1
|
Signaling through the extracellular calcium-sensing receptor (CaSR). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:103-42. [PMID: 22453940 DOI: 10.1007/978-94-007-2888-2_5] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The extracellular calcium ([Formula: see text])-sensing receptor (CaSR) was the first GPCR identified whose principal physiological ligand is an ion, namely extracellular Ca(2+). It maintains the near constancy of [Formula: see text] that complex organisms require to ensure normal cellular function. A wealth of information has accumulated over the past two decades about the CaSR's structure and function, its role in diseases and CaSR-based therapeutics. This review briefly describes the CaSR and key features of its structure and function, then discusses the extracellular signals modulating its activity, provides an overview of the intracellular signaling pathways that it controls, and, finally, briefly describes CaSR signaling both in tissues participating in [Formula: see text] homeostasis as well as those that do not. Factors controlling CaSR signaling include various factors affecting the expression of the CaSR gene as well as modulation of its trafficking to and from the cell surface. The dimeric cell surface CaSR, in turn, links to various heterotrimeric and small molecular weight G proteins to regulate intracellular second messengers, lipid kinases, various protein kinases, and transcription factors that are part of the machinery enabling the receptor to modulate the functions of the wide variety of cells in which it is expressed. CaSR signaling is impacted by its interactions with several binding partners in addition to signaling elements per se (i.e., G proteins), including filamin-A and caveolin-1. These latter two proteins act as scaffolds that bind signaling components and other key cellular elements (e.g., the cytoskeleton). Thus CaSR signaling likely does not take place randomly throughout the cell, but is compartmentalized and organized so as to facilitate the interaction of the receptor with its various signaling pathways.
Collapse
|
2
|
Gamba G, Friedman PA. Thick ascending limb: the Na(+):K (+):2Cl (-) co-transporter, NKCC2, and the calcium-sensing receptor, CaSR. Pflugers Arch 2009; 458:61-76. [PMID: 18982348 PMCID: PMC3584568 DOI: 10.1007/s00424-008-0607-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 10/21/2008] [Indexed: 01/12/2023]
Abstract
The thick ascending limb of Henle's loop is a nephron segment that is vital to the formation of dilute and concentrated urine. This ability is accomplished by a consortium of functionally coupled proteins consisting of the apical Na(+):K(+):2Cl(-) co-transporter, the K(+) channel, and basolateral Cl(-) channel that mediate electroneutral salt absorption. In thick ascending limbs, salt absorption is importantly regulated by the calcium-sensing receptor. Genetic or pharmacological disruption impairing the function of any of these proteins results in Bartter syndrome. The thick ascending limb is also an important site of Ca(2+) and Mg(2+) absorption. Calcium-sensing receptor activation inhibits cellular Ca(2+) absorption induced by parathyroid hormone, as well as passive paracellular Ca(2+) transport. The present review discusses these functions and their genetic and molecular regulation.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14000 Mexico City, Mexico
| | - Peter A. Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
3
|
|
4
|
Procino G, Carmosino M, Tamma G, Gouraud S, Laera A, Riccardi D, Svelto M, Valenti G. Extracellular calcium antagonizes forskolin-induced aquaporin 2 trafficking in collecting duct cells. Kidney Int 2005; 66:2245-55. [PMID: 15569313 DOI: 10.1111/j.1523-1755.2004.66036.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Urinary concentrating defects and polyuria are the most important renal manifestations of hypercalcemia and the resulting hypercalciuria. In this study, we tested the hypothesis that hypercalciuria-associated polyuria in kidney collecting duct occurs through an impairment of the vasopressin-dependent aquaporin 2 (AQP2) water channel targeting to the apical membrane possibly involving calcium-sensing receptor (CaR) signaling. METHODS AQP2-transfected collecting duct CD8 cells were used as experimental model. Quantitation of cell surface AQP2 immunoreactivity was performed using an antibody recognizing the extracellular AQP2 C loop. Intracellular cyclic adenosine monophosphate (cAMP) accumulation was measured in CD8 cells using a cAMP enzyme immunoassay kit. To study the translocation of protein kinase C (PKC), membranes or cytosol fractions from CD8 cells were subjected to Western blotting using anti-PKC isozymes antibodies. The amount of F-actin was determined by spectrofluorometric techniques. Intracellular calcium measurements were performed by spectrofluorometric analysis with Fura-2/AM. RESULTS We demonstrated that extracellular calcium (Ca2+ o) (5 mmol/L) strongly inhibited forskolin-stimulated increase in AQP2 expression in the apical plasma membrane. At least three intracellular pathways activated by extracellular calcium were found to contribute to this effect. Firstly, the increase in cAMP levels in response to forskolin stimulation was drastically reduced in cells pretreated with Ca2+ o compared to untreated cells. Second, Ca2+ o activated PKC, known to counteract vasopressin response. Third, quantification of F-actin demonstrated that Ca2+ o caused a nearly twofold increase in F-actin content compared with basal conditions. All these effects were mimicked by a nonmembrane permeable agonist of the extracellular CaR, Gd3+. CONCLUSION Together, these data demonstrate that extracellular calcium, possibly acting through the endogenous CaR, antagonizes forskolin-induced AQP2 translocation to the apical plasma membrane in CD8 cells. In hypercalciuria, this mechanism might blunt water reabsorption and prevent further calcium concentration, thus protecting against a potential risk of urinary calcium-containing stone formation.
Collapse
Affiliation(s)
- Giuseppe Procino
- Department of General and Environmental Physiology, University of Bari, Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
5
|
|
6
|
Abstract
Renal cyclooxygenase 1 and 2 activity produces five primary prostanoids: prostaglandin E2, prostaglandin F2alpha, prostaglandin I2, thromboxane A2, and prostaglandin D2. These lipid mediators interact with a family of distinct G protein-coupled prostanoid receptors designated EP, FP, IP, TP, and DP, respectively, which exert important regulatory effects on renal function. The intrarenal distribution of these prostanoid receptors has been mapped, and the consequences of their activation have been partially characterized. FP, TP, and EP1 receptors preferentially couple to an increase in cell calcium. EP2, EP4, DP, and IP receptors stimulate cyclic AMP, whereas the EP3 receptor preferentially couples to Gi, inhibiting cyclic AMP generation. EP1 and EP3 mRNA expression predominates in the collecting duct and thick limb, respectively, where their stimulation reduces NaCl and water absorption, promoting natriuresis and diuresis. The FP receptor is highly expressed in the distal convoluted tubule, where it may have a distinct effect on renal salt transport. Although only low levels of EP2 receptor mRNA are detected in the kidney and its precise intrarenal localization is uncertain, mice with targeted disruption of the EP2 receptor exhibit salt-sensitive hypertension, suggesting that this receptor may also play an important role in salt excretion. In contrast, EP4 receptor mRNA is predominantly expressed in the glomerulus, where it may contribute to the regulation of glomerular hemodynamics and renin release. The IP receptor mRNA is highly expressed near the glomerulus, in the afferent arteriole, where it may also dilate renal arterioles and stimulate renin release. Conversely, TP receptors in the glomerulus may counteract the effects of these dilator prostanoids and increase glomerular resistance. At present there is little evidence for DP receptor expression in the kidney. These receptors act in a concerted fashion as physiological buffers, protecting the kidney from excessive functional changes during periods of physiological stress. Nonsteroidal anti-inflammatory drug (NSAID)-mediated cyclooxygenase inhibition results in the loss of these combined effects, which contributes to their renal effects. Selective prostanoid receptor antagonists may provide new therapeutic approaches for specific disease states.
Collapse
Affiliation(s)
- M D Breyer
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
7
|
Abstract
The cloning of the G protein-coupled, extracellular calcium (Ca(2+)o)-sensing receptor (CaR) has identified a central mediator of the mechanism governing systemic Ca(2+)o homeostasis. This system enables organisms to adapt successfully to wide variations in dietary Ca(2+)o intake while maintaining near constancy of Ca(2+)o. Whereas discussions of Ca(2+)o homeostasis have generally focused on the key role of Ca(2+)o-elicited changes in parathyroid hormone secretion, the presence of the CaRs in effector tissues of this system enables direct regulation of processes (e.g. renal tubular Ca(2+) reabsorption and possibly bone formation and resorption) that add additional layers of homeostatic control. As we understand more about how the CaR regulates these tissues, we may find that it participates in other processes relevant to mineral ion homeostasis, including the control of the 1-hydroxylation and activation of vitamin D3 or reabsorption of phosphate in the renal proximal tubule. Regardless, the remarkable sensitivity of the CaR to small changes in Ca(2+)o allows adjustments in the response of the Ca(2+)o homeostatic system to increases or decreases in the intake of dietary Ca(2+), for instance, that cause barely detectable alterations in Ca(2+)o. Furthermore, the CaR likely participates in coordinating interactions among several different homeostatic control systems (including those for water, Mg(2+)o, Na(+), extracellular volume, and/or blood pressure), despite the fact that these systems are often considered to function largely independently of mineral ion metabolism.
