1
|
Bourgeois S, Houillier P. State of knowledge on ammonia handling by the kidney. Pflugers Arch 2024; 476:517-531. [PMID: 38448728 PMCID: PMC11006756 DOI: 10.1007/s00424-024-02940-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
The disposal of ammonia, the main proton buffer in the urine, is important for acid-base homeostasis. Renal ammonia excretion is the predominant contributor to renal net acid excretion, both under basal condition and in response to acidosis. New insights into the mechanisms of renal ammonia production and transport have been gained in the past decades. Ammonia is the only urinary solute known to be produced in the kidney and selectively transported through the different parts of the nephron. Both molecular forms of total ammonia, NH3 and NH4+, are transported by specific proteins. Proximal tubular ammoniagenesis and the activity of these transport processes determine the eventual fate of total ammonia produced and excreted by the kidney. In this review, we summarized the state of the art of ammonia handling by the kidney and highlighted the newest processes described in the last decade.
Collapse
Affiliation(s)
- Soline Bourgeois
- Institut of Physiology, University of Zurich, Zurich, Switzerland.
| | - Pascal Houillier
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- Centre National de La Recherche Scientifique (CNRS), EMR 8228, Paris, France
| |
Collapse
|
2
|
Wang Q, Guan K, Lv Y, Zhang Y, Yu Z, Kan Q. Disturbance of hepatocyte growth and metabolism in a hyperammonemia microenvironment. Arch Biochem Biophys 2021; 716:109109. [PMID: 34932992 DOI: 10.1016/j.abb.2021.109109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND We found through previous research that hyperammonemia can cause secondary liver damage. However, whether hepatocytes are target cells of ammonia toxicity and whether hyperammonemia affects hepatocyte metabolism remain unknown. AIMS The purpose of the current study is to examine whether the hepatocyte is a specific target cell of ammonia toxicity and whether hyperammonemia can interfere with hepatocyte metabolism. METHODS Cell viability and apoptosis were analyzed in primary hepatocytes and other cells that had been exposed to ammonium chloride. Western blotting was adopted to examine the expression of proteins related to ammonia transport. We also established a metabolomics method based on gas chromatography-mass spectrometry to understand the characteristics of the hepatocyte metabolic spectrum in a hyperammonemia microenvironment, to screen and identify differential metabolites, and to determine the differential metabolic pathway. Different technologies were used to verify the differential metabolic pathways. RESULTS Hepatocytes are target cells of ammonia toxicity. The mechanism is related to the ammonia transporter. Hyperammonemia interferes with hepatocyte metabolism, which leads to TCA cycle, urea cycle, and RNA synthesis disorder. CONCLUSIONS This study demonstrates that hepatocyte growth and metabolism are disturbed in a hyperammonemia microenvironment, which further deteriorates hepatocyte function.
Collapse
Affiliation(s)
- Qiongye Wang
- Departments of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kelei Guan
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanjun Lv
- Department of Respiratory. the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingxuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery. the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Departments of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Quancheng Kan
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
3
|
Abstract
Acid-base homeostasis is critical to maintenance of normal health. Renal ammonia excretion is the quantitatively predominant component of renal net acid excretion, both under basal conditions and in response to acid-base disturbances. Although titratable acid excretion also contributes to renal net acid excretion, the quantitative contribution of titratable acid excretion is less than that of ammonia under basal conditions and is only a minor component of the adaptive response to acid-base disturbances. In contrast to other urinary solutes, ammonia is produced in the kidney and then is selectively transported either into the urine or the renal vein. The proportion of ammonia that the kidney produces that is excreted in the urine varies dramatically in response to physiological stimuli, and only urinary ammonia excretion contributes to acid-base homeostasis. As a result, selective and regulated renal ammonia transport by renal epithelial cells is central to acid-base homeostasis. Both molecular forms of ammonia, NH3 and NH4+, are transported by specific proteins, and regulation of these transport processes determines the eventual fate of the ammonia produced. In this review, we discuss these issues, and then discuss in detail the specific proteins involved in renal epithelial cell ammonia transport.
Collapse
Affiliation(s)
- I David Weiner
- Division of Nephrology, Hypertension and Renal Transplantation, University of Florida College of Medicine, Gainesville, Florida; and Nephrology and Hypertension Section, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Jill W Verlander
- Division of Nephrology, Hypertension and Renal Transplantation, University of Florida College of Medicine, Gainesville, Florida; and Nephrology and Hypertension Section, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| |
Collapse
|
4
|
Weiner ID, Verlander JW. Ammonia transport in the kidney by Rhesus glycoproteins. Am J Physiol Renal Physiol 2014; 306:F1107-20. [PMID: 24647713 PMCID: PMC4024734 DOI: 10.1152/ajprenal.00013.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/14/2014] [Indexed: 12/26/2022] Open
Abstract
Renal ammonia metabolism is a fundamental element of acid-base homeostasis, comprising a major component of both basal and physiologically altered renal net acid excretion. Over the past several years, a fundamental change in our understanding of the mechanisms of renal epithelial cell ammonia transport has occurred, replacing the previous model which was based upon diffusion equilibrium for NH3 and trapping of NH4(+) with a new model in which specific and regulated transport of both NH3 and NH4(+) across renal epithelial cell membranes via specific membrane proteins is required for normal ammonia metabolism. A major advance has been the recognition that members of a recently recognized transporter family, the Rhesus glycoprotein family, mediate critical roles in renal and extrarenal ammonia transport. The erythroid-specific Rhesus glycoprotein, Rh A Glycoprotein (Rhag), was the first Rhesus glycoprotein recognized as an ammonia-specific transporter. Subsequently, the nonerythroid Rh glycoproteins, Rh B Glycoprotein (Rhbg) and Rh C Glycoprotein (Rhcg), were cloned and identified as ammonia transporters. They are expressed in specific cell populations and membrane domains in distal renal epithelial cells, where they facilitate ammonia secretion. In this review, we discuss the distribution of Rhbg and Rhcg in the kidney, the regulation of their expression and activity in physiological disturbances, the effects of genetic deletion on renal ammonia metabolism, and the molecular mechanisms of Rh glycoprotein-mediated ammonia transport.
Collapse
Affiliation(s)
- I David Weiner
- Nephrology and Hypertension Section, North Florida/South Georgia Veterans Health System, Gainesville Florida; and Division of Nephrology, Hypertension, and Transplantation, University of Florida College of Medicine, Gainesville, Florida
| | - Jill W Verlander
- Nephrology and Hypertension Section, North Florida/South Georgia Veterans Health System, Gainesville Florida; and
| |
Collapse
|
5
|
Lee HW, Verlander JW, Handlogten ME, Han KH, Weiner ID. Effect of collecting duct-specific deletion of both Rh B Glycoprotein (Rhbg) and Rh C Glycoprotein (Rhcg) on renal response to metabolic acidosis. Am J Physiol Renal Physiol 2013; 306:F389-400. [PMID: 24338819 DOI: 10.1152/ajprenal.00176.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Rhesus (Rh) glycoproteins, Rh B and Rh C Glycoprotein (Rhbg and Rhcg, respectively), are ammonia-specific transporters expressed in renal distal nephron and collecting duct sites that are necessary for normal rates of ammonia excretion. The purpose of the current studies was to determine the effect of their combined deletion from the renal collecting duct (CD-Rhbg/Rhcg-KO) on basal and acidosis-stimulated acid-base homeostasis. Under basal conditions, urine pH and ammonia excretion and serum HCO3(-) were similar in control (C) and CD-Rhbg/Rhcg-KO mice. After acid-loading for 7 days, CD-Rhbg/Rhcg-KO mice developed significantly more severe metabolic acidosis than did C mice. Acid loading increased ammonia excretion, but ammonia excretion increased more slowly in CD-Rhbg/Rhcg-KO and it was significantly less than in C mice on days 1-5. Urine pH was significantly more acidic in CD-Rhbg/Rhcg-KO mice on days 1, 3, and 5 of acid loading. Metabolic acidosis increased phosphenolpyruvate carboxykinase (PEPCK) and Na(+)/H(+) exchanger NHE-3 and decreased glutamine synthetase (GS) expression in both genotypes, and these changes were significantly greater in CD-Rhbg/Rhcg-KO than in C mice. We conclude that 1) Rhbg and Rhcg are critically important in the renal response to metabolic acidosis; 2) the significantly greater changes in PEPCK, NHE-3, and GS expression in acid-loaded CD-Rhbg/Rhcg-KO compared with acid-loaded C mice cause the role of Rhbg and Rhcg to be underestimated quantitatively; and 3) in mice with intact Rhbg and Rhcg expression, metabolic acidosis does not induce maximal changes in PEPCK, NHE-3, and GS expression despite the presence of persistent metabolic acidosis.
