Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 108(25): 10361-6, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21633011

ABSTRACT

The heteromeric inwardly rectifying Kir4.1/Kir5.1 K(+) channel underlies the basolateral K(+) conductance in the distal nephron and is extremely sensitive to inhibition by intracellular pH. The functional importance of Kir4.1/Kir5.1 in renal ion transport has recently been highlighted by mutations in the human Kir4.1 gene (KCNJ10) that result in seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME)/epilepsy, ataxia, sensorineural deafness, and renal tubulopathy (EAST) syndrome, a complex disorder that includes salt wasting and hypokalemic alkalosis. Here, we investigated the role of the Kir5.1 subunit in mice with a targeted disruption of the Kir5.1 gene (Kcnj16). The Kir5.1(-/-) mice displayed hypokalemic, hyperchloremic metabolic acidosis with hypercalciuria. The short-term responses to hydrochlorothiazide, an inhibitor of ion transport in the distal convoluted tubule (DCT), were also exaggerated, indicating excessive renal Na(+) absorption in this segment. Furthermore, chronic treatment with hydrochlorothiazide normalized urinary excretion of Na(+) and Ca(2+), and abolished acidosis in Kir5.1(-/-) mice. Finally, in contrast to WT mice, electrophysiological recording of K(+) channels in the DCT basolateral membrane of Kir5.1(-/-) mice revealed that, even though Kir5.1 is absent, there is an increased K(+) conductance caused by the decreased pH sensitivity of the remaining homomeric Kir4.1 channels. In conclusion, disruption of Kcnj16 induces a severe renal phenotype that, apart from hypokalemia, is the opposite of the phenotype seen in SeSAME/EAST syndrome. These results highlight the important role that Kir5.1 plays as a pH-sensitive regulator of salt transport in the DCT, and the implication of these results for the correct genetic diagnosis of renal tubulopathies is discussed.


Subject(s)
Kidney Tubules/physiology , Kidney Tubules/physiopathology , Phenotype , Potassium Channels, Inwardly Rectifying/metabolism , Acidosis/genetics , Acidosis/physiopathology , Amiloride/pharmacology , Animals , Diuretics/pharmacology , Furosemide/pharmacology , Humans , Hydrochlorothiazide/pharmacology , Hypokalemia/genetics , Hypokalemia/physiopathology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Mice , Mice, Knockout , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Sodium Channel Blockers/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Syndrome , Kir5.1 Channel
2.
Kidney Int ; 80(3): 256-62, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21326170

ABSTRACT

Modern dietary habits are characterized by high-sodium and low-potassium intakes, each of which was correlated with a higher risk for hypertension. In this study, we examined whether long-term variations in the intake of sodium and potassium induce lasting changes in the plasma concentration of circulating steroids by developing a mathematical model of steroidogenesis in mice. One finding of this model was that mice increase their plasma progesterone levels specifically in response to potassium depletion. This prediction was confirmed by measurements in both male mice and men. Further investigation showed that progesterone regulates renal potassium handling both in males and females under potassium restriction, independent of its role in reproduction. The increase in progesterone production by male mice was time dependent and correlated with decreased urinary potassium content. The progesterone-dependent ability to efficiently retain potassium was because of an RU486 (a progesterone receptor antagonist)-sensitive stimulation of the colonic hydrogen, potassium-ATPase (known as the non-gastric or hydrogen, potassium-ATPase type 2) in the kidney. Thus, in males, a specific progesterone concentration profile induced by chronic potassium restriction regulates potassium balance.


Subject(s)
Adrenal Glands/metabolism , Hypokalemia/metabolism , Kidney/metabolism , Potassium, Dietary/metabolism , Progesterone/biosynthesis , Adrenal Glands/drug effects , Adrenal Glands/enzymology , Aldosterone/biosynthesis , Analysis of Variance , Animals , Cell Line , Chronic Disease , Corticosterone/biosynthesis , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Hormone Antagonists/pharmacology , Humans , Hypokalemia/enzymology , Hypokalemia/genetics , Kidney/drug effects , Kidney/enzymology , Male , Mice , Mice, Knockout , Mifepristone/pharmacology , Models, Biological , Potassium, Dietary/administration & dosage , Potassium, Dietary/urine , Progesterone/blood , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Sodium, Dietary/metabolism , Time Factors , Up-Regulation
3.
Proc Natl Acad Sci U S A ; 107(30): 13526-31, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20624970

