You are on page 1of 11

Pediatric Nephrology (2021) 36:479–489

https://doi.org/10.1007/s00467-020-04496-5

REVIEW

Renin-angiotensin system in mammalian kidney development


Ihor V. Yosypiv 1

Received: 26 December 2019 / Revised: 30 January 2020 / Accepted: 31 January 2020 / Published online: 18 February 2020
# IPNA 2020

Abstract
Mutations in the genes of the renin-angiotensin system result in congenital anomalies of the kidney and urinary tract (CAKUT),
the main cause of end-stage renal disease in children. The molecular mechanisms that cause CAKUT are unclear in most cases.
To improve the care of children with CAKUT, it is critical to determine the underlying mechanisms of CAKUT. In this review, we
discuss recent advances that have helped to better understand how disruption of the renin-angiotensin system during kidney
development contributes to CAKUT.

Keywords Kidney development . Renin-angiotensin . Prorenin receptor . Ureteric bud . Mesenchyme

Introduction Synopsis of kidney development

The renin-angiotensin system is essential for normal kid- Development of the permanent (metanephric) kidney in mam-
ney development [1–9]. Dysregulation of the renin- mals begins during the fifth week of gestation in humans and
angiotensin system during embryogenesis results in di- on embryonic (E) day E10.5 in mice (of a 19-day gestation)
verse forms of congenital anomalies of the kidney and from the outgrowth of the ureteric bud (UB) from the Wolffian
urinary tract (CAKUT) in mammals [1–9]. Mutations of (nephric) duct (WD) which invades the surrounding group of
the renin-angiotensin system genes in humans are associ- cells within the metanephric mesenchyme (MM) [11].
ated with renal tubular dysgenesis (RTD, OMIM 267430) Nascent UB tips interact reciprocally with self-renewing
[10]. Affected patients exhibit severe hypotension, and nephron progenitor cells (NPCs) of the MM and undergo suc-
most children with RTD die in the perinatal period, most cessive branching to form the renal collecting system, whereas
likely due to pulmonary hypoplasia (a consequence of MM cells condense around UB tips and differentiate into
oligohydramnios). Recent advances in the kidney and uri- nephrons. Stromal mesenchyme, which surrounds the MM
nary tract development field have expanded our under- and the UB, interacts with these compartments and gives rise
standing of the molecular mechanisms through which to different types of cells that include glomerular mesangial
renin-angiotensin system regulates normal kidney devel- cells, fibroblasts, vascular smooth muscle cells, renin cells,
opment. Here, we provide a comprehensive overview of pericytes, and mesenchyme surrounding the ureter [11, 12].
the molecular and cellular mechanisms by which intact The urinary bladder development begins during the fifth week
renin-angiotensin system directs renal morphogenesis of gestation in humans and at E11.5 in the developing mouse
and outline lineage-specific roles of how aberrations of fetus from the superior region of the urogenital sinus. UB-
the renin-angiotensin system during kidney development derived ureters initially join the bladder indirectly, via the
contribute to CAKUT. portions of WD (common WDs). Later in gestation, the ure-
ters detach from the WDs and fuse with the bladder to estab-
lish urine flow. Separation of ureters from the Wolffian duct
followed by ureter-bladder fusion is achieved through spatio-
* Ihor V. Yosypiv temporally regulated apoptosis of common WDs [13, 14].
iiosipi@tulane.edu Developmental defects that disrupt normal kidney or bladder
1
development result in a diverse array of congenital anomalies
Section of Pediatric Nephrology, Department of Pediatrics,
Hypertension and Renal Center of Excellence, Tulane University
of the kidney and urinary tract (CAKUT) which include renal
Health Sciences Center, 1430 Tulane Avenue, New agenesis, kidney hypodysplasia, and ureter anomalies that can
Orleans, LA 70112, USA cause reflux of the urine or urinary tract obstruction [11].
480 Pediatr Nephrol (2021) 36:479–489

Expression, activity, and signaling II (Ang II), the major effector peptide of the renin-
of the renin-angiotensin system during mammalian angiotensin system, acts via the Agtr1 and Agtr2 receptors.
kidney development In rodent kidney, Agtr2 is detected earlier than Agtr1, peaks
early during gestation, and rapidly declines after birth [23, 33].
In the rodent kidney, angiotensinogen (ATG), the substrate for Unlike the Agtr2, Agtr1 levels are low in early gestation, in-
enzyme renin which generates angiotensin I (Ang I) from crease during gestation, peak perinatally, and decline gradual-
ATG, is detected in the UB, proximal tubules, and renal stro- ly thereafter [23, 33]. On E12, AGTR1 protein expression in
ma [15] (Table 1). In humans, ATG mRNA is localized in the the mouse kidney is low in the UB and the adjacent metaneph-
proximal tubule on the eighth week of pregnancy and remains ric mesenchyme [15]. In the E14 and E15 mouse kidney,
expressed thereafter [17]. Expression of renin mRNA in the AGTR1 immunoreactivity becomes localized on the
embryonic mouse kidney is detected at E14.5 in a small num- basolateral and luminal sides of UBs and their derivatives.
ber of cells scattered throughout the kidney [18]. On E15.5, At E16, AGTR1 is detected in UB branches, glomeruli,
renin becomes abundantly expressed in the renal arteries, as microvessels, stromal mesenchyme, and proximal tubules.
well as in the arcuate and interlobar arteries. Later in gestation, AGTR2 protein and mRNA are detected in the mouse embry-
renin localizes in the juxtaglomerular cells [18]. Renin mRNA onic kidney from E11.5 and are confined to the mesenchyme
is expressed in the human metanephros at the glomerular vas- adjacent to UB tips and in the UB [22]. At E14, AGTR2
cular pole and in arteries adjacent to glomeruli on the sixth immunostaining is present in the developing nephrons, med-
week of gestation [17]. Aside from cleaving ATG to produce ullary stroma, and branching UB. On E16, AGTR2 is abun-
Ang I, renin acts via the prorenin receptor (PRR (also called dantly expressed in tubules localized in the inner cortex and
ATP6AP2)) that triggers several intracellular signaling path- look like proximal tubules [22]. In the human embryonic kid-
ways, including p38 kinase in kidney mesangial and vascular ney, AGTR1 and AGTR2 mRNAs are expressed on E30-E35
smooth muscle (VSMC) cells, extracellular signal-regulated of gestation [17]. At this time, AGTR1 mRNA is expressed in
kinases 1 and 2 (Erk1/2), and phosphatidylinositol 3-kinase/ glomeruli and proximal tubules, whereas AGTR2 mRNA is
Akt (PI3K/Akt) in human embryonic kidney cells grown present in the mesenchyme [17]. Similar to temporal trends of
in vitro [28, 29]. The PRR is a single trans-membrane domain Agtr2 mRNA expression in rodents, AGTR2 mRNA levels
receptor encoded by the Atp6ap2 (ATPase-associated decrease after 20 weeks of gestation in humans. In contrast
protein2) gene located on the X chromosome in humans to AGTR2 mRNA expression profile, AGTR1 mRNA is
(ATP6AP2) and rodents (Atp6Aap2) [30]. In addition to being expressed for the whole duration of metanephric kidney de-
a receptor for renin/prorenin, PRR co-purifies with the vacu- velopment [17]. Immunoreactive kidney tissue Ang II con-
olar proton pump V-ATPase and is considered to be its acces- tents are higher in the developing rodent kidney compared to
sory protein [31]. Atp6ap2 mRNA and protein are expressed the adult kidney (rat kidney: P1 667 ± 75, adult 103 ± 6 fmol/g
in the mouse metanephros on E12.5 [24]. PRR immunoreac- tissue; mouse kidney: E17.0 800 ± 60, P1 975 ± 136, adult
tivity is detected in the UB and developing nephrons on E13.5 200 ± 50 pg/g tissue) [6, 34]. In summary, ATG, renin, ACE,
[24]. Later during gestation (on E16.5 and E18.5), PRR is Agtr1, and Agtr2 are expressed during early metanephric kid-
most abundantly present in the tubules which resemble ney development in humans and rodents, indicating a role for
collecting ducts (CDs) as well as in the glomerular mesangium kidney tissue Ang II in normal kidney organogenesis.
[24]. Kidney PRR protein expression is high during gestation
and declines gradually after birth in the mouse [24]. In the Lineage-specific requirement of renin-angiotensin
neonatal human kidney, PRR localizes mainly in the glomer- system signaling during murine kidney development
uli, proximal tubules, collecting ducts, and arteries [27]. PRR
expression in neonatal kidneys is correlated inversely with Studies utilizing knockout mouse models have revealed a crit-
gestational age and is increased in neonates compared to older ical role of the renin-angiotensin system during kidney devel-
children. These data are consistent with the hypothesis that opment and greatly amplified our understanding of how dis-
PRR plays a role in nephrogenesis in humans. ruptions in the renin-angiotensin system result in CAKUT.
Ang I is converted to Ang II, the major effector peptide of Genetic inactivation of the Atg, renin, Atp6ap2, Ace,
the renin-angiotensin system, by angiotensin-converting en- Agtr1a;Agtr1b, or Agtr2 in mice is either lethal (Atp6ap2),
zyme (ACE). In the rat kidney, Ace mRNA is present on indicating an essential role of the PRR in embryonic develop-
E16 in the developing glomeruli and proximal tubule [20]. ment or leads to a spectrum of abnormalities in the develop-
During invasion of the S-shaped bodies by capillaries, which ment of the metanephros [1–9] (Table 2). Complete deletion
occurs around 20 weeks of gestation in humans, ACE immu- of renin (Ren1c), Atg, Ace, or Agtr1a;Agtr1b in mice leads to
nostaining is present in glomerular endothelial cells [17]. hypoplastic inner medulla and hydronephrosis that become
Later during gestation, at E30-E35, ACE immunoreactivity apparent and progress following birth. Agtr1a; Agtr1b-defi-
is localized in proximal tubules and CDs [32]. Angiotensin cient mice (lacking genes encoding both Agtr1 and Agtr2
Pediatr Nephrol (2021) 36:479–489 481

