Animal Feed Science and Technology 174 (2012) 79–85

Contents lists available at SciVerse ScienceDirect

Animal Feed Science and Technology

journal homepage: www.elsevier.com/locate/anifeedsci

Zearalenone enhances reproductive tract development, but does not

alter skeletal muscle signaling in prepubertal gilts

a,∗ a b b c b

W.T. Oliver , J.R. Miles , D.E. Diaz , J.J. Dibner , G.E. Rottinghaus , R.J. Harrell

a

USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, United States

b

Novus International, Inc., St. Charles, MO, United States

c

Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, MO, United States

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

Article history: is a potent mycotoxin that has estrogenic properties. In vitro results indicate

Received 23 March 2011

that zearalenone metabolites down-regulate proteins associated with protein synthesis

Received in revised form 24 February 2012

(protein kinase B, Akt) and cellular proliferation (extracellular signal-regulated kinase, ERK).

Accepted 27 February 2012

The objectives of this study were to determine the effect of zearalenone on (1) growth

performance and signaling for protein synthesis, and (2) reproductive tract development.

At 28 d of age, gilts were randomly assigned to consume a commercial basal diet (C) or

Keywords:

Mycotoxin C+1.5 mg/kg zearalenone (n = 10) for 4 wk, at which time gilts were euthanized, urine col-

lected, and tissue collected. No differences were observed in average daily gain, average

Skeletal muscle

Swine daily feed intake, or gain:feed (P>0.28). Reproductive tract weight (2.4-fold) and uterine

Uterus endometrial gland development (50%) were increased in zearalenone fed gilts (P<0.01). In

Zearalenone uterus, receptor (ER)-␣ expression was unchanged (P>0.28), but gilts consuming

zearalenone had 2.0- and 3.5-fold higher abundance of ER-␤ mRNA and protein, respec-

tively (P<0.01). No differences were observed in Akt, mammalian target of rapamycin, or

ERK abundance or phosphorylation in muscle (P>0.36). Zearalenone had no effect on growth

performance or skeletal muscle signaling in prepubertal gilts, but zearalenone increased

reproductive tract size and glandular development, possibly due, in part, to altering the

expression of ER-␤.

Published by Elsevier B.V.

1. Introduction

The mycotoxin zearalenone is a potent estrogenic secondary metabolite produced by several Fusarium species (Riley and

Petska, 2005). Zearalenone is a resorcylic acid lactone that is most commonly found in many cereal crops and swine are

one of the most sensitive species with tolerance limits in feed as low as 50 ␮g/kg proposed (BML, 2000). Zearalenone and

its metabolites ␣-zearalenol and ␤-zearalenol have been shown, in vitro, to down-regulate signaling for protein synthesis

and cellular proliferation (Wollenhaupt et al., 2004). In addition, zearalenone, as well as ␣-zearalenol and its metabolite

Abbreviations: ADFI, average daily feed intake; ADG, average daily gain; Akt, protein kinase B; BW, body weight; C, commercial basal diet; COX2,

prostaglandin-endoperoxide synthase 2; DON, deoxynivalenol; ER, ; ERK, extracellular signal-regulated kinase; ESI, electrospray ioniza-

tion; G:F, gain to feed ratio; GLM, general linear model; HPLC/MS, high-performance liquid chromatography/mass spectrometry; mTOR, mammalian target

of rapamycin; tcRNA, total cellular ribonucleic acid.

Mention of trade names, proprietary products, or specified equipment does not constitute a guarantee or warranty by the USDA and does not imply

approval to the exclusion of other products that may be suitable. USDA is an equal opportunity provider and employer.

Corresponding author at: USDA, ARS, P.O. Box 166, U.S. Meat Animal Research Center, Clay Center, NE, United States. Tel.: +1 402 762 4206;

fax: +1 402 762 4209.

E-mail address: [email protected] (W.T. Oliver).

0377-8401/$ – see front matter. Published by Elsevier B.V. doi:10.1016/j.anifeedsci.2012.02.012

80 W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85

Table 1

a

Composition and calculated nutrient content of the diets.

Item Diets

b

Phase 2 Phase 3

Control Zearalenone Control Zearalenone

Ingredients, g/kg

Maize 477.5 470.0 622.2 614.7

Soybean meal, 465 g/kg 240.0 240.0 299.9 299.9

Lactose 120.0 120.0 0.0 0.0

Poultry meal 37.0 37.0 0.0 0.0

Fishmeal 35.0 35.0 0.0 0.0

Choice white grease 35.0 35.0 35.0 35.0

Monocalcium phosphate 13.3 13.3 14.1 14.1

Limestone 4.0 4.0 8.9 8.9

c

Mecadox, 5.5 g/kg 10.0 10.0 5.0 5.0

Lysine HCl,788 g/kg 3.0 3.0 3.0 3.0

l-Threonine,985 g/kg 14.0 14.0 1.1 1.1

Methionine hydroxy analog, 880 g/kg 2.2 2.2 1.8 1.8

Salt 4.0 4.0 5.0 5.0

CuSO4 1.0 1.0 0.0 0.0

d

Vitamin premix 2.5 2.5 2.5 2.5

e

Mineral premix 1.5 1.5 1.5 1.5

Toxin 0.0 7.5 0.0 7.5

f

Calculated nutrient content

Metabolizable energy, MJ/kg 14.3 14.2 14.4 14.3

Crude protein, g/kg 208.0 207.0 196.0 195.0

Total ileal digestible lysine, g/kg 12.5 12.0 12.5 12.0

Ca, g/kg 8.6 8.6 7.2 7.2

Available P, g/kg 5.2 5.2 3.7 3.7

a

Expressed on as-fed basis.

