ArticlePDF Available

Lack of Connexin 40 Causes Displacement of Renin-Producing Cells from Afferent Arterioles to the Extraglomerular Mesangium

Authors:
  • Medizinische Fakultät Mannheim der Universität Heidelberg

Abstract and Figures

In the adult kidney, renin-producing cells are typically located in the walls of afferent arterioles at the transition into the glomerular capillary network. The mechanisms that are responsible for restricting renin expression to the juxtaglomerular position are largely unknown. This study showed that in mice that lack connexin 40 (Cx40), the predominant connexin of renin-producing cells, renin-positive cells are absent in the vessel walls and instead are found in cells of the extraglomerular mesangium, glomerular tuft, and periglomerular interstitium. Blocking macula densa transport function by acute administration of loop diuretics strongly enhances renin secretion in vivo and in isolated perfused kidneys of wild-type mice. This effect of loop diuretics is markedly attenuated in vivo and even blunted in vitro in Cx40-deficient mice. Even after prolonged stimulation of renin secretion by severe sodium depletion, renin expression is not seen in juxtaglomerular cells or in cells of more proximal parts of the arterial vessel wall as occurs normally. Instead, renin remains restricted to the extra-/periglomerular interstitium in Cx40-deficient mice. In contrast to the striking displacement of renin-expressing cells in the adult kidney, renin expression in the vessels of the developing kidney was found to be normal. This is the first evidence to indicate that cell-to-cell communication via gap junctions is essential for the correct juxtaglomerular positioning and recruitment of renin-producing cells. Moreover, these findings support the notion that gap junctions are relevant for the macula densa signaling to renin-producing cells.
Content may be subject to copyright.
Lack of Connexin 40 Causes Displacement of Renin-
Producing Cells from Afferent Arterioles to the
Extraglomerular Mesangium
Lisa Kurtz,* Frank Schweda,* Cor de Wit,
Wilhelm Kriz,
Ralph Witzgall,*
Richard Warth,* Alexander Sauter,* Armin Kurtz,* and Charlotte Wagner*
*Institut fu¨r Physiologie and Anatomie, Universita¨t Regensburg, Regensburg,
Institut fu¨r Physiologie, Universita¨t
Lu¨beck, Lu¨beck, and
Institut fu¨r Anatomie und Embryologie, Universita¨t Heidelberg, Heidelberg, Germany
In the adult kidney, renin-producing cells are typically located in the walls of afferent arterioles at the transition into the
glomerular capillary network. The mechanisms that are responsible for restricting renin expression to the juxtaglomerular
position are largely unknown. This study showed that in mice that lack connexin 40 (Cx40), the predominant connexin of
renin-producing cells, renin-positive cells are absent in the vessel walls and instead are found in cells of the extraglomerular
mesangium, glomerular tuft, and periglomerular interstitium. Blocking macula densa transport function by acute adminis-
tration of loop diuretics strongly enhances renin secretion in vivo and in isolated perfused kidneys of wild-type mice. This
effect of loop diuretics is markedly attenuated in vivo and even blunted in vitro in Cx40-deficient mice. Even after prolonged
stimulation of renin secretion by severe sodium depletion, renin expression is not seen in juxtaglomerular cells or in cells of
more proximal parts of the arterial vessel wall as occurs normally. Instead, renin remains restricted to the extra-/periglomerular
interstitium in Cx40-deficient mice. In contrast to the striking displacement of renin-expressing cells in the adult kidney, renin
expression in the vessels of the developing kidney was found to be normal. This is the first evidence to indicate that cell-to-cell
communication via gap junctions is essential for the correct juxtaglomerular positioning and recruitment of renin-producing
cells. Moreover, these findings support the notion that gap junctions are relevant for the macula densa signaling to
renin-producing cells.
J Am Soc Nephrol 18: 1103–1111, 2007. doi: 10.1681/ASN.2006090953
T
he aspartyl-protease renin is the regulating key enzyme
of the renin-angiotensin-aldosterone system, which
controls BP and extracellular volume. Renin is predom-
inantly produced by the kidneys. Renin-producing cells of the
kidneys show a high degree of plasticity. The cells are com-
monly considered to be a special subset of transformed vascu-
lar smooth muscle cells (VSMC) that are related to myofibro-
blasts or pericytes (1). During angiogenesis and vasculogenesis
of the kidneys, renin-producing cells cover most of the arterial
vascular tree (1– 6). During vessel maturation, renin expression
in proximal parts of the arteriole is progressively silenced so
that in the adult kidney, it is condensed in juxtaglomerular cells
of the ultimate part of the afferent arterioles (1). In this segment
of the arteriole, renin-producing cells largely replace the typical
VSMC. Because of the high number of renin storage vesicles,
the cells achieve a cobble-stone like “epithelioid” appearance.
Chronic stimulation of the renin-angiotensin-aldosterone sys-
tem by extracellular volume depletion or increased sympathetic
nerve activity is associated with re-emergence of renin expres-
sion in cells in the walls of larger renal arteries in a pattern that
is not fully predictable (7–12). The cellular mechanisms that are
responsible for initiating or terminating renin expression dur-
ing vasculogenesis, for directing renin expression to the glo-
merular vascular pole in the adult kidney, and for restarting
renin expression in some but not all cells of larger vessels are
not yet understood. Although local humoral as well as biome-
chanical factors have been considered in this context, no clear
concept has emerged.
In this study, we considered the role of cell-to-cell commu-
nication as a novel determinant of focal renal renin expression.
Previous work established that cell-to-cell communication
through gap junctions is important for the positioning and
differentiation of resident cells (13–19). Within the juxtaglomer-
ular apparatus (JGA), renin-producing cells not only form nu-
merous gap junctions among each other but also are connected
to extraglomerular cells and to endothelial cells of the afferent
arteriole (20–22). The gap junctions of renin-producing cells are
likely formed by connexin 40 (Cx40), which is expressed with
high density in these cells (23–26). The prominent expression of
Cx40 in the JGA is conspicuous, because with the exception of
the electrical conduction system of the heart, Cx40 is almost
exclusively expressed in the endothelium, where it contributes
to propagation of vasodilation (27–31). In the (juxta)glomerular
region, however, Cx40 is expressed by both endothelial and
nonendothelial cells such as renin-producing and intra- and
Received September 3, 2006. Accepted January 18, 2007.
Published online ahead of print. Publication date available at www.jasn.org.
Address correspondence to: Dr. Armin Kurtz, Physiologisches Institut der Uni-
versita¨t Regensburg, D-93040 Regensburg, Germany. Phone: 49-0-941-9432980;
Fax: 49-0-941-9434315; E-mail: armin.kurtz@vkl.uni-regensburg.de
Copyright © 2007 by the American Society of Nephrology ISSN: 1046-6673/1804-1103
extraglomerular mesangial cells (23–26). Other typical endothe-
lial connexins, such as Cx43, are found in larger kidney vessels
but not in the juxtaglomerular endothelial and renin-producing
cells (23–26). In this study, we used mice that lacked Cx40 to
explore the possibility that gap junctional coupling may be
required for the differentiation and positioning of renin-pro-
ducing cells during renal angiogenesis and vasculogenesis, for
the condensation of renin to the JGA of the adult kidney, and
for the recruitment of renin-expressing cells during prolonged
challenges. Moreover, it was of interest for us to assess the role
of Cx40 for the signaling of macula densa cells to renin-pro-
ducing cells, a process that is considered to involve gap junc-
tional function (32).
Materials and Methods
All animal experiments were conducted according to the National
Institutes of Health guidelines for the care and use of animals in
research. Kidneys were sampled from eight 12- to 20-wk-old male
homozygous Cx40/ mice (33), from three fetuses (days 17 to 19),
and from three pups (postnatal day 1). Age-matched wild-type (wt)
mice served as controls. In addition, five adult male Cx40/ mice
were examined. Furthermore, five adult male mice of the Cx40/
and Cx40/ strains were pretreated with a low-salt diet (0.02%
wt/wt) for 1 wk. In addition, these mice received the angiotensin-
converting enzyme inhibitor (ACE) enalapril (10 mg/kg) via the drink-
ing water for the last3dofthedietary treatment. The genetic back-
ground of Cx40-deficient and wt mice was considered identical because
Cx40-deficient mice were backcrossed for seven generations on a
C57Bl/6 background. The genotype of the mice was verified by PCR as
described previously (34).
For assessment of the macula densa control of renin secretion in vivo,
Cx40/ and Cx40/ mice of either gender (25 to 30 g body wt)
were used (Cx40/ seven male and seven female mice; Cx40/
eight male and eight female mice). For determination of plasma renin
concentration (PRC), blood samples were taken from the tail vein. Ten
days after baseline blood collection, all mice received a single injection
of furosemide (40 mg/kg body wt; Dimazon, Intervet, Germany) and a
blood sample was taken 60 min thereafter from the tail vein.
Immunohistochemistry for Renin and
-Smooth Muscle Actin
The expression of renin and
-smooth muscle actin (
-SMA) was
localized by immunohistochemistry. In brief, kidneys were fixed in
methyl-Carnoy solution (60% methanol, 30% chloroform, and 10% gla-
cial acetic acid) as described previously (35). Immunolabeling was
performed on 5-
m paraffin sections. After blocking with 10% horse
serum and 1% BSA in PBS, sections were incubated with anti-renin or
anti–
-SMA antibodies (Beckman Coulter, Immunotech, Marseilles,
France) overnight at 4°C, followed by incubation with a fluorescence
secondary antibody.
