SlideShare a Scribd company logo
1 of 8
Download to read offline
1499
Chapter 204
Renin–Angiotensin System
Minolfa C. Prieto, Alexis A Gonzalez, Ryousuke Satou, Hiroyuki Kobori
and L. Gabriel Navar
ABSTRACT
Renal angiotensin (Ang) II is regulated independently of the sys-
temic renin–angiotensin system. Intrarenal Ang II concentrations
are higher in renal interstitium and proximal tubular compartments
than in the circulation. The transcript for angiotensinogen (AGT) is
in proximal tubule cells and for renin in juxtaglomerular and col-
lecting duct cells. Secretion of their proteins increases intratubular
formation of angiotensin peptides. Ang II is internalized via AT1R
into renal endosomes. When inappropriately stimulated, Ang II
augments synthesis and secretion of AGT in proximal tubules and
renin in collecting ducts, which increase further intrarenal Ang II
and cause hypertension. Ang II has pleiotropic actions on cortical
and medullary blood flow, vascular tone of afferent and efferent
arterioles, mesangial cell function, and sensitivity of the tubulo-
glomerular feedback mechanism via the activation of AT1R. AT2R
and Ang III partially oppose these actions. Ang II also regulates
sodium transport in proximal and distal nephrons via the activation
of luminal and basolateral AT1R.
INTRODUCTION
The contribution of the intrarenal renin–angiotensin system
(RAS) to the regulation of renal function is unique because
renal functional, and transport alterations caused by angio-
tensin II (Ang II) influence overall sodium balance and
cardiovascular status and thus contribute to blood pressure
regulation. When inappropriately stimulated, an enhanced
intrarenal RAS markedly influences renal function by act-
ing on vascular and tubular components causing the devel-
opment and maintenance of hypertension and associated
cardiovascular dysfunction.12,18–20,33
This chapter covers the intrarenal RAS, the formation
of several angiotensin peptides by various enzymes, and the
characterization of angiotensin receptors including AT1a,
AT1b, and AT2 for Ang II and Ang III, and the Mas receptor
for Ang 1–7. In addition, we describe some newly discov-
ered aspects of the intrarenal RAS including the regulation
of proximal tubule angiotensinogen (AGT), collecting duct
renin, and the renin/(pro)renin receptor.
DISTRIBUTION OF ANGIOTENSINOGEN,
RENIN, (PRO)RENIN RECEPTORS,
ANGIOTENSIN CONVERTING ENZYMES,
AND ANG TYPE 1 RECEPTORS IN THE
KIDNEY
All components of the RAS are present in the kidney and are
compartmentalized to the tubular and interstitial networks.
Because kidney Ang II tissue concentrations are greater
than can be explained by the concentrations delivered in
the arterial blood flow, it is generally acknowledged that
most of the Ang II in the intrarenal components is generated
locally from AGT delivered to the kidney as well as that
locally produced by proximal tubule cells. The presence of
abundant sources of renin,AGT, and angiotensin-converting
enzyme (ACE) in the kidney provides an environment con-
ducive to high levels of Ang II synthesis.19,21
Angiotensinogen
AGT, the only known substrate for renin, is a 60-kD glyco-
protein synthesized and released from the liver, and cleaved
in the circulation to form the biologically inactive, Ang I
(Fig. 1). In addition, renal AGT mRNA and protein are
localized to proximal tubule cells indicating that much of
the intratubular Ang II is derived from locally formed and
secreted AGT. AGT is regulated by an amplification mecha-
nism such that AGT mRNA and protein are stimulated by
Ang II, which maintains or increases the production of Ang
II in Ang II-dependent hypertension. Producing its metabo-
lites intracellularly and secreting them into the tubule lumen,
AGT is taken up by megalin-dependent endocytosis and is
then secreted into the tubular lumen.12,19,24 More recent
studies have evaluated the molecular mechanisms respon-
sible for the regulation of AGT expression and synthesis in
proximal tubular cells.21 Activation of AT1 receptors stimu-
lates intrarenal NF-κB, which is a key factor in the aug-
mentation of AGT expression in proximal tubule cells.1,20
The activation of NF-κB by Ang II leads to the stimulation
Handbook of Biologically Active Peptides. http://dx.doi.org/10.1016/B978-0-12-385095-9.00204-9
Copyright © 2013 Elsevier Inc. All rights reserved.
Chapter | 204  Renin–Angiotensin System
1500
of intrarenal proinflammatory cytokine expressions such as
interleukin-6, contributing to the increase in AGT expres-
sion via activation of a JAK-STAT pathway.29 Therefore,
Ang II increases intrarenal AGT levels via both direct and
indirect mechanisms mediated by activated NF-κB and the
JAK-STAT pathway. By contrast, tumor necrosis factor α,
which is also an Ang II-induced proinflammatory factor in
the kidney, suppresses AGT expression by increasing p50/
p50 complex in proximal tubular cells.30 This inhibitory
action on NF-κB provides a counterregulatory influence
limiting the magnitude of Ang II-induced AGT augmen-
tation in renal proximal tubular cells, which may explain
why higher doses of Ang II infusion in mice fail to augment
intrarenal AGT levels.20
Proximal tubuleAGT concentrations in anesthetized rats
are in the range of 300 nM, which greatly exceeds the free
Ang I and Ang II tubular fluid concentrations. Because of
its molecular size, it is unlikely that much plasma AGT fil-
ters across the glomerular membrane, further supporting the
concept that proximal tubule cells secrete AGT directly into
the tubule.12 In response to chronic Ang II infusions there is
an augmentation of AGT mRNA leading to increased syn-
thesis and secretion with some of the secreted AGT spilling
over to the distal nephron segments as reflected by increased
urinary AGT excretion.13 Under conditions of elevated salt
intake combined with chronic Ang II infusion, the urinary
AGT excretion response is elevated to much greater lev-
els than those seen in Ang II-infused rats fed a normal salt
diet.14 By contrast, normal rats fed a high salt diet exhibit
urinary AGT excretion rates similar to those observed in
rats fed a normal salt diet. Thus, during chronic Ang II infu-
sion, salt-sensitive hypertension develops and high salt par-
adoxically stimulates intrarenal RAS activity, as reflected
by exacerbation of the urinary AGT despite inhibition of
plasma renin activity.14 Formation of Ang I and II in the
tubular lumen subsequent to AGT secretion is probable
because some renin is filtered and/or secreted from juxta-
glomerular apparatus (JGA) cells.12 Once Ang I is formed,
conversion readily occurs because there is abundant ACE
associated with the proximal tubule brush border.32 Intact
AGT in urine reflects its presence throughout the nephron
and, to the extent that renin and ACE are available along
the nephron, substrate availability supports continued Ang
I generation and Ang II conversion in distal segments.19,25
Renin and (Pro)renin Receptor
Renin, synthesized by the JGA cells, is the primary source
of both circulating and intrarenal renin levels. Juxtaglo-
merular apparatus cells secrete renin into the renal inter-
stitium, thus providing a stimulus for the local generation
of Ang I, and this secreted active form contains 339–343
amino acid residues after proteolytic removal of the 43
amino acid residue at the N-terminus of prorenin. Circu-
lating active renin and prorenin are derived mainly from
the kidney; and circulating prorenin is taken up by some
tissues where it may contribute to the local synthesis of
angiotensin peptides. The only well-established role of
FIGURE 1  Intrarenal renin–angiotensin system cascade. Known and postulated enzymatic pathways responsible for formation and metabolism of
angiotensin peptides.
1501
SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides
renin is to cleave Ang I from AGT. However, the recently
cloned (pro)renin receptor ((P)RR) binds renin or pro-
renin, thus enhancing renin activity and fully activating the
biologically inactive prorenin peptide, which leads to an
increase in the catalytic efficiency of Ang I formation from
AGT. Activation of the (P)RR by renin or prorenin also
directly elicits intracellular signals that stimulate ERK1/
ERK2 and downstream pathways.22
There is growing awareness of the possible roles of renin
in other intrarenal structures where renin has been local-
ized. Renin producing cells have a unique origin that allows
their abundance along the afferent arteriole to be regulated
by various stimuli. ACE inhibition induces a recruitment of
cells in afferent arterioles beyond the JGA even though they
were not expressing the renin gene in the basal state.31 Posi-
tive renin immunoreactivity has also been observed in the
cells of glomeruli and in proximal and distal tubular seg-
ments. More recently, increased attention has been focused
on the presence of renin mRNA and protein expression in
distal nephron segments, specifically in the principal cells
of connecting tubules and collecting ducts, suggesting local
formation and secretion into the distal tubular fluid. Renin
synthesis in principal cells of collecting ducts is further
increased by chronic infusions of Ang II.25,26 Rats chroni-
cally infused with Ang II for 13 days show a suppression of
plasma renin activity but increases in urinary prorenin con-
tent and renin excretion.16 In addition, the urinary renin and
prorenin protein levels, examined by Western blot, are aug-
mented about 10-fold in Ang II-infused rats. Concomitant
AT1R blockade with candesartan prevents increases in both
urinary and tissue renin and in prorenin levels. Although
there is marked suppression of plasma renin activity,
increased renin synthesis by CD cells and secretion into the
luminal fluid lead to increased urinary levels of renin and
prorenin.16
The presence of renin in distal nephron segments pro-
vides the requisite pathway for Ang I generation from prox-
imally delivered AGT.25,26 Renin synthesis is augmented
in the principal cells of connecting tubules and cortical
and medullary collecting ducts of chronic Ang II-infused
rats and mice, Cyp1a1Ren2 transgenic rats, and in both
kidneys of two-kidney one clip Goldblatt hypertensive
rats.9,25,26 The AT1R via a PKC pathway directly mediates
the stimulation of renin synthesis in rat inner medullary col-
lecting duct because inhibition of PKC with calphostin C
prevents the Ang II-mediated increases in renin transcript
and prorenin protein levels in rat inner medullary collecting
duct (IMCD) cells while activation of PKC with phorbol
12-myristate 13-acetate increases renin expression to the
same extent as Ang II.7 Further, renin activity and renin and
prorenin protein levels are increased in the urine of these
rats6 indicating that both renin and prorenin are secreted by
the collecting duct cells into the lumen of distal nephron
segments and are also augmented in chronic Ang II-infused
hypertensive rats. The discovery of the (P)RR provides new
insights regarding its possible role in a setting of activated
intrarenal RAS. The (P)RR is expressed in the kidneys6
particularly in mesangial cells, podocytes, and intercalated
type A cells of distal nephron segments.2,6,11 Chronic infu-
sion of Ang II for 14 days in Sprague–Dawley rats increases
renal (P)RR transcript levels and augments the soluble form
of the (P)RR, s(P)RR.6 The augmentation of s(P)RR in the
urine of Ang II hypertensive rats suggests that s(P)RR can
bind renin and prorenin secreted by the principal collecting
duct cells and can catalyze additional formation of Ang I.
The existence of an inducible and functional form of s(P)
RR in the renal medulla and in the urine of chronic Ang
II-infused rats reflects increased s(P)RR in collecting duct
tubular fluid and further supports the potential role of this
soluble form to enhance intratubular renin activity. Thus,
the interaction between the (P)RR and renin in the lumen of
the collecting ducts may contribute substantially to facili-
tate intratubular formation of Ang I in hypertension.
Angiotensin-Converting Enzymes
As in other organs, ACE is located on endothelial cells
throughout the vasculature. ACE is also on membranes of
both proximal and distal nephron segments with the greatest
abundance found on the brush border of proximal tubules,
particularly in the S3 segment.21,32 Marked upregulation of
ACE in the brush border occurs in kidneys from Goldblatt
hypertensive rats and Ang II infused hypertensive rats.32
Humans predominantly express ACE in the brush border
of proximal tubular segments with very little ACE expres-
sion on vascular endothelial cells or in the vasculature of
the glomerular tuft. The lower renal vascular endothelial
expression in humans helps explain the much lowerAng I to
Ang II conversion rates that have been reported for human
kidneys as compared with that of other species. In rats, ACE
activity is present in tubular fluid throughout the nephron
except in the distal tubule, with activity higher at the initial
portion of the proximal tubule, decreasing toward the distal
nephron, and increasing again in the collecting ducts and
urine. Therefore, intratubular Ang II formation may occur
not only in the proximal tubule but also beyond the distal
convoluted tubule. Renal tissue ACE activity is critical to
maintain the steady-state Ang II levels in the kidney. The
presence of ACE only in the kidney is sufficient to increase
kidneyAng II levels and promote the development of hyper-
tension in response to chronicAng I infusions.8 Tissue-ACE
knockout mice exhibit 80% lower intrarenal Ang II levels
compared with wild-type mice.3
ACE2, which acts on Ang II to form Ang 1–7 or on Ang
I to form Ang 1–9 (Fig. 1), is a membrane-associated and
secreted enzyme expressed predominantly on endothelium,
Chapter | 204  Renin–Angiotensin System
1502
but highly restricted in humans to heart, kidneys, and tes-
tes.21 Ang 1–7 may serve as an endogenous antagonist of
the Ang II induced actions mediated via AT1 receptors and
changes in ACE2 activity could affect the balance of Ang
peptides found in the kidney. This probably explains the ele-
vatedAng II levels in theACE2 knockout mice.28 Collectrin,
a novel homolog of ACE2 has been identified in mouse, rat,
and human. Both ACE2 and collectrin have tissue-restricted
expression in the kidney. Collectrin is localized on the lumi-
nal surface and in the cytoplasm of collecting duct cells,
and its mRNA is expressed in the renal collecting ducts
cells while ACE2 is present throughout the endothelium,
in proximal tubular epithelial cells and in medullary tis-
sue.21 In Ang-II-dependent hypertension, intrarenal ACE is
increased, but ACE2 levels are decreased leading to another
mechanism maintaining increased Ang II levels.26
Intrarenal Angiotensin II Receptors
Ang II receptors are widely distributed in various regions
and cell types of the kidney. Two major categories of Ang II
receptors, AT1 and AT2, have been described, pharmacologi-
cally characterized and cloned.18,21 As depicted in Figure 2,
