 3-kinase- and PKC-dependent pathway in human renal proximal tubule cells. Am J Physiol Endocrinol Metab 2016; 310:E565-71. [PMID: 26786777 PMCID: PMC4824137 DOI: 10.1152/ajpendo.00360.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/08/2016] [Indexed: 12/31/2022]
Abstract
The natriuretic effect of gastrin suggests a role in the coordinated regulation of sodium balance by the gastrointestinal tract and the kidney. The renal molecular targets and signal transduction pathways for such an effect of gastrin are largely unknown. Recently, we reported that gastrin induces NHE3 phosphorylation and internalization via phosphatidylinositol (PI) 3-kinase and PKCα. In this study, we show that gastrin induced the phosphorylation of human Na(+),K(+)-ATPase at serine 16, resulting in its endocytosis via Rab5 and Rab7 endosomes. The gastrin-stimulated phosphorylation of Na(+),K(+)-ATPase was dependent on PI 3-kinase because the phosphorylation was blocked by the PI 3-kinase inhibitor wortmannin. The phosphorylation of Na(+),K(+)-ATPase was also blocked by chelerythrine, a pan-PKC inhibitor, Gö-6976, a conventional PKC (cPKC) inhibitor, and BAPTA-AM, an intracellular calcium chelator, suggesting the importance of cPKC and intracellular calcium in the gastrin signaling pathway. The gastrin-mediated phosphorylation of Na(+),K(+)-ATPase was also inhibited by U-73122, a phospholipase C (PLC) inhibitor. These results suggest that gastrin regulates sodium hydrogen exchanger and pump in renal proximal tubule cells at the apical and basolateral membranes.
Collapse
Affiliation(s)
- Tianbing Liu
- Center for Molecular Physiology Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Prasad R Konkalmatt
- Departments of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, Maryland; Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC; and
| | - Yu Yang
- Center for Molecular Physiology Research, Children's Research Institute, Children's National Medical Center, Washington, DC; Departments of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
25
|
Jose PA, Yang Z, Zeng C, Felder RA. The importance of the gastrorenal axis in the control of body sodium homeostasis. Exp Physiol 2016; 101:465-70. [PMID: 26854262 DOI: 10.1113/ep085286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022]
Abstract
NEW FINDINGS What is the topic of this review? Sensing the amount of ingested sodium is one mechanism by which sodium balance is regulated. This review describes the role of gastrin in the cross-talk between the stomach and the kidney following the ingestion of sodium. What advances does it highlight? Neural mechanisms and several gut hormones, including cholecystokinin and uroguanylin, have been suggested to mediate the natriuresis after an oral sodium load. It is proposed that gastrin produced by G-cells via its receptor, cholecystokinin B receptor, interacts with renal D1 -like dopamine receptors to increase renal sodium excretion. Hypertension develops with chronically increased sodium intake when sodium that accumulates in the body can no longer be sequestered, extracellular fluid volume is expanded, and compensatory neural, hormonal and pressure-natriuresis mechanisms fail. Sensing the amount of ingested sodium, by the stomach, is one mechanism by which sodium balance is regulated. The natriuresis following the ingestion of a certain amount of sodium may be due to an enterokine, gastrin, secreted by G-cells in the stomach and duodenum and released into the circulation. Circulating gastrin levels are 10- to 20-fold higher than those for cholecystokinin. Of all the gut hormones circulating in the plasma, gastrin is the one that is reabsorbed to the greatest extent by renal tubules. Gastrin, via its receptor, the cholecystokinin type B receptor (CCKBR), is natriuretic in mammals, including humans, by inhibition of renal sodium transport. Germline deletion of gastrin (Gast) or Cckbr gene in mice causes salt-sensitive hypertension. Selective silencing of Gast in the stomach and duodenum impairs the ability to excrete an oral sodium load and also increases blood pressure. Thus, the gastrorenal axis, mediated by gastrin, can complement pronatriuretic hormones, such as dopamine, to increase sodium excretion after an oral sodium load. These studies in mice may be translatable to humans because the chromosomal loci of CCKBR and GAST are linked to human essential hypertension. Understanding the role of genes in the regulation of renal function and blood pressure may lead to the tailoring of antihypertensive treatment based on genetic make-up.