Table 1 Expression of the renin-angiotensin system components during metanephric kidney development

E12 E14 E15 E16 E19 References

ATG
Mouse UB, SM UB, SM, PT PT, UB, SM PT, UB, SM [15]
Rat UB, SM PT, UB, SM PT [16]
Human From 8 weeks and throughout gestation [17]
PT
Renin
Mouse Precursor cells M of entire M of entire M close to V, G V, G [18]
present in M/SM kidney kidney
Rat V V V [19]
Human E30-E35
At the vascular pole of the glomerulus and [17]
arteries located next to glomeruli
ACE
Rat PT, G, CD PT, G, CD [20]
Human E30-E35 until birth
PT, CD [17]
AGTR1
Mouse UB, M UB, G UB, V PT, UB, SM, G PT, DT [15, 26]
Rat G, UB, SM SM PT, CD, G [21]
Human E30-E35 until birth [17]
G, PT, JG cells
AGTR2
Mouse UB, M UB, M, SM UB, SM Inner cortical [22]
tubules
M M, under renal M between M between [23]
capsule CDs CDs
Under renal Under renal
capsule capsule
Rat M Condensed M Medulla, G, V [21]
Human E30-E35 until birth [17]
M
PRR/ATP6AP2
Mouse UB, CM, SM UB, G CDs CDs, G [24–26]
Human From E33
G, PT, CD, arteries [27]

ATG angiotensinogen, ACE angiotensin-converting enzyme, AGTR1/AGTR2 angiotensin II receptors, PRR/ATP6AP2 prorenin receptor, UB ureteric
bud, M mesenchyme, CM cap mesenchyme, SM stromal mesenchyme, PT proximal tubule, G glomeruli, V renal vessels, CD collecting duct

receptors) exhibit hypoplasia of the ureteral smooth muscle, development of the medulla by promoting vasa recta forma-
resulting in impaired ureteral peristalsis and increased pres- tion [45]. A total of 3.1% of Agtr2-knockout mice have duplex
sure within the renal pelvis [6]. Thus, small medulla and renal collecting systems, vesicoureteral reflux, and
hydronephrosis seen in Ren1c-, Atg-, Ace-, or Agtr1a; hydronephrosis [41]. The number of UB tips is decreased in
Agtr1b-knockout mice may result from functional urinary Agtr2-null compared with Agtr2+/+ mouse kidneys, indicating
tract obstruction causing increased backward pressure to the that Agtr2 signaling promotes early UB branching morpho-
renal tissue. Given that activation of the Agtr1 by Ang II genesis [44]. Inductive effects of the Agtr2 on UB branching
counteracts shrinkage of the developing papillas grown are mediated by increased UB cell proliferation and survival
in vitro, Agtr1 signaling may also promote structural develop- [44]. In terms of the effect of the global deletion of Ren1c-,
ment of the medulla by induction of medullary CD elongation Atg-, Ace-, or Agtr1a; Agtr1b genes on renal function, mutant
[44]. In addition, Agtr1 signaling may contribute to structural mice have polyuria and a decreased urine concentrating ability
482 Pediatr Nephrol (2021) 36:479–489