Collapse
Affiliation(s)
- E M Brown
- Endocrine-Hypertension Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| |
Collapse
|
8
|
Féraille E, Doucet A. Sodium-potassium-adenosinetriphosphatase-dependent sodium transport in the kidney: hormonal control. Physiol Rev 2001; 81:345-418. [PMID: 11152761 DOI: 10.1152/physrev.2001.81.1.345] [Citation(s) in RCA: 340] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tubular reabsorption of filtered sodium is quantitatively the main contribution of kidneys to salt and water homeostasis. The transcellular reabsorption of sodium proceeds by a two-step mechanism: Na(+)-K(+)-ATPase-energized basolateral active extrusion of sodium permits passive apical entry through various sodium transport systems. In the past 15 years, most of the renal sodium transport systems (Na(+)-K(+)-ATPase, channels, cotransporters, and exchangers) have been characterized at a molecular level. Coupled to the methods developed during the 1965-1985 decades to circumvent kidney heterogeneity and analyze sodium transport at the level of single nephron segments, cloning of the transporters allowed us to move our understanding of hormone regulation of sodium transport from a cellular to a molecular level. The main purpose of this review is to analyze how molecular events at the transporter level account for the physiological changes in tubular handling of sodium promoted by hormones. In recent years, it also became obvious that intracellular signaling pathways interacted with each other, leading to synergisms or antagonisms. A second aim of this review is therefore to analyze the integrated network of signaling pathways underlying hormone action. Given the central role of Na(+)-K(+)-ATPase in sodium reabsorption, the first part of this review focuses on its structural and functional properties, with a special mention of the specificity of Na(+)-K(+)-ATPase expressed in renal tubule. In a second part, the general mechanisms of hormone signaling are briefly introduced before a more detailed discussion of the nephron segment-specific expression of hormone receptors and signaling pathways. The three following parts integrate the molecular and physiological aspects of the hormonal regulation of sodium transport processes in three nephron segments: the proximal tubule, the thick ascending limb of Henle's loop, and the collecting duct.
Collapse
Affiliation(s)
- E Féraille
- Division of Nephrology, Geneva University Hospital, Geneva, Switzerland.
| | | |
Collapse
|
9
|
Abstract
The cloning of a G protein-coupled extracellular Ca(2+) (Ca(o)(2+))-sensing receptor (CaR) has elucidated the molecular basis for many of the previously recognized effects of Ca(o)(2+) on tissues that maintain systemic Ca(o)(2+) homeostasis, especially parathyroid chief cells and several cells in the kidney. The availability of the cloned CaR enabled the development of DNA and antibody probes for identifying the CaR's mRNA and protein, respectively, within these and other tissues. It also permitted the identification of human diseases resulting from inactivating or activating mutations of the CaR gene and the subsequent generation of mice with targeted disruption of the CaR gene. The characteristic alterations in parathyroid and renal function in these patients and in the mice with "knockout" of the CaR gene have provided valuable information on the CaR's physiological roles in these tissues participating in mineral ion homeostasis. Nevertheless, relatively little is known about how the CaR regulates other tissues involved in systemic Ca(o)(2+) homeostasis, particularly bone and intestine. Moreover, there is evidence that additional Ca(o)(2+) sensors may exist in bone cells that mediate some or even all of the known effects of Ca(o)(2+) on these cells. Even more remains to be learned about the CaR's function in the rapidly growing list of cells that express it but are uninvolved in systemic Ca(o)(2+) metabolism. Available data suggest that the receptor serves numerous roles outside of systemic mineral ion homeostasis, ranging from the regulation of hormonal secretion and the activities of various ion channels to the longer term control of gene expression, programmed cell death (apoptosis), and cellular proliferation. In some cases, the CaR on these "nonhomeostatic" cells responds to local changes in Ca(o)(2+) taking place within compartments of the extracellular fluid (ECF) that communicate with the outside environment (e.g., the gastrointestinal tract). In others, localized changes in Ca(o)(2+) within the ECF can originate from several mechanisms, including fluxes of calcium ions into or out of cellular or extracellular stores or across epithelium that absorb or secrete Ca(2+). In any event, the CaR and other receptors/sensors for Ca(o)(2+) and probably for other extracellular ions represent versatile regulators of numerous cellular functions and may serve as important therapeutic targets.
Collapse
Affiliation(s)
- E M Brown
- Endocrine-Hypertension Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
10
|
|
11
|
Abstract
Prostaglandin E(2) is a major renal cyclooxygenase metabolite of arachidonate and interacts with four G protein-coupled E-prostanoid receptors designated EP(1), EP(2), EP(3), and EP(4). Through these receptors, PGE(2) modulates renal hemodynamics and salt and water excretion. The intrarenal distribution and function of EP receptors have been partially characterized, and each receptor has a distinct role. EP(1) expression predominates in the collecting duct where it inhibits Na(+) absorption, contributing to natriuresis. The EP(2) receptor regulates vascular reactivity, and EP(2) receptor-knockout mice have salt-sensitive hypertension. The EP(3) receptor is also expressed in vessels as well as in the thick ascending limb and collecting duct, where it antagonizes vasopressin-stimulated salt and water transport. EP(4) mRNA is expressed in the glomerulus and collecting duct and may regulate glomerular tone and renal renin release. The capacity of PGE(2) to bidirectionally modulate vascular tone and epithelial transport via constrictor EP(1) and EP(3) receptors vs. dilator EP(2) and EP(4) receptors allows PGE(2) to serve as a buffer, preventing excessive responses to physiological perturbations.
Collapse
Affiliation(s)
- M D Breyer
- Division of Nephrology and Departments of Medicine, Veterans Affairs Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
12
|
Yamaguchi T, Chattopadhyay N, Brown EM. G protein-coupled extracellular Ca2+ (Ca2+o)-sensing receptor (CaR): roles in cell signaling and control of diverse cellular functions. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1999; 47:209-53. [PMID: 10582088 DOI: 10.1016/s1054-3589(08)60113-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- T Yamaguchi
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|
13
|
Abstract
The physiological relevance of calcium in many vital processes requires that its concentration in extracellular fluids be kept within a narrow range. The near-constancy of this parameter emphasizes the remarkable sensitivity of cells sensing changes in extracellular calcium concentration to minimal fluctuations (< 2%) and the level of sophistication of the homeostatic system (1). The identification of a cell surface, Ca2+ (polyvalent cation)-sensing receptor (CaR), has shed considerable light on the molecular aspects of hypercalcemia on cell function (2). Activation of the receptor by calcium triggers an intracellular cascade of second messengers producing a variety of biological effects, many of which have yet to be understood. This suggests, for the first time, that Ca2+ can exert its effects in a hormone-like fashion without crossing the plasma membrane. The demonstration that inherited genetic disorders of Ca2+ homeostasis are associated with mutations that reduce or enhance responsiveness of the receptor to extracellular Ca2+ concentration clearly proposes CaR as the main regulator of divalent mineral ion excretion (3). This hypothesis is confirmed by the assessment of the presence of the receptor in all regions involved in Ca2+ homeostasis (e.g., parathyroid glands, kidney, calcitonin-secreting C cells, bone-derived cell lines, and intestine) (1,4-8). Recently, the receptor has also been found in regions not normally involved in mineral ion metabolism, such as the brain, eye, stomach, and pancreas (9-13). This clearly indicates a much broader relevance of CaR in the maintenance of local ionic homeostasis and, possibly, in the involvement in vital processes such as the regulation of cell fate.
Collapse
Affiliation(s)
- D Riccardi
- School of Biological Sciences, University of Manchester, United Kingdom
| | | |
Collapse
|
14
|
Jensen BL, Mann B, Skøtt O, Kurtz A. Differential regulation of renal prostaglandin receptor mRNAs by dietary salt intake in the rat. Kidney Int 1999; 56:528-37. [PMID: 10432392 DOI: 10.1046/j.1523-1755.1999.00564.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND In this study, we tested the hypothesis that prostaglandin (PG) receptor expression in the rat kidney is subject to physiological regulation by dietary salt intake. METHODS Rats were fed diets with 0.02 or 4% NaCl for two weeks. PG receptor expression was assayed in kidney regions and cells by ribonuclease protection assay and reverse transcription-polymerase chain reaction analysis. Functional correlates were studied by measurement of PGE2-induced cAMP formation and renin secretion in juxtaglomerular (JG) cells isolated from animals on various salt intakes. RESULTS EP1 and EP3 receptors were predominantly expressed, and the EP2 receptor was exclusively expressed in the rat kidney medulla. The EP4 receptor was strongly expressed in glomeruli and in renin-secreting JG granular cells. IP receptor transcripts were found mainly in cortex. Maintaining rats on a low- or high-NaCl diet did not affect the expression of EP1 or IP receptors, whereas EP4 transcripts in glomeruli were increased twofold by salt deprivation. Consistent with this, we found that PGE2-evoked cAMP production and renin secretion by JG cells from salt-deprived animals were significantly higher compared with cells obtained from salt-loaded animals. In the outer medulla, EP3 transcripts correlated directly with salt intake, and mRNA abundance was increased twofold by a high-NaCl diet. CONCLUSIONS Our results suggest that subtype-specific, regional changes in PG receptor expression are involved in the renal adaptation to changes in salt intake. The results are in accord with the general concept that renocortical PGE2 stimulates renin secretion and maintains renal blood flow during low-salt states, whereas medullary PGE2 promotes salt excretion in response to a high salt intake.
