Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Soc Nephrol ; 32(11): 2885-2899, 2021 11.
Article in English | MEDLINE | ID: mdl-34607910

ABSTRACT

BACKGROUND: Over the last decade, advances in genetic techniques have resulted in the identification of rare hereditary disorders of renal magnesium and salt handling. Nevertheless, approximately 20% of all patients with tubulopathy lack a genetic diagnosis. METHODS: We performed whole-exome and -genome sequencing of a patient cohort with a novel, inherited, salt-losing tubulopathy; hypomagnesemia; and dilated cardiomyopathy. We also conducted subsequent in vitro functional analyses of identified variants of RRAGD, a gene that encodes a small Rag guanosine triphosphatase (GTPase). RESULTS: In eight children from unrelated families with a tubulopathy characterized by hypomagnesemia, hypokalemia, salt wasting, and nephrocalcinosis, we identified heterozygous missense variants in RRAGD that mostly occurred de novo. Six of these patients also had dilated cardiomyopathy and three underwent heart transplantation. We identified a heterozygous variant in RRAGD that segregated with the phenotype in eight members of a large family with similar kidney manifestations. The GTPase RagD, encoded by RRAGD, plays a role in mediating amino acid signaling to the mechanistic target of rapamycin complex 1 (mTORC1). RagD expression along the mammalian nephron included the thick ascending limb and the distal convoluted tubule. The identified RRAGD variants were shown to induce a constitutive activation of mTOR signaling in vitro. CONCLUSIONS: Our findings establish a novel disease, which we call autosomal dominant kidney hypomagnesemia (ADKH-RRAGD), that combines an electrolyte-losing tubulopathy and dilated cardiomyopathy. The condition is caused by variants in the RRAGD gene, which encodes Rag GTPase D; these variants lead to an activation of mTOR signaling, suggesting a critical role of Rag GTPase D for renal electrolyte handling and cardiac function.


Subject(s)
Cardiomyopathy, Dilated/genetics , Hypercalciuria/genetics , Kidney Diseases/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/genetics , TOR Serine-Threonine Kinases/metabolism , Cardiomyopathy, Dilated/metabolism , Female , HEK293 Cells , Humans , Hypercalciuria/metabolism , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Male , Models, Molecular , Natriuresis/genetics , Nephrocalcinosis/metabolism , Pedigree , Protein Conformation , Renal Tubular Transport, Inborn Errors/metabolism , Seizures/genetics , Seizures/metabolism , Signal Transduction , Exome Sequencing , Whole Genome Sequencing
2.
J Am Soc Nephrol ; 29(10): 2546-2561, 2018 10.
Article in English | MEDLINE | ID: mdl-30185469

ABSTRACT

BACKGROUND: Recent evidence emphasizes the critical role of inflammation in the development of diabetic nephropathy. Angiotensin-converting enzyme (ACE) plays an active role in regulating the renal inflammatory response associated with diabetes. Studies have also shown that ACE has roles in inflammation and the immune response that are independent of angiotensin II. ACE's two catalytically independent domains, the N- and C-domains, can process a variety of substrates other than angiotensin I. METHODS: To examine the relative contributions of each ACE domain to the sodium retentive state, renal inflammation, and renal injury associated with diabetic kidney disease, we used streptozotocin to induce diabetes in wild-type mice and in genetic mouse models lacking either a functional ACE N-domain (NKO mice) or C-domain (CKO mice). RESULTS: In response to a saline challenge, diabetic NKO mice excreted 32% more urinary sodium compared with diabetic wild-type or CKO mice. Diabetic NKO mice also exhibited 55% less renal epithelial sodium channel cleavage (a marker of channel activity), 55% less renal IL-1ß, 53% less renal TNF-α, and 53% less albuminuria than diabetic wild-type mice. This protective phenotype was not associated with changes in renal angiotensin II levels. Further, we present evidence that the anti-inflammatory tetrapeptide N-acetyl-seryl-asparyl-lysyl-proline (AcSDKP), an ACE N-domain-specific substrate that accumulates in the urine of NKO mice, mediates the beneficial effects observed in the NKO. CONCLUSIONS: These data indicate that increasing AcSDKP by blocking the ACE N-domain facilitates sodium excretion and ameliorates diabetic kidney disease independent of intrarenal angiotensin II regulation.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/deficiency , Amino Acid Substitution , Angiotensin II/metabolism , Animals , Catalytic Domain/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Epithelial Sodium Channels/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Natriuresis/genetics , Natriuresis/physiology , Oligopeptides/antagonists & inhibitors , Oligopeptides/metabolism , Peptidyl-Dipeptidase A/genetics , Protein Domains , Renin-Angiotensin System/physiology
3.
Am J Physiol Renal Physiol ; 314(6): F1138-F1144, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29357420

