You are on page 1of 113

An International Journal

Aims and scope. Animal Reproduction Science publishes original research and timely reviews on topics relating to reproduction and fertility in
animals. This includes both fundamental research and applied studies, including management practices that increase our understanding of the
biology and manipulation of reproduction. The focus is on animals that are useful to humans including food and fiber producing; companion/
recreational; captive; and endangered species including zoo animals. Animal Reproduction Science has no page charges and only publishes
papers after rigorous peer review.
The journal’s scope includes the study of reproductive physiology and endocrinology, reproductive cycles, natural and artificial control of reproduction,
preservation and use of gametes and embryos, pregnancy and parturition, infertility and sterility, diagnostic and therapeutic techniques but excludes in
vitro studies of oocytes and embryos.

Editors-in-Chief
A.C.O. Evans, Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
Phone: (+353) 1 7166271 Fax: (+353) 1 7166253
G.N. Hinch, School of Environmental and Rural Science, University of New England, Armidale, NSW 2351, Australia
Phone: (+61) 2 67732202 Fax: (+61) 2 67733922
J.E. Kinder, Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210-1095, USA
Phone: +1 614 292 3232 Fax: +1 614 292 2929
Reviews Editor:
K.L. Macmillan, University of Melbourne, Veterinary Clinical Centre, 250 Princes Highway, Werribee, VIC 3030, Australia
Book Review Editor
N. Prathalingam, Newcastle Fertility Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom

Editorial Advisory Board


B.M. Alexander (Laramie, WY, USA) W. Jöchle (Manahawkin, NJ, USA)
L.L. Anderson (Ames, IA, USA) A.L. Johnson (University Park, PA, USA)
D.T. Armstrong (Adelaide, Australia) R.J. Mapletoft (Saskatoon, SK, Canada)
C. Aurich (Vienna, Austria) W.M.C. Maxwell (Sydney, NSW, Australia)
F.W. Bazer (College Station, TX, USA) J.F. Mee (Fermoy, Co. Cork, Ireland)
P.J. Broadbent (Aberdeen, UK) M. Mihm (Glasgow, UK)
H. Cardenas (Columbus, OH, USA) J.C.F. Moraes (Bage, Brazil)
J. Cavalieri (Townsville, QLD, Australia) T. Nagai (Ibaraki, Japan)
D. Cavestany (Montevideo, Uruguay) R.L. Nebel (Blacksburg, VA, USA)
P. Comizzoli (Washington DC, USA) J.E. Parks (Ithaca, NY, USA)
R.A. Cushman (Clay Center, NE, USA) N. Parvizi (Neustadt, Germany)
R.A. Dailey (Morgantown, WV, USA) R. Pursley (East Lansing, MI, USA)
J.A. Delgadillo (Torren, Coahuila, Mexico) J.P. Ravindra (Bangalore, India)
M.G. Diskin (Athenry, Co. Galway, Ireland) S. Revell (Ruthin, UK)
M.A. Driancourt (Nouzilly, France) R.R. Santos (Utrecht, The Netherlands)
A.D. Ealy (Gainesville, FL, USA) D.C. Sharp (Gainesville, FL, USA)
T. Fair (Dublin, Ireland) J.F. Smith (Hamilton, New Zealand)
W. Flowers (Raleigh, NC, USA) D.L. Thompson (Baton Rouge, LA, USA)
J. Ford (Clay Center, NE, USA) Y. Tsunoda (Nara, Japan)
D.L. Garner (Graeagle, CA, USA) A. Turner (Burwood, Victoria, Australia)
P.J. Hansen (Gainesville, FL, USA) S. Uzbekova (Nouzilly, France)
W.V. Holt (London, UK) J.L. Vallet (Clay Center, NE, USA)
J.S.M. Hutchinson (Aberdeen, UK) P.J. Verma (Clayton, Victoria, Australia)
K. Imakawa (Tokyo, Japan) T. Zhang (Luton, UK)

Publication Information. Animal Reproduction Science (ISSN 0378-4320). For 2010 volumes 117-123 (28 issues) are scheduled for
publication. Subscription prices are available upon request from the Publisher or from the Elsevier Customer Service Department nearest
you or from this journal’s website (http://www.elsevier.com/locate/anirep). Further information is available on this journal and other
Elsevier products through Elsevier’s website: (http://www.elsevier.com). Subscriptions are accepted on a prepaid basis only, and are
entered on a calender year basis. Issues are sent by surface mail (surface within Europe, air delivery outside Europe). Priority rates are
available upon request. Claims for missing issues should be made within six months of our publication (mailing) date.
Animal Reproduction Science 121 (2010) 189–196

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Review article

Impact of maternal nutrition on ovine foetoplacental development:


A review of the role of insulin-like growth factors
U.M. Igwebuike a,b,∗
a
Department of Veterinary Anatomy, University of Nigeria, Nsukka, Nigeria
b
Abdus Salam International Centre for Theoretical Physics, Trieste, Italy

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

Article history: The intrauterine environment may impact the conceptus (embryo/foetus and associated
Received 15 September 2009 extra-embryonic membranes) during ‘critical periods’ of development, when rapid cell divi-
Received in revised form 29 March 2010
sions occur in various tissues of the body, and may alter expression of the foetal genome,
Accepted 6 April 2010
and this may have lifelong consequences; a phenomenon known as foetal programming.
Available online 13 April 2010
Among intrauterine environmental factors, nutrition appears to play the most critical role
in influencing placental and foetal growth. Changes in maternal nutritional status during
Keywords:
pregnancy often results in permanent structural and functional deficits in foetal, as well as,
Maternal nutrition during pregnancy
Insulin-like growth factors postnatal growth of animals. Maternal undernutrition or overnutrition during pregnancy
Foetal growth can impair foetal growth. Alterations of the insulin-like growth factor cascade are spec-
Placental growth ulated to play a significant role in intrauterine nutrition-associated compromised foetal
Sheep growth and foetal programming. Insulin-like growth factors, IGF-1 and IGF-2 are nutrition-
ally sensitive proteins that are believed to modulate foetal and placental growth. The role
of these growth factors in nutrition-associated deficits in ovine foetal and placental growth
is the subject of this review.
© 2010 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
2. Significance of maternal nutrition during gestation for foetoplacental growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
2.1. Impact of maternal nutrition during pregnancy on foetal growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
2.2. Impact of maternal nutrition during pregnancy on placental growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
3. Insulin-like growth factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
3.1. Expression of insulin-like growth factors in ovine foetoplacental tissues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
3.2. Role of insulin-like growth factors in nutrition-associated foetal growth deficits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
3.3. Role of insulin-like growth factors in placental growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194

∗ Correspondence address: Department of Veterinary Anatomy, University of Nigeria, Nsukka, Nigeria. Tel.: +234 8038726150.
E-mail addresses: abuchi2002@yahoo.com, udensi.igwebuike@unn.edu.ng.

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.04.007
190 U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196

1. Introduction 2.1. Impact of maternal nutrition during pregnancy on


foetal growth
The intrauterine environment is a major determinant
of foetal growth, since foetal developmental pathways With regard to maternal nutrition influencing birth
are dependent on such signaling molecules as hormones, weight in the sheep, much information is available,
nutrients and metabolites, which must be present at but occasional discrepancies arise, most likely due to
the right time and in the right proportions. Whilst the study–study differences between breed of sheep, sample
foetal genome undoubtedly determines growth potential size, definition of ‘optimum’ or 100% requirement (Russel,
in utero, there is evidence that its role may be subor- 1971; Russel and Foot, 1973; Robinson, 1977; Mellor and
dinate (Snow, 1989). The intrauterine environment may Matheson, 1979; Wallace et al., 1996; Heasman et al.,
impact the conceptus during ‘critical periods’ of develop- 2000). However, there is growing evidence that mater-
ment, when rapid cell divisions occur in various tissues nal nutritional status can alter the epigenetic state of
of the body, and may alter expression of the foetal the foetal genome and imprint gene expression. Epige-
genome, and this may have lifelong consequences (Godfrey netic alterations (stable alterations of gene expression
and Robinson, 1998). This phenomenon is referred to as through covalent modifications of DNA and core histones)
foetal programming (Lucas, 1991). Experimental studies in early embryos may be carried forward to subsequent
have documented many examples of foetal programming, developmental stages (Waterland and Jirtle, 2004). Thus,
including long-term effects of alterations in the dam’s diet nutritional insult during a critical period of gestation may
during pregnancy on postnatal growth, relative organ size leave a permanent “memory” throughout life. Either mater-
and lipid metabolism of the offspring (Barker, 1994). The nal undernutrition or overnutrition during pregnancy is
role of intrauterine environment in foetal development has capable of producing impaired foetal growth.
been supported by embryo transfer studies, which have Maternal undernutrition during pregnancy may result
shown that it is the recipient mother rather than the donor from dietary deficiencies of essential nutrients (e.g., energy,
mother that strongly influences foetal growth (Brooks et al., protein and micronutrients). There is evidence (Wu et al.,
1995). Among intrauterine environmental factors, nutri- 1998) that foetal growth is most vulnerable to maternal
tion appears to play the most critical role in influencing dietary deficiencies of these nutrients. A 30% reduction in
placental and foetal growth (Barker and Clark, 1997). Stud- maternal nutrition from at least 45 days before until 7
ies have shown that the allocation of cells to the inner days after conception resulted in an increase, in late ges-
and the outer cell masses during early embryonic stage tation, in arterial blood pressure and in the activation of
of development is influenced by nutrition and hormones the foetal pituitary–adrenal axis in twin but not singleton
(Kleeman et al., 1994; Walker et al., 1996). The inner cell ovine foetuses (Edwards and McMillen, 2002a,b). The dif-
mass gives rise to the embryo proper, while the extra- ferential effects of preconceptional undernutrition in twin
embryonic membranes (placenta) arise from the outer cell and singleton pregnancies was attributed to the fact that
mass. The varying critical periods during which organs and foetal growth trajectory in early pregnancy is related to
systems mature indicate that inappropriate foetal nutrition foetal number, which may be important in determining the
at different developmental stages may likely have spe- differential impact of periconceptional undernutrition on
cific short- and long-term effects (Godfrey and Robinson, singletons and twins.
1998). Overnutrition during pregnancy can result from
increased intake of energy and/or protein by the dam.
2. Significance of maternal nutrition during It has been demonstrated that maternal overnutrition
gestation for foetoplacental growth retards placental and foetal growth, and increases foetal
and neonatal mortality in some animal species including
The foetus acquires nutrients from the maternal com- sheep (Wallace et al., 2003). In another study (Gardner
partment either directly as products of digestion following et al., 2007), maternal energy intake from early to mid-
maternal food ingestion, or as mobilized components of the gestation had little influence on lamb birth weight, but
maternal body reserves (McCrabb et al., 1992). Changes in late gestation intake was positively associated with weight
maternal nutritional status during pregnancy often results at term. It was suggested that the lack of effect of early
in permanent structural and functional deficits in foetal, as to mid-gestation intake is most likely through maternal
well as postnatal growth of animals (Robinson et al., 1999; ‘buffering’; that is, the response is dependent on maternal
Symonds et al., 2001; Osgerby et al., 2002). The effect of pre-pregnancy condition. Therefore, it would appear that
maternal nutrient intake on foetal development depends maternal body condition prior to pregnancy and energy
upon the timing, duration, and severity of the nutritional intake during late gestation are important factors in terms
insult (Robinson et al., 1999). The placenta is pivotal to of birth weight in sheep.
foetal development, mediating the supply of substrate and Furthermore, maternal nutritional status during preg-
waste products between the dam and the foetus (Bell et nancy may also affect the growth of specific foetal organs
al., 1999). The significance of placental function to foetal such as the liver and skeletal muscles (Brameld et al., 2000).
growth has been demonstrated in sheep, with restrictions Studies have shown that the foetal ovary is highly sensitive
to placental growth or blood supply by pre-mating carun- to maternal nutrition (Da Silva et al., 2003). Maternal over-
clectomy (Robinson et al., 1979), uterine artery ligation nutrition can induce alterations in ovarian development
(Emmanouilides et al., 1968) or heat stress (Galan et al., during late gestation (day 131 of gestation) independent
1999) resulting in foetal growth retardation. of changes in foetal weight or indeed in ovary weight
U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196 191

(Da Silva et al., 2002). Moreover, it has been reported mother (McCrabb et al., 1987, 1991; Faichney and White,
that in sheep, low maternal nutrition during pregnancy 1987).
resulted in delayed ovarian germ cell degeneration at day
42 (Borwick et al., 1997) and delayed onset of meiosis at 3. Insulin-like growth factors
day 65 of gestation (Rae et al., 2001), at a stage when
nutrient requirements for foetal body growth are mini- Insulin-like growth factors, IGF-1 and IGF-2 are nutri-
mal. It is thought that undernutrition-associated restriction tionally sensitive proteins that are believed to modulate
of placental growth, or placenta-mediated changes in the foetal and placental growth (Baker et al., 1993; Liu et
foetal endocrine environment may induce these detri- al., 1993). They act via the type-1 insulin-like growth
mental effects on ovarian development (Da Silva et al., factor receptor (IGF-1R) to instigate cellular prolifera-
2003). tion, differentiation and metabolism (Jones and Clemmons,
1995). This IGF-1R is structurally similar to the insulin
2.2. Impact of maternal nutrition during pregnancy on receptor, consisting of two ␣ and two ␤ subunits, which
placental growth combine to form a heterodimer (LeRoith et al., 1995). A
type-2 insulin-like growth factor receptor (IGF-2R) has
Ehrhardt and Bell (1995) observed that placental growth also been identified, but it acts as a degradative path-
in terms of mass and net cellular proliferation is maxi- way, removing excess IGF-2 from the circulation (Braulke,
mal in the first half of gestation in sheep. These authors 1999). This receptor, which is identical to the mannose-6-
demonstrated that a rapid increase in DNA synthesis and phosphate receptor, was originally isolated from placenta
tissue mass per nuclei number occurred between days 40 as a molecule that could bind IGF-2, but not IGF-1 (Braulke,
and 50 followed by proliferative growth to day 70. It was 1999). The IGFs act as progression factors in the cell cycle to
found that net cellular proliferation ceased by day 80 with increase DNA synthesis and cell differentiation in cultured
no change in tissue dry matter content observed there- embryos and several different foetal cell lines in vitro (Han
after. The indication is that placental size and even function and Fowden, 1994; Gardner et al., 1999).
later in gestation may be substantially influenced at this
early stage in sheep. The placenta of sheep is cotyledonary. 3.1. Expression of insulin-like growth factors in ovine
It exhibits discrete areas of attachment, the placentomes, foetoplacental tissues
which are interspersed among the interplacentomal areas
(Igwebuike, 2009). The placentomes are formed by inter- Local production of members of the IGF system has been
action of patches of the chorioallantois (cotyledons) with reported in the uterus and placenta of the ewe (Stevenson
raised aglandular areas of the endometrium called the et al., 1994; Reynolds et al., 1997). Polymerase chain reac-
caruncles. Intercaruncular areas of the endometrium are tion techniques have revealed the presence of mRNAs
intensely glandular (Bazer, 1975; Gray et al., 2001). Attach- for both IGF-1 and IGF-2 in sheep embryos throughout
ment of foetal cotyledons to the maternal caruncles occurs preimplantation development from single-cell stage to the
between days 25 and 30 of gestation, and growth restric- hatched blastocyst (Watson et al., 1994). Similarly, mRNAs
tion during this period can limit the eventual number of for these two IGFs were also expressed in both the mucosa
placentomes that form the placenta (Wathes et al., 1998). and muscle layers of the oviduct (Stevenson and Wathes,
Later in gestation, the size and morphology of the placen- 1996) within which early embryonic development takes
tomes are affected rather than their number (Vatnick et al., place. The mucosal expression of mRNA for IGF-1 was
1991; Wallace et al., 1996). maximal, but the concentrations for IGF-2 mRNA were
The consequences of variable nutrition in the first half much lower. Thus, locally produced IGF-1 is more likely
of pregnancy for ruminant placental growth have not been than IGF-2 to influence early stage of embryonic devel-
extensively studied. However, available evidence support opment. Following implantation, maternal uterine IGF-1
the idea that placental growth, and by implication, foetal mRNA expression was low (Reynolds et al., 1997). This
growth is most vulnerable to maternal nutritional status makes it unlikely that locally produced IGF-1 is impor-
during the peri-implantation period and the period of rapid tant in placental development. In contrast, IGF-2 mRNA
placental development (the first trimester of gestation) expression in maternal caruncles, placentome capsule and
(Wu et al., 2004). High nutrient intakes in early pregnancy endometrial stroma continued throughout the first half of
enhanced placental and, hence, foetal growth in ewes that gestation. Expression of IGF-2 mRNA in foetal mesodermal
were poorly nourished around the time of conception; con- tissues increased between days 14 and 35, and remained
versely, in ewes well nourished around conception, high high until parturition (Wathes et al., 1998). In addition to
intakes in early pregnancy suppressed placental growth, these local productions of the IGFs, the uterus will also
decreasing placental and foetal size (Robinson et al., 1994). be exposed to maternal circulating IGFs. Plasma levels of
Similarly, maternal feed restriction during the period from IGF-2 are about 5-fold higher than those of IGF-1 in sheep
early to mid-gestation also impacted placental growth (Gallaher et al., 1995). Similarly, plasma concentrations of
in the ewe. Placental weights were greater in ewes that IGF-2 in ovine foetal circulation were 3–10-fold higher than
received reduced nutrition in early and mid-gestation com- those of IGF-1 during late gestation (Owens et al., 1994). It
pared to control animals (Owens et al., 1986). It has been has been suggested that the placenta of ruminants is both
suggested that the increase in placental weight associated the source of foetal plasma IGF-2 and a site for IGF-1 clear-
with undernutrition during pregnancy is an attempt to ance from foetal circulation (Bassett et al., 1990; Holland et
compensate for the reduction in nutrient supply from the al., 1997). Abundance of IGF mRNA varies widely between
192 U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196

foetal tissues and with gestational age. Igf-2 gene expres- impact the foetal IGF system and therefore affect organo-
sion was particularly high in the lungs and kidneys while genesis and foetal development during critical stages of the
IGF-1 mRNA abundance was highest in liver and skeletal gestation period. Early to mid-gestational nutrient restric-
muscles (Delhanty and Han, 1993; Kind et al., 1995). Igf-1 tion significantly reduced foetal crown-rump length (CRL),
gene expression was up- and down-regulated during late changed the expression of IGF-1 and IGF-2 receptors in
gestation in liver and skeletal muscles respectively (Li et foetal myocardium, and played a role in cardiac ventric-
al., 1996; Forhead et al., 2002). The igf-2 gene expression ular enlargement in foetal sheep (Dong et al., 2005). On
was suppressed in liver and skeletal muscles, but not in the the other hand, administration of IGF-1 to foetal sheep
lungs and kidneys towards term in sheep (Li et al., 1993; increased the weight of specific foetal organs such as
Lu et al., 1994). spleen, lungs, heart, pituitary and adrenal glands (Lok et
al., 1996). Expression of insulin-like growth factor genes
3.2. Role of insulin-like growth factors in (igf genes) is developmentally regulated in a tissue-specific
nutrition-associated foetal growth deficits manner, and may be altered by nutritional and endocrine
conditions in utero (Ren et al., 1999; Fazio et al., 2000). In
Growth factors such as insulin-like growth factors (IGFs) general, the igf-1 gene is more responsive to these stimuli
may play an important role in regulating foetoplacental than the igf-2 gene. Thus, while the igf-1 gene is thought
growth (Dunk and Ahmed, 2000). Insulin-like growth fac- to regulate foetal growth and development in relation to
tors regulate nutrient partitioning by mediating maternal the nutrient supply, the igf-2 gene appears to produce the
and placental metabolism, as well as transplacental nutri- constitutive drive for intrauterine growth via its placental
ent transfer (Vernon et al., 1981; Liu et al., 1994; Kniss et and paracrine actions on foetal tissues (Fowden, 2003).
al., 1994). IGF-I influences amino acid and carbohydrate Six IGF binding proteins (IGFBPs) designated IGFBP-
metabolism in addition to the transplacental transfer of 1 to -6 control the biological activities of the IGFs by
amino acids and glucose from the mother to foetus (Kniss modulating the interaction of the IGFs with the IGF recep-
et al., 1994; Harding et al., 1994; Liu et al., 1994). Synthe- tors (Ferry et al., 1999). This indicates that the effects of
sis of insulin-like growth factors, or that of their receptors IGFs on foetal growth can be amplified or attenuated by
or binding proteins, may be affected by undernutrition. these IGFBPs, which are themselves regulated by nutri-
Indeed, maternal undernutrition during periconceptional tional and endocrine signals (Fowden, 2003). The IGFBPs
and gestational periods was shown to cause significant may serve not only to transport IGFs in the circulation but
falls in foetal insulin, insulin-like growth factors (IGFs) and also prolong the half-lives of the IGFs. In addition, it has
insulin-like growth factor binding protein-3 (IGFBP-3) lev- been postulated that IGFBPs can modulate IGF action by
els in sheep (Gallaher et al., 1998; Osgerby et al., 2002). In intercepting receptor interaction and ligand transduction
addition, plasma IGF-1 levels were reduced and elevated, (Wetterau et al., 1999) or by influencing the bioavailabil-
in nutrition restricted and overfed foetuses respectively ity of the IGFs (Jones and Clemmons, 1995). Studies with
(Dong et al., 2008). Maternal nutrient restriction in early to transgenic mice models demonstrated that elevated lev-
mid-gestation with re-feeding thereafter results in alter- els of IGFBP-1 are causal in the pathogenesis of growth
ations in hepatic and skeletal muscle expression of mRNA restriction, likely through a decrease in the bioavailabil-
for IGF-I, IGF-II and/or growth hormone receptor (GHR) ity of IGFs (Murphy, 2000; Watson et al., 2002). Thus,
in the foetus which may subsequently relate to altered decreased foetal growth occurred in mice that have ele-
organ and tissue function (Brameld et al., 2000). The foetal vated circulating concentrations of IGFBP-1 (Watson et
crown-rump length (CRL) was positively correlated with al., 2002) and in those with a paracrine model of expres-
maternal IGF-I levels (Osgerby et al., 2002), and change sion (Murphy, 2000). IGFBP-2, IGFBP-3 and IGFBP-6 mRNAs
in plasma IGF-1 levels correlates well with the absolute were all expressed in ovine placentome capsule, and IGFBP-
foetal weights under nutrition restriction (29.9% reduc- 3 and IGFBP-6 mRNAs were also present in the stroma
tion) and overfeeding (22.2% increase) (Dong et al., 2008). of the maternal villi, while IGFBP-3 and IGFBP-5 were
Moreover, concentrations of IGFs in the foetus in vivo are both expressed in both the luminal and glandular epithe-
positively correlated to birth weight in a number of species, lium of intercotyledonary endometrium (Osgerby et al.,
including sheep (Kind et al., 1995). Thus, alterations of the 2003). Foetal expression of IGFBPs is tissue specific and
insulin-like growth factor cascade are speculated to play developmentally regulated in most species including sheep
a significant role in intrauterine nutrition-associated com- (Delhanty and Han, 1993; Carr et al., 1995). IGFBP expres-
promised foetal growth and foetal programming. sion in the foetus may be affected by both nutritional and
Gene manipulation experiments have shown that IGF- endocrine conditions in utero. These conditions have more
1 affects foetal growth directly, but suggest that the pronounced effects on IGFBP-1, IGFBP-2 and IGFBP-4 than
growth-promoting actions of IGF-2 on the foetus may IGFBP-3 (Fowden, 2003). Osborn et al. (1992) reported that
be indirect, being mediated via changes in the growth tissue expression and plasma levels of IGFBP-1 were ele-
and nutrient-transport capacity of the placenta. How- vated by foetal nutrient restriction induced by maternal
ever, more detailed comparison of growth rates in various dietary restriction in sheep, but increase in foetal glucose
IGF mutants has shown that foetal growth is determined levels lowered hepatic expression and plasma levels of the
by the actions of IGF-1 on the type-1 IGF receptor, and IGFBP-1. In summary, IGFBP-1 and IGFBP-2 are increased
of IGF-2 on both the type-1 IGF and insulin receptors during nutrient restriction, but the reverse is true for IGFBP-
(Eggenschwiler et al., 1997; Louvi et al., 1997; Efstratiadis, 3 and IGFBP-4 (Osborn et al., 1992; Gallaher et al., 1995).
1998). It is possible that maternal undernutrition may These nutrition-induced changes in foetal IGFBP expres-
U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196 193

Fig. 1. A summary of the role of nutritionally sensitive insulin-like growth factors (IGFs) in ovine foetoplacental development.

sion may be related to the concomitant changes in the embryo carrying null mutations for igf-2 gene. In con-
foetal endocrine environment. It has been shown that hep- trast, igf-1 gene deletion did not influence placental weight.
atic expression and plasma concentrations of IGFBP-1 are Conversely, placentomegaly occurred when IGF-2 was
also reduced by insulin and increased by cathecolamines over-expressed (Ludwig et al., 1996). The growth stimula-
(Gallaher et al., 1994; Hooper et al., 1994). tory effect of IGF-2 on the placenta may be paracrine and/or
endocrine, but do not appear to be mediated via the type-1
3.3. Role of insulin-like growth factors in placental IGF receptor (Fowden, 2003). Placental growth was found
growth to be normal in double mutant mice lacking both type-1
IGF and insulin receptors, which suggests that IGF-2 may
Ovine placental growth is maximal during the first half act through an unknown placenta-specific receptor (Louvi
of pregnancy. Maternal plasma IGF-I was positively cor- et al., 1997). The existence of another type of IGF receptor in
related with the total placentome number, with levels the placenta may also explain the unusual characteristics of
being higher in high than low condition ewes (Osgerby IGF-1 binding observed in the ovine trophoblast between
et al., 2002). This suggests that high circulating maternal days 45 and 75 of gestation, when no igf-1 receptor gene
IGF-I concentrations may stimulate placentome forma- expression can be detected in the placentomes (Lacroix et
tion in ewes in good condition. Confirmation that the IGF al., 1995; Wathes et al., 1998).
system can control placental growth came from gene dele- In addition to their influence on placental growth,
tion studies (DeChiara et al., 1991; Baker et al., 1993) insulin-like growth factors also determine uptake and
that showed a deficiency in placental growth of mouse utilization of nutrients by placental and foetal tissues.
194 U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196

Administration of IGF-1 to either the foetus or dam was Edwards, L.J., McMillen, I.C., 2002b. Periconceptional nutrition pro-
shown to alter the transfer and partitioning of glucose and grams development of the cardiovascular system in the fetal
sheep. Am. J. Physiol. Regul. Integr. Comp. Physiol. 283, R669–
amino acids between ovine foetal and uteroplacental tis- R679.
sues (Harding et al., 1994; Liu et al., 1994). A summary of the Efstratiadis, A., 1998. Genetics of mouse growth. Int. J. Dev. Biol. 42,
role of insulin-like growth factors in ovine foetoplacental 955–976.
Eggenschwiler, J., Ludwig, T., Fisher, P., Leighton, P.A., Tilghmen, S.M.,
development is given in Fig. 1. Efstratiadis, A., 1997. Mouse mutant embryos over-expressing IGF-II
In conclusion, maternal nutritional status during preg- exhibit phenotypic features of Beckman-Wiedemann and Simpson-
nancy is an important intrauterine environmental factor Golobi-Behmel syndromes. Genes Dev. 11, 3128–3142.
Emmanouilides, G.C., Townsend, D.E., Bauer, R.A., 1968. Effects of sin-
governing placental growth, as well as foetal growth and gle umbilical artery ligation in the lamb fetus. Pediatrics 42, 919–
programming. This influence of maternal nutrition on 927.
foetoplacental development may be mediated via its regu- Ehrhardt, R.A., Bell, A.W., 1995. Growth and metabolism of the ovine pla-
centa during mid-gestation. Placenta 16, 727–741.
lation of the insulin-like growth factor system axis.
Faichney, G.J., White, G.A., 1987. Effects of maternal nutritional status on
fetal and placental growth and on fetal urea synthesis in sheep. Aust.
J. Biol. Sci. 40, 365–377.
References Fazio, S., Palmieri, E.A., Biondi, B., Cittadini, A., Sacca, L., 2000. The role
of the GH-IGF-I axis in the regulation of myocardial growth: from
Baker, J., Liu, J.-P., Robertson, E.J., Efstratiadis, A., 1993. Role of insulin-like experimental models to human evidence. Eur. J. Endocrinol. 142, 211–
growth factors in embryonic and post-natal growth. Cell 75, 73–82. 216.
Barker, D.J.P., 1994. Mothers, Babies and Disease in Later Life. BMJ Pub- Ferry, R.J., Katz, L.E.L., Grimberg, A., Cohen, P., Weinzimer, S.A., 1999. Cel-
lishing Group, London. lular actions of insulin-like growth factor binding proteins. Horm.
Barker, D.J.P., Clark, P.M., 1997. Fetal undernutrition and disease in later Metab. Res. 31, 192–202.
life. Rev. Reprod. 2, 105–112. Forhead, A.J., Li, J., Gilmour, R.S., Dauncey, M.J., Fowden, A.L., 2002. Thy-
Bassett, N.S., Brier, B.H., Hodgkinson, S.C., Dans, S.R., Henderson, H.W., roid hormones and the mRNA of the GH receptor and IGFs in skeletal
Gluckman, P.D., 1990. Plasma clearance of radiolabelled IGF-I in the muscle of fetal sheep. J. Physiol. 282, E80–86.
late gestation ovine fetus. J. Dev. Physiol. 14, 73–79. Fowden, A.L., 2003. The insulin-like growth factors and feto-placental
Bazer, F.W., 1975. Uterine protein secretions: relationship to development growth. Placenta 24, 803–812.
of the conceptus. J. Anim. Sci. 41, 1376–1382. Galan, H.L., Hussey, M.J., Barbera, A., Ferrazzi, E., Chung, M., Hobbins, J.C.,
Bell, A.W., Hay, W.W., Ehrhardt, R.A., 1999. Placental transport of nutri- Battaglia, F.C., 1999. Relationship of fetal growth to the duration of
ents and its implications for fetal growth. J. Reprod. Fertil. Suppl. 54, heat stress in an ovine model of placental insufficiency. Am. J. Obstet.
401–410. Gynecol. 180, 1278–1282.
Borwick, S.C., Rhind, S.M., McMillen, S.R., Racey, P.A., 1997. Effect of Gallaher, B.W., Breier, B.H., Blum, W.F., McCutcheon, S.N., Gluckman, P.D.,
undernutrition of ewes from the time of mating on fetal ovarian devel- 1995. A homologous radioimmunoassay for ovine insulin-like growth
opment in mid gestation. Reprod. Fertil. Dev. 9, 711–715. factor-binding protein-2: ontogenesis and the response to growth
Brameld, J.M., Mostyn, A., Dandrea, J., Stephenson, T.J., Dawson, J.M., But- hormone, placental lactogen and insulin-like growth factor-1 treat-
tery, P.J., Symonds, M.E., 2000. Maternal nutrition alters the expression ment in sheep. J. Endocrinol. 144, 75–82.
of insulin-like growth factors in fetal sheep liver and skeletal muscle. Gallaher, B.W., Breier, B.H., Keven, C.L., Harding, J.E., Gluckman, P.D.,
J. Endocrinol. 167, 429–437. 1998. Fetal programming of insulin-like growth factor (IGF)-I and IGF-
Braulke, T., 1999. Type-2 IGF receptor: a multi-ligand binding protein. binding protein-3: evidence for an altered response to undernutrition
Horm. Metab. Res. 31, 242–246. in late gestation following exposure to periconceptual undernutrition
Brooks, A.A., Johnson, M.R., Steer, P.J., Pawson, M.E., Abdalla, H.I., 1995. in the sheep. J. Endocrinol. 159, 501–508.
Birth weight: nature or nurture? Early Hum. Dev. 42, 29–35. Gallaher, B.W., Oliver, M.H., Eichorn, K., Kessler, U., Kiess, W., Harding, J.E.,
Carr, J.M., Owens, J.A., Gran, P.A., Walton, P.E., Owens, P.C., Wallace, J.C., Gluckman, P.D., Breier, B.H., 1994. Circulating insulin-like growth fac-
1995. Circulating insulin-like growth factors (IGFs) IGF binding pro- tor II/mannose-6-phosphate receptor and insulin-like growth factor
teins (IGFBPs) and tissue mRNA levels of IGFBP2 and IGFBP-4 in the binding proteins in fetal sheep plasma are regulated by glucose and
ovine fetus. J. Endocrinol. 145, 545–557. insulin. Eur. J. Endocrinol. 131, 398–404.
Da Silva, P., Aitken, R.P., Rhind, S.M., Racey, P.A., Wallace, J.M., 2002. Impact Gardner, D.S., Buttery, P.J., Daniel, Z., Symonds, M.E., 2007. Factors affect-
of maternal nutrition during pregnancy on pituitary gonadotrophin ing birth weight in sheep: maternal environment. Reproduction 133
gene expression and ovarian development in growth-restricted and (1), 297–307.
normally grown late gestation sheep fetuses. Reproduction 123, Gardner, R.L., Squire, S., Zainan, S., Hills, S., Graham, C.F., 1999. Insulin-
769–777. like growth factor-2 regulation of conceptus composition: effects of
Da Silva, P., Aitken, R.P., Rhind, S.M., Racey, P.A., Wallace, J.M., the trophectoderm and inner cell mass genotypes in the mouse. Biol.
2003. Effect of maternal overnutrition during pregnancy on pitu- Reprod. 60, 190–195.
itary gonadotrophin gene expression and gonadal morphology in Godfrey, K., Robinson, S., 1998. Maternal nutrition, placental growth and
female and male sheep at day 103 of gestation. Placenta 24, 248– fetal programming. Proc. Nutr. Soc. 57 (1), 105–111.
257. Gray, C.A., Bartol, F.F., Tarleton, B.J., Wiley, A.A., Johnson, G.A., Bazer, F.W.,
DeChiara, T.M., Robertson, E.J., Efstratiadis, A., 1991. Parental imprinting Spencer, T.E., 2001. Developmental biology of uterine glands. Biol.
of the mouse insulin-like growth factor II gene. Cell 64, 849–859. Reprod. 62, 1311–1323.
Delhanty, P.J.D., Han, V.K.M., 1993. The expression of insulin-like growth Han, V.K.M., Fowden, A.L., 1994. Paracrine regulation of fetal growth. In:
factor (IGF)-binding 2 and IGF-II genes in the tissues of the developing Ward, R.H.T., Smith, S.K., Donnai, D. (Eds.), Early Fetal Growth and
ovine fetus. Endocrinology 132, 41–51. Development. RCOG Press, London, pp. 275–292.
Dong, F., Ford, S.P., Fang, C.X., Nijland, M.J., Nathanielsz, P.W., Ren, J., Harding, J.E., Liu, L., Evans, P.C., Gluckman, P.D., 1994. Insulin-like growth
2005. Maternal nutrient restriction during early to mid gestation up- factor 1 alters feto-placental protein and carbohydrate metabolism in
regulates cardiac insulin-like growth factor (IGF) receptors associated fetal sheep. Endocrinology 134, 1509–1514.
with enlarged ventricular size in fetal sheep. Growth Horm. IGF Res. Heasman, L., Brameld, J., Mostvn, A., Budge, H., Dawson, J., Buttery, P.,
15, 291–299. Stephenson, T., Symonds, M.E., 2000. Maternal nutrient restriction
Dong, F., Ford, S.P., Nijland, M.J., Nathanielsz, P.W., Ren, J., 2008. Influence during early to mid gestation alters the relationship between insulin-
of maternal undernutrition and overfeeding on cardiac ciliary neu- like growth factor I and bodyweight at term in fetal sheep. Reprod.
rotrophic factor receptor and ventricular size in fetal sheep. J. Nutr. Fertil. Dev. 12, 345–350.
Biochem. 19 (6), 409–414. Holland, M.D., Hossner, M.D., Williams, S.E., Wallace, C.R., Niswerder, G.D.,
Dunk, C., Ahmed, A., 2000. Growth factor regulation of placental angiogen- Odde, K.G., 1997. Serum concentrations of insulin-like growth factors
esis. In: Baker, P., Kingdom, J. (Eds.), Intrauterine Growth Retardation. and placental lactogen during gestation in cattle: fetal profiles. Dom.
Springer, London, pp. 149–166. Anim. Endocrinol. 14, 231–239.
Edwards, L.J., McMillen, I.C., 2002a. Impact of maternal undernutrition Hooper, S.A., Bocking, A.D., White, S.E., Fraher, L.J., McDonald, T.J., Han,
during the periconceptional period, fetal number, and fetal sex on V.K.M., 1994. Catecholamines stimulate the synthesis and release of
the development of the hypothalamo–pituitary–adrenal axis in sheep insulin-like growth factor binding protein-1 (IGFBP1) by fetal sheep
during late gestation. Biol. Reprod. 66, 1562–1369. liver in vivo. Endocrinology 134, 1104–1112.
U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196 195

Igwebuike, U.M., 2009. A review of uterine structural modifications that Osgerby, J.C., Wathes, D.C., Howard, D., Gadd, T.S., 2002. The effect of
influence conceptus implantation and development in sheep and maternal undernutrition on ovine fetal growth. J. Endocrinol. 173,
goats. Anim. Reprod. Sci. 112, 1–7. 131–141.
Jones, J.I., Clemmons, D.R., 1995. Insulin-like growth factors and their Owens, J.A., Falconer, I., Robinson, J.S., 1986. Effect of restriction of placen-
binding proteins: biological actions. Endocr. Rev. 16, 3–34. tal growth on umbilical and uterine blood flows. Am. J. Physiol. 250,
Kind, K.L., Owens, J.A., Robinson, J.S., Quinn, K.J., Grant, P.A., Walton, R427–R434.
P.E., Gilmour, R.S., Owens, P.C., 1995. Effect of restriction of placental Owens, J.A., Kind, K.L., Carbone, F., Robinson, J.C., Owens, P.C., 1994. Cir-
growth on the expression insulin-like growth factors in fetal sheep: culating insulin-like growth factor-I and II and substrate in fetal
relationship to fetal growth, circulating insulin-like growth factors sheep following restriction of placental growth. J. Endocrinol. 140,
and binding proteins. J. Endocrinol. 146, 23–34. 5–13.
Kleeman, D.O., Walker, S.K., Seamark, R.F., 1994. Enhanced fetal growth Rae, M.T., Palassio, S., Kyle, C.E., Brooks, A.N., Lea, R.G., Miller, D.W., Rhind,
in sheep administered progesterone during the first three days of S.M., 2001. Effect of maternal undernutrition during pregnancy on
pregnancy. J. Reprod. Fertil. 102, 411–417. early ovarian development and subsequent follicular development in
Kniss, D.A., Shubert, P.J., Zimmerman, P.D., Landon, M.B., Gabbe, S.G., 1994. sheep fetuses. Reproduction 122, 915–922.
Insulin-like growth factors: their regulation of glucose and amino acid Ren, J., Samson, W.K., Sowers, J.R., 1999. Insulin-like growth factor I as a
transport in placental trophoblasts isolated from first trimester chori- cardiac hormone: physiological and pathophysiological implications
onic villi. J. Reprod. Med. 39, 249–256. in heart disease. J. Mol. Cell Cardiol. 31, 2049–2061.
Lacroix, M.C., Servety, J.L., Kann, G., 1995. IGF-I and IGF-II receptors in Reynolds, T.S., Stevenson, K.R., Wathes, D.C., 1997. Pregnancy-specific
the sheep placenta: evolution during the course of pregnancy. J. alterations in the expression of the insulin-like growth factor system
Endocrinol. 144, 179–191. during early placental development in the ewe. Endocrinology 138,
LeRoith, D., Werner, H., Beitner-Johnson, D., Roberts, C.T., 1995. Molecu- 886–897.
lar and cellular aspects of the insulin-like growth factor 1 receptor. Robinson, J.J., 1977. The influence of maternal nutrition on ovine foetal
Endocr. Rev. 16, 143–163. growth. Proc. Nutr. Soc. 36, 9–16.
Li, J., Owens, J.A., Owens, P.C., Saunders, J.C., Fowden, A.L., Gilmour, R.S., Robinson, J.J., Sinclair, K.D., McEvoy, T.G., 1999. Nutritional effects on foetal
1996. The ontogeny of hepatic growth hormone receptor and insulin- growth. Anim. Sci. 68, 315–331.
like growth factor-I gene expression in the sheep fetus during late Robinson, J.S., Kingston, E.J., Jones, C.T., Thorburn, G.D., 1979. Studies on
gestation: developmental regulation by cortisol. Endocrinology 137, experimental growth retardation in sheep: the effect of removal of
1650–1657. endometrial caruncles on fetal size and metabolism. J. Dev. Physiol. 1,
Li, J., Saunders, J.C., Gilmour, R.S., Silver, M., Fowden, A.L., 1993. Insulin-like 379–398.
growth factor-II messenger ribonucleic acid expression in fetal tissue Robinson, J.S., Owens, J.A., de Barro, T., Lok, F., Chidzanja, S., 1994. Maternal
of the sheep during late gestation: effects of cortisol. Endocrinology nutrition and fetal growth. In: Ward, R.H.T., Smith, S.K., Donnai, D.
132, 2083–2089. (Eds.), Early Fetal Growth and Development. RCOG Press, London, pp.
Liu, J.-P., Baker, J., Perkins, A.S., Robertson, E.J., Efstratiadis, A., 1993. 317–334.
Mice carrying null mutations of the genes encoding insulin-like Russel, A.J., 1971. Relationships between energy intake and productivity
growth factor I (Igf-1) and type-1 IGF receptor (Igf1r). Cell 75, 59– in hill sheep. Proc. Nutr. Soc. 30, 197–204.
72. Russel, A.J., Foot, J.Z., 1973. The effect of level of nutrition at two stages of
Liu, L., Harding, J.E., Evans, P.C., Gluckman, P.D., 1994. Maternal insulin- pregnancy on the performance of primiparous ewes. Proc. Nutr. Soc.
like growth factor-I infusion alters feto-placental carbohydrate and 32, 27A–28A.
protein metabolism in pregnant sheep. Endocrinology 135, 895– Snow, M.H.L., 1989. Effects of genome on fetal size at birth. In: Sharp, F.,
900. Fraser, R.B., Milner, R.D.G. (Eds.), Fetal Growth, Proceedings of the 20th
Lok, F., Owens, J.A., Mundy, L., Robinson, J.S., Owens, P.C., 1996. Insulin- Study Group. RCOG Press, London, pp. 1–11.
like growth factor I promotes growth selectivity in fetal sheep in late Stevenson, K.R., Gilmour, R.S., Wathes, D.C., 1994. Localization of insulin-
gestation. Am. J. Physiol. 270, R1148–1155. like growth factor-I and -II messenger ribonucleic acid and type 1
Louvi, A., Accili, D., Efstratiadis, A., 1997. Growth-promoting interaction IGF receptors in the ovine uterus during the estrous cycle and early
of IGF-II with the insulin receptor during mouse embryonic develop- pregnancy. Endocrinology 134, 1655–1664.
ment. Dev. Biol. 189, 33–48. Stevenson, K.R., Wathes, D.C., 1996. Insulin-like growth factors and their
Lu, F., Han, V.K.M., Milne, W.K., Fraser, M., Carter, A.M., Berdusco, E.T.M., binding proteins in the ovine oviduct during the oestrous cycle. J.
Challis, J.R.G., 1994. Regulation of insulin-like growth factor-II gene Reprod. Fertil. 108, 31–40.
expression in the ovine fetal adrenal gland by adrenocorticotrophic Symonds, M.E., Budge, H., Stephenson, T., McMillen, I.C., 2001. Fetal
hormone and cortisol. Endocrinology 134, 2628–2635. endocrinology and development – manipulation and adaptation to
Lucas, A., 1991. Programming by early nutrition in man. In: Bock, Whelan, long-term nutritional and environmental challenges. Reproduction
J. (Eds.), The Childhood Environment and Adult Disease. John Wiley 121, 853–862.
and Sons, Chichester, pp. 38–55. Vatnick, I., Schoknecht, P.A., Darrigrand, R., Bell, A.W., 1991. Growth and
Ludwig, T., Eggenschwiler, J., Fisher, P., D’Ercole, A.J., Davenport, M.L., metabolism of the placenta after unilateral fetectomy in twin preg-
Efstratiadis, A., 1996. Mouse mutants lacking the type-2 IGF recep- nant ewes. J. Dev. Physiol. (Oxf.) 15, 351–356.
tor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null Vernon, R.G., Clegg, R.A., Flint, D.J., 1981. Metabolism of sheep adi-
backgrounds. Dev. Biol. 177, 517–535. pose tissue during pregnancy and lactation. Biochem. J. 200, 307–
McCrabb, G.J., Egan, A.R., Hosking, B.J., 1991. Maternal undernutrition 314.
during mid-pregnancy in sheep: placental size and its relationship Walker, S.K., Hartwich, K.M., Seamark, R.F., 1996. The production of unusu-
to calcium transfer during late pregnancy. Br. J. Nutr. 65, 157– ally large offspring following embryo manipulation: concepts and
168. challenges. Theriogenology 45, 111–120.
McCrabb, G.J., Egan, A.R., Hosking, B.J., 1992. Maternal undernutrition dur- Wallace, J.M., Aitken, R.P., Cheyne, M.A., 1996. Nutrient partitioning and
ing mid-pregnancy in sheep: variable effects on placental growth. J. fetal growth in rapidly growing adolescent ewes. J. Reprod. Fertil. 107,
Agric. Sci. 118, 127–132. 183–190.
McCrabb, G.J., Hosking, B.J., Egan, A.R., 1987. Nutrition manipulation of Wallace, J.M., Bourke, D.A., Aitken, R.P., Milne, J.S., Hay, W.W.,
placental size not the key to higher foetal growth. Proc. Nutr. Soc. 2003. Placental glucose transport in growth-restricted pregnancies
Aust. 12, 87–91. induced by overnourished adolescent sheep. J. Physiol. 547, 85–
Mellor, D.J., Matheson, I.C., 1979. Daily changes in the curved crown-rump 94.
length of individual sheep fetuses during the last 60 days of preg- Waterland, R.A., Jirtle, R.L., 2004. Early nutrition, epigenetic changes at
nancy and effects of different levels of maternal nutrition. Quart. J. transposons and imprinted genes, and enhanced susceptibility to
Exp. Physiol. Cogn. Med. Sci. 64, 119–131. adult chronic diseases. Nutrition 20, 63–68.
Murphy, L.J., 2000. Overexpression of insulin-like growth factor binding Wathes, D.C., Reynolds, T.S., Robinson, R.S., Stevenson, K.R., 1998. Role of
protein-1 in transgenic mice. Pediatr. Nephrol. 14, 567–571. the insulin-like growth factor system in uterine function and placental
Osborn, B.H., Fowlkes, J., Han, V.K.M., Freemark, M., 1992. Nutritional development in ruminants. J. Dairy Sci. 81, 1778–1789.
regulation of insulin-like growth factor-binding protein gene expres- Watson, C.S., Benson, M., Bialek, P., Yee, S.P., Han, V.K.M., 2002. Hep-
sion in the ovine fetus and pregnant ewe. Endocrinology 131, 1743– atic overexpression of insulin-like growth factor binding protein-1
1750. in fetal mice: a transgenic model of intrauterine growth restriction.
Osgerby, J.C., Gadd, T.S., Wathes, D.C., 2003. Effect of maternal body condi- In: 49th Annual Meeting of the Society for Gynecologic Inves-
tion on placental and fetal growth and insulin-like growth factor axis tigation, Los Angeles, USA, J. Soc Gynecol. Investig. 9 (Suppl),
in Dorset ewes. Reproduction 125, 717–731. p. 295A.
196 U.M. Igwebuike / Animal Reproduction Science 121 (2010) 189–196

Watson, A.J., Watson, P.H., Arcellia-Paulilo, M., Warnes, D., Walker, S.K., Wu, G., Pond, W.G., Flynn, S.P., Ott, T.L., Bazer, F.W., 1998. Maternal dietary
Schultz, G.A., Armstrong, D.T., Seamark, R.F., 1994. A growth factor protein deficiency decreases nitric oxide synthase and ornithine
phenotype map for ovine preimplantation development. Biol. Reprod. decarboxylase activities in placenta and endometrium of pigs during
50, 725–733. early gestation. J. Nutr. 128, 2395–2402.
Wetterau, L.A., Moore, M.G., Lee, K.W., Shim, M.L., Cohen, P., 1999. Novel Wu, G., Bazer, F.W., Cudd, T.A., Meininger, C.J., Spencer, T.E., 2004. Maternal
aspects of the insulin-like growth factor binding proteins. Mol. Genet. nutrition and fetal development. J. Nutr. 134, 2169–2172.
Metab. 68, 161–181.
Animal Reproduction Science 121 (2010) 197–207

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Review article

Disorders of sex development in the dog—Adoption of a new


nomenclature and reclassification of reported cases
T. Poth a,∗ , W. Breuer a , B. Walter b , W. Hecht c , W. Hermanns a
a
Institute of Veterinary Pathology, Ludwig-Maximilians-University, Veterinaerstrasse 13, D-80539 Munich, Germany
b
Clinic for Veterinary Surgery and Gynaecology for Small Animals, Ludwig-Maximilians-University, Veterinaerstrasse 13, D-80539 Munich, Germany
c
Institute of Veterinary Pathology, Justus-Liebig-University, Frankfurter Strasse 96, D-35392 Giessen, Germany

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

Article history: Intersexuality is a rare congenital abnormality in domestic animals. It is reported in numer-
Received 4 June 2009 ous species including the swine, goat, horse, cat, and dog. The present work provides an
Received in revised form 1 April 2010
overview of the variety of intersexual conditions known in different dog breeds. Each case
Accepted 9 April 2010
was reclassified based on the described gonadal constitution, reproductive tract abnormal-
Available online 24 April 2010
ities and karyogram, and categorised according to the stages normal sex development is
undergoing resulting in three main categories: (1) sex chromosome disorders, (2) disorders
Keywords:
of gonadal sex development, and (3) disorders of phenotypic sex development. Reclassifi-
Disorders of sex development
Sex reversal cation disclosed that the current classification scheme and terminology are inconsistently
Ovotestis used in literature masking the real occurrence and frequency of various intersex conditions
True hermaphroditism in dogs. For establishment of an individual, precise and definite diagnosis, introduction of
Pseudohermaphroditism a new nomenclature is proposed as recently recommended for humans. The new termi-
Dog nology is based on the gonosomal constellation and gonadal constitution, contributes to a
systematic classification of canine intersex cases, and replaces the common but confusing
diagnoses “true hermaphrodite” and “pseudohermaphrodite”.
The literature survey was supplemented by adding the results from own investigations
in a German Pinscher and Berger Picard dog with bilateral ovotestes and ambiguous exter-
nal genitalia. The diagnostic approach and clinical, pathomorphological and cytogenetic
findings were described in detail.
© 2010 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
2. Normal sex development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
3. Canine intersexuality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
3.1. Sex chromosome disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
3.2. Disorders of gonadal sex development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
3.3. Disorders of phenotypical sex development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202

Abbreviations: ADM, androgen-dependent masculinisation; AIS, androgen insensitivity syndrome; C, chimaera; DSD, disorder(s) of sex development; H&E,
haematoxylin & eosin; MIS, Müllerian inhibiting substance; M, mosaic; PMDS, persistent Müllerian duct syndrome; SR, sex reversal; SRY, sex-determining
region of the Y chromosome; TFS, testicular feminisation syndrome; XXSR, XX sex reversal; XYSR, XY sex reversal.
∗ Corresponding author. Tel.: +49 89 21802543; fax: +49 89 21802544.
E-mail address: Poth@patho.vetmed.uni-muenchen.de (T. Poth).

