Você está na página 1de 8

Am J Physiol Regul Integr Comp Physiol 296: R1124–R1131, 2009.

First published January 21, 2009; doi:10.1152/ajpregu.90947.2008.

Acupuncture and exercise restore adipose tissue expression


of sympathetic markers and improve ovarian morphology in rats
with dihydrotestosterone-induced PCOS
Louise Mannerås,1 Stefan Cajander,2 Malin Lönn,3 and Elisabet Stener-Victorin1
1
Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg,
Gothenburg, Sweden; 2 Department of Pathology and Cytology, Sunderby County Hospital, Luleå, Sweden; and 3 Institute
of Medicine, Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
Submitted 22 November 2008; accepted in final form 15 January 2009

Mannerås L, Cajander S, Lönn M, Stener-Victorin E. Acu- thermore, increased ovarian sympathetic nerve activity stimu-
puncture and exercise restore adipose tissue expression of sympa- lates androgen secretion in rats (19, 47), and PCOS is associ-
thetic markers and improve ovarian morphology in rats with ated with an increase in ovarian catecholaminergic nerve fibers
dihydrotestosterone-induced PCOS. Am J Physiol Regul Integr (20, 48) and altered catecholamine metabolism (15, 49), sug-
Comp Physiol 296: R1124 –R1131, 2009. First published January
gesting increased sympathetic nervous system activity. In-
21, 2009; doi:10.1152/ajpregu.90947.2008.—Altered activity of the
sympathetic nervous system, which innervates adipose and ovarian creased sympathetic nerve activity in the ovaries might con-
tissue, may play a role in polycystic ovary syndrome (PCOS). We tribute to PCOS by stimulating androgen secretion (19, 47).
hypothesize that electro-acupuncture (EA) and physical exercise re- Ovarian surgeries reduce the amount of androgen-producing
duce sympathetic activity by stimulating ergoreceptors and somatic theca cells and the regulation of the hypothalamic-pituitary-
afferent pathways in muscles. Here we investigated the effects of ovarian axis is changed, which increases ovulatory response in
low-frequency EA and physical exercise on mRNA expression of women with PCOS (21). Additionally, ovarian surgeries may
sympathetic markers in adipose tissue and on ovarian morphology in improve ovulatory dysfunction via disruption of ovarian sym-
female rats that received dihydrotestosterone (DHT) continuously, pathetic innervation.
starting before puberty, to induce PCOS. At age 11 wk, rats with Recently, we performed direct intra-neural recordings of
DHT-induced PCOS were randomly divided into three groups: PCOS, sympathetic nerve activity in PCOS patients (58). This
PCOS plus EA, and PCOS plus exercise. The latter two groups
received 2-Hz EA (evoking muscle twitches) three times/week or had
novel study showed that PCOS is associated with increased
free access to a running wheel for 4 –5 wk. In mesenteric adipose sympathetic nervous system activity, which correlated with
tissue, expression of ␤3-adrenergic receptor (ADRB3), nerve growth the elevated testosterone levels that characterize this syn-
factor (NGF), and neuropeptide Y (NPY) mRNA was higher in drome. Increased sympathetic outflow might explain the
untreated PCOS rats than in controls. Low-frequency EA and exercise increased prevalence of vascular disease in women with
downregulated mRNA expression of NGF and NPY, and EA also PCOS and might contribute to its etiology.
downregulated expression of ADRB3, compared with untreated rats Obesity and metabolic disturbances exacerbate many of the
with DHT-induced PCOS. EA and exercise improved ovarian mor- typical symptoms in women with PCOS and increase the risk
phology, as reflected in a higher proportion of healthy antral follicles of long-term health consequences. Adipose tissue participates
and a thinner theca interna cell layer than in untreated PCOS rats. in the integrative physiology of whole body glucose and fat
These findings support the theory that increased sympathetic activity metabolism and is innervated by the autonomic nervous sys-
contributes to the development and maintenance of PCOS and that the
effects of EA and exercise may be mediated by modulation of
tem, mainly the sympathetic nervous system, which modulates
sympathetic outflow to the adipose tissue and ovaries. its metabolic and endocrine functions (6). Alterations in sym-
pathetic activity in adipose tissue can affect the metabolic and
sympathetic activity; ␤3-adrenergic receptor; androgen receptor; endocrine functions of other tissues by altering the secretion of
nerve growth factor; neuropeptide Y fatty acids and adipokines. Indeed, in obese women with
metabolic syndrome, circulatory concentrations and gene ex-
THE AUTONOMIC NERVOUS SYSTEM has been suggested to contrib-
pression of nerve growth factor (NGF), a marker of sympa-
ute to polycystic ovary syndrome (PCOS) (15, 19, 20, 61). thetic activity, are increased in subcutaneous adipose tissue (7).
Features of PCOS and the related metabolic syndromes, such However, the expression of genes encoding sympathetic mark-
as hyperandrogenemia, hyperinsulinemia, insulin resistance, ers and the influence of therapy on those markers has not been
and abdominal obesity, are associated with disturbed activity studied in PCOS.
of the sympathetic nervous system (13, 33). Women with Since PCOS appears to be associated with increased sym-
PCOS have increased sympathetic and decreased parasympa- pathetic nerve activity, interventions thought to modulate sym-
thetic components of heart rate variability, an indirect measure pathetic nerve activity, such as physical exercise (43) and
of cardiac autonomic control (61), and abnormal heart rate low-frequency electro-acupuncture (EA) (39, 51), might be
recovery, another measure of autonomic function (17). Fur- beneficial. In uncontrolled studies of women with well-defined
PCOS and women with undefined ovulatory dysfunction, acu-

Address for reprint requests and other correspondence: E. Stener-Victorin,


Institute of Neuroscience and Physiology, Dept. of Physiology, Sahlgrenska The costs of publication of this article were defrayed in part by the payment
Academy, Univ. of Gothenburg, Box 434, SE-405 30 Göteborg, Sweden of page charges. The article must therefore be hereby marked “advertisement”
(e-mail: elisabet.stener-victorin@neuro.gu.se). in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