ABSTRACT

Tissue kallikrein (TK) is a serine protease synthetized in renal tubular cells located upstream from the collecting duct where renal potassium balance is regulated. Because secretion of TK is promoted by K+ intake, we hypothesized that this enzyme might regulate plasma K+ concentration ([K+]). We showed in wild-type mice that renal K+ and TK excretion increase in parallel after a single meal, representing an acute K+ load, whereas aldosterone secretion is not modified. Using aldosterone synthase-deficient mice, we confirmed that the control of TK secretion is aldosterone-independent. Mice with TK gene disruption (TK-/-) were used to assess the impact of the enzyme on plasma [K+]. A single large feeding did not lead to any significant change in plasma [K+] in TK+/+, whereas TK-/- mice became hyperkalemic. We next examined the impact of TK disruption on K+ transport in isolated cortical collecting ducts (CCDs) microperfused in vitro. We found that CCDs isolated from TK-/- mice exhibit net transepithelial K+ absorption because of abnormal activation of the colonic H+,K+-ATPase in the intercalated cells. Finally, in CCDs isolated from TK-/- mice and microperfused in vitro, the addition of TK to the perfusate but not to the peritubular bath caused a 70% inhibition of H+,K+-ATPase activity. In conclusion, we have identified the serine protease TK as a unique kalliuretic factor that protects against hyperkalemia after a dietary K+ load.


Subject(s)
Adaptation, Physiological/physiology , Kidney/physiology , Potassium/metabolism , Tissue Kallikreins/metabolism , Adaptation, Physiological/drug effects , Aldosterone/metabolism , Aldosterone/urine , Animals , Biological Transport , Cytochrome P-450 CYP11B2/deficiency , Cytochrome P-450 CYP11B2/genetics , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Kidney/metabolism , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/physiology , Mice , Mice, Knockout , Potassium/blood , Potassium/urine , Potassium, Dietary/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , Tissue Kallikreins/genetics
4.
J Clin Invest ; 120(6): 1895-904, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484819

ABSTRACT

Ammonia absorption by the medullary thick ascending limb of Henle's loop (MTALH) is thought to be a critical step in renal ammonia handling and excretion in urine, in which it is the main acid component. Basolateral Na+/H+ exchangers have been proposed to play a role in ammonia efflux out of MTALH cells, which express 2 exchanger isoforms: Na+/H+ exchanger 1 (NHE1) and NHE4. Here, we investigated the role of NHE4 in urinary acid excretion and found that NHE4-/- mice exhibited compensated hyperchloremic metabolic acidosis, together with inappropriate urinary net acid excretion. When challenged with a 7-day HCl load, NHE4-/- mice were unable to increase their urinary ammonium and net acid excretion and displayed reduced ammonium medulla content compared with wild-type littermates. Both pharmacologic inhibition and genetic disruption of NHE4 caused a marked decrease in ammonia absorption by the MTALH. Finally, dietary induction of metabolic acidosis increased NHE4 mRNA expression in mouse MTALH cells and enhanced renal NHE4 activity in rats, as measured by in vitro microperfusion of MTALH. We therefore conclude that ammonia absorption by the MTALH requires the presence of NHE4 and that lack of NHE4 reduces the ability of MTALH epithelial cells to create the cortico-papillary gradient of NH3/NH4+ needed to excrete an acid load, contributing to systemic metabolic acidosis.


Subject(s)
Acidosis/metabolism , Kidney/physiology , Loop of Henle/metabolism , Quaternary Ammonium Compounds/metabolism , Rodentia/metabolism , Absorption , Acidosis/genetics , Ammonia/metabolism , Ammonia/urine , Animals , Biological Transport/genetics , Epithelial Cells/metabolism , Female , Kidney/metabolism , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Rodentia/genetics , Specific Pathogen-Free Organisms
5.
Biomol Concepts ; 1(1): 41-7, 2010 May 01.
Article in English | MEDLINE | ID: mdl-25961984

ABSTRACT

Steroids are well-known mediators of many different physiological functions. Their best characterized mechanism of action involves interaction with well-defined nuclear receptors and regulation of gene transcription. However, rapid effects of steroids have been reported which are incompatible with their classical long-term/slow effects. Although the concept of membrane-bound receptors for steroids which can transduce their rapid effects has been proposed many years ago, it is only recently that such proteins have been identified and characterized. In this review, we will discuss recent data regarding the rapid action of progesterone mediated by newly characterized membrane-bound receptors belonging to the progestin and adiponectin receptor family.

6.
FEBS Lett ; 583(10): 1644-8, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19401201

ABSTRACT

Renal functions are regulated by steroid sex hormones, but the exhaustive identification of their receptors along the nephron is still lacking. Here, we have localized all known nuclear or membrane-bound sex hormone receptors and some of their activators along the nephron of male and female mice. Almost all receptors are present in male and female kidney, some of them having very restricted localization. Only one gene tested among 11 (ARA54) exhibits a gender difference in the level of its expression. This first "renal map" of sex steroid receptor expression may serve as a pre-requisite for investigating the role of these hormones on kidney functions.