ABSTRACT

Many physiological functions have a circadian rhythm, including blood pressure (BP). BP is highest during the active phase, whereas during the rest period, BP dips 10-20%. Patients that do not experience this dip at night are termed "nondippers." Nondipping hypertension is associated with increased risk of cardiovascular disease. The mechanisms underlying nondipping hypertension are not understood. Without the circadian clock gene Per1, C57BL/6J mice develop nondipping hypertension on a high-salt diet plus mineralocorticoid treatment (HS/DOCP). Our laboratory has shown that PER1 regulates expression of several genes related to sodium (Na) transport in the kidney, including epithelial Na channel (ENaC) and Na chloride cotransporter (NCC). Urinary Na excretion also demonstrates a circadian pattern with a peak during active periods. We hypothesized that PER1 contributes to circadian regulation of BP via a renal Na-handling-dependent mechanism. Na-handling genes from the distal nephron were inappropriately regulated in KO mice on HS/DOCP. Additionally, the night/day ratio of Na urinary excretion by Per1 KO mice is decreased compared with WT (4 × vs. 7×, P < 0.001, n = 6 per group). Distal nephron-specific Per1 KO mice also show an inappropriate increase in expression of Na transporter genes αENaC and NCC. These results support the hypothesis that PER1 mediates control of circadian BP rhythms via the regulation of distal nephron Na transport genes. These findings have implications for the understanding of the etiology of nondipping hypertension and the subsequent development of novel therapies for this dangerous pathophysiological condition.


Subject(s)
Blood Pressure , Circadian Rhythm , Hypertension/metabolism , Kidney Tubules, Distal/metabolism , Natriuresis , Period Circadian Proteins/metabolism , Renal Elimination , Animals , Blood Pressure/genetics , Circadian Rhythm/genetics , Desoxycorticosterone Acetate , Disease Models, Animal , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Genetic Predisposition to Disease , Hypertension/genetics , Hypertension/physiopathology , Kidney Tubules, Distal/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Natriuresis/genetics , Period Circadian Proteins/deficiency , Period Circadian Proteins/genetics , Phenotype , Renal Elimination/genetics , Sodium Chloride, Dietary , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Time Factors , Up-Regulation
4.
Am J Physiol Renal Physiol ; 311(5): F991-F998, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27582096

ABSTRACT

Recent studies suggested a direct link between circadian rhythms and regulation of sodium excretion. Endothelin-1 (ET-1) regulates sodium balance by promoting natriuresis through the endothelin B receptor (ETB) in response to increased salt in the diet, but the effect that the time of day has on this natriuretic response is not known. Therefore, this study was designed to test the hypothesis that ETB receptor activation contributes to the diurnal control of sodium excretion and that sex differences contribute to this control as well. Twelve-hour urine collections were used to measure sodium excretion. On day 3 of the experiment, a NaCl load (900 µeq) was given by oral gavage either at Zeitgeber time [ZT] 0 (inactive period) or ZT12 (active period) to examine the natriuretic response to the acute salt load. Male and female ETB-deficient (ETB def) rats showed an impaired natriuretic response to a salt load at ZT0 compared with their respective transgenic controls (Tg cont). Male ETB def rats showed a delayed natriuretic response to a salt load given at ZT12 compared with male Tg cont, a contrast to the prompt response shown by female ETB def rats. Treatment with ABT-627, an ETA receptor antagonist, improved the natriuretic response seen within the first 12 h of a ZT0 salt load in both sexes. These findings demonstrate that diurnal excretion of an acute salt load 1) requires ET-1 and the ETB receptor, 2) is more evident in male vs. female rats, and 3) is opposed by the ETA receptor.


Subject(s)
Natriuresis/genetics , Receptor, Endothelin B/metabolism , Sodium/metabolism , Animals , Atrasentan , Endothelin Receptor Antagonists/pharmacology , Endothelin-1/metabolism , Female , Male , Natriuresis/drug effects , Pyrrolidines/pharmacology , Rats , Rats, Inbred WKY , Rats, Transgenic , Receptor, Endothelin B/genetics , Sex Factors , Sodium/pharmacology , Sodium/urine , Time Factors
5.
Am J Physiol Renal Physiol ; 308(12): F1409-20, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25925253

ABSTRACT

Caffeine is one of the most widely consumed behavioral substances. We have previously shown that caffeine- and theophylline-induced inhibition of renal reabsorption causes diuresis and natriuresis, an effect that requires functional adenosine A1 receptors. In this study, we tested the hypothesis that blocking the Gi protein-coupled adenosine A1 receptor via the nonselective adenosine receptor antagonist caffeine changes Na(+)/H(+) exchanger isoform 3 (NHE3) localization and phosphorylation, resulting in diuresis and natriuresis. We generated tubulus-specific NHE3 knockout mice (Pax8-Cre), where NHE3 abundance in the S1, S2, and S3 segments of the proximal tubule was completely absent or severely reduced (>85%) in the thick ascending limb. Consumption of fluid and food, as well as glomerular filtration rate, were comparable in control or tubulus-specific NHE3 knockout mice under basal conditions, while urinary pH was significantly more alkaline without evidence for metabolic acidosis. Caffeine self-administration increased total fluid and food intake comparably between genotypes, without significant differences in consumption of caffeinated solution. Acute caffeine application via oral gavage elicited a diuresis and natriuresis that was comparable between control and tubulus-specific NHE3 knockout mice. The diuretic and natriuretic response was independent of changes in total NHE3 expression, phosphorylation of serine-552 and serine-605, or apical plasma membrane NHE3 localization. Although caffeine had no clear effect on localization of the basolateral Na(+)/bicarbonate cotransporter NBCe1, pretreatment with DIDS inhibited caffeine-induced diuresis and natriuresis. In summary, NHE3 is not required for caffeine-induced diuresis and natriuresis.


