16
REVIEW Dopamine, kidney, and hypertension: studies in dopamine receptor knockout mice Xiaoyan Wang & Van Anthony M. Villar & Ines Armando & Gilbert M. Eisner & Robin A. Felder & Pedro A. Jose Received: 21 January 2008 / Revised: 18 April 2008 / Accepted: 7 May 2008 / Published online: 10 July 2008 # IPNA 2008 Abstract Dopamine is important in the pathogenesis of hypertension because of abnormalities in receptor-mediated regulation of renal sodium transport. Dopamine receptors are classified into D 1 -like (D 1 ,D 5 ) and D 2 -like (D 2 ,D 3 ,D 4 ) subtypes, all of which are expressed in the kidney. Mice deficient in specific dopamine receptors have been gener- ated to provide holistic assessment on the varying physio- logical roles of each receptor subtype. This review examines recent studies on these mutant mouse models and evaluates the impact of individual dopamine receptor subtypes on blood pressure regulation. Keywords Dopamine receptor . Hypertension . Knockout mice . Renal function Introduction Hypertension, which affects a quarter of all middle-aged adults, is one of the most common risk factors of cardiovascular disease. High blood pressure is a phenotype expressed by several disorders, and results from an aberrant interaction between genetic and environmental factors. Hypertension occurs because of the increased activity of pro-hypertensive systems and/or decreased activity of anti- hypertensive systems [14], one of which is the dopaminer- gic system [1, 2]. Therefore, a dysfunction in the dopaminergic pathway may be a mechanism in the devel- opment of essential hypertension. Mammalian dopamine receptors are G protein-coupled receptors (GPCRs) belonging to the α group of the rhodopsin family (Class A, Family A, or Family 1) and include five subtypes identified by molecular cloning [5, 6]. The D 1 and D 5 receptor subtypes are known as D 1 -like receptors and couple to G proteins G s and G olf to stimulate adenylyl cyclase activity. The D 2 ,D 3 , and D 4 receptor subtypes are known as D 2 -like receptors and couple to G I and G o to inhibit adenylyl cyclase activity [1, 2]. D 1 -like receptors are typically expressed post-synaptically/post- junctionally, whereas D 2 -like receptors are also expressed pre-synaptically/pre-junctionally. Distinct cells within the same organ may co-express different receptor subtypes, rendering cell-specific assignment of receptor subtypes difficult [2, 714]. Dopamine receptor agonists and antag- onists have poor selectivity for individual dopamine receptors and, consequently, hinder investigation of the specific roles of each dopamine receptor subtype. To circumvent this problem, gene-targeted mutant mouse models lacking one or more dopamine receptor subtypes [813, 1526] have been generated. Studies using these mouse models have shed light on the specific physiological Pediatr Nephrol (2008) 23:21312146 DOI 10.1007/s00467-008-0901-3 X. Wang : V. A. M. Villar : I. Armando : P. A. Jose (*) Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA e-mail: [email protected] G. M. Eisner Department of Medicine, Georgetown University Medical Center, Washington, DC, USA R. A. Felder Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA, USA P. A. Jose Department of Physiology and Biophysics, Georgetown University Medical Center, Washington, DC, USA

Dopamine, kidney, and hypertension: studies in dopamine receptor knockout mice

Embed Size (px)

Citation preview

REVIEW

Dopamine, kidney, and hypertension: studies in dopaminereceptor knockout mice

Xiaoyan Wang & Van Anthony M. Villar &

Ines Armando & Gilbert M. Eisner & Robin A. Felder &

Pedro A. Jose

Received: 21 January 2008 /Revised: 18 April 2008 /Accepted: 7 May 2008 / Published online: 10 July 2008# IPNA 2008

Abstract Dopamine is important in the pathogenesis ofhypertension because of abnormalities in receptor-mediatedregulation of renal sodium transport. Dopamine receptorsare classified into D1-like (D1, D5) and D2-like (D2, D3, D4)subtypes, all of which are expressed in the kidney. Micedeficient in specific dopamine receptors have been gener-ated to provide holistic assessment on the varying physio-logical roles of each receptor subtype. This reviewexamines recent studies on these mutant mouse modelsand evaluates the impact of individual dopamine receptorsubtypes on blood pressure regulation.

Keywords Dopamine receptor . Hypertension .

Knockout mice . Renal function

Introduction

Hypertension, which affects a quarter of all middle-agedadults, is one of the most common risk factors ofcardiovascular disease. High blood pressure is a phenotypeexpressed by several disorders, and results from an aberrantinteraction between genetic and environmental factors.Hypertension occurs because of the increased activity ofpro-hypertensive systems and/or decreased activity of anti-hypertensive systems [1–4], one of which is the dopaminer-gic system [1, 2]. Therefore, a dysfunction in thedopaminergic pathway may be a mechanism in the devel-opment of essential hypertension.

Mammalian dopamine receptors are G protein-coupledreceptors (GPCRs) belonging to the α group of therhodopsin family (Class A, Family A, or Family 1) andinclude five subtypes identified by molecular cloning [5, 6].The D1 and D5 receptor subtypes are known as D1-likereceptors and couple to G proteins Gs and Golf to stimulateadenylyl cyclase activity. The D2, D3, and D4 receptorsubtypes are known as D2-like receptors and couple to GI

and Go to inhibit adenylyl cyclase activity [1, 2]. D1-likereceptors are typically expressed post-synaptically/post-junctionally, whereas D2-like receptors are also expressedpre-synaptically/pre-junctionally. Distinct cells within thesame organ may co-express different receptor subtypes,rendering cell-specific assignment of receptor subtypesdifficult [2, 7–14]. Dopamine receptor agonists and antag-onists have poor selectivity for individual dopaminereceptors and, consequently, hinder investigation of thespecific roles of each dopamine receptor subtype. Tocircumvent this problem, gene-targeted mutant mousemodels lacking one or more dopamine receptor subtypes[8–13, 15–26] have been generated. Studies using thesemouse models have shed light on the specific physiological

Pediatr Nephrol (2008) 23:2131–2146DOI 10.1007/s00467-008-0901-3

X. Wang :V. A. M. Villar : I. Armando : P. A. Jose (*)Department of Pediatrics, Georgetown University Medical Center,Washington, DC, USAe-mail: [email protected]

G. M. EisnerDepartment of Medicine, Georgetown University Medical Center,Washington, DC, USA

R. A. FelderDepartment of Pathology,University of Virginia Health Sciences Center,Charlottesville, VA, USA

P. A. JoseDepartment of Physiology and Biophysics,Georgetown University Medical Center,Washington, DC, USA

role of each receptor at the cell, organ, and whole animallevel.

The long-term regulation of blood pressure is accom-plished by both renal and non-renal mechanisms [1, 2, 27–31]. Abnormalities in renal epithelial ion transport playimportant roles in the pathogenesis of essential hypertension,especially in salt-sensitive hypertension. In this review, wesummarize the role of the dopamine receptor subtypes in theregulation of blood pressure in humans and rodents.

Renal distribution of the dopamine receptors

D1 receptor

Human D1 receptor mRNA is expressed in cultured humanrenal proximal tubule cells [32]. In the human kidney [33],D1 receptor immunostaining is found in the apical andbasolateral membrane of the proximal and distal convolutedtubules, medullary thick ascending limb of Henle, maculadensa, and cortical collecting duct, but not in the glomer-ulus and juxtaglomerular cell. D1 receptor protein is alsopresent in the large intrarenal arteries in humans [33], butnot in renal veins. Immunoblotting studies have revealedthe presence of several forms of D1 receptors, varying insize from 50 to 210 kDa, in rat and human renal proximaltubules [34], which is in agreement with data obtained inbrain tissue [35].

Rat Radioligand binding studies of the rat have shown thepresence of D1-like receptors in the renal cortex, but not inglomeruli or medulla [36–41]. However, ligands to bothD1-like receptors have similar affinities and, therefore,cannot pharmacologically distinguish the D1 from the D5

receptor. D1 receptor mRNA is expressed in the ratproximal and distal tubule, arteriole, juxtaglomerularapparatus, but not in the glomerulus or medulla [42–44].It is also expressed in cultured renal proximal tubule cellsfrom the rat [45], mouse (unpublished studies), opossum[46], human [32], and pig [47]. D1 receptor immunostain-ing is present in the rat apical and basolateral membrane ofthe proximal and distal convoluted tubules, medullary thickascending limb of Henle, macula densa, and corticalcollecting duct [7, 48]. Most of the D1 receptors in the ratproximal tubules are located in the S3 segment (unpub-lished observations, 2007). The protein expression of D1

receptors in the inner medullary collecting duct has notbeen documented, but dopamine does not stimulate cyclicadenosine monophosphate (cAMP) production in thisnephron segment [49]. The expression of D1 receptors inthe juxtaglomerular cell [7, 44, 48] has been reported in therat, but not in the human or mouse kidney. Consistent withthe results from mRNA studies and adenylyl cyclase

activation in response to D1-like receptor agonist [2], D1

receptor proteins are not expressed in the rat glomerulus.

Mouse The distribution of D1 receptor protein in the renaltubular segments in the mouse ([2], unpublished studies) issimilar to that in the human and rat kidney.

D2 receptor

There are two D2 receptor isoforms, D2short and D2long,which have been suggested to function pre- and post-synaptically, respectively [50]. The D2short receptor isexpressed more in pre- than in post-synaptic junctions[51, 52].

Rat D2long mRNA is expressed in renal tubules andglomeruli [53]. Immunoblotting [54] revealed that D2

receptor protein is expressed in rat kidney cortex at a majorband of 63 kDa and a minor protein band of 95 kDa(probably a D2 receptor dimer). By indirect immunofluores-cence microscopy, D2 receptor immunostaining is found inthe rat renal cortical proximal tubule, collecting duct, and themesangial cell of the glomerulus [54]. The D2 receptor isexpressed in the intercalated—but not in the principal cell—of the medullary collecting duct. D2 receptor protein is alsoexpressed in an opossum proximal tubule cell line, whichalso exhibits distal tubular cell characteristics [55]. There areno reports of renal tubular D2 receptor expression in thehuman and mouse.

D3 receptor

Human There are no published studies on the location ofthe D3 receptor in non-embryonic human kidney.

Rat Radioligand binding of 7-OH-DPAT, a selective D2 andD3 receptor ligand, has been reported in rat cortical tubulesand mesangial cells [56]. D3 receptor mRNA is expressedin the glomeruli, renal cortical and medullary tubular, andintrarenal vascular tissues [53]. In renal proximal tubules,D3 receptor protein is mainly located in the apical andsubapical areas, but not in the basolateral membrane [57].However, results on the expression of the rat D3 receptoralong the nephron and blood vessels have not beenconsistent. Nurnberger et al. [57] found D3 receptor proteinto be limited to the proximal tubule, whereas O’Connell etal. [58] found the receptor protein in the glomerulus, renalblood vessel, renal proximal tubule, the apical membrane ofthe distal convoluted tubule, in the intercalated cell of thecortical collecting duct, and in the macula densa of the ratkidney, but not in juxtaglomerular apparatus. However, D3

2132 Pediatr Nephrol (2008) 23:2131–2146

receptor mRNA and functional D3 receptors have beendescribed in primary cultures of rat juxtaglomerular cells[59]. The discrepancy between the findings in the kidney insitu and juxtaglomerular cells in vitro suggests that theexpression of D3 receptors is conditional (e.g. culture-dependent). Two species of D3 receptor (45 and 90 kDa) areexpressed in rat renal proximal tubule brush bordermembranes [60] and cells [61], as detected by immuno-blotting studies.

Mouse D3 receptor immunostaining is observed at theapical membrane of the S1 segment of the proximalconvoluted tubule. The staining is also found in the maculadensa, cytoplasm of the thick ascending limb of Henle,distal convoluted tubule, and the glomerulus, but not in themedullary collecting duct [62].

D4 receptor

Human D4 receptor mRNA is expressed in the humankidney [63].

Rat D4 receptor mRNA is not expressed in the glomerulusor the loop of Henle [63], but the D4 receptor protein isfound in cultured rat juxtaglomerular cells [59]. Immuno-blotting analysis of the renal cortex has revealed a minorprotein band of 49 kDa and a major protein band of 38 kDa[54]. D4 receptor immunostaining is present in the S1segment of the proximal tubule, the distal convolutedtubule [64], and especially in the cortical and medullarycollecting ducts, where it is more prominent at thebasolateral area than at the luminal areas. D4 receptorstaining is weak in the glomerulus [54].