b

A common phase 1 diet was fed for one week prior to the initiation of treatments.

c

For control of enteritis and improved growth performance.

d

Provided the following per kilogram of diet: vitamin A, 9000 IU; vitamin D3, 200 IU; vitamin E, 19 IU; vitamin K, 2.2 mg; thiamine, 2.2 mg; riboflavin,

4.4 mg; , 33 mg; pantothenic acid, 22 mg; vitamin B12, 0.028 mg; vitamin B6, 2.2 mg; folic acid, 1.35 mg; and biotin, 0.11 mg.

e

Provided the following per kilogram of diet: Fe, 78 mg; Cu, 7 mg; Co, 0.80 mg; Zn, 168 mg; Mn, 60 mg; I, 0.79 mg; and Se, 0.13 mg.

f

Calculated nutrient content based on standard feed tables (NRC, 1998).

␣-zearalanol, accumulate in muscle tissue, in vivo (Zöllner et al., 2002). Thus, one objective of the current experiment

was to determine the effect of zearalenone contaminated feed on growth performance and protein synthesis signaling in

prepubertal gilts.

The effect of zearalenone on the reproductive performance of pigs is highly dependent on their reproductive status

(Diekman and Green, 1992) and prepubertal gilts are particularly susceptible to zearalenone-contaminated feed (Kordic

et al., 1992). In gilts, high levels of zearalenone (>22.0 mg/kg, Tiemann and Dänicke, 2007) can have deleterious effects on

reproductive performance including a decrease in corpora lutea, decreased ovary size, decreased fertility, and an increase

in abortion rates. More moderate doses (>2.0 mg/kg, Kordic et al., 1992; Döll et al., 2004) will increase vulva and total

reproductive tract size in gilts. The effects of zearalenone and its metabolites are mediated through the estrogen receptors

(ER), and these metabolites have an affinity for both the -␣ and -␤ isoforms of the ER (Mueller et al., 2004; Takemura

et al., 2007). Thus, the second objective of this experiment was to determine the effect of zearalenone contaminated feed

on the reproductive tract development, including the abundance of key regulators of reproductive tract development and

maintenance.

2. Materials and methods

2.1. Animal care and treatments

All animal procedures were reviewed and approved by NOVUS International Animal Care and Use Committee. Gilts

(n = 10 per treatment; PIC cross, landrace × large white females × large white × duroc × pietran males) were weaned from

their sow at 21 d of age, blocked by weight and placed in a pen containing two pigs per pen. A three-phase feeding program

was implemented for 35 d with diets formulated to meet or exceed NRC requirements (NRC, 1998). Dietary ingredients

were screened, and found negative, throughout the purchasing process for levels of most common mycotoxins, including

zearalenone. Gilts were allowed to adjust for 1 wk on a commercial diet, at which time gilts were randomly assigned to

consume a control basal diet or the control diet + 1.5 mg/kg zearalenone for 4 wk (Table 1). Gilts were weighed gravimetrically

at d 0, 7, 14, 21, and 28 of treatment. In addition, vulva height, width, and length were measured as the gilts were weighed.

W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85 81

Table 2

a

Primer sequences for real time RT-PCR analysis.

mRNA Primer Sequence Fragment size (bp)



PR F 5 -AAGTCACTGCCAGGTTTTCG 209 

R 5 -TGCCACATGGTAAGGCATAA



COX2 F 5 -TCGACCAGAGCAGAGAGATGAGAT 134 

R 5 -ACCATAGAGCGCTTCTAACTCTGC 

ER␣ F5-AGCACCCTGAAGTCTCTGGA 160 R5-TGTGCCTGAAGTGAGACAGG



ER␤ F 5 -GGCAACGACTTCAAGGTTTC 239 

R 5 -CTGCTGCTGGGAGGAGATAC



ERK1 F 5 -CAGTCTCTGCCCTCCAAGAC 218 

R 5 -GGGTAGATCATCCAGCTCC



ERK2 F 5 -AGGGGCGGTTTCTGATAGTT 225 

R 5 -GAGGAACAGGGTCAGCAGAG



mTOR F 5 -GAAGAGCACGACCTGGAGAG 249 

R 5 -GTCCAGCTTCTCCCCTTTCT 

18S F 5 -ATGGCCGTTCTTAGTTGGTG 217 

R 5 -CGCTGAGCCAGTCAGTGTAG

a ◦ ◦ ◦ ◦

The PCR conditions included denaturation (95 C, 2 min) followed by 40 amplification cycles of 95 C for 15 s, 60 C for 15 s, and 70 C for 45 s. F, forward

primer; R, reverse primer; PR, progesterone receptor; COX2, prostaglandin-endoperoxide synthase 2; ER␣, ; ER␤, estrogen receptor

beta; ERK, extracellular signal-related kinase; mTOR, mammalian target of rapamycin; 18S, 18S ribosomal protein.