Subject(s)
Caffeine/pharmacology , Diuresis/drug effects , Diuretics/pharmacology , Kidney Tubules/drug effects , Natriuresis/drug effects , Sodium-Hydrogen Exchangers/drug effects , Animals , Diuresis/genetics , Female , Glomerular Filtration Rate/drug effects , Kidney Tubules/metabolism , Male , Mice , Natriuresis/genetics , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
6.
Biochem Biophys Res Commun ; 456(2): 670-5, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25498502

ABSTRACT

The Na(+)/Ca(2+) exchanger (NCX) is a bidirectional transporter that is controlled by membrane potential and transmembrane gradients of Na(+) and Ca(2+). Although two isoforms of NCX1 and NCX2 are coexpressed on the basolateral membrane of the distal nephron, the functional significance of these isoforms is not entirely clear. Therefore, we used NCX1- and NCX2-heterozygote knockout mice (KO) and their double KO, as well as isoform-selective NCX inhibitors, to determine the roles of NCX isoforms in urine formation and electrolyte excretion in mice. NCX inhibitors, particularly NCX2-sensitive inhibitors, caused a dose-dependent natriuresis and in a higher dose, moreover, hypercalciuria. Consistently, NCX1-KO possessed normal renal function similar to wild-type mice (WT), whereas NCX2-KO and double KO exhibited moderate natriuresis and hypercalciuria. Notably, renal responses to YM-244769 were equivalently observed in NCX1-KO and WT, but disappeared in NCX2-KO and double KO. Thus, functional inhibition of NCX2 initially causes natriuresis, and further inhibition of NCX2 produces hypercalciuria, suggesting that the functional significance of NCX2 lies in Na(+) and Ca(2+) reabsorption of the kidney.


Subject(s)
Hypercalciuria/physiopathology , Natriuresis/physiology , Sodium-Calcium Exchanger/metabolism , Aniline Compounds/pharmacology , Animals , Gene Knockout Techniques , Hypercalciuria/genetics , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuresis/drug effects , Natriuresis/genetics , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenyl Ethers/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Thiourea/analogs & derivatives , Thiourea/pharmacology
7.
Am J Physiol Cell Physiol ; 302(1): C188-94, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21918182

ABSTRACT

Collecting duct (CD)-derived endothelin-1 (ET-1) acting via endothelin B (ETB) receptors promotes Na(+) excretion. Compromise of ET-1 signaling or ETB receptors in the CD cause sodium retention and increase blood pressure. Activity of the epithelial Na(+) channel (ENaC) is limiting for Na(+) reabsorption in the CD. To test for ETB receptor regulation of ENaC, we combined patch-clamp electrophysiology with CD-specific knockout (KO) of endothelin receptors. We also tested how ET-1 signaling via specific endothelin receptors influences ENaC activity under differing dietary Na(+) regimens. ET-1 significantly decreased ENaC open probability in CD isolated from wild-type (WT) and CD ETA KO mice but not CD ETB KO and CD ETA/B KO mice. ENaC activity in WT and CD ETA but not CD ETB and CD ETA/B KO mice was inversely related to dietary Na(+) intake. ENaC activity in CD ETB and CD ETA/B KO mice tended to be elevated under all dietary Na(+) regimens compared with WT and CD ETA KO mice, reaching significance with high (2%) Na(+) feeding. These results show that the bulk of ET-1 inhibition of ENaC activity is mediated by the ETB receptor. In addition, they could explain the Na(+) retention and elevated blood pressure observed in CD ET-1 KO, CD ETB KO, and CD ETA/B KO mice consistent with ENaC regulation by ET-1 via ETB receptors contributing to the antihypertensive and natriuretic effects of the local endothelin system in the mammalian CD.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Receptor, Endothelin B/deficiency , Receptor, Endothelin B/genetics , Up-Regulation/genetics , Amiloride/pharmacology , Animals , Endothelin-1/metabolism , Endothelin-1/physiology , Epithelial Sodium Channel Blockers , Female , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Knockout , Natriuresis/genetics , Sodium/metabolism
8.
J Cell Mol Med ; 16(11): 2701-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22686466

ABSTRACT

Renal medullary hypoxia-inducible factor (HIF)-1α and its target genes, such as haem oxygenase and nitric oxide synthase, have been indicated to play an important role in the regulation of sodium excretion and blood pressure. HIF prolyl hydroxylase domain-containing proteins (PHDs) are major enzymes to promote the degradation of HIF-1α. We recently reported that high salt intake suppressed the renal medullary PHD2 expression and thereby activated HIF-1α-mediated gene regulation in the renal medulla in response to high salt. To further define the functional role of renal medullary PHD2 in the regulation of renal adaptation to high salt intake and the longer term control of blood pressure, we transfected PHD2 expression plasmids into the renal medulla in uninephrectomized rats and determined its effects on pressure natriuresis, sodium excretion after salt overloading and the long-term control of arterial pressure after high salt challenge. It was shown that overexpression of PHD2 transgene increased PHD2 levels and decreased HIF-1α levels in the renal medulla, which blunted pressure natriuresis, attenuated sodium excretion, promoted sodium retention and produced salt sensitive hypertension after high salt challenge compared with rats treated with control plasmids. There was no blood pressure change in PHD2-treated rats that were maintained in low salt diet. These results suggested that renal medullary PHD2 is an important regulator in renal adaptation to high salt intake and a deficiency in PHD2-mediated molecular adaptation in response to high salt intake in the renal medulla may represent a pathogenic mechanism producing salt sensitive hypertension.