Table 2 Renin-angiotensin
system gene mutations associated Gene/model Renal phenotype Reference
with murine and human CAKUT
Murine models
Atg−/− Vascular thickening, interstitial fibrosis, delayed glomerular [1, 2]
maturation, hypoplastic papilla hydronephrosis, reduced
ability to concentrate urine
Ren1−/− Arterial wall thickening, interstitial fibrosis, [3]
glomerulosclerosis, hypoplastic papilla hydronephrosis
Hoxb7Cre;Renflox/flox No structural renal phenotype [35]
Foxd1Cre;Renflox/flox Absence of renin, hypertrophic arteries, hydronephrosis
Atp6ap2−/− Embryonic lethal [36]
Hoxb7Cre;Atp6ap2flox/flox Kidney hypoplasia, thin cortex, reduced UB tip and nephron [37]
number, polyuria impaired urine-concentrating ability
Aqp2Cre;Atp6ap2flox/flox No structural renal phenotype, polyuria, reduced urine [38]
osmolality
V-ATPaseB1Cre;Atp6ap2flox/flox No structural renal phenotype
Six2Cre;Atp6ap2flox/flo Reduced developing nephron volume and size, delayed [25]
nephrogenesis, small cystic kidneys, podocyte foot
process effacement
Nphs2Cre;Atp6ap2flox/flox Foot process effacement, early postnatal death from kidney [8, 9]
failure, severe proteinuria
Foxd1Cre;Atp6ap2flox/flox Decreased number of glomeruli, expansion of Six2+ nephron [26]
progenitors, marked decrease in arterial and arteriolar
development
Ace−/− Arterial wall thickening, hypoplastic papilla and medulla, [4]
hydronephrosis, reduced ability to concentrate urine
Agtr1a;Agtr1b−/− Decreased kidney weight, hypoplastic papilla and medulla, [5, 7]
hydronephrosis, delayed glomerular maturation, arterial
wall thickening, interstitial fibrosis, tubular atrophy
reduced ability to concentrate urine
Hoxb7Cre;Agtr1aflox/flox No structural abnormalities, reduced Aqp2 levels and urine [39]
concentrating
Aqp2Cre;Agtr1aflox/flox ability after water deprivation
Foxd1Cre;Agtr1flox/flox No structural or functional abnormalities [40]
Agtr2−/− Duplicated ureters, hydronephrosis, decreased UB branching [22, 41]
Human CAKUT
ATG−/− RTD: Reduced number of proximal tubules, short proximal [10, 42]
tubules without brush border, atrophic loops of Henle and
collecting ducts, closely packed glomeruli, marked
thickening and disorganization of interlobular and
preglomerular arteries
AGTR1−/− RTD, similar to AGT phenotype, PUV [10, 42]
AGTR2−/− UPJ obstruction, megaureter, MCDK, hydronephrosis, PUV [43]
ACE−/− RTD, similar to AGT phenotype, renal hypodysplasia, PUV [10, 42]
REN−/− RTD, similar to AGT phenotype [10, 42]

Agtr1/AGTR1 angiotensin II receptor type 1, Agtr2/AGTR2 angiotensin II receptor type 2, Ace/ACE angiotensin-
converting enzyme, Aqp2 aquaporin 2, PUV posterior urethral valves, CAKUT congenital anomalies of the kidney
and urinary tract, UPJ uretero-pelvic junction, RTD renal tubular dysgenesis, Ren1 renin 1, Atg/ATG
angiotensinogen, Ang angiotensin, Atp6ap2 gene that encodes prorenin receptor (PRR), UB ureteric bud, CD
collecting duct, PC principal cell of collecting duct, IC intercalated cell of collecting duct

[3, 4, 7]. AGTR1a subtype is a murine homolog to the human nephron or impaired water reabsorption. In this regard, re-
AGTR1. Complete deletion of the Agtr1a subtype in mice duced expression of the thiazide-sensitive sodium-chloride
results in normal renal tissue structure, low blood pressure, cotransporter and the alpha-subunit of the amiloride-
polyuria, and impaired capability to concentrate the urine sensitive epithelial Na+ channel are observed in Agtr1a-
[46]. Reduced ability to concentrate the urine in Agtr1a-null knockout compared with wild-type mice [47]. In addition,
mice may result from decreased expression of solute trans- Agtr1a−/− mice have reduced expression of AQP2 protein in
porters that are important for salt reabsorption along the the inner medulla and decreased vasopressin levels in the
Pediatr Nephrol (2021) 36:479–489 483

circulation [48]. Thus, decreased urine concentrating ability in branching and increased UB cell apoptosis [37].
Agtr1a−/− mice may be due to impaired interactions between Hoxb7Cre+;Atp6ap2flox/flox mice have reduced expression of
AGTR1a and arginine vasopressin (AVP) receptor in the inner genes crucial for nephrogenesis and UB branching morpho-
medulla. Targeted inactivation of the individual components genesis. Genes with reduced expression include glial cell-
of the renin-angiotensin system in mice has allowed circum- derived neurotrophic factor (GDNF), its receptor rearranged
vention of embryonic lethality and determination of renin- during transfection (Ret), and genes known to be downstream
angiotensin system functions in the UB, nephrogenic, and targets of Ret such as Wnt11, Etv4, and Etv5 (Fig. 1).
stromal compartments (Table 2). Following birth, Hoxb7Cre+;Atp6ap2flox/flox mice have small
dysplastic kidneys with thin cortex and reduced number of
UB lineage nephrons. Reduced nephron endowment in this model is like-
ly due to decreased UB tip number. Later in life,
Targeted deletion of the Ang II Agtr1a subtype A (Agtr1a−/−) Hoxb7Cre+;Atp6ap2flox/flox mice have increased levels of se-
in the UB lineage in mice using Hoxb7Cre (in all UB-derived rum creatinine and develop chronic kidney disease. In addi-
CD cells) or aquaporin 2 (Aqp2) Aqp2Cre (in principal cells tion to renal structural anomalies, Hoxb7Cre+;Atp6ap2flox/flox
(PC)) allows determination of the role of the AGTR1 in the mice exhibit reduced expression of winged helix transcription
regulation of urine-concentrating ability in the absence of sys- factor Foxi1 (a transcription factor that promotes differentia-
temic AGTR1A-mediated effects observed in mice with com- tion of the CD intercalated cells (IC) from precursor UB cells),
plete inactivation of the Agtr1a. Deletion of the Agtr1a in basolateral membrane chloride-bicarbonate transporter anion
either all CD cells or in PC does not alter systemic blood exchanger 1 (AE1), V-ATPase α4 subunit, and Aqp2 mRNA
pressure, kidney morphology, or baseline urinary concentrat- [37]. Functionally, Hoxb7Cre+ ;Atp6ap2 flox/flox mice are
ing ability [39]. However, both strains of mutant mice exhibit polyuric, have reduced capacity to concentrate, and acidify
dampened increase in osmolality of the urine following ad- the urine after 48 h of NH4Cl administration in drinking water
ministration of desmopressin, a synthetic analogue of the despite no difference in water intake compared with control
antidiuretic hormone arginine vasopressin, or after water dep- wild-type mice [37]. To explore in-depth the underlying
rivation [39]. These changes may result from reduced expres- mechanisms by which UB PRR regulates UB branching and
sion of AQP2 in the medulla and not from changes in salt CD morphogenesis, we performed a whole-genome analysis
delivery into renal tubules or altered expression of the vaso- of RNA expressed in UB cells FACS-isolated from kidneys of
pressin V2 receptor mRNA in the renal medulla [39]. Hoxb7Cre+;Atp6ap2flox/flox and control mice on day of birth
Activation of the AGTR1 is also reported to induce alkalini- (P0) and carried out gene ontology analysis to identify func-
zation of the CD lumen in the rabbit [49, 50]. These findings tional groups of genes that are differentially expressed [51].
indicate that AGTR1 activation by Ang II may facilitate func- Expression of several genes expressed specifically in the UB
tional maturation of CD cells that regulate acid-base and water cells and previously demonstrated to regulate UB branching
balance. was altered in mutants. Genes with decreased expression in-
Renin is detected in the developing mouse CD on E15.5 cluded Wnt11, Bmp7, Etv4, and Gfrα1, whereas levels of
[18]. Global deletion of the Ren1c gene in mice results in Wnt9b, β-catenin, and Fgfr2 mRNA were increased.
hydronephrosis and inability to concentrate the urine, suggest- Because intact β-catenin activity in the UB lineage is critical
ing that CD renin might be responsible for this phenotype [3]. for normal UB branching, we next determined β-catenin sig-
Sequeira-Lopez et al. examined whether UB/CD renin plays a naling in UB cells in vitro and in vivo. Infection of immortal-
role in the development of the CD derivatives during kidney ized UB cells with adenovirus-driven short hairpin (sh) anti-
development [35]. Conditional Ren1c deletion in the UB/CD sense shPRR in vitro increased levels of active form of β-
lineage was achieved using Hoxb7-cre mice, which were catenin as well as the expression of direct β-catenin target,
crossed to Ren1c flox/flox mice. However, lack of renin in the axin2. The same effect of disrupted PRR signaling in the
UB/CD did not affect kidney or renal vascular morphology, UB on β-catenin activity in UB epithelia was observed
expression, and distribution of renin in the kidney. The authors in vivo in double-transgenic Hoxb7Cre+;Atp6ap2flox/flox/
conclude that renin is dispensable in the renal collecting sys- BatGal+ mice. In BatGal+ transgenic mice, activation of β-
tem for kidney development. catenin-responsive promoter by endogenous β-catenin in-
As UB-derived CD is the predominant nephron site of PRR creases expression of nuclear β-galactosidase (BatGal) [51].
expression, we used a cre-lox conditional targeting approach In addition to genes known to regulate UB branching morpho-
to delete Atp6ap2 (gene that encodes PRR protein) specifical- genesis, the functional groups of genes with decreased expres-
ly in the UB lineage using Hoxb7Cre driver and examine the sion in Hoxb7Cre+;Atp6ap2flox/flox UB cells included genes
role of the UB PRR on kidney development in mice that regulate metabolism of amino acids, molecular transport,
(Hoxb7Cre+;Atp6ap2flox/flox) [37]. Knockout and production of energy. These findings show that UB PRR
Hoxb7Cre+;Atp6ap2flox/flox metanephroi have decreased UB directs UB branching and modulates CD function by
484 Pediatr Nephrol (2021) 36:479–489