Mouse There are no reports on the expression of the D4

receptor in the mouse kidney. Our unpublished studiesindicate that the D4 receptor is expressed throughout thenephron, except at the thick ascending limb of Henle(Fig. 1).

D5 receptor

Human D5 receptor protein is expressed in culturedproximal tubule cells from the human kidney [65].

Rat The D5 receptor protein is expressed in the proximaland distal convoluted tubules, and arteriole, but not in thejuxtaglomerular cell, glomerulus, or macula densa [7, 66–68]. The D5 receptor may be expressed preferentially overthe D1 receptor in the thick ascending limb of Henle and thecortical collecting duct, while the D1 receptor is preferen-

tially expressed in the proximal tubule [69, 70]. Immortal-ized rat renal proximal tubule cells express D5 receptors ofmolecular sizes similar to those of the D1 receptor [71, 72].The D5 receptor is also expressed in the opossum kidney,but not in the opossum kidney cell line [46].

Mouse The D5 receptor immunostaining is located in theapical and brush border membrane of mouse renal corticaltubule [72].

Role of the dopamine receptors in renal physiology

The D1-like receptors

Dopamine produced by the renal proximal tubule plays animportant role in the regulation of sodium excretion,especially when there is a surfeit of sodium in the system[1, 2]. The D1-like dopamine receptors, D1R and D5R,account for 50–70% of renal sodium excretion [1, 2] duringmoderate sodium intake. The inhibition of sodium transportexerted by endogenous renal dopamine is due mainly torenal tubular effects [2, 38, 73], in contrast to exogenouslyadministered dopamine and D1-like receptor agonists whichregulate sodium excretion by both hemodynamic andtubular mechanisms [1, 2, 14, 38, 74–76].

As aforementioned, there are no available ligands that candistinguish the D1 from the D5 receptor. However, it is likelythat the natriuretic effect of dopamine is exerted mainly

Renal Dopamine Receptors and Apical Sodium TransportersDCT

(NCC):D1-5

PCT, PST(NHE3, NaPi2):

D1-5

TAL(NKCC2,NHE3):

D1&D5

CNT,CCD(ENaCs):

D1-5

MCD(ENaCs):

D4

Cortex

Medulla

Fig. 1 Distribution of dopamine receptor subtypes (D1–5) and apicalsodium transporters in rat tubules. PCT Proximal convoluted tubule,PST proximal straight tubule, TAL thick ascending limb of Henle’sloop, DCT distal convoluted tubule, CNT cortical connecting tubule,CCD cortical collecting duct, MCD medullar collecting duct, NHE3sodium hydrogen exchanger type 3, NaPi2 sodium phosphatecotransporter type 2, NKCC2 sodium potassium two chloridecotransporter, NCC sodium chloride cotransporter, ENaC epithelialsodium channel

Pediatr Nephrol (2008) 23:2131–2146 2133

through the D1 receptor in renal proximal tubule cells sincethe cAMP production caused by dopamine in these cells ismostly due to D1 receptor activity [77]; the inhibition ofrenal ion transport by D1-like receptors is, in part, mediatedby cAMP. Chronic intrarenal cortical infusion of D1 receptorantisense oligodeoxynucleotides, which selectively decreaseD1 receptor protein expression without affecting the D5

receptor expression [78], provides direct evidence of thespecific D1 receptor role in the inhibition of renal sodiumtransport. In this study, the selective renal cortical inhibitionof D1 receptor expression decreased sodium excretion duringa normal or high salt intake.

The stimulation of D1-like receptors decreases sodiumtransport in the proximal tubule by inhibiting the activity ofseveral sodium transporters, including the sodium hydrogenexchanger 3 (NHE3, SLC9A3), sodium phosphate cotrans-porter (NaPi-IIa/SLC34A1 and NaPi-IIc/SLC34A3), and Cl-/HCO3

- exchanger (SLC26A6) at the apical membrane, andelectrogenic Na+/HCO3

- co-transporter (NBCe1A, SLC4A4),and Na+-K+ ATPase at the basolateral membrane [16, 75,79–86]. In the medullary thick ascending limb of Henle, D1-like receptors decrease sodium transport [87] by inhibitingNa+–K+ ATPase activity, although dopamine increasesNKCC2 (SLC12A1) activity in this nephron segment [88].We have suggested that the D1-like receptor stimulation ofNKCC2 may be important in K+ recycling. Eicosanoids (20-hydroxyeicosatetraenoic acid, 20-HETE) may act synergisti-cally with D1-like receptors to inhibit NKCC2 activity. Gprotein-dependent, cAMP/protein kinase A (PKA)-depen-dent, and protein kinase C (PKC)-independent mechanismsare involved in the D1-like receptor inhibition of Na+/Pi2,NHE3, Na/HCO3

- co-transporter and Cl-/HCO3- exchanger

activity, including their translocation out of the brush bordermembranes [82, 89–94]. On the other hand, D1-like receptorsinhibit Na+–K+ ATPase activity via cAMP/PKA, certainPKC isoforms, and 20-HETE that internalizes Na+–K+

ATPase subunits [2, 83, 95–104]. The mechanism of theinhibitory effect of D1-like receptors on Na+–K+ ATPase isnephron-segment specific. Protein kinase A is involved inD1-like receptor-mediated inhibition of sodium transportersin the cortical collecting duct, while PKC is involved in theproximal convoluted tubule, and the eicosanoids areinvolved in all nephron segments [1, 95–98, 100–103,105–107]. D1-like receptors inhibit NHE3 activity via PKAin rat renal proximal tubule cells [91], while PKC may alsobe involved in opossum kidney cells [107]. Interestingly, theability of D1-like receptors to stimulate calcium uptake inrenal proximal tubule cells, via cAMP and PKC, may play arole in the inhibition of NHE3 activity [108].

In addition to the direct inhibition of sodium excretionby the D1-like receptors per se, these receptors interact withthe D2-like receptors to enhance their inhibitory effect. Wehave reported that the increase in sodium excretion induced

by Z-1046, a dopamine receptor agonist with a rank orderpotency D4 ≥ D3 > D2 > D5 > D1, is blocked by either a D1-like or D2-like receptor antagonist [60]. D2-like receptorsmay potentiate the inhibitory effect of D1-like receptors onNa+-Pi co-transport, NHE3, and Na+-K+ ATPase activitiesin renal proximal tubules (see below).

D1 receptor and renal function

The D1 receptor interacts with other systems, such as therenin–angiotensin–aldosterone and the sympathetic nervoussystems, to tightly regulate renal and cardiovascular func-tions. Angiotensin II exerts its effects on angiotensin II type Ireceptors (AT1R) and opposes the effects of dopamineexerted mainly through the D1-like receptors. D1-like andAT1 receptors have contrasting effects on the generation ofsecond messengers, i.e. D1-like receptors stimulate adenylylcyclases, while the AT1 receptor inhibits them. Both D1-likeand AT1 receptors stimulate phospholipase C activity;however, they stimulate different PKC isoforms [1, 2].Activation of AT1 receptors stimulates all renal proximaltubular ion-transporting proteins that are inhibited by the D1-like dopamine receptors [106, 109–111]. Thus, the effect ofthe D1-like receptors on natriuresis is enhanced whenangiotensin II production is decreased or when AT1 receptorsare blocked [112, 113]. The D1-like receptors can alsocounteract the renal vasoconstrictor effect of angiotensin II[114]. Activation of the D1-like receptors decreases AT1

receptor expression and the number of angiotensin II bindingsites in renal proximal tubule cells from normotensiveWistar–Kyoto (WKY) rats [72, 115]. The use of antisenseoligonucleotides against D1 or D5 receptors points to D5R asthe major negative regulator of AT1R expression [65].However, the D1 receptor can also inhibit AT1 receptorfunction by direct physical interaction [116]. In contrast toAT1 receptors, AT2 receptors participate in the natriuresismediated by the D1-like receptors [117].

D5 receptor and renal function

Activation of D1-like receptors induces diuresis andnatriuresis in WKY rats [1, 2, 38, 39, 117]. Due to thelack of selective D1 and D5 receptor ligands, the relativecontribution of D1 and D5 receptors on natriuresis inresponse to D1-like receptor agonist stimulation is notknown. It is presumed that both D1 and D5 receptors areinvolved because both receptors increase cAMP productionand cAMP mediates the D1-like receptor-mediated inhibi-tion of ion transport [77]. However, as aforementioned,cAMP production is increased to a greater extent by the D1

receptor than by the D5 receptor in renal proximal tubulecells [77] and, thus, the D1 receptor, conceivably, mediatesmost of the natriuretic effect of dopamine [78].

2134 Pediatr Nephrol (2008) 23:2131–2146

D2-like receptors and renal function

The D2-like receptor family consists of the D2, D3, and D4

dopamine receptors. These receptors variably regulatesodium transport [1, 2]. The D2-like receptors can modulaterenal function by regulating dopamine transporter activity[118] and renal dopamine production [119]. Bromocriptine, aD2-like receptor agonist with a tenfold greater affinity for D2

over D3 receptors, stimulates Na+–K+ ATPase activity byincreasing its alpha subunit in the plasma membrane [120,121]. Bromocriptine also increases chloride transport in themedullary thick ascending limb of Henle in rat renalproximal tubules [122]. LY171555, a D2-like receptoragonist with a higher affinity to the D2 receptors, stimulatesNa+–K+ ATPase activity in murine fibroblasts heterologouslyexpressing D2Long [123]. Interestingly, dopamine engendersantinatriuresis in sodium-depleted women [124]. Sulpiride, aD2-like receptor antagonist with equal affinity to all D2-likereceptors, impairs the dopamine-mediated natriuresis involume-expanded women [125]. Thus, activation of D2-likereceptors results in an antinatriuresis in conditions of volumedepletion or in a natriuresis in volume-expanded states.Whether this is a direct effect of the D2-like receptors or dueto their interaction with the D1-like receptors remains to bedetermined. D2-like receptors have also been shown topossess vasodilatory effects, presumably by a pre-junctionalinhibition of catecholamine release [2].

D2 receptor and renal function

In the LTK2 cells expressing the D2Long receptor, activationof D2-like receptors decreases cAMP levels, resulting inincreased Na+–K+ ATPase activity [123]. The selectiveagonists for the D2 receptor, bromocriptine, and LY171555inhibit adenylyl cyclase activities. The resulting decrease incAMP production/PKA activation probably mediates thestimulation of Na+–K+ ATPase activity [120, 123]. The D2

receptor can inhibit adenylyl cyclase activity even in theabsence of adenylyl cyclase V [126], the isoform notexpressed in the kidney [127]. D1 receptor-mediatedactivation of PKC could lead to a D2long-mediatedsensitization of adenylyl cyclase VI [128]. In Chinesehamster ovary (CHO) cells heterologously expressingtenfold more D2 receptors than D1 receptors, stimulationof either receptor results in the potentiation of arachidonicacid release compared to those cells expressing eitherreceptor only [129, 130]; arachidonic acid cytochrome P-450 products have been shown to inhibit renal sodiumtransport [83, 98, 131, 132]. This synergism between D1

and D2 receptors in the production of cAMP, phospholipaseC, and arachidonic acid products might explain theobserved D2 receptor-mediated natriuretic effect. The D1

and D2 receptors can heterodimerize [133] and interact with

each other [130, 134] to increase c-fos [135] and inhibitNa+–K+ ATPase activity [136]. It is conceivable that theD2-like receptors stimulate sodium transport under con-ditions of “low” sodium intake and inhibit Na+–K+ ATPaseactivity in renal proximal tubules cells under conditions of“moderate” sodium excess (in concert with D1-like recep-tors) to synergistically increase sodium excretion [124, 125,137–139].

D3 receptor and renal function

The ability of D3 receptors to mediate natriuresis is sup-ported by the decreased ability of D3 receptor-deficient miceto excrete an acute saline load [17] and corroborated byseveral pharmacological studies. The D3 receptor agonists 7-OH-DPAT and PD128907 [140] have been shown todecrease renal sodium transport [141]. Acute intravenousadministration of 7-OH-DPAT in salt-resistant Dahl ratsincreases glomerular filtration rate and sodium and waterexcretion without affecting blood pressure [141]. PD128907,a selective D3 receptor agonist with a 120-fold selectivityover the D2 receptor, infused directly into the renal artery,dose-dependently increases fractional sodium excretion inWKY rats [but not in spontaneously hypertensive rats(SHR)] fed a normal or high salt diet [142]. Quinerolane, aD2-like receptor agonist with 21-fold selectivity for the D3

over the D2 receptor, opens K+ channels, resulting inhyperpolarization and inhibition of Na+–K+ ATPase activity[143]. The renal effects of D3 receptor activation aremediated by actions on post-junctional glomerular andtubular receptors, and pre-synaptic modulation of norepi-nephrine release because renal denervation attenuates theeffects of 7-OH-DPAT [144].