R1124 0363-6119/09 $8.00 Copyright © 2009 the American Physiological Society http://www.ajpregu.org
Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS R1125
puncture exerted long-lasting beneficial effects on endocrine weekly. The study was concluded after ⬃12 wk of DHT exposure,
parameters and anovulation without negative side effects (8, when the rats were 15–16 wk of age.
40, 57). In overweight women with PCOS, exercise together EA. Low-frequency EA was administered to conscious rats every
with other lifestyle modification decreases central fat and second weekday for 4 –5 wk (12–14 treatments). The treatment
procedure has been described in detail previously (35). Each treatment
increases insulin sensitivity and ovulatory function (23, 27). lasted 15 min during the first week, 20 min during weeks 2–3, and
However, those studies were not focused on parameters related 25 min during weeks 4 –5. Acupuncture needles were inserted
to the autonomic nervous system or the mechanisms underly- bilaterally into the abdominal and hindlimb muscles, in somatic
ing the effects of EA. segments corresponding to the innervation of the ovaries. The
In rats with polycystic ovaries induced with estradiol valer- intensity was adjusted to produce local muscle contractions and
ate (EV), we found that low-frequency (2 Hz) EA (3, 37, varied from 0.8 –1.3 mA.
53–56) and physical exercise (36) restored the ovarian expres- Before needle insertion, rats were lightly anesthetized with 2%
sion of markers of sympathetic nervous system activity, con- isoflurane (Isoba Vet; Schering-Plough, Stockholm, Sweden) in a 1:1
sistent with the notion that EA and exercise inhibit sympathetic mixture of oxygen and air for 2–3 min. After needle insertion, the rats
hyperactivity. In rats with PCOS induced with dihydrotestos- were suspended in a fabric harness during EA treatment. To avoid
possible acute effects of EA, rats were not treated for 24 h before the
terone (DHT), we showed that low-frequency EA and physical experiment was ended.
exercise each reduced insulin resistance and the expression of Physical exercise. Rats in the PCOS exercise group were placed
adipose tissue genes associated with insulin resistance, obesity, individually in cages with an exercise wheel and allowed to exercise
and inflammation (35). EA did so without affecting adiposity voluntarily for 4 –5 wk. The exercise wheels were locked 24 h before
or adipose tissue cellularity. the experiment was ended to avoid possible acute effects of physical
Hyperandrogenism is the central feature of PCOS and may exercise.
be a key factor in the excessive sympathetic tone associated Three times per week, rats in the PCOS group and the PCOS
with the syndrome. Our DHT-induced rat PCOS model de- exercise group were anesthetized, suspended in a harness, and handled
velop obesity and display ovarian dysfunction, both of which in the same way as rats in the PCOS EA group but without needle
are associated with high sympathetic activity. Therefore, we insertion or EA stimulation. All rats were conscious during the
handling/treatment procedure.
aimed to investigate the mRNA expression of the ␤3-adrener- Vaginal smears. The stage of cyclicity was determined by mi-
gic receptor (Adrb3), Ngf, NGF receptor (Ngfr), and neuropep- croscopic analysis of the predominant cell type in vaginal smears
tide Y (Npy), all markers of sympathetic nerve activity, and of the obtained daily from rats at 11 wk of age to the end of the experi-
androgen receptor (Ar) in adipose tissue in our rat model of ment (38).
DHT-induced PCOS (34). We also assessed the effects of Tissue collection, RNA isolation and cDNA synthesis and real-
low-frequency EA and physical exercise on the expression of time RT-PCR. Rats were decapitated at 15–16 wk of age (i.e., after
those genes and on ovarian morphology. 4 –5 wk of treatment and 11–12 wk after pellet implantation). The
mesenteric fat depot was dissected, snap frozen in liquid nitrogen,
and stored at ⫺80°C for mRNA analyses. mRNA isolation and
MATERIALS AND METHODS
real-time RT-PCR were performed as previously described in
Animals detail by using a low-density array card (35). Primers and probes
for rat genes corresponding to the TaqMan Gene Expression Assay
Six Wistar dams, each with 8 –9 female pups, were purchased from numbers and GenBank accession numbers (Table 1). Gene expres-
Charles River (Frankfurt, Germany). Pups were raised with a lactating sion values were calculated with the 2⫺⌬⌬Ct method (31), where Ct is
dam until 21 days of age and were then housed four to five per cage cycle threshold. The ovaries were excised, fixed in neutral buffered
under controlled conditions (21–22°C, 55– 65% humidity, 12:12-h 4% formaldehyde for 24 h, placed in 70% ethanol, dehydrated, and
light-dark cycle). Rats were fed commercial chow containing 18.7% embedded in paraffin.
protein, 4.7% fat, 63% carbohydrates, vitamins, minerals (B&K Uni- Ovarian morphology. The ovaries were longitudinally and serially
versal, Sollentuna, Sweden), and tap water ad libitum. Animals were sectioned at 4 ␮m; every 20th section (n ⫽ 5 per ovary) was mounted
cared for in accordance with the principles of the Guide to the Care on a glass slide, stained with hematoxylin and eosin, and analyzed
and Use of Experimental Animals (49a). The study was approved by under a conventional birefringence microscope by two persons
the Animal Ethics Committee of the University of Gothenburg. blinded to the origin of the sections. The slides were scanned with
ScanScope (Aperio Technologies, Vista, CA) and analyzed with
Study Procedure ImageScope virtual microscopy software (Aperio Technologies). The
diameter of the ovaries in the section with the largest ovarian cross
The study procedure has been described (35). In brief, at 21 days of section was measured with a calibrated scale tool in the virtual
age, rats were randomly divided into two groups. The DHT-induced microscope. Antral follicles, distinguished by an antrum within the
PCOS rats (n ⫽ 36) were implanted subcutaneously in the neck with granulosa cell layers enclosing the oocyte, were counted by two
90-day continuous-release pellets (Innovative Research of America, persons (to avoid duplicate counting) and classified as atretic or
Sarasota, FL) containing 7.5 mg of DHT (daily dose, 83 ␮g), which healthy. Follicles were considered atretic if at least two pyknotic
induces metabolic disturbances and other characteristics of PCOS in granulosa cells were observed or if the oocyte showed obvious signs
adulthood (34); controls (n ⫽ 13) received pellets containing 7.5 mg of degeneration (22). The thickness of theca interna cell layer was
of vehicle. Forty days later, to compensate for weight gain, an measured in the largest healthy and atretic antral follicles. Corpora
additional pellet was implanted that released 3.5 mg of DHT or lutea were noted but not counted.
placebo for 60 days (i.e., an additional daily dose of 58 ␮g). A
microchip (AVID, Norco, CA) with an identification number was Statistical Analyses
inserted along with the pellets. After 7 wk of DHT exposure, rats in
the PCOS group were randomly subdivided into three treatment All statistical evaluations were performed with SPSS software
groups: PCOS (n ⫽ 12), PCOS plus exercise (n ⫽ 13), and PCOS plus (version 13.0; SPSS, Chicago, IL). Values are reported as means ⫾
EA (n ⫽ 11). The treatments lasted 4 –5 wk. The rats were weighed SE. The Kruskal-Wallis test was used for comparisons of all groups,
AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org
Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
R1126 ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS

Table 1. Genes presented on the TaqMan low-density array with their respective TaqMan gene expression assay numbers
and GenBank accession numbers
Gene Symbol Gene Description TaqMan Gene Expression Assay No. GenBank Accession No.

Putative Reference Genes


18S 18S ribosomal RNA Hs99999901_s1 X03205
Actb ␤-Actin Rn00667869_m1 NM_012583.2
Hprt Hypoanthine guanine phosphoribosyl transferase Rn01527840_m1 NM_012583-2
Ppia Peptidylprolyl isomerase A Rn00690933_m1 NM_017101.1
Target Genes
Adrb3 Adrenergic receptor, beta 3 Rn00565393_m1 NM_013108.1
Ar Androgen receptor Rn00560747_m1 NM_012502.1
Ngfr Nerve growth factor receptor Rn00561634_m1 NM_012610.1
Ngf Nerve growth factor, beta Rn01533872_m1 XM_227525.3
Npy Neuropeptide Y Rn00561681_m1 NM_012614.1

and if significant, a Mann-Whitney U-test was performed between untreated DHT-induced PCOS rats (Table 2). However, both
individual groups. P ⬍ 0.05 was considered significant. the PCOS exercise and PCOS EA rats had still a signifi-
RESULTS
cantly higher proportion of atretic antral follicles compared
with controls (Fig. 2). In the PCOS EA group, fresh corpora
Exercise and EA Influence Ovarian Morphology lutea were observed in five of eleven rats (45%) (Fig. 1,
E–F), indicating ovulation. In the PCOS exercise group,
All control rats had a normal estrus cycle of 4 days. Rats in healthy follicles were observed, but no fresh corpora lutea
the PCOS group were acyclic, while those in the PCOS (Fig. 1, G–H).
exercise and PCOS EA groups exhibited irregular cycles.
Ovarian weight and area were lower in the PCOS group than in PCOS Increases Gene Expression of Sympathetic Markers in
the controls, and neither variable was affected by exercise or Mesenteric Adipose Tissue
low-frequency EA (Table 2).
Morphologically, ovaries from control rats exhibited fol- In mesenteric adipose tissue, expression of Adrb3, Ngf, and
licles in various stages of development, ranging from pri- Npy mRNA was higher in rats with DHT-induced PCOS than
mordial follicles to mature preovulatory follicles, as well as controls (Fig. 3). Expression of Ar and Ngfr mRNA did not
several corpora lutea, many resulting from recent ovulations differ in DHT-induced PCOS rats and controls (Fig. 3).
(Fig. 1, A–B). Some antral follicles contained pyknotic granu-
losa cells. EA and Physical Exercise Influence the Expression of
In rats with DHT-induced PCOS, small follicles in early Sympathetic Markers and AR in Mesenteric Adipose Tissue
development were observed, as were antral follicles with Repeated low-frequency EA and physical exercise for
various degrees of atresia: some follicles had nuclear pyk- 4 –5 wk each lowered DHT-induced changes in the expres-
nosis in few cells, and others exhibited massive degenera- sion of Ngf and Npy mRNA in mesenteric adipose tissue; EA
tion and granulosa cells in the antrum, as well as degener- also lowered the expression of Adrb3 and Ar mRNA (Fig.
ated cells containing pyknotic nuclei scattered throughout 3). Neither exercise nor EA influenced Ngfr mRNA expres-
the membrane granulosa (Fig. 1, C–D). Corpora lutea were sion (Fig. 3). Furthermore, the expression of selected genes
absent in all PCOS rats. Although the number of antral after low-frequency EA and physical exercise were not
follicles was similar in DHT-induced PCOS and control significantly different from the corresponding level in con-
rats, DHT-induced PCOS rats had a higher proportion of trol rats.
atretic antral follicles (Fig. 2) in which the thickness of
theca interna was increased (Table 2). DISCUSSION
Rats in the PCOS exercise and PCOS EA groups had a
lower proportion of atretic antral follicles (Fig. 2), and the Increased sympathetic activity may be an important etiolog-
theca interna cell layer in those follicles was thinner than in ical factor in PCOS (15, 17, 19, 20, 48, 49, 58, 61). Like the

Table 2. Effects of physical exercise and electro-acupuncture on ovarian weight and area and thickness of the theca interna
of antral follicles
Variable Control PCOS PCOS Exercise PCOS EA Kruskal-Wallis

Average weight of one ovary, mg 82.64⫾3.04 35.39⫾2.17* 39.06⫾2.74* 37.39⫾3.27* P ⬍ 0.001


Ovarian area, mm2 24.00⫾1.43 7.74⫾0.43* 8.64⫾0.62* 6.61⫾1.13* P ⬍ 0.001
Thickness of TI, ␮m
Atretic antral follicles 23.40⫾2.45 31.71⫾1.34† 26.84⫾1.03‡ 22.86⫾1.65§ P ⬍ 0.001
Healthy antral follicles 19.95⫾1.80 18.53⫾0.98 19.23⫾1.02 16.77⫾1.18 NS
Values are means ⫾ SE, n ⫽ 10 –12/group. PCOS, polycystic ovary syndrome; TI, theca interna; NS, not significant. *P ⬍ 0.001 vs. control (Mann-Whitney
U-test); †P ⬍ 0.01 vs. control (Mann-Whitney U-test); ‡P ⬍ 0.05 vs. PCOS (Mann-Whitney U-test); §P ⬍ 0.01 vs. PCOS (Mann-Whitney U-test).

AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org


Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS R1127

Fig. 1. A: ovary from a normal cycling control


rat. B: high-power view shows a recently ovu-
lated collapsed follicle with the apical rupture
hole (broken arrow) closed by granulosa cells
and perifollicular edema (thin arrow) typical of
ovulation and (on the left) an antral follicle
containing macrophages (thick arrow) and a
trapped oocyte (long thick arrow). C: ovary
from a rat with dihydrotestosterone (DHT)-
induced polycystic ovary syndrome (PCOS)
with small early atretic follicles (EAF) and
antral follicles with various degrees of atresia.
D: high-power view shows degenerating py-
knotic granulosa cells in the antral part of the
membrane granulosa in EAF. E: ovary from a
rat in PCOS EA group with healthy follicles
(HF), atretic follicles (AF), and fresh corpora
lutea (FCL). F: high-power view shows FCL
and a HF. G: ovary from a rat in the PCOS
exercise group with HF, EAF, and old AF
(arrows). H: high-power view shows a HF and
an EAF.

metabolic syndrome, PCOS is characterized by insulin resis- is increased in mesenteric adipose tissue, a fat depot that
tance and obesity, which are associated with enhanced activity affects metabolic status by releasing free fatty acids to the liver
of the sympathetic nervous system (28, 59). Recently, it was via the portal vein. This finding supports the notion that
demonstrated that sympathetic activation is greater in subjects autonomic nervous system is involved in the pathogenesis of
with central obesity, a feature of PCOS, than in those with PCOS. Low-frequency EA and physical exercise each influ-
peripheral obesity (18). These findings underscore the impor- enced ovarian morphology and downregulated the expression
tance of evaluating new treatment strategies that attenuate of several markers of sympathetic nervous activity in mesen-
sympathetic activity in patients with PCOS. teric adipose tissue, indicating that these interventions exert
Using a rat model of DHT-induced PCOS, we show here their effects by modulating sympathetic outflow to the adipose
that the expression of several markers of sympathetic activity tissue and ovaries.
AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org
Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
R1128 ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS

one) and metabolic function (decreases in body mass index and


fasting serum insulin) in women with PCOS (1, 14).

Markers of Sympathetic Activity in Mesenteric Adipose


Tissue. Effect of Treatment?
The autonomic nervous system has a distinct organization in
different adipose tissue depots. The same set of neurons con-
trols the various intra-abdominal adipose tissue depots,
whereas subcutaneous adipose tissue located outside the ab-
dominal compartment receives input from other autonomic
neurons (45).
NPY. NPY, a phenotypic marker for sympathetic neurons, is
colocalized and coreleased with norepinephrine at peripheral
sites of action and is therefore considered a good indicator of
Fig. 2. Ratio of healthy and atretic antral follicles, counted from 5 consecutive sympathetic activity (42). In coculture with adipocytes, sym-
slides. Values are means ⫾ SE, n ⫽ 10 –12/group. P ⬍ 0.001 (Kruskal Wallis), pathetic neurons secrete increased amounts of NPY, suggesting
†††
P ⬍ 0.001; ††P ⬍ 0.01 vs. control (Mann-Whitney U-test); *P ⬍ 0.05 vs.
PCOS (Mann-Whitney U-test).
cross talk between these two cell types (60). Adipose tissue and
adipocytes both produce and secrete NPY (29), which is
important in adipose tissue remodeling that results in abdom-
Mechanisms Behind the Effects of Low-Frequency EA and inal obesity and metabolic syndrome (30). Consistent with
Physical Exercise these findings, mesenteric expression of Npy mRNA was
higher in our DHT-induced rat PCOS model than in controls.
Most likely, low-frequency EA and physical exercise ex- Furthermore, in women with PCOS, circulating levels of NPY
erted their effects in rats with DHT-induced PCOS by stimu- are elevated independently of body mass index (4). Both
lating ergoreceptors and somatic afferents in the muscles, low-frequency EA and physical exercise resulted in pro-
which results in modulation of spinal reflexes and central nounced downregulation of DHT-induced increase of in mes-
sympathetic outflow. Previously, we showed that low-fre- enteric Npy mRNA expression, indicating decreased sympa-
quency EA increases ovarian blood flow and that this effect is thetic activity. Moreover, the mesenteric Npy expression after
mediated as a reflex response via ovarian sympathetic nerves, low-frequency EA and physical exercise were not significantly
which in turn is controlled via central nervous system pathways different from the corresponding level in control rats.
(50). Furthermore, reduction in central sympathetic activity, as NGF and NGFR. The development and maintenance of
reflected by a drop in blood pressure, after low-frequency sympathetic nerves are facilitated by the target-derived neuro-
electrical muscle stimulation is mediated by the release of trophin NGF and its low-affinity receptor (NGFR). NGF is
␤-endorphin (24, 25). Neurons that express proopiomelanocor- secreted from adipocytes and can therefore be considered to be
tin (the precursor of ␣-MSH and ␤-endorphin) and other an adipokine (46), which may be involved in communication
neuropeptides reside in the hypothalamic arcuate nucleus, a between adipocytes and sympathetic neurons. NGF expression
site for the regulation of metabolism and reproduction (9). is closely linked to inflammatory conditions, as TNF-␣ stim-
Hypothetically, low-frequency EA and physical exercise could ulates NGF production by adipocyte (46). Overweight women
each modulate sympathetic outflow, metabolism, and release of and women with metabolic syndrome have high levels of
gonatropin-releasing hormone and lutenizing hormone (LH) circulating NGF (7), which are associated with insulin resis-
through effects on ␤-endorphin and other neuropeptides (9, tance, as well as increased levels of IL-6 and leptin. The high
51). Evidence that ␤-endorphin is involved in PCOS comes circulatory levels of NGF may reflect the increased levels of
from recent trials in which naltrexone, a ␮-receptor antagonist, NGF mRNA in adipose tissue in obese women (7). NGF
improved endocrine function (induced ovulation and decreases production in adipose tissue is also increased in genetic models
in LH, LH-to-follicle-stimulating hormone ratio, and testoster- of obesity in animals (44).