Subject(s)
Nephrons/metabolism , Receptors, Steroid/metabolism , Animals , Female , Gonadal Steroid Hormones/metabolism , Kidney/metabolism , Male , Mice , RNA, Messenger/metabolism , Receptors, Steroid/analysis
7.
Biochim Biophys Acta ; 1783(12): 2234-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18722485

ABSTRACT

Sex hormones have broader effects than regulating reproductive functions. Recent identification of membrane progestin receptors expressed in kidney prompted us to investigate their putative involvement in the renal effects of this hormone. We first focused our investigations on mPRalpha and gamma by analyzing three parameters 1/ their distribution along the mouse nephron and their subcellular location in native kidney, 2/ the ability of progesterone to stimulate ERK pathway and/or Ca(2+) release from internal stores in native kidney structures and 3/ the cellular localization of mPRalpha and its molecular determinants in heterologous expression system. We observed that 1/ mPRalpha expression is restricted to proximal tubules of both male and female mice whereas mPRgamma exhibits a much broader expression all along the nephron except the glomerulus, 2/ mPRalpha and gamma are not localized at the plasma membrane in native kidney, 3/ this expression does not permit either progesterone-induced ERK phosphorylation or Ca(2+) release and 4/ in HEK transfected cells, mPRalpha localizes in the endoplasmic reticulum (ER) due to a C-terminal ER retention motif (-KXX). Therefore, we have characterized mPRs in kidney but their role in renal physiology remains to be elucidated.


Subject(s)
Cell Membrane/metabolism , Kidney Tubules, Proximal/metabolism , Progesterone/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Progesterone/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Female , Humans , Immunoenzyme Techniques , Kidney/metabolism , Kidney Tubules, Proximal/cytology , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Peptide Fragments , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subcellular Fractions , Triiodobenzoic Acids/pharmacology
8.
J Am Soc Nephrol ; 19(10): 1965-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18650486

ABSTRACT

Although adult kidney cells are quiescent, enlargement of specific populations of epithelial cells occurs during repair and adaptive processes. A prerequisite to the development of regenerative therapeutics is to identify the mechanisms and factors that control the size of specific populations of renal cells. Unfortunately, in most cases, it is unknown whether the growth of cell populations results from transdifferentiation or proliferation and whether proliferating cells derive from epithelial cells or from circulating or resident progenitors. In this study, the mechanisms underlying the enlargement of the acid-secreting cell population in the mouse kidney collecting duct in response to metabolic acidosis was investigated. Acidosis led to two phases of proliferation that preferentially affected the acid-secreting cells of the outer medullary collecting duct. All proliferating cells displayed polarized expression of functional markers. The first phase of proliferation, which started within 24 h and peaked at day 3, was dependent on the overexpression of growth differentiation factor 15 (GDF15) and cyclin D1 and was abolished when phosphatidylinositol-3 kinase and mammalian target of rapamycin were inhibited. During this phase, cells mostly divided along the tubular axis, contributing to tubule lengthening. The second phase of proliferation was independent of GDF15 but was associated with induction of cyclin D3. During this phase, cells divided transversely. In summary, acid-secreting cells proliferate as the collecting duct adapts to metabolic acidosis, and GDF15 seems to be an important determinant of collecting duct lengthening.


Subject(s)
Acidosis, Renal Tubular/metabolism , Acidosis, Renal Tubular/pathology , Cytokines/metabolism , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/pathology , Acid-Base Equilibrium/physiology , Acidosis, Renal Tubular/etiology , Animals , Cell Proliferation , Cell Transdifferentiation/physiology , Cyclin D3 , Cyclins/metabolism , Cytokines/genetics , Female , Growth Differentiation Factor 15 , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
9.
J Am Soc Nephrol ; 17(8): 2153-63, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16825334

ABSTRACT

Recent studies indicate that pendrin, an apical Cl-/HCO3- exchanger, mediates chloride reabsorption in the connecting tubule and the cortical collecting duct and therefore is involved in extracellular fluid volume regulation. The purpose of this study was to test whether pendrin is regulated in vivo primarily by factors that are associated with changes in renal chloride transport, by aldosterone, or by the combination of both determinants. For achievement of this goal, pendrin protein abundance was studied by semiquantitative immunoblotting in different mouse models with altered aldosterone secretion or tubular chloride transport, including NaCl loading, hydrochlorothiazide administration, NaCl co-transporter knockout mice, and mice with Liddle's mutation. The parallel regulation of the aldosterone-regulated epithelial sodium channel (ENaC) was examined as a control for biologic effects of aldosterone. Major changes in pendrin protein expression were found in experimental models that are associated with altered renal chloride transport, whereas no significant changes were detected in pendrin protein abundance in models with altered aldosterone secretion. Moreover, in response to hydrochlorothiazide administration, pendrin was downregulated despite a marked secondary hyperaldosteronism. In contrast, alpha-ENaC was markedly upregulated, and the molecular weight of a large fraction of gamma-ENaC subunits was shifted from 85 to 70 kD, consistent with previous results from rat models with elevated plasma aldosterone levels. These results suggest that factors that are associated with changes in distal chloride delivery govern pendrin expression in the connecting tubule and cortical collecting duct.