Collapse
MESH Headings
- Animals
- Antisense Elements (Genetics)
- Cells, Cultured
- Cloning, Molecular
- Cyclic AMP/analysis
- DNA, Complementary
- Dinoprostone/metabolism
- Gene Expression Regulation/drug effects
- Juxtaglomerular Apparatus/cytology
- Juxtaglomerular Apparatus/drug effects
- Juxtaglomerular Apparatus/physiology
- Kidney Glomerulus/chemistry
- Kidney Glomerulus/metabolism
- Kidney Medulla/chemistry
- Kidney Medulla/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- RNA, Messenger/analysis
- Rats
- Rats, Sprague-Dawley
- Receptors, Epoprostenol
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP3 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
- Sodium Chloride/pharmacology
- Sodium, Dietary/pharmacology
Collapse
Affiliation(s)
- B L Jensen
- Institute of Physiology I, University of Regensburg, Germany.
| | | | | | | |
Collapse
|
15
|
Brown EM, Vassilev PM, Quinn S, Hebert SC. G-protein-coupled, extracellular Ca(2+)-sensing receptor: a versatile regulator of diverse cellular functions. VITAMINS AND HORMONES 1999; 55:1-71. [PMID: 9949679 DOI: 10.1016/s0083-6729(08)60933-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- E M Brown
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
16
|
Aarab L, Siaume-Perez S, Chabardès D. Cell-specific coupling of PGE2 to different transduction pathways in arginine vasopressin- and glucagon-sensitive segments of the rat renal tubule. Br J Pharmacol 1999; 126:1041-9. [PMID: 10193786 PMCID: PMC1571213 DOI: 10.1038/sj.bjp.0702390] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The aim of the present study was to investigate the transduction pathways elicited by prostaglandin E2 (PGE2) to inhibit hormone-stimulated adenosine 3':5'-cyclic monophosphate (cyclic AMP) accumulation in the outer medullary collecting duct (OMCD) and medullary thick ascending limb (MTAL) microdissected from the rat nephron. 2. In the OMCD, 0.3 microM PGE2 and low concentrations of Ca2+ ionophores (10 nM ionomycin or 50 nM A23187) inhibited by about 50% a same pool of arginine vasopressin (AVP)-stimulated cyclic AMP content through a same process insensitive to Bordetella pertussis toxin (PTX). 3. Sulprostone, an agonist of the EP1/EP3 subtypes of the PGE2 receptor, decreased AVP-dependent cyclic AMP accumulation in OMCD and MTAL samples. The concentration eliciting half-maximal inhibition was of about 50 nM in OMCD and 0.1 nM in MTAL. 4. In MTAL, 1 nM sulprostone and PGE2 inhibited by about 90% a same pool of AVP-dependent cyclic AMP content through a PTX-sensitive, Ca2+ -independent pathway. 5. In the OMCD, PGE2 decreased by about 50% glucagon-dependent cyclic AMP synthesis by a process sensitive to PTX and Ca2+ -independent. Sulprostone 1 nM induced the same level of inhibition. 6. These results demonstrate that PGE2 decrease hormone-dependent cyclic AMP accumulation through a G(alpha)i-mediated inhibition of adenylyl cyclase activity in MTAL cells and glucagon-sensitive cells of the OMCD or through a PTX-insensitive increase of intracellular Ca2+ concentration in AVP-sensitive cells of the OMCD.
Collapse
Affiliation(s)
- Lotfi Aarab
- CNRS URA 1859, Service de Biologie Cellulaire, Département de Biologie Cellulaire et Moléculaire, CEA Saclay, France
| | | | - Danielle Chabardès
- CNRS URA 1859, Service de Biologie Cellulaire, Département de Biologie Cellulaire et Moléculaire, CEA Saclay, France
- Author for correspondence:
| |
Collapse
|
17
|
Recent Insights Into the Coordinate Regulation of Body Water and Divalent Mineral Ion Metabolism. Am J Med Sci 1998. [DOI: 10.1016/s0002-9629(15)40430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Baum MA, Harris HW. Recent insights into the coordinate regulation of body water and divalent mineral ion metabolism. Am J Med Sci 1998; 316:321-8. [PMID: 9822114 DOI: 10.1097/00000441-199811000-00005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Traditionally, arginine vasopressin modulation of renal water, sodium, and urea excretion has been considered somewhat in isolation from factors that control divalent mineral ion homeostasis. Similarly, previous considerations of divalent mineral ion metabolism have focused mainly on the role of hormones, eg, parathyroid hormone and various forms of vitamin D, as principal modifiers of renal calcium handling. Recent data, however, have now suggested the existence of novel linkages that coordinate control of water and divalent mineral ion homeostasis. This article summarizes these data and highlights the fundamental roles of the extracellular calcium polyvalent cation-sensing receptor (CaR) as an integrator of water and divalent mineral ion homeostasis on a cellular, organ-specific, and whole-body basis. Organs where CaRs may integrate water and divalent mineral ion metabolism include endocrine tissues that express CaRs, the brain, various nephron segments of the kidney, bone, and the gastrointestinal tract. These new data suggest that considerable regulatory overlap exists between water and divalent mineral ion homeostasis.
Collapse
Affiliation(s)
- M A Baum
- Division of Nephrology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
19
|
Bapty BW, Dai LJ, Ritchie G, Canaff L, Hendy GN, Quamme GA. Mg2+/Ca2+ sensing inhibits hormone-stimulated Mg2+ uptake in mouse distal convoluted tubule cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:F353-60. [PMID: 9729507 DOI: 10.1152/ajprenal.1998.275.3.f353] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The distal convoluted tubule plays a significant role in renal magnesium conservation. An immortalized mouse distal convoluted tubule (MDCT) cell line has been extensively used to study the cellular mechanisms of magnesium transport in this nephron segment. MDCT cells possess an extracellular polyvalent cation-sensing mechanism responsive to Mg2+, Ca2+, and neomycin. The present studies determined the effect of Mg2+/Ca2+ sensing on hormone-mediated cAMP formation and Mg2+ uptake in MDCT cells. MDCT cells were Mg2+ depleted by culturing in Mg2+-free media for 16 h, and Mg2+ uptake was measured by microfluorescence after placing the depleted cells in 1.5 mM MgCl2. The mean rate of Mg2+ uptake was 164 +/- 5 nM/s in control MDCT cells. Activation of Mg2+/Ca2+ sensing with neomycin did not affect basal Mg2+ uptake (155 +/- 5 nM/s). We have previously reported that treatment of MDCT cells with either glucagon or arginine vasopressin (AVP) stimulated Mg2+ entry. In the present studies, the addition of extracellular Mg2+ or Ca2+ inhibited glucagon- and AVP-stimulated cAMP formation and Mg2+ uptake in concentration-dependent manner with half-maximal concentrations of approximately 1.5 and 3.0 mM, respectively. Exogenous cAMP or forskolin stimulated Mg2+ uptake in the presence of Mg2+/Ca2+ sensing activation. We infer from these studies that Mg2+/Ca2+-sensing mechanisms located in the distal convoluted tubule may play a role in control of distal magnesium absorption.
Collapse
Affiliation(s)
- B W Bapty
- Department of Medicine, University of British Columbia, University Hospital, Koerner Pavilion, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | | | | | | | |
Collapse
|
20
|
Breyer MD, Zhang Y, Guan YF, Hao CM, Hebert RL, Breyer RM. Regulation of renal function by prostaglandin E receptors. KIDNEY INTERNATIONAL. SUPPLEMENT 1998; 67:S88-94. [PMID: 9736261 DOI: 10.1046/j.1523-1755.1998.06718.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Prostaglandin E2 is the major cyclooxygenase product of arachidonic acid metabolism produced along the nephron. This autacoid interacts with four distinct, G-protein-coupled E-prostanoid receptors designated EP1-EP4. The intrarenal distribution of each receptor has been mapped and the consequences of receptor activation examined. EP3 receptor mRNA is expressed highly in the medullary thick ascending limb (mTAL) and collecting duct (CD). EP3 receptor activation inhibits cAMP generation via Gi, thus inhibiting vasopressin-stimulated water reabsorption in the CD. EP3 receptor activation also may contribute to PGE2-mediated inhibition of NaCl absorption in the mTAL. The EP1 receptor is coupled to increased cell [Ca2+]. EP1 mRNA expression is restricted to the CD, and receptor activation inhibits Na+ absorption. PGE2 also increases cAMP generation in the cortical thick ascending limb and CD; this may be due to EP4 receptor activation. EP4 mRNA is readily detected in the CD with little detectable EP2 expression. The EP4 receptor appears to be expressed both on luminal and basolateral membranes. EP4 receptor activation also may contribute to the regulation of renin release by the juxtaglomerular apparatus. The consequences of renal EP-receptor activation for salt and water balance may be determined by the relative renal expression of each of these receptors.
