You are on page 1of 12

Domestic Animal Endocrinology 59 (2017) 11–22

Contents lists available at ScienceDirect

Domestic Animal Endocrinology


journal homepage: www.domesticanimalendo.com

Glucocorticoid metabolism in equine follicles and oocytes


D. Scarlet a, *, N. Ille b, R. Ertl c, B.G. Alves d, G.D.A. Gastal d, S.O. Paiva d,
M.O. Gastal d, E.L. Gastal d, C. Aurich b
a
Division of Obstetrics, Gynecology and Andrology, Department for Small Animals and Horses, University of Veterinary Medicine
Vienna, 1210 Vienna, Austria
b
Center for Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, University of Veterinary
Medicine Vienna, 1210 Vienna, Austria
c
Vetcore Facility, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
d
Department of Animal Science, Food and Nutrition, Southern Illinois University, Carbondale, IL 62901, USA

a r t i c l e i n f o a b s t r a c t

Article history: The objective of this study was to determine whether (1) systemic and intrafollicular
Received 12 July 2016 cortisol concentrations in horses are directly related and (2) supraphysiological levels of
Received in revised form 30 September 2016 glucocorticoids affect in vitro maturation (IVM) rates of oocytes. Specifically, we studied
Accepted 23 October 2016
the (1) changes in the intrafollicular cortisol and progesterone in context with granulosa
cell gene expression during maturation of equine follicles (from 5–9 mm, 10–14 mm, 15–19
Keywords:
mm, 20–24 mm, and 25 mm in diameter) and (2) effects of cortisol supplementation on
Cortisol
IVM rates and gene expression of equine cumulus–oocyte complexes (COCs). For these
Horse
Ovary purposes, follicular fluid, granulosa cells, and COCs were collected from 12 mares (mean
Follicle age 8.6  0.5 yr) by transvaginal aspiration. Cortisol and progesterone concentrations in
Oocyte follicular fluid from follicles 25 mm were greater (P < 0.05) than in all other follicle
classes and were positively correlated (r ¼ 0.8; P < 0.001). Plasma concentrations of
cortisol and progesterone did not differ before and after follicle aspiration (P > 0.05). In
granulosa cells, gene expression of NR3C1, HSD11B1, HSD11B2, and CYP21A2 did not differ
(P > 0.05) among different follicle classes. Maturation rates were similar (P > 0.05) among
groups, regardless of the cortisol concentration in the IVM medium. In cumulus cells,
messenger RNA expression of genes involved in glucocorticoid mechanism and apoptosis
was either increased (NR3C1 and BCL2) or decreased (HSD11B2) by treatment (P < 0.01). In
oocytes, gene expression of maturation markers (BMP15 and GDF9) was affected (P <
0.001) by cortisol treatment. This study demonstrates the involvement of glucocorticoids
in follicle and oocyte maturation and cortisol modulation by HSD11B2 in equine COCs. Our
data provide further information for understanding the normal ovarian endocrine physi-
ology which might in turn also help improve equine assisted reproduction techniques.
Ó 2016 Elsevier Inc. All rights reserved.

1. Introduction reproductive function in a variety of species [1,2]. How-


ever, glucocorticoids are also involved in tissue differ-
Glucocorticoids are mediators of a systemic stress entiation and maturation [3]. A well-regulated balance
response. Increased glucocorticoid release and synthesis between beneficial and detrimental effects of glucocor-
in response to acute or chronic stress impairs ticoid hormones might therefore be of utmost impor-
tance for reproductive tissue development as well as
maintenance of its function.
* Corresponding author. Tel.: þ43-1-250776426; fax: þ43-1- The ovary is a target tissue for glucocorticoids in many
250775491.
species, including rats [4], mice [5], pigs [6,7], humans [8],
E-mail address: dragos.scarlet@vetmeduni.ac.at (D. Scarlet).

0739-7240/$ – see front matter Ó 2016 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.domaniend.2016.10.004
12 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22

