You are on page 1of 17

Chronobiology International

The Journal of Biological and Medical Rhythm Research

ISSN: 0742-0528 (Print) 1525-6073 (Online) Journal homepage: https://www.tandfonline.com/loi/icbi20

Neuropeptide changes in the suprachiasmatic


nucleus are associated with the development of
hypertension

Ajda Yilmaz, Frederik N Buijs, Andries Kalsbeek & Ruud M Buijs

To cite this article: Ajda Yilmaz, Frederik N Buijs, Andries Kalsbeek & Ruud M Buijs (2019):
Neuropeptide changes in the suprachiasmatic nucleus are associated with the development of
hypertension, Chronobiology International, DOI: 10.1080/07420528.2019.1613424

To link to this article: https://doi.org/10.1080/07420528.2019.1613424

Published online: 29 May 2019.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=icbi20
CHRONOBIOLOGY INTERNATIONAL
https://doi.org/10.1080/07420528.2019.1613424

Neuropeptide changes in the suprachiasmatic nucleus are associated with the


development of hypertension
Ajda Yilmaza, Frederik N Buijsa,b, Andries Kalsbeeka,c, and Ruud M Buijsa,b
a
Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam BA, The Netherlands;
b
Department of Cell Biology and Physiology, Institute for Biomedical Research, Universidad Nacional Autonoma de Mexico, Mexico City,
Mexico (Present address RMB); cDepartment of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam AZ,
The Netherlands

ABSTRACT ARTICLE HISTORY


Human postmortem studies as well as experimental animal studies indicate profound changes in Received 30 January 2019
neuropeptide expression in the suprachiasmatic nucleus (SCN) in several pathological conditions Revised 23 April 2019
including hypertension. In addition, animal experimental observations show that the SCN pep- Accepted 27 April 2019
tides, vasopressin (AVP) and vasoactive intestinal peptide (VIP) are essential for adequate rhyth- KEYWORDS
micity. These data prompted us to investigate whether changes in these neuronal populations Circadian rhythm;
could be the cause or consequence of hypertension. Changes in blood pressure and levels of vasopressin; vasoactive
neuropeptide expression in the SCN were determined during development of hypertension in intestinal peptide; nucleus
spontaneously hypertensive rats (SHR), in 2K1C reno-vascular induced hypertensive animals and tractus solitarius; blood
their respective controls. During the pre-hypertensive stage (5 weeks of age), the VIP and AVP pressure
content was higher and the somatostatin (SOM) content was lower in the SHR SCN. At the onset
of hypertension (12 weeks of age), when blood pressure levels had just reached about 140 mmHg,
AVP and SOM content in the SCN was not different anymore in SHRs compared to control, but VIP
was still higher. After 16 weeks, the AVP content was decreased, but SOM was increased and the
overall level of VIP in the SCN was still higher in SHRs compared to controls. None of the
aforementioned changes in the SCN was observed after induction of hypertension in the 2K1C
model. However, while VIP was increased in the NTS projecting medial region of the SCN in SHR
animals only after the establishment of hypertension, VIP was decreased in the same region in the
2K1C induced hypertensive rats. Consequently, the present findings confirm previous studies in
human and rat indicating that changes in the SCN are strongly associated with the development
of hypertension. In addition, the changes in peptide content in the 2K1C animals indicate that the
SCN is also able to respond to increases in blood pressure.

Introduction SCN or to aging restores their circadian rhythm of


activity (LeSauter and Silver 1999; Li and Satinoff
The Suprachiasmatic Nucleus (SCN) seems the sole
1998; Ralph et al. 1993).
center for the control of bio-behavioral rhythms,
The SCN can be divided into a part that receives
including heart rate, blood pressure, sleep – wake,
direct information from the retina (core) and
activity and hormone secretion (Janssen et al. 1994;
a part that does not receive this direct input
Scheer et al. 2003; Silver and Schwartz 2005). First,
(shell). The retinorecipient part contains vasoac-
animals bearing bilateral lesions of the SCN become
tive intestinal peptide (VIP) and gastrin-releasing
arrhythmic for many rhythms including blood pres-
peptide (GRP) as major neurotransmitters,
sure, heart rate, and activity as well as hormone secre-
whereas the shell area contains arginine vasopres-
tion (Scheer et al. 2001; Janssen et al. 1994; Zucker
sin (AVP) and more medially somatostatin (SOM)
et al. 1983). Secondly, SCN neurons show endogenous
(Romijn et al. 1996, 1997) as its main neurotrans-
rhythmicity in electrical activity and metabolism, both
mitters. Intricate interactions between the core and
in in vivo and in vitro conditions (Silver and Schwartz
shell region (Varadarajan et al. 2018) enable the
2005). Finally, transplantation of embryonic SCN tis-
SCN to synchronize its endogenous activity to
sue into the animals arrhythmic due to lesions of the

CONTACT Ruud M Buijs ruudbuijs@gmail.com Department of Cell Biology and Physiology, Institute for Biomedical Research, Universidad Nacional
Autonoma de Mexico, Mexico City, Mexico
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/icbi.
© 2019 Taylor & Francis Group, LLC
2 A. YILMAZ ET AL.

environmental or bodily changes, such as the light- with age, and blood pressure rising progressively
dark cycle or the daily rhythm in blood pressure, with maturation (Yamori et al. 1974). They have
respectively (Brown and Piggins 2009; Buijs et al. increased blood pressure, increased heart weight and
2014). The core region containing mainly VIP is corticosterone levels which are also observed in
essential not only for conveying the entrainment human hypertensives (Dunn and Pfeffer 1999; Eilam
signals but also for maintaining the endogenous et al. 1991; Filipovský et al. 1996; Watlington et al.
network oscillatory activity (Harmar 2003). 1992). Besides, SHRs show neurotransmitter changes,
Developmentally the VIP containing core region especially in the hypothalamus which could be the
can be divided in two separate sub-compartments, underlying mechanism for the development of hyper-
per-1 expressing self-oscillatory medial compart- tension. They have decreased levels of AVP and oxy-
ment and GRP- co-localized lateral compartment tocin (OXT) (Morris and Keller 1982) as well as
(Ban et al. 1997). These sub-compartments still are changed corticotrophin-releasing hormone (CRH)
detectable in adult animals functionally and levels in the hypothalamic paraventricular nucleus
neuro-anatomically (Kawamoto et al. 2003; (PVN) (Hattori et al. 1986), an important relay center
Mazuski et al. 2018). The early developing medial- for the regulation of autonomic and endocrine output
VIP region has endogenous oscillatory activity in influenced by the SCN. Interestingly, SHRs also exhi-
embryonic and early postnatal life, and it lacks bit abnormalities in their circadian physiology. They
light induced Per-1 and GRP expression in adult show increased phase advances and decreased phase
(Ban et al. 1997). The lateral-VIP region, on the delays in their activity rhythm as a response to noc-
other hand, expresses light-induced per-1 but lacks turnal light as compared to normotensive WKYs
diurnal variation of VIP expression in constant (Peters et al. 1994). They have an inverted daily
conditions since peptide content of this region is rhythm in blood pressure, but not heart rate after
dependent on the changes in environmental light the establishment of hypertension (Minami et al.
(Kawamoto et al. 2003). This might show sensory 1988; Munakata et al. 1990). The amplitude of their
information dependency of lateral-VIP region for daily locomotor activity as well as their blood pressure
VIP expression in the SCN. rhythm is increased when they get hypertensive
Many human postmortem studies indicate the (Canal-Corretger et al. 2001; Yilmaz et al. 2018).
involvement of the SCN in pathophysiology and SHRs have more VIP in their SCN (Peters et al.
show alterations of its subdivisions in different con- 1994) and the number of AVP neurons was decreased
ditions. Aging affects the rhythmicity and number of in wildtype hosts 8 months after receiving an SCN
AVP neurons in the SCN and differentially affects the graft of hypertensive animals (Eilam et al. 1994).
number of VIP cells in male and female subjects Considering these findings, we aimed to investigate
(Zhou et al. 1995). Additionally, the AVP cell number whether the observed neurochemical changes of the
is decreased in depression, Alzheimer disease (Swaab SCN are the cause or consequences of hypertension.
2003) and multiple system atrophy (MSA) Therefore, we conducted experiments with SHRs and
(Benarroch et al. 2006). Finally, AVP and VIP immu- their normotensive controls, using the SHRs as
noreactivity was diminished in the SCN of hyperten- a model of central nervous system mediated, top-
sive patients as compared to normotensive controls down control of hypertension. In addition, we
(Goncharuk et al. 2001), implying the involvement of induced hypertension in normotensive animals by
the SCN also in the etiology of this disease. In view of using the 2 Kidney 1 Clamp (2K1C) protocol, which
the striking animal experimental observations that is a model for peripherally mediated, bottom-up con-
both AVP and VIP are essential for adequate SCN trol of hypertension (Li and Satinoff 1998; Zheng and
rhythmicity, it becomes very attractive to hypothesize Patel 2017). Following this, we evaluated the AVP,
that the diminishment of these neuronal populations SOM and VIP levels in the SCN with semi-
in these diseases is directly associated with the quantitative immunohistochemistry before, during
observed pathology. and after the animals established hypertension by
Spontaneously hypertensive rats (SHR) are paying special attention to changes in the VIP con-
a widely accepted model of essential hypertension in taining medial and lateral subdivisions. Before sacri-
humans. They develop hypertension endogenously fice, intra-carotid blood pressure and heart rate
CHRONOBIOLOGY INTERNATIONAL 3