most of the Ang-II-mediated hypertensinogenic actions are
generally attributed to the AT1 receptor.5,18 In rodents, there
are two AT1 receptor subtypes, with the AT1a being the pre-
dominant subtype in all nephron segments, while the AT1b
is more abundant in the glomerulus.18,21 In mature kidneys,
AT1a receptors have been localized to the renal microvascu-
lature in cortex and medulla, smooth muscle cells of afferent
and efferent arterioles, thick ascending limb of the loop of
Henle, proximal tubular apical and basolateral membranes,
mesangial cells, distal tubules, collecting ducts, and macula
densa cells. The afferent arteriole has both AT1a and AT1b
receptors, whereas the efferent arteriole primarily expresses
AT1a receptors.10 The essential role of the AT1a receptor in
mice is apparent from studies showing that 2-kidney, 1-clip
(2K1C) Goldblatt hypertension does not develop in AT1a
knockout mice.4
Vascular and tubular receptors respond differently dur-
ing high Ang II states.18 High Ang II levels associated with
a low salt diet decrease glomerular AT1 receptor expression
but increase proximal tubular AT1 receptor levels. Glomeru-
lar AT1 receptors and AT1a receptor protein are reduced in
both clipped and contralateral kidneys of 2K1C Goldblatt
and of 2 kidney-1-wrap hypertensive models and in kidneys
of Ang II infused rats.18 In the TGR(mRen2) harboring
the mouse renin2 gene, AT1 receptor binding is increased
in vascular smooth muscle of afferent and efferent arteri-
oles, JGA, glomerular mesangial cells, proximal tubular
cells, and renomedullary interstitial cells suggesting that
the upregulation of AT1 receptors contributes to the patho-
genesis of hypertension in these rats. In Ang II infused rats
studied with in vitro autoradiography, there are differential
responses with significant decreases in glomeruli and inner
stripe but not in proximal tubules.18
The AT2 receptor is highly expressed in human and
rodent kidney mesenchyme during fetal life and decreases
dramatically after birth.AT2 receptors have been localized to
the glomerular epithelial cells, proximal tubules, collecting
ducts, and parts of the renal vasculature of the adult rat. AT2
receptor activation is believed to counteract AT1 receptor
FIGURE 2  Major Ang II receptor subtypes and their renal actions.
1503
SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides
effects by stimulating the formation of bradykinin and nitric
oxide leading to increases in interstitial fluid concentration
of cyclic guanosine monophosphate (cGMP). AT2 receptor
activation influences proximal tubule sodium reabsorption
either by a cell-membrane-receptor-mediated mechanism or
via an interstitial nitric-oxide-cGMP pathway.17,18
INTRARENAL LEVELS OF ANGIOTENSIN II
Interstitial Angiotensin II
Intrarenal Ang II is not distributed in a homogeneous
fashion but is compartmentalized in both a regional and
segmental manner.19 Medullary Ang II levels are higher
than cortical levels in normal rats and increase further in
Ang II infused hypertensive rats.18 Within the cortex, there
is distribution of Ang II in the interstitial fluid, tubular
fluid, and intracellular compartments. The interstitial and
intratubular compartments contribute to the disproportion-
ately high intrarenal Ang II levels. Ang II concentrations
in interstitial fluid are much higher than the plasma con-
centrations with values in the range of 3–5 pmoles/mL.19
Renal interstitial fluid Ang II levels increase further in
hypertensive rats infused with Ang II for 2 weeks. The
high renal interstitial values indicate local regulation of
Ang II formation in the renal interstitial compartment and
an enhancement of interstitial Ang II production in Ang II
dependent hypertension.12
Tubular Ang II
As shown in Figure 3, proximal tubule fluid concentrations
of Ang I and Ang II are also much greater than the plasma
concentrations and have been found to be in the range of
5–10 pmoles/ml.19 In perfused tubules, where the contri-
bution resulting from glomerular filtration is prevented,
Ang II concentrations are similar to those in nonperfused
tubules indicating that the proximal tubule secretes Ang II
or a precursor into the proximal tubule fluid. In addition to
AGT, proximal tubule cells have renin that can act on AGT
to generate Ang I. Evidence that renin is augmented in the
urine of chronic Ang II-infused rats also indicates secretion
of renin by the collecting duct cell into the tubular lumen16
and provides a pathway for Ang I generation from proxi-
mally delivered AGT, leading to concentrations of Ang II in
distal tubular fluid in the picomolar range.34
Intracellular Angiotensin II
Ang II is internalized via AT1–receptor-mediated endo-
cytosis and endosomal accumulation of Ang II in inter-
microvillar clefts and endosomes is increased further in
Ang II infused hypertensive rats.35 AT1 receptor block-
ade prevents the endosomal accumulation even though
plasma Ang II increases. The presence of Ang II in renal
endosomes indicates that some of the internalized Ang II
remains intact and contributes to the total Ang II content
measured in tissue homogenates.35 Endocytosis of the Ang
II-AT1 receptor complex is required for the full expression
of functional responses coupled to the activation of signal
transduction pathways, but the full extent of the functions
exerted by intracellular Ang II has not been determined.15
In Ang-II-dependent hypertension, a higher fraction of
the total kidney Ang II is internalized into intracellular
endosomes consisting of both light endosomes and inter-
microvillar clefts via an AT1-receptor-mediated process.
AT1 receptor blockade prevents the augmentation of intra-
cellular Ang II.35
FIGURE 3  Intratubular processing of the renin–angiotensin system. All components of the renin–angiotensin system are present in the kidney provid-
ing multiple pathways for enhanced intrarenal Ang II formation. PT—proximal tubule; DT—distal tubule; CD—collecting duct; AGT—angiotensinogen;
JGA—juxtaglomerular apparatus; AA—afferent arteriole; EA—efferent arteriole; ACE—angiotensin converting enzyme; PRR—(pro)renin receptor (modi-
fied from Ref. 19).
Chapter | 204  Renin–Angiotensin System
1504
Our understanding of the functions of internalized Ang
II remains incomplete. Ang II may be recycled and secreted
to exert further actions by binding to Ang II receptors on
the cell membranes. Ang II may also act on cytosolic recep-
tors to activate intracellular signaling mechanisms.15 Ang II
may also migrate to the nucleus to exert genomic effects.21
Nuclear binding sites of the AT1 subtype in renal cells have
been reported. Because Ang II exerts a positive stimulation
on AGT mRNA and protein production, intracellular Ang II
may have genomic actions to regulate AGT or renin mRNA
expression in proximal tubule cells either directly or via
NF-κB.1,21,30
BIOLOGICAL ACTIONS
OF INTRARENAL ANG II
In view of the near ubiquitous distribution of Ang II recep-
tors in the various structures of the kidney, it is not surpris-
ing that Ang II exerts pleiotropic effects at multiple levels
within the kidney. As shown in Figure 2, the actions of Ang
II span both cortical and medullary, vascular and tubular
and can be stimulatory or inhibitory.18 At the level of the
vasculature, Ang II exerts important direct and indirect
effects to regulate vascular tone of the afferent and efferent
arterioles and of the mesangial cells.17 Ang II also regulates
afferent arteriolar tone through its influence on the tubulo-
glomerular feedback mechanism with elevated Ang II levels
increasing the sensitivity of the feedback mechanism, thus
regulating the amount of filtrate that escapes the proximal
nephron and enters the distal nephron segments. In the post-
glomerular circulation, Ang II regulates both the cortical
and medullary capillary blood flow and helps to regulate
the relative blood flowing to the medullary tissues through
its control of the tone of the pericytes.17
Interestingly, although Ang II constricts both afferent
and efferent arterioles, the mechanisms are different. The
afferent arteriolar constrictor responses to Ang II involve
activation of L-type Ca++ channels to provide sustained
Ca++ entry and vasoconstriction.17 By contrast, efferent
arterioles of normal rats do not vasodilate or vasodilate
much less in response to L-type Ca++ channel blockers
and the vasoconstrictor effects of Ang II are not blocked
by L-type Ca++ channel blockers. Rather, T-type channels
have greater functional significance in regulating the effer-
ent arteriolar constrictor responses to Ang II.17 T-type Ca++
channel blockers markedly attenuate the efferent vasocon-
strictor responses to Ang II. Storage operated Ca++ chan-
nels in efferent and afferent arterioles are also activated by
Ang II. Thus, although both afferent and efferent arterioles
respond to Ang II, the mechanisms for Ca++ entry and the
activation pathways are different. These differences may be
attributable to the differential abundance of AT1A and AT1B
receptors on the afferent and efferent arterioles.10
Virtually, all aspects of the filtration and tubular reab-
sorptive processes are influenced by Ang II. Ang II directly
regulates the glomerular filtration coefficient with high Ang
II concentrations reducing and low levels or blockade of
AT1 receptors increasing the filtration coefficient.17,18 At
the level of the proximal tubule, Ang II binds to receptors at
both the apical and basolateral membranes, stimulating the
activity of the sodium/hydrogen exchanger and the sodium
bicarbonate co-transporter, thereby exerting an important
influence on net proximal reabsorption rate.18 Transport
mechanisms in the proximal tubule, loop of Henle, and dis-
tal nephron segments are also influenced by Ang II.18,27,34
Under conditions in which the activity of the RAS is
stimulated and greater amounts of AGT are secreted from
the proximal tubule, the locus of influence shifts to include
the distal nephron segments where the interaction between
the (P)RR and renin/prorenin may contribute substantially
to increase intrarenal and intratubular formation of Ang II
(Fig. 4).7,20 The in vivo demonstration that increased urinary
Ang II concentrations in mice infused chronically with Ang
II enhance distal sodium reabsorption34 emphasizes further
the importance that renin and the (P)RR may have in the
distal nephron segments contributing to the increases in
intratubular Ang II formation. Ang II stimulates the sodium/
hydrogen exchanger and the sodium chloride cotransporter
in distal tubules, and the amiloride sensitive sodium channel
in principal cells of connecting tubules and collecting duct
segments.20,23 This enhanced stimulatory effect allows the
kidney to have maximum capability to conserve sodium by
markedly decreasing fractional sodium reabsorption.34 At
the whole kidney level, this is manifested as a marked sup-
pression of the pressure natriuresis relationship. Ang II also
regulates acid base balance through its effects on the Na+/
H+ exchanger as well as by influencing the activity of the
H+ ATPase in intercalated cells.2 Collectively, the actions of
Ang II allow the kidney to maximize sodium conservation
without compromising its ability to regulate its many other
functions.
CONCLUDING COMMENTS
AND PERSPECTIVE
Recent findings have stimulated interest in the molecular
mechanisms regulating the various components of the intra-
renal RAS and particularly the interstitial, intracellular, and
intratubular concentrations of Ang II and related peptides.
Intratubular and interstitial Ang II levels are regulated inde-
pendently of the circulatingAng II and exert powerful actions
via the stimulation of AT1 receptors on the vascular, glomer-
ular, and tubular structures to provide a synchronous cascade
of effects contributing to the ability of the kidney to retain
>99% of the filtered sodium. From a functional perspective,
the effects of Ang II on proximal nephron reabsorption as
1505
SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides
well as on distal nephron transport function, coupled with
the associated actions of elevated aldosterone levels, mark-
edly increase the sodium retaining capability of the kidney.
When activated in a physiologically appropriate setting under
conditions of volume contraction or salt deficient states,
these actions can be life saving. When inappropriately main-
tained or augmented, however, these effects contribute to the
development and maintenance of hypertension. Further, the
sustained increases in intrarenal Ang II in a setting of hyper-
tension can lead to progressive renal injury, cell proliferation,
and fibrosis associated with activation of several major cyto-
kines and growth factors.12,33
ACKNOWLEDGMENTS
The authors thank Debbie Olavarrieta for the preparation of the man-
uscript and figures. The authors’ research related to this review has
been supported by research grants from NHLBI, NIDDK, NCRR, and
AHA.
REFERENCES
	 1.	
Acres OW, Satou R, Navar LG, Kobori H. Contribution of a Nuclear
Factor-{kappa}B binding site to human angiotensinogen Promoter
activity in renal proximal tubular cells. Hypertension 2011;57:
608–13.
	 2.	
Advani A, Kelly DJ, Cox AJ, White KE, Advani SL, Thai K, et al. The
(Pro)renin receptor: site-specific and functional linkage to the vacuolar
H+-ATPase in the kidney. Hypertension 2009;54:261–9.
	 3.	
Bernstein KE, Xiao HD, Adams JW, Frenzel K, Li P, Shen XZ, et al.
Establishing the role of angiotensin-converting enzyme in renal func-
tion and blood pressure control through the analysis of genetically
modified mice. J Am Soc Nephrol 2005;16:583–91.
	 4.	
Cervenka L, Vaneckova I, Huskova Z, Vanourkova Z, Erbanova M,
Thumova M, et  al. Pivotal role of angiotensin II receptor subtype
1A in the development of two-kidney, one-clip hypertension: study
in angiotensin II receptor subtype 1A knockout mice. J Hypertens
2008;26:1379–89.
	 5.	
Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R,
Kumar AP, et  al. Angiotensin II causes hypertension and cardiac
hypertrophy through its receptors in the kidney. Proc Natl Acad Sci
U S A 2006;103:17985–90.
	 6.	
Gonzalez AA, Lara LS, Luffman C, Seth DM, Prieto MC. Soluble
form of the (pro)renin receptor is augmented in the collecting duct and
urine of chronic angiotensin II-dependent hypertensive rats. Hyperten-
sion 2011;57:859–64.
	 7.	
Gonzalez AA, Liu L, Lara LS, Seth DM, Navar LG, Prieto MC.
Angiotensin II stimulates renin in inner medullary collecting duct
cells via protein Kinase C and Independent of epithelial sodium
channel and Mineralocorticoid receptor activity. Hypertension
2011;57:594–9.
	 8.	
Gonzalez-Villalobos RA, Billet S, Kim C, Satou R, Fuchs S,
Bernstein KE, et al. Intrarenal angiotensin-converting enzyme induces
hypertension in response to Angiotensin I infusion. J Am Soc Nephrol
2011;22:449–59.
	 9.	
Gonzalez-Villalobos RA, Satou R, Ohashi N, Semprun-Prieto LC,
Katsurada A, Kim C, et  al. Intrarenal mouse renin-angiotensin
system during ANG II-induced hypertension and ACE inhibition.
Am J Physiol Ren Physiol 2010;298:F150–7.
	