cattle [9], sheep [10], and horses (own unpublished results). oocytes to high concentrations of cortisol during IVM im-
Intracellular glucocorticoid activity is regulated by 11b- pairs maturation and the expression of maturation-
hydroxysteroid dehydrogenase (HSD11B) enzyme types 1 associated genes.
and 2. Although HSD11B1 has a bidirectional activity in
activating and deactivating conversion of cortisone to 2. Materials and methods
cortisol, HSD11B2 only inactivates conversion of cortisol to
cortisone [11]. All experimental procedures were performed according
Glucocorticoid effects on oocyte maturation are still to the United States Government Principles for
controversial. This may be because of the fact that systemic the Utilization and Care of Vertebrate Animals Used
concentration and intracellular or intracompartmental ac- in Testing, Research, and Training (https://grants.nih.gov/
tivity of glucocorticoids are not necessarily correlated [11]. grants/olaw/references/phspol.htm#USGovPrinciples). The
In humans and cattle, high intrafollicular concentrations of research protocol (#14-055) was approved by the Institu-
the biological active metabolite cortisol were found around tional Animal Care and Use Committee of Southern Illinois
the time of the LH peak [12,13]. In these species, oocyte University. The study was conducted in the northern
in vitro maturation (IVM) and blastocyst development were hemisphere during the months of October and November.
improved by adding cortisol to the culture medium [14– Quarter Horse mares (n ¼ 12: BW ¼ 533  7.3 kg) were aged
16]. Conversely, IVM of pig [7] and lamb [10] oocytes was between 7 and 12 yr (mean age: 8.6  0.5 yr), were repro-
inhibited by glucocorticoids. ductively sound, and still undergoing normal estrous cycles.
Cortisol is not produced de novo by the ovaries [17] and This was proven by the fact that in all mares, at least 1
reaches ovarian follicles via blood circulation, where it is ovulation was inferred by the presence of 1 collapsed antral
largely bound to corticosteroid-binding globulin (CBG) [18]. follicle and subsequent development of a CL after the end of
Only the free, unbound cortisol fraction is considered bio- the study. Animals were kept on pasture under natural light
logically active [19]. Earlier, the increased expression of conditions and were fed additional orchardgrass–alfalfa
HSD11B1 in the granulosa cells from human pre-ovulatory mixed hay and had free access to mineral salt and fresh
follicles in response to the mid-cycle surge of gonadotro- water in a sheltered area. The ovaries and uteri of all mares
phins has been associated with metabolization of cortisol were scanned with a transrectal ultrasound scanner (Aloka
from cortisone [20]. As ovulation has been previously SSD-900; Aloka Co, Ltd, Wallingford, CT, USA) equipped
described as a controlled inflammatory reaction, it was with a multi-frequency 5- to 10-MHz linear array trans-
suggested that the local high levels of free cortisol act as ducer (Aloka UST-5821–7.5).
anti-inflammatory agents and thus limit tissue damage to
the follicle and the developing corpus luteum (CL) [19]. This 2.1. Animal preparation for transvaginal aspiration
suggestion is in line with the finding that HSD11B1 acts
rather as a dehydrogenase during porcine [21] and bovine For follicular ablation–aspiration or ovum pickup
[9] follicle maturation, inactivating cortisol to cortisone. (OPU) procedures, mares were restrained in a palpation
Cortisol is also present in follicular fluid (FF) of mares [22], chute to restrict movement. The mare’s tail was wrapped
but its intrafollicular dynamics during folliculogenesis has in a plastic palpation sleeve to minimize contamination.
not been investigated so far. Feces were removed from the rectum, the perineal area
Cortisol is not only involved in the stress response but is was washed with povidone–iodine solution and alcohol,
also an important mediator of other events such as and a Foley catheter was placed in the urinary bladder.
apoptosis [23]. Previous studies indicated that the pro- Approximately 10 min before the start of each procedure,
apoptotic gene BAX is upregulated in cumulus cells from rectal relaxation was induced with hyoscine N-butyl bro-
aged mares [24]. Also, oocytes originating from older mares mide (Buscopan; 0.2 mg/kg intravenously (i.v.); Sigma, St.
showed a lower expression of GDF9 and BMP15 [25], two Louis, MO, USA). Approximately 5 min before the start of
genes co-expressed during follicular development and each procedure, sedation and analgesia were induced with
associated with developmental competence of oocytes. a combination of detomidine (Dormosedan; 0.02–
However, the effects of cortisol supplementation on the 0.04 mg/kg (i.v.); Zoetis, Florham Park, NJ, USA) and
expression of genes involved in the apoptotic pathway or in butorphanol tartrate (Dolorex; 0.05 mg/kg (i.v.); Merck
oocyte maturation have not been studied so far. Animal Health, Summit, NJ, USA). Mares were treated with
The objective of this study was to determine whether antibiotics (Penicillin, Agri-Cillin; 12,000 IU/kg intramus-
(1) the equine ovary possesses mechanisms to modulate cularly; AgriLabs, St. Joseph, MO, USA) for 3 d after each
cortisol activity and (2) supraphysiological concentrations procedure.
of glucocorticoids affect IVM rates of oocytes. Specifically,
we studied the (1) changes in the intrafollicular concen- 2.2. Experiment 1: hormone concentrations in FF and plasma
trations of cortisol and progesterone in context with and gene expression of the granulosa cells
granulosa cell gene expression during maturation of equine
follicles (from 5–9 mm, 10–14 mm, 15–19 mm, 20–24 mm 2.2.1. Aspiration of FF and collection of granulosa cells
and 25 mm in diameter) and (2) effects of cortisol sup- A total of six mares were used for the collection of FF and
plementation on IVM rates and gene expression of equine granulosa cells. Antral follicles were measured and aspi-
cumulus–oocyte complexes (COCs). We hypothesized that rated with 18-G needles using transvaginal ultrasonogra-
systemic and intrafollicular cortisol concentrations in phy [26]. A different needle was used for each follicle class
horses are not directly related and that exposing equine from each animal to avoid contamination. Aspirated
D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22 13

follicles were grouped according to their diameter into the San Diego, CA, USA). Serial dilutions of equine plasma and
following classes: 5 to 9 mm, 10 to 14 mm, 15 to 19 mm, 20 FF showed good parallelism to the CBG standard curve
to 24 mm and 25 mm. In total, 4 to 7 FF samples from each (deviation within 20% of expected values). The mean re-
follicle class (mean: 5.4  0.5) were collected. To obtain as covery of standard added to equine plasma and FF sam-
many different follicle samples as possible from each ani- ples before assaying was 90% for both. The intra- and
mal, 1 to 2 follicle aspirations were performed per animal, inter-assay coefficients of variation were 10.2% and 8.1%,
allowing at least 2-d interval between procedures. During respectively, whereas the minimal detectable concentra-
aspiration of FF, follicles were flushed 2 to 3 times with tion was 3.12 mg/mL.
their own FF to retrieve the granulosa cells. Collected FF
was first filtered through a 70-mm cell strainer (Fisher Sci- 2.3. Experiment 2: effects of cortisol supplementation on IVM
entific, Pittsburgh, PA, USA) for isolation of granulosa cells. of equine oocytes
After centrifugation (1,200  g, 10 min), the supernatant
(pure FF) was removed and stored in 0.5-mL aliquots at 2.3.1. Follicle ablation
20 C until analysis. Granulosa cells were eluted from the At the beginning of the experiment, ablation of all fol-
cell strainer with PBS, and the fluid was analyzed in a Petri licles was performed to induce a new follicular wave and to
dish under a stereomicroscope to confirm the absence of avoid atretic and/or pre-ovulatory follicles. Afterward, 1
oocytes. After centrifugation (1,200  g, 10 min), the su- injection of prostaglandin F2a (Lutalyse; 0.01 mg/kg; Zoetis)
pernatant was discarded, and the granulosa cells were was given to regress any CL present. Mares (n ¼ 12) were
stored at 80 C. Before RNA extraction, granulosa cell then examined by transrectal ultrasound on a daily basis to
samples were washed twice with Buffer EL (79,217; Qiagen, establish the best time for OPU, according to the number of
Hilden, Germany) to remove remaining erythrocytes. In follicles 8 mm and 25 mm present in each mare. Two to
total, 4 to 7 granulosa cell samples (mean: 5.4  0.5) per 3 OPU procedures per mare were performed at 5- to 10-
follicle class were collected. d intervals. In 2 cases, luteal tissue was observed during
Blood samples were collected from the mare’s jugular the first OPU procedure; therefore, these mares received
vein into heparinized tubes at the beginning and at the end another injection of prostaglandin F2a (Lutalyse) after the
of the aspiration period. After centrifugation at 1,200  g procedure.
for 10 min, plasma was collected and stored at 20 C until
further analysis. 2.3.2. Ovum pickup
All follicles were punctured and vigorously flushed at
2.2.2. Hormone assays least 6 times with 30- to 100-mL Vigro complete embryo
Total cortisol concentrations in blood and FF were flush with BSA (Bioniche, Pullman, WA, USA) containing 2
determined with a commercial enzyme-linked immuno- IU/mL heparin (H3149; Sigma), pre-warmed at 38 C.
sorbent assay (DE1887; Demeditec, Kiel-Wellsee, Germany). Flushing fluid was collected in 50-mL Falcon tubes, filtered
According to the manufacturer, cross-reactivity is 100% with in 70-mm cell strainers (Fisher Scientific), and the
cortisol and <9% with progesterone. Serial dilutions of remainder in the filter was eluted with flushing medium in
equine plasma and FF showed very good parallelism to the Petri dishes to permit searching the COCs.
cortisol standard curve (deviation within 10% of expected
values). The mean recovery of the standard added to equine 2.3.3. In vitro maturation of COCs
plasma and FF before assaying was 83% and 95%, respec- After collecting the COCs (n ¼ 93), they were washed in
tively. Plasma and FF were not diluted before analysis. The pre-warmed HEPES-buffered (20 mM) synthetic oviduct
intra- and inter-assay coefficients of variation were 6.1% and fluid medium (Caisson Labs, North Logan, UT, USA) and kept
10.9%, respectively, whereas the sensitivity of the assay was at 38 C in air atmosphere for 2 to 4 h before maturation. All
0.02 ng/mL. COCs with <3 layers of cumulus cells, as well as expanded
Progesterone concentrations were determined with a COCs and damaged oocytes (in total, n ¼ 11), were excluded
commercial enzyme-linked immunosorbent assay (ADI- from culture. After arrival at the laboratory, COCs (n ¼ 82)
901–011; Enzo Life Sciences, Farmingdale, NY, USA) as from different mares were pooled and randomly assigned
described for equine plasma [27]. The antiserum cross- either to the control group (standard IVM medium) or 1 of
reacts 100% with progesterone and 100% with 5a-preg- the treatment groups and cultured for 28 h at 38.5 C. In total,
nane-3,20-dione. According to the manufacturer, 14 to 19 COC samples (mean: 16.4  0.9) were assigned to
cross-reactivity is 3.5% with 17-OH progesterone and <1% each treatment group. For the treatment groups, cortisol
with all other steroids tested. Serial dilutions of equine (H4001; Sigma) was added at the following concentrations:
plasma and FF showed good parallelism to the progester- 0.1, 1, 5, and 10 mg/mL. The COCs were grouped (2–3 per
one standard curve (deviation within 20% of expected group) and cultured in 500-mL droplets. The standard IVM
values). The mean recovery of the standard added to equine medium consisted of advanced DMEM-F12, 15% serum
plasma and FF samples before assaying was 96% and 74%, replacement, 50-ng/mL epidermal growth factor, 100-ng/mL
respectively. Plasma and FF were diluted 1:100 before IGF, and 0.1% penicillin–streptomycin (all from Life Tech-
analysis. The intra- and inter-assay coefficients of variation nologies, Carlsbad, CA, USA), to which 0.1 IU/mL each of FSH
were 5.3% and 5.5%, respectively, and the minimal detect- and LH (Sioux Biochemical, Sioux Center, IA, USA) were
able concentration was 0.081 ng/mL. added as previously described [28].
CBG concentrations were assessed using an equine- After IVM, COCs were denuded by incubating them for 5
specific commercial assay (MBS047353; MyBioSource, to 10 min in 0.5% hyaluronidase (H3884; Sigma)
14 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22

