Вы находитесь на странице: 1из 11

Clinical Science (2016) 130, 9–18 doi: 10.

1042/CS20150654

Sex, the brain and hypertension: brain oestrogen


receptors and high blood pressure risk factors
Meredith Hay*

*Department of Physiology, Evelyn F. McKnight Brain Institute, Saver Heart Center, University of Arizona, Tucson, AZ, U.S.A.

Abstract
Hypertension is a major contributor to worldwide morbidity and mortality rates related to cardiovascular disease.
There are important sex differences in the onset and rate of hypertension in humans. Compared with age-matched
men, premenopausal women are less likely to develop hypertension. However, after age 60, the incidence of
hypertension increases in women and even surpasses that seen in older men. It is thought that changes in levels
of circulating ovarian hormones as women age may be involved in the increase in hypertension in older women. One
of the key mechanisms involved in the development of hypertension in both men and women is an increase in
sympathetic nerve activity (SNA). Brain regions important for the regulation of SNA, such as the subfornical organ,
the paraventricular nucleus and the rostral ventral lateral medulla, also express specific subtypes of oestrogen
receptors. Each of these brain regions has also been implicated in mechanisms underlying risk factors for
hypertension such as obesity, stress and inflammation. The present review brings together evidence that links
actions of oestrogen at these receptors to modulate some of the common brain mechanisms involved in the ability
of hypertensive risk factors to increase SNA and blood pressure. Understanding the mechanisms by which
oestrogen acts at key sites in the brain for the regulation of SNA is important for the development of novel,
sex-specific therapies for treating hypertension.

Key words: blood pressure, central nervous system, oestrogen, PVN, RVLM, sex, SFO.

INTRODUCTION central nervous system (CNS) regulation of SNA, heart rate and
ultimately BP involves a number of key brain regions in the hy-
Hypertension is a worldwide health challenge and a key factor in pothalamus and brain stem, including the nucleus of the solitary
the morbidity and mortality related to cardiovascular disease [1]. tract (NTS), nucleus ambiguus, area postrema, caudal ventrolat-
Sex differences in the rate and onset of hypertension have been eral medulla (CVLM) and rostral ventrolateral medulla (RVLM),
well established. In both women and men, there is an age-related parabrachial nucleus (PBN), the subfornical organ (SFO) and the
increase in blood pressure (BP). However, women under the age paraventricular nucleus of the hypothalamus (PVN). All these
of 50 have a lower incidence of hypertension than age-matched regions of the brain are known to express oestrogen receptors
men [2,3]. After age 55, the incidence of hypertension in women (ERs), and oestrogen modulation of these neurons and associated
markedly increases and almost 70 % of women aged over 65 in glia and oestrogen modulation of brain neurotransmission have
the U.S.A. are hypertensive [4]. Contributing risk factors to the been shown to decrease SNA and decrease BP [17]. The present
development of hypertension as women age include changes in review surveys the current understanding of the role of increased
levels of circulating ovarian hormones [5,6], obesity [7–10], salt SNA in the risk factors contributing to hypertension and the role
intake [11–13], stress [14,15] and the immune system [16]. of ERs in the brain on the regulation of SNA, and attempts to
One mechanism that is common to all these hypertensive risk shed some possible mechanistic light on how ovarian sex steroid
factors is an increase in sympathetic nerve activity (SNA). The actions on discrete brain regions responsible for the regulation of

Abbreviations: ACTH, adrenocorticotrophic hormone; Aldo, aldosterone; AngII, angiotensin-II; AT1, AngII type 1; BMI, body mass index; BP, blood pressure; CEE, conjugated equine
oestrogen; CNS, central nervous system; CO, cardiac output; CRF, corticotrophin-releasing factor; CVLM, caudal ventrolateral medulla; ER, oestrogen receptor; ERT, oestrogen
replacement therapy; GluN1, glutamate-receptor subunit 1; HPA, hypothalamic–pituitary–adrenal; HRT, hormone replacement therapy; MAPK, mitogen-activated protein kinase; NMDA,
N-methyl-D-aspartate; NO, nitric oxide; nNOS, neuronal nitric oxide synthase; NTS, nucleus of the solitary tract; OC, oral contraceptive; OVX, ovariectomized; PBN, parabrachial nucleus;
PVN, paraventricular nucleus; ROS, reactive oxygen species; RVLM, rostral ventral lateral medulla; SFO, subfornical organ; SNA, sympathetic nerve activity; WT, wild-type.

Correspondence: Meredith Hay (mhay@arizona.edu).


C 2016 Authors; published by Portland Press Limited 9
M. Hay

SNA may attenuate the development of hypertension. An under- creased BP is that OCs may result in a blunting of the baroreflex
standing of these mechanisms and their contribution to this dev- regulation of BP. However, further studies are needed to test this
astating disease are essential to the development of sex-specific hypothesis fully.
therapies for treatment and management of hypertension. With regard to HRT and changes in SNA and BP, the results
are less clear [29–31]. Factors that influence the effects of HRT on
the regulation of BP include the age of the woman when she starts
SEX, OVARIAN HORMONES AND HRT, the type of hormone being used as a replacement, how the
SYMPATHETIC OUTFLOW HRT is taken (oral vs transdermal) and any predisposing factors
for hypertension that may exist in the individual [6,31]. Results
Although it is well known that increases in SNA result in in- include the finding of the Women’s Health Initiative (WHI) trial
creases in vasomotor tone [18,19], the correlation between rest- that hormone replacement with conjugated equine oestrogens
ing sympathetic outflow and resting arterial BP in humans is (CEEs) results in a small increase in BP [33]. However, other
not what would be expected. In studies of both young men and studies have shown that CEEs result in a lower BP after 10 years
young women in which direct recordings of SNA were obtained of treatment [34]. It has been suggested that the method of HRT
in awake individuals, those with a higher SNA were found to have delivery influences CEEs’ effects on BP. Orally taken oestrogens
lower resting BP than those with a lower SNA [20,21]. In young have been shown to increase or have no effect whereas trans-
men, this higher SNA correlates with lower cardiac output (CO) dermal oestrogens reduce SNA and BP [29]. The role of the
[19], thus explaining the normal BP in the presence of higher brain, and consequently the SNA, in the effects of HRT on BP
SNA in young men. However, in young women the correlation control in experiments on direct recordings of muscle SNA have
of SNA, vascular tone and CO is absent [20,22]. Further studies been more conclusive. In studies in postmenopausal women, it
have found that young women have enhanced vascular β-receptor was found that transdermal but not oral estradiol significantly
function [23], which is thought to offset the increased SNA seen decreased SNA and had minimal effects on the BP [30,35]. It
in them [24]; thus this may contribute to the maintenance of has been suggested that estradiol’s effect to decrease SNA is due
normal BP in young women. These sex differences in the rela- to oestrogen’s direct effect on CNS regions known to regulate
tionship between SNA and BP is lost as women enter menopause. SNA.
In studies examining SNA in older women, a tight correlation was
found between SNA and BP [21]. It has been suggested that de-
creases in circulating ovarian hormones may modulate vascular BRAIN OESTROGEN RECEPTORS,
β-receptor expression, thus unmasking the vasoconstrictor ef- SYMPATHETIC OUTFLOW AND BLOOD
fects of increased SNA. Therefore, the postmenopausal increase PRESSURE REGULATION
in sympathetic outflow in women most probably contributes to
the increased incidence of hypertension in women aged over 50. Using animal models of hypertension, we have gained somewhat
Studies on the effects of ageing on SNA and BP in women have more clarity on the CNS mechanisms that may underlie the effects
found that, independent of the menopause, there is an increase in of oestrogen replacement on SNA and BP.
SNA as women age [25]. In that study, for each decade increase Earlier results from my laboratory have shown that brain ER
in life, women exhibited significantly greater increases in resting activation in key regions known to be important for the regulation
SNA compared with age-matched men. For both older men and of SNA are able to protect against hypertension induced by either
older women, there is a positive correlation between SNA and angiotensin-II (AngII) or aldosterone (Aldo) [36–38]. In both
BP, with this correlation being much stronger in older women. males and ovariectomized (OVX) females, central administration
In addition, older women have been shown to have an impaired of oestrogen attenuates AngII- or Aldo-induced hypertension.
baroreflex function compared with younger women [26]. These But, identification of the sites of action of oestrogen in the brain
results, together, suggest that an increase in SNA may be a key and the central cellular mechanisms required for oestrogen to
factor in the development of hypertension as women age. modulate the development of neurogenic hypertension are still
The effects of oral contraceptives (OCs) and hormone replace- being elucidated.
ment therapy (HRT) on resting BP and SNA have been examined
in a number of studies [27–31]. With regard to OCs, these studies The rostral ventral lateral medulla
support the conclusion that their use increases the risk of hyper- A key area of the brain stem in regulation of SNA and thus BP is
tension. This conclusion would suggest that OC use would also the RVLM. In both human and animal studies, increases in RVLM
result in an increase in resting SNA. To test this hypothesis, a activity are known to be associated with SNA and SNA-related
recent study examined the interrelationship of resting arterial BP, changes in BP [39–43]. The RVLM expresses both ERα and
SNA, total peripheral resistance (TPR) and CO in two groups of ERβ receptors [44], and activation of these receptors via direct
women between the ages of 18 and 35, either taking OCs or not injection of oestrogen into the RVLM results in a decrease in
[32]. The women taking OCs had a higher BP compared with SNA [45]. Importantly, however, it is the ERβ receptors and not
the women not taking OCs, but both groups had similar levels of the ERα receptors within the RVLM that contribute to estradiol’s
SNA. This result was unexpected given the higher resting BP in ability to inhibit increases in SNA and BP [46,47] caused by
the women taking OCs. One mechanism suggested as possibly AngII or Aldo. Recent studies selectively knocked down ERβ
contributing to the lack of change in SNA in the presence of in- receptors in the RVLM using siRNA in intact female rats, in which