10.	
Harrison-Bernard LM, Cook AK, Oliverio MI, Coffman TM. Renal
segmental microvascular responses to ANG II in AT1A receptor null
mice. Am J Physiol Ren Physiol 2003;284:F538–45.
	
11.	
IchiharaA, Sakoda M, Kurauchi-MitoA, KaneshiroY, Itoh H. Involve-
ment of (pro)renin receptor in the glomerular filtration barrier. J Mol
Med 2008;86:629–35.
	
12.	
Kobori H, Nangaku M, Navar LG, Nishiyama A. The intrarenal renin–
angiotensin system: from physiology to the pathobiology of hyperten-
sion and kidney disease. Pharmacol Rev 2007;59:251–87.
FIGURE 4  Renin and (pro)renin receptor interaction in the collecting duct. Prorenin and renin stimulation in the principal cells of the collecting ducts
may increase Ang I formation in the tubular lumen and increase Ang II generation. The presence of (pro)renin receptor at the surface of the intercalated
cells along with its soluble form in the tubular lumen increases the catalytic efficiency for renin to form Ang I. In addition, the pro(renin) receptor bound
to the membrane of the intercalated cells may anchor prorenin secreted by the principal cells to reduce the washout of prorenin or renin into the urine.
ANG—angiotensin; AGT—angiotensinogen; AT1R—angiotensin II type 1 receptor; PRR—(pro)renin receptor; sPRR—soluble form of the (pro)renin
receptor; ACE—angiotensin converting enzyme. See color plate 54.
Chapter | 204  Renin–Angiotensin System
1506
	
13.	
Kobori H, Prieto-Carrasquero MC, OzawaY, Navar LG. AT1 receptor
mediated augmentation of intrarenal angiotensinogen in angiotensin
II-dependent hypertension. Hypertension 2004;43:1126–32.
	