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.04.011
198 T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207

4. Reclassification of canine intersex cases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202


5. New nomenclature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
6. Diagnostic approach in intersex patients—report on two phenotypically female dogs with ovotestes . . . . . . . . . . . . . . . . . . . . . . . . . . 204
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206

1. Introduction each stage depending upon successful completion of the


previous one: establishment of chromosomal, gonadal, and
With the exception of bovine freemartinism, intersex- phenotypic sex (Meyers-Wallen and Patterson, 1986, 1989;
uality is considered an uncommon congenital disorder in Meyers-Wallen, 2001; Feldman and Nelson, 2004; Schlafer
domestic animals (Schlafer and Miller, 2007). The affected and Miller, 2007).
individuals have parts of or all genital organs of both sexes Chromosomal sex is determined at fertilisation result-
resulting in a variety of phenotypes (Meyers-Wallen and ing in XY and XX zygotes. Initially, early male and
Patterson, 1989; Simpson et al., 1998; Feldman and Nelson, female embryos have similar undifferentiated gonads,
2004; Schlafer and Miller, 2007). mesonephric (Wolffian) as well as paramesonephric (Mül-
Most intersex dogs are actually subcategorised into lerian) ducts, and primitive external genitalia consisting
true hermaphrodites and pseudohermaphrodites, respec- of the urogenital sinus, the genital tubercle, and geni-
tively. True hermaphroditism is defined by the presence tal swellings. The end of the undifferentiated period is
of both ovarian and testicular tissue in any possible com- marked by gonadal differentiation, which is normally
bination: (1) bilateral ovotestes, (2) unilateral ovotestis determined by the genetic sex. In male embryos, the SRY-
with contralateral ovary or testis, and (3) testis with gene representing the sex-determining region of the Y
contralateral ovary. Correspondingly, the three different chromosome initiates the differentiation of the primor-
forms are termed as bilateral, unilateral, and lateral true dial gonads into testes and is supported by the autosomal
hermaphroditism. Pseudohermaphrodites have one type of gene SOX9. The production of two testicular hormones,
gonadal tissue, either ovaries or testes, but the opposite testosterone and antiparamesonephric hormone (Mülle-
phenotype. The affected individuals are classified as a male rian inhibiting substance, MIS), induces the development
or female pseudohermaphrodite according to their gonadal of male genitalia. MIS is secreted by the sustentacular cells
sex. and causes regression of the embryonic paramesonephric
The establishment of an appropriate diagnosis in dogs ducts. Testosterone is secreted by the interstitial cells, and
may be problematical due to the variety of intersexual stimulates the mesonephric ducts to form epididymides
conditions and the presence of ambiguous external gen- and ductus deferentia. Testosterone is converted into dihy-
italia in most cases that initially allow no conclusions drotestosterone within the tissue of the primitive external
about the underlying disorder. Authors of previous stud- genitalia and promotes the formation of the prostate, pro-
ies have already concluded that the determination of both static urethra, penis, and scrotum. These developmental
the gonadal sex and the chromosomal sex are required procedures are mediated via binding of both hormones to
for establishment of a precise diagnosis and classification the cellular androgen receptor, which is located within the
(Hare, 1976; Chaffaux et al., 1980; Meyers-Wallen and target tissue.
Patterson, 1986; Melniczek et al., 1999). The absence of a Y chromosome and therefore a func-
In contrast to human medicine, a standardised and pre- tional SRY-gene results in the development of ovaries
cise nomenclature is missing in veterinary medicine. The and female genitalia. The genetic basis in ovary deter-
existing terminology is inconsistently used, independently mination is only incompletely understood. In contrast to
from the underlying disorder. male embryos, the paramesonephric duct system persists
The objectives of the present work were (1) to review in female embryos, and develops into the uterine tubes,
the literature on canine intersexuality, (2) to categorise uterus, and cranial portion of the vagina, whereas the
and reclassify the reported intersex cases according to a mesonephric ducts regress. The undifferentiated primitive
systematic scheme, (3) to propose a new nomenclature external genitalia being uninfluenced by androgens differ-
for various intersex conditions, which would enable an entiate into the caudal portion of the vagina, labia vulvae,
exact diagnosis and help to avoid the confusing terms “true vestibule, and clitoris.
hermaphrodite” and “pseudohermaphrodite”, and (4) to
illustrate the diagnostic approach and detailed clinical and 3. Canine intersexuality
pathomorphological findings using the case studies of two
dogs with bilateral ovotestes and ambiguous genitalia. To survey the variety of known disorders of sex devel-
opment (DSD) in dogs and establish precise diagnoses
2. Normal sex development in affected patients, categorising of the intersex condi-
tions is useful. DSD can occur at any stage of normal sex
An understanding of normal sex development is a nec- development, and lead to various genital tract abnormal-
essary basis from which to study the variety of intersex ities ranging from conditions with apparently normal to
conditions known in dogs. Generally, normal sex develop- ambiguous genitalia and concomitant sterility or fertility
ment happens in an orderly sequence of three stages, with (Meyers-Wallen and Patterson, 1989; Feldman and Nelson,
T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207 199

2004). Therefore, DSD can be categorised according to the 3.2. Disorders of gonadal sex development
chronological stages normal sex development is undergo-
ing resulting in three main categories: sex chromosome Affected dogs have either an XY or an XX chromosome
disorders, disorders of gonadal sex development, and dis- constitution, but chromosomal and gonadal sex disagree
orders of phenotypic sex development as described in the (Meyers-Wallen and Patterson, 1986). This abnormality
following chapters and demonstrated in Table 1. results in two kinds of sex-reversed conditions: either
XX sex reversal (XXSR) or XY sex reversal (XYSR). Both
3.1. Sex chromosome disorders conditions cause the development of ambiguous external
genitalia in varying degrees, which is positively correlated
Reported abnormalities in dogs include congenital with the amount of testicular tissue within the gonads
changes of sex chromosome number and XX/XY chi- (Meyers-Wallen and Patterson, 1989; Feldman and Nelson,
maerism/mosaicism resulting from random events. The 2004).
three kinds of X chromosomal aneuploidy are based on XXSR is subdivided into two categories currently
mitotic nondisjunction during meiosis and cause underde- termed as XX true hermaphroditism and XX male
veloped rather than ambiguous genitalia (Meyers-Wallen syndrome (Meyers-Wallen and Patterson, 1986, 1989;
and Patterson, 1986). Monosomy X is known as Turner’s Feldman and Nelson, 2004; Schlafer and Miller, 2007).
syndrome in women. Affected bitches are infertile and have XX true hermaphrodites are characterised by having both,
afollicular ovaries with partly hypoplastic female genital ovarian and testicular tissue occurring in one of three
organs in combination with normal developed (Löfstedt possible variations: bilateral, unilateral, and lateral, in
et al., 1992) or ambiguous external genitalia (Smith et which the former combination prevails (Selden et al.,
al., 1989). Trisomy X results in the development of infer- 1984; Meyers-Wallen and Patterson, 1986, 1989; Feldman
tile bitches with infantile genitalia (Johnston, 1989) or a and Nelson, 2004). In addition to an often uninfluenced
malformed uterus (Switonski et al., 2000). The XXY chro- paramesonephric duct system in affected animals, the
mosomal constitution is termed as Klinefelter’s syndrome mesonephric ducts may partly be developed. In contrast
in man. In dogs, the syndrome is associated with the devel- to XX true hermaphrodites, XX males have no ovarian
opment of sterile individuals, either a phenotypical male tissue but bilateral non-spermatogenic testes in an ovar-
with hypoplastic testes (Clough et al., 1970; Marshall et ian position, and derivates of both, the mesonephric and
al., 1982; Nie et al., 1998; Reimann-Berg et al., 2008) and paramesonephric duct system resulting in a simultane-
optionally a uterus (Marshall et al., 1982) or a phenotypical ous formation of both, female and male genital organs
female with afollicular ovaries, hypoplastic epididymides, in various degrees of differentiation (Meyers-Wallen and
and a malformed female genital tract (Bosu et al., 1978). Patterson, 1986). XX males are sterile, whereas XX true
In XX/XY chimaera or mosaics, more than one cell line hermaphrodites can reproduce depending on the grade of
exists. Chimaerism is caused by fusion of different zygotes masculinisation (Selden et al., 1978, 1984).
or cells originating from different zygotes, whereas XX/XY Testis formation in females with a normal XX chromo-
mosaicism results from non-disjunction in a single zygote some constitution seems to be contradictory. The aetiology
or cells deriving from a single zygote, respectively (Meyers- and pathogenesis of this phenomenon is only partially
Wallen and Patterson, 1986; Feldman and Nelson, 2004). understood, but some hypotheses are offered in litera-
Generally, the phenotype of chimaera and mosaics depends ture. In humans, 80% of XX males and 10% of XX true
on the proportion of testicular tissue within the gonads and hermaphrodites show a translocation of the Y-linked SRY-
its hormonal activity (Feldman and Nelson, 2004). Bitches gene (McElreavey et al., 1992; Sarafoglou and Ostrer,
with an XX/X0 karyotype are infertile, and have normal 2000). But all hitherto tested XXSR dogs were SRY-negative
female external genitalia (Mayenco-Aguirre et al., 1999; (Melniczek et al., 1999; Meyers-Wallen et al., 1999; Kuiper
Switonski et al., 2003). In the dog, XX/XY and XX/XXY and Distl, 2004; Switonski et al., 2004; Nowacka et al.,
chimaerism or mosaicism are associated with develop- 2005; De Lorenzi et al., 2008; Buijtels et al., 2009; present
ment of ambiguous genitalia and of either testes (Dain study) indicating an SRY-independent mechanism for testis
and Walker, 1979; Chaffaux et al., 1980; Allen et al., 1981; induction in dogs with an XX chromosome constitution
Meyers-Wallen and Patterson, 1986; Genero et al., 1998), (Meyers-Wallen et al., 1995, 1999; De Lorenzi et al., 2008).
ovaries (Weaver et al., 1979; Chaffaux et al., 1980) or uni- Further hypotheses in the dog include the presence of auto-
lateral ovotestis/bilateral ovotestes (Hare, 1976; Bosu et somal genes with Y-effect as proven for other species as the
al., 1978; Dain and Walker, 1979; Johnston, 1989). A fur- SXR gene in female carrier mice (Schlafer and Miller, 2007)
ther case with 78,XX/77,X0/79,XXY mosaicism, ovotestes or mutations influencing the transcription of PISRT1 and
and ambiguous genitalia was described by Pullen (1970). FOXL2 in XX goats (polled intersex syndrome, Pailhoux et
Canine XY/XXY chimaerism or mosaicism is associated al., 2001). In contrast, studies on a multigenerational pedi-
with bilateral cryptorchidism, absence of spermatogenesis, gree of SRY-negative XXSR dogs disclosed no mutations
and ambiguous genitalia (Goldschmidt et al., 2001). There is involving the region of PISRT1 and FOXL2 (Kothapalli et al.,
one report on a sex-reversed dog tentatively designated as 2005). Further candidate genes (WT1, DMRT1, GATA4, FOG2,
being a 78,XY/78,XYrcp (X;autosome) mosaic. The authors LHX1/LIM1, SF1, SOX9, LHX9, RSPO1) known to play a role in
speculated, whether this kind of translocation was respon- the sex determination pathway and therefore in the devel-
sible for the disordered sex development resulting in the opment of XXSR or XYSR in humans (Sarafoglou and Ostrer,
presence of ovotestes and an enlarged clitoris with bone 2000; Cotinot et al., 2002) or in animal models (Cotinot et
formation (Schelling et al., 2001). al., 2002) could also be excluded as being causative in the
200 T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207

Table 1
Various disorders of sex development (DSD) known in the dog; the cases were reclassified according to the reported gonadal constitution, genital tract
abnormalities and the karyogram, renamed adopting a new terminology based on gonosomal constellation and gonadal constitution, and categorised
appropriate to the stages of normal sex development.

Gonosomal Currently used Proposed Genital tract Affected dog breeds


constellation nomenclature nomenclature abnormalities

I. Sex chromosome disorders


(a) X aneuploidy
77,X0 Monosomy X, X0 –a O, PD, NG/AG, I Dobermann Pinscher, Miniature
syndrome American Eskimo (Smith et al.,
1989; Löfstedt et al., 1992)
79,XXX Trisomy X, triple-X –a O, PD, NG/AG, I Airedale Terrier, Crossbreed
syndrome (Johnston, 1989; Switonski et al.,
2000)
79,XXY Canine Klinefelter’s Ovarian XXY O, MD, PD, AG, I Great Dane (Bosu et al., 1978)
syndrome, XXY syndrome
syndrome, male PH
Testicular XXY T, (CO), MD, PD, German Shorthaired Pointer,
syndrome NG/AG, I Miniature Schnauzer, Norwich
Terrier, West Highland White
Terrier (Clough et al., 1970;
Marshall et al., 1982; Nie et al.,
1998; Reimann-Berg et al., 2008)

(b) Chimaerism and mosaicism


78,XX/77,X0 XX/X0 C or M 78,XX/77,X0 C O, PD, I Bearded Collie, Munsterlander, Toy
or M Poodle (Mayenco-Aguirre et al.,
1999; Switonski et al., 2003)b
78,XX/78,XY XX/XY C or M, TH, 78,XX/78,XY OT, OT + T, MD, American Eskimo, Pug, Schipperke
THC ovotesticular C (PD), AG (Hare, 1976d ; Bosu et al., 1978;
or M Johnston, 1989)b
XX/XY C or M with 78,XX/78,XY T, (CO), (MD), Belgium Shepherd, German
testes, male PH testicular C or (PD), (H), AG, Shepherd, Old English Sheepdog
M (I) (Chaffaux et al., 1980;
Meyers-Wallen and Patterson,
1986; Genero et al., 1998)b
XX/XY C or M, 78,XX/78,XY O, PD, AG, (I) Dachshund, Toy Spaniel (Weaver et
female PH ovarian C or M al., 1979; Chaffaux et al., 1980)b
XX/XY C or M –c ?; AG Border Terrier, Fila Brasileiro
(Meyers-Wallen et al., 1999;
Kuiper and Distl, 2004)b
78,XX/79,XXY XX/XXY C or M 78,XX/79,XXY OT, PD, AG Irish Red Setter (Dain and Walker,
ovotesticular C 1979)b
or M
XX/XXY C or M, 78,XX/79,XXY T, CO, MD, PD, Labrador-Setter-crossbreed,
male PH testicular C or AG, (I) Cocker Spaniel (Dain and Walker,
M 1979; Allen et al., 1981)b
78,XX/77,X0/79,XXY XX/X0/XXY M, TH 78,XX/77,X0/79, OT, MD, PD, AG Beagle (Pullen, 1970)
XXY
ovotesticular M
78,XY/79,XXY XY/XXY C or M 78,XY/79,XXY T, CO, I Poodle (Goldschmidt et al., 2001)b
testicular C or
M
78,XY/78,XYrcp XY/XYrcp M, TH 78,XY/78,XYrcp OT, MD, PD, AG Yorkshire Terrier (Schelling et al.,
(X;autosome) ovotesticular M 2001)

II. Disorders of gonadal sex development


(a) XX sex reversal
78,XX TH, XX TH 78,XX OT, OT + O, American Cocker Spaniel, Basset,
ovotesticular OT + T, O + T, Beagle, Brussels Griffon, Collie,
DSD (MD), PD, AG, Crossbreed, Doberman Pinscher,
(I) German Pinscher, German
Shorthaired Pointer, Norwegian
Elkhound, Poodle, Pug, Soft Coated
Wheaten Terrier, Kerry Blue
Terriere Vizslae , Weimaraner
(Hare, 1976d ; Selden et al., 1978;
Tangner et al., 1982; Thomas et al.,
1986; Randolph et al., 1987;
Fitzgerald and Murphy, 1990;
Melniczek et al., 1999;
Meyers-Wallen et al., 1999; De
Lorenzi et al., 2008; present study)
T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207 201

Table 1 (Continued )

Gonosomal Currently used Proposed Genital tract Affected dog breeds


constellation nomenclature nomenclature abnormalities

XX male, male PH 78,XX T, CO, MD, PD, American Cocker Spaniel,


testicular DSD (H), AG, I American Pit Bull Terriere
American Staffordshire Terrier,
Basset Artésien Normand, Beagle,
Border Colliee , Crossbreed, English
Cocker Spaniele , French Bull Dog,
German Shorthaired Pointer, Jack
Russel Terrier, Kerry Blue Terrier,
Norwegian Elkhound, Picard,
Podenco, PON, Pug, Soft Coated
Wheaten Terriere Walker hound,
Weimaranere (Stewart et al., 1972;
Selden et al., 1978; Williamson,
1979; Chaffaux et al., 1990d ;
Williams et al., 1997; Tammer et
al., 1998; Melniczek et al., 1999;
Meyers-Wallen et al., 1999; Kuiper
and Distl, 2004; Nowacka et al.,
2005; De Lorenzi et al., 2008;
Buijtels et al., 2009)
(b) XY sex reversal
78,XY TH 78,XY O + T, MD, PD, Fox Terrier, Yorkshire Terrier
ovotesticular (H), AG (Chaffaux et al., 1986; Jurka et al.,
DSD 2009)

III. Disorders of phenotypic sex development


(a) Male with PMDS
78,YX Male PH, male PH 78,XY T, (CO), MD, PD, American Cocker Spaniel, Basset,
with PMDS DSD/PMDS (H), AG, (I) Bull Terrier, Collie, Crossbreed,
Dachshund, English Cocker Spaniel,
German Shepherd, Labrador
Retriever, Miniature Schnauzer,
Pekinese, Poodle, Standard
Schnauzer, West Highland White
Terrier, Yorkshire Terrier (Hare,
1976d ; Kelly et al., 1976; Chaffaux
et al., 1980d ; Marshall et al., 1982;
Svendsen et al., 1985; Chaffaux et
al., 1990d ; Nickel et al., 1992;
Volpe et al., 2000; Kuiper and Distl,
2004; Alam et al., 2007;
Nowacka-Woszuk et al., 2007;
Whyte et al., 2009)

(b) Male with failure of ADM/incomplete TFS


78,XY Male PH 78,XY DSD T, (CO), MD, Labrador Retriever, Crossbreed
AG, (I) (Hare, 1976d ; Peter et al., 1993;
Wernham and Jerram, 2006)

(c) Virilised female due to exogenous steroid exposure


78,XX Female PH 78,XX DSD O, PD, (H), AG Australian Shepherd crossbreed,
Beagle, Crossbreed, Greyhound
(Hare, 1976d ; Medleau et al., 1983;
Olson et al., 1989)

(d) Virilised female of unknown cause


78,XX Female PH 78,XX DSD O, PD, AG Briard, Cairn Terrier, Cocker
Spaniel, Collie crossbreed, German
Shepherd, Yorkshire Terrier (Hare,
1976d ; Chaffaux et al., 1980d ; Allen
et al., 1981; Van Schouwenburg
and Louw, 1982; Holt et al., 1983)

ADM = androgen-dependent masculinisation; AG = ambiguous genitalia; C = chimaera; CO = cryptorchidism; H = hypospadias; I = infertility;


M = mosaic; MD = mesonephric duct derivates; NG = normal genitalia; O = ovary/ovaries; OT = ovotestis/-es; PD = paramesonephric duct derivates;
PH = pseudohermaphrodite; PMDS = persistent Müllerian duct syndrome; PON = Polski Owczarek Nizinny; SR = sex reversal; T = testis/testes; TH = true
hermaphrodite; THC = true hermaphrodite chimaera; TFS = testicular feminisation syndrome.
a
Renaming is redundant.
b
No differentiation between chimaera or mosaic.
c
Renaming failed due to incomplete information.
d
Review article.
e
SRY-negative, karyotyping not done.
202 T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207

canine model (Kothapalli et al., 2005; Nowacka et al., 2005; show uni- or bilateral cryptorchidism, and can be fertile
Pujar et al., 2005; De Lorenzi et al., 2008). In the meantime, a in a case of scrotal testis/testes (Chaffaux et al., 1980).
new study indicates that another gene locus, which is likely They usually have ambiguous external genitalia besides
located in the linked region of CFA29, may be causative for paramesonephric duct derivates and tend to get urinary
sex reversal in dogs (Pujar et al., 2007). It should be consid- tract infections, prostatitis, cystic endometrial hyperpla-
ered that more than one mechanism could be involved in sia, pyometra, and testicular tumours (Hare, 1976; Kelly
the development of SRY-negative sex reversal syndrome in et al., 1976; Chaffaux et al., 1980; Marshall et al., 1982;
mammals (Meyers-Wallen et al., 1999; Pujar et al., 2007). Svendsen et al., 1985; Meyers-Wallen and Patterson, 1986,
Studies on human pedigrees with familiar occurrence of 1989; Nickel et al., 1992; Simpson et al., 1998; Volpe et al.,
both XX male syndrome and XX true hermaphroditism in 2000).
siblings led to suggest a genetic aetiology (Sarafoglou and In individuals suffering from defects in androgen-
Ostrer, 2000; Cotinot et al., 2002). In the dog, an autoso- dependent masculinisation, the paramesonephric ducts
mal recessive trait has been proven in the American Cocker regress normally, but masculinisation of the androgen-
Spaniel resulting in both, XX true hermaphrodites and XX responsive tissue happened insufficiently or failed result-
males (Selden et al., 1984), and is presumed to be likely in ing amongst others in ambiguous genitalia and hypospa-
several other dog breeds including the German Shorthaired dias (Meyers-Wallen and Patterson, 1986, 1989; Simpson
Pointer, Beagle, English Cocker Spaniel, Kerry Blue Terrier, et al., 1998; Wernham and Jerram, 2006; Sonne et al., 2008).
Norwegian Elkhound, Pug, Weimaraner, and German Shep- Hypospadias is described in several dog breeds including
herd (Stewart et al., 1972; Williamson, 1979; Randolph the Boston Terrier, in which the condition is suspected to
et al., 1987; Meyers-Wallen and Patterson, 1989; Meyers- be inherited. It can occur independently or in combination
Wallen et al., 1995, 1999; Melniczek et al., 1999; Switonski with other genital tract abnormalities (Selden et al., 1978;
et al., 2004). Chaffaux et al., 1980; Hayes and Wilson, 1986; Jurka et al.,
XYSR results in the presence of ovarian tissue in addi- 2009). The X-linked inherited testicular feminisation syn-
tion to testicular tissue, and its occurrence in dogs is rarely drome (TFS) occurring in humans and rodents is based on
reported. Chaffaux et al. (1986) and Jurka et al. (2009) a defect of the androgen receptor, and is also assumed to
described each a case of true hermaphroditism showing an exist in dogs (Meyers-Wallen and Patterson, 1986, 1989),
XY chromosomal constitution, one testis with an attached but has only been reported once as an incomplete form
epididymis, a contralateral ovary, a uterus and ambiguous in a mixed-breed dog (Peter et al., 1993). Deficiency of
external genitalia and occasional hypospadias. A further 5-alpha-reductase that converts testosterone into dihy-
dog with an XY karyotype, ovotestes, epididymides, a drotestosterone is autosomal recessive inherited in men
uterus and ambiguous external genitalia was reported, but but has not been detected in dogs (Meyers-Wallen and
due to the presence of a translocation involving the X Patterson, 1986).
chromosome, the animal was finally classified as a mosaic Female pseudohermaphrodites have an XX chromo-
(Schelling et al., 2001; Table 1). some constitution and bilateral ovaries but an androgen-
dependent masculinisation of the genitalia ranging from
3.3. Disorders of phenotypical sex development mild clitoral enlargement to nearly male external gen-
italia. Beyond a cranial vagina and uterus present in
In affected dogs, chromosomal and gonadal sex match, all cases, a prostate gland could possibly be developed
but the external genitalia have characteristics of the (Meyers-Wallen and Patterson, 1986). Most cases result
opposite gender (Meyers-Wallen and Patterson, 1986). from administration of steroids to pregnant bitches dur-
They are currently classified either as female or male ing critical stages of development inducing virilisation of
pseudohermaphrodites depending on their gonadal sex female foetuses (Hare, 1976; Chaffaux et al., 1980; Medleau
(Meyers-Wallen and Patterson, 1986; Simpson et al., 1998; et al., 1983; Meyers-Wallen and Patterson, 1989; Olson
Schlafer and Miller, 2007). et al., 1989; Melniczek et al., 1999; Feldman and Nelson,
Male pseudohermaphroditism occurs more frequently 2004; Schlafer and Miller, 2007). Other underlying causes
than female pseudohermaphroditism (Hare, 1976; such as the adrenogenital syndrome in women and sev-
Chaffaux et al., 1980; Simpson et al., 1998; Table 1) and eral animal species has not yet been identified in the dog
is based either on the failure of paramesonephric duct (Meyers-Wallen and Patterson, 1989; Feldman and Nelson,
regression (persistent Müllerian duct syndrome, PMDS) 2004).
or on the failure of androgen-dependent masculinisation
due to enzyme deficiencies in testosterone biosynthesis 4. Reclassification of canine intersex cases
or insensitivity of their receptors in the target tissue
(androgen insensitivity syndrome, AIS) (Meyers-Wallen Reported intersex cases in dogs were reclassified to give
and Patterson, 1986; Feldman and Nelson, 2004; Schlafer an overview of the diversity of canine DSD, the frequency
and Miller, 2007). of various conditions, and the dog breeds involved. In the
PMDS is the predominant form in the dog (Table 1), present work, reported canine intersex cases were reclas-
and is autosomal recessive inherited in the Miniature sified based on the described genital tract abnormalities
Schnauzer (Meyers-Wallen and Patterson, 1989; Wu et al., and cytogenetic findings and categorised according to the
2009). In contrast to XX males, dogs suffering from PMDS chronological stages of normal sex development (Table 1).
have a normal XY chromosome constitution (Meyers- As far as possible, only the earliest report on an affected
Wallen and Patterson, 1986). Affected animals commonly dog breed was considered for each kind of DSD. Those cases
T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207 203

Table 2
Unclassified cases of canine DSD.

True hermaphrodite
(a) Bilateral Afghan Hound, American Staffordshire Terrier, Berger Picard, Boxer,
German Shepherd crossbreed, (Chaffaux et al., 1980a ; Meyers-Wallen et
al., 1999; De Rooster et al., 2006; Redden, 2007; present study)
(b) Lateral Miniature Schnauzer (Stünzi and Stünzi, 1950)

Male pseudohermaphrodite
(a) Persistent Müllerian ducts Afghan Hound, Australian Terrier, Golden Retriever, Pug, Springer Spaniel,
Toy Terrier, Terrierb (Hare, 1976a ; Axelson, 1978; Chaffaux et al., 1980a ;
Röcken, 1988)
(b) Failure of ADM Greenland dog, Pit Bull Terrier, Pug (Stewart et al., 1972; Fischer, 1983;
Sonne et al., 2008)
(c) Unknown Siberian Husky (Nowacka-Woszuk and Switonski, 2009)

Female pseudohermaphrodite
(a) Exogenous steroid exposure Boxer (Hare, 1976a )
(b) Unknown Pekinese-Poodle-crossbreed, Saluki (Hinsch, 1979; Chaffaux et al., 1980a )

Incomplete information impedes reclassification and adoption of a new nomenclature. ADM = androgen-dependent masculinisation.
a
Review article.
b
Not otherwise specified.

that could not be reclassified due to incomplete informa- present review revealed that at least 27 dog breeds are
tion are summarised in Table 2. The case selection was involved in XXSR exceeding the group of male pseudo-
limited to those dog breeds that were not affected from hermaphrodites.
an equivalent intersex DSD already listed in Table 1. There In contrast to the frequent occurrence of sex reversal in
are further reports on dog breeds such as the Miniature purebred dogs, intersexual conditions are less frequently
Pinscher and Shetland Sheepdog with ambiguous geni- reported in crossbreed dogs (Hare, 1976; Chaffaux et al.,
talia (Schneck, 1975; Nowacka et al., 2005), but these are 1980; Tables 1 and 2). Purebred dogs originate from a
excluded from both, Tables 1 and 2, due to insufficient small gene pool, therefore, a recessive mode of inher-
information. itance results in a frequent expression (Meyers-Wallen
This literature review has revealed that the classifica- et al., 1999). Furthermore, reclassification disclosed that
tion and terminology are inconsistently used, leading to numerous dog breeds including amongst others the Cocker
confusion, and masking the occurrence and frequency of Spaniel, Beagle, German Shepherd, German Shorthaired
various intersex conditions in dogs. One possible expla- Pointer, Poodle, Pug, and Yorkshire Terrier are affected
nation could be the fact that aberrations originating from with various kinds of DSD originating either from differ-
different stages of sex development and therefore repre- ent stages or from the same stage of sex development
senting separate conditions of DSD, can result in similar (Table 1). It should be considered that favourite dog breeds
alterations on internal and/or external genitalia. Ovotestis are predominantly involved and some pedigrees, e.g. of the
formation, for example, can be caused by both, sex chro- American Cocker Spaniel are used for research in inher-
mosome disorders (chimaerism/mosaicism) and disorders ited intersex conditions (Selden et al., 1978, 1984), which
at the stage of gonadal sex development (sex reversal). increases the chance to detect rare DSD.
Another example is PMDS representing a disorder of phe-
notypical sex development that can easily be confused 5. New nomenclature
due to anatomical similarities with the XX male syndrome
(disorder of gonadal sex development) or some kinds Beside the consequent use of a systematic classifica-
of chimaerism/mosaicism (sex chromosome disorder) tion scheme in dogs with DSD, the introduction of a new
(Table 1). Reclassification disclosed that most canine chi- nomenclature in veterinary medicine as was recently rec-
maera or mosaics were previously termed as female/male ommended for humans (Hughes, 2008) would facilitate a
pseudohermaphrodites or true hermaphrodites, respec- definite and individual diagnosis. The new terminology is
tively (Pullen, 1970; Hare, 1976; Bosu et al., 1978; Weaver based on the gonosomal constellation and gonadal consti-
et al., 1979; Chaffaux et al., 1980; Allen et al., 1981; tution and replaces the common but confusing and inexact
Johnston, 1989; Genero et al., 1998). Additionally, many main categories “true hermaphroditism” and “pseudo-
of the intersex cases previously diagnosed as male pseudo- hermaphroditism”, which are used independently from the
hermaphrodites (Stewart et al., 1972; Hare, 1976; Chaffaux gonosomal constellation (Table 1).
et al., 1980; Allen et al., 1981) could be reclassified as XX In Table 1, the new terminology was proposed for nearly
males. This finding was consistent with investigations of all known varieties of canine DSD, apart from the cur-
Meyers-Wallen and Patterson (1986). Furthermore, a dog rently used terms “monosomy X” and “trisomy X”, which
suffering from XXY syndrome was also originally diagnosed are regarded as being unmistakable diagnoses. In some
as male pseudohermaphrodite (Marshall et al., 1982). conditions, the previous nomenclature was amended by
Male pseudohermaphroditism was previously consid- considering the constitution of the gonadal tissue result-
ered as being the main intersex condition in dogs (Hare, ing, e.g. in “ovarian” or “testicular XXY syndrome” instead
1976; Chaffaux et al., 1980; Simpson et al., 1998), but the of “canine Klinefelter’s syndrome” or in “78,XX/78,XY tes-
204 T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207

ticular C or M” instead of “78,XX/78,XY C or M”. In other shamrock-formed glans clitoridis with 6.0 mm in diame-
conditions, the currently used terms would be replaced, ter, and contained a large central os clitoridis surrounded
e.g. “true hermaphroditism” by “78,XX/78,XY ovotesticular by the corpus cavernosum. Subsequent to the macroscopic
C or M”, “78,XX ovotesticular DSD”, and “78,XY ovotesticu- examination, removed organs were fixed in 7% buffered
lar DSD”, respectively, or “male pseudohermaphrodite” by formaldehyde. Representative specimens were embedded
“78,XY DSD”. The latter can be completed by adding the in paraffin (Paraplast® ), cut into 4–6 ␮m-thick sections and
underlying syndrome as, for example, “78,XY DSD/PMDS”. stained with haematoxylin & eosin (H&E) according to stan-
dard protocols.
6. Diagnostic approach in intersex patients—report Histologically, the gonadal parenchyma was distinctly
on two phenotypically female dogs with ovotestes subdivided in a small peripheral zone of ovarian tis-
sue and a medullar part of non-spermatogenic testicular
Most dogs with DSD are admitted to veterinary prac- tissue by a small band of connective tissue containing
titioners due to the presence of ambiguous external lymphatic and blood vessels (Fig. 1). The ovarian part
genitalia, infertility or absence of oestrus signs. If external showed numerous small follicles that were mostly at stages
genitalia appear normal or show only mild abnormali- of degeneration. They could be identified as being pri-
ties, diagnosis of an intersex condition in dogs can be mordial, primary, secondary, and tertiary follicles (latter
problematical. A precise and extensive clinical, patho- only found in dog 2) consisting of highly vacuolated fol-
morphological, and cytogenetic examination determining licular cells and frequently multiple oocytes (polyovular
phenotypic sex, gonadal constitution, and gonosomal con- follicles, Fig. 2). Several granulosa cell cords and follicu-
stellation is essential for a definitive diagnosis as also lar cysts (latter only found in dog 2) were also present.
recommended by authors of previous studies (Hare, 1976; The medullar testicular tissue was composed of numerous
Chaffaux et al., 1980; Meyers-Wallen and Patterson, 1986; normal appearing interstitial cells and hypoplastic seminif-
Melniczek et al., 1999). The chapters above provide erous tubules. The tubules were lined with a single layer of
a structured classification scheme, and propose a new highly vacuolated sustentacular cells, but spermatogenic
nomenclature aimed at facilitating individual and exact cells were absent (Fig. 3). Hypoplastic epididymal tissue
diagnoses when confronted with intersex patients. Addi- showing the convoluted tubule lined with a flattened cili-
tional pedigree analysis should be carried out to disclose a ated epithelium (Fig. 3) as well as a physiologically formed
hereditary disorder (Meyers-Wallen and Patterson, 1986), uterine tube lined with a ciliated columnar epithelium
and to exclude affected parents and siblings from breeding. were attached to each gonad. The endometrium appeared
Below, this diagnostic approach is used for two intersex inactive resembling the stage seen in juvenile bitches. Myo-
dogs presented at the Faculty of Veterinary Medicine of the and perimetrium revealed no pathological findings.
Ludwig-Maximilians-University. For chromosomal analysis, whole blood cultures of
A five month old German Pinscher (dog 1) and an eight dog 1 (1 ml blood/10 ml culture) were set up in RPMI
month old Berger Picard (dog 2) were presented for clini- 1640 medium (PAA Laboratories, Pasching, Austria) sup-
cal examination due to concerns about an enlarged clitoris. plemented with pokeweed mitogen (0.8 ␮g/ml f.c.),
Both dogs were phenotypical females, had no signs of Penicillin/Streptomycin (100 U/ml f.c./0.1 mg/ml f.c.), and
oestrus before presentation and showed normal female- fetal calf serum (15%). Metaphases were arrested using
like urination. Physical examination of external genitalia colcemid (0.1 ␮g/ml f.c.) for two hours and spreads were
revealed a normal sized vulva in physiological position and prepared according to Barch (1991) after 72 h culture time
confirmed clitoromegaly in both cases. The enlarged clitoris at 37 ◦ C (all additives from Biochrom, Berlin, Germany).
protruded from the rima vulvae, had a palpable os clitoridis, Giemsa (5% in Sörensen buffer pH 6.8) stained metaphases
and was about 2.5 cm in length (dog 1) and about 4.0 cm were microphotographed at 1000× microscopic magnifica-
in length (dog 2), respectively. Endoscopy of the vagina tion. Among 35 metaphases scored, 33 displayed a diploid
revealed no atypical findings, and the urethral tuber- chromosome number of 2n = 78. Two metaphases harbour-
cles were positioned ventrally near the vaginovestibular ing 75 and 77 chromosomes were classified as artefacts
junction. Both dogs underwent laparotomy under general due to experimental impacts. All metaphases screened pre-
anaesthesia. They had a bicornuated uterus in a physiolog- sented an XX gonosomal constellation.
ical anatomical position and gonads enclosed in the bursa For detection of the sex-determining region of the Y
ovarica at each horn tip. Routine gonadohysterectomy was chromosome (SRY-gene), genomic DNA was extracted from
performed in both bitches, and the clitoris was additionally peripheral blood lymphocytes of dog 1 using the nexttecTM
excised in dog 2. Genomic DNA Isolation Kit (Bio&Sell e.K., Nuremberg, Ger-
The genital tract of both dogs revealed similar path- many). SRY-specific PCR was performed using primers
omorphological findings. The gonads measured approx- published by Meyers-Wallen et al. (1995). As an ampli-
imately 1.5 cm × 1.0 cm × 0.5 cm in dog 1 and approx- fication control, a microsatellite (cph 17) (Fredholm and
imately 2.0 cm × 1.5 cm × 0.7 cm in dog 2. Every gonad Winterø, 1995) was simultaneously amplified using a
had a testis-like appearance with an attached structure multiplex master mix (Qiagen® , Hilden, Germany). Total
resembling epididymides. The physiologically developed volume was 25 ␮l. Cycling conditions were 95 ◦ C, 15 min
uterus had a maximum length of 9.0 cm in dog 1 and (1×); 94 ◦ C, 1 min, 55 ◦ C, 1.5 min, 72 ◦ C, 1 min (35×); 72 ◦ C,
12.0 cm in dog 2, and was composed of two uterine 5 min (1×). Amplicons were separated in an ethidium
horns, corpus and cervix. The clitoris removed from dog bromide containing agarose gel (2%) in TBE buffer at
2 measured 4.0 cm × 1.0 cm × 1.0 cm. It had a knobbly and 4 V/cm. PCR analysis detected no SRY-specific product
T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207 205

within the limit of detection (detection limit was about


one male genome in 10,000 female equivalents, data not
shown).
The clinical and pathomorphological findings includ-
ing bilateral ovotestes, ambiguous external genitalia, and a
markedly enlarged clitoris with bone formation are similar
in both dogs and initially consistent with those described
for true hermaphrodites in literature (Meyers-Wallen
and Patterson, 1986, 1989; Feldman and Nelson, 2004;
Schlafer and Miller, 2007). The clinical history of both cases
matches the literature reports describing clitoromegaly
with bone formation as the most obvious indication for
a disordered sex development in a bitch (Meyers-Wallen
and Patterson, 1986; Meyers-Wallen et al., 1995, 1999;
Simpson et al., 1998). Masculinisation of external geni-
Fig. 1. Dog 2, DSD, ovotestis, histopathology: parenchyma is distinctly
talia in dogs usually occur at about 4 months of age as the
subdivided in a small peripheral zone of ovarian tissue (1) and a larger
medullar zone of testicular tissue (2); both zones are separated by a small testicular parts of the gonads become endocrinologically
band of connective tissue (3); bar = 1 mm; haematoxylin & eosin (H&E). functional, and the degree is likely related to the amount of
testosterone during target organ sensitivity. The extent of
androgen-dependent masculinisation in both bitches can
be explained by the large amount of testicular tissue within
the gonads as described for previous cases (Meyers-Wallen
and Patterson, 1989; Simpson et al., 1998; Feldman and
Nelson, 2004). The findings in the presented dogs agree
with reports describing the presence of bilateral ovotestes
as the most frequent combination (Hare, 1976; Chaffaux et
al., 1980; Selden et al., 1984; Meyers-Wallen and Patterson,
1986, 1989; Feldman and Nelson, 2004) and the centrally
positioned testicular tissue as the most common location
within gonads in true hermaphrodites (Meyers-Wallen and
Patterson, 1989). Furthermore, the presence of an uninflu-
enced developed paramesonephric duct system regardless
of coexisting testicular tissue within the gonads is a known
Fig. 2. Dog 1, 78,XX ovotesticular DSD, ovotestis, histopathology: phenomenon in dogs (Selden et al., 1984; Meyers-Wallen
the ovarian part contains cellular stroma, numerous atretic folli- et al., 1987). The coexistence of epididymic tissue in both
cles (1 = primary, 2 = polyovular follicle) and granulosa cell cords (3); dogs results from stimulation of the mesonephric duct sys-
bar = 200 ␮m; H&E.