a b UB lineage

Gdnf, Ret, Wnt11, E12.5: Reduced number of UB tips and


Six2+ NP Gdnf Etv4/5 nephrons
Ret E18.5: Small kidneys
Wnt11 Foxi1, AE1,
P30: Polyuria, reduced capacity to
V-ATPase α4
Hoxb7+ concentrate and acidify the urine
RV
UB
Hoxb7Cre+;Atp6ap2flox/flox

Foxd1+ Stroma

c MM lineage
Lef1,Jag1, Wnt4, P0: Reduced nephron number, Nephrin, podocin, P7: Podocyte foot process
Lhx1, Fgf8, Six2, block in nephrogenesis after RV WT1, V-ATPase C effacement
Cited1, Sall1, Notch1 stage, small cystic kidneys, early P21: Proteinuria, reduced
postnatal death renal function
Six2Cre;Atp6ap2flox/+:
Six2Cre+;Atp6ap2flox/flox P60: Focal glomerulosclerosis, Nphs2Cre+;Atp6ap2flox/flox
proteinuria, reduced renal funcon
d Stromal lineage
E16.5: Absence of renin Sall1, Jag1, WT1, E18.5: Decreased RV and
Absence of renal P0: Neonatal mortality expansion of Six2+ NPs
renin, expanded renal glomerular number, decre-
P14: Thickened renal arteries and ase in arterial and arteriolar
α-SMA expression arterioles Foxd1, Meis1, development
P60: Hydronephrosis, reduced renal α-SMA, renin
function and urine osmilality
Foxd1Cre+;Renflox/flox Foxd1Cre+;Atp6ap2flox/flox
Fig. 1 Lineage-specific roles of the renin-angiotensin system signaling in nephrogenesis by maintaining appropriate balance between NP mainte-
the developing kidney. a Schematic of major lineages and select lineage- nance and induction of nephron differentiation. Podocyte ATP6AP2 di-
specific molecular markers in the developing kidney. NP nephron pro- rects normal podocyte development and function via maintenance of
genitors, UB ureteric bud, RV renal vesicle, GDNF glial cell-derived normal V-ATPase expression and function in podocytes. Lef1, Jag1,
neurotrophic factor. b In the UB lineage, absence of the UB prorenin Wnt4, Lhx1, Fgf8, Notch1, WT1—nephron differentiation markers.
receptor (ATP6AP2) signaling causes a reduction of Gdnf-Ret-Wnt11 Cited1, Six2, Sall1—NP markers. d In the Foxd1+ stromal lineage, renin
pathway gene expression, causing decreased UB branching. Foxi1, and ATP6AP2 are critical for normal renal arterial tree development.
AE1 (chloride-bicarbonate anion exchanger 1), and V-ATPase direct ter- ATP6AP2 promotes arterial development by maintaining expression of
minal differentiation and function of UB-derived collecting duct cells. c stromal markers Foxd1 and Meis1. ATP6AP2 also facilitates normal
In the metanephric mesenchyme (MM) lineage, ATP6AP2 promotes nephron differentiation by inhibiting excessive expansion of Six2+ NPs

regulating the expression of genes that guide UB branching renin vs. PRR deficiency in the UB/CD lineage for kidney
and determine terminal differentiation of the CD cells. development may result from the difference in signaling path-
Moreover, UB/CD PRR is important for the function of CD ways downstream of renin and PRR, where UB PRR affects
cells involved in acid-base and water homeostasis. renal development primarily as a subunit of V-ATPase [30].
Ramkumar et al. investigated how deletion of Atp6ap2 in An additional possibility is that lack of renal structural pheno-
IC or in PC of the CD affected renal morphology and water type in Hoxb7Cre+;Ren1c flox/flox mice may be due to a rescue
and Na+ handling in mice [38]. IC- or PC-targeted Atp6ap2 by renin-independent Ang peptide generation from AGT by
knockout (KO) was achieved by crossing mice expressing Cre proteases [52].
recombinase under the control of the B1 subunit of the V-
ATPase (Atp6v1b1) or Aqp2 promoters, respectively, with Nephrogenic lineage
Atp6ap2-floxed mice. No gross alterations in kidney tissue
histology or anatomy were found in IC- or PC-mutant mice During renal morphogenesis, PRR expression is prominent
at 21 days of age [38]. Expression of the renal epithelial med- within the metanephric mesenchyme and its derivatives. To
ullary channel ENaC-α was reduced in PC-mutant mice. This determine the role of the PRR (ATP6AP2) within Six2+
was accompanied by an increased loss of Na+ in the urine in nephrogenic progenitor population, we deleted Atp6ap2 in
PC-mutant compared to control mice. In addition, expression Six2+ nephron progenitors and their epithelial derivatives in
of renal medullary AQP2 and urine osmolality was reduced, mice using Six2 Cre -mediated ablation of the Atp6ap2
whereas urine volume was increased in PC-KO compared (Six2Cre+;Atp6ap2flox/flox) [25]. We did not observe changes
with control mice. In contrast, ENaC or AQP2 expression, in UB tree volume or in the number of UB tips in
Na+ balance, urine volume, and osmolality did not differ in Atp6ap2-mutant compared with control mice on E12.5. In
IC-KO compared with control mice. Thus, PC PRR regulates contrast, average size of developing glomeruli and glomerular
ENaC activity, water transport, and urinary Na+ excretion. volume were reduced in Six2Cre+;Atp6ap2flox/flox kidneys at
One possible explanation for a discrepancy in the impact of E12.5. The number of renal vesicles (RV) did not differ
Pediatr Nephrol (2021) 36:479–489 485

