You are on page 1of 13

Available online at www.sciencedirect.

com

Domestic Animal Endocrinology 36 (2009) 32–44

Trilostane-induced inhibition of cortisol secretion results in reduced


negative feedback at the hypothalamic–pituitary axis
Takahiro Teshima a,∗ , Yasushi Hara a , Susumu Takekoshi b , Yoshinori Nezu a ,
Yasuji Harada a , Takuya Yogo a , Akira Teramoto c ,
Robert Y. Osamura b , Masahiro Tagawa a
a Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine,

Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
b Department of Pathology, Tokai University School of Medicine, 143 Shimokasuya, Isehara-shi, Kanagawa 259-1193, Japan
c Department of Neurosurgery, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan

Received 4 September 2008; received in revised form 28 September 2008; accepted 1 October 2008

Abstract
Cushing’s disease caused by pituitary corticotroph adenoma in dogs is usually treated by medical treatment, and the efficacy of this
treatment has been reported. However, controversy remains as to whether reduced negative feedback through the inhibition of cortisol
secretion, similar to Nelson’s syndrome, may appear as an adverse effect. The purpose of this study was to investigate the effect
of reduced negative feedback through the inhibition of cortisol secretion by daily trilostane administration on the pituitary–adrenal
axis in clinically normal dogs. Dogs were administered 5 mg/kg trilostane twice a day every day for 8 weeks (n = 8) or 16 weeks
(n = 3). After the initiation of trilostane administration, plasma adrenocorticotropic hormone (ACTH) concentrations were increased
remarkably. As assessed by magnetic resonance imaging (MRI) during administration, the pituitary became enlarged. After trilostane
administration, the cytoplasmic areas of the pituitary corticotrophs were increased and the ratio of pituitary corticotrophs to all cells
in the anterior lobe was greater in the trilostane-treated dogs than that in untreated animals. In addition, histological examinations
revealed bilateral adrenal cortical hyperplasia. Using real-time PCR quantification, the expression of proopiomelanocortin (POMC)
mRNA in the pituitary and ACTH receptor (ACTH-R) mRNA in the adrenal gland was greater in the dogs treated with trilostane
than in untreated dogs. These results indicate that reduced negative feedback induced hyperfunction of the pituitary corticotrophs
and pituitary enlargement in healthy dogs. These changes suggest that the inhibition of cortisol secretion by trilostane may increase
the risk for accelerating the growth of corticotroph adenomas in dogs with Cushing’s disease.
© 2008 Elsevier Inc. All rights reserved.

Keywords: Corticotroph; Cushing’s disease; Dog; Nelson’s syndrome; Trilostane

1. Introduction corticism, a major endocrine disorder in dogs, and


the majority of these cases, which represent Cush-
Pituitary-dependent hyperadrenocorticism (PDH) ing’s disease, had adenomas of the pituitary corticotroph
accounts for approximately 80–85% of hyperadreno- [1,2]. In veterinary clinical medicine, the prevalence of
advanced imaging modalities for diagnosis, such as com-
puter tomography (CT) and magnetic resonance imaging
∗ Corresponding author. Tel.: +81 422 31 4151; (MRI), has enabled a detailed visualization of the pitu-
fax: +81 422 33 8836. itary, and, in combination with endocrinological tests,
E-mail address: teshima@bg8.so-net.ne.jp (T. Teshima). has enabled the accurate diagnosis of Cushing’s disease.

0739-7240/$ – see front matter © 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.domaniend.2008.10.002
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 33

In regard to the treatment of PDH dogs, although sur- Committee at Nippon Veterinary and Life Science Uni-
gical treatment by transsphenoidal hypophysectomy has versity.
been reported to be useful [3–5], most dogs with PDH
are treated clinically to inhibit cortisol excess [6]. 2.2. Trilostane administration
Recently, trilostane has been used increasingly for
the treatment of PDH dogs, and its efficacy has been In the trilostane administration group, adrenocor-
reported [7–12]. Trilostane, a competitive inhibitor of tical function was inhibited by the administration of
3␤-hydroxysteroid dehydrogenase, inhibits the synthesis trilostane (DESOPAN® Tab.; Mochida Pharmaceutical,
of several steroids in the adrenal cortex including corti- Tokyo, Japan). Trilostane was capsuled at a dosage of
sol [13,14]. Mitotane, which has been used for a long 5 mg/kg body weight and administered orally twice a day
time, inhibits the secretion of cortisol by cell necrosis (10 mg/kg/day) with meals every day for 8 weeks (TS
due to cytotoxicity on the zona fasciculata (ZF) and zona group) and 16 weeks (TL group). The dosage was estab-
reticularis (ZR) in the adrenal cortex [15–17]. However, lished from several reports of trilostane [8–12,18–21].
trilostane inhibits cortisol secretion by competitively During the administration period, the general condition
inhibiting enzymes and adverse effects are less frequent of dogs was monitored.
than those that appear with mitotane treatment.
2.3. Endocrine tests and sample collection
Although there are previous reports demonstrating
that the clinical symptoms of PDH dogs were improved Blood samples for hormone measurements were col-
by treatment with trilostane [7–9,12], neither mitotane or lected from the jugular vein and transferred to ice-chilled
trilostane administration is aimed at a radical cure of the tubes containing EDTA and plain serum. Plasma and
disease arising from adenomas of the pituitary. More- serum were separated by centrifugation at 4 ◦ C for
over, reduced negative feedback due to the inhibition of 15 min and stored at −80 ◦ C until assayed.
cortisol secretion by mitotane or trilostane may increase The adrenocorticotropic hormone (ACTH)-stimu-
the risk for accelerating growth of the corticotroph lation test was performed by collecting blood samples
adenoma in dogs with Cushing’s disease. However, no for measurement of the cortisol concentration at 0 and
studies have investigated the effect of cortisol inhibition 60 min after the intravenous administration of 0.25 mg of
by trilostane on the pituitary corticotroph that is the synthetic ACTH (Cortrosyn® ; Daiichi Sankyo, Tokyo,
cause the disease. In this study, in order to investigate the Japan) [22].
effect of inhibition of cortisol secretion from the adrenal The corticotropin releasing hormone (CRH)-
cortex by trilostane on the pituitary corticotroph, stimulation test was performed by collecting blood
we examined the effect of daily administration of samples for measurement of the ACTH concentration
trilostane on the hypothalamic–pituitary–adrenal at 0 and 30 min after the intravenous administration
(HPA) axis in normal Beagle of 1.5 ␮g ovine corticotropin-releasing factor (Peptide
dogs. institute, Inc., Osaka, Japan) per kg body weight [23,24].
In the trilostane administration group, the ACTH-
2. Materials and methods stimulation test was performed prior to trilostane
administration, every week until 8 weeks during the
2.1. Dogs trilostane administration, and every 2 weeks after 8
weeks (TL group). The CRH-stimulation test was per-
Twenty-one healthy Beagles were used. The group formed prior to trilostane administration, at 30 (4 weeks)
characteristics were as follows: 9 males and 12 females; days, 58 (8 weeks) and 86 (12 weeks) days (TL group),
1–6 years old (mean: 2.1 years old); and 7.0–15.0 kg and after completion of the trilostane administration
body weight (mean: 9.8 kg). Dogs were randomly period. All ACTH- and CRH-stimulation tests were per-
assigned to a control group (n = 10, 4 males and 6 formed at 4 h after the administration of the morning
females, mean age was 2.8 years, and mean weight was dose of trilostane.
9.8 kg), a short term of trilostane administration group
(TS group) (n = 8, 3 males and 5 females, mean age 2.4. Hormone determination
was 1.5 years, and mean weight was 9.8 kg), and a long
term of trilostane administration group (TL group) (n = 3, Serum cortisol concentrations were measured using a
2 males and 1 female, mean age was 1.4 years, and competitive immunoassay (Immulite® Cortisol; Diag-
mean weight was 10.0 kg). Protocols for all experiments nostics Products Corporation, Los Angeles, USA) as
involving the use of dogs were approved by the Bioethics described previously [25]. The intra-assay coefficients of
34 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