Collapse
Affiliation(s)
- Hyun-Wook Lee
- Div. of Nephrology, Hypertension, and Transplantation, Univ. of Florida College of Medicine, PO Box 100224, Gainesville, FL 32610.
| | | | | | | | | |
Collapse
|
6
|
Abstract
Renal ammonia metabolism and transport mediates a central role in acid-base homeostasis. In contrast to most renal solutes, the majority of renal ammonia excretion derives from intrarenal production, not from glomerular filtration. Renal ammoniagenesis predominantly results from glutamine metabolism, which produces 2 NH4(+) and 2 HCO3(-) for each glutamine metabolized. The proximal tubule is the primary site for ammoniagenesis, but there is evidence for ammoniagenesis by most renal epithelial cells. Ammonia produced in the kidney is either excreted into the urine or returned to the systemic circulation through the renal veins. Ammonia excreted in the urine promotes acid excretion; ammonia returned to the systemic circulation is metabolized in the liver in a HCO3(-)-consuming process, resulting in no net benefit to acid-base homeostasis. Highly regulated ammonia transport by renal epithelial cells determines the proportion of ammonia excreted in the urine versus returned to the systemic circulation. The traditional paradigm of ammonia transport involving passive NH3 diffusion, protonation in the lumen and NH4(+) trapping due to an inability to cross plasma membranes is being replaced by the recognition of limited plasma membrane NH3 permeability in combination with the presence of specific NH3-transporting and NH4(+)-transporting proteins in specific renal epithelial cells. Ammonia production and transport are regulated by a variety of factors, including extracellular pH and K(+), and by several hormones, such as mineralocorticoids, glucocorticoids and angiotensin II. This coordinated process of regulated ammonia production and transport is critical for the effective maintenance of acid-base homeostasis.
Collapse
Affiliation(s)
- I David Weiner
- Nephrology and Hypertension Section, NF/SGVHS, Gainesville, Florida, USA.
| | | |
Collapse
|
7
|
Stewart AK, Shmukler BE, Vandorpe DH, Rivera A, Heneghan JF, Li X, Hsu A, Karpatkin M, O'Neill AF, Bauer DE, Heeney MM, John K, Kuypers FA, Gallagher PG, Lux SE, Brugnara C, Westhoff CM, Alper SL. Loss-of-function and gain-of-function phenotypes of stomatocytosis mutant RhAG F65S. Am J Physiol Cell Physiol 2011; 301:C1325-43. [PMID: 21849667 PMCID: PMC3233792 DOI: 10.1152/ajpcell.00054.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 08/11/2011] [Indexed: 11/22/2022]
Abstract
Four patients with overhydrated cation leak stomatocytosis (OHSt) exhibited the heterozygous RhAG missense mutation F65S. OHSt erythrocytes were osmotically fragile, with elevated Na and decreased K contents and increased cation channel-like activity. Xenopus oocytes expressing wild-type RhAG and RhAG F65S exhibited increased ouabain and bumetanide-resistant uptake of Li(+) and (86)Rb(+), with secondarily increased (86)Rb(+) influx sensitive to ouabain and to bumetanide. Increased RhAG-associated (14)C-methylammonium (MA) influx was severely reduced in RhAG F65S-expressing oocytes. RhAG-associated influxes of Li(+), (86)Rb(+), and (14)C-MA were pharmacologically distinct, and Li(+) uptakes associated with RhAG and RhAG F65S were differentially inhibited by NH(4)(+) and Gd(3+). RhAG-expressing oocytes were acidified and depolarized by 5 mM bath NH(3)/NH(4)(+), but alkalinized and depolarized by subsequent bath exposure to 5 mM methylammonium chloride (MA/MA(+)). RhAG F65S-expressing oocytes exhibited near-wild-type responses to NH(4)Cl, but MA/MA(+) elicited attenuated alkalinization and strong hyperpolarization. Expression of RhAG or RhAG F65S increased steady-state cation currents unaltered by bath Li(+) substitution or bath addition of 5 mM NH(4)Cl or MA/MA(+). These oocyte studies suggest that 1) RhAG expression increases oocyte transport of NH(3)/NH(4)(+) and MA/MA(+); 2) RhAG F65S exhibits gain-of-function phenotypes of increased cation conductance/permeability, and loss-of-function phenotypes of decreased and modified MA/MA(+) transport, and decreased NH(3)/NH(4)(+)-associated depolarization; and 3) RhAG transports NH(3)/NH(4)(+) and MA/MA(+) by distinct mechanisms, and/or the substrates elicit distinct cellular responses. Thus, RhAG F65S is a loss-of-function mutation for amine transport. The altered oocyte intracellular pH, membrane potential, and currents associated with RhAG or RhAG F65S expression may reflect distinct transport mechanisms.
Collapse
Affiliation(s)
- Andrew K Stewart
- Beth Israel Deaconess Medical Center, 330 Brookline Ave., Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Han KH, Lee HW, Handlogten ME, Bishop JM, Levi M, Kim J, Verlander JW, Weiner ID. Effect of hypokalemia on renal expression of the ammonia transporter family members, Rh B Glycoprotein and Rh C Glycoprotein, in the rat kidney. Am J Physiol Renal Physiol 2011; 301:F823-32. [PMID: 21753075 DOI: 10.1152/ajprenal.00266.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypokalemia is a common electrolyte disorder that increases renal ammonia metabolism and can cause the development of an acid-base disorder, metabolic alkalosis. The ammonia transporter family members, Rh B glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg), are expressed in the distal nephron and collecting duct and mediate critical roles in acid-base homeostasis by facilitating ammonia secretion. In the current studies, the effect of hypokalemia on renal Rhbg and Rhcg expression was examined. Normal Sprague-Dawley rats received either K(+)-free or control diets for 2 wk. Rats receiving the K(+)-deficient diet developed hypokalemia and metabolic alkalosis associated with significant increases in both urinary ammonia excretion and urine pH. Rhcg expression increased in the outer medullary collecting duct (OMCD). In OMCD intercalated cells, hypokalemia resulted in more discrete apical Rhcg expression and a marked increase in apical plasma membrane immunolabel. In principal cells, in the OMCD, hypokalemia increased both apical and basolateral Rhcg immunolabel intensity. Cortical Rhcg expression was not detectably altered by immunohistochemistry, although there was a slight decrease in total expression by immunoblot analysis. Rhbg protein expression was decreased slightly in the cortex and not detectably altered in the outer medulla. We conclude that in rat OMCD, hypokalemia increases Rhcg expression, causes more polarized apical expression in intercalated cells, and increases both apical and basolateral expression in the principal cell. Increased plasma membrane Rhcg expression in response to hypokalemia in the rat, particularly in the OMCD, likely contributes to the increased ammonia excretion and thereby to the development of metabolic alkalosis.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Anatomy Department, Ewha Womans University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Endre ZH, Pickering JW, Storer MK, Hu WP, Moorhead KT, Allardyce R, McGregor DO, Scotter JM. Breath ammonia and trimethylamine allow real-time monitoring of haemodialysis efficacy. Physiol Meas 2010; 32:115-30. [PMID: 21149927 DOI: 10.1088/0967-3334/32/1/008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Non-invasive monitoring of breath ammonia and trimethylamine using Selected-ion-flow-tube mass spectroscopy (SIFT-MS) could provide a real-time alternative to current invasive techniques. Breath ammonia and trimethylamine were monitored by SIFT-MS before, during and after haemodialysis in 20 patients. In 15 patients (41 sessions), breath was collected hourly into Tedlar bags and analysed immediately (group A). During multiple dialyses over 8 days, five patients breathed directly into the SIFT-MS analyser every 30 min (group B). Pre- and post-dialysis direct breath concentrations were compared with urea reduction, Kt/V and creatinine concentrations. Dialysis decreased breath ammonia, but a transient increase occurred mid treatment in some patients. Trimethylamine decreased more rapidly than reported previously. Pre-dialysis breath ammonia correlated with pre-dialysis urea in group B (r(2) = 0.71) and with change in urea (group A, r(2) = 0.24; group B, r(2) = 0.74). In group B, ammonia correlated with change in creatinine (r(2) = 0.35), weight (r(2) = 0.52) and Kt/V (r(2) = 0.30). The ammonia reduction ratio correlated with the urea reduction ratio (URR) (r(2) = 0.42) and Kt/V (r(2) = 0.38). Pre-dialysis trimethylamine correlated with Kt/V (r(2) = 0.21), and the trimethylamine reduction ratio with URR (r(2) = 0.49) and Kt/V (r(2) = 0.36). Real-time breath analysis revealed previously unmeasurable differences in clearance kinetics of ammonia and trimethylamine. Breath ammonia is potentially useful in assessment of dialysis efficacy.