14.	
Lara LS, McCormack M, Semprum-Prieto LC, Shenouda S,
Majid DS, Kobori H, et al. AT1 receptor-mediated augmentation
of angiotensinogen, oxidative stress, and inflammation in ANG
II-salt hypertension. Am J Physiol Ren Physiol 2012;302:F85–94.
	
15.	
Li XC, Hopfer U, Zhuo JL. AT1 receptor-mediated uptake of angio-
tensin II and NHE-3 expression in proximal tubule cells through a
microtubule-dependent endocytic pathway. Am J Physiol Ren Physiol
2009;297:F1342–52.
	
16.	
Liu L, Gonzalez AA, McCormack M, Seth DM, Kobori H,
Navar LG, et  al. Increased renin excretion associated with aug-
mented urinary angiotensin (Ang) II levels in chronic angiotensin
II-infused hypertensive rats. Am J Physiol Ren Physiol 2011;301:
F1195–201.
	
17.	
Navar LG,Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD.
The renal Microcirculation. In: Tuma RF, Duran WN, Ley K, editors.
Handbook of physiology: microcirculation. Academic Press; 2008. p.
550–683.
	
18.	
Navar LG, Harrison-Bernard LM, Imig JD, Mitchell KD. Renal actions
of angiotensin II and AT1 receptor blockers. In: Epstein M, Brunner
HR, editors. Angiotensin II receptor antagonists. Philadelphia: Hanley
 Belfus, Inc.; 2000. p. 189–214.
	
19.	
Navar LG, Kobori H, Prieto MC, Gonzalez-Villalobos RA. Intra-
tubular renin-angiotensin system in hypertension. Hypertension
2011;57:355–62.
	
20.	
Navar LG, Prieto MC, Satou R, Kobori H. Intrarenal angiotensin II
and its contribution to the genesis of chronic hypertension. Curr Opin
Pharmacol 2011;11:180–6.
	
21.	
Navar LG, Prieto-Carrasquero MC, Kobori H. Molecular aspects of
the renal renin–angiotensin system. In: Re R, DiPette DJ, Schiffrin EL,
Sowers JR, editors. Molecular mechanisms in hypertension. Taylor 
Francis Group; 2006. p. 3–14.
	
22.	
Nguyen G, Muller DN. The biology of the (pro)renin receptor. J Am
Soc Nephrol 2010;21:18–23.
	
23.	
Peti-Peterdi J, Warnock DG, Bell PD. Angiotensin II directly stimu-
lates ENaC activity in the cortical collecting duct via AT(1) receptors.
J Am Soc Nephrol 2002;13:1131–5.
	
24.	
Pohl M, Kaminski H, Castrop H, Bader M, Himmerkus N, Bleich M,
et al. Intrarenal renin angiotensin system Revisited: role of megalin-
dependent endocytosis along the proximal nephron. J Biol Chem
2010;285:41935–46.
	
25.	
Prieto-Carrasquero MC, Botros FT, Kobori H, Navar LG. Collecting
duct renin: a major player in angiotensin II-dependent hypertension.
J Am Soc Hypertens 2009;3:96–104.
	
26.	
Prieto-Carrasquero MC, Botros FT, Pagan J, Kobori H, Seth DM,
Casarini DE, et al. Collecting duct renin is upregulated in both kid-
neys of 2-kidney, 1-clip goldblatt hypertensive rats. Hypertension
2008;51:1590–6.
	
27.	
Riquier-BrisonAD, Leong PK, Pihakaski-Maunsbach K, Mcdonough
AA.Angiotensin II stimulates trafficking of NHE3, NaPi2, and associ-
ated proteins into the proximal tubule microvilli. Am J Physiol Ren
Physiol 2010;298:F177–86.
	
28.	
Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP,
de B I, et al. Angiotensin-(1-7) is an endogenous ligand for the G pro-
tein-coupled receptor Mas. Proc NatlAcad Sci U SA 2003;100:8258–63.
	
29.	
Satou R, Gonzalez-Villalobos RA, Miyata K, Ohashi N, Urushihara M,
Acres OW, et al. IL-6 augments angiotensinogen in primary cultured
renal proximal tubular cells. Mol Cell Endocrinol 2009;311:24–31.
	
30.	
Satou R, Miyata K, KatsuradaA, Navar LG, Kobori H. Tumor necrosis
factor-{alpha} suppresses angiotensinogen expression through forma-
tion of a p50/p50 homodimer in human renal proximal tubular cells.
Am J Physiol Cell Physiol 2010;299:C750–9.
	
31.	
Sequeira-Lopez ML, Weatherford ET, Borges GR, Monteagudo MC,
Pentz ES, Harfe BD, et al. The microRNA-processing enzyme dicer
maintains juxtaglomerular cells. J Am Soc Nephrol 2010;21:460–7.
	
32.	
Vio CP, Jeanneret VA. Local induction of angiotensin-converting
enzyme in the kidney as a mechanism of progressive renal diseases.
Kidney Int Suppl 2003;86:S57–63.
	
33.	
Wolf G. The renin–angiotensin system and progression of renal dis-
eases. Hamburg: Karger; 2002.
	
34.	
Zhao D, Seth DM, Navar LG. Enhanced distal nephron sodium
reabsorption in chronic angiotensin II-Infused mice. Hypertension
2009;54:120–6.
	
35.	
Zhuo JL, Imig JD, Hammond TG, Orengo S, Benes E, Navar LG.
Ang II accumulation in rat renal endosomes during Ang II-induced
hypertension: role of AT(1) receptor. Hypertension 2002;39:116–21.

More Related Content

Similar to 6. lectura sistema renina angiotensina

drugsaffectingrenin-angiotensinsystem-161117183104.pdf
drugsaffectingrenin-angiotensinsystem-161117183104.pdfdrugsaffectingrenin-angiotensinsystem-161117183104.pdf
drugsaffectingrenin-angiotensinsystem-161117183104.pdf
KalyanAcharya10
 
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
Jackfrimpong
 
Chen et al PNAS-2010
Chen et al PNAS-2010Chen et al PNAS-2010
Chen et al PNAS-2010
Zhen Chen
 
MetabolicAcidosisinA.pdf
MetabolicAcidosisinA.pdfMetabolicAcidosisinA.pdf
MetabolicAcidosisinA.pdf
Hosneara18
 

Similar to 6. lectura sistema renina angiotensina (20)

d1. Angiotensin.pdf
d1. Angiotensin.pdfd1. Angiotensin.pdf
d1. Angiotensin.pdf
 
Function of the renin
Function of the reninFunction of the renin
Function of the renin
 
Raas
RaasRaas
Raas
 
Renin angiotensin aldosterone- Updates
Renin angiotensin aldosterone-   UpdatesRenin angiotensin aldosterone-   Updates
Renin angiotensin aldosterone- Updates
 
Drugs affecting renin-angiotensin system
Drugs affecting renin-angiotensin systemDrugs affecting renin-angiotensin system
Drugs affecting renin-angiotensin system
 
drugsaffectingrenin-angiotensinsystem-161117183104.pdf
drugsaffectingrenin-angiotensinsystem-161117183104.pdfdrugsaffectingrenin-angiotensinsystem-161117183104.pdf
drugsaffectingrenin-angiotensinsystem-161117183104.pdf
 
Ras system
Ras systemRas system
Ras system
 
UPDATES OF RENIN ANGIOTENSIN SYTEM INTERVENTION
UPDATES OF RENIN ANGIOTENSIN SYTEM INTERVENTIONUPDATES OF RENIN ANGIOTENSIN SYTEM INTERVENTION
UPDATES OF RENIN ANGIOTENSIN SYTEM INTERVENTION
 
Pharmacology of Bradykinin , pentagastrin, cholecystokinin and angiotensin
Pharmacology of Bradykinin , pentagastrin, cholecystokinin and angiotensinPharmacology of Bradykinin , pentagastrin, cholecystokinin and angiotensin
Pharmacology of Bradykinin , pentagastrin, cholecystokinin and angiotensin
 
Renin Angiotensin Aldosterone System (RAAS).pptx
Renin Angiotensin Aldosterone System (RAAS).pptxRenin Angiotensin Aldosterone System (RAAS).pptx
Renin Angiotensin Aldosterone System (RAAS).pptx
 
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
Lecture 15,16--Drugs Used in Congestive Heart Failure 1 and 2 Syllabus(1)_mer...
 