10 
C 2016 Authors; published by Portland Press Limited
Brain oestrogen receptors and high blood pressure risk factors

infusion of Aldo has a minimal effect on SNA and BP compared sponsible for oestrogen’s protective effects against AngII-induced
with OVX females or males [36]. However, after knockdown hypertension [59]. These studies employed selective knockdown
of ERβ receptors within the RVLM, Aldo results in significant of the ERα receptor in the SFO using adenovirus-Cre injected
hypertension in intact females [47]. These results support the into the SFO of ERα flox mice. In the animals that had the ERα
conclusion that oestrogen acting on ERβ receptors in the RVLM receptor knocked down selectively in the SFO, AngII induced a
may be partially responsible for oestrogen’s protection against significantly greater increase in both the hypertensive response
some forms of hypertension. and the SNA, as measured by ganglionic blockade, than seen in
Sex differences in the expression of both AngII type 1 (AT1) control animals. These results suggest that the ERα receptors in
receptors and NADPH oxidase subunits have been identified in the SFO are required to protect against the increase in SNA and
the RVLM [48]. NADPH oxidase is the key enzyme involved BP caused by AngII.
in reactive oxygen species (ROS) production and known to be
essential for the development of AngII-dependent hypertension The paraventricular nucleus
[49]. Using electron microscopy and immunolabelling in males, The PVN in the hypothalamus is a key region of the brain involved
female rats in pro-oestrus and female rats in di-oestrus, the fe- in the regulation of SNA. It is the parvocellular neurons of the
males were found to have more AT1 labelling in the dendrites PVN that receive neuronal input from the SFO, and then relay
of the RVLM compared with males, but less NADPH oxidase this information to the RVLM and the interomedial column of
p47 subunit labelling [48]. In addition to the sex differences in the spinal cord region, which controls SNA.
ROS signalling in the RVLM, there are also suggestions that there The study of the role and effects of oestrogen in the modulation
are important sex differences in RVLM neurotransmission [50]. of PVN activity, and subsequent modulation of SNA and BP,
Glutamate neurotransmission and activation of the N-methyl-D- started only recently. Although both ERα and ERβ receptors
aspartate (NMDA) receptor within the RVLM is known to be have been reported as expressed in the PVN, it is the ERβ ones
important in RVLM modulation of BP [50]. Changes in the sub- that seem to be more predominantly expressed and play a major
cellular trafficking of the glutamate-receptor subunit 1 (GluN1) of role in the modulation of the PVN [60,61]. In an Aldo-induced
the NMDA receptor is related to increases in both RVLM activ- model of hypertension, blockade of the ERβ receptors in the
ity and SNA [50]. Recent studies have shown that a low-dose PVN using siRNA knockdown results in an increase in SNA and
infusion of AngII which increases blood pressure in males also BP during Aldo infusion compared with sham injections [47],
results in an increase in trafficking of GluN1 in RVLM neurons suggesting that, in the PVN, ERβ receptor activation protects
of males but not females. This sex difference in AngII-induced intact females against some types of hypertension.
excitatory neurotransmitter receptor trafficking at the level of the Only recently have the cellular and molecular mechanisms
RVLM is most probably a key cellular mechanism underlying sex within the PVN that underlie these protective effects of ERβ
differences in AngII-dependent hypertension. receptor activation started to be studied. Both ROS and nitric ox-
ide (NO) mechanisms have been implicated [17,62,63]. In recent
The subfornical organ studies using male and female mice in which the AT1 receptor
Another region of the brain known to be important in the reg- was co-localized with green fluorescent protein in the PVN [63],
ulation of SNA is the subfornical organ (SFO), which is a cir- the effects of AngII infusion on BP and ROS production in the
cumventricular organ in the forebrain that is directly involved PVN’s AT1-positive cells was determined. As has been shown
in the ability of circulating AngII to increase SNA, resulting in previously [37], AngII infusion resulted in a significant increase
neurogenic hypertension. Studies in animal models have shown in BP in the males but not in the intact females. Importantly,
that increasing circulating AngII results in increases in SFO neur- this increased BP in the males was associated with a greater
onal activity, which then increases SNA via the SFO projections production of ROS in these cells compared with intact females,
to the paraventricular nucleus (PVN) [51,52]. An important cel- suggesting that AngII induces an increase in ROS production in
lular mechanism known to be involved in AngII activation of the PVN of males but not intact females.
SFO neurons and the associated increase in SNA and BP is AT1 In studies in which the PVN of male rats was directly stim-
receptor-induced increases in cellular ROS [49,53]. Blockade of ulated with glutamate injections [64], oestrogen acting at ERβ
the formation of ROS inhibits activation of SFO neurons and the receptors in the PVN has been shown to inhibit the sympatho-
development of AngII-dependent hypertension [54,55]. In studies excitatory and hypertensive effects of glutamate activation in the
using brain slices of the SFO from male mice, AngII-induced pro- PVN. Furthermore, the ability of oestrogen to inhibit activation
duction of ROS in these neurons was blocked by pre-incubation of the PVN depends on NO production, suggesting that estradiol
of the tissue with estradiol, suggesting that oestrogen can inhibit inhibition of SNA at the level of the PVN requires oestrogen ac-
AngII-induced ROS production [56]. tivation of NO production [64]. The role of NO in AngII-induced
In addition to AngII receptors, ERs are known to be expressed increases in SNA and BP has been demonstrated to be import-
in the SFO and have been identified on the same neurons that ant in intact female mice but not in males [65]. In these studies,
express AT1 receptors [57]. Unlike the RVLM, the SFO predom- central blockade of NO production via cerebral infusion of N G -
inately expresses ERα receptors [44]. Activation of SFO ERs has nitro-L-arginine methyl ester (L-NAME) resulted in an increase
been shown to decrease the baseline SFO neuronal activity and in- in the AngII-induced increase in SNA and BP in intact females
hibit SFO neuron activation by AngII [58]. Recent studies tested but not in intact males. Furthermore, intact females have a higher
the hypothesis that it is the ERα receptors in the SFO that are re- resting level expression of neuronal NO synthase (nNOS) within