Fig. 3. Relative expression of ␤3-adrenergic re-


ceptor (ADRB3), androgen receptor (AR), nerve
growth factor (NGF), NGF receptor (NGFR),
and neuropeptide Y (NPY) mRNA in mesenteric
adipose tissue in control rats and in the 3 PCOS
groups. Values are means ⫾ SE, n ⫽ 10/group.
The average cycle threshold (Ct) value for all
genes was 28.6 ⫾ 0.1 (range, 23.8 –34.6).
Kruskal-Wallis analysis: ADRB3, P ⬍ 0.01; AR,
P ⬍ 0.01; NGF, P ⬍ 0.01; NPY, P ⬍ 0.001.

P ⬍ 0.05, ††P ⬍ 0.01 vs. control (Mann-Whit-
ney U-test). ***P ⬍ 0.001 vs. PCOS (Mann-
Whitney U-test).

AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org


Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS R1129
Ngf expression in mesenteric adipose tissue was higher in EA (54, 56) in rats with EV-induced polycystic ovaries. A
rats with DHT-induced PCOS than in controls and was reduced reasonable explanation for this discrepancy might be that rats
by both low-frequency EA and exercise. This result is in line in the present study were conscious during EA but were
with the decrease we observed in the high levels of ovarian anesthetized in our previous studies. Regarding the exercise,
NGF expression after 4 wk of low-frequency EA and exercise rats with EV-induced polycystic ovaries ran longer distances
in rats with EV-induced polycystic ovaries (36, 54, 56). Fur- than rats with DHT-induced PCOS in the present study. We
thermore, the Ngf expression level after low-frequency EA and found no differences in ovarian weight or area after EA or
exercise was not different from the Ngf expression level in exercise treatment. Since corpora lutea were observed in the
control rats. EA group, one would expect increases in ovarian weight and
ADRB3. Lipolysis is under control of stimulatory ␤-ADR area. However, the EA group had fewer antral follicles than
and inhibitory ␣-ADR. Increased release of free fatty acids untreated rats with DHT-induced PCOS, which might explain
from visceral fat cells, due to enhanced lipolysis, to the portal why weight and area were unaffected.
venous system has been hypothesized to cause insulin resis- Furthermore, the theca interna cell layer was thinner in both
tance and other metabolic disturbances. In lean women with the PCOS EA and PCOS exercise groups than in PCOS rats.
PCOS, lipolytic activity in visceral adipose tissue is higher Since the theca interna is innervated by sympathetic neurons
than in lean controls (12). This finding might partly explain the and is important in steroidogenesis (47), the reduced thickness
increased prevalence of insulin resistance in lean women with might reflect inhibition of ovarian sympathetic neurons. Previ-
PCOS, although obesity has an additive role to insulin resis- ously, we showed that low-frequency EA increases ovarian
tance in PCOS (11, 41, 58). Moreover, a relationship between blood flow (50, 52). Transection of the ovarian sympathetic
upper-body obesity, its associated metabolic complications, nerves eliminated this response and also reversed the changes
and increased visceral fat ADRB3 sensitivity has been reported in ovarian morphology induced by EV in rats with polycystic
(26, 32). ovaries (5). These findings confirm the role of the sympathetic
In the present study, expression of Adrb3 mRNA in mesen- nervous system in the control of ovarian function. Further
teric adipose tissue was higher in rats with DHT-induced supporting the involvement of sympathetic nervous system in
PCOS than in controls, supporting the notion that androgens PCOS, women with the syndrome have significantly higher
enhance lipolytic activity in rodents (2). Low-frequency EA NGF levels in the follicular fluid than controls (16).
downregulated Adrb3 gene expression compared with un-
treated PCOS rats, and the Adrb3 expression after low-fre- Perspectives and Significance
quency EA was not different from the corresponding level in Previously, we found that women with PCOS had increased
control rats. Exercise had no effect on Adrb3 gene expression. sympathetic nerve activity related to hormonal and metabolic
Lower Adrb3 expression in visceral adipose tissue might de- features and that both testosterone and cholesterol were inde-
crease lipolytic activity in this depot, a potential explanation pendent predictors of such activity, with testosterone having
for the increased insulin sensitivity induced by EA in our the stronger impact (60). It is not yet clear whether increased
previous study (35). sympathetic activity is an etiologic factor in PCOS or a
AR. The presence of ARs in preadipocytes and adipocytes consequence of hyperandrogenism. However, our findings sup-
suggests that androgens contribute to the control of adipose port the theory that increased sympathetic activity contributes
development and regulation (10). The induction of PCOS in to the development and maintenance of PCOS. Thus, therapies
our rat model by continuous administration of DHT did not aimed at reducing sympathetic nerve activity might alleviate
influence the expression of Ar mRNA in adipose tissue. How- the signs and symptoms of PCOS.
ever, after EA treatment, Ar gene expression in adipose tissue
was lower than in untreated rats with DHT-induced PCOS, and Conclusion
did not differ from Ar gene expression in controls.
In rats with DHT-induced PCOS, low-frequency EA and
exercise each reduced the expression of genes encoding mark-
Exercise and EA Improve Ovarian Morphology
ers of sympathetic activity in adipose tissue and had beneficial
Ovarian morphology in DHT-induced PCOS rats has been effects on ovarian morphology, reflecting modulation of sym-
extensively described elsewhere (34). In evaluating the effects pathetic outflow to the adipose tissue and ovaries.
of different treatment modalities, it is important to be aware
ACKNOWLEDGMENTS
that the rat is a multiovulatory species. Therefore, the ovarian
morphology of our DHT-induced rat PCOS model cannot be We thank Malin Govik and Kristy McKamey-Blom for technical assistance.
directly compared with human polycystic ovaries. For exam- We also thank the SWEGENE Göteborg Genomics Core Facility platform,
University of Gothenburg.
ple, the decreased ovarian size, as well as increased proportion
of atretic follicles, seen in this rat PCOS model, are not in line GRANTS
with human polycystic ovaries. Nevertheless, both exercise and
This study was supported by grants from the Swedish Medical Research
low-frequency EA positively influenced ovarian morphology. Council (project numbers: 2005-72VP-15445-01A, 2005-72VX-15276-01A,
In the PCOS EA groups, we observed fresh corpora lutea in K2007-54X-20325-01-3), Novo Nordisk Foundation, Wilhelm and Martina
45% of rats, indicating ovulation, and a decreased proportion Lundgrens’s Science Fund, Hjalmar Svensson Foundation, Tore Nilson Foun-
of atretic antral follicles, both novel findings. A lower propor- dation, Åke Wiberg Foundation, Herbert and Karin Jacobsson’s Foundation,
Swedish Diabetes Association Research Foundation, Adlerbert Research Foun-
tion of atretic antral follicles was also observed in the PCOS dation, Ekhaga Foundation, and the Swedish federal government under the
exercise group. Previously, we showed that ovarian morpho- LUA/ALF agreement ALFFGBG-10984 and Regional research and develop-
logical changes are partly reversed by exercise (36) but not by ment (FOU) agreement.

AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org


Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
R1130 ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS

REFERENCES implications of ovarian surgery for ovulation induction in polycystic ovary


syndrome. Hum Reprod Update 13: 249 –264, 2007.
1. Ahmed MI, Duleba AJ, El Shahat O, Ibrahim ME, Salem A. Naltrex- 22. Hirshfield AN, Midgley ARJ. Morphometric analysis of follicular devel-
one treatment in clomiphene resistant women with polycystic ovary opment in the rat. Biol Reprod 19: 597– 605, 1978.
syndrome. Hum Reprod 23: 2564 –2569, 2008. 23. Hoeger KM. Role of lifestyle modification in the management of poly-
2. Arner P. Effects of testosterone on fat cell lipolysis. Species differences cystic ovary syndrome. Best Pract Res Clin Endocrinol Metab 20: 293–
and possible role in polycystic ovarian syndrome. Biochimie 87: 39 – 43, 310, 2006.
2005. 24. Hoffmann P, Carlsson S, Skarphedinsson JO, Thoren P. Role of
3. Bai YH, Lim SC, Song CH, Bae CS, Jin CS, Choi BC, Jang CH, Lee different serotonergic receptors in the long-lasting blood pressure depres-
SH, Pak SC. Electro-acupuncture reverses nerve growth factor abundance sion following muscle stimulation in the spontaneously hypertensive rat.
in experimental polycystic ovaries in the rat. Gynecol Obstet Invest 57: Acta Physiol Scand 139: 305–310, 1990.
80 – 85, 2004. 25. Hoffmann P, Thoren P. Electric muscle stimulation in the hind leg of the
4. Baranowska B, Radzikowska M, Wasilewska-Dziubinska E, Kaplin- spontaneously hypertensive rat induces a long-lasting fall in blood pres-
ski A, Roguski K, Plonowski A. Neuropeptide Y, leptin, galanin and sure. Acta Physiol Scand 133: 211–219, 1988.
insulin in women with polycystic ovary syndrome. Gynecol Endocrinol 26. Hoffstedt J, Wahrenberg H, Thorne A, Lönnqvist F. The metabolic
13: 344 –351, 1999. syndrome is related to ␤3-adrenoceptor sensitivity in visceral adipose
5. Barria A, Leyton V, Ojeda SR, Lara HE. Ovarian steroidal response to tissue. Diabetologia 39: 838 – 844, 1996.
gonadotropins and ␤-adrenergic stimulation is enhanced in polycystic 27. Huber-Buchholz MM, Carey DGP, Norman RJ. Restoration of repro-
ovary syndrome: role of sympathetic innervation. Endocrinology 133: ductive potential by lifestyle modification in obese polycystic ovary
2696 –2703, 1993. syndrome: role of insulin sensitivity and luteinizing hormone. J Clin
6. Bartness TJ, Song CK. Thematic review series: Adipocyte biology. Endocrinol Metab 84: 1470 –1474, 1999.
Sympathetic and sensory innervation of white adipose tissue. J Lipid Res 28. Kaaja RJ, Poyhonen-Alho MK. Insulin resistance and sympathetic
48: 1655–1672, 2007. overactivity in women. J Hypertens 24: 131–141, 2006.
7. Bullo M, Peeraully MR, Trayhurn P, Folch J, Salas-Salvado J. Cir- 29. Kos K, Harte AL, James S, Snead DR, O’Hare JP, McTernan PG,
culating nerve growth factor levels in relation to obesity and the metabolic Kumar S. Secretion of neuropeptide Y in human adipose tissue and its
syndrome in women. Eur J Endocrinol 157: 303–310, 2007. role in maintenance of adipose tissue mass. Am J Physiol Endocrinol
8. Chen BY, Yu J. Relationship between blood radioimmunoreactive ␤-en- Metab 293: E1335–E1340, 2007.
dorphin and hand skin temperature during the electro-acupuncture induc- 30. Kuo LE, Kitlinska JB, Tilan JU, Li L, Baker SB, Johnson MD, Lee EW,
tion of ovulation. Acupunct Electrother Res 16: 1–5, 1991. Burnett MS, Fricke ST, Kvetnansky R, Herzog H, Zukowska Z. Neu-
9. Crown A, Clifton DK, Steiner RA. Neuropeptide signaling in the ropeptide Y acts directly in the periphery on fat tissue and mediates stress-
integration of metabolism and reproduction. Neuroendocrinology 86: induced obesity and metabolic syndrome. Nat Med 13: 803– 811, 2007.
175–182, 2007. 31. Livak KJ, Schmittgen TD. Analysis of relative gene expression data
10. Dieudonne MN, Pecquery R, Boumediene A, Leneveu MC, Giudicelli using real-time quantitative PCR and the 2⫺⌬⌬CT method. Methods 25:
Y. Androgen receptors in human preadipocytes and adipocytes: regional 402– 408, 2001.
specificities and regulation by sex steroids. Am J Physiol Cell Physiol 274: 32. Lönnqvist F, Thome A, Nilsell K, Hoffstedt J, Arner P. A pathogenic
C1645–C1652, 1998. role of visceral fat ␤3-adrenoceptors in obesity. J Clin Invest 95: 1109 –
11. Dunaif A, Segal KR, Futterweit W, Dobrjansky A. Profound peripheral 1116, 1995.