2.2. Zearalenone culture method

To culture the zearalenone, shelled maize (100 g) was added to 1 quart wide mouth canning jars and 40 mL of water is

added. The jars were autoclaved for 20 min at 121 C and 138 kilopascal, then inoculated with a sterile aqueous suspension

of Fusarium graminearum R5454 from the Penn State Fusarium culture collection. The jars were shaken, the lids loosened for

additional respiration, and then stacked lying on their sides. They were incubated in the dark for 4 weeks at 23 C. After 4 wk

the jars were autoclaved and the culture material was dried, ground, blended and stored at −20 C. Multiple subsamples of

the culture material were analyzed for zearalenone, aflatoxins, ochratoxin A, fumonisins, DON and T-2 toxin concentration

by HPLC with fluorescence detection.

2.3. Sample collection and analytical procedures

After 4 wk of treatment, gilts were euthanized and urine was collected from the bladder. At this time, the reproductive

tract was removed intact and weighed. A sample of the uterine body and muscle (longissimus dorsi, approximately at the

last rib) were collected and snap-frozen in liquid nitrogen and stored at −80 C for later analyses. Urine was treated with

beta-glucuronidase/sulfatase (Sigma, St. Louis, MO, USA), affinity column cleanup (R-Biopharm, Darmstadt, Germany) and

analyzed by HPLC/MS using ESI in the negative mode with single ion monitoring for zearalenone and ␣- and ␤-zearalenol

(adapted from Songsermsakul et al., 2006). Uterine samples were collected, fixed in 10% formalin for 24 h, and embedded

in paraffin. Cross-sections of the uterine segments were cut approximately 8 ␮m thick with a microtome and were stained

with hematoxylin and eosin-Y. Morphometric measurements were performed by one person using light microscopy with a

computer assisted morphometric system. The area of endometrial and total uterine tissue was measured.

2.4. RNA isolation and real-time PCR

Total cellular RNA (tcRNA) was isolated from the uterus and longissimus dorsi using RNeasy Mini kits (Quiagen, Valencia,

CA, USA) and samples were DNase I treated on the column using DNase I provided by the manufacturer. Total RNA was

quantified using a ND-1000 spectrophotometer (Nano-Drop Technologies, Wilmington, DE, USA). Total cellular RNA samples

(1 ␮g) were reverse transcribed using the iScript cDNA synthesis kit (Bio-Rad, Hercules, CA, USA). Each cDNA sample was

assayed in duplicate (25 ng tcRNA equivalents) for progesterone receptor, prostaglandin-endoperoxide synthase 2 (COX2),

ER-␣, ER-␤, extracellular signal-related kinases (ERK) 1 and 2, and mammalian target of rapamycin (mTOR) with appropriate

0.25 ␮M forward and reverse primers (Table 2), 12.5 ␮L of Taq, SYBR green, and ROX supermix (Bio-Rad, Hercules, CA, USA)

included in a 25 ␮L reaction. Ribosomal 18S primers (Table 2) were used to measure 18S mRNA concentrations, which were

not different between treatments (data not shown). Therefore, 18S was used as the reference control transcript to calculate

relative quantities of each target transcript using the comparative cycle threshold method (Livak and Schmittgen, 2001).

Amplification was performed using a PTC-200 thermocycler fitted with a chromo 4 fluorescence detector (Bio-Rad, Hercules,

◦ ◦ ◦

CA, USA). The PCR conditions included denaturation (95 C, 2 min) followed by 40 amplification cycles of 95 C for 15 s, 60 C

for 15 s, and 70 C for 45 s. Melting curve analysis and gel electrophoresis was used to confirm the amplification of a single

product of the predicted size.

82 W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85

Table 3

a

Urine levels of zearalenone and its metabolites and performance of prepubertal gilts fed control or control + zearalenone (1.5 mg/kg) diets for four weeks.