C 2016 Authors; published by Portland Press Limited 11
M. Hay

the PVN compared with males and OVX females. Together, these nervation of brown adipose and renal and lumbar SNA [26,74,75].
studies suggest that sex differences in AngII hypertension involve Leptin has been shown in men and male animal models to induce
an effect of oestrogen on the increased NOS and NO inhibition increases in SNA and result in an increase in BP. New and in-
of PVN activation, as well as oestrogen suppression of an AngII- triguing studies in an animal model [76] have shown, in female
induced increase in ROS in the PVN. animals, that the effects of leptin on renal, splanchnic and lumbar
As reviewed above, an important role for the ERα and ERβ SNA depends on the levels of estradiol. In this elegant set of stud-
receptors in specific regions of the brain known to be important in ies [26,74,75], SNA in intact and OVX female rats was measured
the regulation of SNA and hypertension has been established in in a number of different sympathetic areas, each important for the
animal models. What is less clear is why some of these brain re- regulation of BP. Leptin was infused directly into the lateral vent-
gions express primarily ERα and others ERβ receptors, and how ricles of the brain and the effects on SNA measured. Centrally
these specific ERs may selectively modulate neurotransmission administered leptin was able to increase only lumbar and renal
in these brain regions. Are these differences in ER expression in SNA when oestrogen was surging systemically (pro-oestrus in
the brain also seen in humans? A common cellular mechanism, the intact rats) or during exogenous estradiol infusion in the OVX
regardless of the site of the brain region expression, includes in- rats. These results offer an interesting and intriguing interaction
hibition of ROS formation and enhancement of nNOS expression between central leptin levels and ERs, and suggest that surges in
[63,64]. How we might exploit this understanding of selective ex- oestrogen may be required to unmask leptin’s sympathoexcitatory
pression of ERs in these key brain regions involved in hyperten- effects in females. However, there are some limitations to these
sion, to develop ER-selective drugs to treat hypertension, needs studies with regard to how they may inform our understanding
further study. of the role of leptin in hypertensive obese women. First, in these
studies in female rats, leptin was infused directly into the cerebral
ventricles at doses much higher than observed in premenopausal
OBESITY, OESTROGEN AND SYMPATHETIC women [10]. Second, it is known that both lean and obese pre-
OUTFLOW menopausal women have a lower SNA than age-matched men,
despite having higher levels of circulating leptin [10]. Thus, the
In western countries, the increases in population rates of altered role of leptin in the regulation of SNA in premenopausal women
metabolic conditions involved in obesity, hypertension, insulin may be less robust than that observed in animal models. Further-
resistance and dyslipidaemia have together been referred to as the more, how this relationship between leptin and SNA may change
metabolic syndrome [66]. It is estimated that approximately 25 % as women age and the effect of menopause on this relationship
of adults in the U.S.A. have been predicted as having the meta- are areas that needs further study.
bolic syndrome, which is associated with a significantly higher Study of exactly where in the brain this interaction between
rate of death due to related cardiovascular disease [67,68]. An un- leptin and SNA takes place and what types of ERs are involved is
derstanding of the underlying mechanisms associated with this only just starting. The expression of the leptin receptor, ObR, has
high rate of mortality from the metabolic syndrome is critical been identified throughout the mammalian brain, including sites
to improving the health of both women and men. The increase in the lateral hypothalamus [77], and key areas involved in the
in the incidence of hypertension in obese individuals has shown regulation of SNA, including the SFO [78] and the NTS [79]. In
good correlation with an increase in SNA and related end-organ studies where the ObR leptin receptor was knocked down in the
damage [8,69]. Specifically, this increase in obesity-related hy- SFO of male mice, the sympathoexcitatory effects of centrally
pertension correlates highly with levels of visceral adiposity but administered leptin were eliminated, suggesting that, in males,
not subcutaneous adiposity [70,71]. However, most of these data leptin’s actions to increase SNA need the SFO. But no such stud-
on the relationship between obesity and SNA have been obtained ies of the effects of leptin on the SFO have been performed in
from either obese men or male animal models. The study of the females. We do know that the SFO expresses predominantly ERα
role of sex and sex steroids, and how obesity and the metabolic receptors [44], and that activation of ERα receptors in the SFO
syndrome may affect SNA and hypertension in women, started has been shown to decrease SFO neuronal activity [58] and atten-
only recently [72]. uate AngII-induced increase in SNA and BP [59]. But, we also
In a study examining SNA, BP and body mass index (BMI) know that, in female animal models, the sympathoexcitatory ef-
in age-matched men and women, women with hypertension had fects of leptin are seen only when circulating oestrogen levels are
an increase in SNA compared with the non-hypertensive control high [76]. Thus, one might hypothesize that either leptin activa-
females, but this increase in SNA was not related to BMI [10]. tion of ObR in the SFO, which increases SFO-induced increases
This is in contrast to age-matched hypertensive men whose SNA in SNA, and activation of ERα receptors in the SFO, which in-
was the same as that in normotensive men, but directly related volves inhibition of SNA, involve separate pathways and separate
to their BMI. This study also measured plasma leptin levels and signalling mechanisms within the SFO, or the oestrogen-induced
observed that women in this study had higher circulating levels of facilitation of leptin sympathoexcitatory effects masks the sym-
leptin compared with the men. These results are consistent with pathoinhibitory effects of oestrogen alone. Such ObR and ERα
other reports that show females having higher levels of circulating receptor interactions at the level of the SFO are only speculative
leptin than males [73]. and most certainly require further experimentation and study.
Leptin is a hormone important to energy homoeostasis and A second possible site of brain interaction of leptin and oes-
involved in regulation of feeding behaviours, as well as SNA in- trogen to regulate SNA is the PVN. In recent studies in fe-

