ArticlePDF Available

Gene expression in the arcuate nucleus of heifers is affected by controlled intake of high- and low-concentrate diets

Authors:

Abstract and Figures

It was hypothesized that a high-concentrate diet fed during early calfhood alters the expression of genes within the arcuate nucleus that subserve reproductive competence. Beef heifers (n = 12) were weaned at approximately 3 mo of age, and after acclimation, were allocated randomly to 1 of 2 nutritional groups: 1) High Concentrate/High Gain (HC/HG), a high concentrate diet fed to promote a gain of 0.91 kg/d; or 2) High Forage/Low Gain (HF/LG), a forage-based diet fed to promote a gain of 0.45 kg/d. Experimental diets were fed under controlled intake for 91 d. At the end of 91 d, heifers were slaughtered by humane procedures, blood samples were collected, brains were removed, liver weights were determined, and rumen fluid was collected for VFA analyses. Tissue blocks containing the hypothalamus were dissected from the brains, frozen, and cut using a cryostat, and frozen sections were mounted on slides. Tissue from the arcuate nucleus (ARC) was dissected from sections for mRNA extraction. Microarray analysis was used to assess genome-wide transcription in the ARC using a 60-mer oligonucleotide 44K bovine expression array. The ADG was greater (P < 0.001) in heifers fed the HC/HG diet than in heifers fed the HF/LG diet. At slaughter, mean propionate to acetate ratios in the ruminal fluid and liver weight as a percentage of BW were increased (P < 0.005) in HC/HG compared with HF/LG heifers. Mean serum concentrations of insulin (P < 0.05) and IGF-1 (P < 0.005) were greater, and leptin tended to be greater (P = 0.1) in HC/HG heifers compared with HF/LG heifers. Approximately 345 genes were observed to be differentially expressed in the HC/HG group with approximately two-thirds of the genes exhibiting increased expression in the HC/HG group. Genes exhibiting decreased expression in the HC/HG group included agouti-related protein and neuropeptide Y, products of which are known to regulate feed intake and energy expenditure. Functional annotation of enriched Gene Ontology terms indicates that a number of biological processes within the hypothalamus are affected by consumption of high-concentrate diets, including those related to control of feed intake, regulation of cellular metabolic processes, receptor and intracellular signaling, and neuronal communication. In summary, dietary treatments shown previously to accelerate the timing of pubertal onset in heifers increased ruminal propionate, promoted enhanced metabolic hormone secretion, and altered gene expression in the ARC.
Content may be subject to copyright.
2222
Key words: agouti-related protein, arcuate nucleus, bovine, leptin, neuropeptide Y, puberty
© 2012 American Society of Animal Science. All rights reserved. J. Anim. Sci. 2012.90:2222–2232
doi:10.2527/jas2011-4684
ABSTRACT: It was hypothesized that a high-concen-
trate diet fed during early calfhood alters the expression
of genes within the arcuate nucleus that subserve repro-
ductive competence. Beef heifers (n = 12) were weaned
at approximately 3 mo of age, and after acclimation, were
allocated randomly to 1 of 2 nutritional groups: 1) High
Concentrate/High Gain (HC/HG), a high concentrate diet
fed to promote a gain of 0.91 kg/d; or 2) High Forage/
Low Gain (HF/LG), a forage-based diet fed to promote a
gain of 0.45kg/d. Experimental diets were fed under con-
trolled intake for 91 d. At the end of 91 d, heifers were
slaughtered by humane procedures, blood samples were
collected, brains were removed, liver weights were deter-
mined, and rumen uid was collected for VFA analyses.
Tissue blocks containing the hypothalamus were dis-
sected from the brains, frozen, and cut using a cryostat,
and frozen sections were mounted on slides. Tissue from
the arcuate nucleus (ARC) was dissected from sections
for mRNA extraction. Microarray analysis was used to
assess genome-wide transcription in the ARC using a
60-mer oligonucleotide 44K bovine expression array.
The ADG was greater (P < 0.001) in heifers fed the HC/
HG diet than in heifers fed the HF/LG diet. At slaughter,
mean propionate to acetate ratios in the ruminal uid and
liver weight as a percentage of BW were increased (P <
0.005) in HC/HG compared with HF/LG heifers. Mean
serum concentrations of insulin (P < 0.05) and IGF-1 (P <
0.005) were greater, and leptin tended to be greater (P =
0.1) in HC/HG heifers compared with HF/LG heifers.
Approximately 345 genes were observed to be differen-
tially expressed in the HC/HG group with approximately
two-thirds of the genes exhibiting increased expression in
the HC/HG group. Genes exhibiting decreased expression
in the HC/HG group included agouti-related protein and
neuropeptide Y, products of which are known to regulate
feed intake and energy expenditure. Functional annota-
tion of enriched Gene Ontology terms indicates that a
number of biological processes within the hypothala-
mus are affected by consumption of high-concentrate
diets, including those related to control of feed intake,
regulation of cellular metabolic processes, receptor and
intracellular signaling, and neuronal communication. In
summary, dietary treatments shown previously to accel-
erate the timing of pubertal onset in heifers increased
ruminal propionate, promoted enhanced metabolic hor-
mone secretion, and altered gene expression in the ARC.
Gene expression in the arcuate nucleus of heifers is affected by
controlled intake of high- and low-concentrate diets1
C. C. Allen,*† B. R. C. Alves,† X. Li, ‡ L. O. Tedeschi,† H. Zhou, ‡ J. C. Paschal,§
P. K. Riggs,† U. M. Braga-Neto,# D. H. Keisler, G. L. Williams,*† and M. Amstalden†2
*Animal Reproduction Laboratory, Texas AgriLife Research, Beeville 78102;
Departments of †Animal Science, ‡Poultry Science, and #Electrical and Computer Engineering,
Texas A&M University, College Station 77843; §Texas AgriLife Extension, Texas AgriLife Research and
Extension Center, Corpus Christi 78406; and Division of Animal Sciences, University of Missouri, Columbia 65211
INTRODUCTION
Age at onset of puberty is largely dependent upon
rate of growth during the prepubertal period. Nutrient
restriction during postnatal development delays puber-
ty (Day et al., 1984) by inhibiting the release of GnRH
(I’Anson et al., 2000). Hormones such as insulin (Adam
and Findlay, 1998), IGF-1 (Hiney et al., 1991; Yelich et
al., 1996), GH (Simpson et al., 1991), and leptin (Garcia
1
Supported by Texas Beef Initiative grant 114443, Texas AgriLife
Research projects H-6881 and TEX09202 of the Texas A&M System,
and Agriculture and Food Research Initiative Competitive Grant no.
2009-65203-05678 from the USDA National Institute of Food and
Agriculture. We acknowledge Kelli Kochan, Randel Franke, and Ray
Villarreal for providing technical assistance and A. F. Parlow, NIDDK
National Hormone and Peptide Program, for providing IGF-1 antiserum.
2Corresponding author: m.amstalden@tamu.edu
Received September 10, 2011.
Accepted January 10, 2012.
Gene expression in arcuate nucleus of heifers 2223
et al., 2003), and nutrients such as glucose (Adam and
Findlay, 1998) and fatty acids (Garcia et al., 2003) have
been implicated in signaling nutritional status. These
signals act within the hypothalamus to regulate feed in-
take, energy expenditure, and neuroendocrine functions,
including reproduction (Hiney et al., 1996; Zieba et al.,
2004; Stanley et al., 2005). Although multiple hypotha-
lamic areas are involved in the pathways mediating the
metabolic regulation of neuroendocrine function, major
metabolic-sensing neurons are located within the arcu-
ate nucleus (ARC) of the hypothalamus. Neuropeptide
Y (NPY)/agouti-related protein (AgRP) neurons present
in the ARC (Broberger et al., 1998; Hahn et al., 1998)
are responsive to changes in metabolic status (McShane
et al., 1993; Stanley et al., 2005) and appear to play a
major role in mediating the effects of nutrition on repro-
duction (Raposinho et al., 1999).
In maturing heifers, accelerated growth during early
calfhood in uences age at puberty. Heifers weaned at
3 mo of age and fed diets that promote rapid rates of
BW gain between 3 and 7 mo of age reach puberty ear-
lier and at lighter BW than heifers fed diets to gain BW
at a reduced rate during the same period (Gasser et al.,
2006a–d). Increased BW gain later during calfhood does
not have the same impact. Therefore, a critical window
for nutritional imprinting of neuroendocrine functions
that regulate age at onset of puberty appears to exist ear-
ly in juvenile development. In the study reported herein,
we examined whether nutritional inputs during early
calfhood result in changes in gene expression within the
ARC that may be critical for the integration of metabolic
and reproductive functions. Greatest emphasis was giv-
en to genes that have been well characterized relative to
their involvement in sensing nutritional inputs.
MATERIALS AND METHODS
All animal-related procedures used in this study
were approved by the Institutional Agricultural Animal
Care and Use Committee of the Texas A&M System.
Animal Procedures
Twelve Angus-sired heifers (½ Angus, ¼ Hereford,
¼ Brahman) were weaned at approximately 3 mo of age,
strati ed by date of birth, and assigned randomly to 1 of
2 dietary treatments (n = 6/treatment) in 2 replicates (n =
6/replicate): 1) High-Forage/Low-Gain (HF/LG): ADG
of 0.45 kg/d (n = 6); and 2) High-Concentrate/High-
Gain (HC/HG): ADG of 0.91 kg/d (n = 6). Diets were
balanced using the Large Ruminant Nutrition System
(http://nutritionmodels.tamu.edu/lrns.htm), which is
based on the Cornell net Carbohydrate and Protein
System as described by Fox et al. (2004). Targeted ADG
was attained by adjustments in DMI based on ADG de-
termined weekly. Ingredients and diet chemical compo-
sitions are presented in Table 1.
Heifers in each group were allocated to pens measur-
ing 25.9 m × 9.5 m and fed an acclimation diet for 2 wk
post-weaning. During the rst week of the acclimation
period, heifers in both treatments were fed the HF diet
up to a maximum of 2.7 kg/heifer daily. During the sec-
ond week of the adaptation period, heifers assigned to the
HC/HG group were fed a diet consisting of 50% HF and
50% HC up to a maximum of 2.7 kg/heifer daily. Heifers
assigned to the HF/LG treatment were fed the HF diet
through the second week of the adaptation period. After
the 2-wk adaptation period, heifers were fed 100% of
their respective treatment diets for 14 wk. Heifers were
weighed weekly for the duration of the experiment.
At the completion of the experimental feeding pe-
riod, heifers were slaughtered by humane procedures
after overnight fasting. A block of tissue containing the
septum, preoptic area, and hypothalamus was dissected
and frozen in liquid-nitrogen vapor. Tissue blocks were
stored at 80°C until further processing. A composite
of ruminal uid sample was obtained from each heifer
immediately post mortem by mixing 3 subsamples of
ruminal uid collected from the dorsal, ventral, and
caudal aspects of the rumen. The ruminal uid samples
were processed as described subsequently and frozen at
20°C for determination of VFA pro le. A single blood
sample (15 mL) was collected and liver weights were
obtained at slaughter. Blood samples were placed on ice
immediately after collection. Serum was obtained from
blood samples by centrifugation (2,200 × g for 20 min at
4°C) and stored at 20°C.
Hormone Assays
Circulating concentrations of insulin, IGF-I, and
leptin were determined in serum samples collected at
Table 1. Ingredients and chemical composition of high-
forage (HF) and high-concentrate (HC) diets fed to heif-
ers during the study
Item HF HC
Ingredients
Cracked corn, % 39.03 50.75
Soybean meal, % 17.61
Chopped coastal Bermuda grass hay, % 3.47 26.95
Dehydrated alfalfa meal pellet, % 57.30 3.83
Calcium carbonate, % 0.86
Calcium monophosphate, % 0.20
Vitamin A/D/E premix, mg/kg 71 73
Chemical composition
ME, Mcal/kg 0.48 0.53
CP, % DM 14.3 17.5
Digestible intake protein, % 66 67
Allen et al.