variation (CV) were 8.8 and 5.8% at cortisol levels of 74 and the enclosed area was calculated by the computer
and 524 nmol/l, respectively. The inter-assay CV were according to the method used in a study on CT [28].
10.0 and 6.3% at cortisol levels of 74 and 524 nmol/l, From the height of the pituitary and the area of the
respectively. The sensitivity of the assay was 5.5 nmol/l. brain obtained in the Gd-T1-weighted image, the pitu-
Plasma ACTH concentrations were measured itary height/brain area (P/B) ratio was calculated, as
using a solid-phase, 2-site chemiluminescent enzyme previously described [28]. Pituitaries with a P/B ratio
immunometric assay (Immulite® ACTH; Diagnostic of more than 0.31 × 10−2 mm−1 were considered to be
Products Corporation, Los Angeles, USA) in duplicate enlarged. The MRI of the pituitary was performed prior
as described previously [26,27]. The intra-assay coef- to trilostane administration and every 2 weeks during the
ficients of variation were 9.6 and 4.9% at ACTH levels trilostane administration.
of 5.3 and 221.8 pmol/l, respectively. The inter-assay
CV were 8.8 and 5.1% at ACTH levels of 5.8 and 2.6. Tissue samples
248.9 pmol/l, respectively. The sensitivity of the assay
was 1.1 pmol/l. In cases that exceeded the measurement Each dog was euthanized by an intravenous injec-
range (>278 pmol/l), samples were diluted using a tion of an overdose pentobarbital solution following the
special diluent (Diagnostics Products Corporation, Los CRH-stimulation test in the control group and after the
Angeles, USA). administration of trilostane in the trilostane administra-
tion group. The pituitary and bilateral adrenal glands
2.5. Magnetic resonance imaging were carefully harvested and weighed before being fixed
in 4% paraformaldehyde for histological examination or
Food was withheld from the trilostane group for 18 h stored at −80 ◦ C for RNA extraction. Each pituitary was
prior to MRI. Following the intramuscular administra- cut at the median sagittal plane and each adrenal gland
tion of 0.25 mg droperidol (DROLEPTAN® ; Daiichi was cut twice transversely, midway between the inden-
Sankyo, Tokyo, Japan) per kg body weight, anesthe- tation of the phrenicoabdominal vein and each pole [29].
sia was induced by intravenous administration of 7 mg
2.7. Histological examination
propofol (Rapinovet® ; Schering-Plough Animal Health,
Tokyo, Japan) per kg body weight and maintained by Samples for histological examination were dehy-
inhalation of isoflurane (Escain® ; Merck, Osaka, Japan) drated and embedded in paraffin. Sections were cut at
and oxygen. 2 ␮m. One section was stained with hematoxylin and
The MRI of the pituitary was performed using a eosin (HE), and adjacent sections of pituitaries were
1.5 T superconducting magnet (VISART; Toshiba Medi- subjected to immunohistochemical staining by standard
cals, Tokyo, Japan). T1-weighted transverse images were enzyme antibody techniques using a monoclonal mouse
made before and after an intravenous bolus injection of antibody to synthetic ACTH1–39 (Dako Japan, Kyoto,
0.1 mmol contrast medium (Omniscan® , gadodiamide- Japan) [30].
hydrate (Gd); Daiichi Sankyo, Tokyo, Japan) per kg body
weight. With the dogs in sternal recumbence, transverse 2.8. Pituitary and adrenal morphometry
scans of the skull were made perpendicular to the skull
base from the rostral clinoid processes to the dorsum Pituitary and adrenal morphometry were per-
sellae using the spin-echo method, which obtained a 350- formed with the “Image J” software version 1.36
ms repetition time (TR), a 15-ms echo time (TE), and (http://rsb.info.nih.gov/ij/). In each dog, the area and
2.0-mm thick consecutive slices. After Gd-T1-weighted numbers of ACTH-positive cells in the anterior pitu-
images of the pituitary were obtained, T1-weighted itary gland were measured. For pituitary morphometry,
imaging of the adrenal gland was performed prior to the anterior pituitary gland was divided equally into five
trilostane administration. With the dogs in supine posi- regions between the dorsal end and the ventral end of the
tion, coronal scans of the adrenal glands were obtained pituitary gland in the median section, and two fields each
perpendicular to the spinous process using the field-echo were randomly selected from the five regions [30]. The
method, which obtained a TR of 200 ms, a TE of 6.8 ms, number of cells and the numbers of ACTH-positive cells
and 5.0-mm thick consecutive slices. per field of the anterior pituitary gland were measured
In each dog, the pituitary height was measured on in 10 fields at 400× magnification for each dog, and the
the image using the largest cross-section of the pitu- ratio of ACTH-positive cells were calculated.
itary. In order to enable adjustments for the size of the In the adrenal glands, the width of the zona glomeru-
dog, the edges of the brain were traced on this image losa (ZG), zona fasciculata, and zona reticularis were
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 35