12 
C 2016 Authors; published by Portland Press Limited
Brain oestrogen receptors and high blood pressure risk factors

male rats, in which α-melanocyte-stimulating hormone (α-MSH) ing two different types of stress and c-fos measurement of PVN
receptors in the PVN were blocked using nanolitre injections of activation, it was found that, in OVX rats, oestrogen replacement
the selective MC3/4 receptor antagonist SHU9119, the sympath- increases plasma corticosteroid response to stress but inhibits
oexcitatory effects of central leptin in female rats in pro-oestrus ACTH-induced stress levels [93], and oestrogen replacement was
were eliminated [76]. These results suggest that, in addition to found to inhibit stress-induced activation of the PVN. In contrast,
the SFO, the PVN may be a key site of integration of leptin and studies using intact females and males found the former had an
ERs in the modulation of SNA and BP. enhanced ACTH response to stress and enhanced c-fos-measured
Although the molecular mechanisms involved in oestrogen’s PVN activation compared with males [94], but found no role for
modulation of leptin’s central sympathoexcitatory effects are not oestrogen in this response. Some of these differences in the results
yet known, a possible candidate would be a direct interaction from the animal studies, and how they correlate with data from
between the leptin ObR and ERs. ER modulation of ObR has women, have suggested the importance of the timing of meas-
been well described in other tissues, including bone [80], cancer urements in the intact animals and the methods of how and when
cells [81] and the hypothalamus [82], in which activation of ERα estradiol should be replaced in the OVX animals [96]. Oestrogen
receptors induces the expression of ObR via activation of the replacement therapy (ERT) in women uses low, chronic and re-
extracellular signal-related kinase (ERK)1/2 [mitogen-activated latively stable levels of oestrogen. In an attempt to mimic ERT
protein kinase (MAPK)1/MAPK3] signalling pathway. Future in women, an animal study using chronic, low-level oestrogen
studies are needed to determine if this molecular regulation of replacement in mature female rats found that oestrogen inhib-
ObR by ERα receptors is involved in the ability of leptin to its both noise stress- and inflammation stress-induced increases
increase SNA in females only when oestrogen levels are high. in ACTH [96]. Furthermore, this study also found that chronic
oestrogen replacement inhibits noise stress-induced increases in
STRESS, THE BRAIN AND HYPERTENSION the activation of both PVN neurons and selective catecholamine-
containing neurons in the ventrolateral medulla. These results
Both acute and chronic stress are major risk factors in the devel- suggest that chronic low-level oestrogen replacement in an an-
opment of cardiovascular disease, and increases in SNA and hy- imal model does inhibit stress-induced activation of the HPA axis
pertension [83,84]. Stress activates the hypothalamic–pituitary– and key neuronal regions involved in SNA increases.
adrenal (HPA) axis, and corticotrophin-releasing factor (CRF)- One cellular mechanism that may underlie oestrogen’s ability
containing neurons in the PVN stimulate adrenocorticotrophic to modulate CRF-containing PVN neurons is by altering neur-
hormone (ACTH) release from the anterior pituitary and an in- onal excitability. In electrophysiological experiments using rat
crease in glucocorticoid and catecholamine release from the ad- brain slices of the RVLM projecting PVN neurons, the effects of
renal gland [85,86]. Stress-induced increases in SNA are also short-term oestrogen replacement on PVN neurons were meas-
thought to involve CRF-containing neurons in the PVN that in- ured [97]. In these experiments, OVX rats were replaced with
nervate and activate neurons in the RVLM to increase SNA. oestrogen using daily, high-dose, subcutaneous injections of oes-
Furthermore, direct injections of CRF into the RVLM results in trogen for 4 days; then PVN brain slices were obtained and the
increases in lumbar SNA [86] and BP [87]. Together, the peri- potassium current measured. It was found that oestrogen can
pheral increase in catecholamine release from the adrenal gland decrease both the expression and the current density of the po-
and the increase in SNA result in an increase in BP and hyper- tassium IA current. It is interesting that an inhibition of IA current
tension. density would be expected to increase PVN excitability, which is
When compared with premenopausal women, postmeno- not what was seen when OVX females were replaced with con-
pausal women have been shown to have an increased HPA re- stant, low-dose, long-duration oestrogen replacement. It remains
sponse and BP response to mental stress [88,89]. Furthermore, to be tested whether, if oestrogen is replaced in an animal model
this increased HPA activation in postmenopausal women is blun- similar to that for postmenopausal women, the effects on PVN
ted by estradiol replacement [88,89]. In animal models sex dif- potassium currents would be different.
ferences in response to stress and ACTH release have been well In addition to the PVN, the RVLM neurons receiving CRF-
documented [90–93]. containing PVN terminals are thought to be important for stress
However, the effects of oestrogen on the HPA axis, the site of activation of SNA and hypertension. If stress increases the activity
action in the brain and oestrogen’s role in modulating the SNA of CRF-containing PVN neurons, then it would be reasonable to
response to stress are still controversial. A number of studies in hypothesize that this would, in turn, result in increases in CRF
animal models have shown that ovariectomy attenuates stress- activation and RVLM activation, and a subsequent increase in
induced increases in circulating corticosteroids and ACTH, and SNA. Studies on OVX rats using c-fos as a measure of cellular
that oestrogen replacement reverses these effects [91,92]. Other activation have shown that stress activation of catecholamine-
studies have shown no effect of oestrogen on measures of the containing ventrolateral cells is inhibited by long-term, low-dose,
HPA axis [94] and still others have shown, similar to the early constant oestrogen replacement [96]. However, these data were
studies in women, that oestrogen replacement inhibits stress- not linked to changes in the SNA or BP. Thus, there is some
induced increases in ACTH [93,95,96]. evidence to suggest that oestrogen may inhibit stress activation
Oestrogen’s site of action within the brain for modulating of RVLM neurons, although direct evidence that this mechanism
stress-induced activation of the HPA axis and SNA is still not is linked to stress-induced increase in SNA and BP has yet to be
clear. One possible site suggested is the PVN [93]. In studies us- established.


C 2016 Authors; published by Portland Press Limited 13
M. Hay

Figure 1 A hypothesis about how hypertensive risk factors such as obesity, stress and inflammation modulate common
central regions of the brain, such as the SFO, PVN and RVLM, causing increases in SNA and BP
It has been suggested that obesity, possibly through leptin-related actions, increases SNA via actions at the SFO and PVN.
In females, this sympathoexcitatory effect of leptin requires high levels of circulating oestrogen. It has been suggested
that stress, activating the HPA axis, increases SNA and BP via actions at the CRF-containing PVN neurons and RVLM.
Evidence suggests that oestrogen may attenuate stress-induced increases in SNA by reducing stress activation of the
PVN and RVLM. It has been suggested that increased inflammation and T-cell activation increase BP partially through an
increased SNA. The T-cell innervation into the SFO in males but not in females in AngII-induced hypertension has been
hypothesized as being involved in inflammation-related hypertension. Oestrogen, acting at specific receptors within the
SFO, PVN and RVLM, is hypothesized as inhibiting the ability of some of these hypertensive risk factors and their respective
mechanisms to increase SNA, and thus attenuate the development of hypertension.

SEX, INFLAMMATION AND HYPERTENSION that the activation of the immune system during AngII hyper-
tension depends on renal sympathetic innervation. Bilateral renal
One of the most recent advances in our understanding of under- denervation not only attenuated AngII hypertension in these mice
lying factors that influence the development and prevalence of but also reduced AngII-induced, T-cell infiltration of the kidney.
hypertension is the role of the adaptive immune system in the de- Unfortunately, this study used only male mice and did not offer
velopment of high BP. In both hypertensive humans and animal any insights into the role of renal SNA in inflammation-induced
models, T-cells have been shown to accumulate in the kidneys AngII hypertension in female mice.
and vasculature [16]. In initial studies in which male mice lacking Only recently has the study of the role of sex and sex hormones
lymphocytes due to a deficiency in the recombination-activating in the effect of the adaptive immune system on BP regulation
gene (Rag-1−/− ) were infused with AngII, the level of hyper- started [102–105]. Recent studies have found that the role of the
tension was significantly less than that observed in wild-type adaptive immune system in the development of AngII hyperten-
(WT) mice [98]. However, when T-cells from a male donor were sion depends on both the sex of the T-cell [104] and the sex of the
given back to the male Rag-1−/− mice, the hypertensive effects of animal receiving T-cell replacement [103]. In a study using intact
AngII were restored, suggesting that AngII-induced hypertension female Rag-1−/− mice, it was found that, after adoptive transfer
requires the adaptive immune system, and T-cells in particular. of WT male T-cells into the intact females, the females were pro-
This role of T-cells and the adaptive immune system has also been tected from AngII hypertension and showed significantly lower
seen in a number of other models of hypertension in male animals BP after AngII infusion compared with male Rag-1−/− mice that
[99,100]. The role of the sympathetic nervous system, and renal had also received WT male T-cell transfer [103].
sympathetic nerve innervation of the kidney in particular in this The sex of the T-cell is also known to be important in this
immune-mediated hypertension, has been shown in an elegant model of inflammation-induced AngII hypertension [104]. A re-
new study in male mice [101]. This study tested the hypothesis cent study has shown that, when male Rag-1−/− mice are given