recordings were performed to establish the level of Blood pressure measurements were performed
hypertension. before the termination of the experiments (see
The results indicate that in the SCN AVP was below). In the second set of experiments, 10-
decreasing and SOM was increasing with the estab- week old male Wistar albino rats underwent sur-
lishment of hypertension, while VIP staining was high gery to induce hypertension (n = 5) or for sham
in SHRs compared to Wistar Kyoto (WKY) rats at all operations (n = 3) (for surgery see below). Two
time points measured. Interestingly, injections into weeks after the induction of hypertension or sham
the NTS (Nucleus Tractus Solitarius – sensory nucleus surgery, blood pressure measurements were done,
of the Vagus Nerve), of CtB (Cholera toxin B), animals were terminated and brains were removed
a neuronal tracer, revealed retrogradely labeled NTS- and tissue processed for further immunohisto-
projecting cells in the SCN corresponding to the chemical analyses.
medial-VIP region, as well as NTS fibers targeting NTS CtB injections were performed on another
the lateral-VIP region of the SCN. By analyzing VIP group of male 8-week old Wistar rats (n = 6) housed
levels in these sub-compartments of the SCN, we individually. All animals undergoing the surgery for
showed that VIP increased in this medial-VIP CtB injections (see NTS CtB injections) were anesthe-
whereas it decreased in the lateral-VIP region in tized with ketamine (50 mg/kg) and xylazine (2 mg/
SHRs after the establishment of hypertension. On kg) (Pisa-Agropecuaria S.A. de C.V.; Atitalaqia,
the contrary, VIP was decreased in the medial but Mexico).
increased in the lateral-VIP region in 2K1C- All experiments were conducted under the
renovascular hypertensive rats after the development approval of the ethical committee of the Royal
of hypertension. Netherlands Academy of Art and Sciences (KNAW)
and Netherlands Institute for Neuroscience and the
Instituto Investigaciones Biomedicas.
Methods
Animals Surgery (2K1C hypertension)
Experiments were conducted with a total of 36 Animals (n = 5) were operated under Hypnorm
male SHR and WKY rats (parents from Harlan, (0.05 ml/100g BW, im, Janssen, High Wycombe,
UK). Additionally, 8 male Wistar Albino rats (10 Buckinghamshire, UK) and Dormicum (0.04 ml/
weeks old, Harlan, Zeist, The Netherlands) were 100g BW, sc, Roche, Almere, The Netherlands)
used for the 2K1C induction of hypertension anesthesia in order to induce hypertension
experiments. All SHR and WKY rats used in this (Okamura et al. 1986). Laparotomy was performed
experiment were bred in our own animal facility in to expose the abdominal aorta by a longitudinal
order to avoid possible interfering effects of stres- incision on the abdominal midline and intestines
sors (i.e., transportation, changes in housing con- in the midline were held aside. The aorta was lifted
ditions, etc.) (Tucker and Johnson 1981) on the between the renal arteries to put a surgical thread
development of hypertension in SHRs. The ani- (4–0 Perma-Hand Seide Ethicon) underneath,
mals were housed as groups, under a 12:12 h light: after separating visible nerve fibers traveling with
dark (LD) cycle (±100 and 5 lux, respectively; the aorta from the vessel. The aorta was tied
lights on 07:00 h), in a temperature controlled together with a blunted injection needle (25
environment (21 ± 2°C) with ad libitum access to gauge, 11/4 inch needle) on top of it. After remov-
tap water and food (Rat Chow). ing the needle, the aorta was narrowed to the
Two sets of experiments were conducted to diameter of the needle. The abdominal wall mus-
reveal the possible neurochemical changes of the cles and the skin were sutured separately. Sham-
SCN after development of hypertension. In the operated animals (n = 3) underwent the same
first set of experiments brains of male SHRs and procedure without narrowing the aorta. Animals
WKYs were collected before, during and after the recovered overnight and were given Temgesic
development of hypertension, namely, at 5, 12 and (Schering-Plough, Maarssen, The Netherlands,
16 weeks of age (n = 6 per group), respectively. 0.01 ml/100 g BW) to reduce post-operative pain.
4 A. YILMAZ ET AL.

Blood pressure measurements, NTS ctb injections three series of adjacent sections – the first for
and tissue collection AVP, second for VIP, third for SOM – were
taken at 240 µm intervals through the complete
Blood pressure measurements were performed by the
SCN. Sets of serial sections of hypertensive and
intra-carotid catheterization method, two weeks after
control hypothalamus, containing 3 SCN sections
the operation for 2K1C and sham-operated rats, in
per animal, were stained in parallel, under the
the first half of the animals’ inactive period (ZT, Z,
same conditions and using the same solutions.
5–6)eitgeber time. For this procedure, animals were
For every time period hypertensive (SHR and
gas anesthetized with isoflurane (Isoflurane;
IndH) and control hypothalami (WKY and sham-
Pharmachemie BV, Haarlem, The Netherlands, 3%
operated controls) were stained at the same time
in oxygen) followed by sodium pentobarbital
in order to ensure that all conditions for staining,
(Nembutal, Ceva Sante Animal B.V, Maassluis, The
including solutions, were the same.
Netherlands, 0.06 ml/100g BW) anesthesia. Blood
Free-floating sections were pretreated with
pressure and heart rate were measured and recorded
absolute methanol and 3% H2O2 for 10 min.
for 10 min via an intra-carotid catheter connected to
After thorough washing in TBS, all sections were
a computer using HDAS software (Maastricht
incubated overnight at 4°C with either AVP
University).
(1:4000; Truus, NIN), VIP (1:4000; Viper, NIN)
To perform NTS CtB injections, after anesthe-
or SOM (1:4000; Somar, NIN) antibodies as
sia, rats were placed in the stereotact with the head
described previously (Romijn et al. 1997, 1996).
fixed at 45°. Dura and arachnoid membranes were
Following the incubation with the primary anti-
dissected exposing the dorsal surface of the
body overnight, sections were incubated for 1
medulla at the level of the area postrema. To target
h with a secondary antibody, biotinylated goat
the NTS, the tip of the micropipette was aligned
anti-rabbit IgG (Vector Laboratories Inc.,
perpendicular to the medulla and placed 0.4 mm
Burlingame, California, USA; 1:400) and then
rostral, 0.5 mm lateral of the obex and 0.5 mm
incubated with ABC (Vector; 1:800) for 1
below the surface of the brainstem. Injections were
h. Finally, sections were incubated with 0.05%
made with 100nl CtB using a glass micropipette.
3.3’- diaminobenzidine tetrachloride and 0.01%
Rats were terminated after 14 days, allowing opti-
hydrogen peroxide for 7 min. For CtB staining,
mal transport of CtB.
sections were incubated in biotinylated donkey-
After receiving an overdose of Nembutal, all
secondary antibody (Jackson Immunoresearch,
animals were transcardially perfused with saline
West Grove, PO, USA; 1:400) for 1.5 h and then
followed by solution of 4% paraformaldehyde in
in an avidin–biotin complex (Vector, Burlingame,
0.1 M phosphate buffer (pH 7.4) at 4°C. Five
CA, USA, 1:500) solution. The staining was per-
weeks old and 12 weeks old SHRs as well as 2K1C-
formed with a solution of 0.025% diaminobenzi-
hypertensive and sham-operated rats were per-
dine (DAB), 10% NiNH4SO4 and 0.01% H2O2
fused at ZT5.5 and rest of the rats were perfused
(Sigma–Aldrich) in Tris-buffered saline (TBS,
at ZT6 after the blood pressure and heart rate
0.01 M, pH 7.6), for 10 min. All sections were
measurements. For further anatomical evaluations,
mounted on gelatinized slides, dried, dehydrated
brains were removed and post-fixed in parafor-
with graded solutions of ethanol, soaked in xylene,
maldehyde solution overnight.
and finally coverslipped in Entellan embedding
agent (Merck).
Immunohistochemistry
Quantitative study and data analyses
After overnight post-fixation in paraformaldehyde,
brain tissue was further incubated for 3 days with The quantitative estimation of the density of the
30% sucrose in 0.1 M Tris-buffered saline (TBS, AVP, SOM and VIP positive neuronal elements in
pH 7.4). After this, SCN containing hypothalamic the hypothalamic SCN sections was performed in
tissue was cut into 40 µm coronal cryostat sections two steps (Goncharuk et al. 2011). The bilateral
at −20°C. For immunohistochemical analyses, results from all SCN sections (3 per animal) were
CHRONOBIOLOGY INTERNATIONAL 5