D4 receptor and renal function

The D4 receptor exerts an antagonistic effect on vasopres-sin- and aldosterone-dependent water and sodium reabsorp-tion in the cortical collecting duct [145, 146]. In the rabbitcortical collecting duct, the D4 receptor-mediated decreasein sodium transport is exerted mainly at the basolateralmembrane despite of greater expression of D4 receptors atthe luminal membrane [146].

The distribution of dopamine receptors and apical sodiumtransporters in rat renal tubules is summarized in Fig. 1.

Aberrant dopamine receptor function and hypertension

D1-like receptors and hypertension

The diuretic and natriuretic responses to administration ofD1-like receptor agonists are impaired in rodents with

Pediatr Nephrol (2008) 23:2131–2146 2135

polygenic hypertension (Dahl salt-sensitive rats and SHRs)[1, 2, 38, 75, 147]. The natriuretic effect of parathyroidhormone and cholecystokinin is not impaired in the SHRs,indicating that the impaired D1-like receptor effect isreceptor-specific [60]. More importantly, the natriureticeffect of dopamine synthesized in the kidney is alsoimpaired in rodent polygenic hypertension [38, 60, 147].In human essential hypertensive subjects, the actions of D1-like receptors on renal hemodynamics and distal renaltubule function (see below) are preserved [148, 149], andfor this reason, the overall natriuretic effect of administeredD1 receptor agonists may be similar or even greater thanthat in normotensive subjects, even when the ability of D1-like receptor agonists to inhibit proximal tubular reabsorp-tion is impaired [148].

The ability of D1-like receptors to inhibit NHE3, Cl-/HCO3

- exchanger, Na+/HCO3- cotransporter, and Na+–K+

ATPase activities is decreased in rodent genetic hyperten-sion which is due, in part, to decreased production ofsecond messengers (e.g. cAMP, diacylglycerol, eicosa-noids) in the renal proximal tubule [16, 37, 71, 75, 86,92, 93, 98, 150–153] and the thick ascending limb of Henle[75]. The D1-like receptor-mediated stimulation of cAMPproduction, however, is not impaired in the corticalcollecting duct of SHRs [154].

The impaired ability of D1-like receptors to stimulatecAMP production in renal proximal tubules is receptorspecific, since the ability of parathyroid hormone tostimulate cAMP production or stimulate G proteins is intact[153, 155, 156] and, at least in young SHRs, β-adrenergicfunction is also intact [157]. Increased serine phosphoryla-tion and decreased expression of D1 receptors at the plasmamembrane [71, 158], are responsible for the impairedability of D1-like receptors to stimulate cAMP productionin renal proximal tubules in genetic hypertension. However,the impairment of D1-like receptor function is organspecific because D1-like receptor function in the brainstriatum is intact [37]. The D1-like receptor defect is geneticbecause it co-segregates with high blood pressure andprecedes the onset of hypertension [16, 37, 159, 160].

Deficiency of the expression of D1-like receptors couldbe involved in human essential hypertension. The humanD1 gene locus on chromosome 5 at q35.1 is linked tohuman essential hypertension [161]. Although there are asyet no reports of the association of polymorphisms of thecoding region of the D1 receptor and essential hypertension,a polymorphism, A-48G, in the 5′-untranslated region isassociated with essential hypertension in Japanese [162]—but not in Caucasians [163]. However, the D1 receptor A-48G polymorphism has been reported to be associated withalbuminuria [164].

The D5 receptor gene locus (chromosome 4p15.1–16.1)is also linked to essential hypertension [165]; the human D5

receptor gene has polymorphisms that code for receptorsabnormally coupled to adenylyl cyclase [166]. The role ofthe D5 receptor in hypertension has been difficult todetermine, mostly due to the fact that the D1 and D5

receptors are pharmacologically indistinguishable. As men-tioned above, the lack of selective ligands makes itimpossible to activate or block only the D1 or D5 receptorin vivo. For this reason, genetic approaches to this problemhave been employed by investigators who used genesilencing techniques to inhibit D1 or D5 receptor expressionand generate D1 or D5 receptor-deficient mice [9, 12, 16,22, 23].

D1 receptor mutant mice

D1 receptor knockout (D1-/-) mice were generated bytargeted mutagenesis with 7.0 kb of the 129/Sv-derivedD1 receptor genomic construct in pPNT [16, 22]. Thehomozygous D1-/- mice have no binding sites for D1-likereceptor antagonists, and in response to D1-like receptoragonist stimulation, they do not increase cAMP accumula-tion although their response to parathyroid hormone isintact. Systolic, diastolic, and mean arterial pressures arehigher in both homozygous and heterozygous mice than inwild-type mice. These data indicate that defective D1

receptor/signal transduction results in increased bloodpressure in mice [16]. However, there is no report availableon the renal function or the activity of the renin-angiotensinII–aldosterone system in D1-/- mice. It is also unknownwhether the hypertension of the null mice is salt-dependent.

D5 receptor-deficient mice

D5 receptor deficient (D5-/-) mice were generated byinjecting into C57BL/6 mouse blastocysts 129/SV embry-onic stem cells containing the targeting construct generatedby ligating the neomycin resistance gene in reverseorientation at the unique SfiI site in the second intracellularloop of the D5 receptor. D5-/- mice are viable, developnormally, and have normal expression of the otherdopamine receptors, including the D1 receptor [23, 167].

Congenic D5-/- mice are hypertensive and salt-sensitive.Blood pressure is also increased in D5+/- mice (Table 1,unpublished data). D5-/- mice have a greater epinephrine/norepinephrine ratio and a greater hypotensive response tothe acute administration of an α-adrenergic blocker [23]than their D5+/+ littermates, indicating that increasedsympathetic activity plays a role in the elevated bloodpressure. However, the percentage decrease in systolicblood pressure, after adrenalectomy, is similar in D5-/- andD5+/+ mice, indicating that central nervous system mech-anisms are involved in the hypertension of D5-/- mice [23,168]. In addition, these central nervous system mecha-

2136 Pediatr Nephrol (2008) 23:2131–2146

nisms, AT1 receptors, and the increased production ofreactive oxygen species (ROS) are also involved in thehypertension of D5-/- mice [14, 66, 72, 169]. However,renal renin protein expression and serum aldosterone levelare not altered in D5-/- mice (unpublished data). D5-/- micealso exhibit proteinuria [170].

D2-like receptors and hypertension

Abnormalities of D2-like receptor function have beenreported in hypertension. One of the polymorphisms ofthe D2 receptor, which results in decreased D2 receptorexpression, is associated with hypertension [171, 172].Moreover, transfer of the segment of chromosome 8 con-taining the D2 receptor gene from the normotensive Brown-Norway rat onto an SHR background decreases bloodpressure [173].

The D3 Ser9Gly receptor variant heterologouslyexpressed in CHO cells impairs D3 receptor-mediatedinhibition of cAMP production, although these cell do

possess the ability to decrease prostaglandin E2 (PGE2)production [174]. However, there is no association betweenthe D3 Ser9Gly, or other D3 receptor gene variants withhypertension [175], despite the chromosome locus of theD3 receptor gene (3q13.3) having been linked to humanessential hypertension [176, 177]. Several studies havedemonstrated that D3 receptor deficiency may be importantin the development of salt-sensitive hypertension. Salt-resistant Dahl rats on a high sodium diet and chronicallytreated with a highly selective D3 receptor antagonist haveincreased blood pressure [178]. The natriuretic effect of a D3

agonist is not evident in hypertensive salt-sensitive Dahl rats[178]. The same research group did not find differences inthe natriuretic effect of a D3 agonist between WKY rats andSHRs [179]. In this study, however, the D3 agonist is givensystemically, and activation of D3 receptors outside thekidney may have had compensating role, thereby obscuring adifferential effect on sodium excretion between WKY andSHRs. When the selective D3 receptor agonist, PD128907, isinfused directly into the renal artery in WKY rats and SHRs

Table 1 Phenotypes of dopamine receptor (DR)-deficient mice and renal mechanism(s) involved in the pathogenesis of hypertension

Mousegenotypes

D1R D5R D2R D3R D4R

-/- +/- -/- +/- -/- +/- -/- +/- -/- +/-

Bloodpressureon 0.8%NaCl diet

Increased[16]

Increased[16]

Increased[23,168,169]

Increased#

Increased[24]

Increased[24]

Increased[17]

Increased[17]

Increased[20]

Normal#

Bloodpressure on4–6%NaCl diet

Increased[68,169]

Increased#

Increased#

Increased#

Kidneydeformities

None # None None # None # None #

Creatinineclearance

& # Normal # Normal[184]

Normal # Normal[20]

Ability toexcrete anacute NaClload

Notimpaired#

Notimpaired[24]

Notimpaired#

Impaired[17]]

Notimpaired[17]

NotImpaired#

Notimpaired#

Ability toexcrete achronicNaCl load

Notimpaired#

Notimpaired#

Impaired[186]

Impaired#

Proteinuria Yes [170] No # No # No #Plasma/Kidneyrenin

Normal # Increased[17]

Increased[17]

Normal[20]

Serum /Urinealdosterone

Normal # Increased[184]

Normal[185]

Normal# Normal #

Renal AT1R Increased[72]

Normal # Increased[17]

Increased[20]

Blank cell indicates no report available; &, indicates normal blood urea nitrogen (BUN); #, indicates unpublished

Pediatr Nephrol (2008) 23:2131–2146 2137

fed a high salt diet, fractional sodium excretion is dose-dependently increased in WKY rats but not in SHRs [142].The mechanisms causing the impaired D3 receptor functionin the kidney in hypertension are not completely known. Thestimulatory effect of the D3 receptor agonist, PD128907, onD3 receptor expression is no longer evident in renal proximaltubule cells from the SHRs [180]. It is possible thatmechanisms which impair D1 receptor function in SHR alsoimpair D3 receptor—for example, by increased G protein-coupled receptor-related kinase 4 (GRK4) activity [181]—orthat impaired D1 receptor function results in impairedsynergistic interaction between the D1 and D3 receptor. Inaddition, the impaired natriuretic function of the D3 receptorin SHRs may, in part, be related to the aberrant D3 and AT1

receptor interaction in renal proximal tubule cells [180]. D3

receptor stimulation decreases AT1 receptor expression inrenal proximal tubule cells from the WKY rat, while D3

receptor stimulation increases AT1 receptor expression in theSHR [180].

A locus near the D4 receptor gene (11p15.5) has beenlinked to hypertension. The most intensively studied D4

receptor polymorphism is a 48-bp repeat located in exon 3of the D4 receptor gene. This variant codes for a 16-aminoacid sequence located in the third intracellular loop of theD4 receptor protein, a region that appears to interact with Gproteins and influence intracellular levels of cAMP [182].The number of repeats at the D4 site varies from two to ten,but the four and seven repeat lengths are the most commonin Caucasians in whom the long variant of the D4 gene isassociated with increased systolic and diastolic bloodpressures [183].

The effects of D4 agonists and antagonists on cardiovas-cular and renal function in genetically hypertensive rats havenot been reported. The D4 receptor protein is increased in therenal cortex—but not in the inner medulla—of SHRs relativeto WKY rats [54].

D2 receptor mutant mice

The D2 receptor gene deficient (D2-/-) mice were generatedby homologous recombination in embryonic stem cells withdeletion of exon 7 and the 5′ half of exon 8. These regionsencode the majority of the putative third intracellular loop,the last two transmembrane domains, and the carboxyterminus [24]. D2-/- mice have decreased initiation ofmovement but otherwise normal basic motor skills withouttremor, ataxia, or abnormal stance or posture. D2-/- andD2-/+ mice have higher systolic and diastolic bloodpressures than D2 wild-type (D2+/+) mice. The ability ofD2-/- mice to excrete an acute saline load is similar to thatof D2+/+ mice regardless of the elevated blood pressure,indicating that D2-/- mice have a blunted pressure-natriure-sis response. α-Adrenergic blockade decreases blood

pressure to a greater extent in D2-/- mice than in D2+/+mice. Epinephrine excretion is greater in D2-/- mice than inD2+/+ mice, and acute adrenalectomy decreases bloodpressure to a similar level in D2-/- and D2+/+ mice.Endothelin B (ETB) receptor expression is greater in D2-/-mice than in D2+/+ mice, and an ETB receptor blockerdecreases blood pressure in D2-/- mice but not D2+/+ mice.D2-/- mice may be hypertensive because of enhancedvascular reactivity caused by increased sympathetic andETB receptor activities [24] as well as by increased ROSproduction [184]. The D2-/- mice also have increasedproduction of aldosterone, and treatment with a mineralo-corticoid receptor blocker normalizes blood pressure inthese mice [184]. In contrast, D2+/- mice have normalurinary aldosterone levels [185].