Collapse
Affiliation(s)
- Z H Endre
- Christchurch Kidney Research Group, Department of Medicine, University of Otago, Christchurch, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Recent studies have identified a new family of ammonia-specific transporters, Rh glycoproteins, which enable NH3-specific transport. The purpose of this review is to summarize recent evidence regarding the role of Rh glycoproteins in renal ammonia transport. RECENT FINDINGS The Rh glycoproteins, RhAG/Rhag, RhBG/Rhbg and RhCG/Rhcg, transport ammonia in the form of molecular NH3, although there is some evidence suggesting the possibility of NH4 transport. RhAG/Rhag is expressed only in erythrocytes, and not in the kidney. Rhbg and Rhcg are expressed in distal nephron sites, from the distal convoluted tubule through the inner medullary collecting duct, with basolateral Rhbg expression and both apical and basolateral Rhcg expression. Whether Rhbg contributes to renal ammonia transport remains controversial. Rhcg expression parallels ammonia excretion in multiple experimental models and genetic deletion studies, both global and collecting duct-specific, demonstrate a critical role for Rhcg in both basal and acidosis-stimulated renal ammonia excretion. X-ray crystallography has defined critical structural elements in Rh glycoprotein-mediated ammonia transport. Finally, Rh glycoproteins may also function as CO2 transporters. SUMMARY No longer can NH3 transport be considered to occur only through diffusive NH3 movement. Transporter-mediated NH3 movement is fundamental to ammonia metabolism.
Collapse
|
11
|
Bishop JM, Verlander JW, Lee HW, Nelson RD, Weiner AJ, Handlogten ME, Weiner ID. Role of the Rhesus glycoprotein, Rh B glycoprotein, in renal ammonia excretion. Am J Physiol Renal Physiol 2010; 299:F1065-77. [PMID: 20719974 DOI: 10.1152/ajprenal.00277.2010] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rh B glycoprotein (Rhbg) is a member of the Rh glycoprotein family of ammonia transporters. In the current study, we examine Rhbg's role in basal and acidosis-stimulated acid-base homeostasis. Metabolic acidosis induced by HCl administration increased Rhbg expression in both the cortex and outer medulla. To test the functional significance of increased Rhbg expression, we used a Cre-loxP approach to generate mice with intercalated cell-specific Rhbg knockout (IC-Rhbg-KO). On normal diet, intercalated cell-specific Rhbg deletion did not alter urine ammonia excretion, pH, or titratable acid excretion significantly, but it did decrease glutamine synthetase expression in the outer medulla significantly. After metabolic acidosis was induced, urinary ammonia excretion was significantly less in IC-Rhbg-KO than in control (C) mice on days 2-4 of acid loading, but not on day 5. Urine pH and titratable acid excretion and dietary acid intake did not differ significantly between acid-loaded IC-Rhcg-KO and C mice. In IC-Rhbg-KO mice, acid loading increased connecting segment (CNT) cell and outer medullary collecting duct principal cell Rhbg expression. In both C and IC-Rhbg-KO mice, acid loading decreased glutamine synthetase in both the cortex and outer medulla; the decrease on day 3 was similar in IC-Rhbg-KO and C mice, but on day 5 it was significantly greater in IC-Rhbg-KO than in C mice. We conclude 1) intercalated cell Rhbg contributes to acidosis-stimulated renal ammonia excretion, 2) Rhbg in CNT and principal cells may contribute to renal ammonia excretion, and 3) decreased glutamine synthetase expression may enable normal rates of ammonia excretion under both basal conditions and on day 5 of acid loading in IC-Rhbg-KO mice.
Collapse
Affiliation(s)
- Jesse M Bishop
- Div. of Nephrology, Hypertension, and Transplantation, P.O. Box 100224, Univ. of Florida College of Medicine, Gainesville, FL 32610-0224, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Han KH, Lee SY, Kim WY, Shin JA, Kim J, Weiner ID. Expression of ammonia transporter family members, Rh B glycoprotein and Rh C glycoprotein, in the developing rat kidney. Am J Physiol Renal Physiol 2010; 299:F187-98. [PMID: 20392801 PMCID: PMC2904167 DOI: 10.1152/ajprenal.00607.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 04/10/2010] [Indexed: 11/22/2022] Open
Abstract
Ammonia metabolism is a primary component of acid-base homeostasis but is incompletely developed at time of birth. Rh B glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg) are recently recognized ammonia transporter family members expressed in the mammalian kidney. This study's purpose was to establish the expression and localization of Rhbg and Rhcg during kidney development. We examined kidneys from fetal days 16 (E16), 18 (E18), and 20 (E20), and from the first 21 days of postnatal development. Rhbg was expressed initially at E18, with expression only in the connecting tubule (CNT); at E20, Rhbg was expressed in both the CNT and the medullary collecting duct (MCD). In contrast, Rhcg was first expressed at E16 with basal expression in the ureteric bud; at E18, it was expressed in a subset of CNT cells with an apical pattern, followed by apical and basolateral expression in the MCD at E20. In the cortex, Rhbg and Rhcg expression increased in the CNT before expression in the cortical collecting duct during fetal development. In the MCD, both Rhbg and Rhcg expression was initially in cells in the papillary tip, with gradual removal from the tip during the late fetal period and transition during the early neonatal period to an adult pattern with predominant expression in the outer MCD and only rare expression in cells in the initial inner MCD. Double-labeling with intercalated cell-specific markers identified that Rhbg and Rhcg were expressed initially in CNT cells, CNT A-type intercalated cells and non-A, non-B intercalated cells, and in MCD A-type intercalated cells. We conclude that expression of Rhbg and Rhcg parallels intercalated cell development and that immature Rhbg and Rhcg expression at birth contributes to incomplete ammonia excretion capacity.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, Ewha Womans University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
13
|
Nakada T, Westhoff CM, Yamaguchi Y, Hyodo S, Li X, Muro T, Kato A, Nakamura N, Hirose S. Rhesus glycoprotein p2 (Rhp2) is a novel member of the Rh family of ammonia transporters highly expressed in shark kidney. J Biol Chem 2009; 285:2653-64. [PMID: 19926789 DOI: 10.1074/jbc.m109.052068] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Rhesus (Rh) glycoproteins are a family of membrane proteins capable of transporting ammonia. We isolated the full-length cDNA of a novel Rh glycoprotein, Rhp2, from a kidney cDNA library from the banded hound shark, Triakis scyllium. Molecular cloning and characterization indicated that Rhp2 consists of 476 amino acid residues and has 12 putative transmembrane spans, consistent with the structure of other family members. The shark Rhp2 gene was found to consist of only one coding exon. Northern blotting and in situ hybridization revealed that Rhp2 mRNA is exclusively expressed in the renal tubules of the sinus zone but not in the bundle zone and renal corpuscles. Immunohistochemical staining with a specific antiserum showed that Rhp2 is localized in the basolateral membranes of renal tubule cells. Double fluorescence labeling with phalloidin or labeling of the Na(+)/K(+)-ATPase further narrowed the location to the second and fourth loops in the sinus zone. Vacuolar type H(+)-ATPase was localized in apical membranes of the Rhp2-expressing tubule cells. Quantitative real-time PCR analysis and Western blotting showed that expression of Rhp2 was increased in response to elevation of environmental salinity. Functional analysis using the Xenopus oocyte expression system showed that Rhp2 has transport activity for methylammonium, an analog of ammonia. This transport activity was inhibited by NH(4)Cl but not trimethylamine-N-oxide and urea. These results suggested that Rhp2 is involved in ammonia reabsorption in the kidney of the elasmobranch group of cartilaginous fish comprising the sharks and rays.