combined for analysis. First, using a microscope 5.64, p < .0001) blood pressure levels were signifi-
from Axioskop (Zeiss, Germany) equipped with cantly higher in SHRs compared to WKYs. In
a black-and-white CCD camera (Sony XC-77, a similar manner, systolic (164.4 ± 8.10 vs 107.7 ±
Japan) and run by the computer program Image- 2.6, p = .002), diastolic (121.8 ± 7.61 vs 79.67 ± 2.03,
Pro Plus 6.3 (Media Cybernetics, Bethesda, MD), p = .006) and mean arterial (137.8 ± 7.27 vs 88.67 ±
we recorded tiled images of the SCN in rat 2.02, p = .002) blood pressure levels were significantly
hypothalamus under a 40x NeoFluar objective higher in the 2K1C hypertensive animals 2 weeks after
(Zeiss), which were further combined into one surgery as compared to their sham-operated controls.
large picture. In order to enhance the contrasts Table 1 indicates blood pressure and heart rate levels
of neuronal elements filled with a final product of the animals as measured at the end of the
of the histochemical reaction, we used an optical experiments.
filter conveying a light with a wavelength of In the same animals, AVP immunoreactivity fol-
430–490 nm. To measure the optical density in lowed an interesting pattern, with SHRs having
the images, special attention was paid to adjusting a significantly higher amount of AVP in their SCN
the saturation of the light current such that the as compared to WKYs at the age of 5 weeks (43.47 ±
non-stained areas of all animals had the same level 3.41 vs 23.56 ± 2.11; p < .0001). At the age of 12 weeks,
of intensity. The second step involved projecting however, this difference in integrated optical density
the large image onto the computer screen where (IOD) of AVP staining in the SCN had disappeared
the SCN was manually outlined. Within the out- (33.76 ± 3.49 vs 41.11 ± 4.52; p = .204), while at the
lined area, the AVP, SOM or VIP immunoreactiv- age of 16 weeks, after the animals had fully developed
ity was analyzed and quantified as integrated hypertension, the IOD for AVP was even lower in
optical density with the program ImageJ (NIH; SHRs’ compared to WKYs’ (21.5 ± 1.97 vs 38.02 ±
Bethesda, MA, USA) (Saderi et al. 2014). The 4.17; p < .0001). No significant differences were
data obtained from the control and hypertensive detected between 2K1C hypertensive animals and
groups were analyzed using one-way ANOVA. their sham-operated controls for IOD of AVP in the
Simple regression analysis was performed to ana- SCN (41.1 ± 4.1 vs 50.19 ± 8.64; p = .291) (Figure 1a
lyze the relationship between parameters obtained and Figure 2).
from neuropeptides and heart rate and blood pres- A similar analysis was performed for SOM. The
sure measurements. Significance was determined IOD of SOM in the SCN of 5 weeks old SHRs was
at p < .05 for all tests performed. All data pre- significantly lower than that of age-matched
sented as mean±S.E.M. WKYs (31.06 ± 3.98 vs 64.85 ± 9.08; p < .0001).
On the other hand, at the age of 12 weeks when
animals started to become hypertensive the differ-
Results
ence in SCN SOM between SHRs and WKYs had
At the age of 5 weeks, no significant differences in disappeared (51.75 ± 5.52 vs 45.27 ± 5.08; p =
cardiovascular parameters were observed between .394). At the age of 16 weeks, on the other hand,
SHRs and WKYs, namely, systolic blood pressure the IOD of SOM in the SHR’s SCN was higher
(SBP; 101.3 ± 7.03 vs 103.0 ± 5.35, p = .854), diastolic than in that of WKYs’ (56.31 ± 6.24 vs 26.52 ±
blood pressure (DBP; 74.33 ± 7.95 vs 78.00 ± 4.38, p = 2.79; p < .0001). No significant differences were
.695), mean arterial pressure (MAP; 81.67 ± 6.65 vs found between 2K1C hypertensive and sham ani-
86.33 ± 4.34, p = .570) and heart rate (HR; 328.0 ± mals for the SOM IOD in the SCN (70.14 ± 9.81 vs
15.86 vs 294.3 ± 10.37, p = .106). However, at the age 70.66 ± 10.78; p = 00973) (Figure 1b and Figure 3).
of 12 weeks and 16 weeks, all three parameters, VIP immunoreactivity was first measured for
namely, systolic (12 weeks; 177.8 ± 10.65 vs 119.7 ± the whole SCN. This IOD analysis revealed that
10.23, p = .004, 16 weeks; 185.3 ± 8.65 vs 107.8 ± 7.29, SHRs had a higher amount of VIP in their SCN
p < .0001), diastolic (12 weeks; 132.4 ± 6.76 vs 88.17 ± compared to WKYs already at the age of 5 weeks,
7.20, p = .002, 16 weeks; 145.5 ± 4.37 vs 84.8 ± 6.28, i.e., before they develop hypertension (309.7 ±
p < .0001) and mean arterial (12 weeks; 148.6 ± 8.75 vs 27.67 vs 237.6 ± 18.73; p = .033). At the age of
98.5 ± 7.82, p = .002, 16 weeks; 158 ± 5.61 vs 92.4 ± 12 weeks, hypertensive SHRs still had a higher
6 A. YILMAZ ET AL.

Table 1. Blood pressures and heart rate variables of SHR, WKY, 2K1C and Sham animals during different stages of the experimental
protocol.
5 weeks 12 weeks 16 weeks 2K1C (12 weeks)
SHR (n = 6) WKY (n = 6) SHR (n = 5) WKY (n = 6) SHR (n = 6) WKY (n = 5) 2K1C (n = 5) Sham (n = 3)
SBP• (mmHg) 101.3 ± 7.03 103.0 ± 5.35 177.8 ± 10.65* 119.7 ± 10.23 185.3 ± 8.65* 107.8 ± 7.29 164.4 ± 8.10* 107.7 ± 2.6
DBP¶ (mmHg) 74.33 ± 7.95 78.00 ± 4.38 132.4 ± 6.76* 88.17 ± 7.20 145.5 ± 4.37* 84.8 ± 6.28 121.8 ± 7.61* 79.67 ± 2.03
MAP‡ (mmHg) 81.67 ± 6.65 86.33 ± 4.34 148.6 ± 8.75* 98.5 ± 7.82 158 ± 5.61* 92.4 ± 5.64 137.8 ± 7.27* 88.67 ± 2.02
HR§ (bpm) 328.0 ± 15.86 294.3 ± 10.37 318.8 ± 14.19 298 ± 20.31 316.3 ± 11.54 312.6 ± 12.86 333.6 ± 16.4 330.3 ± 19.63
Data presented as mean ± S.E.M. * presents significant difference (p< .05, ANOVA) between comparable groups. • SBP; systolic blood pressure
(mmHg), ¶ DBP; diastolic blood pressure (mmHg), ‡ MAP; mean arterial pressure (mmHg), § HR; heart rate (beats per minute-bpm)