Fig. 3. Dog 1, 78,XX ovotesticular DSD, ovotestis, histopathology: the testicular part is composed of hypoplastic non-spermatogenic seminiferous tubules,
which are lined with vacuolated sustentacular cells (1) and interstitial cells (2) that are scattered throughout the stroma (left); bar = 100 ␮m; attached
hypoplastic epididymal tissue without spermatozoa (right); bar = 200 ␮m; H&E.
206 T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207

tem, and was previously reported for true hermaphrodites Chaffaux, S., Cribiu, E.P., Crespeau, F., 1986. Un cas rare d’hermaphrodisme
(Hare, 1976; Bosu et al., 1978; Allen et al., 1981; Selden vrai latéral chez une chienne 78,XY. Rec. Méd. Vét. 162, 463–
470.
et al., 1984; Meyers-Wallen et al., 1987; Randolph et al., Chaffaux, S., Nudelmann, N., Durand, V., Cribiu, E.P., 1990. Le pseudo-
1987; De Rooster et al., 2006). The owner’s anamnesis of hermaphrodisme mâle, a propos du cas d’un chien intersexué 78,XX.
absent oestrus cycles in both bitches is attributed to the Rec. Méd. Vét. 166, 125–132.
Clough, E., Pyle, R.L., Hare, W.C.D., Kelly, D.F., Patterson, D.F., 1970. An XXY
prepubertary stage of the ovarian tissue. sex-chromosome constitution in a dog with testicular hypoplasia and
Genetic analysis performed in dog 1 demonstrated a congenital heart disease. Cytogenetics 9, 71–77.
normal female chromosome constitution of 78,XX and Cotinot, C., Pailhoux, E., Jaubert, F., Fellous, M., 2002. Molecular genetics
of sex determination. Sem. Reprod. Med. 20, 157–168.
the absence of SRY. Regarding the classification scheme Dain, A.R., Walker, R.G., 1979. Two intersex dogs with mosaicism. J.
described earlier and summarised in Table 1, dog 1 was Reprod. Fert. 56, 239–242.
therefore classified as XXSR animal representing a disorder De Lorenzi, L., Gropetti, D., Arrighi, S., Pujar, S., Nicoloso, L., Molteni, L.,
Pecile, A., Cremonesi, F., Parma, P., Meyers-Wallen, V., 2008. Muta-
at the stage of gonadal sex development. Adopting the new
tions in the RSPO1 coding region are not the main cause of canine
nomenclature for intersex individuals (Table 1), the defi- SRY-negative XX sex reversal in several breeds. Sex. Dev. 2, 84–
nite diagnosis of 78,XX ovotesticular DSD was established 95.
for dog 1. De Rooster, H., Vercauteren, G., Görtz, K., Saunders, J., Polis, I., Rijsselaere,
T., 2006. True hermaphroditism in six female littermates after admin-
Unfortunately, classification and establishment of an istration of synthetic androgens to a pregnant bitch. Reprod. Dom.
exact diagnosis failed in dog 2 because the owner objected Anim. 41, 22–26.
to further analysis. Including the present study, the devel- Feldman, E.C., Nelson, R.W., 2004. Infertility, associated breeding disor-
ders, and disorders of sexual development. In: Feldman, E.C., Nelson,
opment of ovotestes was reported for at least 26 dog breeds R.W. (Eds.), Canine and Feline Endocrinology and Reproduction, third
(Tables 1 and 2). Most of these cases were categorised as ed. Saunders, pp. 893–898.
XX sex-reversed animals and could be redefined as indi- Fischer, K., 1983. Pseudohermaphroditism in a dog. Vet. Med. Small. Anim.
Clin. 78, 683–685.
viduals with 78,XX ovotesticular DSD (Table 1). Although Fitzgerald, A.L., Murphy, D.A., 1990. Bilateral ovotestes in an intersex,
XXSR is considered as being most likely for dog 2, the mixed-breed dog. Lab. Anim. Sci. 40, 647–650.
less frequently occurring chimaerism/mosaicism as well as Fredholm, M., Winterø, A.K., 1995. Variation of short tandem repeats
within and between species belonging to the Canidae family. Mamm.
the rare XYSR can only be excluded by cytogenetic analy- Genome 6, 11–18.
sis. Genero, E.R., Moreno-Millán, M., Ocaña-Quero, J.M., 1998. XX/XY chromo-
In both presented cases, no information was available some chimaerism in an intersex dog. Vet. Rec. 142, 340.
Goldschmidt, B., El-Jaick, K.B., Souza, L.M., Carvalho, E.C.Q., Moura,
about the occurrence of sexual abnormalities in parents or
V.L.S., Benevides Filho, I.M., 2001. Cryptorchidism associated with
siblings and exogenous influences like androgen adminis- 78,XY/79,XXY mosaicism in dog. Israel J. Vet. Med. 56, 56–58.
tration to canine foetuses as described by De Rooster et al. Hare, W.C.D., 1976. Intersexuality in the dog. Can. Vet. J. 17, 7–15.
(2006). Further studies in pedigrees of the German Pinscher Hayes, H.M., Wilson, G.P., 1986. Hospital incidence of hypospadias in dogs
in North America. Vet. Rec. 118, 605–607.
and Berger Picard breed are necessary to disclose a hered- Hinsch, G.W., 1979. Intersexes in the dog. Teratology 20, 463–468.
itary genetic defect as proven or assumed for other dog Holt, P.E., Long, S.E., Gibbs, C., 1983. Disorders of urination associated with
breeds (Stewart et al., 1972; Williamson, 1979; Selden et al., canine intersexuality. J. Small. Anim. Pract. 24, 475–487.
Hughes, I.A., 2008. Disorders of sex development: a new definition
1984; Randolph et al., 1987; Meyers-Wallen and Patterson, and classification. Pract. Res. Clin. Endocrinol. Metab. 22, 119–
1989; Meyers-Wallen et al., 1995, 1999; Melniczek et al., 134.
1999; Switonski et al., 2004), and to contribute to a bet- Johnston, S.D., 1989. Premature gonadal failure in female dogs and cats. J.
Reprod. Fertil. Suppl. 39, 65–72.
ter understanding of the mode of inheritance of intersex Jurka, P., Galanty, M., Zielinska, P., Max, A., Sysa, P., 2009. Hypospadias in
conditions in the dog. six dogs. Vet. Rec. 164, 331–333.
Kelly, D.F., Long, S.E., Strohmenger, G.D., 1976. Testicular neoplasia in an
intersex dog. J. Small. Anim. Pract. 17, 247–253.
Acknowledgement Kothapalli, K., Kirkness, E., Pujar, S., Van Wormer, R., Meyers-Wallen, V.N.,
2005. Exclusion of candidate genes for canine SRY-negative XX sex
The authors thank Mrs. Irene Zimmer, DVM, for her pro- reversal. J. Hered. 96, 759–763.
Kuiper, H., Distl, O., 2004. Intersexuality in dogs: causes and genetics.
fessional language assistance. Dtsch. Tierärztl. Wschr. 111, 251–258.
Löfstedt, R.M., Buoen, L.C., Weber, A.F., Johnston, S.D., Huntington,
A., Concannon, P.W., 1992. Prolonged proestrus in a bitch with
References
X chromosomal monosomy (77,X0). J. Am. Vet. Med. Assoc. 200,
1104–1106.
Alam, M.R., Cho, Y.G., Cho, S.J., Lee, J.I., Lee, H.B., Tae, H.J., Kim, I.S., Kim, Marshall, L.S., Oehlert, M.L., Haskins, M.E., Selden, J.R., Patterson, D.F., 1982.
N.S., 2007. Male pseudohermaphroditism in dogs: three case reports. Persistent Müllerian duct syndrome in miniature schnauzers. J. Am.
Vet. Med. (Praha) 52, 74–78. Vet. Med. Assoc. 181, 798–801.
Allen, W.E., Daker, M.G., Hancock, J.L., 1981. Three intersexual dogs. Vet. Mayenco-Aguirre, A.M., Padilla, J.A., Flores, J.M., Daza, M.A., 1999. Canine
Rec. 109, 468–471. gonadal dysgenesis syndrome: a case of mosaicism (77,X0-78,XX).
Axelson, R.D., 1978. Pseudohermaphroditism in a dog. J. Am. Vet. Med. Vet. Rec. 145, 582–584.
Assoc. 172, 584–585. McElreavey, K., Rappaport, R., Vilain, E., Abbas, N., Richaud, F., Lortat-Jacob,
Barch, M.J., 1991. The ACT Cytogenetics Laboratory Manual 2. Raven Press, S., Berger, R., Le Coniat, M., Boucekkine, C., Kucheria, K., Temtamy, S.,
New York. Nihoul-Fekete, C., Brauner, R., Fellous, M., 1992. A minority of 46,XX
Bosu, W.T.K., Chick, B.F., Basrur, P.K., 1978. Clinical, pathologic and cyto- true hermaphrodites are positive for the Y-DNA sequence including
genetic observations on two intersex dogs. Cornell Vet. 68, 375–390. SRY. Hum. Genet. 90, 212–215.
Buijtels, J.J.C.W.M., de Gier, J., van Haeften, T., Kooistra, H.S., Spee, B., Medleau, L., Johnson, C.A., Perry, R.L., Dulisch, M.L., 1983. Female pseu-
Veldhuis Kroeze, E.J.B., Zijlstra, C., Okkens, A.C., 2009. Minimal exter- dohermaphroditism associated with mibolerone administration in a
nal masculinization in a SRY-negative XX male Podenco dog. Reprod. dog. J. Am. Anim. Hosp. Assoc. 19, 213–215.
Dom. Anim. 44, 751–756. Melniczek, J.R., Dambach, D., Prociuk, U., Jezyk, P.F., Henthorn, P.S., Patter-
Chaffaux, S., Mailhac, J.M., Cribiu, E.P., Popescu, C.P., Cotard, J.P., 1980. son, D.F., Giger, U., 1999. Sry-negative XX sex reversal in a family of
L’intersexualité chez le chien (canis familiaris). A propos de quatre Norwegian Elkhounds. J. Vet. Intern. Med. 13, 564–569.
cas. Rec. Méd. Vét. 156, 179–192.
T. Poth et al. / Animal Reproduction Science 121 (2010) 197–207 207

Meyers-Wallen, V.N., Patterson, D.F., 1986. Disorders of sexual devel- Schlafer, D.H., Miller, R.B., 2007. Pathology of the genital system of the non-
opment in the dog. In: Morrow, D.A. (Ed.), Current Therapy in gravid female. In: Grant Maxie, M. (Ed.), Jubb, Kennedy, and Palmer’s
Theriogenology: Diagnosis, Treatment and Prevention of Reproduc- Pathology of Domestic Animals, vol. 3, fifth ed. Elsevier Saunders, pp.
tive Diseases in Animals, second ed. WB Saunders, pp. 567–574. 431–444.
Meyers-Wallen, V.N., Donahoe, P.K., Manganaro, T., Patterson, D.F., 1987. Schneck, G.W., 1975. Letter: hermaphroditism in a Shetland sheepdog.
Müllerian inhibiting substance in sex-reversed dogs. Biol. Reprod. 37, Vet. Rec. 96, 323.
1015–1022. Selden, J.R., Wachtel, S.S., Koo, G.C., Haskins, M.E., Patterson, D.F., 1978.
Meyers-Wallen, V.N., Patterson, D.F., 1989. Sexual differentiation and Genetic basis of XX male syndrome and XX true hermaphroditism:
inherited disorders of sexual development in the dog. J. Reprod. Fertil. evidence in the dog. Science 201, 644–646.
Suppl. 39, 57–64. Selden, J.R., Moorhead, P.S., Koo, G.C., Wachtel, S.S., Haskins, M.E., Patter-
Meyers-Wallen, V.N., Bowman, L., Acland, G.M., Palmer, V.L., Schlafer, D., son, D.F., 1984. Inherited XX sex reversal in the cocker spaniel dog.
Fajt, V., 1995. Sry-negative XX sex reversal in the German Shorthaired Hum. Genet. 67, 62–69.
Pointer dog. J. Hered. 86, 369–373. Simpson, G., England, G., Harvey, M., 1998. BSAVA Manual of Small Animal
Meyers-Wallen, V.N., Schlafer, D., Barr, I., Lovell-Badge, R., Keyzner, A., Reproduction and Neonatology. Iowa State University Press, Ames.
1999. Sry-negative XX sex reversal in purebred dogs. Mol. Reprod. Smith Jr., F.W.K., Buoen, L.C., Weber, A.F., Johnston, S.D., Randolph, J.F.,
Dev. 53, 266–273. Waters, D.J., 1989. X-chromosomal monosomy (77,X0) in a Doberman
Meyers-Wallen, V.N., 2001. Inherited abnormalities of sexual develop- Pinscher with gonadal dysgenesis. J. Vet. Intern. Med. 3, 90–95.
ment in dogs and cats. In: Concannon, P.W., England, G., Verstegen, J. Sonne, C., Dietz, R., Born, E.W., Leifsson, P.S., Andersen, S., 2008. Is there
(Eds.), Recent Advances in Small Animal Reproduction. International a link between hypospadias and organochlorine exposure in East
Veterinary Information Service, Ithaca, New York. Greenland sledge dogs (Canis familiaris)? Ecotoxicol. Environ. Saf. 69,
Nickel, R.F., Ubbink, G., Van der Gaag, I., Van Sluijs, F.J., 1992. Persistent 391–395.
Müllerian duct syndrome in the Basset Hound. Tijdschr. Diergeneeskd Stewart, R.W., Menges, R.W., Selby, L.A., Rhoades, J.D., Crenshaw, D.B.,
117 (Suppl. 1), 31S. 1972. Canine intersexuality in a Pug breeding kennel. Cornell Vet. 62,
Nie, G.J., Johnston, S.D., Hayden, D.W., Buoen, L.C., Stephens, M., 1998. 464–473.
Theriogenology question of the month. Azoospermia associated with Stünzi, B., Stünzi, H., 1950. Hermaphroditus verus unilateralis beim Hund
79,XXY chromosome complement (canine Klinefelter’s syndrome). J. mit blastomatöser Entartung des rudimentären Ovars. Schweiz. Arch.
Am. Vet. Med. Assoc. 212, 1545–1547. Tierheilkd. 92, 67–82.
Nowacka, J., Nizanski, W., Klimowicz, M., Dzimira, S., Switonski, M., 2005. Svendsen, C.K., Thomsen, P.D., Basse, A., 1985. Two cases of male pseu-
Lack of the SOX9 gene polymorphism in sex reversal dogs (78,XX; SRY dohermaphroditism in the dog. Clinical, macroscopic, karyotypic, and
negative). J. Hered. 96, 797–802. therapeutic features. Nord. Vet. Med. 37, 358–363.
Nowacka-Woszuk, J., Nizanski, W., Klimowicz, M., Switonski, M., 2007. Switonski, M., Godynicki, S., Jackowiak, H., Pienkowska, A., Turczuk-Bierla,
Normal male chromosome complement and a lack of the SRY and I., Szymas, J., Golinski, P., Bereszynski, A., 2000. X trisomy in an infer-
SOX9 gene mutations in a male pseudohermaphrodite dog. Anim. tile bitch: cytogenetic, anatomic, and histologic studies. J. Hered. 91,
Reprod. Sci. 98, 371–376. 149–150.
Nowacka-Woszuk, J., Switonski, M., 2009. Variability of CAG tan- Switonski, M., Szczerbal, I., Grewling, J., Antosik, P., Nizanski, W., Yang,
dem repeats in exon 1 of the androgen receptor gene is not F., 2003. Two cases of infertile bitches with 78,XX/77,X mosaic kary-
related with dog intersexuality. Anim. Reprod. Sci., doi:10.1016/j. otype: a need for cytogenetic evaluation of dogs with reproductive
anireprosci.2009.05.001. disorders. J. Hered. 94, 65–68.
Olson, P.N., Seim, H.B., Park, R.D., Grandy, J.L., Freshman, J.L., Carlson, E.D., Switonski, M., Nowacka, J., Skorczyk, A., Chmurzynska, A., Nizanski, W.,
1989. Female pseudohermaphroditism in three sibling Greyhounds. J. 2004. Hereditary sex-reversal syndrome (78,XX; SRY-negative) in
Am. Vet. Med. Assoc. 194, 1747–1749. German Shepherd puppies. Medycyna weterynaryjna 60, 705–707.
Pailhoux, E., Vigier, B., Chaffaux, S., Servel, N., Taourit, S., Furet, J.-P., Fellous, Tammer, I., Herzog, A., Bostedt, H., 1998. Disorders of sexual development
M., Grosclaude, F., Cribiu, E.P., Cotinot, C., Vaiman, D., 2001. A 11.7-kb in the dog: two case reports. Tierärztl. Prax. 26 (K), 390–395.
deletion triggers intersexuality and polledness in goats. Nat. Genet. Tangner, C.H., Breider, M.A., Amoss, M.S., 1982. Lateral hermaphroditism
29, 453–458. in a dog. J. Am. Vet. Med. Assoc. 181, 70–71.
Peter, A.T., Markwelder, D., Asem, E.K., 1993. Phenotypic feminization Thomas, T.N., Olson, P.N., Hoopes, P.J., 1986. Lateral hermaphroditism and
in a genetic male dog caused by nonfunctional androgen receptors. seminoma in a dog. J. Am. Vet. Med. Assoc. 189, 1596–1597.
Theriogenology 40, 1093–1105. Van Schouwenburg, S.J., Louw, G.J., 1982. A case of dysuria as a result of
Pujar, S., Kothapalli, K.S., Kirkness, E., Van Wormer, R.H., Meyers-Wallen, a communication between the urinary bladder and corpus uteri in a
V.N., 2005. Exclusion of Lhx9 as a candidate gene for SRY-negative Cairn Terrier. J. S. Afr. Vet. Assoc. 53, 65–66.
XX sex reversal in the American Cocker Spaniel model. J. Hered. 96, Volpe, P., Izzo, B., Pia Di Meo, G., Perucatti, A., Iannuzzi, L., 2000. Male pseu-
452–454. dohermaphroditism in a dog: a clinical case. Vet. Rec. 146, 532–533.
Pujar, S., Kothapalli, K.S.D., Göring, H.H.H., Meyers-Wallen, V.N., 2007. Weaver, A.D., Harvey, M.J., Munro, C.D., Rogerson, P., McDonald, M.,
Linkage to CFA29 detected in a genome-wide linkage screen of canine 1979. Phenotypic intersex (female pseudohermaphroditism) in a
pedigree segregating SRY-negative XX sex reversal. J. Hered. 98, Dachshund dog. Vet. Rec. 105, 230–232.
438–444. Wernham, B.G.J., Jerram, R.M., 2006. Male pseudohermaphroditism in a
Pullen, C.M., 1970. True bilateral hermaphroditism in a Beagle: a case Labrador Retriever, and a review of mammalian sexual differentiation.
report. Am. J. Vet. Res. 31, 1113–1117. N. Z. Vet. J. 54, 248–252.
Randolph, J.F., Center, S.A., McEntee, M., Goldberg, E.H., 1987. H-Y antigen- Whyte, A., Monteagudo, L.V., Díaz-Otero, A., Lebrero, M.E., Tejedor, M.T.,
positive XX true bilateral hermaphroditism in a German Shorthaired Falceto, M.V., Whyte, J., Gallego, M., 2009. Malformations of the
Pointer. J. Am. Anim. Hosp. Assoc. 24, 417–420. epididymis, incomplete regression of the mesonephric tubles and
Redden, B., 2007. Clinical snapshot. A vulvar mass in a dog. True hyperplasia of Leydig cells in canine persistence of Müllerian duct
hermaphrodite. Compend. Contin. Educ. Vet. 29, 240, 245. syndrome. Anim. Reprod. Sci. 115, 328–333.
Reimann-Berg, N., Murua Escobar, H., Nolte, I., Bullerdiek, J., 2008. Testic- Williams, J., Partington, B.P., Smith, B., Hedlund, C.S., Law, J.M., 1997.
ular tumor in an XXY dog. Cancer Genet. Cytogenet. 183, 114–116. Pyovagina and stump pyometra in neutered XX sex-reversed beagle:
Röcken, H., 1988. Über Hermaphroditismus beim Hund. Berl. Münch. a case report. J. Anim. Hosp. Assoc. 33, 83–90.
Tierärztl. Wschr. 101, 55–57. Williamson, J.H., 1979. Intersexuality in a family of Kerry Blue Terriers. J.
Sarafoglou, K., Ostrer, H., 2000. Familial sex reversal: a review. J. Clin. Hered. 70, 138–139.
Endocrinol. Metab. 85, 483–493. Wu, X., Wan, S., Pujar, S., Haskins, M.E., Schlafer, D.H., Lee, M.M., Meyers-
Schelling, C., Pienkowska, A., Arnold, S., Hauser, B., Switonski, M., 2001. Wallen, V.N., 2009. A single base pair mutation encoding a premature
A male to female sex-reversed dog with a reciprocal translocation. J. stop codon in the MIS type II receptor is responsible for canine Persis-
Reprod. Fertil. Suppl. 57, 435–438. tent Müllerian Duct Syndrome. J. Androl. 30, 46–56.
Animal Reproduction Science 121 (2010) 208–217

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Nutrient intake in the bovine during early and mid-gestation causes


sex-specific changes in progeny plasma IGF-I, liveweight, height and
carcass traits
G.C. Micke a , T.M. Sullivan a , K.L. Gatford b , J.A. Owens b , V.E.A. Perry a,∗
a
School of Veterinary Science, The University of Queensland, St. Lucia, QLD 4072, Australia
b
Research Centre for Early Origins of Adult Health and Disease, Robinson Institute and School of Paediatrics and Reproductive Health, University of Adelaide,
SA 5005, Australia

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

Article history: Fetal and postnatal growth are mediated by insulin-like growth factors (IGFs) and their
Received 15 February 2010 binding proteins (IGFBPs). Maternal nutrient intake during gestation can program the post-
Received in revised form 5 May 2010
natal IGF-axis. This may have significant economic implications for beef cattle production.
Accepted 25 May 2010
We investigated the effect of high (H = 240%) and low (L = 70%) levels of recommended daily
Available online 31 May 2010
crude protein (CP) intake for heifers during the first and second trimesters of gestation in a
two-by-two factorial design on progeny (n = 68) plasma IGF-I, IGF-II, total IGFBP (tIGFBP),
Keywords:
postnatal growth and carcass traits. Calves were heavier at birth following high CP diets
Cattle
Fetal programming during the second trimester (P = 0.03) and this persisted to 29 d. Plasma IGF-I concentrations
IGF of males were greater for HL compared to LL (P < 0.01) and HH (P > 0.04) from 29 to 657 d,
Maternal nutrition and for LH compared to LL from 29 until 379 d (P = 0.02). Exposure to low CP diets during
Metabolites the first trimester resulted in heavier males from 191 d onwards (P = 0.04) but a tendency
Postnatal growth for lighter females from 552 d onwards (P = 0.07) that had lighter carcass weights (P = 0.04).
Longissimus dorsi cross-sectional area of all carcasses was greater following exposure to
low CP diets during the second trimester (P = 0.04). Heifer nutrient intake during the first
and second trimesters causes persistent and sex-specific programming of progeny plasma
IGF-I, postnatal liveweight and carcass weight. Refining heifer nutritional programs during
early gestation may optimize production objectives in progeny.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction like growth factors (IGFs) are associated with traits of


economic importance to cattle production due to their role
Breeder cattle managed under extensive grazing condi- in cell growth, proliferation and metabolism (Hyatt et al.,
tions, and consequently their developing fetus, experience 2004). Specifically, calf plasma IGF-I concentration at birth
a large variation in nutrient availability throughout the is positively associated with calf birth weight (Breier et al.,
year. Maternal nutrient intake during gestation affects 1988) as is average postnatal serum IGF concentration with
progeny postnatal growth and carcass characteristics in average daily gain and linear growth (Lund-Larsen et al.,
cattle as in other species (Greenwood et al., 2006; Larson et 1977). However, there is a negative association between
al., 2009; Martin et al., 2007; Stalker et al., 2006). Insulin- average circulating IGF-I concentrations and improved feed
efficiency (Moore et al., 2005). Six IGF-binding proteins reg-
ulate IGF clearance from circulation, IGF access to receptors
∗ Corresponding author. Tel.: +61 7 4671 2417; fax: +61 7 4671 2781. (Gallaher et al., 1992) and some also act independently on
E-mail addresses: ginamicke@hotmail.com (G.C. Micke),
cells (Firth and Baxter, 2002). Fetal nutrient supply regu-
v.perry@uq.edu.au (V.E.A. Perry). lates the fetal IGF axis (Brameld et al., 2000; Oliver et al.,

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.05.017
G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217 209

1993; Osborn et al., 1992), and can program the IGF axis of 71 heifers (HH = 16; HL = 19; LH = 17; LL = 19) that
after birth in rodents (Olausson et al., 2006) and humans completed the study and gave birth (mean gestation
(Verkauskiene et al., 2005), but few studies have investi- length = 286 ± 0.5 d; range = 278–298 d) (Sullivan et al.,
gated the latter in cattle. Changes in the IGF axis may have 2009b). Three progeny were removed from the study after
long-term consequences for postnatal growth and perfor- birth: one due to mis-mothering; one pre-weaning from
mance. In sheep, maternal nutrient restriction during early sudden death of unknown causes; and one post-weaning
gestation reduces hepatic expression of type 2 IGF recep- from death due to misadventure, leaving 68 progeny dis-
tor in offspring at 3-years of age (Hyatt et al., 2007b) whilst tributed across four treatment groups (HH = 15; HL = 18;
restriction during late gestation increases hepatic expres- LH = 16; LL = 19). Hereafter, all ages refer to the average age
sion of IGF-I, but reduces hepatic expression of types 1 and of progeny on the day of sampling.
2 IGF receptor in offspring at 1-month of age (Hyatt et al., Postnatally, progeny remained with their mothers on
2007a). Despite changes to hepatic IGF gene expression, improved and native pastures until weaning at 191 d in
plasma IGF-I concentration was not altered (Hyatt et al., accordance with standard beef herd management practice
2007b). The aims of this study, therefore, were to deter- (Meat and Livestock Australia, 2004). They were supple-
mine the effect of varying heifer nutrient intake during the mented daily with whole cotton seed (Gossypium spp.)
first and second trimesters of gestation, on plasma IGF-I, -II, allocated at 1 kg/animal whilst grazing native pastures
total IGF binding proteins (tIGFBP), growth of progeny from until 401 d. From 401 d, they were managed as part of
birth until 657 days (d) and their carcass characteristics at a larger group of yearling cattle at Surat, Queensland
680 d. (27◦ 16 S, 149◦ 07 E) due to unforeseen drought conditions
at their property of origin. Each animal was allocated
2. Materials and methods 20 kg/d as fed of a silage-based ration. The ration was 40.7%
dry matter and consisted of 85% corn silage (Zea mays),
All procedures were performed with the prior approval 12% whole cottonseed (Gossypium spp.) and 3% vitamin and
of The University of Queensland Animal Ethics Committee, mineral mix. Progeny commenced an intensive feedlot fin-
approval number SVS/716/06/MLA/AACO. ishing program on 541 d at Dalby, Queensland (27◦ 18 S,
151◦ 26 E), where they remained as one group in their own
2.1. Animals and treatments feedlot pen and were fed commercial feedlot rations prior
to commercial slaughter at 680 d. Male and female progeny
The study was a two-by-two factorial design. The remained together in the same management group at all
study animals are the progeny of two-year-old heifers times throughout the study.
that have been previously described (Micke et al., 2010). Males were castrated at 153 d in accordance with stan-
Heifers (n = 118) were divided into four treatment groups dard beef herd management practice (Newman, 2007). All
on the first day of artificial insemination (AI) accord- progeny were vaccinated against clostridial diseases and
ing to stratification by weight within each composite leptospriosis (Ultravac 7 in 1: Pfizer Animal Health, West
genotype and individually stall fed until parturition. The Ryde, NSW) on 65 and 123 d with a third clostridial vaccine
four treatment groups determined the level of crude given on 544 d (Ultravac 5 in 1: Pfizer Animal Health, West
protein (CP) fed to each heifer during the first (T1) and Ryde, NSW). On 379 and 544 d all progeny were vaccinated
second (T2) trimesters of gestation (HH: T1 = 76.3 MJ ME/d against Bovine herpesvirus 1 (Rhinogard Intranasal Vaccine
and 1.4 kg CP/d; T2 = 82.4 MJ ME and 1.4 kg CP/d, HL: (Live): Q-Vax Pty Ltd., Brookfield, QLD) and Mannheimia
T1 = 76.3 MJ ME/d and 1.4 kg CP/d; T2 = 63.1 MJ ME/d haemolytica (Bovilis MH: Intervet Australia Pty Limited,
and 0.4 kg CP/d, LH: T1 = 62.5 MJ ME/d and 0.4 kg CP/d; Bendigo East, Vic.).
T2 = 82.4 MJ ME/d and 1.4 kg CP/d, LL: T1 = 62.5 MJ ME/d
and 0.4 kg CP/d; T2 = 63.1 MJ ME/d and 0.4 kg CP/d). All 2.2. Data and sample collection
heifers were fed 71.45 MJ ME and 1.06 kg CP/d in the third
trimester (T3). Trimester one was defined as 0 to 93 d after Progeny were weighed at birth (Micke et al., 2010), 15 d,
AI, T2 from 94 to 180 d and T3 from 181 d to parturition. 29 d and then approximately monthly until 657 d. Height
Transition feeding periods from 92 to 97 d and 179 to was defined as the distance between the ground and the
184 d enabled heifers to adjust to the ration changes in a cranial dorsal iliac spine and was measured on 15, 29, 65,
stepwise manner. All heifers consumed their daily feed 123, 191, 286, 379, 462, 552 and 657 d. Progeny blood sam-
allocation each day whilst under supervision. Detailed ples were collected into tubes containing lithium-heparin
composition of the heifer rations is given in Table 1 and the (Vacutainer: Becton Dickinson, Franklin Lakes, NJ) within
rationale underlying ration formulation and their feeding 5 min of birth and then immediately after progeny were
management has been described previously (Micke et al., brought in from grazing or their feedlot pen at 8:00 am on
2010). 29, 94, 191, 379 and 657 d. Samples were stored for 1–2 h
Pregnancy was positively diagnosed in 77 heifers on ice prior to being centrifuged at room temperature at
(HH = 19; HL = 20; LH = 19; LL = 19) at 39 d via transrec- 3000 × g for 10 min. Plasma was harvested and then stored
tal palpation with the aid of a 5 MHz linear rectal at −20 ◦ C until analyses.
probe attached to a real time ultrasound scanner (model At slaughter, each animal was killed by captive bolt
Aloka-500® , Aloka Inc., Tokyo, Japan). During the study, stunning and exsanguination. Standard carcasses (AUS-
six spontaneous abortions (HH = 3; HL = 1; LH = 2; LL = 0) MEAT, 1998) were halved and each side weighed prior to
(Sullivan et al., 2009a) occurred resulting in a total entering the chiller. Carcass weight (HCW) was calculated
210 G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217

Table 1
Details of “high” and “low” treatment group daily rations fed to dams during each trimester of gestationa .

Item Trimester 1 (1 to 93 d) Trimester 2 (94 to 180 d) Trimester 3 (181 d to term)

High Low High Low All

Sorghum (kg) 0.65 1.56 1.00 1.20 1.13


Cottonseed meal (kg) 2.45 0.00 2.50 0.00 1.08
Bambatsi hay (kg) 7.88 2.73 5.79 0.00 0.86
Barley straw (kg) 0.00 5.14 2.21 7.58 7.14
Limestone (kg) 0.07 0.02 0.12 0.06 0.08
Premix (kg)b 0.07 0.06 0.10 0.10 0.10
DMI (kg) 9.95 8.64 10.51 8.10 9.39
DMI (g/kg) BWc 27.81 24.67 25.91 20.18 21.23
Energy (MJ) ME 76.29 62.54 82.43 63.14 71.45
Energy (%) NRCd 243 199 229 176 149
CP (kg) 1.37 0.41 1.40 0.38 1.06
CP (%) NRCd 250 75 228 63 135
a
Data are presented on as fed basis per heifer per day.
b
Premix containing 17 g calcium, 9 g phosphorous, 2.91 g magnesium, 5 g sulphur, 27,200 IU vitamin A, 60 mg vitamin E, 70 mg iron, 150 mg zinc, 100 mg
manganese, 55 mg copper, 0.5 mg selenium, 3.4 mg cobalt and 4.2 mg iodine per 100 g.
c
Average BW at start of trimester.
d
Comparison of ration to NRC (1996) recommended nutrient requirements for pregnant Brangus replacement heifers bred at 23 months with a mature
weight of 475 kg and a calf birth weight of 32 kg.

as the sum total of the weight of both halves. Assessments of neutralized HPLC fraction 3, in an RIA specific for IGF-
of the carcasses were obtained after overnight chilling until I (Francis et al., 1989), using a rabbit polyclonal antibody
the loin temperature was less than 11 ◦ C. Carcass sides to human IGF-I (GroPep, Adelaide, Australia). Total IGFBP
were quartered between the 12th and 13th ribs and no concentrations were measured by analysis of neutralized
less than 20 min later, the cross-section area of longissimus fraction 1 in the same assay. Because IGFBP bind to and
dorsi muscle (LDA), fat depth at the rump (P8) and rib sites, sequester 125 I-IGF-I in this assay, they can be measured
and marbling score (AUS-MEAT, 1998) were assessed using due to their effect of reducing the amount of 125 I-IGF-I
the Meat Standards Australia (MSA) grading system (MSA, in the immunoprecipitated pellet, giving an apparent IGF
1999). concentration that reflects the total amount and binding
affinity of IGFBP present in plasma. The inter-assay CV
for HPLC separation and RIA of IGF-I was 10.9% (n = 18
2.3. Metabolite analyses
assays) and the intra-assay CV for extraction and assay
was 22.0% for a calf QC sample containing 31.4 ng/mL of
Plasma urea and NEFA concentrations were measured
IGF-I.
in singlicate by enzymatic colorimetric analysis on a
Plasma IGF-II concentrations were measured by analysis
Hitachi autoanalyser, using commercially available kits
of HPLC fraction 3 in a RIA specific for IGF-II (Carr et al.,
(UREA/BUN, Roche Diagnostic Systems, Mannheim, Ger-
1995), using a rabbit polyclonal antibody against human
many and NEFA-C, Wako Pure Chemical Industries, Osaka,
IGF-II (GroPep). Inter-assay CV was 9.7% (n = 9) and intra-
Japan, respectively). The inter-assay CV for a calf quality
assay covariance for extraction and assay 21.6% for a calf QC
control (QC) plasma sample containing 5.46 mmol/L urea
sample containing 78.0 ng/mL of IGF-II. The antibodies used
and 0.10 meq/L NEFA was 3 and 5%, respectively (n = 9 ana-
for human IGF-I and -II have 100 and 85% cross-reactivity
lytical runs).
with bovine IGF-I and–II, respectively (GroPep Novozymes,
product information).
2.4. IGF-I, IGF-II and tIGFBP analyses

Concentrations of plasma IGF-I, -II and tIGFBP were 2.5. Statistical analyses
measured by RIA after separation of IGF and tIGFBP by
size-exclusion HPLC under acidic conditions (Owens et al., The individual animal was considered the experimental
1990). Plasma samples were injected onto an HPLC col- unit for analyses. Measures of liveweight (BW), height and
umn, dedicated to bovine plasma samples. Four fractions plasma IGF-I, IGF-II, tIGFBP, NEFA and urea, were analysed
of eluate (fraction 1, containing IGFBP; fraction 2, inter- using repeated measures multifactorial ANOVA to deter-
peak; fraction 3, containing IGF; and fraction 4, post-peak) mine the effects of maternal nutritional treatment group
were routinely collected for each acidified plasma sample, during T1 and T2, progeny sex, time and their interaction
using collection times determined for each column based terms whilst including gestation length as a covariate to
on elution times of 125 I-IGF-I and IGF immunoreactivity. account for differences in length of exposure to the T3
Recovery of 125 I-IGF-I was 90.1 ± 0.9% for 11 HPLC runs diet. Multifactorial ANOVA including maternal nutritional
of calf plasma. Samples were assayed in triplicate in each treatment group during T1 and T2 and progeny sex were
assay, and all samples from the same animal were extracted used to analyse carcass measures, with gestation length
in a single HPLC run and run together in the same assay. included as a covariate to account for differences in length
Plasma IGF-I concentrations were measured by analysis of exposure to the T3 diet. Oneway ANOVA with Bonferroni
G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217 211

adjustment was used to explore significant interactions


between nutritional treatment group during T1 and T2.
Pairwise correlation analyses using Bonferroni adjustment
were used to explore the relationships between plasma
IGF-I, IGF-II, tIGFBP and progeny BW, height and aver-
age daily gain (ADG) in the periods pre- and post- blood
sampling, and plasma IGF-I, -II and tIGFBP with carcass
parameters.
Student t-tests were used to determine whether the
requirement for veterinary treatment during the feedlot
period had a significant effect on any of the outcomes mea-
sured. Average daily gain of male animals that received
veterinary treatment whilst in the feedlot (n = 4) was
lower between 541 and 657 d than those that did not
(1.340 ± 0.054 kg/d vs. 0.830 ± 0.323 kg/d; P < 0.01). These
animals were excluded from analyses of BW during this
period and later carcass measures. There was no effect Fig. 1. Plasma IGF-I of male progeny from birth until 657 d by mater-
of the requirement for veterinary treatment on ADG of nal nutrition during the first and second trimesters of gestation. Values
are unadjusted mean ±SEM. a = LH > LL (P = 0.02); b = HL > LL (P < 0.01) and
female progeny during the feedlot period, or on measures
HL > HH (P = 0.04).
of metabolites, IGF-I, -II or tIGFBP in male or female progeny
at 657 d. Therefore, 64 progeny remained in the study from
3.2. Plasma IGF-II
541 d onwards.
Data are presented as mean ±SEM. A probability of
Maternal nutrition during T1 and T2 did not affect
5% (P < 0.05) was accepted as the level of significance and
plasma IGF-II from birth to 657 d (P > 0.1). Plasma IGF-II of
trends reported at P < 0.1. Data was analysed using Inter-
all animals was higher at birth compared to all other mea-
cooled Stata 9.0 software (StataCorp, College Station TX
surement times (P < 0.01), increased between 29 and 94 d
77845, Texas, USA).
(P < 0.01), remained stable to 379 d, and then decreased by
657 d (P = 0.01). The effect of sex on plasma IGF-II tended to
change with time (P = 0.08) and females had higher plasma
3. Results IGF-II than males from 191 to 657 d (P = 0.02; Fig. 2). There
was no effect of gestation length on plasma IGF-II (P > 0.1).
3.1. Plasma IGF-I
3.3. Plasma tIGFBP
Plasma IGF-I concentrations from birth to 657 d var-
ied in an interaction between maternal nutrition during Maternal nutrition during T1 and T2 did not affect
T1, maternal nutrition during T2, sex and time (four-way plasma tIGFBP from birth to 657 d (P > 0.1). Plasma tIGFBP
interaction, P = 0.04), and we therefore analysed each sex of all animals increased between birth and 29 d (P < 0.01)
separately. In males, the effect of maternal nutrition during before declining slowly to 379 d to levels greater than at
T1 and T2 tended to vary with time (three-way interaction, birth (P < 0.01), then increasing to 657 d (P < 0.01). The effect
P = 0.09). There was no effect of maternal nutrition during of sex on plasma tIGFBP changed with time (P < 0.01). Males
T1 or T2 on plasma IGF-I of males at birth (each P > 0.1). tended to have higher plasma tIGFBP at birth (P = 0.07) and
From 29 to 379 d, plasma IGF-I of males was altered by had higher plasma tIGFBP from 29 until 657 d (P < 0.01)
maternal nutrition in both trimesters (two-way interac- compared to females (Fig. 2). There was no effect of ges-
tion, P = 0.01) and plasma IGF-I concentrations were higher tation length on plasma tIGFBP (P > 0.1).
in LH compared to LL males (two-way interaction, P = 0.02;
Fig. 1). Similarly, effects of maternal nutrition during each 3.4. Plasma metabolites
trimester on plasma IGF-I concentrations of males from
29 to 657 d interacted (two-way interaction, P = 0.04) and Plasma urea from birth until 657 d did not vary with
HL males had higher plasma IGF-I than LL (P < 0.01) and maternal nutrition during T1 or T2 (each P > 0.1). Plasma
HH (P = 0.04) males. There was no effect of maternal nutri- urea was higher in females than males throughout the
tion during T1 or T2 on plasma IGF-I of females over time study (P < 0.01). There was no effect of gestation length on
(each P > 0.1). Plasma IGF-I of male and female progeny fol- plasma urea (P > 0.1).
lowed a similar pattern. It increased between birth and 29 d Plasma NEFA varied with an interaction between
(P < 0.01) before declining to 379 d to levels higher than at maternal nutrition during T2, sex and time (three-way
birth (P < 0.01) and then increased at 657 d to levels higher interaction P < 0.01), and we therefore analysed NEFAs sep-
than at 379 d (P < 0.01). The effect of sex on plasma IGF- arately within each sex. For males, the effect of maternal
I changed with time (P < 0.01) as males had higher plasma nutrition during T2 changed with time (two-way interac-
IGF-I than females at all ages (P < 0.01) except birth (P > 0.1). tion, P < 0.01). At birth but not thereafter, males exposed to
There was no effect of gestation length on plasma IGF-I of low levels of maternal nutrient intake during T2 (HL + LL;
males or females (P > 0.1). 0.60 ± 0.12 meq/L) had greater plasma NEFA than their
212 G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217

Fig. 2. Plasma IGF-II and total IGFBP of male and female progeny from birth until 657 d. Values are unadjusted mean ±SEM.

high group counterparts (LH + HH; 0.258 ± 0.056 meq/L, From 552 to 657 d, female progeny exposed to high
P = 0.01). For females, maternal nutrition during T2 did not maternal nutrition during T1 (HH + HL) tended to be heav-
affect plasma NEFA (P > 0.1). Plasma NEFA was not affected ier than their low counterparts (LH + LL; P = 0.07, Fig. 3).
by progeny sex, maternal nutrition during T1 or gestation Males were heavier than females throughout the study
length (all P > 0.1). (P < 0.01) and there was no effect of gestation length on
BW (P > 0.1).
3.5. Progeny growth Progeny height varied in an interaction between
maternal nutrition during T1, sex and time (three-way
Progeny BW and ADG by trimester treatment group interaction, P < 0.01) and data were therefore analysed sep-
and sex are summarized in Table 2. At birth, progeny arately in each sex. In males, there was no effect of maternal
whose mothers were fed high nutrition during T2 were 8.3% nutrition during T1 (P > 0.1), and in females, the effect of
heavier (HH + LH = 33.05 ± 0.81 kg) than their low coun- maternal nutrition during T1 changed with time (two-way
terparts (HL + LL = 30.76 ± 0.59 kg; P = 0.03; Micke et al., interaction, P < 0.01). From 286 to 657 d, female progeny
2010). Males (33.37 ± 0.64 kg) were heavier than females exposed to high maternal nutrition during T1 (HH + HL)
(30.43 ± 0.59 kg; P < 0.01) and longer gestation length tended to be taller than their low counterparts (LH + LL;
tended (P = 0.05) to be associated with heavier birth weight P = 0.08, Fig. 3). Males were taller than females throughout
calves (Micke et al., 2010). The effect of maternal nutrition the study (P < 0.01). There was no effect of gestation length
during T2 on progeny BW persisted until 29 d (P = 0.03). on height (P > 0.1).
When data from birth until 657 d were considered in
a single analysis, BW varied in an interaction between 3.6. Progeny carcass measures
maternal nutrition during T1, sex and time (three-way
interaction, P < 0.01), and BW was therefore analysed sep- Carcass weight varied in an interaction between mater-
arately in each sex. In males, the effect of maternal nal nutrition during T1 and sex (two-way interaction,
nutrition during T1 changed with time (two-way interac- P = 0.02). In males, there was no effect of maternal nutri-
tion, P < 0.01). From 191 to 657 d, male progeny exposed tional treatment group during T1 on HCW (P > 0.1). Females
to low maternal nutrition during T1 (LH + LL) were heav- exposed to high (HH + HL: 334.53 ± 5.54 kg) compared to
ier than their high counterparts (HH + HL; P = 0.04, Fig. 3). low (LL + LH: 312.92 ± 6.82 kg) maternal nutrition during
In females, the effect of maternal nutrition during T1 T1 had heavier HCW (P = 0.04). Males had heavier HCW
also changed with time (two-way interaction, P < 0.01). (345.38 ± 5.77 kg) than females (323.41 ± 4.73 kg; P = 0.02).

Table 2
Growth of male and female progeny from birth to 657 d by postnatal nutritional period according to maternal nutritional treatment group during the first
two trimesters of gestation. Values are unadjusted means ±SEM.

1st trimester treatment group 2nd trimester treatment group

High Low High Low


Males Females Males Females Males Females Males Females

Pre-weaning n = 16 n = 17 n = 17 n = 18 n = 16 n = 15 n= 17 n = 20
ADG (kg/d) 0.96 ± 0.02 0.91 ± 0.01 0.99 ± 0.02 0.89 ± 0.01 0.96 ± 0.02 0.91 ± 0.01 0.98 ± 0.01 0.89 ± 0.01
BW at 191 d (kg) 213.0 ± 3.5 204.6 ± 2.6 220.8 ± 4.1 200.6 ± 2.6 217.0 ± 5.1 204.9 ± 3.1 217.1 ± 2.6 200.8 ± 2.8
Post-weaning 191 to 541 d n = 16 n = 17 n = 17 n = 18 n = 16 n = 15 n= 17 n = 20
ADG (kg/d) 0.63 ± 0.01 0.51 ± 0.07 0.66 ± 0.02 0.56 ± 0.01 0.65 ± 0.01 0.56 ± 0.01 0.64 ± 0.02 0.52 ± 0.06
BW at 541 d (kg) 408.6 ± 3.6* 383.1 ± 7.0 426.9 ± 6.6 370.4 ± 6.8 418.8 ± 4.1 374.1 ± 6.4 418.8 ± 6.4 378.4 ± 7.3
Post-weaning 541 to 657 d n = 14 n = 17 n = 15 n = 18 n = 15 n = 15 n = 14 n = 20
ADG 541 to 657 d (kg/d) 1.34 ± 0.06 1.58 ± 0.2 1.34 ± 0.09 1.25 ± 0.05 1.32 ± 0.09 1.32 ± 0.07 1.36 ± 0.07 1.48 ± 0.17
BW at 657 d (kg) 589.6 ± 9.7 569.8 ± 9.4 608.3 ± 14.8 540.9 ± 9.7 597.1 ± 15.4 555.8 ± 9.9 601.6 ± 9.2 554.3 ± 10.2
*
Denotes P < 0.05 for same sex comparison in the same trimester.
G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217 213

Fig. 3. Male and female progeny post-weaning liveweight and height by maternal nutrition during the first trimester of gestation. Values are unadjusted
mean ±SEM.