Confocal Microscopy
Sections were analyzed with a confocal microscope (LSM 510; Zeiss,
Go¨ttingen, Germany) using sequential scanning (Plan Apochromat
63/1.4 oil objective, excitation at 488 and 543 nm, emission at 505 to
530 and 560 to 615 nm, respectively).
Bromodeoxyuridine Incorporation
Bromodeoxyuridine (BrdU; Roche, Mannheim, Germany) incorpora-
tion into nuclei was determined as described previously (36). In brief,
BrdU dissolved in sterile isotonic saline (10 mg/ml) was injected twice
daily (0.1 mg BrdU/g body wt per d, intraperitoneally) starting on the
day before the enalapril treatment period. After a total of 7 d, the mice
were anesthetized with an intraperitoneal injection of 100 mg/kg 5-eth-
yl-5-(1-methylbutyl)-2-thiobarbituric acid and 80 mg/kg ketamine-HCl
and fixed by vascular perfusion with 4% paraformaldehyde. After the
removal of kidneys and intestine and the embedding in paraffin, 5-
m
sections were incubated in 4 M HCl for 30 min, neutralized by 0.1 M
sodium borate (pH 8.5), and finally incubated in 0.1% trypsin in PBS.
After blocking with 2% BSA and 0.1% TritonX-100 in PBS, sections were
incubated with anti-BrdU antibody (1:1000) overnight, followed by
incubation with a fluorescence secondary antibody.
Isolated Perfused Mouse Kidney
Male Cx40/ and age-matched Cx40/ mice were used as kid-
ney donors. The isolated perfused mouse kidney model was described
in detail previously (37). Briefly, the mice were anesthetized with an
intraperitoneal injection of 12 mg/kg xylazine (Rompun; Bayer, Wup-
pertal, Germany) and 80 mg/kg ketamine-HCl (Curamed, Karlsruhe,
Germany); the abdominal aorta was cannulated; and the right kidney
was excised, placed in a thermostated moistening chamber, and per-
fused at constant pressure (90 mmHg). Finally, the renal vein was
cannulated and the venous effluent was collected for determination of
renin activity and venous blood flow.
The basic perfusion medium consisted of a modified Krebs-Henseleit
solution supplemented with 6 g/100 ml BSA and with freshly washed
human red blood cells (10% hematocrit). Stock solutions of isoproter-
enol were dissolved in freshly prepared perfusate; stock solution of
bumetanide was made in DMSO. All drugs were infused into the
arterial limb of the perfusion circuit.
For the determination of renin secretion rates, three samples of the
venous effluent were taken in intervals of 2 min during each experi-
mental period. Renin activity in the venous effluent was determined by
RIA (Byk & DiaSorin Diagnostics, Dietzenbach, Germany) as described
previously. Renin secretion rates were calculated as the product of the
renin activity and the venous flow rate (ml/min g kidney weight).
Determination of PRC
For determination of PRC, the blood samples that were taken from
the tail vein were centrifuged and the plasma was incubated for 1.5 h
at 37°C with plasma from bilaterally nephrectomized male rats as renin
substrate. The generated angiotensin I (ng/ml per h) was determined
by RIA (Byk & DiaSorin Diagnostics).
Electron Microscopy
Kidneys were perfusion-fixed and embedded in Epon according to
standard procedures. One-micrometer sections of several blocks in-
cluding a series of 200 1-
m sections were cut with a diamond knife,
stained with methylene blue (38), and studied by light microscopy.
Ultrathin sections were prepared from selected areas and studied by a
Philips 307 transmission electron microscope (Eindhoven, The Nether-
lands).
Three-Dimensional Reconstruction
Serial sections of kidney specimen were fixed and stained for renin
and for
-SMA as described in the previous section. A three-dimen-
sional (3-D) reconstruction of renin immunoreactivity and of
-SMA
immunoreactivity was performed using the Amira 3.1 visualization
program (Mercury Systems, Merignac, France).
1104 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1103–1111, 2007
Results
In adult kidneys of Cx40/ mice, renin-expressing cells
were restricted to the vascular poles of the glomeruli. Renin-
producing cells were integrated into the regular wall of the
afferent arterioles in continuation of the vessel lined by VSMC
(Figure 1, A and B). The latter cells were identified by the
expression of
-SMA (Figure 1, A and B). Renin-producing cells
at the very end of the afferent arterioles showed the typical
epithelioid appearance (juxtaglomerular epithelioid cells) and
expressed no
-SMA. Mixed-phenotype cells that expressed
both renin and
-SMA were found to be located between the
typical SMC and juxtaglomerular epithelioid cells (Figure 1, A
and B). The distribution of renin-expressing cells in Cx40/
kidneys was very similar to that in kidneys of wt mice (data not
shown).
In contrast, the number of renin-expressing cells in adult
kidneys of Cx40-deficient mice was seemingly increased when
compared with wt controls. The number of renin-producing
cells in individual periglomerular regions of Cx40/ kidneys
was variable, ranging from 1 to 20 cells per glomerular region.
Renin-positive cells in Cx40/ kidneys were restricted to the
juxtaglomerular/periglomerular interstitium (Figure 1, C and
D). As confirmed by confocal microscopy, renin-producing
cells in the juxtaglomerular region were not integrated into the
wall of afferent arterioles but instead surrounded the vessel
wall that consisted of endothelial cells and
-SMA–positive
VSMC (Figure 1, E and F). Moreover, renin-producing cells
extended into the region of the extraglomerular mesangium
(EGM) and into the periglomerular interstitial space between
tubules and around glomeruli. The cells did not show the
classic epithelioid appearance but rather appeared mesen-
chyme-like and irregularly shaped. The cytosol of these cells
showed renin immunoreactive granulation. All renin-positive
cells stained negative for
-SMA. No renin-producing cells
were associated with larger kidney vessels (Figure 1, C and D).
Electron microscopic analysis confirmed that walls of affer-
ent arterioles were free of granular cells (Figure 2C). Further-
more, VSMC were found inside the glomerular stalk of
Cx40/ kidneys, whereas they normally terminate at the
entrance into the JGA. VSMC were also observed in the intra-
glomerular segment of the efferent arteriole that is normally
surrounded by mesangial cells and mesangial cell processes. In
contrast to wt kidneys, the majority of extraglomerular mesan-
gial cells in Cx40/ mice contained granules as they are
normally seen in renin-producing cells of the afferent arteriole
(Figure 2, A through D). For example, tracing of an entire
glomerulus in a series of 1-
m sections revealed no cells in the
EGM without granules (Figure 2B). In addition, granular cells
were found in the glomerular stalk to a variable extent. Occa-
sionally, the intraglomerular segment of the efferent arteriole
deep within the glomerular tuft also appeared granulated.
In other sites, EGM cells that looked fairly normal with
elongated processes and without any granules were found
(Figure 2E). The interstitial spaces between these cells seemed
wider and were filled with a translucent amorphous matrix.
Gap junctions were not encountered. Within the intraglomeru-
lar mesangium, foci of mesangiolysis that may be the result of
mesangial cell proliferation and matrix production, leading to
local mesangial expansions, were noted. The resulting narrow-
ing of associated capillaries and their incorporation into the
proliferating mesangium may cause obstruction and eventual
degeneration.
In Cx40/ mice that received a low-sodium (0.02% wt/wt)
diet and the ACEI ramipril, a clear increase of renin-expressing
cells appeared in the walls of the preglomerular vessels (Figure
3, A and B). Cells that expressed both
-SMA and renin could
be seen with increased frequency. In contrast, no expression of
renin was found in the walls of the arterioles or larger vessels
of Cx40/ kidneys (Figure 3, C and D). Instead, the number
of renin-expressing periglomerular cells seemed to be in-
Figure 1. Immunohistochemistry for renin (green) and
-smooth muscle actin (
-SMA; red) on kidney sections of
wild-type (wt; A and B) and of connexin 40 deficient
(Cx40/; C and D) mice. In wt mice, renin immunoreactivity
was found in the walls of afferent arterioles close to the glo-
meruli (dotted, G). Renin-positive cells had a regular form,
appeared thicker, and were negative for
-SMA at the very end
of the afferent arterioles. In Cx40/ kidneys, the number of
renin-positive cells in the juxtaglomerular region was clearly
increased. The cells had a more irregular shape, and they were
not integrated into the walls of the afferent arterioles. Impres-
sion was further corroborated by a confocal analysis (E and F)
of kidney sections from Cx40/ mice, showing that renin-
positive cells were separated from endothelial cells by a layer of
-SMA–positive smooth muscle cells. *, Macula densa; arrow-
heads, endothelial cells. Bars 20
m.
J Am Soc Nephrol 18: 1103–1111, 2007 Connexin 40 and Displacement of Renin-Producing Cells 1105
creased. Cx40/ behaved like Cx40/ kidneys (data not
shown).
When the number of renin-expressing cells in the periglo-
merular region was particularly high, a clear expansion of the
periglomerular cell mass could be readily detected by light
microscopy (Figure 3, E and F). Otherwise the histology of the
kidneys was normal, except for the appearance of sclerotic
glomeruli. To determine whether expansion of periglomerular
cell mass was associated with proliferation of interstitial cells,
we determined the number of cells that underwent an S phase
during treatment with low salt and the ACEI. Using the BrdU
incorporation method, we found single BrdU incorporating
nuclei in proximal tubules but no BrdU incorporation into
nuclei of renin-immunoreactive cells, neither in wt nor in
Cx40/ kidneys (data not shown).