2224
the time of slaughter. Concentrations of insulin were
determined by a single-phase RIA kit (Coat-A-Count;
Siemens, Los Angeles, CA) reported previously for
bovine serum (DiCostanzo et al., 1999; Accorsi et al.,
2005). However, we used a bovine insulin preparation
for standards and references instead of the human insu-
lin standards provided with the kit. Minimum detectable
concentrations were 0.1 ng/mL and intra- and inter-as-
say CV were 9.8% and 14.5%, respectively. Circulating
concentrations of IGF-1 were determined in triplicate
samples as reported previously (Ryan et al., 1994), ex-
cept that we used a rabbit anti-IGF-1 serum provided
by the National Hormone and Pituitary Program (NHPP,
Torrance, CA). Concentrations of IGF-1 in samples col-
lected at slaughter were determined in a single assay and
intra-assay CV was 19%. Circulating concentrations of
leptin were determined in a single RIA as described pre-
viously (Delavaud et al., 2000). Sensitivity of the assay
was 0.1 ng/mL and intra-assay CV averaged 12%.
Volatile Fatty Acid Analysis of Ruminal Fluid
Ruminal uid collected from experimental animals
at slaughter was mixed brie y, transferred to 15-mL
conical tubes, centrifuged, and the aqueous portion was
transferred to a fresh 15-mL conical tube. Samples were
stored at 20°C until analysis for VFA composition us-
ing GLC (Salanitro and Muirhead, 1975).
Tissue Processing
Frozen blocks of diencephalic tissue were cut in
coronal sections of 20 m using a cryostat. Tissue sec-
tions were thaw-mounted on Superfrost/Plus glass mi-
croscope slides (Fisher Scienti c, Waltham, MA) and
frozen immediately. Slides were then stored at 80°C
until processing.
RNA Isolation and Extraction
A single series of tissue sections from the hypothal-
amus of each heifer containing sections 200 m apart
was processed for Cresyl violet staining and observed
using bright- and dark- eld microscopy to determine
the location of the ARC. Location of the ARC was
based upon identi cation of well-established anatomi-
cal markers for rat (Pellegrino et al., 1979) and sheep
(Lehman et al., 1993) brains. A separate series of sec-
tions containing the ARC was used for tissue dissection
and RNA isolation. Using a 25-ga needle, an area of ap-
proximately 1 mm in diameter encompassing the ARC
was scraped from the slides (Figure 1). Scraped tissue
was placed immediately in lysis solution (RNAqueous-
Micro; Ambion, Austin, TX). Sections were maintained
frozen during scraping and, on average, approximately
48 sections from each heifer were used. Total RNA was
collected from tissue scrapes using RNAqueous-Micro
(Ambion) according to manufacturer’s instructions for
the laser-capture micro-dissected tissue protocol, except
that RNA was harvested into 400 L of lysis solution
(RNAqueous-Micro) and precipitated with 200 L of
100% ethanol. Samples were incubated in elution solu-
tion (RNAqueous-Micro) for 5 min before being elut-
ed through the column. Total RNA isolated was treat-
ed with DNase to remove genomic DNA. Quantity of
RNA was determined by spectrophotometry (NanoDrop
ND-1000; ThermoFisher Scienti c, Wilmington, DE).
Quality of RNA was determined using the RNA 6000
Pico Kit (Agilent Technologies, Santa Clara, CA), ac-
cording to manufacturer’s instructions.
Figure 1. Cresyl violet-stained section through the mediobasal hypo-
thalamus depicting an area (asterisk) of the arcuate nucleus dissected for RNA
extraction. Dissection was based on anatomical landmarks: ARC = arcuate
nucleus; FX = fornix; me = median eminence; VMH = ventromedial hypo-
thalamus; 3V = third ventricle. Scale bar = 500 m.
Gene expression in arcuate nucleus of heifers 2225
Microarray Procedure and Analysis
cRNA Labeling. Total RNA (120 ng) from each
heifer was initially reverse transcribed to cDNA us-
ing the 2-color Quick Amp Labeling Kit (Agilent
Technologies). Labeled cRNA was synthesized by in
vitro-transcription of cDNA in presence of either cya-
nine-3 (Cy3) or cyanine-5 (Cy5; RNA Spike-In Kit,
Two-Color; Agilent Technologies) as per manufacturer’s
instructions. One-half of the samples from each dietary
group were labeled with Cy3 and the other half with
Cy5. Labeled cRNA was puri ed using RNeasy Mini
columns (Qiagen, Valencia, CA). Yield and speci city
of cRNA were determined based on uorospectrometry
(NanoDrop ND-1000; ThermoFisher Scienti c).
Microarray Hybridization. Microarray hybrid-
izations were conducted using the Agilent two-dye 4
× 44 K bovine gene expression array (G2514; Agilent
Technologies). Each array contained 44,407 oligo probes,
1,264 of which were positive control probes, and 153
were negative control probes. Each probe was replicat-
ed twice on each array; therefore, each array contained
21,495 unique oligos.
Hybridizations were performed using the Gene
Expression Hybridization Kit (Agilent Technologies)
following manufacturer instructions. Six array hybrid-
izations were conducted using a dye-swap design. Three
of the hybridizations were performed with Cy3-labeled
cRNA from each of 3 heifers in the HC/HG group and
Cy5-labeled cRNA from each of 3 heifers in the HF/LG
group. The remaining 3 hybridizations were performed
with Cy5-labeled cRNA from each of 3 heifers in the
HC/HG group and Cy3-labeled cRNA from each of 3
heifers in the HF/LG group. The dye-swap design result-
ed in each array being hybridized to either Cy3-labeled
HC/HG cRNA and Cy5-labeled HF-LG samples, or
Cy5-labeled HC/HG and Cy3-labeled HF/LG samples.
Arrays were incubated at 65°C with rotation for 17 h in
a microarray hybridization chamber. After hybridization,
arrays were washed according to the Agilent Two-Color
Microarray protocol (Agilent Technologies).
Microarray Imaging, Data Acquisition,
Normalization and Analysis. Agilent arrays were
scanned at 5-m resolution on an Axon GenePix 4100
scanner (Molecular Devices Corporation, Sunnyvale,
CA). Signal intensities were quanti ed using the GenePix
pro 6.0 software (Molecular Devices Corporation,
Downingtown, PA) and following procedures described
previously (Li et al., 2008). Normalization of log-ratios
was accomplished via intensity-dependent nonlinear
location shift using Loess regression. Differential ex-
pression was assessed by tting a linear model to each
probe, expressing the observed log-ratio as the sum of a
probe dye-effect, the true log-ratio between the 2 dietary
groups, plus 0-mean noise. The presence of a signi cant
true non-zero log-ratio between the dietary groups (thus
removing the dye effect) was tested for each probe us-
ing a Bayesian procedure described in Smyth (2004).
The resulting P-value is moderated by using a SD that
borrows information from correlated probes, thereby in-
creasing statistical power. In addition, statistical power
is further increased by taking into consideration in the
analysis the estimated correlation between pairs of du-
plicated spots. The analysis is weighted such that only
probes agged as good quality by the GenePix software
are used. The multiple-testing problem created by test-
ing each probe was addressed by adjusting the P-values
according to the False-Discovery Rate (FDR) method
(Benjamini and Hochberg, 1995).
Bioinformatics. Functional annotation of differen-
tially expressed genes was performed using the Database
for Annotation, Visualization and Integrated Discovery
(DAVID) bioinformatics resources (Huang et al., 2009).
Gene Ontology terms for Biological Process containing
3 or more genes with > 1.5-fold enrichment were con-
sidered signi cant clusters. Terms for Biological Process
were chosen because they represent the most relevant
component of the Gene Ontology annotation for this study.
Quantitative Reverse Transcription-PCR
Four genes exhibiting differential expression in the
microarray experiment were selected for quantitative real-
time reverse transcription-PCR analysis. Glyceraldehyde-
3-phosphate dehydrogenase (GAPDH) and ribosomal
protein L19 (RPL19) were used as reference control
genes. Primers used for real-time reverse transcription-
PCR were designed using Oligo 6 software (Molecular
Biology Insights, Inc., Cascade, CO) and sequences
are shown in Table 2. Speci city of primer sequences
were investigated using the Basic Local Alignment Tool
(BLAST; National Library of Medicine). Ampli cation
ef ciency was > 90% for all primer pairs.
Total RNA (20 to 200 ng) isolated from the ARC was
reverse transcribed to cDNA using the High Capacity
cDNA Reverse Transcription Kit (Applied Biosystems,
Foster City, CA) and oligo (dT)
20 primer (Integrated
DNA Technologies, Coralville, IA). Brie y, PCR reac-
tions contained 2 L cDNA (diluted 1:4 in 25 ng/L
yeast tRNA), 10 L SYBR GreenER PCR master mix
(Invitrogen, Carlsbad, CA), 0.6 L of each forward and
reverse primers, and RNase-free water to a nal volume
of 20 L. Reactions were performed in duplicates for
each sample. Quantitative real-time reverse transcrip-
tion-PCR was carried out in 96-well plates using the
ABI Prism 7900HT sequence detection system (Applied
Biosystems). Cycling conditions were 95°C for 10 min
followed by 40 cycles of 95°C for 15 s and 60°C for
Allen et al.
2226
1 min. A nal cycle with a slow increase in temperature
to 95°C was used to produce a dissociation curve of PCR
products and con rm the absence of non-speci c ampli-
cation. No-template control was used to verify the ab-
sence of primer-dimer ampli cation. Negative control
reactions (no reverse transcriptase) con rmed the ab-
sence of genomic DNA carryover in RNA preparations.
Quantitative reverse transcription-PCR data
were analyzed by relative quanti cation (Livak and
Schmittgen, 2001). Threshold cycle (Ct) data for each
gene was normalized to the geometric mean Ct values
of the control genes GAPDH and RPL19 for each sam-
ple (Ct), and transformed to the average expression
of HF/LG samples (Ct). The VBA applet, geNorm
(Vandesompele et al., 2002) was used to determine sta-
bility of control genes. Mean fold change in the HC/HG
group was compared with the HF/LG group.
Statistical Analysis
Initial and nal BW, cumulative ADG, liver weight
(as percentage of BW), ruminal propionate to acetate ra-
tio, hormone concentrations at slaughter, and mean fold
change of selected differentially expressed genes were
analyzed using the PROC MIXED procedure (SAS Inst.
Inc., Cary, NC). Dietary treatment, replicate, and the
treatment by replicate interaction were used as sources
of variation. Liver weight as a percent of BW and pro-
pionate to acetate ratio were transformed using the arc-
sine of the square root method to normalize data before
performing the statistical analysis.
RESULTS
Performance, Ruminal Volatile Fatty Acid, and
Metabolic Hormone Measures
Mean BW at the beginning of the study did not dif-
fer between groups (HF/LG, 139.7 ± 6.4 kg; HC/HG,
127 ± 6.4 kg; P > 0.1). Body weight increased linearly in
both dietary groups (Figure 2). As expected based on the
experimental design, ADG was greater (P < 0.0001) in
HC/HG heifers (0.98 ± 0.05 kg/d) than in HF/LG heifers
(0.51 ± 0.06 kg/d).
Mean BW, mean liver weight (as a percentage of
BW), and propionate to acetate ratio in the ruminal uid
were greater (P < 0.05) at the time of slaughter in HC/
HG heifers than in HF/LG heifers (Table 3). Mean cir-
culating concentrations of insulin (P < 0.05) and IGF-1
(P < 0.005) were greater at the time of slaughter in HC/
HG heifers than in HF/LG heifers, and circulating con-
centrations of leptin tended to be greater (P = 0.1) in HC/
HG heifers (Table 3).