There was no effect of gestation length on HCW of males or 4. Discussion


females (P > 0.1).
Carcass LDA of all progeny was greater for those This study has shown that male fetuses exposed to
exposed to low (HL + LL: 79.46 ± 1.27 cm2 ) compared to a change from either high to low, or vice versa, level of
high (HH + LH: 76.19 ± 1.12 cm2 ) maternal nutrition during maternal nutrient intake at the end of the first trimester,
T2 (P = 0.04). There was no effect of sex on LDA (P > 0.1). have higher plasma IGF-I during the postnatal period in
There was no effect of maternal nutrition during T1 or T2 comparison to their counterparts exposed to a constant
on LDA corrected for HCW. There was no effect of gestation plane of maternal nutrient intake during the first two
length on LDA. trimesters of gestation. Although low maternal nutrient
Marbling score of all progeny varied with the inter- intake during the second trimester of gestation reduced
action between maternal nutrition during T1 and T2 progeny birth weight, they had caught up in bodyweight
(two-way interaction, P = 0.04). Progeny in the LL group by 65 ± 10 d. Interestingly, subsequent growth was affected
(376.8 ± 18.2) tended to have a higher marbling score than by maternal nutrient intake during the first trimester of
LH (328.8 ± 14.5; P = 0.07). There was no effect of maternal gestation in a sex-specific manner. Specifically, we found
nutritional treatment group during T1 or T2 on carcass fat that male progeny born to heifers fed low nutrient diets
depth at the P8 or rib sites, or on P8 or rib site fat depth during the first trimester of gestation were heavier during
relative to HCW (each P > 0.1). Females had greater mar- the post-weaning period than their high nutrient exposed
bling score (P < 0.01), P8 fat depth (P < 0.01), P8 fat depth counterparts. Female progeny, however, exposed to a high
relative to HCW (P = 0.01), rib fat depth relative to HCW level of maternal nutrient intake during the first trimester
(P = 0.04) and tended to have greater rib fat depth (P = 0.06) of gestation were heavier and taller during the inten-
compared to males. There was no effect of gestation length sive feeding period and had heavier HCW than their low
on marbling score, fat depth at P8 or rib site, or on fat depth counterparts. Furthermore, both BW and postnatal growth
at P8 or rib site relative to HCW (P > 0.1). rates were associated with plasma IGF-I overall, consis-
tent with the IGF axis mediating part of the long-term
effects of variable maternal nutrition on postnatal perfor-
3.7. Relationships between progeny plasma IGF and mance.
tIGFBP and growth
4.1. Nutrition
Progeny plasma IGF-I and BW were positively correlated
at 94 d (r = 0.42; P < 0.01) and 379 d (r = 0.46; P < 0.01) but Protein is often the most limiting nutrient to beef pro-
not at birth (P > 0.1). Plasma IGF-I at 29 d was positively cor- duction in Northern Australian herds (Bortolussi et al.,
related with ADG from 29 to 65 d (r = 0.40; P < 0.01). At 94 d, 2005; Willoughby, 1959), however following unseasonal
plasma IGF-I was positively correlated with ADG from 65 winter rainfall forbs that are high in protein become abun-
to 94 d (r = 0.43; P < 0.01) and 94 to 123 d (r = 0.38; P = 0.03). dant (Perry et al., 2002). The resultant pastures have an
Plasma tIGFBP was positively correlated with BW at 29 d equivalent or higher crude protein content compared to
(r = 0.37; P < 0.01). There were no other significant associa- summer grass pastures. The diets used in the current study,
tions between measures of progeny growth or carcass and therefore, were designed to reflect the nutritive value of
plasma IGF-I, -II or tIGFBP. pastures in this grazing environment.
214 G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217

Exposure of cattle to sudden changes in diet is associ- only postnatally. This contrasts with previous studies
ated with acidosis and rumenitis (Huber, 1976). As such, a where the effect of maternal nutrition on size at birth
stepwise feeding regimen was used to transition heifers to and progeny IGFs were consistent. In rats, lowered birth
their new diets at the end of the first and second trimesters. weight following intrauterine growth restriction is asso-
Clinical signs of digestive and metabolic upsets were not ciated with reduced hepatic expression of IGF-I mRNA
observed. This minimized the potential for acidosis asso- (Woodall et al., 1998) and circulating concentrations of
ciated inflammatory cytokines (Orsi and Tribe, 2008) and IGF-I in neonates (Woodall et al., 1996). Similarly, progeny
altered fetal acid–base balance (Gardner et al., 2002) to postnatal plasma IGF-I is inversely associated with birth
impact upon fetal oxygenation status and mitogenic and weight in sheep following 10 d of maternal nutrient restric-
apoptotic processes. We propose therefore, that in our tion during late gestation (Oliver et al., 2002). The effects
study the observed effects of maternal treatment group to of maternal nutrition on the progeny IGF axis of our
progeny are due to the dietary treatments imposed rather study may have several underlying causes. These poten-
than to physiological adaptations associated with dietary tially include programming via epigenetic modifications
change at the end of the first and second trimesters of ges- of activity of known regulatory influences, such as the
tation. GH axis or other endocrine axes known to be suscep-
The high and low planes of maternal nutrition dif- tible to maternal nutrition and the fetal environment
fered in both CP (3.3–3.6 fold) and energy content (1.2–1.3 (Gatford et al., 2002; Muhlhausler et al., 2009), appetite
fold), with both above the recommended National Research or satiety (De Blasio et al., 2007b; Muhlhausler et al.,
Council (NRC) energy requirements for heifers in first and 2006) or local cellular IGF-I production (Duffield et al.,
second trimesters. Therefore, the difference in CP between 2008).
the high and low diets was much greater than that of The rapid rise in plasma IGF-I and concurrent decline
energy. The degradable intake protein balances for the high in plasma IGF-II during the immediate postnatal period
and low planes of nutrition during the first trimester were seen here is consistent with patterns seen in other
206 g/d and −345 g/d, respectively, and during the second species, including rodents, sheep and humans (Jones and
trimester, 214 g/d and −464 g/d, respectively. The nega- Clemmons, 1995). Consistent with prior reports (Engström
tive degradable intake protein balance for the low plane et al., 2005; Govoni et al., 2003), plasma IGF-I and tIGFBP of
of nutrition in both the first and second trimesters clearly males were greater than females. In humans, plasma IGF-
demonstrates that protein intake was restricted. Protein II increases until the onset of puberty and then remains
intake, especially arginine, is a determinant of placental stable (Yu et al., 1999). The onset of puberty in Brah-
angiogenesis and fetal growth (Kwon et al., 2004). We man × Angus cattle, a similar genotype cross to the cattle
therefore consider that effects of maternal nutrition during in this study, occurs between 320 and 400 d of age and
the first and second trimesters of gestation upon plasma 262–336 kg liveweight (Stewart et al., 1980). This corre-
IGF-I and NEFA, BW, HCW and carcass LDA in this study sponds to the period of decreasing plasma IGF-II in female
reflect the effects of protein intake rather than energy. The calves in this study. In our study, castration of male calves
presence of a small amount of additional energy in high at 153 d prevented any pubertal effects on plasma IGF-II
protein diets was also a feature of the recent study by which may explain the earlier decline in plasma IGF-II in
Larson et al. (2009). males compared to females, despite no reported immedi-
In beef production, as in other production units in which ate effect of castration on plasma IGF-II in sheep (Gatford
dams suckle their young for a defined period, effects of pre- et al., 1996) and pigs (Owens et al., 1999). The similar onto-
natal nutrition may include an impact upon lactation. Milk genic patterns of tIGFBP and IGF-I were not unexpected
intake regulates both progeny postnatal BW (Clutter and given the roles of IGFBPs in forming stable complexes
Nielsen, 1987) and plasma IGF-I (Chellakooty et al., 2006; with circulating IGF-I slowing clearance (Thissen et al.,
Martin et al., 2005), and it is therefore possible that differ- 1994).
ences in milk intake may have partly mediated the effects of
maternal nutrient intake on these outcomes. In the current 4.3. Pre-weaning growth and metabolite responses to
study, however, there was no detectable effect of calf milk maternal nutrient intake during gestation
intake on their BW, height or plasma IGF-I, IGF-II or tIGFBP,
when the sum total of all milk intake by each animal was The majority of neonatal fat mass is present as perirenal
included as a covariate in the analyses (unpublished data). brown adipose tissue (BAT), which provides non-shivering
This was despite low maternal nutrient intake during the thermogenesis during the early neonatal period, a pro-
first trimester of gestation being associated with increased cess regulated by uncoupling protein (UCP) 1 (Clarke et al.,
calf milk intake (Sullivan et al., 2009e). 1997). Maternal nutrient restriction increases exposure of
the fetus to cortisol (Edwards and McMillen, 2001; Hough
4.2. Response of IGF to maternal nutrient intake during et al., 1990). This leads to increased UCP1 abundance in
gestation and ontogeny of IGF fetal perirenal adipose tissue (Mostyn et al., 2003) and thus
potential rate of BAT mobilization following birth. This cas-
Although maternal nutrition during pregnancy altered cade of events could explain the elevated plasma NEFA
progeny birth weight (Micke et al., 2010), this did not at birth observed in the lighter birth weight male calves
explain the elevation in plasma IGF-I in male progeny in our study. Furthermore, since growth restricted fetuses
in response to changing maternal plane of nutrition have increased amounts of adipose tissue at birth and as
between the first and second trimesters, which emerged neonates (Bispham et al., 2003; De Blasio et al., 2007a;
G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217 215

Greenwood et al., 1998), lighter calves may have had a tle, mid- to late-gestation corresponds to the period of
greater fat mass available for mobilization. It is unlikely that secondary muscle fiber formation; the largest contrib-
confounding effects of environmental temperature prior to utor to muscle mass (Gagniere et al., 1999; Robelin et
birth contributed to the change to plasma NEFA observed. al., 1991). The positive effect of low maternal nutrient
This is because calves were born over a short period of intake during the second trimester on carcass LDA in
time with no appreciable change in temperature due to our study is consistent with a prior report in cattle of
season (Bureau-Of-Meterology, 2010); blood samples were a non-significant increase in carcass LDA in association
collected within 5 min of birth; and there was no effect of with low birth weight (Greenwood et al., 2006). In fetal
nutritional treatment group on gestation length (Sullivan sheep, increased sensitivity of skeletal muscle to IGF fol-
et al., 2009e) or time of day of calving (unpublished data). lowing maternal nutrient restriction has been reported
A positive effect of increased maternal nutrient intake (Brameld et al., 2000) and we are currently investigating
on calf BW at weaning has been reported (Cafe et al., 2006; this possibility. Similarly, changes to the fetal environment
Freetly et al., 2005; Stalker et al., 2006) but in our study, during the period of pluripotent stem cell differentiation
increased progeny weight in response to increased mater- (Smas and Sul, 1995) may affect progeny intramuscular
nal feed intake during the second trimester of gestation fat deposition. Greenwood et al. (2006) did not find any
only persisted to 29 d. Changes in plasma IGF and IGFBP appreciable difference in progeny marbling in response to
have been implicated in the increased rate of neonatal feeding maternal diets low in nutrients during early and
growth exhibited by low birth weight babies (Cianfarani mid-gestation, despite using a breed predisposed to mar-
et al., 2002) but this association was not present in our bling. The tendency for increased marbling of LL compared
study. Furthermore, differences in milk intake during the to LH progeny in our study however, supports the recent
early postnatal period induced by variable maternal nutri- suggestion that maternal nutrient intake during gestation
tion during early gestation (Sullivan et al., 2009e) did not and the periconceptal period may affect progeny intramus-
explain the equalization of BW across treatment groups by cular fat deposition (Du et al., 2009; Tong et al., 2009).
65 d. Interestingly, progeny phenotype was altered in
response to maternal nutrient intake during the period
4.4. Post-weaning growth and metabolite responses to of lowest fetal nutrient demand, when the rate of fetal
maternal nutrient intake during gestation cellular proliferation and differentiation was high. This
response may have been mediated by concurrent changes
The effects of maternal nutrient intake during gestation to maternal metabolism (Sullivan et al., 2009d) and pla-
on post-weaning BW of steers in the present study differs cental function (Perry et al., 1999; Sullivan et al., 2009b),
from previous reports (Greenwood et al., 2006; Larson et in addition to possible changes to placental angiogenesis
al., 2009; Martin et al., 2007; Stalker et al., 2006). Such and vascularity (Vonnahme et al., 2007). Furthermore, dur-
inconsistencies may be caused by timing of maternal nutri- ing the third trimester of gestation there were carry-over
ent restriction; the nutritional protocol used; age of the effects of maternal nutrient intake during both the first and
dams; age of progeny at castration; age of the progeny second trimesters on maternal BW, body condition score,
at slaughter or the absence of repeat administration of ADG (Sullivan et al., 2009c), as well as to maternal plasma
anabolic steroids (Larson et al., 2009; Stalker et al., 2006). IGF-I, IGF-II and tIGFBP (Sullivan et al., 2009d). Effects
Previously, studies of the effect of maternal nutrition dur- to progeny attributed to maternal diet during the first
ing gestation on progeny post-weaning performance have trimester of gestation may, therefore, in part be induced
focused on the final trimester of gestation (Larson et al., by altered maternal state during the third trimester.
2009; Martin et al., 2007; Stalker et al., 2006). A single study
commenced between 30 and 90 d of gestation (Greenwood
et al., 2006) but none have begun at conception. Further- 5. Conclusion
more, the heifers in our study remained in positive ADG
throughout gestation (Sullivan et al., 2009c), a distinct con- This study, to our knowledge, has demonstrated for the
trast to the negative effects on maternal BW of the low first time that there is a persistent and sex-specific pro-
nutrient regimens used by previous studies (Cafe et al., gramming of plasma IGF-I in progeny in postnatal life by
2006; Larson et al., 2009; Martin et al., 2007; Stalker et al., heifer nutrient intake during the first and second trimesters
2006). of gestation. This change in abundance of a major growth
Changes to body composition of non-carcass compo- factor may partly explain concomitant effects on postnatal
nents, such as increasing visceral fat mass with ageing, BW and carcass weight, raising the possibility of refining
may explain why males exposed to low levels of mater- heifer nutritional programs during early gestation to opti-
nal nutrient intake during early gestation had heavier BW mize production objectives of their progeny. Advances in
but not carcass weights. This proposal is consistent with animal breeding technologies, such as the ability to selec-
increased abdominal obesity of male offspring following tively breed progeny of a preferred sex, may enable the
maternal nutrient restriction during early to mid-gestation sex-specific effects of maternal nutrition during early ges-
as observed in sheep (Ford et al., 2007) and humans (Ravelli tation to be utilized. Understanding the origins of these
et al., 1976). effects on IGF-I that are not immediately apparent follow-
Postnatal muscle mass accretion is dependent upon fac- ing birth, but rather develop later in postnatal life and
tors affecting fetal myogenesis as demonstrated in sheep are associated with enhanced growth, may reveal novel
(Zhu et al., 2004) and pigs (Dwyer et al., 1994). In cat- approaches to further optimization of production systems.
216 G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217

Acknowledgements IGF1 and leptin mRNA expression in the perirenal adipose tissue of
late gestation fetal sheep. Am. J. Physiol. 294, R1413–R1419.
Dwyer, C.M., Stickland, N.C., Fletcher, J.M., 1994. The influence of maternal
This research was funded by Meat and Livestock nutrition on muscle fibre number development in the porcine fetus
Australia, Sydney, Australia; Australian Agricultural Com- and on subsequent postnatal growth. J. Anim. Sci. 72, 911–917.
pany, Brisbane, Australia; and Western Australian Cattle Edwards, L.J., McMillen, I.C., 2001. Maternal undernutrition increases arte-
rial blood pressure in the sheep fetus during late gestation. J. Physiol.
Industry Compensation Fund, Perth, Australia, with the 533, 561–570.
support of Ridley AgriProducts, Melbourne, Australia; Engström, E., Niklasson, A., Wikland, K.A., Ewald, U., Hellström, A., 2005.
Milne AgriGroup, Perth, Australia; and Network in Genes The role of maternal factors, postnatal nutrition, weight gain, and gen-
der in regulation of serum IGF-I among preterm infants. Pediatr. Res.
and Environment in Development, Adelaide, Australia. We 57, 605–610.
are grateful for the untiring statistical advice received from Firth, S., Baxter, R., 2002. Cellular actions of the insulin-like growth factor
R.J. Soares Magalhaes of The School of Population Health, binding proteins. Endocr. Rev. 23, 824–854.
Ford, S.P., Hess, B.W., Schwope, M.M., Nijland, M.J., Gilbert, J.S., Vonnahme,
The University of Queensland, Herston Road, Queensland,
K.A., Means, W.J., Han, H., Nathanielsz, P.W., 2007. Maternal under-
Australia. nutrition during early to mid-gestation in the ewe results in altered
growth, adiposity, and glucose tolerance in male offspring. J. Anim.
Sci. 85, 1285–1294.
References
Freetly, H.C., Ferrell, C.L., Jenkins, T.G., 2005. Nutritionally altering weight
gain patterns of pregnant heifers and young cows changes the time
AUS-MEAT, 1998. AUS-MEAT National Accreditation Standards and Aus- that feed resources are offered without any differences in production.
tralian Meat Industry Classification System. AUS-MEAT Ltd., Brisbane, J. Anim. Sci. 83, 916–929.
Australia. Gagniere, H., Picard, B., Geay, Y., 1999. Contractile differentiation of
Bispham, J., Gopalakrishnan, G.S., Dandrea, J., Wilson, V., Budge, H., foetal cattle muscles: intermuscular variability. Reprod. Nutr. Dev. 39,
Keisler, D.H., Broughton Pipkin, F., Stephenson, T., Symonds, M.E., 637–655.
2003. Maternal endocrine adaptation throughout pregnancy to nutri- Gallaher, B.W., Breier, B.H., Oliver, M.H., Harding, J.E., Gluckman, P.D.,
tional manipulation: consequences for maternal plasma leptin and 1992. Ontogenic differences in the nutritional regulation of circu-
cortisol and the programming of fetal adipose tissue development. lating IGF binding proteins in sheep plasma. Acta Endocrinol. 126,
Endocrinology 144, 3575–3585. 49–54.
Bortolussi, G., McIvor, J.G., Hodgkinson, J.J., Coffey, S.G., Holmes, C.R., 2005. Gardner, D.S., Fletcher, A.J.W., Bloomfield, M.R., Fowden, A.L., Giussani,
The northern Australian beef industry, a snapshot 2. Breeding herd D.A., 2002. Effects of prevailing hypoxaemia, acidaemia or hypogly-
performance and management. Aust. J. Exp. Agric. 45, 1075–1091. caemia upon the cardiovascular, endocrine and metabolic responses
Brameld, J.M., Mostyn, A., Dandrea, J., Stephenson, T.J., Dawson, J.M., But- to acute hypoxaemia in the ovine fetus. J. Physiol. 540, 351–366.
tery, P.J., Symonds, M.E., 2000. Maternal nutrition alters the expression Gatford, K.L., Clarke, I.J., De Blasio, M.J., McMillen, I.C., Robinson, J.S.,
of insulin-like growth factors in fetal sheep liver and skeletal muscle. Owens, J.A., 2002. Perinatal growth and plasma GH profiles in ado-
J. Endocrinol. 167, 429–437. lescent and adult sheep. J. Endocrinol. 173, 151–159.
Breier, B.H., Gluckman, P.D., Bass, J.J., 1988. Plasma concentrations of Gatford, K.L., Fletcher, T.P., Clarke, I.J., Owens, P.C., Quinn, K.J., Walton,
insulin-like growth factor-I and insulin in the infant calf: ontogeny P.E., Grant, P.A., Hosking, B.J., Egan, A.R., Ponnampalam, E.N., 1996.
and influence of altered nutrition. J. Endocrinol. 119, 43–50. Sexual dimorphism of circulating somatotropin, insulin-like growth
Bureau-Of-Meterology 2010. Climate Statistics for Australian Locations. factor I and II, insulin-like growth factor binding proteins, and insulin:
Bureau of Meterology, Australian Government (8th February 2010; relationships to growth rate and carcass characteristics in growing
http://www.bom.gov.au/jsp/ncc/cdio/weatherData/av?p nccObs lambs. J. Anim. Sci. 74, 1314–1325.
Code=38&p display type=dataFile&p startYear=&p stn num=041521). Govoni, K.E., Hoagland, T.A., Zinn, S.A., 2003. The ontogeny of the soma-
Cafe, L.M., Hennessy, D.W., Hearnshaw, H., Morris, S.T., Greenwood, P.L., totropic axis in male and female Hereford calves from birth to one
2006. Influences of nutrition during pregnancy and lactation on birth year of age. J. Anim. Sci. 81, 2811–2817.
weights and growth to weaning of calves sired by Piedmontese and Greenwood, P.L., Cafe, L.M., Hearnshaw, H., Hennessy, D.W., Morris, S.T.,
Wagyu bulls. Aust. J. Exp. Agric. 46, 245–255. 2006. Long-term consequences of birth weight and growth to weaning
Carr, J.M., Owens, J.A., Grant, P., Walton, P.E., Owens, P.C., Wallace, J.C., on carcass, yield and beef quality characteristics of Peidmontese- and
1995. Circulating insulin-like growth factors (IGFs), IGF-binding pro- Wagyu-sired cattle. Aust. J. Exp. Agric. 46, 257–269.
teins (IGFBPs) and tissue mRNA levels of IGFBP-2 and IGFBP-4 in the Greenwood, P.L., Hunt, A.S., Hermanson, J.W., Bell, A.W., 1998. Effects of
ovine fetus. J. Endocrinol. 145, 545–557. birth weight and postnatal nutrition on neonatal sheep. I. Body growth
Chellakooty, M., Juul, A., Boisen, K.A., Damgaard, I.N., Kai, C.M., Schmidt, and composition and some aspects of energetic efficiency. J. Anim. Sci.
I.M., Petersen, J.H., Skakkebaek, N.E., Main, K.M., 2006. A prospective 76, 2354–2367.
study of serum insulin-like growth factor I (IGF-I) and IGF-binding Hough, R.L., McCarthy, F.D., Kent, H.D., Eversole, D.E., Wahlberg, M.L., 1990.
protein-3 in 942 healthy infants: associations with birth weight, gen- Influence of nutritional restriction during late gestation on produc-
der, growth velocity, and breastfeeding. J. Clin. Endocrinol. Metab. 91, tion measures and passive immunity in beef cattle. J. Anim. Sci. 68,
820–826. 2622–2627.
Cianfarani, S., Geremia, C., Scott, C., Germani, D., 2002. Growth, IGF system, Huber, T.L., 1976. Physiological effects of acidosis on feedlot cattle. J. Anim.
and cortisol in children with intrauterine growth retardation: Is catch- Sci. 43, 902–909.
up growth affected by reprogramming of the hypothalamic-pituitary- Hyatt, M.A., Budge, H., Walker, D., Stephenson, T., Symonds, M.E., 2007a.
adrenal axis? Pediatr. Res. 51, 94–99. Effects of maternal parity and late gestational nutrition on mRNA
Clarke, L., Heasman, L., Firth, K., Symonds, M.E., 1997. Influence of route of abundance for growth factors in the liver of postnatal sheep. Am. J.
delivery and ambient temperature on thermoregulation in newborn Physiol. 292, R1934–R1942.
lambs. Am. J. Physiol. 272, R1931–R1939. Hyatt, M.A., Gopalakrishnan, G.S., Bispham, J., Gentili, S., McMillen, I.C.,
Clutter, A.C., Nielsen, M.K., 1987. Effect of level of beef cow milk production Rhind, S.M., Rae, M.T., Kyle, C.E., Brooks, A.N., Jones, C., Budge, H.,
on pre- and postweaning calf growth. J. Anim. Sci. 64, 1313–1322. Walker, D., Stephenson, T., Symonds, M.E., 2007b. Maternal nutrient
De Blasio, M.J., Gatford, K.L., McMillen, I.C., Robinson, J.S., Owens, restriction in early pregnancy programs hepatic mRNA expression of
J.A., 2007a. Placental restriction of fetal growth increases insulin growth-related genes and liver size in adult male sheep. J. Endocrinol.
action, growth, and adiposity in the young lamb. Endocrinology 148, 192, 87–97.
1350–1358. Hyatt, M.A., Walker, D.A., Stephenson, T., Symonds, M.E., 2004. Ontogeny
De Blasio, M.J., Gatford, K.L., Robinson, J.S., Owens, J.A., 2007b. Placental and nutritional manipulation of the hepatic prolactin-growth
restriction of fetal growth reduces size at birth and alters postna- hormone-insulin-like growth factor axis in the ovine fetus and in
tal growth, feeding activity, and adiposity in the young lamb. Am. J. neonate and juvenile sheep. Proc. Nutr. Soc. 63, 127–135.
Physiol. Regul. Integr. Comp. Physiol. 292, R875–R886. Jones, J.I., Clemmons, D.R., 1995. Insulin-like growth factors and their
Du, M., Tong, J., Zhao, J., Underwood, K.R., Zhu, M., Ford, S.P., Nathanielsz, binding proteins: biological actions. Endocr. Rev. 16, 3–34.
P.W., 2009. Fetal programming of skeletal muscle development in Kwon, H., Ford, S.P., Bazer, F.W., Spencer, T.E., Nathanielsz, P.W., Nijland,
ruminant animals. J. Anim. Sci., doi:10.2527/jas.2009-2311. M.J., Hess, B.W., Wu, G., 2004. Maternal nutrient restriction reduces
Duffield, J.A., Vuocolo, T., Tellam, R., Yuen, B.S., Muhlhausler, B.S., concentrations of amino acids and polyamines in ovine maternal and
McMillen, I.C., 2008. Placental restriction of fetal growth decreases fetal plasma and fetal fluids. Biol. Reprod. 71, 901–908.
G.C. Micke et al. / Animal Reproduction Science 121 (2010) 208–217 217

Larson, D.M., Martin, J.L., Adams, D.C., Funston, R.N., 2009. Winter grazing Ravelli, G.P., Stein, Z.A., Susser, M.W., 1976. Obesity in young men after
system and supplementation during late gestation influence perfor- famine exposure in utero and early infancy. New Engl. J. Med. 295,
mance of beef cows and steer progeny. J. Anim. Sci. 87, 1147–1155. 349–353.
Lund-Larsen, T.R., Sundby, A., Kruse, V., Velle, W., 1977. Relation between Robelin, J., Loucourt, A., Bechet, D., Ferrara, M., Briand, Y., Geay, Y., 1991.
growth rate, serum somatomedin and plasma testosterone in young Muscle differentiation in the bovine fetus: a histological and histo-
bulls. J. Anim. Sci. 44, 189–194. chemical approach. Growth Develop. Aging 55, 151–160.
Martin, J.L., Vonnahme, K.A., Adams, D.C., Lardy, G.P., Funston, R.N., 2007. Smas, C.M., Sul, H.S., 1995. Control of adipocyte differentiation. Biochem.
Effects of dam nutrition on growth and reproductive performance of J. 309, 697–710.
heifer calves. J. Anim. Sci. 85, 841–847. Stalker, L.A., Adams, D.C., Klopfenstein, T.J., Feuz, D.M., Funston, R.N., 2006.
Martin, R.M., Holly, J.M., Davey Smith, G., Ness, A.R., Emmett, P., Rogers, Effects of pre- and postpartum nutrition on reproduction in spring
I., Gunnell, D., 2005. Could associations between breastfeeding and calving cows and calf feedlot performance. J. Anim. Sci. 84, 2582–2589.
insulin-like growth factors underlie associations of breastfeeding with Stewart, T.S., Long, C.R., Cartwright, T.C., 1980. Characterization of cattle
adult chronic disease? The Avon longitudinal study of parents and of a five-breed Diallel. III. Puberty in bulls and Heifers. J. Anim. Sci. 50,
children. Clin. Endocrin. (Oxf.) 62, 728–737. 808–820.
Meat and Livestock Australia, 2004. More Beef from Pastures: the Pro- Sullivan, T.M., Micke, G.C., Greer, R.M., Irving-Rodgers, H.F., Rodgers, R.J.,
ducer’s Manual. Meat and Livestock Australia Ltd., Sydney, Australia. Perry, V.E.A., 2009a. Dietary manipulation of Bos indicus × heifers dur-
Micke, G.C., Sullivan, T.M., Magalhaes, R.J.S., Rolls, P.J., Norman, S.T., Perry, ing gestation affects the reproductive development of their heifer
V.E.A., 2010. Heifer nutrition during early- and mid-pregnancy alters calves. Reprod. Fertil. Develop. 21, 773–784.
fetal growth trajectory and birth weight. Anim. Reprod. Sci. 117, 1–10. Sullivan, T.M., Micke, G.C., Magalhaes, R.S., Martin, G.B., Wallace, C.R.,
Moore, K.L., Johnston, D.J., Graser, H.-U., Herd, R., 2005. Genetic and phe- Green, J.A., Perry, V.E.A., 2009b. Dietary protein during gestation
notypic relationships between insulin-like growth factor-I (IGF-I) and affects circulating indicators of placental function and fetal develop-
net feed intake, fat, and growth traits in Angus beef cattle. Aust. J. ment in heifers. Placenta 30, 348–354.
Agric. Res. 56, 211–218. Sullivan, T.M., Micke, G.C., Magalhaes, R.S., Phillips, N.J., Perry, V.E.A.,
Mostyn, A., Pearce, S., Budge, H., Elmes, M., Forhead, A.J., Fowden, A.L., 2009c. Dietary protein during gestation affects placental development
Stephenson, T., Symonds, M.E., 2003. Influence of cortisol on adi- in heifers. Theriogenology 72, 427–438.
pose tissue development in the fetal sheep during late gestation. J. Sullivan, T.M., Micke, G.C., Perkins, N., Martin, G.B., Wallace, C.R., Gatford,
Endocrinol. 176, 23–30. K.L., Owens, J.A., Perry, V.E.A., 2009d. Dietary protein during gestation
MSA, 1999. Grading for Eating Quality—Development of the Meat Stan- affects maternal IGF, IGFBP, leptin concentrations, and fetal growth in
dards Australia Grading System, vol. 1. Meat Standards Australia, heifers. J. Anim. Sci. 87, 3304–3316.
Newstead, Australia. Sullivan, T.M., Micke, G.C., Perry, V.E.A., 2009e. Influences of diet
Muhlhausler, B.S., Adam, C.L., Findlay, P.A., Duffield, J.A., McMillen, during gestation on potential postpartum reproductive perfor-
I.C., 2006. Increased maternal nutrition alters development of the mance and milk production of beef heifers. Theriogenology,
appetite-regulating network in the brain. FASEB J. 20, 1257–1259. doi:10.1016/j.theriogenology.2009.07.016.
Muhlhausler, B.S., Duffield, J.A., Ozanne, S.E., Pilgrim, C., Turner, N., Mor- Thissen, J.P., Ketelslegers, J.M., Underwood, L.E., 1994. Nutritional regula-
rison, J.L., McMillen, I.C., 2009. The transition from fetal growth tion of the insulin-like growth factors. Endocr. Rev. 15, 80–101.
restriction to accelerated postnatal growth: a potential role for insulin Tong, J.F., Yan, X., Zhu, M.J., Ford, S.P., Nathanielsz, P.W., Du, M., 2009.
signalling in skeletal muscle. J. Physiol. 587, 4199–4211. Maternal obesity downregulates myogenesis and {beta}-catenin sig-
Newman, R., 2007. A Guide to Best Practice Husbandry in Beef Cattle: naling in fetal skeletal muscle. Am. J. Physiol. 296, E917–924.
Branding, Castrating and Dehorning. Meat and Livestock Australia Ltd., Verkauskiene, R., Jaquet, D., Deghmoun, S., Chevenne, D., Czernichow, P.,
Sydney, Australia. Levy-Marchal, C., 2005. Smallness for gestational age is associated
Olausson, H., Lewitt, M., Brismar, K., Uvnas-Moberg, K., Sohlstrom, A.I., with persistent change in insulin-like growth factor I (IGF-I) and the
2006. Maternal food restriction during gestation elevates insulin-like ratio of IGF-I/IGF-binding protein-3 in adulthood. J. Clin. Endocrinol.
growth factor I and insulin-like growth factor binding protein 1 in Metab. 90, 5672–5676.
adult male rat offspring. Nutr. Res. 26, 350–355. Vonnahme, K.A., Zhu, M.J., Borowicz, P.P., Geary, T.W., Hess, B.W.,
Oliver, M.H., Breier, B.H., Gluckman, P.D., Harding, J.E., 2002. Birth weight Reynolds, L.P., Caton, J.S., Means, W.J., Ford, S.P., 2007. Effect of early
rather than maternal nutrition influences glucose tolerance, blood gestational undernutrition on angiogenic factor expression and vas-
pressure, and IGF-I levels in sheep. Pediatr. Res. 52, 516–524. cularity in the bovine placentome. J. Anim. Sci. 85, 2464–2472.
Oliver, M.H., Harding, J.E., Breier, B.H., Evans, P.C., Gluckman, P.D., 1993. Woodall, S.M., Bassett, N.S., Gluckman, P.D., Breier, B.H., 1998. Conse-
Glucose but not a mixed amino acid infusion regulates plasma insulin- quences of maternal undernutrition for fetal and postnatal hepatic
like growth factor-I concentrations in fetal sheep. Pediatr. Res. 34, insulin-like growth factor-I, growth hormone receptor and growth
62–65. hormone binding protein gene regulation in the rat. J. Mol. Endocrinol.
Orsi, N.M., Tribe, R.M., 2008. Cytokine networks and the regulation of 20, 313–326.
uterine function in pregnancy and parturition. J. Neuroendocrinol. 20, Woodall, S.M., Breier, B.H., Johnston, B.M., Gluckman, P.D., 1996. A model
462–469. of intrauterine growth retardation caused by chronic maternal under-
Osborn, B.H., Fowlkes, J., Han, V.K., Freemark, M., 1992. Nutritional regu- nutrition in the rat: effects on the somatotropic axis and postnatal
lation of insulin-like growth factor-binding protein gene expression growth. J. Endocrinol. 150, 231–242.
in the ovine fetus and pregnant ewe. Endocrinology 131, 1743–1750. Willoughby, W.M., 1959. Limitations to animal production imposed by
Owens, P.C., Gatford, K.L., Walton, P.E., Morley, W., Campbell, R.G., 1999. seasonal fluctuations in pasture and by management procedures.
The relationship between endogenous insulin-like growth factors and Aust. J. Agric. Res. 10, 248–268.
growth in pigs. J. Anim. Sci. 77, 2098–2103. Yu, H., Misrty, J., Nicar, M.J., Khosravi, M.J., Diamandis, A., van Doorn, J.,
Owens, P.C., Johnson, R.J., Campbell, R.G., Ballard, F.J., 1990. Growth hor- Juul, A., 1999. Insulin-like growth factors (IGF-I, free IGF-I, and IGF-
mone increases insulin-like growth factor (IGF-I) and decreases IGF-II II) and insulin-like growth factor binding proteins (IGFBP-2, IGFBP-3,
in plasma of growing pigs. J. Endocrinol. 124, 269–275. IGFBP-6, and ALS) in blood circulation. J. Clin. Lab. Anal. 13, 166–172.
Perry, V.E., Norman, S.T., Owen, J.A., Daniel, R.C., Phillips, N., 1999. Low Zhu, M.J., Ford, S.P., Nathanielsz, P.W., Du, M., 2004. Effect of maternal
dietary protein during early pregnancy alters bovine placental devel- nutrient restriction in sheep on the development of fetal skeletal mus-
opment. Anim. Reprod. Sci. 55, 13–21. cle. Biol. Reprod. 71, 1968–1973.
Animal Reproduction Science 121 (2010) 218–224

Contents lists available at ScienceDirect

Animal Reproduction Science


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

mRNA of luteal genes associated with progesterone synthesis,


maintenance, and apoptosis in dairy heifers and lactating dairy cows
T. Pretheeban, A. Balendran, M.B. Gordon, R. Rajamahendran ∗
Department of Animal Science, University of British Columbia, 229-2357, Main Mall, Vancouver, BC, Canada V6T 1Z4

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

Article history: This study was performed to investigate the role of corpus luteum (CL) in reduced preg-
Received 1 February 2010 nancy rates (PR) observed in high producing lactating dairy cows. Development of CL and
Received in revised form 5 May 2010
secretion of progesterone (P4 ) play a key role in early embryo development, implantation,
Accepted 20 May 2010
and maintenance of pregnancy. Time of ovulation was synchronized in dairy heifers and
Available online 1 June 2010
second/third parity lactating dairy cows and CL enucleated surgically under local anesthe-
sia on day 10 of the estrous cycle. Quality of the CL in dairy heifers (n = 5) and lactating dairy
Keywords:
cows (n = 5) was compared by analyzing the expression of candidate genes by mRNA assess-
Corpus luteum
Dairy heifer ments using quantitative real-time PCR. Amounts of mRNA for factors associated with P4
Lactating dairy cow synthesis: 3␤HSD, anti-apoptotic function: BCL2, angiogenesis: VEGF, IGF1, and FGF2, and
Progesterone synthesis luteal maintenance: IL1A were greater (P < 0.05) in CL obtained from dairy heifers compared
Luteal maintenance to that of lactating dairy cows. Also a greater ratio for BAX:BCL2 mRNA was observed in
Angiogenesis lactating cows. Therefore, genes regulating angiogenic, steroidogenic, and luteotropic fac-
tors are highly expressed in heifers compared to lactating dairy cows, whereas apoptosis
seemed to be more evident in CL of lactating cows. These findings suggest that CL of lac-
tating dairy cows have reduced luteotropic as well as steroidogenic capacities on day 10 of
the estrous cycle and might have played a critical role in reduced PR observed in lactating
dairy cows.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction from 51.5% in the first parity to 19.5% in the third/fourth


parity. The causes for the reduced PR in lactating dairy
Reduced pregnancy rates (PR) in high yielding dairy cows seem to be multifactorial and include disruptions in
cows are a world wide problem and a matter of great con- the ovarian follicular dynamics, corpus luteum (CL) func-
cern to dairy producers due to the immense economic tion (Opsomer et al., 1998; Wolfenson et al., 2004), poor
losses (Lucy, 2001). This has led to increased research estrous behavior and/or detection (Lopez et al., 2004),
efforts to further understand the underlying mechanisms reduced oocyte/embryo quality (Lucy, 2007), and com-
of reduced PR in lactating dairy cows. Although PR in promised oviductal and uterine environments (Gustafsson
lactating dairy cows have declined gradually over the and Larsson, 1985; Binelli et al., 1999; Spencer et al.,
past 50 years, PR of dairy heifers have remained constant 2007).
(Pursley et al., 1997). Balendran et al. (2008) showed that Formation of CL and secretion of progesterone (P4 ) are
PR in heifers after first and second artificial insemina- of paramount importance for the early embryonic devel-
tions (AI) was 83.4% and it declined in lactating dairy cows opment, implantation, and maintenance of pregnancy. P4
concentrations during early pregnancy have a marked
effect on PR. Many studies have demonstrated that lower
∗ Corresponding author. Tel.: +1 604 822 4784; fax: +1 604 822 4400. concentration of P4 in milk (Lamming et al., 1989; Mann
E-mail address: raja@interchange.ubc.ca (R. Rajamahendran). et al., 1995) and in plasma (Butler et al., 1996; Mann and

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.05.016
T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224 219

Lamming, 2001) of cows are associated with higher rates 2.2. Corpus luteum enucleation and processing
of pregnancy failures.
The formation, function, and maintenance of CL are Epidural anesthesia (lidocaine; 2%; 4–5 mL) was admin-
regulated by various luteotropic factors before and after istered to lactating dairy cows and an incision was made in
ovulation, as well as by the inhibition of several luteolytic the ventral wall of the vagina and CL removed as previously
factors during the period of active CL function (Stocco et described (Aali et al., 2004). In heifers, CL was removed sur-
al., 2007). These regulatory mechanisms are mediated by gically through a flank incision under paralumbar block and
the cyclic expression of key genes during the lifespan of CL, using lidocaine hydrochloride (lidocaine; 2%; 4–5 mL). In all
and therefore, determine the functional capacities, as well animals CL were located and enucleated by applying gen-
as the lifespan of CL tissue. Major categories of genes that tle pressure to the ovarian area surrounding the gland. All
participate in the formation, maintenance, secretion, and animals were moved and housed in individual free stalls
regression of CL are those involved in extracellular matrix for recovery and observed continually during the first 12 h
and cytoskeleton formation, transcriptional regulation, after surgery, and were kept in individual pens for 3–5 days.
angiogenesis, steroidogenesis, oxygen metabolism, apop- Soon after the enucleation, CL was snap frozen in liquid
tosis, and inflammation (Casey et al., 2005; Goravanahally nitrogen and stored at −80 ◦ C until processing. Blood sam-
et al., 2009). Therefore, it is expected that any alterations ples were taken from the tail vein in all the animals on day
in the coordinated expression of the above genes could 8 post-ovulation and on the day of enucleation (day 10) for
compromise the optimal development and function of the the determination of P4 concentrations. Plasma was sepa-
CL. rated by centrifuging the blood samples at 1000 × g at 4 ◦ C
In this study we compared the mRNA of candidate for 10 min and stored at −20 ◦ C until radioimmunoassay.
genes in the CL during mid luteal phase, between dairy
heifers and second/third parity lactating dairy cows. These 2.3. RNA extraction
genes are involved in steroidogenesis (steroidogenic acute
regulatory protein: StAR, cytochrome P450 family 11 Total RNA was extracted using a single step RNA iso-
sub-family A polypeptide 1: CYP11A1; formerly known lation method (Chomczynski and Sacchi, 1987), using
as P450scc, and 3-beta-hydroxysteroid dehydrogenase: a commercially available total RNA isolation solution,
3␤HSD), angiogenesis (vascular endothelial growth fac- Tri Reagent (Sigma–Aldrich, USA). Briefly, luteal tissues
tor: VEGF, insulin like growth factor: IGF1, and fibroblast (100 mg) were pulverized in liquid nitrogen using mor-
growth factor: FGF2), luteal maintenance (interleukin 1␣: tar and pestle, then immediately transferred into sterile
IL1A and tumor necrotic factor ␣: TNF), and apoptosis or nuclease-free microcentrifuge tubes containing 1000 ␮L of
heat stress (BAX, BCL2, and HSPA1A). We hypothesized Tri Reagent. Contents were mixed thoroughly by vortex-
that expression of aforementioned gene transcripts differ ing and allowed to stand for 5 min at room temperature.
between CL of dairy heifers and lactating dairy cows. Then 200 ␮L of chloroform was added to each tube, and
agitated vigorously for 30 s. The tubes were allowed to
2. Materials and methods stand at room temperature for 15 min and were cen-
trifuged at 12,000 × g for 15 min at 4 ◦ C. The top layer
2.1. Animals and treatments with clear transparent solution was carefully transferred
into a new set of nuclease-free centrifuge tubes and added
This study was conducted at UBC Dairy Education and 750 ␮L of isopropanol to each tube. Contents were mixed
Research Centre in Agassiz, British Columbia, Canada. Dairy by slightly inverting the tube thrice, and again allowed to
heifers (14 months of age) and second or third parity lac- stand at room temperature for 30 min before centrifugation
tating dairy cows (60 days post-partum, BCS – 2.75) with at 12,000 × g for 10 min at 4◦ C. Supernatant was discarded
regular ovarian activity and no reproductive abnormali- and the pellet at the bottom of the tube was washed twice
ties were treated with a protocol to synchronize time of with 1000 ␮L of ice-cold 75% ethanol by centrifugation
ovulation (Aali et al., 2004). Briefly, animals were treated at 7500 × g for 5 min, air dried for 10–15 min thereafter,
with GnRH (buserelin; 100 ␮g; im) followed by dinoprost and finally dissolved in 100 ␮L of sterile DEPC treated
tromethamine (lutalyse; 25 mg; im) 7 days later, and a water. The quantity and quality of RNA were assessed
second dose of GnRH (buserelin; 100 ␮g; im) was admin- by measuring the optical densities using Nanodrop ND-
istered 48 h after dinoprost treatment. All animals were 1000 spectrophotometer and by observing clear bands for
examined once a day from the day of first GnRH treatment 28S, and 18S, ribosomal RNA species on ethidium bromide
until enucleation for ovulation (day 0) and development of stained agarose gel (1%). Total RNA was stored at −80◦ C for
CL using ultrasonography and CL were enucleated on day copy DNA (cDNA) synthesis.
10 from five animals in each group. Animals were housed in
free stall barns and fed a total mixed ration of corn and grass 2.4. Reverse transcription
silage, hay and concentrates. Lactating dairy cows were fed
twice daily and dairy heifers were fed once daily according Reverse transcription (RT) of RNA was performed using
to their dry matter requirements (NRC, 2001). All handling a first-strand cDNA synthesis kit (Cells-to-cDNATM II Kit,
and management of animals used in this study were in Ambion, Austin, USA). The RT reactions were performed
accordance with the guidelines of the Canadian Council on by following the kit manufacturer’s protocol with slight
Animal Care (1993) and approved by the Animal Care and modification to match the experimental conditions. RNA
Use Committee of the University of British Columbia. samples were treated with DNase and first-strand cDNA
220 T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224

Table 1
Details of the primers used for PCR amplification of corpus luteum genes.