Because the extent of periglomerular renin expression was
difficult to estimate from single histologic sections, we per-
formed a 3-D reconstruction of 80 consecutive 5-
m sections
that stained for renin and for
-SMA. With the use of this
technique, no obvious differences between preglomerular ves-
sel trees of wt and Cx40/ kidneys were noted (Figure 4, A
and B). The number of glomeruli within this defined kidney
volume was similar between wt and Cx40/ kidneys. In
low-salt/ACEI-treated wt mice, renin expression was exclu-
sively associated with the walls of the distal parts of the afferent
arterioles, where the renin-producing cells formed cuff-like
Figure 2. (A) High-resolution light microscopy longitudinal sec-
tion (1
m) through the juxtaglomerular apparatus (JGA) and
glomerulus. Granular cells are abundant and are encountered at
abnormal sites in the extraglomerular mesangium (EGM; *) and
the glomerular stalk (arrow). (B) Oblique section through the JGA
showing the abundance of granular cells in the EGM and as
disseminated cells everywhere in the surroundings of the JGA
(arrows). (C) Transmission electron microscopy longitudinal sec-
tions through the JGA. Granular cells (G) are seen within the
EGM; a vascular smooth muscle cell (SM) extends into the glo-
merular stalk. (D) Transmission electron microscopy longitudinal
sections through the JGA. Beneath the macula densa (MD), a
group of EGM cells that contain abundant granules of variable
electron density are seen (arrows); also protogranules (arrow-
heads) are regularly encountered. (E) Transmission electron
microscopy longitudinal sections through the JGA. Beneath the
macula densa are a group of EGM cells and processes that look
fairly normal. Gap junctions have not been encountered be-
tween the EGM cells. AA, afferent arteriole; EA, efferent arte-
riole. Magnifications: 500 in A; 625 in B; 1800 in C; 5700
in D; 4500 in E.
Figure 3. (A through D) Immunohistochemistry for renin
(green) and
-SMA (red) on kidney sections of wt (A and B)
and of Cx40/ (C and D) mice treated with a combination of
low salt and the angiotensin-converting enzyme inhibitor
(ACEI) ramipril. In the kidneys of wt mice, renin expression
was increased in the walls of the preglomerular vessels. Co-
localization of renin and of
-SMA (yellow) was increased. In
kidneys from Cx40/ mice, again no renin immunoreactivity
became visible in the vessel walls. Instead, renin expression
spread out into the periglomerular-peritubular space. (E and F)
Consecutive kidney sections of Cx40/ mice treated with
low salt and the ACEI stained with renin/
-SMA (E) and with
hematoxylin-eosin (F). Bars 20
m.
1106 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1103–1111, 2007
structures at the vascular pole of virtually all glomeruli. In
Cx40/ kidneys, no cuff-like structures were observed. In-
stead, 30% of glomeruli showed a prominent periglomerular
spreading (Figure 4B). In the remaining glomeruli, the number
of renin-expressing cells was markedly lower, but again these
cells were not integrated into the walls of the afferent arterioles.
In view of the aberrant position of renin-producing cells in
the kidneys of adult Cx40/ mice, it was of interest to study
the localization of renin-producing cells during the develop-
ment of the kidneys. Therefore, we performed a 3-D analysis of
the vascular tree and of renin-producing cells of kidneys at day
18 after conception. As shown in Figure 4, C and D, the differ-
ent arteries and arterioles could clearly be identified in embry-
onic kidneys of wt and Cx40/ mice. There were no obvious
differences in the vascular morphology in the two genotypes.
Similarly, the distribution of renin-producing cells in these
fetal kidneys was very similar between the genotypes. The
renin-producing cells were associated with the walls of the
larger arteries. Most notable, renin expression in Cx40/
kidneys was restricted to the vessel wall and did not appear in
the interstitium (Figure 4, C and D). The first clearly recogniz-
able divergence in the localization of renin-producing cells
between the two genotypes appeared with the development of
renin expression in the glomerular regions. Figure 5, B through
D, shows that in the kidneys of 1-d-old Cx40/ pups, renin-
expressing cells are still integrated into the walls of afferent
arterioles, although they start to spread out at the vascular
poles. In wt kidneys of the same age, renin expression remained
restricted to the vessel wall of the preglomerular arteries (Fig-
ure 5A).
Considering these apparent changes of the architecture of the
JGA raises the question about functional changes. It was of
particular interest for us to see whether the normal signaling of
the macula densa cells to renin-producing cells is altered in the
absence of Cx40. To study the macula densa control of renin
secretion, we used a classical maneuver, the acute administra-
tion of loop diuretics, which blocks salt transport in the thick
ascending loop of Henle including the macula densa segment.
In Cx40/ mice, application of furosemide led to a 15-fold
increase of PRC (Figure 6A). Similarly, also in isolated perfused
kidneys of Cx40/ mice, bumetanide caused a strong en-
hancement of renin secretion (Figure 6B). In Cx40/ mice,
basal PRC were sixfold elevated when compared with
Cx40/ mice (Figure 6A). Administration of furosemide in-
creased PRC values only two-fold in Cx40/ mice (Figure
7A). Renin secretion from isolated perfused kidneys of
Cx40/ mice responded well to isoproterenol, whereas the
response to bumetanide was absent (Figure 6B).
Figure 4. (A and B) Three-dimensional (3-D) reconstruction of
serial kidney sections stained for renin/
-SMA of adult wt (A)
and Cx40/ (B) kidneys. Mice were pretreated with low salt
and ACEI (green, renin; red,
-SMA). (C and D) 3-D reconstruc-
tion of serial kidney sections stained for renin and for
-SMA of
wt (C) and of Cx40/ (D) mice at embryonic day 18 (green,
renin; red,
-SMA). Bars 500
m.
Figure 5. Immunohistochemistry for renin (green) and
-SMA
(red) on kidney sections of wt (A) and Cx40/ (B through D)
mice 1 d after birth. Yellow indicates overlap of renin and
-SMA. Bars 20
m.
J Am Soc Nephrol 18: 1103–1111, 2007 Connexin 40 and Displacement of Renin-Producing Cells 1107
Discussion
A main finding in the present study is that the absence of
Cx40 is associated with a major disturbance in the location and
identity of renin-expressing cells. Cell-to-cell communication
through Cx40 seems to be required for the maintenance of the
architecture of the JGA and the direction of renin expression to
cells in the walls of afferent arterioles in a typical juxtaglomer-
ular location (Figure 6, A and B). In the absence of Cx40,
renin-expressing cells are found outside the terminal parts of
the afferent arterioles and extending into the juxta- and peri-
glomerular interstitium. Furthermore, stimulation of renin syn-
thesis and secretion does not induce the normal recruitment of
renin-expressing cells in the walls of upstream preglomerular
arteries.
Given that Cx40 is the main gap junctional protein in the JGA
(25–28), especially in the region of renin production (Figure
Figure 6. Effect of loop diuretics on renin secretion in vivo (A) and
from isolated perfused kidneys (B) in Cx40/ and Cx40/
mice. (A) Data are means SEM of 14 and 16 Cx40/ and
Cx40/ mice, respectively. Furosemide (40 mg/kg) was in-
jected 10 d after baseline blood collection of each mouse. Plasma
renin concentration values were determined 1 h after injection of
furosemide. #P 0.001 versus baseline Cx40/;*P 0.05 versus
baseline Cx40/. (B) For isolated perfused kidneys, five kidneys
of each genotype were used. Data are means SEM. E,
Cx40/; F, Cx40/. The increases of renin secretion that
were elicited by isoproterenol (iso; 3 nM) were significant (P
0.05) for both genotypes. The change of renin secretion that was
elicited by bumetanide (bum) was significant for Cx40/ only
(P 0.05 versus iso 3 nM alone). The change of renin secretion that
was elicited by iso (10 nM) was significant (P 0.05 versus iso 3
nM bum 100
M) for both genotypes.
Figure 7. Sketch summarizing the morphology of the JGA in the
presence (A) and absence (B) of Cx40. mdc, macula densa cells;
JGE, juxtaglomerular epithelioid cells; ec, endothelial cells; mc,
mesangial cells; gcec, glomerular capillary endothelial cells;
pod, podocytes; vsmc, vascular smooth muscle cells; Bc, Bow-
man capsule.
1108 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1103–1111, 2007
6A), our observations are in accordance with numerous find-
ings of a central role of gap junctions in the differentiation of a
variety of cells in the cardiovascular, nervous, or reproductive
system (13–19). There is general agreement that gap junctional
communication favors structural and functional differentiation,
whereas loss of gap junctions leads to dedifferentiation (13–19).
There is also agreement that gap junctional communication is
important for the correct formation of endo- and exocrine
glands and for coordinating the responses of individual gland
cells (39).
Our data suggest that the mislocation of renin-producing
cells in Cx40/ kidneys may begin with typical expression of
renin in the juxtaglomerular region. Endothelial cells of the
afferent arterioles, renin-producing cells, and mesangial cells
form a network of cells that are connected via Cx40 gap junc-
tions (23–26) (Figure 6C) and that are derived from the juxta/
periglomerular mesenchyme as a group of pericytes. Notably,
all of these cell types, including the extraglomerular and intra-
glomerular mesangial cells (40,41), have the capability to syn-
thesize renin, although the expression of renin in the adult
mesangium is a very rare event (1,42–44).