Collapse
Affiliation(s)
- Tsutomu Nakada
- Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Han KH, Mekala K, Babida V, Kim HY, Handlogten ME, Verlander JW, Weiner ID. Expression of the gas-transporting proteins, Rh B glycoprotein and Rh C glycoprotein, in the murine lung. Am J Physiol Lung Cell Mol Physiol 2009; 297:L153-63. [PMID: 19429772 PMCID: PMC2711812 DOI: 10.1152/ajplung.90524.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 05/04/2009] [Indexed: 11/22/2022] Open
Abstract
A family of gas-transporting proteins, the Mep/Amt/Rh glycoprotein family, has been identified recently. These are integral membrane proteins, are widely expressed in sites of gas transport, and are known to transport the gaseous molecule, NH(3), and recent evidence indicates they can transport CO(2). Because the mammalian lung is a critical site for gas transport, the current studies examine the expression of the nonerythroid members of this extended family, Rh B glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg), in the normal mouse lung. Real-time RT-PCR and immunoblot analysis demonstrated both Rhbg and Rhcg mRNA and protein expression, respectively. Immunohistochemistry demonstrated both Rhbg and Rhcg were expressed in bronchial and bronchiolar epithelial cells. Rhbg was expressed by Clara cells, specifically, whereas all bronchial/bronchiolar epithelial cells, with the exception of goblet cells, expressed Rhcg. Rhbg expression was basolateral, whereas Rhcg exhibited apical and intracellular immunolabel, polarized expression similar to that observed in Rhbg- and Rhcg-expressing epithelial cells in other organs. There was no detectable expression of either Rhbg or Rhcg in alveolar endothelial or epithelial cells, in pneumocytes or in vascular tissue. In vitro studies using cultured bronchial epithelial cells confirm Rhbg and Rhcg expression, demonstrate that saturable, not diffusive, transport is the primary mechanism of ammonia/methylammonia transport, and show that the saturable transport mechanism has kinetics similar to those demonstrated previously for Rhbg and Rhcg. These findings suggest Rhbg and Rhcg may contribute to bronchial epithelial cell ammonia metabolism and suggest that they do not contribute to pulmonary CO(2) transport.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, Ewha Womans University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
15
|
Lee HW, Verlander JW, Bishop JM, Igarashi P, Handlogten ME, Weiner ID. Collecting duct-specific Rh C glycoprotein deletion alters basal and acidosis-stimulated renal ammonia excretion. Am J Physiol Renal Physiol 2009; 296:F1364-75. [PMID: 19321595 PMCID: PMC2692449 DOI: 10.1152/ajprenal.90667.2008] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2008] [Accepted: 03/24/2009] [Indexed: 11/22/2022] Open
Abstract
NH3 movement across plasma membranes has traditionally been ascribed to passive, lipid-phase diffusion. However, ammonia-specific transporters, Mep/Amt proteins, are present in primitive organisms and mammals express orthologs of Mep/Amt proteins, the Rh glycoproteins. These findings suggest that the mechanisms of NH3 movement in mammalian tissues should be reexamined. Rh C glycoprotein (Rhcg) is expressed in the collecting duct, where NH3 secretion is necessary for both basal and acidosis-stimulated ammonia transport. To determine whether the collecting duct secretes NH3 via Rhcg or via lipid-phase diffusion, we generated mice with collecting duct-specific Rhcg deletion (CD-KO). CD-KO mice had loxP sites flanking exons 5 and 9 of the Rhcg gene (Rhcg(fl/fl)) and expressed Cre-recombinase under control of the Ksp-cadherin promoter (Ksp-Cre). Control (C) mice were Rhcg(fl/fl) but Ksp-Cre negative. We confirmed kidney-specific genomic recombination using PCR analysis and collecting duct-specific Rhcg deletion using immunohistochemistry. Under basal conditions, urinary ammonia excretion was less in KO vs. C mice; urine pH was unchanged. After acid-loading for 7 days, CD-KO mice developed more severe metabolic acidosis than did C mice. Urinary ammonia excretion did not increase significantly on the first day of acidosis in CD-KO mice, despite an intact ability to increase urine acidification, whereas it increased significantly in C mice. On subsequent days, urinary ammonia excretion slowly increased in CD-KO mice, but was always significantly less than in C mice. We conclude that collecting duct Rhcg expression contributes to both basal and acidosis-stimulated renal ammonia excretion, indicating that collecting duct ammonia secretion is, at least in part, mediated by Rhcg and not solely by lipid diffusion.
Collapse
Affiliation(s)
- Hyun-Wook Lee
- University of Florida College of Medicine, PO Box 100224, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
16
|
Kim HY, Verlander JW, Bishop JM, Cain BD, Han KH, Igarashi P, Lee HW, Handlogten ME, Weiner ID. Basolateral expression of the ammonia transporter family member Rh C glycoprotein in the mouse kidney. Am J Physiol Renal Physiol 2009; 296:F543-55. [PMID: 19129254 DOI: 10.1152/ajprenal.90637.2008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ammonia metabolism and transport are critical for acid-base homeostasis. The ammonia transporter family member Rh C glycoprotein (Rhcg) is expressed in distal renal tubular segments, and its expression is regulated in parallel with renal ammonia metabolism. However, there are inconsistencies in its reported subcellular distribution, with both apical and basolateral Rhcg reported in rat and human kidney and only apical expression in mouse kidney. Because the membrane location of Rhcg is critical for understanding its physiological role, we reassessed mouse Rhcg localization using refined immunolocalization methods. Two antibodies directed against different Rhcg-specific epitopes identified both apical and basolateral Rhcg immunolabel in mouse kidney. Immunogold electron microscopy both confirmed basolateral plasma membrane Rhcg expression and showed that apical immunolabel represented expression in both the apical plasma membrane and in subapical cytoplasmic vesicles. Immunoblots and Northern blots identified similar bands in Balb/c and C57BL/6 kidneys, suggesting basolateral Rhcg may result from alternative trafficking. Basolateral Rhcg intensity was strain dependent, with less basolateral Rhcg expression in the Balb/c mouse compared with the C57BL/6 mouse. In mice with collecting duct-specific Rhcg gene deletion, generated using Cre-loxP techniques, neither apical nor basolateral Rhcg immunolabel was identified in the collecting duct, confirming that basolateral Rhcg was the product of the same gene product as apical Rhcg. Although basolateral Rhcg expression differed between C57BL/6 and Balb/c mice, Rh B glycoprotein, which is exclusively basolateral, was expressed at similar levels in the two strains. We conclude that Rhcg is present in both the apical and basolateral plasma membrane in the mouse kidney, where it is likely to contribute to renal ammonia metabolism.
Collapse
Affiliation(s)
- Hye-Young Kim
- Division of Nephrology, Hypertension, and Transplantation, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lim SW, Ahn KO, Kim WY, Han DH, Li C, Ghee JY, Han KH, Kim HY, Handlogten ME, Kim J, Yang CW, Weiner ID. Expression of ammonia transporters, Rhbg and Rhcg, in chronic cyclosporine nephropathy in rats. Nephron Clin Pract 2008; 110:e49-58. [PMID: 18776723 DOI: 10.1159/000153245] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 06/11/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Cyclosporine (CsA)-induced renal injury causes renal tubular acidosis. The current study was performed to evaluate the influence of CsA-induced renal injury on the ammonia transporter family members, Rh B-glycoprotein (Rhbg) and Rh C-glycoprotein (Rhcg). METHODS Rats were treated daily for 1 or 4 weeks with vehicle (VH) or CsA. Induction of chronic CsA-induced nephropathy was confirmed by demonstrating impaired renal function and characteristic histopathology. Rhbg and Rhcg expression was evaluated with immunoblot, immunohistochemistry, real-time RT-PCR and electron microscopy. RESULTS CsA treatment for 4 weeks developed mild metabolic acidosis and decreased urinary ammonia excretion. Rhcg mRNA expression was unchanged in both the cortex and outer medulla, but Rhcg protein expression in the CsA group was significantly reduced in the cortex and outer medulla. There were no significant differences in Rhbg mRNA and protein expression between the CsA and VH group. CONCLUSION Long-term treatment with CsA in rats results in decreased urinary ammonia excretion accompanied by decreased expression of Rhcg; these changes are likely to mediate the CsA-induced defect in ammonium excretion in the collecting duct.