Subject(s)
Chloride-Bicarbonate Antiporters/analysis , Chlorides/metabolism , Homeostasis/physiology , Kidney/metabolism , Aldosterone/blood , Animals , Chloride-Bicarbonate Antiporters/antagonists & inhibitors , Chlorides/administration & dosage , Disease Models, Animal , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Homeostasis/drug effects , Hydrochlorothiazide/administration & dosage , Immunoblotting , Immunohistochemistry , Kidney/cytology , Kidney/drug effects , Kidney Tubules, Collecting/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Sodium/metabolism
10.
Eur J Nutr ; 45(5): 298-306, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16649091

ABSTRACT

BACKGROUND: Most studies in animals use diets with several features (for example low-fat, rich in micronutriments), likely to be strongly protective against chronic diseases. AIM OF THE STUDY: The present study, performed in wild type outbred mice, was designed to evaluate the validity of a model of 'westernized' (W) diet reproducing, as closely as possible, the overall composition of an average human regime in western countries RESULTS: In contrast to the standard (S) diet, the W diet triggered a marked increase in adiposity with some characteristics of metabolic syndrome (hypercholesterolemia, hyperinsulinemia...). There was an heterogeneity in the propensity to become obese upon exposure to the W diet in female mice. Overweight mice also presented some disturbances of renal function, such as hyperalbuminuria and hypocitraturia. Mice adapted to the W diet showed a reduction of bone mineral density, especially the non-obese ones. CONCLUSION: These data suggest that a model of westernized diet could be appropriate for exploring the effects of mutations, drugs, or specific nutritional factors in animals and could be more relevant for human situations.


Subject(s)
Diet , Disease Models, Animal , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Osteoporosis/epidemiology , Animals , Diet/adverse effects , Female , Humans , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Mice , Obesity/etiology , Obesity/metabolism , Osteoporosis/etiology , Osteoporosis/metabolism , Random Allocation , Urinalysis , Weight Gain
11.
Pflugers Arch ; 451(6): 769-75, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16208521

ABSTRACT

Rat collecting ducts display either an ouabain-insensitive or an ouabain-sensitive K-ATPase activity inhibited by Sch28080 according as animals are fed a normal or a potassium-depleted diet (types I and III K-ATPase, respectively). Two isoforms of H,K-ATPase have been cloned from rat gastric mucosa and colon, respectively. Gastric and colonic H,K-ATPase are expressed in the kidney, suggesting that they might account for types I and III K-ATPases. However, this hypothesis is not fully supported by segmental expression of gastric and colonic H,K-ATPase along the rat collecting duct, as well as by comparison of the pharmacological properties of gastric and colonic H,K-ATPase expressed in Xenopus ovocyte and types I and III K-ATPases in rat collecting ducts. The aim of the present work is to address directly the molecular origin of types I and III K-ATPases in the mouse collecting duct by measuring K-ATPase activities in collecting ducts of wild-type mice and mice genetically deficient in either gastric or colonic H,K-ATPase fed either a regular or a potassium-depleted diet. Like the rat, mouse collecting ducts display type I or III K-ATPase activity when fed a regular or a potassium-depleted diet, respectively. Type I K-ATPase activity is detected in colonic H,K-ATPase-deficient mice but not in gastric H,K-ATPase-deficient animals. Conversely, type III K-ATPase activity disappears in colonic H,K-ATPase-deficient but not in gastric H,K-ATPase-deficient mice. In conclusion, types I and III K-ATPases measured in collecting ducts of normal and potassium-depleted mice reflect the functional expression of gastric and colonic H,K-ATPase, respectively.


Subject(s)
H(+)-K(+)-Exchanging ATPase/metabolism , Imidazoles/pharmacology , Kidney/enzymology , Animals , Colon/enzymology , Diet , H(+)-K(+)-Exchanging ATPase/genetics , Mice , Mice, Knockout , Potassium/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Stomach/enzymology
12.
J Am Soc Nephrol ; 16(12): 3602-10, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16251243