resuspended in DMEM-F12 at 38.5 C, followed by repeated recommended by the manufacturer, using the RNase-Free
pipetting. After cumulus cell removal, oocytes were stained DNase Set (Qiagen). The precipitated RNA was eluted in
in 1- to 2-mg/mL Hoechst 33,342 (Sigma) for 3 min at 37 C 11-mL elution buffer. Total RNA of 10 mL per sample was
to assess chromatin configuration under UV light (Fig. 1) as used as input for complementary DNA synthesis using the
previously described [29]. Oocytes were classified as SuperScript III First-Strand Synthesis System with random
metaphase II (MII), metaphase I (MI), germinal vesicle (GV), hexamer primers (Invitrogen, Carlsbad, CA, USA). Two
or degenerated. Cumulus cells were centrifuged for 5 min replicates were reverse transcribed for each sample and
at 1,500  g, followed by removal of the supernatant. Oo- then pooled for quantitative real-time PCR (qPCR) analysis.
cytes and cumulus cell samples were then stored at 80 C To monitor for residual DNA, additional control samples
until analysis. were included, in which no reverse transcriptase (RT)
enzyme was added. These “minus RT” controls were
2.4. Quantitative real-time PCR consistently negative in qPCR, indicating the absence of any
contaminating DNA.
Total RNA was extracted from single oocytes, cumulus Real-time PCR primers and hydrolysis probes for the
cells, and granulosa cells using the Arcturus PicoPure RNA equine target genes glucocorticoid receptor (NR3C1),
Isolation Kit (Life Technologies, Foster City, CA, USA). The HSD11B1, HSD11B2, 21-hydroxylase (CYP21A2)da gene
cells were lysed in 20-mL PicoPure extraction buffer and which is involved in cortisol biosynthesis, the anti-apoptotic
incubated at 42 C for 30 min. The subsequent RNA isolation gene B-cell lymphoma 2 (BCL2), the pro-apoptotic gene
was performed according to the manufacturer’s in- BCL2-associated X protein (BAX), and the maturation
structions (CapSure HS LCM Caps protocol) with two markers: bone morphogenetic protein 15 (BMP15) and
modifications: 20 mL of 70% ethanol (instead of 10 mL) was growth differentiation factor 9 (GDF9) were designed using
added to the cell extract for RNA precipitation and 100 mL of the PrimerExpress 2.0 software (Life Technologies). Hydro-
Wash Buffer 1 (instead of 40 mL) was used for washing after lysis probes were dual labeled with fluorescent dye FAM on
the DNase treatment. The on-column DNase digestion was the 50 end and the non-fluorescent quencher, Black Hole
performed after step (e) of the RNA isolation protocol, as Quencher 1 on the 30 end (FAM-BHQ1). All assays were

Fig. 1. Horse oocytes labeled with Hoechst 33342 to reveal chromatin configuration. (A) Germinal vesicle (GV). (B) Metaphase I (MI). (C) Metaphase II (MII), with
metaphase and polar body chromatin. (D) Degenerated; the unfocused area of fluorescence is the nucleus of a cumulus cell. 400.
D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22 15

Table 1
Details on quantitative real-time PCR assays.