In another strain of D2-/- mice, blood pressure is onlyincreased when the animals are fed a high-salt diet, and thiscondition is associated with a decrease in renal aromatic L-amino acid decarboxylase (AADC) activity and renaldopamine production [26]; AADC converts L-DOPA(levodopa), which is taken up by renal brush bordermembranes, to dopamine. Sympathetic activity is notincreased in these D2-/- mice. Basal urine flow and sodiumexcretion are lower in D2-/- mice than in D2+/+ mice.However, administration of dopamine increases urinevolume and sodium excretion in D2-/- mice to levelssimilar to those found in D2+/+ mice [118]. The differencesbetween the two strains of D2-/- mice could be related todifferences in their genetic backgrounds.

D3 receptor mutant mice

D3 receptor gene deficient (D3-/-) mice have been generatedby targeted mutagenesis. The targeting construct has a 7-kb129/sv-derived D3 receptor genomic sequence in theGKNeo cassette in antisense orientation at the SalI site inexon 2 [15]. The locomotor phenotype of D3 mutants doesnot resemble that of D2 mutants. D3-/- mice developnormally, are fertile, and may show a transient locomotorhyperactivity in a novel environment. D3-/- and D3-/+ micehave higher systolic and diastolic blood pressures than theirwild-type littermates, either on a mixed C57BL/6 and B129background [17] or in congenic C57BL/6 background [62].In contrast, Staudacher et al. reported that D3-/- mice in acongenic C57BL/6 background and on a low, normal, orhigh salt intake have normal blood pressure [186]. Thisreport has to be interpreted with caution because C57BL/6mice from Jackson Laboratories develop hypertensionwhen fed a high NaCl diet [187], while those from Taconic[169] do not. The two strains of D3-/- mice, however, havea decreased ability to excrete an acute or a chronic sodiumchloride load [17, 186], which may lead to an expansion ofthe extracellular fluid volume. Thus, disruption of the D3

2138 Pediatr Nephrol (2008) 23:2131–2146

receptor results in sodium retention, but the hypertension isalso related to increased renal renin production. Renal reninactivity and AT1 receptor expression are greater in D3-/-than in wild-type mice, and blockade of AT1 receptorsdecreases systolic blood pressure more effectively in D3-/-mice than in wild-type ones [17]. Urinary aldosterone levelsare not altered in D3-/- mice (unpublished data).

D4 receptor mutant mice

D4 receptor deficient (D4-/-) mice backcrossed to C57BL/6Jmice for more than six generations have increased systolicand diastolic blood pressures that are increased yet furtherwith increased sodium intake [20]. D4-/- mice do not havealtered circulating or renal renin levels [20] althoughjuxtaglomerular cells in culture also express D4 receptors[59]. Serum/urinary aldosterone levels are not differentbetween D4-/- and D4+/+ mice on a normal or increasedsodium chloride diet (unpublished data). The expression ofAT1 receptors is increased in homogenates of kidney andbrain of D4-/- mice relative to that shown by similarhomogenates of D4+/+ mice, but AT1 receptor expression inthe heart is similar in the two strains. However, thehypotensive effect of a bolus intravenous injection oflosartan, an AT1 receptor antagonist, dissipates after 10min in D4+/+ mice, whereas the effect has been found topersist for more than 45 min in D4-/- mice. Thus,hypertension brought about by the absence of the D4

receptor is mediated, in part, by increased AT1 receptorexpression [20]. While on a normal salt diet (unpublisheddata), the heterozygote mutant D1, D2, D3, and D5 receptormice have increased blood pressure, while D4+/- mice havenormal blood pressure.

Some phenotypes related to renal function in dopaminereceptor deficient mice are summarized in Table 1.

Development of renal dopamine

Development of renal dopamine synthesis

Dopamine-mediated natriuresis and renal vascular effectsare less in younger animals than in older ones. An inversecorrelation between urinary dopamine and age has beenreported in adults [188, 189]. The excretion of dopamine ishigher in the pediatric population than in elderly subjects[190]. However, urinary dopamine increases postnatally invery low birth weight infants [191, 192]. In rats, intrarenallevels of dopamine have a tendency to increase between 3and 20 days after birth, but significantly decrease between20 to 80 days after birth [193]. Urinary excretions ofdopamine and its deaminated metabolite 3,4-dihydroxyphe-nylacetic acid (DOPAC) [194], as well as dopamine

formation in kidney slices, are markedly decreased in oldrats, which have been attributed to decreased uptake of L-DOPA by tubular brush-border membranes [194, 195]. Theuptake of L-DOPA is the rate-limiting step in the renalsynthesis of dopamine [194]. The activity of aromaticAADC, the enzyme involved in the renal synthesis ofdopamine, however, increases with age [195]. The activityof AADC is higher in the adult dog kidney than in 10-dayor 2-month-old dogs [196]. In the rat, AADC immunore-activity can be observed in renal tubule cells as early asgestational day 18 and is present in proximal tubules in theouter and inner cortex 5 days after birth. Aromatic L-aminoacid decarboxylase mRNA is present in the entire cortex bythe sixth post-natal day [193].

Development of renal dopamine receptors

In the rat, renal dopamine D1-like and D2-like receptordensities approach adult values by 3 weeks of age [197,198]. Rat renal D1 receptors are present by the 15th day ofgestational age and increase with maturation [199]. RenalD1 receptors are also expressed in fetal sheep [200].However, in the sheep, the renal cortical affinity ormaximum receptor of D1-like receptors, as determined byradioligand binding, does not change with maturation[201].

In rats, renal D2-like receptor binding does not changewithin the first 2 weeks of life but increases after 3 weeks[202]. In contrast, renal D2-like receptor binding decreaseswith age in sheep [201].

Renal effects of dopaminergic agonists

The decreased natriuretic action of dopamine and D1-likereceptor agonists in the young and the aged has beenattributed to a decreased renal generation of cAMP andinhibition of NHE3 and Na+/K+ ATPase activity inresponse to D1-like receptor stimulation [198, 201–208].Even though renal D1-like receptor densities are notdifferent between 3- and 10-week-old rats, there is animpaired ability of the D1-like agonist fenoldopam tostimulate adenylyly cyclase activity in the young [198,203]. The decreased inhibitory effect of D1-like receptorson NHE activity in young rats is caused, in part, by theincreased expression and activity of the G protein subunit Gβ/γ [208].

In neonatal dogs, pigs, and sheep, the intravenous orintrarenal arterial administration of dopamine alone maynot always increase renal blood flow or decrease renalvascular resistance [209–213]. The systemic or intrarenalarterial infusion of dopamine alone, even in low doses, mayresult in a renal vasoconstriction that is more pronounced inthe fetal and neonatal animals [209, 211, 213]. This is due

Pediatr Nephrol (2008) 23:2131–2146 2139

to the occupation of renal α adrenergic receptors thatincrease in number in the perinatal dog and sheep. A lowdose of dopamine injected into the renal artery increasesrenal blood flow and decreases renal vascular resistance inadult dogs [214], but not in young puppies [215]. Even inthe presence of α- and β-adrenergic blockade, dopaminedoes not increase renal blood flow in 1- to 2-week-oldpuppies, but it does increase renal blood flow andglomerular filtration rate in older puppies [215]. Similarresults are observed following intravenous dopamineadministration in young piglets [211]. In spite of theabsence of any change in renal blood flow followingintrarenal arterial infusion of dopamine, sodium excretionincreases, but this effect is less evident in the youngerpuppy than in the older one [215]. Dopamine also increasessodium excretion in the fetal sheep without increasing renalblood flow [213, 216]. Thus, in the very young, thenatriuretic effect of dopamine is probably due mainly totubular factors.

The effects of the D2-like receptor agonist are not wellstudied. However, in the developing animal, selective D2-like receptor agonists increase glomerular filtration rate to asimilar extent as that found in younger and older rats [217].In premature infants, in contrast to the findings in adults, apredominantly D2-like receptor antagonist increases urineflow, sodium excretion, and osmolar clearance, suggestingthat D2-like receptor effects may predominate in the humaninfant [218].

Ability to excrete a salt load in human neonates

Pre-term infants have a decreased ability to retain sodium,but both full-term and pre-term infants have a decreasedability to excrete a salt load [219]. In the pediatric age group,a decrease in sodium intake results in an increase in urinarydopamine, while in adults, a high salt intake results in anincrease in urinary dopamine [212, 218]. Low doses ofdopamine, however, can induce a natriuresis in preterminfants, but it is not known if the magnitude of the responseis different from that reported in the adult [220]. Deficits inrenal dopamine function may be responsible for thediminished ability of the aged kidney to excrete a salt load[221]. Whether this is also true in the neonate has not beendetermined.

Overall summary

In summary, dopamine receptors are important players inthe regulation of renal function and blood pressure. Eachdopamine receptor subtype exerts a unique regulatoryfunction and interacts with other GPCRs, especially thoserelated to angiotensin II. All dopamine receptor subtypes

are present in the kidney, showing a distinct distributionalong the nephron. Renal sodium transport is regulated byall subtypes, either directly or indirectly by interaction withother GPCRs, to maintain sodium balance. Deletion of anyof the dopamine receptor subtypes results in elevated bloodpressure and impaired pressure natriuresis in the mutantmice. Abnormalities of dopamine receptors in their genes/proteins or in their regulation of renal function play animportant role in the pathogenesis of hypertension.

Acknowledgment This work is supported in part by grants from theNational Institutes of Health, HL23081, DK39308, HL074940, andHL68686.

References

1. Hussain T, Lokhandwala MF (2003) Renal dopamine receptorsand hypertension. Exp Biol Med (Maywood) 228:134–142

2. Zeng C, Sanada H, Watanabe H, Eisner GM, Felder RA, Jose PA(2004) Functional genomics of the dopaminergic system inhypertension. Physiol Genomics 19:233–246

3. Dickson ME, Sigmund CD (2006) Genetic basis of hypertension:revisiting angiotensinogen. Hypertension 48:14–20

4. Osborn JW, Jacob F, Guzman P (2005) A neural set point for thelong-term control of arterial pressure: beyond the arterialbaroreceptor reflex. Am J Physiol Regul Integr Comp Physiol288:R846–R855

5. Premont RT, Gainetdinov RR (2007) Physiological roles of Gprotein-coupled receptor kinases and arrestins. Annu RevPhysiol 69:511–534

6. Schioth HB, Fredriksson R (2005) The GRAFS classificationsystem of G-protein coupled receptors in comparative perspec-tive. Gen Comp Endocrinol 142:94–101

7. Amenta F, Barili P, Bronzetti E, Ricci A (1999) Dopamine D1-like receptor subtypes in the rat kidney: a microanatomical study.Clin Exp Hypertens 21:17–23

8. Glickstein SB, Schmauss C (2001) Dopamine receptor functions:lessons from knockout mice [corrected]. Pharmacol Ther 91:63–83

9. Holmes A, Lachowicz JE, Sibley DR (2004) Phenotypic analysisof dopamine receptor knockout mice; recent insights into thefunctional specificity of dopamine receptor subtypes. Neuro-pharmacology 47:1117–1134

10. Joseph JD, Wang YM, Miles PR, Budygin EA, Picetti R,Gainetdinov RR, Caron MG, Wightman RM (2002) Dopamineautoreceptor regulation of release and uptake in mouse brainslices in the absence of D3 receptors. Neuroscience 112:39–49

11. Schmitz Y, Schmauss C, Sulzer D (2002) Altered dopaminerelease and uptake kinetics in mice lacking D2 receptors. JNeurosci 22:8002–8009

12. Sibley DR (1999) New insights into dopaminergic receptorfunction using antisense and genetically altered animals. AnnuRev Pharmacol Toxicol 39:313–341

13. Zapata A, Shippenberg TS (2005) Lack of functional D2 receptorsprevents the effects of the D3-preferring agonist (+)-PD 128907on dialysate dopamine levels. Neuropharmacology 48:43–50