IOD of VIP in their SCN than WKYs’ (230.1 ± 4.08, p = .021) (Figure 5a and Figure 5b, Figure
19.11 vs 151.1 ± 12.56; p < .0001). Also at the age 6c and Figure 6d). Interestingly, the IOD ana-
of 16 weeks, a significantly higher IOD for VIP lysis for medial-VIP region in SCN in 2K1C
was measured in the SCN of SHRs compared to and sham animals revealed significant differ-
WKYs (158 ± 11.37 vs 108 ± 14.77; p = .009). ences between groups, but in a direction oppo-
Again, similar as with AVP and SOM, no signifi- site to the results observed in 16 weeks SHR
cant differences in total VIP IOD in the SCN were animals. Significantly lower VIP levels were
found between 2K1C hypertensive and sham ani- detected in the medial-SCN (58.25 ± 5.474 vs
mals (198.8 ± 25.38 vs 230.1 ± 23.41; p = .392) 87.58 ± 14.89, p = .04), but significantly higher
(Figure 1c and Figure 4). VIP detected in lateral-SCN (51.77 ± 6.74 vs
Next to the total analysis, we also performed 28.62 ± 7.41, p = .043) in induced-
IOD analysis at the sub-compartmental level for hypertensive rats compared to aged-matched
the two VIP subdivisions of the SCN sham-operated controls (Figure 5a and Figure
(Kawamoto et al. 2003), in SHRs and WKYs 5b, Figure 6e and Figure 6f).
as well as in the 2K1C hypertensive rats and
their controls. The two sub-divisions being the
medial- and lateral VIP region of the SCN. The
NTS tracing
medial VIP region covers the area where NTS
projecting SCN neurons are located, and the Successful injections into the NTS (n = 4) (Figure
lateral-VIP region overlaps with the SCN area 6b) showed numerous cell bodies in the hypotha-
where the fibers from the NTS terminate (see lamus (paraventricular nucleus and lateral
NTS tracing results). Analyses at the sub- hypothalamus) and distinct groups of neurons in
compartmental level revealed interesting the SCN, mainly in the medial unilateral SCN. In
changes during the development of hyperten- addition also mainly in the unilateral SCN, in the
sion: VIP levels in the medial-SCN were not ventral and lateral part, small caliber fibers were
significantly different at the age of 5 (102.3 ± present in the same area where changes in VIP
6.61 vs 103.9 ± 10.77, p = .899) or 12 weeks immunoreactivity were found following the 2K1C
(101.5 ± 6.79 vs 93.55 ± 7.76, p = .445), but treatment (Figure 6a).
were significantly higher (78.29 ± 7.93 vs 50.16 The interaction of expression levels of SCN
± 4.6, p = .003) at the age of 16 weeks. On the neuropeptides and cardiovascular and body
other hand, in the lateral-VIP region of the weight parameters
SCN the SHRs compared to WKYs showed First of all, we did not observe a significant correla-
a significantly higher VIP-IOD at the age of 5 tion between AVP and VIP in individual SHR rats,
weeks (96.98 ± 8.56 vs 68.10 ± 5.85, p = .019), neither at 5 (Pearson’s r = −0.018, p = .973) nor at 12
while reversing that difference to lower levels in (r = 0.319, p = .537), nor at 16 (r = −0.342, p = .507)
the course of the development of hypertension: weeks of age.
12 weeks (65.35 ± 6.16 vs 48.28 ± 5.92, p = However, there were negative correlations between
.059) and 16 weeks (14.69 ± 2.97 vs 26.78 ± AVP and overall VIP (r = −0.875, P = .023) and VIP
CHRONOBIOLOGY INTERNATIONAL 7

Figure 1. Quantitative (integrated optical density – IOD)


Figure 2. Illustrations of coronal sections of
analysis of AVP, SOM and VIP protein immunoreactivity
Suprachiasmatic Nucleus (SCN) in hypertensive (A-D) and
in the bilateral SCN of hypertensive (HYPT) SHR and 2K1C-
control (E-H) rats stained for Vasopressin (AVP). All sections
induced hypertensive and normotensive (NRMT) WKY and
show the central part (mid-level) of the SCN containing the
sham-operated rats (A, B, C). AVP immunoreactivity was
largest number of AVP immunostained neurons. It is clear by
increased at the age of 5 weeks (5wks- prehypertensive period)
comparing A to E (5 weeks – 5wks), B to F (12 wks) and C to
and then decreased at the age of 16 weeks (hypertensive
G (16 wks), respectively, that spontaneously hypertensive rats
period) in SHRs compared to WKYs (A). In contrast to AVP,
(SHRs; A, B, C) had first an increased and then a decreased
SOM first was decreased (5wks) and then increased (16wks) in
amount of AVP staining intensity compared to normotensive
SHRs compared to WKY (B). VIP was significantly higher for all
Wistar Kyoto rats (WKYs; E, F, G). However, no significant
the time points evaluated (5wks, 12wks, 16wks, respectively) in
difference was observed between 2 Kidney 1 Clamp- induced
SHRs than WKYs (C). No significant differences were detected
hypertensive rats (2K1C; D) and their sham-operated controls
between 2K1C-induced hypertensive and their sham-operated
(H) for AVP immunostaining intensity. * Third ventricle. Scale
controls for any of the neuropeptides examined (A, B, C). * =
Bar = 50 µm (A-H).
p < .05, one-way ANOVA, in SHRs compared to WKYs (A, B, C).

(r = – 0.785, p = .065) at the age of 12 weeks.


medial (r = −0.959, p = .002) expression levels in Conversely, there was a positive correlation between
WKYs at the age of 5 weeks. And a pattern of negative AVP and VIP in 2K1C-hypertensive rats (r = 0.908,
correlation was detected between AVP and VIP lateral p = .033).
8 A. YILMAZ ET AL.

correlation between, VIP lateral expression and


heart rate levels in SHRs (r = −0.834, p = .039) at
the age of 12 weeks. At the age of 16 weeks, there
was a negative correlation between overall VIP
expression levels and heart rate levels (r = −0.967,
p = .002) and there was a positive correlation of
VIP lateral expression and heart rate levels (r =
0.0879, p = .021) in SHRs. There was a strong, but
not significant, positive correlation between
Systolic BP and VIP lateral expression levels (r =
0.767, p = .075) in WKYs at 12 weeks of age. Also
a strong, but not significant, positive correlation
between SOM and HR levels (r = 0.804, p = .054)
was detected in WKYs at the age of 16 weeks.
Conversely, what is observed in WKY at the age
of 12 weeks, there was a very strong negative
correlation between Systolic blood pressure and
VIP lateral expression levels (r = −0.901, p = .037)
in 2K1C-hypertensive rats. Interestingly, perfect
negative correlations were detected in between
SOM expression and HR levels (r = −1.000, p =
.011) in 2K1C-sham animals. This is opposite of
what was observed with 16 weeks old WKY rats.
In addition to this, there was also a negative cor-
relation between SOM expression and Systolic
blood pressure levels (r = −0.997, p = .052) in
2K1C-sham animals.
In addition to these, there might be a small differ-
ence in body weight at the age of 5 weeks (SHR vs
WKY; 109.5 ± 8.66 vs 137.3 ± 9.51) but this is not
Figure 3. Illustrations of coronal sections of significant (p = .056), and absent at the age of 12
Suprachiasmatic Nucleus (SCN) in hypertensive (A-D) and
control (E-H) rats stained for Somatostatin (SOM). All sec- weeks (294.3 ± 11.42 vs 310.7 ± 6.39, p = .241) and
tions show the central part (mid-level) of the SCN where the also at the age of 16 weeks (434.3 ± 6.98 vs 428.3 ±
highest density of SOM immunostaining was observed. In con- 4.08, p = .475). In addition, there was no difference in
trast to AVP (Figure 2), it is clear by comparing A to E (5 body weight between 2K1C-hypertensive and sham
weeks – 5wks), B to F (12 wks) and C to G (16 wks), respectively,
that spontaneously hypertensive rats (SHRs; A, B, C) had first animals (2K1C-hypertensive vs sham; 329.4 ± 10.09
a decreased and then an increased amount of SOM staining vs 350.3 ± 5.23, p = .185). Furthermore, we did not
intensity compared to normotensive Wistar Kyoto rats (WKYs; E, find any correlation between neuropeptide expression
F, G). However, no significant difference was detected between
2 Kidney 1 Clamp-induced hypertensive rats (2K1C; D) and their
levels of animals and the body weight at the age of 5
sham-operated controls (H) for SOM immunostaining intensity. weeks (data not shown). However, there was
Scale Bar = 50 µm (A-H). a positive correlation between VIP expression and
body weight at the age of 12 weeks in SHRs (r =
0.812, p = .0495) but not at the age of 16 weeks (r =
Secondly, we detected a negative correlation −0.514, p = .296). Apart from these, there was
between VIP medial expression and heart rate a positive correlation between systolic blood pressure
levels of SHR animals (r = −0.828, p = .042) at and body weight in WKYs at the age of 12 weeks (r =
the age of 5 weeks. However, there was a negative 0.887, p = .018) but a pattern of negative correlation
CHRONOBIOLOGY INTERNATIONAL 9

Figure 5. Quantitative (integrated optical density – IOD)


analysis of VIP protein expression in the bilateral medial
(A) and lateral-VIP (B) regions of SCN of hypertensive
(HYPT; SHR and 2K1C-induced hypertensive) and normo-
tensive (NRMT; WKY and sham-operated) rats. Protein
expression of VIP was not significantly different at the age of
5 (5wks- prehypertensive period) and at the onset of hyperten-
sion at 12 weeks (12 wks) in SHRs compared to WKYs in the
medial-VIP region . However, at the age of 16 weeks (16wks-
hypertensive period) SHR had significantly more VIP compared
to WKY in medial-SCN. In contrast to this, 2K1C- induced
Figure 4. Illustrations of Coronal sections of hypertensive rats had significantly lower VIP levels compared
Suprachiasmatic Nucleus (SCN) in hypertensive (A-D) and to sham- operated animals in medial- VIP region of the SCN (A).
control (E-H) rats stained for Vasoactive Intestinal Peptide On the other hand, SHRs had more VIP in the lateral region of
(VIP). All sections show the central part (mid-level) of the SCN. the SCN at the age of 5wks compared to WKYs. However no
It is clear by comparing A to E (5 weeks – 5wks), B to F (12 wks) significant differences observed between SHRs and WKYs at the
and C to G (16 wks), respectively, that VIP expression was age of 12 wks. Interestingly, opposite to levels of 2K1C animals
higher at all the time points evaluated in spontaneously hyper- and the results in A, SHRs had less VIP expression in lateral VIP-
tensive rats (SHRs; A, B, C) compared to normotensive Wistar region compared to WKYs’ (B). * = p < .05, one-way ANOVA.
Kyoto rats (WKYs; E, F, G). However, no significant difference
was observed between 2 Kidney 1 Clamp-induced hypertensive
rats (2K1C; D) and their sham-operated controls (H) for VIP expression, the SCN of spontaneously hypertensive
immunostaining intensity. Scale Bar = 50 µm (A-H).
rats (SHRs) is already different from their WKY
controls before the animals have established
between these two parameters observed in 2K1C- hypertension. Furthermore, the present observa-
hypertensive rats (r = −0.833, p = .079). tions show that after the development of hyperten-
sion several of these changes are reversed. These
observations are in agreement with previous
Discussion observations that the functionality of the SHR
Using immunohistochemical analysis, the present SCN had already changed before the animals
study shows that with respect to its neuropeptide established hypertension (Yilmaz et al. 2018).
10 A. YILMAZ ET AL.