Collapse
Affiliation(s)
- M D Breyer
- Department of Veterans Affairs Medical Center and Vanderbilt University, Nashville, Tennessee, USA.
| | | | | | | | | | | |
Collapse
|
21
|
De Jesus Ferreira MC, Bailly C. Extracellular Ca2+ decreases chloride reabsorption in rat CTAL by inhibiting cAMP pathway. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:F198-203. [PMID: 9691008 DOI: 10.1152/ajprenal.1998.275.2.f198] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The effect of activation of the Ca2+-sensing receptor on net Cl flux (JCl) has been investigated on microperfused cortical (C) thick ascending limb (TAL) from rat kidney. Increasing bath Ca2+ from 0.5 to 3 mM or adding 200 microM of the specific Ca2+-sensing receptor agonist neomycin reduced basal as well as antidiuretic hormone (ADH)-stimulated JCl by 27.7 +/- 5.0% and 25.9 +/- 4.1%, respectively. JCl remained unchanged in time control tubules. The effect of neomycin/Ca2+ on JCl was blocked by two protein kinase A inhibitors, H-9 or H-89, but not by a protein kinase C inhibitor, GF-109203X, regardless of whether ADH was present or not. Moreover, H-89 decreased basal JCl and prevented a further effect of 3 mM Ca2+. When JCl was increased by 8-bromo-cAMP plus IBMX, no effect of 3 mM Ca2+ was observed. Inhibitors of phospholipase A2 and cytochrome P-450 monooxygenase failed to modify the effect of 3 mM Ca2+, although these agents dampened significantly the inhibitory effect of bradykinin on medullary TAL. We conclude that extracellular Ca2+ decreases basal and ADH-stimulated Cl reabsorption in CTAL by inhibiting the cAMP pathway, independently of protein kinase C or phospholipase A2 stimulation.
Collapse
Affiliation(s)
- M C De Jesus Ferreira
- Unité de Recherche Associée Centre National de la Recherche Scientifique 1859, Département de Biologie Cellulaire et Moléculaire, Commissariat à l'Energie Atomique-Saclay, 91191 Gif-sur-Yvette, France
| | | |
Collapse
|
22
|
de Jesus Ferreira MC, Héliès-Toussaint C, Imbert-Teboul M, Bailly C, Verbavatz JM, Bellanger AC, Chabardès D. Co-expression of a Ca2+-inhibitable adenylyl cyclase and of a Ca2+-sensing receptor in the cortical thick ascending limb cell of the rat kidney. Inhibition of hormone-dependent cAMP accumulation by extracellular Ca2+. J Biol Chem 1998; 273:15192-202. [PMID: 9614133 DOI: 10.1074/jbc.273.24.15192] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Ca2+-sensing receptor protein and the Ca2+-inhibitable type 6 adenylyl cyclase mRNA are present in a defined segment of the rat renal tubule leading to the hypothesis of their possible functional co-expression in a same cell and thus to a possible inhibition of cAMP content by extracellular Ca2+. By using microdissected segments, we compared the properties of regulation of extracellular Ca2+-mediated activation of Ca2+ receptor to those elicited by prostaglandin E2 and angiotensin II. The three agents inhibited a common pool of hormone-stimulated cAMP content by different mechanisms as follows. (i) Extracellular Ca2+, coupled to phospholipase C activation via a pertussis toxin-insensitive G protein, induced a dose-dependent inhibition of cAMP content (1.25 mM Ca2+ eliciting 50% inhibition) resulting from both stimulation of cAMP hydrolysis and inhibition of cAMP synthesis; this latter effect was mediated by capacitive Ca2+ influx as well as release of intracellular Ca2+. (ii) Angiotensin II, coupled to the same transduction pathway, also decreased cAMP content; however, its inhibitory effect on cAMP was mainly accounted for by an increase of cAMP hydrolysis, although angiotensin II and extracellular Ca2+ can induce comparable release of intracellular Ca2+. (iii) Prostaglandin E2, coupled to pertussis toxin-sensitive G protein, inhibited the same pool of adenylyl cyclase units as extracellular Ca2+ but by a different mechanism. The functional properties of the adenylyl cyclase were similar to those described for type 6. The results establish that the co-expression of a Ca2+-inhibitable adenylyl cyclase and of a Ca2+-sensing receptor in a same cell allows an inhibition of cAMP accumulation by physiological concentrations of extracellular Ca2+.
Collapse
Affiliation(s)
- M C de Jesus Ferreira
- Service de Biologie Cellulaire, Département de Biologie Cellulaire et Moléculaire, CEA Saclay, France
| | | | | | | | | | | | | |
Collapse
|
23
|
Bapty BW, Dai LJ, Ritchie G, Jirik F, Canaff L, Hendy GN, Quamme GA. Extracellular Mg2(+)- and Ca2(+)-sensing in mouse distal convoluted tubule cells. Kidney Int 1998; 53:583-92. [PMID: 9507202 DOI: 10.1046/j.1523-1755.1998.00790.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An immortalized cell line (designated MDCT) has been extensively used to investigate the cellular mechanisms of electrolyte transport within the mouse distal convoluted tubule. Mouse distal convoluted tubule cells possess many of the functional characteristics of the in vivo distal convoluted tubule. In the present study, we show that MDCT cells also possess a polyvalent cation-sensing mechanism that is responsive to extracellular magnesium and calcium. Southern hybridization of reverse transcribed-polymerase chain reaction (RT-PCR) products, sequence determination and Western analysis indicated that the calcium-sensing receptor (Casr) is expressed in MDCT cells. Using microfluorescence of single MDCT cells to determine cytosolic Ca2+ signaling, it was shown that the polyvalent cation-sensing mechanism is sensitive to extracellular magnesium concentration ([Mg2+]o) and extracellular calcium concentration ([Ca2+]o) in concentration ranges normally observed in the plasma. Moreover, both [Mg2+]o and [Ca2+]o were effective in generating intracellular Ca2+ transients in the presence of large concentrations of [Ca2+]o and [Mg2+]o, respectively. These responses are unlike those observed for the Casr in the parathyroid gland. Finally, activation of the polycation-sensitive mechanism with either [Mg2+]o or [Ca2+]o inhibited parathyroid hormone-, calcitonin-, glucagon- and arginine vasopressin-stimulated cAMP release in MDCT cells. These studies indicate that immortalized MDCT cells possess a polyvalent cation-sensing mechanism and emphasize the important role this mechanism plays in modulating intracellular signals in response to changes in [Mg2+]o as well as in [Ca2+]o.
Collapse
Affiliation(s)
- B W Bapty
- Department of Medicine, University of British Columbia, University Hospital, Vancouver, Canada
| | | | | | | | | | | | | |
Collapse
|
24
|
Takeda M, Kobayashi M, Shirato I, Endou H. Involvement of macromolecule synthesis, endonuclease activation and c-fos expression in cisplatin-induced apoptosis of mouse proximal tubule cells. Toxicol Lett 1998; 94:83-92. [PMID: 9574805 DOI: 10.1016/s0378-4274(97)00102-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have previously demonstrated that cisplatin-induced nephrotoxicity is associated with the induction of apoptosis using mouse renal cells derived from the terminal proximal tubule (S3) which is the major target site of cisplatin-induced injury. The purpose of this study was to elucidate the intracellular mechanisms leading to the cisplatin-induced apoptosis of S3 cells. Actinomycin D (an inhibitor of RNA synthesis), cycloheximide (an inhibitor of protein synthesis) and aurintricarboxylic acid (an endonuclease inhibitor) reduced the extent of DNA fragmentation, a biochemical parameter of apoptosis, in cisplatin-treated S3 cells. Furthermore, cisplatin-induced apoptosis of S3 cells was accompanied by an increase in the level of c-fos mRNA expression, which is inhibited by pretreatment of the cells with actinomycin D, but not with cycloheximide or aurintricarboxylic acid. In contrast, outer medullary collecting duct cells treated with cisplatin exhibited morphological changes characteristic of apoptosis and an increase in the level of c-fos mRNA expression, but no increase in the extent of DNA fragmentation. In conclusion, the synthesis of macromolecules such as RNA and protein, endonuclease activation and c-fos expression appear to be involved in the intracellular pathways leading to the induction of apoptosis in cisplatin-treated S3 cells. In addition, the response to cisplatin may be different in different cells.