between mutant and control kidneys on E12.5. In contrast, the the extracellular domain of the full-length PRR and is secreted
number of comma- and S-shaped bodies was reduced in into the plasma where it is able to bind renin [57]. Increased
Six2Cre+;Atp6ap2flox/flox mice at this stage. We conclude that urinary sPRR contents in Het mice may result from renal
glomerular structure volume is reduced in tissue damage (decreased glomerular number, segmental
Six2Cre+;Atp6ap2flox/flox mutants on E12.5 due to a block in thickening of the glomerular basement membrane) [56]. In
nephrogenesis that occurs after the RV phase. Detailed analy- turn, increased levels of sPRR in the urine may induce
sis showed that the expression of genes critical for nephron prorenin/reninactivity in the collecting duct, causing increased
induction, such as Wnt4, Fgf8, Pax8, Lhx1, Lef1, Jag1, and local formation of Ang II which may contribute to increased
Fgf20, is decreased in Six2Cre+;Atp6ap2flox/flox mutants (Fig. blood pressure in Het mice. Collectively, these findings indi-
1) [25]. To determine whether reduced nephrogenesis in cate that NPC PRR plays a critical role in nephrogenesis by
Six2Cre+;Atp6ap2flox/flox mice results from early depletion of regulating the expression of genes important in directing di-
nephron progenitor cells, we examined the expression of mo- verse cellular activities. It is likely that decreased nephron
lecular markers of CM cells with self-perpetuating ability such number and increased excretion of sPRR in the urine play a
as Six2, Cited1, and Sall1 [53]. Reduced expression of neph- role in developmental programming of hypertension in
ron progenitor markers Six2, Cited1, and Sall1 was noted in Six2Cre+;Atp6ap2flox/+ mice.
mutant compared to control kidneys, suggesting that reduced Our observations on the role of NPC PRR in podocyte
nephron induction in Six2Cre+;Atp6ap2flox/flox mice may be morphology and function are in line with data by Oshima
due to premature depletion of nephron progenitors with pre- et al. and Riediger et al., showing that PRR is critical in
cocious propensity of nephron progenitors to differentiation podocytes. Deletion of Atp6ap2 in mouse podocytes with
[25]. Following birth, Six2Cre+;Atp6ap2flox/flox kidneys are re- Cre-recombinase driven by the podocyte-specific promoter
duced in size, lack a well-developed nephrogenic zone, have of the Nphs2 (Nphs2Cre+;Atp6ap2flox/flox) mice causes efface-
reduced number of nephrons, and contain cysts in the outer ment of the podocyte foot processes, decreased expression,
medulla and cortex. Ultrastructural changes observed in and altered subcellular distribution podocin and nephrin, crit-
Six Cre+ ;Atp6ap2 flox/flox kidneys include effacement of ical constituents of the podocyte slit-diaphragm, massive pro-
podocyte foot processes, a characteristic finding observed in teinuria, and early death following birth from kidney injury
renal glomerular proteinuric disease. At 2 months of age, plas- (Table 1) [8, 9]. Additional changes demonstrated in
ma creatinine and urinary excretion of albumin were increased Nphs2Cre+;Atp6ap2flox/flox podocytes include decreased ex-
in heterozygous Six2Cre+;Atp6ap2flox/+ compared with control pression of the V0 c-subunit (Atp6v0c) of the V-ATPase.
mice. Premature cessation of nephrogenesis results in de- Reduced V0 c levels are associated with decreased ability to
creased number of nephrons and is linked to kidney hypopla- acidify lysosomes, increased expression of lysosomal and
sia, proteinuria, increased risk of hypertension, and chronic autophagosomal markers. Observed alterations are consistent
kidney disease in later life [54, 55]. To investigate in-depth with reduced ability of lysosomes and autophagosomes to
the molecular and cellular mechanisms through which neph- process intracellular cargo [8, 9]. Autophagy represents an
ron progenitor cell (NPC) PRR regulates nephrogenesis, we important physiological mechanism for intracellular recycling
searched for genes regulated by PRR in Six2+ NPCs FACS- of proteins and damaged organelles to maintain homeostasis.
isolated from control and Six2Cre+;Atp6ap2flox/flox kidneys on Normal autophagy relies on appropriate pH within the lyso-
E15.5 utilizing microarray analysis. The expression of β-ca- somes and autophagosomes. Given reduced lysosomal acidi-
tenin, Notch1, Jag1, Lhx1, Lef1, and p53, genes known to fication and increased expression of lysosomal and
regulate kidney development, was reduced in mutant com- autophagosomal markers in Nphs2 Cre+ ;Atp6ap2 flox/flox
pared with control kidneys [56]. The groups of downregulated podocytes, the authors conclude that PRR controls acidifica-
genes include genes that control cell and embryo develop- tion of lysosomes and phagosomes to protect podocytes from
ment, regeneration of the kidney tissue, cell organization and decreased protein recycling resulting from compromised au-
assembly, cell structure, proliferation, and apoptosis. Because tophagy. Lineage-specific effects of other renin-angiotensin
Six2Cre+;Atp6ap2flox/flox mice did not survive beyond the first system components within the Six2+ nephrogenic or forkhead
2 days after birth, we investigated the effect of Atp6ap2 gene transcription factor 1 (Foxd1+) stromal precursor populations
dosage reduction in Six2Cre+;Atp6ap2flox/+ (Het) mice on the have not been defined. Additional studies are required to de-
number of nephrons, ultrastructure of the glomerular base- termine the role of renin-angiotensin system actions via the
ment membrane (GBM), and systemic blood pressure. Het AT2R within these lineages.
mice had a reduced number of glomeruli, GBM with irregular
thickening and focal effacement of podocyte foot processes. Stromal lineage
In addition, Het mice had elevated systemic blood pressure
and increased urinary excretion of soluble form of PRR Renin-secreting juxtaglomerular (JG) cells, which originate
(sPRR). sPRR is formed by protease-mediated cleavage of from Foxd1-expressing stromal cells, differentiate into
486 Pediatr Nephrol (2021) 36:479–489