Subject(s)
Hypertension/genetics , Kidney Medulla/physiopathology , Procollagen-Proline Dioxygenase/genetics , Sodium Chloride, Dietary/pharmacology , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Gene Expression Regulation , Hypertension/etiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Kidney Medulla/metabolism , Male , Natriuresis/genetics , Pressure , Procollagen-Proline Dioxygenase/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sodium/urine , Transgenes
9.
Exp Physiol ; 97(6): 785-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22327329

ABSTRACT

Urotensin II (UII), a peptide hormone which influences glomerular filtration rate and urine concentration, and its receptor, UT, are expressed in the adult rat kidney. The ability of the kidney to reabsorb sodium and water starts to develop in utero and matures during early postnatal life in the rat, yet little is known about the ontogeny of the renal UII system. This study mapped renal expression of the urotensin system during the fetal and postnatal periods and determined renal activity of UII in the immature rat. Urotensin II peptide and mRNA were present in Sprague-Dawley (SD) rat metanephroi from the earliest stage examined, embyonic day 19 (E19; rat gestation 22 days); levels increased to peak at 4 weeks of age. In contrast, UT protein and mRNA expression declined rapidly between E19 and birth and remained at a similar level postnatally. Infusion of rat UII [6-60 pmol min(-1) (100 g body weight)(-1)] or rat urotensin-related peptide [6 pmol min(-1) (100 g body weight)(-1)] in anaesthetized 4-week-old SD rats had no influence on measured renal parameters; however, infusion of UT antagonist, SB-706375 (0.01 mg kg(-1) min(-1)), provoked a pronounced diuresis [vehicle 23.5 ± 1.9 versus antagonist 75.3 ± 12.5 µl min(-1) (100 g body weight)(-1); P < 0.001] and natriuresis, accompanied by modest increases in effective renal blood flow and glomerular filtration rate [vehicle 0.4 ± 0.1 versus antagonist 1.1 ± 0.2 ml min(-1) (100 g body weight)(-1); P < 0.0001] and a significant increase in fractional sodium excretion. These results indicate that the endogenous rat UII system may influence renal sodium and water excretion before the onset of full urine concentrating capacity in the SD rat.


Subject(s)
Glomerular Filtration Rate/physiology , Kidney/blood supply , Kidney/physiology , Urotensins/genetics , Urotensins/metabolism , Animals , Female , Fetus/metabolism , Glomerular Filtration Rate/genetics , Kidney/metabolism , Male , Natriuresis/genetics , Natriuresis/physiology , Peptide Hormones/genetics , Peptide Hormones/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Regional Blood Flow/genetics , Regional Blood Flow/physiology , Sodium/metabolism , Urotensins/antagonists & inhibitors , Water/metabolism
10.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-34914636

ABSTRACT

Exchange proteins directly activated by cAMP (Epacs) are abundantly expressed in the renal tubules. We used genetic and pharmacological tools in combination with balance, electrophysiological, and biochemical approaches to examine the role of Epac1 and Epac2 in renal sodium handling. We demonstrate that Epac1-/- and Epac2-/- mice exhibit a delayed anti-natriuresis to dietary sodium restriction despite augmented aldosterone levels. This was associated with a significantly lower response to the epithelial Na+ channel (ENaC) blocker amiloride, reduced ENaC activity in split-opened collecting ducts, and defective posttranslational processing of α and γENaC subunits in the KO mice fed with a Na+-deficient diet. Concomitant deletion of both isoforms led to a marginally greater natriuresis but further increased aldosterone levels. Epac2 blocker ESI-05 and Epac1&2 blocker ESI-09 decreased ENaC activity in Epac WT mice kept on the Na+-deficient diet but not on the regular diet. ESI-09 injections led to natriuresis in Epac WT mice on the Na+-deficient diet, which was caused by ENaC inhibition. In summary, our results demonstrate similar but nonredundant actions of Epac1 and Epac2 in stimulation of ENaC activity during variations in dietary salt intake. We speculate that inhibition of Epac signaling could be instrumental in treatment of hypertensive states associated with ENaC overactivation.


Subject(s)
Calcium Channels/genetics , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/genetics , Kidney Diseases/genetics , Natriuresis/genetics , Sodium/urine , TRPV Cation Channels/genetics , Animals , Biomarkers/urine , Calcium Channels/biosynthesis , Cells, Cultured , Disease Models, Animal , Guanine Nucleotide Exchange Factors/biosynthesis , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA/genetics , TRPV Cation Channels/biosynthesis
11.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1660-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19726707

ABSTRACT

The C57Bl/6J mouse strain, the genetic background of many transgenic and gene knockout models, is salt sensitive and resistant to renal injury. We tested the hypothesis that renal dopaminergic function is defective in C57Bl/6J mice. On normal NaCl (0.8%, 1 wk) diet, anesthetized and conscious (telemetry) blood pressures were similar in C57Bl/6J and SJL/J mice. High NaCl (6%, 1 wk) increased blood pressure (approximately 30%) in C57Bl/6J but not in SJL/J mice and urinary dopamine to greater extent in SJL/J than in C57Bl/6J mice. Absolute and fractional sodium excretions were lower in SJL/J than in C57Bl/6J mice. The blood pressure-natriuresis plot was shifted to the right in C57Bl/6J mice. Renal expressions of D(1)-like (D(1)R and D(5)R) and angiotensin II AT(1) receptors were similar on normal salt, but high salt increased D(5)R only in C57Bl/6J. GRK4 expression was lower on normal but higher on high salt in C57Bl/6J than in SJL/J mice. Salt increased the excretion of microalbumin and 8-isoprostane (oxidative stress marker) and the degree of renal injury to a greater extent in SJL/J than in C57Bl/6J mice. A D(1)-like receptor agonist increased sodium excretion whereas a D(1)-like receptor antagonist decreased sodium excretion in SJL/J but not in C57Bl/6J mice. In contrast, parathyroid hormone had a similar natriuretic effect in both strains. These results show that defective D(1)-like receptor function is a major cause of salt sensitivity in C57Bl/6J mice, decreased renal dopamine production might also contribute. The relative resistance to renal injury of C57Bl/6J may be a consequence of decreased production of reactive oxygen species.


