Você está na página 1de 13

Pflugers Arch - Eur J Physiol (2013) 465:25–37

DOI 10.1007/s00424-012-1126-7

INVITED REVIEW

Regulation of renin secretion by renal juxtaglomerular cells


Ulla G. Friis & Kirsten Madsen & Jane Stubbe &
Pernille B. L. Hansen & Per Svenningsen & Peter Bie &
Ole Skøtt & Boye L. Jensen

Received: 18 May 2012 / Revised: 2 June 2012 / Accepted: 6 June 2012 / Published online: 26 June 2012
# Springer-Verlag 2012

Abstract A major rate-limiting step in the renin–angiotensin– far from threshold that activates L-type voltage-gated calcium
aldosterone system is the release of active renin from channels. Intracellular calcium paradoxically inhibits renin
endocrine cells (juxtaglomerular (JG) cells) in the media secretion likely through attenuated formation and enhanced
layer of the afferent glomerular arterioles. The number degradation of cAMP; by activation of chloride currents and
and distribution of JG cells vary with age and the interaction with calcineurin. Connexin 40 is necessary for
physiological level of stimulation; fetal life and chronic localization of JG cells in the vascular wall and for pressure-
stimulation by extracellular volume contraction is asso- and macula densa-dependent suppression of renin release.
ciated with recruitment of renin-producing cells. Upon
stimulation of renin release, labeled renin granules “dis- Keywords Cyclic AMP . Juxtaglomerular cells . Renin
appear;” the number of granules decrease; cell mem- secretion
brane surface area increases in single cells, and release
is quantal. Together, this indicates exocytosis as the
predominant mode of release. JG cells release few per- Introduction
cent of total renin content by physiological stimulation,
and recruitment of renin cells is preferred to recruitment Renin secretion has been the topic of comprehensive
of granules during prolonged stimulation. Several endo- reviews in 2010 [15] and 2011 [57]. In the present review,
crine and paracrine agonists, neurotransmitters, and cell focus will be on discoveries that have added significant
swelling converge on the stimulatory cyclic AMP knowledge on the cellular mechanisms that govern secretion
(cAMP) pathway. Renin secretion is attenuated in mice of active renin from renin-producing cells in the afferent
deficient in beta-adrenoceptors, prostaglandin E2–EP4 glomerular arterioles. Active renin of renal origin deter-
receptors, Gsα protein, and adenylyl cyclases 5 and 6. mines plasma and, hence, extracellular fluid renin levels.
Phosphodiesterases (PDE) 3 and 4 degrade cAMP in JG Active renin in plasma and tissues disappears after bilateral
cells, and PDE3 is inhibited by cyclic GMP (cGMP) nephrectomy [27]. A nonsecreted intracellular form and a
and couples the cGMP pathway to the cAMP pathway. secreted form of active renin have been detected in the brain,
Cyclic AMP enhances K+-current in JG cells and is but its deletion did not alter blood pressure or circulating
permissive for secretion by stabilizing membrane potential renin [134]. In human plasma, prorenin or other inactive
forms of renin constitute about 80–90 % of circulating total
This article is published as part of the special issue on the Renin- renin [82]. Large amounts of prorenin can be infused in
Angiotensin System. monkeys with no apparent physiologic effect on hemody-
U. G. Friis : K. Madsen : J. Stubbe : P. B. L. Hansen : namic and kidney parameters [71]. Thus, experimental evi-
P. Svenningsen : P. Bie : O. Skøtt : B. L. Jensen (*) dence for the existence of a physiologically significant
Department of Cardiovascular and Renal Research, conversion of prorenin to active renin in the circulation is
Institute of Molecular Medicine, University of Southern Denmark,
not available, and the renin-producing cells of the afferent
J. B. Winslowsvej 21, 3,
5000, Odense C, Denmark glomerular arteriole in the kidney are the only source of
e-mail: bljensen@health.sdu.dk active renin that respond to extracellular volume/blood
26 Pflugers Arch - Eur J Physiol (2013) 465:25–37

pressure-related stimuli. These cells are often called juxta- Mechanisms that govern JG cell differentiation
glomerular (JG) cells referring to the typical location in
physiological sodium-replete states, but renin-producing Renin promoter-driven expression of intravital dyes has
cells can extend far beyond the JG area. JG cells display been used to track renin-expressing cells and their descend-
dual smooth muscle cell-like characteristics and endocrine ants and has shown that the renin lineage of cells differen-
characteristics. Within JG cells, cathepsins, colocalized with tiate into other cell types in adult life (arteriolar smooth
prorenin in the secretory granules, have been suggested to muscle, but also intra- and extraglomerular mesangial cells,
activate prorenin to renin. Data from gene-targeted mice proximal tubule cells, and Bowman’s capsule cells), which,
show that at least cathepsin B is not crucial for this activa- however, retain the capacity to synthesize renin and convert
tion [79], and the prorenin-to-renin-processing enzyme has to a renin cell phenotype upon chronic stimulation of the
not yet been clearly identified as reviewed by Gross et al. system [114]. Thus, recruitment appears to be associated
[40]. The number of JG cells varies considerably with with those cells that expressed renin during embryonic/early
individual’s age and extracellular fluid volume status. Con- postnatal life and suggest that only these cells retain the
ditions of extracellular volume depletion, e.g., low NaCl ability to convert into a renin-producing phenotype. Of
intake [122], alter the number of differentiated renin- interest, renin-lineage cells were also observed in the testis,
producing cells by upstream differentiation of media cells adrenal gland, stomach, and thymus [114]. By which cellu-
to renin-secretory cell, a phenomenon referred to as lar signals do these shifts in phenotype occur? The
recruitment. microRNA-processing enzyme Dicer was deleted specifical-
ly in the JG cell lineage and resulted in a phenotype with
fewer renin-positive cells, lower renin mRNA expression,
Morphology of renin-positive cells lower plasma renin concentration and blood pressure, and
progressive decline in renal function typically observed after
Fully differentiated JG granular cells have a large nucleus, a deletion of renin–angiotensin system components from birth
hypertrophic rough endoplasmic reticulum, and distinct [115]. Thus, microRNAs, which are small noncoding
Golgi apparatus. Two main types of secretory granules can RNAs, appear essential for differentiation and/or mainte-
be discerned in the cytoplasm. Large electron-dense mature nance of JG cells. In particular, miR-330 and miR-125b-5p
granules contain primarily active renin, angiotensin (ANG) I control the smooth muscle phenotype of the renin cell
and ANG II peptides, and cathepsins. In addition, smaller, lineage [77]. A phenotype somewhat similar to that ob-
more electron-lucent protogranules leave the Golgi appara- served after JG cell-specific deletion of Dicer was observed
tus and contain active renin and prorenin [47]. The renin- by deletion of the notch receptor transcription factor RBP-J
secretory granules have several features in common with [14, 115]. After FACS sorting of labeled JG cells, the first
lysosomes: an acid pH, a content of typical lysosomal comprehensive “map” of renin cell-specific gene expression
enzymes like acid phosphatases, and the ability to take up pattern was presented. Pools of highly renin cell- and
extracellular particulate matter and degrade cellular constit- smooth muscle cell-specific genes belonging to transporters,
uents like internalized membrane material [124]. Renal re- enzymes, and transcription factor families were uncovered,
nin gene expression and active renin content show and of specific interest, pathways regulated by calcium and
developmental regulation and peak around the time of birth cyclic AMP (cAMP) were identified [8]. Is there a common
most clearly established in rat, when a large part of the renal stimulus for the recruitment phenomenon at the systemic
arterial vasculature is renin positive [39]. As the individual level? ANG II AT1A receptor deletion in mice combined
matures into adulthood, the localization of renin expression with a low-salt diet uncovers a much more marked recruit-
regresses to include only the most distal or juxtaglomerular ment compared to AT1A-intact mice and is paralleled by a
part of the afferent arterioles, but maintains the capacity to more marked decrease in blood pressure. When blood pres-
switch to a fetal pattern of increased renin cell distribution. sure was kept high by inhibition of nitric oxide synthase or
Recruitment of renin-positive cells has been observed after by infusion of alpha-adrenergic agonists during low dietary
pharmacological inhibition of renin, ANG II type 1 (AT1) NaCl intake in AT1A−/− mice, the renin cell recruitment was
receptors, angiotensin-converting enzyme (ACE), and after attenuated, suggesting that pressure is a more important
targeted disruption of genes encoding components in the signal compared to low transepithelial NaCl transport rate
cascade from angiotensinogen over renin to aldosterone [74]. Recruitment of JG cells during fetal life depends on
synthase, mineralocorticoid receptor, and Na+ transporters intact beta-adrenoceptors [81] and on the adenylyl-
within the thick ascending limb extending to the collecting stimulating Gsα protein [81], which indicates that renal
ducts (e.g., [29]). The molecular and cellular mechanisms by nerves, together with pressure, might have separate roles
which the changes in cell phenotype associated with recruit- for recruitment. The observation that salt depletion or ACE
ment occur are currently being unraveled. inhibition of hydronephrotic mice with no macula densa
Pflugers Arch - Eur J Physiol (2013) 465:25–37 27