Because renin expression in the EGM of Cx40/ kidneys is
abundant, Cx40 gap junctions do not seem to be a general
prerequisite for the expression of renin in the pericytes, a
conclusion that is confirmed by normal fetal development.
Because endothelial cells in the afferent arteriole normally form
Cx40 gap junctions with renin-producing cells but not with
VSMC, one may speculate that the lack of Cx40 gap junctions
leads to a transformation of originally renin-producing peri-
cytes into VSMC. As a consequence, remaining renin-produc-
ing cells are displaced to the periphery of the afferent arterioles,
initiating the process of dispersion of renin expression to cells
outside the afferent arteriolar vessel wall. Our findings suggest
that this proliferation period ends once the formation of the
glomeruli is complete.
One may hypothesize that contact between endothelial cells
and renin-producing pericytes via Cx40 gap junctions could
provide an essential anchor for the juxtaglomerular position of
renin-producing cells. Once a stable contact is established, en-
dothelium-coupled pericytes may send a signal through Cx40
gap junctions to neighboring pericytes to suppress their prolif-
eration and renin secretion. The inherent capability to produce
renin, however, would be preserved. In fact, it is known that
long-lasting stimulation of the renin-angiotensin system can
lead to the recruitment of renin production not only in the
larger vessels but also in the EGM (42–47).
The displacement of renin-producing cells raises the question
of whether the physiologic regulation of the renin system is
affected by the absence of Cx40. It is known that Cx40/
mice are hypertensive (48). Although first evidence suggests
that the hypertension in Cx40/ mice is not primarily renin
dependent (48), our observation of elevated PRC values (Figure
7A) could suggest that an exaggerated renin secretion could
contribute to the hypertension. The enhanced basal renin secre-
tion in Cx40/ mice could result from an interruption of the
inhibitory macula densa signaling to renin-producing cells.
Such an explanation would be supported by our findings that
the stimulatory effect of loop diuretics on renin secretion is
absent in isolated kidneys of Cx40/ mice (Figure 7B) and is
markedly attenuated in vivo (Figure 7A). A possible explanation
for why the renin response to loop diuretics in vivo is not
blunted as in vitro is that additional factors may contribute to
the effects of loop diuretics in vivo, such as inhibition of NKCC1
(49,50) or activation of the sympathetic nervous system, which
may be less relevant in vitro. In fact, our in vitro findings suggest
that Cx40/ kidneys respond more sensitively to
-adreno-
receptor activation than do Cx40/ kidneys (Figure 7B).
Altogether, our data show that the macula densa signaling to
renin-producing cells is markedly deteriorated in the absence of
Cx40/, suggesting an important role of this connexin in
macula densa signaling and supporting the conclusions of a
recent elegant imaging study (32).
For further elucidation of the possible underlying mecha-
nisms of the crucial effect of Cx40 for the position of renin-
producing cells and for the macula densa control function, two
strategies will be used. First, in mice with an endothelium-
specific deletion of Cx40, we would expect an interruption of
the communication between endothelial cells and renin-pro-
ducing cells without affecting the communication between re-
nin-producing cells and mesangial cells. Second, replacement
of Cx40 with other related connexins should provide informa-
tion about the connexin subtype that is causal in determining
the position of renin-producing cells.
Acknowledgments
This work was financially supported by the Sonderforschungsbe-
reich 699, by the Deutche Forschungsgemeinschaft grant Wi 2071/1-1,
and by the “Prof. Dr. Karl und Gerhard Schiller-Stiftung, Frankfurt/
Main.”
We thank Jurgen Schnermann for critical reading of the manuscript
and for helpful comments. The expert technical assistance provided by
Anna Ba´ngui, Katharina Machura, and Hiltraud Hosser is gratefully
acknowledged.
Disclosures
None.
References
1. Taugner R, Hackenthal E: The Juxtaglomerular Apparatus,
Springer, Heidelberg, 1989
2. Minuth W, Hackenthal E, Poulsen K: Renin immunocyto-
chemistry of the differentiating juxtaglomerular apparatus.
Anat Embryol 162: 173–181, 1981
3. Gomez RA, Chevalier RL, Sturgill BC: Maturation of the
intrarenal renin distribution in Wistar-Kyoto rats. J Hyper-
tens 4(Suppl 1): S31–S33, 1986
4. Richoux JP, Amsaguine S, Grignon G: Earliest renin-con-
taining cell differentiation during ontogenesis in the rat.
An immunocytological study. Histochemistry 88: 41–46,
1997
5. Reddi V, Zaglul A, Pentz ES, Gomez RA: Renin-expressing
cells are associated with branching of the developing kid-
ney vasculature. J Am Soc Nephrol 9: 63–71, 1998
6. Sequeira Lopez ML, Pentz ES, Robert B, Abrahamson DR,
J Am Soc Nephrol 18: 1103–1111, 2007 Connexin 40 and Displacement of Renin-Producing Cells 1109
Gomez RA: Embryonic origin and lineage of juxtaglomer-
ular cells. Am J Physiol 281: F345–F356, 2001
7. Cantin M, Araujo-Nascimento MD, Benchimol S, Des-
ormeaux Y: Metaplasia of smooth muscle cells into juxta-
glomerular cells in the juxtaglomerular apparatus, arteries,
and arterioles of the ischemic (endocrine) kidney: An ul-
trastructural-cytochemical and autoradiographic study.
Am J Pathol 87: 581– 602, 1977
8. Taugner R, Hackenthal E, Nobiling R, Harlacher M, Reb G:
The distribution of renin in the different segments of the
renal arterial tree: Immunocytochemical investigation in
the mouse kidney. Histochemistry 73: 75– 88, 1981
9. Taugner R, Marin-Grez M, Keilbach R, Hackenthal E, No-
biling R: Immunoreactive renin and angiotensin II in the
afferent glomerular arterioles of rats with hypertension
due to unilateral renal artery constriction. Histochemistry
76: 61–69, 1982
10. Tufro-McReddie A, Johns DW, Geary KM, Dagli H, Everett
AD, Chevalier RL, Carey RM, Gomez RA: Angiotensin II
type 1 receptor: Role in renal growth and gene expression
during normal development. Am J Physiol 266: F911–F918,
1994
11. Hubert C, Gasc JM, Berger S, Schutz G, Corvol P: Effects of
mineralocorticoid receptor gene disruption on the compo-
nents of the renin-angiotensin system in 8-day-old mice.
Mol Endocrinol 13: 297–306, 1999
12. Fuchs S, Germain S, Philippe J, Corvol P, Pinet F: Expres-
sion of renin in large arteries outside the kidney revealed
by human renin promoter/LacZ transgenic mouse. Am J
Pathol 161: 717–725, 2002
13. Kruger O, Plum A, Kim JS, Winterhager E, Maxeiner S,
Hallas G, Kirchhoff S, Traub O, Lamers WH, Willecke K:
Defective vascular development in connexin 45-deficient
mice. Development 127: 4179 4193, 2000
14. Montecino-Rodriguez E, Leathers H, Dorshkind K: Expres-
sion of connexin 43 (Cx43) is critical for normal hemato-
poiesis. Blood 96: 917–924, 2000
15. Montoro RJ, Yuste R: Gap junctions in developing neocor-
tex: A review. Brain Res Brain Res Rev 47: 216 –326, 2004
16. Wong RC, Pebay A, Nguyen LT, Koh KL, Pera MF: Pres-
ence of functional gap junctions in human embryonic stem
cells. Stem Cells 22: 883– 889, 2004
17. Oviedo-Orta E, Howard Evans W: Gap junctions and con-
nexin-mediated communication in the immune system.
Biochim Biophys Acta 1662: 102–112, 2004
18. Malassine A, Cronier L: Involvement of gap junctions in
placental functions and development. Biochim Biophys Acta
1719: 117–124, 2005
19. Pointis G, Segretain D: Role of connexin-based gap junc-
tion channels in testis. Trends Endocrinol Metab 16: 300–306,
2005
20. Taugner R, Schiller A, Kaissling B, Kriz W: Gap junctional
coupling between the JGA and the glomerular tuft. Cell
Tissue Res 186: 279 –285, 1978
21. Taugner R, Kirchheim H, Forssmann WG: Myoendothelial
contacts in glomerular arterioles and in renal interlobular
arteries of rat, mouse and Tupaia belangeri. Cell Tissue Res
235: 319–325, 1984
22. Forsmann WG, Taugner R: Studies on the juxtaglomerular
apparatus. V. The juxtaglomerular apparatus in Tupaia
with special reference to intercellular contacts. Cell Tissue
Res 177: 291–305, 1977
23. Taugner R, Buhrle CP, Nobiling R: Ultrastructural changes
associated with renin secretion from the juxtaglomerular
apparatus of mice. Cell Tissue Res 237: 459 472, 1984
24. Hwan Seul K, Beyer EC: Heterogeneous localization of
connexin40 in the renal vasculature. Microvasc Res 59: 140
148, 2000
25. Haefliger JA, Demotz S, Braissant O, Suter E, Waeber B,
Nicod P, Meda P: Connexins 40 and 43 are differentially
regulated within the kidneys of rats with renovascular
hypertension. Kidney Int 60: 190 –201, 2001
26. Arensbak B, Mikkelsen H-B, Gustafsson F, Christensen T,
Holstein-Rathlou NH: Expression of connexin 37, 40, and
43 mRNA and protein in renal preglomerular arterioles.