determined at 10 different places. The number of cells reverse transcribed RNA, 10 ␮l SYBR GREEN PCR
per field of ZF and ZR were measured in 10 fields at Master Mix, 0.4 ␮l (10 ␮mol/l) of respective forward
400× magnification for each dog. and reverse primer, and 8.2 ␮l DEPC treated water per
reaction was prepared. The PCR reaction was divided
2.9. RNA extraction and reverse transcription into three stages: (1) 2 min at 50 ◦ C; (2) 2 min at
95 ◦ C, and (3) 15 s at 95 ◦ C, and 60 s at 60 ◦ C. Stage
Total RNA was extracted from pituitaries and adrenal 3 was repeated 40 times. All samples were assayed
glands in tubes containing 2 ml Trizol (Invitrogen Japan, in duplicate. The specificity and the size of the PCR
Tokyo, Japan). The tissues were disrupted using a products were assessed by adding a melt curve at the
standard homogenizer. For purification of total RNA, end of the amplifications and a single melting curve
samples were treated with RNeasy Mini Kit (QIAGEN, peak was observed. Data were quantitatively analyzed
Tokyo, Japan). The yield of RNA was quantified by mea- using a serial dilution of control samples included in
suring the optical density of a sample diluted to 1:20 each reaction to produce a standard curve. To normal-
at 260 and 280 nm. 0.5 ␮g of total RNA of pituitary ize the expression level of each gene in each sample,
and adrenal gland were then reverse transcribed with the expression level of three commonly used refer-
the SuperScriptTM III first-Strand Synthesis SuperMix ence genes (glyceraldehyde-3-phosphate dehydrogenase
(Invitrogen Japan, Tokyo, Japan). Reverse transcribed (GAPDH), beta glucuronidase (GUS), and ribosomal
samples were diluted to 20 ␮l in DEPC treated water protein S5 (RPS5)) was determined for each sample. The
(Invitrogen Japan, Tokyo, Japan). most stable reference gene for normalization of the rel-
ative concentrations of mRNA was identified using the
2.10. Real-time quantitative RT-PCR GeNorm Visual basic application for Microsoft Excel
(http://medgen.ugent.be/∼jvdesomp/genorm/) [33]. We
Real-time quantitative RT-PCR analysis for proopi- found that GAPDH was the most stably expressed refer-
omelanocortin (POMC) and ACTH receptor (ACTH-R) ence gene. The relative expression of each gene in each
mRNAs were performed using the Applied Biosys- sample was calculated by dividing by that of GAPDH.
tems 7500 Sequence Detections System (Applied
Biosystems Japan, Tokyo, Japan). Sequences of the 2.11. Statistical analysis
canine POMC and ACTH-R were obtained from NCBI
(http://www.ncbi.nlm.nih.gov/). Corresponding primer All results were presented as median and range. In the
pairs (forward and reverse) for the Sybergreen sys- trilostane administration group, changes in serum corti-
tem, as listed in Table 1, were designed using the sol concentration, plasma ACTH concentration, and P/B
NCBI database (http://www.ncbi.nlm.nih.gov/) and used ratio were analyzed by two-way ANOVA followed by
according to methods described in the literature [31,32]. Tukey–Kramer’s post hoc tests. Regarding excised tis-
The PCR reaction was performed in MicroAmp Strio sue weights of pituitary and bilateral adrenal glands, the
Tubes (Applied Biosystems, Tokyo, Japan) with an ratios and area of ACTH-positive cells in the anterior
end-volume of 20 ␮l. A master mix containing 1.0 ␮l pituitary gland, the width of ZG, ZF, and ZR, the num-

Table 1
Primer sequences.
Gene Sequence (5 → 3 ) Length (bp) Melting temperature (◦ C) Reference

POMC For GGCCTCTGTGGAAGTGAGTG 80 84.8 Accession No. AY024339


Rev ACGCCAGCAGGTTACTTTCC
ACTH-R For GATCTTGACGCTTCCCAGAG 147 82.4 Accession No. XM541098
Rev ATGTAGCAGGCGCAGTAAGG
GAPDH For GATGGGCGTGAACCATGAG 131 84.0 Sieber-Ruckstuhl et al. [31]
Rev TCATGAGGCCCTCCACGAT
GUS For CCTCCTGCCGTATTACCCTTG 117 81.4 Accession No. NM001003191
Rev TCTGGACGAAGTAACCCTTGG
RPS5 For TCACTGGTGAGAACCCCCT 141 85.8 Brinkhof et al. [32]
Rev CCTGATTCACACGGCGTAG
36 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

450

579.3 (408.0–636.9)
857.1 (710.2–886.6)
30.4 (27.6–41.4)
33.1 (30.4–77.3)
400

350

16 weeks
Cortisol (nmol/l)

300
250

27.6 (27.6–30.4)
33.1 (30.4–63.5)
200

14 weeks
150

∗ ∗ ∗ ∗

nd
nd
100
∗ ∗ ∗ ∗ ∗ ∗
∗ ∗
50

390.1 (315.9–405.4)
484.0 (316.6–624.4)
30.4 (27.6–41.4)
33.1 (30.4–35.1)
0
0 1 2 3 4 5 6 7 8 10 12 14 16
Time (weeks)

12 weeks
Fig. 1. Changes in serum cortisol concentrations. Serum cortisol
concentrations before (䊉) and after ACTH stimulation () during

33.1 (27.6–126.9)
trilostane administration. The post-stimulation serum cortisol con-

30.4 (27.6–82.8)
centrations decrease significantly during trilostane administration. *,
P < 0.01 vs. at time 0.

10 weeks

nd
nd
ber of cells per field of ZF and ZR, and the expression
of POMC and ACTH receptor mRNA were analyzed

204.2 (42.5–1623.0)
670.7 (238.5–758.6)
27.6 (24.8–186.5)
29.0 (24.8–65.5)
by one-way ANOVA, and then differences among the
means were analyzed using Tukey–Kramer’s post hoc
tests. Statistical analyses were performed using Excel

8 weeks
2003 with the add-in software Statcel 2. Differences were
considered significant when P < 0.05.