14. Zeng C, Felder RA, Jose PA (2006) A new approach fortreatment of hypertension: modifying D1 dopamine receptorfunction. Cardiovasc Hematol Agents Med Chem 4:369–377

15. Accili D, Fishburn CS, Drago J, Steiner H, Lachowicz JE, ParkBH, Gauda EB, Lee EJ, Cool MH, Sibley DR, Gerfen CR,Westphal H, Fuchs S (1996) A targeted mutation of the D3

2140 Pediatr Nephrol (2008) 23:2131–2146

dopamine receptor gene is associated with hyperactivity in mice.Proc Natl Acad Sci USA 93:1945–1949

16. Albrecht FE, Drago J, Felder RA, Printz MP, Eisner GM,Robillard JE, Sibley DR, Westphal HJ, Jose PA (1996) Role ofthe D1A dopamine receptor in the pathogenesis of genetichypertension. J Clin Invest 97:2283–2288

17. Asico LD, Ladines C, Fuchs S, Accili D, Carey RM, Semeraro C,Pocchiari F, Felder RA, Eisner GM, Jose PA (1998) Disruption ofthe dopamine D3 receptor gene produces renin-dependent hyper-tension. J Clin Invest 102:493–498

18. Azdad K, Piet R, Poulain DA, Oliet SH (2003) Dopamine D4

receptor-mediated presynaptic inhibition of GABAergic transmis-sion in the rat supraoptic nucleus. J Neurophysiol 90:559–565

19. Baik JH, Picetti R, Saiardi A, Thiriet G, Dierich A, Depaulis A, LeMeur M, Borrelli E (1995) Parkinsonian-like locomotor impair-ment in mice lacking dopamine D2 receptors. Nature 377:424–428

20. Bek MJ, Wang X, Asico LD, Jones JE, Zheng S, Li X, EisnerGM, Grandy DK, Carey RM, Soares-da-Silva P, Jose PA (2006)Angiotensin-II type 1 receptor-mediated hypertension in D4

dopamine receptor-deficient mice. Hypertension 47:288–29521. Dickinson SD, Sabeti J, Larson GA, Giardina K, Rubinstein M,

Kelly MA, Grandy DK, Low MJ, Gerhardt GA, Zahniser NR(1999) Dopamine D2 receptor-deficient mice exhibit decreaseddopamine transporter function but no changes in dopaminerelease in dorsal striatum. J Neurochem 72:148–156

22. Drago J, Gerfen CR, Lachowicz JE, Steiner H, Hollon TR, LovePE, Ooi GT, Grinberg A, Lee EJ, Huang SP, Bartlett PF, Jose PA,Sibley DR, Westphal H (1994) Altered striatal function in amutant mouse lacking D1A dopamine receptors. Proc Natl AcadSci USA 91:12564–12568

23. Hollon TR, Bek MJ, Lachowicz JE, Ariano MA, Mezey E,Ramachandran R, Wersinger SR, Soares-da-Silva P, Liu ZF,Grinberg A, Drago J, Young WS 3rd, Westphal H, Jose PA,Sibley DR (2002) Mice lacking D5 dopamine receptors haveincreased sympathetic tone and are hypertensive. J Neurosci22:10801–10810

24. Li XX, Bek M, Asico LD, Yang Z, Grandy DK, Goldstein DS,Rubinstein M, Eisner GM, Jose PA (2001) Adrenergic andendothelin B receptor-dependent hypertension in dopaminereceptor type-2 knockout mice. Hypertension 38:303–308

25. Rubinstein M, Phillips TJ, Bunzow JR, Falzone TL, DziewczapolskiG, Zhang G, Fang Y, Larson JL, McDougall JA, Chester JA, Saez C,Pugsley TA, Gershanik O, Low MJ, Grandy DK (1997) Micelacking dopamine D4 receptors are supersensitive to ethanol,cocaine, and methamphetamine. Cell 90:991–1001

26. Ueda A, Ozono R, Oshima T, Yano A, Kambe M, Teranishi Y,Katsuki M, Chayama K (2003) Disruption of the type 2dopamine receptor gene causes a sodium-dependent increase inblood pressure in mice. Am J Hypertens 16:853–858

27. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, KumarAP, Kim HS, Smithies O, Le TH, Coffman TM (2006)Angiotensin II causes hypertension and cardiac hypertrophythrough its receptors in the kidney. Proc Natl Acad Sci USA103:17985–17990

28. Granger JP, Schnackenberg CG (2000) Renal mechanisms ofangiotensin II-induced hypertension. Semin Nephrol 20:417–425

29. Nasjletti A (1998) The role of eicosanoids in angiotensin-dependent hypertension. Hypertension 31:194–200

30. Raizada MK, Der Sarkissian S (2006) Potential of gene therapystrategy for the treatment of hypertension. Hypertension 47:6–9

31. Touyz RM (2005) Intracellular mechanisms involved in vascularremodelling of resistance arteries in hypertension: role ofangiotensin II. Exp Physiol 90:449–455

32. Watanabe H, Xu J, Bengra C, Jose PA, Felder RA (2002)Desensitization of human renal D1 dopamine receptors by Gprotein-coupled receptor kinase 4. Kidney Int 62:790–798

33. Ozono R, O’Connell DP, Wang ZQ, Moore AF, Sanada H,Felder RA, Carey RM (1997) Localization of the dopamine D1

receptor protein in the human heart and kidney. Hypertension30:725–729

34. Yu P, Yang Z, Jones JE, Wang Z, Owens SA, Mueller SC, FelderRA, Jose PA (2004) D1 dopamine receptor signaling involvescaveolin-2 in HEK-293 cells. Kidney Int 66:2167–2180

35. Jarvie KR, Booth G, Brown EM, Niznik HB (1989) Glycopro-tein nature of dopamine D1 receptors in the brain and parathyroidgland. Mol Pharmacol 36:566–574

36. Felder RA, Jose PA (1988) Dopamine1 receptors in rat kidneysidentified with 125I-Sch 23982. Am J Physiol 255:F970–F976

37. Felder RA, Kinoshita S, Ohbu K, Mouradian MM, Sibley DR,Monsma FJ Jr, Minowa T, Minowa MT, Canessa LM, Jose PA(1993) Organ specificity of the dopamine1 receptor/adenylylcyclase coupling defect in spontaneously hypertensive rats. Am JPhysiol 264:R726–R732

38. Felder RA, Seikaly MG, Cody P, Eisner GM, Jose PA (1990)Attenuated renal response to dopaminergic drugs in spontane-ously hypertensive rats. Hypertension 15:560–569

39. Hedge SS, Ricci A, Amenta F, Lokhandwala MF (1989)Evidence from functional and autoradiographic studies for thepresence of tubular dopamine-1 receptors and their involvementin the renal effects of fenoldopam. J Pharmacol Exp Ther251:1237–1245

40. Huo T, Healy DP (1989) Autoradiographic localization ofdopamine DA1 receptors in rat kidney with [3H]Sch 23390.Am J Physiol 257:F414–F423

41. Takemoto F, Satoh T, Cohen HT, Katz AI (1991) Localization ofdopamine-1 receptors along the microdissected rat nephron.Pflugers Arch 419:243–248

42. O’Connell DP, Aherne AM, Lane E, Felder RA, Carey RM(1998) Detection of dopamine receptor D1A subtype-specificmRNA in rat kidney by in situ amplification. Am J Physiol 274:F232–F241

43. Yamaguchi I, Jose PA, Mouradian MM, Canessa LM, MonsmaFJ Jr, Sibley DR, Takeyasu K, Felder RA (1993) Expression ofdopamine D1A receptor gene in proximal tubule of rat kidneys.Am J Physiol 264:F280–F285

44. Yamaguchi I, Yao L, Sanada H, Ozono R, Mouradian MM, JosePA, Carey RM, Felder RA (1997) Dopamine D1A receptors andrenin release in rat juxtaglomerular cells. Hypertension 29:962–968

45. Zeng C, Luo Y, Asico LD, Hopfer U, Eisner GM, Felder RA,Jose PA (2003) Perturbation of D1 dopamine and AT1 receptorinteraction in spontaneously hypertensive rats. Hypertension42:787–792

46. Nash SR, Godinot N, Caron MG (1993) Cloning and character-ization of the opossum kidney cell D1 dopamine receptor:expression of identical D1A and D1B dopamine receptor mRNAsin opossum kidney and brain. Mol Pharmacol 44:918–925

47. Kruse MS, Adachi S, Scott L, Holtback U, Greengard P, Aperia A,Brismar H (2003) Recruitment of renal dopamine 1 receptorsrequires an intact microtubulin network. Pflugers Arch 445:534–539

48. O’Connell DP, Botkin SJ, Ramos SI, Sibley DR, Ariano MA,Felder RA, Carey RM (1995) Localization of dopamine D1Areceptor protein in rat kidneys. Am J Physiol 268:F1185–F1197

49. Maeda Y, Terada Y, Nonoguchi H, Knepper MA (1992)Hormone and autacoid regulation of cAMP production in ratIMCD subsegments. Am J Physiol 263:F319–F327

50. Monsma FJ Jr, McVittie LD, Gerfen CR, Mahan LC, Sibley DR(1989) Multiple D2 dopamine receptors produced by alternativeRNA splicing. Nature 342:926–929

51. Jomphe C, Tiberi M, Trudeau LE (2006) Expression of D2

receptor isoforms in cultured neurons reveals equipotent autor-eceptor function. Neuropharmacology 50:595–605

Pediatr Nephrol (2008) 23:2131–2146 2141

52. Lindgren N, Usiello A, Goiny M, Haycock J, Erbs E, Greengard P,Hokfelt T, Borrelli E, Fisone G (2003) Distinct roles of dopamineD2L and D2S receptor isoforms in the regulation of proteinphosphorylation at presynaptic and postsynaptic sites. Proc NatlAcad Sci USA 100:4305–4309

53. Gao DQ, Canessa LM, Mouradian MM, Jose PA (1994)Expression of the D2 subfamily of dopamine receptor genes inkidney. Am J Physiol 266:F646–F650

54. Shin Y, Kumar U, Patel Y, Patel SC, Sidhu A (2003) Differentialexpression of D2-like dopamine receptors in the kidney of thespontaneously hypertensive rat. J Hypertens 21:199–207

55. Narkar VA, Hussain T, Pedemonte C, Lokhandwala MF (2001)Dopamine D2 receptor activation causes mitogenesis via p44/42mitogen-activated protein kinase in opossum kidney cells. J AmSoc Nephrol 12:1844–1852

56. Barili P, Ricci A, Baldoni E, Mignini F, Amenta F (1997)Pharmacological characterizsation and autoradiographic local-isation of a putative dopamine D3 receptor in the rat kidney. EurJ Pharmacol 338:89–95

57. Nurnberger A, Rabiger M, Mack A, Diaz J, Sokoloff P,Muhlbauer B, Luippold G (2004) Subapical localization of thedopamine D3 receptor in proximal tubules of the rat kidney. JHistochem Cytochem 52:1647–1655

58. O’Connell DP, Vaughan CJ, Aherne AM, Botkin SJ, Wang ZQ,Felder RA, Carey RM (1998) Expression of the dopamine D3

receptor protein in the rat kidney. Hypertension 32:886–89559. Sanada H, Yao L, Jose PA, Carey RM, Felder RA (1997)

Dopamine D3 receptors in rat juxtaglomerular cells. Clin ExpHypertens 19:93–105

60. Ladines CA, Zeng C, Asico LD, Sun X, Pocchiari F, Semeraro C,Pisegna J, Wank S, Yamaguchi I, Eisner GM, Jose PA (2001)Impaired renal D1-like and D2-like dopamine receptor interactionin the spontaneously hypertensive rat. Am J Physiol Regul IntegrComp Physiol 281:R1071–R1078

61. Zeng C, Asico LD, Wang X, Hopfer U, Eisner GM, Felder RA,Jose PA (2003) Angiotensin II regulation of AT1 and D3

dopamine receptors in renal proximal tubule cells of spontane-ously hypertensive rats. Hypertension 41:724–729

62. Wang X, Armando I, Asico LD, Jones JE, Escano CS, Jose PA(2005) Hypertension in dopamine receptor D3 deficient mice isassociated with increased Na transporters in kidney (abstract). JAm Soc Nephrol 16:350A