Subject(s)
Blood Pressure , Dopamine/urine , Kidney/metabolism , Mice, Inbred C57BL/metabolism , Natriuresis , Receptors, Dopamine D1/metabolism , Sodium Chloride, Dietary/metabolism , Albuminuria/etiology , Albuminuria/metabolism , Animals , Benzazepines/pharmacology , Blood Pressure/drug effects , Blood Pressure/genetics , Dinoprost/analogs & derivatives , Dinoprost/urine , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Fenoldopam/pharmacology , Genotype , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL/genetics , Natriuresis/drug effects , Natriuresis/genetics , Oxidative Stress , Parathyroid Hormone/metabolism , Phenotype , Receptor, Angiotensin, Type 1/metabolism , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/genetics , Receptors, Dopamine D5/metabolism , Sodium Chloride, Dietary/adverse effects , Species Specificity
12.
J Am Heart Assoc ; 8(8): e012192, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30957627

ABSTRACT

Background Dopamine D5 receptor (D5R) plays an important role in the maintenance of blood pressure by regulating renal sodium transport. Our previous study found that human D5R mutant F173L transgenic ( hD 5 R F173L-TG) mice are hypertensive. In the present study, we aimed to investigate the mechanisms causing this renal D5R dysfunction in hD 5 R F173L-TG mice. Methods and Results Compared with wild-type D5R-TG ( hD 5 R WT-TG) mice, hD 5 R F173L-TG mice have higher blood pressure, lower basal urine flow and sodium excretion, and impaired agonist-mediated natriuresis and diuresis. Enhanced reactive oxygen species production in hD 5 R F173L-TG mice is caused, in part, by decreased expression of antioxidant enzymes, including thioredoxin 1 (Trx1). Na+-K+-ATPase activity is increased in mouse renal proximal tubule cells transfected with hD 5 R F173L, but is normalized by treatment with exogenous recombinant human Trx1 protein. Regulation of Trx1 by D5R occurs by the phospholipase C/ protein kinase C (PKC) pathway because upregulation of Trx1 expression by D5R does not occur in renal proximal tubule cells from D1R knockout mice in the presence of a phospholipase C or PKC inhibitor. Fenoldopam, a D1R and D5R agonist, stimulates PKC activity in primary renal proximal tubule cells of hD5R WT -TG mice, but not in those of hD 5 R F173L-TG mice. Hyperphosphorylation of hD5RF173L and its dissociation from Gαs and Gαq are associated with impairment of D5R-mediated inhibition of Na+-K+-ATPase activity in hD 5 R F173L-TG mice. Conclusions These suggest that hD 5 R F173L increases blood pressure, in part, by decreasing renal Trx1 expression and increasing reactive oxygen species production. Hyperphosphorylation of hD5RF173L, with its dissociation from Gαs and Gαq, is the key factor in impaired D5R function of hD 5 R F173L-TG mice.


Subject(s)
Blood Pressure/genetics , Hypertension/genetics , Natriuresis/genetics , Reactive Oxygen Species/metabolism , Receptors, Dopamine D5/genetics , Thioredoxins/genetics , Animals , Blood Pressure/drug effects , Chromogranins/metabolism , Diuresis/drug effects , Diuresis/genetics , Dopamine Agonists/pharmacology , Fenoldopam/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation , Humans , Hypertension/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Natriuresis/drug effects , Protein Kinase C/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D5/metabolism , Signal Transduction , Thioredoxins/metabolism , Thioredoxins/pharmacology , Type C Phospholipases/metabolism
13.
Am J Hypertens ; 31(4): 415-421, 2018 03 10.
Article in English | MEDLINE | ID: mdl-29182730

ABSTRACT

BACKGROUND: Salt sensitivity of blood pressure (SSBP) increases the risk of cardiovascular complications, and the heritability of SSBP is about 50% in Chinese population. However, studies identifying genes involved in BP responses to acute sodium loading and diuresis shrinkage are still limited. METHOD: A total of 342 essential hypertensives from Beijing were recruited in our study. A modified Sullivan's acute oral saline load and diuresis shrinkage test was conducted to each individual. Medical history and lifestyle risk factors were obtained by questionnaire. Generalized linear model was used to examine the associations of 29 single-nucleotide polymorphisms (SNPs) with SSBP and false discovery rate (FDR) was used to correct P values for multiple testing. RESULTS: In the process of acute sodium loading, after adjusting for age and 24-hour urinary sodium concentration, SNPs in CYP11B2, PRKG1, SLC8A1 genes were significantly associated with systolic BP (SBP) rising in the additive and recessive model; SNPs in CYP4A11, PRKG1, SLC8A1, and ADRB2 genes were significantly associated with diastolic BP (DBP) rising. In the process of diuresis shrinkage, SNPs of CLCNKA, eNOS, PRKG1 gene were associated with SBP and DBP decreasing. After FDR correction, rs434082 in SLC8A1 gene was still significantly associated with blood pressure rising during salt load. In the additive model, A allele increased DBP of 2.8 mm Hg (FDR_q = 0.029) and MAP of 3.1 mm Hg (FDR_q = 0.029) after adjusting for age and 24-hour urinary sodium concentration. CONCLUSION: SLC8A1 gene may contribute to BP change in the process of acute sodium loading in a Han Chinese population.