Variable Treatment P value

Control Zearalenone

Zearalenone, ␮g/kg 4.0 ± 85.3 292.4 ± 76.2 <0.025

␣-zearalenol, ␮g/kg 1.3 ± 21.9 113.3 ± 19.6 <0.002

-zearalenol, ␮g/kg 0.0 ± 3.5 14.6 ± 3.1 <0.007

Live weight, kg

d 0 6.45 ± 0.21 6.97 ± 0.19 0.10

± ±

d 7 9.27 0.50 10.48 0.44 0.11

d 14 12.19 ± 0.46 12.96 ± 0.41 0.25

d 21 17.85 ± 0.91 19.70 ± 0.81 0.17

d 28 21.55 ± 0.94 23.26 ± 0.85 0.21

Average daily gain, g/d

d 0–14 442 ± 28 461 ± 25 0.63

d 14–28 624 ± 42 686 ± 37 0.30

d 0–28 539 ± 28 582 ± 25 0.29

Average daily feed intake, g/d

d 0–14 608 ± 67 601 ± 60 0.93

d 14–28 1068 ± 47 1068 ± 42 0.99

d 0–28 838 ± 48 834 ± 43 0.96

Gain:feed, g/g

d 0–14 0.77 ± 0.07 0.77 ± 0.06 0.96

d 14–28 0.58 ± 0.02 0.64 ± 0.02 0.06

d 0–28 0.65 ± 0.03 0.70 ± 0.03 0.28

a

Values are least squares means ± SEM; for zearalenone, ␣-zearalenol, ␤-zearalenol, live weight, and average daily gain, n = 10; for average daily feed

intake and gain:feed, n = 5.

2.5. Western immunoblotting

Uterine and muscle samples were analyzed for total and amino acid phosphorylation of Akt, ERK 1/2, and mTOR by

Western immunoblot analysis. In addition, uterine samples were analyzed for total ER-␣ and -␤. Samples were powdered

at liquid nitrogen temperature and a crude soluble protein extract was prepared in buffer that contained 50 mM Tris HCl

(pH 7.5); 3 mM EDTA; 100 mM NaCl; protease inhibitors (Complete Mini (1836153), 1 tab/10 mL, Roche Applied Science,

Indianapolis, IN, USA); phosphatase inhibitors (Phosphatase Inhibitor Cocktail 1 (P2850) and 2 (P5726), each at 1:100 v:v;

Sigma–Aldrich, St. Louis, MO, USA); and 1% IGEPAL A-630 (USB Corp., Cleveland, OH, USA). The protein concentration of the

extracts was determined using the bicinchoninic acid reagent (Smith et al., 1985). Sixty micrograms of soluble protein were

resolved on 10% SDS-PAGE gels and electroblotted onto polyvinylidene fluoride membranes (PALL Co., Pensacola, FL, USA).

Pooled samples of known phosphorylation status were used on each block as a positive control. For Akt, ERK 1/2, and mTOR,

the membranes were probed for phosphorylated protein using specific anti-phosphoprotein antibodies (1:2000, Akt-Ser

202 204 2448

473, 1:2000, ERK-Thr /Tyr , 1:2000, mTOR-Ser ; Cell Signaling Technology, Danvers, MA, USA). After washing in Tris-

buffered saline with 0.1% Tween (TBS-T), the blots were reprobed for total protein with anti-Akt (1:2000, detects isoforms

1–3; Cell Signaling Technology, Danvers, MA, USA), anti-ERK (1:2000, detects isoforms 1 and 2; Cell Signaling Technology,

Danvers, MA, USA), and anti-mTOR (1:2000, Cell Signaling Technology, Danvers, MA, USA). For the ERs, the membranes were

probed for total protein with anti-ER-␣ (1/2000, Santa Cruz Biotechnology, Santa Cruz, CA, USA) and anti-ER-␤ (1/1000,

Santa Cruz Biotechnology, Santa Cruz, CA, USA) antibodies.

2.6. Statistical analysis

Data were subjected to analysis of variance using the GLM procedure of SAS (Version 9.2, 1998; SAS Inst., Cary, NC, USA).

Data were evaluated for the effects of diet, week, and their interaction. For performance measures, initial body weight was as

a covariate. Similarly to previous experiments (Oliver et al., 2005; Oliver and Miles, 2010), pen of pigs was the experimental

unit for evaluation of the effects of diet on feed intake and feed efficiency and individual pig was the experimental unit for

all other statistical procedures. The significance level for all tests was set at P<0.05.

3. Results

3.1. Zearalenone and metabolites

Zearalenone and the zearalenone metabolites ␣- and ␤-zearalenol increased in urine from gilts consuming zearalenone

contaminated feed, compared to gilts consuming the control diet (Table 3; P<0.007). Prepubertal gilts weighed 6.71 ± 0.20 kg

initially (P=0.10) and gained 560 ± 26 g/d (Table 3; P=0.29) during the 4-wk study, which resulted in an ending BW of

W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85 83

Table 4

Reproductive tract measurements and estrogen receptor-␣ and -␤ abundance of prepubertal gilts fed control or control + zearalenone (1.5 mg/kg) diets for a

four weeks.