Gene symbol NCBI/Ensembl accession number Oligo sequence 50 -30 Amplicon size (bp) PCR efficiency (%) R2 value

NR3C1 NM_001195191.1 F: GGCACCAGCAGAGGCAAT 71 101.8 0.998


XM_005599142.1 R: GAAAACTCCAAATCCGGCAAA
XM_005599145.1 P: TGTATACCACAGACCAAAGAACCTTTGAC
XM_005599143.1
XM_005599144.1
HSD11B1 XM_001490175.2 F: CTGGGATACTTGACACAAATGCA 71 90 0.997
R: CAGAGCTCCCCCTTTGATGA
P: CTCCAAAGGAAGAATGTGCCCTGGC
HSD11B2 NM_001081926.2 F: TCCAAGGGTCGCATTGTGA 61 100.6 0.999
R: CCAAGCATGGATATGGCATGT
P: TGTGGGCAGCCCGGCAGG
CYP21A2 ENSECAT00000017464 F: GGCATTCTTGCTTCACCACC 67 91 0.998
R: GCCCAGCTCACGATCCAA
P: ATTCAGCAGCGACTGCAGGACGAG
BCL2 XM_001490436.2 F: TTGGAAAGCCTACCACTAATTGC 74 92.6 0.998
R: CCGTGTTTATAGGCACAGGAGAT
P: CCCACCTGAGCGGCTCCACC
BAX XM_014729721.1 F: AGGATGCGTCCACCAAGAAG 80 93.2 0.994
XM_014729717.1 R: CCTCTGCAGCTCCATGTTACTG
P: CTCAAGCGCATCGGAGATGAGCTG
BMP15 XM_001496223.2 F: AGCCCTTGACCAATGTAGCAA 79 94.5 0.999
R: CGGTTGGATCTCAGAGGAAAGT
P: CAGAGGCCCCTGGCATATACAGACCC
GDF9 XM_001504427.2 F: AGGCCACCTCTACAACACTGTCC 113 99.5 0.999
R: CCAGGTTAAACAGCAGGTCCAC
P: CCCCCTGTGCCCAGCACAAGC
SNRPD3 XM_001489060.4 F: ACGCACCTATGTTAAAGAGCATG 120 99.4 0.996
XM_008511652.1 R: CACGTCCCATTCCACGTC
PSMB4 XM_001492317.4 F: CTTGGTGTAGCCTATGAAGCCC 82 93.1 0.991
XM_005610132.1 R: CCAGAATTTCTCGCAGCAGAG
XM_008515015.1
XM_005613704.1

Abbreviations: BAX, BCL2-associated X protein; BCL2, B-cell lymphoma 2; BMP15, bone morphogenetic protein 15; CYP21A2, 21-hydroxylase; GDF9, growth
differentiation factor 9; HSD11B1, 11beta-hydroxysteroid dehydrogenase type 1; HSD11B2, 11beta-hydroxysteroid dehydrogenase type 2; NCBI, National
Center for Biotechnology Information; NR3C1, glucocorticoid receptor; PSMB4, proteasome subunit beta type IV; SNRPD3, small nuclear ribonucleoprotein
D3 polypeptide.
R2: correlation coefficient of standard curve; F and R: forward and reverse primers; P: hydrolysis probe.

validated by the generation of standard curves to determine Real-time PCR was performed in 20-mL reaction mixes
the PCR reaction efficiencies (E), which were calculated ac- including 200 mM of each dNTP, 1 buffer B2 (Solis BioDyne,
cording to the formula E ¼ 101/s  1, where s is the slope of Tartu, Estonia), 3-mM MgCl2, 300 nM of each primer, 200-
the graph obtained by plotting the Cq value against the log10 nM probe, 50-nM ROX reference dye (Invitrogen), 1 unit
of the cDNA input. For Cq values generated at E < 1, HOT FIREPol DNA polymerase (Solis BioDyne), and 2-mL
efficiency-corrected Cq values were obtained from the term cDNA template. All samples were analyzed in duplicates on a
Cq  log10 ðE þ 1Þ=log10 ð2Þ [30]. Detailed assay data are lis- Viia7 Real-Time PCR System (Life Technologies) using the
ted in Table 1. following temperature profile: 95 C for 10 min, 45 cycles of

Fig. 2. (A) Cortisol, (B) progesterone, and (C) corticosteroid-binding globulin (CBG) concentrations in plasma of mares before and after ovum pickup (OPU) and in
follicular fluid (FF) from the following follicle classes: 5 to 9 mm, 10 to 14 mm, 15 to 19 mm, 20 to 24 mm and 25 mm. Values are means  SEM. Significant
differences in FF concentrations are marked with an asterisk. No significant differences were seen in plasma concentrations.
16 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22
D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22 17

