You are on page 1of 18

P1: sbs/mkv P2: sbs/vks QC: sbs

December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

Annu. Rev. Physiol. 1997. 59:395–412


Copyright c 1997 by Annual Reviews Inc. All rights reserved

BIOLOGICAL FUNCTIONS OF
ANGIOTENSIN AND ITS RECEPTORS
T. Matsusaka and I. Ichikawa
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

Vanderbilt University Medical Center, Nashville, Tennessee 37232-2584

KEY WORDS: aldosterone, JG cells, extracellular matrix, heart failure, renal failure

ABSTRACT
Angiotensin receptors are present in a number of organs and systems including
heart, kidney, gonad, and placenta; pituitary and adrenal glands; the peripheral
vessels, and the central nervous system. This octapeptide exerts diverse effects
that include induction of cell hypertrophy and/or hyperplasia and a stimulation
of hormone synthesis and ion transport in the heart, kidney, and adrenal, pri-
marily through type 1 (AT1) receptors. In the kidney, several heterogeneous cell
populations—endothelial, epithelial, and vascular—carry AT1 receptors. Some
studies suggest that AT2 receptors are also functional, but the cell type carrying
this receptor and the nature of its specific function have not been fully eluci-
dated. Although studies indicate that AT1 receptors are affected in response to
physiological and pathophysiological manipulations, the functional significance
of these modulations remains largely uncertain. Nevertheless, recent human ge-
netic studies indicate that polymorphisms in AT1 receptors, as well as in other
angiotensin-related genes, have significant impact on organ remodeling processes
of the heart and the kidney.

A Wide Variety of Renal Cells Have Angiotensin Receptors


A decade ago, Mendelsohn et al examined localization of angiotensin (Ang) II
receptors in the adult rat kidney by using 125 I-labeled [Sar1 ] Ang II as a probe
(57). High-density Ang II binding was observed in the glomerulus and in the
inner zone of the outer medulla, outlining vasa recta bundles. The glomerular
binding is consistent with mesangial localization. A recent electron micro-
scope autoradiography study revealed that the medullary binding is localized
exclusively to type 1 renomedullary interstitial cells (99). Studies have failed
to demonstrate a similar signal in the inner stripe of the outer medulla of mice
(37). Moderate to low binding occurred in tubules of the cortex and within the

395
0066-4278/97/0315-0395$08.00
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

396 MATSUSAKA & ICHIKAWA

inter bundle area of the inner zone of the outer medulla (57). An Ang type
1 receptor (AT1) antagonist, losartan, almost completely inhibits the binding
of 125 -I[Sar1 , Ile8 ] Ang II in adult rat kidneys, whereas type 2 receptor (AT2)
antagonist PD123177 has little effect, which indicates that most of the Ang II
receptors in adult kidneys are AT1 receptors, with a limited number of AT2
receptors (81).
Mice and rats, but not humans, have two subtypes of AT1, namely, AT1A and
AT1B. These receptor subtypes are likely the result of gene duplication, which
occurred in rodents prior to the phylogenic divergence between the mouse and
the rat (96). Gasc et al studied AT1A and AT1B expression in rats by in situ
hybridization using specific probes for the 30 noncoding region of their mRNAs
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

(28). AT1A mRNA was observed in mesangial cells, juxtaglomerular (JG)


cells, epithelial cells of the proximal tubule, in the vasa recta, and occasionally
in a few interstitial cells of the kidney cortex. AT1B mRNA was expressed
at a lower level in mesangial cells, in JG cells, and in smooth muscle cells
of the renal pelvis. It should be noted, however, that a negative result does
not necessarily mean absence of the receptor but instead may reflect technical
limitations in demonstrating expression.
In Vivo Function of AT1 Receptors on JG Cells
Needs Further Clarification
The highly expressed AT1 receptor on JG cells is closely associated with renin
expression (43, 55a), consistent with the possibility that Ang II directly sup-
presses the release and production of renin (i.e. via short-loop feedback). In-
deed, it was shown that Ang II suppresses renin release from rat kidney slices
(61) and isolated perfused JG apparatus (54). Moreover, losartan was shown to
induce renin mRNA in freshly isolated renin-producing cells cultured in vitro
(88). A sub-pressor dose of Ang II infusion partially attenuated the renal renin
mRNA stimulated by treatment with enalapril, an Ang I-converting enzyme
(ACE) inhibitor (ACEi) (40). However, the in vivo significance of this short-
loop feedback mechanism has not been established because the release and pro-
duction of renin can be regulated primarily by baroreceptor and macula densa
mechanisms (Figure 1). In fact, in renovascular hypertension or during dietary
sodium restriction, renin release and production continues to be stimulated de-
spite a marked increase in Ang II. In this connection, marked hyperplasia and
hypertrophy of renin-producing cells have been noted in angiotensinogen null-
mutant mice (64). The effect of removing Ang II’s inhibition on renin-producing
cells cannot be distinguished from systemic effects such as low blood pressure,
which characterize the angiotensinogen null-mutant mice. Thus the biologi-
cal significance of short-loop feedback and the functional role of the intense
expression of AT1 on the renin production by JG cells remain unclear (55a).
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 397


Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

Figure 1 Regulatory mechanism of renin production and release. AT1 receptor is by far the
most intensely expressed in renin-producing cells. The renin release and production are regulated
primarily by baroreceptor and macula densa mechanisms. The significance of the direct feedback
regulation by Ang II has not been confirmed in vivo. It remains to be determined why renin-
producing cells need to express high-density AT1.

The Pathogenic Role of Angiotensin in Progressive Renal


Disease Is Multifactorial
In addition to regulating systemic and local hemodynamics, Ang II may be
involved in the accumulation of extracellular matrix (ECM), the hallmark of
glomerulosclerosis and interstitial fibrosis, which are ubiquitous in progressive
renal diseases. During advancement of glomerulosclerosis, both the quantity
and the quality of ECM change. Administration of a dose of AT1 receptor
antagonist (which did not lower blood pressure), to stroke-prone spontaneous
hypertensive rats and deoxycorticosterone acetate-salt hypertensive rats, re-
markably attenuated the typical morphological pattern of nephrosclerosis and
the elevation of renal mRNA of TGF-β, type I, III, and IV collagens, and
fibronectin, which occur in untreated controls (46). These observations sug-
gest that Ang II can enhance gene expression of ECM components by a blood
pressure-independent action. Indeed, in in vitro experiments, Ang II was found
to stimulate synthesis of mRNA and protein of type I collagen, fibronectin and
biglycan in mesangial cells via the AT1 receptor, and this was shown to be me-
diated, at least in part, by secretion of TGF-β (42). Lee et al have shown that in
the rat remnant kidney, angiotensinogen mRNA as well as TGF-β 1, fibronectin
and laminin B1 transcripts are abnormally expressed in endothelial cells, par-
ticularly in dilated glomerular capillaries prior to the development of sclerosis
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