40.0 (27.6–143.5)
29.0 (21.3–38.6)
3. Results
6 weeks

nd
nd
Changes of serum cortisol and plasma ACTH concen-
Serum cortisol and plasma ACTH concentrations during trilostane administration.

trations during trilostane administration are summarized


543.4 (153.6–894.1)
38.9 (24.8–168.3)

138.2 (22.9–557.0)
28.8 (18.6–77.3)

in Table 2.
4 weeks

3.1. Effect on serum cortisol and plasma ACTH


Trilostane administration

concentrations
60.7 (27.6–173.8)
46.9 (27.6–85.5)

After the initiation of trilostane administration, post-


stimulation serum cortisol concentrations decreased
2 weeks

significantly compared to that observed before adminis-


nd
nd

tration (Fig. 1). The basal plasma ACTH concentrations


in the control group and the trilostane administra-
408.3 (324.0–510.4)
38.6 (27.6–107.6)

43.6 (22.4–75.5)

tion group before the administration of trilostane


Before trilostane

5.2 (3.3–10.1)
administration

were 5.2 (3.3–10.8) pmol/l and 5.2 (3.3–10.1) pmol/l,


respectively. The post-stimulation plasma ACTH con-
centrations in the control group and trilostane group
before the administration of trilostane were 40.9
post-ACTH concentration

post-CRH concentration

(19.1–66.9) pmol/l and 43.6 (22.4–75.5) pmol/l, respec-


Basal concentration

Basal concentration

tively. No significant difference was observed in the


nd = not determined.
Cortisol (nmol/l)

basal plasma ACTH concentrations and the plasma


ACTH (pmol/l)

ACTH concentrations after CRH-stimulation between


Table 2

the control and trilostane groups. After the initiation of


trilostane administration, both the basal plasma ACTH
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 37

1000 1.63 (1.52–1.88) g in the control, TS and TL group,


900 a, b, c, d respectively. The weights of the pituitary and bilateral
800 adrenal glands were significantly higher in the TS and TL
700 group than in the control group (pituitary, and right and

TH (pmol/l)

a
600 a a left adrenal gland, P < 0.01, respectively). The weight
500 of the pituitary of the TL group was also significantly
400 ∗ higher than that of the TS group (P < 0.05).
ACT

300
200 3.4. Pituitary and adrenal morphometry
100
0 The ACTH-positive cell area in the control, TS,
0 4 8 12 16
and TL group were 78.54 (58.37–228.95) ␮m2 , 93.66
Time (weeks) (72.03–253.22) ␮m2 , and 107.87 (78.87–289.77) ␮m2 ,
respectively. The ACTH-positive cell areas were signif-
Fig. 2. Changes in plasma ACTH concentrations. Plasma ACTH con-
centrations before (䊉) and after CRH-stimulation () during trilostane
icantly (P < 0.01) greater in the TS and TL group than in
administration. The basal and post-stimulation ACTH concentrations the control group. The ACTH-positive cell areas of the
increase significantly during trilostane administration. *, P < 0.05 vs. TL group were also significantly greater than that of the
before stimulation at time 0. a, P < 0.01 vs. after stimulation at time TS group (P < 0.01) (Figs. 5 and 6). The ACTH-positive
0. b, P < 0.01 vs. after stimulation at 4 weeks. c, P < 0.05 vs. after cell ratios were 22.2 (18.0–28.2)%, 28.7 (24.2–38.1)%,
stimulation at 8 weeks. d, P < 0.05 vs. after stimulation at 12 weeks.
and 32.7 (32.1–38.9)% in the control, TS, and TL group,
respectively. The ACTH-positive cell ratio of the TS and
concentrations and the plasma ACTH concentrations
TL group were significantly (P < 0.05) higher than that
after CRH-stimulation increased significantly compared
of the control group.
to those before trilostane administration (Fig. 2). Finally,
The MR images prior to trilostane administration
the plasma ACTH concentrations after CRH-stimulation
revealed no abnormalities in size and shape of the
at 16 weeks increased significantly compared to that at
bilateral adrenal glands in all dogs. The width of
12 weeks (P < 0.05).
ZG in the control, TS, and TL group were 0.132
(0.100–0.234) mm, 0.146 (0.112–0.329) mm, and 0.148
3.2. P/B ratio (0.108–0.305) mm, respectively. The width of ZF in the
control in the control, TS and TL group were 0.786
Before the administration of trilostane, the P/B ratio (0.560–1.719) mm, 1.839 (0.726–3.229) mm, and 1.607
was 0.269 (0.256–0.288; ×10−2 mm−1 ) in the trilostane (1.147–2.863) mm, respectively. The width of ZR in the
administration group. During the administration of control in the control, TS, and TL group were 0.403
trilostane, the P/B ratios showed a significant increase (0.257–0.888) mm, 0.858 (0.288–1.986) mm, and 1.101
after trilostane administration (Figs. 3 and 4). The (0.688–1.739) mm, respectively. The width of ZG, ZF,
P/B ratios at every 2 weeks were 0.288 (0.261–0.330; and ZR were significantly (P < 0.01, respectively) greater
2 weeks), 0.314 (0.264–0.332; 4 weeks), 0.329 in the TS and TL group than in the control group. The
(0.282–0.354; 6 weeks), 0.336 (0.294–0.368; 8 weeks), number of cells per field of ZF in the control, TS, and TL
0.342 (0.321–0.353; 10 weeks), 0.331 (0.321–0.355; group were 158.0 (114–194) cells, 110 (72–180) cells,
12 weeks), 0.348 (0.321–0.356; 14 weeks), and 0.350 and 107 (94–141) cells, respectively. The number of
(0.323–0.389; 16 weeks), respectively. Finally, the cells per field of ZR in the control, TS, and TL group
increase of the P/B ratios at 16 weeks increased signifi- were 260 (188–394) cells, 209 (151–289) cells, and 204
cantly compared to that at 8 weeks (P < 0.05). (160–235) cells, respectively. The number of cells of
ZF and ZR were significantly lower (P < 0.01, respec-
3.3. Tissue weights tively) in the TS and TL group than in the control group
(Fig. 7).
The weight of the pituitary was 55 (50–70) mg,
90 (70–120) mg, and 120 (110–130) mg in the con- 3.5. Levels of POMC and ACTH receptor mRNA
trol, TS, and TL group, respectively. The weight of expression
the right adrenal gland was 0.65 (0.41–0.83) g, 1.49
(1.24–3.14) g, and 1.92 (1.91–1.96) g, and left adrenal The levels of POMC mRNA expression in the TS
gland was 0.60 (0.42–0.84) g, 1.39 (1.11–2.88) g, and and TL group were about 3.3-fold (median 3.48, range
38 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

Fig. 3. Changes in pituitary gland observed by Gd-T1-weighted transverse images. Gd-T1-weighted transverse images of a dog in the TS group
before (A) and after 8 weeks of trilostane administration (B). Before trilostane administration, pituitary gland height is 4.4 mm and P/B ratio is
0.273. After 8 weeks of trilostane administration, pituitary gland height is 5.6 mm and P/B ratio is 0.339. Gd-T1-weighted transverse images of a
dog in the TL group before (C) and after 16 weeks of trilostane administration (D). Before trilostane administration, pituitary gland height is 4.7 mm
and P/B ratio is 0.261. After 16 weeks of trilostane administration, pituitary gland height is 6.8 mm and P/B ratio is 0.389. Bar = 10 mm.