Collapse
Affiliation(s)
- Pedro A Jose
- Department of Medicine, The George Washington University School of Medicine, Washington, DC, USA.,Department of Physiology, The George Washington University School of Medicine, Washington, DC, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medicine Centre, Peking Union Medical College, Beijing, PR China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, PR China
| | - Robin A Felder
- Department of Pathology, The University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
26
|
Jiang X, Chen W, Liu X, Wang Z, Liu Y, Felder RA, Gildea JJ, Jose PA, Qin C, Yang Z. The Synergistic Roles of Cholecystokinin B and Dopamine D5 Receptors on the Regulation of Renal Sodium Excretion. PLoS One 2016; 11:e0146641. [PMID: 26751218 PMCID: PMC4709046 DOI: 10.1371/journal.pone.0146641] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/21/2015] [Indexed: 01/07/2023] Open
Abstract
Renal dopamine D1-like receptors (D1R and D5R) and the gastrin receptor (CCKBR) are involved in the maintenance of sodium homeostasis. The D1R has been found to interact synergistically with CCKBR in renal proximal tubule (RPT) cells to promote natriuresis and diuresis. D5R, which has a higher affinity for dopamine than D1R, has some constitutive activity. Hence, we sought to investigate the interaction between D5R and CCKBR in the regulation of renal sodium excretion. In present study, we found D5R and CCKBR increase each other’s expression in a concentration- and time-dependent manner in the HK-2 cell, the specificity of which was verified in HEK293 cells heterologously expressing both human D5R and CCKBR and in RPT cells from a male normotensive human. The specificity of D5R in the D5R and CCKBR interaction was verified further using a selective D5R antagonist, LE-PM436. Also, D5R and CCKBR colocalize and co-immunoprecipitate in BALB/c mouse RPTs and human RPT cells. CCKBR protein expression in plasma membrane-enriched fractions of renal cortex (PMFs) is greater in D5R-/- mice than D5R+/+ littermates and D5R protein expression in PMFs is also greater in CCKBR-/- mice than CCKBR+/+ littermates. High salt diet, relative to normal salt diet, increased the expression of CCKBR and D5R proteins in PMFs. Disruption of CCKBR in mice caused hypertension and decreased sodium excretion. The natriuresis in salt-loaded BALB/c mice was decreased by YF476, a CCKBR antagonist and Sch23390, a D1R/D5R antagonist. Furthermore, the natriuresis caused by gastrin was blocked by Sch23390 while the natriuresis caused by fenoldopam, a D1R/D5R agonist, was blocked by YF476. Taken together, our findings indicate that CCKBR and D5R synergistically interact in the kidney, which may contribute to the maintenance of normal sodium balance following an increase in sodium intake.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
| | - Wei Chen
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
| | - Xing Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
| | - Zihao Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
| | - Yunpeng Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
| | - Robin A. Felder
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - John J. Gildea
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Pedro A. Jose
- Division of Nephrology, Departments of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail: (PAJ); (CQ); (ZWY)
| | - Chuan Qin
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
- * E-mail: (PAJ); (CQ); (ZWY)
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China
- CollaborativeInnovation Center for Cardiovascular Disorders, Beijing, P. R. China
- * E-mail: (PAJ); (CQ); (ZWY)
| |
Collapse
|
27
|
Muscella A, Vetrugno C, Calabriso N, Cossa LG, De Pascali SA, Fanizzi FP, Marsigliante S. [Pt(O,O'-acac)(γ-acac)(DMS)] alters SH-SY5Y cell migration and invasion by the inhibition of Na+/H+ exchanger isoform 1 occurring through a PKC-ε/ERK/mTOR Pathway. PLoS One 2014; 9:e112186. [PMID: 25372487 PMCID: PMC4221608 DOI: 10.1371/journal.pone.0112186] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/13/2014] [Indexed: 11/19/2022] Open