398 MATSUSAKA & ICHIKAWA

(48). The AT1 receptor antagonist inhibited this gene expression. These find-
ings suggest that Ang II is locally up-regulated in the injured endothelium and
may trigger a cascade involving TGF-β and ECM protein synthesis.
Other studies suggest that Ang II is also involved in interstitial fibrosis. Klahr
and colleagues observed that ACEi and AT1 antagonist significantly blunted
the increase of TGF-β1 and collagen IV mRNAs in experimentally obstructed
rat kidneys, in which a marked interstitial fibrosis would otherwise develop
(45). Johnson et al showed that a chronic infusion of Ang II led to develop-
ment of tubulointerstitial injury with type IV collagen deposition (41). In vitro
studies by Wolf et al demonstrated that Ang II can induce type IV collagen and
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

TGF-β in MCT cells, a proximal tubular cell line (94). It was further shown that
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

interstitial cells express a high level of AT1 receptors (28, 99). In fibroblasts of
cardiac origin, Ang II was shown to induce hypertrophy and hyperplasia along
with the expression of TGF-β1 mRNA (77). Ang II may, therefore, stimulate
the synthesis of ECM proteins in the renal interstitium directly, or via induction
of growth factors.
Ang II may also modulate degradation of ECM. The importance of degrada-
tion mechanisms in the advancement of glomerulosclerosis is supported by the
experimental demonstration that attenuation of ECM accumulation could lead
to a reversal of established glomerulosclerosis (35). ECM proteins are degraded
by a number of proteinases, among which matrix metalloproteinases (MMPs)
play major roles. MMPs are secreted in inactive form, and plasmin is required
for their conversion to active enzymes. Plasmin is generated from plasminogen
by the action of plasminogen activators (PAs), which are also activated by plas-
min, thereby forming a positive feedback loop. This system, which also plays
a central role in fibrinolysis, is tightly regulated by PA inhibitors (PAIs). The
importance of PAI-1 in ECM degradation in the kidney was shown by a study
in which administration of PAI-1-neutralizing antibodies to cultured mesan-
gial cells resulted in a several-fold increase in ECM degradation (6). It has
been shown that Ang II increases PAI-1 mRNA in cultured vascular smooth
muscle cells and vascular endothelial cells (26). In vivo infusion of Ang II
in humans resulted in increased circulating levels of PAI-1 (75). Moreover,
ACEi administration lowers plasma PAI-1 levels in humans (95). Thus Ang II
has a capacity to attenuate fibrinolytic activity and ECM degradation via the
PAI-1-PA-plasmin cascade. Overall, Ang II has the potential to attenuate ECM
degradation via induction of PAI-1.

Angiotensin Is Required for Kidney Ontogeny


AT1 is expressed in maturing glomeruli of the mouse fetus at day 16 (out of 20)
of gestation (44). Ang II exerts a potent mitogenic action on mesangial cells
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 399

isolated from the fetus but not on cells derived from the adult (72). The pos-
sibility thus exists that Ang II may participate in the proliferation of glomeru-
lar mesangial cells during nephrogenesis. This hypothesis has been proven
in angiotensinogen null-mutant mice, which had a modest delay in glomeru-
lar maturation (64). Because glomerular maturity was comparable between
homozygous-mutant neonates produced by homozygous versus heterozygous
mothers, Ang II in the maternal circulation appears to have little impact on fetal
glomerular maturation. The delay of glomerular maturity was most prominent
at 1 week of age, and there was no difference in maturity at 3 weeks of age,
indicating that Ang II is necessary for full glomerular maturation in the early
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

postnatal period. Gomez and colleagues found that administration of an AT1


Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

antagonist to newborn rats induced an arrest in nephrovascular maturation and


attenuated the increase in glomerular size and number (89) and thus proposed
that Ang II is a renal vascular growth factor. In angiotensinogen null-mutant
mice, the glomerular number was found to be unaffected, however (64).
AT2 is intensely expressed in mesenchymal cells in the fetal mouse kidney
(44). Moreover, the site of AT2 expression closely overlaps with that of a
group of cells undergoing apoptosis following nephrogenesis. In this regard,
it has been shown that AT2 exerts antiproliferative and apoptotic functions in
some experimental settings (62). Therefore, the possibility was raised that AT2
receptors might be involved in nephrogenesis by eliminating undifferentiated
mesenchymal cells surrounding the stalk of the ureter epithelium (44). Gomez
and colleagues reported that an AT2 antagonist increased glomerulogenesis
in organ culture (65). However, AT2 null-mutant mice were found to lack a
comparable abnormality in glomerular morphology (34) (Table 1). Although
the fate of fetal mesenchymal cells in the AT2 null-mutant mice has not been
examined in full, the AT2 receptor does not appear to be essential for normal
nephrogenesis in vivo.

Significance of Local Production and Action of Angiotensin


in the Heart Awaits In Vivo Verification
In 1979, an ACEi, captopril, was found to have a remarkable inhibitory effect
on the myocardial hypertrophy of spontaneous hypertensive rats (SHR), which
cannot be achieved by normalization of blood pressure with hydralazine (5).
This early finding echoed a number of in vitro studies showing trophic effects
of Ang II on cells, including cardiomyocytes and vascular smooth muscle cells,
and led to the notion that Ang II is a cardiovascular growth factor. The anti-
cardiovascular-hypertrophic effect of ACEi or AT1 antagonist was also observed
in rats with abdominal aorta constriction (51). Moreover, Ang II infusion in
rats was shown to induce cardiac hypertrophy, which can be prevented by
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

400 MATSUSAKA & ICHIKAWA

Table 1 Phenotypes of homozygous


angiotensinogen null-mutant mice

Hypotension
Growth failure
Glomerulus
Delayed maturation
Resistance to glomerular sclerosis
Impaired size autoregulation
Tubulointerstitium
Interstitial fibrosis
Tubular dilatation
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

Hypoplastic papilla
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

Reduced Na/K ATPase


Renal vasculature
JGA hypertrophy
Arterial wall thickening
Adrenal
Hyperkalemia

an AT1 antagonist, but not by hydralazine (24). Although it is difficult to


distinguish systemic versus local effects of Ang II in vivo, these studies suggest
that inhibition of Ang II may have a salutary effect on cardiac hypertrophy to a
degree greater than expected from its systemic blood pressure–lowering effect.
In addition to preventing cardiac hypertrophy accompanied by pressure over-
load, AT1 antagonists and ACEi preserve cardiac function after myocardial in-
farction both in rats and in humans (68). The salutary effect of ACEi may reflect
the capacity of ACEi to reduce not only afterload but also preload; both effects
becoming evident shortly after the start of ACEi treatment. One potentially
important mechanism through which Ang II may be involved in the process
of heart failure is the regulation of ECM turnover. Pressure overload causes
not only cardiomyocyte hypertrophy but also reactive fibrosis (93). However,
a disproportional accumulation of collagen leads to increased ventricular stiff-
ness and hence diastolic dysfunction (93). As in the kidney, Ang II was shown
to promote fibrosis in the heart. Sadoshima et al showed that Ang II induces a
mitogenic response and up-regulation of TGF-β1 mRNA in cultured neonatal
cardiac fibroblasts via AT1 receptor (77). Chronic infusion of Ang II into rats
induces fibrosis and increases the content of type I collagen and fibronectin
mRNA in the heart (21). It was shown that treatment with ACEi attenuates the
reactive fibrosis in rats following myocardial infarction (58).
In these settings, Ang II is thought to be generated locally in the heart be-
cause a discrepancy exists between the concentration of plasma Ang II and that
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 401