14 
C 2016 Authors; published by Portland Press Limited
Brain oestrogen receptors and high blood pressure risk factors

T-cells from WT female donors, they do not develop the same they may share common mechanisms within the brain to increase
level of hypertension after AngII infusion as male Rag-1−/− mice sympathetic outflow and hypertension, it is hoped that new ideas
given T-cells from WT male donors [105]. These results suggest for testing the interactions may emerge.
that not only is the sex of the host important, but a critical determ-
inant in the development of inflammation-induced hypertension ACKNOWLEDGEMENTS
is also the sex of the T-cell [105]. The author would like to acknowledge the extraordinary contribu-
The role of the sympathetic nervous system in this protection tions of her long-time collaborators and colleagues Drs Baojian
in females against inflammation-induced AngII hypertension has Xue and Alan Kim Johnson from the University of Iowa, and Kathryn
not yet been determined. However, the above referenced stud- Sandberg from Georgetown University.
ies [103–105], also using male and female Rag-1−/− mice, did
observe a significant increase in T-cell infiltration into the SFO
FUNDING
in the male Rag-1−/− mice compared with the female Rag-1−/−
mice. These results suggest that T-cell infiltration into the SFO This work has been supported by the National Institutes of Health
may be important to inflammation-induced AngII hypertension. [HL098207] and the University of Arizona.
Additional studies are needed to investigate the role of ovarian
hormones in inflammation-induced hypertension and the effect
of ovarian hormones on T-cell infiltration into key end-organs REFERENCES
such as the kidney, vasculature and brain.
1 Kearney, P.M., Whelton, M., Reynolds, K., Muntner, P., Whelton,
P.K. and He, J. (2005) Global burden of hypertension: analysis
CONCLUSION of worldwide data. Lancet 365, 217–223
CrossRef PubMed
2 Cutler, J.A., Sorlie, P.D., Wolz, M., Thom, T., Fields, L.E. and
The present review links compelling evidence for the role of the Roccella, E.J. (2008) Trends in hypertension prevalence,
brain in the effects of sex and sex hormones on the regulation of awareness, treatment, and control rates in United States adults
between 1988–1994 and 1999–2004. Hypertension 52,
SNA and the development of hypertension. Specific regions of the
18–27. CrossRef
brain known to be required for the development of neurogenic 3 Yanes, L.L., Romero, D.G., Iliescu, R. and Reckelhoff, J.F.
hypertension, such as the SFO, PVN and RVLM, also express (2011) A single pill to treat postmenopausal hypertension? Not
ERs. Each of these key regions express unique populations of yet. Curr. Top. Med. Chem. 11, 1736–1741 CrossRef
4 Sandberg, K. and Ji, H. (2012) Sex differences in primary
ERs, either ERα or ERβ or both, and the activity of the neurons
hypertension. Biol. Sex Differ. 3, 7 CrossRef PubMed
in these regions can be affected by selective ER modulators. 5 Maranon, R. and Reckelhoff, J.F. (2013) Sex and gender
There is significant evidence from animal models to support the differences in control of blood pressure. Clin. Sci. (Lond.) 125,
conclusion that activation of these key brain ERs can modulate 311–318 CrossRef PubMed
6 Maric-Bilkan, C., Gilbert, E.L. and Ryan, M.J. (2014) Impact of
cell activation in these regions, and alter the responses of these ovarian function on cardiovascular health in women: focus on
neurons to hormones, peptides and neurotransmitters known to hypertension. Int. J. Women’s Health 6, 131–139 CrossRef
be involved in increases in SNA and BP. 7 Esler, M.D., Eikelis, N., Lambert, E. and Straznicky, N. (2008)
The present review also, for the first time, endeavours to link Neural mechanisms and management of obesity-related
hypertension. Curr. Cardiol. Rep. 10, 456–463
some common mechanisms underlying known hypertensive risk CrossRef PubMed
factors of obesity, stress and inflammation. Figure 1 illustrates 8 Lambert, E.A., Straznicky, N.E., Dixon, J.B. and Lambert, G.W.
how the risk factors of obesity, stress and inflammation all have (2015) Should the sympathetic nervous system be a target to
improve cardiometabolic risk in obesity? Am J Physiol Heart
verified links to the SFO, PVN or RVLM to increase SNA, and
Circ Physiol. 309, H244–H258 CrossRef PubMed
result in hypertension. In each of these brain regions there is 9 Lambert, E.A., Rice, T., Eikelis, N., Straznicky, N.E., Lambert,
compelling evidence to support oestrogen acting on selective re- G.W., Head, G.A., Hensman, C., Schlaich, M.P. and Dixon, J.B.
ceptors in these key regions to modulate increases in SNA caused (2014) Sympathetic activity and markers of cardiovascular risk
in nondiabetic severely obese patients: the effect of the initial
by the risk factors, and thus decrease the probability of hyper- 10 % weight loss. Am. J. Hypertens. 27, 1308–1315
tension. In some cases such as obesity, it is hypothesized that CrossRef PubMed
circulating oestrogen may facilitate leptin-induced increases in 10 Lambert, E., Straznicky, N., Eikelis, N., Esler, M., Dawood, T.,
SNA and hypertension. The sites in the brain involved in this Masuo, K., Schlaich, M. and Lambert, G. (2007) Gender
differences in sympathetic nervous activity: influence of body
interaction between leptin and oestrogen have not been proven mass and blood pressure. J. Hypertens. 25, 1411–1419
but may include both the PVN and the SFO. In other cases, such CrossRef PubMed
as stress-induced increases in SNA, oestrogen is hypothesized to 11 Kim, J.M., Kim, T.H., Lee, H.H., Lee, S.H. and Wang, T. (2014)
act on the PVN and RVLM to reduce stress-induced activation Postmenopausal hypertension and sodium sensitivity. J.
Menopausal Med. 20, 1–6 CrossRef PubMed
of these regions, and thus attenuate stress-induced increases in 12 He, F.J., Li, J. and Macgregor, G.A. (2013) Effect of longer term
SNA and hypertension. All these hypotheses need further rigor- modest salt reduction on blood pressure: Cochrane systematic
ous testing, but offer intriguing mechanisms by which oestrogen review and meta-analysis of randomised trials. BMJ 346, f1325
CrossRef PubMed
acting in the brain can modulate the development of hyperten-
13 Lima, R., Wofford, M. and Reckelhoff, J.F. (2012) Hypertension
sion. By reviewing the role of the brain and ovarian hormones in postmenopausal women. Curr. Hypertens. Rep. 14,
in each of these seemingly independent risk factors, and how 254–260 CrossRef PubMed