Histochem Cell Biol 115: 479 487, 2001
27. Zhang J, Hill CE: Differential connexin expression in pre-
glomerular and postglomerular vasculature: Accentuation
during diabetes. Kidney Int 68: 1171–1185, 2005
28. Haefliger JA, Nicod P, Meda P: Contribution of connexins
to the function of the vascular wall. Cardiovasc Res 62:
345–356, 2004
29. Sohl G, Willecke K: Gap junctions and the connexin protein
family. Cardiovasc Res 62: 228 –232, 2004
30. de Wit C, Wolfle SE, Hopfl B: Connexin-dependent com-
munication within the vascular wall: Contribution to the
control of arteriolar diameter. Adv Cardiol 42: 268–283, 2006
31. Boyett MR, Inada S, Yoo S, Li J, Liu J, Tellez J, Greener ID,
Honjo H, Billeter R, Lei M, Zhang H, Efimov IR, Dobrzyn-
ski H: Connexins in the sinoatrial and atrioventricular
nodes. Adv Cardiol 42: 175–197, 2006
32. Peti-Peterdi J: Calcium wave of tubuloglomerular feed-
back. Am J Physiol Renal Physiol 291: F473–F480, 2006
33. Kirchhoff S, Nelles E, Hagendorff A, Kruger O, Traub O,
Willecke K: Reduced cardiac conduction velocity and pre-
disposition to arrhythmias in connexin40-deficient mice.
Curr Biol 8: 299 –302, 1998
34. de Wit C, Roos F, Bolz S-S, Kirchhoff S, Kruger O, Willecke K,
Pohl U: Impaired conduction of vasodilation along arterioles
in connexin40-deficient mice. Circ Res 86: 649 655, 2000
35. Mann B, Hartner A, Jensen BL, Hilgers KF, Hocherl K,
Kramer BK, Kurtz A: Acute upregulation of COX-2 by
renal artery stenosis. Am J Physiol 280: F119–F125, 2001
36. Vogetseder A, Karadeniz A, Kaissling B, Le Hir M: Tubular
cell proliferation in the healthy rat kidney: Histochem Cell
Biol 124: 97–104, 2005
37. Schweda F, Wagner C, Kramer BK, Schnermann J, Kurtz A:
Preserved macula densa-dependent renin secretion in A1
adenosine receptor knockout mice. Am J Physiol Renal
Physiol 284: F770–F777, 2003
38. Richardson KC: The fine structure of autonomic nerves
after vital staining with methylene blue. Anat Rec 164:
359–377, 1969
39. Michon L, Nlend Nlend R, Bavamian S, Bischoff L, Bou-
card N, Caille D, Cancela J, Charollais A, Charpantier E,
Klee P, Peyrou M, Populaire C, Zulianello L, Meda P:
Involvement of gap junctional communication in secretion.
Biochim Biophys Acta 1719: 82–101, 2005
40. Kazimierczak J: Development of the renal corpuscle and
the juxtaglomerular apparatus. Acta Pathol Microbiol Immu-
nol Scand 218: S81–S227, 1971
41. Osanthanondh V, Potter EL: Developing human kidney as
shown by microdissection. V. Development of vascular
pattern of glomerulus. Arch Pathol 82: 403–411, 1966
1110 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1103–1111, 2007
42. Zhuo J, Anderson WP, Song K, Mendelsohn FA: Autora-
diographic localization of active renin in the juxtaglomer-
ular apparatus of the dog kidney: Effects of sodium intake.
Clin Exp Pharmacol Physiol 23: 291–298, 1996
43. Hugo C, Shankland SJ, Bowen-Pope DF, Couser WG, John-
son RJ: Extraglomerular origin of the mesangial cell after
injury. A new role of the juxtaglomerular apparatus. J Clin
Invest 100: 786–794, 1997
44. Taugner R, Waldherr R, Seyberth HW, Erdos EG, Menard
J, Schneider D: The juxtaglomerular apparatus in Bartter’s
syndrome and related tubulopathies. An immunocyto-
chemical and electron microscopic study. Virchows Arch A
Pathol Anat Histopathol 412: 459 470, 1988
45. Spangler WL: Ultrastructure of the juxtaglomerular appa-
ratus in the dog in a sodium-balanced state. Am J Vet Res
40: 802–828, 1979
46. Barajas L: Anatomy of the juxtaglomerular apparatus. Am J
Physiol 237: F333–F343, 1979
47. Christensen JA, Bohle A, Mikeler E, Taugner R: Renin-
positive granulated Goormaghtigh cells. Immunohisto-
chemical and electron-microscopic studies on biopsies
from patients with pseudo-Bartter syndrome. Cell Tissue
Res 255: 149–153, 1989
48. de Wit C, Roos F, Bolz SS, Pohl U: Lack of vascular con-
nexin40 is associated with hypertension and irregular arte-
riolar vasomotion. Physiol Genomics 13: 169 –177, 2003
49. Castrop H, Lorenz JN, Hansen PB, Friis U, Mizel D, Op-
permann M, Jensen BL, Briggs J, Skott O, Schnermann J:
Contribution of the basolateral isoform of the Na-K-2Cl-
cotransporter (NKCC1/BSC2) to renin secretion. Am J
Physiol Renal Physiol 289: F1185–F1192, 2005
50. Wall SM, Knepper MA, Hassell KA, Fischer MP, Sho-
deinde A, Shin W, Pham TD, Meyer JW, Lorenz JN, Beier-
waltes WH, Dietz JR, Shull GE, Kim YH: Hypotension in
NKCC1 null mice: role of the kidneys. Am J Physiol Renal
Physiol 290: F409–F416, 2006
J Am Soc Nephrol 18: 1103–1111, 2007 Connexin 40 and Displacement of Renin-Producing Cells 1111
... Intercellular communication via gap junctions and, in particular, through Cx40-formed channels, has been demonstrated to play a central role in the reninmediated control of blood pressure. All vascular Cxs are found in the juxtaglomerular apparatus, but only Cx37 and Cx40 are expressed in renin-producing cells [136][137][138][139][140][141]. Consistent with the renin-dependent hypertension observed in Cx40 knockout mice, global deletion of this Cx disrupts the negative feedback control that inhibits renin production, and secretion in response to increments of blood pressure or angiotensin II, leading to an increase in plasma renin concentration [122,142,143]. ...
... Consistent with the renin-dependent hypertension observed in Cx40 knockout mice, global deletion of this Cx disrupts the negative feedback control that inhibits renin production, and secretion in response to increments of blood pressure or angiotensin II, leading to an increase in plasma renin concentration [122,142,143]. Interestingly, the dysregulation of renin-angiotensin system observed in Cx40 knockout mice was also associated with structural changes in the juxtaglomerular apparatus architecture [140,144], since ablation of Cx40 resulted in striking changes in the cellular distribution of reninexpressing cells, which were no longer present in the media layer of afferent arterioles at the juxtaglomerular position (Fig. 4), but, in these animals, renin-expressing cells were found in the extraglomerular mesangium and periglomerular interstitium [140,144]. This cellular relocalization was also observed after disrupting the expression of Cx40 in adult mice using a tamoxifeninducible knockout mouse model [145], excluding the participation of secondary developmental alterations triggered by embryonic deletion of Cx40. ...
... Consistent with the renin-dependent hypertension observed in Cx40 knockout mice, global deletion of this Cx disrupts the negative feedback control that inhibits renin production, and secretion in response to increments of blood pressure or angiotensin II, leading to an increase in plasma renin concentration [122,142,143]. Interestingly, the dysregulation of renin-angiotensin system observed in Cx40 knockout mice was also associated with structural changes in the juxtaglomerular apparatus architecture [140,144], since ablation of Cx40 resulted in striking changes in the cellular distribution of reninexpressing cells, which were no longer present in the media layer of afferent arterioles at the juxtaglomerular position (Fig. 4), but, in these animals, renin-expressing cells were found in the extraglomerular mesangium and periglomerular interstitium [140,144]. This cellular relocalization was also observed after disrupting the expression of Cx40 in adult mice using a tamoxifeninducible knockout mouse model [145], excluding the participation of secondary developmental alterations triggered by embryonic deletion of Cx40. ...
Article
Vascular system is a complex network in which different cell types and vascular segments must work in concert to regulate blood flow distribution and arterial blood pressure. Although paracrine/autocrine signaling is involved in the regulation of vasomotor tone, direct intercellular communication via gap junctions plays a central role in the control and coordination of vascular function in the microvascular network. Gap junctions are made up by connexin (Cx) proteins, and among the four Cxs expressed in the cardiovascular system (Cx37, Cx40, Cx43, and Cx45), Cx40 has emerged as a critical signaling pathway in the vessel wall. This Cx is predominantly found in the endothelium, but it is involved in the development of the cardiovascular system and in the coordination of endothelial and smooth muscle cell function along the length of the vessels. In addition, Cx40 participates in the control of vasomotor tone through the transmission of electrical signals from the endothelium to the underlying smooth muscle and in the regulation of arterial blood pressure by renin-angiotensin system in afferent arterioles. In this review, we discuss the participation of Cx40-formed channels in the development of cardiovascular system, control and coordination of vascular function, and regulation of arterial blood pressure.