also [4] leads to renin cell recruitment, and increased renin against intracytoplasmic solubilization as a significant path-
tissue content supports that pressure/vascular wall signals way for renin release [3, 121]. The data indicate that the
are sufficient to regulate renin cell recruitment in the com- initial fusion pore and small contact point are rapidly en-
plete absence of signals from the tubular epithelium. larged, and a more long-lived conformation is established
with folded or endocytosed membrane material. Quantita-
tive stereological counting of renin granules in rat kidneys
Mode of renin release was used to test the hypothesis that exocytosis would be
expected to result in fewer granules. Acute stimulation of
The renin storage granules are evenly distributed in the renin release in rats with a chronically prestimulated renin–
cytoplasm, and there is no apparent morphological polari- angiotensin–aldosterone system (RAAS) (by combined
zation of the release process towards the vascular lumen or ACEI and low NaCl) yielded a significant reduction in renin
renal interstitium. Thus, renin is released to plasma and a granule numbers [92]. This reduction in granule number
certain fraction of this renin is filtered at the glomerulus and agrees with the concept that mature granules fuse with the
probably taken up by the proximal tubule [72]. Some renin cell membrane and empty their contents by exocytosis and
is secreted into the interstitial compartment, which is thus “disappear.” Stereological counting of renin granules at
reflected in a high concentration of active renin in renal the electronmicroscopic level in the spontaneous mutant of
lymph [133]. the C57 strain “beige” mutant mice with impaired systemic
granule biogenesis showed that this mice strain had 1–2
Morphology of renin release renin granules per cell with multiple fingerlike processes
and a significantly lower plasma renin concentration than
The renin release process exhibits features of exocytosis that control mice [50]. Each granule was about 300 times larger
include close contact between mature granules and the plas- compared to a control mouse renin granulum [50]. If it is
ma membrane, membrane-limited omega-shaped recesses, assumed that granules contain an amount of renin that
and local changes of the granular matrix at the site of corresponds to the size of the granule, the observation of
membrane alignment; whereas, fusion areas, where the plasma renin concentrations in low–normal range suggests
two trilaminar membranes have merged, are an extremely that parts of granules or granule contents can be released or
rare observation, and plane fusion between the two mem- that renin is released very slowly from the granule matrix.
branes has not typically been observed in ultrathin sections
[121–123]. After intense stimulation of renin release in Renin release mode-functional data
mice, a contact area is observed between the renin storage
granule membrane and the plasma membrane, which is Collection of superfusion fluid with high time resolution
limited to a small protrusion of the granule [120, 121]. from single rat afferent arterioles showed that renin is re-
Within the protrusion, the granule matrix is less dense and leased in quanta with a renin content in each quantum,
a local increase of granule volume becomes apparent [120, which corresponds to the calculated content of one renin
121]. In vitro, similar changes in renin granule morphology granule [117]. The concept of exocytosis is supported by
were observed when renin secretion was stimulated. The data obtained with the whole-cell patch-clamp technique by
granules were often clustered close to the plasma mem- which cell capacitance, Cm, an accepted measure for cell
brane, and sites of putative fusion were observed [120]. A membrane surface area, can be recorded continuously dur-
frequent observation during intense stimulation of renin ing maneuvers predicted to stimulate renin release [30]. A
secretion is the emergence of large membrane-bounded range of extra- and intracellular factors known to alter renin
omega-shaped membrane recesses, which label for renin secretion by classic biochemical measurement of enzymic
protein and contain modified granule matrix. These recesses renin activity (cAMP, cell swelling, and isoprenaline) in-
are closely associated with the plasma membrane [3, 84, creased Cm 5–10 % over minutes in single JG cells compat-
121]. Because the plasma membrane-associated structures ible with addition of surface membrane and, thus, fusion of
label for renin, they are considered to represent a late stage renin granules [30]. Real-time imaging of JG cell granules
of the release process, when most of the granule content has in an in vitro system with high resolution demonstrated
been released. The tracking of renin protein from early rapid “disappearance” of granules compatible with fusion
protogranules through electron-dense mature granules to and release of granule contents [89].
membrane-bound recesses strongly suggests that the Renin secretion from isolated perfused kidneys also dem-
recesses represent empty granules that have undergone exo- onstrate very transient renin responses upon osmotic stimuli
cytotic membrane fusion. Moreover, in immunohistochem- that could be repeated after a lag period of minutes and,
ical studies, renin immunopositivity was not typically secondly, that the peak transient responses depended on the
observed within the cytoplasmic matrix, which speaks prestimulatory level of renin secretion [66]. These findings
28 Pflugers Arch - Eur J Physiol (2013) 465:25–37

all fit the concept that there exists a “readily releasable pool” days. In rats with chronic stimulation of the RAAS by
of renin granules available for prompt exocytosis. The iso- enalapril and low salt, a 36-fold elevated level of circulating
lated perfused kidney data further support that the pool of active renin was observed [92]. Upon acute stimulation of
readily releasable granules is inhibited by calcium-dependent renin secretion by lowering perfusion pressure to 40 mmHg
stimuli and supported by the cAMP pathway [66]. Of note, for 5 min, there was a significant and fairly similar twofold
supraphysiological stimulation of renin release from single JG increase in plasma renin concentration in both control and
cells led to significant decrease in Cm and, hence, cell mem- prestimulated rats. Thus, a similar order of magnitude of
brane area, which suggests either regular “loss” or by well- relative stimulation in response to acute stimulation al-
known “compensatory” membrane retrieval [30]. After mod- though the prestimulatory absolute levels of plasma and
erate stimulation of renin release and prolonged recording tissue renin differed markedly. In mice with constitutively
time, an increase of Cm was followed by a compensatory lower renin stores and lower JG cell number (JG cell-
decrease in Cm to the prestimulatory level [31]. If this reflects specific Gsα−/−, COX-2−/−, and dense core vesicle pro-
membrane retrieval, it would be compatible with the observa- tein−/−), similar observations have been made. The absolute
tion that renin cells take up external markers and internalize level of renin secretion (delta plasma renin) in response to
particles into renin granules that behave like lysosomes; ex- acute stimuli (furosemide, hydralazine, and isoproterenol
ogenous tracers, like ferritin, reach juvenile renin granules boli) corresponds to the preexisting level of renin expres-
without labeling the Golgi complex, and data show a large sion, while fold changes are rather similar [17, 53, 55].
membrane turnover in renin granules [123, 124]. What con- Thus, across species and kidney renin levels, there is a
trols intracellular renin granule trafficking? Targeted deletion significant correlation between the number of renin-
of “dense core vesicle proteins,” which are granule-associated producing cells and the absolute release potential. In total,
proteins known from pancreatic beta cells, leads to lower this results in a rather constant relative release potential.
levels of renin expression and plasma renin concentra- In agreement, stereological quantification of granule-
tion [55]. However, since these proteins do not coloc- containing volume of afferent arterioles in rat kidney showed
alize with renin, the lower renin levels were interpreted that the volume of the afferent arteriole that contained renin
as an indirect effect. Data showed that the deleted dense did not decrease despite decrease in granule numbers [92].
core vesicle proteins impaired catecholamine vesicle This observation suggests that a limited pool of renin granules
processing in mice and thereby attenuated sympathetic is released from each JG cell instead of complete depletion of
stimulation of renin [55]. The vesicle-associated mem- individual JG cells. Recruitment of stored granules beyond
brane protein (VAMP) 2 and VAMP3 were recently this acute release potential is limited. The capacitance meas-
shown in JG cells and knock-down in vitro of VAMP2, urements of surface membrane changes in JG cells were
but not VAMP3 attenuated cAMP-mediated renin release compared to granulum size and granulum pool size, and this
from isolated cells [78]. Further studies are warranted has yielded a rough estimate of the number of stored granules
along this line. that are released at ca. 5 % upon standard stimuli [30]. The
mouse afferent arteriole (NMRI strain) contains on average
1,900 granules/arteriole by stereological counting on standard
Renin stores, granule numbers, and renin release rodent diet [50]. In the rat, stereological counting yielded on
average 445 renin-secretory granules per afferent arteriole on
There is a constant, constitutive release of prorenin from JG a standard diet [92]. Chronic stimulation increased the number
granular cells, probably through vesicles that are different 20 times to ca. 9,000 granules [92]. Calculations have sug-
from mature storage granules [41]. Rapid changes in plasma gested that, in the rat, the release of one renin granulum per
active renin are not reflected by similar rapid changes in afferent arteriole per 5 min maintains basal PRC [117]. A
plasma prorenin, which argues against cosecretion of active similar estimation has yielded one each 20 min per arteriole
renin and prorenin [3]. The rate of prorenin release primarily in the mouse [121], and with ca. 1,900 granules per arteriole in
reflects the rate of renin synthesis [91]. What is the physi- mice, this provides theoretically the mouse with sufficient
ological significance of renin cell differentiation during renin granules for 26 days and the rat with a smaller potential.
chronic stimulation of the system? Is renin release depen- So despite the large stores of active renin within the kidney,
dent on renin cell recruitment because absolute stores of the full secretory capacity of preexisting JG cells is not
active renin are limited or because the release process can- exploited. Recruitment of more JG cells is preferred over
not recruit stored renin granules to an extent that suffices to enhanced intracellular granule recruitment processes. The
meet the circulatory demand in situations with extracellular evolutionary background for this dominant negative regula-
volume depletion or low renal perfusion pressure? The first tion of renin secretion could be the danger of evoking a
question can be easily answered since kidney tissue renin hypertensive crisis by uncontrolled renin release. These obser-
content suffices to maintain circulating levels for hours to vations contribute to the understanding of how it is possible
Pflugers Arch - Eur J Physiol (2013) 465:25–37 29