0.39 a, b, c, 2.09–4.45) and 4.0-fold (median 4.53, range 2.63–4.85)


e, f greater than that in the control group (P < 0.01). The
0.37
levels of ACTH-R mRNA expression in the TS and
P/B ratio x 10-2 (mm-1)

a, b, c
aa, b
b, c a, b
0.35 a, b b, d TL group were about 2.5-fold (median 2.14, range
a, b 1.52–4.35) and 2.7-fold (median 2.76, range 2.40–3.18)
0.33
a greater than that in the control group (P < 0.05)
0.31 (Fig. 8).
0.29
4. Discussion
0.27
27

0.25 Cortisol secretion from the adrenal cortex is regulated


0 2 4 6 8 10 12 14 16
Time (weeks) by the HPA axis, and a negative feedback mecha-
nism functions to control cortisol concentrations in vivo
Fig. 4. Changes in P/B ratios during the trilostane administration. P/B [34,35]. Several reports have shown that the inhibition
ratio of more than 0.31 × 10−2 mm−1 is considered to be pituitary
of cortisol secretion in dogs with Cushing’s disease
enlarged. a, P < 0.01 vs. time 0. b, P < 0.01 vs. 2 weeks. c, P < 0.01 vs.
4 weeks. d, P < 0.05 vs. 4 weeks. e, P < 0.01 vs. 6 weeks. f, P < 0.05 vs. by mitotane treatment suppressed negative feedback,
8 weeks. and may accelerate the growth of pituitary adenoma
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 39

Fig. 5. Immunohistochemical staining of the anterior pituitary gland using anti-ACTH antibody. The ACTH-positive cells in the control (A, B), TS
(C, D), and TL group (E, F). Bar = 100 ␮m (A, C, E) and 50 ␮m (B, D, F).

Fig. 6. Comparison of the ACTH-positive cell area and cell ratio in the anterior pituitary gland. *, P < 0.05 vs. control group (CT). **, P < 0.01 vs.
CT. #, P < 0.01 vs. TS group.
40 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

Fig. 7. Comparison of the width and numbers of cells in the adrenal cortex. Hematoxylin and eosin staining of a section from the adrenal gland (A).
Left: TS group; right: control group. The adrenal cortex is enlarged in the TS and TL group. Bar = 1.0 mm. The width of the zona glomerulosa (ZG),
zona fasciculata (ZF), and zona reticularis (ZR) (B). The number of cells per field of ZF and ZR (C). **, P < 0.01 vs. control group. #, P < 0.01 vs.
TS group.

Fig. 8. Comparison of the relative levels of POMC mRNA and ACTH receptor mRNA expression. The relative levels of proopiomelanocortin
(POMC) mRNA expression in the pituitary gland and ACTH receptor (ACTH-R) mRNA expression in the adrenal gland. *, P < 0.05 vs. control
group (CT). **, P < 0.01 vs. CT.
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 41

[2,36–38]. In humans, a number of reports demonstrated size of the pituitary in dogs [28], revealed that the P/B
the occurrence of Nelson’s syndrome, in which pituitary ratio was greater than 0.31, which was judged to indicate
adenoma growth was accelerated, resulting from the dis- an enlarged pituitary. Immunohistochemical analysis
appearance of cortisol secretion and negative feedback revealed that there was a significant change in the
that occurred after bilateral adrenalectomy in Cushing’s cytoplasmic areas of corticotrophs and the ratio of corti-
disease patients [39–46]. Therefore, Nelson’s syndrome cotrophs to all cells in the anterior lobe of the pituitary in
was re-evaluated recently [47–49], and this phenomenon the TS and TL group compared with the control group,
caused by trilostane and mitotane in humans has been and, furthermore, the cytoplasmic areas of corticotrophs
termed “chemical Nelson’s syndrome” [50]. We have in the TL group were greater than that of the TS group.
experienced some PDH dogs in which large pituitary In addition, the weight of the pituitary after the end of
adenomas were found after trilostane treatment. These trilostane administration showed a significant increase in
dogs were diagnosed with PDH without diagnostic imag- the TS and TL groups. These results suggest that hyper-
ing, such as CT or MRI, and treated by trilostane. trophy and hyperplasia of the corticotroph may account
However, several months later, the doses of trilostane for the enlargement of the pituitary, and that the longer
needed increased or neurological signs were observed. cortisol secretion is suppressed, the size of the pituitary
However, assessing the potential risk in PDH dogs may become larger with increasing the plasma ACTH
treated with trilostane is difficult. Whether changes sim- concentrations.
ilar to those observed in Nelson’s syndrome appear in Monoclonal expansion of the corticotroph may
dogs with Cushing’s disease is controversial, and no induce adenomatous formation [51], but the hyperplasia
studies have investigated the effect of cortisol inhibi- of the corticotrophs observed in this study was not mon-
tion by trilostane on the normal pituitary corticotroph. oclonal. In humans and rats, adrenalectomy and chronic
In this study, we investigated the effect of trilostane on increased secretion of CRH has been shown to result
the pituitary–adrenal axis by inhibiting adrenal cortex in corticotroph hyperplasia and adenomatous formation
function through the daily administration of trilostane in [52–54]. However, adenomatous formation of the cor-
healthy Beagle dogs. ticotroph in dogs has not been examined. In this study,
The dose of trilostane in the trilostane administration since cortisol secretion was inhibited, the production and
group was determined based on reports of the dura- secretion of ACTH were increased through reduced neg-
tion of action and its use in PDH dogs [7–12,18–21]. ative feedback, although adenomatous formation was not
In addition, the doses tended to increase as the therapy observed in the trilostane administration group. Never-
continued [8,9,11,12,21]. Since the duration of trilostane theless, hypertrophy and hyperplasia of the corticotroph
is less than 20 h [14], we attempted to suppress cor- and enlargement of the pituitary were observed, probably
tisol secretion of the adrenal cortex continuously by resulted in reduced negative feedback. Although CRH,
the twice daily administration in this experiment. The- which stimulates the production and secretion of ACTH
ACTH stimulation test revealed that cortisol secretion from the corticotrophs, was not measured in the present
was sufficiently suppressed during trilostane adminis- experiment, an increase in ACTH concentrations and
tration, and adverse effects such as lethargy, anorexia, POMC mRNA expression suggests that CRH secretion
vomiting and diarrhea were not observed. With regard to from the hypothalamus was increased in the trilostane
ACTH secretion from the pituitary, the CRH-stimulation administration group. In humans with Cushing’s dis-
test showed that ACTH concentrations sequentially ease, CRH secretion from the hypothalamus is believed
increased remarkably during the trilostane adminis- to increase after the adrenalectomy [49]. In regards
tration, which suggests an increased production and to dogs with Cushing’s disease, no study has exam-
secretion of ACTH at the pituitary corticotroph. This ined whether CRH concentrations are increased by the
may be attributable to the suppressed negative feed- suppressed secretion of cortisol. However, if CRH con-
back due to the decreased cortisol secretion from the centrations are also increased by the suppressed secretion
adrenal cortex induced by trilostane administration. of cortisol in dogs with Cushing’s disease, the risk of
Indeed, increased plasma ACTH concentrations have occurrence of chemical Nelson’s syndrome may become
also been observed in PDH dogs treated with trilostane high.
[18,19]. After the end of trilostane administration, the level
MRI during the trilostane administration revealed of ACTH-R mRNA expression was increased, and the
that the pituitary increased in size during the period of size of the bilateral adrenal glands was increased in
trilostane administration. After 4 weeks of treatment, the trilostane administration group. The ACTH-R is
evaluation using the P/B ratio, a common marker for the observed principally in cells of the adrenal cortex and
42 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