pericytes, glomerular mesangium, and mural cells of the kid- descendants, in contrast to characteristic renal structural and
ney arterioles [35]. To determine the role of the stromal renin functional consequences observed in mice with global
in kidney development, Sequeira-Lopez et al. deleted renin in Agtr1a;Agtr1b inactivation, does not disturb normal kidney
Foxd1+ stromal cells and all their descendants by crossing development [7, 40]. It is conceivable that normal kidney
Ren1c flox/flox mice with Foxd1-cre mice development in Foxd1 Cre+/Agtr1flox/flox mice is compensated
(Foxd1Cre+;Ren1cflox/flox) [35]. It remains controversial by AT1R activity in compartments beyond the stromal lineage,
whether Foxd1+ cells can form renal endothelial cells [35, i.e., in cells expressing renin, renal mesangial cells, interstitial,
58]. Removal of renin in the Foxd1+ cells resulted in neonatal or vascular smooth muscle cells.
mortality. The kidneys of mice that survived immediately after
birth showed a marked decrease in renin expression, thick Renin-angiotensin system inhibitor fetopathy
arteries, reduced plasma renin levels, and hydronephrosis.
Thus, renin generated in the Foxd1+ stromal compartment is Use of renin-angiotensin system inhibitors (RASi) such as
crucial for normal kidney and renal vasculature development ACE inhibitors (ACEi), Ang II receptor blockers (ARBs),
and that main source of circulating renin is the kidney and direct renin inhibitors (DRI) is associated with an in-
vasculature. creased risk of fetopathy in humans. The severity of fetopathy
We recently investigated the role of the stromal PRR in appears to be related to the timing of exposure to renin-
kidney development in mice using conditional deletion of angiotensin system-inhibiting drugs during gestation and to
the Atp6ap2 in Foxd1+ stromal progenitors by crossing the drug type taken by pregnant women. Pulmonary hypopla-
Atp6ap2 flox/flox mice with Foxd1-cre mice sia requiring mechanical ventilation and renal injury that need
(Foxd1Cre+;Atp6ap2flox/flox) [26]. Similar to renal replacement therapy occur more frequently in children
Foxd1Cre+;Ren1cflox/flox mice, removal of PRR in the Foxd1+ with RASi exposure after the first trimester compared to chil-
cells resulted in death within several days after birth. The dren exposed during the first trimester [59]. A recent study
kidneys of living mice showed decreased number of renal reports that fetopathy was not observed when RASi were
arteries and arterioles, as evidenced from decreased expres- discontinued before 20 weeks of gestational [60]. In contrast,
sion of α smooth muscle actin (α-SMA), in the developing 90% of pregnancies had oligohydramnios and 31% had signs
interstitium. Expression of renin in the renal arterioles and of RASi fetopathy when exposed to RASi after the first tri-
arteries was markedly reduced in Foxd1Cre+;Atp6ap2flox/flox mester [60]. The rate of fetopathy was higher for ARBs
compared with control mice. These data indicate that PRR (29.2%) than for ACEi (3.2%). Considering a broad-
plays a critical role in the development of cells that express spectrum of significant adverse effects of RASi on fetal
renin and that renin-expressing cells are required for the cor- wellbeing, use of all drugs that block the renin-angiotensin
rect formation of the kidney arteries. Expression levels of system should be avoided during all trimesters of pregnancy.
Foxd1 and Meis1, molecular markers of renal stroma, were All patients of potential childbearing age should be counseled
also reduced in Foxd1 Cre+ ;Atp6ap2 flo x/flox kidneys. about the potential fetal risks associated with RASi and the
Subsequently, expanded expression of Six2+ nephron progen- importance of contraception when taking these drugs.
itor population accompanied by slower differentiation of the
developing nephrons was observed in mutants (Fig. 1) [26].
These findings demonstrate that PRR present in renal stromal Conclusion
mesenchyme is critical for normal differentiation of the renal
arteries. In turn, deficient arterial development in Mutations in the renin-angiotensin system genes are associat-
Foxd1Cre+;Atp6ap2flox/flox mice may cause excessive expan- ed with renal malformations in mammals and humans. These
sion of nephron progenitors to impede proper formation of the observations indicate an important role for the renin-
kidney. angiotensin system in normal kidney development. Recent
Schrankl el al. studied the role of the stromal Ang II AT1R studies in mice have demonstrated lineage-specific roles for
in kidney development in mice with targeted inactivation of discrete components of the renin-angiotensin system in the
the Agtr1a;Agtr1b receptor subtypes in the Foxd1+ cells UB, nephrogenic mesenchyme, and stromal lineages during
(Foxd1 Cre+/Agtr1flox/flox) [40]. The major AT1A receptor sub- embryonic development. The role of the PRR signaling in
type was expressed in vascular smooth muscle cells, renin, cell-specific lineages (e.g., in PCs and ICs of the CD, in
mesangial, and interstitial cells, which all derive from podocytes) has helped to define the cell-autonomous and
Foxd1+ stromal progenitors. The kidneys showed neither non-cell autonomous mechanisms underlying CAKUT and
structural nor functional (blood pressure and glomerular filtra- renal functional derangements. As aberrant renin-angiotensin
tion rate) abnormalities in comparison with wild-type mice. system impairs normal kidney development, these findings
These findings suggest that targeted deletion of the further underscore a need to avoid the use of inhibitors of
Agtr1a;Agtr1b in renal stromal progenitors and their the renin-angiotensin system during pregnancy and advocate
Pediatr Nephrol (2021) 36:479–489 487