Variable Treatment P value

Control Zearalenone

Estrogen receptor-␣

mRNA, relative units 0.25 ± 0.08 0.24 ± 0.06 0.65

Protein, arbitrary units 83.1 ± 8.06 81.8 ± 5.49 0.32

Estrogen receptor-␤

± ±

mRNA, relative units 0.39 0.08 0.79 0.09 <0.01

Protein, arbitrary units 15.12 ± 4.42 51.89 ± 5.86 <0.01

Reproductive tract weight, g 20.91 ± 4.30 50.62 ± 3.81 <0.001

Endometrial gland development, glandular:total uterine area 0.18 ± 0.02 0.27 ± 0.02 0.02

Vulva height, mm

d 0 11.09 ± 0.70 10.96 ± 0.63 0.9

d 7 11.48 ± 0.80 14.64 ± 0.72 0.03

d 14 13.65 ± 1.36 22.09 ± 1.21 0.002

± ±

d 21 13.25 1.36 18.77 1.22 0.02

d 28 13.73 ± 1.02 23.89 ± 0.91 <0.001

Vulva width, mm

d 0 9.21 ± 0.55 8.36 ± 0.49 0.28

d 7 10.55 ± 0.66 12.39 ± 0.59 0.08

d 14 13.06 ± 0.80 20.28 ± 0.71 <0.001

d 21 12.53 ± 0.91 17.92 ± 0.82 0.004

d 28 13.43 ± 0.65 23.49 ± 0.58 <0.001

Vulva length, mm

d 0 8.32 ± 0.76 7.71 ± 0.68 0.57

d 7 13.57 ± 0.63 11.00 ± 0.57 0.62

d 14 13.05 ± 0.74 15.23 ± 0.66 0.06

d 21 12.09 ± 0.77 14.61 ± 0.69 <0.05

d 28 14.00 ± 1.07 19.44 ± 0.96 0.007

a

Values are least squares means ± SEM; n = 10.

23.91 ± 0.90 kg (P=0.21), regardless of treatment. Average daily feed intake did not differ between gilts consuming the

control and zearalenone diets (836 ± 45 g/d; P=0.96), which resulted in similar feed efficiency (G:F; 0.68 ± 0.03; P=0.28).

3.2. Reproductive tract measurements

Vulva measurements (height, width, and length) did not differ between groups at the initiation of treatments (Table 4;

P>0.28). By d 7, gilts consuming zearalenone contaminated diets had larger vulva heights (14.64 ± 0.72 vs. 11.48 ± 0.80 mm;

P=0.03) compared to control gilts. Vulva heights were 74% larger at d 28 of study in zearalenone treated compared to control

gilts (23.89 ± 0.91 vs. 13.73 ± 1.02 mm; P<0.001). Vulva width and length increased in zearalenone treated gilts compared to

control gilts by d 14 and 21 of treatment, respectively (P<0.05). By 28 d of age, vulva width and length were increased by 75

and 39%, respectively, in zearalenone treated compared to control gilts (23.49 ± 0.58 vs. 13.43 ± 0.65 mm and 19.44 ± 0.96

vs. 14.00 ± 1.07 mm; P<0.008). After 4 wk of consuming zearalenone contaminated diets, prepubertal gilts had a 2.4-fold

increase in total reproductive tract weight compared to gilts consuming the control diet (Table 4; 50.6 ± 3.8 vs. 20.9 ± 4.3 g;

P<0.001). In addition, endometrial gland development, expressed as a ratio of glandular to total uterine area, was increased

50% in pigs consuming the zearalenone contaminated diets compared to control gilts (Table 4; 0.264 ± 0.027 vs. 0.117 ± 0.016;

P<0.02).

3.3. Cellular signaling

No differences in ERK 1/2 (0.82 ± 0.11 vs. 0.85 ± 0.13 relative units) or mTOR (0.73 ± 0.08 vs. 0.81 ± 0.10 relative units)

mRNA abundance were observed in skeletal muscle of pigs consuming the zearalenone or control diets (P>0.43). Sim-

ilarly, no difference was observed in the total abundance of ERK 1/2 (81 ± 8 vs. 88 ± 9 arbitrary units), Akt (92 ± 8 vs.

86 ± 7 arbitrary units), or mTOR (62 ± 4 vs. 63 ± 4 arbitrary units) protein (P>0.61). In addition, the activity of ERK 1/2

(0.22 ± 0.4 vs. 0.28 ± 0.05), Akt (0.41 ± 0.06 vs. 0.40 ± 0.04), and mTOR (0.39 ± 0.04 vs. 0.41 ± 0.04), as measured by the ratio

of phosphorylated protein to total protein, was not different between zearalenone and control gilts (P>0.22).

In the uterus, no differences in the mRNA abundance of the progesterone receptor, COX2, ERK 1/2, or mTOR were observed

between zearalenone treated and control gilts (data not shown; P>0.44). In addition, ER-␣ mRNA and protein abundance

was similar between zearalenone and control gilts (Table 4; P>0.32). However, the mRNA and protein abundance of ER-␤

were increased 2.0- and 3.5-fold, respectively, in prepubertal gilts consuming zearalenone compared to gilts consuming the

control diet (Table 4; P<0.01).

84 W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85

4. Discussion

In vivo, zearalenone is predominantly converted into ␣- and ␤-zearalenol and excreted as zearalenone, ␣-zearalenol, or

-zearalenol in feces and urine (Zöllner et al., 2002). In the current experiment, zearalenone delivery to prepubertal gilts

was confirmed by analyzing zearalenone and its metabolites, ␣- and ␤-zearalenol, in urine. As expected, control gilts had

very low or undetectable levels while zearalenone gilts excreted high levels of these compounds.