95 C for 15 s, and 60 C for 1 min. For data normalization, the (r ¼ 0.56; P < 0.01; Fig. 3B) between plasma CBG and
geometric mean of the gene pair composed of SNRPD3 and progesterone.
PSMB4 was chosen based on its tested suitability for the adult Gene expression of NR3C1, HSD11B1, HSD11B2, and
equine ovary (own unpublished results). Real-time PCR for CYP21A2 in the granulosa cells was not different among fol-
SNRPD3 and PSMB4 was done with the fluorescent DNA dye licle classes (Fig. 4). Gene expression of NR3C1 was negatively
SYBR Green. Reaction conditions have been described [31]. correlated with CBG concentration (r ¼ 0.56; P < 0.05;
Only samples with mean Cq of SNRPD3 and PSMB4 <35 cycles Fig. 3C) in the respective FF, whereas gene expression of
and Cq <38 cycles for all other genes of interest were HSD11B1 and CYP21A2 was highly positively correlated be-
considered for further analysis. The mean Cq values of the tween each other (r ¼ 0.88; P < 0.01; Fig. 3D). All 4 genes
normalization factor were 30.6  0.1 for oocyte, 27.5  0.2 for analyzed were either expressed at a low level (HSD11B1 and
cumulus cell, and 27.2  0.7 for granulosa cell samples. Target HSD11B2) or not expressed at all (NR3C1 and CYP21A2) in the
gene expression levels were then normalized to the geo- granulosa cells originating from follicles 25 mm (Fig. 4).
metric mean of SNRPD3 and PSMB4 and relative expression
changes were calculated using the comparative 2DDCt 3.2. Experiment 2: effects of cortisol supplementation on IVM
method [32]. The qPCR data of this work comply with the of equine oocytes
Minimum Information for Publication of Quantitative Real-
Time PCR Experiments guidelines [33]. During 27 OPU procedures, 192 follicles between 8 and
26 mm were aspirated and 93 COCs were collected, resulting
2.5. Statistical analyses in a recovery rate of 48.4%. From the total of 93 COCs, 82 were
considered suitable according to previously mentioned
For statistical analyses, the computer software SPSS criteria and were submitted to culture. There were no sig-
version 24 (IBM-SPSS, Armonk, NY, USA) was used. Data nificant differences in the percentage of oocytes classified as
were tested for normal distribution by Kolmogorov–Smirnov MII, MI, GV, and degenerated among groups (Fig. 5), regard-
test. All data were normally distributed, and parametric tests less of the concentration of cortisol in the IVM medium.
were used throughout. For the analysis of hormone con- In oocytes, messenger RNA (mRNA) for HSD11B2, BAX,
centrations and gene expression in the granulosa cells, BMP15, and GDF9 was present, whereas NR3C1, HSD11B1, and
Pearson’s correlation and one-way ANOVA (general linear BCL2 were absent. Neither treatment nor maturation stage
model) with follicle class as between subject factor and had any significant effect on HSD11B2 (Fig. 6A) or BAX (Fig. 6B)
progesterone and cortisol as covariate were used. For the gene expression. In contrast, gene expression of BMP15 and
analysis of gene expression in oocytes and cumulus cells, GDF9 was significantly affected by maturation stage and
Pearson’s correlation and one-way ANOVA (general linear treatment group: it was lower in MII oocytes in comparison
model) with treatment as between subject factor and with non-matured oocytes (P < 0.01) and increased (P <
maturation status as covariate were used. Chi-square anal- 0.001) with rising cortisol concentrations in the IVM medium
ysis was used to analyze the IVM rates of oocytes per group. (Fig. 6C,D). Nevertheless, BMP15 and GDF9 levels were highly
Data are shown as mean  SEM. A P value <0.05 was positively correlated (r ¼ 0.9; P < 0.001; Fig. 3E) and also
considered statistically significant. correlated (r ¼ 0.4; P < 0.001; Fig. 3F,G) with BAX levels.
In cumulus cells, only NR3C1, HSD11B2, BCL2, and BAX
3. Results mRNA could be detected, whereas HSD11B1, BMP15, and
GDF9 transcripts were not expressed in these samples. All
3.1. Experiment 1: hormone concentrations in FF and plasma expressed genes with exception of BAX were affected by
and gene expression of the granulosa cells treatment (P < 0.01), but not by maturation stage (Fig. 7).
Levels of NR3C1 (P < 0.01) and BCL2 (P < 0.001) were
Cortisol (115.4  13.3 ng/mL) and progesterone (22.1 greater in all treatment groups than in the control
 3.1 ng/mL) concentrations in FF were greater (P < 0.05) in (Fig. 7A,C), whereas HSD11B2 levels were lower (P < 0.05)
follicles 25 mm than in all other groups (Fig. 2A,B). Irre- in the cumulus cells exposed to cortisol at a concentration
spective of follicular size, concentrations of cortisol and of 5 mg/mL compared with all other treatments, excepting
progesterone in FF were positively correlated (r ¼ 0.8; the 10-mg/mL treatment group (Fig. 7B).
P < 0.001; Fig. 3A). Concentrations of cortisol (118.6  7.8 vs
120.3  12.2 ng/mL), progesterone (2.4  0.5 vs 2.5 4. Discussion
 0.4 ng/mL), and CBG (11.1  5.1 vs 9.9  3.2 mg/mL) in
plasma did not differ (P > 0.05) before and after the follicle In the present study, intrafollicular concentrations of
aspiration period (Fig. 2). There was a negative correlation total cortisol increased in dominant follicles compared with

=
Fig. 3. Scatter plot graph showing correlation between (A) progesterone (y-axis, nanogram/milliliter) and cortisol (x-axis, nanogram/milliliter) concentrations in
FF (r ¼ 0.8; P < 0.001), (B) progesterone (y-axis, nanogram/milliliter) and CBG (x-axis, microgram/milliliter) concentrations in plasma of mares (r ¼ 0.56; P <
0.01) before and after ovum pickup, (C) gene expression of glucocorticoid receptor (NR3C1) in granulosa cells (y-axis) and CBG concentrations (x-axis, microgram/
milliliter) in FF (r ¼ 0.56; P < 0.05), (D) gene expression of 21-hydroxylase (CYP21A2, y-axis) and 11ß-hydroxysteroid dehydrogenase type 1 (HSD11B1, x-axis) in
granulosa cells (r ¼ 0.88; P < 0.01), (E) gene expression of bone morphogenetic protein 15 (BMP15, y-axis) and growth differentiation factor 9 (GDF9, x-axis) in
equine oocytes (r ¼ 0.9; P < 0.001), (F) gene expression of BCL2-associated X protein (BAX, y-axis) and GDF9 (x-axis) in equine oocytes (r ¼ 0.4; P < 0.001), and
(G) gene expression of BAX (y-axis) and BMP15 (x-axis) in equine oocytes (r ¼ 0.4; P < 0.001). Follicular fluid and granulosa cells were collected from the
following follicle classes: 5–9 mm, 10–14 mm, 15–19 mm, 20–24 mm, and 25 mm.
18 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22

Fig. 4. Relative gene expression in granulosa cells from the following follicle classes: 5–9 mm, 10–14 mm, 15–19 mm, 20–24 mm, and 25 mm. Graphs show
average log10-fold changes in every gene evaluated by quantitative real-time PCR (qPCR). None of the genes was expressed differently among follicle classes.
Values are means  SEM. NR3C1, glucocorticoid receptor; HSD11B1, 11b-hydroxysteroid dehydrogenase type 1; HSD11B2, 11b-hydroxysteroid dehydrogenase type
2; CYP21A2, 21-hydroxylase. Data were normalized against the geometric mean of SNRPD3 and PSMB4. All 4 genes analyzed were either expressed at a low level
(HSD11B1 and HSD11B2) or not expressed at all (NR3C1 and CYP21A2) in the granulosa cells originating from follicles 25 mm.

smaller follicles, which is in agreement with findings in that cortisol and progesterone concentrations in FF were
humans and cattle [12,13]. Nevertheless, CYP21A2, a key highly positively correlated irrespective of follicular size,
enzyme for de novo production of cortisol was absent from this suggests an intrafollicular displacement of cortisol
granulosa cells of dominant follicles. Together with the fact from CBG in favor of progesterone, as shown in humans
[19]. This can be explained by a similar affinity of CBG to
both hormones. In contrast, concentrations of cortisol in
plasma of the mares remained constant, excluding the
possibility of an increase in FF cortisol because of increasing
cortisol concentrations in the circulation. The negative
correlation between plasma CBG and progesterone also
supports the free hormone hypothesis. Previously, our
group demonstrated a clear acute stress response of horses
to parturition [27] or transport [34]. Therefore, concern has
been raised regarding stress of the mare induced by prep-
aration for OPU and by the procedure itself, which might
exert detrimental effects on oocyte quality. In this study, we
were able to show that OPU itself does not induce a chronic
stress response in mares as cortisol concentration in
plasma was not increased at the end of the period where
mares were exposed to repeated OPU procedures. We did
Fig. 5. Comparison of maturation status between in vitro matured equine not determine the acute stress response of mares to indi-
oocytes in the absence or in the presence of cortisol. Values are means 
SEM. MII, metaphase II; MI, metaphase I; GV, germinal vesicle. No significant
vidual OPU procedures in the present study. The presence
differences were seen among treatments. No degenerated oocytes were seen of the gene encoding 21-hydroxylase (CYP21A2) in gran-
in the control group. ulosa cells originating from follicles <25 mm in diameter is
D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22 19