Microarray Gene Expression
A total of 346 probes presented a signi cant non-
zero log ratio between the HC/HG and HF/LG groups at
a signi cance level of 0.05 after multiple-test correction
by the FDR method. Therefore, 346 unique sequences
Figure 2. Mean (± SEM) BW of heifers weaned at 3 mo of age and
fed either a High-Forage/Low-Gain (HF/LG; n = 6) or a High-Concentrate/
High-Gain (HC/HG; n = 6) diet for 14 wk. Body weight of HC/HG heifers
was greater than HF/LG heifers beginning at wk 11 (P < 0.06) and continuing
through wk 14 (P < 0.01 to P < 0.03).
Table 2. Gene name, symbol, primer sequence, and accession number for all genes validated by quantitative reverse
transcription PCR
Gene name Gene symbol Primer sequences (forward/reverse) Accession number
Agouti-related protein AGRP GAAGAGGATAACGAACAG
CAGGGGTTCGTGGTGGGTA
NM_173983
GH receptor GHR ATCACCACAGAAAGCCTTACCACTA
GACAGGTATCTCAGAACTTGGAAC
NM_176608
Neuropeptide Y NPY AAGCAGAGATACGGGAAACGA
ATTGGGAGGACTGGCAGACT
NM_001014845
Prolactin-releasing hormone receptor PRLHR AGGGAGTGAAGGAGCAATAAGCA
GAAGGTAATGGGTTTGAATGGACTA
NM_001030300
Glyceraldehyde-3- phosphate dehydrogenase GAPDH CAGCGACACTCACTCTTCTACCTT
GAACTCTTCCTCTCGTGCTCCT
NM_001034034
Ribosomal protein L19 RPL19 ACCCCAATGAGACCAATGAA
GCAGTACCCTTTCGCTTACCTAT
NC_007317
Gene expression in arcuate nucleus of heifers 2227
(genes) were observed to be differentially expressed in
the ARC of heifers in the HC/HG group compared with
the HF/LG group (adjusted P < 0.05). Among the dif-
ferentially expressed genes, 229 exhibited increased and
117 exhibited decreased expression in HC/HG heifers
compared with HF/LG heifers. Functional annotation of
differentially expressed genes identi ed enriched Gene
Ontology (GO) terms for biological process in clusters
that included responses to hormones and nutrients, feed-
ing behavior, intracellular signaling, cell to cell commu-
nication and synaptic transmission (Tables 4 and 5). One
of the clusters included down-regulated genes in the ARC
of HC/HG heifers that are involved in regulating feed-
ing behavior. Genes in this cluster included AGRP, NPY,
and prolactin-releasing hormone receptor (PRLHR).
Additional clusters of down-regulated genes in HC/HG
heifers include genes involved in the response to hor-
mones and nutrients, and metabolic processes. Genes
in these clusters included GH receptor (GHR) and sig-
nal transducer and activator of transcription 1 (STAT1),
deiodinase type II (DIO2), and retinol-binding protein 1
(RBP1). Clusters representing genes that were up reg-
ulated in HC/HG heifers included signaling molecules
(phospholipase D2, adenyl cyclase type 1, sphingosine
kinase, tyrosine kinase, and dynamin 2), and molecules
involved in synaptic transmission (voltage-gated potas-
sium channel-interacting protein 2, gamma amino butyr-
ic acid A receptor delta). Genes associated with control
of feed intake were also up-regulated in HC/HG heifers
and included pro-opiomelanocortin (POMC), the gene
encoding the precursor molecule for alpha-melanocyte-
stimulating hormone (α-MSH). A partial list of up-reg-
ulated and down-regulated genes with greatest changes
in the ARC of HC/HG heifers is depicted in Table 6. A
complete list of differentially expressed genes identi ed
by the microarray analysis is available (see Supplemental
Tables; available in the online version of this paper).
Quantitative Reverse Transcription
PCR of Selected Genes
Neuropeptide Y, AGRP, GHR, and PRLHR genes
were selected for further analysis from the 346 differ-
entially expressed genes because of their known roles
in signaling metabolic status in the hypothalamus and
regulating energy expenditure. Quantitative reverse
transcription-PCR analysis of these genes con rmed
microarray observations and indicated that mean expres-
sion of NPY (P < 0.001), AGRP (P < 0.001), GHR (P <
0.02), and PRLHR (P < 0.06) genes was less in HC/HG
heifers than in HF/LG heifers (Figure 3).
DISCUSSION
The ARC is a major area of the hypothalamus that
integrates complex signaling pathways regulating met-
abolic and reproductive functions. The ARC region of
the hypothalamus is populated by important metabolic-
sensing neurons (Broberger et al., 1998; Hahn et al.,
1998), as well as neurons implicated in pubertal devel-
opment (Redmond et al., 2011). Because the initiation
of puberty is greatly in uenced by nutritional balance
and metabolic status, we sought to examine an array of
genes within the ARC that may be responsive to, or may
mediate the effects of, nutritional inputs known to affect
the process of sexual maturation. The precocious puber-
ty model in heifers described by Gasser et al. (2006a–d)
provides an ideal approach for examining the involve-
ment of the ARC in the nutritional regulation of neuro-
endocrine function during early juvenile development.
The microarray and computational methods used in the
Table 4. Functional annotation of most signi cant enriched
Gene Ontology terms for Biological Process of down-regu-
lated genes in the arcuate nucleus of heifers gaining BW at
high rates (High-Concentrate/High-Gain group)
Biological
process term
Fold
enrichment
No. of
genes
Cellular response to insulin stimulus 9.82 4
Feeding behavior 9.68 4
Hormone metabolic process 7.88 5
Cellular response to hormone stimulus 6.68 5
Regulation of hormone concentrations 5.53 5
Response to peptide hormone stimulus 5.42 5
Gland development 4.95 4
Response to extracellular stimulus 4.55 6
Response to nutrient quantities 4.24 5
Response to endogenous stimulus 3.30 8
Response to hormone stimulus 3.19 7
Table 3. Mean (± SEM) BW, liver weight, rumen pro-
pionate to acetate ratio, and serum concentrations of
metabolic hormones at the time of slaughter in heifers
fed High-Forage/Low-Gain (HF/LG; n = 6) or High-
Concentrate/High-Gain (HC/HG; n = 6) diets for 14 wk
beginning at 16 ± 1 wk of age.
Dietary treatment
Variable HF/LG HC/HG
BW, kg 172.12 ± 9.19a218.49 ± 11.47b
Liver weight, % of BW11.07 ± 0.04c1.42 ± 0.04d
Propionate:acetate1 0.23 ± 0.01c0.31 ± 0.01d
Insulin, ng/mL 0.95 ± 0.07a1.16 ± 0.11b
IGF-1, ng/mL 52.01 ± 4.39c103.75 ± 9.9d
Leptin, ng/mL 2.55 ± 0.35e3.62 ± 0.43f
1Liver weight as percentage of BW and propionate to acetate ratio was
transformed using the arcsine of the square root method to normalize data
before performing statistical analysis. Data shown in the table represents un-
transformed data.
a-fRow means without a common superscript differ (a,b P = 0.05; c,d P <
0.005; e,f P = 0.1).
Allen et al.
2228
current studies provide an opportunity to investigate
gene networks that may be involved in integrating nutri-
ent metabolism and reproductive function. Results from
these studies support the premise that a considerable
number of genes expressed within the ARC are regu-
lated by nutrient intake concurrent with the development
of distinct metabolic and hormonal states. Interestingly,
this differential gene expression was observed in heif-
ers that were under positive nutrient balance, except that
groups differed in the rate at which BW increased.
Using microarray gene expression analysis, we iden-
ti ed a total of 346 genes that were differentially ex-
pressed between HC/HG and HF/LG heifers at the end of
the 14-wk feeding period (approximately at 7 mo of age).
This observation demonstrates that the heifer is exqui-
sitely sensitive to nutritional input during early calf-hood.
Importantly, nutrient requirements for maintenance were
met, and both groups of heifers were in positive energy
balance. However, nutrient availability for growth dif-
fered between groups. Therefore, differences in gene
expression within the ARC between heifers growing at
distinct rates indicate that the state of nutrient suf ciency
to support growth involves functional changes within the
hypothalamus early in development. We propose that
those changes may be important for early maturation of
the reproductive neuroendocrine axis. In mice, neuronal
Table 5. Functional annotation of most signi cant enriched
Gene Ontology terms for Biological Process of up-regulated
genes in the arcuate nucleus of heifers gaining BW at high
rates (High-Concentrate/High-Gain group)
Biological
process term
Fold
enrichment
No. of
genes
Regulation of oxidoreductase activity 9.46 4
Receptor metabolic process 9.15 3
Synaptic vesicle transport 8.60 3
Regulation of amine transport 8.60 3
Cell maturation 6.31 5
Activation of immune response 6.04 6
Feeding behavior 5.48 4
Positive regulation of defense response 5.18 4
Regulation of Rho protein signal transduction 4.78 5
Developmental maturation 4.68 5
Regulation of MAPKKK cascade 4.34 5
Positive regulation of immune response 3.91 6
Positive regulation of response to stimulus 3.61 9
Regulation of cell morphogenesis 3.61 5
Regulation of small GTPase mediated signal transduction 3.38 9
Synaptic transmission 3.17 10
Membrane invagination 3.01 7
Phospholipic metabolic process 2.99 6
Regulation of phosphorylation 2.84 14
Transmission of nerve impulse 2.70 10
Cell-cell signaling 2.68 17
Regulation of protein kinase cascade 2.66 7
Positive regulation of cell communication 2.59 9
Positive regulation of signal transduction 2.57 8
Membrane organization 2.48 10
Regulation of kinase activity 2.38 9
Intracellular signaling cascade 1.81 24
Figure 3. Normalized mean (± SEM) expression of neuropeptide-Y
(NPY), agouti-related protein (AGRP), growth hormone receptor (GHR), and
prolactin-releasing hormone receptor (PRLHR) genes in the arcuate nucleus
of heifers fed a high-forage diet to gain BW at a slow rate (HF/LG; n = 6) and
heifers fed a high-concentrate diet to gain BW at a rapid rate (HC/HG; n = 6).
Expression is relative to mean normalized expression of HF/LG samples in
the experiment. Statistical differences between HF/LG and HC/HG are indi-
cated: *P < 0.001; **P < 0.02; ***P < 0.06.
Table 6. Partial list of differentially-expressed genes in
the arcuate nucleus of heifers fed high-concentrate diets
to gain BW at a rapid rate (High-Concentrate/High-Gain
group)1
Gene symbol Gene description Fold change2
Up-regulated
IGFLR1 IGF-like family receptor 1 1.54
GTF2H5 General transcription factor IIH, polypeptide 5 1.02
GPR45 G protein-coupled receptor 45 1.01
AGOUTI Agouti protein 0.91
LOC513294 Similar to SEC14p-like protein TAP3 0.88
LOC539711 Similar to dedicator of cytokinesis 6 0.82
DSPP Dentin sialophosphoprotein 0.80
TNS4 Tensin 4 0.74
TLR6 Toll-like receptor 6 0.74
RRAD Ras-related associated with diabetes 0.71
Down-regulated
AGRP Agouti related protein -2.36
ORM1 Alpha-1 acid glycoprotein -2.10
NPY Neuropeptide Y -1.65
LOC538993 KIAA0748 ortholog -0.92
CRYM Crystallin, mu -0.90
COL9A3 Collagen, type IX, alpha 3 -0.88
CB456458 Idothyronine, Type II (DIO2) transcript variant 1 -0.83
CRH Corticotropin releasing hormone -0.72
LOC519502 Similar to glutamate receptor interacting protein 1 -0.70
LOC525861 Similar to Homeobox protein orthodpedia -0.68
1Complete list is provided as Supplemental Tables; available in the online
version of this paper.
2Fold change was calculated as the ratio between the mean of intensities
of High-Forage/Low-Gain and High-Concentrate/High-Gain samples trans-
formed to the natural logarithm.
Gene expression in arcuate nucleus of heifers 2229
projections originating in the ARC toward hypothalamic
regions that regulate metabolism and feed intake are es-
tablished during the rst 2 wk after birth (Bouret et al.,
2004). This observation indicates that the early juvenile
period of development is critical for development of hy-
pothalamic pathways that control neuroendocrine func-
tions during maturation and adulthood.