There are few reports on the effect of zearalenone or its metabolites on growth performance in pigs. Döll et al. (2003)

observed decreased feed intake and growth rates in gilts fed diets with zearalenone (1.2 mg/kg of maize), but the decrease in

performance was likely due the presence of the related Fusarium toxin deoxynivalenol (DON, 8.6 mg/kg of maize). It is well

established that DON decreases feed intake and growth (Rotter et al., 1996). However, ␣-zearalanol (also called ),

which is converted from ␣-zearalenol (Zöllner et al., 2002), has long been used successfully as an implant to improve

growth performance in cattle (Simms et al., 1988). Generally, cattle administered zeranol implants do not have altered feed

intake and body composition differences in response to implants are breed specific (Williams et al., 1991). To the authors

knowledge, no genetic differences have been reported in response to the consumption of zearalenone or its metabolites

in swine. Although zearalenone, ␣-zearalenol, and zeranol are present in muscle of pigs consuming zearalenone (Zöllner

et al., 2002), prepubertal gilts consuming zearalenone-contaminated feed in the current experiment gained at a similar rate

and consumed a similar amount of feed compared to gilts consuming the control diet. This is likely because pigs produce

only trace amounts (<1 ␮g/L urine) of zeranol from ␣-zearalenol (Zöllner et al., 2002). In gilts, Jiang et al. (2010) observed

increased ADG and ADFI in pigs consuming zearalenone contaminated feeds. This differs from the current study and is

possibly due to the fact that the gilts in the Jiang et al. (2010) study were older than those in the current study. The age of the

pigs is unknown, but their initial body weight was twice that of gilts in the current experiment. In addition, gilts consuming

zearalenone in the study by Jiang et al. (2010) appear to be heavier at the initiation of treatments, compared to control pigs

(12.12 vs. 11.01 kg; SEM = 0.131), which confounds the performance data.

Contrary to the anabolic effects of zeranol in cattle, ␣- and ␤-zearalenol have been shown, in vitro, to reduce the phospho-

rylation of proteins involved in protein synthesis and cellular proliferation (Wollenhaupt et al., 2004). In addition, Abid-Essefi

et al. (2004) observed that zearalenone inhibited protein and DNA synthesis in Vero (kidney) and Caco-2 (human colon

cancer) cell lines. Cellular proliferation and protein synthesis are driven by a complex interaction of signaling proteins.

Wollenhaupt et al. (2004) observed changes in phosphorylation of ERK 1/2, Akt, and the translation initiation factor 4E-BP1

by ␤-, and to a lesser extent, ␣-zearalenol that would result in decreased cellular proliferation and protein synthesis. Mam-

malian target of rapamycin is considered a master protein kinase that is regulated, in part, by Akt and contributes to the

control of protein synthesis by regulating components of cellular signaling that directly affect protein synthesis (Avruch

et al., 2005). In the current experiment, no differences in skeletal muscle ERK1/2, Akt, or mTOR were observed between gilts

consuming zearalenone and gilts consuming the C diet, which is in agreement with the lack of a performance difference

in the current study. The gilts in the current study were gaining at a high rate (>500 g/d). Thus, it is likely that the normal

anabolic signals (insulin like growth factor-I, insulin) not present in the in vitro studies precluded any potential changes

in signaling for reduced cellular proliferation and protein synthesis that may have occurred due to zearalenone and its

metabolites.

The effect of zearalenone on the reproductive performance of pigs is highly dependent on their reproductive status

(Diekman and Green, 1992) and prepubertal gilts are particularly susceptible to zearalenone-contaminated feed (Kordic

et al., 1992). In the current experiment, prepubertal gilts had large increases in edematous swelling of the vulva throughout

the study and increased total uterus weight and endometrial gland development after 4 wk consuming zearalenone con-

taminated diets. An increase in vulva size is well documented in pigs consuming zearalenone (Kordic et al., 1992). Similarly,

zearalenone has been shown to increase the size of the reproductive tract in prepubertal gilts (Döll et al., 2004). However,

Döll et al. (2004) did not observe histological differences in the uterine glands of these gilts. These data likely differ from the

current experiment due to the lower dose of zearalenone used (0.42 vs. 1.5 mg/kg). In addition, it is likely that pigs in the

current study had considerably less developed uteri before exposure to zearalenone and its metabolites compared to the

older gilts (70 d, 32.6 kg vs. 28 d, 6.6 kg) used in the study by Döll et al. (2004).

The effect of zearalenone and its metabolites on the in vivo transcription and translation of reproductive genes and

mRNA is not well established. In the current experiment, we did not observe any changes in the expression of mRNA for

progesterone receptor, COX2, or ER-␣. In addition, protein abundance for ER-␣ was unchanged. Zearalenone, ␣-zearalenol,

␤-zearalenol, and zeranol readily bind to ER, with ␣-zearalanol and zeranol having the highest affinities for the receptor

(Le Guevel and Pakdel, 2001; Leffers et al., 2001). Upon ligand binding to the ER, translocation of the ER to the nucleus

occurs where the ER binds to ER-responsive elements to initiate transcription of specific genes in target tissues (Edwards,

2000; Nettles and Greene, 2005). As observed in the current study, estrogen-mediated responses include maintenance and

development of secondary female sex characteristics, as well as uterine hypertrophy. Contrary to ER-␣, we observed 2.0-

and 3.5-fold increases in ER-␤ mRNA and protein, respectively, in the uterus of prepubertal gilts consuming zearalenone.