Brain renin angiotensin system
Brain renin        angiotensin systemBrain renin        angiotensin system
Brain renin angiotensin system
 
.-11 - Renal Modulation_ The Renin-Angiotensin Systemªªªª.pdf
.-11 - Renal Modulation_ The Renin-Angiotensin Systemªªªª.pdf.-11 - Renal Modulation_ The Renin-Angiotensin Systemªªªª.pdf
.-11 - Renal Modulation_ The Renin-Angiotensin Systemªªªª.pdf
 
Biochemical properties of renin and prorenin binding
Biochemical properties of renin and prorenin bindingBiochemical properties of renin and prorenin binding
Biochemical properties of renin and prorenin binding
 
RAAS mbbs
RAAS mbbsRAAS mbbs
RAAS mbbs
 
Renin in Vertebrates-1.pdf
Renin in Vertebrates-1.pdfRenin in Vertebrates-1.pdf
Renin in Vertebrates-1.pdf
 
Chen et al PNAS-2010
Chen et al PNAS-2010Chen et al PNAS-2010
Chen et al PNAS-2010
 
MetabolicAcidosisinA.pdf
MetabolicAcidosisinA.pdfMetabolicAcidosisinA.pdf
MetabolicAcidosisinA.pdf
 
Presentation final
Presentation finalPresentation final
Presentation final
 
PCPSeminar_Shraddha
PCPSeminar_ShraddhaPCPSeminar_Shraddha
PCPSeminar_Shraddha
 

More from Paulo Osmar

1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
Paulo Osmar
 
3. ejercicios heredabilidad
3. ejercicios heredabilidad3. ejercicios heredabilidad
3. ejercicios heredabilidad
Paulo Osmar
 
5. lectura corazón
5. lectura   corazón5. lectura   corazón
5. lectura corazón
Paulo Osmar
 
1. lectura potencial de membrana
1. lectura   potencial de membrana1. lectura   potencial de membrana
1. lectura potencial de membrana
Paulo Osmar
 
4. lectura caracteristicas del sistema nerviosos para produccion de movimiento
4. lectura   caracteristicas del sistema nerviosos para produccion de movimiento4. lectura   caracteristicas del sistema nerviosos para produccion de movimiento
4. lectura caracteristicas del sistema nerviosos para produccion de movimiento
Paulo Osmar
 

More from Paulo Osmar (6)

1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
1. Lectura - Terminos de Uso Comun Nutricion y Alimentacion.PDF
 
3. ejercicios heredabilidad
3. ejercicios heredabilidad3. ejercicios heredabilidad
3. ejercicios heredabilidad
 
5. lectura corazón
5. lectura   corazón5. lectura   corazón
5. lectura corazón
 
1. lectura potencial de membrana
1. lectura   potencial de membrana1. lectura   potencial de membrana
1. lectura potencial de membrana
 
4. lectura caracteristicas del sistema nerviosos para produccion de movimiento
4. lectura   caracteristicas del sistema nerviosos para produccion de movimiento4. lectura   caracteristicas del sistema nerviosos para produccion de movimiento
4. lectura caracteristicas del sistema nerviosos para produccion de movimiento
 
Control musculo esqueletico
Control musculo esqueleticoControl musculo esqueletico
Control musculo esqueletico
 

Recently uploaded

Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptx
Silpa
 
development of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virusdevelopment of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virus
NazaninKarimi6
 
Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.
Silpa
 
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 bAsymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Sérgio Sacani
 
POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.
Silpa
 
Conjugation, transduction and transformation
Conjugation, transduction and transformationConjugation, transduction and transformation
Conjugation, transduction and transformation
Areesha Ahmad
 

Recently uploaded (20)

CURRENT SCENARIO OF POULTRY PRODUCTION IN INDIA
CURRENT SCENARIO OF POULTRY PRODUCTION IN INDIACURRENT SCENARIO OF POULTRY PRODUCTION IN INDIA
CURRENT SCENARIO OF POULTRY PRODUCTION IN INDIA
 
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptxClimate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
 
Thyroid Physiology_Dr.E. Muralinath_ Associate Professor
Thyroid Physiology_Dr.E. Muralinath_ Associate ProfessorThyroid Physiology_Dr.E. Muralinath_ Associate Professor
Thyroid Physiology_Dr.E. Muralinath_ Associate Professor
 
Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptx
 
GBSN - Microbiology (Unit 1)
GBSN - Microbiology (Unit 1)GBSN - Microbiology (Unit 1)
GBSN - Microbiology (Unit 1)
 
Selaginella: features, morphology ,anatomy and reproduction.
Selaginella: features, morphology ,anatomy and reproduction.Selaginella: features, morphology ,anatomy and reproduction.
Selaginella: features, morphology ,anatomy and reproduction.
 
An introduction on sequence tagged site mapping
An introduction on sequence tagged site mappingAn introduction on sequence tagged site mapping
An introduction on sequence tagged site mapping
 
development of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virusdevelopment of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virus
 
Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.
 
Introduction of DNA analysis in Forensic's .pptx
Introduction of DNA analysis in Forensic's .pptxIntroduction of DNA analysis in Forensic's .pptx
Introduction of DNA analysis in Forensic's .pptx
 
Chemistry 5th semester paper 1st Notes.pdf
Chemistry 5th semester paper 1st Notes.pdfChemistry 5th semester paper 1st Notes.pdf
Chemistry 5th semester paper 1st Notes.pdf
 
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 bAsymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
 
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceuticsPulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
 
Site Acceptance Test .
Site Acceptance Test                    .Site Acceptance Test                    .
Site Acceptance Test .
 
POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.
 
Conjugation, transduction and transformation
Conjugation, transduction and transformationConjugation, transduction and transformation
Conjugation, transduction and transformation
 
GBSN - Microbiology (Unit 2)
GBSN - Microbiology (Unit 2)GBSN - Microbiology (Unit 2)
GBSN - Microbiology (Unit 2)
 
GBSN - Microbiology (Unit 3)
GBSN - Microbiology (Unit 3)GBSN - Microbiology (Unit 3)
GBSN - Microbiology (Unit 3)
 
Velocity and Acceleration PowerPoint.ppt
Velocity and Acceleration PowerPoint.pptVelocity and Acceleration PowerPoint.ppt
Velocity and Acceleration PowerPoint.ppt
 
300003-World Science Day For Peace And Development.pptx
300003-World Science Day For Peace And Development.pptx300003-World Science Day For Peace And Development.pptx
300003-World Science Day For Peace And Development.pptx
 