required for biological responses (25). Although it is controversial whether


cardiovascular tissues can synthesize functionally significant amounts of renin
(92, 25), the cardiovascular tissue can uptake circulating renin or utilize cir-
culating Ang I (25). In vitro experiments showed that stretch induces Ang II
release from cultured neonatal cardiomyocytes and that AT1 antagonist atten-
uates the expression of c-fos, which is activated by the stretch, thus indicating
that Ang II can become an autocrine mediator for stretch-induced hypertrophy
(78). A possible mechanism of Ang II up-regulation in cardiovascular diseases
is an overproduction of ACE in the lesion. In this regard, ACE has been shown
to be produced in an abnormally high amount in the reactive fibrous heart tissue
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

of rats undergoing renovascular hypertension, myocardial infarction, or pericar-


Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

diotomy (15, 82). It remains uncertain, however, whether the locally activated
ACE is sufficient to account for the cardiovascular lesion.
Stimulatory Effect of Angiotensin on Aldosterone Occurs
Through AT1 Receptors and Involves Activation
of Synthesis and Cell Proliferation
Ang II is the major regulator of aldosterone synthesis, and this function is me-
diated by the AT1 receptor (16). Consistent with this function, AT1 is intensely
expressed in the adrenal gland. Gasc et al showed AT1A and AT1B expression
in the rat adrenal by in situ hybridization (28). Zona glomerulosa cells express
a high level of AT1A and AT1B mRNA, with AT1B mRNA expression being
higher than that of At1a.
Ang II is thought to stimulate the secretion of aldosterone in at least three
ways: (a) induction of enzymes that are required for aldosterone synthesis,
(b) stimulation of the proliferation of adrenocortical cells, (c) induction of AT1
receptors (Figure 2). Cholesterol side chain cleavage (P450scc) and aldosterone
synthase (P450aldo) catalyze the early and late rate-limiting steps, respectively.
ACEi was shown to almost completely attenuate the enzyme activity, mRNA,
and protein of both P450scc and P450aldo, which are increased during dietary
sodium restriction in rats (2, 86). Holland et al, utilizing an adrenocortical cell
line (33), showed that the Ang II response element is present within the initial
425 base pairs (bp) of the mouse P450aldo gene promoter.
In dietary sodium-restricted rats, the number of glomerulosa cells is in-
creased, and enalapril completely inhibits the increase, indicating that Ang
II has a proliferative effect on zona glomerulosa cells in vivo (59). Tian et al
showed that Ang II stimulates thymidine incorporation and proliferation in
cultured bovine glomerulosa cells via the AT1 receptor (84). In rats fed a low-
sodium diet, the major site of cell replication (assessed by BrdU-incorporation)
was not in the zona glomerulosa but in the transitional zone, which is located
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

402 MATSUSAKA & ICHIKAWA


Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

Figure 2 Postulated mechanism of stimulation of aldosterone synthesis during dietary sodium


restriction. Dietary sodium restriction activates the renin-angiotensin system. Ang II stimulates
aldosterone synthesis by increasing P450scc and P450aldo, inducing cell proliferation, and up-
regulation of AT1 receptors. Sodium restriction may lead to a mild hyperkalemia, which in turn
stimulates aldosterone secretion independently of the intra-adrenal renin-angiotensin system.

between the zona glomerulosa and zona fasciculata (59). This finding indi-
cated that under sodium-deficient conditions, the major proliferating adrenal
cells are not the glomerulosa cells, which are already differentiated, but stem
cells, which are localized in the transitional zone.
Dietary sodium deficiency is the most common stimulus for increasing aldos-
terone secretion under physiological conditions. As discussed above, blockade
of Ang II markedly attenuates aldosterone secretion, as well as expression of
P450scc and P450aldo, and glomerulosa cell proliferation during sodium re-
striction. Ang II is, therefore, a potent mediator of aldosterone secretion during
sodium depletion, although it is not known whether the increase in aldosterone
is entirely dependent on Ang II. Several early observations discounted this
notion. For example, plasma renin and Ang II levels often dissociate from
aldosterone secretion (9). Nephrectomized rats respond to sodium depletion
with a significant rise in aldosterone secretion (66). However, more recently
these phenomena have been attributed to either one or a combination of the
two mechanisms involving Ang II; that is, activation of the intra-adrenal renin-
angiotensin system (23, 71) and/or alteration of the adrenal sensitivity to Ang
II (1).
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 403

Potassium’s Stimulatory Effect on Aldosterone Is Believed


to Involve Activation of Adrenal Renin-Angiotensin
and/or AT1 Receptor Up-Regulation
Twenty-five years ago, potassium was proposed to be an important mediator
for the zona glomerulosa response to sodium depletion (11). Indeed, extracel-
lular potassium concentration is a major modulator of aldosterone synthesis.
Nevertheless, studies have shown that the presence of Ang II is required for a
full steroidgenic effect of potassium (Figure 2). Thus treatment of rats with
captopril inhibits the potassium-induced increase in aldosterone and P450aldo,
but not P450scc (86, 63). Moreover, although potassium supplement decreases
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

plasma renin activity, it increases adrenal renin (63). Therefore, it has been pro-
posed that locally generated Ang II in the adrenal glomerulosa cells mediates
the aldosterone secretion induced by potassium. Local Ang II is shown to be
necessary not only for potassium-stimulated aldosterone secretion but also for
basal ACTH-stimulated and sodium depletion-induced aldosterone secretion
(29, 66).

Data on Renal AT1 Receptor Regulation Are Inconsistent,


and Its Biological Significance Remains Uncertain
In classic terms, renin was thought to be the only rate-limiting factor for the
renin-angiotensin system (RAS). Recently, it was shown that angiotensinogen,
as well as ACE, is a rate-limiting factor. The importance of the number of Ang II
receptors in regulating the activity of RAS has not been established. However,
recent gene targeting studies have shown that the mice heterozygous for the
AT1A receptor gene null mutation have blood pressure that is intermediate
between wild-type and homozygous-mutant mice (37, 55a), suggesting that
receptor regulation is functionally important.
Radioligand receptor-binding studies in rats showed that dietary sodium re-
striction leads to a decrease in glomerular Ang II receptor binding (7, 17).
Because this down-regulation by sodium restriction can be aborted by a con-
current administration of an ACEi (79), it is speculated that the down-regulation
is driven by the high plasma Ang II prevailing during sodium-restricted condi-
tions. Indeed, both acute and chronic infusion of Ang II was shown to cause
a significant reduction in Ang II binding sites in the rat glomerulus (8, 17).
Because the same stimulus does not alter cardiac Ang II receptor binding (22)
and increases adrenal receptor binding (see below), this down-regulation ap-
pears unique to the glomerulus. In concert with these in vivo observations,
several-day exposure of glomerular mesangial cells to Ang II down-regulated
Ang II binding sites by 23% (17). Because the in vivo down-regulation of
Ang II binding sites could not be reversed experimentally by dissociation of
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