Collapse
Affiliation(s)
- M Takeda
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
25
|
Sands JM, Naruse M, Baum M, Jo I, Hebert SC, Brown EM, Harris HW. Apical extracellular calcium/polyvalent cation-sensing receptor regulates vasopressin-elicited water permeability in rat kidney inner medullary collecting duct. J Clin Invest 1997; 99:1399-405. [PMID: 9077550 PMCID: PMC507956 DOI: 10.1172/jci119299] [Citation(s) in RCA: 238] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
During antidiuresis, increases in vasopressin (AVP)-elicited osmotic water permeability in the terminal inner medullary collecting duct (tIMCD) raise luminal calcium concentrations to levels (> or = 5 mM) above those associated with the formation of calcium-containing precipitates in the urine. Calcium/polycation receptor proteins (CaRs) enable cells in the parathyroid gland and kidney thick ascending limb of Henle to sense and respond to alterations in serum calcium. We now report the presence of an apical CaR in rat kidney tIMCD that specifically reduces AVP-elicited osmotic water permeability when luminal calcium rises. Purified tIMCD apical membrane endosomes contain both the AVP-elicited water channel, aquaporin 2, and a CaR. In addition, aquaporin 2-containing endosomes also possess stimulatory (G(alpha q)/G(alpha 11) and inhibitory (G(alpha i1, 2, and 3)) GTP binding proteins reported previously to interact with CaRs as well as two specific isoforms (delta and zeta) of protein kinase C. Immunocytochemistry using anti-CaR antiserum reveals the presence of CaR protein in both rat and human collecting ducts. Together, these data provide support for a unique tIMCD apical membrane signaling mechanism linking calcium and water metabolism. Abnormalities in this mechanism could potentially play a role in the pathogenesis of renal stone formation.
Collapse
Affiliation(s)
- J M Sands
- Renal Division, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Hebert SC. Extracellular calcium-sensing receptor: implications for calcium and magnesium handling in the kidney. Kidney Int 1996; 50:2129-39. [PMID: 8943500 DOI: 10.1038/ki.1996.539] [Citation(s) in RCA: 168] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- S C Hebert
- Brigham & Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
27
|
KUROKAWA KIYOSHI. How is plasma calcium concentration held constant? Nephrology (Carlton) 1996. [DOI: 10.1111/j.1440-1797.1996.tb00157.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
|
29
|
|
30
|
Hartle JE, Prpic V, Siddhanti SR, Spurney RF, Quarles LD. Differential regulation of receptor-stimulated cyclic adenosine monophosphate production by polyvalent cations in MC3T3-E1 osteoblasts. J Bone Miner Res 1996; 11:789-99. [PMID: 8725176 DOI: 10.1002/jbmr.5650110610] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Extracellular cations have paradoxical trophic and toxic effects on osteoblast function. In an effort to explain these divergent actions, we investigated in MC3T3-E1 osteoblasts if polyvalent cations differentially modulate the agonist-stimulated cyclic adenosine monophosphate (cAMP) pathway, an important regulator of osteoblastic function. We found that a panel of cations, including gadolinium, aluminum, calcium, and neomycin, inhibited prostaglandin E1 (PGE)-stimulated cAMP accumulation but paradoxically potentiated parathyroid hormone (PTH)-stimulated cAMP production. In contrast, these cations had no effect on forskolin- or cholera toxin-induced increases in cAMP, suggesting actions proximal to adenylate cyclase and possible modulation of receptor interactions with G proteins. Phorbol 12-myristate 13-acetated (PMA) mimicked the effects of cations on PGE1- and PTH-stimulated cAMP accumulation in MC3T3-E1 cells, respectively, diminishing and augmenting the responses. Moreover, down-regulation of protein kinase C (PKC) by overnight treatment with PMA prevented gadolinium (Gd3+) from attenuating PGE1- and enhancing PTH-stimulated cAMP production, indicating involvement of PKC-dependent pathways. Cations, however, activated signal transduction pathways not coupled to phosphatidylinositol-specific phospholipase C (PI-PLC), since there was no corresponding increase in inositol phosphate formation or intracellular calcium concentrations. In addition, pertussis toxin treatment failed to prevent Gd(3+)-mediated suppression of PGE1-stimulated cAMP, suggesting actions independent of Gm. Thus, polyvalent cations may either stimulate or inhibit hormone-mediated cAMP accumulation in osteoblasts. These differential actions provide a potential explanation for the paradoxical trophic and toxic effects of cations on osteoblast function that occur in vivo under different hormonal conditions.
Collapse
Affiliation(s)
- J E Hartle
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | |
Collapse
|
31
|
Abstract
Extracellular or plasma calcium ion concentration is held constant at 5 mg/dl through the actions of parathyroid hormone (PTH), vitamin D, and calcitonin, on their target organs, kidney and bone. The thresholds of renal tubular calcium reabsorption and bone resorption and formation are both set at 5 mg/dl. The set point of PTH secretion is also set at 5 mg/dl plasma calcium ion. Therefore, the sensing system (parathyroid cell) and the effectors, kidney and bone, are all set to maintain plasma calcium at 5 mg/dl, perhaps through membrane-bound calcium sensor proteins. The effectiveness of this system depends upon the presence of bone remodeling, which allows a swift shift of plasma calcium from and to bone in response to PTH and calcitonin, respectively. In this regard, directing hematopoiesis to bone marrow that provides bone resorbing osteoclasts is critical. It is likely that this shift of hematopoiesis occurs through evolution at the transition from the aquatic to the terrestrial life, and this event is directed by expression of "homing molecule" in bone marrow stromal cells. This brief review provides a factual and conceptual framework of the current understanding of the milieu interieur of the calcium ion.
Collapse
Affiliation(s)
- K Kurokawa
- First Department of Internal Medicine, University of Tokyo Faculty of Medicine, Japan
| |
Collapse
|
32
|
Brown EM, Hebert SC. A cloned extracellular Ca(2+)-sensing receptor: molecular mediator of the actions of extracellular Ca2+ on parathyroid and kidney cells? Kidney Int 1996; 49:1042-6. [PMID: 8691723 DOI: 10.1038/ki.1996.152] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- E M Brown
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
33
|
Ohtomo Y, Aperia A, Sahlgren B, Johansson BL, Wahren J. C-peptide stimulates rat renal tubular Na+, K(+)-ATPase activity in synergism with neuropeptide Y. Diabetologia 1996; 39:199-205. [PMID: 8635672 DOI: 10.1007/bf00403963] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This study was performed in order to test the hypothesis that the connecting peptide of proinsulin, C-peptide, might in itself possess biological activity. Renal tubular Na+, K(+)-ATPase, which is a well-established target for many peptide hormones, was chosen as a model. Rat C-peptide (I) was found to stimulate Na+, K(+)-ATPase activity in single, proximal convoluted tubules dissected from rat kidneys. C-peptide increased the Na+ affinity of the enzyme and all subsequent studies were performed at non-saturating Na+ concentrations. C-peptide stimulation of Na+, K(+)-ATPase activity occurred in a concentration-dependent manner in the dose range 10(-8)-10(-6) mol/l. The presence of neuropeptide Y, 5 x 10(-9) mol/l, enhanced this effect and stimulation of Na+, K(+)-ATPase activity then occurred in the C-peptide dose range 10(-11)-10(-8) mol/l. C-peptide stimulation of Na+, K(+)-ATPase activity was abolished in tubules pretreated with pertussis toxin. It was also abolished in the presence of FK 506, a specific inhibitor of the Ca2(+)-calmodulin-dependent protein phosphatase 2B. These results indicate that C-peptide stimulates Na+, K(+)-ATPase activity, probably by activating a receptor coupled to a pertussis toxin-sensitive G-protein with subsequent activation of Ca2(+)-dependent intracellular signalling pathways.