Figure 6. SCN–NTS interactions after CTB injection into the NTS and illustrations of coronal sections of Suprachiasmatic
Nucleus (SCN) in hypertensive and control rats stained for Vasoactive Intestinal Peptide (VIP) indicating differences in
staining intensity in medial-VIP region of the SCN. Site of the CTB injections in the NTS (B). CtB (green) cell bodies are present in
the medial- region of the SCN (thick white arrow), together with the fibers in the lateral- region of SCN (white arrows) (A). The VIP
expression was higher in the medial- but was lower in the lateral-VIP region of the SCN in spontaneously hypertensive rats (SHRs; C)
compared to normotensive Wistar Kyoto rats (WKYs; D) at the age of 16 weeks. However, VIP expression was lower in medial- but
higher in lateral- VIP region of the SCN in 2 Kidney 1 Clamp-induced hypertensive rats (2K1C; E) compared to their sham-operated
controls (sham; F). Scale Bar = 50 µm (A-F).

Taken together these observations indicate that the SCN and NTS fibers in the lateral-VIP part of
early changes in the SCN may contribute to the the SCN drew our attention to VIP staining differ-
development of hypertension. On the other hand, ences in these two sub-divisions of the SCN.
the reversal of these changes after the development Interestingly, both in the SHR and the 2K1C ani-
of hypertension indicates that the SCN may not mals these two sub-compartments showed oppo-
only control, but is also sensitive for the changes in site changes during the development of
blood pressure, an observation that fits with earlier hypertension. In the 2K1C rats, these opposite
studies indicating the NTS as a possible source for changes canceled each other out when analyzing
this feedback (Buijs et al. 2014). The presence of the whole SCN. The presence of these opposite
NTS-projecting neurons in the medial-VIP part of alterations in SCN areas associated with the output
CHRONOBIOLOGY INTERNATIONAL 11

to and input from the NTS suggests that these development of hypertension in SHRs; the absence of
might be the result of the feedback from the NTS changes in the 2K1C also suggests that.
and the following adjustment to the increase in In stark contrast to SOM–IR in SHRs, we found
blood pressure. that with the establishment of hypertension the
In the current study three SCN neurotransmit- AVP content in the SCN of SHRs was drastically
ter systems, i.e. VIP, SOM and AVP, were studied reduced compared to WKYs, despite the initial
by means of semi-quantitative IOD analyses of increased staining intensity before the develop-
identically and simultaneously stained bilateral ment of hypertension. Although AVP levels in
SCN sections. The observed changes are therefore the SCN and PVN were increased in the SHRs
not likely due to variations in fixation, age or other compared to WKYs after the acute immobilization
factors since the direction of the changes observed stress at the age of 25 weeks (Negro-Vilar and
were not parallel and differed for each of the three Saavedra 1980), a number of studies indicated
neuropeptides investigated. decreased levels of AVP in the hypothalamus, par-
In agreement with earlier studies (Avidor et al. ticularly in the PVN, when animals become hyper-
1989; Eilam et al. 1991), we observed in the pre- tensive (Morris et al. 1983, 1981). Besides, these
sent study that VIP immunoreactivity (VIP-IR) changes in AVP levels seemed to be site-specific in
was increased in SHRs. However, our study pro- SHRs, since no differences in the neuropeptide
vides more elaborate information than these pre- content were detected in supraoptic nucleus or
vious studies. Because in the current study we median eminence when compared to WKYs’.
included animals at different stages of the devel- Moreover, also a reduction in the number of
opment of hypertension, namely, before (5 weeks) AVP neurons in the SCN and PVN was observed
and at the onset (12 weeks), as well as after the 3 months after transplantation of a hypothalamic
establishment of hypertension (16 weeks). Hence, graft from SHRs to normotensive WKYs (Eilam
we can conclude that elevated levels of VIP espe- et al. 1994). Therefore, the alterations that take
cially before the onset of hypertension might be place within the AVP (and SOM) cells of the
the indicator of a possible important role of this SCN may be associated with the development of
neurotransmitter in the development of hypertension; also here in the 2K1C induced
hypertension. hypertension, no AVP modifications could be
In addition to the VIP changes in the SCN of observed. In human postmortem tissue, the loss
hypertensive rats, also changes in SOM and AVP of AVP content in the SCN correlates with
expression were observed. The SCN projects to and a higher CRF mRNA content in the PVN in the
receives input from a number of stress-related areas same hypertensive patients (Goncharuk et al. 2001,
(Buijs et al. 1993). Importantly, the SOM and AVP 2002). Besides, Kalsbeek et al. (1992) demonstrated
containing dorsal region of SCN receives mainly that AVP from the SCN provides a strong inhibi-
stress-related inputs from limbic and other hypotha- tory input to the PVN for corticosterone release,
lamic areas (Moga and Moore 1997). Although in which is also related with inhibition of the sympa-
normal conditions no differences in SOM and AVP thetic tone to the adrenal (Buijs et al. 1999; Leon-
hypothalamic content, including the SCN, were Mercado et al. 2017). ICV injections of CRF or
found, after stress exposure, the SOM content failed increased amounts of cortisol cause a prominent
to decrease in the SCN of SHRs as compared to hypertension in animals and men, respectively
WKYs’ (Negro-Vilar and Saavedra 1980). In the cur- (Knowlton et al. 1952; Morimoto et al. 1993).
rent study, a strong positive correlation in individual Hence, the decreased AVP content in the SCN of
WKYs and a perfect negative correlation in individual adult SHRs might exacerbate the hypertension in
2K1C-hypertensive rats were observed for SOM and SHR by increasing sympathetic tone and corticos-
heart rate levels (see results). However, we observed terone release.
that with increase in blood pressure also the SOM In spite of the resonant aforementioned changes in
content of the SCN increased. Hence, it is tempting to the SCN of spontaneously hypertensive rats and
suggest that SOM in the SCN is associated with the humans, none of the neurotransmitter systems
12 A. YILMAZ ET AL.