... Cx40 KO mice have hypertension and decreased vascular response to ACh in skeletal muscle arterioles [54]. Hypertension is caused by increased renin release [55,56]. The RSC are physically disorganized and do not respond to changes in blood pressure or Ang II indicating a lack of Ca 2+ sensitivity. ...
... The RSC are physically disorganized and do not respond to changes in blood pressure or Ang II indicating a lack of Ca 2+ sensitivity. The response to MD cell stimulation is also attenuated [55]. As suggested above Cx40 couples the RSC together and coordinates their renin release. ...
... It is likely that, as in pancreatic islets [57 ], the cells of the juxtaglomerular apparatus control each other in an on/off system. Inhibition of gap junctional communication in the pancreas inhibits the control of b-cells [57 ] much like deletion of Cx40 abolishes the control of RSC [55,56]. ...
... Thus, the study on mice with deletion of the gene for Cx37 reported the relation of this connexin to the regulation of renin expression [42]. Cx40 is recognized as the predominant connexin of renin-producing cells in mice, although there are some inconsistencies in its role in fetal mice kidneys [34,43]. Based on a study on Cx40 -/-mice, Cx40 might be connected to renin release and renin-dependent hypertension [6,44]. ...
... Similar to our results, a study on human kidneys has shown evidence of Cx37, Cx40 and Cx43 expression in the extraglomerular mesangium of JGA [53]. In contrast, studies on developing and adult mice showed only Cx40-renin co-expression [43], while the displacement of renin-producing cells to extraglomerular mesangium was observed only in the absence of Cx40 [34]. In addition, in mice lacking Cx40, renin-producing cells were disorganized and dysfunctional [54,55], while in Cx37 null mice investigators have failed to show any changes in renin positioning and activity [42]. ...
Article
Full-text available
Our study analyzed the expression pattern of different connexins (Cxs) and renin positive cells in the juxtaglomerular apparatus (JGA) of developing, postnatal healthy human kidneys and in nephrotic syndrome of the Finnish type (CNF), by using double immunofluorescence, electron microscopy and statistical measuring. The JGA contained several cell types connected by Cxs, and consisting of macula densa, extraglomerular mesangium (EM) and juxtaglomerular cells (JC), which release renin involved in renin-angiotensin- aldosteron system (RAS) of arterial blood pressure control. During JGA development, strong Cx40 expression gradually decreased, while expression of Cx37, Cx43 and Cx45 increased, postnatally showing more equalized expression patterning. In parallel, initially dispersed renin cells localized to JGA, and greatly increased expression in postnatal kidneys. In CNF kidneys, increased levels of Cx43, Cx37 and Cx45 co-localized with accumulations of renin cells in JGA. Additionally, they reappeared in extraglomerular mesangial cells, indicating association between return to embryonic Cxs patterning and pathologically changed kidney tissue. Based on the described Cxs and renin expression patterning, we suggest involvement of Cx40 primarily in the formation of JGA in developing kidneys, while Cx37, Cx43 and Cx45 might participate in JGA signal transfer important for postnatal maintenance of kidney function and blood pressure control.
... Absence of Cx40 impairs, but normally does not block, conducted vascular responses in other vascular beds (de Wit et al., 2006;Jobs et al., 2012;Zechariah et al., 2020). Cx40 is also located within the mesangium where it plays significant roles in transmission of TGF signals from the macula densa to the afferent arteriole, affecting vascular resistance, and to the juxtaglomerular apparatus, where it is important to structural organization and tubular regulation of renin secretion (Kurtz et al., 2007;Kurtz et al., 2010). A Cx40 knockout (KO) mouse was generated 25 years ago and has become a mainstay of microvascular research (Simon et al., 1998). ...
Article
Full-text available
Introduction: Tubuloglomerular feedback (TGF) is the negative feedback component of renal blood flow (RBF) autoregulation. Neighbouring nephrons often exhibit spontaneous TGF oscillation and synchronization mediated by endothelial communication, largely via connexin40 (Cx40). Methods: We had a knockout (KO) rat made that lacks Cx40. One base pair was altered to create a stop codon in exon 1 of Gja5, the gene that encodes Cx40 (the strain is WKY-Gja5 5em1Mcwi ). Blood pressure (BP)-RBF transfer functions probed RBF dynamics and laser speckle imaging interrogated the dynamics of multiple efferent arterioles that reach the surface (star vessels). Results: The distribution of wild type (WT), heterozygote, and KO pups at weaning approximated the Mendelian ratio of 1:2:1; growth did not differ among the three strains. The KO rats were hypertensive. BP-RBF transfer functions showed low gain of the myogenic mechanism and a smaller TGF resonance peak in KO than in WT rats. Laser speckle imaging showed that myogenic mechanism had higher frequency in KO than in WT rats, but similar maximum spectral power. In contrast, the TGF frequency was similar while peak power of its oscillation was much smaller in KO than in WT rats. In WT rats, plots of instantaneous TGF phase revealed BP-independent TGF synchronization among star vessels. The synchronization could be both prolonged and widespread. In KO rats TGF synchronization was not seen, although BP transients could elicit short-lived TGF entrainment. Discussion: Despite the reduced TGF spectral power in KO rats, there was sufficient TGF gain to induce oscillations and therefore enough gain to be effective locally. We conclude that failure to synchronize is dependent, at least in part, on impaired conducted vasomotor responses.
... Each gene in each EC subtype has a role in kidney function and is an identity (marker). For example, Gja5, also known as connexin 40, controls renin release by regulating the communication between ECs and the granular cells in the JGA (Kurtz et al., 2007). ...
Article
Full-text available
Vascular endothelial cells (ECs) that constitute the inner surface of blood vessels are essential for new vessel formation and organ homeostasis. ECs display remarkable phenotypic heterogeneity across different organs and the vascular tree during angiogenesis and homeostasis. Recent advances in single cell RNA sequencing (scRNA-seq) technologies have allowed a new understanding of EC heterogeneity in both mice and humans. In particular, scRNA-seq has identified new molecular signatures for arterial, venous and capillary ECs in different organs, as well as previously unrecognized specialized EC subtypes, such as the aerocytes localized in the alveolar capillaries of the lung. scRNA-seq has also revealed the gene expression profiles of specialized tissue-resident EC subtypes that are capable of clonal expansion and contribute to adult angiogenesis, a process of new vessel formation from the pre-existing vasculature. These specialized tissue-resident ECs have been identified in various different mouse tissues, including aortic endothelium, liver, heart, lung, skin, skeletal muscle, retina, choroid, and brain. Transcription factors and signaling pathways have also been identified in the specialized tissue-resident ECs that control angiogenesis. Furthermore, scRNA-seq has also documented responses of ECs in diseases such as cancer, age-related macular degeneration, Alzheimer’s disease, atherosclerosis, and myocardial infarction. These new findings revealed by scRNA-seq have the potential to provide new therapeutic targets for different diseases associated with blood vessels. In this article, we summarize recent advances in the understanding of the vascular endothelial cell heterogeneity and endothelial stem cells associated with angiogenesis and homeostasis in mice and humans, and we discuss future prospects for the application of scRNA-seq technology.
... Cx37 and Cx40 appear to exist in symbiosis, with protein expression regulating plasma membrane insertion and channel gating (383). Multiple studies using knockout animals have reported that Cx40, but not Cx37, is indispensable for EC conduction (91,130,250,294,576,577,593). For instance, global or EC-specific Cx40 knockout mice develop hypertension; however, these mice also demonstrated downregulation of Cx37 in EC (91,250). ...
Chapter
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
... Within the JGA, gap junctions are formed by four different connexin (Cx) isoforms, Cx37, Cx40, Cx43, and Cx45, with reninproducing cells expressing only Cx37 and Cx40 [49,50,118]. Expression of Cx40 seems to be crucial for determining the juxtaglomerular position of renin-producing cells, as well as for the blood pressure-dependent regulation of renin synthesis and secretion [47,51,61,112,113]. Inducible deletion of Cx40 in adult mice results in misplaced renin cells that are no longer located in the media layer of afferent arterioles but in the extraglomerular mesangium [22]. ...
Article
Full-text available
The protease renin, the key enzyme of the renin–angiotensin–aldosterone system, is mainly produced and secreted by juxtaglomerular cells in the kidney, which are located in the walls of the afferent arterioles at their entrance into the glomeruli. When the body’s demand for renin rises, the renin production capacity of the kidneys commonly increases by induction of renin expression in vascular smooth muscle cells and in extraglomerular mesangial cells. These cells undergo a reversible metaplastic cellular transformation in order to produce renin. Juxtaglomerular cells of the renin lineage have also been described to migrate into the glomerulus and differentiate into podocytes, epithelial cells or mesangial cells to restore damaged cells in states of glomerular disease. More recently, it could be shown that renin cells can also undergo an endocrine and metaplastic switch to erythropoietin-producing cells. This review aims to describe the high degree of plasticity of renin-producing cells of the kidneys and to analyze the underlying mechanisms.
... Based on these findings, we would hypothesize that EPO induction in juxtaglomerular renin + cells requires cell reprogramming due to prolonged HIF-2 stabilization. Indeed, during chronic HIF-2 stabilization through PHD2 and PHD3 co-deletion, the typical renin cell markers connexin 40 and Akr1b7 were downregulated along with renin in juxtaglomerular cells (Kurtz et al. 2007;Brunskill Figure 13. Cortical details showing co-RNAscopes for EPO, renin and Cx40 (top) or CD73 (bottom) mRNA on kidney sections of Aldo-KO mice and Aldo-KO mice with different PHD deletions in renin-producing cells Circles indicate glomeruli. ...