Collapse
Affiliation(s)
- Sun Woo Lim
- Department of Internal Medicine, Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Bens M, Vandewalle A. Cell models for studying renal physiology. Pflugers Arch 2008; 457:1-15. [DOI: 10.1007/s00424-008-0507-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 03/22/2008] [Indexed: 12/24/2022]
|
19
|
Worrell RT, Merk L, Matthews JB. Ammonium transport in the colonic crypt cell line, T84: role for Rhesus glycoproteins and NKCC1. Am J Physiol Gastrointest Liver Physiol 2008; 294:G429-40. [PMID: 18032481 DOI: 10.1152/ajpgi.00251.2006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although colonic lumen NH(4)(+) levels are high, 15-44 mM normal range in humans, relatively few studies have addressed the transport mechanisms for NH(4)(+). More extensive studies have elucidated the transport of NH(4)(+) in the kidney collecting duct, which involves a number of transporter processes also present in the distal colon. Similar to NH(4)(+) secretion in the renal collecting duct, we show that the distal colon secretory model, T84 cell line, has the capacity to secrete NH(4)(+) and maintain an apical-to-basolateral NH(4)(+) gradient. NH(4)(+) transport in the secretory direction was supported by basolateral NH(4)(+) loading on NKCC1, Na(+)-K(+)-ATPase, and the NH(4)(+) transporter, RhBG. NH(4)(+) was transported on NKCC1 in T84 cells nearly as well as K(+) as determined by bumetanide-sensitive (86)Rb-uptake. (86)Rb-uptake and ouabain-sensitive current measurement indicated that NH(4)(+) is transported by Na(+)-K(+)-ATPase in these cells to an equal extent as K(+). T84 cells expressed mRNA for the basolateral NH(4)(+) transporter RhBG and the apical NH(4)(+) transporter RhCG. Net NH(4)(+) transport in the secretory direction determined by (14)C-methylammonium (MA) uptake and flux occurred in T84 cells suggesting functional RhG protein activity. The occurrence of NH(4)(+) transport in the secretory direction within a colonic crypt cell model likely serves to minimize net absorption of NH(4)(+) because of surface cell NH(4)(+) absorption. These findings suggest that we rethink the present limited understanding of NH(4)(+) handling by the distal colon as being due solely to passive absorption.
Collapse
Affiliation(s)
- Roger T Worrell
- Department of Surgery, University of Cincinnati, Cincinnati, OH 45219-0581, USA.
| | | | | |
Collapse
|
20
|
Han KH, Kim HY, Croker BP, Reungjui S, Lee SY, Kim J, Handlogten ME, Adin CA, Weiner ID. Effects of ischemia-reperfusion injury on renal ammonia metabolism and the collecting duct. Am J Physiol Renal Physiol 2007; 293:F1342-54. [PMID: 17686949 DOI: 10.1152/ajprenal.00437.2006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute renal injury induces metabolic acidosis, but its specific effects on the collecting duct, the primary site for urinary ammonia secretion, the primary component of net acid excretion, are incompletely understood. We induced ischemia-reperfusion (I/R) acute renal injury in Sprague-Dawley rats by clamping the renal pedicles bilaterally for 30 min followed by reperfusion for 6 h. Control rats underwent sham surgery without renal pedicle clamping. I/R injury decreased urinary ammonia excretion significantly but did not persistently alter urine volume, Na+, K+, or bicarbonate excretion. Histological examination demonstrated cellular damage in the outer and inner medullary collecting duct, as well as in the proximal tubule and the thick ascending limb of the loop of Henle. A subset of collecting duct cells were damaged and/or detached from the basement membrane; these cells were present predominantly in the outer medulla and were less frequent in the inner medulla. Immunohistochemistry identified that the damaged/detached cells were A-type intercalated cells, not principal cells. Both TdT-mediated dUTP nick-end labeling (TUNEL) staining and transmission electron microscopic examination demonstrated apoptosis but not necrosis. However, immunoreactivity for caspase-3 was observed in the proximal tubule, but not in collecting duct intercalated cells, suggesting that mechanism(s) of collecting duct intercalated cell apoptosis differ from those operative in the proximal tubule. We conclude that I/R injury decreases renal ammonia excretion and is associated with intercalated cell-specific detachment and apoptosis in the outer and inner medullary collecting duct. These effects likely contribute to the metabolic acidosis frequently observed in acute renal injury.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, Ewha Womans University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kim HY, Baylis C, Verlander JW, Han KH, Reungjui S, Handlogten ME, Weiner ID. Effect of reduced renal mass on renal ammonia transporter family, Rh C glycoprotein and Rh B glycoprotein, expression. Am J Physiol Renal Physiol 2007; 293:F1238-47. [PMID: 17652373 DOI: 10.1152/ajprenal.00151.2007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidneys can maintain acid-base homeostasis, despite reduced renal mass, through adaptive changes in net acid excretion, of which ammonia excretion is the predominant component. The present study examines whether these adaptations are associated with changes in the ammonia transporter family members, Rh B glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg). We used normal Sprague-Dawley rats and a 5/6 ablation-infarction model of reduced renal mass; control rats underwent sham operation. After 1 wk, glomerular filtration rate, assessed as creatinine clearance, was decreased, serum bicarbonate was slightly increased, and Na+and K+were unchanged. Total urinary ammonia excretion was unchanged, but urinary ammonia adjusted for creatinine clearance, an index of per nephron ammonia metabolism, increased significantly. Although reduced renal mass did not alter total Rhcg protein expression, both light microscopy and immunohistochemistry with quantitative morphometric analysis demonstrated hypertrophy of both intercalated cells and principal cells in the cortical and outer medullary collecting duct that was associated with increased apical and basolateral Rhcg polarization. Rhbg expression, analyzed using immunoblot analysis, immunohistochemistry, and measurement of cell-specific expression, was unchanged. We conclude that altered subcellular localization of Rhcg contributes to adaptive changes in single-nephron ammonia metabolism and maintenance of acid-base homeostasis in response to reduced renal mass.
Collapse
Affiliation(s)
- Hye-Young Kim
- Division of Nephrology, Hypertension and Transplantation, University of Florida College of Medicine, Gainesville, Florida, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The Rh system is one of the most important and complex blood group systems because of the large number of antigens and the serious complications for the fetus of a woman sensitized by transfusion or pregnancy. Major advances in our understanding of the Rh system have occurred with the cloning of the genes and with functional evidence that the Rh blood group proteins belong to an ancient family of membrane proteins involved in ammonia transport. The arrangement and configuration of the genes at the RH locus promotes genetic exchange, generating new antigens. Importantly, RH genetic testing can now be applied to clinical transfusion medicine and prenatal practice. This includes testing for RHD zygosity, confirmation or resolution of D antigen status, and detection of altered RHD and RHCE genes in individuals at risk for producing antibodies to high-incidence Rh antigens, particularly sickle cell disease (SCD) patients. The Rh proteins form a core complex that is critical to the structure of the erythrocyte membrane, and they may play a physiologic role in the sequestration of blood ammonia. The Rh family of proteins now includes non-erythroid homologs present in many other tissues, and comparative genomics reveal Rh homologs in all domains of life.
Collapse
Affiliation(s)
- Connie M Westhoff
- American Red Cross and Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, University of Pennsylvania, Philadelphia, PA 19123, USA.
| |
Collapse
|
23
|
Abstract
Acid-base homeostasis to a great extent relies on renal ammonia metabolism. In the past several years, seminal studies have generated important new insights into the mechanisms of renal ammonia transport. In particular, the theory that ammonia transport occurs almost exclusively through nonionic NH(3) diffusion and NH(4)(+) trapping has given way to a model postulating that a variety of proteins specifically transport NH(3) and NH(4)(+) and that this transport is critical for normal ammonia metabolism. Many of these proteins transport primarily H(+) or K(+) but also transport NH(4)(+). Nonerythroid Rh glycoproteins transport ammonia and may represent critical facilitators of ammonia transport in the kidney. This review discusses the underlying aspects of renal ammonia transport as well as specific proteins with important roles in renal ammonia transport.
Collapse
Affiliation(s)
- I. David Weiner
- Nephrology Section, North Florida/South Georgia Veterans Health System, University of Florida, Gainesville, Florida 32608
- Division of Nephrology, Hypertension and Transplantation, University of Florida, Gainesville, Florida 32608
| | - L. Lee Hamm
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
| |
Collapse
|
24
|
Yang B, Zhao D, Solenov E, Verkman AS. Evidence from knockout mice against physiologically significant aquaporin 8-facilitated ammonia transport. Am J Physiol Cell Physiol 2006; 291:C417-23. [PMID: 16624991 DOI: 10.1152/ajpcell.00057.2006] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aquaporin (AQP)8-facilitated transport of NH3has been suggested recently by increased NH3permeability in Xenopus oocytes and yeast expressing human or rat AQP8. We tested the proposed roles of AQP8-facilitated NH3transport in mammalian physiology by comparative phenotype studies in wild-type vs. AQP8-null mice. AQP8-facilitated NH3transport was confirmed in mammalian cell cultures expressing rat or mouse AQP8, in which the fluorescence of a pH-sensing yellow fluorescent protein was measured in response to ammonia (NH3/NH4+) gradients. Relative AQP8 single-channel NH3-to-water permeability was ∼0.03. AQP8-facilitated NH3and water permeability in a native tissue was confirmed in membrane vesicles isolated from testes of wild-type vs. AQP8-null mice, in which BCECF was used as an intravesicular pH indicator. A series of in vivo studies were done in mice, including 1) serum ammonia measurements before and after ammonia infusion, 2) renal ammonia clearance, 3) colonic ammonia absorption, and 4) liver ammonia accumulation and renal ammonia excretion after acute and chronic ammonia loading. Except for a small reduction in hepatic ammonia accumulation and increase in ammonia excretion in AQP8-null mice loaded with large amounts of ammonia, there were no significant differences in wild-type vs. AQP8-null mice. Our results support the conclusion that AQP8 can facilitate NH3transport but provide evidence against physiologically significant AQP8-facilitated NH3transport in mice.