6. lectura sistema renina angiotensina

  • 1. 1499 Chapter 204 Renin–Angiotensin System Minolfa C. Prieto, Alexis A Gonzalez, Ryousuke Satou, Hiroyuki Kobori and L. Gabriel Navar ABSTRACT Renal angiotensin (Ang) II is regulated independently of the sys- temic renin–angiotensin system. Intrarenal Ang II concentrations are higher in renal interstitium and proximal tubular compartments than in the circulation. The transcript for angiotensinogen (AGT) is in proximal tubule cells and for renin in juxtaglomerular and col- lecting duct cells. Secretion of their proteins increases intratubular formation of angiotensin peptides. Ang II is internalized via AT1R into renal endosomes. When inappropriately stimulated, Ang II augments synthesis and secretion of AGT in proximal tubules and renin in collecting ducts, which increase further intrarenal Ang II and cause hypertension. Ang II has pleiotropic actions on cortical and medullary blood flow, vascular tone of afferent and efferent arterioles, mesangial cell function, and sensitivity of the tubulo- glomerular feedback mechanism via the activation of AT1R. AT2R and Ang III partially oppose these actions. Ang II also regulates sodium transport in proximal and distal nephrons via the activation of luminal and basolateral AT1R. INTRODUCTION The contribution of the intrarenal renin–angiotensin system (RAS) to the regulation of renal function is unique because renal functional, and transport alterations caused by angio- tensin II (Ang II) influence overall sodium balance and cardiovascular status and thus contribute to blood pressure regulation. When inappropriately stimulated, an enhanced intrarenal RAS markedly influences renal function by act- ing on vascular and tubular components causing the devel- opment and maintenance of hypertension and associated cardiovascular dysfunction.12,18–20,33 This chapter covers the intrarenal RAS, the formation of several angiotensin peptides by various enzymes, and the characterization of angiotensin receptors including AT1a, AT1b, and AT2 for Ang II and Ang III, and the Mas receptor for Ang 1–7. In addition, we describe some newly discov- ered aspects of the intrarenal RAS including the regulation of proximal tubule angiotensinogen (AGT), collecting duct renin, and the renin/(pro)renin receptor. DISTRIBUTION OF ANGIOTENSINOGEN, RENIN, (PRO)RENIN RECEPTORS, ANGIOTENSIN CONVERTING ENZYMES, AND ANG TYPE 1 RECEPTORS IN THE KIDNEY All components of the RAS are present in the kidney and are compartmentalized to the tubular and interstitial networks. Because kidney Ang II tissue concentrations are greater than can be explained by the concentrations delivered in the arterial blood flow, it is generally acknowledged that most of the Ang II in the intrarenal components is generated locally from AGT delivered to the kidney as well as that locally produced by proximal tubule cells. The presence of abundant sources of renin,AGT, and angiotensin-converting enzyme (ACE) in the kidney provides an environment con- ducive to high levels of Ang II synthesis.19,21 Angiotensinogen AGT, the only known substrate for renin, is a 60-kD glyco- protein synthesized and released from the liver, and cleaved in the circulation to form the biologically inactive, Ang I (Fig. 1). In addition, renal AGT mRNA and protein are localized to proximal tubule cells indicating that much of the intratubular Ang II is derived from locally formed and secreted AGT. AGT is regulated by an amplification mecha- nism such that AGT mRNA and protein are stimulated by Ang II, which maintains or increases the production of Ang II in Ang II-dependent hypertension. Producing its metabo- lites intracellularly and secreting them into the tubule lumen, AGT is taken up by megalin-dependent endocytosis and is then secreted into the tubular lumen.12,19,24 More recent studies have evaluated the molecular mechanisms respon- sible for the regulation of AGT expression and synthesis in proximal tubular cells.21 Activation of AT1 receptors stimu- lates intrarenal NF-κB, which is a key factor in the aug- mentation of AGT expression in proximal tubule cells.1,20 The activation of NF-κB by Ang II leads to the stimulation Handbook of Biologically Active Peptides. http://dx.doi.org/10.1016/B978-0-12-385095-9.00204-9 Copyright © 2013 Elsevier Inc. All rights reserved.
  • 2. Chapter | 204  Renin–Angiotensin System 1500 of intrarenal proinflammatory cytokine expressions such as interleukin-6, contributing to the increase in AGT expres- sion via activation of a JAK-STAT pathway.29 Therefore, Ang II increases intrarenal AGT levels via both direct and indirect mechanisms mediated by activated NF-κB and the JAK-STAT pathway. By contrast, tumor necrosis factor α, which is also an Ang II-induced proinflammatory factor in the kidney, suppresses AGT expression by increasing p50/ p50 complex in proximal tubular cells.30 This inhibitory action on NF-κB provides a counterregulatory influence limiting the magnitude of Ang II-induced AGT augmen- tation in renal proximal tubular cells, which may explain why higher doses of Ang II infusion in mice fail to augment intrarenal AGT levels.20 Proximal tubuleAGT concentrations in anesthetized rats are in the range of 300 nM, which greatly exceeds the free Ang I and Ang II tubular fluid concentrations. Because of its molecular size, it is unlikely that much plasma AGT fil- ters across the glomerular membrane, further supporting the concept that proximal tubule cells secrete AGT directly into the tubule.12 In response to chronic Ang II infusions there is an augmentation of AGT mRNA leading to increased syn- thesis and secretion with some of the secreted AGT spilling over to the distal nephron segments as reflected by increased urinary AGT excretion.13 Under conditions of elevated salt intake combined with chronic Ang II infusion, the urinary AGT excretion response is elevated to much greater lev- els than those seen in Ang II-infused rats fed a normal salt diet.14 By contrast, normal rats fed a high salt diet exhibit urinary AGT excretion rates similar to those observed in rats fed a normal salt diet. Thus, during chronic Ang II infu- sion, salt-sensitive hypertension develops and high salt par- adoxically stimulates intrarenal RAS activity, as reflected by exacerbation of the urinary AGT despite inhibition of plasma renin activity.14 Formation of Ang I and II in the tubular lumen subsequent to AGT secretion is probable because some renin is filtered and/or secreted from juxta- glomerular apparatus (JGA) cells.12 Once Ang I is formed, conversion readily occurs because there is abundant ACE associated with the proximal tubule brush border.32 Intact AGT in urine reflects its presence throughout the nephron and, to the extent that renin and ACE are available along the nephron, substrate availability supports continued Ang I generation and Ang II conversion in distal segments.19,25 Renin and (Pro)renin Receptor Renin, synthesized by the JGA cells, is the primary source of both circulating and intrarenal renin levels. Juxtaglo- merular apparatus cells secrete renin into the renal inter- stitium, thus providing a stimulus for the local generation of Ang I, and this secreted active form contains 339–343 amino acid residues after proteolytic removal of the 43 amino acid residue at the N-terminus of prorenin. Circu- lating active renin and prorenin are derived mainly from the kidney; and circulating prorenin is taken up by some tissues where it may contribute to the local synthesis of angiotensin peptides. The only well-established role of FIGURE 1  Intrarenal renin–angiotensin system cascade. Known and postulated enzymatic pathways responsible for formation and metabolism of angiotensin peptides.
  • 3. 1501 SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides renin is to cleave Ang I from AGT. However, the recently cloned (pro)renin receptor ((P)RR) binds renin or pro- renin, thus enhancing renin activity and fully activating the biologically inactive prorenin peptide, which leads to an increase in the catalytic efficiency of Ang I formation from AGT. Activation of the (P)RR by renin or prorenin also directly elicits intracellular signals that stimulate ERK1/ ERK2 and downstream pathways.22 There is growing awareness of the possible roles of renin in other intrarenal structures where renin has been local- ized. Renin producing cells have a unique origin that allows their abundance along the afferent arteriole to be regulated by various stimuli. ACE inhibition induces a recruitment of cells in afferent arterioles beyond the JGA even though they were not expressing the renin gene in the basal state.31 Posi- tive renin immunoreactivity has also been observed in the cells of glomeruli and in proximal and distal tubular seg- ments. More recently, increased attention has been focused on the presence of renin mRNA and protein expression in distal nephron segments, specifically in the principal cells of connecting tubules and collecting ducts, suggesting local formation and secretion into the distal tubular fluid. Renin synthesis in principal cells of collecting ducts is further increased by chronic infusions of Ang II.25,26 Rats chroni- cally infused with Ang II for 13 days show a suppression of plasma renin activity but increases in urinary prorenin con- tent and renin excretion.16 In addition, the urinary renin and prorenin protein levels, examined by Western blot, are aug- mented about 10-fold in Ang II-infused rats. Concomitant AT1R blockade with candesartan prevents increases in both urinary and tissue renin and in prorenin levels. Although there is marked suppression of plasma renin activity, increased renin synthesis by CD cells and secretion into the luminal fluid lead to increased urinary levels of renin and prorenin.16 The presence of renin in distal nephron segments pro- vides the requisite pathway for Ang I generation from prox- imally delivered AGT.25,26 Renin synthesis is augmented in the principal cells of connecting tubules and cortical and medullary collecting ducts of chronic Ang II-infused rats and mice, Cyp1a1Ren2 transgenic rats, and in both kidneys of two-kidney one clip Goldblatt hypertensive rats.9,25,26 The AT1R via a PKC pathway directly mediates the stimulation of renin synthesis in rat inner medullary col- lecting duct because inhibition of PKC with calphostin C prevents the Ang II-mediated increases in renin transcript and prorenin protein levels in rat inner medullary collecting duct (IMCD) cells while activation of PKC with phorbol 12-myristate 13-acetate increases renin expression to the same extent as Ang II.7 Further, renin activity and renin and prorenin protein levels are increased in the urine of these rats6 indicating that both renin and prorenin are secreted by the collecting duct cells into the lumen of distal nephron segments and are also augmented in chronic Ang II-infused hypertensive rats. The discovery of the (P)RR provides new insights regarding its possible role in a setting of activated intrarenal RAS. The (P)RR is expressed in the kidneys6 particularly in mesangial cells, podocytes, and intercalated type A cells of distal nephron segments.2,6,11 Chronic infu- sion of Ang II for 14 days in Sprague–Dawley rats increases renal (P)RR transcript levels and augments the soluble form of the (P)RR, s(P)RR.6 The augmentation of s(P)RR in the urine of Ang II hypertensive rats suggests that s(P)RR can bind renin and prorenin secreted by the principal collecting duct cells and can catalyze additional formation of Ang I. The existence of an inducible and functional form of s(P) RR in the renal medulla and in the urine of chronic Ang II-infused rats reflects increased s(P)RR in collecting duct tubular fluid and further supports the potential role of this soluble form to enhance intratubular renin activity. Thus, the interaction between the (P)RR and renin in the lumen of the collecting ducts may contribute substantially to facili- tate intratubular formation of Ang I in hypertension. Angiotensin-Converting Enzymes As in other organs, ACE is located on endothelial cells throughout the vasculature. ACE is also on membranes of both proximal and distal nephron segments with the greatest abundance found on the brush border of proximal tubules, particularly in the S3 segment.21,32 Marked upregulation of ACE in the brush border occurs in kidneys from Goldblatt hypertensive rats and Ang II infused hypertensive rats.32 Humans predominantly express ACE in the brush border of proximal tubular segments with very little ACE expres- sion on vascular endothelial cells or in the vasculature of the glomerular tuft. The lower renal vascular endothelial expression in humans helps explain the much lowerAng I to Ang II conversion rates that have been reported for human kidneys as compared with that of other species. In rats, ACE activity is present in tubular fluid throughout the nephron except in the distal tubule, with activity higher at the initial portion of the proximal tubule, decreasing toward the distal nephron, and increasing again in the collecting ducts and urine. Therefore, intratubular Ang II formation may occur not only in the proximal tubule but also beyond the distal convoluted tubule. Renal tissue ACE activity is critical to maintain the steady-state Ang II levels in the kidney. The presence of ACE only in the kidney is sufficient to increase kidneyAng II levels and promote the development of hyper- tension in response to chronicAng I infusions.8 Tissue-ACE knockout mice exhibit 80% lower intrarenal Ang II levels compared with wild-type mice.3 ACE2, which acts on Ang II to form Ang 1–7 or on Ang I to form Ang 1–9 (Fig. 1), is a membrane-associated and secreted enzyme expressed predominantly on endothelium,