63. Matsumoto T, Ozono R, Sasaki N, Oshima T, Matsuura H,Kajiyama G, Carey Matsumoto M, Hidaka K, Tada S, Tasaki Y,Yamaguchi T (1995) Full-length cDNA cloning and distribu-tion of human dopamine D4 receptor. Mol Brain Res 29:157–162

64. Ricci A, Marchal-Victorion S, Bronzetti E, Parini A, Amenta F,Tayebati SK (2002) Dopamine D4 receptor expression in ratkidney: evidence for pre- and postjunctional localization.Histochem Cytochem 50:1091–1096

65. Gildea JJ, Wang X, Jose PA, Felder RA (2008) D5 dopaminereceptor inhibition of the angiotensin type I receptor. Hyperten-sion 51:1–7

66. Yang Z, Asico LD, Yu P, Wang Z, Jones JE, Bai RK, Sibley DR,Felder RA, Jose PA (2005) D5 dopamine receptor regulation ofphospholipase D. Am J Physiol Heart Circ Physiol 288:H55–H61

67. Yao L, Ruan X, Arendshorst WJ, Jose PA (1995) Dopaminereceptor subtype (D1A and D1B) expression in rat renalmicrovessels (abstract). Pediatr Res 37:374A

68. Zheng S, Yu P, Zeng C, Wang Z, Yang Z, Andrews PM, FelderRA, Jose PA (2003) Gα12- and Gα13-protein subunit linkage ofD5 dopamine receptors in the nephron. Hypertension 41:604–610

69. Amenta F (1997) Light microscope autoradiography of periph-eral dopamine receptor subtypes. Clin Exp Hypertens 19:27–41

70. Yao LP, Huque E, Baraniuk J, Carey RM, Felder RA, Jose PA(1996) Dopamine receptor subtype expression (D1A and D1B)in rat nephron segments (abstract). J Invest Med 44:305A

71. Yu P, Asico LD, Luo Y, Andrews P, Eisner GM, Hopfer U,Felder RA, Jose PA (2006) D1 dopamine receptor hyper-phosphorylation in renal proximal tubules in hypertension.Kidney Int 70:1072–1079

72. Zeng C, Yang Z, Wang Z, Jones J, Wang X, Altea J, Mangrum AJ,Hopfer U, Sibley DR, Eisner GM, Felder RA, Jose PA (2005)Interaction of AT1 and D5 dopamine receptors in renal proximaltubule cells. Hypertension 45:804–810

73. Carey RM, Siragy HM, Ragsdale NV, Howell NL, Felder RA,Peach MJ, Chevalier RL (1990) Dopamine-1 and dopamine-2mechanisms in the control of renal function. Am J Hypertens3:59S–63S

74. Felder RA, Wang X, Gildea J, Bengra C, Sasaki M, Zeng C,Jones JE, Zheng W, Asico LD, Jose PA (2003) Human renalangiotensin type 1 receptor regulation by the D1 dopaminereceptor (abstract). Hypertension 42:438A

75. Nishi A, Eklöf AC, Bertorello AM, Aperia A (1993) Dopamineregulation of renal Na+,K+-ATPase activity is lacking in Dahlsalt-sensitive rats. Hypertension 21:767–771

76. Teirstein PS, Price MJ, Mathur VS, Madyoon H, Sawhney N,Baim DS (2006) Differential effects between intravenous andtargeted renal delivery of fenoldopam on renal function andblood pressure in patients undergoing cardiac catheterization.Am J Cardiol 97:1076–1081

77. Sanada H, Xu J, Watanabe H, Jose PA, Felder RA (2000)Differential expression and regulation of dopamine-1 (D-1) anddopamine-5 (D-5) receptor function in human kidney (abstract).Am J Hypertens 13:156A

78. Wang ZQ, Felder RA, Carey RM (1999) Selective inhibition ofthe renal dopamine subtype D1A receptor induces antinatriuresisin conscious rats. Hypertension 33:504–510

79. Aperia A, Bertorello A, Seri I (1987) Dopamine causesinhibition of Na+-K+-ATPase activity in rat proximal convolutedtubule segments. Am J Physiol 252:F39–F45

80. Asghar M, Hussain T, Lokhandwala MF (2003) Overexpressionof PKC-βI and -δ contributes to higher PKC activity in theproximal tubules of old Fischer 344 rats. Am J Physiol RenalPhysiol 285:F1100–F1107

81. Bacic D, Kaissling B, McLeroy P, Zou L, Baum M, Moe OW(2003) Dopamine acutely decreases apical membrane Na/Hexchanger NHE3 protein in mouse renal proximal tubule.Kidney Int 64:2133–2141

82. Banday AA, Hussain T, Lokhandwala MF (2004) Renal dopamineD1 receptor dysfunction is acquired and not inherited in obeseZucker rats. Am J Physiol Renal Physiol 287:F109–F116

83. Grider JS, Ott CE, Jackson BA (2003) Dopamine D1 receptor-dependent inhibition of NaCl transport in the rat thick ascendinglimb: mechanism of action. Eur J Pharmacol 473:185–190

84. Hu MC, Fan L, Crowder LA, Karim-Jimenez Z, Murer H, MoeOW (2001) Dopamine acutely stimulates Na+/H+ exchanger(NHE3) endocytosis via clathrin-coated vesicles: dependence onprotein kinase A-mediated NHE3 phosphorylation. J Biol Chem276:26906–26915

85. Gomes P, Soares-da-Silva P (2004) Dopamine acutely decreasestype 3 Na(+)/H(+) exchanger activity in renal OK cells throughthe activation of protein kinases A and C signalling cascades.Eur J Pharmacol 488:51–59

86. Xu J, Li XX, Albrecht FE, Hopfer U, Carey RM, Jose PA (2000)D1 receptor, Gsα, and Na+/H+ exchanger interactions in thekidney in hypertension. Hypertension 36:395–399

87. Fryckstedt J, Meister B, Aperia A (1992) Control of electrolytetransport in the kidney through a dopamine- and cAMP-regulatedphosphoprotein, DARPP-32. J Auton Pharmacol 12:183–189

2142 Pediatr Nephrol (2008) 23:2131–2146

88. Aoki Y, Albrecht FE, Bergman KR, Jose PA (1996) Stimulationof Na+-K+-2Cl- cotransport in rat medullary thick ascendinglimb by dopamine. Am J Physiol 271:R1561–R1567

89. Bacic D, Capuano P, Baum M, Zhang J, Stange G, Biber J,Kaissling B, Moe OW, Wagner CA, Murer H (2005) Activationof dopamine D1-like receptors induces acute internalization ofthe renal Na+/phosphate cotransporter NaPi-IIa in mouse kidneyand OK cells. Am J Physiol Renal Physiol 288:F740–F747

90. Baines AD, Drangova R (1998) Does dopamine use severalsignal pathways to inhibit Na-Pi transport in OK cells? J Am SocNephrol 9:1604–1612

91. Felder CC, Campbell T, Albrecht F, Jose PA (1990) Dopamineinhibits Na+-H+ exchanger activity in renal BBMV by stimula-tion of adenylate cyclase. Am J Physiol 259:F297–F303

92. Kunimi M, Seki G, Hara C, Taniguchi S, Uwatoko S, Goto A,Kimura S, Fujita T (2000) Dopamine inhibits renal Na+:HCO3

-

cotransporter in rabbits and normotensive rats but not inspontaneously hypertensive rats. Kidney Int 57:534–543

93. Pedrosa R, Jose PA, Soares-Da-Silva P (2004) Defective D1-likereceptor-mediated inhibition of Cl-/HCO3

- exchanger in immor-talized SHR proximal tubular epithelial cells. Am J PhysiolRenal Physiol 286:F1120–F1126

94. Kocinsky HS, Girardi AC, Biemesderfer D, Nguyen T, MentoneS, Orlowski J, Aronson PS (2005) Use of phospho-specificantibodies to determine the phosphorylation of endogenous Na+/H+ exchanger NHE3 at PKA consensus sites. Am J PhysiolRenal Physiol 289:F249–258

95. Asghar M, Hussain T, Lokhandwala MF (2001) Activation ofdopamine D1-like receptor causes phosphorylation of alpha1-subunit of Na+,K+-ATPase in rat renal proximal tubules. Eur JPharmacol 411:61–66

96. Horiuchi A, Takeyasu K, Mouradian MM, Jose PA, Felder RA(1993) D1A dopamine receptor stimulation inhibits Na+-K+

ATPase activity through protein kinase A. Mol Pharmacol43:281–285

97. Efendiev R, Cinelli AR, Leibiger IB, Bertorello AM, PedemonteCH (2006) FRET analysis reveals a critical conformational changewithin the Na,K-ATPase alpha1 subunit N-terminus during GPCR-dependent endocytosis. FEBS Lett 580:5067–5070

98. Hussain T, Lokhandwala MF (1996) Altered arachidonic acidmetabolism contributes to the failure of dopamine to inhibit Na+,K+-ATPase in kidney of spontaneously hypertensive rats. ClinExp Hypertens 18:963–974

99. Kirchheimer C, Mendez CF, Acquier A, Nowicki S (2007) Roleof 20-HETE in D1/D2 dopamine receptor synergism resulting inthe inhibition of Na+-K+-ATPase activity in the proximal tubule.Am J Physiol Renal Physiol 292:F1435–F1442

100. Li D, Aperia A, Celsi G, da Cruz e Silva EF, Greengard P,Meister B (1995) Protein phosphatase-1 in the kidney: evidencefor a role in the regulation of medullary Na+-K+-ATPase. Am JPhysiol 269:F673–F680

101. Nowicki S, Kruse MS, Brismar H, Aperia A (2000) Dopamine-induced translocation of protein kinase C isoforms visualized inrenal epithelial cells. Am J Physiol Cell Physiol 279:C1812–C1818

102. Ominato, Satoh MT, Katz AI (1996) Regulation of Na-KATPaseactivity in the proximal tubule: role of the protein kinase Cpathway and of eicosanoids. J Membr Biol 152:235–243

103. Satoh T, Cohen HT, Katz AI (1992) Intracellular signaling in theregulation of renal Na-K-ATPase. I. Role of cyclic AMP andphospholipase A2. J Clin Invest 89:1496–1500

104. Hussain T, Abdul-Wahab R, Kotak DK, Lokhandwala MF(1998) Bromocriptine regulates angiotensin II response onsodium pump in proximal tubules. Hypertension 32:1054–1059

105. Satoh T, Cohen HT, Katz AI (1993) Different mechanisms ofrenal Na-K-ATPase regulation by protein kinases in proximaland distal nephron. Am J Physiol 265:F399–F405

106. Yao LP, Li XX, Yu PY, Xu J, Asico LD, Jose PA (1998)Dopamine D1 receptor and protein kinase C isoforms inspontaneously hypertensive rats. Hypertension 32:1049–1053

107. Pedrosa R, Gomes P, Soares-da-Silva P (2004) Distinct signal-ling cascades downstream to Gsalpha coupled dopamine D1-likeNHE3 inhibition in rat and opossum renal epithelial cells. CellPhysiol Biochem 14:91–100

108. Han JY, Heo JS, Lee YJ, Lee JH, TaubM, HanHJ (2007) Dopaminestimulates 45Ca2+ uptake through cAMP, PLC/PKC, and MAPKsin renal proximal tubule cells. J Cell Physiol 211:486–494

109. Sheikh-Hamad D, Wang YP, Jo OD, Yanagawa N (1993)Dopamine antagonizes the actions of angiotensin II in renalbrush-border membrane. Am J Physiol 264:F737–F743

110. Gesek FA, Schoolwerth AC (1990) Hormonal interactions withthe proximal Na+-H+ exchanger. Am J Physiol 258:F514–F521

111. Efendiev R, Budu CE, Cinelli AR, Bertorello AM, PedemonteCH (2003) Intracellular Na+ regulates dopamine and angiotensinII receptors availability at the plasma membrane and their cellularresponses in renal epithelia. J Biol Chem 278:28719–28726

112. Chen C, Lokhandwala MF (1995) Potentiation by enalaprilat offenoldopam-evoked natriuresis is due to blockade of intrarenalproduction of angiotensin-II in rats. Naunyn SchmiedebergsArch Pharmacol 352:194–200

113. Clark KL, Hilditch A, Robertson MJ, Drew GM (1991) Effectsof dopamine DA1-receptor blockade and angiotensin convertingenzyme inhibition on the renal actions of fenoldopam in theanaesthetized dog. J Hypertens 9:1143–50

114. Chatziantoniou C, Ruan X, Arendshorst WJ (1993) Interactionsof cAMP-mediated vasodilators with angiotensin II in rat kidneyduring hypertension. Am J Physiol 265:F845–F852