Subject(s)
Blood Pressure/genetics , Essential Hypertension/genetics , Polymorphism, Single Nucleotide , Sodium Chloride, Dietary/adverse effects , Sodium-Calcium Exchanger/genetics , Aged , Asian People/genetics , Beijing/epidemiology , Essential Hypertension/diagnosis , Essential Hypertension/ethnology , Essential Hypertension/physiopathology , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Natriuresis/genetics , Phenotype , Risk Factors , Sodium Chloride, Dietary/administration & dosage
14.
Am J Hypertens ; 31(8): 910-918, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29617893

ABSTRACT

BACKGROUND: Pathogenic variations in HSD11B2 gene triggers the apparent mineralocorticoid excess syndrome (AME). There is scarce information regarding the phenotypes of subjects carrying heterozygous pathogenic variants in HSD11B2 gene. We investigated if serum cortisol/cortisone (F/E) ratio and cortisone are useful for identifying partial 11ßHSD2 deficiency in those heterozygous subjects. METHODS: We studied two patients diagnosed with AME and their families carrying either D223N or R213C mutation. We also evaluated 32 healthy control subjects (13 children and 19 adults) to obtain normal references ranges for all measured variables. Case 1: A boy carrying D223N mutation in HSD11B2 gene and Case 2: A girl carrying R213C mutation. We assessed serum F/E ratio and cortisone by HPLC-MS/MS, aldosterone, plasma-renin-activity(PRA), electrolytes, and HSD11B2 genetic analyses. RESULTS: The normal values (median [interquartile range]) in children for serum F/E and cortisone (µg/dl) were 2.56 [2.21-3.69] and 2.54 [2.35-2.88], and in adults were 4.42 [3.70-4.90] and 2.23 [1.92-2.57], respectively. Case 1 showed a very high serum F/E 28.8 and low cortisone 0.46 µg/dl. His mother and sister were normotensives and heterozygous for D223N mutation with high F/E (13.2 and 6.0, respectively) and low cortisone (2.0 and 2.2, respectively). Case 2 showed a very high serum F/E 175 and suppressed cortisone 0.11 µg/dl. Her parents and sister were heterozygous for the R213C mutation with normal phenotype, but high F/E and low cortisone. Heterozygous subjects showed normal aldosterone, PRA, but lower fractional excretion of sodium and urinary Na/K ratio than controls. CONCLUSION: Serum F/E ratio and cortisone allow to identify partial 11ßHSD2 deficiencies, as occurs in heterozygous subjects, who would be susceptible to develop arterial hypertension.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Cortisone/blood , Hydrocortisone/blood , Mineralocorticoid Excess Syndrome, Apparent/blood , Adolescent , Adult , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Cross-Sectional Studies , Female , Genetic Predisposition to Disease , Heredity , Heterozygote , Humans , Male , Middle Aged , Mineralocorticoid Excess Syndrome, Apparent/diagnosis , Mineralocorticoid Excess Syndrome, Apparent/enzymology , Mineralocorticoid Excess Syndrome, Apparent/genetics , Mutation , Natriuresis/genetics , Pedigree , Phenotype , Predictive Value of Tests
15.
Circ Res ; 95(11): 1100-8, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15528469

ABSTRACT

Alpha-adducin polymorphism in humans is associated with abnormal renal sodium handling and high blood pressure. The mechanisms by which mutations in adducin affect the renal set point for sodium excretion are not known. Decreases in Na+,K+-ATPase activity attributable to endocytosis of active units in renal tubule cells by dopamine regulates sodium excretion during high-salt diet. Milan rats carrying the hypertensive adducin phenotype have a higher renal tubule Na+,K+-ATPase activity, and their Na+,K+-ATPase molecules do not undergo endocytosis in response to dopamine as do those of the normotensive strain. Dopamine fails to promote the interaction between adaptins and the Na+,K+-ATPase because of adaptin-mu2 subunit hyperphosphorylation. Expression of the hypertensive rat or human variant of adducin into normal renal epithelial cells recreates the hypertensive phenotype with higher Na+,K+-ATPase activity, mu2-subunit hyperphosphorylation, and impaired Na+,K+-ATPase endocytosis. Thus, increased renal Na+,K+-ATPase activity and altered sodium reabsorption in certain forms of hypertension could be attributed to a mutant form of adducin that impairs the dynamic regulation of renal Na+,K+-ATPase endocytosis in response to natriuretic signals.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Cytoskeletal Proteins/physiology , Hypertension/genetics , Kidney Tubules/enzymology , Microfilament Proteins/physiology , Natriuresis/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex mu Subunits/chemistry , Amino Acid Substitution , Animals , Blood Pressure/genetics , Blood Pressure/physiology , Cell Line/drug effects , Cell Line/enzymology , Cytoskeletal Proteins/genetics , Dopamine/pharmacology , Endocytosis/drug effects , Endosomes/enzymology , Epithelium/enzymology , Humans , Hypertension/enzymology , Hypertension/physiopathology , Kidney Tubules/drug effects , Microfilament Proteins/genetics , Mutagenesis, Site-Directed , Natriuresis/drug effects , Natriuresis/genetics , Opossums , Phosphoprotein Phosphatases/metabolism , Protein Interaction Mapping , Protein Subunits , Rats , Rats, Mutant Strains , Recombinant Fusion Proteins/physiology , Structure-Activity Relationship , Transfection
16.
Int Rev Cytol ; 194: 1-66, 2000.
Article in English | MEDLINE | ID: mdl-10494624