for the renin–angiotensin system to be involved in short-term potential in a range far from the threshold for activation
regulation of blood pressure via acute effects on peripheral of voltage-gated calcium channels. On the other hand,
resistance, while at the same time, the system can be involved significant depolarization (as e.g., after ANG II) may
in the long-term regulation of blood pressure by its effects on depolarize JG cells to level of Vm that activate L-type
sodium and water homeostasis. After chronic stimulation of calcium channels and attenuate cAMP-mediated stimula-
the system, the circulating renin and ANG II concentrations tion of renin release.
are high. In this situation, the ANG II receptor expression is
downregulated in the peripheral blood vessels, whereby, a
hypertensive crisis is avoided. The renin–angiotensin system Intracellular signaling pathways that control exocytosis
maintains its ability to regulate short-term blood pressure in juxtaglomerular cells
(e.g., after changing position), also in a situation with massive
chronic stimulation, because the relative increase in renin Cyclic AMP pathway
release is the same as in the control situation.
Several physiologically relevant para- and endocrine agonists
and neurotransmitters coupled to activation of adenylyl cy-
Membrane potential and its influence on renin secretion clase stimulate renin release and increase cAMP in JG cells.
Mice with deficiency in the adenylyl cyclase activating Gsα
Stimulus–secretion coupling often depends on electrical sig- protein in JG cells display lower renin stores and plasma renin
nals that initiate the process. JG cells in situ are polarized (−50 concentration [17]. There is an attenuated renin secretion
to −70 mV) [9–12, 26], and membrane potential influences the response from isolated Gsα protein-deficient-isolated JG cells
release process. Bührle et al. observed that addition of agonists to β1-adrenoceptor agonist isoproterenol and PGE2 [1, 17].
with intracellular coupling to cAMP had no effect on mem- Adenylyl cyclases 5 and 6 catalyze cAMP formation in JG
brane potential (Vm), while calcium-dependent agonists led to cells [1, 86, 87] (Fig. 1). Phosphodiesterases (PDE) 3 and 4 are
depolarization [9, 11]. It was concluded that stimulation of expressed in renal vessels and JG cells [97] and use primarily
renin release was independent of membrane potential, while cAMP as a substrate. PDE3 and PDE4 degrade cAMP in JG
inhibitory signals could depend on membrane depolarization. cells since selective PDE3 and PDE4 inhibitors stimulate
The first whole-cell patch clamp study on JG cells in isolated renin secretion in conscious rabbits and humans [20–22, 93]
mouse afferent arterioles showed depolarization by inhibition and in isolated perfused rat kidney [61] (Fig. 1).
of inward-rectifier K channels (KIR) after addition of ANG II
[63]. Subsequent studies confirmed that JG cells express KIR Cyclic GMP pathway and coupling to cAMP pathway
channel subunits [70]. At normal resting membrane potential
in JG cells, the dominant potassium conductance is through Agonists that raise intracellular cGMP in granular cells,
calcium- and cAMP-sensitive BKCa channels (KCa1.1, ZERO such as nitric oxide (NO) and atrial natriuretic peptide
variant) and, to a lesser degree, through voltage-gated KV [60], have been reported to have either an inhibitory [44]
channels [33]. Setting Vm in JG cells at different levels did or a biphasic [108] effect on renin release rate in vitro. In
not per se induce changes in Cm and, hence, exocytosis [33]. vivo and within the isolated perfused kidney preparation, it
Depolarization in one, but not other studies showed activation is a rather consistent finding that NO donors promptly
of classical voltage-gated calcium currents [33, 64, 103] stimulate renin secretion [102]. NO donors depend on an
which could be blocked by selective L-type antagonists [33, intact protein kinase A (PKA) pathway for stimulation of
64, 103]. Both RNA and protein for the L-type, voltage-gated renin release, which suggests an effect mediated by cAMP
calcium channel (CaV) 1.2 was observed in single JG cells rather than cGMP [61]. NO donors potently increase cGMP
[33]. A functional role of voltage-gated calcium channels was in JG granular cells, but cAMP is also stimulated [108]. The
indicated by the observation that depolarizing the membrane putative connection between the two pathways in the regu-
potential inhibited cAMP-mediated increases in cell mem- lation of renin secretion was established by discovery of
brane capacitance in an L-type channel blocker-sensitive marked stimulation of renin secretion by pharmacological
way [33]. So what is the integrated role of Vm in block of PDE3 [20–22, 61]. PDE3A and PDE3B were
stimulus–secretion coupling in JG cells? JG cells in the demonstrated in afferent arterioles [97], and while PDE3
vascular wall are quite polarized −50 to −60 mV and thus at degrades cAMP, it is inhibited by cGMP (Fig. 1). Sensitivity
prevailing conditions far from levels of Vm that activate towards PDE3 blockers and PDE3 expression was detected
the L-type calcium current which is maximal at also at the level of the single JG cell where intracellular
+10 mV. It is likely that the hyperpolarizing influence dialysis with cGMP-enhanced exocytosis as determined by
of cAMP-coupled agonists through KCa1.1, ZERO vari- a cell capacitance increase dependent of protein kinase A
ant is permissive and serves to stabilize membrane activity [32]. It is conceivable that at least part of the
30 Pflugers Arch - Eur J Physiol (2013) 465:25–37

stimulation of renin release in response to NO is caused by Calcium-regulated pathways


inhibition of cAMP degradation.
Two cGMP-dependent protein kinases have been identi- The paradoxical inhibitory role of calcium in renin release is
fied, and both are expressed by JG granular cells, in particular, reviewed comprehensively by others in the current issue and
the cGMP-regulated protein kinase type II (cGKII) [36]. In is, therefore, not detailed in the present review. What is
mice with targeted disruption of cGKII, cGMP-dependent current status on mechanisms for this effect? Agonists that
agonists are unable to inhibit renin secretion [131], while in raise [Ca2+]i in granular cells prevent the stimulatory action
granular cells which have been transfected to overexpress of cAMP-dependent agonists on renin release [94]. Depo-
cGKII, there is marked inhibition of cAMP-stimulated renin larization of the membrane potential prevents cAMP-
release [37]. Thus, the renin-secretory response to cGMP- stimulated exocytosis in JG cells in a way sensitive to
dependent agonists is determined by the dominance of the calcium channel blockers [33]. These observations are com-
cGKII pathway compared to the PDE3 pathway. Most data patible with an enhanced degradation or reduced formation
suggest that PDE3 is constitutively expressed, whereas cGKII of cAMP by calcium in JG cells. Both adenylate cyclase and
is regulated [36], but at present, the significance of this is not cAMP phosphodiesterases exist in calcium-sensitive iso-
established. Altogether, cGMP has complex effects on renin forms, and experimental evidence indicates that calcium-
secretion, which are determined by the prevalence of two stimulated PDE1 is present in JG cells and contribute to
mutually antagonistic target pathways for cGMP in stimu- the inhibition of renin release by calcium [85] (Fig. 1).
lus–secretion coupling (Fig. 1). At present, the data suggest Calcium-inhibitable adenylyl cyclase V is present in JG
that the stimulatory pathway for cGMP depends on cAMP. cells, and chelation of calcium increases cAMP formation

Fig. 1 Signaling pathways in the stimulus–secretion coupling for calcium from intracellular stores (ER). Intracellular cGMP increases
renin in renal juxtaglomerular granular cells. Arrow indicates stimula- through soluble guanylyl cyclase and membrane receptor-activated
tion and a line with foot indicates suppression. Cyclic AMP, cGMP, and guanylyl cyclase activity through nitric oxide and atrial natriuretic
free ionized calcium are crucial second messengers in the pathways peptide. Cyclic GMP can either stimulate renin release by inhibition
that govern exocytosis of renin. Several agonists (e.g., PGE2, norepi- of PDE3 or inhibit renin release through cGMP-regulated protein
nephrine, and PGI2), acting through specific receptors (R), converge on kinase. Gi-coupled agonists like angiotensin II, extracellular calcium,
Gsα protein-dependent activation of adenylyl cyclases (AC5 and 6) or endothelin cause an increase of intracellular free calcium concentra-
and synthesis of cAMP. Protein kinase A is necessary for cAMP- tion by release of calcium from intracellular stores and by membrane
mediated stimulation of exocytosis measured as an increase of cell depolarization (Vm) with subsequent activation of transmembrane cal-
capacitance (Cm). Moderate cell swelling occurs through aquaporin 1- cium influx through CaV. Membrane depolarization is caused by inhi-
mediated water uptake and also leads to PGE2-EP2/4 receptor activa- bition of anomalous inward-rectifier K channel and by stimulation of
tion of the cAMP pathway. PKA increases K+ current though BKCa calcium-activated chloride channels. This could lead to coordinated
channels (ZERO variant). This leads to hyperpolarization of membrane efflux of KCl and cell shrinkage. Calcium suppresses renin secretion
potential (Vm), which is permissive for renin release by preventing likely through serial activation of calmodulin and calcineurin and by
activation of voltage-gated calcium channels (CaV). Cyclic AMP is parallel inhibition of cAMP. Cyclic AMP degradation by PDE1 is
degraded through phosphodiesterases 3 that is inhibited by cGMP, and enhanced, and cAMP formation is inhibited through AC5/6
PDE1 that is stimulated by Ca2+. Cyclic AMP prevents liberation of
Pflugers Arch - Eur J Physiol (2013) 465:25–37 31