Fig. 6. Relative gene expression in oocytes from matured (hatched columns) and nonmatured (open columns) COCs after IVM in the absence or the presence
of cortisol. Graphs show average log10-fold changes in every gene determined by quantitative real-time PCR (qPCR). Significant differences for treatment and
maturation are indicated for each gene. Significant differences among treatment groups are indicated by different superscripts and refer to matured and
nonmatured COCs combined. Values are means  SEM. Note different scales on y-axis. HSD11B2, 11b-hydroxysteroid dehydrogenase type 2; BAX, BCL2-
associated X protein; BMP15, bone morphogenetic protein 15; GDF9, growth differentiation factor 9. Data were normalized against the geometric mean of
SNRPD3 and PSMB4.

an important finding of this study and is in contrast with estrous cycle, antral follicles of smaller size can become
findings in other species [17]. Progesterone and 17a- atretic near ovulation. In the present study, gene expression
hydroxyprogesterone can be converted to 11- of NR3C1, HSD11B1, and HSD11B2 did not change with
deoxycorticosterone and 11-deoxycortisol, respectively, increasing follicular size. However, the high correlation
only through the action of 21-hydroxylase. Typically, between HSD11B1 and CYP21A2 in granulosa cells suggests
expression of CYP21A2 is exclusively attributed to adreno- that cortisol synthesis in equine ovarian follicles is pri-
cortical cells and especially those of the zona fasciculata marily regulated by HSD11B1. As CYP21A2 was only found in
reticularis [35]. In horses, it has been previously shown that follicles <25 mm and cortisol concentrations remained
the ovarian follicle is an extra-adrenal site of preferential largely unchanged up to this follicle class, HSD11B1 seems
11-deoxycorticosterone biosynthesis by an enzyme having to be essential for maintaining constant cortisol concen-
steroid 21-hydroxylase activity [36]. Nevertheless, in the trations because of its oxidative activity [11]. In contrast,
present study, CYP21A2 mRNA was exclusively found in HSD11B1 and HSD11B2 were only determined in very few
follicles <25 mm in diameter. This makes de novo pro- samples from follicles >25 mm. Their minimal level or
duction of cortisol as a reason for higher concentrations in absence in dominant follicles allows physiologically high
equine FF collected from large follicles highly unlikely. On levels of cortisol to be present in FF in vivo, suggesting an
the other hand, CBG concentration was not decreased in important role in oocyte maturation and probably also for
follicles of this size class. The mechanism contributing to an mediation of the inflammatory-like reaction induced by the
increase in total cortisol concentration in large follicles in LH surge in the follicular wall that precedes ovulation [37].
the present study therefore remains unclear. Our results indicate that the equine COC exclusively
Another mechanism for modulating the local glucocor- expresses HSD11B2, which exerts an oxidative activity. The
ticoid concentration within maturing ovarian follicles is the mRNA expression of HSD11B2 in the oocytes was largely
action of the two metabolizing enzymes, HSD11B1 and unchanged, regardless of maturation stage or addition of
HSD11B2. In cattle, expression of HSD11B1 increases as cortisol to the IVM medium. The same has been observed
follicular maturation progresses, whereas mRNA expres- also in bovine oocytes [38], whereas the oxidative activity
sion of HSD11B2 and NR3C1 in the granulosa cells is low and of HSD11B enzymes, mainly HSD11B2, was increased dur-
remains largely unchanged [9]. During the physiological ing IVM of pig oocytes [6]. In the present study, HSD11B2
20 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22

Fig. 7. Relative gene expression in cumulus cells from matured (hatched columns) and nonmatured (open columns) COCs after IVM in the absence or the
presence of cortisol. Graphs show average log10-fold changes in every gene determined by quantitative real-time PCR (qPCR). Significant differences for treatment
and maturation are indicated for each gene. Significant differences among treatment groups are indicated by different superscripts and refer to matured and
nonmatured COCs combined. Values are means  SEM. Note different scales on y-axis. NR3C1, glucocorticoid receptor; HSD11B2, 11b-hydroxysteroid dehydro-
genase type 2; BCL2, B-cell lymphoma 2; BAX, BCL2-associated X protein. Data were normalized against the geometric mean of SNRPD3 and PSMB4.

was also expressed in equine cumulus cells, and its slightly lower than in the study from Galli et al [28], most
expression was downregulated as cortisol concentrations likely as a consequence of culturing COCs in small groups
in the IVM medium increased. In contrast, bovine and (2–3 COCs in 500 mL). This was done with the aim to easily
human cumulus cells express only HSD11B1, and its separate oocytes and corresponding cumulus cells, ac-
expression and activity are upregulated as IVM progressed cording to their maturation stage, for subsequent molecu-
[38,39]. This might be necessary to modulate in vivo lar analysis. Nevertheless, the number of follicles and COCs
cortisol production by the pre-ovulatory follicles [12,19]. available each day was limited, as OPU was performed
However, in equine COCs, HSD11B2 expression remained when the largest follicle reached 25 mm to avoid retrieving
unchanged as maturation progressed, probably also oocytes from atretic follicles, not every 14 d as recom-
because de novo production of cortisol in the dominant mended [28].
follicles is absent as has been shown in the present study. Cortisol addition affected HSD11B2 expression in the
For the equine oocyte, no information is available about the cumulus cells, but not in the oocytes, suggesting a critical
physiological role of HSD11B2, but it may protect oocytes role of the cumulus cells in regulating local glucocorticoid
from adverse effects of cortisol. In our study, NR3C1 was metabolism in the equine COC. Interestingly, the anti-
either absent from equine oocytes or expression was too apoptotic gene BCL2 is upregulated in the cumulus cells in
low to be reliably quantified. However, presence of response to high cortisol concentrations, whereas HSD11B2
HSD11B2 in the oocytes suggests that changes in the expression is downregulated. This suggests that, when
follicular environment may be translated from cumulus– nonphysiological conditions occur and the metabolizing
granulosa cells or FF and further affect oocyte development. capacity of the COC is inhibited, anti-apoptotic mechanisms
In the present study, a concentration of cortisol (0.1 mg/mL) are being activated to protect the oocyte. An involvement of
proven to be beneficial for bovine oocytes [14], as well as glucocorticoids in granulosa cell apoptosis is not a new
concentrations with detrimental effects (eg, 10 mg/mL) for finding [23]; however, it has received no attention so far in
pig oocytes [7] were tested. In equine oocytes, presence of horses. In this study, the anti-apoptotic gene BCL2 was found
cortisol in the IVM medium in concentrations 100 times exclusively in cumulus cells. However, as RNA transcripts
higher than those physiologically present in large follicles move through the transzonal projections of the cumulus
did not affect IVM rates, as has been shown in mice [40] and cells to the oocyte, the oocyte is also affected [41]. Although
lambs [10], indicating differences in glucocorticoid sensi- not necessarily all proteins are translated from the cumulus
tivity among species. In our study, the MII rates were cells to the oocyte [42], transfer of relevant transcripts for
D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22 21