ABSTRACT

Renal tubular calcium (RTCa) transport is one of the main factors that determine serum Ca concentration and urinary Ca excretion. The distal convoluted and connecting tubules reabsorb a significant fraction (10%) of filtered Ca. These tubule segments also synthesize in large abundance tissue kallikrein (TK), a major kinin-forming enzyme. Tested was the hypothesis that TK and kinins are involved in controlling RTCa transport by studying TK (TK-/-) or kinin B2 receptor (B2-/-)-deficient mice on different Ca diets. On a 0.9% wt/wt Ca diet, 129Sv or C57Bl/6 TK-/- mice excreted significantly more Ca in urine than their wild-type (WT) littermates. There was no difference between TK-/- and WT mice for plasma concentrations of Ca, Mg, creatinine, parathyroid hormone, or 1,25-dihydroxyvitamin D. On a low Ca (LCa) diet (0.01% wt/wt), urinary Ca excretion decreased in both TK-/- and WT mice but still remained higher in TK-/- mice compared with WT. The plasma Ca concentration was unchanged in C57Bl/6 TK-/- mice but decreased significantly in 129Sv TK-/- mice. Taken together, these data demonstrate that TK deficiency led to impaired RTCa absorption. On the LCa diet, renal TK gene expression doubled in WT mice. No change in urinary Ca excretion was observed in B2-/- mice, even after treatment with a kinin B1-receptor antagonist, and these mice adapted normally to the LCa diet. TK deficiency had no effect on the renal abundance of distal Ca transporter mRNA. These data suggest that TK may be a physiologic regulator of RTCa transport, acting through a non-kinin-mediated mechanism.


Subject(s)
Calcium/metabolism , Kidney Tubules/physiology , Tissue Kallikreins/deficiency , Tissue Kallikreins/genetics , Analysis of Variance , Animals , Blotting, Northern , Disease Models, Animal , Female , Gene Expression Regulation , Immunohistochemistry , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Phenotype , Probability , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Statistics, Nonparametric
13.
Am J Physiol Renal Physiol ; 289(6): F1281-90, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16077082

ABSTRACT

NH(4)(+) transport by the distal nephron and NH(4)(+) detoxification by the liver are critical for achieving regulation of acid-base balance and to avoid hyperammonemic hepatic encephalopathy, respectively. Therefore, it has been proposed that rhesus type B glycoprotein (Rhbg), a member of the Mep/Amt/Rh NH(3) channel superfamily, may be involved in some forms of distal tubular acidosis and congenital hyperammonemia. We have tested this hypothesis by inactivating the RHbg gene in the mouse by insertional mutagenesis. Histochemical studies analyses confirmed that RHbg knockout (KO) mice did not express Rhbg protein. Under basal conditions, the KO mice did not exhibit encephalopathy and survived well. They did not exhibit hallmarks of distal tubular acidosis because neither acid-base status, serum potassium concentration, nor bone mineral density was altered by RHbg disruption. They did not have hyperammonemia or disturbed hepatic NH(3) metabolism. Moreover, the KO mice adapted to a chronic acid-loading challenge by increasing urinary NH(4)(+) excretion as well as their wild-type controls. Finally, transepithelial NH(3) diffusive permeability, or NH(3) and NH(4)(+) entry across the basolateral membrane of cortical collecting duct cells, measured by in vitro microperfusion of collecting duct from KO and wild-type mice, was identical with no apparent effect of the absence of Rhbg protein. We conclude that Rhbg is not a critical determinant of NH(4)(+) excretion by the kidney and of NH(4)(+) detoxification by the liver in vivo.


Subject(s)
Ammonia/metabolism , Glycoproteins/genetics , Kidney/physiology , Membrane Transport Proteins/genetics , Acidosis, Renal Tubular/physiopathology , Ammonia/urine , Animals , Anion Exchange Protein 1, Erythrocyte/metabolism , Bone Density , Mice , Mice, Knockout , Mutagenesis, Insertional , Proton-Translocating ATPases/biosynthesis
14.
J Bone Miner Res ; 20(5): 799-808, 2005 May.
Article in English | MEDLINE | ID: mdl-15824853