Gene name Primer sequences (5 –3 ) Ta (◦ C) Gene accession number

GAPDH F – TGTTCCAGTATGATTCCACCC 58 U85042


R – AGGAGGCATTGCTGACAATC
VEGF F – TGTAATGACGAAAGTCTGCAG 60 M32976
R – TCACCGCCTCGGCTTGTCACA
IGF1 F – TCAGTTCGTGTGCGGAGACA 56 NM001077828
R – ACTTCCTTCTGAGCCTTGGG
FGF2 F – TACAACTTCAAGCAGAAGAG 56 NM174056
R – CAGCTCTTAGCAGACATTGG
StAR F – CATGGTGCTCCGCCCCTTGGCT 58 BC110213
R – CATTGCCCACAGACCTCTTGA
3␤HSD F – TGTTGGTGGAGGAGAAGG 58 BC111203
R – GGCCGTCTTGGATGATCT
CYP11A1 F – AACGTCCCTCCAGAACTGTACC 58 BC133389
R – CTTGCTTATTGCTCCCTCTGCC
IL1A F – CTCTCTCAATCAGAAGTCCTTCTATG 58 NM174092
R – CATGTCAAATTTCACTGCCTCCTCC
TNF F – GAAGCTGGAAGACAACCA 60 NM173966
R – TCCCAAAGTAGACCTGCC
BAX F – TGCTTCAGGGTTTCATCCAG 58 U92569
R – AACATTTCAGCCGCCACTC
BCL2 F – TTCGCCGAGATGTCCAGTCAGC 62 U92434
R – GTTGACGCTCTCCACACACA
HSPA1A F – CACTTCGTGGAGGAGTTCA 58 AY149619
R – GGTTGATGCTCTTGTTGAGG

F: forward primer; R: reverse primer; Ta : annealing temperature.

was synthesized by incubating a 20 ␮L reaction mixture gel. The gels were photographed under ultraviolet illumi-
containing 2.5 ␮M of random decamers, 1.25 ␮M of oligo nation. At the beginning of the experiment, all the primer
dT primers, 0.25 mM of deoxyribonucleoside triphosphate sets were standardized using semi-quantitative RT-PCR
mixture, 2 ␮L of 10× RT buffer (pH 7.4), 0.5 U/␮L of RNase and commercially available JumpStart RED Taq Ready Mix
inhibitor, 0.5 U/␮L of M-MLV reverse transcriptase, 2 ␮g of PCR reaction mix (Jumpstart; Sigma–Aldrich Canada Ltd.,
total RNA and nuclease-free water at 42 ◦ C for 1 h. Inactiva- Oakville, ON) from slaughter house CL samples. Amplifica-
tion of reverse transcriptase was performed by incubating tion of single bands was confirmed and the PCR products
the reaction mixture at 94 ◦ C for 10 min and the product were sequenced to confirm their identity. The primer
was stored at −20◦ C to use in the PCR amplification. sequence, fragment size, annealing temperature, number
of PCR cycles, and gene accession number are provided in
Table 1.
2.5. Quantitative real-time PCR

Amount of mRNA was compared for all the genes in 2.6. Progesterone RIA
this study using quantitative real-time PCR (Q-RT-PCR)
and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) Radioimmunoassay was performed using a Coat-A-
as the housekeeping gene. PCR was performed using the Count progesterone kit (Diagnostic Products, Los Angeles,
iCycler, CFX96 Real-Time PCR Detection System [Bio-Rad USA). Reference standards or serum samples (100 ␮L) were
laboratories (Canada) Ltd.], iQ SYBR Green Super Mix [Bio- added to P4 antibody coated tubes. Buffered I125 -labelled P4
Rad laboratories (Canada) Ltd.] and gene specific primers (1.0 mL) was added to all tubes and vortexed. After 3 h of
(Table 1). For each sample PCR reaction consisted of 2.5 ␮L incubation, tubes were decanted and counted for radioac-
of cDNA (fractions of 1/20 of original cDNA), 10 ␮M of tivity on a gamma counter for 1 min and P4 concentrations
each primer in a final volume of 25 ␮L per reaction. Reac- were detected. Coefficient of variation (CV) within (intra)
tion conditions included an initial step of 95 ◦ C (5 min), and between (inter) assays were 7% and 9%, respectively.
followed by 40 cycles of 95 ◦ C (15 s), 60 ◦ C (1 min), and The sensitivity of the assay was 0.03 ng/mL.
72 ◦ C (20 s). Melt curve analysis was performed for each
gene to confirm the presence of a single peak. Q-RT-PCR
data (Ct values) were analyzed using the comparative Ct 2.7. Statistical analysis
method (Livak and Schmittgen, 2001). Values (Ct values)
of all genes for each sample were adjusted according to Data analysis for mRNA expression and P4 concentration
corresponding housekeeping/control gene (GAPDH) values were performed by one-way ANOVA. Mean separation pro-
(Ct values). Calculations of relative quantitation were per- cedure was performed when analysis of variance showed
formed using the sample with the lowest value in the significant F-values using Fisher’s least significant differ-
group as a control group and using the formula (2−Ct ). ence. The results were reported as the mean values for
The PCR products were confirmed by gel electrophoresis each set of data ± SEM and statistical significance declared
using ethidium bromide (0.4 ␮g/mL) stained 1% agarose at probability level (P) less than 0.05.
T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224 221

Fig. 1. Amounts of mRNA of genes associated with (A) apoptosis and heat shock, (B) steroidogenesis, (C) angiogenesis and (D) luteal maintenance, in dairy
heifers and lactating dairy cows. The data are represented as mean ± SEM. *P < 0.05.

3. Results 3.4. mRNA of genes associated with luteal maintenance

3.1. mRNA of apoptotic molecules and heat shock protein When the mRNA of genes associated with luteal mainte-
nance was analyzed, IL1A was highly expressed in the CL of
The quantitative RT-PCR results of luteal tissue mRNA dairy heifers, compared to that of lactating cows (P < 0.05).
for apoptotic molecules (BAX and BCL2) and heat shock TNF, mRNA was not different between these two groups
protein (HSPA1A) are shown in Fig. 1A. Analysis of vari- (Fig. 1D).
ance revealed greater mRNA of BCL2 (P < 0.05) in heifers,
compared to that of lactating cows. However, amounts of 3.5. Progesterone assay
BAX and HSPA1A did not differ between the two groups.
The ratio of BAX:BCL2 was greater in lactating dairy cows, Plasma concentrations of P4 on days 8 and 10 of the
in comparison to that of dairy heifers. estrous cycle (mid luteal phase) did not show any signifi-
cant differences between dairy heifers and lactating dairy
cows (data not shown).
3.2. mRNA of steroidogenic enzymes

The amounts of mRNA for the steroidogenic enzymes 4. Discussion


in CL were examined using quantitative real-time PCR. No
differences were observed for the StAR and CYP11A1 mRNA Investigation of reduced PR in high producing dairy
between dairy heifers and lactating dairy cows. There was cows is a complex procedure and comparing the quality of
a greater amount of mRNA for 3␤HSD (P < 0.05) in dairy CL using gene expression profiles will give an insight into
heifers compared to that of lactating dairy cows (Fig. 1B). some of the underlying causes at molecular level. Proper
formation, function, and maintenance of CL are essential
for adequate P4 secretion which is critically important for
3.3. mRNA of genes associated with angiogenesis the early embryo development, implantation, and mainte-
nance of pregnancy in many animal species including cattle
The quantitative real-time PCR results of luteal tissue (Niswender et al., 2000; Spencer et al., 2007).
mRNA for genes associated with luteal angiogenesis (VEGF, Apoptosis is an important process that occurs at all
IGF1, and FGF2) was greater (P < 0.05) in dairy heifers com- stages of the CL lifespan. The degree of apoptosis differs
pared to that of lactating dairy cows (Fig. 1C). between different stages especially between CL develop-
222 T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224

ment and regression. The apoptotic molecules of BCL2 IGF1 (Schams and Berisha, 2002). We compared amounts
family (activator, BAX and inhibitor, BCL2) are a determi- of mRNA of the aforementioned genes in CL of heifers and
nant of apoptotic status and the fate of cells in distinct lactating dairy cows, because inadequate luteal function
tissues including CL (Korsmeyer, 1995; Tilly, 1996). Find- is associated with decreased luteal vascularity (Reynolds,
ings in the present study indicated greater BCL2 mRNA 1986). Lesser amounts of mRNA found in the present study
and a lesser ratio of BAX:BCL2 mRNA in heifers compared for all three factors in lactating dairy cows may indicate
to lactating dairy cows. This indicates a possible occur- reduced vascularity in the CL of cows compared to that of
rence of increased rate of apoptosis in CL of lactating heifers. Furthermore, FGF2 acts as mitogen and induces the
dairy cows compared to that of dairy heifers. It is fur- secretion of P4 in CL of cattle during the mid luteal phase
ther supported by the studies in human CL, where they (Miyamoto et al., 1992). The greater amounts of BCL2 mRNA
have demonstrated the greatest amounts of BCL2 mRNA in in the present study could also be due to the anti-apoptotic
the midluteal functional CL and greatest amounts of BAX effects of FGF2 and VEGF in endothelial cells (Pardo et al.,
mRNA in the regressing CL (Rueda et al., 1997; Sugino et 2002; Bufalo et al., 2003). It is speculated that stimulation
al., 2000). of steroidogenesis and inhibition of apoptosis in the CL of
Heat shock proteins are the molecular chaperones that lactating dairy cows would be compromised due to the
help other proteins to undergo folding, transport, and lesser amounts of IGF1 mRNA compared to that of heifers
assembly into multi-protein complexes, or to refold after (Neuvians et al., 2003; Townson, 2006).
heat shock or other stresses (Didelot et al., 2007). It has been The role of cytokines such as IL1A and TNF in CL survival
widely recognized that HSPA1A promotes cell survival in a and luteolysis is an active area of research. Treatment of
variety of tissues by preventing apoptosis caused by differ- endometrium with IL1A increases the production of PGE2 ,
ent means such as heat shock, UV radiation, and inadequate a luteotrophic factor and thereby the PGE2 :PGF2␣ ratio in
nutrition (Beere, 2004, 2005). There is evidence for possi- cows (Tanikawa et al., 2005; Okuda and Sakumoto, 2006;
ble protective actions of HSPA1A during induced luteolysis Skarzynski et al., 2008). Also, it increases the production of
in sheep (McPherson et al., 1993), as well as a media- P4 and the lifespan of the CL. Greater amounts of IL1A mRNA
tor in luteal regression in rats (Narayansingh et al., 2004). in heifers in the present study suggests a greater probabil-
When considering the stressors, especially heat shock, even ity of survival and function of their CL compared to that of
though both heifers and cows are managed under similar lactating dairy cows. Furthermore, endometrial production
conditions the effect of milk production causes an increase of PGE2 increases during early and mid luteal phases in cat-
in the body temperature in lactating dairy cows (Sartori tle (Miyamoto et al., 2000; Murakami et al., 2001). This is
et al., 2002). Therefore, it was expected that an induced further supported from recent findings, that the expres-
expression of HSPA1A mRNA in cows compared to heifers sion of the IL1A gene is greater in the endometrium of
because of HSPA1A’s inducible nature by which it resists dairy heifers as indicated by amounts of mRNA and pro-
subsequent apoptotic stimuli (Dimmeler and Zeiher, 1997). tein compared to lactating dairy cows (Pretheeban et al.,
However, in the present study, amount of HSPA1A mRNA 2009). Luteal survival and P4 secretion were also enhanced
was not different between heifers and lactating dairy cows. after treating endometrium with TNF in cattle (Miyamoto
This might reflect the susceptibility of CL in lactating dairy et al., 2000; Skarzynski et al., 2000, 2003) during the mid
cows for apoptosis, compared to dairy heifers and also evi- lueal phase of the estrous cycle. However, the present study
dent from the expression of apoptotic genes in the present did not reveal any difference in the expression of luteal
study. Furthermore, it is shown that nitric oxide mediated TNF mRNA on day 10 between heifers and lactating dairy
inhibition of apoptosis in rat pre-ovulatory granulosa cells cows.
is associated with an induction of HSPA1A (Yoon et al., Timing of post-ovulatory P4 rise, luteal survival and
2002). continuous P4 production are important for the establish-
Steroid biosynthesis in CL is controlled by the expres- ment and maintenance of pregnancy rather than the final
sion of various factors, mainly by StAR, CYP11A1, and concentrations of P4 during pre-implantation development
3␤HSD, which are recognized as luteal markers (Stocco et (Goff, 2004; Inskeep, 2004; Robinson et al., 2005). In the
al., 2007). In the present study, there were greater amounts present study, the P4 concentrations during the mid luteal
of 3␤HSD mRNA in CL of heifers compared to that of lac- phase did not differ between dairy heifers and lactating
tating dairy cows, which suggests an enhanced capability dairy cows. Optimal P4 concentration for enhancing PR in
of P4 production by heifer CL. A gradual decline in amounts cattle is not well established (Nogueira et al., 2004); not
of 3␤HSD mRNA will lead to a decrease in P4 production only sub-optimal concentrations of P4 can cause luteol-
which in turn could trigger the release of intraluteal PGF2␣ ysis but greater concentrations during early luteal phase
(Stocco and Deis, 1998; Arosh et al., 2004). Casey et al. can also trigger premature release of PGF2␣ and luteoly-
(2005) have demonstrated a sharp reduction in the amount sis (Nogueira et al., 2004). Furthermore, P4 concentration
of StAR, CYP11A1, and 3␤HSD mRNA in regressed CL when during the previous estrous cycle can affect the embryo
compared to non-regressed CL of same stage. Even though, survival of the preceding cycle (Shrestha et al., 2004).
not significant, the amount of mRNA for StAR and CYP11A1 Factors such as feed intake and milk yield can alter the
in the present study tends to be less in lactating dairy cows metabolism of P4 (McCann and Hansel, 1986; Wiltbank et
(P = 0.15 and 0.20, respectively). al., 2000; Rabiee et al., 2001) and thereby the peripheral
Angiogenesis plays an important role in the formation, P4 concentrations might differ considerably from the local
maintenance, and function of CL and several angiogenic fac- concentrations needed for the establishment and mainte-
tors are involved in this process including VEGF, FGF2, and nance of pregnancy.
T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224 223

5. Conclusion Goravanahally, M.P., Salem, M., Yao, J., Inskeep, E.K., Flores, J.A., 2009.
Differential gene expression in the bovine corpus luteum during
transition from early phase to midphase and its potential role in acqui-
From the present study, it is concluded that mRNA sition of luteolytic sensitivity to prostaglandin F2 alpha. Biol. Reprod.
of genes in the mid luteal phase CL differs between 80, 980–988.
dairy heifers and lactating dairy cows. Genes regulat- Gustafsson, H., Larsson, K., 1985. Embryonic mortality in heifers after arti-
ficial insemination and embryo transfer-differences between virgin
ing angiogenic, steroidogenic, and luteotropic factors are and repeat breeding heifers. Res. Vet. Sci. 39, 271–274.
highly expressed in heifers compared to lactating dairy Inskeep, E.K., 2004. Preovulatory, postovulatory, and postmaternal recog-
cows, whereas apoptosis seemed to be more evident nition effects of concentrations of progesterone on embryonic survival
in the cow. J. Anim. Sci. 82 (E. Suppl.), E24–E39.
in CL of lactating cows. Collectively these findings sug- Korsmeyer, S., 1995. Regulators of cell death. Trends Genet. 11, 101–105.
gest that CL of second/third parity lactating dairy cows Lamming, G.E., Darwash, A.O., Back, H.L., 1989. Corpus luteum func-
is compromised in luteotropic as well as steroidogenic tion in dairy cows and embryo mortality. J. Reprod. Fertil. Suppl. 37,
245–252.
capacities on day 10 of the estrous cycle. However, the post-
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data
transcription regulation of aforementioned factors should using real-time quantitative PCR and the 2-[delta] [delta] CT method.
be further studied in order to determine whether they have Methods 25, 402–408.
a role in the reduction of PR observed in lactating dairy Lopez, H., Satter, L.D., Wiltbank, M.C., 2004. Relationship between level of
milk production and estrous behavior of lactating dairy cows. Anim.
cows. Reprod. Sci. 81, 209–223.
Lucy, M.C., 2001. Reproductive loss in high-producing dairy cattle: where
will it end? J. Dairy Sci. 84, 1277–1293.
Acknowledgements Lucy, M.C., 2007. Fertility in high-producing dairy cows: reasons for
decline and corrective strategies for sustainable improvement. Soc.
The authors would like to acknowledge the support Reprod. Fertil. Suppl. 64, 237–254.
Mann, G.E., Lamming, G.E., 2001. Relationship between the maternal
provided by Natural Science and Engineering Council of endocrine environment, early embryo development and the inhi-
Canada (NSERC), Western Canada Genetic Centre (WEST- bition of the luteolytic mechanism in the cow. Reproduction 121,
GEN) and British Columbia Investment Agriculture, Canada 175–180.
Mann, G.E., Lamming, G.E., Fray, M.D., 1995. Plasma oestradiol during early
for this study. pregnancy in the cow and the effects of treatment with buserelin.
Anim. Reprod. Sci. 37, 121–131.
McCann, J.P., Hansel, W., 1986. Relationship between insulin and glucose
References metabolism and pituitary ovarian functions in fasted heifers. Biol.
Reprod. 34, 630–641.
Aali, M., Small, J.A., Giritharan, G., Ramakrishnappa, N., Cheng, K.M., McPherson, L.A., Van Kirk, E.A., Murdoch, W.J., 1993. Localization of stress
Rajamahendran, R., 2004. In vitro assessment of corpus luteum func- protein-70 in ovine corpora lutea during prostaglandin-induced lute-
tions in cattle following Ovsynch and CIDR ovulation synchronization olysis. Prostaglandins 46, 433–440.
protocols. Can. J. Anim. Sci. 84, 721–724. Miyamoto, A., Okuda, K., Schweigert, F.J., Schams, D., 1992. Effects of basic
Arosh, J.A., Banu, S.K., Chapdelaine, P., Madore, E., Sirois, J., Fortier, M.A., fibroblast growth factor, transforming growth factor-␤ and nerve
2004. Prostaglandin biosynthesis, transport, and signaling in corpus growth factor on the secretory function of the bovine corpus luteum
luteum: a basis for autoregulation of luteal function. Endocrinology in vitro. J. Endocrinol. 135, 103–114.
145, 2551–2560. Miyamoto, Y., Skarzynski, D.J., Okuda, K., 2000. Is tumor necrosis factor
Balendran, A., Gordon, M., Pretheeban, T., Singh, R., Perera, R., Rajamahen- alpha a trigger for the initiation of endometrial prostaglandin F2 alpha
dran, R., 2008. Decreased fertility with increasing parity in lactating release at luteolysis in cattle? Biol. Reprod. 62, 1109–1115.
dairy cows. Can. J. Anim. Sci. 88, 425–428. Murakami, S., Miyamoto, Y., Skarzynski, D.J., Okuda, K., 2001. Effects of
Beere, H.M., 2004. The stress of dying: the role of heat shock proteins in tumor necrosis factor-alpha on secretion of prostaglandin E2 and F2
the regulation of apoptosis. J. Cell Sci. 117, 2641–2651. alpha in bovine endometrium throughout the estrous cycle. Theri-
Beere, H.M., 2005. Death versus survival: functional interaction between ogenology 55, 1667–1678.
the apoptotic and stress-inducible heat shock protein pathways. J. Narayansingh, R.M., Senchyna, M., Vijayan, M.M., Carlson, J.C., 2004.
Clin. Invest. 115, 2633–2639. Expression of prostaglandin G/H synthase (PGHS) and heat shock
Binelli, M., Hampton, J., Buhi, W.C., Thatcher, W.W., 1999. Persistent dom- protein-70 (HSP-70) in the corpus luteum of prostaglandin F2␣
inant follicle alters pattern of oviductal secretory proteins from cows treated immature superovulated rats. Can. J. Physiol. Pharmacol. 82,
at estrus. Biol. Reprod. 61, 127–134. 363–371.
Bufalo, D.D., Trisciuoglio, D., Scarsella, M., Zangemeister-Wittke, U., Zupi, National Research Council, 2001. Nutrient Requirements of Dairy Cattle,
G., 2003. Treatment of melanoma cells with a bcl-2/bcl-xL anti- 7th ed. National Academy of Science, Washington, DC.
sense oligonucleotide induces antiangiogenic activity. Oncogene 22, Neuvians, T.P., Pfaff, M.W., Berisha, B., Schams, D., 2003. The mRNA expres-
8441–8447. sion of the members of the IGF-system in bovine corpus luteum during
Butler, W.R., Calaman, J.J., Beam, S.W., 1996. Plasma and milk urea nitrogen induced luteolysis. Domest. Anim. Endocrinol. 25, 359–372.
in relation to pregnancy rate in lactating dairy cattle. J. Anim. Sci. 74, Niswender, G.D., Juengel, J.L., Silva, P.J., Rollyso, M.K., McIntush, E.W.,
858–865. 2000. Mechanisms controlling the function and life span of the corpus
Canadian Council on Animal Care, 1993. Guide to the Care and Use of luteum. Physiol. Rev. 80, 1–29.
Experimental Animals, vol. 1., 2nd ed. CCAC, Ottawa, ON. Nogueira, G.M.F., Melo, S.D., Carvalho, M.L., Fuck, E.J., Trinca, L.A., Barros,
Casey, O.M., Morris, D.G., Powell, R., Sreenan, J.M., Fitzpatrick, R., 2005. C.M., 2004. Do high progesterone concentrations decrease pregnancy
Analysis of gene expression in non-regressed and regressed bovine rates in embryo recipients synchronized with PGF2a and eCG? Theri-
corpus luteum tissue using a customized ovarian cDNA array. Theri- ogenology 61, 1283–1290.
ogenology 64, 1963–1976. Okuda, K., Sakumoto, R., 2006. Regulation of uterine function by cytokines
Chomczynski, P., Sacchi, N., 1987. Single-step method of RNA isolation by in cows: Possible actions of tumor necrosis factor-alpha, interleukin-1
acid guanidinium thiocyanate–phenol–chloroform extraction. Anal. alpha and interferon-tau. Anim. Sci. J. 77, 266–274.
Biochem. 162, 156–159. Opsomer, G., Coryn, M., Deluyker, H., de Kruif, A., 1998. An analysis of
Didelot, C., Lanneau, D., Brunet, M., Joly, A.L., de Thonel, A., Chiosis, G., ovarian dysfunction in high yielding dairy cows after calving based
Garrido, C., 2007. Anti cancer therapeutic approaches based on intra- on progesterone profiles. Reprod. Domest. Anim. 33, 193–204.
cellular and extracellular heat shock proteins. Curr. Med. Chem. 14, Pardo, O.E., Arcaro, A., Salerno, G., Raguz, S., Downward, J., Seckl, M.J., 2002.
2839–2847. Fibroblast growth factor-2 induces translational regulation of Bcl-XL
Dimmeler, S., Zeiher, A.M., 1997. Nitric oxide and apoptosis. Nitric Oxide and Bcl-2 via a MEK-dependent pathway: correlation with resistance
1, 275–281. to etoposide-induced apoptosis. J. Biol. Chem. 277, 12040–12046.
Goff, K.A., 2004. Steroid hormone modulation of prostaglandin secretion Pretheeban, T., Gordon, M., Singh, R., Rajamahendran, R., 2009. Endome-
in the ruminant endometrium during the estrous cycle. Biol. Reprod. trial gene and protein expressions in dairy cows and heifers. Biol.
71, 11–16. Reprod. 81 (1 Suppl.), 305.
224 T. Pretheeban et al. / Animal Reproduction Science 121 (2010) 218–224

Pursley, J.R., Wiltbank, M.C., Stevenson, J.S., Ottbre, J.S., Garverick, H.A., response to tumor necrosis factor alpha: cell type specificity and
Anderson, L.L., 1997. Pregnancy rates per artificial insemination for intracellular mechanisms. Biol. Reprod. 62, 1116–1120.
cows and heifers inseminated at a synchronized ovulation or synchro- Spencer, T.E., Johnson, G.A., Bazer, F.W., Burghardt, R.C., Palmarini, M.,
nized estrus. J. Dairy Sci. 80, 295–300. 2007. Pregnancy recognition and conceptus implantation in domestic
Rabiee, A.R., Macmillan, K.L., Schwarzenberger, F., 2001. Excretion rate of ruminants: roles of progesterone, interferons and endogenous retro-
progesterone in milk and faeces in lactating dairy cows with two levels viruses. Reprod. Fertil. Dev. 19, 65–78.
of milk yield. Reprod. Nutr. Dev. 41, 309–319. Stocco, C., Telleria, C., Gibori, G., 2007. The molecular control of cor-
Reynolds, L.P., 1986. Utero-ovarian interactions during early pregnancy: pus luteum formation, function, and regression. Endocr. Rev. 28,
role of conceptus-induced vasodilation. J. Anim. Sci. 62, 47–61. 117–149.
Robinson, R.S., Hammond, A.J., Hunter, M.G., Mann, G.E., 2005. The induc- Stocco, C.O., Deis, R.P., 1998. Participation of intraluteal progesterone
tion of a delayed post-ovulatory progesterone rise in dairy cows: a and prostaglandin F2␣ in LH-induced luteolysis in pregnant rat. J.
novel model. Domest. Anim. Endocrinol. 28, 285–295. Endocrinol. 156, 253–259.
Rueda, B.R., Tilly, K.I., Hansen, T.R., Jolly, P.D., Hoyer, P.B., Tilly, J.L., 1997. Sugino, N., Suzuki, T., Kashida, S., Karube, A., Takiguchi, S., Kato, H.,
Increased Bax and interleukin-1b-converting enzyme messenger RNA 2000. Expression of Bcl-2 and Bax in the human corpus luteum
levels coincide with apoptosis in the corpus luteum during structural during the menstrual cycle and in early pregnancy: regulation
regression. Biol. Reprod. 56, 186–193. by human chorionic gonadotropin. J. Clin. Endocrinol. Metab. 85,
Sartori, R., Sartor-Bergfelt, R., Mertens, S.A., Guenther, J.N., Parrish, J.J., 4379–4386.
Wiltbank, M.C., 2002. Fertilization and early embryonic development Tanikawa, M., Acosta, T.J., Fukui, T., Murakami, S., Korzekwa, A., Skarzynski,
in heifers and lactating cows in summer and lactating and dry cows D.J., Park, C.K., Okuda, K., 2005. Regulation of prostaglandin synthe-
in winter. J. Dairy Sci. 85, 2803–2812. sis by interleukin-1 alpha in bovine endometrium during the estrous
Schams, D., Berisha, B., 2002. Angiogenic factors (VEGF, FGF and IGF) in cycle. Prostaglandins Other Lipid Mediat. 78, 279–290.
the bovine corpus luteum. J. Reprod. Dev. 48, 233–242. Tilly, J.L., 1996. Apoptosis and ovarian function. Rev. Reprod. 1, 162–172.
Shrestha, H.K., Nakao, T., Higaki, T., Suzuki, S., Akita, M., 2004. Resump- Townson, H.D., 2006. Immune cell-endothelial cell interactions in the
tion of postpartum ovarian cyclicity in high-producing Holstein cows. bovine corpus luteum. Integr. Comp. Biol. 46, 1055–1059.
Theriogenology 61, 637–649. Wiltbank, M.C., Fricke, P.M., Sangsritavong, S., Sartori, R., Ginther, O.J.,
Skarzynski, D.J., Bah, M.M., Deptula, K.M., Woclawek-Potocka, I., 2000. Mechanisms that prevent and produce double ovulation in dairy
Korzekwa, A., Shibaya, M., Pilawski, W., Okuda, K., 2003. Roles of tumor cattle. J. Dairy Sci. 83, 2998–3007.
necrosis factor-␣ of the estrous cycle in cattle: an in vivo study. Biol. Wolfenson, D., Inbar, G., Roth, Z., Kaim, M., Bloch, A., Braw-Tal,
Reprod. 69, 1907–1913. R., 2004. Follicular dynamics and concentrations of steroids and
Skarzynski, D.J., Majewska, M., Bah, M.M., Hapunik, J., Piotrowska, K.K., gonadotropins in lactating cows and nulliparous heifers. Theriogenol-
Okuda, K., 2008. Is interleukin 1␣ a luteotropic or luteolytic agent in ogy 62, 1042–1055.
cattle? Biol. Reprod. 78, 60. Yoon, S.J., Choi, K.H., Lee, K.A., 2002. Nitric oxide-mediated inhibition of
Skarzynski, D.J., Miyamoto, Y., Okuda, K., 2000. Production of follicular apoptosis is associated with HSP70 Induction and Bax sup-
prostaglandin F2 alpha by cultured bovine endometrial cells in pression. Mol. Reprod. Dev. 61, 504–510.
Animal Reproduction Science 121 (2010) 225–235

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Expression analysis of circadian genes in oocytes and preimplantation


embryos of cattle and rabbits
Tomoko Amano a,∗ , Kaori Tokunaga a , Reiko Kakegawa a , Ayaka Yanagisawa a ,
Atsushi Takemoto a , Atsuhiro Tatemizo a , Tatsuya Watanabe a , Yuki Hatanaka a ,
Akinori Matsushita a , Masao Kishi b , Masayuki Anzai b , Hiromi Kato b , Tasuku Mitani b ,
Satoshi Kishigami a , Kazuhiro Saeki a , Yoshihiko Hosoi a ,
Akira Iritani a,b , Kazuya Matsumoto a
a
Department of Genetic Engineering, College of Biology-Oriented Science and Technology, Kinki University, 930 Nishimitani, Kinokawa, Wakayama
649-6493, Japan
b
Institute of Advanced Technology, Kinki University, 14-1 Akasaka, Kainan, Wakayama 642-0017, Japan

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

Article history: We previously showed that circadian genes clock, bmal1, cry1, cry2, per1, and per2
Received 5 January 2010 are expressed and function as maternal mRNA regulating events in the oocytes and
Received in revised form 14 May 2010
preimplantation embryos of mice. Recent evidence indicates however that either or both
Accepted 27 May 2010
expression profiles of circadian genes in some tissues, and transcript sequences of circadian
Available online 4 June 2010
genes, differ to generate the physiological differences between diurnal and nocturnal
species. We therefore investigated the expression profiles of circadian genes in oocytes
Keywords:
and preimplantation embryos of species other than mice, namely cattle and rabbits,
Circadian genes
Oocytes representing diurnal and nocturnal species, respectively, and determined the protein
Preimplantation embryos sequences of circadian genes in these species. Quantitative real-time PCR revealed that all
Rabbits circadian genes considered in this study were present in the oocytes and preimplantation
Cattle embryos of both species, and the transcript amounts of clock, cry1 and per1 contained in
oocytes were significantly higher than in preimplantation embryos of both species. The
transcripts of clock, cry1, and per1 of cattle and rabbits were determined by primer walking,
and functional domains in the estimated amino acid sequences were compared between
cattle and rabbits and with those of humans and mice. The sequences of clock, cry1, and
per1 in cattle and rabbits closely resembled those in mice (85–100% homologies), and no
difference based on diurnality or nocturnality was observed. These findings suggest that
circadian genes in the oocytes and preimplantation embryos of mammals fulfill the same
functions across species as maternal mRNA.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction tors that regulate the circadian clock in mammals (Dunlap,


1999; Reppert and Weaver, 2001). They are expressed
Circadian genes (clock, bmal11, cry1, cry2, per1, and in most organs, tissues, and cells and are classified
per2) are known to encode transcription regulation fac- into two groups: transcription-promoting factors CLOCK
and BMAL1; and transcription-suppression factors CRY1,
CRY2, PER1, and PER2. Since the transcription activity of
∗ Corresponding author. Tel.: +81 0736 77 0345.
each group alternatively oscillates, peaking nearly every
E-mail addresses: amino@waka.kindai.ac.jp,
12 h, numerous genes that have their transcription reg-
amano@waka.kindai.ac.jp (T. Amano). ulated by circadian genes are repetitively transcribed

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.05.020
226 T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235

every 24 h, thereby regulating many circadian physiologi- tion embryos may different between species, especially
cal phenomena (Dunlap, 1999; Reppert and Weaver, 2001; between that of diurnal and nocturnal species, and
Albrecht, 2007). Moreover, the transcription rhythms of these genes may be involved in the physiological differ-
circadian genes in most parts of the body are synchro- ences in oocytes and preimplantation embryos of each
nized with those in the suprachiasmatic nucleus (SCN), species. These possibilities warrant investigation into the
which converts light/dark information transmitted by expression profiles of circadian genes in the oocytes and
the optic nerve from the environment into transcrip- preimplantation embryos of other nocturnal and diurnal
tion oscillations of circadian genes, although the factors species.
involved in this synchronization have not been fully clar- In fact, the species differences in the oocyte maturation
ified (Dunlap, 1999; Reppert and Weaver, 2001; Albrecht, time and the development of preimplantation embryos
2007). from the 1-cell to blastocyst stage are well known (Hosoi
Studies to date have suggested that the transcription et al., 1981; Dominko and First, 1997; Polanski et al., 1998;
rhythms of circadian genes in diurnal and nocturnal species Latham et al., 1999). These differences explain the inter-
are the same in the SCN but differ in peripheral organs, est in identifying maternal mRNAs that are differently
tissues, and cells. These differing peripheral transcription expressed between the oocytes and blastocysts of each
patterns are thought to be one of the basic mechanisms species and able to modulate the speed of cell-cycle pro-
underlying the physiological differences between noctur- gression, which is an important factor in regulating the
nal and diurnal species (Smale et al., 2003; Mahoney speed of oocyte maturation and preimplantation embryos
et al., 2004; Schwartz et al., 2004; Doyle et al., 2008; development. Since the circadian genes are reported to be
Yan et al., 2008; Chappell et al., 2009). For example, the differently expressed between the peripheral organs, tis-
transcript quantities of bmal1 and per 2 in the skele- sues, and cells of diurnal and nocturnal species (Smale et
tal muscle of nocturnal and diurnal species peak at al., 2003; Mahoney et al., 2004; Schwartz et al., 2004; Doyle
opposite times of the day (Yan et al., 2008). Moreover, et al., 2008; Yan et al., 2008; Chappell et al., 2009) and
although the transcription patterns of circadian genes in to play a role in the modulation of cell cycle progression,
the SCN in the nocturnal lab-rat species Rattus norvegi- including meiosis (Matsuo et al., 2003; Nagoshi et al., 2004;
cus and diurnal grass-rat species Arvicanthis niloticus are Amano et al., 2009), mRNAs of circadian genes appear to
the same, the daily rhythm in activity of their GnRH neu- be good candidates for maternal mRNAs that are differ-
rons, which is controlled by circadian genes transcribed ently expressed between the oocytes and blastocysts of
within the GnRH neurons themselves, shows a phase dif- each species and able to modulate the speed of cell-cycle
ference of 12 h (Mahoney et al., 2004; Chappell et al., progression.
2009). Further, it has been shown that a point mutation in the
In mammalian ovaries, when the follicle is recruited CSNK1E-binding domain of per2 brings forward the timing
for growth, the oocytes in the follicle undergo a burst of of the sleep phase in humans, suggesting that inter-species
transcription and translation activity. Some of the tran- differences in the timing of daily active and inactive phases
scripts transcribed are long-lived and accumulate in the may be explained by differences in the functional domain
oocytes as maternal mRNA. This maternal mRNA is impor- sequences of proteins derived from circadian genes (Toh
tant for promoting the subsequent processes of meiosis et al., 2001; Ebisawa, 2007). Although this also suggests
and development of preimplantation embryos (Gosden, the importance of considering differences between diurnal
2002; Lonergan et al., 2003; Mtango et al., 2008). More- and nocturnal species in protein sequences derived from
over, maternal mRNA is abundant in immature oocytes circadian genes when assessing for universal functionality
(GV oocytes), but begins to decrease due to consumption of these genes in the oocytes and preimplantation embryos
with the start of oocyte maturation and is almost com- of mammals, the source animals used to obtain the protein
pletely depleted by the blastocyst stage (Nothias et al., sequences derived from circadian genes are often limited
1995; Latham, 1999; Gosden, 2002; Hamatani et al., 2004, to common experimental animals, which are commonly
2008). nocturnal species such as mice and rats. Sequence infor-
In a previous study, we found transcripts of circadian mation of circadian genes provided by on-line databases is
genes in the oocytes and preimplantation embryos of mice, therefore limited.
although their quantities did not oscillate every 24 h like In this study, to compare differences between diurnal
that observed in other cells (Amano et al., 2009). We and nocturnal species in expression profiles of circa-
also found that the expression patterns of some circadian dian genes in oocytes and preimplantation embryos, we
genes, such as clock, bmal1, cry1, and per1, in the oocytes investigated the expression profiles of circadian genes
and preimplantation embryos of mice are consistent with in the oocytes and preimplantation embryos of cattle
those of maternal mRNA, such as an abundance of these and rabbits, representing diurnal and nocturnal mam-
transcripts in GV oocytes and the subsequent decrease in mals, respectively (Gonyou and Stricklin, 1984; Linnane et
transcript amounts with the beginning of oocyte matura- al., 2001). Results were compared with those previously
tion, and involvement of cry1 in oocyte maturation (Amano analyzed for mice. We also determined the amino acid
et al., 2009). sequences of the functional domains of proteins derived
Although these findings suggest circadian genes play a from circadian genes in cattle and rabbits, which have
role in the oocyte and early preimplantation embryo other not been available in on-line databases, using transcript
than regulation of the circadian rhythm, the expression sequences, and compared them to those of humans and
profiles of circadian genes in oocytes and preimplanta- mice.
T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235 227

2. Materials and methods Briefly, cattle ovaries were collected at a local abattoir
and transported to the laboratory within 1 h after col-
2.1. Media lection in a sterile saline solution (0.85% NaCl) kept at
20–25 ◦ C. COCs were aspirated from antral follicles 2–8 mm
For cattle, 25 mM Hepes-buffered TCM199 medium in diameter using a 21-gauge needle attached to a 10-
with Hanks’ salts (H199-H; Gibco, Invitrogen Life Tech- ml disposable syringe and then pooled in a 15-ml test
nologies, Tokyo, Japan) supplemented with 5% (v/v) FBS tube placed in a water bath at 37 ◦ C. The sediment in
was used to handle the cumulus-oocyte complexes (COCs), the test tube was collected and put into a 35-mm dis-
oocytes, and preimplantation embryos in air (handling posable dish containing handling medium, and the COCs
medium). Twenty-five millimolar Hepes-buffered TCM199 were collected by a capillary tube attached to a rubber
medium to which 5% (v/v) FBS, 0.5 mM sodium pyru- tube operated by mouth suction. For the collection of
vate, 25 mg/ml gentamicin, 0.02 Armour unit (AU)/ml FSH immature oocytes, 60 COCs were collected and the oocytes
(Antrin; Kawasaki Seiyaku, Tokyo, Japan), and 1 mg/ml were detached from the cumulus cells by vigorous pipet-
estradiol-17b were added was used for the maturation ting. The immature oocytes were then separated into 3
of oocytes (maturation medium). Brackett and Oliphant groups of 20 each, and each group was collected into a
medium (Brackett and Oliphant, 1975) excluding glu- 1.5-ml disposable tube. The tubes were then frozen with
cose and supplemented with 10 mg/ml heparin was used liquid nitrogen immediately after collection and stored at
for in vitro fertilization (fertilization medium). SOFaa −80 ◦ C until mRNA extraction. For production of mature
medium from which potassium phosphate monobasic was oocytes, COCs collected from ovaries were separated into
removed was used for the development of the preim- groups of 10 each and put into a 50-␮l drop of matu-
plantation embryos (development medium) (Tervit et ration medium covered with mineral oil. The maturation
al., 1972; Takahashi and First, 1992; Saeki et al., 1998). media containing COCs were then cultured for 21 h in
The handling medium was warmed before use to 39 ◦ C, an incubator (humidified atmosphere of 5% CO2 in air at
while the maturation and fertilization media were incu- 39 ◦ C), and the cumulus cells of COCs were subsequently
bated before use in CO2 (humidified atmosphere of 5% detached from the oocytes by vigorous pipetting and
CO2 in air at 37 ◦ C) and CO2 /O2 (humidified atmosphere hyaluronidase treatment. For mRNA extraction, 60 oocytes
of 5% CO2 , 5% O2 and 90% N2 set at 38 ◦ C) incubators, that had first polar bodies were collected as matured
respectively. oocytes, and the matured oocytes were separated into 3
For rabbits, M2 medium (Quinn et al., 1982) was groups of 20 each and collected into 1.5-ml disposable
used to handle the COCs, oocytes, and preimplanta- tubes. The tubes were then frozen in liquid nitrogen imme-
tion embryos in air (handling medium). Brackett and diately after collection and stored at −80 ◦ C until mRNA
Oliphant medium to which 3 mg/ml bovine serum albu- extraction.
min (BSA; A-7638; Sigma-Aldrich, St. Louis, MO) was added
was used for in vitro fertilization (fertilization medium). 2.2.3. Production of cattle preimplantation embryos
Three milligrams per milliliter BSA-supplemented TCM In vitro fertilization and culturing of preimplanta-
199 medium (680557; Nissui Pharmaceutical, Tokyo, tion embryos was performed according to a previously
Japan) was used for the development of the preimplan- described method with slight modifications (Saeki et al.,
tation embryos (development medium). The handling 1998). Briefly, approximately 200 mature oocytes obtained
medium was warmed before use to 38 ◦ C, while the fer- by the above methods were separated into groups of 20–25
tilization developmental media were incubated before each, and each group was put into a 50 ␮l drop of fer-
use in CO2 (humidified atmosphere of 5% CO2 in air tilization medium covered with mineral oil. The drops
at 37 ◦ C) and CO2 /O2 (humidified atmosphere of 5% of fertilization medium containing matured oocytes were
CO2 , 5% O2 and 90% N2 set at 39 ◦ C) incubators, placed in a CO2 incubator (humidified atmosphere of 5%
respectively. CO2 in air at 37 ◦ C) until insemination. Cattle spermatozoa
stored in a storage straw in liquid nitrogen were thawed in
2.2. Collection of oocytes and preimplantation embryos a water bath set at 37 ◦ C, put on a discontinuous gradient
from cattle and rabbits percoll solution (Amersham Biosciences, Uppsala, Sweden;
Pousette et al., 1986) in a 15-ml conical tube, and cen-
2.2.1. Animals and their care trifuged at 1900 rpm for 30 min. After centrifugation, the
Sexually mature male and female New Zealand White supernatant containing percoll solution and spermatozoa
and Japanese White rabbits (1–2 years old) were individ- of low fertilization ability was removed, and the remaining
ually housed under a lighting schedule with 14 h of light spermatozoa sediment was suspended into 10 ml of fer-
(08:00–22:00) followed by 10 h of darkness. All proce- tilization medium. The spermatozoa concentration in the
dures involving animals conformed to the Kinki University suspension was adjusted by addition of an adequate vol-
Guidelines for the Care and Use of Laboratory Animals. ume of fertilization medium, and the final concentration of
spermatozoa in the drops of fertilization medium contain-
2.2.2. Collection of immature oocytes from cattle and in ing COCs was adjusted to 2.0 × 106 /ml by addition of 50 ␮l
vitro maturation of suspension. The time of spermatozoa addition to the fer-
Collection of immature oocytes from cattle and in vitro tilization medium was set as 0 h post-insemination (hpi).
production of mature oocytes was performed according The co-culture of COCs and spermatozoa was continued for
to a previously described method (Saeki et al., 1998). 9 h, the time required for 60% of oocytes to be fertilized.
228 T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235

After co-culture, the spermatozoa and cumulus cells were 2.2.5. Production of rabbit preimplantation embryos
put into 10 ml of developmental medium in a 15-ml coni- Production of rabbit preimplantation embryos was per-
cal tube. The tube was vortexed to remove the spermatozoa formed according to previously described methods with
and cumulus cells from the oocytes, and the oocytes, now slight modifications (Hosoi et al., 1981). Briefly, 9 female
defined as 1-cell embryos, were subsequently collected. rabbits were injected with 80 IU PMSG at 19:00 and 60 IU of
The 1-cell embryos were then separated into groups of 20 hCG was injected 72 h later. The rabbits were sacrificed 14 h
each, 3 of which were separately put into 1.5-ml disposable thereafter, and 107 COCs were obtained from the fallopian
tubes immediately after collection, frozen in liquid nitro- tubes. The COCs were separated into groups of 20–22 each
gen, and stored at −80 ◦ C until mRNA extraction. Each of and put into 50-␮l drops of fertilization medium covered
the remaining groups was transferred to 100 ␮l of devel- with mineral oil. The fertilization medium drops contain-
opmental medium covered with mineral oil and incubated ing COCs were incubated in a CO2 incubator (humidified
for a determined time in an incubator (humidified atmo- atmosphere of 5% CO2 in air at 37 ◦ C) until insemination.
sphere of 5% CO2 , 5% O2 and 90% N2 at 39 ◦ C). Incubation of Approximately 300 ␮l of sperm was collected from the
1-cell embryos was continued until 30, 36, 60, 84, 144, and epididymis of a male rabbit and suspended in 5 ml of fertil-
168 hpi, at which time respective 2-cell embryos, 4-cell ization medium in a 15-ml disposal tube. The suspension
embryos, early 8-cell embryos, late 8-cell embryos, com- was then centrifuged to remove the supernatant contain-
pacted morulae (CM), and blastocysts (BL) were collected ing seminal plasma, and after removal of the supernatant,
and accordingly named. 5 ml of fertilization medium was added to the remaining
Embryos of a certain developmental stage at each time- sediment to suspend the spermatozoa. The spermatozoa
point were collected from 2 or 3 drops of developmental in suspension were incubated in a CO2 incubator (humid-
medium, and the drops that had been used for the collec- ified atmosphere of 5% CO2 in air at 37 ◦ C) for 10 h to
tion of the embryos were discarded. The embryos collected induce spermatozoa capacitation. The spermatozoa con-
at each time-point were put into 1.5-ml disposable tubes, centration in the suspension was adjusted by addition of
frozen with liquid nitrogen immediately after collection, an adequate volume of fertilization medium, and the final
and the tubes were then stored at −80 ◦ C until mRNA concentration of spermatozoa in the drops of fertilization
extraction. The in vitro fertilization and culture procedure medium containing COCs was adjusted to 1.5 × 106 /ml by
was repeated until 60 embryos were collected from each addition of 50 ␮l of suspension. The drops containing both
post-insemination time-point. COCs and spermatozoa were incubated in an incubator
(humidified atmosphere of 5% CO2 at 37 ◦ C) for 6 h, and
2.2.4. Collection of immature and matured oocytes from then the oocyte-attached cumulus cells and spermatozoa
rabbits were removed by hyaluronidase treatment and vigorous
Collection of immature and matured oocytes from rab- pipetting. Eighty oocytes having two pronuclei were then
bits was performed according to a previously described collected as 1-cell embryos. Groups of 20 were put into 1.5-
method with slight modifications (Hosoi et al., 1981; ml disposable tubes, immediately frozen in liquid nitrogen,
Kanaya et al., 2007). Briefly, for the collection of immature and the tubes were then stored at −80 ◦ C until mRNA
oocytes, 5 female rabbits were injected with 80 IU of preg- collection.
nant mare serum gonadotrophin (PMSG; Sankyo Lifetech)
at 19:00 and sacrificed by intravenous injection of pen- 2.3. mRNA extraction and cDNA synthesis
tobarbital 72 h later. Ovaries were collected immediately
after sacrifice and the COCs were aspirated from antral fol- mRNA extraction and cDNA synthesis were performed
licles 1-5 mm in diameter using a 24-gauge needle attached according to previously described methods with slight
to a 5-ml disposable syringe. Collected COCs were trans- modifications (Amano et al., 2009). Briefly, the mRNA of
ferred to a 35-mm disposable dish containing handling oocytes and preimplantation embryos was extracted using
medium, and the cumulus cells were detached from the a QuickPrep micro mRNA purification kit (GE Healthcare,
oocytes by vigorous pipetting. Sixty oocytes with germinal CT). Four hundred microliters of extraction buffer produced
vesicles were collected as immature oocytes. Groups of 20 by the manufacturer was added to each tube contain-
each were then put into 1.5-ml tubes, frozen in liquid nitro- ing oocytes and preimplantation embryos, and the tubes
gen, and the tubes were then stored at −80 ◦ C until mRNA were vortexed to extract the total RNA. For the tubes used
extraction. in the collection of cattle embryos, 20 ␮l of extraction
For the collection of mature oocytes, 7 female rab- buffer was added per embryo. The total volume of RNA
bits were injected with 80 IU PMSG at 19:00, and 60 IU extract obtained at each post-insemination time-point was
of human chorionic gonadotropin (hCG; Sankyo Lifetech) 1200 ␮l, and the total volume was divided into 3 sets of
was injected 72 h later. The rabbits were then sacrificed 400 ␮l, each containing the total RNA of 20 embryos, and
14 h thereafter, and COCs were obtained from the fal- put into 1.5-ml disposable tubes. The total RNA extract in
lopian tubes. Cumulus cells were detached from oocytes each tube was applied to the column attached to the kit
by hyaluronidase treatment and vigorous pipetting, and to extract the mRNA. The extracted mRNA in each tube
80 oocytes with extruded polar bodies were collected as was incubated with DNase at 37 ◦ C for 30 min to remove
matured oocytes. Groups of 20 mature oocytes each were genomic DNA, and cDNA was then synthesized from the
put into 1.5-ml disposable tubes, immediately frozen in liq- purified mRNA using a SuperScriptTM III First Strand Syn-
uid nitrogen, and the tubes were then stored at −80 ◦ C until thesis Kit with random hexamer (Invitrogen). The total
mRNA collection. volume of each cDNA sample solution was adjusted to 10 ␮l
T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235 229

Table 1
Primers used for real-time quantitative PCR analysis of circadian gene transcripts of cattle and the amplicon characteristics.