C 2016 Authors; published by Portland Press Limited 15
M. Hay

14 Lambert, E.A. and Lambert, G.W. (2011) Stress and its role in 32 Harvey, R.E., Hart, E.C., Charkoudian, N., Curry, T.B., Carter,
sympathetic nervous system activation in hypertension and the J.R., Fu, Q., Minson, C.T., Joyner, M.J. and Barnes, J.N. (2015)
metabolic syndrome. Curr. Hyperten. Rep. 13, 244–248 Oral contraceptive use, muscle sympathetic nerve activity, and
CrossRef systemic hemodynamics in young women. Hypertension 66,
15 Esler, M., Eikelis, N., Schlaich, M., Lambert, G., Alvarenga, M., 590–597 CrossRef PubMed
Dawood, T., Kaye, D., Barton, D., Pier, C and Guo, L. (2008) 33 Anderson, G.L., Limacher, M., Assaf, A.R., Bassford, T.,
Chronic mental stress is a cause of essential hypertension:
Beresford, S.A., Black, H., Bonds, D., Brunner, R., Brzyski, R.
presence of biological markers of stress. Clin. Exp. Pharmacol.
and Caan, B. (2004) Effects of conjugated equine estrogen in
Physiol. 35, 498–502 CrossRef PubMed
16 Harrison, D.G., Guzik, T.J., Lob, H.E., Madhur, M.S., Marvar, P.J., postmenopausal women with hysterectomy: the Women’s
Thabet, S.R., Vinh, A. and Weyand, C.M. (2011) Inflammation, Health Initiative randomized controlled trial. JAMA 291,
immunity, and hypertension. Hypertension 57, 132–140 1701–1712 CrossRef PubMed
CrossRef PubMed 34 Fung, M.M., Poddar, S., Bettencourt, R., Jassal, S.K. and
17 Xue, B., Johnson, A.K. and Hay, M. (2013) Sex differences in Barrett-Connor, E. (2011) A cross-sectional and 10-year
angiotensin II- and aldosterone-induced hypertension: the prospective study of postmenopausal estrogen therapy and
central protective effects of estrogen. Am. J. Physiol. Regul. blood pressure, renal function, and albuminuria: the Rancho
Integr. Comp. Physiol. 305, R459–R463 Bernardo Study. Menopause 18, 629–637 CrossRef PubMed
CrossRef PubMed 35 Weitz, G., Elam, M., Born, J., Fehm, H.L. and Dodt, C. (2001)
18 Seals, D.R. (1989) Sympathetic neural discharge and vascular Postmenopausal estrogen administration suppresses muscle
resistance during exercise in humans. J. Appl. Physiol. 66, sympathetic nerve activity. J. Clin. Endocrinol. Metab. 86,
2472–2478 PubMed
344–348 PubMed
19 Charkoudian, N., Joyner, M.J., Johnson, C.P., Eisenach, J.H.,
36 Xue, B., Badaue-Passos, D., Guo, F., Gomez-Sanchez, C.E., Hay,
Dietz, N.M. and Wallin, B.G. (2005) Balance between cardiac
output and sympathetic nerve activity in resting humans: role in M. and Johnson, A.K. (2009) Sex differences and central
arterial pressure regulation. J. Physiol. 568 Pt 1, 315–321 protective effect of 17beta-estradiol in the development of
CrossRef PubMed aldosterone/NaCl-induced hypertension. Am. J. Physiol. Heart
20 Hart, E.C., Charkoudian, N., Wallin, B.G., Curry, T.B., Eisenach, Circ. Physiol. 296, H1577–H1585 CrossRef PubMed
J.H. and Joyner, M.J. (2009) Sex differences in sympathetic 37 Xue, B., Pamidimukkala, J. and Hay, M. (2005) Sex differences
neural-hemodynamic balance: implications for human blood in the development of angiotensin II-induced hypertension in
pressure regulation. Hypertension 53, 571–576 conscious mice. Am. J. Physiol. Heart Circ. Physiol. 288,
CrossRef PubMed H2177–H2184 CrossRef PubMed
21 Hart, E.C. and Charkoudian, N. (2014) Sympathetic neural 38 Xue, B., Pamidimukkala, J., Lubahn, D.B. and Hay, M. (2007)
regulation of blood pressure: influences of sex and aging. Estrogen receptor-alpha mediates estrogen protection from
Physiology 29, 8–15 CrossRef PubMed angiotensin II-induced hypertension in conscious female mice.
22 Joyner, M.J., Wallin, B.G. and Charkoudian, N. (2015) Sex
Am. J. Physiol. Heart Circ. Physiol. 292, H1770–H1776
differences and blood pressure regulation in humans. Exp.
CrossRef PubMed
Physiol. doi: 10.1113/EP085146
39 Macefield, V.G. and Henderson, L.A. (2010) Real-time imaging
23 Hart, E.C. and Charkoudian, N. (2011) Sympathetic neural
mechanisms in human blood pressure regulation. Curr. of the medullary circuitry involved in the generation of
Hypertens. Rep. 13, 237–243 CrossRef PubMed spontaneous muscle sympathetic nerve activity in awake
24 Hart, E.C., Joyner, M.J., Wallin, B.G. and Charkoudian, N. subjects. Hum. Brain Mapp. 31, 539–549 PubMed
(2012) Sex, ageing and resting blood pressure: gaining insights 40 Henderson, L.A., Richard, C.A., Macey, P.M., Runquist, M.L., Yu,
from the integrated balance of neural and haemodynamic P.L., Galons, J.P. and Harper, R.M. (1985) Functional magnetic
factors. J. Physiol. 590 Pt 9, 2069–2079 CrossRef PubMed resonance signal changes in neural structures to baroreceptor
25 Narkiewicz, K., Phillips, B.G., Kato, M., Hering, D., reflex activation. J. Appl. Physiol. 96, 693–703 CrossRef
Bieniaszewski, L. and Somers, V.K. (2005) Gender-selective 41 Lundblad, L.C., Fatouleh, R.H., McKenzie, D.K., Macefield, V.G.
interaction between aging, blood pressure, and sympathetic and Henderson, L.A. (2015) Brain stem activity changes
nerve activity. Hypertension 45, 522–525 CrossRef PubMed associated with restored sympathetic drive following CPAP
26 Credeur, D.P., Holwerda, S.W., Boyle, L.J., Vianna, L.C., Jensen, treatment in OSA subjects: a longitudinal investigation. J.
A.K. and Fadel, P.J. (2014) Effect of aging on carotid baroreflex
Neurophysiol. 114, 893–901 CrossRef PubMed
control of blood pressure and leg vascular conductance in
42 Campos, R.R., Oliveira-Sales, E.B., Nishi, E.E., Paton, J.F. and
women. Am. J. Physiol. Heart Circ. Physiol. 306, H1417–H425
CrossRef PubMed Bergamaschi, C.T. (2015) Mechanisms of renal sympathetic
27 Brito, M.B., Nobre, F. and Vieira, C.S. (2011) Hormonal activation in renovascular hypertension. Exp. Physiol. 100,
contraception and cardiovascular system. Arq. Bras. Cardiol. 496–501 CrossRef PubMed
96, e81–e89 CrossRef PubMed 43 Barman, S.M. and Gebber, G.L. (2007) Role of ventrolateral
28 Maguire, K. and Westhoff, C. (2011) The state of hormonal medulla in generating the 10-Hz rhythm in sympathetic nerve
contraception today: established and emerging discharge. Am. J. Physiol. Regul. Integr. Comp. Physiol. 293,
noncontraceptive health benefits. Am. J. Obstet. Gynecol. 205 R223–R233 CrossRef PubMed
4 Suppl, S4–S8 CrossRef PubMed 44 Spary, E.J., Maqbool, A. and Batten, T.F. (2009) Oestrogen
29 Akkad, A.A., Halligan, A.W., Abrams, K. and al-Azzawi, F. (1997) receptors in the central nervous system and evidence for their
Differing responses in blood pressure over 24 hours in role in the control of cardiovascular function. J. Chem.
normotensive women receiving oral or transdermal estrogen Neuroanat. 38, 185–196 CrossRef PubMed
replacement therapy. Obstet. Gynecol. 89, 97–103
45 Saleh, M.C., Connell, B.J. and Saleh, T.M. (2000) Autonomic
CrossRef PubMed
and cardiovascular reflex responses to central estrogen
30 Vongpatanasin, W., Tuncel, M., Mansour, Y., Arbique, D. and
Victor, R.G. (2001) Transdermal estrogen replacement therapy injection in ovariectomized female rats. Brain Res 879,
decreases sympathetic activity in postmenopausal women. 105–114 CrossRef PubMed
Circulation 103, 2903–2908 CrossRef PubMed 46 Shih, C.D. (2009) Activation of estrogen receptor
31 Cannoletta, M. and Cagnacci, A. (2014) Modification of blood beta-dependent nitric oxide signaling mediates the hypotensive
pressure in postmenopausal women: role of hormone effects of estrogen in the rostral ventrolateral medulla of
replacement therapy. Int. J. Women’s Health 6, 745–757 anesthetized rats. J. Biomed. Sci. 16, 60 CrossRef PubMed