and renin release [1, 86] and JG cell membrane capacitance Cellular pathways for major systemic controllers
[75]. Blockers of calmodulin enhance renin release and JG of renin
cell membrane capacitance [25, 75]. Calcium is not likely to
control renin solely through cAMP, because release rate can “Baroreceptor”
be changed without alterations in cAMP [65]. Calcium
could affect renin secretion by a direct interaction with ion It was suggested by Tobian [125] and demonstrated by
channels in the cell membrane. In fact, the most direct effect of Skinner et al. [116] that there existed a renal baroreceptor
a rise in [Ca2+]i reported is activation of a chloride conduc- by which arterial blood pressure negatively controlled renin
tance [63, 103] (Fig. 1). A calcium–calmodulin-dependent release. In the denervated, nonfiltering dog kidney, Blaine et
phosphatase, calcineurin, appears also to control exocytosis al. [5] demonstrated that pressure-regulated renin release
of renin by yet undefined interactions as judged by sensitivity was intact. There is now agreement that the baroreceptor
of renin release from isolated JG cells to clinically well- mechanism resides in the renal vasculature itself. The baro-
established blockers of calcineurin, e.g., cyclosporine [58, receptor has been extensively characterized with respect to
75] (Fig. 1). The second messenger cyclic ADP-ribose “dose” (pressure)–response relationships [56] and calcium
through Ca2+ signaling appears also to be a potential pathway dependence [100, 101]. Data suggest that pressure-sensitive
that exist at least in the renin-producing AS4-1 cell line [138]. renin release is independent of voltage-gated calcium chan-
nels [101, 103], but dependent on extracellular calcium
Osmotic forces [100]. It has been difficult to establish reproducible data
on baroreceptor function in vitro with isolated arterioles.
Renin secretion from in vitro preparations exhibits a high Flow appears to be required to obtain pressure-sensitive
sensitivity to changes in extracellular osmolality such that a renin responses with isolated renal microvessels [6, 96]. In
modest, e.g., ~5–10 % reduction in the extracellular osmo- contrast, it has been shown that mechanical stretch leads to
lality, leads to rapid increases in renin secretion [28, 104, significant inhibition of renin release and reduced renin
127]. Changes in osmolality induced by nonpermeant sol- mRNA abundance in cultured rat granular cells [13]. In-
utes are more efficient in inducing changes in renin release creased pressure leads to elevated intracellular calcium con-
than permeant solutes, which implies that it is the osmotic centration in a dose-dependent way and sensitive to L-type
effect rather than a specific ionic effect that causes the calcium channel blockers and cation channel blockers in JG
change in renin release. JG cells with permeabilized cell cells in situ within isolated murine arterioles [68]. Are
membranes retain the ability to release renin upon exposure mediators involved in the response? Adenosine acting
to a moderate decrease in osmolality, which indicates that through A1 receptors is necessary for high pressure-
renin granules are osmotically sensitive [52]. In contrast, in induced suppression of renin secretion, while lowering of
more complex models, i.e., the perfused kidney and in vivo pressure led to adenosine-independent stimulation of renin
settings, an increase in osmolality stimulates renin secretion secretion [112]. Further dissection of the mechanism was
[2, 66]. The extracellular osmolality in the JG area has been obtained by data showing that the integral position of JG
suggested to vary depending on transepithelial NaCl trans- cells within the vascular wall is an absolute requirement for
port rate by the rather water impermeable macula densa pressure sensitivity of renin release [130]. Mice deficient in
[88]. JG cell capacitance is rapidly increased by a decrease the gap junction protein connexin 40 (Cx40−/−) dis-
in extracellular osmolality or by introduction of slightly played an unexpected phenotype where JG cells were
hyperosmotic solution into the cytoplasm [30]. Thus, cell ectopically localized, dissociated from the vascular wall
swelling, and not granule swelling, is necessary for the [67]. Disruption of Cx40 led to significantly elevated
stimulus–secretion response to hypoosmolality, at least in blood pressure, loss of inhibition of renin release by
single JG cells. The response depended on presence of the ANG II, and perfusion pressure [130]. This observation
water channel aquaporin 1 (AQP1) and on intact cAMP- has clinical implications since the human juxtaglomeru-
PKA signaling [34]. An autocrine loop was suggested based lar apparatus displays a similar connexin expression as
on the dependence of cell swelling-induced renin release on rodents [62], and a human mutation in Cx40, associated
phospholipase A2 (PLA2) and COX-2 activities [34]. The with hypertension, yields hypertension and ectopic JG
findings indicate that slight hypoosmotic extracellular fluid cells when introduced in the mouse [73]. Loss of JG
leads to AQP1-mediated water influx in JG cells, PLA2/ cell “homing” is associated with hypertension and com-
COX-2/prostanoid-dependent cAMP formation, and activa- plete absence of pressure sensitivity of renin secretion.
tion of PKA, which promotes exocytosis of renin. The Together, the data show that pressure-mediated suppres-
difference between renin release responses to changes in sion of renin release depends on adenosine, on extracel-
osmolality in vivo and in reduced in vitro settings is not lular calcium, and on Cx40-dependent proper vascular
clear at present. localization of JG cells.
32 Pflugers Arch - Eur J Physiol (2013) 465:25–37

Renal sympathetic nerves—catecholamines juxtaglomerular cells [35, 51, 111]. Mice with targeted
deletion of PGE2-EP4 receptors display low sensitivity of
Renin mRNA abundance and renin release are tonically renin secretion to chronic furosemide [83] and in isolated
stimulated through a β-adrenoceptor mechanism indepen- perfused kidneys from EP2 and EP4−/− mice, PGE2-mediat-
dent of tubular function or glomerular filtration rate. Renin ed renin secretion is attenuated [111]. In the isolated per-
release and renin mRNA abundance are decreased by renal fused juxtaglomerular apparatus, stimulation of renin
denervation [46], while α1-adrenoceptor agonists have the secretion by low luminal NaCl concentration was abolished
opposite effect in vivo [45]. In addition to pharmacological by COX-2 blockers [126]. At more integrated levels, a full
data, gene-targeted mice have further supported the notion dependence of renin secretion on intact COX-2 function has
that β1-adrenoceptors are necessary for catecholamine- been difficult to establish. COX-2−/− mice display constitu-
mediated stimulation of renin secretion in vivo and that tively low renin expression and exhibit impaired stimulation
β1-agonist-mediated renin release depends on G protein of renin by low NaCl intake [135]. Furosemide infusion
Gsα and on adenylyl cyclase types 5 and 6 (Fig. 1) [1, yielded a smaller increase in plasma renin in COX-2−/−
17]. In vitro, β1-adrenoceptor occupation initiates cAMP compared to wild type [53]. However, combined stimulation
formation in JG cells and increases renin release and renin by low salt and AT1 inhibition in COX-2−/− greatly en-
mRNA level [18, 24] and cell membrane capacitance, doc- hanced renin stores and markedly increased the release of
umenting exocytosis [30]. At present, there are no system- renin in response to furosemide [53]. Together, there is good
atic publications documenting the effect of catheter-based evidence that COX-2-PGE2 through EP2/EP4 receptors sup-
radiofrequency ablation of renal sympathetic nerves in ports renin release and renin mRNA in conditions with
patients with treatment-resistant hypertension on plasma attenuated transepithelial transport of NaCl in the cortical
concentration of renin–angiotensin system components. In loop of Henle. At integrated levels, the findings indicate that
one minor Dutch study, plasma renin activity did not COX-2 rather controls the pool size of renin, perhaps
change, while plasma aldosterone fell [129]. New data from through effects on the number of JG cells [135], while the
many ongoing trials will provide this information in the near effect on renin secretion is redundant and not mandatory.
future. Adenosine A1 analogues inhibit renin release from the
isolated JGA when applied to the abluminal side of the
Macula densa and tubular control of renin secretion preparation [132] and cause depolarization and calcium
increases in JG cells [69]. Adenosine inhibits renin release
It has been established that whole body sodium chloride both in vivo and in vitro [48, 59, 118]. In general, vasocon-
content exerts a negative feedback on renin secretion strictor A1 receptors are activated by nanomolar concentra-
through the macula densa mechanism [72, 119, 128]. As tions of adenosine, but very high concentrations of
defined by Schnermann [98], a true mediator of responses in adenosine are necessary to inhibit low NaCl-stimulated re-
the JG apparatus (JGA) must be able to exert a graded effect nin release from the isolated, perfused juxtaglomerular ap-
on renin secretion, and this effect must be initiated by NaCl paratus [48, 59, 118]. On the other hand, mice with targeted
transport-dependent signals which stimulate or inhibit the deletion of adenosine A1 receptors display elevated basal
local concentration of the mediator. Improper positioning of renin plasma concentration [7] and impaired suppression of
JG cells as observed in Cx40−/− mice leads to impaired renin by high salt intake [54, 95] and by high perfusion
stimulation by furosemide [67]. The proper localization of pressure [112], while stimulation by furosemide was intact
JG cells is a prerequisite for the macula densa signal. A [112, 113]. Current data support a significant role for aden-
close contact between macula densa cells and JG cells is osine in both the TGF response [99] and in suppression of
necessary. A diffusible mediator appears unable to reach renin secretion in response to high NaCl and elevated per-
“displaced” JG cells although still in the glomerular tuft fusion pressure, whereas, stimulation of renin by low trans-
area. Especially, three paracrine mediator candidates have epithelial NaCl transport across the loop of Henle is not
been in focus: adenosine, NO, and prostaglandins. Macula dependent on adenosine.
densa associated COX-2 mRNA, and protein is regulated
concordantly with renin under most conditions tested (e.g., Nitric oxide
[42, 49, 137]). The sensed variable appears to be the apical
concentration of chloride that correlates inversely to COX-2 Macula densa cells selectively express neuronal nitric oxide
expression in cultured TAL/macula densa cells through a synthase or NOS-I [80]. NOS-I expression in the macula
p38 protein kinase-dependent pathway [19, 136]. In isolated densa correlates to renin expression (e.g., [110]). NO syn-
JG apparatus, a low luminal NaCl concentration leads to thase inhibition reduces renin secretion and renin mRNA
release of PGE2 [90]. PGE2 and prostacyclin (PGI2) stimu- level under acute [102] or chronic [107] low renal perfusion
late renin secretion through EP2/EP4/IP receptors on pressure, under a low dietary NaCl intake [109], under
Pflugers Arch - Eur J Physiol (2013) 465:25–37 33