studied differed at the gross level between 2K1C reno- abnormal blood pressure and heart rate
vascular hypertensive and sham-operated rats. rhythms and abnormal patterns of locomotor
However, when analyzing at the sub-compartmental activity (Sheward et al. 2010). All these obser-
level changes could be observed. The medial- and vations imply the involvement of VIP signaling
lateral-VIP regions of the SCN showed changes in in cardiovascular and locomotor activity regula-
neuropeptide expression depending on the type of tion, besides light responsiveness and maintain-
hypertension. Previous evidence shows that SCN’s ing the synchrony within the SCN. In
inputs and outputs are highly specialized (Kreier accordance with the above literature, SHRs
et al. 2006; Moga and Moore 1997). As also illustrated have abnormalities in their cardiovascular
here, there is a direct input from the NTS to the SCN rhythms (Minami et al. 1988; Munakata et al.
in the lateral-VIP region and is related with receiving 1990) and they have shorter but increased
blood pressure information (Buijs et al. 2014). In the amplitude of their locomotor activity rhythms
current study, we also identified an adjacent region in in constant darkness (Yilmaz et al. 2018). In the
the SCN, containing SCN neurons projecting to the current study, correlational analysis revealed
NTS. As this SCN area containing the NTS-projecting significant negative correlations between VIP
neurons overlaps with the medial-VIP region and is expression levels and HR levels in SHRs but
differentially affected by the centrally- or peripherally a positive, not significant, correlation between
mediated hypertension, i.e., SHR or 2K1C, it might VIP lateral expression levels and systolic blood
also be related with blood pressure regulation. In pressure (SBP) levels is observed in WKYs at
SHRs, medial-VIP was increased but lateral-VIP was the age of 12 weeks. In contrast to WKYs,
decreased after the establishment of hypertension (16 a strong negative correlation between VIP lat-
weeks of age). However, exactly the opposite pattern eral expression levels and SBP is observed in
was observed after induction of hypertension in 2K1C 2K1C-hypertensive rats (see results). Moreover,
reno-vascular hypertensive rats; the VIP content was by taking into account the overall neuro-
decreased in medial, but was increased in lateral-VIP anatomical changes described here it is highly
region compared to sham-operated controls (Figure 5 tempting to speculate a crucial role of the inter-
and Figure 6). It is tempting to correlate these changes play between two different VIP sub-
with the changes of running wheel activity, that are compartments and the AVP region of the SCN
increased in SHR in constant dark conditions as for regulating blood pressure control in health
compared to normotensive rats, while 2K1C rats’ and disease (Figure 7).
activity level for wheel running was decreased com- Apart from the above-described hypertension
pared to sham-operated controls (Yilmaz et al. 2018). related changes, changes of the SCN have been
Taken together the results may imply that the VIP reported in many other human pathologies, includ-
levels in the different VIP regions of the SCN are ing aging, dementia, depression, Alzheimer’s dis-
correlated with the level of activity. ease, Parkinson’s and MSA (Buijs et al. 2016;
In mammals, the SCN has at least two func- Swaab 2003). Interestingly, high blood pressure or
tionally distinct compartments (Shinohara et al. hypertension itself is comorbid to all these pathol-
1995). The retinorecipient core region contains ogies (Fanciulli et al. 2016; Gabin et al. 2017; Nagai
VIP and self-oscillatory dorsal cap- shell- et al. 2010; Pinto 2007; Scalco et al. 2005;
region contains mainly AVP (Reghunandanan Tsukamoto et al. 2013). As also shown here, the
and Reghunandanan 2006; van Den Pol 1980). SCN importantly influences the cardiovascular sys-
Interestingly, interactions between these two tem (Buijs et al. 2014; Scheer et al. 2001).
regions and coupling within the SCN are main- Interestingly, one of the common findings in most
tained mainly by VIP signaling. One of the VIP of the aforementioned diseases is a decrease in the
receptors, Vipr2, has a major role in light sig- number of AVP neurons in the SCN. Taken
naling in the SCN and signaling between core together the observed changes in the SCN open
and shell. Interestingly, Vipr2 knockout mice new possibilities to study the underlying mechan-
(Vipr2-/-) lose synchrony within the SCN isms of important human pathologies where cardi-
(Aton et al. 2005; Harmar 2003) and also have ovascular system changes are comorbid.
CHRONOBIOLOGY INTERNATIONAL 13

Figure 7. Schematic illustration of the proposed functional organization of the SCN and its sub-compartments in the case
of blood pressure regulation and light responsiveness. When the light input reaches the SCN, it effects the VIP neurons and this
causes a decrease of the peptide levels in the lateral-VIP region; when peptide levels decrease in the lateral-VIP region this causes
a dis-inhibition and further increase in the level or secretion of AVP in the dorsal cap region (Romijn et al. 1996), AVP then inhibits
VIP expression in the medial-VIP region and this inhibition results in a dis-inhibition and thereby activation of NTS neurons.
Activation of the NTS neurons causes a further activation of vagal efferents (Niijima et al. 1992) resulting in decreased heart rate (HR)
and blood pressure (BP) during the light or animal’s rest period (Scheer et al. 2005). However, SCN also regulates short-term blood
pressure responses through the NTS (Buijs et al. 2014). When HR and BP reach higher levels, the NTS efferents excite the lateral-VIP
neurons and these neurons in turn inhibit the inhibitory medial-VIP neurons. Hence, this mechanism including the activation of NTS
neurons and vagal efferents, HR and BP decrease to normal levels. On the other hand, in the case of hypertension, the inhibitory
signal onto the medial-VIP neurons is lost, either through AVP or NTS afferents to this region. Consequently, the medial-VIP neurons
become over-activated (as observed with the IOD results in SHRs in the current study). Ultimately, over-activation of the medial-VIP
neurons causes a long-term inhibition of the vagal system and therefore results in increased HR and BP, which characterizes the
essential hypertension. Besides, involvement of SCN-VIP system in hypertension is indicated by the experimentations with VIPR2
knock out (VIPR2 KO) and also VIP KO mice. VIPR2 KO mice which become completely arrhythmic for HR and BP in constant
conditions (Hannibal et al. 2011; Sheward et al. 2010) and VIP KO male mice have a hypertensive phenotype (Said et al. 2007). 3V;
third ventricle, OCh; Optic chiasm. Ajda Yilmaz, Frederik N Buijs, Andries Kalsbeek & Ruud M Buijs.

Acknowledgments Reference
We thank Joop van Heerikhuize for his technical assistance Aton SJ, Colwell CS, Harmar AJ, Waschek J, Herzog ED,
and Dr. Valeri D. Goncharuk for his valuable advice on Aton SJ, Colwell CS, Harmar AJ, Waschek J, Herzog ED.
image analysis. 2005. Vasoactive intestinal polypeptide mediates circadian
rhythmicity and synchrony in mammalian clock neurons.
Nat Neurosci. 8:476–483. doi:10.1038/nn1419. Cited in
Declaration of interest PubMed; PMID: 15750589.
The authors report no conflict of financial or academic interest. Avidor R, Eilam R, Malach R, Gozes I. 1989. VIP-mRNA is
The authors alone are responsible for the content and writing of increased in hypertensive rats. Brain Res. 503:304–307.
this article. doi:10.1016/0006-8993(89)91679-X. Cited in PubMed;
PMID: 2605522.
Ban Y, Shigeyoshi Y, Okamura H. 1997. Development of
Funding vasoactive intestinal peptide mRNA rhythm in the rat
suprachiasmatic nucleus. J Neurosci. 17:3920–3931. Cited
This project was financially supported by Hartstichting (grant in PubMed; PMID: 9133410.
number 00B180) and Mexican CONACyT 220598 and Benarroch EE, Schmeichel AM, Sandroni P, Low PA,
CONACyT Frontera 1802 and DGAPA PAPIIT IG200417. Parisi JE. 2006. Differential involvement of
14 A. YILMAZ ET AL.