caution for their use in early infancy. Opportunities for pre- 3. Takahashi N, Lopez ML, Co Whig JE Jr, Taylor MA, Hatada T,
Riggs E, Lee G, Gomez RA, Kim HS, Smithies O (2005) Ren1c
vention and education of healthcare providers and women
homozygous null mice are hypotensive and polyuric, but heterozy-
about the use of RASi during pregnancy and in early infancy gotes are indistinguishable from wild-type. J Am Soc Nephrol 16:
should be optimized. 125–132
4. Esther CR Jr, Howard TE, Marino EM, Goddard JM, Capecchi
MR, Bernstein KE (1996) Mice lacking angiotensin-converting en-
State of the field zyme have low blood pressure, renal pathology, and reduced male
fertility. Lab Investig 7:953–965
5. Oliverio MI, Kim HS, Ito M, Le T, Audoly L, Best CF, Hiller S,
Recent studies have expanded our knowledge of the role of Kluckman K, Maeda N, Smithies O, Coffman TM (1998) Reduced
the renin-angiotensin system in kidney development and in growth, abnormal kidney structure, and type 2 (AT2) angiotensin
the pathogenesis of CAKUT. However, a better understanding receptor-mediated blood pressure regulation in mice lacking both
of the time course of developmental origins of renal anomalies AT1A and AT1B receptors for angiotensin II. Proc Natl Acad Sci U S
A 95:15496–15501
and diseases linked to renin-angiotensin system gene muta- 6. Miyazaki Y, Tsuchida S, Nishimura H, Pope JC IV, Harris RC,
tions or its drug-induced inhibition is needed. This will poten- McKanna JM, Inagami T, Hogan BL, Fogo A, Ichikawa I (1998)
tially identify new biomarkers to more precisely predict affect- Angiotensin induces the urinary peristaltic machinery during the
ed offspring and ensure early initiation of monitoring, genetic perinatal period. J Clin Invest 102:1489–1497
7. Tsuchida S, Matsusaka T, Chen X, Okubo S, Niimura F, Nishimura
counseling, and discontinuation of nephrotoxic drugs. More H, Fogo A, Utsunomiya H, Inagami T, Ichikawa I (1998) Murine
detail has to be uncovered regarding the specific signaling double nullizygotes of the angiotensin type 1A and 1B receptor
pathways affected, which could drive the development of nov- genes duplicate severe abnormal phenotypes of angiotensinogen
el treatment strategies. A greater understanding of differences nullizygotes. J Clin Invest 101:755–760
8. Oshima Y, Kinouchi K, Ichihara A, Sakoda M, Kurauchi-Mito A,
in the responses of each sex to renin-angiotensin system ab- Bokuda K, Narita T, Kurosawa H, Sun-Wada GH, Wada Y, Yamada
errations during embryonic development is needed. As the T, Takemoto M, Saleem MA, Quaggin SE, Itoh H (2011) Prorenin
postnatal kidney is composed of > 25 distinct cell types, fur- receptor is essential for normal podocyte structure and function. J
ther work is needed to explore how disruptions in the renin- Am Soc Nephrol 22:2203–2212
9. Riediger F, Quack I, Qadri F, Hartleben B, Park JK, Potthoff SA,
angiotensin system signaling during renal organogenesis im- Sohn D, Sihn G, Rousselle A, Fokuhl V, Maschke U, Purfürst B,
pact specific renal cell identity and plasticity. As induced plu- Schneider W, Rump LC, Luft FC, Dechend R, Bader M, Huber TB,
ripotent stem cells (iPSCs) represent a valuable tool to inves- Nguyen G, Muller DN (2011) Prorenin receptor is essential for
tigate mechanisms underlying human disease, iPSC-derived podocyte autophagy and survival. J Am Soc Nephrol 22:2193–
2202
kidney organoids can be used to model human CAKUT asso- 10. Gribouval O, Gonzales M, Neuhaus T (2005) Mutations in genes in
ciated with renin-angiotensin system gene mutations or RASi- the renin-angiotensin system are associated with autosomal reces-
induced toxicity [61]. In addition, gene-editing tools such as sive renal tubular dysgenesis. Nat Genet 37:964–968
clustered regularly interspaced short palindromic repeat 11. Costantini F, Kopan R (2010) Patterning a complex organ:
branching morphogenesis and nephron segmentation in kidney de-
(CRISPR)/CRISPR-associated protein 9 (Cas9)) can be uti- velopment. Dev Cell 18:698–712
lized to generate mutated control iPSC lines or correct poten- 12. Li W, Hartwig S, Rosenblum ND (2014) Developmental origins
tially pathogenic mutations in iPSCs derived from patients and functions of stromal cells in the normal and diseased mamma-
with CAKUT associated with renin-angiotensin system gene lian kidney. Dev Dyn 243:853–863
13. Batourina E, Tsai S, Lambert S, Sprenkle P, Viana R, Dutta S,
mutations.
Hensle T, Wang F, Niederreither K, McMahon AP, Carroll TJ,
Mendelsohn CL (2005) Apoptosis induced by vitamin A signaling
Compliance with ethical standards is crucial for connecting the ureters to the bladder. Nat Genet 37:
1082–1089
Conflict of interest The authors declare that they have no conflict of 14. Hoshi M, Reginensi A, Joens MS, Fitzpatrick JAJ, McNeill H, Jain
interest. S (2018) Reciprocal spatiotemporally controlled apoptosis regu-
lates Wolffian duct cloaca fusion. J Am Soc Nephrol 29:775–783
15. Iosipiv IV, Schroeder M (2003) A role for angiotensin II AT1 re-
ceptors in ureteric bud cell branching. Am J Phys 285:F199–F207
16. Prieto M, Dipp S, Meleg-Smith S, El-Dahr SS (2001) Ureteric bud
References derivatives express angiotensinogen and AT1 receptors. Physiol
Genomics 6:29–37
1. Nagata M, Tanimoto K, Fukamizu A, Kon Y, Sugiyama F, Yagami 17. Schutz S, Le Moullec J-M, Corvol P, Gasc JM (1996) Early expres-
K, Murakami K, Watanabe T (1996) Nephrogenesis and renovas- sion of all the components of the renin–angiotensin system in hu-
cular development in angiotensinogen-deficient mice. Lab Investig man development. Am J Pathol 149:2067–2079
75:745–753 18. Jones CA, Sigmund CD, McGowan RA, Kane-Haas CM, Gross
2. Niimura F, Labosky PA, Kakuchi J, Okubo S, Yoshida H, Oikawa KW (1990) Expression of murine renin genes during fetal develop-
T, Ichiki T, Naftilan AJ, Fogo A, Inagami T (1995) Gene targeting ment. Mol Endocrinol 4:375–383
in mice reveals a requirement for angiotensin in the development 19. Gomez RA, Lynch KR, Sturgill BC, Elwood JP, Chevalier RL,
and maintenance of kidney morphology and growth factor regula- Carey RM, Peach MJ (1989) Distribution of renin mRNA and its
tion. J Clin Invest 96:2947–2954 protein in the developing kidney. Am J Phys 257:F850–F858
488 Pediatr Nephrol (2021) 36:479–489