16 
C 2016 Authors; published by Portland Press Limited
Brain oestrogen receptors and high blood pressure risk factors

47 Xue, B., Zhang, Z., Beltz, T.G., Johnson, R.F., Guo, F., Hay, M. 62 Xue, B., Singh, M., Guo, F., Hay, M. and Johnson, A.K. (2009)
and Johnson, A.K. (2013) Estrogen receptor-β in the Protective actions of estrogen on angiotensin II-induced
paraventricular nucleus and rostroventrolateral medulla plays hypertension: role of central nitric oxide. Am. J. Physiol. Heart
an essential protective role in aldosterone/salt-induced Circ. Physiol. 297, H1638–H1646 CrossRef PubMed
hypertension in female rats. Hypertension 61, 1255–1262 63 Marques-Lopes, J., Lynch, M.K., Van Kempen, T.A., Waters,
CrossRef PubMed E.M., Wang, G., Iadecola, C. et al. (2015) Female protection
48 Pierce, J.P., Kievits, J., Graustein, B., Speth, R.C., Iadecola, C. from slow-pressor effects of angiotensin II involves prevention
and Milner, T.A. (2009) Sex differences in the subcellular of ROS production independent of NMDA receptor trafficking in
distribution of angiotensin type 1 receptors and NADPH oxidase hypothalamic neurons expressing angiotensin 1A receptors.
subunits in the dendrites of C1 neurons in the rat rostral Synapse 69, 148–165 CrossRef PubMed
ventrolateral medulla. Neuroscience 163, 329–338
64 Gingerich, S. and Krukoff, T.L. (2006) Estrogen in the
CrossRef PubMed
paraventricular nucleus attenuates L-glutamate-induced
49 Zimmerman, M.C., Lazartigues, E., Lang, J.A., Sinnayah, P.,
increases in mean arterial pressure through estrogen receptor
Ahmad, I.M., Spitz, D.R. and Davisson, R.L. (2002) Superoxide
beta and NO. Hypertension 48, 1130–1136 CrossRef PubMed
mediates the actions of angiotensin II in the central nervous
system. Circ. Res. 91, 1038–1045 CrossRef PubMed 65 Xue, B., Singh, M., Guo, F., Hay, M. and Johnson, A.K. (2009)
50 Van Kempen, T.A., Dodos, M., Woods, C., Marques-Lopes, J., Protective actions of estrogen on angiotensin II-induced
Justice, N.J., Iadecola, C., Pickel, V.M., Glass, M.J. and Milner, hypertension: role of central nitric oxide. Am. J. Physiol. Heart
T.A. (2015) Sex differences in NMDA GluN1 plasticity in rostral Circ. Physiol. 297, H1638–H1646 CrossRef PubMed
ventrolateral medulla neurons containing corticotropin-releasing 66 Alberti, K.G., Zimmet, P. and Shaw, J. (2006) Metabolic
factor type 1 receptor following slow-pressor angiotensin II syndrome – a new world-wide definition. A Consensus
hypertension. Neuroscience 307, 83–97 CrossRef PubMed Statement from the International Diabetes Federation. Diabet.
51 Ferguson, A.V. (2009) Angiotensinergic regulation of autonomic Med. 23, 469–480 CrossRef PubMed
and neuroendocrine outputs: critical roles for the subfornical 67 Malik, S., Wong, N.D., Franklin, S.S., Kamath, T.V., L’Italien,
organ and paraventricular nucleus. Neuroendocrinology 89, G.J., Pio, J.R. and Williams, G.R. (2004) Impact of the
370–376 CrossRef PubMed metabolic syndrome on mortality from coronary heart disease,
52 Ciriello, J. (1997) Afferent renal inputs onto subfornical organ cardiovascular disease, and all causes in United States adults.
neurons responsive to angiotensin II. Am. J. Physiol. 272 5 Pt Circulation 110, 1245–1250 CrossRef PubMed
2, R1684–R1689 PubMed 68 Ford, E.S. (2005) Prevalence of the metabolic syndrome
53 Lob, H.E., Schultz, D., Marvar, P.J., Davisson, R.L. and Harrison, defined by the International Diabetes Federation among adults
D.G. (2012) Role of the NADPH oxidases in the subfornical in the U.S. Diabetes Care 28, 2745–2749 CrossRef PubMed
organ in angiotensin II-induced hypertension. Hypertension 61, 69 Esler, M., Straznicky, N., Eikelis, N., Masuo, K., Lambert, G.
382–37 CrossRef PubMed and Lambert, E. (2006) Mechanisms of sympathetic activation
54 Li, S., Case, A.J., Yang, R.F., Schultz, H.D. and Zimmerman,
in obesity-related hypertension. Hypertension 48, 787–796
M.C. (2013) Over-expressed copper/zinc superoxide dismutase
CrossRef PubMed
localizes to mitochondria in neurons inhibiting the angiotensin
70 Alvarez, G.E., Ballard, T.P., Beske, S.D. and Davy, K.P. (2004)
II-mediated increase in mitochondrial superoxide. Redox Biol 2,
Subcutaneous obesity is not associated with sympathetic
8–14 CrossRef PubMed
55 Wang, G., Sarkar, P., Peterson, J.R., Anrather, J., Pierce, J.P., neural activation. Am. J. Physiol. Heart Circ. Physiol. 287,
Moore, J.M., Feng, J., Zhou, P., Milner, T.A. and Pickel, V.M. H414–H418 CrossRef PubMed
(2013) COX-1-derived PGE2 and PGE2 type 1 receptors are vital 71 Alvarez, G.E., Beske, S.D., Ballard, T.P. and Davy, K.P. (2002)
for angiotensin-II-induced formation of reactive oxygen species Sympathetic neural activation in visceral obesity. Circulation
and Ca2+ influx in the subfornical organ. Am. J. Physiol. Heart 106, 2533–2536 CrossRef PubMed
Circ. Physiol. 305, H1451–H461 72 Brooks, V.L., Shi, Z., Holwerda, S.W. and Fadel, P.J. (2015)
56 Xue, B., Zhao, Y., Johnson, A.K. and Hay, M. (2008) Central Obesity-induced increases in sympathetic nerve activity: sex
estrogen inhibition of angiotensin II-induced hypertension in matters. Auton. Neurosci. 187, 18–26 CrossRef PubMed
male mice and the role of reactive oxygen species. Am. J. 73 Karastergiou, K., Smith, S.R., Greenberg, A.S. and Fried, S.K.
Physiol. Heart Circ. Physiol. 295, H1025–H1032 (2012) Sex differences in human adipose tissues – the biology
CrossRef PubMed of pear shape. Biol. Sex Differ. 3, 13 CrossRef PubMed
57 Rosas-Arellano, M.P., Solano-Flores, L.P. and Ciriello, J. (1999) 74 Friedman, J.M. and Mantzoros, C.S. (2015) 20 years of leptin:
Co-localization of estrogen and angiotensin receptors within from the discovery of the leptin gene to leptin in our therapeutic
subfornical organ neurons. Brain Res. 837, 254–262 armamentarium. Metabolism 64, 1–4 CrossRef PubMed
CrossRef PubMed 75 Haynes, W.G. (2005) Role of leptin in obesity-related
58 Ciriello, J. and Roder, S. (2013) 17β-Estradiol alters the hypertension. Exp. Physiol. 90, 683–688 CrossRef PubMed
response of subfornical organ neurons that project to 76 Shi, Z. and Brooks, V.L. (2015) Leptin differentially increases
supraoptic nucleus to plasma angiotensin II and hypernatremia. sympathetic nerve activity and its baroreflex regulation in
Brain Res. 1526, 54–64 CrossRef PubMed
female rats: role of oestrogen. J. Physiol. 593, 1633–1647
59 Xue, B., Zhang, Z., Beltz, T.G., Guo, F., Hay, M. and Johnson,
CrossRef PubMed
A.K. (2015) Genetic knockdown of estrogen receptor-alpha in
77 Wauman, J. and Tavernier, J. (2011) Leptin receptor signaling:
the subfornical organ augments ANG II-induced hypertension in
pathways to leptin resistance. Front. Biosci. 16, 2771–2793
female mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308,
R507–R516 CrossRef PubMed CrossRef
60 Chung, W.C., Pak, T.R., Suzuki, S., Pouliot, W.A., Andersen, 78 Young, C.N., Morgan, D.A., Butler, S.D., Mark, A.L. and
M.E. and Handa, R.J. (2007) Detection and localization of an Davisson, R.L. (2013) The brain subfornical organ mediates
estrogen receptor beta splice variant protein (ERbeta2) in the leptin-induced increases in renal sympathetic activity but not its
adult female rat forebrain and midbrain regions. J. Comp. metabolic effects. Hypertension 61, 737–744
Neurol. 505, 249–267 CrossRef PubMed CrossRef PubMed
61 Bingham, B., Williamson, M. and Viau, V. (2006) Androgen and 79 Mark, A.L., Agassandian, K., Morgan, D.A., Liu, X., Cassell,
estrogen receptor-beta distribution within spinal-projecting and M.D. and Rahmouni, K. (2009) Leptin signaling in the nucleus
neurosecretory neurons in the paraventricular nucleus of the tractus solitarii increases sympathetic nerve activity to the
male rat. J. Comp. Neurol. 499, 911–923 CrossRef PubMed kidney. Hypertension 53, 375–380 CrossRef PubMed