hypothalamic vasopressin neurons in multiple system and plasma cortisol levels according to degree of obesity in
atrophy. Brain. 129:2688–2696. doi:10.1093/brain/ middle-aged men. J Hypertens.. 14:229–235. Cited in
awl109. Cited in PubMed; PMID: 16670175. PubMed; PMID: 8728301.
Brown TM, Piggins HD. 2009. Spatiotemporal Heterogeneity Gabin JM, Tambs K, Saltvedt I, Sund E, Holmen J. 2017.
in the Electrical Activity of Suprachiasmatic Nuclei Association between blood pressure and Alzheimer disease
Neurons and their Response to Photoperiod. J Biol measured up to 27 years prior to diagnosis: the HUNT
Rhythms. 24:44–54. doi:10.1177/0748730408327918. Cited Study. Alzheimers Res Ther 9(37). doi:10.1186/s13195-
in PubMed; PMID: 19227579. 017-0262-x. Cited in PubMed; PMID: 28569205.
Buijs FN, Cazarez F, Basualdo MC, Scheer FAJL, Goncharuk VD, Buijs RM, Jhamandas JH, Swaab DF. 2011.
Perusquía M, Centurion D, Buijs RM. 2014. The supra- Vasopressin (VP) and neuropeptide FF (NPFF) systems in
chiasmatic nucleus is part of a neural feedback circuit the normal and hypertensive human brainstem. J Comp
adapting blood pressure response. Neuroscience. Neurol. 519:93–124. doi:10.1002/cne.22507. Cited in
266:197–207. doi:10.1016/j.neuroscience.2014.02.018. PubMed; PMID: 21120930.
Cited in PubMed; PMID: 24583038. Goncharuk VD, van Heerikhuize J, Dai JP, Swaab DF,
Buijs FN, León-Mercado L, Guzmán-Ruiz M, Guerrero- Buijs RM. 2001. Neuropeptide changes in the suprachias-
Vargas NN, Romo-Nava F, Buijs RM. 2016. The circadian matic nucleus in primary hypertension indicate functional
system: a regulatory feedback network of periphery and impairment of the biological clock. J Comp Neurol.
brain. Physiology (Bethesda). 31:170–181. doi: 1 10.1152/ 431:320–330. doi:10.1002/1096-9861. Cited in PubMed;
physiol.00037.2015. Cited in PubMed; PMID: 27053731. PMID: 11170008.
Buijs RM, Markman M, Nunes-Cardoso B, Hou YX, Shinn S. Goncharuk VD, Van Heerikhuize J, Swaab DF, Buijs RM.
1993. Projections of the suprachiasmatic nucleus to 2002. Paraventricular nucleus of the human hypothalamus
stress-related areas in the rat hypothalamus: a light and in primary hypertension: activation of
electron microscopic study. J Comp Neurol. 335:42–54. corticotropin-releasing hormone neurons. J Comp
doi:10.1002/cne.903350104. Cited in PubMed; PMID: Neurol. 443:321–331. doi:10.1002/cne.10124. Cited in
7691904. PubMed; PMID: 11807841.
Buijs RM, Wortel J, Van Heerikhuize JJ, Feenstra MG, Ter Hannibal J, Hsiung HM, Fahrenkrug J. 2011. Temporal phas-
Horst GJ, Romijn HJ, Kalsbeek A. 1999. Anatomical and ing of locomotor activity, heart rate rhythmicity, and core
functional demonstration of a multisynaptic suprachias- body temperature is disrupted in VIP receptor 2-deficient
matic nucleus adrenal (cortex) pathway. Eur J Neurosci. mice. Am J Physiol Regul Integr Comp Physiol. 300:R519–
11:1535–1544. Cited in PubMed; PMID: 10215906. R530. doi:10.1152/ajpregu.00599.2010. Cited in PubMed;
Canal-Corretger MM, Witte K, Lemmer B. 2001. Circadian PMID: 21178124.
pacemaker function and entrainment during maturation of Harmar AJ. 2003. An essential role for peptidergic signalling
transgenic hypertensive TGR(mREN2)27 and in the control of circadian rhythms in the suprachiasmatic
Sprague-Dawley rats. Chronobiol Int. 18:627–640. nuclei. J Neuroendocrinol. 15:335–338. doi:10.1046/j.1365-
doi:10.1081/CBI-100106077. Cited in PubMed; PMID: 2826.2003.01005.x. Cited in PubMed; PMID: 12622830.
11587086. Hattori T, Hashimoto K, Ota Z. 1986. Brain corticotropin releas-
Dunn FG, Pfeffer MA. 1999. Left ventricular hypertrophy in ing factor in the spontaneously hypertensive rat.
hypertension. N Engl J Med. 340:1279–1280. doi:10.1056/ Hypertension. 8:1027–1031. doi:10.1161/01.HYP.8.11.1027.
NEJM199904223401610. Cited in PubMed; PMID: Cited in PubMed; PMID: 3490439.
10210713. Janssen BJA, Tyssen CM, Duindam H, Rietveld WJ. 1994.
Eilam R, Malach R, Bergmann F, Segal M. 1991. Suprachiasmatic lesions eliminate 24-h blood pressure
Hypertension induced by hypothalamic transplantation variability in rats. Physiol Behav. 55:307–311.
fromgenetically hypertensive to normotensive rats. doi:10.1016/0031-9384(94)90138-4. Cited in PubMed;
J Neurosci. 11:401–411. Cited in PubMed; PMID: 1992008. PMID: 8153170.
Eilam R, Malach R, Segal M. 1994. Selective elimination of Kalsbeek A, Buijs RM, van Heerikhuize JJ, Arts M, van der
hypothalamic neurons by grafted hypertension-inducing Woude TP. 1992. Vasopressin-containing neurons of the
neural tissue. J Neurosci. 14:4891–4902. Cited in suprachiasmatic nuclei inhibit corticosterone release. Brain
PubMed; PMID: 7913956. Res. 580:62–67. Cited in PubMed; PMID: 1363851.
Fanciulli A, Göbel G, Ndayisaba JP, Granata R, Duerr S, Kawamoto K, Nagano M, Kanda F, Chihara K, Shigeyoshi Y,
Strano S, Colosimo C, Poewe W, Pontieri FE, Two OH. 2003. Types of VIP neuronal components in rat
Wenning GK. 2016. Supine hypertension in Parkinson’s suprachiasmatic nucleus. J Neurosci Res. 74:852–857.
disease and multiple system atrophy. Clin Auton Res. doi:10.1002/jnr.10751. Cited in PubMed; PMID:
26:97–105. doi:10.1007/s10286-015-0336-4. Cited in 14648589.
PubMed; PMID: 26801189. Knowlton AI, Loeb EN, Stoerk HC, White JP, Heffernan JF.
Filipovský J, Ducimetiére P, Eschwége E, Richard JL, 1952. Induction of arterial hypertnesion in normal and
Rosselin G, Claude JR. 1996. The relationship of blood adrenalectomized ratsgiven cortisone acetate. J Exp Med
pressure with glucose, insulin, heart rate, free fatty acids 96(3):187–205. Cited in PubMed; PMID: 14955574.
CHRONOBIOLOGY INTERNATIONAL 15

Kreier F, Kap YS, Mettenleiter TC, Van Heijningen C, Van Nagai M, Hoshide S, Kario K. 2010. Hypertension and
Der Vliet J, Kalsbeek A, Sauerwein HP, Fliers E, Dementia. Febr Am J Hypertens. 23:116–124.
Romijn JA, Buijs RM. 2006. Tracing from fat tissue, liver, doi:10.1038/ajh.2009.212. Cited in PubMed; PMID:
and pancreas: A neuroanatomical framework for the role 19927134.
of the brain in type 2 diabetes. Endocrinology. Navar LG, Zou L, Von Thun A, Tarng Wang C, Imig JD,
147:1140–1147. doi:10.1210/en.2005-0667. Cited in Mitchell KD. 1988. Unraveling the mystery of goldblatt
PubMed; PMID: 16339209. hypertension. News Physiol Sci. 13:170–176. Cited in
Leon-Mercado L, Herrera Moro Chao D, Basualdo M, Del C, PubMed; PMID: 11390784.
Kawata M, Escobar C, Buijs RM. 2017. The arcuate Negro-Vilar A, Saavedra JM. 1980. Changes in brain soma-
nucleus: A site of fast negative feedback for corticosterone tostatin and vasopressin levels after stress in spontaneously
secretion in male rats. Eneuro. 4. doi: 10.1523/ hypertensive and Wistar-Kyoto rats. Brain Res Bull.
ENEURO.0350-16.2017. Cited in PubMed; PMID: 5:353–358. doi:10.1016/S0361-9230(80)80004-9. Cited in
28275717. PubMed; PMID: 6105906.
LeSauter J, Silver R. 1999. Localization of a suprachiasmatic Niijima A, Nagai K, Nagai N, Nakagawa H. 1992. Light
nucleus subregion regulating locomotor rhythmicity. enhances sympathetic and suppresses vagal outflows and
J Neurosci. 19:5574–5585. Cited in PubMed; PMID: 10377364. lesions including the suprachiasmatic nucleus eliminate
Li H, Satinoff E. 1998. Fetal tissue containing the suprachias- these changes in rats. J Auton Nerv Syst. 40:155–160.
matic nucleus restores multiple circadian rhythms in old Cited in PubMed; PMID: 1464695.
rats. Am J Physiol. 275:R1735–44. Cited in PubMed; Okamura T, Miyazaki M, Inagami T, Toda N. 1986. Vascular
PMID: 9843862. renin-angiotensin system in two-kidney, one clip hyper-
Mazuski C, Abel JH, Chen SP, Hermanstyne TO, Jones JR, tensive rats. Hypertension. 8:560–565. doi:10.1161/01.
Simon T, Doyle FJ, Herzog ED, Mazuski C, Abel JH, et al. HYP.8.7.560. Cited in PubMed; PMID: 3013774.
2018. Entrainment of circadian rhythms depends on firing Peters RV, Zoeller RT, Hennessey AC, Stopa EG,
rates and neuropeptide release of VIP SCN neurons. Anderson G, Albers HE. 1994. The control of circadian
Neuron. 99:555–563.e5. doi:10.1016/j.neuron.2018.06.029. rhythms and the levels of vasoactive intestinal peptide
Cited in PubMed; PMID: 30017392. mRNA in the suprachiasmatic nucleus are altered in spon-
Minami N, Imai Y, Munakata M, Sasaki S, Sekino H, Abe K, taneously hypertensive rats. Brain Res. 639:217–227.
Yoshinaga K. 1988. Reversed circadian rhythm of blood doi:10.1016/0006-8993(94)91733-7. Cited in PubMed;
pressure in adult spontaneously hypertensive rats. Journal PMID: 8205475.
of Hypertension. 4:S70–73. Cited in PubMed; PMID: Pinto E. 2007. Blood pressure and ageing. Postgrad Med J.
3241281. 83:109–114. doi:10.1136/pgmj.2006.048371. Cited in
Moga MM, Moore RY. 1997. Organization of neural inputs PubMed; PMID: 17308214.
to the suprachiasmatic nucleus in the rat. J Comp Neurol. Ralph MR, Joyner AL, Lehman MN. 1993. Culture and trans-
389:508–534. doi:10.1002/(SICI)1096-9861. Cited in plantation of the mammalian circadian pacemaker. J Biol
PubMed; PMID: 9414010. Rhythms 8(Suppl):S83–7. Cited in PubMed; PMID: 8274766.
Morimoto A, Nakamori T, Morimoto K, Tan N, Murakami N. Reghunandanan V, Reghunandanan R. 2006.
1993. The central role of corticotrophin-releasing factor Neurotransmitters of the suprachiasmatic nuclei.
(CRF-41) in psychological stress in rats. J Physiol. J Circadian Rhythms. 4:2. doi:10.1016/S0361-9230(80)
460:221–229. Cited in PubMed; PMID: 8487193. 80004-9. Cited in PubMed; PMID: 6105906.
Morris M, Keller M. 1982. A specific deficiency in paraven- Romijn HJ, Sluiter AA, Pool CW, Wortel J, Buijs RM. 1996.
tricular vasopressin and oxytocin in the spontaneously Differences in colocalization between Fos and PHI, GRP,
hypertensive rat. Brain Res. 249:173–176. Cited in VIP and VP in neurons of the rat suprachiasmatic nucleus
PubMed; PMID: 7139293. after a light stimulus during the phase delay versus the
Morris M, Keller M, Sundberg DK. 1983. Changes in para- phase advance period of the night. J Comp Neurol.
ventricular vasopressin and oxytocin during the develop- 372:1–8. doi:10.1002/(SICI)1096-9861. Cited in PubMed;
ment of spontaneous hypertension. Hypertension. Jul-Aug; PMID: 8841917.
5(4):476–481. Cited in PubMed; PMID: 6862574. Romijn HJ, Sluiter AA, Pool CW, Wortel J, Buijs RM. 1997.
Morris M, Wren JA, Sundberg DK. 1981. Central neural Evidence from confocal fluorescence microscopy for
peptides and catecholamines in spontaneous and DOCA/ a dense, reciprocal innervation between AVP-,
salt hypertension. Peptides Summer(2):207–211. Cited in somatostatin-, VIP/PHI-, GRP- and VIP/PHI/
PubMed; PMID: 6117062. GRP-immunoreactive neurons in the rat suprachiasmatic
Munakata M, Imai Y, Minami N, Sasaki S, Ichijyo T, nucleus. Eur J Neurosci. 9:2613–2623. doi:10.1111/j.1460-
Yoshizawa M, Sekino H, Abe K, Yoshinaga K. 1990. 9568.1997.tb01691.x. Cited in PubMed; PMID: 9517467.
Cosinor analysis of changes in circadian blood pressure Saderi N, Buijs FN, Salgado-Delgado R, Merkenstein M,
rhythm with aging in spontaneously hypertensive rats. Basualdo MC, Ferri GL, Escobar C, Buijs RM. 2014.
Tohoku. J Exp Med. 161:55–64. Cited in PubMed; PMID: A role for VGF in the hypothalamic arcuate and paraven-
2396257. tricular nuclei in the control of energy homeostasis.
16 A. YILMAZ ET AL.