Gene NCBI accession number (mouse) Forward or reverse Primer sequence 5 to 3 Amplicon length (bp)

Clock NM 007715 F GCCATGCAGCATCTCAAAGA 83


R CTTAGTTCTTCTTGCCGATCA

Bmal1 NM 007489 F CACTACGGAGTCGGTGGTTCA 74


R GGTGTTCGTGGAGACAATGTATTC

Cry1 NM 007771 F ACCATCCGCTGCGTCTAC 78


R CAAAAATCGCCACCTGTTGA

Cry2 NM 009963 F GCGATACCTGCCTAAACTGAAAG 86


R TGGCTGCCTTCTGAATGGA

Per1 NM 011065 F CCACACTGCAATACGCGCT 101


R TGTCCATGGCACACGGTTC

Per2 NM 011066 F TGCACGAGATTGAGGGCAT 84


R TACAGGGTCTTCCCGGTCAC

using TE, and the obtained cDNA samples were stored at then applied to the corresponding standard curve for that
−20 ◦ C until use in real-time quantitative polymerase chain gene.
reaction (real-time PCR). After each run, the reaction tubes were gradually heated
above the amplicons’ melting temperatures, confirming
2.4. Quantification of circadian gene transcripts in
the specificity of the amplified products. The real-time
oocytes and preimplantation embryos
PCR reaction described above was also performed using
Real-time PCR was performed using SYBR® Premix Ex the series of cDNA samples obtained from one insemina-
TaqTM II (Perfect Real Time; TaKaRa) and an ABI 7300 instru- tion, and the reaction used to plot the standard curve for
ment. Primers amplifying 71–127 bp DNA fragments of the each target gene was performed at every run. To check the
open reading frame (ORF) of each transcript of cattle and amplification efficiency and reproducibility of each run of
rabbit clock, bmal1, cry1, and per1 and cattle cry2 and per2 real-time PCR, slopes and correlation coefficients (R2) were
were designed based on transcript sequences from the calculated for the obtained standard curves. We confirmed
primer walking method described below (Tables 1 and 2). that the slopes of the standard curves were between −3.4
A 0.5 ␮l cDNA sample, which corresponded to one oocyte and −3.6 and the R2 for all standard curves were >0.95. In
or embryo, was added to each reaction tube. The amplifi- addition, to check the reproducibility of the real-time PCR
cation profiles of the target DNA sequences in each tube quantification, quantification reactions of each target gene
were checked by measuring the fluorescence intensity of transcript were performed twice for each cDNA sample in
the dye, which emits fluorescence when intercalated to each run, and we confirmed that the ratio of duplicated
double-stranded DNA and therefore increases with ampli- data obtained from each target gene in the same cDNA
fication of target DNA. To quantify the target DNA sequence sample did not exceed the range of 1:1.7–1:1. To express
in each tube, Ct (threshold cycle), defined as the cycle the data concisely, the duplicated data were averaged and
number at which the increase in fluorescence starts, was these data for each gene from each of the three samples
determined for each tube. Transcript quantities were then of GV oocytes, MII oocytes, and preimplantation embryos
calculated by first measuring the Cts of solutions containing obtained at each collection time-point were further aver-
107 , 106 , 105 , 104 , 103 and 102 copies/␮l of DNA fragment aged and used as the representative amount of transcripts
for each gene. Then, for each gene, the Cts were plotted for each gene (Figs. 1 and 2).
against a log2 scale of each cDNA fragment concentration 2.5. Protein sequence determination for CLOCK, CRY1,
to obtain a standard curve. To quantify the transcript con- and PER1 in cattle and rabbits
centration of each circadian gene, the Ct of each circadian
gene in each cDNA sample from a oocyte or preim- In this study, the amino acid sequences of CLOCK, CRY1,
plantation embryo was measured by real-time PCR and and PER1 for cattle and rabbits were estimated from the

Table 2
Primers used for real-time quantitative PCR analysis of circadian gene transcripts of rabbits and the amplicon characteristics.

Gene NCBI accession number (mouse) Forward or reverse Primer sequence 5 to 3 Amplicon length (bp)

Clock NM 007715 F CCTGCAAATTTACCGATTCCA 71


R TGCTGCATGGCTCCTAATTG

Bmal1 NM 007489 F AGGAGGCAAGAAGATTTTAAATGGA 76


R CTCCTGAGCCTGTCCTGGTAATA

Cry1 NM 007771 F ACCAAGATCCGGGTTTCAGA 82


R AACTGAAGCACTTACACGTTTGGA

Per1 NM 011065 F ACCAAGATCCGGGTTTCAGA 127


R TGTGCCGCGTGGTGAAG
230 T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235

Fig. 1. Expression profiles of circadian genes in the oocytes and preimplantation embryos of cattle. Mean (±SEM) quantity of transcripts of circadian
genes per 1 oocyte or preimplantation embryo of cattle. Abbreviations: GV, GV oocytes; MII, MII oocytes; E8-cell, early 8-cell embryos; L8-cell, late 8-cell
embryos; CM, compacted morulae; BL, blastocysts. Statistical significant differences between each developmental stage are denoted by differing letters
(all differences with P < 0.05).

mRNA sequences by primer walking using a DNASIS sys- white screening method. Competent cells (E-coli JM109
tem (Hitachi Lifescience Solutions, Tokyo, Japan). Briefly, competent cells; TaKaRa) were transformed by plasmid
forward and reverse primers were designed to amplify vectors (pGEM T-easy Vector; Promega, WI), into which the
approximately 500 bp at the 5 end of the ORF of each tran- DNA fragments were inserted. The cells were inoculated
script of clock, cry1 and per1 for cattle and rabbits by PCR on an LB plate containing carbenicilln with X-gal and iso-
based on known mouse clock, cry1, and per1 sequences propyl ␤-D-1-thiogalactopyranoside, and DNA fragments
(NCBI accession numbers NM 007715, NM 007771, and were selected from the white colonies. The sequences of
NM 011065, respectively). cDNA synthesized from total these DNA fragments were determined by the dye ter-
RNA extracted from cattle liver and rabbit kidney were minator method using a Big dye terminator (ABI) and
used as the PCR templates. Ex Taq (TaKaRa, Shiga, Japan) DNA sequencer (3730 DNA Analyzer, ABI), and the consis-
was used as the DNA polymerase and conditions were tency of the DNA-fragments derived from three different
set in accordance with the manufacturer’s instructions. white colonies on the same LB plate was confirmed. A
The amplified DNA fragments were cloned by the blue- forward primer was then designed approximately 500 bp

Fig. 2. Expression profiles of circadian genes in the oocytes and preimplantation embryos of rabbits. Mean (±SEM) quantity of transcripts of circadian
genes per 1 oocytes or preimplantation embryo of rabbits. Abbreviations: GV, GV oocytes; MII, MII oocytes; E8-cell, early 8-cell embryos; L8-cell, late 8-cell
embryos; CM, compacted morulae; BL, blastocysts. Statistical significant differences between each developmental stage are denoted by differing letters
(all differences with P < 0.05).
T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235 231

from the 5 end of each gene transcript ORF. The cor- and that of bmal1 was less than 100 copies/oocyte (Fig. 1).
responding reverse primer was designed approximately Although the levels of clock, cry1, and per1 significantly
500 bp from the 3 end based on the known mouse declined after fertilization (Fig. 1, P < 0.05), no significant
sequences. The sequence determination for clock, cry1, declines were observed in the levels of bmal1, cry2 and per2
and per1 genes in cattle and rabbits was stopped when transcripts in cattle (Fig. 1).
it reached the base corresponding to the 3 end of each
ORF sequence for that of mice. The functional domains of 3.2. Expression profiles of circadian genes in oocytes and
CLOCK, PER1, and CRY1 of cattle and rabbits were esti- preimplantation embryos of rabbits
mated according to that of mice, which are available at
Uniprot (http://www.uniprot.org/) and have the accession Transcripts of clock, bmal1, cry1, and per1 were detected
numbers O08785, O35973, and P97784, respectively. No in the oocytes and 1-cell embryos of rabbits (Fig. 2). Tran-
suitable primers could be designed for rabbit per1 and so scripts of cry1 were abundant in GV and MII oocytes
the last 200–300 bp of the transcript was not identified (approximately 60,000 and 30,000 copies/oocyte) with
(Fig. 3C). the levels 2- to 300-fold that of other circadian genes
(Fig. 2). The transcripts of per1 and clock were also abun-
2.6. Statistical analysis dant in GV and MII oocytes, with 10,000–30,000 and
4000–6000 copies/oocyte, respectively (Fig. 2). In contrast,
Overall differences between groups were determined the level of bmal1 transcripts was low (approximately 200
by one-way ANOVA, and Fisher’s protected LSD multiple copies/oocytes) in GV and MII oocytes (Fig. 2). Although the
comparison test was applied when the results of one-way transcripts of bmal1, cry1 and per1 declined with matura-
ANOVA were significant (P < 0.05). All statistical analyses tion (P < 0.05, Fig. 2), only the transcripts of cry1 declined
were performed using STATVIEW (Abacus Concepts, Pis- after fertilization (Fig. 2, P < 0.05).
cataway, NJ).
3.3. Comparison between human, cattle, rabbit, and
3. Results mouse major functional domains of circadian gene
proteins
3.1. Expression profiles of circadian genes in oocytes and
preimplantation embryos of cattle The transcripts of clock, cry1, and per1 were abundant
in the oocytes and preimplantation embryos of both cattle
Transcripts of clock, bmal1, cry1, cry2, per1, and per2 and rabbits (Figs. 1 and 2) and so we focused on analyzing
were detected in the oocytes and all stages of preimplanta- the respective amino acid sequences of these genes.
tion embryos of cattle (Fig. 1). Transcripts of per1 were most The functional domains sequences were well conserved
abundant in GV and MII oocytes (approximately 20,000 between species, and the sequence homology of cattle
copies/oocyte) with the level 10- to 100-fold that of other and of rabbits to that of mice was 85–100% (Table 3)
circadian genes detected in oocytes (Fig. 1). The next most with no sequences specific to diurnal or nocturnal species
abundant transcripts in GV and MII oocytes were cry1 (Fig. 3A–C). As point mutations at the amino acid residues
and clock, with 3000–4000 and 1000–2000 copies/oocyte, 656, 658, 659, 669, 670, and 672 in the Q-rich domain
respectively (Fig. 1). In contrast, the levels of cry2 and per2 of mouse CLOCK attenuate its transcription-promoting
transcripts in oocytes were less than 1000 copies/oocyte activity by inducing failure of histone acetyltransferase

Table 3
Homologies of functional domains in CLOCK, CRY1 and PER1 of human, cattle and rabbit to those of mice.

a. CLOCK

HLH PAS1 PAS2 PAC Q-rich domain

Human 97.4% 100% 100% 100% 97.0%


Cattle 97.4% 100% 100% 100% 97.6%
Rabbit 97.4% 100% 100% 100% 85.5%

b. CRY1

DNA photolyase superfamily FAD-binding superfamily Region required for inhibition of


CLOCK-BMAL1-mediated transcription

Human 96.6% 100% 100%


Cattle 99.4% 100% 100%
Rabbit 96.0% 98.9% 99.0%

c. PER1

PAS1 PAS2 PAC Nuclear export signal 1 CSNK1E-binding domain Nuclear-localization signal

Human 97.0% 100% 100% 100% 91.4% 83.2%


Cattle 95.6% 100% 100% 100% ND ND
Rabbit 94.1% 97.0% 100% 92.3% 89.5% 88.2%
232 T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235
T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235 233

recruitment (Doi et al., 2006), we compared these amino 6 h (Figs. 1 and 2). Many studies have however shown that
acid residues between species. We found that the 656 until zygotic gene activation (ZGA), which occurs at the 4-
amino acid residue in humans, cattle, and mice, shown as to -8 cell stage in cattle and the 8 to 16 cell stage in rab-
amino acid residue 664 in Fig. 3A, was proline while in that bits, transcription activity is almost totally suppressed. This
of rabbits was alanine. suggests transcripts of circadian genes detected in oocytes
and preimplantation embryos of cattle and rabbits up to
4. Discussion ZGA are present as maternal mRNA and not transcribed
during either or both meiosis and the developmental pro-
Here, we provided evidence that circadian genes cess after fertilization (Nothias et al., 1995; Latham, 1999;
function universally across species as maternal mRNA Memli and First, 2000; Parcheco-Trigon et al., 2002). More-
orchestrating events in the oocytes and preimplantation over, our data showed that transcripts of all circadian genes
embryos of mammals. This indicates that circadian genes considered in this study were almost totally depleted after
may have unique although as-yet unknown roles in the the 8-cell stage in cattle (Fig. 1) and the 2-cell stage in
physiological mechanisms of oocytes and preimplantation mice preimplantation embryos, which are the respective
embryos in mammals. stages that ZGA starts in these species. This suggests circa-
It has been previously shown that transcripts of all cir- dian genes are not transcribed in preimplantation embryos
cadian genes are present in the oocytes of mice, and that of mammals after ZGA and are not involved in circadian
clock, bmal1, cry1, and per1 transcripts are more abun- clock regulation in the preimplantation embryos after ZGA
dant in oocytes and preimplantation embryos up until (Amano et al., 2009).
the 2-cell stage than at subsequent developmental stages. It has previously been shown that bmal1 transcript
These findings are consistent with those observed in the levels are abundant in mouse oocytes and preimplan-
oocytes and preimplantation embryos of cattle and rabbits tation embryos up until the 2-cell stage (approximately
in this study (Figs. 1 and 2). Considering these simi- 20,000 copies/oocyte or embryo) (Amano et al., 2009).
lar expression profiles of circadian genes in the oocytes Here, however, we found levels of these to be very low
and preimplantation embryos of cattle, rabbits, and mice, in the oocytes and preimplantation embryos of all stages
which between them represent both diurnal and nocturnal in cattle and rabbits (50–200 copies/oocyte or embryo,
mammalian species, it is reasonable to assume that circa- Figs. 1 and 2). Further, the relative transcript amounts
dian gene transcripts play a regulatory role universal to for each circadian gene were different in oocytes and
all mammals in the development of oocytes and preim- preimplantation embryos for cattle, rabbits, and mice, with
plantation embryos (Amano et al., 2009). Moreover, it has per1 > cry1 > clock in cattle, cry1 > per1 > clock in rabbits, and
been shown that maternal mRNA important to the devel- clock > bmal1 > cry1 > per1 in mice (Figs. 1 and 2). Although
opment of oocytes and preimplantation embryos, such as the mechanisms underlying and physiological meaning
spindlin, zar1 and gdf9 is abundant in oocytes and then of these differences are not known, several physiological
decreases immediately after fertilization due to rapid con- characteristics of oocytes and preimplantation embryos are
sumption (Oh et al., 1997; Gosden, 2002; Xumei et al., known to differ between species. For example, the timing
2003; Hanrahan et al., 2004). Our results therefore sug- of ZGA during preimplantation embryo development varies
gest that the transcripts of all circadian genes are present among species and is thought to depend on differences in
as maternal mRNA and that those of clock, cry1, and per1, the oocyte and preimplantation embryo maternal mRNA of
which are abundant in oocytes and decrease immedi- each species. This suggests that differing expression pro-
ately after fertilization, are important for the development files of circadian genes in oocytes and preimplantation
of oocytes and preimplantation embryos in cattle and embryos between different species may play a role in deter-
rabbits. mining their different physiological characteristics, such as
In several mammal species, including mice and cat- the timing of ZGA (Nothias, 1995; Latham, 1999).
tle, it has been shown that oocytes and preimplantation Amino acid sequences of each functional domain in
embryos must reach each developmental stage at a cer- CLOCK, CRY1, and PER1 were largely the same for humans,
tain time-point, and those that reach each stage either cattle, rabbits, and mice and there were no sequences par-
before or after this time will not develop further properly. ticularly unique to diurnal or nocturnal species, suggesting
(Dominko and First, 1997; Polanski et al., 1998; Lonergan that the functions of circadian genes in the oocytes and
et al., 2003). For this reason, although quantification of preimplantation embryos of mammals are virtually univer-
transcripts within intervals of at least 6 h is required to sal (Fig. 3 and Table 3). The Q-rich domain in rabbit CLOCK
precisely detect oscillatory transcription that repeats every and the CSNK1E-binding domain and nuclear-localization
24 h, we collected oocytes and preimplantation embryos in signal in PER1 of humans and cattle, however, showed
a stage-specific manner that required intervals longer than low homologies to those of mice (less than 90%) (Fig. 3

Fig. 3. Comparison of the amino acid sequences of proteins derived from circadian genes of humans, cattle, rabbits, and mice. (A) CLOCK amino acid
sequence. Numbered underlined sequences indicate functional domains: 1, HLH domain; 2, PAS domain 1; 3, PAS domain 2; 4, PAC domain; 5, Q-rich
domain. (B) CRY1 amino acid sequence. Numbered underlined sequences indicate functional domains: 1, DNA photolyase superfamily; 2, FAD-binding
superfamily; 3, Region required for inhibition of CLOCK-BMAL1-mediated transcription. (C) PER1 amino acid sequence. Numbered underlined sequences
indicate functional domains: 1, PAS domain1; 2, PAS domain 2; 3, PAC domain; 4, nuclear export signal; 5, CSNK1E-binding domain; 6, nuclear-localization
signal.
234 T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235

and Table 3). These domains are known to regulate the References
transcription activity of circadian gene-regulated genes;
for example, the Q-rich domain in CLOCK recruits his- Akashi, M., Tsuchiya, Y., Yoshino, T., Nishida, E., 2002. Control of intra-
cellular dynamics of mammalian period proteins by casein kinase I
tone acetyltransferase to promote transcription activity epsilon (CKIepsilon) and CKIdelta in cultured cells. Mol. Cell. Biol. 22,
(Doi et al., 2006), and the CSNK1E-binding domain in 1693–1703.
PER1 includes sites regulating the speed of its own degra- Albrecht, U., 2007. Orchestration of gene expression and physiology by
the circadian clock. J. Physiol. Paris. 100, 243–251.
dation depending on its phosphorylation state (Akashi Amano, T., Matsushita, A., Hatanaka, Y., Watanabe, T., Oishi, K., Ishida,
et al., 2002). Further, the nuclear-localization signal in N., Anzai, M., Mitani, T., Kato, H., Kishigami, S., Saeki, K., Hosoi, Y.,
mouse PER1 transfers it to the nucleus, with CRY1 and Iritani, A., Matsumoto, K., 2009. Expression and functional analyses
of circadian genes in mouse oocytes and preimplantation embryos:
2 suppressing transcription activity (Loop et al., 2005).
Cry1 is involved in the meiotic process independently of circadian
In addition, as a mutation at amino acid residue 656 in clock regulation. Biol. Reprod. 80, 473–483.
mouse CLOCK from proline to alanine has been shown to Brackett, B.G., Oliphant, G., 1975. Capacitation of rabbit spermatozoa in
vitro. Biol. Reprod. 12, 260–274.
degrade histone acetyltransferase activity and attenuate
Chappell, P.E., White, R.S., Mellon, P.L., 2009. Circadian gene expression
the transcription-activation ability of mouse CLOCK (Doi et regulates pulsatile gonadotropin-releasing hormone (GnRH) secre-
al., 2006), our results showing alanine was the 656 amino tory patterns in the hypothalamic GnRH-secreting GT1-7cell line. J.
acid residue in rabbit CLOCK may indicate that mouse Neuroendocrinol. 21, 339–345.
Doi, M., Hirayama, J., Sassone-Corsi, P., 2006. Circadian regulator CLOCK
CLOCK has a stronger transcription-promoting ability than is a histone acetyltransferase. Cell 125, 497–508.
that of rabbit. Whether circadian genes are involved in Dominko, T., First, N.L., 1997. Timing of meiotic progression in bovine
transcription suppression in oocytes and preimplantation oocytes and its effect on early embryo development. Mol. Reprod. Dev.
47, 456–467.
embryos remains unclear, and the possibility exists that the Doyle, S.E., Yoshikawa, T., Hillson, H., Menaker, M., 2008. Retinal path-
observed functional domain sequence differences of cir- ways influence temporal niche. Proc. Natl. Acad. Sci. U.S.A. 105,
cadian gene proteins between species might only reflect 12645–12646.
Dunlap, J.C., 1999. Molecular bases for circadian clocks. Cell 96, 271–290.
differences between individual animals used for that anal- Ebisawa, T., 2007. Circadian rhythms in the CNS and peripheral clock dis-
ysis. Nonetheless, variations in these domains may reflect orders: human sleep disorders and clock genes. J. Pharmacol. Sci. 103,
differences in physiological characteristics of oocytes and 150–154.
Gonyou, H.W., Stricklin, W.R., 1984. Diurnal behavior patterns of feedlot
preimplantation embryos between species, such as the
bulls during winter and spring in northern latitudes. J. Anim. Sci. 58,
time required for preimplantation embryos to reach a cer- 1075–1083.
tain developmental stage (Amano et al., 2009). Gosden, R.G., 2002. Oogenesis as a foundation for embryogenesis. Mol.
Cell. Endocrinol. 186, 149–153.
In summary, our results suggest that, rather than act-
Hamatani, T., Carter, M.G., Sharov, A.A., Ko, M.S., 2004. Dynamics of global
ing as regulators of the circadian clock, circadian genes gene expression changes during mouse preimplantation develop-
function universally across species as maternal mRNA ment. Dev. Cell. 6, 117–131.
in mammals. This finding indicates that circadian genes Hamatani, T., Yamada, M., Akutsu, H., Kuji, N., Mochimaru, Y., Takano, M.,
Toyoda, M., Miyado, K., Umezawa, A., Yoshimura, Y., 2008. What can
may have unique although as-yet unknown roles in the we learn from gene expression profiling of mouse oocytes? Reproduc-
physiological mechanisms of oocytes and preimplantation tion 135, 581–592.
embryos. Our results not only provide new information on Hanrahan, J., Gregan, S.M., Mulsant, P., Mullen, M., Davis, G.H., Powell,
R., Galloway, S.M., 2004. Mutations in the genes for oocyte-derived
the functions of maternal mRNA in general, but also shed growth factors GDF9 and BMP15 are associated with both increased
light on the specific functions of circadian genes as mater- ovulation rate and sterility in Cambridge and Belclare sheep (Ovis
nal mRNA. aries). Biol. Reprod. 70, 900–909.
Hosoi, Y., Niwa, K., Hatanaka, S., Iritani, A., 1981. Fertilization in vitro of
rabbit eggs by epididymal spermatozoa capacitated in a chemically
defined medium. Biol. Reprod. 24, 637–642.
Acknowledgements Kanaya, H., Hashimoto, S., Teramura, T., Morimoto, Y., Matsumoto, K.,
Saeki, K., Iritani, A., Hosoi, Y., 2007. Mitochondrial dysfunction of in
vitro grown rabbit oocytes results in preimplantation embryo arrest
We thank Ms. Madoka Ozawa, Ms. Yukihi Mera, Mr. Keiji after activation. J. Reprod. Fertil. 53, 631–637.
Masukura, and Mr. Hajime Takahashi for their excellent Latham, K.E., 1999. Mechanism and control of embryonic genome activa-
technical support in rabbit oocyte collection and in vitro tion in mammalian embryos. Int. Rev. Cytol. 193, 71–124.
Linnane, M.I., Brereton, A.J., Giller, P.S., 2001. Seasonal changes in circadian
fertilization. This study was supported by a grant from the grazing patterns of Kerry cows (Bos Taurus) in semi-feral conditions
Wakayama Prefecture Collaboration of Regional Entities for in Killarney National Park, Co. Kerry, Ireland. Appl. Anim. Behav. Sci.
the Advancement of Technology Excellence Project (CRE- 71, 277–292.
Loop, S., Katzer, M., Pieler, T., 2005. mPER1-mediated nuclear export of
ATE Project) of the Japan Science and Technology Agency
mCRY1/2 is an important element in establishing circadian rhythm.
and a Grant-In-Aid for Young Scientists (19780210) from EMBO Rep. 6, 341–347.
the Japanese Ministry of Education, Culture, Sports, and Lonergan, P., Rizos, D., Gutierrey-Adan, A., Fair, T., Boland, M.P., 2003.
Oocyte and embryo quality: effect of origin, culture conditions and
Technology.
gene expression patterns. Reprod. Dom. Anim. 38, 259–267.
Contributions. Conceived and designed the experiments: Mahoney, M.M., Sisk, C., Ross, H.E., Smale, L., 2004. Circadian regulation
Amano T, Kishi M, Anzai M, Kato H, Mitani T, Kishigami of gonadotropin-releasing hormone neurons and the preovulatory
S, Saeki K, Hosoi Y, Iritani A, Matsumoto K; quantified surge in luteinizing hormone in the diurnal rodent, Arvicanthis nilog-
icus, and in a nocturnal rodunt, Rattus norvegicus. Biol. Reprod. 70,
the transcripts: Matsushita A, Kakegawa R, Yanagisawa A, 1049–1054.
Watanabe T, Hatanaka Y; sequenced the DNA and analyzed Matsuo, T., Yamaguchi, S., Mitsui, S., Emi, A., Shimoda, F., Okamura, H.,
the data: Tokunaga K and Takemoto A; produced the cat- 2003. Control mechanism of the circadian clock for timing of cell
division in vivo. Science 10, 255–259.
tle samples: Tatemizo A and Saeki K; produced the rabbit Memli, E., First, N.L., 2000. Zygotic and embryonic gene expression in
samples: Anzai M, Kishi M, Kato H, Mitani T, Kishigami S, cow: a review of timing and mechanisms of early gene expression
Hosoi Y; wrote the paper: Amano T. as compared with other species. Zygote 8, 87–96.
T. Amano et al. / Animal Reproduction Science 121 (2010) 225–235 235

Mtango, N.R., Potireddy, S., Latham, K.E., 2008. Oocyte quality and mater- Reppert, S.M., Weaver, D.R., 2001. Molecular analysis of mammalian cir-
nal control of development. Int. Rev. Cell. Mol. Biol. 268, 223–290. cadian rhythms. Annu. Rev. Physiol. 63, 647–676.
Nagoshi, E., Saini, C., Bauer, C., Laroche, T., Naef, F., Schibler, U., 2004. Saeki, K., Nagao, Y., Kishi, M., Nagai, M., Iritani, A., 1998. Timing of comple-
Circadian gene expression in individual fibroblasts: cell-autonomous tion of the first meiotic division in bovine oocytes after maintenance
and self-sustained oscillators pass time to daughter cells. Cell 119, of meiotic arrest with cycloheximide and their subsequent develop-
693–705. ment. J. Vet. Med. Sci. 60, 523–526.
Nothias, J.Y., Majumder, S., Kaneko, K.J., DePamphilis, M.L., 1995. Regula- Schwartz, M.D., Nunez, A.A., Smale, L., 2004. Differences in the suprachi-
tion of gene expression at the beginning of mammalian development. asmatic nucleus and lower subparaventricular zone of diurnal and
J. Biol. Chem. 270, 22077–22080. nocturnal rodents. Neuroscience 127, 13–23.
Oh, B., Hwang, S.Y., Solter, D., Knowles, B., 1997. Spindlin, a major maternal Smale, L., Lee, T., Nunez, A., 2003. Mammalian diurnality: some facts and
transcript expressed in the mouse during the transition from oocyte gaps. J. Biol. Rhythms 18, 356–366.
to embryo. Development 124, 493–503. Takahashi, Y., First, N.L., 1992. In vitro development of bovine one-cell
Parcheco-Trigon, S., Hennequet-Antier, C., Oudin, J.F., Piumi, F., Renard, J.P., embryos: Influence of glucose, lactate, pyruvate, amino acids and vita-
Duranthon, V., 2002. Molecular characterization of genomic activities mins. Theriogenology 37, 963–978.
at the onset of zygotic transcription in mammals. Biol. Reprod. 67, Tervit, H.R., Whittingham, D.G., Rowson, L.E., 1972. Successful culture in
1907–1918. vitro of sheep and cattle ova. J. Reprod. Fertil. 30, 493–497.
Polanski, Z., Ledan, E., Brunet, S., Louvet, S., Verlhac, M.H., Kublak, J.Z., Toh, K.L., Jones, C.R., He, Y., Eide, E.J., Hinz, W.A., Virshup, D.M.,
Maro, B., 1998. Cyclin synthesis controls the progression of meiotic Ptácek, L.J., Fu, Y.H., 2001. An hPer2 phosphorylation site muta-
maturation in mouse oocytes. Development 125, 4989–4997. tion in familial advanced sleep phase syndrome. Science 291,
Pousette, A., Akerlöf, E., Rosenborg, L., Fredricsson, B., 1986. Increase in 1040–1043.
progressive motility and improved morphology of human spermato- Xumei, W., Viveiros, M.M., Eppig, J.J., Bai, Y., Fitzpatrick, S.L., Matzuk, M.M.,
zoa following their migration through Percoll gradients. Int. J. Androl. 2003. Zygote arrest1 (Zar1) is a novel maternal-effect gene critical for
9, 1–13. the oocyte-to-embryo transition. Nat. Genet. 33, 187–191.
Quinn, P., Barros, C., Whittingham, D.G., 1982. Preservation of hamster Yan, J., Wang, H., Liu, Y., Shao, C., 2008. Analysis of gene regulatory net-
oocytes to assay the fertilizing capacity of human spermatozoa. J. works in the mammalian circadian rhythm. PLoS. Comput. Biol. 10,
Reprod. Fertil. 66, 161–168. e1000193.
Animal Reproduction Science 121 (2010) 236–241

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Resumption of ovarian cyclicity and fertility response in bull-exposed


postpartum buffaloes
P.P. Gokuldas a,∗ , M.C. Yadav a , H. Kumar a , G. Singh b , S. Mahmood a , A.K.S. Tomar c
a
Division of Animal Reproduction, Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India
b
Division of Physiology & Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India
c
LPM Section, Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India

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

Article history: A study was undertaken to evaluate the effect of bull exposure on resumption of ovar-
Received 22 January 2010 ian cyclicity and fertility response in postpartum buffaloes raised under standard farm
Received in revised form 9 June 2010
conditions. A total of 24 Murrah buffaloes was randomly grouped to receive one of the
Accepted 16 June 2010
following treatments: (1) exposure to a vasectomised bull from 40th to 90th day postpar-
Available online 1 July 2010
tum (bull-exposed, BE, n = 11) and (2) isolated from bull (non-exposed, NE, n = 13). Changes
in the progesterone concentration were used to assess the resumption of ovarian cyclic-
Keywords:
ity. Postpartum interval to resumption of ovarian cyclicity (47 ± 2.58 days vs. 56 ± 2.37
Biostimulation
Buffalo days, p < 0.05) as well as behavioral estrus (57 ± 3.61 days vs. 71 ± 5.13 days, p < 0.05) was
Bull exposure shorter in bull-exposed animals than control animals. Similarly, animals in the BE group
Ovarian cyclicity had significantly shorter interval to postpartum ovulation (48 ± 2.69 days vs. 57 ± 2.37 days,
Fertility p < 0.05). Reduced incidence of silent ovulation was observed in BE group compared to NE
Postpartum group (18.18% vs. 50%). More than half proportion of animals in BE group conceived by 60
Progesterone days postpartum compared to a very low proportion of animals in NE group (54% vs. 15%,
p < 0.05). Furthermore, first service conception rate in BE animals was significantly greater
than NE animals (100% vs. 37.50%, p < 0.05). In conclusion, continuous bull exposure to buf-
faloes during later postpartum period accelerates resumption of ovarian cyclicity, reduces
incidence of silent ovulation and enhances first service conception rate. These results indi-
cate that introduction of bulls to buffalo herd could be a rational management strategy for
reducing the postpartum anestrus by enhancing reproductive function in buffaloes.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction anestrus and repeat breeding, besides delayed age at


first calving and higher incidence of silent ovulations.
In tropical countries like India, the domestic buffalo pro- Delayed postpartum estrus and conception affect produc-
duction is an indispensable livestock resource to millions tivity and cause economic losses to buffalo producers.
of smallholder farmers. Indian buffalo constitute less than Various attempts to regulate postpartum reproduction
40% of the bovine population, but accounts for more than through use of hormones and biological agents have so
half (34.5 million tonnes) of the total milk production in the far failed to be consistently effective and besides, some
country (Ingawale and Dhoble, 2004). However, buffaloes of these biologicals are not easily available, unsustainable
are considered difficult breeder because of their inher- or too expensive for smallholder farmers. For harnessing
ent susceptibility to environmental stress which cause the reproductive potential of buffaloes, there is a need to
develop rational management strategies that will curtail
postpartum reproductive problems, without reducing milk
∗ Corresponding author. Tel.: +91 9927305130; fax: +91 581 2303284.
yield or calf growth rate, and without significant increases
E-mail address: dasgokul2001@gmail.com (P.P. Gokuldas).
in production costs.

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.005
P.P. Gokuldas et al. / Animal Reproduction Science 121 (2010) 236–241 237

Table 1
Characteristics (mean ± SEM) of buffaloes exposed or non-exposed to vasectomised bull.

Groups Body weight (kg) Age (years) Parity Calf body weight (kg) Calf sex ratio

Bull-exposed (BE) (n = 11) 586 ± 25a 8.64 ± 0.93b 3.0 ± 0.33c 30.82 ± 0.81a 1.2:1
Non-exposed (NE) (n = 13) 570 ± 20a 6.69 ± 0.76b 2.77 ± 0.31c 31.31 ± 1.26a 1.17:1
Overall (n = 24) 578 ± 22 7.66 ± 0.85 2.88 ± 0.32 31.07 ± 1.02 1.18:1

Values bearing common superscripts within a column did not differ significantly (p > 0.05) by Student’s t-test.

Biostimulation by bull exposure represents a relatively The elapsed time period within which reproductive
inexpensive management tool. It has been reported that performance was monitored in this study included from
the exposure of bulls to postpartum cows can be used 40 to 90 days postpartum. Twenty-four Murrah buffaloes
to reduce the interval from calving to the resumption of with similar body weight, age, parity, calf body weight
luteal activity in primiparous (Gifford et al., 1989; Custer and calf sex ratio were selected for the study. There were
et al., 1990; Fernandez et al., 1993) as well as multiparous no significant differences between the groups in terms of
cows (Zalesky et al., 1984; Alberio et al., 1987; Naasz and these parameters (Table 1). The buffaloes were grouped
Miller, 1990; Burns and Spitzer, 1992). Presence of male at random to receive one of the following treatments: (1)
before the breeding season stimulated onset of estrus in exposure to a vasectomised bull (bull-exposed, BE) and (2)
seasonally anestrus ewes (Knight and Peterson, 1978) in isolated from bull (non-exposed, NE). The animals of the
goats (Shelton, 1960) and in suckled sows (Rowlinson et BE group (n = 11) were continuously exposed to a vasec-
al., 1975). While there is overwhelming evidence to support tomised bull which was tethered in a semi-open shed from
the effect of bull exposure in inducing earlier return to post- 40th to 90th day postpartum whereas, the buffaloes in the
partum estrus in cattle, there exists a paucity of information NE group (n = 13) were kept in another semi-open shed at
concerning this application in postpartum buffaloes. The a distance of approximately 0.13 km without bull exposure
objective of the present study was to study the effect of for the similar period. Buffaloes in this group had no visual
direct bull exposure on resumption of ovarian cyclicity and or tactile contact with mature bulls or their excretory prod-
fertility response in postpartum buffaloes. ucts during the same period. Onset of estrus in all animals
was checked twice daily (8:30 h and 17:00 h) by teaser bull
2. Materials and methods parading and confirmed by examination of genitalia per
rectum. Animals in estrus were inseminated using 0.5 ml
2.1. Experimental materials and procedures of frozen–thawed semen of fertile buffalo bulls. Pregnancy
was checked by per rectal examination after 60 days.
This study was conducted during the winter season with
24 postpartum Murrah buffaloes maintained at the Cat- 2.2. Progesterone analysis
tle and Buffalo Farm, Indian Veterinary Research Institute,
Bareilly, Uttar Pradesh. The farm is situated at 28◦ 22 (N) Changes in the plasma progesterone concentration
latitude and 79◦ 24 (E) longitude, at an altitude of 169.2 m were used to assess the resumption of ovarian cyclic-
above mean sea level. Agro-ecologically, the experimen- ity. Blood samples were collected by jugular venipuncture
tal site is situated in the upper Gangetic plain region with in heparinised glass tubes. Samples were collected at
extreme conditions where temperature varies from 37.5 ◦ C 4 days interval beginning from 40th day after calving
to 39.5 ◦ C in the month of May to June (summer) and 5 ◦ C to until 90th day. After collection, plasma from each blood
10 ◦ C in December to February (winter). All buffaloes were sample was separated through centrifugation (600 × g
between the second and the fourth parity and were free for 10 min) and stored in 1.5 ml aliquots at −20 ◦ C until
from reproductive disorders. They were maintained under assayed. The progesterone levels in the plasma samples
similar managemental conditions and stall feeding with were estimated using solid phase 125 I Radioimmunoas-
free access to water. The animals were hand-milked twice say kits (Immunotech, Beckman Coulter Company, France).
daily and limited suckling by a calf preceded each milking. The assay sensitivity was 0.05 ng/ml and had an intra-

Table 2
Effect of bull exposure on some reproductive criteria (mean ± SEM) in postpartum buffaloes.

Reproductive criteria BE group (n = 11) NE group (n = 13) p value

Postpartum interval (days) to:


(1) resumption of ovarian activity 47.4 ± 2.58a 56.0 ± 2.37b <0.05
(2) first ovulation 48.1 ± 2.69a 57.0 ± 2.37b <0.05
(3) first behavioral estrus 54.5 ± 1.79 59.6 ± 4.74 0.29
(4) first behavioral estrus(adjusted) 57.7 ± 3.61a 71.3 ± 5.13b <0.05

Proportion of animals resuming cyclicity*


by days 60 postpartum 81.82%a 38.46%b <0.05
by days 90 postpartum 90.91% 61.54% 0.11

Incidence of silent ovulations* 18.18% 50.00% 0.12

Values bearing different superscripts within a row differ significantly (p < 0.05) by Student’s t-test. BE-Bull-exposed; NE-Non-exposed.
*
Percentages within rows that lack a common superscript differ significantly (p < 0.05) by normal deviate test.
238 P.P. Gokuldas et al. / Animal Reproduction Science 121 (2010) 236–241

and inter-assay coefficient of variation of <10%. A total of


760 plasma samples was thawed at room temperature and
assayed for progesterone.
An increase in baseline progesterone concentration in
at least two consecutive samples that exceeded 1 ng/ml
was used as the criterion for resumption of ovarian
cyclicity (Stumpf et al., 1992; Berardinelli and Joshi, 2005).
Postpartum intervals to estrus or conception for buffaloes
that had not exhibited estrus or conceived by the end of
experimental period (90 days postpartum) were calculated
by assuming that estrus or conception in these animals had
occurred on the last day of the experimental period and
subtracting this date from their calving date (Fernandez et Fig. 1. Cumulative percentages of cyclic buffaloes in bull-exposed and
al., 1996). These intervals were then included in the data non-exposed group. Bars that lack common letter between groups differ
set of buffaloes that actually showed estrus or conceived by significantly (p < 0.05) by normal deviate test.
the end of the experiment (adjusted interval) for statistical
analysis. Inspection of cumulative proportions of buffaloes that
resumed estrous activity at weekly intervals during the
experiment indicated that maximum response in terms of
2.3. Statistical analysis
resumption of cyclicity occurred during 60–70 days post-
calving (20–30 days of bull exposure, Fig. 1). By 60 days
Data were analyzed as per Snedecor and Cochran (1989).
postpartum, a significantly higher proportion (81.82% vs.
Student’s t-test was used to compare the means of post-
38.46%) of BE animals resumed cyclicity as compared to
partum interval to ovulation, first behavioral estrus, and
animals in NE group. In accordance with the findings of
conception between the groups and a p value of <0.05 was
Abdalla (2003), animals in the BE group had significantly
used to define statistical significance. The proportional data
shorter interval to postpartum ovulation (48.1 ± 2.69 days
were analyzed by Normal Deviate test. Student’s t-test and
vs. 57.0 ± 2.37 days, Table 2). Ruminant animals are spon-
Duncan’s multiple range tests were used to compare the
taneous ovulators but may experience seasonal cyclicity.
means of progesterone between the groups.
However, the influence of male and mating stimuli in
hastening ovulation through nervous and hormonal mech-
3. Results and discussion anisms associated with it has been proven even in these
species (Randel et al., 1975; Kutty, 2004). Interestingly,
In the present study, the postpartum interval to resump- in the present study, bull exposure stimulated a major-
tion of ovarian cyclicity (47.4 ± 2.58 days vs. 56.0 ± 2.37 ity of animals to ovulate before 50 days postpartum. This
days, p < 0.05) as well as adjusted interval to behav- is important as it allows them to experience one or two
ioral estrus (57.7 ± 3.61 days vs. 71.3 ± 5.13 days, p < 0.05, estrous cycles for successful breeding before 70–90 days
Table 2) was significantly shorter for buffaloes exposed postpartum, by which the buffaloes must be pregnant to
to bull than non-exposed buffaloes. These results are maintain a calving interval of 13–14 months (El-Wishy,
consistent with the reports of Burns and Spitzer (1992) 2007).
and Rekwot et al. (2000) in cattle and Abdalla (2003) in Presence of males has been found to enhance the estrous
buffaloes. The finding that early resumption of ovarian expression in sows (Langendijk et al., 2000) and goats
cyclicity in BE animals possibly indicates a rapid response (Kunowska and Ryniewicz, 2001). Various workers have
of these animals to male stimuli. reported significantly reduced proportion of silent ovula-
However, in contrast to these results, some work- tion in bull-exposed cattle (22% vs. 89%, Alberio et al., 1987)
ers recorded no significant difference in the interval to as well as buffaloes (57.14% vs. 85.71%, Barman, 2008).
resumption of ovarian activity and estrus between bull- This is further confirmed by a lower incidence of silent
exposed and non-exposed suckling beef cows (Alberio et ovulation in bull-exposed buffaloes (18.18%) compared
al., 1987) and buffaloes (Barman, 2008). In these stud- to non-exposed animals (50.00%) in this study (Table 2).
ies, animals were exposed to bull in the early postpartum Remarkably, the presence of bulls increased overall inci-
period (10 days and 3 days postpartum, respectively). dence of overt estrus, while decreasing the incidence of
Tauck (2005) opined that postpartum cows would not silent estrus. As has been seen in this study, lack of appro-
respond to the bull pheromone when bulls were intro- priate male stimuli seems to be a contributory factor for
duced to cows shortly after parturition. The initiation of weak expression of estrus and higher incidence of silent
bull exposure post-calving appears to be a decisive factor ovulation in non-exposed buffaloes.
in the biostimulatory effect as neuro-endocrine restora- Table 3 depicts various parameters in relation to fertil-
tion of cyclicity takes place gradually over 20–30 days after ity response in experimental animals. Animals in BE group
parturition (Alberio et al., 1987). In the present study, bull conceived earlier with an insignificant difference of 6 days
exposure at progressively later stage (40 days postpartum) compared to NE group (55.4 ± 1.8 vs. 61.5 ± 5.4, p > 0.05).
would have strengthened the biostimulatory effect and However, difference of adjusted interval from calving
consequently exposed animals responded more rapidly to to conception between the two groups (13.2 days) was
the male stimuli. found to be statistically significant (68.0 ± 5.3 vs. 81.2 ± 4.1,
P.P. Gokuldas et al. / Animal Reproduction Science 121 (2010) 236–241 239

Table 3
Effect of bull exposure on certain fertility parameters in postpartum Murrah buffaloes.