Article
Full-text available
Activation of the hypoxia‐signalling pathway induced by deletion of the ubiquitin‐ligase von Hippel–Lindau protein causes an endocrine shift of renin‐producing cells to erythropoietin (EPO)‐expressing cells. However, the underlying mechanisms have not yet been investigated. Since oxygen‐regulated stability of hypoxia‐inducible transcription factors relevant for EPO expression is dependent on the activity of prolyl‐4‐hydroxylases (PHD) 2 and 3, this study aimed to determine the relevance of different PHD isoforms for the EPO expression in renin‐producing cells in vivo. For this purpose, mice with inducible renin cell‐specific deletions of different PHD isoforms were analysed. Our study shows that there are two subgroups of renal renin‐expressing cells, juxtaglomerular renin⁺ cells and platelet‐derived growth factor receptor‐β⁺ interstitial renin⁺ cells. These interstitial renin⁺ cells belong to the cell pool of native EPO‐producing cells and are able to express EPO and renin in parallel. In contrast, co‐deletion of PHD2 and PHD3, but not PHD2 deletion alone, induces EPO expression in juxtaglomerular and hyperplastic renin⁺ cells and downregulates renin expression. A strong basal PHD3 expression in juxtaglomerular renin⁺ cells seems to prevent the hypoxia‐inducible transcription factor‐2‐dependent phenotype shift into EPO cells. In summary, PHDs seem important for the stabilization of the juxtaglomerular renin cell phenotype. Moreover, these findings reveal tubulointerstitial cells as a novel site of renal renin expression and suggest a high endocrine plasticity of these cells. Our data concerning the distinct expression patterns and functions of PHD2 and PHD3 provide new insights into the regulation of renin‐producing cells and highlight the need for selective PHD inhibitors. Key points Renal renin‐expressing cells can be clearly distinguished into two subgroups, the typical juxtaglomerular renin‐producing cells and interstitial renin⁺ cells. Interstitial renin⁺ cells belong to the cell pool of native erythropoietin (EPO)‐producing cells, show a fast EPO response to acute hypoxia‐inducible factor‐2 (HIF‐2) stabilization and are able to express EPO and renin in parallel. Only co‐deletion of the prolyl‐4‐hydroxylases (PHD) 2 and 3, but not PHD2 deletion alone, induces EPO expression in juxtaglomerular renin⁺ cells. Chronic HIF‐2 stabilization in juxtaglomerular renin‐expressing cells leads to their phenotypic shift into EPO‐producing cells. A strong basal PHD3 expression in juxtaglomerular renin⁺ cells seems to prevent a HIF‐2‐dependent phenotype shift into EPO cells suggesting PHD3 fulfils a stabilizer function for the juxtaglomerular renin cell phenotype.
... 50 Deletion of several components of the cAMP pathway, including deletion of the β-adrenergic receptor, results in arrested vascular development and alterations in renin secretion. 51,68,69 Lack of connexin 40 in renin cells leads to malignant hypertension, 70 and deletion of the Von Hippel-Lindau gene in renin cells leads to a remarkable phenotypic switch: the former renin-producing cells stop making renin and transcribe erythropoietin. 71 As a result, the animals display secondary erythrocytosis. ...
Article
Full-text available
A lthough renin cells are crucial for blood pressure homeo-stasis, little is known about their nature. We now know that renin cells are precursors that appear early and in multiple tissues during embryonic development. They participate in morphogenetic events, vascular development and injury, tissue repair, and regeneration. When confronted to a homeostatic threat, renin cell descendants have the capability to switch the renin gene on or off. This poorly understood switch or molecular memory enables the organism to maintain constancy of the internal milieu and tissue perfusion. Here, we discuss briefly the major events that govern the acquisition and maintenance of renin cell identity and how manipulations that alter the fate of renin cells can lead to serious disease. We also advance the concept that renin cells are at the center of an ancestral system of defense linking the endocrine, the immune, and the repair responses of the organism. Renin-Angiotensin System The renin-angiotensin system (RAS) is crucial in the regulation of blood pressure and fluid-electrolyte homeostasis. 1,2 In the traditional view of the RAS, renin is released by the kidney juxtaglomerular cells, and on reaching the circulation, it acts on its only known substrate, angiotensinogen, produced mainly in the liver to yield angiotensin I (Ang I), a decapep-tide and Des-Ang I-angiotensinogen, a large molecule of unclear function. Thereafter, Ang I is hydrolyzed by angio-tensin-converting enzyme to yield the octapeptide Ang II, a fast acting and powerful vasoconstrictor that regulates peripheral vascular resistance, renal hemodynamics, and sodium reabsorption via several mechanisms, including the stimulation of aldosterone secretion by the adrenal glands. Most of the known cardiovascular and renal actions of the RAS are achieved by the actions of Ang II on its receptors, mainly AT1 receptors. It should be noted that for the system to operate properly, it needs to respond accurately and rapidly to changes in the composition and volume of the extracel-lular fluid and to variations in systemic blood pressure. The key regulated event in this enzymatic cascade is the tightly controlled, minute-to-minute regulation of renin release by the juxtaglomerular cells. This is possible because juxtaglo-merular cells are sensors strategically located in the juxtaglo-merular apparatus (JGA), where they receive and interpret signals that convey the composition and volume of the extra-cellular fluid and the level of perfusion pressure. The JGA is composed of the afferent and efferent arterioles, the macula densa, and the extraglomerular mesangium or polkissen. 1,3,4 In the adult unstressed mammalian kidney, juxtaglomerular cells are located in the afferent arteriole at the entrance to the glomerulus, where they make contact with macula densa cells, extraglomerular mesangial cells, and other renin and smooth muscle cells along the arteriole. 3,4 Juxtaglomerular cells have a myoepithelioid appearance; they are densely innervated by sympathetic terminals arising from the renal nerve; and they contain granules from where renin is stored and released in response to a diverse number of stimuli emanating from nearby cells, sympathetic terminal, and from the circulation. 5 Three major mechanisms control renin release by juxtaglomerular cells: (1) the renal baroreceptor mechanism, whereby renin release is elicited by a decrease in renal perfu-sion pressure as it occurs during hypotension, shock, hemorrhage , or cardiac failure. The nature of the renal baroreceptor has not been determined since its original 1959 description by Tobian et al, 6 (2) the macula densa mechanism, whereby renin release is stimulated by a decrease in sodium chloride in the distal tubule as it occurs during sodium depletion, and (3) the β-receptor-mediated mechanism, whereby stimulation of β-receptors elicited by sympathetic terminals or via circulating catecholamines such as during hypoxia results in increased renin release. Interestingly, the renal baroreceptor mechanism continues to function in the absence of the other 2 mechanisms: in the denervated, nonfiltering kidney, the barorecep-tor mechanism continues to operate suggesting that the renal baroreceptor mechanism is independent from the influence of the macula densa or the β-receptor. 2 Under normal circumstances , however, these mechanisms operate together to finely regulate renin output. For instance, the β-receptor mechanism, the baroreceptor mechanism, and the macula densa mechanism are all activated during hemorrhage, a situation where there is decreased perfusion pressure, decreased delivery of sodium chloride to the macula densa, and stimulation of the sympathetic system. It should be noted that Ang II exerts a negative feedback on renin release, a typical case where the byproduct of an enzymatic reaction controls its own production , in this case governed by the underlying physiological status of the animal. When angiotensin production and its actions are limited, such as when animals are exposed to angiotensin-converting enzyme inhibitors or AT1 receptor blockers, renin synthesis and release is increased. This is accomplished in great part by an increase in the number of cells that synthesize and release renin as described below. 7-9
... Since RBP-J also controls the expression of smooth muscle genes and their master regulators, this suggests that the Notch pathway is intimately involved in renin cell plasticity and recruitment of smooth muscle cells. Likewise, the observation that connexin-40, a transmembrane protein involved in gap junctions, is highly expressed in JGCs 37 and that its knockout leads to the loss of JGC and appearance of RPCs in the periglomerular interstitium 48 suggests that cell-cell communication is also essential for JGC positioning. ...
Article
Full-text available
Cellular plasticity is a topical subject with interest spanning a wide range of fields from developmental biology to regenerative medicine. Even the nomenclature is a subject of debate, and the underlying mechanisms are still under investigation. On top of injury repair, cell plasticity is a constant, physiological process in adult organisms and tissues, in response to homeostatic challenges. In this review we discuss two examples of plasticity for the maintenance of homeostasis in the renal system – namely the renin‐producing juxtaglomerular cells (JG cells) and cortical collecting duct (CCD) cells. JG cells show plasticity through recruitment mechanisms, answering the demand for an increase in renin production. In the CCD, cells appear to have the ability to transdifferentiate between principal and intercalated cells to help maintain the highly regulated solute transport levels of that segment. These two cases highlight the complexity of plasticity processes and the role they can play in the kidney.