404 MATSUSAKA & ICHIKAWA

bound endogenous Ang II, the Ang II receptor down-regulation was attributed
to a phenomenon other than increased prior occupancy of the receptor by en-
dogenous Ang II (8).
Although it is widely believed that the Ang II receptor in adult rat or hu-
man kidneys is AT1, it remains inconclusive whether the down-regulation of
glomerular Ang II binding sites is entirely reflective of AT1 because there is cur-
rently no perfect receptor subtype-specific ligand available for study. Losartan,
the most widely used synthetic AT1 receptor antagonist, binds to rat glomeruli
and human mesangial cells; however, the number of sites and the dissociation
constant value are greater than those for Ang II itself (17). These findings sug-
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

gest that losartan binds not only to AT1 but also to non-Ang II receptor(s). This
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

explains why chronic Ang II administration down-regulates Ang II-binding


sites but not those of losartan (17).
The changes in renal Ang II receptor-binding characteristics in response to
dietary salt restriction or Ang II infusion are not necessarily accompanied by
parallel changes in receptor mRNA. Indeed, the studies are highly conflicting.
In vitro exposure of mesangial cells to Ang II for up to 24 h does not affect AT1
mRNA expression (17). However, Makita et al reported a down-regulation of
AT1 mRNA in rat mesangial cells exposed to Ang II (55). In rabbits, a sodium-
deficient diet for 4 weeks led to a 43% increase in AT1 mRNA in glomeruli
(18). These diverse observations on the level of AT1 gene transcripts may be
attributed to (a) a technical difficulty in quantitating AT1 mRNA because the
tissue level of mRNA is uniformly low, and thus commonly requires amplifi-
cation of the message, which in itself may become a potential source of error;
(b) heterogeneity of renal cells in which AT1 mRNA may be differentially reg-
ulated; and/or (c) activation of multiple factors during salt restriction, including
reduced renal perfusion pressure, local Ang II release, and plasma aldosterone
(90), which have various potentials to modulate AT1 at mRNA and/or protein
levels.
In obstructed kidneys of the rat, renal AT1 mRNA began to fall appreciably
within 6 h of the onset of obstruction (69). Similarly, a rat model of cyclosporine
A toxicity characterized by hypertension and progressive renal scarring has been
shown to down-regulate AT1 mRNA (87). In all these conditions, ACEi or AT1
antagonists can exert renal tissue-protective effects. Teleologically, therefore,
down-regulation of Ang receptors may be viewed as an organ-specific homeo-
static mechanism to minimize the unwanted actions of Ang II.
In models of hypertension, the status of renal Ang II receptors is less con-
sistent. An elevated AT1 receptor density in the renal outer medulla of a rat
hypertensive strain of SHR was noted by a ligand-binding assay (80). By
quantitative autoradiography, however, Correa et al (19) reported that, whereas
Ang II receptor density is significantly higher in 1 week old SHR rats than in
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 405

age-matched Wistar-Kyoto (WKY) controls, the density at the glomeruli and


the outer stripe of outer medulla decreases to the level below that of adult WKY
controls. In contrast, in a rat model of renovascular hypertension, AT1 mRNA
was found to be decreased in clipped kidneys (i.e. kidneys with a high local
Ang II level), and unchanged in contralateral kidneys (22). In a similar study,
AT1A mRNA was lower in both kidneys of models of hypertensive rats (30).
However, in another study by Modrall et al (60), although AT1 mRNA was
down-regulated shortly after the onset of hypertension, it increased seven- to
eightfold in both clipped and unclipped kidneys when a marked hypertension
was fully developed.
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

Overall, as in physiological conditions, the current literature is somewhat


Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

confusing with regard to the status of renal Ang II receptors in pathophysi-


ological conditions. Several factors may contribute to this confusion. Most
fundamentally, it has been neither established nor tested whether the alterations
in Ang II receptor density reported for various experimental conditions, either
physiological or pathophysiological, are of any biological significance.
Cardiac AT1 Receptors May Be Up-Regulated During
the Process of Hypertrophy
Several studies indicate that when the heart is undergoing hypertrophy, Ang
receptors, particularly AT1, are up-regulated. Thus during the neonatal period,
rat cardiac AT1A mRNA and the number of Ang II-binding sites are elevated
two- to fourfold over adult levels (83). Moreover, the cardiac hypertrophy in-
duced in rats with 2-kidney 1-clip Goldblatt hypertension is associated with
increased cardiac AT1A mRNA and Ang II-binding sites (83). Likewise, in
spontaneously hypertensive rats, increased AT1B mRNA was shown in the car-
diac ventricle (38). Following myocardial infarction in rats, Meggs et al (56)
documented cellular hypertrophy in the non-infarcted myocardium. In these
cells, a nearly twofold increase in the density of Ang II was also noted. In
contrast to these observations, Lopez et al (53) found decreased AT1 binding
capacity and decreased AT1/AT2 ratio in the hypertrophied left ventricle follow-
ing a partial aortic banding. Thus it is likely that, in addition to physical forces,
other factors including the level of Ang II, impact on the status of myocardial
Ang II receptors in different in vivo models of cardiac hypertrophy.
ECF Volume Depletion and Angiotensin Up-Regulation
of Adrenal AT1
Aguilera et al first documented that dietary salt restriction in rats increases the
sensitivity of the adrenal gland to release aldosterone in response to Ang II.
This adaptation was accompanied by increased binding of Ang II to the adrenal
(3). Subsequently, a greater than fourfold increase in Ang II receptor-binding
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

406 MATSUSAKA & ICHIKAWA

sites was documented in the adrenal zona glomerulosa of rats placed on a salt-
restricted diet (12). These changes in Ang II binding capacity are accompanied
by parallel changes in the transcript of the AT1 gene, the predominant Ang
II receptor in the adrenal gland. Thus dietary salt restriction increases adrenal
AT1A and AT1B mRNA by 60 and 110%, respectively (52). Of note, the Ang II
receptor number in the rat adrenal gland was shown to be up-regulated by Ang
II infusion, whereas it was down-regulated by administration of an ACEi (74).
Moreover, an infusion of Ang II in rats in vivo up-regulates AT1 mRNA without
altering its expression in other organs, and losartan administration reduces AT1
mRNA (36). Kitami et al further showed that AT1B mRNA in the rat adrenal
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

was down-regulated by an AT1 antagonist (47). The up-regulation of Ang II


Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

receptor in the adrenal during salt restriction is, therefore, attributed to high
Ang II levels.
In contrast to these results from the rat, where Ang II leads to up-regulation
of its own receptors on glomerulosa cells and increases steroidgenic responsive-
ness, addition of this peptide to cultured bovine fasciculata and glomerulosa
cells was shown to result in both down-regulation and desensitization (67).
Moreover, in monkeys, sodium restriction causes reduction in Ang II recep-
tors in the zona glomerulosa (70). Thus the response of adrenal AT1 recep-
tors to salt restriction and Ang II appears to be different between rodents and
bovine/primate groups. One notion is that a gene duplication occurred in AT1
to produce two subtypes (i.e. AT1A and AT1B) in rodents before the species
divergence between these two groups (96) and that AT1A and AT1B in rodents
are regulated differently. Thus it appears that the different responses of the two
species groups reflect a phylogenic divergence that occurred in the regulatory
regions of the AT1 receptor genes.
Certain Gene Polymorphisms in the Renin Angiotensin
System May Significantly Impact Organ Remodeling
Processes in the Kidney and Heart
Rigat et al identified a polymorphism within intron 16 of the human ACE gene,
consisting of the presence or absence of an Alu sequence 287 bp fragment (76).
The presence of this fragment defines the insertion or the I allele, whereas its
absence defines the deletion or the D allele. This ACE genotype was thus clas-
sified as II, ID, or DD. The ACE polymorphism has a significant association
not only with the plasma level of ACE, but also with intracellular ACE as mea-
sured by ACE activity in T lymphocytes (20). Cambien et al proposed that the
ID polymorphism is a marker for increased risk of myocardial infarction (MI)
(13). Adult males with the DD genotype were found to have one-and–one-third
times the risk for MI when compared with those with other genotypes. Similar
findings have been reported by several subsequent studies (73), whereas some
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 407