Collapse
Affiliation(s)
- Y Ohtomo
- Department of Woman and Child Health, St. Göran's Children's Hospital, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
34
|
Brown EM, Segre GV, Goldring SR. Serpentine receptors for parathyroid hormone, calcitonin and extracellular calcium ions. BAILLIERE'S CLINICAL ENDOCRINOLOGY AND METABOLISM 1996; 10:123-61. [PMID: 8734454 DOI: 10.1016/s0950-351x(96)80346-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The cloning of the receptors for PTH, CT and extracellular calcium ions represents a significant advance in the elucidation of the mechanisms through which extracellular calcium ions are regulated. All are members of the superfamily of GPCR, and the inclusion of the Ca2+o-sensing receptor in this superfamily documents that extracellular calcium ions can serve as an extracellular first messenger, in addition to subserving their better known role as a key intracellular second messenger. Furthermore, it has proved possible to identify several human diseases that result from inactivating or activating mutations in the PTH or Ca2+o-sensing receptor. Finally, the availability of these cloned receptors will enable many more studies on structure-function relationships for these receptors as well as clarifying their tissue distribution, regulation and roles in health and disease. It may also be possible to design novel therapeutic agents that permit manipulation of the receptors when their function is abnormal.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- GTP-Binding Proteins/physiology
- Humans
- Mutation
- Receptor, Parathyroid Hormone, Type 1
- Receptors, Calcitonin/chemistry
- Receptors, Calcitonin/genetics
- Receptors, Calcitonin/physiology
- Receptors, Calcium-Sensing
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Receptors, Parathyroid Hormone/chemistry
- Receptors, Parathyroid Hormone/genetics
- Receptors, Parathyroid Hormone/physiology
- Second Messenger Systems
Collapse
Affiliation(s)
- E M Brown
- Endocrine-Hypertension Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
35
|
Abstract
The importance of intracellular calcium in regulating cell function is well recognized. No less important, but less well understood (and probably appreciated), is the fundamental role played by extracellular calcium, Ca2+o, in the modulation of cell function. The recent cloning of Ca2+o-sensing, G-protein-coupled receptors from bovine (and human) parathyroid and rat kidney (and brain) has clearly demonstrated that Ca2+o can function as a traditional 'first messenger'. The identification of 'inactivating' and 'activating' mutations in this Ca2+o-sensing receptor in two hypercalcemic disorders and in an autosomal dominant form of hypocalcemia, respectively, has underscored the physiological relevance of this receptor in Ca2+ homeostasis in man. These advances have significantly enhanced our understanding of the molecular mechanisms involved in extracellular calcium sensing in parathyroid and kidney. Moreover, the localization of the Ca2+o-sensing receptor in tissues previously not known to have Ca2+o-sensing capability has suggested novel and potentially quite important roles for Ca2+o in regulating the function of cells not apparently directly involved in Ca2+ homeostasis.
Collapse
Affiliation(s)
- S C Hebert
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | |
Collapse
|
36
|
Brown EM, Pollak M, Seidman CE, Seidman JG, Chou YH, Riccardi D, Hebert SC. Calcium-ion-sensing cell-surface receptors. N Engl J Med 1995; 333:234-40. [PMID: 7791841 DOI: 10.1056/nejm199507273330407] [Citation(s) in RCA: 177] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- E M Brown
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Firsov D, Aarab L, Mandon B, Siaume-Perez S, de Rouffignac C, Chabardès D. Arachidonic acid inhibits hormone-stimulated cAMP accumulation in the medullary thick ascending limb of the rat kidney by a mechanism sensitive to pertussis toxin. Pflugers Arch 1995; 429:636-46. [PMID: 7792141 DOI: 10.1007/bf00373984] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The possible regulation of adenosine 3',5'-cyclic monophosphate (cAMP) accumulation by arachidonic acid (AA) was studied in segments, microdissected from the rat kidney, which are sensitive to arginine vasopressin (AVP). In the presence of 5 microM indomethacin, the addition of 5 microM AA did not impair AVP-dependent cAMP accumulation (measured during 4 min at 35 degrees C) in the cortical or outer medullary collecting tubule, but decreased this response in the thick ascending limb with an inhibition much more pronounced in the medullary portion (MTAL) than in the cortical portion. In MTAL, the response to 10 nM AVP was inhibited by 34.4 +/- 9.6% (SEM) and 65.8 +/- 5.4% with 1 microM and 5 microM AA, respectively, N = 5 experiments. AVP-, glucagon- and calcitonin-sensitive cAMP levels in MTAL were inhibited by 5 microM AA to a similar extent. AA-induced inhibition was unaffected by the presence of inhibitors of AA metabolism: (1) either 10 microM indomethacin or 50 microM ibuprofen added to all media; (2) a 10-min pre-incubation and a 4-min incubation of MTAL samples with 10 microM eicosa-5,8,11,14-tetrayonic acid, (3) a 1-h preincubation with either 30 microM SKF-525A, 20 microM ketoconazole, or 20 microM nordihydroguariaretic acid. In contrast to AA, 11 other saturated or unsaturated fatty acids had no inhibitory effect on the AVP-dependent cAMP level. In fura-2-loaded MTAL samples, AA induced a slow increase of the intracellular calcium concentration ([Ca2+]i) which reached 21.0 +/- 3.8 nM and 92.9 +/- 21.4 nM over basal values (n = 11) at 2 min and 4 min, respectively, after the beginning of the superfusion of 5 microM AA. AA-induced inhibition of AVP-dependent cAMP accumulation was due neither to the increase in [Ca2+]i elicited by AA, nor to an activation of protein kinase C because this inhibition: (1) was not blocked when MTAL samples were incubated either in zero Ca2+ medium, or in the presence of 1,2-bis(2-aminophenoxy)ethane-N, N, N', N'-tetraacetic acid (BAPTA) to chelate [Ca2+]i, and (2) it was not reproduced by a pre-treatment of MTAL segments with a phorbol ester. Pre-incubation of MTAL (6 h at 35 degrees C) with 500 ng/ml pertussis toxin (PTX) prevented AA-induced inhibition: in the presence of PTX inhibition was 24.7 +/- 6.6% vs 10 nM AVP, as compared to 81.6 +/- 4.0% in control groups, i.e in the absence of PTX, N = 6.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- D Firsov
- Laboratoire de Physiologie Cellulaire, Collège de France, Paris
| | | | | | | | | | | |
Collapse
|
38
|
Brown EM, Pollak M, Hebert SC. Sensing of extracellular Ca2+ by parathyroid and kidney cells: cloning and characterization of an extracellular Ca(2+)-sensing receptor. Am J Kidney Dis 1995; 25:506-13. [PMID: 7872334 DOI: 10.1016/0272-6386(95)90118-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The ability of the parathyroid cell to sense minute fluctuations in the extracellular ionized calcium concentration (Ca2+ o) is essential for maintaining mineral ion homeostasis. However, the mechanism(s) through which the parathyroid cell and other cells recognize and respond to changes in Ca2+ o has remained unclear. We recently isolated a cDNA encoding a Ca2+ o-sensing receptor from bovine parathyroid using expression cloning in Xenopus laevis oocytes. The receptor shows pharmacologic properties that are almost identical to those of the receptor on the parathyroid cell and, like the latter, stimulates phospholipase C in a G-protein-dependent manner. The amino acid sequence of the cloned receptor deduced from this cDNA predicts a protein with a molecular mass of 121 kd, which has three principal structural domains. The first is a 613 amino acid, putatively extracellular amino terminus which has several regions rich in acidic amino acids that may potentially be involved in binding Ca2+ and other polycationic agonists. The second comprises seven membrane-spanning segments that are characteristic of the superfamily of G-protein-coupled receptors, and the third is a 222 amino acid cytoplasmic tail. Transcripts for this Ca2+ o-sensing receptor are present in the parathyroid as well as in the kidney, thyroid, and brain. We next investigated the hypercalcemic disorders, familial hypocalciuric hypercalcemia and neonatal severe hyperparathyroidism, as possible examples of inherited abnormalities in this Ca2+ o-sensing receptor, since both disorders show abnormal Ca2+ o-sensing and/or handling in the kidney and parathyroid.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- E M Brown
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02186
| | | | | |
Collapse
|
39
|
Riccardi D, Park J, Lee WS, Gamba G, Brown EM, Hebert SC. Cloning and functional expression of a rat kidney extracellular calcium/polyvalent cation-sensing receptor. Proc Natl Acad Sci U S A 1995; 92:131-5. [PMID: 7816802 PMCID: PMC42831 DOI: 10.1073/pnas.92.1.131] [Citation(s) in RCA: 372] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The maintenance of a stable extracellular concentration of ionized calcium depends on the integrated function of a number of specialized cells (e.g., parathyroid and certain kidney epithelial cells). We recently identified another G protein-coupled receptor (BoPCaRI) from bovine parathyroid that responds to changes in extracellular Ca2+ within the millimolar range and provides a key mechanism for regulating the secretion of parathyroid hormone. Using an homology-based strategy, we now report the isolation of a cDNA encoding an extracellular Ca2+/polyvalent cation-sensing receptor (RaKCaR) from rat kidney. The predicted RaKCaR protein shares 92% identity with BoPCaR1 receptor and features a seven membrane-spanning domain, characteristic of the G protein-coupled receptors, which is preceded by a large hydrophilic extracellular NH2 terminus believed to be involved in cation binding. RaKCaR cRNA-injected Xenopus oocytes responded to extracellular Ca2+, Mg2+, Gd3+, and neomycin with characteristic activation of inositol phospholipid-dependent, intracellular Ca(2+)-induced Cl- currents. In rat kidney, Northern analysis revealed RaKCaR transcripts of 4 and 7 kb, and in situ hybridization showed localization primarily in outer medulla and cortical medullary rays. Our results provide important insights into the molecular structure of an extracellular Ca2+/polyvalent cation-sensing receptor in rat kidney and provide another basis on which to understand the role of extracellular divalent cations in regulating kidney function in mineral metabolism.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Blotting, Northern
- Chlorides/metabolism
- Cloning, Molecular
- Female
- Gene Expression
- Gene Library
- In Situ Hybridization
- Kidney/metabolism
- Male
- Molecular Sequence Data
- Oocytes/physiology
- Organ Specificity
- Protein Structure, Secondary
- RNA, Complementary
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptors, Calcium-Sensing
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/physiology
- Xenopus laevis
Collapse
Affiliation(s)
- D Riccardi
- Laboratory of Molecular Physiology and Biophysics, Brigham and Women's Hospital, Boston, MA
| | | | | | | | | | | |
Collapse
|
40
|
Ohtomo Y, Meister B, Hökfelt T, Aperia A. Coexisting NPY and NE synergistically regulate renal tubular Na+, K(+)-ATPase activity. Kidney Int 1994; 45:1606-13. [PMID: 7523751 DOI: 10.1038/ki.1994.211] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The sympathetic renal nerves are of central importance for the regulation of sodium balance. Sodium excretion decreases following renal nerve activation and increases following denervation. These effects have been attributed to norepinephrine (NE) acting on alpha-adrenergic receptors. In the present study, using isolated permeabilized rat renal proximal convoluted tubule (PCT) cells, neuropeptide Y (NPY) was shown to stimulate Na+, K(+)-ATPase activity. This 36-amino acid peptide is a messenger molecule in the sympathetic nervous system which is co-stored with NE and dopamine-beta-hydroxylase (DBH), the NE synthesizing enzyme in the renal nerves. The effect is likely to be mediated via the NPY Y2 receptor, a pertussis toxin (PTX)-sensitive G-protein, and calcium. It is partially antagonized by alpha-adrenergic antagonists, and enhanced by the subthreshold doses of alpha-adrenergic agonists. Our results suggest an important role for this peptide in the regulation of the sodium balance in the kidney.