Collapse
Affiliation(s)
- Baoxue Yang
- 1246 Health Sciences East Tower, Box 0521, University of California-San Francisco, San Francisco, CA 94143-0521, USA.
| | | | | | | |
Collapse
|
25
|
Han KH, Croker BP, Clapp WL, Werner D, Sahni M, Kim J, Kim HY, Handlogten ME, Weiner ID. Expression of the ammonia transporter, rh C glycoprotein, in normal and neoplastic human kidney. J Am Soc Nephrol 2006; 17:2670-9. [PMID: 16928804 PMCID: PMC4319185 DOI: 10.1681/asn.2006020160] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent studies have identified the presence of a novel Mep/Amt/Rh glycoprotein family of proteins that may play an important role in transmembrane ammonia transport. One of the mammalian members of this family, Rh C glycoprotein (RhCG), transports ammonia, is expressed in distal nephron sites that are critically important for ammonia secretion, exhibits increased expression in response to chronic metabolic acidosis, and originally was cloned as a tumor-related protein. The purpose of our studies was to determine the localization of RhCG in the normal and neoplastic human kidney. Immunoblot analysis of human renal cortical protein lysates demonstrated RhCG protein expression with a molecular weight of approximately 52 kD. Immunohistochemistry revealed both apical and basolateral Rhcg expression in the distal convoluted tubule, connecting segment, and initial collecting tubule and throughout the collecting duct. Co-localization with calbindin-D28k, H(+)-ATPase, aquaporin-2, and pendrin showed that distal convoluted tubule and connecting segment cells, A-type intercalated cells, and non-A, non-B cells express RhCG and that B-type intercalated cells, principal cells, and inner medullary collecting duct cells do not. In renal neoplasms, RhCG was expressed by chromophobe renal cell carcinoma and renal oncocytoma but not by clear cell renal cell carcinoma or by papillary renal cell carcinomas. These studies suggest that RhCG contributes to both apical and basolateral membrane ammonia transport in the human kidney. Furthermore, renal chromophobe renal cell carcinoma and renal oncocytoma seem to originate from the A-type intercalated cell.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, Ewha Womans University, Seoul, Korea
| | - Byron P. Croker
- Department of Pathology, University of Florida
- Department of Pathology, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - William L. Clapp
- Department of Pathology, University of Florida
- Department of Pathology, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | | | - Manisha Sahni
- Division of Nephrology, Hypertension and Transplantation, University of Florida
| | - Jin Kim
- Department of Anatomy and Medical Research Center for Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Korea
| | - Hye-Young Kim
- Division of Nephrology, Hypertension and Transplantation, University of Florida
| | - Mary E. Handlogten
- Division of Nephrology, Hypertension and Transplantation, University of Florida
| | - I. David Weiner
- Division of Nephrology, Hypertension and Transplantation, University of Florida
- Nephrology Section, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| |
Collapse
|
26
|
Goralski KB, Bose R, Sitar DS. NH4+ modulates renal tubule amantadine transport independently of intracellular pH changes. Eur J Pharmacol 2006; 541:87-94. [PMID: 16750190 DOI: 10.1016/j.ejphar.2006.04.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 04/21/2006] [Accepted: 04/28/2006] [Indexed: 01/11/2023]
Abstract
A bicarbonate-dependent organic cation transporter, unique from rOCT1 and rOCT2, primarily mediates amantadine uptake into renal proximal tubules. We examined whether intracellular pH regulates bicarbonate-dependent amantadine transporter function in these tubules. NH(4)Cl treatment resulted in immediate intracellular alkalinization of tubules for up to 30s followed by gradual acidification that was maximal at 5min. Proximal tubule amantadine uptake was similarly inhibited (60%) by NH(4)Cl during both the early intracellular alkalinization and later acidification phases. Sodium propionate treatment resulted in immediate intracellular acidification of proximal tubules without inhibiting amantadine uptake. NH(4)Cl inhibition of bicarbonate-dependent amantadine uptake was dose-dependent, competitive and sex-dependent. NH(4)Cl, NH(4)NO(3), (NH(4))(2)SO(4) and (NH(4))(2)HPO(4) inhibited amantadine uptake into proximal tubules similarly. NH(4)Cl also stimulated efflux of amantadine and tetraethylammonium from preloaded proximal tubules, suggesting mediation of a facilitated process. These data suggest the potential for direct modulation of organic cation transporters by NH(4)(+) in rat kidney proximal tubules.
Collapse
Affiliation(s)
- Kerry B Goralski
- Department of Pharmacology and Therapeutics, University of Manitoba Winnipeg, Manitoba, Canada R3E 0W3
| | | | | |
Collapse
|
27
|
Chambrey R, Goossens D, Quentin F, Eladari D. Rh glycoproteins in epithelial cells: lessons from rat and mice studies. Transfus Clin Biol 2006; 13:154-8. [PMID: 16563831 DOI: 10.1016/j.tracli.2006.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Rhesus glycoproteins are a recently discovered family of ammonium transporters and a new branch of the Mep/AMT proteins superfamily that was identified more than 15 years ago in lower organisms and plants. Despite many ex vivo studies showing evidences that Rh glycoproteins can accelerate transmembrane NH3 or NH4+ transfer, their role in normal and disease physiology remains unknown. This review focuses on some of the different studies carried out in animal models to gain insight into Rh glycoprotein function. Immunolocalization studies have added new evidence that this protein family is related to ammonium transport or metabolism in epithelial cells. However, the absence of distal tubular acidosis or hyperammonemia in Rhbg KO mice have raised new questions about the physiological significance of these proteins.
Collapse
Affiliation(s)
- R Chambrey
- Inserm U652, IFR58, institut des Cordeliers, faculté de médecine René-Descartes, université Paris-Descartes, 15, rue de l'Ecole-de-Médecine, 75006 Paris, France.
| | | | | | | |
Collapse
|
28
|
Van Kim CL, Colin Y, Cartron JP. Rh proteins: Key structural and functional components of the red cell membrane. Blood Rev 2006; 20:93-110. [PMID: 15961204 DOI: 10.1016/j.blre.2005.04.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rh (Rhesus) proteins (D, CcEe) are expressed in red cells (RBC) in association with other membrane proteins (RhAG, LW, CD47 and GPB). By interacting with the spectrin-based skeleton through protein 4.2 and ankyrin, the Rh complex contributes to the maintenance of the mechanical properties of the erythrocyte membrane. The RH system is one of the most immunogenic and polymorphic human blood group system. Molecular basis of most Rh phenotypes, including the Rh(null) phenotype associated with hemolytic anemia, have been determined. The demonstration that the RHD-positive locus is composed of the RHD and RHCE genes, whereas the RHD gene is deleted in most RhD-negative individuals, allowed fetal RhD genotyping by non-invasive PCR assays for antenatal diagnosis of pregnancy at risk for Rh hemolytic disease of the newborn. In mammals, the Rh protein family includes two non-erythroid members, RhBG and RhCG, mainly expressed in liver and kidney, two organs specialized in ammonia genesis and excretion. Functional analyses in heterologous systems revealed that RhAG, RhBG and RhCG can mediate ammonium (NH(3) and/or NH(4)(+)) transport across the cell membrane and might represent mammalian specific ammonium transporters. Furthermore, recent studies performed in human and murine red blood cells (RBC) indicate that RhAG facilitates CH(3)NH(2)/NH(3) movement across the membrane and represents a potential example of gas channel. The crystallographic structure of the bacterial ammonia channel AmtB and functional studies showing that AmtB conducts NH(3) into reconstituted vesicles is fully consistent with these latter studies. In RBCs, RhAG may transport NH(3) to detoxifying organs like kidney and liver and with non-erythroid tissues orthologs may contribute to regulation of the acid-base balance.