furosemide treatment [105], and under ANG II receptor Acknowledgments The authors wish to thank Annette K. Rasmussen
Mette Fredenslund, Lis Teusch, Gitte Dybmose, and Inge Andersen for
blockade [106]. Targeted deletion of NOS-I led to lower
their skillful technical assistance. Work in the authors’ lab has been
plasma renin concentration and lower secretion rates from supported by grants from Carlsbergfondet, the Danish Research Council
isolated perfused kidneys and attenuated furosemide- for Health and Disease, the Strategic Research Council, the Danish Heart
stimulated renin secretion [16]. The stimulation of renin Foundation, the NOVO Nordisk Foundation, the Lundbeck Foundation,
AP Moller Foundation, Helen and Ejnar Bjørnows Foundation, the
secretion induced by a low luminal NaCl concentration
Foundation for the Promotion of Medical Science, and the Foundation
was abolished in the presence of an NO synthesis inhibitor of 17.12.1981.
[43]. NO is of significance for a low NaCl-induced renin
release at the level of the juxtaglomerular apparatus. NO is
permissive for renin secretion, in particular, during low References
NaCl conditions and likely through inhibition of PDE3
(Fig. 1). PDE3 inhibitor administration to nNOS−/− mice 1. Aldehni F, Tang T, Madsen K, Plattner M, Schreiber A, Friis UG,
restored plasma renin concentration from low to normal Hammond HK, Han PL, Schweda F (2011) Stimulation of renin
independent of blood pressure [95]. secretion by catecholamines is dependent on adenylyl cyclases 5
and 6. Hypertension 57:460–468
2. Beierwaltes WH (2006) A different vision of the osmolar regu-
ANG II lation of renin secretion. Am J Physiol Renal Physiol 290:F795–
F796
ANG II AT1 receptors are present on JG cells, and ANG II 3. Berka JL, Alcorn D, Ryan GB, Skinner SL (1992) Renin process-
ing studied by immunogold localization of prorenin and renin in
exposure elicits an increase in the intracellular concentration granular juxtaglomerular cells in mice treated with enalapril. Cell
of calcium and membrane depolarization, which display Tissue Res 268:141–148
rapid desensitization in JG cells [10, 63, 64]. ANG II is 4. Berka JL, Alcorn D, Ryan GB, Skinner SL, Weaver DA (1993)
thought to exert direct negative feedback on renin secretion Renin processing in cultured juxtaglomerular cells of the hydro-
nephrotic mouse kidney. J Histochem Cytochem 41:365–373
by the so-called “short loop feedback inhibition.” Renin
5. Blaine EH, Davis JO (1971) Evidence for a renal vascular mech-
secretion from isolated perfused kidneys in the absence of anism in renin release: new observations with graded stimulation
nerve input and variation in perfusion pressure is highly by aortic constriction. Circ Res 28(Suppl 2):118–126
sensitive to ANG II infusion (e.g., [100]). In vitro, the 6. Bock HA, Hermle M, Brunner FP, Thiel G (1992) Pressure
dependent modulation of renin release in isolated perfused glo-
inhibition of renin release has been difficult to establish
meruli. Kidney Int 41:275–280
clearly with some exceptions [38]. Despite the presence of 7. Brown RD, Thoren P, Steege A, Mrowka R, Sallstrom J, Skott O,
AT1 receptors, recruitment was observed in JG cells in Fredholm BB, Persson AE (2006) Influence of the adenosine A1
chimeric mice bearing both AT1+/+ and AT1−/− JG cells, receptor on blood pressure regulation and renin release. Am J
Physiol Regul Integr Comp Physiol 290:R1324–R1329
thus questioning the direct feedback of ANG II on JG cells 8. Brunskill EW, Sequeira-Lopez ML, Pentz ES, Lin E, Yu J,
[76]. Cross-transplanted mice with AT1A receptor deficien- Aronow BJ, Potter SS, Gomez RA (2011) Genes that confer the
cy only in the kidney exhibit a small, but significant lower identity of the renin cell. J Am Soc Nephrol 22:2213–2225
blood pressure, while renin expression was not elevated 9. Buhrle CP, Hackenthal E, Helmchen U, Lackner K, Nobiling R,
Steinhausen M, Taugner R (1986) The hydronephrotic kidney of
compared to the AT1A intact mice [23]. This observation
the mouse as a tool for intravital microscopy and in vitro electro-
questions the existence of a short-loop feedback, although physiological studies of renin-containing cells. Lab Investig
plasma renin concentration was not measured. Moreover, in 54:462–472
mice with complete AT1A deletion, renin expression was 10. Buhrle CP, Hackenthal E, Nobiling R, Skott O, Baumbach L,
Taugner R (1987) Tachyphylaxis of juxtaglomerular epithelioid
elevated, and this corresponded with a lower blood pressure.
cells to angiotensin II. Differences between the electrical mem-
This indicates that blood pressure is more important to brane response and renin secretion. Pflugers Arch 410:55–62
determine the renin response after impaired ANG II signal- 11. Buhrle CP, Scholz H, Hackenthal E, Nobiling R, Taugner R
ing than short-loop ANG II feedback on renin. Salt- (1986) Epithelioid cells: membrane potential changes induced
by substances influencing renin secretion. Mol Cell Endocrinol
loading mice with kidney-only AT1 receptor deletion 45:37–47
raised blood pressure and lowered renin expression con- 12. Buhrle CP, Scholz H, Nobiling R, Taugner R (1986) Junctional
firming this notion [23]. Quantification of JG cell re- transmission in renin-containing and smooth muscle cells of the
cruitment in salt-depleted AT1−/− and wild type shows afferent arteriole. Pflugers Arch 406:578–586
13. Carey RM, McGrath HE, Pentz ES, Gomez RA, Barrett PQ
that recruitment, as well as renin expression, was atten-
(1997) Biomechanical coupling in renin-releasing cells. J Clin
uated by increasing blood pressure through, e.g., phen- Invest 100:1566–1574
ylephrine or L-NAME infusion [74]. Taken together, the 14. Castellanos Rivera RM, Monteagudo MC, Pentz ES, Glenn ST,
current findings indicate that although functional ANG Gross KW, Carretero O, Sequeira-Lopez ML, Gomez RA (2011)
Transcriptional regulator RBP-J regulates the number and plas-
II receptors are associated with JG cells, the effect of
ticity of renin cells. Physiol Genomics 43:1021–1028
ANG II on renin to a significant extent is mediated by blood 15. Castrop H, Hocherl K, Kurtz A, Schweda F, Todorov V, Wagner
pressure. C (2010) Physiology of kidney renin. Physiol Rev 90:607–673
34 Pflugers Arch - Eur J Physiol (2013) 465:25–37