signals cells in the ZF to synthesize and secrete cortisol. cortisol may occur in dogs with Cushing’s disease,
In many species, ACTH has been reported to up-regulate and ACTH secretion may be suppressed by negative
the level of ACTH-R mRNA and the number of ACTH feedback by cortisol in the corticotroph adenoma as
binding sites in adrenocortical cells [55–58]. Moreover, well.
in murine adrenal glands, a high level of in vivo ACTH is Since changes in the HPA axis during the trilostane
a major factor up-regulating ACTH-R gene expression, administration were examined in this study, it remains to
while low circulating ACTH may not be a substantial be clarified how enlargement of the pituitary and hyper-
factor for ACTH-R gene expression [58]. With regard to function of the pituitary corticotroph by suppression of
ACTH-R mRNA expression, the remarkably increased cortisol secretion would change after cessation of the
ACTH may result in an up-regulation of ACTH-R trilostane administration. Taking into account the phar-
mRNA expression in the trilostane administration group. macology of trilostane, a competitive enzyme inhibitor,
Histological analysis using HE staining revealed that these changes may return after cessation of administra-
each zona of the adrenal cortex was thickened in the TS tion. Clinically, however, it is difficult to end trilostane
and TL groups. In addition, an enlargement of cytoplas- administration in PDH dogs, and life-time treatment is
mic areas and a decrease in cell number per area were thought to be necessary.
observed in ZF and ZR, where cortisol was produced In conclusion, due to the suppression of cortisol secre-
and secreted, suggesting the appearance of hyperplastic tion by trilostane, reduced negative feedback induced
changes. Two potential causes for these changes in the hyperfunction of the pituitary corticotroph, which caused
adrenal cortex were considered. First, since the ACTH- an enlarged pituitary due to corticotroph hypertrophy
R mRNA expression and serum ACTH concentrations and hyperplasia in healthy dogs. Moreover, the longer
were raised, stimulation of the adrenal cortex by ACTH cortisol secretion was suppressed, the more corticotroph
was increased. This may lead to hypertrophy and hyper- hyperfunction increased and the larger the pituitary
plasia in the cells of the adrenal cortex. Second, the became. These changes may help to understand whether
mechanism of action of trilostane may be involved. With changes similar to Nelson’s syndrome appear in dogs
inhibition of steroid synthesis in the adrenal cortex, pre- with Cushing’s disease.
cursors of steroids may accumulate in the cells, which
may lead to hypertrophy in the adrenal cortex [59,60].
Although the precursors of steroids were not measured Acknowledgment
in this study, trilostane administration has been reported
to increase precursors of steroids in dogs, humans, and The authors thank IDEXX Laboratories, KK (Tokyo,
guinea pigs [13,19,61]. Despite the reports that necro- Japan) for support in hormone determination.
sis was observed in the adrenal cortex of the PDH dogs
treated with trilostane [59,62], no such changes were
References
observed in the adrenal cortex in the TS and TL groups
in this study. [1] Guptill L, Scott-Moncrieff JC, Widmer WR. Diagnosis of canine
It is not possible to conclude, based on the present hyperadrenocorticism. Vet Clin North Am Small Anim Pract
experiments using healthy dogs, that similar changes 1997;27:215–35.
observed in the pituitary corticotroph in the trilostane [2] Feldman EC, Nelson RW. Canine hyperadrenocorticism (Cush-
group will be observed in dogs with Cushing’s dis- ing’s syndrome). In: Feldman EC, Nelson RW, editors. Canine
and feline endocrinology and reproduction. Philadelphia: WB
ease. However, the negative feedback of cortisol on Saunders Co.; 2004. p. 252–357.
ACTH secretion from the pituitary is presumed to [3] Hanson JM, van’t HM, Voorhout G, Teske E, Kooistra HS, Meij
function even in dogs with Cushing’s disease. The BP. Efficacy of transsphenoidal hypophysectomy in treatment of
dexamethasone suppression test revealed that cortisol dogs with pituitary-dependent hyperadrenocorticism. J Vet Intern
secretion is not suppressed by low-dose dexamethasone Med 2005;19:687–94.
[4] Hanson JM, Teske E, Voorhout G, Galac S, Kooistra HS, Meij BP.
(0.01 mg/kg), but is suppressed by high-dose dexam- Prognostic factors for outcome after transsphenoidal hypophy-
ethasone (0.1 mg/kg) due to negative feedback in most sectomy in dogs with pituitary-dependent hyperadrenocorticism.
dogs with Cushing’s disease [63–65]. Moreover, a cor- J Neurosurg 2007;107:830–40.
relation was found between the size of the pituitary [5] Hara Y, Tagawa M, Masuda H, Sako T, Koyama H, Orima
gland and resistance to dexamethasone, and the dex- H, et al. Transsphenoidal hypophysectomy for four dogs with
pituitary ACTH-producing adenoma. J Vet Med Sci 2003;65:
amethasone resistance was associated with high plasma 801–4.
ACTH concentrations in dogs with Cushing’s disease [6] Braddock JA. Medical treatment of hyperadrenocorticism in the
[28]. Considering these findings, negative feedback by dog. Aust Vet J 2003;81:31–3.
T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44 43