showed variant results (10, 50). The DD genotype has also been associated
with idiopathic dilated cardiomyopathy and idiopathic ischemic cardiomyopa-
thy (73). Some studies (50) refute these observations, however.
Tiret et al found a synergistic effect of ACE ID polymorphism and angiotensin
II type 1 receptor gene polymorphism on the risk for MI (85). The polymor-
phism studied by Tiret consists of a nucleic acid substitution of A to C at position
1166 (A1166C) in the 30 untranslated region of an AT1 exon (85). A1166C ho-
mozygosity, which is, by itself, not predictive of MI, is, in a synergistic fashion
with the ACE gene DD genotype, associated with increased risk of MI. Because
this A to C substitution does not alter a potential mRNA polyadenylation or
Access provided by University of Pennsylvania on 03/03/15. For personal use only.

destabilization signal, this polymorphic locus is, as the ACE ID locus, believed
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

to be a marker for an adjacent functional variant locus.


Genetic variations in several RAS genes have been evaluated for potential as-
sociation with hypertension. Although the ACE, renin, and AT1 receptor genes
have heretofore failed to be linked or associated with hypertension, Jeunemaitre
et al showed that variation in the angiotensinogen gene is linked to hyperten-
sion (39). The mutation in which threonine (T) is substituted for methionine
(M) at residue 235 was significantly more frequent in hypertensives than in
controls in two genetically distinct human populations in Salt Lake City and
Paris. The prevalence of the T variant was 36% in control, 47% in unrelated
index cases, and 51% in severely hypertensive individuals (39). Moreover, T
allele homozygotes had ≈20% higher levels of plasma angiotensinogen.
The significance of ACE gene polymorphism varies among different organs
or tissues depending on the local availability of Ang I for conversion to Ang II
by ACE. It has been shown that the Ang II/Ang I ratio is markedly higher in the
kidney; this ratio averaged 0.3 and ∼2.0 in the plasma and kidney, respectively
(4). It is possible, therefore, that the ACE ID polymorphism may be particularly
relevant at the renal tissue level. Diabetic nephropathy and IgA nephropathy
are recognized as the two most common causes of chronic renal failure. In
both diseases, 30–50% of patients end up with chronic renal failure. Moreover,
several studies (14, 49) have shown that ACE inhibitor effectively attenuates the
progressive decline in renal function even in normotensive patients. The RAS
is, therefore, believed to be involved in the progressive process of renal tissue
destruction in these diseases. Several recent studies have shown a significant
association in IgA nephropathy between ACE gene polymorphism and the
progression to chronic renal failure. A study of 53 Japanese patients with IgA
nephropathy (98) showed that, whereas the frequency of ACE DD genotype in
patients with stable renal function (16%) was not significantly different from
that in the general Japanese population (∼10%), the ACE DD genotype in
patients with declining renal function was significantly more frequent (43%).
Other studies (32) reached similar conclusions, although variant results have
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

408 MATSUSAKA & ICHIKAWA

also been obtained (27). Similar results were obtained in patients with diabetic
nephropathy (97).
The DD genotype was shown in preliminary studies to serve as a significant
risk factor for progressive loss of renal function in polycystic kidney disease
(31) and other renal diseases (91).

ACKNOWLEDGMENTS
Our scientific endeavors are supported by the Center of Excellence in Pediatric
Nephrology and Urology and funded by National Institutes of Health grant DK
44747. Some of the studies cited in this article were also supported in part
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

by National Institutes of Health grant DK 37868. TM is the recipient of a


fellowship from the National Kidney Foundation.

Literature Cited

1. Aguilera G. 1993. Factors controlling nism of sodium modulation of glomerular


steroid biosynthesis in the zona glomeru- angiotensin receptors in the rat. J. Clin.
losa of the adrenal. J. Steroid Biochem. Invest. 74:1593–600
Mol. Biol. 45:147–51 9. Blair-West JR, Cain MD, Catt KJ, Cogh-
2. Aguilera G, Catt KJ. 1978. Regulation lan JP, Denton DA, et al. 1971. The
of aldosterone secretion by the renin- dissociation of aldosterone secretion and
angiotensin system during sodium restric- systemic renin and angiotensin II lev-
tion in rats. Proc. Natl. Acad. Sci. USA els during the correction of sodium de-
75:4057–61 ficiency. Acta Endocrinol. 66:229–47
3. Aguilera G, Hauger RL, Catt KJ. 1978. 10. Bohn M, Berge KE, Bakken A, Erikssen
Control of aldosterone secretion during J, Berg K. 1993. Insertion/deletion (I/D)
sodium restriction: adrenal receptor reg- polymorphism at the locus for angiotensin
ulation and increased adrenal sensitivity I-converting enzyme and myocardial in-
to angiotensin II. Proc. Natl. Acad. Sci. farction. Clin. Genet. 44:298–301
USA 75:975–79 11. Boyd JE, Palmore WP, Mulrow PJ. 1971.
4. Allan DR, McKnight JA, Kifor I, Co- Role of potassium in the control of aldos-
letti CM, Hollenberg NK. 1994. Convert- terone secretion in the rat. Endocrinology
ing enzyme inhibition and renal tissue 88:556–65
angiotensin II in the rat. Hypertension 12. Bradshaw B, Moore TJ. 1988. Abnormal
24:516–22 regulation of adrenal angiotensin II recep-
5. Antonaccio MJ, Rubin B, Horovitz ZP, tors in spontaneously hypertensive rats.
Laffan RJ, Goldberg ME, et al. 1979. Ef- Hypertension 11:49–54
fect of chronic treatment with captopril 13. Cambien F, Poirier O, Lecerf L, Evans A,
(SQ 14,225), an orally active inhibitor of Cambou JP, et al. 1992. Deletion poly-
angiotensin I-converting enzyme in spon- morphism in the gene for angiotensin-
taneously hypertensive rats. Jpn. Pharma- converting enzyme is a potent risk fac-
col. 29:285–94 tor for myocardial infarction. Nature
6. Baricos WH, Cortez SL, el-Dahr SS, 359:641–44
Schnaper HW. 1995. ECM degradation 14. Cattran DC, Greenwood C, Ritchie S.
by cultured human mesangial cells is me- 1994. Long-term benefits of angiotensin-
diated by a PA/plasmin/MMP-2 cascade. converting enzyme inhibitor therapy in
Kidney Int. 47:1039–47 patients with severe immunoglobulin A
7. Beaufils M, Sraer J, Lepreux C, Ar- nephropathy: a comparison to patients
daillou R. 1976. Angiotensin II binding receiving treatment with other antihyper-
to renal glomeruli from sodium-loaded tensive agents and to patients receiving no
and sodium-depleted rats. Am. J. Physiol. therapy. Am. J. Kidney Dis. 23:247–54
230:1187–93 15. Challah M, Nicoletti A, Arnal JF, Philippe
8. Bellucci A, Wilkes BM. 1984. Mecha- M, Laboulandine I, et al. 1995. Cardiac
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 409