Collapse
Affiliation(s)
- Y Ohtomo
- Department of Pediatrics, St. Göran's Children's Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
41
|
Baudouin-Legros M, Bouthier M, Teulon J. [Arginine]vasopressin hydrolyses phosphoinositides in the medullary thick ascending limb of mouse nephron. Pflugers Arch 1993; 425:381-9. [PMID: 8134254 DOI: 10.1007/bf00374862] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
NaCl reabsorption across the thick ascending limb of Henle's loop (TAL) is stimulated by several hormones, in particular vasopressin acting through V2 receptors and cyclic AMP production. This study used suspensions of medullary TAL (mTAL) tubules from the mouse nephron to investigate the possibility that, besides activating adenylyl cyclase, vasopressin also stimulates phospholipase C via V1 receptor occupancy. Two different methods, phosphoinositide labelling and inositol trisphosphate (InsP3) radioimmunoassay, were used to show that [arginine]vasopressin (AVP) rapidly stimulated the formation of InsP3, which peaked at 200%-250% of control within the first minute of incubation with 10 nmol/l vasopressin at 37 degrees C, and declined to basal level after 5-10 min. Dose/response curves for InsP3, established at 30 degrees C and 37 degrees C using radioimmunoassay, showed a half-maximal stimulation of InsP3 production at about 1 nmol/l AVP and a maximal response at 10 nmol/l. Similar values were obtained for the response to AVP in terms of cAMP accumulation. InsP3 content in the presence of higher concentrations of AVP (1 mumol/l) was significantly lower (P < 0.001) than in the presence of 10 nmol/l AVP, giving a bell-shaped appearance to the dose/response curve at 37 degrees C but not at 30 degrees C. The V2 receptor agonist, 1-deamino-[8-D-Arg]vasopressin (dAVP) did not stimulate the formation of InsP3, and the V1 receptor antagonist d(CH2)5[Tyr(Me)2]AVP inhibited AVP-induced InsP3 formation, which therefore appeared to be mediated by V1 receptor occupancy. Under the same conditions, AVP also induced the formation of diradylglycerol via V1 receptor activation, with an analogous dose/response curve.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- M Baudouin-Legros
- INSERM U323, Faculté de Médecine Necker-Enfants Malades, Paris, France
| | | | | |
Collapse
|
42
|
Griffiths NM, Brick-Ghannam C, Siaume-Perez S, Chabardès D. Effect of prostaglandin E2 on agonist-stimulated cAMP accumulation in the distal convoluted tubule isolated from the rabbit kidney. Pflugers Arch 1993; 422:577-84. [PMID: 7682323 DOI: 10.1007/bf00374005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The effects of calcitonin, vasoactive intestinal peptide (VIP), parathyroid hormone (PTH) and isoprenaline on intracellular cAMP accumulation were determined in the distal tubule (DCT) microdissected from collagenase-treated rabbit kidney. In DCTb (the initial "bright" portion) calcitonin (10 ng/ml) elicited a highly reproducible response 203.7 +/- 19.1 fmol cAMP mm-1 4 min-1 (SE,N = 13) whereas VIP-induced cAMP accumulation was less and more variable from one experiment to another (1 microM, 97.2 +/- 17.8 fmol mm-1 4 min-1, SE, N = 12). When used in combination, these two agonists were non-additive, indicating stimulation of a single pool of cAMP in DCTb. In DCTg, ("granular") which consists of at least two cell types, PTH (100 nM) elicited a marked, reproducible accumulation of cAMP (154.3 +/- 27.0 fmol mm-1 4 min-1; SE, N = 5). Isoprenaline (1 microM) and VIP (1 microM) induced much smaller increases in cAMP levels 20.9 +/- 2.7 and 29.4 +/- 4.1 fmol mm-1 4 min-1 (SE, N = 5) respectively, and, when used in combination, were non-additive, demonstrating that VIP and isoprenaline are active on the same cell type. In DCTb, prostaglandin E2 (PGE2) inhibited both calcitonin- and VIP-stimulated cAMP accumulation (calcitonin 57.8 +/- 2.7% inhibition, SE, N = 16; VIP, 80.6 +/- 2.1% inhibition, SE, N = 5). The EC50 values for calcitonin were 1.21 +/- 0.33 ng/ml and 1.83 +/- 0.25 ng/ml (SD, N = 3) in the absence and presence of PGE2 (300 nM) respectively with an IC50 for PGE2 of 26.3 +/- 6.3 nM (SE, N = 4).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- N M Griffiths
- Laboratoire de Physiologie Cellulaire, URA 219 CNRS, Collège de France, Paris
| | | | | | | |
Collapse
|
43
|
Pharmacological Identification of Vasopressin Receptors in Isolated Renal Tubule. ACTA ACUST UNITED AC 1993. [DOI: 10.1016/b978-0-12-185275-7.50024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
44
|
Brown EM. Kidney and Bone: Physiological and Pathophysiological Relationships. Compr Physiol 1992. [DOI: 10.1002/cphy.cp080239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Cellular mechanisms of adrenaline-induced hyperpolarization in renal epitheloid MDCK cells. Biochem J 1991; 274 ( Pt 1):243-8. [PMID: 2001240 PMCID: PMC1149944 DOI: 10.1042/bj2740243] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The effects of adrenaline on the potential difference across the cell membrane, on formation of inositol phosphates and on intracellular Ca2+ ([Ca2+]i) were analysed in cells without or with pretreatment with pertussis toxin or phorbol 12-myristate 13-acetate (PMA). In untreated cells, adrenaline leads to a sustained hyperpolarization, a stimulation of Ins(1,4,5)P3 and Ins(1,3,4,5,)P4 formation and a transient increase in [Ca2+]i from 78 +/- 7 to 555 +/- 43 nM, followed by a plateau of 260 +/- 23 microM. In the absence of extracellular Ca2+ the effect of adrenaline on both potential difference and [Ca2+]i is transient. In cells pretreated with pertussis toxin, the effects of adrenaline on InsP3 and [Ca2+]i are still preserved, but the effect on potential difference is transient. In cells pretreated with PMA, the effect of adrenaline on InsP3 formation is severely decreased and that on [Ca2+]i abolished, whereas a transient hyperpolarizing effect is still present. This transient hyperpolarization is abolished by additional pretreatment with pertussis toxin. The observations suggest that adrenaline hyperpolarizes the cell membrane of MDCK cells by several distinct mechanisms. First, adrenaline stimulates the formation of InsP3 and InsP4, which at least in part accounts for the release of intracellular Ca2+ and the entry of Ca2+ from the extracellular fluid. Stimulation of phospholipase C is not mediated by pertussis-toxin-sensitive G-proteins, but apparently is inhibited by activation of protein kinase C. Second, adrenaline hyperpolarizes the cell membrane by a mechanism independent from increase in [Ca2+]i which is sensitive to pertussis toxin but is, at least in part, insensitive to PMA.