[7] Bell R, Neiger R, McGrotty Y, Ramsey IK. Study of [24] Reusch CE, Steffen T, Hoerauf A. The efficacy of l-deprenyl in
the effects of once daily doses of trilostane on corti- dogs with pituitary-dependent hyperadrenocorticism. J Vet Intern
sol concentrations and responsiveness to adrenocorticotrophic Med 1999;13:291–301.
hormone in hyperadrenocorticoid dogs. Vet Rec 2006;159: [25] Fracassi F, Gandini G, Diana A, Preziosi R, Ingh TS, Famigli-
277–81. Bergamini P, et al. Acromegaly due to a somatroph adenoma in a
[8] Alenza DP, Arenas C, Lopez ML, Melian C. Long-term efficacy dog. Domest Anim Endocrinol 2007;32:43–54.
of trilostane administered twice daily in dogs with pituitary- [26] Granger N, de Fornel P, Devauchelle P, Segond S, Delisle
dependent hyperadrenocorticism. J Am Anim Hosp Assoc F, Rosenberg D. Plasma pro-opiomelanocortin, pro-
2006;42:269–76. adrenocorticotropin hormone, and pituitary adenoma size
[9] Braddock JA, Church DB, Robertson ID, Watson AD. Trilostane in dogs with Cushing’s disease. J Vet Intern Med 2005;19:
treatment in dogs with pituitary-dependent hyperadrenocorticism. 23–8.
Aust Vet J 2003;81:600–7. [27] Scott-Moncrieff JC, Koshko MA, Brown JA, Hill K, Refsal KR.
[10] Barker EN, Campbell S, Tebb AJ, Neiger R, Herrtage ME, Reid Validation of a chemiluminescent enzyme immunometric assay
SW, et al. A comparison of the survival times of dogs treated with for plasma adrenocorticotropic hormone in the dog. Vet Clin
mitotane or trilostane for pituitary-dependent hyperadrenocorti- Pathol 2003;32:180–7.
cism. J Vet Intern Med 2005;19:810–5. [28] Kooistra HS, Voorhout G, Mol JA, Rijnberk A. Correlation
[11] Reine NJ. Medical management of pituitary-dependent hypera- between impairment of glucocorticoid feedback and the size of
drenocorticism: mitotane versus trilostane. Clin Tech Small Anim the pituitary gland in dogs with pituitary-dependent hyperadreno-
Pract 2007;22:18–25. corticism. J Endocrinol 1997;152:387–94.
[12] Neiger R, Ramsey I, O’Connor J, Hurley KJ, Mooney CT. [29] Meij BP, Mol JA, van den Ingh TS, Bevers MM, Hazewinkel
Trilostane treatment of 78 dogs with pituitary-dependent hyper- HA, Rijnberk A. Assessment of pituitary function after transsphe-
adrenocorticism. Vet Rec 2002;150:799–804. noidal hypophysectomy in beagle dogs. Domest Anim Endocrinol
[13] Potts GO, Creange JE, Hardomg HR, Schane HP. Trilostane, 1997;14:81–97.
an orally active inhibitor of steroid biosynthesis. Steroids [30] Taoda T, Hara Y, Takekoshi S, Itoh J, Teramoto A, Osamura RY,
1978;32:257–67. et al. Effect of mitotane on pituitary corticotrophs in clinically
[14] Plumb DC. Trilostane. In: Plumb DC, editor. Veterinary drug normal dogs. Am J Vet Res 2006;67:1385–94.
handbook. 5th ed. Ames: Iowa State University Press; 2005. p. [31] Sieber-Ruckstuhl NS, Meli ML, Boretti FS, Gönczi E, Lutz H,
778–9. Reusch CE. Quantitative real-time PCR for the measurement
[15] Peterson ME. Medical treatment of canine pituitary-dependent of 11beta-HSD1 and 11beta-HSD2 mRNA levels in tissues of
hyperadrenocorticism (Cushing’s disease). Vet Clin North Am healthy dogs. Horm Metab Res 2007;39:548–54.
Small Anim Pract 2001;31:1005–14. [32] Brinkhof B, Spee B, Rothuizen J, Penning LC. Development and
[16] Kintzer PP, Peterson ME. Mitotane (o,p’-DDD) treatment of 200 evaluation of canine reference genes for accurate quantification
dogs with pituitary-dependent hyperadrenocorticism. J Vet Intern of gene expression. Anal Biochem 2006;356:36–43.
Med 1991;5:182–90. [33] Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De
[17] den Hertog E, Braakman JC, Teske E, Kooistra HS, Rijnberk Paepe A, et al. Accurate normalization of real-time quantitative
A. Results of non-selective adrenocorticolysis by o,p’-DDD in RT-PCR data by geometric averaging of multiple internal control
129 dogs with pituitary-dependent hyperadrenocorticism. Vet Rec genes. Genome Biol 2002;18(3) [RESEARCH0034].
1999;144:12–7. [34] Keller-Wood ME, Dallman MF. Corticosteroid inhibition of
[18] Witt AL, Neiger R. Adrenocorticotropic hormone levels in ACTH secretion. Endocr Rev 1984;5:1–24.
dogs with pituitary-dependent hyperadrenocorticism following [35] Bertoy EH, Feldman EC, Nelson RW, Dublin AB, Reid
trilostane therapy. Vet Rec 2004;154:399–400. MH, Feldman MS. One-year follow-up evaluation of magnetic
[19] Sieber-Ruckstuhl NS, Boretti FS, Wenger M, Maser-Gluth C, resonance imaging of the brain in dogs with pituitary-
Reusch CE. Cortisol, aldosterone, cortisol precursor, androgen dependent hyperadrenocorticism. J Am Vet Med Assoc 1996;208:
and endogenous ACTH concentrations in dogs with pituitary- 1268–73.
dependant hyperadrenocorticism treated with trilostane. Domest [36] Nelson RW, Feldman EC, Shinsako J. Effect of o,p’DDD therapy
Anim Endocrinol 2006;31:63–75. on endogenous ACTH concentrations in dogs with hypophysis-
[20] Wenger M, Sieber-Ruckstuhl NS, Müller C, Reusch CE. Effect dependent hyperadrenocorticism. Am J Vet Res 1985;46:1534–7.
of trilostane on serum concentrations of aldosterone, cortisol, [37] Peterson ME, Orth DN, Halmi NS, Zielinski AC, Davis DR,
and potassium in dogs with pituitary-dependent hyperadrenocor- Chavez FT, et al. Plasma immunoreactive proopiomelanocortin
ticism. Am J Vet Res 2004;65:1245–50. peptides and cortisol in normal dogs and dogs with Addi-
[21] Ruckstuhl NS, Nett CS, Reusch CE. Results of clinical exam- son’s disease and Cushing’s syndrome: basal concentrations.
inations, laboratory tests, and ultrasonography in dogs with Endocrinology 1986;119:720–30.
pituitary-dependent hyperadrenocorticism treated with trilostane. [38] Sarfaty D, Carrillo JM, Peterson ME. Neurologic, endocrino-
Am J Vet Res 2002;63:506–12. logic, and pathologic findings associated with large pituitary
[22] Hansen BL, Kemppainen RJ, MacDonald JM. Synthetic ACTH tumors in dogs: eight cases (1976–1984). J Am Vet Med Assoc
(cosyntropin) stimulation tests in normal dogs: comparison of 1988;193:854–6.
intravenous and intramuscular administration. J Am Anim Hosp [39] Nelson DH, Meakin JW, Dealy Jr JB, Matson DD, Emerson Jr
Assoc 1994;30:38–41. K, Thorn GW. ACTH-producing tumor of the pituitary gland. N
[23] Meij BP, Mol JA, Bevers MM, Rijnberk A. Residual pitu- Engl J Med 1958;259:161–4.
itary function after transsphenoidal hypophysectomy in dogs [40] Nelson DH, Meakin JW, Thorn GW. ACTH-producing pituitary
with pituitary-dependent hyperadrenocorticism. J Endocrinol tumors following adrenalectomy for Cushing’s syndrome. Ann
1997;155:531–9. Intern Med 1960;52:560–9.
44 T. Teshima et al. / Domestic Animal Endocrinology 36 (2009) 32–44