angiotensin converting enzyme overpro- of IgA nephropathy. Lancet 346:571


duction indicates interstitial activation in 28. Gasc JM, Shanmugam S, Sibony M, Cor-
renovascular hypertension. Cardiovasc. vol P. 1994. Tissue-specific expression of
Res. 30:231–39 type 1 angiotensin II receptor subtypes.
16. Chang RS, Lotti VJ. 1990. Two distinct An in situ hybridization study. Hyperten-
angiotensin II receptor binding sites in rat sion 24:531–37
adrenal revealed new by selective nonpep- 29. Gupta P, Franco-Saenz R, Mulrow PJ.
tide ligands. Mol. Pharmacol. 37:347–51 1995. Locally generated angiotensin II
17. Chansel D, Bizet T, Vandermeersch S, in the adrenal gland regulates basal,
Pham P, Levy B, Ardaillou R. 1994. corticotropin-, and potassium-stimulated
Differential regulation of angiotensin II aldosterone secretion. Hypertension 25:
and losartan binding sites in glomeruli 443–48
and mesangial cells. Am. J. Physiol. 30. Haefliger JA, Bergonzelli G, Waeber G,
266:F384–93 Aubert JF, Nussberger J, et al. 1995. Renin
18. Cheng HF, Becker BN, Burns KD, Har- and angiotensin II receptor gene expres-
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

ris RC. 1995. Angiotensin II upregulates sion in kidneys of renal hypertensive rats.
type-1 angiotensin II receptors in renal Hypertension 26:733–37
proximal tubule. J. Clin. Invest. 95:2012– 31. Harden PN, Geddes C, Boulton Jones M,
19 Briggs JD, Junor JR, et al. 1994. Polymor-
19. Correa FM, Viswanathan M, Ciuffo GM, phism in the ACE gene in patients with
Tsutsumi K, Saavedra JM. 1995. Kid- renal disease. J. Am. Soc. Nephrol. 5:330
ney angiotensin II receptors and convert- (Abstr.)
ing enzyme in neonatal and adult Wistar- 32. Harden PN, Geddes C, Rowe PA, McIlroy
Kyoto and spontaneously hypertensive JH, Boulton Jones M, et al. 1995. Poly-
rats. Peptides 16:19–24 morphisms in angiotensin-converting-
20. Costerousse O, Allegrini J, Lopez M, enzyme gene and progression of IgA
Alhenc-Gelas F. 1993. Angiotensin I nephropathy. Lancet 345:1540–42
converting enzyme in human circula- 33. Holland OB, Carr BC, Braisier AR. 1995.
ting mononuclear cells: genetic polymor- Aldosterone synthase gene regulation by
phism of expression in T lymphocytes. angiotensin. Endocr. Res. 21:455–62
Biochem J. 290:33–40 34. Ichiki T, Labosky PA, Shiota C, Okuyama
21. Crawford DC, Chobanian AV, Brecher P S, Imagawa Y, et al. 1991. Effect on
1994. Angiotensin II induces fibronectin blood pressure and exploratory behaviour
expression associated with cardiac fibro- of mice lacking angiotensin II type-2 re-
sis in the rat. Circ. Res. 74:727–39 ceptor. Nature 377:748–50
22. Della Bruna R, Ries S, Himmelstoss C, 35. Ikoma M, Kawamura T, Kakinuma Y,
Kurtz A. 1995. Expression of cardiac an- Fogo A. Ichikawa I. 1991. Cause of vari-
giotensin II AT1 receptor genes in rat able therapeutic efficiency of angiotensin
hearts is regulated by steroids but not by converting enzyme inhibitor on glomeru-
angiotensin II. J. Hypertens. 13:763–69 lar lesions. Kidney Int. 40:195–202
23. Doi Y, Atarashi K, Franco-Saenz R, Mul- 36. Inagami T, Iwai N, Sasaki K, Guo DF,
row PJ. 1984. Effect of changes in sodium Furuta H, et al. 1993. Angiotensin II re-
or potassium balance, and nephrectomy ceptors: cloning and regulation. Arzneim.
on adrenal renin and aldosterone concen- Forsch. 43:226–28
trations. Hypertension 6:I124–29 37. Ito M, Oliverio MI, Mannon PJ, Best CF,
24. Dostal DE, Baker KM. 1992. Angiotensin Maeda N, et al. 1995. Regulation of blood
II stimulation of left ventricular hypertro- pressure by the type 1A angiotensin II re-
phy in adult rat heart. Mediation by the ceptor gene. Proc. Natl. Acad. Sci. USA
AT1 receptor. Am. J. Hypertens. 5:276– 92:3521–25
80 38. Iwai N, Inagami T. 1992. Regulation of
25. Dzau VJ, Re R. 1994. Tissue angiotensin the expression of the rat angiotensin II
system in cardiovascular medicine: a receptor mRNA. Biochem. Biophys. Res.
paradigm shift? Circulation 89:493–98 Commun. 182:1094–99
26. Feener EP, Northup JM, Aiello LP, King 39. Jeunemaitre X, Soubrier F, Kotelevtsev
GL. 1995. Angiotensin II induces plas- YV, Lifton RP, Williams CS, et al. 1992.
minogen activator inhibitor-1 and -2 Molecular basis of human hypertension:
expression in vascular endothelial and role of angiotensinogen. Cell 71:169–80
smooth muscle cells. J. Clin. Invest. 40. Johns DW, Peach MJ, Gomez RA, In-
95:1353–62 agami T, Carey RM. 1990. Angiotensin
27. Fukushima T, Nomura S, Kawai S, Osawa II regulates renin gene expression. Am. J.
G. 1995. ACE genotype and progression Physiol. 259:F882–87
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

410 MATSUSAKA & ICHIKAWA

41. Johnson RJ, Alpers CE, Yoshimura A, analysis. Hypertension 24:538–48


Lombardi D, Pritzl P, et al. 1992. Renal 53. Lopez JJ, Lorell BH, Ingelfinger JR,
injury from angiotensin II mediated hy- Weinberg EO, Schunkert H, et al. 1994.
pertension. J. Hypertens. 19:464–74 Distribution and function of cardiac an-
42. Kagami S, Border WA, Miller DE, Noble giotensin AT1- and AT2-receptor sub-
NA. 1994. Angiotensin II stimulates ma- types in hypertrophied rat hearts. Am. J.
trix protein synthesis through induction of Physiol. 267:H844–52
transforming growth factor-β expression 54. Lorenz JN, Weihprecht H, He X, Skøtt
in rat glomerular mesangial cells. J. Clin. O, Briggs JP, Schnermann J. 1993. Effect
Invest. 93:2431–37 of adenosine and angiotensin on macula
43. Kakinuma Y, Fogo A, Inagami T, densa-stimulated renin secretion. Am. J.
Ichikawa I. 1993. Intrarenal localization Physiol. 265:F187–94
of angiotensin II type 1 receptor mRNA 55. Makita N, Iwai N, Inagami T, Badr K.
in the rat. Kidney Int. 43:1229–35 1992. Two distinct pathways in the down-
44. Kakuchi J, Ichiki T, Kiyama S, Hogan regulation of type-1 angiotensin II re-
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