Article
Full-text available
Gap junctions are intercellular channels that allow both chemical and electrical signaling between two adjacent cells. Gap junction intercellular communication has been implicated in the regulation of various cellular processes, including cell migration, cell proliferation, cell differentiation, and cell apoptosis. This study aimed to determine the presence and functionality of gap junctions in human embryonic stem cells (hESCs). Using reverse transcription--polymerase chain reaction and immunocytochemistry, we demonstrate that human ES cells express two gap junction proteins, connexin 43 and connexin 45. Western blot analysis revealed the presence of three phosphorylated forms (nonphosphorylated [NP], P1, and P2) of connexin 43, NP being prominent. Moreover, scrape loading/dye transfer assay indicates that human ES cells are coupled through functional gap junctions that are inhibited by protein kinase C activation and extracellular signal-regulated kinase inhibition.
Article
Gap junctions are intercellular channels, formed by individual structural units known as connexins (Cx), that allow the intercellular exchange of various messenger molecules. The finding that numbers of Cx43-type gap junctions in bone marrow are elevated during establishment and regeneration of the hematopoietic system has led to the hypothesis that expression of Cx43 is critical during the initiation of blood cell formation. To test this hypothesis, lymphoid and myeloid development were examined in mice with a targeted disruption of the gene encoding Cx43. Because Cx43−/− mice die perinatally, initial analyses were performed on Cx43−/−, Cx43+/−, and Cx43+/+ embryos and newborns. The data indicate that lack of Cx43 expression during embryogenesis compromises the terminal stages of primary T and B lymphopoiesis. Cx43−/− embryos and neonates had a reduced frequency of CD4+ and T-cell receptor-expressing thymocytes and surface IgM+cells compared to their Cx43+/+ littermates. Surprisingly, Cx43+/− embryos/neonates also showed defects in B- and T-cell development similar to those observed in Cx43−/− littermates, but their hematopoietic system was normal at 4 weeks of age. However, the regeneration of lymphoid and myeloid cells was severely impaired in the Cx43+/− mice after cytoablative treatment. Taken together, these data indicate that loss of a single Cx43 allele can affect blood cell formation. Finally, the results of reciprocal bone marrow transplants between Cx43+/+ and Cx43+/− mice and examination of hematopoietic progenitors and stromal cells in vitro indicates that the primary effects of Cx43 are mediated through its expression in the hematopoietic microenvironment.
Article
The zonation of the expression and regulation of the cytosolic aspartate aminotransferase (cAspAT) mRNAs in the liver acinus was investigated in diabetic and/or adrenalectomized rats. Dexamethasone increased cAspAT activity two- to threefold alone and up to sixfold in combination with streptozotocin-induced diabetes. Northern blot analysis showed that the cAspAT mRNAs were increased by those treatments; the effect of streptozotocin was reversed by the administration of insulin. In situ hybridization experiments showed that basal cAspAT mRNAs were uniformly distributed within the liver acinus. However, cAspAT mRNAs were induced by glucocorticoids specifically in the periportal zone and by streptozotocin in a larger area including the periportal and intermediary zone. The alpha 2u-globulin mRNAs which are specifically expressed in the perivenous hepatocytes are also induced by glucocorticoids in this zone, suggesting that the specific regulation of the cAspAT gene by glucocorticoids in the periportal zone is not due to the absence of functional glucocorticoid receptors in the other zones. We conclude that the regulation of the cAspAT housekeeping gene is zone specific in the liver. Furthermore, this zonation depends on the gene and on the type of hormonal or pharmacological treatment.
Article
The intrarenal distribution of renin in the mouse kidney was evaluated in a semiquantitative immunocytochemical study using an antiserum against pure mouse renin and the PAP technique. The bulk of renin positive cells was found in the media of the afferent arteriole. When examining the geometry of renin distribution about 35% of the afferent vessels were seen to be renin positive only over a distance of 20 m, about 60% over a distance of 30 m. In the remaining afferent arterioles, renin was also found upstream over distances up to 100 or even 200 m. These results are discussed with regard to the stimuli for renin secretion, especially the macula densa signal. — At the vascular pole of the glomerulus, virtually 100% of the afferent, and 20%–40% of the efferent arterioles were found to be renin positive at an antiserum dilution of 1:1,000. As some efferent vessels — especially those of the juxtamedullar region — show scattered activity occasionally over a distance of more than 100 m, it is suggested that the figure of 20%–40% should be taken as a minimal count for renin positive efferent arterioles. — To compare the renin content of superficial and juxtamedullary, afferent and efferent arterioles in normal salt and salt depleted mice, the fraction of positive renin reactions close to the vascular pole was determined at antiserum concentrations of 10–3, 10–4, 210–4 and 10–5. By this semiquantitative immunocytochemical method the afferent arterioles of superficial glomeruli could be shown to contain significantly higher renin concentrations than those of juxtamedullar glomeruli. This result was in agreement with biochemical renin estimations in mouse kidney slices taken from cortical and juxtamedullar sites. Sodium deprivation was followed by only a slight elevation of the fraction of positive superficial afferent arterioles (confirmed by the biochemical data). In contrast, sodium deprivation induced a highly significant increase of the number of positive superficial efferent vessels. This result is discussed with regard to (controversial) reports on a preferential efferent vasoconstrictor tone sustained by angiotensin II especially under the condition of sodium depletion. Juxtamedullar vasa afferentia and efferentia did not respond significantly to sodium restriction. —The Goormaghtigh cell field was found to be renin negative in superficial as well as in juxtamedullar glomeruli both in normal salt and salt deprived mice. Inspecting nearly 5,000 glomeruli, only 5 clearly renin positive mesangial cells were seen close to the glomerular stalk. In contrast, renin positive media cells could not seldom be seen in interlobular arteries and at the point of their branching into afferent arterioles.
Article
The juxtaglomerular apparatus (JGA) in the rabbit kidney was examined by transmission electron microscopy and by freeze fracturing. It was found, that the Goormaghtigh cells of the JGA are extensively coupled with the mesangial cells within the glomerular tuft by gap junctions. A broad band of gap junctions starting within the Goormaghtigh cells, traversing the transitional area at the root of the glomerular tuft and continuing along the mesangial cells has been revealed by freeze fracturing. No gap Junctional connections to the macula densa cells have been found. In accordance with data from literature it may be stated that all smooth muscle derived cell groups at the vascular pole of the glomerulus (smooth muscle cells of the vas afferens and efferens, granular cells, Goormaghtigh cells, mesangial cells) are extensively coupled by gap junctions with each other. It is supposed that this cell system may act as a synchronized functional unit.
Article
A comparative immunocytochemical and electron microscopic study was performed on renal biopsies from two children with classical Bartter's syndrome (BS) and three children with a recently described variant, the so-called hyperprostaglandin E-syndrome (HES). Compared to age-matched controls, kidney specimens from patients with BS and HES disclosed a marked hypertrophy and hyperplasia of the juxtaglomerular apparatus (JGA). In addition, in HES focal tubular and interstitial calcifications accompanied by interstitial fibrosis and tubular atrophy were noted. On immunocytochemistry, chronic stimulation of the JGA in BS and HES was characterized by an increase in the number of renin-positive cells, particularly in the media of afferent arterioles, but also in efferent arterioles and in the glomerular stalk. The length of the renin-positive portion of the preglomerular arterioles was significantly increased when compared to controls (10032 vs. 4917 m;phypertrophy of the RER and of Golgi complexes with paracrystalline deposits in dilated RER cisterns and protogranules indicated an increased renin synthesis. Renin could be identified in mature secretory granules as well as protogranules by immune electron microscopy. Angiotensinogen was present in hypertrophied epithelial cells of Bowman's capsule. Converting-enzyme reactivity was observed in controls as well as in BS and HES in the brush border of the proximal tubule. In contrast to previous reports, Angiotensin II was completely negative in control as well as in diseased kidneys. We conclude from our results that both BS and HES are characterized by a marked activation of the JGA and severe stimulation of the renin-angiotensin system. Since activation of this system, however, leads - independently of the primary stimulus - to qualitatively very similar morphological reactions, these results do not implicate a common pathogenetic mechanism to both conditions.
Article
Intercellular channels of gap junctions are formed in vertebrates by the protein family of connexins and allow direct exchange of ions, metabolites and second messenger molecules between apposed cells (reviewed in [1-3]). In the mouse, connexin40 (Cx40) protein has been detected in endothelial cells of lung and heart and in certain heart muscle cells: atrial myocytes, cells of the atrial ventricular (AV) node and cells of the conductive myocardium, which conducts impulses from the AV node to ventricular myocyctes [3]. We have generated mice homozygous for targeted disruption of the Cx40 gene (Cx40-/-mice). The electrocardiograph (ECG) parameters of Cx40-/- mice were very prolonged compared to those of wild type (Cx40+/+) mice, indicating that Cx40-/- mice have lower atrial and ventricular conduction velocities. For 6 out of 31 Cx40-/- animals, different types of atrium-derived abnormalities in cardiac rhythm were recorded, whereas continuous sinus rhythm was observed for the 26 Cx40+/+ and 30 Cx40+/- mice tested. The expression levels of other connexins expressed in heart (Cx37, Cx43 and Cx45) were the same in Cx40-/- and Cx40+/+ mice. Our results demonstrate the function of Cx40 in the regulation and coordination of heart contraction and show that cardiac arrhythmogenesis can not only be caused by defects in the ion channels primarily involved in cellular excitation but also by defects in intercellular communication through gap junction channels. As the distribution of Cx40 protein is similar in mouse and human hearts, further functional analysis of Cx40 should yield relevant insights into arrhythmogenesis in human patients.