Clustering of differentially expressed genes in juve-
nile heifers growing at distinct rates provides evidence
of important biological functions affected by nutritional
input. One includes the control of feed intake. Peptide
products of genes included in this cluster are well known
for stimulating (NPY and AGRP) or inhibiting (-MSH)
feeding behavior (Stanley et al., 2005). In addition, clus-
tering of differentially expressed genes involved in the
regulation of cellular metabolic processes, receptor and
intracellular signaling, and neuronal communication pro-
vides evidence of a broad array of biological functions
that are regulated within the ARC in response to changes
in nutritional status. It is evident that the expression of
many differentially expressed genes is not limited to
neurons. The methodology used in the current study did
not allow us to isolate neurons from other cell types, nor
speci c populations of neurons within the ARC nucleus.
Nevertheless, the integrative approach used in the cur-
rent study maximized our ability to investigate, at the
cellular level, complex functional changes that occur
within the ARC nucleus that may be critical for the regu-
lation of various physiological functions. This is particu-
larly important because the control of neuroendocrine
functions involves interactions among neurons, glia and
vascular elements (Haydon and Camignoto, 2006; Ojeda
et al., 2006; Roth et al., 2007). Moreover, the approach
used in this study will direct further investigations of nu-
tritionally responsive, cellular components of the neuro-
endocrine system in a reductive manner.
Of the differentially expressed genes identi ed in
the microarray study, expression of NPY, AGRP, GHR,
and PRLHR genes were con rmed by quantitative PCR.
Neuropeptide Y and AGRP are known to be intimately
involved in signaling metabolic status to the central
reproductive axis (Pierroz et al., 1996; Schioth et al.,
2001; Vulliémoz et al., 2005) through neuronal path-
ways emanating from within the ARC (Turi et al., 2003).
Neuropeptide Y projections are observed in close prox-
imity to GnRH neurons and dendrites in the pre-optic
area (POA; Campbell et al., 2001) and GnRH bers in
the median eminence (Li et al., 1999). Neuropeptide Y5
receptor has been localized in GnRH neurons (Li et al.,
1999; Campbell et al., 2001), indicating direct actions of
NPY upon GnRH neurons. Neuropeptide Y expression
increases during negative energy balance (McShane et
al., 1993) and decreases during adequate or excess en-
ergy (Sanacora et al., 1990). Therefore, because of its in-
volvement in the control of reproductive function in sex-
ually mature animals (Catze ies et al., 1993; Gazal et al.,
1998; Thomas et al., 1999), NPY is a major focal point
in the efforts to understand mechanisms associated with
the metabolic control of sexual maturation. Importantly,
treatment of female rats with NPY delays sexual matura-
tion and disrupts reproductive cyclicity (Catze ies et al.,
1993). Furthermore, McShane et al. (1992) have reported
that intracerebroventricular administration of NPY to
estradiol-implanted and non-implanted, ovariectomized
ewes reduced release of LH. Others have reported that
NPY administration suppresses circulating concentra-
tions of LH by inhibiting pulsatile release of GnRH and
LH in ovariectomized, estradiol-implanted cows (Gazal
et al., 1998; Thomas et al., 1999). Additionally, acute
central administration of NPY inhibits secretion of LH
in mature, ovariectomized cows pretreated with leptin
(Garcia et al., 2004), suggesting that NPY signaling may
be downstream of the actions of leptin. Based on previous
reports (Gasser et al., 2006a–d), the nutritional strategies
employed in the current experiment were designed to
promote a relatively rapid rate of BW gain, which, when
associated with high concentrate diets, have been shown
to markedly hasten puberty in heifers. Thus, our obser-
vation of decreased NPY expression in the ARC of HC/
HG heifers compared with HF/LG support the hypoth-
esis that NPY neuronal pathways may serve as metabolic
integrators of the pubertal process. Speci cally, elevated
NPY expression in the ARC may serve as a metabolic
‘brake’ for pubertal onset. However, the neuroendocrine
mechanisms and pathways by which NPY exerts this
function remain unclear, but may involve differential
NPY projections that are established during early calf-
hood development (Alves et al., 2011). In addition, al-
terations in hypothalamic neuronal circuitry associated
with nutritional inputs have been observed during early
postnatal development in rodents (Bouret et al., 2008).
The majority of the NPY neurons in the ARC co-
express AGRP (Broberger et al., 1998), which functions
in concert with NPY to increase nutrient intake and de-
crease energy expenditure during times of inadequate
nutrition. In the current study, differential expression of
AGRP between HC/HG and HF/LG heifers was parallel
to that of NPY. Agouti-related protein is an antagonist of
melanocortin receptors (MCR) and blocks the actions of
-MSH (Ollmann et al., 1997). Hypothalamic -MSH is
cleaved from its polypeptide precursor, POMC, during
post-translational processing and upon binding to MCR
suppresses appetite and feeding behavior. In the current
study, expression of POMC gene was increased in heif-
ers fed high concentrate diets to gain BW at rapid rates.
This is in agreement with the opposing roles of NPY/
AGRP and -MSH in the control of feeding and energy
expenditure (Stanley et al., 2005). It has been suggested
Allen et al.
2230
previously that AGRP exerts negative modulatory ef-
fects on GnRH neurons (Schioth et al., 2001; Vulliémoz
et al., 2005) and thus ultimately, decreases secretion of
gonadotropins from the adenohypophysis. Melanocortin
4 receptor (MC4R) is expressed in immortalized hy-
pothalamic GnRH-secreting GT-1 cells (Khong et al.,
2001), but it is unclear if they are expressed speci -
cally on GnRH neurons. Based on in vitro experiments,
AGRP does not appear to affect gonadotropin secretion
directly in cultured pituitary cells (Stanley et al., 1999).
The apparent role of AGRP in regulating gonadotropin
secretion is to function in synchrony with NPY to in-
crease appetite and feeding behavior by blocking the
actions of -MSH. Similar to NPY, AGRP expression
was more abundantly expressed in HF/LG than in HC/
HG heifers. Thus, AGRP may bolster the actions of NPY
as a metabolic ‘brake’ on pubertal onset by suppressing
GnRH release. Previously, it has been demonstrated that
nutritional inputs during early postnatal development af-
fect neuronal development of AGRP projections in the
hypothalamus (Bouret et al., 2008).
The current study also revealed that GHR expres-
sion was decreased in HC/HG heifers relative to HF/LG.
Growth hormone is the cognate ligand for GHR, and
GH secretion is regulated by an intricate interaction be-
tween GHRH and somatostatin actions on somatotrophs
(Tannenbaum and Ling, 1984). Only a small proportion
of GHRH neurons in the ARC express GHR (Burton et
al., 1995); however, the majority (98%) of NPY neurons
in the ARC express GHR mRNA (Chan et al., 1996). In
addition, evidence indicates that NPY neurons act as an
intermediate step in the regulation of GH (Chan et al.,
1996). In rats, hypophysectomy caused a signi cant re-
duction of NPY mRNA expression in the ARC, and treat-
ment with GH restored mRNA expression to that of con-
trol animals (Chan et al., 1996). This indicates a direct
regulation of NPY expression by GH, likely via GHR on
NPY neurons in the ARC. Concentrations of GH in cir-
culation tend to decrease during the juvenile growth and
are low at puberty, particularly in heifers gaining BW at
rapid rates (Yelich et al., 1995). Therefore, it is possible
that the increased nutritional status promoted in the HC/
HG group supports alterations in the NPY system with
downstream effects on GH secretion and GH feedback
signaling on NPY neurons. Previous reports from our
group (Thomas et al., 1999) and from others (McMahon
et al., 1999; Morrison et al., 2003) have demonstrated
clear stimulatory effects of NPY on release of GH in
cattle and sheep. Thus, the interrelationships between
NPY and GH release seem to reinforce a metabolic sta-
tus that may be permissive (if suppressed) or restrictive
(if maintained elevated) to reproductive maturation.
The PRLHR gene (originally denominated GPR10)
has been found to be highly expressed in regions of the
brain that are involved in the control of nutrient intake and
energy expenditure, including the hypothalamus (Fujii et
al., 1999). PRLHR was found to be abundant in the ade-
nohypophysis (Fujii et al., 1999) and its endogenous pep-
tide ligand, prolactin-releasing hormone (PRLH), was
found to induce prolactin release, and reduce feed intake
and BW (Lawrence et al., 2000). Moreover, intracerebro-
ventricular injection of PRLH increases body temperature
and oxygen consumption, indicating that activation of
PRLHR increases energy expenditure. Further, PRLHR-
de cient female mice show increased circulating leptin
and increased hypothalamic expression of corticotropin-
releasing hormone (CRH) and POMC genes (Bjursell et
al., 2007). These observations corroborate our ndings in
which expression of POMC was greater in HC/HG than in
HF/LG heifers (see Supplemental Tables; available in the
online version of this paper).
The greater circulating concentrations of leptin and
insulin in HC/HG compared with HF/LG heifers con-
rm the elevated nutritional status in that group, and the
greater concentrations of IGF-1 in circulation, support
greater potential for increased somatic growth. The in-
crease in mean circulating concentrations of leptin in
HC/HG heifers is congruent with the observed decrease
in expression of NPY and AGRP, because leptin has
been shown to suppress NPY and AGRP gene expression
and release (Aubert et al., 1998; Belgardt et al., 2009;
Olofsson et al., 2009). Leptin also positively modulates
GH secretion under fasting conditions by direct actions
at the anterior pituitary (Zieba et al., 2003) and attenu-
ates the stimulatory effects of NPY on GH release in
cattle (Garcia et al., 2004). In addition, numerous stud-
ies have demonstrated the close association between se-
cretion patterns of leptin and insulin. Similar to leptin,
insulin also has negative regulatory effects on NPY and
AGRP expression (Wang and Leibowitz, 1997).
The foregoing changes in metabolic hormones and
in expression of key regulatory genes in the ARC are
also congruent with the observed changes of ruminal
VFA content. In ruminants, propionate is readily con-
verted to glucose in the liver, whereas acetate is not (Van
Soest, 1982). Increased hepatic function may explain in-
creased liver weight in HC/HG heifers, supporting in-
creased hepatic IGF-1 synthesis and maintaining greater
concentrations of IGF-1 in circulation.
In summary, our results have con rmed marked
differences in gene expression within the ARC in heif-
ers nutritionally programmed to hasten pubertal onset.
Among the numerous differently-expressed genes, NPY,
AGRP, GHR and PRLHR exhibited decreased expres-
sion under nutritional conditions that promoted a rela-
tively rapid rate of gain. These genes are known to be
involved in the neuroendocrine control of metabolic
functions, and interact to regulate the reproductive neu-
Gene expression in arcuate nucleus of heifers 2231
roendocrine axis. Therefore, nutritional inputs during
juvenile development regulate expression of an array of
genes in the ARC that are involved in various biological
processes; most notably, response to hormones and nu-
trients, feeding behavior and neural transmission. Fine
regulation of these biological processes may be critical
for timing the onset of puberty in heifers.
LITERATURE CITED
Accorsi, P. A., N. Govoni, R. Gaiani, C. Pezzi, E. Seren, and C. Tamanini. 2005.
Leptin, GH, PRL, insulin and metabolic parameters throughout the dry
period and lactation in dairy cows. Reprod. Dom. Anim. 40:217–223.
Adam, C. L., and P. A. Findlay. 1998. Inhibition of luteinizing hormone secre-
tion and expression of c-fos and corticotrophin-releasing factor genes
in the paraventricular nucleus during insulin-induced hypoglycaemia in
sheep. J. Neuroendocrinol. 10:777–783.
Alves, B. R. C., S. Liu, E. Stevenson, J. Thorson, R. C. Cardoso, L. O. Tedeschi,
D. H. Keisler, G. L. Williams, and M. Amstalden. 2011. Accelerated
body weight gain during the juvenile period reduces neuropeptide Y
close contacts with GnRH neurons in heifers. Proc. Soc. Study Reprod.