Abstract
We previously showed that [Pt(O,O'-acac)(γ-acac)(DMS)] ([Pt(acac)2(DMS)]) exerted substantial cytotoxic effects in SH-SY5Y neuroblastoma cells, and decreased metalloproteases (MMPs) production and cells migration in MCF-7 breast cancer cells. The ubiquitously distributed sodium-hydrogen antiporter 1 (NHE1) is involved in motility and invasion of many solid tumours. The present study focuses on the effects of [Pt(acac)2(DMS)] in SH-SY5Y cell migration and also on the possibility that NHE1 may be involved in such effect. After sublethal [Pt(acac)2(DMS)] treatment cell migration was examined by wounding assay and cell invasion by transwell assay. NHE1 activity was measured in BCECF-loaded SH-SY5Y as the rate of Na+-dependent intracellular pH recovery in response to an acute acid pulse. Gelatin zymography for MMP-2/9 activities, Western blottings of MMPs, MAPKs, mTOR, S6 and PKCs and small interfering RNAs to PKC-ε/-δ mRNA were performed. Sublethal concentrations of [Pt(acac)2(DMS)] decreases NHE1 activity, inhibits cell migration and invasion and decreases expression and activity of MMP-2 and -9. [Pt(acac)2(DMS)] administered to SH-SY5Y cells provokes the increment of ROS, generated by NADPH oxidase, responsible for the PKC-ε and PKC-δ activation. Whilst PKC-δ activates p38/MAPK, responsible for the inhibition of MMP-2 and -9 secretion, PKC-ε activates a pathway made of ERK1/2, mTOR and S6K responsible for the inhibition of NHE1 activity and cell migration. In conclusion, we have shown a drastic impairment in tumour cell metastatization in response to inhibition of NHE1 and MMPs activities by [Pt(acac)2(DMS)] occurring through a novel mechanism mediated by PKC-δ/-ε activation.
Collapse
Affiliation(s)
- Antonella Muscella
- Cell Pathology Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
| | - Carla Vetrugno
- Neuropathology Unit, Istituto di Neurologia sperimentale e Divisione di Neuroscienze, Istituto Scientifico IRCCS San Raffaele (sezione di Lecce), Milano, Italy
| | - Nadia Calabriso
- Cell Physiology Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
| | - Luca Giulio Cossa
- Cell Physiology Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
| | - Sandra Angelica De Pascali
- General and Inorganic Chemistry Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
| | - Francesco Paolo Fanizzi
- General and Inorganic Chemistry Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
| | - Santo Marsigliante
- Cell Physiology Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Salento University, Lecce, Italy
- * E-mail:
| |
Collapse
|
28
|
Crajoinas RO, Pessoa TD, Rodrigues MV, Malnic G, Girardi ACC. Changes in the activity and expression of protein phosphatase-1 accompany the differential regulation of NHE3 before and after the onset of hypertension in spontaneously hypertensive rats. Acta Physiol (Oxf) 2014; 211:395-408. [PMID: 24666699 DOI: 10.1111/apha.12288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 11/18/2013] [Accepted: 03/18/2014] [Indexed: 12/14/2022]
Abstract
AIM The Na(+) /H(+) exchanger NHE3 activity decreases in the proximal tubule of spontaneously hypertensive rats (SHRs) as blood pressure increases, and this reduction is correlated with higher NHE3 phosphorylation levels at the PKA consensus site serine 552. This study tested the hypothesis that this lowered NHE3 activity is associated with an increase in PKA activity and expression, and/or a decrease in protein phosphatase-1 (PP1) activity and expression. METHODS Proximal tubule NHE3 activity was measured as the rate of bicarbonate reabsorption by stationary microperfusion. NHE3 phosphorylation and protein expression were determined by immunoblotting. PKA and PP1 activities were determined using specific substrates under optimal enzymatic