BLM, Fogo A, et al. 1995. Developmental ceptor gene in rat glomerular mesangial
expression of renal angiotensin II receptor cells. Biochem. Biophys. Res. Commun.
genes in the mouse. Kidney Int. 47:140– 185:142–46
47 55a. Matsusaka T, Nishimura H, Utsunomiya
45. Kaneto H, Morrissey J, Klahr S. 1993. In- H, Kakuchi J, Niimura F, et al. 1996.
creased expression of TGF-β1 mRNA in Chimeric mice carrying regional targeted
the obstructed kidney of rats with unilat- deletion of the angiotensin type 1A re-
eral ureteral ligation. Kidney Int. 44:313– ceptor gene: evidence against the role
21 for local angiotensin in the in vivo feed-
46. Kim S, Ohta K, Hamaguchi A, Omura back regulation of renin synthesis in jux-
T, Yukimura T, et al. 1994. Contribution taglomerular cells. J. Clin. Invest. In press
of renal angiotensin II type 1 receptor to 56. Meggs LG, Coupet J, Huang H, Cheng
gene expressions in hypertension-induced W, Li P, et al. 1993. Regulation of an-
renal injury. Kidney Int. 46:1346–58 giotensin II receptors on ventricular my-
47. Kitami Y, Okura T, Marumoto K, Waka- ocytes after myocardial infarction in rats.
miya R, Hiwada K. 1992. Differential Circ. Res. 72:1149–62
gene expression and regulation of type- 57. Mendelsohn FAO, Dunbar M, Allen A.
1 angiotensin II receptor subtypes in the Chou ST, Millan MA, et al. 1986. An-
rat. Biochem. Biophys. Res. Commun. giotensin II receptors in the kidney. Fed.
188:446–52 Proc. 45:1420–25
48. Lee LK, Meyer TW, Pollock AS, Lovett 58. Michel JB, Lattion AL, Salzmann JL, de
DH. 1995. Endothelial cell injury initi- Lourdes Cerol M, Philippe M, et al. 1988.
ates glomerular sclerosis in the rat rem- Hormonal and cardiac effects of convert-
nant kidney. J. Clin. Invest. 96:953–64 ing enzyme inhibition in rat myocardial
49. Lewis EJ, Hunsicker LG, Bain RP, Ro- infarction. Circ. Res. 62:641–50
hde RD. 1993. The effect of angiotensin- 59. Mitani F, Suzuki H, Hata J, Ogishima
converting enzyme inhibition on diabetic T, Shimada H, Ishimura Y. 1994. A
nephropathy. N. Engl. J. Med. 329:1456– novel cell layer without corticosteroid-
62 synthesizing enzymes in rat adrenal cor-
50. Lindpaintner K, Lee M, Larson MG, Rao tex: histochemical detection and pos-
S, Pfeffer MA, et al. 1996. Absence of as- sible physiological role. Endocrinology
sociation or genetic linkage between the 135:431–38
angiotensin converting-enzyme gene and 60. Modrall JG, Quinones MJ, Frankhouse
left ventricular mass. N. Engl. J. Med. JH, Hsueh WA, Weaver FA, Kedes L.
334:1023–28 1995. Upregulation of angiotensin II type
51. Linz W, Scholkens BA, Ganten D. 1989. 1 receptor gene expression in chronic
Converting enzyme inhibition specifi- renovascular hypertension. J. Surg. Res.
cally prevents the development and in- 59:135–40
duces regression of cardiac hypertrophy 61. Naftilan AJ, Oparil S. 1978. Inhibition of
in rats. Clin. Exp. Hypertens. A11:1325– renin release from rat kidney slices by the
50 angiotensins. Am. J. Physiol. 235:F62–68
52. Llorens-Cortes C, Greenberg B, Huang 62. Nakajima M, Hutchinson HG, Fujinaga
H, Corvol P. 1994. Tissular expression M, Hayashida W, Morishita R, et al.
and regulation of type 1 angiotensin II 1995. The angiotensin II type 2 (AT2) re-
receptor subtypes by quantitative reverse ceptor antagonizes the growth effects of
transcriptase-polymerase chain reaction the AT1 receptor: gain-of-function study
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

BIOLOGY OF ANGIOTENSIN 411

using gene transfer. Proc. Natl. Acad. Sci. 74. Regitz-Zagrosek V, Auch-Schwelk W,
USA 92:10663–67 Neuss M, Fleck E. 1994. Regulation of the
63. Nakamaru M, Misono KS, Naruse M, angiotensin receptor subtypes in cell cul-
Workman RJ, Inagami T. 1985. A role ture, animal models and human diseases.
for the adrenal renin-angiotensin system Eur. Heart J. 15:92–97 (Suppl. D)
in the regulation of potassium-stimulated 75. Ridker PM, Gaboury CL, Conlin PR,
aldosterone production. Endocrinology Seely EW, Wiliams GH, Vaughan DE.
117:1772–78 1993. Stimulation of plasminogen activa-
64. Niimura F, Labosky PA, Kakuchi J, tor inhibitor (PAI-1) in vivo by infusion
Okubo S, Yoshida H, Oikawa T, et al. of angiotension II. Evidence of a potential
1995. Gene targeting in mice reveals a interaction between the renin-angiotensin
requirement for angiotensin in the devel- system and fibrinolytic function. Circula-
opment and maintenance of kidney mor- tion 7:1969–73
phology and growth factor regulation. J. 76. Rigat B, Hubert C, Alhenc-Gelas F, Cam-
Clin. Invest. 96:2947–54 bien F, Corvol P, Soubrier F. 1990. An
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