Neuroscience. 265:184–195. doi:10.1016/j.neu- disease. Brain Behav. 3:710–714. doi:10.1002/brb3.179.


roscience.2014.01.060. Cited in PubMed; PMID: 24508747. Cited in PubMed; PMID: 24363973.
Said SI, Hamidi SA, Dickman KG, Szema AM, Lyubsky S, Tucker DC, Johnson AK. 1981. Behavioral correlates of spon-
Lin RZ, Jiang YP, Chen JJ, Waschek JA, Kort S. 2007. taneous hypertension. Neurosci Biobehav Rev. 5:463–471.
Moderate pulmonary arterial hypertension in male Cited in PubMed; PMID: 7033843.
mice lacking the vasoactive intestinal peptide gene. van Den Pol AN. 1980. The hypothalamic suprachiasmatic
Circulation. 115(10):1260–1268. doi:10.1161/ nucleus of rat: intrinsic anatomy. J Comp Neurol.
CIRCULATIONAHA.106.681718. Mar 13; doi: Cited 191:661–702. doi:10.1002/cne.901910410. Cited in
in PubMed; PMID: 17309917. PubMed; PMID: 6158529.
Scalco AZ, Scalco MZ, Azul JBS, Lotufo Neto F. 2005. Varadarajan S, Tajiri M, Jain R, Holt R, Ahmed Q, LeSauter J,
Hypertension and depression. Clinics (Sao Paulo). Silver R. 2018 5:ENEURO.0205-18.2018. Connectome of
60:241–250. doi:10.1590/S1807-59322005000300010. Cited the Suprachiasmatic Nucleus: new evidence of the
in PubMed; PMID: 15962086. core-shell relationship. eneuro. doi: 10.1523/
Scheer FAJL, Kalsbeek A, Buijs RM. 2003. Cardiovascular control ENEURO.0205-18.2018. Cited in PubMed; PMID:
by the suprachiasmatic nucleus: neural and neuroendocrine 30283813.
mechanisms in human and rat. Biol Chem. 384:697–709. Watlington CO, Kramer LB, Schuetz EG, Zilai J,
doi:10.1515/BC.2003.078. Cited in PubMed; PMID: 12817466. Grogan WM, Guzelian P, Gizek F, Schoolwerth AC.
Scheer FAJL, Pirovano C, Ejw VS, Buijs RM. 2005. Environmental 1992. Corticosterone 6 beta-hydroxylation correlates with
light and suprachiasmatic nucleus interact in the regulation of blood pressure in spontaneously hypertensive rats. Am
body temperature. Neuroscience. 132:465–477. doi:10.1016/j. J Physiol. 262:F927–31. doi:10.1152/ajprenal.1992.262.6.
neuroscience.2004.12.012. Cited in PubMed; PMID: 15802197. F927. Cited in PubMed; PMID: 1621817.
Scheer FAS, Ter Horst GJ, van Der Vliet J, Buijs RM. 2001. Yamori Y, Tomimoto K, Ooshima A, Hazama F, Okamoto K.
Physiological and anatomic evidence for regulation of the 1974. Proceedings: developmental course of hypertension
heart by suprachiasmatic nucleus in rats. Am J Physiol in the SHR-substrains susceptible to hypertensive cerebro-
Heart Circ Physiol. 280:H1391–H1399. Cited in PubMed; vascular lesions. Jpn Heart J. 15:209–210. doi:10.1536/
PMID: 11179089. ihj.15.209. Cited in PubMed; PMID: 4546183.
Sheward WJ, Naylor E, Knowles-Barley S, Armstrong JD, Yilmaz A, Kalsbeek A, Buijs RM. 2018. Functional changes of
Brooker GA, Seckl JR, Turek FW, Holmes MC, Zee PC, the SCN in spontaneous hypertension but not after the
Harmar AJ. 2010. Circadian control of mouse heart rate induction of hypertension. Chronobiol Int. 1–15.
and blood pressure by the suprachiasmatic nuclei: beha- doi:10.1080/07420528.2018.1469035. Cited in PubMed;
vioral effects are more significant than direct outputs. PMID: 29787305.
PLoS One. 5:1–9. doi:10.1371/journal.pone.0009783. Cited Zheng H, Patel KP. 2017. Integration of renal sensory affer-
in PubMed; PMID: 20339544. ents at the level of the paraventricular nucleus dictating
Shinohara K, Honma S, Katsuno Y, Abe H, Honma K. 1995. sympathetic outflow. Auton Neurosci. 204:57–64. doi:
Two distinct oscillators in the rat suprachiasmatic nucleus in 10.1016/j.autneu.2016.08.008. Cited in PubMed; PMID:
vitro. Proc Natl Acad Sci U S A. 92:7396–7400. doi:10.1073/ 27527558.
pnas.92.16.7396. Cited in PubMed; PMID: 7638204. Zhou JN, Hofman MA, Swaab DF. 1995. VIP neurons in the
Silver R, Schwartz WJ. 2005. The suprachiasmatic nucleus is human SCN in relation to sex, age, and
a functionally heterogeneous timekeeping organ.Methods Alzheimers-disease. Neurobiol Aging. 16:571–576.
Enzymology. 393: 451–465. doi:10.1016/S0076-6879(05) doi:10.1016/0197-4580(95)00043-e. Cited in PubMed;
93022-X. Cited in PubMed; PMID: 15817305. PMID: 8544907.
Swaab D. 2003. Suprachiasmatic nucleus (SCN) and pineal Zucker I, Boshes M, Dark J. 1983. Suprachiasmatic nuclei
gland. Handb Clin Neurol 79(Part 1):63–125. 3rd Ser Vol influence circannual and circadian rhythms of ground
1) Hum Hypothal basic clincal Asp. squirrels. Am J Physiol Integr Comp Physiol. 244:R472–
Tsukamoto T, Kitano Y, Kuno S. 2013. Blood pressure fluc- R480. doi:10.1152/ajpregu.1983.244.4.R472. Cited in
tuation and hypertension in patients with Parkinson’s PubMed; PMID: 6837764.

You might also like