meiosis resumption from apoptotic cumulus cells might be [3] Schmid W, Cole TJ, Blendy JA, Schutz G. Molecular genetic analysis
of glucocorticoid signalling in development. J Steroid Biochem Mol
missing. In contrary to BCL2, the pro-apoptotic gene BAX was
Biol 1995;53:33–5.
present in cumulus cells and oocytes as well. Recently, [4] Schreiber JR, Nakamura K, Erickson GF. Rat ovary glucocorticoid
expression of several genes involved in the apoptotic pro- receptor: identification and characterization. Steroids 1982;39:569–
cess was analyzed in equine oocytes [24]. In that study, BAX 84.
[5] Hirst MA, Northrop JP, Danielsen M, Ringold GM. High level
was upregulated in cumulus cells originating from older expression of wild type and variant mouse glucocorticoid re-
mares (aged >18 yr), and the authors suggested that aging is ceptors in Chinese hamster ovary cells. Mol Endocrinol 1990;4:
associated with increasing follicular apoptosis rates. Ac- 162–70.
[6] Webb RJ, Sunak N, Wren L, Michael AE. Inactivation of glucocorti-
cording to the current National Center for Biotechnology coids by 11beta-hydroxysteroid dehydrogenase enzymes increases
Information database, the DNA sequence used in the pre- during the meiotic maturation of porcine oocytes. Reproduction
vious study [24] is no longer associated with the BAX gene, 2008;136:725–32.
[7] Yang JG, Chen WY, Li PS. Effects of glucocorticoids on maturation of
but with the transmembrane BAX inhibitor motif containing pig oocytes and their subsequent fertilizing capacity in vitro. Biol
6 (TMBIM6), which is not a pro-apoptotic gene, but an anti- Reprod 1999;60:929–36.
apoptotic gene. In our study, different reference sequences [8] Rae MT, Niven D, Critchley HO, Harlow CR, Hillier SG. Antiin-
flammatory steroid action in human ovarian surface epithelial cells.
for primer design and a homogenous group of mares (aged J Clin Endocrinol Metab 2004;89:4538–44.
7–12 yr) were used. Under these conditions, we observed no [9] Tetsuka M, Nishimoto H, Miyamoto A, Okuda K, Hamano S. Gene
significant differences with regard to BAX expression in expression of 11beta-HSD and glucocorticoid receptor in the bovine
(Bos taurus) follicle during follicular maturation and atresia: the role
matured vs non-matured COCs. However, an involvement of of follicular stimulating hormone. J Reprod Dev 2010;56:616–22.
BAX in oocyte maturation cannot be completely excluded [10] Gonzalez R, Ruiz-Leon Y, Gomendio M, Roldan ER. The effect of
because of its positive correlation with GDF9 and BMP15. glucocorticoids on ERK-1/2 phosphorylation during maturation of
lamb oocytes and their subsequent fertilization and cleavage ability
Oocyte-secreted paracrine factors GDF9 and BMP15 are
in vitro. Reprod Toxicol 2010;29:198–205.
key members of the tumor necrosis factor b superfamily and [11] Michael AE, Thurston LM, Rae MT. Glucocorticoid metabolism and
can regulate female fertility in mammals [43]. Our study reproduction: a tale of two enzymes. Reproduction 2003;126:425–41.
confirmed a highly positive correlation between GDF9 and [12] Acosta TJ, Tetsuka M, Matsui M, Shimizu T, Berisha B, Schams D,
Miyamoto A. In vivo evidence that local cortisol production in-
BMP15 in equine oocytes and a predictive value for matura- creases in the preovulatory follicle of the cow. J Reprod Dev 2005;
tion. Although their expression was strictly localized in the 51:483–9.
oocytes, this does not exclude a role in cell signaling to the [13] Harlow CR, Jenkins JM, Winston RM. Increased follicular fluid total
and free cortisol levels during the luteinizing hormone surge. Fertil
cumulus cells. Interestingly, mRNA levels of GDF9 and BMP15 Steril 1997;68:48–53.
were increased in non-matured compared with MII oocytes, [14] da Costa NN, Brito KN, Santana P, Cordeiro Mda S, Silva TV,
suggesting an essential role of tumor necrosis factor b su- Santos AX, Ramos Pdo C, Santos Sdo S, King WA, Miranda Mdos S,
Ohashi OM. Effect of cortisol on bovine oocyte maturation and
perfamily members in cumulus–oocyte communication and embryo development in vitro. Theriogenology 2016;85:323–9.
for inducing cumulus expansion and meiosis resumption. [15] Lewicka S, von Hagens C, Hettinger U, Grunwald K, Vecsei P,
Our data show that cortisol supplementation downregulates Runnebaum B, Rabe T. Cortisol and cortisone in human follicular
fluid and serum and the outcome of IVF treatment. Hum Reprod
their gene expression and thus may influence maturation if 2003;18:1613–7.
high nonphysiological concentrations occur. [16] Thurston LM, Norgate DP, Jonas KC, Gregory L, Wood PJ, Cooke BA,
In conclusion, this study addressed for the first time the Michael AE. Ovarian modulators of type 1 11beta-hydroxysteroid
dehydrogenase (11betaHSD) activity and intra-follicular cortisol:
involvement of glucocorticoids in follicular growth and
cortisone ratios correlate with the clinical outcome of IVF. Hum
oocyte maturation in horses. We have demonstrated herein Reprod 2003;18:1603–12.
complex mechanisms of glucocorticoid regulation within [17] Omura T, Morohashi K. Gene regulation of steroidogenesis. J Steroid
Biochem Mol Biol 1995;53:19–25.
the growing ovarian follicle and in maturing equine COCs.
[18] Gayrard V, Alvinerie M, Toutain PL. Interspecies variations of
The results clearly suggest that cortisol seems to be corticosteroid-binding globulin parameters. Domest Anim Endo-
necessary for oocyte development and maturation and that crinol 1996;13:35–45.
glucocorticoid action in equine COCs is modulated by [19] Andersen CY. Possible new mechanism of cortisol action in female
reproductive organs: physiological implications of the free hormone
HSD11B2. Our data elucidated at least in part the involve- hypothesis. J Endocrinol 2002;173:211–7.
ment of glucocorticoids in equine ovarian function. A better [20] Tetsuka M, Thomas FJ, Thomas MJ, Anderson RA, Mason JI,
understanding of these mechanisms might help in further Hillier SG. Differential expression of messenger ribonucleic acids
encoding 11beta-hydroxysteroid dehydrogenase types 1 and 2 in
development of equine-assisted reproduction techniques. human granulosa cells. J Clin Endocrinol Metab 1997;82:2006–9.
[21] Sunak N, Green DF, Abeydeera LR, Thurston LM, Michael AE.
Acknowledgments Implication of cortisol and 11beta-hydroxysteroid dehydrogenase
enzymes in the development of porcine (Sus scrofa domestica)
ovarian follicles and cysts. Reproduction 2007;133:1149–58.
The authors thank Mahsa Adib and Ashani Hamilton for [22] Short RV. Steroids present in the follicular fluid of the mare. J
their help in performing quantitative real-time PCR ana- Endocrinol 1960;20:147–56.
[23] Sasson R, Tajima K, Amsterdam A. Glucocorticoids protect against
lyses and handling the animals. apoptosis induced by serum deprivation, cyclic adenosine 30 ,50 -
monophosphate and p53 activation in immortalized human
References granulosa cells: involvement of Bcl-2. Endocrinology 2001;142:
802–11.
[24] Cox L, Vanderwall DK, Parkinson KC, Sweat A, Isom SC. Expression
[1] Breen KM, Karsch FJ. New insights regarding glucocorticoids, stress profiles of select genes in cumulus-oocyte complexes from young
and gonadotropin suppression. Front Neuroendocrinol 2006;27: and aged mares. Reprod Fertil Dev 2015;27:914–24.
233–45. [25] Campos-Chillon F, Farmerie TA, Bouma GJ, Clay CM, Carnevale EM.
[2] Kalantaridou SN, Makrigiannakis A, Zoumakis E, Chrousos GP. Stress Effects of aging on gene expression and mitochondrial DNA in the
and the female reproductive system. J Reprod Immunol 2004;62:61–8. equine oocyte and follicle cells. Reprod Fertil Dev 2015;27:925–33.
22 D. Scarlet et al. / Domestic Animal Endocrinology 59 (2017) 11–22