65. Norwood VF, Garmey M, Gomez RA. insertion/deletion polymorphism in the


1995. Type 2 angiotensin receptor (AT-2) angiotensin I-converting enzyme gene
inhibits in vitro glomerulogenesis. J. Am. accounting for half the variance of serum
Soc. Nephrol. 6:704 (Abstr.) enzyme levels. J. Clin. Invest. 86:1343–
66. Palmore WP, Marieb NJ, Mulrow PJ. 46
1969. Stimulation of aldosterone secre- 77. Sadoshima J, Izumo S. 1993. Molec-
tion by sodium depletion in nephrec- ular characterization of angiotensin II-
tomized rats. Endocrinology 84:1342–51 induced hypertrophy of cardiac myocytes
67. Penhoat A, Jaillard C, Crozat A, Saez J. and hyperplasia of cardiac fibroblasts.
1988. Regulation of angiotensin II recep- Critical role of the AT1 receptor subtype.
tors and steroidogenic responsiveness in Circ. Res. 73:413–23
cultured bovine fasciculata and glomeru- 78. Sadoshima J, Xu Y, Slayter, HS, Izumo S.
losa adrenal cells. Eur. J. Biochem. 1993. Autocrine release of angiotensin II
172:247–54 mediates stretch-induced hypertrophy of
68. Pfeffer JM, Fischer TA, Pfeffer MA. cardiac myocytes in vitro. Cell 75:977–84
1995. Angiotensin-converting enzyme in- 79. Skorecki KL, Ballerman BJ, Rennke HG,
hibition and ventricular remodeling after Brenner BM. 1983. Angiotensin II recep-
myocardial infarction. Annu. Rev. Phys- tor regulation in isolated renal glomeruli.
iol. 57:805–26 Fed. Proc. 42:3064–70
69. Pimentel JL, Montero A, Wang S, Yosipiv 80. Song K, Kurobe Y, Kanehara H, Okunishi
I, El-Dahr S, Martinez-Maldonado M. H, Wada T, et al. 1994. Quantitative local-
1995. Sequential changes in renal expres- ization of angiotensin II receptor subtypes
sion of renin-angiotensin system genes in in spontaneously hypertensive rats. Blood
acute unilateral ureteral obstruction. Kid- Press 5:21–26 (Suppl.)
ney Int. 48:1247–53 81. Song K, Zhuo J, Allen AM, Paxinos G,
70. Platia MP, Catt KJ, Hodgen GD, Aguil- Mendelsohn FAO. 1991. Angiotensin II
era G. 1986. Regulation of primate angi- receptor subtypes in rat brain and periph-
otensin II receptors during altered sodium eral tissues. Cardiology 79:45–54 (Suppl.
intake. Hypertension 8:1121–26 1)
71. Pratt RE, Bradshaw B, Moore TJ, Dzau 82. Sun Y, Cleutjens JPM, Diaz-Arias AA,
VJ. 1988. Altered expression of the Weber KT. 1994. Cardiac angiotensin
adrenal renin-angiotensin system in spon- converting enzyme and myocardial fibro-
taneously hypertensive rats (SHR). Circu- sis in the rat. Cardiovasc. Res. 28:1423–
lation 78:II59 (Abstr.) 32
72. Ray PE, Bruggerman LA, Horikoshi S, 83. Suzuki J, Matsubara H, Urakami M, Inada
Aguilera G, Klotman PE. 1994. Angi- M. 1993. Rat angiotensin II (type 1A) re-
otensin II stimulates human fetal mesan- ceptor mRNA regulation and subtype ex-
gial cell proliferation and fibronectin pression in myocardial growth and hyper-
biosynthesis by binding to AT1 receptors. trophy. Circ. Res. 73:439–47
Kidney Int. 45:177–84 84. Tian Y, Balla T, Baukal AJ, Catt KJ.
73. Raynolds MV, Bristow MR, Bush EW, 1995. Growth responses to angiotensin II
Abraham WT, Lowes BD, et al. 1993. in bovine adrenal glomerulosa cells. Am.
Angiotensin-converting enzyme DD ge- J. Physiol. 268:E135–44
notype in patients with ischemic or id- 85. Tiret L, Bonnardeaux A, Poirier O, Ri-
iopathic dilated cardiomyopathy. Lancet card S, Marques-Vidal P, et al. 1994. Syn-
342:1073–75 ergistic effects of angiotensin-converting
P1: sbs/mkv P2: sbs/vks QC: sbs
December 10, 1996 14:16 Annual Reviews MATSCHPT.TXT-CH17 AR25-17

412 MATSUSAKA & ICHIKAWA

enzyme and angiotensin II type 1 receptor dence. Circulation 89:493–98


gene polymorphisms on risk of myocar- 93. Weber KT, Brilla CG. 1993. Structural ba-
dial infarction. Lancet 344:910–12 sis for pathologic left ventricular hyper-
86. Tremblay A, Lehoux JG. 1992. Influence trophy. Clin. Cardiol. 16:II-10–14
of captopril on adrenal cytochrome P- 94. Wolf G, Mueller E, Stahl RA, Ziyadeh
450s and adrenodoxin expression in high FN. 1993. Angiotensin II-induced hyper-
potassium or low sodium intake. J. Steroid trophy of cultured murine proximal tubu-
Biochem. Mol. Biol. 41:799–808 lar cells is mediated by endogenous trans-
87. Tufro-McReddie A, Gomez RA, Norling forming growth factor-β. J. Clin. Invest.
LL, Omar AA, Moore LC, Kaskel FJ. 92:1366–72
1993. Effect of CsA on the expression 95. Wright RA, Flapan AD, Alberti KG, Lud-
of renin and angiotensin type 1 recep- lam CA, Fox KA. 1994. Effects of cap-
tor genes in the rat kidney. Kidney Int. topril therapy on endogenous fibrinolysis
43:615–22 in men with recent uncomplicated my-
88. Tufro-McReddie A, Johns DW, Geary ocardial infarction. J. Am. Coll. Cardiol.
Access provided by University of Pennsylvania on 03/03/15. For personal use only.
Annu. Rev. Physiol. 1997.59:395-412. Downloaded from www.annualreviews.org

KM, Dagli H, Everett AD, et al. 1994. 24:67–73


Angiotensin II type 1 receptor: role in 96. Yoshida H, Kakuchi J, Guo D, Furuta H,
renal growth and gene expression dur- Iwai N, et al. 1992. Analysis of the evolu-
ing normal development. Am. J. Physiol. tion of angiotensin II type 1 receptor gene
266:F911–18 in mammals (mouse, rat, bovine and hu-
89. Tufro-McReddie A, Romano LM, Har- man). Biochem. Biophys. Res. Commun.
ris JM, Ferder L, Gomes RA. 1995. An- 186:1042–49
giotensin II regulates nephrogenesis and 97. Yoshida H, Kuriyama S, Atsumi Y,
renal vascular development. Am. J. Phys- Tomonari H, Mitarai T, et al. 1996. An-
iol. 269:F110–15 giotensin I converting enzyme gene poly-
90. Ullian ME, Schelling RJ, Linas SL. 1992. morphism in non-insulin dependent dia-
Aldosterone enhances angiotensin II re- betes mellitus. Kidney Int. 50:657–64
ceptor binding and inositol phosphate re- 98. Yoshida H, Mitarai T, Kawamura T, Kita-
sponses. Hypertension 20:67–73 jima T, Miyazaki Y, et al. 1995. Role of
91. van Essen GG, Rensma PL, Apperloo AJ, the deletion of polymorphism of the an-
Scheffer H, Sluiter WJ, et al. 1995. ACE- giotensin converting enzyme gene in the
gene polymorphism predicts rate of loss progression and therapeutic responsive-
of renal function in non-diabetic chronic ness of IgA nephropathy. J. Clin. Invest.
renal failure. J. Am. Soc. Nephrol. 6:407 96:2162–69
(Abstr.) 99. Zhuo J, Alcorn D, McCausland J,
92. von Lutterotti N, Catanzaro DF, Sealey Mendelsohn FAO. 1994. Localization and
JE, Laragh JH. 1994. Renin is not syn- regulation of angiotensin II receptors in
thesized by cardiac and extrarenal vascu- renomedullary interstitial cells. Kidney
lar tissues: a review of experimental evi- Int. 46:1483–85

You might also like