conditions. RESULTS The PKA activator, 6-MB-cAMP, increased the phosphorylation levels of NHE3 at serine 552 in the renal cortex; this increase happens to a much greater extent in young pre-hypertensive SHRs (Y-SHRs) compared to adult SHRs with established hypertension (A-SHRs). Likewise, the inhibitory effect of 6-MB-cAMP on NHE3 transport activity was much more pronounced in the proximal tubules of Y-SHRs than in those of A-SHRs. Renal cortical activity of PKA was not significantly different between Y-SHRs and A-SHRs. On the other hand, Y-SHRs exhibited higher protein phosphatase 1 (PP1) activity, and their expression of the PP1 catalytic subunit PP1α in the renal cortex was also higher than in A-SHRs. CONCLUSION Collectively, these results support the idea that the lower NHE3 transport activity and higher phosphorylation occurring after the development of hypertension in SHRs are due, at least in part, to reduced PP1-mediated dephosphorylation of NHE3 at serine 552.
Collapse
Affiliation(s)
- R. O. Crajoinas
- Laboratory of Genetics and Molecular Cardiology; Heart Institute (InCor); University of São Paulo Medical School; São Paulo Brazil
| | - T. D. Pessoa
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - M. V. Rodrigues
- Laboratory of Genetics and Molecular Cardiology; Heart Institute (InCor); University of São Paulo Medical School; São Paulo Brazil
| | - G. Malnic
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - A. C. C. Girardi
- Laboratory of Genetics and Molecular Cardiology; Heart Institute (InCor); University of São Paulo Medical School; São Paulo Brazil
| |
Collapse
|
29
|
Hendus-Altenburger R, Kragelund BB, Pedersen SF. Structural dynamics and regulation of the mammalian SLC9A family of Na⁺/H⁺ exchangers. CURRENT TOPICS IN MEMBRANES 2014; 73:69-148. [PMID: 24745981 DOI: 10.1016/b978-0-12-800223-0.00002-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mammalian Na⁺/H⁺ exchangers of the SLC9A family are widely expressed and involved in numerous essential physiological processes. Their primary function is to mediate the 1:1 exchange of Na⁺ for H⁺ across the membrane in which they reside, and they play central roles in regulation of body, cellular, and organellar pH. Their function is tightly regulated through mechanisms involving interactions with multiple protein and lipid-binding partners, phosphorylations, and other posttranslational modifications. Biochemical and mutational analyses indicate that the SLC9As have a short intracellular N-terminus, 12 transmembrane (TM) helices necessary and sufficient for ion transport, and a C-terminal cytoplasmic tail region with essential regulatory roles. No high-resolution structures of the SLC9As exist; however, models based on crystal structures of the bacterial NhaAs support the 12 TM organization and suggest that TMIV and XI may form a central part of the ion-translocation pathway, whereas pH sensing may involve TMII, TMIX, and several intracellular loops. Similar to most ion transporters studied, SLC9As likely exist as coupled dimers in the membrane, and this appears to be important for the well-studied cooperativity of H⁺ binding. The aim of this work is to summarize and critically discuss the currently available evidence on the structural dynamics, regulation, and binding partner interactions of SLC9As, focusing in particular on the most widely studied isoform, SLC9A1/NHE1. Further, novel bioinformatic and structural analyses are provided that to some extent challenge the existing paradigm on how ions are transported by mammalian SLC9As.
Collapse
Affiliation(s)
- Ruth Hendus-Altenburger
- Section for Biomolecular Sciences, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Section for Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Birthe B Kragelund
- Section for Biomolecular Sciences, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stine Falsig Pedersen
- Section for Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|