[41] Kelly WF, MacFarlane IA, Longson D, Davies D, Sutcliffe factor increases pituitary corticotroph number. Endocrinology
H. Cushing’s disease treated by total adrenalectomy: long-term 1987;120:381–8.
observations of 43 patients. Q J Med 1983;52:224–31. [55] Mountjoy KG, Bird IM, Rainey WE, Cone RD. ACTH induces
[42] Barnett AH, Livesey JH, Friday K, Donald RA, Espiner EA. up-regulation of ACTH receptor mRNA in mouse and human
Comparison of preoperative and postoperative ACTH concen- adrenocortical cell lines. Mol Cell Endocrinol 1994;99:R17–20.
trations after bilateral adrenalectomy in Cushing’s disease. Clin [56] Penhoat A, Jaillard C, Saez JM. Corticotropin positively regulates
Endocrinol 1983;18:301–5. its own receptors and cAMP response in cultured bovine adrenal
[43] Thomas JP, Hall R. Medical management of pituitary disease. cells. Proc Natl Acad Sci U S A 1989;86:4978–81.
Clin Endocrinol Metab 1983;12:771–88. [57] Lebrethon MC, Naville D, Begeot M, Saez JM. Regulation of
[44] Kasperlik-Załuska AA, Nielubowicz J, Wisławski J, Hartwig W, corticotropin receptor number and messenger RNA in cultured
Załuska J, Jeske W, et al. Nelson’s syndrome: incidence and human adrenocortical cells by corticotropin and angiotensin II. J
prognosis. Clin Endocrinol 1983;19:693–8. Clin Invest 1994;93:1828–33.
[45] Thomas Jr CG, Smith AT, Benson M, Griffith J. Nelson’s [58] Kijima H, Kubo M, Shimizu C, Ishizuka T, Takano K, Nagai S,
syndrome after Cushing’s disease in childhood: a continuing prob- et al. Effects of hypophysectomy and in vivo administration of
lem. Surgery 1984;96:1067–77. ACTH or dexamethasone on the level of ACTH receptor mRNA
[46] Pereira MA, Halpern A, Salgado LR, Mendonça BB, Nery M, in adrenal glands and adipose tissues of mice. Endocr Regul
Liberman B, et al. A study of patients with Nelson’s syndrome. 2004;38:87–95.
Clin Endocrinol 1998;49:533–9. [59] Reusch CE, Sieber-Ruckstuhl N, Wenger M, Lutz H, Perren A,
[47] Gil-Cárdenas A, Herrera MF, Díaz-Polanco A, Rios JM, Pantoja Pospischil A. Histological evaluation of the adrenal glands of
JP. Nelson’s syndrome after bilateral adrenalectomy for Cushing’s seven dogs with hyperadrenocorticism treated with trilostane. Vet
disease. Surgery 2007;141:147–52. Rec 2007;160:219–24.
[48] Assié G, Bahurel H, Coste J, Silvera S, Kujas M, Dugué MA, et al. [60] Mantis P, Lamb CR, Witt AL, Neiger R. Changes in ultra-
Corticotroph tumor progression after adrenalectomy in Cushing’s sonographic appearance of adrenal glands in dogs with
disease: a reappraisal of Nelson’s Syndrome. J Clin Endocrinol pituitary-dependent hyperadrenocorticism treated with trilostane.
Metab 2007;92:172–9. Vet Radiol Ultrasound 2003;44:682–5.
[49] Assié G, Bahurel H, Bertherat J, Kujas M, Legmann P, Bertagna [61] Lambert F, Corcelle-Cerf F, Lammerant J, Kolanowski J. On
X. The Nelson’s syndrome . . . revisited. Pituitary 2004;7: the specificity of the inhibitory effect of trilostane and aminog-
209–15. lutethimide on adrenocortical steroidogenesis in guinea pig. Mol
[50] Teramoto A. Diagnosis and treatment of Cushing’s disease. Jpn J Cell Endocrinol 1984;37:115–20.
Neurosurg 2001;10:86–91. [62] Chapman PS, Kelly DF, Archer J, Brockman DJ, Neiger R.
[51] Asa SL, Ezzat S. The cytogenesis and pathogenesis of pituitary Adrenal necrosis in a dog receiving trilostane for the treatment of
adenomas. Endocr Rev 1998;19:798–827. hyperadrenocorticism. J Small Anim Pract 2004;45:307–10.
[52] Suda T, Tomori N, Tozawa F, Mouri T, Demura H, [63] Mack RE, Feldman EC. Comparison of two low-dose dexam-
Shizume K. Effects of bilateral adrenalectomy on immunore- ethasone suppression protocols as screening and discrimination
active corticotropin-releasing factor in the rat median emi- tests in dogs with hyperadrenocorticism. J Am Vet Med Assoc
nence and intermediate-posterior pituitary. Endocrinology 1990;197:1603–6.
1983;113:1182–4. [64] Feldman EC. Evaluation of a six-hour combined dexamethasone
[53] Scheithauer BW, Kovacs K, Randall RV. The pituitary gland suppression/ACTH stimulation test in dogs with hyperadrenocor-
in untreated Addison’s disease. A histologic and immunocy- ticism. J Am Vet Med Assoc 1986;189:1562–6.
tologic study of 18 adenohypophyses. Arch Pathol Lab Med [65] Feldman EC, Nelson RW, Feldman MS. Use of low- and high-
1983;107:484–7. dose dexamethasone tests for distinguishing pituitary-dependent
[54] Gertz BJ, Contreras LN, McComb DJ, Kovacs K, Tyrrell JB, from adrenal tumor hyperadrenocorticism in dogs. J Am Vet Med
Dallman MF. Chronic administration of corticotropin-releasing Assoc 1996;209:772–5.

You might also like