  • 4. Chapter | 204  Renin–Angiotensin System 1502 but highly restricted in humans to heart, kidneys, and tes- tes.21 Ang 1–7 may serve as an endogenous antagonist of the Ang II induced actions mediated via AT1 receptors and changes in ACE2 activity could affect the balance of Ang peptides found in the kidney. This probably explains the ele- vatedAng II levels in theACE2 knockout mice.28 Collectrin, a novel homolog of ACE2 has been identified in mouse, rat, and human. Both ACE2 and collectrin have tissue-restricted expression in the kidney. Collectrin is localized on the lumi- nal surface and in the cytoplasm of collecting duct cells, and its mRNA is expressed in the renal collecting ducts cells while ACE2 is present throughout the endothelium, in proximal tubular epithelial cells and in medullary tis- sue.21 In Ang-II-dependent hypertension, intrarenal ACE is increased, but ACE2 levels are decreased leading to another mechanism maintaining increased Ang II levels.26 Intrarenal Angiotensin II Receptors Ang II receptors are widely distributed in various regions and cell types of the kidney. Two major categories of Ang II receptors, AT1 and AT2, have been described, pharmacologi- cally characterized and cloned.18,21 As depicted in Figure 2, most of the Ang-II-mediated hypertensinogenic actions are generally attributed to the AT1 receptor.5,18 In rodents, there are two AT1 receptor subtypes, with the AT1a being the pre- dominant subtype in all nephron segments, while the AT1b is more abundant in the glomerulus.18,21 In mature kidneys, AT1a receptors have been localized to the renal microvascu- lature in cortex and medulla, smooth muscle cells of afferent and efferent arterioles, thick ascending limb of the loop of Henle, proximal tubular apical and basolateral membranes, mesangial cells, distal tubules, collecting ducts, and macula densa cells. The afferent arteriole has both AT1a and AT1b receptors, whereas the efferent arteriole primarily expresses AT1a receptors.10 The essential role of the AT1a receptor in mice is apparent from studies showing that 2-kidney, 1-clip (2K1C) Goldblatt hypertension does not develop in AT1a knockout mice.4 Vascular and tubular receptors respond differently dur- ing high Ang II states.18 High Ang II levels associated with a low salt diet decrease glomerular AT1 receptor expression but increase proximal tubular AT1 receptor levels. Glomeru- lar AT1 receptors and AT1a receptor protein are reduced in both clipped and contralateral kidneys of 2K1C Goldblatt and of 2 kidney-1-wrap hypertensive models and in kidneys of Ang II infused rats.18 In the TGR(mRen2) harboring the mouse renin2 gene, AT1 receptor binding is increased in vascular smooth muscle of afferent and efferent arteri- oles, JGA, glomerular mesangial cells, proximal tubular cells, and renomedullary interstitial cells suggesting that the upregulation of AT1 receptors contributes to the patho- genesis of hypertension in these rats. In Ang II infused rats studied with in vitro autoradiography, there are differential responses with significant decreases in glomeruli and inner stripe but not in proximal tubules.18 The AT2 receptor is highly expressed in human and rodent kidney mesenchyme during fetal life and decreases dramatically after birth.AT2 receptors have been localized to the glomerular epithelial cells, proximal tubules, collecting ducts, and parts of the renal vasculature of the adult rat. AT2 receptor activation is believed to counteract AT1 receptor FIGURE 2  Major Ang II receptor subtypes and their renal actions.
  • 5. 1503 SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides effects by stimulating the formation of bradykinin and nitric oxide leading to increases in interstitial fluid concentration of cyclic guanosine monophosphate (cGMP). AT2 receptor activation influences proximal tubule sodium reabsorption either by a cell-membrane-receptor-mediated mechanism or via an interstitial nitric-oxide-cGMP pathway.17,18 INTRARENAL LEVELS OF ANGIOTENSIN II Interstitial Angiotensin II Intrarenal Ang II is not distributed in a homogeneous fashion but is compartmentalized in both a regional and segmental manner.19 Medullary Ang II levels are higher than cortical levels in normal rats and increase further in Ang II infused hypertensive rats.18 Within the cortex, there is distribution of Ang II in the interstitial fluid, tubular fluid, and intracellular compartments. The interstitial and intratubular compartments contribute to the disproportion- ately high intrarenal Ang II levels. Ang II concentrations in interstitial fluid are much higher than the plasma con- centrations with values in the range of 3–5 pmoles/mL.19 Renal interstitial fluid Ang II levels increase further in hypertensive rats infused with Ang II for 2 weeks. The high renal interstitial values indicate local regulation of Ang II formation in the renal interstitial compartment and an enhancement of interstitial Ang II production in Ang II dependent hypertension.12 Tubular Ang II As shown in Figure 3, proximal tubule fluid concentrations of Ang I and Ang II are also much greater than the plasma concentrations and have been found to be in the range of 5–10 pmoles/ml.19 In perfused tubules, where the contri- bution resulting from glomerular filtration is prevented, Ang II concentrations are similar to those in nonperfused tubules indicating that the proximal tubule secretes Ang II or a precursor into the proximal tubule fluid. In addition to AGT, proximal tubule cells have renin that can act on AGT to generate Ang I. Evidence that renin is augmented in the urine of chronic Ang II-infused rats also indicates secretion of renin by the collecting duct cell into the tubular lumen16 and provides a pathway for Ang I generation from proxi- mally delivered AGT, leading to concentrations of Ang II in distal tubular fluid in the picomolar range.34 Intracellular Angiotensin II Ang II is internalized via AT1–receptor-mediated endo- cytosis and endosomal accumulation of Ang II in inter- microvillar clefts and endosomes is increased further in Ang II infused hypertensive rats.35 AT1 receptor block- ade prevents the endosomal accumulation even though plasma Ang II increases. The presence of Ang II in renal endosomes indicates that some of the internalized Ang II remains intact and contributes to the total Ang II content measured in tissue homogenates.35 Endocytosis of the Ang II-AT1 receptor complex is required for the full expression of functional responses coupled to the activation of signal transduction pathways, but the full extent of the functions exerted by intracellular Ang II has not been determined.15 In Ang-II-dependent hypertension, a higher fraction of the total kidney Ang II is internalized into intracellular endosomes consisting of both light endosomes and inter- microvillar clefts via an AT1-receptor-mediated process. AT1 receptor blockade prevents the augmentation of intra- cellular Ang II.35 FIGURE 3  Intratubular processing of the renin–angiotensin system. All components of the renin–angiotensin system are present in the kidney provid- ing multiple pathways for enhanced intrarenal Ang II formation. PT—proximal tubule; DT—distal tubule; CD—collecting duct; AGT—angiotensinogen; JGA—juxtaglomerular apparatus; AA—afferent arteriole; EA—efferent arteriole; ACE—angiotensin converting enzyme; PRR—(pro)renin receptor (modi- fied from Ref. 19).
  • 6. Chapter | 204  Renin–Angiotensin System 1504 Our understanding of the functions of internalized Ang II remains incomplete. Ang II may be recycled and secreted to exert further actions by binding to Ang II receptors on the cell membranes. Ang II may also act on cytosolic recep- tors to activate intracellular signaling mechanisms.15 Ang II may also migrate to the nucleus to exert genomic effects.21 Nuclear binding sites of the AT1 subtype in renal cells have been reported. Because Ang II exerts a positive stimulation on AGT mRNA and protein production, intracellular Ang II may have genomic actions to regulate AGT or renin mRNA expression in proximal tubule cells either directly or via NF-κB.1,21,30 BIOLOGICAL ACTIONS OF INTRARENAL ANG II In view of the near ubiquitous distribution of Ang II recep- tors in the various structures of the kidney, it is not surpris- ing that Ang II exerts pleiotropic effects at multiple levels within the kidney. As shown in Figure 2, the actions of Ang II span both cortical and medullary, vascular and tubular and can be stimulatory or inhibitory.18 At the level of the vasculature, Ang II exerts important direct and indirect effects to regulate vascular tone of the afferent and efferent arterioles and of the mesangial cells.17 Ang II also regulates afferent arteriolar tone through its influence on the tubulo- glomerular feedback mechanism with elevated Ang II levels increasing the sensitivity of the feedback mechanism, thus regulating the amount of filtrate that escapes the proximal nephron and enters the distal nephron segments. In the post- glomerular circulation, Ang II regulates both the cortical and medullary capillary blood flow and helps to regulate the relative blood flowing to the medullary tissues through its control of the tone of the pericytes.17 Interestingly, although Ang II constricts both afferent and efferent arterioles, the mechanisms are different. The afferent arteriolar constrictor responses to Ang II involve activation of L-type Ca++ channels to provide sustained Ca++ entry and vasoconstriction.17 By contrast, efferent arterioles of normal rats do not vasodilate or vasodilate much less in response to L-type Ca++ channel blockers and the vasoconstrictor effects of Ang II are not blocked by L-type Ca++ channel blockers. Rather, T-type channels have greater functional significance in regulating the effer- ent arteriolar constrictor responses to Ang II.17 T-type Ca++ channel blockers markedly attenuate the efferent vasocon- strictor responses to Ang II. Storage operated Ca++ chan- nels in efferent and afferent arterioles are also activated by Ang II. Thus, although both afferent and efferent arterioles respond to Ang II, the mechanisms for Ca++ entry and the activation pathways are different. These differences