Collapse
|
46
|
Chabardès D, Montégut M, Zhou Y, Siaume-Perez S. Two mechanisms of inhibition by prostaglandin E2 of hormone-dependent cell cAMP in the rat collecting tubule. Mol Cell Endocrinol 1990; 73:111-21. [PMID: 1702742 DOI: 10.1016/0303-7207(90)90124-q] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The effect of exogenous prostaglandin E2 (PGE2) on hormone-dependent adenosine 3',5'-cyclic monophosphate (cAMP) accumulation was investigated by microradioimmunoassay in collecting tubules microdissected from the cortex (CCT) or outer medulla (MCT) of the rat kidney. Two phosphodiesterase inhibitors were used: either a xanthine derivative (isobutyl-methylxanthine (IBMX, 1 mM] active on all forms of phosphodiesterase or Ro 20-1724 (50 microM) active on the phosphodiesterase type III. A prostaglandin synthesis inhibitor was added to all media. In the presence of IBMX, 0.3 microM PGE2 inhibited by 39.1% the response induced in the CCT by the beta-adrenergic agonist isoproterenol (1 microM). Under the same experimental conditions, arginine vasopressin (AVP)-stimulated cAMP accumulation in CCT or MCT was not affected by PGE2. In the presence of Ro 20-1724, 0.3 microM PGE2 did not modify the response to 1 nM AVP in CCT but inhibited this response in MCT samples (mean inhibition: 52.7%). The inhibition by PGE2 was dose dependent with a maximum at 0.3 microM, observed for all concentrations of AVP tested (from 50 pM to 1 nM) and did not affect the concentration of AVP inducing half-maximal cAMP accumulation. In a second experimental series performed in the presence of adenosine deaminase, an A1-adenosine agonist [theta)-N6-(R-phenylisopropyl)adenosine (PIA, 0.1 microM] also decreased the response to 1 nM AVP in the MCT. The addition of an A1-adenosine antagonist relieved the effect of PIA but did not modify the inhibition observed with PGE2. Thus PGE2 decreased the synthesis of cAMP in beta-adrenergic sensitive cells in rat CCT and might affect the catabolism of AVP-dependent cAMP level rather than its synthesis in rat MCT.
Collapse
Affiliation(s)
- D Chabardès
- Laboratoire de Physiologie Cellulaire, URA 219 CNRS, Collège de France, Paris
| | | | | | | |
Collapse
|
47
|
Dublineau I, Pradelles P, de Rouffignac C, Elalouf JM. In vitro desensitization of isolated nephron segments to vasopressin. Proc Natl Acad Sci U S A 1990; 87:7583-7. [PMID: 1699229 PMCID: PMC54792 DOI: 10.1073/pnas.87.19.7583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent studies have demonstrated that in vivo administration of 1-deamino-8-D-arginine-vasopressin, an analog of arginine-8-vasopressin, induces homologous desensitization to vasopressin in the thick ascending limb of the loop of Henle. Desensitization has been documented by a decreased physiological response to vasopressin in vivo and by a reduced cAMP accumulation in the cortical thick ascending limb (CTAL). By measuring cAMP content in single isolated medullary thick ascending limbs (MTALs), we now report that desensitization can occur all along the thick ascending limb and, more importantly, that it can also be induced in vitro. In a first series of experiments, we observed that 1 hr after in vivo injection of 1-deamino-8-D-arginine-vasopressin, MTALs were desensitized by 80% to vasopressin, whereas the effects of the other hormones acting on the same cyclase pool (glucagon, calcitonin) were fully maintained. In a second set of experiments, desensitization was induced in vitro by vasopressin, the natural hormone. A 60-min preincubation of MTALs with vasopressin caused a marked (up to 86%) and highly reproducible desensitization. The process was dose and time dependent. The apparent Ka for desensitization was 0.2 nM, and the half-maximal effect was obtained within 20 min. The desensitization induced in vitro by vasopressin was again essentially homologous in nature, with 80% of the maximal stimulation of cAMP accumulation being obtained in the presence of glucagon. Desensitization to vasopressin was observed in the presence and absence of indomethacin, indicating that it is independent of prostaglandin synthesis. It is concluded that (i) vasopressin and its analog 1-deamino-8-D-arginine-vasopressin cause marked desensitization in the CTAL and MTAL and (ii) the low vasopressin concentrations required to induce desensitization and the rapid onset of the process suggest that it has a physiological significance.
Collapse
Affiliation(s)
- I Dublineau
- Service de Biologie Cellulaire, Centre d'Etudes Nucléaires de Saclay, Gif-sur-Yvette, France
| | | | | | | |
Collapse
|
48
|
Teitelbaum I. Cyclic adenosine monophosphate and diacylglycerol. Mutually inhibitory second messengers in cultured rat inner medullary collecting duct cells. J Clin Invest 1990; 86:46-51. [PMID: 2164048 PMCID: PMC296688 DOI: 10.1172/jci114713] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Studies were performed to examine interactions between the adenylyl cyclase (AC) and phospholipase C (PLC) signaling systems in cultured rat inner medullary collecting duct cells. Stimulation of AC by either arginine vasopressin (AVP) or forskolin or addition of exogenous cAMP inhibits epidermal growth factor (EGF)-stimulated PLC. This inhibition is mediated by activation of cAMP-dependent kinase as it is prevented by pretreatment with the A-kinase inhibitor, N-[2-(methylamino)ethyl]-5-isoquinoline-sulfonamide (H8) but not by the C-kinase inhibitor, 1-(5-isoquinolinylsulfonyl)-2-methylpiperazine (H7). Exposure to EGF eliminates AVP-stimulated cAMP generation. This is not mediated by a cyclooxygenase product as inhibition by EGF is observed even in the presence of the cyclooxygenase inhibitor, flurbiprofen. Inhibition by EGF is not due to an increase in inositol trisphosphate (IP3) as exposure of saponin-permeabilized cells to exogenous IP3 is without effect. Inhibition by EGF is prevented by pretreatment with the C-kinase inhibitor, H7, but not by the A-kinase inhibitor, H8. Exposure to the synthetic diacylglycerol (DAG), dioctanoylglycerol, also inhibits AVP-stimulated AC activity; therefore, inhibition by EGF is due to activation of protein kinase C. Thus, in cultured rat inner medullary collecting duct cells, cAMP and DAG function as mutually inhibitory second messengers with each impairing formation of the other.
Collapse
Affiliation(s)
- I Teitelbaum
- Department of Medicine, University of Colorado School of Medicine, Denver 80262
| |
Collapse
|
49
|
Tomita K, Nonoguchi H, Marumo F. Effects of endothelin on peptide-dependent cyclic adenosine monophosphate accumulation along the nephron segments of the rat. J Clin Invest 1990; 85:2014-8. [PMID: 1693379 PMCID: PMC296672 DOI: 10.1172/jci114667] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We investigated the tubular action of endothelin in rat nephron segments. The effects of endothelin on arginine vasopressin (AVP)-, parathyroid hormone-, glucagon-, calcitonin-, and isoproterenol-dependent cAMP accumulation were studied. The following nephron segments were microdissected: glomerulus (Gl), proximal convoluted tubule (PCT), cortical and medullary thick ascending limbs of Henle's loop (cTAL and mTAL, respectively), cortical collecting duct (CCD), outer medullary collecting duct (OMCD), and inner medullary collecting duct (IMCD). Endothelin dose dependently (10(-8)-10(-10)M) inhibited AVP-dependent cAMP accumulation in CCD, OMCD, and IMCD. This effect was independent of the presence or absence of phosphodiesterase inhibitor, 3-isobutyl-1-methylxanthine, Ca channel blocker nicardipine, or indomethacin, but was abolished in the presence of protein kinase C inhibitor H-7. Protein kinase C stimulator dioctanoyl glycerol mimicked the effect of endothelin. On the other hand, endothelin had no inhibitory effect on AVP-dependent cAMP accumulation in cTAL or mTAL, parathyroid hormone-dependent cAMP accumulation in Gl and PCT, or glucagon-, calcitonin-, and isoprotereol-dependent cAMP accumulation in OMCD. We conclude that endothelin specifically inhibits AVP-dependent cAMP accumulation in CCD, OMCD, and IMCD through activating protein kinase C. This effect possibly has a role in maintaining urine volume to counteract the decrease in GFR caused by endothelin itself.
Collapse
Affiliation(s)
- K Tomita
- 2nd Department of Internal Medicine, Tokyo Medical & Dental University, Japan
| | | | | |
Collapse
|
50
|
|