16. Castrop H, Schweda F, Mizel D, Huang Y, Briggs J, Kurtz A, requires aquaporin-1 and cyclooxygenase-2. J Am Soc Nephrol
Schnermann J (2004) Permissive role of nitric oxide in macula 20:2154–2161
densa control of renin secretion. Am J Physiol Renal Physiol 286: 35. Friis UG, Stubbe J, Uhrenholt TR, Svenningsen P, Nusing RM,
F848–F857 Skott O, Jensen BL (2005) Prostaglandin E2 EP2 and EP4 recep-
17. Chen L, Kim SM, Oppermann M, Faulhaber-Walter R, Huang Y, tor activation mediates cAMP-dependent hyperpolarization and
Mizel D, Chen M, Lopez ML, Weinstein LS, Gomez RA, Briggs exocytosis of renin in juxtaglomerular cells. Am J Physiol Renal
JP, Schnermann J (2007) Regulation of renin in mice with Cre Physiol 289:F989–F997
recombinase-mediated deletion of G protein Gsalpha in juxtaglo- 36. Gambaryan S, Hausler C, Markert T, Pohler D, Jarchau T, Walter
merular cells. Am J Physiol Renal Physiol 292:F27–F37 U, Haase W, Kurtz A, Lohmann SM (1996) Expression of type II
18. Chen M, Schnermann J, Smart AM, Brosius FC, Killen PD, cGMP-dependent protein kinase in rat kidney is regulated by
Briggs JP (1993) Cyclic AMP selectively increases renin mRNA dehydration and correlated with renin gene expression. J Clin
stability in cultured juxtaglomerular granular cells. J Biol Chem Invest 98:662–670
268:24138–24144 37. Gambaryan S, Wagner C, Smolenski A, Walter U, Poller W,
19. Cheng HF, Wang JL, Zhang MZ, McKanna JA, Harris RC (2000) Haase W, Kurtz A, Lohmann SM (1998) Endogenous or overex-
Role of p38 in the regulation of renal cortical cyclooxygenase-2 pressed cGMP-dependent protein kinases inhibit cAMP-
expression by extracellular chloride. J Clin Invest 106:681–688 dependent renin release from rat isolated perfused kidney, micro-
20. Chiu YJ, Hu SH, Reid IA (1999) Inhibition of phosphodiesterase dissected glomeruli, and isolated juxtaglomerular cells. Proc Natl
III with milrinone increases renin secretion in human subjects. J Acad Sci U S A 95:9003–9008
Pharmacol Exp Ther 290:16–19 38. Geary KM, Hunt MK, Peach MJ, Gomez RA, Carey RM (1992)
21. Chiu N, Park I, Reid IA (1996) Stimulation of renin secretion by Effects of angiotensin converting enzyme inhibition, sodium
the phosphodiesterase IV inhibitor rolipram. J Pharmacol Exp depletion, calcium, isoproterenol, and angiotensin II on renin
Ther 276:1073–1077 secretion by individual renocortical cells. Endocrinology
22. Chiu T, Reid IA (1996) Role of cyclic GMP-inhibitable phospho- 131:1588–1594
diesterase and nitric oxide in the beta adrenoceptor control of 39. Gomez RA, Lynch KR, Sturgill BC, Elwood JP, Chevalier RL,
renin secretion. J Pharmacol Exp Ther 278:793–799 Carey RM, Peach MJ (1989) Distribution of renin mRNA and its
23. Crowley SD, Gurley SB, Oliverio MI, Pazmino AK, Griffiths R, protein in the developing kidney. Am J Physiol 257:F850–F858
Flannery PJ, Spurney RF, Kim HS, Smithies O, Le TH, Coffman 40. Gross KW, Gomez RA, Sigmund CD (2010) Twists and turns in
TM (2005) Distinct roles for the kidney and systemic tissues in the search for the elusive renin processing enzyme: focus on
blood pressure regulation by the renin–angiotensin system. J Clin “cathepsin B is not the processing enzyme for mouse prorenin”.
Invest 115:1092–1099 Am J Physiol Regul Integr Comp Physiol 298:R1209–R1211
24. Della Bruna R, Kurtz A, Corvol P, Pinet F (1993) Renin mRNA 41. Hackenthal E, Paul M, Ganten D, Taugner R (1990) Morphology,
quantification using polymerase chain reaction in cultured juxta- physiology, and molecular biology of renin secretion. Physiol
glomerular cells. Short-term effects of cAMP on renin mRNA Rev 70:1067–1116
and secretion. Circ Res 73:639–648 42. Harris RC, McKanna JA, Akai Y, Jacobson HR, Dubois RN,
25. Della Bruna R, Pinet F, Corvol P, Kurtz A (1992) Calmodulin Breyer MD (1994) Cyclooxygenase-2 is associated with the
antagonists stimulate renin secretion and inhibit renin synthesis in macula densa of rat kidney and increases with salt restriction. J
vitro. Am J Physiol 262:F397–F402 Clin Invest 94:2504–2510
26. Fishman MC (1976) Membrane potential of juxtaglomerular 43. He XR, Greenberg SG, Briggs JP, Schnermann JB (1995) Effect
cells. Nature 260:542–544 of nitric oxide on renin secretion. II. Studies in the perfused
27. Fordis CM, Megorden JS, Ropchak TG, Keiser HR (1983) Ab- juxtaglomerular apparatus. Am J Physiol 268:F953–F959
sence of renin-like activity in rat aorta and microvessels. Hyper- 44. Henrich WL, McAllister EA, Smith PB, Campbell WB (1988)
tension 5:635–641 Guanosine 3′,5′-cyclic monophosphate as a mediator of inhibition
28. Frederiksen O, Leyssac PP, Skinner SL (1975) Sensitive osmometer of renin release. Am J Physiol 255:F474–F478
function of juxtaglomerular cells in vitro. J Physiol 252:669–679 45. Holmer SR, Kaissling B, Putnik K, Pfeifer M, Kramer BK,
29. Friberg P, Sundelin B, Bohman SO, Bobik A, Nilsson H, Riegger GA, Kurtz A (1997) Beta-adrenergic stimulation of renin
Wickman A, Gustafsson H, Petersen J, Adams MA (1994) expression in vivo. J Hypertens 15:1471–1479
Renin-angiotensin system in neonatal rats: induction of a renal 46. Holmer S, Rinne B, Eckardt KU, Le Hir M, Schricker K, Kaissling
abnormality in response to ACE inhibition or angiotensin II B, Riegger G, Kurtz A (1994) Role of renal nerves for the expres-
antagonism. Kidney Int 45:485–492 sion of renin in adult rat kidney. Am J Physiol 266:F738–F745
30. Friis UG, Jensen BL, Aas JK, Skott O (1999) Direct demonstra- 47. Hunt MK, Ramos SP, Geary KM, Norling LL, Peach MJ, Gomez
tion of exocytosis and endocytosis in single mouse juxtaglomer- RA, Carey RM (1992) Colocalization and release of angiotensin
ular cells. Circ Res 84:929–936 and renin in renal cortical cells. Am J Physiol 263:F363–F373
31. Friis UG, Jensen BL, Hansen PB, Andreasen D, Skott O (2000) 48. Itoh S, Carretero OA, Murray RD (1985) Possible role of aden-
Exocytosis and endocytosis in juxtaglomerular cells. Acta Phys- osine in the macula densa mechanism of renin release in rabbits. J
iol Scand 168:95–99 Clin Invest 76:1412–1417
32. Friis UG, Jensen BL, Sethi S, Andreasen D, Hansen PB, Skott O 49. Jensen BL, Kurtz A (1997) Differential regulation of renal cyclo-
(2002) Control of renin secretion from rat juxtaglomerular cells oxygenase mRNA by dietary salt intake. Kidney Int 52:1242–1249
by cAMP-specific phosphodiesterases. Circ Res 90:996–1003 50. Jensen BL, Rasch R, Nyengaard JR, Skott O (1997) Giant renin
33. Friis UG, Jorgensen F, Andreasen D, Jensen BL, Skott O (2003) secretory granules in beige mouse renal afferent arterioles. Cell
Molecular and functional identification of cyclic AMP-sensitive Tissue Res 288:399–406
BKCa potassium channels (ZERO variant) and L-type voltage- 51. Jensen BL, Schmid C, Kurtz A (1996) Prostaglandins stimulate
dependent calcium channels in single rat juxtaglomerular cells. renin secretion and renin mRNA in mouse renal juxtaglomerular
Circ Res 93:213–220 cells. Am J Physiol 271:F659–F669
34. Friis UG, Madsen K, Svenningsen P, Hansen PB, Gulaveerasingam 52. Jensen BL, Skott O (1993) Osmotically sensitive renin release
A, Jorgensen F, Aalkjaer C, Skott O, Jensen BL (2009) from permeabilized juxtaglomerular cells. Am J Physiol 265:
Hypotonicity-induced renin exocytosis from juxtaglomerular cells F87–F95
Pflugers Arch - Eur J Physiol (2013) 465:25–37 35