Abstr.191:44.
Aubert, M. L., D. D. Pierroz, N. M. Gruaz, V. d’Alleves, B. A. M. Vuagnat, F.
P. Pralong, W. F. Blum, and P. C. Sizonenko. 1998. Metabolic control of
sexual function and growth: Role of neuropeptide Y and leptin. Mol. Cell.
Endocrinol. 140:107–113.
Belgardt, B. F., T. Okamura, and J. C. Bruning. 2009. Hormone and glucose
signaling in POMC and AgRP neurons. J. Physiol. 587:5305–5314.
Benjamini, Y., and Y. Hochberg. 1995. Controlling the false discovery rate: A
practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B
Stat. Methodol. 57:289–300.
Bjursell, M., M. Lennerås, M. Göransson, A. Elmgren, and M. Bohlooly-Y.
2007. GPR10 de ciency in mice results in altered energy expenditure and
obesity. Biochem. Biophys. Res. Commun. 363:633–638.
Bouret, S. G., S. J. Draper, and R. B. Simerly. 2004. Formation of projection
pathways from the arcuate nucleus of the hypothalamus to hypothalamic
regions implicated in the neuronal control of feeding in mice. J. Neurosci.
24:2797–2805.
Bouret S. G., J. N. Gorski, C. M. Patterson, S. Chen, B. E. Levin, and R. B.
Simerly. 2008. Hypothalamic neuronal projections are permanently dis-
rupted in diet-induced obese rats. Cell. Metab. 7:179–185.
Broberger, C., J. Johansen, C. Johansson, M. Schalling, and T. Hokfelt. 1998.
The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry
in normal, anorectic and monosodium glutamate-treated mice. Proc. Natl.
Acad. Sci. USA 95:15043–15048.
Burton, K. A., E. B. Kabigting, R. A. Steiner, and D. K. Clifton. 1995.
Identi cation of target cells for growth hormone’s action in the arcuate
nucleus. Am. J. Physiol. 269:E716–E722.
Campbell, R. E., J. M. ffrench-Mullen, M. A. Cowley, M. S. Smith, and K. L.
Grove. 2001. Hypothalamic circuitry of neuropeptide Y regulation of
neuroendocrine function and food intake via the Y5 receptor subtype.
Neuroendocrinology 74:106–119.
Catze ies, C., D. D. Pierroz, F. Rohner-Jeanrenaud, J. Rivier, P. C. Sizonenko,
and M. L. Aubert. 1993. Neuropeptide Y administered chronically into
the lateral ventricle profoundly inhibits both the gonadotropic and the so-
matotropic axis in intact adult female rats. Endocrinology 132:224–234.
Chan, Y. Y., D. K. Clifton, and R. A. Steiner. 1996. Role of NPY neurons in GH-
dependent feedback signalling to the brain. Horm. Res. Suppl. 1:12–14.
Day, M. L., K. Imakawa, M. Garcia-Winder, D. D. Zalesky, B. D. Schanbacher,
R. J. Kittok, and J. E. Kinder. 1984. Endocrine mechanisms of puberty
in heifers: Estradiol negative feedback regulation of luteinizing hormone
secretion. Biol. Reprod. 31:332–341.
Delavaud, C., F. Bocquier, Y. Chilliard, D. H. Keisler, A. Gertler, and G. Kann.
2000. Plasma leptin determination in ruminants: Effect of nutritional sta-
tus and body fatness on plasma leptin concentration assessed by a speci c
RIA in sheep. J. Endocrinol. 165:519–526.
DiCostanzo, A., J. E. Williams, and D. H. Keisler. 1999. Effects of short- or long-
term infusions of acetate or propionate on luteinizing hormone, insulin,
and metabolite concentrations in beef heifers. J. Anim. Sci. 77:3050–3056.
Fox, D. G., L. O. Tedeschi, T. P. Tylutki, J. B. Russell, M. E. Van Amburgh, L. E.
Chase, A. N. Pell, and T. R. Overton. 2004. The Cornell net carbohydrate
and protein system model for evaluating herd nutrition and nutrient excre-
tion. Anim. Feed Sci. Technol. 112:29–79.
Fujii, R., S. Fukusumi, M. Hosoya, Y. Kawamata, Y. Habata, S. Hinuma, M.
Sekiguchi, C. Kitada, T. Kurokawa, O. Nishimura, H. Onda, Y. Sumino,
and M. Fujino. 1999. Tissue distribution of prolactin-releasing peptide
(PrRP) and its receptor. Regul Pept. 83:1–10.
Garcia, M. R., M. Amstalden, D. H. Keisler, N. Raver, A. Gertler, and G. L.
Williams. 2004. Leptin attenuates the acute effects of centrally adminis-
tered neuropeptide Y on somatotropin but not gonadotropin secretion in
ovariectomized cows. Domest. Anim. Endocrinol. 26:189–200.
Garcia, M. R., M. Amstalden, C. D. Morrison, D. H. Keisler, and G. L. Williams.
2003. Age at puberty, total fat and conjugated linoleic acid content of car-
cass, and circulating metabolic hormones in beef heifers fed a diet high in
linoleic acid beginning at four months of age. J. Anim. Sci. 81:261–268.
Gasser, C. L., E. J. Behlke, D. E. Grum, and M. L. Day. 2006a. Effect of timing
of feeding a high-concentrate diet on growth and attainment of puberty in
early-weaned heifers. J. Anim. Sci. 84:3118–3122.
Gasser, C. L., G. A. Bridges, M. L. Mussard, D. E. Grum, J. E. Kinder, and M.
L. Day. 2006b. Induction of precocious puberty in heifers III: Hastened
reduction of estradiol negative feedback on secretion of luteinizing hor-
mone. J. Anim. Sci. 84:2050–2056.
Gasser, C. L., C. R. Burke, M. L. Mussard, E. J. Behlke, D. E. Grum, J. E.
Kinder, and M. L. Day. 2006c. Induction of precocious puberty in heifers
II: Advanced ovarian follicular development. J. Anim. Sci. 84:2042–2049.
Gasser, C. L., D. E. Grum, M. L. Mussarg, F. L. Fluharty, J. E. Kinder, and M. L.
Day. 2006d. Induction of precocious puberty in heifers I: Enhanced secre-
tion of luteinizing hormone. J. Anim. Sci. 84:2035–2041.
Gazal, O. S., L. S. Leshin, R. L. Stanko, M. G. Thomas, D. H. Keisler, L. L.
Anderson, and G. L. Williams. 1998. Gonadotropin-releasing hormone
secretion into third-ventricle cerebrospinal uid of cattle: Correspondence
with the tonic and surge release of luteinizing hormone and its tonic inhibi-
tion by suckling and neuropeptide Y. Biol. Reprod. 56:676–683.
Hahn, T. M., F. Breininger, D. G. Baskin, and M. W. Schwartz. 1998. Co-
expression of Agrp and NPY in fasting-activated hypothalamic neurons.
Nat. Neurosci. 1:271–272.
Haydon P. G., and G. Carmignoto. 2006. Astrocyte control of synaptic transmis-
sion and neurovascular coupling. Physiol. Rev. 86:1009–1031.
Hiney, J. K., S. R. Ojeda, and W. L. Dees. 1991. Insulin-like growth factor 1:
A possible metabolic signal involved in the regulation of female puberty.
Neuroendocrinology 54:420–423.
Hiney, J. K., V. Srivastava, C. L. Nyberg, S. R. Ojeda, and W. L. Dees. 1996.
Insulin-like growth factor-1 of peripheral origin acts centrally to acceler-
ate the initiation of female puberty. Endocrinology 127:3717–3728.
Huang, D. W., B. T. Sherman, and R. A. Lempicki. 2009. Systematic and inte-
grative analysis of large gene lists using DAVID bioinformatics resources.
Nat. Protoc. 4:44–57.
I’Anson, H., J. M. Manning, C. G. Herbosa, J. Pely, C. R. Friedman, R. I. Wood,
D. C. Bucholtz, and D. L. Foster. 2000. Central inhibition of gonadotro-
pin-releasing hormone secretion in growth-restricted hypogonadotropic
female sheep. Endocrinology 141:520–527.
Khong, K., S. E. Kurtz, R. L. Sykes, and R. D. Cone. 2001. Expression of
functional melanocortin-4 receptor in the hypothalamic GT1-1 cell line.
Neuroendocrinology 74:193–201.
Lawrence, C. B., F. Celsi, J. Brennand, and S. M. Luckman. 2000. Alternative
role for prolactin-releasing peptide in the regulation of food intake. Nat.
Neurosci. 3:645–646.
Allen et al.
2232
Lehman, M. N., F. J. Ebling, S. M. Moenter, and F. J. Karsch. 1993.
Distribution of estrogen receptor-immunoreactive cells in the sheep brain.
Endocrinology 133:876–886.
Li, C., P. Chen, and M. S. Smith. 1999. Morphological evidence for direct in-
teraction between arcuate nucleus neuropeptide Y (NPY) neurons and
gonadotropin-releasing hormone neurons and the possible involvement of
NPY Y1 receptors. Endocrinology 140:5382–5390.
Li X., H. Chiang, J. Zhu, S. E Dowd, and H. Zhou. 2008. Characterization of
a newly developed chicken 44k Agilent microarray. BMC Genomics
9:60–74.
Livak, K. J., and T. D. Schmittgen. 2001. Analysis of relative gene expression
data using real-time quantitative PCR and the 2-Ct method. Methods
25:402–408.
McMahon, C. D., D. F. Buxton, T. H. Elsasser, D. R. Gunter, L. G. Sanders, B. P.
Steele, and J. L. Sartin. 1999. Neuropeptide Y restores appetite and alters
concentrations of GH after central administration to endotoxic sheep. J.
Endocrinol. 161:333–339.
McShane, T. M., T. May, J. L. Miner, and D. H. Keisler. 1992. Central actions
of neuropeptide Y may provide a neuromodulatory link between nutrition
and reproduction. Biol. Reprod. 46:1151–1157.
McShane, T. M., S. L. Petersen, S. McCrone, and D. H. Keisler. 1993. In uence
of food restriction on neuropeptide-Y, proopiomelanocortin, and lutein-
izing hormone-releasing hormone gene expression in sheep hypothalami.
Biol. Reprod. 49:831–839.
Morrison, C. D., J. A. Daniel, J. H. Hampton, P. R. Buff, T. M. McShane, M.
G. Thomas, and D. H. Keisler. 2003. Luteinizing hormone and growth
hormone secretion in ewes infused intracerebroventricularly with neuro-
peptide Y. Domest. Anim. Endocrinol. 24:69–80.
Ojeda S. R., A. Lomniczi, C. Mastronardi, S. Heger, C. Roth, A. S. Parent, V.
Matagne, and A. E. Mungenast. 2006. Minireview: The neuroendocrine
regulation of puberty: Is the time ripe for a systems biology approach?
Endocrinology 147:1166–1174.
Ollmann, M. M., B. D. Wilson, Y. Yang, J. A. Kerns, Y. Chen, I. Gantz, and G. S.
Barsh. 1997. Antagonism of central melanocortin receptors in vitro and in
vivo by agouti-related protein. Science 278:136–138.
Olofsson, L. E., A. A. Pierce, and A. W. Xu. 2009. Functional requirement of
AgRP and NPY neurons in ovarian cycle-dependent regulation of food
intake. Proc. Nat. Acad. Sci. 106:15932–15937.
Pellegrino, L. J., A. S. Pellegrino, and A. J. Cushman. 1979. A stereotaxic atlas
of the rat brain. Plenum Press, New York.
Pierroz, D. D., C. Catze ies, A. C. Aebi, J. E. Rivier, and M. L. Aubert. 1996.