may be attributable to the differential abundance of AT1A and AT1B receptors on the afferent and efferent arterioles.10 Virtually, all aspects of the filtration and tubular reab- sorptive processes are influenced by Ang II. Ang II directly regulates the glomerular filtration coefficient with high Ang II concentrations reducing and low levels or blockade of AT1 receptors increasing the filtration coefficient.17,18 At the level of the proximal tubule, Ang II binds to receptors at both the apical and basolateral membranes, stimulating the activity of the sodium/hydrogen exchanger and the sodium bicarbonate co-transporter, thereby exerting an important influence on net proximal reabsorption rate.18 Transport mechanisms in the proximal tubule, loop of Henle, and dis- tal nephron segments are also influenced by Ang II.18,27,34 Under conditions in which the activity of the RAS is stimulated and greater amounts of AGT are secreted from the proximal tubule, the locus of influence shifts to include the distal nephron segments where the interaction between the (P)RR and renin/prorenin may contribute substantially to increase intrarenal and intratubular formation of Ang II (Fig. 4).7,20 The in vivo demonstration that increased urinary Ang II concentrations in mice infused chronically with Ang II enhance distal sodium reabsorption34 emphasizes further the importance that renin and the (P)RR may have in the distal nephron segments contributing to the increases in intratubular Ang II formation. Ang II stimulates the sodium/ hydrogen exchanger and the sodium chloride cotransporter in distal tubules, and the amiloride sensitive sodium channel in principal cells of connecting tubules and collecting duct segments.20,23 This enhanced stimulatory effect allows the kidney to have maximum capability to conserve sodium by markedly decreasing fractional sodium reabsorption.34 At the whole kidney level, this is manifested as a marked sup- pression of the pressure natriuresis relationship. Ang II also regulates acid base balance through its effects on the Na+/ H+ exchanger as well as by influencing the activity of the H+ ATPase in intercalated cells.2 Collectively, the actions of Ang II allow the kidney to maximize sodium conservation without compromising its ability to regulate its many other functions. CONCLUDING COMMENTS AND PERSPECTIVE Recent findings have stimulated interest in the molecular mechanisms regulating the various components of the intra- renal RAS and particularly the interstitial, intracellular, and intratubular concentrations of Ang II and related peptides. Intratubular and interstitial Ang II levels are regulated inde- pendently of the circulatingAng II and exert powerful actions via the stimulation of AT1 receptors on the vascular, glomer- ular, and tubular structures to provide a synchronous cascade of effects contributing to the ability of the kidney to retain >99% of the filtered sodium. From a functional perspective, the effects of Ang II on proximal nephron reabsorption as
  • 7. 1505 SECTION | XV  Handbook of Biologically Active Peptides: Renal Peptides well as on distal nephron transport function, coupled with the associated actions of elevated aldosterone levels, mark- edly increase the sodium retaining capability of the kidney. When activated in a physiologically appropriate setting under conditions of volume contraction or salt deficient states, these actions can be life saving. When inappropriately main- tained or augmented, however, these effects contribute to the development and maintenance of hypertension. Further, the sustained increases in intrarenal Ang II in a setting of hyper- tension can lead to progressive renal injury, cell proliferation, and fibrosis associated with activation of several major cyto- kines and growth factors.12,33 ACKNOWLEDGMENTS The authors thank Debbie Olavarrieta for the preparation of the man- uscript and figures. The authors’ research related to this review has been supported by research grants from NHLBI, NIDDK, NCRR, and AHA. REFERENCES 1. Acres OW, Satou R, Navar LG, Kobori H. Contribution of a Nuclear Factor-{kappa}B binding site to human angiotensinogen Promoter activity in renal proximal tubular cells. Hypertension 2011;57: 608–13. 2. Advani A, Kelly DJ, Cox AJ, White KE, Advani SL, Thai K, et al. The (Pro)renin receptor: site-specific and functional linkage to the vacuolar H+-ATPase in the kidney. Hypertension 2009;54:261–9. 3. Bernstein KE, Xiao HD, Adams JW, Frenzel K, Li P, Shen XZ, et al. Establishing the role of angiotensin-converting enzyme in renal func- tion and blood pressure control through the analysis of genetically modified mice. J Am Soc Nephrol 2005;16:583–91. 4. Cervenka L, Vaneckova I, Huskova Z, Vanourkova Z, Erbanova M, Thumova M, et  al. Pivotal role of angiotensin II receptor subtype 1A in the development of two-kidney, one-clip hypertension: study in angiotensin II receptor subtype 1A knockout mice. J Hypertens 2008;26:1379–89. 5. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP, et  al. Angiotensin II causes hypertension and cardiac hypertrophy through its receptors in the kidney. Proc Natl Acad Sci U S A 2006;103:17985–90. 6. Gonzalez AA, Lara LS, Luffman C, Seth DM, Prieto MC. Soluble form of the (pro)renin receptor is augmented in the collecting duct and urine of chronic angiotensin II-dependent hypertensive rats. Hyperten- sion 2011;57:859–64. 7. Gonzalez AA, Liu L, Lara LS, Seth DM, Navar LG, Prieto MC. Angiotensin II stimulates renin in inner medullary collecting duct cells via protein Kinase C and Independent of epithelial sodium channel and Mineralocorticoid receptor activity. Hypertension 2011;57:594–9. 8. Gonzalez-Villalobos RA, Billet S, Kim C, Satou R, Fuchs S, Bernstein KE, et al. Intrarenal angiotensin-converting enzyme induces hypertension in response to Angiotensin I infusion. J Am Soc Nephrol 2011;22:449–59. 9. Gonzalez-Villalobos RA, Satou R, Ohashi N, Semprun-Prieto LC, Katsurada A, Kim C, et  al. Intrarenal mouse renin-angiotensin system during ANG II-induced hypertension and ACE inhibition. Am J Physiol Ren Physiol 2010;298:F150–7. 10. Harrison-Bernard LM, Cook AK, Oliverio MI, Coffman TM. Renal segmental microvascular responses to ANG II in AT1A receptor null mice. Am J Physiol Ren Physiol 2003;284:F538–45. 11. IchiharaA, Sakoda M, Kurauchi-MitoA, KaneshiroY, Itoh H. Involve- ment of (pro)renin receptor in the glomerular filtration barrier. J Mol Med 2008;86:629–35. 12. Kobori H, Nangaku M, Navar LG, Nishiyama A. The intrarenal renin– angiotensin system: from physiology to the pathobiology of hyperten- sion and kidney disease. Pharmacol Rev 2007;59:251–87. FIGURE 4  Renin and (pro)renin receptor interaction in the collecting duct. Prorenin and renin stimulation in the principal cells of the collecting ducts may increase Ang I formation in the tubular lumen and increase Ang II generation. The presence of (pro)renin receptor at the surface of the intercalated cells along with its soluble form in the tubular lumen increases the catalytic efficiency for renin to form Ang I. In addition, the pro(renin) receptor bound to the membrane of the intercalated cells may anchor prorenin secreted by the principal cells to reduce the washout of prorenin or renin into the urine. ANG—angiotensin; AGT—angiotensinogen; AT1R—angiotensin II type 1 receptor; PRR—(pro)renin receptor; sPRR—soluble form of the (pro)renin receptor; ACE—angiotensin converting enzyme. See color plate 54.
  • 8. Chapter | 204  Renin–Angiotensin System 1506 13. Kobori H, Prieto-Carrasquero MC, OzawaY, Navar LG. AT1 receptor mediated augmentation of intrarenal angiotensinogen in angiotensin II-dependent hypertension. Hypertension 2004;43:1126–32. 14. Lara LS, McCormack M, Semprum-Prieto LC, Shenouda S, Majid DS, Kobori H, et al. AT1 receptor-mediated augmentation of angiotensinogen, oxidative stress, and inflammation in ANG II-salt hypertension. Am J Physiol Ren Physiol 2012;302:F85–94. 15. Li XC, Hopfer U, Zhuo JL. AT1 receptor-mediated uptake of angio- tensin II and NHE-3 expression in proximal tubule cells through a microtubule-dependent endocytic pathway. Am J Physiol Ren Physiol 2009;297:F1342–52. 16. Liu L, Gonzalez AA, McCormack M, Seth DM, Kobori H, Navar LG, et  al. Increased renin excretion associated with aug- mented urinary angiotensin (Ang) II levels in chronic angiotensin II-infused hypertensive rats. Am J Physiol Ren Physiol 2011;301: F1195–201. 17. Navar LG,Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The renal Microcirculation. In: Tuma RF, Duran WN, Ley K, editors. Handbook of physiology: microcirculation. Academic Press; 2008. p. 550–683. 18. Navar LG, Harrison-Bernard LM, Imig JD, Mitchell KD. Renal actions of angiotensin II and AT1 receptor blockers. In: Epstein M, Brunner HR, editors. Angiotensin II receptor antagonists. Philadelphia: Hanley Belfus, Inc.; 2000. p. 189–214. 19. Navar LG, Kobori H, Prieto MC, Gonzalez-Villalobos RA. Intra- tubular renin-angiotensin system in hypertension. Hypertension 2011;57:355–62. 20. Navar LG, Prieto MC, Satou R, Kobori H. Intrarenal angiotensin II and its contribution to the genesis of chronic hypertension. Curr Opin Pharmacol 2011;11:180–6. 21. Navar LG, Prieto-Carrasquero MC, Kobori H. Molecular aspects of the renal renin–angiotensin system. In: Re R, DiPette DJ, Schiffrin EL, Sowers JR, editors. Molecular mechanisms in hypertension. Taylor Francis Group; 2006. p. 3–14. 22. Nguyen G, Muller DN. The biology of the (pro)renin receptor. J Am Soc Nephrol 2010;21:18–23. 23. Peti-Peterdi J, Warnock DG, Bell PD. Angiotensin II directly stimu- lates ENaC activity in the cortical collecting duct via AT(1) receptors. J Am Soc Nephrol 2002;13:1131–5. 24. Pohl M, Kaminski H, Castrop H, Bader M, Himmerkus N, Bleich M, et al. Intrarenal renin angiotensin system Revisited: role of megalin- dependent endocytosis along the proximal nephron. J Biol Chem 2010;285:41935–46. 25. Prieto-Carrasquero MC, Botros FT, Kobori H, Navar LG. Collecting duct renin: a major player in angiotensin II-dependent hypertension. J Am Soc Hypertens 2009;3:96–104. 26. Prieto-Carrasquero MC, Botros FT, Pagan J, Kobori H, Seth DM, Casarini DE, et al. Collecting duct renin is upregulated in both kid- neys of 2-kidney, 1-clip goldblatt hypertensive rats. Hypertension 2008;51:1590–6. 27. Riquier-BrisonAD, Leong PK, Pihakaski-Maunsbach K, Mcdonough AA.Angiotensin II stimulates trafficking of NHE3, NaPi2, and associ- ated proteins into the proximal tubule microvilli. Am J Physiol Ren Physiol 2010;298:F177–86. 28. Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP, de B I, et al. Angiotensin-(1-7) is an endogenous ligand for the G pro- tein-coupled receptor Mas. Proc NatlAcad Sci U SA 2003;100:8258–63. 29. Satou R, Gonzalez-Villalobos RA, Miyata K, Ohashi N, Urushihara M, Acres OW, et al. IL-6 augments angiotensinogen in primary cultured renal proximal tubular cells. Mol Cell Endocrinol 2009;311:24–31. 30. Satou R, Miyata K, KatsuradaA, Navar LG, Kobori H. Tumor necrosis factor-{alpha} suppresses angiotensinogen expression through forma- tion of a p50/p50 homodimer in human renal proximal tubular cells. Am J Physiol Cell Physiol 2010;299:C750–9. 31. Sequeira-Lopez ML, Weatherford ET, Borges GR, Monteagudo MC, Pentz ES, Harfe BD, et al. The microRNA-processing enzyme dicer maintains juxtaglomerular cells. J Am Soc Nephrol 2010;21:460–7. 32. Vio CP, Jeanneret VA. Local induction of angiotensin-converting enzyme in the kidney as a mechanism of progressive renal diseases. Kidney Int Suppl 2003;86:S57–63. 33. Wolf G. The renin–angiotensin system and progression of renal dis- eases. Hamburg: Karger; 2002. 34. Zhao D, Seth DM, Navar LG. Enhanced distal nephron sodium reabsorption in chronic angiotensin II-Infused mice. Hypertension 2009;54:120–6. 35. Zhuo JL, Imig JD, Hammond TG, Orengo S, Benes E, Navar LG. Ang II accumulation in rat renal endosomes during Ang II-induced hypertension: role of AT(1) receptor. Hypertension 2002;39:116–21.