53. Kim SM, Chen L, Mizel D, Huang YG, Briggs JP, Schnermann J 72. Leyssac PP (1986) Changes in single nephron renin release are
(2007) Low plasma renin and reduced renin secretory responses mediated by tubular fluid flow rate. Kidney Int 30:332–339
to acute stimuli in conscious COX-2-deficient mice. Am J Phys- 73. Lubkemeier I, Machura K, Kurtz L, Neubauer B, Dobrowolski R,
iol Renal Physiol 292:F415–F422 Schweda F, Wagner C, Willecke K, Kurtz A (2011) The connexin
54. Kim SM, Mizel D, Huang YG, Briggs JP, Schnermann J (2006) 40 A96S mutation causes renin-dependent hypertension. J Am
Adenosine as a mediator of macula densa-dependent inhibition of Soc Nephrol 22:1031–1040
renin secretion. Am J Physiol Renal Physiol 290:F1016–F1023 74. Machura K, Neubauer B, Steppan D, Kettl R, Gross A, Kurtz A
55. Kim SM, Theilig F, Qin Y, Cai T, Mizel D, Faulhaber-Walter R, (2012) Role of blood pressure to mediate the influence of salt
Hirai H, Bachmann S, Briggs JP, Notkins AL, Schnermann J intake on renin expression in the kidney. Am J Physiol Renal
(2009) Dense-core vesicle proteins IA-2 and IA-2{beta} affect Physiol 302:F1278–F1285
renin synthesis and secretion through the {beta}-adrenergic path- 75. Madsen K, Friis UG, Gooch JL, Hansen PB, Holmgaard L, Skott
way. Am J Physiol Renal Physiol 296:F382–F389 O, Jensen BL (2010) Inhibition of calcineurin phosphatase pro-
56. Kirchheim HR, Finke R, Hackenthal E, Lowe W, Persson P motes exocytosis of renin from juxtaglomerular cells. Kidney Int
(1985) Baroreflex sympathetic activation increases threshold 77:110–117
pressure for the pressure-dependent renin release in conscious 76. Matsusaka T, Nishimura H, Utsunomiya H, Kakuchi J, Niimura
dogs. Pflugers Arch 405:127–135 F, Inagami T, Fogo A, Ichikawa I (1996) Chimeric mice carrying
57. Kurtz A (2011) Renin release: sites, mechanisms, and control. ‘regional’ targeted deletion of the angiotensin type 1A receptor
Annu Rev Physiol 73:377–399 gene. Evidence against the role for local angiotensin in the in vivo
58. Kurtz A, Della Bruna R, Kuhn K (1988) Cyclosporine A enhan- feedback regulation of renin synthesis in juxtaglomerular cells. J
ces renin secretion and production in isolated juxtaglomerular Clin Invest 98:1867–1877
cells. Kidney Int 33:947–953 77. Medrano S, Monteagudo MC, Sequeira-Lopez ML, Pentz ES,
59. Kurtz A, Della Bruna R, Pfeilschifter J, Bauer C (1988) Role of Gomez RA (2012) Two microRNAs, miR-330 and miR-125b-5p,
cGMP as second messenger of adenosine in the inhibition of mark the juxtaglomerular cell and balance its smooth muscle
renin release. Kidney Int 33:798–803 phenotype. Am J Physiol Renal Physiol 302:F29–F37
60. Kurtz A, Della Bruna R, Pfeilschifter J, Taugner R, Bauer C 78. Mendez M, Gross KW, Glenn ST, Garvin JL, Carretero OA
(1986) Atrial natriuretic peptide inhibits renin release from juxta- (2011) Vesicle-associated membrane protein-2 (VAMP2) medi-
glomerular cells by a cGMP-mediated process. Proc Natl Acad ates cAMP-stimulated renin release in mouse juxtaglomerular
Sci U S A 83:4769–4773 cells. J Biol Chem 286:28608–28618
61. Kurtz A, Gotz KH, Hamann M, Wagner C (1998) Stimulation of 79. Mercure C, Lacombe MJ, Khazaie K, Reudelhuber TL (2010)
renin secretion by nitric oxide is mediated by phosphodiesterase Cathepsin B is not the processing enzyme for mouse prorenin.
3. Proc Natl Acad Sci U S A 95:4743–4747 Am J Physiol Regul Integr Comp Physiol 298:R1212–R1216
62. Kurtz L, Madsen K, Kurt B, Jensen BL, Walter S, Banas B, Wagner 80. Mundel P, Bachmann S, Bader M, Fischer A, Kummer W, Mayer
C, Kurtz A (2010) High-level connexin expression in the human B, Kriz W (1992) Expression of nitric oxide synthase in kidney
juxtaglomerular apparatus. Nephron Physiol 116:p1–p8 macula densa cells. Kidney Int 42:1017–1019
63. Kurtz A, Penner R (1989) Angiotensin II induces oscillations of 81. Neubauer B, Machura K, Chen M, Weinstein LS, Oppermann M,
intracellular calcium and blocks anomalous inward rectifying Sequeira-Lopez ML, Gomez RA, Schnermann J, Castrop H,
potassium current in mouse renal juxtaglomerular cells. Proc Natl Kurtz A, Wagner C (2009) Development of vascular renin ex-
Acad Sci U S A 86:3423–3427 pression in the kidney critically depends on the cyclic AMP
64. Kurtz A, Penner R (1990) Effects of angiotensin II on intracellu- pathway. Am J Physiol Renal Physiol 296:F1006–F1012
lar calcium and electrical function of mouse renal juxtaglomerular 82. Nielsen AH, Poulsen K (1988) Is prorenin of physiological and
cells. Kidney Int Suppl 30:S51–S54 clinical significance? J Hypertens 6:949–958
65. Kurtz A, Pfeilschifter J, Bauer C (1984) Is renin secretion gov- 83. Nusing RM, Treude A, Weissenberger C, Jensen B, Bek M,
erned by the calcium permeability of the juxtaglomerular cell Wagner C, Narumiya S, Seyberth HW (2005) Dominant role of
membrane? Biochem Biophys Res Commun 124:359–366 prostaglandin E2 EP4 receptor in furosemide-induced salt-losing
66. Kurtz A, Schweda F (2006) Osmolarity-induced renin secretion tubulopathy: a model for hyperprostaglandin E syndrome/antena-
from kidneys: evidence for readily releasable renin pools. Am J tal Bartter syndrome. J Am Soc Nephrol 16:2354–2362
Physiol Renal Physiol 290:F797–F805 84. Ogawa K, Yamasato M, Taniguchi K (1995) Exocytosis of secre-
67. Kurtz L, Schweda F, de Wit C, Kriz W, Witzgall R, Warth R, Sauter tory granules in the juxtaglomerular granular cells of kidneys.
A, Kurtz A, Wagner C (2007) Lack of connexin 40 causes displace- Anat Rec 243:336–346
ment of renin-producing cells from afferent arterioles to the extra- 85. Ortiz-Capisano MC, Liao TD, Ortiz PA, Beierwaltes WH (2009)
glomerular mesangium. J Am Soc Nephrol 18:1103–1111 Calcium-dependent phosphodiesterase 1C inhibits renin release
68. Lai EY, Wang Y, Persson AE, Manning RD Jr, Liu R (2011) from isolated juxtaglomerular cells. Am J Physiol Regul Integr
Pressure induces intracellular calcium changes in juxtaglomerular Comp Physiol 297:R1469–R1476
cells in perfused afferent arterioles. Hypertens Res 34:942–948 86. Ortiz-Capisano MC, Ortiz PA, Harding P, Garvin JL, Beierwaltes
69. Laske-Ernst J, Stehle A, Vallon V, Quast U, Russ U (2008) Effect WH (2007) Adenylyl cyclase isoform v mediates renin release
of adenosine on membrane potential and Ca2+ in juxtaglomerular from juxtaglomerular cells. Hypertension 49:618–624
cells. Comparison with angiotensin II. Kidney Blood Press Res 87. Ortiz-Capisano MC, Ortiz PA, Harding P, Garvin JL, Beierwaltes
31:94–103 WH (2007) Decreased intracellular calcium stimulates renin re-
70. Leichtle A, Rauch U, Albinus M, Benohr P, Kalbacher H, Mack lease via calcium-inhibitable adenylyl cyclase. Hypertension
AF, Veh RW, Quast U, Russ U (2004) Electrophysiological and 49:162–169
molecular characterization of the inward rectifier in juxtaglomer- 88. Persson BE, Sakai T, Marsh DJ (1988) Juxtaglomerular intersti-
ular cells from rat kidney. J Physiol 560:365–376 tial hypertonicity in Amphiuma: tubular origin-TGF signal. Am J
71. Lenz T, Sealey JE, Maack T, James GD, Heinrikson RL, Marion Physiol 254:F445–F449
D, Laragh JH (1991) Half-life, hemodynamic, renal, and hormon- 89. Peti-Peterdi J, Fintha A, Fuson AL, Tousson A, Chow RH (2004)
al effects of prorenin in cynomolgus monkeys. Am J Physiol 260: Real-time imaging of renin release in vitro. Am J Physiol Renal
R804–R810 Physiol 287:F329–F335
36 Pflugers Arch - Eur J Physiol (2013) 465:25–37