115. Cheng HF, Becker BN, Harris RC (1996) Dopamine decreasesexpression of type-1 angiotensin II receptors in renal proximaltubule. J Clin Invest 97:2745–2752

116. Zeng C, Luo Y, Asico LD, Hopfer U, Eisner GM, Felder RA,Jose PA (2003) Perturbation of D1 dopamine and AT1 receptorinteraction in spontaneously hypertensive rats. Hypertension42:787–792

117. Salomone LJ, Howell NL, McGrath HE, Kemp BA, Keller SR,Gildea JJ, Felder RA, Carey RM (2007) Intrarenal dopamine D1-like receptor stimulation induces natriuresis via an angiotensintype-2 receptor mechanism. Hypertension 49:155–161

118. Lee FJ, Pei L, Moszczynska A, Vukusic B, Fletcher PJ, Liu F(2007) Dopamine transporter cell surface localization facilitatedby a direct interaction with the dopamine D2 receptor. EMBO J26:2127–2136

119. Ozono R, Ueda A, Oishi Y, Yano A, Kambe M, Katsuki M,Oshima T (2003) Dopamine D2 receptor modulates sodiumhandling via local production of dopamine in the kidney. JCardiovasc Pharmacol 42:S75–S79

120. Hussain T, Abdul-Wahab R, Lokhandwala MF (1997) Bromo-criptine stimulates Na+, K+-ATPase in renal proximal tubules viathe cAMP pathway. Eur J Pharmacol 321:259–263

121. Narkar VA, Hussain T, Lokhandwala MF (2002) Activation ofD2-like receptors causes recruitment of tyrosine-phosphorylatedNKA alpha 1-subunits in kidney. Am J Physiol Renal Physiol283:F1290–F1295

122. Grider J, Kilpatrick E, Ott C, Jackson B (1998) Effect ofdopamine on NaCl transport in the medullary thick ascendinglimb of the rat. Eur J Pharmacol 342:281–284

123. Yamaguchi I, Walk SF, Jose PA, Felder RA (1996) DopamineD2L receptors stimulate Na+/K+ ATPase activity in murine LTK2cells. Mol Pharmacol 49:373–378

124. Agnoli GC, Borgatti R, Cacciari M, Garutti C, Ikonomu E, Lenzi P(1989) Antagonistic effects of sulpiride-racemic and enantiomers-on renal response to low-dose dopamine infusion in normal women.Nephron 51:491–498

Pediatr Nephrol (2008) 23:2131–2146 2143

125. Agnoli GC, Cacciari M, Garutti C, Ikonomu E, Lenzi P,Marchetti G (1987) Effects of extracellular fluid volume changeson renal response to low-dose dopamine infusion in normalwomen. Clin Physiol 7:465–479

126. Robinson SW, Caron MG (1997) Selective inhibition of adenylylcyclase type V by the dopamine D3 receptor. Mol Pharmacol52:508–14

127. Bek MJ, Zheng S, Xu J, Yamaguchi I, Asico LD, Sun XG, JosePA (2001) Differential expression of adenylyl cyclases in the ratnephron. Kidney Int 60:890–899

128. Beazely MA, Watts VJ (2005) Activation of a novel PKCisoform synergistically enhances D2L dopamine receptor-medi-ated sensitization of adenylate cyclase type 6. Cell Signal17:647–653

129. Kanterman RY, Mahan LC, Briley EM, Monsma FJ Jr, SibleyDR, Axelrod J, Felder CC (1991) Transfected D2 dopaminereceptors mediate the potentiation of arachidonic acid release inChinese hamster ovary cells. Mol Pharmacol 39:364–369

130. Piomelli D, Pilon C, Giros B, Sokoloff P, Martres MP, SchwartzJC (1991) Dopamine activation of the arachidonic acid cascadeas a basis for D1/D2 receptor synergism. Nature 353:164–167

131. Aperia A, Eklof AC, Holtback U, Nowicki S, Sundelof M,Greengard P (1998) The renal dopamine system. Adv Pharmacol42:870–873

132. Sarkis A, Lopez B, Roman RJ (2004) Role of 20-hydroxyeico-satetraenoic acid and epoxy- eicosatrienoic acids in hypertension.Curr Opin Nephrol Hypertens 13:205–214

133. Dziedzicka-WasylewskaM, Faron-Gorecka A, Andrecka J, Polit A,Kusmider M, Wasylewski Z (2006) Fluorescence studies revealheterodimerization of dopamine D1 and D2 receptors in the plasmamembrane. Biochemistry 45:8751–8759

134. Pollack A (2004) Coactivation of D1 and D2 dopamine receptors:in marriage, a case of his, hers, and theirs. Sci STKE 2004:pe50

135. Cho DI, Quan W, Oak MH, Choi HJ, Lee KY, Kim KM (2007)Functional interaction between dopamine receptor subtypes forthe regulation of c-fos expression. Biochem Biophys ResCommun 357:1113–1138

136. Bertorello AM, Hopfield JF, Aperia A, Greengard P (1990)Inhibition by dopamine of (Na+-K+)ATPase activity in neostriatalneurons through D1 and D2 dopamine receptor synergism.Nature 347:386–388

137. Bertorello A, Aperia A (1990) Inhibition of proximal tubule Na+-K+-ATPase activity requires simultaneous activation of DA1 andDA2 receptors. Am J Physiol 259:F924–F928

138. Eklof AC (1997) The natriuretic response to a dopamine DA1agonist requires endogenous activation of dopamine DA2receptors. Acta Physiol Scand 160:311–314

139. Jose PA, Asico LD, Eisner GM, Pocchiari F, Semeraro C, FelderRA (1998) Effects of costimulation of dopamine D1- and D2-likereceptors on renal function. Am J Physiol 275:R986–R994

140. Levesque D (1996) Aminotetralin drugs and D3 receptorfunctions. What may partially selective D3 receptor ligands tellus about dopamine D3 receptor functions? Biochem Pharmacol52:511–518

141. Luippold G, Kuster E, Joos TO, Muhlbauer B (1998) DopamineD3 receptor activation modulates renal function in anesthetizedrats. Naunyn Schmiedebergs Arch Pharmacol 358:690–693

142. Zeng C, Asico LD, Zheng S, Hopfer U, Eisner GM, Felder RA,Jose PA (2004) Role of Gα12- and Gα13-protein subunit linkageof D3 dopamine receptors in the impaired natriuretic effect of D3

dopamine receptors in SHRs (abstract). Am J Hypertens 17:96A143. Gomes P, Soares-Da-Silva P (2002) D2-like receptor-mediated

inhibition of Na+-K+-ATPase activity is dependent on theopening of K+ channels. Am J Physiol Renal Physiol 283:F114–F123

144. Muhlbauer B, Kuster E, Luippold G (2000) Dopamine D3

receptors in the rat kidney: role in physiology and pathophysi-ology. Acta Physiol Scand 168:219–223

145. Li L, Schafer JA (1998) Dopamine inhibits vasopressin-dependent cAMP production in the rat cortical collecting duct.Am J Physiol 275:F62–F67

146. Saito O, Ando Y, Kusano E, Asano Y (2001) Functionalcharacterization of basolateral and luminal dopamine receptorsin rabbit CCD. Am J Physiol Renal Physiol 281:F114–F122

147. Chen CJ, Lokhandwala MF (1992) An impairment of renaltubular DA-1 receptor function as the causative factor fordiminished natriuresis to volume expansion in spontaneouslyhypertensive rats. Clin Exp Hypertens 14:615–628

148. O’Connell DP, Ragsdale NV, Boyd DG, Felder RA, Carey RM(1997) Differential human renal tubular responses to dopaminetype 1 receptor stimulation are determined by blood pressurestatus. Hypertension 29:115–122

149. Nielsen CB, Pedersen EB (1997) Abnormal distal tubularsodium reabsorption during dopamine infusion in patients withessential hypertension evaluated by the lithium clearancemethods. Clin Nephrol 47:304–309

150. Chen C, Beach RE, Lokhandwala MF (1993) Dopamine fails toinhibit renal tubular sodium pump in hypertensive rats. Hyper-tension 21:364–372

151. Felder CC, Jose PA, Axelrod J (1989) The dopamine-1 agonist,SKF 82526, stimulates phospholipase-C activity independent ofadenylate cyclase. J Pharmacol Exp Ther 248:171–175

152. Albrecht FE, Xu J, Moe OW, Hopfer U, Simonds WF, Orlowski J,Jose PA (2000) Regulation of NHE3 activity by G protein subunitsin renal brush-border membranes. Am J Physiol Regul IntegrComp Physiol 278:R1064–1073

153. Kinoshita S, Sidhu A, Felder RA (1989) Defective dopamine-1receptor adenylate cyclase coupling in the proximal convolutedtubule from the spontaneously hypertensive rat. J Clin Invest84:1849–1856

154. Ohbu K, Felder RA (1993) Nephron specificity of dopaminereceptor-adenylyl cyclase defect in spontaneous hypertension.Am J Physiol 264:F274–F279

155. Hussain T, Lokhandwala MF (1997) Renal dopamine DA1receptor coupling with GS and Gq/11 proteins in spontaneouslyhypertensive rats. Am J Physiol 272:F339–F346

156. Jose PA, Eisner GM, Felder RA (1993) Dopaminergic defect inhypertension. Pediatr Nephrol 7:859–864

157. Michel MC, Siepmann F, Buscher R, Philipp T, Brodde OE(1993) Ontogenesis of sympathetic responsiveness in spontaneous-ly hypertensive rats. I. Renal alpha 1-, alpha 2-, and beta-adrenergicreceptors and their signaling. Hypertension 22:169–177

158. Sanada H, Jose PA, Hazen-Martin D, Yu PY, Xu J, Bruns DE,Phipps J, Carey RM, Felder RA (1999) Dopamine-1 receptorcoupling defect in renal proximal tubule cells in hypertension.Hypertension 33:1036–1042

159. Lao YS, Hendley ED, Felder RA, Jose PA (2002) Elevated renalcortical calmodulin- dependent protein kinase activity and bloodpressure. Clin Exp Hypertens 24:289–300

160. Ohbu K, Hendley ED, Yamaguchi I, Felder RA (1993) Renaldopamine-1 receptors in hypertensive inbred rat strains with andwithout hyperactivity. Hypertension 21:485–490

161. Krushkal J, Xiong M, Ferrell R, Sing CF, Turner ST, Boerwinkle E(1998) Linkage and association of adrenergic and dopaminereceptor genes in the distal portion of the long arm of chromosome5 with systolic blood pressure variation. Hum Mol Genet 7:1379–1383

162. Sato M, Soma M, Nakayama T, Kanmatsuse K (2000) DopamineD1 receptor gene polymorphism is associated with essentialhypertension. Hypertension 36:183–186

2144 Pediatr Nephrol (2008) 23:2131–2146

163. Beige J, Bellmann A, Sharma AM, Gessner R (2004) Ethnicorigin determines the impact of genetic variants in dopaminereceptor gene (DRD1) concerning essential hypertension. Am JHypertens 17:1184–1187

164. Rao F, Wessel J, Wen G, Zhang L, Rana BK, Kennedy BP,Greenwood TA, Salem RM, Chen Y, Khandrika S, Hamilton BA,Smith DW, Holstein-Rathlou NH, Ziegler MG, Schork NJ,O’Connor DT (2007) Renal albumin excretion: twin studiesidentify influences of heredity, environment, and adrenergicpathway polymorphism. Hypertension 49:1015–1031

165. Allayee H, de Bruin TW, Michelle Dominguez K, Cheng LS, IppE, Cantor RM, Krass KL, Keulen ET, Aouizerat BE, Lusis AJ,Rotter JI (2001) Genome scan for blood pressure in Dutchdyslipidemic families reveals linkage to a locus on chromosome4p. Hypertension 38:773–778

166. Cravchik A, Gejman PV (1999) Functional analysis of the humanD5 dopamine receptor missense and nonsense variants: differencesin dopamine binding affinities. Pharmacogenetics 9:199–206

167. Holmes A, Hollon TR, Gleason TC, Liu Z, Dreiling J, SibleyDR, Crawley JN (2001) Behavioral characterization of dopamineD5 receptor null mutant mice. Behav Neurosci 115:1129–1144

168. Zeng C, Yang Z, Asico LD, Jose PA (2007) Regulation of bloodpressure by D5 dopamine receptors. Cardiovasc Hematol AgentsMed Chem 5:241–248