ABSTRACT

The natriuretic peptide (NP) system consists of three types of hormones [atrial NP (ANP), brain or B-type NP (BNP), and C-type NP (CNP)] and three types of receptors [NP receptor (R)-A, NPR-B, and NPR-C]. ANP and BNP are circulating hormones secreted from the heart, whereas CNP is basically a neuropeptide. NPR-A and NPR-B are membrane-bound guanylyl cyclases, whereas NPR-C is assumed to function as a clearance-type receptor. ANP, BNP, and CNP occur commonly in all tetrapods, but ventricular NP replaces BNP in teleost fish. In elasmobranchs, only CNP is found, even in the heart, suggesting that CNP is an ancestral form. A new guanylyl cyclase-uncoupled receptor named NPR-D has been identified in the eel in addition to NPR-A, -B, and -C. The NP system plays pivotal roles in cardiovascular and body fluid homeostasis. ANP is secreted in response to an increase in blood volume and acts on various organs to decrease both water and Na+, resulting in restoration of blood volume. In the eel, however, ANP is secreted in response to an increase in plasma osmolality and decreases Na+ specifically, thereby promoting seawater adaptation. Therefore, it seems that the family of NPs were originally Na(+)-extruding hormones in fishes; however, they evolved to be volume-depleting hormones promoting the excretion of both Na+ and water in tetrapods in which both are always regulated in the same direction. Vertebrates expanded their habitats from fresh water to the sea or to land during evolution. The structure and function of osmoregulatory hormones have also undergone evolution during this ecological evolution. Thus, a comparative approach to the study of the NP family affords new insights into the essential function of this osmoregulatory hormone.


Subject(s)
Natriuresis/physiology , Peptides/chemistry , Peptides/physiology , Amino Acid Sequence , Animals , Evolution, Molecular , Humans , Molecular Sequence Data , Natriuresis/genetics , Peptides/genetics , Receptors, Peptide/genetics , Receptors, Peptide/physiology , Sequence Homology, Amino Acid , Vertebrates , Water-Electrolyte Balance
17.
Pharmacogenomics ; 6(7): 665-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16207143

ABSTRACT

Adducin is a heterodimeric cytoskeleton protein consisting of an alpha-subunit and either a beta- or gamma-subunit. In rats and humans, mutation of the alpha-adducin subunit leads to the stimulation of the sodium (Na(+)), potassium (K(+))-adenosine triphosphate (ATP)-ase activity in renal tubular cells, increased renal Na(+) reabsorption, and, subsequently, hypertension. Ouabain is a hormone that is released by the hypothalamus and, possibly, the adrenal glands. In renal tubular cells it modulates Na(+)/K(+)-ATPase activity and regulates natriuresis. Plasma ouabain levels increase with the number of copies of the mutated alpha-adducin allele. Rostafuroxin is a digitoxygenin derivative that selectively displaces ouabain from the Na(+)/K(+)-ATPase receptor and lowers blood pressure in rats and humans. In this short editorial review, we summarize the recent experimental, clinical and epidemiological evidence that contributed to our understanding of the pathogenetic mechanisms that lead to hypertension associated with the alpha-adducin Gly460Trp polymorphism and its interaction with ouabain. We propose that a pharmacogenomic approach, as applied in an ongoing Phase II dosage study of rostafuroxin, will be a critical step in moving the adducin hypothesis from experimental and observational studies to clinical application.


Subject(s)
Calmodulin-Binding Proteins/genetics , Hypertension, Renal/etiology , Alleles , Androstanols/pharmacology , Animals , Blood Pressure/genetics , Blood Pressure/physiology , Calmodulin-Binding Proteins/chemistry , Forecasting , Heterozygote , Humans , Hypertension, Renal/enzymology , Hypertension, Renal/genetics , Hypertension, Renal/metabolism , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Mutation , Natriuresis/drug effects , Natriuresis/genetics , Ouabain/metabolism , Ouabain/pharmacology , Polymorphism, Genetic , Protein Subunits/genetics , Rats , Sodium-Potassium-Exchanging ATPase/metabolism
18.
J Mol Med (Berl) ; 79(10): 574-80, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11692154

ABSTRACT

A higher prevalence of hypertension has been associated with the G-->A/GT (Gly40Ser) polymorphism of the glucagon receptor gene (GCGR) in two population studies. As the mutated receptor is less responsive to glucagon, it has been speculated that the increased susceptibility to hypertension is due to deprivation of the recognized natriuretic effect of the hormone. To test this hypothesis we determined the frequency of the polymorphic variant and evaluated the segmental renal sodium handling by the clearances of uric acid and of exogenous lithium in the Olivetti Heart Study participants (n=971). The polymorphic variant was present only in heterozygous form in 37 individuals (3.8%). After controlling for age and body mass index, the carriers of the variant were twice more likely to be hypertensive and almost three times more likely to be on antihypertensive treatment at the time of examination. Compared to participants carrying the wild type, those carrying the Gly40Ser allele had higher serum uric acid and lower fractional excretion of uric acid and exogenous lithium, independently of age, body mass, and current pharmacological treatment. We conclude that the Gly40Ser polymorphism of the GCGR gene is associated with higher risk of hypertension and with enhanced proximal tubular sodium reabsorption, a factor possibly contributing to hypertension in this group.