90. Peti-Peterdi J, Komlosi P, Fuson AL, Guan Y, Schneider A, Qi Z, 111. Schweda F, Klar J, Narumiya S, Nusing RM, Kurtz A (2004)
Redha R, Rosivall L, Breyer MD, Bell PD (2003) Luminal NaCl Stimulation of renin release by prostaglandin E2 is mediated by
delivery regulates basolateral PGE2 release from macula densa EP2 and EP4 receptors in mouse kidneys. Am J Physiol Renal
cells. J Clin Invest 112:76–82 Physiol 287:F427–F433
91. Pratt RE, Flynn JA, Hobart PM, Paul M, Dzau VJ (1988) Differ- 112. Schweda F, Segerer F, Castrop H, Schnermann J, Kurtz A (2005)
ent secretory pathways of renin from mouse cells transfected with Blood pressure-dependent inhibition of Renin secretion requires
the human renin gene. J Biol Chem 263:3137–3141 A1 adenosine receptors. Hypertension 46:780–786
92. Rasch R, Jensen BL, Nyengaard JR, Skott O (1998) Quantitative 113. Schweda F, Wagner C, Kramer BK, Schnermann J, Kurtz A
changes in rat renin secretory granules after acute and chronic (2003) Preserved macula densa-dependent renin secretion in A1
stimulation of the renin system. Cell Tissue Res 292:563–571 adenosine receptor knockout mice. Am J Physiol Renal Physiol
93. Reid IA (1999) Role of phosphodiesterase isoenzymes in the 284:F770–F777
control of renin secretion: effects of selective enzyme inhibitors. 114. Sequeira Lopez ML, Pentz ES, Nomasa T, Smithies O, Gomez
Curr Pharm Des 5:725–735 RA (2004) Renin cells are precursors for multiple cell types that
94. Ritthaler T, Scholz H, Ackermann M, Riegger G, Kurtz A, Kramer switch to the renin phenotype when homeostasis is threatened.
BK (1995) Effects of endothelins on renin secretion from isolated Dev Cell 6:719–728
mouse renal juxtaglomerular cells. Am J Physiol 268:F39–F45 115. Sequeira-Lopez ML, Weatherford ET, Borges GR, Monteagudo
95. Sallstrom J, Jensen BL, Skott O, Gao X, Persson AE (2010) MC, Pentz ES, Harfe BD, Carretero O, Sigmund CD, Gomez RA
Neuronal nitric oxide synthase supports renin release during (2010) The microRNA-processing enzyme dicer maintains juxta-
sodium restriction through inhibition of phosphodiesterase 3. glomerular cells. J Am Soc Nephrol 21:460–467
Am J Hypertens 23:1241–1246 116. Skinner SL, McCubbin JW, Page IH (1963) Renal baroceptor
96. Salomonsson M, Skott O, Persson AE (1991) Influence of intra- control of renin secretion. Science 141:814–816
luminal arterial pressure on renin release. Acta Physiol Scand 117. Skott O (1986) Episodic release of renin from single isolated
141:285–286 superfused rat afferent arterioles. Pflugers Arch 407:41–45
97. Sandner P, Kornfeld M, Ruan X, Arendshorst WJ, Kurtz A (1999) 118. Skott O, Baumbach L (1985) Effects of adenosine on renin
Nitric oxide/cAMP interactions in the control of rat renal vascular release from isolated rat glomeruli and kidney slices. Pflugers
resistance. Circ Res 84:186–192 Arch 404:232–237
98. Schnermann J (1998) Juxtaglomerular cell complex in the regu- 119. Skott O, Briggs JP (1987) Direct demonstration of macula densa-
lation of renal salt excretion. Am J Physiol 274:R263–R279 mediated renin secretion. Science 237:1618–1620
99. Schnermann J, Weihprecht H, Briggs JP (1990) Inhibition of 120. Skott O, Taugner R (1987) Effects of extracellular osmolality on
tubuloglomerular feedback during adenosine1 receptor blockade. renin release and on the ultrastructure of the juxtaglomerular
Am J Physiol 258:F553–F561 epithelioid cell granules. Cell Tissue Res 249:325–329
100. Scholz H, Hamann M, Gotz KH, Kurtz A (1994) Role of calcium 121. Taugner R, Buhrle CP, Nobiling R (1984) Ultrastructural changes
ions in the pressure control of renin secretion from the kidneys. associated with renin secretion from the juxtaglomerular appara-
Pflugers Arch 428:173–178 tus of mice. Cell Tissue Res 237:459–472
101. Scholz H, Kurtz A (1992) Disparate effects of calcium channel 122. Taugner R, Mannek E, Nobiling R, Buhrle CP, Hackenthal E,
blockers on pressure dependence of renin secretion and flow in Ganten D, Inagami T, Schroder H (1984) Coexistence of renin
the isolated perfused rat kidney. Pflugers Arch 421:155–162 and angiotensin II in epitheloid cell secretory granules of rat
102. Scholz H, Kurtz A (1993) Involvement of endothelium-derived kidney. Histochemistry 81:39–45
relaxing factor in the pressure control of renin secretion from 123. Taugner R, Metz R (1986) Development and fate of the secretory
isolated perfused kidney. J Clin Invest 91:1088–1094 granules of juxtaglomerular epithelioid cells. Cell Tissue Res
103. Scholz H, Kurtz A (1995) Differential regulation of cytosolic 246:595–606
calcium between afferent arteriol ar smooth muscle cells from 124. Taugner R, Whalley A, Angermuller S, Buhrle CP, Hackenthal E
mouse kidney. Pflugers Arch 431:46–51 (1985) Are the renin-containing granules of juxtaglomerular ep-
104. Schricker K, Della Bruna R, Kurtz A (1993) Extracellular calci- ithelioid cells modified lysosomes? Cell Tissue Res 239:575–587
um exerts a dual effect on renin secretion from isolated mouse 125. Tobian L (1960) Physiology of the juxtaglomerular cells. Ann
juxtaglomerular cells. Pflugers Arch 423:14–20 Intern Med 52:395–410
105. Schricker K, Hamann M, Kurtz A (1995) Nitric oxide and pros- 126. Traynor TR, Smart A, Briggs JP, Schnermann J (1999) Inhibition
taglandins are involved in the macula densa control of the renin of macula densa-stimulated renin secretion by pharmacological
system. Am J Physiol 269:F825–F830 blockade of cyclooxygenase-2. Am J Physiol 277:F706–F710
106. Schricker K, Hegyi I, Hamann M, Kaissling B, Kurtz A (1995) 127. Twu BM, Cannon ME, Hsu CH (1989) The role of osmolarity in
Tonic stimulation of renin gene expression by nitric oxide is renin release from afferent arterioles. J Hypertens 7:757–761
counteracted by tonic inhibition through angiotensin II. Proc Natl 128. Vander AJ, Miller R (1964) Control of renin secretion in the
Acad Sci U S A 92:8006–8010 anesthetized dog. Am J Physiol 207:537–546
107. Schricker K, Holmer S, Hamann M, Riegger G, Kurtz A (1994) 129. Voskuil M, Verloop WL, Blankestijn PJ, Agostoni P, Stella PR,
Interrelation between renin mRNA levels, renin secretion, and blood Doevendans PA (2011) Percutaneous renal denervation for the
pressure in two-kidney, one clip rats. Hypertension 24:157–162 treatment of resistant essential hypertension; the first Dutch ex-
108. Schricker K, Kurtz A (1993) Liberators of NO exert a dual effect perience. Neth Heart J 19:319–323
on renin secretion from isolated mouse renal juxtaglomerular 130. Wagner C, de Wit C, Kurtz L, Grunberger C, Kurtz A, Schweda F
cells. Am J Physiol 265:F180–F186 (2007) Connexin40 is essential for the pressure control of renin
109. Schricker K, Kurtz A (1996) Blockade of nitric oxide formation synthesis and secretion. Circ Res 100:556–563
inhibits the stimulation of the renin system by a low salt intake. 131. Wagner C, Pfeifer A, Ruth P, Hofmann F, Kurtz A (1998) Role of
Pflugers Arch 432:187–191 cGMP-kinase II in the control of renin secretion and renin ex-
110. Schricker K, Potzl B, Hamann M, Kurtz A (1996) Coordinate pression. J Clin Invest 102:1576–1582
changes of renin and brain-type nitric-oxide-synthase (b-NOS) 132. Weihprecht H, Lorenz JN, Schnermann J, Skott O, Briggs JP
mRNA levels in rat kidneys. Pflugers Arch 432:394–400 (1990) Effect of adenosine1-receptor blockade on renin release
Pflugers Arch - Eur J Physiol (2013) 465:25–37 37

from rabbit isolated perfused juxtaglomerular apparatus. J Clin 136. Yang T, Park JM, Arend L, Huang Y, Topaloglu R, Pasumarthy
Invest 85:1622–1628 A, Praetorius H, Spring K, Briggs JP, Schnermann J (2000)
133. Wilcox CS, Peart WS (1987) Release of renin and angiotensin II Low chloride stimulation of prostaglandin E2 release and
into plasma and lymph during hyperchloremia. Am J Physiol cyclooxygenase-2 expression in a mouse macula densa cell
253:F734–F741 line. J Biol Chem 275:37922–37929
134. Xu D, Borges GR, Davis DR, Agassandian K, Sequeira Lopez 137. Yang T, Singh I, Pham H, Sun D, Smart A, Schnermann JB,
ML, Gomez RA, Cassell MD, Grobe JL, Sigmund CD (2011) Briggs JP (1998) Regulation of cyclooxygenase expression in
Neuron- or glial-specific ablation of secreted renin does not affect the kidney by dietary salt intake. Am J Physiol 274:F481–
renal renin, baseline arterial pressure, or metabolism. Physiol F489
Genomics 43:286–294 138. Yi F, Zhang AY, Li N, Zhang F, Xia M, Li PL (2007) Role
135. Yang T, Endo Y, Huang YG, Smart A, Briggs JP, Schnermann J of cyclic ADP-ribose-Ca2+ signaling in mediating renin pro-
(2000) Renin expression in COX-2-knockout mice on normal or duction and release in As4.1 cells. Cell Physiol Biochem
low-salt diets. Am J Physiol Renal Physiol 279:F819–F825 19:293–302

Você também pode gostar