Collapse
Affiliation(s)
- Caroline Le Van Kim
- Inserm U76; Institut National de la Transfusion Sanguine, 6 Rue Alexandre Cabanel, 75015 Paris, France.
| | | | | |
Collapse
|
29
|
Abstract
A novel family of proteins, the Mep/AMT/Rh glycoprotein family may mediate important roles in transmembrane ammonia transport in a wide variety of single-celled and multicellular organisms. Results from our laboratory have examined the expression of the non-erythroid proteins, Rh B Glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg), in a wide variety of mammalian tissues. In the kidney, Rhbg and Rhcg are present in distal nephron sites responsible for ammonia secretion. In the mouse kidney, Rhbg immunoreactivity is exclusively basolateral and Rhcg immunoreactivity is exclusively apical, whereas in the rat kidney Rhcg exhibits both apical and basolateral expression. Chronic metabolic acidosis increases Rhcg expression in the outer and inner medulla of the rat kidney; these changes, at least in the outer medullary collecting duct, involve changes in total cellular protein expression in both principal and intercalated cell and changes in its subcellular localization. In the liver, Rhbg is present in the basolateral plasma membrane of the perivenous hepatocyte and Rhcg is present in bile duct epithelia. In the gastrointestinal tract, Rhbg and Rhcg exhibit cell-specific, axially heterogeneous, and polarized expression. These patterns of expression are consistent with Rhbg and Rhcg mediating important roles in mammalian ammonia biology. The lack of the effect of chronic metabolic acidosis on Rhbg expression raises the possibility that Rhbg may function either as ammonia sensing-protein or that it may mediate roles other than ammonia transport.
Collapse
Affiliation(s)
- I D Weiner
- Nephrology and Hypertension Section, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.
| |
Collapse
|
30
|
Seshadri RM, Klein JD, Smith T, Sands JM, Handlogten ME, Verlander JW, Weiner ID. Changes in subcellular distribution of the ammonia transporter, Rhcg, in response to chronic metabolic acidosis. Am J Physiol Renal Physiol 2006; 290:F1443-52. [PMID: 16434569 DOI: 10.1152/ajprenal.00459.2005] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The primary mechanism by which the kidneys mediate net acid excretion is through ammonia metabolism. In the current study, we examined whether chronic metabolic acidosis, which increases ammonia metabolism, alters the cell-specific and/or the subcellular expression of the ammonia transporter family member, Rhcg, in the outer medullary collecting duct in the inner stripe (OMCDi). Chronic metabolic acidosis was induced in normal SD rats by HCl ingestion for 7 days; controls were pair-fed. The subcellular distribution of Rhcg was determined using immunogold electron microscopy and morphometric analyses. In intercalated cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane. Intracellular Rhcg decreased significantly, and basolateral Rhcg was unchanged. Because apical plasma membrane length increased in parallel with apical Rhcg immunolabel, apical plasma membrane Rhcg density was unchanged. In principal cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane while decreasing the intracellular proportion. In contrast to the intercalated cell, chronic metabolic acidosis did not significantly alter apical boundary length; accordingly, apical plasma membrane Rhcg density increased. In addition, basolateral Rhcg immunolabel increased in response to chronic metabolic acidosis. These results indicate that in the rat OMCDi 1) chronic metabolic acidosis increases apical plasma membrane Rhcg in both the intercalated cell and principal cell where it may contribute to enhanced apical ammonia secretion; 2) increased apical plasma membrane Rhcg results from both increased total protein and changes in the subcellular distribution of Rhcg; 3) the mechanism of Rhcg subcellular redistribution differs in intercalated and principal cells; and 4) Rhcg may contribute to regulated basolateral ammonia transport in the principal cell.
Collapse
Affiliation(s)
- Ramanathan M Seshadri
- Division of Nephrology, Hypertension, and Transplantation, Univ. of Florida College of Medicine, Gainesville, FL, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Zidi-Yahiaoui N, Mouro-Chanteloup I, D'Ambrosio AM, Lopez C, Gane P, Le Van Kim C, Cartron JP, Colin Y, Ripoche P. Human Rhesus B and Rhesus C glycoproteins: properties of facilitated ammonium transport in recombinant kidney cells. Biochem J 2006; 391:33-40. [PMID: 15929723 PMCID: PMC1237136 DOI: 10.1042/bj20050657] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The mammalian Rh (Rhesus) protein family belongs to the Amt/Mep (ammonia transporter/methylammonium permease)/Rh superfamily of ammonium transporters. Whereas RhCE, RhD and RhAG are erythroid specific, RhBG and RhCG are expressed in key organs associated with ammonium transport and metabolism. We have investigated the ammonium transport function of human RhBG and RhCG by comparing intracellular pH variation in wild-type and transfected HEK-293 (human embryonic kidney) cells and MDCK (Madin-Darby canine kidney) cells in the presence of ammonium (NH4+/NH3) gradients. Stopped-flow spectrofluorimetry analysis, using BCECF [2',7'-bis-(2-carboxyethyl)-5(6)-carboxyfluorescein] as a pH-sensitive probe, revealed that all cells submitted to inwardly or outwardly directed ammonium gradients exhibited rapid alkalinization or acidification phases respectively, which account for ammonium movements in transfected and native cells. However, as compared with wild-type cells known to have high NH3 lipid permeability, RhBG- and RhCG-expressing cells exhibited ammonium transport characterized by: (i) a five to six times greater kinetic rate-constant; (ii) a weak temperature-dependence; and (iii) reversible inhibition by mercuric chloride (IC50: 52 microM). Similarly, when subjected to a methylammonium gradient, RhBG- and RhCG-expressing cells exhibited kinetic rate constants greater than those of native cells. However, these constants were five times higher for RhBG as compared with RhCG, suggesting a difference in substrate accessibility. These results, indicating that RhBG and RhCG facilitate rapid and low-energy-dependent bi-directional ammonium movement across the plasma membrane, favour the hypothesis that these Rh glycoproteins, together with their erythroid homologue RhAG [Ripoche, Bertrand, Gane, Birkenmeier, Colin and Cartron (2005) Proc. Natl. Acad. Sci. U.S.A. 101, 17222-17227] constitute a family of NH3 channels in mammalian cells.
Collapse
Affiliation(s)
- Nedjma Zidi-Yahiaoui
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Isabelle Mouro-Chanteloup
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Anne-Marie D'Ambrosio
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Claude Lopez
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Pierre Gane
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Caroline Le Van Kim
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Jean-Pierre Cartron
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| | - Yves Colin
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
- To whom correspondence should be addressed (email )
| | - Pierre Ripoche
- INSERM, U665, Paris, F-75015, France, Institut National de la Transfusion Sanguine, Paris, F-75015, France, and Université Paris 7/Denis Diderot, Paris, F-75005, France
| |
Collapse
|
32
|
Lytton SD, Fischer W, Nagel W, Haas R, Beck FX. Production of ammonium by Helicobacter pylori mediates occludin processing and disruption of tight junctions in Caco-2 cells. MICROBIOLOGY-SGM 2005; 151:3267-3276. [PMID: 16207910 DOI: 10.1099/mic.0.28049-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tight junctions, paracellular permeability barriers that define epithelial cell polarity, play an essential role in transepithelial transport, cell-cell adhesion and lymphocyte transmigration. They are also important for the maintenance of innate immune defence and intestinal antigen uptake. Ammonium (NH4+) is elevated in the gastric aspirates of Helicobacter pylori-infected patients and has been implicated in the disruption of tight-junction functional integrity and the induction of gastric mucosal damage during H. pylori infection. The precise mechanism of the effect of ammonium and the molecular targets of ammonium in host tissue are not yet identified. To study the effects of ammonium on epithelial tight junctions, the human colon carcinoma cell line Caco-2 was cultured on permeable supports and the transepithelial resistance (TER) was measured at different time intervals following exposure to ammonium salts or H. pylori-derived ammonium. A biphasic response to treatment with ammonium was found. Acute exposure to ammonium salts or NH3/NH4+ derived from urea metabolism by wild-type H. pylori resulted in a 20-30 % decrease in TER. After 24 h, the NH4Cl-treated cells showed a partial recovery of TER. In contrast, the control culture, or cultures that were exposed to supernatants derived from urease-deficient H. pylori, showed no significant decrease in TER. Occludin-specific immunoblots revealed the expression of a low-molecular-weight form of occludin of 42 kDa upon NH3/NH4+ exposure. The results indicate that modulation of tight-junction function by H. pylori is ammonium-dependent and linked to the accumulation of a low-molecular-weight and detergent-soluble form of occludin.