Subject(s)
Hypertension/genetics , Kidney/metabolism , Receptors, Glucagon/genetics , Sodium/metabolism , Adult , Aged , Amino Acid Substitution , Blood Pressure/genetics , Creatinine/blood , Glomerular Filtration Rate , Humans , Hypertension/physiopathology , Kidney/physiopathology , Lithium/urine , Male , Middle Aged , Multivariate Analysis , Natriuresis/genetics , Point Mutation , Polymorphism, Genetic , Sodium/urine , Uric Acid/blood , Uric Acid/urine
19.
Cardiovasc Res ; 51(3): 429-41, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11476733

ABSTRACT

Natriuretic peptides and nitric oxide play important roles in cardiovascular and renal physiology and disease. The natriuretic peptides - atrial natriuretic peptide, brain natriuretic peptide, and C-type natriuretic peptide - comprise a family of proteins that participate in the integrated control of intravascular volume and arterial blood pressure. The natriuretic peptides differentially bind distinct classes of receptors that signal through different mechanisms. Membrane-bound, guanylyl cyclase-coupled natriuretic peptide receptors (A- and B-types) mediate natriuretic peptide effects through the production of 3',5'-cyclic guanosine monophosphate (cGMP). C-Type natriuretic peptide receptors, which lack the guanylyl cyclase domain, alter target cell function through G(i) protein-coupled inhibition of membrane adenylyl cyclase activity, and also serve to clear circulating natriuretic peptides. The expression of the natriuretic peptides and their receptors are subject to complex controls. Similar structural and regulatory diversity exists for the nitric oxide synthases. The three nitric oxide synthase genes are regulated by a variety of mechanisms ranging from alternative splicing and alternative promoter usage to complex post-translational controls. This review highlights the molecular diversity of the natriuretic peptides and nitric oxide synthases and explores recent insights into their regulation.


Subject(s)
Natriuresis/genetics , Nitric Oxide Synthase/genetics , Peptides/genetics , Atrial Natriuretic Factor/genetics , Gene Expression Regulation , Humans , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, C-Type/genetics , Receptors, Peptide/genetics
20.
Hypertension ; 66(1): 68-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941340

ABSTRACT

The gene SLC4A5 encodes the Na(+)-HCO3 (-) cotransporter electrogenic 2, which is located in the distal nephron. Genetically deleting Na(+)-HCO3 (-) cotransporter electrogenic 2 (knockout) causes Na(+)-retention and hypertension, a phenotype that is diminished with alkali loading. We performed experiments with acid-loaded mice and determined whether overactive epithelial Na(+) channels (ENaC) or the Na(+)-Cl(-) cotransporter causes the Na(+) retention and hypertension in knockout. In untreated mice, the mean arterial pressure was higher in knockout, compared with wild-type (WT); however, treatment with amiloride, a blocker of ENaC, abolished this difference. In contrast, hydrochlorothiazide, an inhibitor of Na(+)-Cl(-) cotransporter, decreased mean arterial pressure in WT, but not knockout. Western blots showed that quantity of plasmalemmal full-length ENaC-α was significantly higher in knockout than in WT. Amiloride treatment caused a 2-fold greater increase in Na(+) excretion in knockout, compared with WT. In knockout, but not WT, amiloride treatment decreased plasma [Na(+)] and urinary K(+) excretion, but increased hematocrit and plasma [K(+)] significantly. Micropuncture with microelectrodes showed that the [K(+)] was significantly higher and the transepithelial potential (Vte) was significantly lower in the late distal tubule of the knockout compared with WT. The reduced Vte in knockout was amiloride sensitive and therefore revealed an upregulation of electrogenic ENaC-mediated Na(+) reabsorption in this segment. These results show that, in the absence of Na(+)-HCO3 (-) cotransporter electrogenic 2 in the late distal tubule, acid-loaded mice exhibit disinhibition of ENaC-mediated Na(+) reabsorption, which results in Na(+) retention, K(+) wasting, and hypertension.


Subject(s)
Epithelial Sodium Channels/physiology , Hypertension, Renal/metabolism , Sodium-Bicarbonate Symporters/deficiency , Amiloride/pharmacology , Amiloride/therapeutic use , Animals , Antihypertensive Agents/therapeutic use , Disease Models, Animal , Diuretics/therapeutic use , Epithelial Sodium Channels/drug effects , Hematocrit , Hydrochlorothiazide/therapeutic use , Hydrogen-Ion Concentration , Hypertension, Renal/drug therapy , Hypertension, Renal/genetics , Hypokalemia/etiology , Kidney Tubules, Distal/metabolism , Membrane Potentials/drug effects , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Natriuresis/drug effects , Natriuresis/genetics , Polymorphism, Single Nucleotide , Potassium/metabolism , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/physiology
SELECTION OF CITATIONS
SEARCH DETAIL