Chronic administration of neuropeptide Y into the lateral ventricle inhib-
its both the pituitary-testicular axis and growth hormone and insulin-like
growth factor-1 secretion in intact adult male rats. Endocrinology 137:3–
12.
Raposinho, P. D., P. Broqua, D. D. Pierroz, A. Hayward, Y. Dumont, R. Quirion,
J. Junien, and M. L. Aubert. 1999. Evident that the inhibition of luteiniz-
ing hormone secretion exerted by central administration of neuropeptide
Y (NPY) in the rat is predominantly mediated by the NPY-Y5 receptor
subtype. Endocrinology 140:4046–4055.
Redmond, J. S., G. M. Baez-Sandoval, K. M. Spell, T. E. Spencer, C. A. Lents,
G. L. Williams, and M. Amstalden. 2011. Developmental changes in
hypothalamic Kiss1 expression during the activation of the pulsatile re-
lease of luteinizing hormone in maturing ewe lambs. J. Neuroendocrinol.
23:815–822.
Roth, C. L., C. Mastronardi, A. Lomniczi, H. Wright, R. Cabrera, A. E.
Mungenast, S. Heger, H. Jung, C. Dubay, and S. R. Ojeda. 2007.
Expression of a tumor-related gene network increases in the mammalian
hypothalamus at the time of female puberty. Endocrinology 148:5147–
5161.
Ryan, D. P., R. A. Spoon, M. K. Grif th, and G. L. Williams. 1994. Ovarian
follicular recruitment, granulosa cell steriodogenic potential, and growth
hormone/insulin-like growth factor-I relationships in suckled beef cows
consuming high lipid diets: Effects of graded differences in body con-
dition maintained during the puerperium. Domest. Anim. Endocrinol.
11:161–174.
Salanitro, J. P., and P. A. Muirhead. 1975. Quantitative method for the gas chro-
matographic analysis of short-chain monocarboxylic and dicarboxylic
acids in fermentation media. Appl. Microbiol. 29:374–381.
Sanacora, G., M. Kershaw, J. A. Finkelstein, and J. D. White. 1990. Increased
hypothalamic content of preproneuropeptide Y messenger ribonucleic
acid in genetically obese Zucker rats and its regulation by food depriva-
tion. Endocrinology 127:730–737.
Schioth, H. B., Y. Kakizaki, A. Kohsaka, T. Suda, and H. Watanobe. 2001.
Agouti-related peptide prevents steroid-induced luteinizing hormone and
prolactin surges in female rats. Neuroreport 12:687–690.
Simpson, R. B., J. D. Armstrong, R. W. Harvey, D. C. Miller, E. P. Heimer,
and R. M. Campbell. 1991. Effect of active immunization against growth
hormone-releasing factor on growth and onset of puberty in beef heifers.
J. Anim. Sci. 69:4914–4924.
Smyth, G. K. 2004. Linear models and empirical Bayes methods for assess-
ing differential expression in microarray experiments. Stat. Appl. Genet.
Mole. Biol. 1(E-pub):Article 3.
Stanley, S., K. Wynne, B. McGowan, and S. Bloom. 2005. Hormonal regulation
of food intake. Physiol. Rev. 85:1131–1158.
Stanley, S. A., C. J. Small, M. S. Kim, M. M. Heath, L. J. Seal, S. H. Russell, M.
A. Ghatei, and S. R. Bloom. 1999. Agouti related peptide (Agrp) stimu-
lates the hypothalamo pituitary gonadal axis in vivo & in vitro in male rats.
Endocrinology 140:5459–5462.
Tannenbaum, G. S., and N. Ling. 1984. The interrelationship of growth hor-
mone (GH)-releasing factor and somatostatin in generation of the ultra-
dian rhythm of GH secretion. Endocrinology 115:1952–1957.
Thomas, M. G., O. S. Gazal, G. L. Williams, R. L. Stanko, and D. H. Keisler.
1999. Injection of neuropeptide Y into the third cerebroventricle differ-
entially in uences pituitary secretion of luteinizing hormone and growth
hormone in ovariectomized cows. Domest. Anim. Endocrinol. 16:159–
169.
Turi, G. F., Z. Liposits, S. M. Moenter, C. Fekete, and E. Hrabovszky. 2003.
Origin of neuropeptide Y-containing afferents to gonadotropin-releasing
hormone neurons in male mice. Endocrinology 144:4967–4974.
Van Soest, P. J. 1982. Nutritional ecology of the ruminant. Ithaca, New York:
Cornell University Press.
Vandesompele, J., K. De Preter, F. Pattyn, B. Poppe, N. Van Roy, A. De Paepe,
and F. Speleman. 2002. Accurate normalization of real-time quantitative
RT-PCR data by geometric averaging of multiple internal control genes.
Genome Biol. 3:RESEARCH0034.
Vulliémoz, N. R., E. Xiao, L. Xia-Zhang, S. L. Wardlaw, and M. Ferin. 2005.
Central infusion of agouti-related peptide suppresses pulsatile luteinizing
hormone release in the ovariectomized rhesus monkey. Endocrinology
146:784–789.
Wang, J., and K. L. Leibowitz. 1997. Central insulin inhibits hypothalamic
galanin and neuropeptide Y gene expression and peptide release in intact
rats. Brain Res. 777:231–236.
Yelich, J. V., R. P. Wettemann, H. G. Dolezal, K. S. Lusby, D. K. Bishop, and
L. J. Spicer. 1995. Effects of growth rate on carcass composition and lipid
partitioning at puberty and growth hormone, insulin-like growth factor
I, insulin and metabolites before puberty in beef heifers. J. Anim. Sci.
73:2390–2405.
Yelich, J. V., R. P. Wettemann, T. T. Marston, and L. J. Spicer. 1996. Luteinizing
hormone, growth hormone, insulin-like growth factor-1, insulin and me-
tabolites before puberty in heifers fed to gain at two rates. Domest. Anim.
Endocrinol. 13:325–338.
Zieba, D. A., M. Amstalden, S. Morton, J. L. Gallino, J. F. Edwards, P. G.
Harms, and G. L. Williams. 2003. Effects of leptin on basal and GHRH-
stimulated GH secretion from the bovine adenohypophysis are dependent
on nutritional status. J. Endocrinol. 178:83–89.
Zieba, D. A., M. Amstalden, S. Morton, M. N. Maciel, D. H. Keisler, and G.
L. Williams. 2004. Regulatory roles of leptin in the hypothalamic-hy-
pophyseal axis before and after sexual maturation in cattle. Biol. Reprod.
71:804–812.
... Seminal plasma does not simply protect sperm but can also influence the reproductive events independent of sperm in some species other than cattle. Seminal fluid regulation of female reproductive physiology is shown in hamsters, pigs, and human studies [68,73,[123][124][125][126][127][128][129][130][131][132][133][134]. Mating with males lacking seminal vesicles resulted in reduced blastocyst rate, reduced conception rates, and offspring that experienced sexdependent phenotypic changes postnatally [120,125,135]. ...
... AGE/RAGE signaling has been shown to increase oxidative stress and inflammation. AGE impaired testicular function and thus affected spermatogenesis and sperm quality in rodents [134]. AGE hindered trophoblast invasion. ...
... In heifers, increased rates of BW gain during a prepubertal period can advance puberty [145,146]. Functional changes in the NPY and POMC neuronal pathways appear to contribute to attaining puberty [134]. In humans, miR-21-3p has been implicated in adipose tissue browning and diabetes [147]. ...
Article
Full-text available
Recent advances in high-throughput in silico techniques portray experimental data as exemplified biological networks and help us understand the role of individual proteins, interactions, and their biological functions. The objective of this study was to identify differentially expressed (DE) sperm and seminal plasma microRNAs (miRNAs) in high- and low-fertile Holstein bulls (four bulls per group), integrate miRNAs to their target genes, and categorize the target genes based on biological process predictions. Out of 84 bovine-specific, prioritized miRNAs analyzed by RT-PCR, 30 were differentially expressed in high-fertile sperm and seminal plasma compared to low-fertile sperm and seminal plasma, respectively (p ≤ 0.05, fold regulation ≥ 5 magnitudes). The expression levels of DE-miRNAs in sperm and seminal plasma followed a similar pattern. Highly scored integrated genes of DE-miRNAs predicted various biological and molecular functions, cellular process, and pathways. Further, analysis of the categorized genes showed association with pathways regulating sperm structure and function, fertilization, and embryo and placental development. In conclusion, highly DE-miRNAs in bovine sperm and seminal plasma could be used as a tool for predicting reproductive functions. Since the identified miRNA-mRNA interactions were mostly based on predictions from public databases, the causal regulations of miRNA-mRNA and the underlying mechanisms require further functional characterization in future studies.
... ADG = average daily gain; EPD = expected progeny difference; BW = body weight; DMI = dry matter intake; FE = feed efficiency 1 ADG × EPD = Interaction between ADG and EPD 2 From 3 rd to 7 th month of age; HG heifers were fed ad libitum diet and MG heifers were fed with restriction 3 From 7 th month of age to puberty; all heifers were fed ad libitum diet independent of genetic factors. This idea did not agree with several studies that showed anticipation in puberty when the nutritional plans were increased in the juvenile age, specifically between three and nine months of age (Allen et al., 2012;Moriel et al., 2014;Cardoso et al., 2020). Another possibility was that the HG (0.9 kg d −1 ) was not high enough to cause metabolic imprinting to induce precocious puberty. ...
... Treatments P-value HG MG P NP P NP ADG EPD ADG × EPD 1 IGF-1 (ng/mL) 83.1 ± 8.0 74.4 ± 6.2 76.2 ± 6.0 65.9 ± 6.6 0.26 0.16 0.91 Insulin (µUI/mL) 12.2 ± 2.0 ab 12.4 ± 1.6 ab 15.7 ± 1.6 a 9.1 ± 1.7 b 0.96 0.07 0.05 heifers at 15 months of age, since insulin increases with increasing energy intake (Allen et al. 2012). Another nutritional mediator with an effect on puberty, such as insulin, is IGF-1 (Moriel et al. 2014;Ferraz Jr et al. 2018) . ...
Article
Full-text available
The early attainment of puberty in heifers is essential for the profitability of the cow-calf farm. This study aimed to evaluate the impact of juvenile average daily gain (ADG) and sire’s expected progeny difference (EPD) on puberty of crossbred beef heifers. Sixty Angus × Nellore heifers early weaned (age = 102 ± 4.3 d; initial BW = 103 ± 4.7 kg) were used in a 2 × 2 factorial arrangement. The factor 1 was the sire’s EPD for scrotal circumference, in which heifers born from sires with positive EPD were considered precocious (P), and heifers born from sires with negative EPD were considered non-precocious (NP). The factor 2 was the high (HG; ADG = 0.9 kg; ad libitum) and medium ADG (MG; ADG = 0.7 kg) from 3rd to 7th month of age (1st phase). After 1st phase until puberty, all heifers were fed ad libitum (2nd phase). Statistical analysis was performed by SAS. There was an interaction between factors for DMI in the 1st phase (P = 0.02), which PHG heifers had higher DMI than NPHG. There was no effect on puberty rate, BW, age and BCS at puberty comparing HG vs. MG. However, favorable sires’ EPD for scrotal circumference induced a higher proportion of puberty (P 87% vs. NP 59%) at 15 months of age. Thus, the ADG in juvenile age did not affect puberty rate, but sires with positive EPD increased puberty rate of Angus × Nellore heifers in heifers fed a high gain diet.
... ADG = average daily gain; EPD = expected progeny difference; BW = body weight; DMI = dry matter intake; FE = feed efficiency 1 ADG × EPD = Interaction between ADG and EPD 2 From 3 rd to 7 th month of age; HG heifers were fed ad libitum diet and MG heifers were fed with restriction 3 From 7 th month of age to puberty; all heifers were fed ad libitum diet independent of genetic factors. This idea did not agree with several studies that showed anticipation in puberty when the nutritional plans were increased in the juvenile age, specifically between three and nine months of age (Allen et al., 2012;Moriel et al., 2014;Cardoso et al., 2020). Another possibility was that the HG (0.9 kg d −1 ) was not high enough to cause metabolic imprinting to induce precocious puberty. ...