ABSTRACT

UNLABELLED: Chronic thiazide treatment is associated with high BMD. We report that patients and mice with null mutations in the thiazide-sensitive NaCl cotransporter (NCC) have higher renal tubular Ca reabsorption, higher BMD, and lower bone remodeling than controls, as well as abnormalities in Ca metabolism, mainly caused by Mg depletion. INTRODUCTION: Chronic thiazide treatment decreases urinary Ca excretion (UVCa) and increases BMD. To understand the underlying mechanisms, Ca and bone metabolism were studied in two models of genetic inactivation of the thiazide-sensitive NaCl cotransporter (NCC): patients with Gitelman syndrome (GS) and Ncc knockout (Ncc(-/-)) mice. MATERIALS AND METHODS: Ca metabolism was analyzed in GS patients and Ncc(-/-) mice under conditions of low dietary Ca. BMD was measured by DXA in patients and mice, and bone histomorphometry was analyzed in mice. RESULTS: GS patients had low plasma Mg. They exhibited reduced UVCa, but similar serum Ca and GFR as control subjects, suggesting increased renal Ca reabsorption. Blood PTH was lower despite lower serum ionized Ca, and Mg repletion almost corrected both relative hypoparathyroidism and low UVCa. BMD was significantly increased in GS patients at both lumbar (+7%) and femoral (+16%) sites, and osteocalcin was reduced. In Ncc(-/-) mice, serum Ca and GFR were unchanged, but UVCa was reduced and PTH was elevated; Mg repletion largely corrected both abnormalities. Trabecular and cortical BMD were higher than in Ncc(+/+) mice (+4% and +5%, respectively), and despite elevated PTH, were associated with higher cortical thickness and lower endosteal osteoclastic surface. CONCLUSIONS: Higher BMD is observed in GS patients and Ncc(-/-) mice. Relative hypoparathyroidism (human) and bone resistance to PTH (mice), mainly caused by Mg depletion, can explain the low bone remodeling and normal/low serum Ca despite increased renal Ca reabsorption.


Subject(s)
Benzothiadiazines , Bone Density , Kidney Diseases/genetics , Kidney/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Symporters/chemistry , Thiadiazines/pharmacology , Adolescent , Adult , Age Factors , Aged , Animals , Body Weight , Bone and Bones/metabolism , Calcium/metabolism , Case-Control Studies , Diuretics , Female , Humans , Hypoparathyroidism , Magnesium/blood , Magnesium/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Mutation , Phenotype , Sodium Chloride/pharmacology , Sodium Chloride Symporters , Symporters/metabolism , Syndrome , Tibia/metabolism , Time Factors , Transgenes
15.
Diabetes ; 54(4): 991-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15793237

ABSTRACT

The renin-angiotensin system with its active metabolite angiotensin (Ang) II has been related not only to hypertension but also to obesity and insulin resistance. Recent evidence obtained in vitro suggests that the type 2 Ang II receptor (AT2R) mediates the trophic action of Ang II on adipocyte differentiation and lipogenesis. We used AT2R(y/-) mice to delineate a potential role of AT2R in adipose tissue development and metabolism. AT2R(y/-) mice had a normal adiposity but displayed a striking adipose tissue phenotype characterized by small adipocytes and an increase in cell number. In muscle, the expression of several genes involved in lipid metabolism, including fatty acid translocase, uncoupling protein-3, peroxisome proliferator-activated receptors (alpha, delta), and carnitine palmitoyl transferase-1, was increased in AT2R-deficient mice. In response to high-fat feeding, these mice were protected against obesity and obesity-related glucose intolerance, as assessed by glucose tolerance tests. Moreover, lipid oxidation assessed by indirect calorimetry was higher in AT2R-deficient mice than in wild-type mice, irrespective of the diet. This suggests that AT2R-dependent signaling exerts a direct or indirect negative control on lipid utilization in muscles. These data support the idea that AT2R-dependent Ang II signaling increases adipose cell mass and glucose intolerance and thus could participate to the deleterious effects of a high-fat diet.


Subject(s)
Adipocytes/physiology , Cell Size , Insulin Resistance/physiology , Obesity/physiopathology , Receptor, Angiotensin, Type 2/physiology , Angiotensin II/pharmacology , Animals , Dietary Fats , Energy Metabolism/physiology , Hypertension/physiopathology , Imidazoles/pharmacology , Insulin Resistance/genetics , Mice , Mice, Knockout , Muscle, Skeletal , Obesity/genetics , Obesity/pathology , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/genetics
16.
J Am Soc Nephrol ; 15(9): 2276-88, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15339977

ABSTRACT

Gitelman's syndrome, an autosomal recessive renal tubulopathy caused by loss-of-function mutations in the thiazide-sensitive NaCl co-transporter (NCC) of the distal convoluted tubule (DCT), is characterized by mild renal Na(+) wasting, hypocalciuria, hypomagnesemia, and hypokalemic alkalosis. For gaining further insights into the pathophysiology of Gitelman's syndrome, the impact of NCC ablation on the morphology of the distal tubule, on the distribution and abundance of ion transport proteins along its length, and on renal tubular Na(+) and Ca(2+) handling in a gene-targeted mouse model was studied. NCC-deficient mice had significantly elevated plasma aldosterone levels and exhibited hypocalciuria, hypomagnesemia, and compensated alkalosis. Immunofluorescent detection of distal tubule marker proteins and ultrastructural analysis revealed that the early DCT, which physiologically lacks epithelial Na(+) (ENaC) and Ca(2+) (TRPV5) channels, was virtually absent in NCC-deficient mice. In contrast, the late DCT seemed intact and retained expression of the apical ENaC and TRPV5 as well as basolateral Na(+)-Ca(2+) exchanger. The connecting tubule exhibited a marked epithelial hypertrophy accompanied by an increased apical abundance of ENaC. Ca(2+) reabsorption seemed unaltered in the distal convolution (i.e., the DCT and connecting tubule) as indicated by real-time reverse transcription-PCR, Western blotting, and immunohistochemistry for TRPV5 and Na(+)-Ca(2+) exchanger and micropuncture experiments. The last experiments further indicated that reduced glomerular filtration and enhanced fractional reabsorption of Na(+) and Ca(2+) upstream and of Na(+) downstream of the DCT provide some compensation for the Na(+) transport defect in the DCT and contribute to the hypocalciuria. Thus, loss of NCC leads to major structural remodeling of the renal distal tubule that goes along with marked changes in glomerular and tubular function, which may explain some of the clinical features of Gitelman's syndrome.