20. Jung FF, Bouyounes B, Barrio R, Tang SS, Diamant D, Ingelfinger collecting duct directly modulate the concentration of urine. J Am
JR (1993) Angiotensin converting enzyme in renal ontogeny: hy- Soc Nephrol 22:2237–2246
pothesis for multiple roles. Pediatr Nephrol 7:834–840 40. Schrankl J, Neubauer B, Fuchs M, Gerl K, Wagner C, Kurtz A
21. Norwood VF, Craig MR, Harris JM, Gomez RA (1997) Differential (2019) Apparently normal kidney development in mice with con-
expression of angiotensin II receptors during early renal morpho- ditional disruption of ANGII-AT1 receptor genes in FoxD1 positive
genesis. Am J Phys 272:R662–R668 stroma cell precursors. Am J Physiol Ren Physiol 16:F1191–F1200
22. Song R, Spera M, Garrett C, El-Dahr SS, Yosypiv IV (2010) 41. Oshima K, Miyazaki Y, Brock JW, Adams MC, Ichikawa I, Pope
Angiotensin II AT2 receptor regulates ureteric bud morphogenesis. JC (2001) Angiotensin type II receptor expression and ureteral bud-
Am J Phys 298:F807–F817 ding. J Urol 166:1848–1852
23. Kakuchi J, Ichiki T, Kiyama S, Hogan BL, Fogo A, Inagami T, 42. Lacoste M, Cai Y, Guicharnaud L, Mounier F, Dumez Y, Bouvier
Ichikawa I (1995) Developmental expression of renal angiotensin R, Dijoud F, Gonzales M, Chatten J, Delezoide AL, Daniel L,
II receptor genes in the mouse. Kidney Int 47:140–147 Joubert M, Laurent N, Aziza J, Sellami T, Amar HB, Jarnet C,
24. Song R, Preston G, Yosypiv IV (2013) Ontogeny of the (pro)renin Frances AM, Daïkha-Dahmane F, Coulomb A, Neuhaus TJ,
receptor. Pediatr Res 74:5–10 Foliguet B, Chenal P, Marcorelles P, Gasc JM, Corvol P, Gubler
25. Song R, Preston G, Kidd L, Bushnell D, Sims-Lucas S, Bates CM, MC (2006) Renal tubular dysgenesis, a not uncommon autosomal
Yosypiv IV (2016) Prorenin receptor is critical for nephron progen- recessive disorder leading to oligohydramnios: role of the renin-
itors. Dev Biol 409:382–391 angiotensin system. J Am Soc Nephrol 17:2253–2263
26. Yosypiv IV, Lopez M, Song R, Goes Martini A (2019) Stromal 43. Nishimura H, Yerkes E, Hohenfellner K, Miyazaki Y, Ma J, Hunley
prorenin receptor is critical for normal kidney development. Am J TE, Yoshida H, Ichiki T, Threadgill D, Phillips JA 3rd, Hogan BM,
Phys Regul Integr Comp Phys 316:R640–R650 Fogo A, Brock JW 3rd, Inagami T, Ichikawa I (1999) Role of the
27. Terada T, Urushihara M, Saijo T, Nakagawa R, Kagami S (2017) angiotensin type 2 receptor gene in congenital anomalies of the
(Pro)renin and (pro)renin receptor expression during kidney devel- kidney and urinary tract, CAKUT, of mice and men. Mol Cell 3:
opment in neonates. Eur J Pediatr 176:183–189 1–10
44. Song R, Preston G, Khalili A, El-Dahr SS, Yosypiv IV (2012)
28. Huang Y, Noble NA, Zhang J, Xu C, Border WA (2007) Renin-
Angiotensin II regulates growth of the developing papillas
stimulated TGF-beta1 expression is regulated by a mitogen-
ex vivo. Am J Physiol Ren Physiol 302:F1112–F1120
activated protein kinase in mesangial cells. Kidney Int 72:45–52
45. Madsen K, Marcussen N, Pedersen M, Kjaersgaard G, Facemire C,
29. Sakoda M, Ichihara A, Kaneshiro Y, Takemitsu T, Nakazato Y, Nabi
Coffman TM, Jensen BL (2010) Angiotensin II promotes develop-
AH, Nakagawa T, Suzuki F, Inagami T, Itoh H (2007) (Pro)renin
ment of the renal microcirculation through AT1 receptors. J Am Soc
receptor-mediated activation of mitogen-activated protein kinases
Nephrol 21:448–459
in human vascular smooth muscle cells. Hypertens Res 30:1139–
46. Oliverio MI, Delnomdedieu M, Best CF, Li P, Morris M, Callahan
1146
MF, Johnson GA, Smithies O, Coffman TM (2000) Abnormal wa-
30. Nguyen G, Delarue F, Burcklé C, Bouzhir L, Giller T, Sraer JD
ter metabolism in mice lacking the type 1A receptor for ANG II.
(2002) Pivotal role of the renin/prorenin receptor in angiotensin II
Am J Phys 278:F75–F82
production and cellular responses to renin. J Clin Invest 109:1417–
47. Brooks HL, Allred AJ, Beutler KT, Coffman TM, Knepper MA
1427
(2002) Targeted proteomic profiling of renal Na(+) transporter
31. Ludwig J, Kerscher S, Brandt U, Pfeiffer K, Getlawi F, Apps DK, and channel abundances in angiotensin II type 1a receptor knockout
Schägger H (1998) Identification and characterization of a novel mice. Hypertension 39:470–473
9.2-kDa membrane sector-associated protein of vacuolar proton- 48. Li XC, Shao Y, Zhuo JL (2012) AT1a receptor signaling is required
ATPase from chromaffin granules. J Biol Chem 273:10939–10947 for basal and water deprivation-induced urine concentration in
32. Mounier F, Hinglais N, Sich M, Gros F, Lacoste M, Deris Y, AT1a receptor-deficient mice. Am J Physiol Ren Physiol 303:
Alhenc-Gelas F, Gubler MC (1987) Ontogenesis of angiotensin-I F746–F756
converting enzyme in human kidney. Kidney Int 32:684–690 49. Weiner ID, New AR, Milton AE, Tisher CC (1995) Regulation of
33. Garcia-Villalba P, Denkers ND, Wittwer CT, Hoff C, Nelson RD, luminal alkalinization and acidification in the cortical collecting
Mauch TJ (2003) Real-time PCR quantification of AT1 and AT2 duct by angiotensin II. Am J Phys 269:F730–F738
angiotensin receptor mRNA expression in the developing rat kid- 50. Tojo A, Tisher CC, Madsen KM (1994) Angiotensin II regulates
ney. Nephron Exp Nephrol 94:e154–e159 H+-ATPase activity in rat cortical collecting duct. Am J Phys 267:
34. Yosipiv IV, Dipp S, El-Dahr SS (1994) Ontogeny of somatic F1045–F1051
angiotensin-converting enzyme. Hypertension 23:369–374 51. Song R, Janssen A, Li Y, El-Dahr SS, Yosypiv IV (2017) Prorenin
35. Sequeira-Lopez ML, Nagalakshmi VK, Li M, Sigmund CD, receptor controls renal branching morphogenesis via Wnt/β-catenin
Gomez RA (2015) Vascular versus tubular renin: role in kidney signaling. Am J Physiol Ren Physiol 312:F407–F417
development. Am J Phys Regul Integr Comp Phys 309:R650– 52. Yosipiv I, El-Dahr SS (1996) Activation of angiotensin-generating
R657 systems in the developing rat kidney. Hypertension 27:281–286
36. Sihn G, Burckle C, Rousselle A, Reimer T, Bader M (2013) 53. Kopan R, Chen S, Little M (2014) Nephron progenitor cells:
(Pro)renin receptor: subcellular localizations and functions. Front shifting the balance of self-renewal and differentiation. Curr Top
Biosci 5:500–508 Dev Biol 107:293–331
37. Song R, Preston G, Ichihara A, Yosypiv IV (2013) Deletion of the 54. Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME (1989)
prorenin receptor from the ureteric bud causes renal hypodysplasia. Growth in utero, blood pressure in childhood and adult life, and
PLoS One 8:e63835 mortality from cardiovascular disease. BMJ 298:564–567
38. Ramkumar N, Stuart D, Mironova E, Abraham N, Gao Y, Wang S, 55. Brenner BM, Garcia DL, Anderson S (1988) Glomeruli and blood
Lakshmipathi J, Stockand JD, Kohan DE (2018) Collecting duct pressure. Less of one, more the other? Am J Hypertens 1:335–347
principal, but not intercalated, cell prorenin receptor regulates renal 56. Song R, Kidd L, Janssen A, Yosypiv IV (2018) Conditional abla-
sodium and water excretion. Am J Physiol Ren Physiol 315:F607– tion of the prorenin receptor in nephron progenitor cells results in
F617 developmental programming of hypertension. Phys Rep 6:e13644
39. Stegbauer J, Gurley SB, Sparks MA, Woznowski M, Kohan DE, 57. Cousin C, Bracquart D, Contrepas A, Corvol P, Muller L, Nguyen
Yan M, Lehrich RW, Coffman TM (2011) AT1 receptors in the G (2009) Soluble form of the (pro)renin receptor generated by
Pediatr Nephrol (2021) 36:479–489 489

intracellular cleavage by furin is secreted in plasma. Hypertension 60. Weber-Schoendorfer C, Kayser A, Tissen-Diabaté T, Winterfeld U,
53:1077–108250 Eleftheriou G, Te Winkel B, Diav-Citrin O, Greenall A,
58. Sims-Lucas S, Schaefer C, Bushnell D, Ho J, Logar A, Prochownik Hoeltzenbein M, Schaefer C (2020) Fetotoxic risk of AT1 blockers
E, Gittes G, Bates CM (2013) Endothelial progenitors exist within exceeds that of angiotensin-converting enzyme inhibitors: an obser-
the kidney and lung mesenchyme. PLoS One 8:e65993 vational study. J Hypertens 38:133–141
59. Nadeem S, Hashmat S, Defreitas MJ, Westreich KD, Shatat IF, 61. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda
Selewski DT, Onder AM, Chiang M, Weaver DJ Jr, Steinke J, K, Yamanaka S (2007) Induction of pluripotent stem cells from
Barcia J, Hernandez J, Hidalgo G, Ingraham SE, Abitbol CL, Pan adult human fibroblasts by defined factors. Cell 131:861–872
C, Greenbaum LA (2015) Renin angiotensin system blocker
fetopathy: a midwest pediatric nephrology consortium report. J Publisher’s note Springer Nature remains neutral with regard to jurisdic-
Pediatr 167:881–885 tional claims in published maps and institutional affiliations.

You might also like