Estrogen receptor-␤ is widely distributed in tissues, although not highly expressed in the uterus (Shughure et al., 1998; Wang

et al., 1999). In addition, the uteri of ER- knockout mice appear comparable to wildtype mice and respond to estrogen due

to the presence of ER-␣. These mice have decreased fertility compared to wildtype control mice, but are capable of successful

pregnancies (Krege et al., 1998). Thus, ER␣ is the predominant isoform in the uterus, and it is likely that uterine hypertrophy

observed in the current study was largely mediated through this isoform. However, due to the changes in ER-␤ in gilts

W.T. Oliver et al. / Animal Feed Science and Technology 174 (2012) 79–85 85

consuming zearalenone contaminated feed, it is also likely that ER-␤ was, in part, responsible for uterine growth. Thus, a

larger role for the ER-␤ in uterine growth and development cannot be ruled out.

Contrary to some reports, zearalenone did not increase growth performance in the current study. In addition, activation

of key proteins, as measured by protein phosphorylation, involved in protein synthesis was not down-regulated. Thus, in

spite of cell culture-based evidence, zearalenone and its metabolites, at these doses, do not regulate skeletal muscle protein

synthesis in prepubertal gilts, in vivo. This study confirms earlier reports showing the estrogenic effects on prepubertal gilts,

in that reproductive tract size and endometrial gland development increased in gilts consuming zearalenone contaminated

feed. Zearalenone did not affect several mRNA abundances associated with uterine growth and maintenance, including ER-␣.

However, novel to the current experiment, ER-␤ mRNA and protein abundances increased in gilts consuming zearalenone.

While it is likely that the ER-␣ is the main estrogenic mediator in uterine tissue, the changes in ER-␤ observed indicates a

major role in the growth and (or) maintenance of the uterus in response to zearalenone contaminated feed in prepubertal

gilts. Thus, we conclude that zearalenone had no effect on growth performance or skeletal muscle signaling in prepubertal

gilts, but increased reproductive tract size and glandular development which was due, in part, to altering the expression of ER-␤.

References

Abid-Essefi, S., Ouanes, Z., Hassen, W., Baudrimont, I., Creppy, E., Bacha, H., 2004. Cytotoxicity, inhibition of DNA and protein syntheses and oxidative

damage in cultured cells exposed to zearalenone. Toxicol. In Vitro 18, 467–474.

Avruch, J., Lin, Y., Long, X., Murthy, S., Ortiz-Vega, S., 2005. Recent advances in the regulation of the TOR pathway by insulin and nutrients. Curr. Opin. Clin.

Nutr. Metab. Care 8, 67–72.

BML, 2000. The German Federal Ministry of Nutrition, Agriculture and Forestry: orientation values for critical concentrations of deoxynivalenol and

zearalenone in diets for pigs, ruminants and gallinaceous poultry. VDM 27/00, 2–3.

Diekman, M.A., Green, M.L., 1992. Mycotoxins and reproduction in domestic livestock. J. Anim. Sci. 70, 1615–1627.

Döll, S., Dänicke, S., Ueberschär, K.H., Valenta, H., Schnurrbusch, U., Ganter, M., Klobasa, F., Flachowsky, G., 2003. Effects of graded levels of Fusarium toxin

contaminated maize in diets for female weaned piglets. Arch. Anim. Nutr. 57, 311–334.

Döll, S., Dänicke, S., Schnurrbusch, U., 2004. The effect of increasing concentrations of Fusarium toxins in piglet diets on histological parameters of the

uterus and vagina. Arch. Anim. Nutr. 58, 413–417.

Edwards, D.P., 2000. The role of coactivators and corepressors in the biology and mechanism of action of hormone receptors. J. Mammary Gland

Biol. Neoplasia 5, 307–324.

Jiang, S.Z., Yang, Z.B., Yang, W.R., Yao, B.Q., Zhao, H., Liu, F.X., Chen, C.C., Chi, F., 2010. Effects of feeding purified zearalenone contaminated diets with or

without clay enterosorbent on growth, nutrient availability, and genital organs in post-weaning female pigs. Asian-Aust. J. Anim. Sci. 23, 74–81.

Kordic, B., Pribicevic, S., Muntanol-Cvetkovic, M., Nicolic, P., Nicolic, B., 1992. Experimental studies of the effects of known quantities of zearalenone on

swine reproduction. J. Environ. Pathol. Toxicol. Oncol. 11, 53–55.

Krege, J.H., Hodgin, J.B., Couse, J.F., Enmark, E., Warner, M., Mahler, J.F., Sar, M., Korach, K.S., Gustafsson, J.A., Smithies, O., 1998. Generation and reproductive

phenotypes of mice lacking estrogen receptor ␤. Proc. Natl. Acad. Sci. U.S.A. 95, 15677–15682.