Subject(s)
Alkalosis/metabolism , Calcium/metabolism , Disease Models, Animal , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/pathology , Magnesium/metabolism , Sodium/metabolism , Animals , Carrier Proteins/physiology , Kidney Diseases/genetics , Mice , Receptors, Drug/physiology , Sodium Chloride Symporters , Solute Carrier Family 12, Member 3 , Symporters/physiology , Syndrome
17.
Circ Res ; 94(12): 1623-9, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15131008

ABSTRACT

The vascular kallikrein-kinin system contributes to about one third of flow-dependent dilation in mice carotid arteries, by activating bradykinin B2 receptors coupled to endothelial nitric oxide (NO) release. Because the bradykinin/NO pathway may mediate some of the effects of angiotensin II AT2 receptors, we examined the possible contribution of AT2 receptors to the kinin-dependent response to flow. Changes in outer diameter after increases in flow rate were evaluated in perfused arteries from wild-type animals (TK+/+) and in tissue kallikrein-deficient mice (TK-/-) in which the presence of AT2 receptor expression was verified. Saralasin, a nonselective angiotensin II receptor antagonist, impaired significantly flow-induced dilation in TK+/+, whereas it had no effect in TK-/- mice. In both groups, blockade of AT1 receptors with losartan or candesartan did not affect the response to flow. Inhibition of AT2 receptors with PD123319 reduced significantly flow-induced dilation in TK+/+ mice, but had no significant effect in TK-/- mice. Combining PD123319 with the bradykinin B2 receptor antagonist HOE-140 had no additional effect to AT2 receptor blockade alone in TK+/+ arteries. Flow-dependent-dilation was also impaired in AT2 receptor deficient mice (AT2-/-) when compared with wild-type littermates. Furthermore, HOE-140 significantly reduced the response to flow in the AT2+/+, but not in AT2-/- mice. In conclusion, this study demonstrates that the presence of functional AT2 receptors is necessary to observe the contribution of the vascular kinin-kallikrein system to flow-dependent dilation.


Subject(s)
Bradykinin/analogs & derivatives , Carotid Arteries/drug effects , Receptor, Angiotensin, Type 2/physiology , Tissue Kallikreins/physiology , Vasodilation/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Acetylcholine/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Bradykinin/pharmacology , Bradykinin B2 Receptor Antagonists , Carotid Arteries/physiology , Carotid Arteries/ultrastructure , Hemorheology , Imidazoles/pharmacology , Losartan/pharmacology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mesenteric Arteries/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylephrine/pharmacology , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/deficiency , Receptor, Angiotensin, Type 2/genetics , Receptor, Bradykinin B2/physiology , Tetrazoles/pharmacology , Tissue Kallikreins/deficiency , Tissue Kallikreins/genetics , Vasodilation/drug effects , Vasodilator Agents/pharmacology
18.
Kidney Int ; 64(6): 2155-62, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633138

ABSTRACT

BACKGROUND: The observation of renin expression in connecting tubule, a segment that also expresses tissue kallikrein (KLK-1), raises two questions. Are the genes expressed in the same or in different cells of connecting tubule? Does this topography support the hypothesis that KLK-1 activates prorenin or is it more likely that it affords coordinated gene regulation? METHODS: Renin and KLK-1 were examined by immunostaining and in situ hybridization. Renin activation by KLK-1 was investigated in vitro. In vivo, excretion of prorenin and active renin was compared in mice homozygous for targeted inactivation of KLK-1 (TK(-/-)) and normal littermates (TK(+/+)). RESULTS: Using in situ immunostaining for renin and in situ hybridization for KLK-1 mRNA, we found that connecting tubule cells expressing renin also expressed KLK-1. We confirmed in vitro activation of prorenin by KLK-1, but found no difference in the ratio of active renin to prorenin in urine of TK(-/-) and TK(+/+) animals. Compared to TK(+/+) controls, TK(-/-) mice exhibited significantly lower 24-hour excretion of prorenin (5.05 +/- 1.16 mg Ang I/hour vs. 9.39 +/- 1.96 mg Ang I/hour, P < 0.05) and active renin (1.98 +/- 0.25 mg Ang I/hour vs. 3.58 +/- 0.39 mg Ang I/hour, P < 0.05), with no difference in either urine volumes or plasma renin concentrations. CONCLUSION: Direct interaction between renin and KLK-1, not ruled out in vitro, is not supported in vivo. By contrast, lower excretion of active renin and prorenin in TK(-/-) compared to TK(+/+) suggest coordinated regulation of the two proteins in their participation to collecting duct function.