Parameter BE group (n = 11) NE group (n = 13) p value

(1) Proportion of animals conceiving by


days 60 postpartum 54.55%a 15.39%b <0.05
days 90 postpartum 63.63% 30.77% 0.09

(2) First service conception rate 100%a 37.50%b <0.05


(3) Calving to conception* 55.4 ± 1.8 61.5 ± 5.4 0.22
(4) Calving to conception (adjusted)* 68.0 ± 5.3a 81.2 ± 4.0b <0.05

Percentages bearing different superscripts within a row differ significantly (p < 0.05) by normal deviate test. BE-Bull-exposed; NE-Non-exposed.
*
Means (±SEM) bearing different superscripts within a row differ significantly (p < 0.05) by Student’s t-test.

p < 0.05). More than half proportion of animals in BE group 1 ng/ml was observed as early as on 44th day postpartum
conceived by 60 days postpartum compared to a very low in biostimulated buffaloes indicates early resumption of
proportion of animals in NE group (54.55% vs. 15.39%). cyclicity in these animals. Similarly, higher progesterone
Other noteworthy finding in the present study was signif- levels (above 2.5 ng/ml) observed in BE animals during
icantly higher first service conception rate in bull-exposed later postpartum period (after 60 days) probably indicates
buffaloes compared to non-exposed buffaloes (100% vs. enhanced conception rate as well as higher proportion of
37.50%, Table 3). Conception data indicate that not only bull cyclic animals in BE group.
exposure significantly reduced calving to conception inter- Exposure of postpartum animals with bull may be
val but that a significant proportion of buffaloes conceived affecting time of ovulation, fertilization rates, corpus
at bull induced estrus associated with the first postpar- luteum development, progesterone secretion and embry-
tum estrous cycle. In support of this contention, Ebert et onic survival. In the present study, higher levels of
al. (1972) reported higher conception rate after first ser- progesterone were observed during the critical period of
vice (68%) in the bull-exposed group than the control group conception in BE buffaloes. Higher plasma progesterone
(48%). This is further supported by the findings of Abdalla concentration during the estrous cycle preceding insem-
(2003), who reported significantly higher proportion of ination is closely related to the occurrence of conception in
conception in buffaloes (84% vs. 63%) exposed continu- postpartum dairy cows (Folman et al., 1973). Secretion of
ously (from day 7 to 150 days postpartum) to bull than endogenous LH also may improve potential development
those exposed twice daily (30 min per session for same of the corpus luteum and increase progesterone secretion
period) to bull. Considering the fertility response of bull (Kawate et al., 2000). Maurer and Echternkamp (1982)
induced estrous cycles, the results of this study further reaf- found higher embryo survival in cows with higher pre-
firm that bull exposure can enhance the conception rates ovulatory surges of LH and earlier postovulatory rises of
and reduce calving to conception interval in postpartum luteal progesterone. Interestingly, introduction of a male
buffaloes. These findings and the observation that, higher to females has been shown to induce a rapid release of
proportion of animals became cyclic within 20 days of bull LH, mating behavior and ovulation in mice (Bronson and
exposure, thus indicated bull-mediated stimulatory effect Desjardins, 1974) and goats (Claus et al., 1990). There-
on reproductive function in postpartum buffaloes. fore, the possibility that added increment of LH secretion
Apparently higher progesterone levels, even though sta- might have become beneficial to the events associated with
tistically non-significant, were observed in bull-exposed conception in bull-exposed buffaloes cannot be ignored.
animals throughout the experiment (Fig. 2). This is in agree- Further work needs to be done to elucidate the bull bios-
ment with the results reported by Hornbuckle et al. (1995) timulatory effect on hormone secretion and its mechanism
in cattle. The finding that mean progesterone rises above for improving conception.

Fig. 2. Mean (±SEM) plasma progesterone concentration in bull-exposed and non-exposed group of buffaloes.
240 P.P. Gokuldas et al. / Animal Reproduction Science 121 (2010) 236–241

It is known that resumption of ovarian cycling activity Berardinelli, J.G., Joshi, P.S., 2005. Introduction of bulls at different days
is characterized by the release of GnRH by the hypotha- postpartum on resumption of ovarian cycling activity in primiparous
beef cows. J. Anim. Sci. 83, 2106–2110.
lamus in low amplitude high frequency temporal release Bronson, F.H., Desjardins, C., 1974. Circulating concentrations of FSH,
pattern (Wright et al., 1992). As a result, the anterior pitu- LH, estradiol and progesterone associated with acute, male-induced
itary releases low amplitude, high frequency pulses of LH puberty in female mice. Endocrinology 94, 1658–1664.
Burns, P.D., Spitzer, J.C., 1992. Influence of biostimulation on reproduction
which in turn stimulates ovulation and resumption ovar- in postpartum beef cows. J. Anim. Sci. 70, 358–362.
ian cycling activity. Unlike in mice (Singh, 2001) and goats Claus, R., Over, R., Dehnhard, M., 1990. Effect of male odour on LH secretion
(Iwata et al., 2000) aspects of biostimulation such as the and the induction of ovulation in seasonally anestrous goats. Anim.
Reprod. Sci. 22, 27–31.
behavioral and pheromonal signal and its translation into Custer, E.E., Berardinelli, J.G., Short, R.E., Wehman, M., Adair, R., 1990.
a hormonal response remain unclear in cattle and buf- Postpartum interval to estrus and patterns of LH and progesterone
faloes. Fernandez et al. (1996) observed that bull exposure in first-calf suckled beef cows exposed to mature bulls. J. Anim. Sci.
68, 1370–1377.
caused an increase in the pulse frequency of luteinizing
Ebert, J.J., Conteras, P., Saelzer, P., 1972. Influence of a teaser bull on
hormone from the anterior pituitary. These data support puerperium and fertility in dairy cows. In: Proceedings of the 8th
the hypothesis that bull exposure stimulates the percep- International Congress on Animal Reproduction and Artificial Insem-
tion of pheromones which cause a relatively immediate ination, vol. 3, Munich, pp. 1743–1748.
El-Wishy, A.B., 2007. The postpartum buffalo II. Acyclicity and anestrus.
stimulation of the HPO axis and the release of LH which in Anim. Reprod. Sci. 97, 216–236.
turn stimulates ovulation and resumption ovarian cycling Fernandez, D., Berardinelli, J.G., Short, R.E., Adair, R., 1996. Acute and
activity. The findings of the present study such as, bull chronic changes in luteinizing hormone secretion and postpartum
interval to estrus in first-calf suckled beef cows exposed continuously
exposure rapidly induced estrus in higher proportion of or intermittently to mature bulls. J. Anim. Sci. 74, 1098–1103.
animals, reduced incidence of silent estrus and enhanced Fernandez, D., Berardinelli, J.G., Short, R.E., Adair, R., 1993. The time
conception rate, further corroborate the fact that biostimu- required for the presence of bulls to alter the interval from parturi-
tion to resumption of ovarian activity and reproductive performance
latory effect of bull did influence postpartum reproduction in first calf-suckled beef cows. Theriogenology 39, 411–419.
in buffaloes. The bull-effect observed in the present study Folman, Y., Rosenberg, M., Herz, Z., Davidson, M., 1973. The relationship
might have occurred through olfactory cues (pheromones) between plasma progesterone concentration and conception in post-
partum dairy cows maintained on two levels of nutrition. J. Reprod.
transmitted from the bull to the female as they were in con- Fertil. 34, 267–278.
tinuous and close proximity to the pheromonal source for a Gifford, D.R., D’Occhio, M.J., Sharpe, P.H., Weatherly, T., Pittar, R.Y., Reeve,
sufficient period during which the stimuli could influence D.V., 1989. Return to cyclic ovarian activity following parturition in
mature cows and first-calf beef heifers exposed to bulls. Anim. Reprod.
the HPO axis to initiate resumption of ovarian activity.
Sci. 19, 209–214.
Hornbuckle, T., Weston, P.G., Hixon, J.E., 1995. Effects of bull exposure on
4. Conclusions the cyclic activity of beef cows. Theriogenology 61, 1521–1532.
Ingawale, M.V., Dhoble, R.L., 2004. Buffalo reproduction in India: an
Continuous bull exposure of buffaloes during later overview. Buffalo Bull. 23 (1), 4–7.
Iwata, E., Wakabayashi, Y., Kakuma, Y., Kikusui, T., Takeuchi, Y., Mori, Y.,
postpartum period (40 days post-calving) accelerates post- 2000. Testosterone dependent primer pheromone production in the
partum resumption of ovarian cyclic activity, reduces sebaceous gland of male goat. Biol. Reprod. 62, 806–810.
incidence of silent ovulation and enhances first service Kawate, N., Morita, N., Tsuji, M., Tamada, H., Inaba, T., Sawada, T., 2000.
Roles of pulsatile release of LH in the development and mainte-
conception rate. Biostimulation through bull stimuli offers
nance of corpus luteum function in the goat. Theriogenology 54 (7),
an efficient strategy to increase the number of pregnant 1133–1143.
animals during the earlier days of mating period after Knight, T.W., Peterson, A.J., 1978. The ovarian and hormonal response
calving. Incorporation of bull biostimulation into buffalo of ewe to the stimulation by the ram early in the breeding season.
Theriogenology 10, 343–353.
production system may help to optimize overall repro- Kunowska, M., Ryniewicz, Z., 2001. The preliminary investigations on
ductive performance and fertility response in postpartum estrus period in goats. J. Anim. Sci. 38, 47–56.
buffaloes. Kutty, I.C., 2004. Emerging fertility problems in cattle and buffaloes bred
through artificial insemination. Intas Polivet 5 (11), 269–274.
Langendijk, P., Soede, N.M., Bouwman, E.G., Kemp, B., 2000. Responsive-
Acknowledgements ness to boar stimuli and change in vulvar reddening in relation to
ovulation in weaned sows. J. Anim. Sci. 78, 3019–3026.
The authors thank the Director, IVRI, Bareilly, India for Maurer, R.R., Echternkamp, S.E., 1982. Hormonal asynchrony and embry-
providing necessary facilities and logistics support to con- onic development. Theriogenology 17, 11–16.
Naasz, C.D., Miller, H.L., 1990. Effects of bull exposure on postpartum inter-
duct the study. Thanks are also due to the Indian Council of val and reproductive performance in beef cows. Can. J. Anim. Sci. 70,
Agricultural Research, Government of India for the financial 537–543.
support provided in the form of Institutional Fellowship to Randel, R.D., Short, R.E., Christensen, D.S., Bellows, R.A., 1975. Effect of cli-
toral massage after artificial insemination on conception in the bovine.
the first author. J. Anim. Sci. 40, 1119–1123.
Rekwot, P.I., Ogwu, D., Oyedipe, E.O., 2000. Influence of bull biostim-
References ulation, season and parity on resumption of ovarian activity of
zebu (Bos indicus) cattle following parturition. Anim. Reprod. Sci. 63,
Abdalla, E.B., 2003. Improving the reproductive performance of Egyptian 1–11.
buffalo cows by changing the management system. Anim. Reprod. Sci. Rowlinson, P., Boughton, H.G., Bryant, M.J., 1975. Studies on reproductive
75, 1–8. performance of buffaloes. Anim. Prod. 21, 233–241.
Alberio, R.H., Schiersmann, G., Carou, N., Mestre, J., 1987. Effect of a teaser Shelton, M., 1960. Influence of the presence of a male goat on the initia-
bull on ovarian and behavioral activity of suckling beef cows. Anim. tion of estrous cycling and ovulation of Angora does. J. Anim. Sci. 19,
Reprod. Sci. 14, 263–271. 368–379.
Barman, P., 2008. Identification of urinary compounds, response of estrual Singh, P.B., 2001. Chemosensation and genetic individuality. Reproduction
and diestrual urine on male libido and effect of bull exposure on ovar- 121, 529–539.
ian cyclicity in postpartum buffaloes. Ph.D. Thesis submitted to Indian Snedecor, G.W., Cochran, W.G., 1989. Statistical Methods, 6th ed. The Iowa
Veterinary Research Institute, Izatnagar, U.P., pp. 68–70. State University Press, USA.
P.P. Gokuldas et al. / Animal Reproduction Science 121 (2010) 236–241 241

Stumpf, T.T., Wolfe, M.W., Day, M.L., Kittok, R.J., Kinder, J.E., 1992. Weight Wright, I.A., Rhind, S.M., Whyte, T.K., Smith, A.J., 1992. A note on the effects
changes prepartum and presence of bulls postpartum interact to of pattern intake and body condition on the duration of the post-
affect duration of postpartum anestrus in cows. J. Anim. Sci. 70, partum anoestrous period and LH profiles in beef cows. Anim. Prod.
3133–3137. 54, 143–146.
Tauck, S.A., 2005. Factors associated with the biostimulatory effect of bulls Zalesky, D.D., Day, M.L., Garcia-Winder, M., Imakawa, K., Kittok, R.J.,
on resumption of ovarian cycling activity and breeding performance D’Occhio, M.J., Kinder, J.E., 1984. Influence of exposure to bulls on
of first-calf suckled beef cows. M.S. Thesis submitted to Montana State resumption of estrous cycles following parturition in beef cows. J.
University, Bozeman, Montana, pp. 3–75. Anim. Sci. 59, 1135–1139.
Animal Reproduction Science 121 (2010) 242–248

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Short-term grazing of lucerne and chicory increases ovulation rate in


synchronised Merino ewes
B.J. King a,b,∗ , S.M. Robertson a,b , J.F. Wilkins a , M.A. Friend a,b
a
EH Graham Centre for Agricultural Innovation (Industry & Investment NSW and Charles Sturt University), Locked Bag 588,
Wagga Wagga, NSW 2678, Australia
b
Co-operative Research Centre for Future Farm Industries, Nedlands, WA, Australia

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

Article history: This study evaluated the ability of short-term grazing of live = green pasture to increase
Received 18 November 2009 ovulation rate during late summer when annual pasture is generally dead and of low qual-
Received in revised form 17 June 2010
ity. Ovulation rates, measured by the number of corpora lutea, were compared between
Accepted 28 June 2010
4 nutritional treatments: senesced phalaris (Phalaris aquatica), phalaris plus 500 g lupin
Available online 3 August 2010
grain per day, lucerne (Medicago sativa) or chicory (Chicorum intybus) pastures. The study
used 100 Merino ewes per treatment, divided between 2 replicates. The experiment was
Keywords:
repeated in 3 years; February 2006, and January 2007 and 2008. Oestrus was synchronised
Sheep
Ovulation rate and the ewes grazed the pastures for 9 days prior to ovulation at times corresponding to
Nutrition days 8–17 of the cycle in 2006, and days 6–14 in 2007 and 2008. The proportion of ewes
Flushing producing multiple ovulations was higher (P < 0.05) in the lucerne and chicory (0.36, 0.38)
Lupins than the phalaris (0.27), and intermediate in the lupin (0.33) treatment. Regression analysis
Lucerne showed that the proportion of ewes with multiple ovulations increased with the quantity
Chicory of live herbage (P < 0.04). Responses were achieved even at low levels of live herbage with
90% of the maximum proportion of multiples occurring at 350 kg DM/ha. It is concluded
that providing short-term grazing of live chicory or lucerne to ewes can increase ovulation
rates relative to ewes grazing senesced phalaris and to levels similar to those achieved by
lupin grain supplementation.
Crown Copyright © 2010 Published by Elsevier B.V. All rights reserved.

1. Introduction may be inexpensive and avoids the use of chemicals and


hormones.
Increasing ewe reproductive capacity provides poten- Increased nutrition or flushing prior to mating has long
tial for increasing profitability in sheep meat production been known to increase ovulation rates or the number
systems (Warn et al., 2006). This increase in reproductive of lambs born. This operates through either or both a
rate needs to be achieved cost effectively and in a way that ‘dynamic’ effect – a rising plane of nutrition and gaining
satisfies an increasing consumer demand for a “clean, green weight at and for some weeks prior to mating (Gunn et
and ethical” product (Martin et al., 2004). Using nutrition al., 1984), or a ‘static’ effect – that of the resultant higher
to manipulate ovulation rate provides a useful strategy that liveweight or condition at the time of mating (Coop, 1962;
Edey, 1968). An ‘acute’ effect of nutrition has also been
shown, whereby ‘short-term’ or ‘spike’ feeding with lupin
∗ Corresponding author at: School of Animal and Veterinary Sciences,
grain for 4–6 days increases ovulation rates without affect-
Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, Australia.
ing liveweight or body condition (Knight et al., 1975; Smith
Tel.: +61 02 6933 2427; fax: +61 02 6933 2911. and Stewart, 1990). This short-term feeding targets a criti-
E-mail address: bking@csu.edu.au (B.J. King). cal period in the luteal phase of the oestrous cycle around

0378-4320/$ – see front matter. Crown Copyright © 2010 Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.007
B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248 243

days 10–14 of the oestrous cycle (Stewart and Oldham, phalaris with and without short-term lupin grain supple-
1986), or during the period 6 days before luteolysis (Nottle mentation.
et al., 1990). The benefit of this strategy is that limited feed
resources can be used more efficiently than if a longer feed- 2. Materials and methods
ing period is required.
It is clear that the ovulatory response to short-term This experiment was conducted with the approval of
feeding with lupins is generally repeatable (Scaramuzzi et the Charles Sturt University Animal Ethics committee. The
al., 2006) although responses are variable, and can pro- experiment was conducted on a property south-east of
vide increases in ovulation rate of up to 60% (Stewart and Wagga Wagga NSW (latitude 147.5; longitude −35.2). The
Oldham, 1986; Teleni et al., 1989; Nottle et al., 1990, 1997; climate is typical Mediterranean with hot dry summers and
Wilkins, 1997). Lupin grain has been the most common feed moderately cold winters. Rainfall is an average 600 mm per
supplement used in previous studies, but is expensive and annum falling evenly throughout the year, although sum-
not readily available in all localities. However, the use of mer rainfall is typically less reliable.
supplements is an additional cost which may be unneces-
sary if similar responses could be obtained using existing 2.1. Animals and experimental procedures
pasture resources. Since research has provided increasing
evidence that the effect of short-term feeding on ovulation A schematic representation of the experimental design
rate is driven by dietary energy (e.g. Teleni et al., 1989; is presented in Fig. 1. The experimental design com-
Vinoles et al., 2005), other feed alternatives that provide prised 4 nutritional treatments (senesced phalaris pasture,
similar nutrition, but are less risky to metabolic health senesced phalaris pasture plus 500 g/ewe/day lupin grain
and are more cost effective than lupin supplementation, (Lupinus angustifolius), chicory and lucerne), in a ran-
become possible. Indeed, the value of short-term grazing domised block design with 2 replicates of each treatment
(up to 18 days) of high quality leguminous pastures such combination. The experiment was repeated in each of 3
as the perennial shrub tagasaste (Chamaecytisus palmen- years (2006, 2007 and 2008). In each year, 400 medium to
sis) (Wilkins, 1997) and Lotus spp. (Ramirez-Restrepo et al., large-framed 5-year-old Merino ewes of CentrePlus blood-
2005; Vinoles et al., 2009) have already been shown to have line (a dual-purpose bloodline selected for both meat and
positive effects on prolificacy. If similar responses to those wool characteristics) were stratified and randomly allo-
obtained with lupin grain could be achieved with com- cated to treatment groups (n = 100) according to body
monly grown summer-active pasture species, short-term condition (scale 0 (emaciated) to 5 (obese) (Jefferies, 1961))
grazing to increase ovulation rates in summer/autumn and liveweight. In 2007 and 2008, when some older ewes
joined ewes could be adopted by a substantial number of were replaced with younger ewes, animals were also allo-
sheep producers. cated according to age group.
Lucerne (Medicago sativa) and chicory (Chicorum inty- Ewes were weighed and condition scored (without
bus) are perennial pastures that provide good quality fasting) at the beginning and end of the nutritional treat-
nutrition during the summer and autumn (Holst et al., ment periods. Oestrous cycles were synchronised using
1998) when traditional annual pastures senesce and an intravaginal CIDR® (Controlled Internal Release Device;
become low in nutritive value (Thomas et al., 2010). 0.3 g progesterone, EZI-breed® NZ) inserted for 11–13 days.
Phalaris (Phalaris aquatic) is widely sown as pasture in In 2006, CIDRs were inserted on day −11 and removed
the higher rainfall temperate regions of Australia, but it on day 0. It was expected that the mean time of ovula-
is summer-dormant and less able to provide quality sum- tion would occur approximately 3 days after CIDRs were
mer feed for finishing lambs and improved ewe nutrition removed. That is, the mean time of onset of oestrus in the
at joining. Lucerne is suited to medium and high rainfall flock would occur around 36 h after CIDR removal (Kohno
areas and in addition to extending the supply of quality et al., 2005) and ovulation would occur approximately 24 h
pasture, can be sown as a pasture phase in crop rotations after that. All treatment groups grazed senesced pastures
to provide a disease break and fix nitrogen. Chicory is more off plots until day −6 when they were introduced to pas-
tolerant of acid soils than lucerne, thereby providing an ture plots and remained there until day 3. Lupin grain was
alternative where lucerne cannot be grown (Upjohn et al., fed daily to the relevant group while grazing the phalaris
2005). Lucerne and chicory are also desirable because, as pasture. After 2006, the protocol was modified to remove
perennials, they show potential to reduce ground water the possibility that high levels of feeding, late in the luteal
recharge and thus contribute to the control of dryland phase of the oestrous cycle, may inhibit ovulatory response
salinity (Dear and Ewing, 2008). Although these improved (Stewart and Oldham, 1986). Therefore, in 2007 and 2008,
pastures have proven effective in producing high growth CIDRs were inserted at day −13 and removed on day 0. In
rates in lambs over the summer period (Holst et al., 1998) these years, due to drought conditions, the ewes were sup-
in central N.S.W., the potential of these forages to increase plementary fed a maintenance ration (SCA, 1990) of wheat
ovulation rate in a fashion similar to supplementation with grain until day −8 when they were placed on the pasture
of lupin grain and the leguminous species reported by oth- plots and were removed from plots at the end of the CIDR
ers, is not known. treatment. In all years, the ewes grazed plots for a period
The field study described in this paper was conducted of 9 days, corresponding to days 8–17 (2006) or days 6–14
over 3 years. It aimed to investigate the effects of short- (2007 and 2008) of the 17-day oestrous cycle. On removal
term grazing of lucerne and chicory on ovulation rate in from pasture treatment plots, ewes were returned as one
Merino ewes compared to the existing perennial pasture, flock to annual pasture or low quality hay until the day
244 B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248

Fig. 1. Schematic representation of experimental design in 2006 and 2007/2008. Ewes were synchronised with CIDRs for either 11 (2006) or 13 days
(2007/2008). Where day 0 is the day of CIDR removal, ewes grazed nutritional treatments for 9 days in all years from day −6 to day 3 (2006) and day −8 to
day 0 (2007/2008). At the end of this period, all the ewes were united into one flock and grazed senesced annual pasture until ovulation rate was measured
via laparoscopy at day 5–6 after expected ovulation (2006) or via trans-rectal ultrasound at day 8–11 after expected time of ovulation.

of ovulation, after which grain feeding re-commenced if 2.2.1. Pasture biomass


required. Pastures were measured 2–4 days before ewes were put
Ovulation rate, as determined by the number of cor- on plots, and again 3 or 9 days after ewes were removed
pora lutea per ewe ovulating, was measured on 6 March from plots. Live and dead pasture biomass was visually
2006 via laparoscopy (Oldham and Lindsay, 1980) at day estimated using the method of Haydock and Shaw (1975)
5–6 after the estimated average time of ovulation in the as described by Cayley and Bird (1996), using an elec-
flock. On 29–31 January 2007 and 2008 ovulation rate was tric handpiece to cut calibration quadrats to ground level.
measured using transrectal ultrasonography (Vinoles et al., The quantity of live pasture pre-grazing in the chicory and
2004) with the ewe in a standing position at day 8–11 after lucerne treatments was higher (P < 0.05) than in the lupin
the estimated average time of ovulation. Transrectal ultra- and phalaris treatments (Table 1). Higher levels of herbage
sound provides a less invasive technique that has a high were estimated in 2008 than in other years, but in the
predictive value and sensitivity for the diagnosis of func- lupin and phalaris treatments all except about 100 kg was
tional corpora lutea (Dickie et al., 1999; Vinoles et al., 2004). clammy goosefoot (Chenopodium pumilio) which is gener-
ally considered relatively unpalatable. Over the 9 days of
grazing the quantity of pasture decreased, with most live
2.2. Pastures leaf material consumed 3 days before ewes were removed
from plots. The larger quantities of live lucerne and chicory
The pastures used as the nutritional treatments com- remaining post-grazing were stem, with no or little leaf;
promised senesced phalaris pasture (P. aquatica cv. the live material in lupin and phalaris in 2008 was clammy
Australian), senesced phalaris pasture, chicory (C. intybus goosefoot. The quantity of dead pasture was usually above
cv. Punall), and lucerne (M. sativa cv. Aurora) all with a levels that would limit pasture intake by sheep (SCA, 1990),
subclover (Trifolium subterranean) component. The phalaris both pre and post-grazing.
pasture was an existing 5-year-old stand that had been
sown with subclover (cv. Coolamon). The lucerne and
chicory pastures were sown in spring 2005 at sowing rates 2.2.2. Pasture/feed quality
of 4 and 3.5 kg seed/ha, respectively, along with 2 kg/ha In 2007 and 2008 samples for herbage quality were
subclover. The phalaris plots were 2.5 ha, and the lucerne taken using the ‘toe-cut’ method (Cayley and Bird, 1996)
and chicory plots 2.2 ha. and these and lupin grain samples were tested for crude

Table 1
Mean quantity of live and dead pasture (kg DM/ha) pre- and post-grazing for the different treatment groups averaged over 2006–2008.

Live pasture (kg DM/ha) Dead pasture (kg DM/ha)

Treatment Pre-grazing Post-grazing Pre-grazing Post-grazing

Phalaris 86a 39a 2220d 1869d


Phalaris + lupin 167a 156bc 1737c 1634c
Lucerne 761b 208cd 974a 928a
Chicory 884c 260d 1508b 1416b

Values with different superscript letters (a, b, c, d) within columns indicate means differ at P < 0.05.
B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248 245

Table 2 Table 3
Mean crude protein (CP %), dry organic matter digestibility (DOMD %) Mean daily maximum, minimum and mean temperature and total rainfall
and metabolisable energy (ME, MJ/kg DM) of live pasture post-grazing for during each experimental period 2006–2008.
different treatments, combining data for 2007–2008.
2006 2007 2008
Treatment CP (%) DOMD (%) ME (MJ/kg DM)
Mean (◦ C) 24.3 25.4 24.6
Phalaris 21.0d 66b 10.5c Mean minimum (◦ C) 16.3 17.2 18.1
Phalaris + lupin 19.6c 63b 9.8b Mean maximum (◦ C) 32.4 33.3 31.4
Lucerne 13.0b 47a 6.5a Rainfall (mm) 1 25 85
Chicory 10.5a 50a 7.0a

Values with different superscript letters (a, b, c, d) within columns indicate


means differ at P < 0.05.
of ewes ovulating, the proportion of ewes recording mul-
tiple ovulations (2+ ovulations) were analysed by logistic
regression. Condition score, measured at the start of the
protein (CP), neutral detergent fibre (NDF), dry matter nutritional treatments, was used as a co-variate for the
digestibility (DMD), and digestibility of organic matter proportion of multiple ovulations. Liveweight, body condi-
(DOMD) (Department of Primary Industries, Hamilton, tion and pasture biomass data were analysed using residual
Victoria). Values were estimated using near infrared spec- maximum likelihood (REML). Non-linear regression was
troscopy (NIR) and metabolisable energy (ME) calculated. used to predict the relationship between pasture biomass
The quality of the live component of pastures was simi- and the mean percentage of ewes with multiple ovulations
lar (P > 0.05) between all treatments pre-grazing: mean CP in each group (treatment/replicate/year). Quality parame-
17.2%; DOMD 62% and ME 9.8 MJ ME/kg DM. The quality ters for dead pasture for 2007 and 2008 were analysed by
of live pasture declined with grazing in the chicory and ANOVA; while live pasture parameters were analysed by
lucerne, but not in the phalaris or phalaris plus lupin treat- REML because there was insufficient live to test in 2007.
ments. This was partially due to a higher presence of the
unpalatable goosefoot in the phalaris plots. However, the
3. Results
ewes also consumed most of the leaf, leaving mainly stem
in the lucerne and chicory paddocks. Post-grazing quality
3.1. Liveweight and condition score
of live pasture is shown in Table 2.
The dead component of chicory pastures was higher
Ewe liveweight pre-grazing averaged 54.5 kg and did
(P < 0.05) than other pastures for crude protein, digestibil-
not differ between treatments, but increased with each
ity, ME and lower in NDF pre-grazing and generally
successive year (P < 0.001) being 51 ± 7 s.e., 52 ± 6 and
post-grazing. Mean values for all pastures pre- and post-
61 ± 7 kg respectively, in 2006, 2007 and 2008. Mean
grazing, respectively were CP 12.0, 9.62%; DOMD 52, 42%;
liveweight increased (P < 0.05) by 2 kg during grazing, with
and ME 7.56, 5.62 MJ ME/kg DM. Quality between years was
ewes grazing phalaris only increasing by 1 kg compared
generally similar (P > 0.05).
with 2–3 kg in others. Mean condition score pre-grazing
The lupin grain contained a mean 29.5% CP, 80% DOMD
in 2006 at 3.2 was higher (P < 0.001) than in later years
and 13.3 MJ ME/kg DM.
(3.0) but was similar (P > 0.05) between treatments. Post-
grazing the condition score of ewes which grazed phalaris
2.3. Weather was lower (P < 0.05) than for other treatments.

Temperature and rainfall data were recorded for the 3.2. Ovulation rate
period of grazing and up to measurement of ovulation rate
at a meteorological station 10 km from the experimen- On average, 8% of ewes did not ovulate, and this was sim-
tal site. Mean temperatures were similar between years ilar (P > 0.05) between treatments and years. The majority
(Table 3). of multiple ovulations were twins; only 1.8% of ovulating
ewes had triplet ovulations. In ovulating ewes, both lucerne
2.4. Statistical analyses and chicory increased (P < 0.05) the proportion of ewes with
multiple ovulations compared with phalaris pasture (0.36
Data were analysed using Genstat® 9th edition (Payne vs. 0.38 vs. 0.27 respectively; Table 4). Feeding lupins pro-
et al., 2006). The proportions of ewes failing to ovulate and, duced an intermediate response (0.33). The proportion of

Table 4
The mean ovulation rate and proportion of ewes with multiple ovulations for ewes grazing 4 pastures averaged over 2006–2008, excluding non-ovulating
ewes.

Treatment Number of ewes Mean Proportion of ewes with


ovulations/ewea 2+ ovulations

Phalaris 266 1.28 0.27a


Phalaris + lupin 270 1.35 0.33ab
Lucerne 278 1.41 0.36b
Chicory 274 1.39 0.38b

Values with different superscript letters (a, b) indicate means differ at P < 0.05.
a
Mean ovulations per ewe and the percentage of ewes with multiple ovulations are not the same due to the occurrence of some triple ovulating ewes.
246 B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248

Grazing both chicory and lucerne increased ovulation rates


to the same or higher extent when compared to ewes given
supplements of 500 g/h/day of lupin grain per ewe. The
ovulatory response is closely related to the quantity of live
pasture available and the pattern of feeding may also be
important.
The increases in ovulation rate of up to 10% in plots
where sufficient live pasture was available were within the
range achieved with lupin feeding in previous flushing tri-
als of either short-term (14–29%) (Stewart and Oldham,
1986; Teleni et al., 1989) or longer-term (−14 to 21%)
(Croker et al., 1985) flushing trials, indicating the suitability
Fig. 2. Predicted lines of regression between proportion of ovulating ewes of short-term grazing as an alternative. Although there are
with multiple ovulations and live pasture on offer pre- () and post- reports of grazing lucerne reducing ovulation rate, this was
grazing (), combining data for 2007 and 2008. due to ingestion of coumestans when lucerne is infected
with fungus (Smith et al., 1979), and this situation can be
ewes with multiple ovulations over all treatments differed avoided.
between years (P < 0.05): in 2006, 0.41; in 2007, 0.27; and There are few reports of short-term flushing using
in 2008, 0.33. However, there was an interaction (P < 0.05) live pasture. Wilkins (1997) investigated the potential of
between treatment and year largely due to a change in grazing for 18 days prior to mating using the perennial
ranking between chicory and lucerne between years. Data leguminous shrub tagasaste (C. palmensis). This plant pro-
is not shown because it was the quantity of pasture, rather vides fodder typically testing as 15–18% crude protein and
than the treatment, influencing this response, as presented around 70% in vitro digestibility (Oldham et al., 1994). He
below. found that the ovulation rates of ewes grazing tagasaste
in both of 2 experiments (1.34 and 1.22) were higher than
3.3. Relationship between quantity of pasture and that of control ewes grazing dead standing pasture (1.14
ovulation rate and 1.06), but not as high as that measured in ewes supple-
mented with lupins for 9 or 18 days prior to mating (1.80
Logistic regression showed no difference (P > 0.05) and 1.72). Similarly, the number of lambs born per ewe
between treatments in the relationship between the quan- has been increased by grazing Lotus corniculatus either for
tity of live pasture on offer and the proportion of ewes with 12 days (Vinoles et al., 2009) or several weeks (Ramirez-
multiple ovulations. This indicates that it was the quan- Restrepo et al., 2005). Thus previous evidence supports
tity of live pasture, rather than the species, that resulted in the current study which indicated that both legume and
different ovulatory responses in this study. non-legume live pastures can increase ovulation rate. It
Excluding the lupin treatments, where the response is likely that the quantity of live pasture will be the most
may not reflect the quantity of pasture, and 2006 data limiting factor determining ovulatory response, due to the
where ovulation rate was measured in March rather than improvement in feed quality when compared with senes-
January, the proportion of ewes with multiple ovulations cent pasture.
was significantly increased with increasing quantities of The proportion of ewes with multiple ovulations being
live herbage both pre- and post-grazing (Fig. 2). The regres- higher in 2006 than in later years was probably due to
sion equations, where y is proportion of ewes with multiple several factors: measurement via laparoscopy rather than
ovulations, were: ultrasound providing a more accurate estimate (Dickie et
al., 1999), ewes being in higher condition, and because ovu-
Pre-flushing live biomass: lation rate was measured well into, rather than at the start
y = 0.3492 − 0.276 × 0.99410biomass (P = 0.04) of the breeding season. From this study it is not possible to
Post-flushing live biomass: determine whether the ovulatory response to short-term
y = 0.3483 − 0.2203 × 0.9736biomass (P = 0.04) grazing varies with time of year.
The variation in ovulatory responses in different treat-
The proportion of ewes with multiple ovulations at 90% ments between years was largely explained by the
of the maximum predicted by the regression was achieved quantities of live pasture available. This is similar to previ-
with a pre-grazing live pasture on offer of 350 kg DM/ha, ous studies that show that increases in ovulation rate due
and post-grazing 70 kg DM/ha. There was a poor relation- to lupin supplementation are rate responsive (Lightfoot et
ship (P > 0.05) between the quantity of dead pasture and al., 1976). The data here suggest that significant responses
ovulation rate. in ovulation rate can be achieved when as low as 350 kg
DM/ha live pasture is available pre-grazing to synchronised
4. Discussion ewes.
In this study the pattern of nutrition would also have
This study indicates that short-term grazing of live pas- varied between years. Much or all of the live pasture leaf
tures can increase ovulation rate in oestrus synchronised was eaten before the end of the grazing period, at times sev-
ewes by increasing the proportion of ewes with multiple eral days before, such that both the quantity and quality of
ovulations compared with those grazing senesced pasture. live pasture after about day 10 of the oestrous cycle would
B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248 247

have been rapidly declining with mainly stem remaining. time of year and, to evaluate whether short-term feeding
A varying pattern between different paddocks probably is effective in unsynchronised ewes.
contributed to the varying response in ovulation rate, but
the regressions demonstrate that even small quantities Acknowledgements
of live pasture pre-grazing are associated with significant
increases in ovulation rate (Fig. 2). Although large quanti- This work was funded by EverGraze® - More livestock
ties of live stem of low quality were available post-grazing from perennials. EverGraze is a Future Farm Industries CRC,
in both lucerne and chicory in 2008, we can only specu- Meat and Livestock Australia and Australian Wool Innova-
late that ovulation rates could have been further increased tion research and delivery partnership. The studies were
if the quality of remaining pasture was higher. It is likely conducted under the CRC for Plant-based Management
because the ovulation rates achieved are considered below of Dryland Salinity. The authors wish to thank Profes-
the ewe’s genetic potential. In practice, producers need to sor Graeme Martin for his advice on experimental design,
adjust stocking rates so that sufficient live pasture remains Mr John Broster for his technical assistance and Dr Car-
at the end of the critical grazing period. olina Vinoles for conducting rectal ultrasounds in 2007. Dr
If a higher response is obtainable with better quality Raquel Waller and Mr Matthew Lieschke assisted with field
and quantity of pasture, then short-term flushing is more work in 2006.
likely to have commercial value. In our case, while the 10%
increase in ovulation rate was statistically significant, only References
approximately 10.5 more lambs would be born per 100
ewes joined, once embryo mortality has been accounted Cayley, J.W.D., Bird, P.R., 1996. Techniques for Measuring Pastures. Agri-
culture Victoria, Hamilton.
for. In this case, the economic benefit of the short-term
Coop, I.E., 1962. Liveweight-productivity relationships in sheep I.
feeding, after allowing for the additional costs of synchro- Liveweight and reproduction. NZ J. Agric. Res. 5, 249–264.
nisation (synchronisation materials, additional labour and Croker, K.P., Johns, M.A., Johnson, T.J., 1985. Reproductive performance
of Merino ewes supplemented with sweet lupin seed in southern
increased ram percentage) would depend on lamb values.
Western Australia. Aust. J. Exp. Agric. 25, 21–26.
From a practical viewpoint, synchronisation is a sub- Dear, B.S., Ewing, M.A., 2008. The search for new pasture plants to achieve
stantial cost and effort and is unlikely to be adopted unless more sustainable production systems in southern Australia. Aust. J.
the ovulatory response is large. However, the advantage Exp. Agric. 48, 387–396.
Dickie, A.M., Paterson, C., Anderson, J.L.M., Boyd, J.S., 1999. Determination
of synchronisation is that it allows exposure of all ewes of corpora lutea numbers in Booroola-Texel ewes using transrectal
to live pasture at the “critical” stage of the oestrous cycle ultrasound. Theriogenology 51, 1209–1224.
in a short grazing period. This is particularly beneficial Edey, T.N., 1968. Bodyweight and ovulation rate in sheep. Proc. Aust. Soc.
Anim. Prod. 24, 190.
in drought years such as those in this study, where the Gunn, R.G., Doney, J.M., Smith, W.F., 1984. The effect of level of pre-mating
quantity of feed was limited. It also allows the reallocation nutrition on ovulation rate in Scottish Blackface ewes in different body
of limited feed resources to other livestock classes, such conditions at mating. Anim. Prod. 39, 235–239.
Haydock, K.P., Shaw, N.H., 1975. The comparative yield method for esti-
as finishing lambs. Furthermore, the short joining period mating dry matter yield of pasture. Aust. J. Exp. Agric. Anim. Husb. 15,
associated with synchronisation facilitates the “focussed” 663–670.
feeding of pregnant ewes such as “colostrum feeding” Holst, P.J., Kemp, D.R., Goodacre, M., Hall, D.G., 1998. Summer lamb pro-
duction from puna chicory (Cichorium intybus) and lucerne (Medicago
(Martin et al., 2004). However, producers in extensive
sativa). Proc. Aust. Soc. Anim. Prod. 22, 145–148.
sheep production systems are more likely to use short- Jefferies, B.C., 1961. Body condition scoring and its use in management.
term feeding if synchronisation is not required. Although Tas. J. Agric. 32, 19–21.
Kohno, H., Okamoto, C., Iida, K., Takeda, T., Kaneko, E., Kawashima, C.,
this study has shown the principle that short-term feeding
Miyamoto, A., Fukui, Y., 2005. Comparison of estrus induction and
of live pasture increases ovulation rates in synchronised subsequent fertility with two different intravaginal devices in ewes
ewes, its effectiveness in unsynchronised ewes is not during the non-breeding season. J. Reprod. Dev. 51, 805–812.
known. Knight, T.W., Oldham, C.M., Lindsay, D.R., 1975. Studies in ovine infertility
in agricultural regions of Western Australia: The influence of a sup-
plement of lupins (Lupinus angustifolius cv. Uniwhite) at joining on the
5. Conclusion reproductive performance of ewes. Aust. J. Agric. Res. 26, 567–575.
Lightfoot, R.J., Marshall, T., Croker, K.P., 1976. Effects of rate and duration
of lupin grain supplementation on ovulation and fertility of Merino
Short-term feeding of synchronised ewes on live pas- ewes. Proc. Aust. Soc. Anim. Prod. 11, 5.
ture is a method of increasing ovulation rate in ewes Martin, G.B., Milton, J.T.B., Davidson, R.H., Banchero Hunzicker, G.E., Lind-
without risking triplet pregnancies. It may be a more cost- say, D.R., Blache, D., 2004. Natural methods for increasing reproductive
efficiency in small ruminants. Anim. Reprod. Sci. 82/83, 231–245.
effective alternative to both longer-term grazing or lupin Nottle, M.B., Kleeman, D.O., Seamark, R.F., 1997. Effect of previous under-
supplementation, which has been shown to give highly nutrition on the ovulation rate of merino ewes supplemented with
variable responses, where suitable pastures already exist lupin grain. Anim. Reprod. Sci. 49, 29–36.
Nottle, M.B., Seamark, R.F., Setchell, B.P., 1990. Feeding lupin grain for six
in the grazing system. However, it is also important to cal-
days prior to a cloprostenol-induced luteolysis can increase ovulation
culate whether the extra lambs born will lead to increased rate in sheep irrespective of when in the oestrous cycle supplemen-
profit margins after considering the cost of synchronisa- tation commences. Reprod. Fertil. Dev. 2, 189–192.
Oldham, C.M., Lindsay, D.R., 1980. Laparoscopy in the ewe: a photographic
tion. On the other hand, these costs may be out-weighed
record of the ovarian activity of ewes experiencing normal or abnor-
by the benefits that growing perennial rather than annual mal oestrous cycles. Anim. Reprod. Sci. 3, 119–124.
pastures have to the local environment in their ability to Oldham, C.M., Allen, G.M., Fortune, J.A., 1994. Production from cows and
reduce groundwater discharge. Further studies are needed calves set-stocked on tagasaste, a perennial leguminous fodder shrub.
Proc. Aust. Soc. Anim. Prod. 20, 85–88.
to define optimum quantities of live pasture pre- and post- Payne, R.W., Harding, S.A., Murray, D.A., Soutar, D.M., Baird, D.B., Welham,
grazing, to determine whether the response varies with S.J., Kane, A.F., Gilmour, A.R., Thompson, R., Webster, R., Tunnicliffe
248 B.J. King et al. / Animal Reproduction Science 121 (2010) 242–248

Wilson, G., 2006. Genstat Release 9 Lawes Agricultural Trust, Rotham- in ewes to increased availability of glucose, acetate and amino acids.
stead. Reprod. Fertil. Dev. 1, 117–125.
Ramirez-Restrepo, C.A., Barry, T.N., Lopez-Villalobos, N., Kemp, P.D., Har- Thomas, D.T., Milton, J.T.B., Revell, C.K., Ewing, M.A., Dynes, R.A., Murray,
vey, T.G., 2005. Use of Lotus corniculatus containing condensed tannins K., Lindsay, D.R., 2010. Preference of sheep among annual legumes is
to increase reproductive efficiency in ewes under commercial dryland more closely related to plant nutritive value as plants mature. Anim.
farming conditions. Anim. Feed Sci. Technol. 121, 23–43. Prod. Sci. 50, 114–123.
SCA (Ed.), 1990. Feeding standards for Australian livestock: Ruminants. Upjohn, B., Parker, M., Kemp, D., 2005. Chicory (Chicorium intybus), Agnote
CSIRO, Melbourne. DPI-398, 3rd ed. NSW Department of Primary Industries.
Scaramuzzi, R.J., Campbell, B.K., Downing, J.A., Kendall, N.R., Khalid, M., Vinoles, C., Forsberg, M., Martin, G.B., Cajarville, C., Repetto, J., Meikle, A.,
Munoz-Gutierrez, M., Somchit, A., 2006. A review of the effects of 2005. Short-term nutritional supplementation of ewes in low body
supplementary nutrition in the ewe on the concentrations of repro- condition affects follicle development due to an increase in glucose
ductive and metabolic hormones and the mechanisms that regulate and metabolic hormones. Reproduction 129, 299–309.
folliculogenesis and ovulation rate. Reprod. Nutr. Dev. 46, 339–354. Vinoles, C., Meikle, A., Forsberg, M., 2004. Accuracy of evaluation of ovarian
Smith, J.F., Jagusch, K.T., Brunswick, L.F.C., Kelly, R.W., 1979. Coumestans structures by transrectal ultrasonography in ewes. Anim. Reprod. Sci.
in lucerne and ovulation in ewes. NZ J. Agric. Res. 22, 411–416. 80, 69–79.
Smith, J.F., Stewart, J.F., 1990. Effects of nutrition on the ovulation rate Vinoles, C., Meikle, A., Martin, G.B., 2009. Short-term nutritional treat-
of ewes. In: Oldham, C.M., Martin, G.B., Purvis, I.W. (Eds.), Reproduc- ments grazing legumes or feeding concentrates increase prolificacy
tive Physiology of Merino Sheep – Concepts and Consequences, School in Corriedale ewes. Anim. Reprod. Sci. 113, 82–92.
of Agriculture (Animal Science). The University of Western Australia, Warn, L., Webb Ware, J., Salmon, L., Donnelly, J., Alcock, D., 2006. Analysis
Perth, pp. 85–101. of the profitability of sheep wool and meat enterprises in southern
Stewart, R., Oldham, C.M., 1986. Feeding lupins to ewes for four days dur- Australia. Final Report for Project 1.2.6., Sheep Co-operative Research
ing the luteal phase can increase ovulation rate. Proc. Aust. Soc. Anim. Centre.
Prod. 16, 367–370. Wilkins, J.F., 1997. Method of stimulating ovulation rate in Merino ewes
Teleni, E., Rowe, J.B., Croker, K.P., Murray, P.J., King, W.R., 1989. Lupins may affect conception but not embryo survival. Anim. Reprod. Sci. 47,
and energy-yielding nutrients in ewes. II. Responses in ovulation rate 31–42.
Animal Reproduction Science 121 (2010) 249–258