... ADG = average daily gain; EPD = expected progeny difference; SC = Scrotal Circumference Table 3 Effect of juvenile ADG and sires' expected progeny difference (EPD) for scrotal circumference (SC) on serum concentrations of IGF-1 and Insulin in Angus × Nellore heifers Heifers Precocious (P-born from sires with positive EPD for SC) and Non-precocious (NP-born from sires with negative EPD for SC) submitted to High ADG (HG) and Medium ADG (MG) from 3 rd to 7 th month of age. ADG = average daily gain; EPD = expected progeny difference 1 Interaction between factors ADG and EPD (Allen et al. 2012). Another nutritional mediator with an effect on puberty, such as insulin, is IGF-1 (Moriel et al. 2014;Ferraz Jr et al. 2018). ...
Article
Full-text available
This study aimed to determine the effect of growth rates on the hormonal status and puberty onset. Forty-eight Nellore heifers were weaned at 3.0 ± 0.1 (means ± standard error of the mean) months old were blocked according to body weight at weaning (84 ± 2 kg) and randomly assigned to treatments. The treatments were arranged in 2 × 2 factorial according to the feeding program. The first program was high (H; 0.79 kg/day) or control (C; 0.45 kg/day) average daily gain (ADG) from 3rd to 7th month of age (growing phase I). The second program was also high (H; 0.70 kg/day) or control (C; 0.50 kg/day) ADG from the 7th month until puberty (growing phase II), resulting in four treatments: HH (n = 13), HC (n = 10), CH (n = 13), and CC (n = 12). To achieve desired gains, heifers in high ADG program were fed ad libitum dry matter intake (DMI), and the control group was offered around 50% of ad libitum DMI of high group. All heifers received a diet with similar composition. Puberty was assessed weekly by ultrasound examination, and the largest follicle diameter was evaluated every month. Blood samples were collected to quantify leptin, insulin growth factor-1 (IGF1) and luteinizing hormone (LH). At 7 months of age, heifers in high ADG were 35 kg heavier than the control. Heifers in the HH had greater DMI compared with CH in phase II. The puberty rate at 19 months old was greater in the HH treatment (84%) than in the CC (23%), but there was no difference between HC (60%) and CH (50%) treatments. Heifers from HH treatment had greater serum leptin concentration than others at 13 months old, and serum leptin was greater in HH compared with CH and CC at 18 months old. High heifers in phase I had greater serum IGF1 concentration than the control. In addition, HH heifers had a greater diameter of the largest follicle than CC. There was no interaction between phases and age in any variable relative to the LH profile. However, the heifers’ age was the main factor that increased the frequency of LH pulse. In conclusion, increasing ADG was associated with greater ADG, serum leptin and IGF-1 concentration, and puberty onset; however, LH concentration was affected mainly by age of the animal. The increasing growth rate at younger age made heifers more efficient.
... Previous research using the bovine model has demonstrated that significant modifications in dietary energy status during the early juvenile period (∼4-9 months of age) impact the development of neuronal signaling pathways that regulate the GnRH pulse generator [1,2], the expression of genes in several hypothalamic nuclei [3,4], DNA methylation patterns in the arcuate nucleus [5], and timing of puberty [6]. However, little in-depth research has been conducted to determine whether prenatal nutrition, independently or interactively with postnatal nutrition, affects the pubertal process or specific aspects of adult ovarian function. ...
... Although there is evidence that both over and undernutrition during pregnancy can negatively impact the offspring's metabolic functions [33,34], the postnatal juvenile period is more sensitive to dietary manipulations in which rate of gain can readily advance or delay pubertal onset. The positive effect on pubertal onset of an early postnatal high concentrate diet reflected by the 2-month difference in age at puberty between postnatal L and H heifers in the current experiment is similar to that observed previously by our group [3,6] and others [35,36], demonstrating that the juvenile period (ranging from 4 to 9 months of age) represents a period of significant hypothalamic plasticity relative to the nutritional programming of puberty. Although the exact physiological mechanisms involved in the nutritional acceleration of puberty are not completely clear, an earlier decrease in the sensitivity of the negative feedback effects of E2 is associated with a decreased inhibition of kisspeptin neuronal activity in the arcuate nucleus (ARC) [37,38]. ...
Article
Objectives were to test the hypothesis that pre and postnatal nutrition in the bovine female, independently or interactively, affect age at puberty and functional characteristics of the estrous cycle of sexually mature offspring. Brangus and Braford (n = 97) beef cows bearing a female fetus were fed to achieve body condition scores of 7.5–8 (H, obese), 5.5–6 (M, moderate) or 3–3.5 (L, thin) by the start of the third trimester and maintained until parturition. Heifer offspring were weaned and fed to gain weight at either a high (H; 1 kg/d) or low (L; 0.5 kg/d) rate between 4 and 8 months of age, then fed the same diet during a common feeding period until puberty which resulted in compensatory growth of heifers in the L group. Heifers (n = 95) from the H postnatal diet reached puberty two months earlier (12 ± 0.4 months; P = 0.0002) than those from the L postnatal diet (14 ± 0.4 months). Estrous cycles of a subgroup of postpubertal heifers (n = 53) were synchronized to evaluate antral follicle count (AFC), rate of growth and size of the pre-ovulatory follicle, size of corpus luteum and ovary, endometrial thickness, and plasma concentrations of progesterone and estradiol-17β (E2). Although there was a trend for postnatal H heifers to have greater AFC and plasma concentrations of E2 compared to L heifers, neither pre nor postnatal nutrition affected any other physiological or hormonal variables, including short-term fertility. Postnatal nutritional effects on pubertal age remained the dominant observed feature.
... There is a growing body of evidence supporting the relevance of early life nutrition, and thus the crosstalk between metabolism and hypothalamic responses are important for molecular events associated with sexual maturation [20,29]. However, these studies focus on alterations within the hypothalamus and to our knowledge, no reports exist about the nutritional effects during early life on the molecular level of the female reproductive tissues in dairy cattle. ...
Article
Full-text available
Background Nutrition has not only an impact on the general wellbeing of an animal but can also affect reproductive processes. In cattle, feeding regimes can influence the age of puberty onset and alter gonadal development. We analyzed effects of different milk replacer (MR) feeding regimes during rearing on ovarian physiology with specific emphasis on the numbers as well as gene expression characteristics of granulosa cells (GCs) at the age of puberty onset. Two groups of calves received either 10% or 20% of bodyweight MR per day during their first 8 weeks. After weaning, both groups were fed the same mixed ration ad libitum until slaughter at 8 months. Results Animals of the 20% feeding group had a significantly higher body weight, but the proportion of animals having a corpus luteum at the time of slaughter was not different between groups, suggesting a similar onset of puberty. Calves of the 10% group showed a constant GC count regardless of the number of follicles (r = 0.23) whereas in the 20% group increasing numbers of GCs were detected with a higher follicle count (r = 0.71). As a first effort to find a possible molecular explanation for this unexpected limitation of GC numbers in the 10% group, we comparatively analyzed GC transcriptomes in both diet groups. The mRNA microarray analysis revealed a total of 557 differentially expressed genes comparing both groups (fold change > |1.5| and p < 0.05). OAS1X, MX2 and OAS1Z were among the top downregulated genes in the 20% vs. the 10% group, whereas top upregulated genes comprised BOLA and XCL1. All of these genes are known to be regulated by interferon. Subsequent signaling pathway analysis revealed the involvement of several immune response mechanisms in accordance with a number of interferons as upstream regulators. Conclusions The results indicate that the plane of MR feeding in early life has an impact on the number and physiology of GCs later in life. This might influence the overall reproductive life initiated by the onset of puberty in cattle. In addition, the observed alterations in GCs of calves fed less MR might be a consequence of interferon regulated immunological pathways.
... Even if calves had the capacity to select forage of greater quality, as it occurs in lambs, the CP and fibre content of the forage during summer is limiting for optimum growth [22]. Moreover, in female calves it has been suggested that there is a pre-weaning nutritional programming window that affects the onset of puberty [23,24], causing endocrinal changes associated to greater reproductive efficiency [25,26]. Therefore, calf supplementation is recommended in both the short and long-term if the technique is economic viable. ...
Article
Full-text available
Objective: The objective was to test if creep feeding (CF) improves the average daily gain (ADG) and weaning weight of calves submitted to temporary weaning (TW) and if the combination of CF and TW improves conception and pregnancy rates of cows. Methods: Primiparous (n = 74) and primiparous and multiparous (n = 104) cows grazing native grasslands were used in experiment 1 and 2; respectively. The experimental design was in plots divided into complete random blocks with two replications. CF was the big plot and TW the small plot, thus four experimental groups were formed: 1) -CF-TW (n = 21 and 27); 2) -CF+TW (n = 16 and 24); 3) +CF-TW (n = 20 and 26); 4) +CF+TW (n=17 and 27) with cow-calf pairs for experiments 1 and 2; respectively. Nose plate application for TW had a duration of 14 and 15 days for experiment 1 and 2: respectively. In experiment 1, calves were fed at 1 % of live weight for 112 days using a commercial supplement with 18.4 % crude protein. In experiment 2, the supplementation lasted 98 days, and was carried out with corn dried distillers grains with soluble (DDGS) at 40 % of the potential intake on a daily basis. Results: TW reduced ADG during the TW period and the following 14 days, but the negative effect of TW was maintained until the final weaning only in experiment 2. CF increased ADG during TW period in both experiments. TW promoted an earlier conception of the dams (12 days in -CF treatment and 19 days in +CF treatment, P<0.01) and CF increased pregnancy rate in experiment 1, being the effects not consistent between experiments. Conclusion: CF consistently promoted an increase in ADG during the period of TW and increased final weaning weight of calves, therefore it is economically viable.
... The increase in weight indicates to the female organism that the environment is favourable for the beginning of reproductive activity. This indicator is controlled by a complex hormonal system that involves leptin and insulin-like growth factor-1 (IGF-1), responsible for activation of the neuroendocrine pathways that lead the physiological system towards reproduction (Allen et al., 2012). ...
Article
The aim of this study was to evaluate over two reproductive years the factors that influence the probability of pregnancy in 185 multiparous beef cows with early-weaned calves. The effect of the following parameters were evaluated: the year; the individual and maternal heterozygosity; the percentage of Zebu in the genotype; the weight at calving, weaning and at the end of the breeding season; the body condition score at calving; the age of the cow, the Julian calving date and milk production. Logistic regression was used to analyse the variables using the LOGISTIC procedure. The variables included in the regression equation were tested for multicollinearity, and the parameters evaluated using odds ratio statistics, in which changes in the chances of pregnancy were estimated as a function of the additions to each regressor variable. The average birth rate was 74.1%. For each year beyond the mean age of the herd under evaluation (6 years) the probability of pregnancy is expected to increase by 36.6%. Every seven days from 31 October onwards the probability of pregnancy reduces by 14.3%. For every ten kilograms above the mean body weight at the end of breeding (414.5 kg) the probability of pregnancy increases by 7%. Milk production has no influence on the pregnancy of beef cows with early-weaned calves. The weight at the end of the breeding season is a good indicator of the reproductive performance of the cow, which has a higher probability of pregnancy the greater the age and the earlier calving occurs during the calving season.