C 2016 Authors; published by Portland Press Limited 17
M. Hay

80 Wang, S.J., Li, X.F., Jiang, L.S. and Dai, L.Y. (2012) Estrogen 93 Figueiredo, H.F., Ulrich-Lai, Y.M., Choi, D.C. and Herman, J.P.
stimulates leptin receptor expression in ATDC5 cells via the (2007) Estrogen potentiates adrenocortical responses to
estrogen receptor and extracellular signal-regulated kinase stress in female rats. Am. J. Physiol. Endocrinol. Metab. 292,
pathways. J. Endocrinol. 213, 163–172 E1173–E1182 CrossRef PubMed
CrossRef PubMed 94 Babb, J.A., Masini, C.V., Day, H.E. and Campeau, S. (2013) Sex
81 Strong, A.L., Ohlstein, J.F., Biagas, B.A., Rhodes, L.V., Pei, D.T., differences in activated corticotropin-releasing factor neurons
Tucker, H.A., Llamas, C., Bowles, A.C., Dutreil, M.F. and Zhang, within stress-related neurocircuitry and
S. (2015) Leptin produced by obese adipose stromal/stem hypothalamic–pituitary–adrenocortical axis hormones following
cells enhances proliferation and metastasis of estrogen restraint in rats. Neuroscience 234, 40–52
receptor positive breast cancers. Breast Cancer Res. 17, 112 CrossRef PubMed
CrossRef PubMed 95 Ochedalski, T., Subburaju, S., Wynn, P.C. and Aguilera, G.
82 Meli, R., Pacilio, M., Raso, G.M., Esposito, E., Coppola, A., (2007) Interaction between oestrogen and oxytocin on
Nasti, A., Di Carlo, C., Nappi, C. and Di Carlo, R. (2004) hypothalamic–pituitary–adrenal axis activity. J. Neuroendocrinol.
Estrogen and raloxifene modulate leptin and its receptor in 19, 189–197 CrossRef PubMed
hypothalamus and adipose tissue from ovariectomized rats. 96 Dayas, C.V., Xu, Y., Buller, K.M. and Day, T.A. (2000) Effects of
Endocrinology 145, 3115–3121 CrossRef PubMed chronic oestrogen replacement on stress-induced activation of
83 Hering, D., Lachowska, K. and Schlaich, M. (2015) Role of the hypothalamic–pituitary–adrenal axis control pathways. J.
sympathetic nervous system in stress-mediated cardiovascular Neuroendocrinol. 12, 784–794 CrossRef PubMed
disease. Curr. Hypertens. Rep. 17, 594 97 Lee, S.K., Ryu, P.D. and Lee, S.Y. (2013) Estrogen replacement
84 Schiöler, L., Söderberg, M., Rosengren, A., Järvholm, B. and modulates voltage-gated potassium channels in rat
Torén, K. (2015) Psychosocial work environment and risk of presympathetic paraventricular nucleus neurons. BMC Neurosci
ischemic stroke and coronary heart disease: a prospective 14, 134 CrossRef PubMed
longitudinal study of 75 236 construction workers. Scand. J. 98 Guzik, T.J., Hoch, N.E., Brown, K.A., McCann, L.A., Rahman, A.,
Work Environ. Health 41, 280–287 CrossRef PubMed Dikalov, S., Goronzy, J., Weyand, C. and Harrison, D.G. (2007)
85 Aguilera, G. (1994) Regulation of pituitary ACTH secretion Role of the T cell in the genesis of angiotensin II induced
during chronic stress. Front. Neuroendocrinol. 15, 321–350 hypertension and vascular dysfunction. J. Exp. Med. 204,
CrossRef PubMed 2449–2460 CrossRef PubMed
86 Bardgett, M.E., Sharpe, A.L. and Toney, G.M. (2014) Activation 99 Mattson, D.L., Lund, H., Guo, C., Rudemiller, N., Geurts, A.M.
of corticotropin-releasing factor receptors in the rostral and Jacob, H. (2013) Genetic mutation of recombination
ventrolateral medulla is required for glucose-induced activating gene 1 in Dahl salt-sensitive rats attenuates
sympathoexcitation. Am. J. Physiol. Endocrinol. Metab. 307, hypertension and renal damage. Am. J. Physiol. Regul. Integr.
E944–E953 CrossRef PubMed Comp. Physiol. 304, R407–R414 CrossRef PubMed
87 Milner, T.A., Reis, D.J., Pickel, V.M., Aicher, S.A. and Giuliano, R. 100 De Miguel, C., Guo, C., Lund, H., Feng, D. and Mattson, D.L.
(1993) Ultrastructural localization and afferent sources of (2011) Infiltrating T lymphocytes in the kidney increase
corticotropin-releasing factor in the rat rostral ventrolateral oxidative stress and participate in the development of
medulla: implications for central cardiovascular regulation. J. hypertension and renal disease. Am. J. Physiol. Renal Physiol.
Comp. Neurol. 333, 151–167 CrossRef PubMed 300, F734–F742 CrossRef PubMed
88 Lindheim, S.R., Legro, R.S., Bernstein, L., Stanczyk, F.Z., Vijod, 101 Xiao, L., Kirabo, A., Wu, J., Saleh, M.A., Zhu, L., Wang, F.,
M.A., Presser, S.C. and Lobo, R.A. (1992) Behavioral stress Takahashi, T., Loperena, R., Foss, J.D. and Mernaugh, R.L.
responses in premenopausal and postmenopausal women and (2015) Renal denervation prevents immune cell activation and
the effects of estrogen. Am. J. Obstet. Gynecol. 167, renal inflammation in angiotensin II-induced hypertension. Circ.
1831–1836 CrossRef PubMed Res. 117, 547–557 CrossRef PubMed
89 Lindheim, S.R., Legro, R.S., Morris, R.S., Wong, I.L., Tran, D.Q., 102 Tipton, A.J., Baban, B. and Sullivan, J.C. (2012) Female
Vijod, M.A., Stanczyk, F.Z. and Lobo, R.A. (1994) The effect of spontaneously hypertensive rats have greater renal
progestins on behavioral stress responses in postmenopausal anti-inflammatory T lymphocyte infiltration than males. Am. J.
women. J. Soc. Gynecol. Investig. 1, 79–83 PubMed Physiol. Regul. Integr. Comp. Physiol. 303, R359–R367
90 Veldhuis, J.D., Sharma, A. and Roelfsema, F. (2013) 103 Pollow, D.P., Uhrlaub, J., Romero-Aleshire, M.J., Sandberg, K.,
Age-dependent and gender-dependent regulation of Nikolich-Zugich, J., Brooks, H.L. and Hay, M. (2014) Sex
hypothalamic–adrenocorticotropic–adrenal axis. Endocrinol. differences in T-lymphocyte tissue infiltration and development
Metab. Clin. North Am. 42, 201–225 CrossRef PubMed of angiotensin II hypertension. Hypertension 64, 384–390
91 Handa, R.J. and Weiser, M.J. (2014) Gonadal steroid hormones CrossRef PubMed
and the hypothalamo–pituitary–adrenal axis. Front. 104 Ji, H., Zheng, W., Li, X., Liu, J., Wu, X., Zhang, M.A., Umans,
Neuroendocrinol. 35, 197–220 CrossRef PubMed J.G., Hay, M., Speth, R.C., Dunn, S.E. et al. (2014) Sex-specific
92 Weiser, M.J. and Handa, R.J. (2009) Estrogen impairs T-cell regulation of angiotensin II-dependent hypertension.
glucocorticoid dependent negative feedback on the Hypertension 64, 573–582 CrossRef PubMed
hypothalamic–pituitary–adrenal axis via estrogen receptor alpha 105 Sandberg, K., Ji, H. and Hay, M. (2015) Sex-specific immune
within the hypothalamus. Neuroscience 159, 883–895 modulation of primary hypertension. Cell Immunol. 294,
CrossRef PubMed 95–101 CrossRef PubMed

Received 15 September 2015/6 October 2015; accepted 21 October 2015


Version of Record published 30 November 2015, doi: 10.1042/CS20150654

18 
C 2016 Authors; published by Portland Press Limited
Copyright of Clinical Science is the property of Portland Press Ltd. and its content may not be
copied or emailed to multiple sites or posted to a listserv without the copyright holder's
express written permission. However, users may print, download, or email articles for
individual use.

Вам также может понравиться