[26] Gastal EL, Gastal MO, Wiltbank MC, Ginther OJ. Follicle deviation [34] Deichsel K, Pasing S, Erber R, Ille N, Palme R, Aurich J, Aurich C.
and intrafollicular and systemic estradiol concentrations in mares. Increased cortisol release and transport stress do not influence
Biol Reprod 1999;61:31–9. semen quality and testosterone release in pony stallions. Ther-
[27] Nagel C, Erber R, Bergmaier C, Wulf M, Aurich J, Mostl E, Aurich C. iogenology 2015;84:70–5.
Cortisol and progestin release, heart rate and heart rate variability [35] Ishimura K, Fujita H. Light and electron microscopic immunohisto-
in the pregnant and postpartum mare, fetus and newborn foal. chemistry of the localization of adrenal steroidogenic enzymes.
Theriogenology 2012;78:759–67. Microsc Res Tech 1997;36:445–53.
[28] Galli C, Colleoni S, Duchi R, Lagutina I, Lazzari G. Developmental [36] Bijault C, Dehennin L. Steroid 21-hydroxylase activity in equine
competence of equine oocytes and embryos obtained by in vitro ovarian follicles evidenced by isotope dilution-mass spectrometry. J
procedures ranging from in vitro maturation and ICSI to embryo Steroid Biochem Mol Biol 1991;38:165–72.
culture, cryopreservation and somatic cell nuclear transfer. Anim [37] Hillier SG, Tetsuka M. An anti-inflammatory role for glucocorticoids
Reprod Sci 2007;98:39–55. in the ovaries? J Reprod Immunol 1998;39:21–7.
[29] Hinrichs K, Choi YH, Love LB, Varner DD, Love CC, Walckenaer BE. [38] Tetsuka M, Takagi R, Ambo N, Myat TS, Zempo Y, Onuma A.
Chromatin configuration within the germinal vesicle of horse oo- Glucocorticoid metabolism in the bovine cumulus-oocyte complex
cytes: changes post mortem and relationship to meiotic and matured in vitro. Reproduction 2016;151:73–82.
developmental competence. Biol Reprod 2005;72:1142–50. [39] Smith MP, Mathur RS, Keay SD, Hall L, Hull MGR, Jenkins JM. Peri-
[30] Kenkel CD, Aglyamova G, Alamaru A, Bhagooli R, Capper R, ovulatory human oocytes, cumulus cells, and ovarian leukocytes
Cunning R, deVillers A, Haslun JA, Hedouin L, Keshavmurthy S, express type 1 but not type 2 11b-hydroxysteroid dehydrogenase
Kuehl KA, Mahmoud H, McGinty ES, Montoya-Maya PH, Palmer CV, RNA. Fertil Steril 2000;73:825–30.
Pantile R, Sanchez JA, Schils T, Silverstein RN, Squiers LB, Tang PC, [40] Andersen CY. Effect of glucocorticoids on spontaneous and
Goulet TL, Matz MV. Development of gene expression markers of follicle-stimulating hormone induced oocyte maturation in
acute heat-light stress in reef-building corals of the genus Porites. mouse oocytes during culture. J Steroid Biochem Mol Biol 2003;
PLoS One 2011;6:e26914. 85:423–7.
[31] Scarlet D, Ertl R, Aurich C, Steinborn R. The orthology clause in the [41] Macaulay AD, Gilbert I, Scantland S, Fournier E, Ashkar F,
next generation sequencing era: novel reference genes identified by Bastien A, Saadi HA, Gagne D, Sirard MA, Khandjian EW,
RNA-seq in humans improve normalization of neonatal equine Richard FJ, Hyttel P, Robert C. Cumulus cell transcripts transit to
ovary RT-qPCR data. PLoS One 2015;10:e0142122. the bovine oocyte in preparation for maturation. Biol Reprod
[32] Livak KJ, Schmittgen TD. Analysis of relative gene expression data 2016;94:16.
using real-time quantitative PCR and the 2(-Delta Delta C(T)) [42] Macaulay AD, Gilbert I, Caballero J, Barreto R, Fournier E, Tossou P,
Method. Methods 2001;25:402–8. Sirard MA, Clarke HJ, Khandjian EW, Richard FJ, Hyttel P, Robert C.
[33] Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, The gametic synapse: RNA transfer to the bovine oocyte. Biol
Mueller R, Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, Wittwer CT. Reprod 2014;91:90.
The MIQE guidelines: minimum information for publication of quan- [43] Otsuka F, McTavish KJ, Shimasaki S. Integral role of GDF-9 and BMP-
titative real-time PCR experiments. Clin Chem 2009;55:611–22. 15 in ovarian function. Mol Reprod Dev 2011;78:9–21.

You might also like