Subject(s)
Kidney Tubules, Collecting/metabolism , Renin/metabolism , Tissue Kallikreins/metabolism , Animals , CHO Cells , Cricetinae , Immunohistochemistry/methods , In Situ Hybridization , Kidney Tubules, Collecting/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling , Tissue Kallikreins/pharmacology
19.
Exp Nephrol ; 10(5-6): 374-82, 2002.
Article in English | MEDLINE | ID: mdl-12381922

ABSTRACT

Transforming growth factors beta (TGF-betas) are peptides involved in autocrine and paracrine control of cell growth and differentiation. In the kidneys, TGF-beta(2) has been shown to localize specifically in renin-producing cells in various conditions stimulating the renin response. To test in vivo the functional role of TGF-beta(2), the renin response was investigated in mice heterozygous for a null mutation of the TGF-beta(2) gene, which had a twofold reduction in the amount of TGF-beta(2) mRNA. Although the increase in plasma renin concentration triggered by dehydration was not different from wild-type mice, renal renin mRNA and protein levels were higher in mutant mice under hydrated or dehydrated conditions. These data suggest that TGF-beta(2) exerts an inhibitory effect on renin synthesis and release from the juxtaglomerular apparatuses.


Subject(s)
Gene Deletion , Heterozygote , Kidney/metabolism , Mutation/physiology , Renin/metabolism , Transforming Growth Factor beta/genetics , Animals , Arterioles/metabolism , Body Water/metabolism , Dehydration/metabolism , Fetus/metabolism , Genotype , Juxtaglomerular Apparatus/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , RNA, Messenger/metabolism , Renal Circulation , Renin/genetics , Transforming Growth Factor beta2 , Water Deprivation/physiology
20.
Hypertension ; 40(1): 90-5, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12105144

ABSTRACT

To clarify the role of the kallikrein-kinin system in cardiovascular homeostasis, the systemic and regional hemodynamics of kinin B2 receptor-deficient (B2-/-) and tissue kallikrein-deficient (TK-/-) mice were compared with their wild-type (WT) littermates on a pure C57BL/6 genetic background. B2-/-, TK-/-, and WT adult mice were normotensive and displayed normal hemodynamic (left ventricular [LV] pressure, cardiac output, total peripheral resistance, dP/dt(max)) and echocardiographic (septum and LV posterior wall thickness, LV diameter, LV mass, and LV fractional shortening) parameters. However, heart rate was lower in B2-/- mice compared with TK-/- and WT mice. In addition, B2-/- mice, but not TK-/- mice, exhibited lower coronary and renal blood flows and greater corresponding vascular resistances than did WT mice, indicating a tonic physiological vasodilating effect of bradykinin in these vascular beds. However, maximal coronary vasodilatation capacity, estimated after dipyridamole infusion, was similar in the 3 groups of mice. B2-/- mice were significantly more sensitive than were TK-/- mice to the vasoconstrictor effects of angiotensin II and norepinephrine. Finally, renin mRNA levels were significantly greater in B2-/- mice and smaller in TK-/- mice compared with WT mice. Taken together, these results indicate that under basal conditions, the kinin B2 receptor is not an important determinant of blood pressure in mice but is involved in the control of regional vascular tone in the coronaries and the kidneys. The phenotypic differences observed between TK-/- and B2-/- mice could be underlain by tissue kallikrein kinin-independent effect and/or kinin B1 receptor activation.


Subject(s)
Cardiovascular Physiological Phenomena , Kallikreins/deficiency , Receptors, Bradykinin/deficiency , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Blotting, Northern , Bradykinin/pharmacology , Cardiac Output/drug effects , Cardiac Output/physiology , Cardiovascular Physiological Phenomena/drug effects , Dose-Response Relationship, Drug , Female , Genotype , Heart Ventricles/drug effects , Hemodynamics/drug effects , Homeostasis/genetics , Kallikreins/genetics , Ketamine/pharmacology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Norepinephrine/pharmacology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1 , Receptor, Bradykinin B2 , Receptors, Angiotensin/genetics , Receptors, Bradykinin/genetics , Regional Blood Flow/drug effects , Renin/genetics , Ventricular Function , Xylazine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...