Le Guevel, R., Pakdel, F., 2001. Assessment of oestrogenic potency of chemicals used as growth promoter by in vitro methods. Hum. Reprod. 16, 1030–1036.

Leffers, H., Naesby, M., Vendelbo, B., Skakkeback, N.E., Jorgensen, M., 2001. Oestrogenic potencies of Zeranol, oestradiol, diethylstilboestrol, Bisphenol-A,

and : implications for exposure assessment of potential endocrine disrupters. Hum. Reprod. 16, 1037–1045.

Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-time quantitative pcr and the 2(-delta delta c(t)) method. Methods

25, 402–408.

Mueller, S.O., Simon, S., Chae, K., Metzler, M., Korach, K.S., 2004. and their human metabolites show distinct agonistic and antagonistic

properties on estrogen receptor ␣ (ER␣) and ER␤ in human cells. Toxicol. Sci. 80, 14–25.

Nettles, K.W., Greene, G.L., 2005. Ligand control of coregulator recruitment to nuclear receptors. Annu. Rev. Physiol. 37, 309–333.

National Research Council, 1998. Nutrient Requirements of Swine. Tenth ed. National Academy Press, Washington, DC.

Oliver, W.T., Touchette, K.J., Coalson, J.A., Whisnant, C.S., Brown, J.A., Mathews Oliver, S.A., Odle, J., Harrell, R.J., 2005. Pigs weaned from the sow at 10 d of

age respond to porcine somatotropin and dietary energy source of manufactured liquid diets. J. Anim. Sci. 83, 1002–1009.

Oliver, W.T., Miles, J.R., 2010. A low-fat liquid diet increases protein accretion and alters cellular signaling for protein synthesis in 10-day-old pigs. J. Anim.

Sci. 99, 2576–2584.

Riley, R.T., Petska, J., 2005. Mycotoxins: metabolism, mechanisms and biochemical markers. In: Diaz, D.E. (Ed.), The Mycotoxin Blue Book. Nottingham

University Press, UK, pp. 279–294.

Rotter, B.A., Prelusky, D.B., Pestka, J.J., 1996. Toxicology of deoxynivalenol (vomitoxin). J. Toxicol. Environ. Health 48, 1–34.

Shughure, P.J., Lane, M.V., Scrimo, P.J., Merchenthaler, I., 1998. Tissue distribution and quantitative analysis of estrogen receptor-␣ (ER-␣) and ␤ (ER-␤)

mRNA in the rat pituitary, gonad, and reproductive tract. 63, 498–504.

Simms, D.D., Goehring, T.B., Brandt Jr., R.T., Kuhl, G.L., Higgins, J.J., Laudert, S.B., Lee, R.W., 1988. Effect of sequential implanting with zeranol on steer lifetime

performance. J. Anim. Sci. 66, 2736–2741.

Smith, P.K., Krohn, R.I., Hermanson, G.T., Mallia, A.K., Gartner, F.H., Provenzano, M.D., Fujimoto, E.K., Goeke, N.M., Olson, B.J., Klenk, D.C., 1985. Measurement

of protein using bicinchoninic acid. Anal. Biochem. 150, 76–85.

Songsermsakul, P., Sontag, G., Cichna-Markl, M., Zentek, J., Razzazi-Fazeli, E., 2006. Determination of zearalenone and its metabolites in urine, plasma and

faeces of horses by HPLC-APCI–MS. J. Chromatogr. B 843, 252–261.

Takemura, H., Shim, J., Sayama, K., Tsubura, A., Zhu, B.T., Shimoi, K., 2007. Characterization of the estrogenic activities of zearalenone and zeranol in vivo

and in vitro. J. Steroid Biochem. Mol. Biol. 103, 170–177.

Tiemann, U., Dänicke, S., 2007. In vivo and in vitro effects of the mycotoxins zearalenone and deoxynivalenol on different non-reproductive and reproductive

organs in female pigs: a review. Food Addit. Contam. 24, 306–314.

Wang, H., Masironi, B., Eriksson, H., Sahlin, L., 1999. A comparative study of estrogen receptors ␣ and ␤ in the rat uterus. Biol. Reprod. 61, 955–964.

Williams, J.E., Ireland, S.J., Mollett, T.A., Hancock, D.L., Beaver, E.E., Hannah, S., 1991. Influence of zeranol and breed on growth, composition of gain, and

plasma hormone. J. Anim. Sci. 69, 1688–1696.

Wollenhaupt, K., Jonas, L., Tiemann, U., Tomek, W., 2004. Influence of the mycotoxins ␣- and ␤-zearalenol (ZOL) on regulators of cap-dependent translation

control in pig endometrial cells. Reprod. Toxicol. 19, 189–199.

Zöllner, P., Jodlbauer, J., Kleinova, M., Kahlbacher, H., Kuhn, T., Hochsteiner, W., Lindner, W., 2002. Concentration levels of zearalenone and its metabolites

in urine, muscle, tissue, and liver samples of pigs fed with mycotoxin-contaminated oats. J. Agric. Food Chem. 50, 2494–2501.