Article
Perinatal nutrition modulates the hypothalamic neurocircuitries controlling GnRH release, thus programming pubertal maturation in female mammals. Objectives of experiments reported here were to test the hypotheses that prenatal nutrition during mid- to late gestation interacts with postnatal nutrition during the juvenile period in heifer offspring to alter expression of leptin receptor (LepR) variants (ObRa, ObRb, ObRc, ObRt), and lipoprotein transporter molecules (LRP1 and 2) in the choroid plexus, leptin transport across the blood–brain barrier, and hypothalamic-hypophyseal responsiveness to exogenous ovine leptin (oleptin) during fasting. Nutritional programming of heifers employed a 3 x 2 factorial design of maternal (High, H; Low, L; and Moderate, M) x postnatal (H and L) dietary treatments. Results (Expt. 1) demonstrated that prepubertal heifers born to L dams, regardless of postnatal diet, had reduced expression of the short isoform of ObRc compared to H and M dams, with sporadic effects of undernutrition (L or LL) on ObRb, ObRt, and LRP1. Intravenous administration of oleptin to a selected postpubertal group (HH, MH, LL) of ovariectomized, estradiol implanted heifers fasted for 56 hours (Expt. 2) did not create detectable increases in third ventricle CSF but increased gonadotropin secretion in all nutritional groups tested. Previous work has shown that leptin enhances gonadotropin secretion during fasting via effects at both hypothalamic and anterior pituitary levels in cattle. Given the apparent lack of robust transfer of leptin across the blood–brain barrier in the current study, effects of leptin at the adenohypophyseal level may predominate in this experimental model.
Article
The availability of high-quality semen from genetically elite bulls is essential to support continued genetic gain and the sustainability of cattle production worldwide. While reducing the age at which usable semen is available also reduces the generation interval, it is dependent on timely onset of puberty in young bulls. There is now good evidence that hastened sexual development in bulls is achieved through enhancing nutrition in early life. This review will cover the physiological and molecular-based response to prevailing diet in key organs that orchestrate the ontogeny of sexual development in the bull calf. Given the central importance of the interaction between metabolic status and neuronal function to the progression of sexual development, we will discuss how communication between metabolic organs, reproductive organs and the brain are mediated via molecular and physiological processes. The availability of high-throughput nucleic acid and protein sequencing technologies and innovative data analytics have allowed us to improve our understanding of molecular regulation of puberty and sexual development. Analysing data from a number of organs, simultaneously, allows for a better understanding of the underlying biology and biochemical interactions that are influencing sexual development. Specifically, we can determine how early life nutritional interventions augment changes in potential key molecules regulating sexual development. Ultimately, a greater understanding of the inherent regulation of postnatal sexual development in the bull calf and how strategically targeted nutritional management can advance the ontogeny of this process, will facilitate the timely availability of high-quality semen from genetically elite animals, thus supporting more economically and environmentally sustainable beef and dairy production systems.
Article
Pubertal attainment is an intricate biological process that involves maturation of the reproductive neuroendocrine axis and increased pulsatile release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone. Nutrition is a critical environmental factor controlling the timing of puberty attainment. Nutrient restriction during early postnatal development delays puberty, whereas increased feed intake and adiposity during this period hasten pubertal maturation by imprinting the hypothalamus. Moreover, the dam’s nutrition during gestation can program the neuroendocrine system in the developing fetus and has the potential to advance or delay puberty in the offspring. Leptin, a hormone produced primarily by adipose cells, plays an important role in communicating energy status to the brain and regulating sexual maturation. Leptin’s regulation of GnRH release is mediated by an upstream neuronal network since GnRH neurons do not contain the leptin receptor. Two groups of neurons located in the arcuate nucleus of the hypothalamus that express neuropeptide Y (NPY), an orexigenic peptide, and alpha melanocyte-stimulating hormone (αMSH), an anorexigenic peptide, are central elements of the neural circuitry that relay inhibitory (NPY) and excitatory (αMSH) inputs to GnRH neurons. Moreover, KNDy neurons, neurons in the arcuate nucleus that co-express kisspeptin, neurokinin B (NKB), and dynorphin, also play a role in the metabolic regulation of puberty. Our studies in beef heifers demonstrate that increased rates of BW gain during early postweaning (4–9 mo of age) result in reduced expression of NPY mRNA, increased expression of proopiomelanocortin and kisspeptin receptor mRNA, reduced NPY inhibitory inputs to GnRH neurons, and increased excitatory αMSH inputs to KNDy neurons. Finally, our most recent data demonstrate that nutrition of the cow during the last two trimesters of gestation can also induce transcriptional and structural changes in hypothalamic neurocircuitries in the heifer progeny that likely persist long-term after birth. Managerial approaches, such as supplementation of the dam during gestation (fetal programming), creep feeding, early weaning, and stair-step nutritional regimens have been developed to exploit brain plasticity and advance pubertal maturation in heifers.
Article
Full-text available
We tested the hypothesis that recombinant ovine leptin would attenuate the acute effects of neu-ropeptide Y (NPY) on secretion of GH and gonadotropins (LH and FSH) in cows. Ovariectomized cows (n = 6) fitted with third ventricle guide cannulas were assigned randomly to each of three groups in a Latin square arrangement: (1) control; saline treatment only, (2) NPY; saline followed by NPY, and (3) L-NPY; leptin pretreatment followed by NPY. Treatments were: s.c. injection of saline or leptin (30 g/kg BW) at time 0, i.v. injection of saline or leptin (30 g/kg BW) at 70 min, and intracerebroventricular (i.c.v.) injection of saline or NPY (500 g) at 90 min. Plasma leptin was elevated (P < 0.01) at least four-fold throughout the experiment in the L-NPY group. Mean plasma concentrations of LH declined within 1 h and were lower (P < 0.03) than controls in both the NPY and L-NPY groups beginning 2 h after NPY injection. An acute increase in plasma concentrations of GH was observed within 1 h after NPY in the NPY group and mean values were greater (P < 0.01) than controls. However, in the L-NPY group, leptin pretreatment attenuated the NPY effect on GH. Treatments had no effect on FSH secretion. Results confirm suppressive and stimulatory effects of Supported by USDA-NRI 00-35203-9132 and the Texas Agricultural Experiment Station. NPY on LH and GH secretion, respectively, and indicate that leptin can attenuate the acute effects of NPY on GH secretion in cattle.
Article
Full-text available
DAVID bioinformatics resources consists of an integrated biological knowledgebase and analytic tools aimed at systematically extracting biological meaning from large gene/protein lists. This protocol explains how to use DAVID, a high-throughput and integrated data-mining environment, to analyze gene lists derived from high-throughput genomic experiments. The procedure first requires uploading a gene list containing any number of common gene identifiers followed by analysis using one or more text and pathway-mining tools such as gene functional classification, functional annotation chart or clustering and functional annotation table. By following this protocol, investigators are able to gain an in-depth understanding of the biological themes in lists of genes that are enriched in genome-scale studies.
Article
The common approach to the multiplicity problem calls for controlling the familywise error rate (FWER). This approach, though, has faults, and we point out a few. A different approach to problems of multiple significance testing is presented. It calls for controlling the expected proportion of falsely rejected hypotheses — the false discovery rate. This error rate is equivalent to the FWER when all hypotheses are true but is smaller otherwise. Therefore, in problems where the control of the false discovery rate rather than that of the FWER is desired, there is potential for a gain in power. A simple sequential Bonferronitype procedure is proved to control the false discovery rate for independent test statistics, and a simulation study shows that the gain in power is substantial. The use of the new procedure and the appropriateness of the criterion are illustrated with examples.
Conference Paper
Increased body weight gain during the juvenile period leads to early maturation of the reproductive neuroendocrine system and accelerates age at puberty. Although the mechanisms and pathways involved in this process are unclear, leptin and leptin-sensitive cells in the central nervous system may play critical roles. Leptin inhibits the expression of neuropeptide Y (NPY), an orexigenic peptide implicated in mediating the inhibitory effects of undernutrition on GnRH release. Leptin is also involved in regulating neural plasticity of hypothalamic pathways that control feed intake. In an earlier study, we observed that heifers fed to gain weight at a high rate during the juvenile period had elevated circulating concentrations of leptin and reduced NPY expression in the arcuate nucleus. In this study, we hypothesized that those nutritional regimens that accelerate the onset of puberty in heifers lead to diminished NPY innervation of GnRH neurons. Crossbred beef heifers were weaned at approximately 4 mo of age and fed diets to promote an average daily gain of 0.5 (Low Gain, LG; n=6) or 1.0 kg (High Gain, HG; n=6) for 15 wk. At the end of the experimental feeding period, heifers were humanely slaughtered and a block of tissue containing the preoptic area (POA) and hypothalamus was collected. Tissue blocks were cut in 50-µm sections and a series of sections 200 µm apart were processed for double immunofluorescent detection of NPY and GnRH. Sections were examined using an epifluorescent microscope and the proportion of GnRH neurons in direct apposition to NPY-containing varicosities, as well as the number of NPY close contacts per GnRH soma and proximal dendrites, were recorded and compared between treatments. Mean body weight was greater in the HG group at week 10 of the experiment and remained greater until slaughter. Mean circulating concentrations of leptin increased during the experiment in HG heifers and were greater in HG than in LG heifers for the last 3 wk of the experiment. Analysis of GnRH/NPY labeling in tissue sections from 3 heifers per group indicated that approximately 60% of the GnRH neurons in the POA and hypothalamus were in close proximity to NPY-containing fibers. Nutritional treatment had no effect on this proportion. The number of NPY close contacts per GnRH soma and proximal dendrites in the POA and anterior hypothalamus averaged 4.7 ± 0.8 and did not differ between LG and HG heifers. However, the number of NPY close contacts per GnRH soma and proximal dendrites in the mediobasal hypothalamus (MBH) was less in HG heifers (4.2 ± 1.4) than in LG heifers (8.2 ± 0.9). In addition, the proportion of GnRH neurons highly innervated by NPY fibers (10 or more contacts/soma and proximal dendrites) was less in HG heifers (18%) than in LG heifers (71%). Results from the current experiment provided evidence for structural plasticity of the NPY circuitry in response to elevated body weight gain during the juvenile period. Because in sheep, pulsatile release of LH has been associated with activation of GnRH neurons in the MBH, diminished NPY innervation of GnRH neurons in the MBH may be involved in the functional maturation of the neuroendocrine system and in facilitating the onset of puberty in females. Supported by USDA-NIFA-AFRI 2009-65203-05678. (platform)
Article
Neuropeptide Y (NPY) is known to be involved in the central regulation of appetite, sexual behavior, and reproductive functions. Whereas central administration of NPY strongly stimulates feeding in satiated animals, diet restriction produces overexpression of NPY in the arcuate and paraventricular nuclei that might reflect behavioral adaptations to shortage of food. Previous studies indicated that central administration of NPY resulted in controversial actions on LH secretion, either stimulatory or inhibitory. In order to analyze the chronic effect on pituitary function of centrally administered NPY, stainless steel cannulae were implanted in the right lateral ventricles of intact 45-day-old Sprague-Dawley female rats. Ten days later, Alzet osmotic minipumps filled with saline or different concentrations of NPY adjusted to deliver 3, 6, 12, or 18 micrograms/day were connected to the intracerebroventricular (icv) cannulae, implanted sc dorsally, and the effects of these treatments evaluated after 7 days. C...
Article
The distribution of immunoreactive (IR) estrogen receptor (ER)-containing cells was studied in the brains of adult Suffolk ewes using a rat monoclonal antibody (H222) which recognizes the human estrogen receptor. IR cells were characterized by dense nuclear reaction product, and in some instances, cytoplasmic immunostaining which filled dendrite-like processes. The greatest densities of ER-IR cells were found in the medial preoptic area, the mediobasal hypothalamus, and in a number of limbic system structures (amygdala, bed nucleus of the stria terminalis, lateral septum). ER-IR cells were found at lower densities in several other subregions of the hypothalamus and limbic system, and in the periaqueductal gray of the caudal midbrain. Cytoplasmic ER immunoreactivity was most prominent among ER-IR cells in the ventrolateral-ventromedial nucleus, the bed nucleus of the stria terminalis, the midbrain periaqueductal gray, and some ER-IR cells in the substantia innominata. The distribution of ER-containing cells in the sheep brain closely parallels that seen in other mammals. ER-IR cells are found in sites such as the medial preoptic area and ventrolateral-ventromedial hypothalamus which have been implicated as targets in this species and others for the influence of estradiol on sexual behavior and reproductive neuroendocrine function.