insulin resistance, independent of obesity, in polycystic ovary syndrome. 33. Mancia G, Bousquet P, Elghozi JL, Esler M, Grassi G, Julius S, Reid
Diabetes 38: 1165–1174, 1989. J, Van Zwieten PA. The sympathetic nervous system and the metabolic
12. Ek I, Arner P, Ryden M, Holm C, Thorne A, Hoffstedt J, Wahrenberg syndrome. J Hypertens 25: 909 –920, 2007.
H. A unique defect in the regulation of visceral fat cell lipolysis in the 34. Mannerås L, Cajander S, Holmäng A, Seleskovic Z, Lystig T, Lönn
polycystic ovary syndrome as an early link to insulin resistance. Diabetes M,. Stener-Victorin E. A new rat model exhibiting both ovarian and
51: 484 – 492, 2002. metabolic characteristics of polycystic ovary syndrome. Endocrinology
13. Fagius J. Sympathetic nerve activity in metabolic control—some basic 148: 3781–3791, 2007.
concepts. Acta Physiol Scand 177: 337–343, 2003. 35. Mannerås L, Jonsdottir IH, Holmäng A, Lönn M, Stener-Victorin E.
14. Fruzzetti F, Bersi C, Parrini D, Ricci C, Genazzani AR. Effect of Low frequency electro-acupuncture and physical exercise improve meta-
long-term naltrexone treatment on endocrine profile, clinical features, and bolic disturbances and modulate gene expression in adipose tissue in rats
insulin sensitivity in obese women with polycystic ovary syndrome. Fertil with dihydrotestosterone-induced polycystic ovary syndrome. Endocrinol-
Steril 77: 936 –944, 2002. ogy 149: 3559 –3568, 2008.
15. Garcia-Rudaz C, Armando I, Levin G, Escobar ME, Barontini M. 36. Manni L, Cajander S, Lundeberg T, Naylor AS, Aloe L, Holmäng A,
Peripheral catecholamine alterations in adolescents with polycystic ovary Jonsdottir IH, Stener-Victorin E. Effect of exercise on ovarian mor-
syndrome. Clin Endocrinol 49: 221–228, 1998. phology and expression of nerve growth factor and ␣1- and ␤2-adrenergic
16. Garcia-Rudaz MC, Mayerhofer A, Ojeda SR, Dissen GA. An excessive receptors in rats with steroid-induced polycystic ovaries. J Neuroendocri-
ovarian production of nerve growth factor (NGF) facilitates the develop- nol 17: 846 – 858, 2005.
ment of polycystic ovarian morphology in mice and is a discernible feature 37. Manni L, Lundeberg T, Holmäng A, Aloe L, Stener-Victorin E. Effect
ovarian syndrome in humans. Proc Endocrine Society’s 90th Annual of electro-acupuncture on ovarian expression of ␣1- and ␤2-adrenoceptors,
Meeting. San Francisco, CA, 2008. and p75 neurotrophin receptors in rats with steroid-induced polycystic
17. Giallauria F, Palomba S, Manguso F, Vitelli A, Maresca L, Tafuri D, ovaries. Reprod Biol Endocrinol 3: 21, 2005.
Lombardi G, Colao A, Vigorito C, Orio F. Abnormal heart rate recovery 38. Marcondes FK, Bianchi FJ, Tanno AP. Determination of the estrous
after maximal cardiopulmonary exercise stress testing in young over- cycle phases of rats: Some helpful considerations. Braz J Biol 62: 609 –
weight women with polycystic ovary syndrome. Clin Endocrinol (Oxf) 68: 614, 2002.
88 –93, 2008. 39. Middlekauff HR, Hui K, Yu JL, Hamilton MA, Fonarow GC, Morigu-
18. Grassi G, Dell’Oro R, Facchini A, Quarti Trevano F, Bolla GB, chi J, MacLellan WR, Hage A. Acupuncture inhibits sympathetic acti-
Mancia G. Effect of central and peripheral body fat distribution on vation during mental stress in advanced heart failure patients. J Card Fail
sympathetic and baroreflex function in obese normotensives. J Hypertens 8: 399 – 406, 2002.
22: 2363–2369, 2004. 40. Mo X, Li D, Pu Y, Xi G, Le X, Fu Z. Clinical studies on the mechanism
19. Greiner M, Paredes A, Araya V, Lara HE. Role of stress and sympa- for acupuncture stimulation of ovulation. J Tradit Chin Med 13: 115–119,
thetic innervation in the development of polycystic ovary syndrome. 1993.
Endocrine 28: 319 –324, 2005. 41. Morales AJ, Laughlin GA, Butzow T, Maheshwari H, Baumann G,
20. Heider U, Pedal I, Spanel-Borowski K. Increase in nerve fibers and loss Yen SS. Insulin, somatotropic, and luteinizing hormone axes in lean and
of mast cells in polycystic and postmenopausal ovaries. Fertil Steril 75: obese women with polycystic ovary syndrome: common and distinct
1141–1147, 2001. features. J Clin Endocrinol Metab 81: 2854 –2864, 1996.
21. Hendriks ML, Ket JCF, Hompes PGA, Homburg R, Lambalk CB. 42. Morris MJ, Russell AE, Kapoor V, Cain MD, Elliott JM, West MJ,
Why does ovarian surgery in PCOS help? Insight into the endocrine Wing LM, Chalmers JP. Increases in plasma neuropeptide Y concentra-

AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org


Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.
ACUPUNCTURE AND EXERCISE RESTORE SYMPATHETIC MARKERS IN PCOS R1131
tions during sympathetic activation in man. J Auton Nerv Syst 17: 143– 53. Stener-Victorin E, Lindholm C. Immunity and ␤-endorphin concentra-
149, 1986. tions in hypothalamus and plasma in rats with steroid-induced polycystic
43. Mueller PJ. Exercise training and sympathetic nervous system activity: ovaries: effect of low-frequency electroacupuncture. Biol Reprod 70:
evidence for physical activity dependent neural plasticity. Clin Exp Phar- 329 –333, 2004.
macol Physiol 34: 377–384, 2007. 54. Stener-Victorin E, Lundeberg T, Cajander S, Aloe L, Manni L,
44. Nisoli E, Tonello C, Benarese M, Liberini P, Carruba MO. Expression Waldenström U, Janson P. Steroid-induced polycystic ovaries in rats:
of nerve growth factor in brown adipose tissue: implications for thermo- effect of electro-acupuncture on concentrations of endothelin-1 and nerve
genesis and obesity. Endocrinology 137: 495–503, 1996. growth factor (NGF), and expression of NGF mRNA in the ovaries, the
45. Nogueiras R, Wilson H, Rohner-Jeanrenaud F, Tschöp MH. Central adrenal glands, and the central nervous system. Reprod Biol Endocrinol 1:
nervous system regulation of adipocyte metabolism. Regul Pept 149: 33, 2003.
26 –31, 2008. 55. Stener-Victorin E, Lundeberg T, Waldenström U, Bileviciute-Ljungar
46. Peeraully MR, Jenkins JR, Trayhurn P. NGF gene expression and I, Janson PO. Effects of electro-acupuncture on corticotropin-releasing
secretion in white adipose tissue: regulation in 3T3–L1 adipocytes by factor in rats with experimentally-induced polycystic ovaries. Neuropep-
hormones and inflammatory cytokines. Am J Physiol Endocrinol Metab tides 35: 227–231, 2001.
287: E331–E339, 2004. 56. Stener-Victorin E, Lundeberg T, Waldenström U, Manni L, Aloe L,
47. Ricu M, Paredes A, Greiner M, Ojeda SR, Lara HE. Functional
Gunnarsson S, Janson PO. Effects of electro-acupuncture on nerve
development of the ovarian noradrenergic innervation. Endocrinology
growth factor and ovarian morphology in rats with experimentally induced
149: 50 –56, 2008.
polycystic ovaries. Biol Reprod 63: 1497–1503, 2000.
48. Semenova II. Adrenergic innervation of ovaries in Stein-Leventhal syn-
57. Stener-Victorin E, Waldenström U, Tägnfors U, Lundeberg T, Lind-
drome [in Russian]. Vestn Akad Med Nauk SSSR 24: 58 – 62, 1969.
49. Shoupe D, Lobo RA. Evidence for altered catecholamine metabolism in stedt G, Janson PO. Effects of electro-acupuncture on anovulation in
polycystic ovary syndrome. Am J Obstet Gynecol 150: 566 –571, 1984. women with polycystic ovary syndrome. Acta Obstet Gynecol Scand 79:
49a.Statens jordbruksverks föreskrifter om uppfödning, förvaring, tillhanda- 180 –188, 2000.
hållande och användning m.m. av försöksdjur. L 50, DFS 2004: 15, 58. Sverrisdottir YB, Mogren T, Kataoka J, Janson PO, Stener-Victorin
www.sjv.se. E. Is polycystic ovary syndrome associated with high sympathetic nerve
50. Stener-Victorin E, Fujisawa S, Kurosawa M. Ovarian blood flow activity and size at birth? Am J Physiol Endocrinol Metab 294: E576 –
responses to electroacupuncture stimulation depend on estrous cycle and E581, 2008.
on site and frequency of stimulation in anesthetized rats. J Appl Physiol 59. Troisi RJ, Weiss ST, Parker DR, Sparrow D, Young JB, Landsberg L.
101: 84 –91, 2006. Relation of obesity and diet to sympathetic nervous system activity.
51. Stener-Victorin E, Jedel E, Mannerås L. Acupuncture in polycystic Hypertension 17: 669 – 677, 1991.
ovary syndrome: current experimental and clinical evidence. J Neuroen- 60. Turtzo LC, Marx R, Lane MD. Cross-talk between sympathetic neurons
docrinol 20: 290 –298, 2008. and adipocytes in coculture. Proc Natl Acad Sci USA 98: 12385–12390, 2001.
52. Stener-Victorin E, Kobayashi R, Kurosawa M. Ovarian blood flow 61. Yildirir A, Aybar F, Kabakci G, Yarali H, Oto A. Heart rate variability
responses to electro-acupuncture stimulation at different frequencies and in young women with polycystic ovary syndrome. Ann Noninvasive
intensities in anaesthetized rats. Auton Neurosci 108: 50 –56, 2003. Electrocardiol 11: 306 –312, 2006.

AJP-Regul Integr Comp Physiol • VOL 296 • APRIL 2009 • www.ajpregu.org


Downloaded from www.physiology.org/journal/ajpregu by ${individualUser.givenNames} ${individualUser.surname} (152.118.148.234) on January 15, 2018.
Copyright © 2009 American Physiological Society. All rights reserved.

Você também pode gostar