169. Yang Z, Asico LD, Yu P, Wang Z, Jones JE, Escano CS, WangX, Quinn MT, Sibley DR, Romero GG, Felder RA, Jose PA(2006) D5 dopamine receptor regulation of reactive oxygenspecies production, NADPH oxidase, and blood pressure. Am JPhysiol Regul Integr Comp Physiol 290:R96–R104

170. Wang Z, Armando I, Asico LD, Escano C, Wang X, Lu Q,Felder RA, Schnackenberg CG, Sibley DR, Eisner GM, Jose PA(2007) The elevated blood pressure of human GRK4gammaA142V transgenic mice is not associated with increased ROSproduction. Am J Physiol Heart Circ Physiol 292:H2083–2092

171. Thomas GN, Critchley JA, Tomlinson B, Cockram CS, Chan JC(2001) Relationships between the taqI polymorphism of thedopamine D2 receptor and blood pressure in hyperglycaemic andnormoglycaemic Chinese subjects. Clin Endocrinol (Oxf)55:605–611

172. Thomas GN, Tomlinson B, Critchley JA (2000) Modulation ofblood pressure and obesity with the dopamine D2 receptor geneTaqI polymorphism. Hypertension 36:177–182

173. Kren V, Pravenec M, Lu S, Krenova D, Wang JM, Wang N,Merriouns T, Wong A, St Lezin E, Lau D, Szpirer C, Szpirer J,Kurtz TW (1997) Genetic isolation of a region of chromosome8 that exerts major effects on blood pressure and cardiac mass inthe spontaneously hypertensive rat. J Clin Invest 99:577–581

174. Hellstrand M, Danielsen EA, Steen VM, Ekman A, Eriksson E,Nilsson CL (2004) The ser9gly SNP in the dopamine D3 receptorcauses a shift from cAMP related to PGE2 related signaltransduction mechanisms in transfected CHO cells. J Med Genet41:867–871

175. Soma M, Nakayama K, Rahmutula D, Uwabo J, Sato M,Kunimoto M, Aoi N, Kosuge K, Kanmatsuse K (2002) Ser9Glypolymorphism in the dopamine D3 receptor gene is notassociated with essential hypertension in the Japanese. Med SciMonit 8:CR1–CR4

176. James K, Weitzel LR, Engelman CD, Zerbe G, Norris JM,Framingham Heart Study (2003) Genome scan linkage resultsfor longitudinal systolic blood pressure phenotypes in subjectsfrom the Framingham Heart Study. BMC Genet 4[Suppl 1]:S83

177. Rice T, Rankinen T, Province MA, Chagnon YC, Perusse L,Borecki IB, Bouchard C, Rao DC (2000) Genome-wide linkageanalysis of systolic and diastolic blood pressure: the QuebecFamily Study. Circulation 102:1956–1963

178. Luippold G, Zimmermann C, Mai M, Kloor D, Starck D, Gross G,Muhlbauer B (2001) Dopamine D3 receptors and salt-dependenthypertension. J Am Soc Nephrol 12:2272–2279

179. Luippold G, Piesch C, Osswald H, Muhlbauer B (2003) DopamineD3 receptor mRNA and renal response to D3 receptor activation inspontaneously hypertensive rats. Hypertens Res 26:855–861

180. Zeng C, Liu Y, Wang Z, He D, Huang L, Yu P, Zheng S, JonesJE, Asico LD, Hopfer U, Eisner GM, Felder RA, Jose PA (2006)Activation of D3 dopamine receptor decreases AT1 angiotensinreceptor expression in rat renal proximal tubule cells. Circ Res99:494–500

181. Sanada H, Yatabe J, Midorikawa S, Katoh T, Hashimoto S,Watanabe T, Xu J, Luo Y, Wang X, Zeng C, Armando I, FelderRA, Jose PA (2006) Amelioration of genetic hypertension bysuppression of renal G protein-coupled receptor kinase type 4expression. Hypertension 47:1131–1139

182. Van Tol HHM, Wu CM, Guan H-C, Ohara K, Bunzow JR,Civelli O, Kennedy J, Seeman P, Niznik HB, Jovanovic V (1992)Multiple dopamine D4 receptor variants in the human population.Nature 358:149–152

183. Sen S, Nesse R, Sheng L, Stoltenberg SF, Gleiberman L,Burmeister M, Weder AB (2005) Association between aDopamine-4 receptor polymorphism and blood pressure. Am JHypertens 18:1206–1210

184. Armando I, Wang X, Villar VA, Jones JE, Asico LD, Escano C,Jose PA (2007) Reactive oxygen species-dependent hypertensionin dopamine D2 receptor-deficient mice. Hypertension 49:672–678

185. Armando I, Wang X, Pascua AM, Villar VAM, Luo Y, Jones JE,Asico L, Escano C, Jose PA (2007) Regulation of renal NADPHoxidase and inflammation by dopamine D-2 receptor (abstract).Circulation 116[Suppl]:124

186. Staudacher T, Pech B, Tappe M, Gross G, Muhlbauer B,Luippold G (2007) Arterial blood pressure and renal sodiumexcretion in dopamine D3 receptor knockout mice. HypertensRes 30:93–101

187. Sugiyama F, Churchill GA, Higgins DC, Johns C, Makaritsis KP,Gavras H, Paigen B (2001) Concordance of murine quantitativetrait loci for salt-induced hypertension with rat and human loci.Genomics 71:70–77

188. Goldstein DS, Grossman E, Armando I, Wolfovitz E, Folio CJ,Holmes C, Keiser HR (1993) Correlates of urinary excretion ofcatechols in humans. Biog Amines 10:3–17

189. Gerlo EAM, Schoors DF, Dupont MG (1991) Age- and sex-related differences for the urinary excretion of norepinephrine,epinephrine, and dopamine in adults. Clin Chem 37:875–878

190. Lakatua DJ, Nicolau GY, Bogdan C, Plinga L, Jachimowicz A,Sackett-Lundeen L, Petrescu E, Ungureanu E, Haus E (1987)Chronobiology of catecholamine excretion in different agegroups. Prog Clin Biol Res 227B:31–50

191. Vanpée M, Herin P, Lagercrantz H, Aperia A (1997) Effect ofextreme prematurity on renal dopamine and norepinephrineexcretion during the neonatal period. Pediatr Nephrol 11:46–8

192. Sulyok E, Gyódi G, Ertl T, Bódis J, Hartmann G (1985) Theinfluence of NaCl supplementation on the postnatal developmentof urinary excretion of noradrenaline, dopamine, and serotonin inpremature infants. Pediatr Res 19:5–8

193. Meister B, Fried G, Holgert H, Aperia A, Hökfelt T (1992)Ontogeny of aromatic L-amino acid decarboxylase-containingtubule cells in rat kidney. Kidney Int 42:617–623

194. Armando I, Nowicki S, Aguirre J, Barontini M (1995) Adecreased tubular uptake of DOPA results in defective dopamineproduction in aged rats. Am J Physiol 268:F1087–F1092

195. Soares da Silva P, Fernandes MH (1991) A study of the renalsynthesis of dopamine in aged-rats. Acta Physiol Scand143:287–293

Pediatr Nephrol (2008) 23:2131–2146 2145

196. Soares-da-Silva P, Vieira-Coelho MA, Pestana M, FernandesMH, Guimarães JT (1995) Ontogeny of the cell outwarddopamine transporter in canine renal tissues. Fundam ClinPharmacol 9:255–262

197. Tenore G, Barili P, Sabbatini M, Tayebati SK, Amenta F (1997)Postnatal development of dopamine D1-like and D2-likereceptors in the rat kidney: a radioligand binding study. MechAgeing Dev 95:1–11

198. Kinoshita S, Felder RA (1990) Ontogeny of DA1 receptor-adenylate cyclase coupling in proximal convoluted tubules. Am JPhysiol 259:F971–F976

199. Svennilson J, Aperia A (1999) Dopamine in the developingkidney. Int J Dev Biol 43:441–443

200. Massmann GA, Zhang J, Rose JC, Figueroa JP (2006) Acute andlong-term effects of clinical doses of antenatal glucocorticoids in thedeveloping fetal sheep kidney. J Soc Gynecol Invest 13:174–180

201. Felder RA, Nakamura KT, Robillard JE, Kanadjian M, Jose PA(1988) Dopamine receptors in the developing sheep kidney.Pediatr Nephrol 2:156–162

202. Cantalamessa F, Barili P, Cavagna R, Sabbatini M, Tenore G,Amenta F (1998) Influence of neonatal treatment with thepyrethroid insecticide cypermethrin on the development ofdopamine receptors in the rat kidney. Mech Ageing Dev103:165–78

203. Fryckstedt J, Svensson LB, Lindén M, Aperia A (1993) Theeffect of dopamine on adenylate cyclase and Na+,K+-ATPaseactivity in the developing rat renal cortical and medullary tubulecells. Pediatr Res 34:308–311

204. Hussain T, Kansra V, Lokhandwala MF (1999) Renal dopaminereceptor signaling mechanisms in spontaneously hypertensiveand Fischer 344 old rats. Clin Exp Hypertens 21:25–36

205. Kansra V, Hussain T, Lokhandwala MF (1997) Alterations indopamine DA1 receptor and G proteins in renal proximal tubulesof old rats. Am J Physiol 273:F53–F59

206. Li XX, Xu J, Zheng S, Albrecht FE, Robillard JE, Eisner GM,Jose PA (2001) D1 dopamine receptor regulation of NHE3during development in spontaneously hypertensive rats. Am JPhysiol Regul Integr Comp Physiol 280:R1650–R1660

207. Kaneko S, Albrecht F, Asico LD, Eisner GM, Robillard JE,Jose PA (1992) Ontogeny of DA1 receptor-mediated natriuresisin the rat: in vivo and in vitro correlations. Am J Physiol 263:R631–R638

208. Li XX, Albrecht FE, Robillard JE, Eisner GM, Jose PA (2000)Gβ regulation of Na/H exchanger-3 activity in rat renal proximaltubules during development. Am J Physiol Regul Integr CompPhysiol 278:R931–936

209. Buckely NM, Brazeau P, Frazier ID (1983) Cardiovasculareffects of dopamine in developing swine. Biol Neonate 43:50–60

210. Fiser DH, Fewell JE, Hill DE, Brown AL (1988) Cardiovascularand renal effects of dopamine and dobutamine in healthy,conscious piglets. Crit Care Med 16:340–345

211. Gootman NB, Buckley BJ, Gootman PM, Nagelberg JS (1982)Age-related effects of single injections of dopamine on cardiovas-cular function in developing swine. Dev Pharmacol Ther 4:139–150

212. Vane DW, Weber TR, Caresky J, Grosfeld JL (1982) Systemicand renal effects of dopamine in the infant pig. J Surg Res32:477–483

213. Nakamura KT, Felder RA, Jose PA, Robillard JE (1987) Effectsof dopamine in the renal vascular bed of fetal, newborn, andadult sheep. Am J Physiol 252:R490–R497

214. Kohli JD, Goldberf LI (1987) Dopamine receptors: a classifica-tion based on physiological studies. In: Creese I, Fraser CM(eds) Dopamine receptors. Liss, New York, pp 97–114

215. Pelayo JC, Fildes RD, Jose PA (1984) Age-dependent renaleffects of intrarenal dopamine infusion. Am J Physiol 247:R212–R216

216. Segal JL, Smith FG, Guillery EN, Jose PA, Robillard JE (1992)Ontogeny of renal response to specific dopamine DA1-receptorstimulation in sheep. Am J Physiol 263:R868–R873

217. Seri I, Aperia A (1988) Contribution of dopamine2 receptors todopamine-induced increase in glomerular filtration rate. Am JPhysiol 254:F196–F201

218. Sulyok E (1988) Dopaminergic control of neonatal salt and watermetabolism. Pediatr Nephrol 2:163–165

219. Holtback U, Aperia A (2003) Molecular determinants of sodiumand water balance during early human development. SeminNeonatol 8:291–299

220. Cheung PY, Barrington KJ (1996) Renal dopamine receptors:mechanisms of action and developmental aspects. CardiovascRes 31:2–6

221. Vieira-Coelho MA, Hussain T, Kansra V, Serrao MP, GuimaraesJT, Pestana M, Soares-Da-Silva P, Lokhandwala MF (1999)Aging, high salt intake, and renal dopaminergic activity inFischer 344 rats. Hypertension 34:666–672

2146 Pediatr Nephrol (2008) 23:2131–2146