Collapse
Affiliation(s)
- Simon D Lytton
- Physiologisches Institut der Ludwig-Maximilians-Universität, D-80336 München, Germany
| | - Wolfgang Fischer
- Max von Pettenkofer-Institut der Ludwig-Maximilians-Universität, D-80336 München, Germany
| | - Wolfram Nagel
- Physiologisches Institut der Ludwig-Maximilians-Universität, D-80336 München, Germany
| | - Rainer Haas
- Max von Pettenkofer-Institut der Ludwig-Maximilians-Universität, D-80336 München, Germany
| | - Franz X Beck
- Physiologisches Institut der Ludwig-Maximilians-Universität, D-80336 München, Germany
| |
Collapse
|
33
|
Seshadri RM, Klein JD, Kozlowski S, Sands JM, Kim YH, Han KH, Handlogten ME, Verlander JW, Weiner ID. Renal expression of the ammonia transporters, Rhbg and Rhcg, in response to chronic metabolic acidosis. Am J Physiol Renal Physiol 2005; 290:F397-408. [PMID: 16144966 DOI: 10.1152/ajprenal.00162.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic metabolic acidosis induces dramatic increases in net acid excretion that are predominantly due to increases in urinary ammonia excretion. The current study examines whether this increase is associated with changes in the expression of the renal ammonia transporter family members, Rh B glycoprotein (Rhbg) and Rh C glycoprotein (Rhcg). Chronic metabolic acidosis was induced in Sprague-Dawley rats by HCl ingestion for 1 wk; control animals were pair-fed. After 1 wk, metabolic acidosis had developed, and urinary ammonia excretion increased significantly. Rhcg protein expression was increased in both the outer medulla and the base of the inner medulla. Intercalated cells in the outer medullary collecting duct (OMCD) and in the inner medullary collecting duct (IMCD) in acid-loaded animals protruded into the tubule lumen and had a sharp, discrete band of apical Rhcg immunoreactivity, compared with a flatter cell profile and a broad band of apical immunolabel in control kidneys. In addition, basolateral Rhcg immunoreactivity was observed in both control and acidotic kidneys. Cortical Rhcg protein expression and immunoreactivity were not detectably altered. Rhcg mRNA expression was not significantly altered in the cortex, outer medulla, or inner medulla by chronic metabolic acidosis. Rhbg protein and mRNA expression were unchanged in the cortex, outer and inner medulla, and no changes in Rhbg immunolabel were evident in these regions. We conclude that chronic metabolic acidosis increases Rhcg protein expression in intercalated cells in the OMCD and in the IMCD, where it is likely to mediate an important role in the increased urinary ammonia excretion.
Collapse
Affiliation(s)
- Ramanathan M Seshadri
- University of Florida College of Medicine, P. O. Box 100224, Gainesville, FL 32610-0224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mak DOD, Dang B, Weiner ID, Foskett JK, Westhoff CM. Characterization of ammonia transport by the kidney Rh glycoproteins RhBG and RhCG. Am J Physiol Renal Physiol 2005; 290:F297-305. [PMID: 16131648 PMCID: PMC4321823 DOI: 10.1152/ajprenal.00147.2005] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The erythrocyte Rh-associated glycoprotein (RhAG) was recently found to mediate transport of ammonia/ammonium when expressed in Xenopus laevis oocytes and yeast Saccharomyces cerevisiae. Nonerythroid homologs, RhBG and RhCG, are expressed in the mammalian kidney connecting segment and the collecting duct, major sites of urinary ammonia secretion. This study characterizes the transport properties of murine RhBG and RhCG by ammonium analog [14C]methylamine (MA) uptake and two-electrode voltage clamping of X. laevis oocytes. Both RhBG and RhCG mediated transport of ammonia, but differed in affinity for substrate (K(m) = 2.5 and 10 mM, respectively). The rates of RhBG- and RhCG-mediated transport were sensitive to the concentration of the protonated MA species and were stimulated by extracellular alkalosis and inhibited by acidosis, suggesting a role for H+ in the transport process. Whereas expression of RhBG or RhCG caused a small increase in plasma membrane conductance, [14C]MA uptake was not affected by depolarization of oocytes with 100 mM extracellular K+ or by clamping the membrane potential between 0 and -100 mV, indicating that RhBG- and RhCG-mediated transport was independent of the membrane potential. These results strongly suggest that RhBG and RhCG transport ammonia by an electroneutral process that involves NH4(+)/H+ exchange resulting in net NH3 translocation. The polarized localization of RhBG and RhCG in kidney tubules and the different substrate affinities may enable these proteins to participate in transepithelial ammonia secretion and to therefore play an important role in whole animal acid-base regulation.
Collapse
Affiliation(s)
- Don-On Daniel Mak
- American Red Cross, 700 Spring Garden St., Philadelphia, PA 19123, USA
| | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Connie M Westhoff
- American Red Cross and the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
36
|
Handlogten ME, Hong SP, Westhoff CM, Weiner ID. Apical ammonia transport by the mouse inner medullary collecting duct cell (mIMCD-3). Am J Physiol Renal Physiol 2005; 289:F347-58. [PMID: 15798090 DOI: 10.1152/ajprenal.00253.2004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The collecting duct is the primary site of urinary ammonia secretion; the current study determines whether apical ammonia transport in the mouse inner medullary collecting duct cell (mIMCD-3) occurs via nonionic diffusion or a transporter-mediated process and, if the latter, presents the characteristics of this apical ammonia transport. We used confluent cells on permeable support membranes and examined apical uptake of the ammonia analog [14C]methylammonia ([14C]MA). mIMCD-3 cells exhibited both diffusive and saturable, transporter-mediated, nondiffusive apical [14C]MA transport. Transporter-mediated [14C]MA uptake had a Kmof 7.0 ± 1.5 mM and was competitively inhibited by ammonia with a Kiof 4.3 ± 2.0 mM. Transport activity was stimulated by both intracellular acidification and extracellular alkalinization, and it was unaltered by changes in membrane voltage, thereby functionally identifying an apical, electroneutral NH4+/H+exchange activity. Transport was bidirectional, consistent with a role in ammonia secretion. In addition, transport was not altered by Na+or K+removal, not inhibited by luminal K+, and not mediated by apical H+-K+-ATPase, Na+-K+-ATPase, or Na+/H+exchange. Finally, mIMCD-3 cells express the recently identified ammonia transporter family member Rh C glycoprotein (RhCG) at its apical membrane. These studies indicate that the renal collecting duct cell mIMCD-3 has a novel apical, electroneutral Na+- and K+-independent NH4+/H+exchange activity, possibly mediated by RhCG, that is likely to mediate important components of collecting duct ammonia secretion.
Collapse
Affiliation(s)
- Mary E Handlogten
- Division of Nephrology, Hypertension and Transplantation, University of Florida College of Medicine, Gainesville, 32610-0224, USA
| | | | | | | |
Collapse
|
37
|
Handlogten ME, Hong SP, Zhang L, Vander AW, Steinbaum ML, Campbell-Thompson M, Weiner ID. Expression of the ammonia transporter proteins Rh B glycoprotein and Rh C glycoprotein in the intestinal tract. Am J Physiol Gastrointest Liver Physiol 2005; 288:G1036-47. [PMID: 15576624 DOI: 10.1152/ajpgi.00418.2004] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ammonia metabolism is important in multiple aspects of gastrointestinal physiology, but the mechanisms of ammonia transport in the gastrointestinal tract remain incompletely defined. The present study examines expression of the ammonia transporter family members Rh B glycoprotein (RhBG) and Rh C glycoprotein (RhCG) in the mouse gastrointestinal tract. Real-time RT-PCR amplification and immunoblot analysis identified mRNA and protein for both RhBG and RhCG were expressed in stomach, duodenum, jejunum, ileum, and colon. Immunohistochemistry showed organ and cell-specific expression of both RhBG and RhCG. In the stomach, both RhBG and RhCG were expressed in the fundus and forestomach, but not in the antrum. In the forestomach, RhBG was expressed by all nucleated squamous epithelial cells, whereas RhCG was expressed only in the stratum germinativum. In the fundus, RhBG and RhCG immunoreactivity was present in zymogenic cells but not in parietal or mucous cells. Furthermore, zymogenic cell RhBG and RhCG expression was polarized, with apical RhCG and basolateral RhBG immunoreactivity. In the duodenum, jejunum, ileum, and colon, RhBG and RhCG immunoreactivity was present in villous, but not in mucous or crypt cells. Similar to the fundic zymogenic cell, RhBG and RhCG expression in villous epithelial cells was polarized when apical RhCG and basolateral RhBG immunoreactivity was present. Thus the ammonia transporting proteins RhBG and RhCG exhibit cell-specific, axially heterogeneous, and polarized expression in the intestinal tract suggesting they function cooperatively to mediate gastrointestinal tract ammonia transport.
Collapse
Affiliation(s)
- Mary E Handlogten
- Medical Service, North Florida/South Georgia Veterans Health System, USA
| | | | | | | | | | | | | |
Collapse
|