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Comparative studies on bull and stallion seminal DNase activity


and interaction with semen extender and spermatozoa
Abdorrahman S. Alghamdi a,∗ , Bethany J. Funnell b , Scott L. Bird b , G. Cliff Lamb c ,
Aaron K. Rendahl d , Patrick C. Taube e , Douglas N. Foster f
a
100 E. Normal St., Department of Agricultural Sciences, Truman State University, Kirksville, MO 63501, United States
b
North Central Research and Outreach Center, University of Minnesota, Grand Rapid, MN, United States
c
North Florida Research and Education Center, University of Florida, Marianna, FL, United States
d
School of Statistics, University of Minnesota, St. Paul, MN, United States
e
Pfizer Animal Health Inc., Kalamazoo, MI, United States
f
Department of Animal Science, University of Minnesota, St. Paul, MN, United States

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

Article history: We performed a series of comparative studies of bull and stallion seminal plasma (SP) and its
Received 17 August 2009 role on sperm–neutrophil binding as well as the interaction between semen extender and
Received in revised form 8 June 2010
seminal DNase. Because of contrasting roles of SP on sperm–neutrophil binding between
Accepted 14 June 2010
horses and cattle, it was suspected there were some species-specific differences on sperm
Available online 30 June 2010
interaction with SP proteins due to the variations in the natural location of semen deposition
(uterus compared to vagina). Bull frozen-thawed sperm removed from egg yolk extender
Keywords:
showed similar results to fresh sperm, but this also caused extensive sperm agglutination
Bull
Stallion unless SP or egg yolk was included. If similar agglutination occurs after AI with frozen bull
Seminal plasma semen, it could interfere with sperm transport or sperm functions. Commonly used bull
Sperm semen extenders were poor media for seminal DNase activity on plasmid DNA degradation,
Neutrophil raising the prospect that the same may be true with other SP factors important to fertility.
DNase DNase activity per mg SP protein of bulls was less than that of horses (P < 0.05), but DNase
Semen extender activity associated with bull sperm was greater (P < 0.05) indicating a different affinity of
Binding DNase to spermatozoa. This could be related to the fact that bull sperm naturally migrate
from the vagina to the uterus leaving the bulk of SP behind. In such migration, sperm
cells needed to carry DNase and other SP factors along. Incorporation of egg yolk in bull
semen and introducing SP into the uterus of cattle with current AI protocols may contribute
to reduced fertility. Modifications of semen extender and/or semen processing should be
examined to allow sperm cells a maximum potential for fertilization.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction et al., 2007; Calvete et al., 1996; Lazarevic et al., 1995; Nauc
and Manjunath, 2000; Penna et al., 2007; Robertson, 2007;
Seminal plasma contains many cytokines, hormones, Sharkey et al., 2007; Therien et al., 1995, 1997). While bull
enzymes, and growth factors that modify various physi- epididymal sperm fertilize the egg, fertilization rate was
ological and immunological functions in different species 10% greater for ejaculated sperm (Amman and Griel, 1974),
(Alghamdi and Foster, 2005; Alghamdi et al., 2004; Aljabari and non-return rate was 69% and 75% for epididymal and
ejaculated sperm, respectively, but was not statically sig-
nificant (Igboeli and Foote, 1968). In human and horses,
∗ Corresponding author. Tel.: +1 660 785 4548. ejaculation and SP improved fertility rate compared to epi-
E-mail address: abdo@Truman.edu (A.S. Alghamdi). didymal sperm (Masaya et al., 1996; Morris et al., 2002;

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.003
250 A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258

Heise et al., 2010). Bull semen is naturally deposited into 2.1. Frozen semen
the vagina and sperm migrate into the uterus leaving
the bulk of the SP behind, but stallion sperm and SP are Frozen bull semen from young beef sires was purchased
deposited directly into the uterus. The site of semen depo- from Select Sires (Plain City, OH; DeJarnette et al., 2000)
sition results in different barriers for sperm transport to and only samples with a post-thaw motility of ≥40% were
the site of fertilization including neutrophil influx into used. Semen from stallions of known fertility was frozen as
the female reproductive tract (FRT; Alghamdi et al., 2001, previously described (Alghamdi et al., 2002) and all sam-
2004; Mattner, 1968; Rozeboom et al., 1998; Taylor, 1982; ples had a minimum of 35% post-thaw motility. The data
Tremellen et al., 1998). While neutrophils combat micro- reported here on frozen semen included only sperm sam-
bial contamination and eliminate excess/dead sperm, their ples that were first removed from freezing extender by a
presence at the time of semen deposition reduces fertil- ninefold dilution followed by centrifugation to remove the
ity including in horses and cattle (Alghamdi et al., 2004; freezing extender that included egg yolk, glycerol and SP.
Kasimanickam et al., 2005, 2004; Matilsky et al., 1993; Ou
and Su, 2000; Rozeboom et al., 2000). SP improves the fer- 2.2. Seminal plasma preparation
tility of stallion sperm deposited in the neutrophil-laden
uteri by reducing sperm–neutrophil binding (Alghamdi et Semen (Bos taurus) was collected by electro ejacula-
al., 2004; Alghamdi and Foster, 2005). tion from five Angus bulls (18–24 months old) that passed
In contrast to horses, bull sperm binding to neutrophils a breeding soundness examination (BSE) and were used
and neutrophil extracellular traps (NETs) formation were for breeding and in vitro fertilization at the University of
lesser in the absence than in the presence of SP or SP pro- Minnesota North Central Research and Outreach Center.
tein (SP-P; Alghamdi et al., 2009). Bull sperm selectively The collection method of semen for SP preparation did not
bind certain SP factors, including seminal DNase, and carry influence these data because identical results were found
them into the uterus and oviduct to help accomplish their from SP collected with artificial vagina (data not shown).
presumed roles (Kawano and Yoshida, 2007; McCauley All bull semen samples had excellent motility (raw semen
et al., 1999; Miller et al., 1990). Extension of bull semen examined immediately after collection) and had a mini-
always targets sperm number resulting in different pro- mum of 80% motility after centrifugation. All bulls were
portions of SP in the final insemination dose (Bergeron et housed in a small lot without access to pasture with an
al., 2004), and semen extenders commonly contain pro- open-faced barn for shelter. Semen (Equus caballus) was
tein and lipoprotein not naturally found in semen (e.g. collected by artificial vagina from three Arabian stallions
egg yolk, milk). This is especially true for preserved semen (5–30 years old) of normal fertility, which were housed in
to help sperm withstand cold shock and preserve mem- box stalls. The SP was isolated by centrifugation of undi-
brane integrity. Egg yolk replaces/reduces sperm binding luted semen at 400 × g for 10 min to remove the bulk of the
to some SP factors, interferes with assays performed on sperm then at 3000 × g for 20 min to remove the remaining
sperm (Alghamdi et al., 2009; Amirat et al., 2005; Bergeron sperm.
et al., 2004; Wall and Foote, 1999), and stimulates antibody
production in the FRT, which was associated with reduced 2.3. Seminal plasma protein isolation
fertility in cows (Griffin et al., 1971, 1974; Coulter et al.,
1976). Pooled SP from each of three males (bulls or stallion)
The different locations of natural semen deposition was prepared as above, treated with proteinase inhibitor
between bulls and stallions may explain the contrasting cocktail (Calbiochem, Cat #539131) and frozen at −20 ◦ C
role of SP on sperm–neutrophil binding between these two until all samples had been collected. The samples were
species. The ability of egg yolk to negate the role of SP thawed on ice, buffered with 2× Dulbecco phosphate
on sperm–neutrophil binding (Alghamdi et al., 2009) and buffered saline without Ca and Mg (PBS, pH 7), and total
to reduce sperm binding to some SP factors necessary for SP proteins were precipitated by ammonium sulfate (33%
upstream function (Bergeron et al., 2004) indicates a role (w/v)). Precipitated SP protein (SP-P) was collected by cen-
for semen extenders on sperm interactions with SP factors trifugation at 3000 × g for 20 min, re-suspended in PBS, and
important for fertility. We hypothesized semen extenders dialyzed (3500 MWCO; Slide-A-Lyzer; Pierce) against two
would influence seminal DNase activity in vitro and this changes of 4 l PBS for 2 h each followed by a third change
enzyme would have greater binding to bull than stallion over night. Protein concentration was determined (Protein
sperm. We used different experiments to investigate the Assay, BioRad) and the samples were divided to aliquots
interactions between frozen bull sperm with neutrophils and frozen at −80 ◦ C until needed.
and the DNase activity and binding to sperm incubated in
different media. 2.4. Semen extenders

Bull semen was diluted with different extenders


2. Materials and methods depending on the experiment: (1) Tris–citrate (Tris); (2)
Tris–citrate–egg yolk (EY; similar to 1 except that it
All sample collection was conducted in accordance to included 20% egg yolk by volume); (3) skim milk (milk;
the guidelines of the Institutional Animal Care and Use heated at 95 ◦ C for 10 min and cleared by centrifugation at
Committee, Research Subjects’ Protection Program at the 10,000 × g for 5 min with lipids floating on top removed by
University of Minnesota. aspiration using a wide mouth pasture pipette); and (4)
A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258 251

AndroMed (Minitube of America, Verona, WI, Reference MgCl2 10 mM; CaCl2 mM; pH 7.2; Hashida et al., 1982). The
# 13503-1250), which substitutes egg yolk with soybean reaction was performed at room temperature for 15 min
extracts (for semen extender composition see: Kommisrud followed by inactivation of DNase with 50 ␮M EGTA. To
et al., 1996; Amirat et al., 2005; Bergeron et al., 2004). All determine the role of semen extender on DNase activity,
extenders were glycerol-free except those used for semen stallion and bull SP (10% (v/v)) and SP-P (2 mg ml−1 ) were
freezing, but the thawed sperm were removed from the examined with plasmid DNA. The buffer was replaced by
freezing extender and suspended in glycerol-free exten- the corresponding extenders and the reaction was con-
ders (see Section 2.1 above). ducted as previously described. Similar experiments were
performed to examine the effects of divalent cations (Mg2+
2.5. Neutrophil isolation from peripheral blood and Ca2+ ) on DNase activity using these extenders. In other
experiments, SP and SP-P were replaced by purified cat-
Heparinized blood from healthy Angus cows (3½–5½ tle pancreatic DNase I (Roche) to determine whether the
years old) or heifers (12–18 months old) was subjected to results obtained with the different extenders were due to
centrifugation at 600 × g for 10 min and the blood plasma their composition or potential interactions between exten-
was removed keeping the buffy coat and RBC layers. The ders and SP/SP-P. In all the experiments above, treated DNA
red blood cells were lysed with four volumes of sterile was then separated on a 1.2% agarose gel. Plasmid DNA
water for 40 s and the isotonicity was restored by dilu- samples treated with 2 mg ml−1 BSA served as a negative
tion with an equal volume of 2× PBS. The WBC pellet was control.
re-suspended in PBS and layered over lymphocyte sepa-
ration media (LSM; Mediatech Inc., Herndon, VA). After 2.6.3. Experiment 3: DNase binding to sperm cells
centrifugation at 500 × g for 30 min, the LSM and buffy To examine sperm binding to seminal DNase, frozen-
coat (peripheral blood mononuclear cells; PBMCs) were thawed bull and stallion sperm were washed from the
removed and pelleted neutrophils were then washed, re- freezing extender by a 1:9 dilution followed by centrifu-
suspended in PBS, counted, adjusted to 1 × 108 cell ml−1 , gation with PBS containing 20% milk extender (v/v). Sperm
and used within 1.5 h after collection. cells (3 × 106 ) from each species were set aside to be tested
for DNase activity without further treatment. The remain-
2.6. Experiments ing sperm cells were divided (within species) to include
the same sperm number in 100 ␮l reaction for the fol-
2.6.1. Experiment 1: sperm–neutrophil binding lowing treatments: (1) sperm alone; (2) sperm with 20%
We have previously shown a profound influence of SP (v/v); and (3) 20% SP without sperm (washing con-
SP, SP-P, and EY on fresh bull sperm–neutrophil binding trol). After incubation at 37 ◦ C for 30 min, 900 ␮l of DNase
(Alghamdi et al., 2009) and because egg yolk is almost buffer was added, the samples were subjected to cen-
exclusively used with frozen bull semen, the binding trifugation at 2000 × g for 5 min, and the supernatant was
between neutrophils and frozen-thawed bull sperm was removed leaving 20 ␮l in the bottom of the tube. In the first
investigated. Frozen-thawed semen was diluted at a ratio group, 10 ␮l of DNase buffer containing 2 ␮g of plasmid
of 1:9 with Tris, EY, milk or AndroMed extenders and sperm was added and incubated at room temperature for 15 min
were recovered by centrifugation and used for the bind- before stopping DNase activity by the addition of EGTA. In
ing assay. Frozen-thawed sperm cells (3 × 107 ml−1 ) were the second group, 980 ␮l DNase buffer was added to the
incubated with either 0% or 40% SP (v/v) using the four 20 ␮l left from the first wash and subjected to a second
semen extenders for 30 min at room temperature. Neu- centrifugation and treated as described for the first group.
trophils (5 × 106 ml−1 ) were added to sperm preparations The third group was washed a third time before adding
and incubated for 3 h. Sperm binding was evaluated at 30 the plasmid DNA. After EGTA addition, samples were sub-
and 180 min after mixing the two cell types as the percent- jected to centrifugation at 10,000 × g for 2 min and 20 ␮l
age of neutrophils that bound to at least one sperm cell of the supernatant was separated on an agarose gel as
counted under light microscope at a magnification of 400× above.
(Alghamdi et al., 2009).
2.7. Time lapse analysis
2.6.2. Experiment 2: DNase activity in SP and SP-P
In horses, SP digests neutrophil extracellular traps To show the effect of extender and SP on NETs forma-
(NETs) and frees entangled sperm resulting in reduced tion, we used the same method described for binding. At
binding (Alghamdi and Foster, 2005). However, bull SP the 180-min time point, samples were treated with Sytox
increased neutrophil binding to fresh (Alghamdi et al., Green and 25 ␮l was placed on a glass slide and covered
2009) and frozen bull sperm (Experiment 1) suggesting with a cover slip. Time lapse analysis was performed with
seminal DNase was either absent from bull samples or was fluorescent microscopy at 1 s intervals. To show that puri-
inhibited by unknown factors. Because a large proportion fied DNase could digest the formed NETs, an extensively
of sperm binding was due to NETs, bull SP DNase activ- formed sperm–neutrophil cluster was placed on a Petri
ity in comparison to SP from stallions was investigated as dish and covered with 20 ␮l DNase buffer containing 30
previously described (Alghamdi and Foster, 2005). Briefly, units of purified DNase and Sytox Green. The sample was
crude SP-P from bulls and stallions (2, 1, and 0.5 mg ml−1 ) then covered with mineral oil and time lapse analysis was
were used with 2 ␮g plasmid DNA substrate (5 kb) in performed at 5 min intervals. Un-stimulated neutrophils
a DNase digestion buffer (Tris–HCl 50 mM; NaCl 5 mM; were used as a negative control to demonstrate the absence
252 A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258

of NETs formation and time lapse was performed every uated in the four semen extenders commonly used with
15 min. bull semen as well as a DNase buffer using a plasmid DNA
substrate with SP and SP-P from both stallions and bulls.
2.8. Statistical analysis Fig. 2d shows that all four extenders served as poor media
for bull seminal DNase activity compared to the DNase
Data were analyzed using the R 2.5.1 program (R buffer. Although AndroMed showed some degraded DNA,
Development Core Team, 2007) with Repeated Measure the DNA smear did not increase even when the incubation
ANOVA. The model included the extender, SP, the day of time was extended for 45 min. Milk had a lesser rate of
sample collection, and time points of data collection as DNase activity as shown by the presence of a DNA smear
independent variables. The day of sample collection rep- at 45 min (data not shown), although not at 15 min. Stal-
resents different frozen semen and neutrophil samples lion seminal DNase, however, was not inhibited by milk
for each independent experiment. Means were compared extender (the most commonly used semen extender in this
with Tukey HSD method to find simultaneous 95% con- species), had reduced activity in egg yolk extender, and
fidence intervals. All data are presented as means ± SE. was completely inhibited in AndroMed and Tris extenders
Sperm–neutrophil binding experiment was performed in (Fig. 2d).
three independent replicates in which semen from a dif- The poor performance of the four extenders examined
ferent bull and neutrophils from a different cow were indicated the need for modification for DNase to function.
collected for the experiment in three different days. The Because bull seminal DNase is Mg2+ - and Ca2+ -dependent
number of replications was chosen based on power anal- (Hashida et al., 1982), a simple modification would be the
ysis using variations gleaned previously (Alghamdi et al., addition of these cations to the extenders. Because DNase
2009). All other experiments were replicated in at least five buffer included 10 mM Mg2+ and 2 mM Ca2+ for optimal
different days and the figures are representative of these activity, this concentration was used as a standard, but
replications. samples at 2× and 0.5× concentrations were also included.
Bull seminal DNase activity can be improved in milk exten-
3. Results der by these cations, but no improvement was seen for the
other extenders (Fig. 3a). Because SP and SP-P contains a
3.1. Experiment 1 complex mixture of proteins, we examined purified DNase
I activity (0.1 U) on plasmid DNA suspended in the four
Similar to the results obtained with fresh spermatozoa extenders above as well as DNase buffer. Similar to stal-
(Alghamdi et al., 2009), EY extender invariably prevented lion seminal DNase (see Fig. 2d), purified DNase I activity
frozen-thawed spermatozoa from binding to neutrophils in milk extender was less than DNase buffer, but became
with either SP concentration and at either time point (Fig. 1, similar when supplemented with Ca2+ and Mg2+ (Fig. 3b).
and supplemental video 1). The absence of SP resulted in However, Tris, EY, and AndroMed all showed essentially no
a lesser (P < 0.05) sperm–neutrophil binding in all exten- DNase activity.
ders except for AndroMed at 180 min. Interestingly, when
sperm cells were removed from the freezing extender and 3.3. Experiment 3
re-suspended in any extender other than EY, more than 50%
(mean ± SE; 66.2 ± 2.2) of sperm cells bound to each other To examine the hypothesis that bull sperm cells bind
(Fig. 2a) unless SP was added, a phenomenon not observed and carry seminal DNase into the cervix and uterus, we
in any experiment performed with fresh sperm. used frozen semen that had been washed from freezing
extender and that was treated with either buffer alone or
3.2. Experiment 2 buffer and 20% (v/v) SP for 30 min at 37 ◦ C. Frozen-thawed
stallion and bull sperm cells that were removed from
DNase activity can be examined with plasmid DNA (or the freezing extender and examined directly showed less
any DNA with a defined unit size) by the appearance of (P < 0.05) DNase activity (Fig. 3c; lanes 4 and 5). A second
a DNA smear after treatment with DNase (Fig. 2b; if the wash was enough to eliminate DNase activity from these
reaction time is allowed to proceed long enough, DNA can frozen-thawed spermatozoa. However, washed sperm cells
be degraded completely). Bull and horses SP-P was com- incubated with SP had DNase activity after a single wash;
pared using a plasmid DNA substrate and it was found although stallion sperm cells incubated with SP did not
that SP from both species contained DNase, but the enzy- appear to retain DNase activity being similar in samples
matic activity per mg crude preparation was greater in SP-P without sperm cells (washing control). When those sperm
from stallions than from bulls (Fig. 2c; P < 0.05). Because cells were washed twice before incubation with plasmid
we previously showed that stallion seminal DNase reduced DNA, stallion sperm retained less DNase activity than that
sperm–neutrophil binding (Alghamdi and Foster, 2005), of bulls. After three washes, stallion sperm showed no DNA
it was paradoxical to find that bull SP increased bind- degradation, but bull sperm still contained DNase activity
ing despite the fact that it contained DNase activity. It indicating bull sperm bind to DNase with greater affinity
was suspected the lesser DNase activity in bull SP may be than stallion sperm (Fig. 3c).
due to other SP molecules that inhibit DNase, interactions Based on the previous studies with stallions, it was
between semen extender and SP/SP-P, or that the removal expected bull seminal DNase would reduce sperm bind-
of bull sperm cells from SP resulted in the removal of DNase ing to neutrophils. Because the bull SP results were the
bound to sperm cells. Seminal DNase activity was first eval- opposite of those expectations, the influence of purified
A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258 253

Fig. 1. The role of semen extenders and SP on the binding of frozen bovine sperm to peripheral neutrophils in vitro. (a) Frozen-thawed sperm were washed
from freezing extender and re-suspended in the four extenders with either 0% or 40% SP (v/v) and the binding was evaluated at 30 and 180 min. Different
letters indicates a difference (P < 0.05; n = 3). (b) Frozen-thawed sperm re-suspended in EY (left panels) or Tris (right panels) with 0% SP (top panels) or 40%
SP (bottom panels) treated with Sytox Green, incubated for 2 h at room temperature and photographed at one frame/s using fluorescent microscopy. Note
the greater NETs formation with Tris extender and 40% SP (green streaks; bottom right panel). Results with AndroMed and milk are similar to those with
Tris. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)

DNase I on bull sperm–neutrophil clustering was exam- to the media. Purified DNase I digested the formed NETs
ined. The rationale was based on the ability of bull sperm and freed entangled bull sperm when DNase buffer was
to bind and carry DNase (Miller et al., 1990) into the uterus, used in a manner similar to that observed for stallions
which was supported by the observation that incubation (Fig. 4a and supplementary video 2). To ascertain that neu-
of bull sperm with SP appears to restore DNase activity trophils will not form NETs without being activated with SP,
(Fig. 3b). It is possible that some factors in whole SP may time lapse analysis was also performed with neutrophils
interfere with DNase activity in addition to the effects due alone and NET formation was virtually absent (Fig. 4b and
254 A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258

Fig. 2. (a) Agglutination of bull spermatozoa removed from freezing extender. (b) DNase activity causes a DNA smear indicating degradation. Plasmid DNA
was treated with 1, 0.1, 0.01 and 0 U of DNase I (lanes 1–4, respectively) for 15 min at room temperature. (c) DNase activity per mg crude SP-P from bull and
stallion. Different concentrations of SP-P (2, 1, and 0.5 mg) were incubated with plasmid DNA substrate for 15 min. (d) The four different extenders were
compared to DNase buffer using bull and stallion SP (10% (v/v); first lane within each extender) and SP-P (2 mg ml−1 ; second lane within each extender).
Note how milk inhibited DNase activity from the bull but not the stallion.

supplementary video 3). Unlike stallion sperm, bull sperm previous findings in horses (Alghamdi and Foster, 2005).
cells without SP did not stimulate NETs formation (Fig. 1b A plausible explanation may be provided by Lusignan et
and supplementary video 1). al. (2007) who demonstrated that whole boar SP failed
to induce sperm capacitation, but purified bull seminal
4. Discussion plasma protein (BSP) and a homologous protein (pB1) from
boar SP did. It is possible that some factor(s) in the whole
The surprising finding that seminal DNase from bull SP bull SP and SP-P masked the DNase activity. This is sup-
and SP-P did not digest NETs and that bull SP/SP-P gener- ported by the fact that purified DNase was able to digest
ally promoted sperm–neutrophil binding was contrary to NETs and release entrapped bull sperm cells, and that the
A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258 255

Fig. 3. (a) Supplementation of milk extender (but not Tris, EY or AndroMed) improved the DNase activity of bull SP. (b) Purified DNase I (0.1 U) was
incubated with plasmid DNA suspended in the following extenders: DNase buffer, Tris, EY, milk, AndroMed, and milk supplemented with Mg and Ca (lanes
1–6, respectively; control BSA in lane 7). (c) Bull but not stallion spermatozoa bind DNase from SP; (1) sperm alone; (2) sperm with SP; and (3) SP alone
(washing control). Sperm removed from freezing extender and used without further washing are shown in lanes 4 and 5. Control BSA was used in all
experiments.

DNase activity in bull SP-P was less than that of stallions. The specificity of such interactions can be appreciated by
It should be noted that SP from all species is sequestered the absence of NETs formation when cattle and horse neu-
in different organs, is mixed during ejaculation, and the trophils are incubated alone. However, when neutrophils
contents of accessory sex glands are not released at the are incubated with sperm cells, stallion SP prevents, while
same time. All these phenomena may have evolved due bull SP exacerbates binding and NETs formation.
to the presence of certain factors that mask others until Semen extender had a clear influence on the binding of
needed, or alternatively activate others at a specific time frozen-thawed sperm to neutrophils similar to that seen
or location. With the complex composition of SP we need with fresh semen (Alghamdi et al., 2009) and EY exten-
to decipher the interactions of SP, sperm cells, and the FRT. der negated the effects of SP. Frozen spermatozoa removed
256 A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258

Fig. 4. (a) Purified DNase digested NETs and freed entangled bull sperm, but this required the use of DNase digestion buffer and covered with mineral
oil. Time lapse analysis was performed under fluorescent microscopy with frames acquired every 5 min (see supplementary video 3; left and right panels
represent first and last frames, respectively). (b) Neutrophils alone do not form NETs. First frame was acquired by light microscopy (left panel) then frames
were acquired every 15 min by fluorescent microscopy (right panel; see supplementary video 3).

from the freezing extender (including egg yolk and variable incubation of these sperm cells in any extender containing
amounts of SP) resulted in low sperm–neutrophil binding, SP (or EY extender) prevented sperm agglutination pre-
but the addition of SP increased the binding significantly. sumably by replacing those SP factors that masked the
Surprisingly, the removal of frozen-thawed spermatozoa adhesion molecules. More attention needs to be focused
from freezing extender resulted in extensive sperm agglu- on this possibility due to the potential implication of such
tination unless SP was included in the extender or EY agglutination on sperm transport and fertilization. For
was re-used to suspend those spermatozoa. The ability of example, if uterine secretions resulted in the removal of EY
egg yolk molecules to replace some SP proteins or reduce from inseminated bull sperm, similar agglutination in the
their binding to sperm cells (Bergeron et al., 2004) may be uterus may lead to hindered sperm transport or increased
responsible for this agglutination. It is possible that egg yolk sperm elimination. Mounting evidence suggests that egg
replaced some SP factors and when egg yolk was removed, yolk may be counter productive to fertility and a thorough
some adhesion molecules were exposed, leading to sperm examination of this component is overdue (Griffin et al.,
agglutination. This possibility is supported by the fact that 1971, 1974; Coulter et al., 1976; Hanzen et al., 1994; Amirat
A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258 257

et al., 2004; Roasa et al., 2007; Leroy et al., 2005; Alghamdi uterine neutrophils and those with clear uteri at the time
et al., 2009). of insemination.
Stallion SP-P had greater DNase activity per mg crude
protein than that of bulls, but bull sperm cells incubated Acknowledgements
with SP retained greater DNase activity after repeated
washing. It is possible that sperm cells from stallions have This project was supported by a grant from Pfizer
evolved to rely on soluble seminal DNase for degradation Animal Health Inc., Kalamazoo MI; Grant Number 1743-
of NETs because both sperm and SP are deposited into 410-6320. We thank Linda Foster and Robyn Gangl for
the uterus. However, bull sperm cells that must migrate laboratory assistance, and Dr. Scott Madill for assistance
through the cervix leave SP behind and therefore need to with sample collection.
carry this enzyme along to assist sperm navigation through
the NETs in the uterus. The ability of bull sperm cells to Appendix A. Supplementary data
bind DNase may have resulted in the removal of DNase
from prepared SP samples and may have contributed to the Supplementary data associated with this arti-
lesser DNase activity per mg SP-P compared to horses. Nev- cle can be found, in the online version, at
ertheless, with the exception of milk extender, the other doi:10.1016/j.anireprosci.2010.06.003.
three semen extenders commonly used with bovine semen
appear to serve as poor media for bull seminal DNase activ- References
ity (at least for up to the 45 min incubation time tested).
Surprisingly, purified bovine pancreatic DNase I and equine Alghamdi, A.S., Foster, D.N., 2005. Seminal DNase frees spermato-
zoa entangled in neutrophil extracellular traps. Biol. Reprod. 73,
seminal DNase function well in milk extender showing
1174–1181.
clear DNA degradation within 15 min and the latter even Alghamdi, A., Troedsson, M.H., Laschkewitsch, T., Xue, J.L., 2001. Uterine
showed activity in EY extender with increased incuba- secretion from mares with post-breeding endometritis alters sperm
motion characteristics in vitro. Theriogenology 55, 1019–1028.
tion time. Bull seminal DNase activity improved in milk
Alghamdi, A.S., Troedsson, M.H., Xue, J.L., Crabo, B.G., 2002. Effect of semi-
extender supplemented with Ca2+ and Mg2+ , but this sup- nal plasma concentration and various extenders on postthaw motility
plementation did not improve activity in the other three and glass wool–Sephadex filtration of cryopreserved stallion semen.
extenders. Am. J. Vet. Res. 63, 880–885.
Alghamdi, A.S., Foster, D.N., Troedsson, M.H., 2004. Equine seminal plasma
The results with frozen bull semen in the present reduces sperm binding to polymorphonuclear neutrophils (PMNs)
study were similar to those with fresh semen (Alghamdi and improves the fertility of fresh semen inseminated into inflamed
et al., 2009) in that the absence of SP resulted in uteri. Reproduction 127, 593–600.
Alghamdi, A.S., Lovaas, B.J., Bird, S.L., Lamb, G.C., Rendahl, A.K., Taube, P.C.,
low sperm–neutrophil binding, while the presence of SP Foster, D.N., 2009. Species-specific interaction of seminal plasma on
increased the binding substantially and EY extender was sperm–neutrophil binding. Anim. Reprod. Sci. 114, 331–344.
capable of interfering with this binding. Furthermore, the Aljabari, B., Calogero, A.E., Perdichizzi, A., Vicari, E., Karaki, R., Lahloub, T.,
Zatari, R., El-Abed, K., Nicoletti, F., Miller, E.J., Pavlov, V.A., Al-Abed, Y.,
composition of the most commonly used bull semen exten- 2007. Imbalance in seminal fluid MIF indicates male infertility. Mol.
ders are not capable of supporting bull seminal DNase Med. 13, 199–202.
activity and maybe similar for other SP factors important to Amirat, L., Tainturier, D., Jeanneau, L., Thorin, C., Gerard, O., Courtens, J.L.,
Anton, M., 2004. Bull semen in vitro fertility after cryopreservation
fertility mandating a re-examination of these extenders as
using egg yolk LDL: a comparison with Optidyl, a commercial egg yolk
more SP factors (other than DNase) are identified and their extender. Theriogenology 61, 895–907.
functions are characterized. It is possible that bull semen Amirat, L., Anton, M., Tainturier, D., Chatagnon, G., Battut, I., Courtens, J.L.,
2005. Modifications of bull spermatozoa induced by three extenders:
deposition into the vagina has resulted in altered sperm
Biociphos, low density lipoprotein and Triladyl, before, during and
interactions with SP factors such that spermatozoa can after freezing and thawing. Reproduction 129, 535–543.
carry these factors into the uterus and oviduct while SP left Amman, R.P., Griel, L.C., 1974. Fertility of bovine spermatozoa from rete
in the vagina promotes neutrophil activities for the elimi- testis, cauda epididymids, and ejaculated semen. J. Dairy Sci. 57,
212–219.
nation of spermatozoa that failed to migrate into the uterus. Bergeron, A., Crete, M.H., Brindle, Y., Manjunath, P., 2004. Low-density
The ability of stallion SP to reduce sperm–neutrophil bind- lipoprotein fraction from hen’s egg yolk decreases the binding of the
ing may reflect the fact that semen in this species is major proteins of bovine seminal plasma to sperm and prevents lipid
efflux from the sperm membrane. Biol. Reprod. 70, 708–717.
deposited into the uterus with an altered composition of Calvete, J.J., Varela, P.F., Sanz, L., Romero, A., Mann, K., Topfer-Petersen, E.,
SP to allow sperm cells sufficient time to reach the oviduct 1996. A procedure for the large-scale isolation of major bovine seminal
by reducing and/or delaying the neutrophil elimination plasma proteins. Protein Expr. Purif. 8, 48–56.
Coulter, G.H., Foote, R.H., Schiavo, J.J., Braun, R.K., 1976. Antibodies to egg
of spermatozoa. In addition, the estrous period of cattle yolk in blood serum of rabbits and cattle and cervical mucus of cattle
is much shorter than that of horses, so the potential for inseminated artificially. Theriogenology 6, 585–589.
repeated inseminations under natural conditions in the for- DeJarnette, J.F., Barnes, D.A., Marshall, C.E., 2000. Effects of pre- and post-
thaw thermal insults on viability characteristics of cryopreserved
mer is less than for the latter. If the deposition of bull SP
bovine semen. Theriogenology 53, 1225–1238.
into the uterus (as is performed with current AI) results Griffin, J.F., Nunn, W.R., Hartigan, P.J., 1971. An immune response to egg-
in an increased or faster neutrophil influx, the increased yolk semen diluent in dairy cows. J. Reprod. Fertil. 25, 193–199.
Griffin, J.F., Hartigan, P.J., McGilligan, C.A., Nunn, W.R., 1974. Antibodies to
binding may indicate that a significant number of sperm
semen diluent and infertility in rabbits. Theriogenology 1, 55–61.
cells is eliminated before reaching the site of fertilization. Hanzen, C., Laurent, Y., Ward, W.R., 1994. Comparison of reproductive
Future research need to include testing the influence of performance in Belgian dairy and beef cattle. Theriogenology 41,
bull SP deposited in the uterus on sperm transport to the 1099–1114.
Hashida, T., Tanaka, Y., Matsunami, N., Yoshihara, K., Kamiya, T., Tanigawa,
oviduct, influx of neutrophils to the uterine lumen, and Y., Koide, S.S., 1982. Purification and properties of bull seminal plasma
ultimately the fertility rate in both cows with pre-existing Ca2+ , Mg2+ -dependent endonuclease. J. Biol. Chem. 257, 13114–13119.
258 A.S. Alghamdi et al. / Animal Reproduction Science 121 (2010) 249–258

Heise, A., Hahn, W., Volkmann, D.H., Thompson, P.N., Gerber, D., 2010. Morris, L., Tiplady, C., Allen, W.R., 2002. The in vivo fertility of cauda epi-
Influence of seminal plasma on fertility of fresh and frozen-thawed didymal spermatozoa in the horse. Theriogenology 58, 643–646.
stallion epididymal spermatozoa. Anim. Reprod. Sci. 118, 48–53. Nauc, V., Manjunath, P., 2000. Radioimmunoassays for bull seminal
Igboeli, G., Foote, R.H., 1968. Maturation changes in bull epididymal sper- plasma proteins (BSP-A1/-A2, BSP-A3, and BSP-30-kilodaltons), and
matozoa. J. Dairy Sci. 51, 1703–1705. their quantification in seminal plasma and sperm. Biol. Reprod. 63,
Kasimanickam, R., Duffield, T.F., Foster, R.A., Gartley, C.J., Leslie, 1058–6610.
K.E., Walton, J.S., Johnson, W.H., 2004. Endometrial cytology and Ou, M.C., Su, C.S., 2000. Implications of asymptomatic endocervical leuko-
ultrasonography for the detection of subclinical endometritis in post- cytosis in infertility. Gynecol. Obstet. Invest. 49, 124–126.
partum dairy cows. Theriogenology 62, 9–23. Penna, N., Mondaini, S., Amuchastegui, S., Degli Innocenti, M., Carini,
Kasimanickam, R., Duffield, T.F., Foster, R.A., Gartley, C.J., Leslie, K.E., Wal- G., Giubilei, B., Fibbi, E., Colli, M., Maggi, M., Adorini, L., 2007. Sem-
ton, J.S., Johnson, W.H., 2005. A comparison of the cytobrush and inal plasma cytokines and chemokines in prostate inflammation:
uterine lavage techniques to evaluate endometrial cytology in clin- interleukin 8 as a predictive biomarker in chronic prostatitis/chronic
ically normal postpartum dairy cows. Can. Vet. J. 46, 255–259. pelvic pain syndrome and benign prostatic hyperplasia. Eur. Urol. 51,
Kawano, N., Yoshida, M., 2007. Semen-coagulating protein, SVS2, in 524–533.
mouse seminal plasma controls sperm fertility. Biol. Reprod. 76, Roasa, L.M., Choi, Y.H., Love, C.C., Romo, S., Varner, D.D., Hinrichs, K., 2007.
353–361. Ejaculate and type of freezing extender affect rates of fertilization of
Kommisrud, E., Steine, T., Graffer, T., 1996. Comparison of fertility rates horse oocytes in vitro. Theriogenology 68, 560–566.
following insemination with different numbers of spermatozoa per Robertson, S.A., 2007. Seminal fluid signaling in the female reproductive
insemination dose of frozen bovine semen. Reprod. Domes. Anim. 31, tract: lessons from rodents and pigs. J. Anim. Sci. 85, E36–44.
359–362. Rozeboom, K.J., Troedsson, M.H., Crabo, B.G., 1998. Characterization of
Lazarevic, M., Skibinski, G., Kelly, R.W., James, K., 1995. Immunomodu- uterine leukocyte infiltration in gilts after artificial insemination. J.
latory effects of extracellular secretory vesicles isolated from bovine Reprod. Fertil. 114, 195–199.
semen. Vet. Immunol. Immunopathol. 44, 237–250. Rozeboom, K.J., Troedsson, M.H., Hodson, H.H., Shurson, G.C., Crabo, B.G.,
Leroy, J.L., Opsomer, G., De Vliegher, S., Vanholder, T., Goossens, L., Geldhof, 2000. The importance of seminal plasma on the fertility of subsequent
A., Bols, P.E., de Kruif, A., Van Soom, A., 2005. Comparison of embryo artificial inseminations in swine. J. Anim. Sci. 78, 443–448.
quality in high-yielding dairy cows, in dairy heifers and in beef cows. R Development Core Team, 2007. R: A Language and Environment for Sta-
Theriogenology 64, 2022–2036. tistical Computing. R Foundation for Statistical Computing, Vienna,
Lusignan, M.F., Bergeron, A., Crete, M.H., Lazure, C., Manjunath, P., 2007. Austria, ISNB 3-900051007-0 http://www.R-project.org.
Induction of epididymal boar sperm capacitation by pB1 and BSP- Sharkey, D.J., Macpherson, A.M., Tremellen, K.P., Robertson, S.A., 2007.
A1/-A2 proteins, members of the BSP protein family. Biol. Reprod. 76, Seminal plasma differentially regulates inflammatory cytokine gene
424–432. expression in human cervical and vaginal epithelial cells. Mol. Hum.
Masaya, K., Kiyomi, M., Mikio, N., Tomoko, H., Tsukasa, S., Akihiko, O., 1996. Reprod. 13, 491–501.
The fertilizing ability of human epididymal sperm. J. Assist. Reprod. Taylor, N.J., 1982. Investigation of sperm-induced cervical leucocytosis by
Genet. 13, 652–656. a double mating study in rabbits. J. Reprod. Fertil. 66, 157–160.
Matilsky, M., Ben-Ami, M., Geslevich, Y., Eyali, V., Shalev, E., 1993. Cervical Therien, I., Bleau, G., Manjunath, P., 1995. Phosphatidylcholine-binding
leukocytosis and abnormal post-coital test: a diagnostic and thera- proteins of bovine seminal plasma modulate capacitation of sperma-
peutic approach. Hum. Reprod. 8, 244–246. tozoa by heparin. Biol. Reprod. 52, 1372–1379.
Mattner, P.E., 1968. The distribution of spermatozoa and leucocytes in Therien, I., Soubeyrand, S., Manjunath, P., 1997. Major proteins of
the female genital tract in goats and cattle. J. Reprod. Fertil. 17, 253– bovine seminal plasma modulate sperm capacitation by high-density
261. lipoprotein. Biol. Reprod. 57, 1080–1088.
McCauley, T.C., Zhang, H., Bellin, M.E., Ax, R.L., 1999. Purification and char- Tremellen, K.P., Seamark, R.F., Robertson, S.A., 1998. Seminal transforming
acterization of fertility-associated antigen (FAA) in bovine seminal growth factor beta1 stimulates granulocyte-macrophage colony-
fluid. Mol. Reprod. Dev. 54, 145–153. stimulating factor production and inflammatory cell recruitment in
Miller, D.J., Winer, M.A., Ax, R.L., 1990. Heparin-binding proteins from the murine uterus. Biol. Reprod. 58, 1217–1225.
seminal plasma bind to bovine spermatozoa and modulate capaci- Wall, R.J., Foote, R.H., 1999. Fertility of bull sperm frozen and stored in
tation by heparin. Biol. Reprod. 42, 899–915. clarified egg yolk–Tris–glycerol extender. J. Dairy Sci. 82, 817–821.
Animal Reproduction Science 121 (2010) 259–266

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Effect of number of motile, frozen-thawed boar sperm and number of


fixed-time inseminations on fertility in estrous-synchronized gilts夽
Karl W. Spencer a , Phil H. Purdy b , Harvey D. Blackburn b , Scott F. Spiller b , Terry S. Stewart c ,
Robert V. Knox a,∗
a
Department of Animal Sciences, University of Illinois, 1207 W Gregory Dr. BLD. 165, Urbana, IL 61801, United States
b
USDA-ARS National Center for Genetic Resources Preservation, National Animal Germplasm Program, 1111 S. Mason St., Fort Collins, CO 80521-4500,
United States
c
Purdue University, 915 W State St., 3-234 Lilly Hall, West Lafayette, IN 47907, United States

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

Article history: There are advantages for use of frozen-thawed boar sperm (FTS) as a tool for preser-
Received 6 November 2009 vation and transfer of valuable genetic material, despite its practical limitations. It was
Received in revised form 11 June 2010
hypothesized that increasing the number of motile FTS and number of fixed-time artificial
Accepted 5 July 2010
inseminations (AI) would improve pregnancy rate and litter size. Semen from six boars
Available online 13 July 2010
was frozen in 0.5 mL straws at 500 × 106 cells/mL. Gilts ∼170 days of age, were induced
into estrus with PG600® and synchronized using MATRIXTM (synthetic progestagen). Fol-
Keywords:
lowing last feeding of MATRIX (LFM), gilts were checked twice daily for estrus. At onset of
Frozen sperm
Boar estrus, gilts were randomly assigned in a 3 × 2 factorial treatment design to receive 1 × 109
Pregnancy motile FTS (n = 19), 2 × 109 motile FTS (n = 19), 4 × 109 motile FTS (n = 19) in a single AI
Litter size at 32 h after onset of estrus, or 1 × 109 motile FTS (n = 18), 2 × 109 motile FTS (n = 17), or
Ovulation 4 × 109 motile FTS (n = 19) in each of the two AI at 24 and 32 h following onset of estrus.
AI Ultrasonography was performed at 12 h intervals after estrus to estimate time of ovulation.
Reproductive tracts were collected 28–34 days following AI. Estrus occurred at 139 ± 2 h
(mean ± SE) after LFM and ovulation at 33 ± 1 h following onset of estrus. Dose and num-
ber of inseminations did not interact or individually influence pregnancy rate at slaughter
(73 ± 4.2%) or numbers of normal fetuses (10.8 ± 0.5). However, number of fetuses tended
(P = 0.14) to increase with double AI but not with dose. Boar did not affect pregnancy rate
but did affect number of normal fetuses and embryonic survival (P < 0.01). Longer intervals
from insemination to ovulation reduced pregnancy rate (P < 0.05), number of normal fetuses
(P < 0.001), and embryonic survival (P < 0.01). Ovarian abnormalities at slaughter were asso-
ciated with reduced pregnancy rate (P < 0.001). The results of this experiment indicate that
a double insemination using 2 × 109 motile sperm would produce the greatest number of
piglets with fewest numbers of frozen sperm used, while double AI with 1 × 109 motile
sperm would be most practical for pig production with limited genetic resources. Fertil-
ity was also influenced by boar, interval from insemination to ovulation, and gilt ovarian
abnormalities.
© 2010 Elsevier B.V. All rights reserved.

夽 Mention of a trade name or proprietary product does not constitute a guaranty or warranty by the USDA and does not imply approval to the
exclusion of other products that may be suitable. USDA, Agricultural Research Service, Northern Plains Area, is an equal opportunity/affirmative action
employer. All agency services are available without discrimination.
∗ Corresponding author at: University of Illinois, 360 ASL, 1207 W Gregory Dr., MC-630, Urbana, IL 61801 Tel.: +1 217 244 5177; fax: +1 217 333 8286.
E-mail address: rknox@illinois.edu (R.V. Knox).

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.07.002
260 K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266

1. Introduction in fertilization rates similar to those using liquid semen


(Waberski et al., 1994).
Due to reduced fertility in comparison to liquid We hypothesized that improved pregnancy rates and
extended semen, frozen-thawed boar sperm is used in litter sizes would result from use of increased numbers of
less than 1% of all inseminations (Johnson et al., 2000). motile, frozen-thawed sperm when using multiple instead
Despite its limited use in commercial production, frozen of single timed inseminations in order to compensate for
boar sperm has potential for application if improvements variation in the time of ovulation and reduced fertile lifes-
can be achieved in post-thaw sperm quality and AI fertility. pan of frozen sperm. The objectives of this study were to
Frozen boar semen is valued for maintaining swine genet- test the impact of numbers of motile, frozen-thawed boar
ics in the cryopreserved state to allow for preservation and sperm in either a single or double AI, in relation to interval
regeneration of valued genetic lines, for providing for an from last estrous synchronization treatment to estrus, time
emergency sperm supply, and for facilitating opportuni- of ovulation, and other sources of variation that included
ties for global gene distribution (Almlid and Hofmo, 1996; boar, ovulation rate, quality of insemination, and other
Johnson et al., 2000; Eriksson et al., 2002; Roca et al., 2006; uncontrolled causes of variation.
Bailey et al., 2008).
Extensive research has been conducted on methods 2. Materials and methods
to improve post-thaw fertility of frozen boar sperm. One
major limitation is that fertility following freezing is influ- 2.1. Animals, induction and estrous synchrony, ovulation
enced by boar and ejaculate and involves variability in and insemination determination
plasma membrane integrity (Waterhouse et al., 2006). To
preserve membrane integrity and sperm viability, several The experiment was performed at the University of
freezing methods have proven successful (Larsson, 1978; Illinois swine research farm in six sequential replicates
Bwanga, 1991; Johnson et al., 2000; Grobfeld et al., 2008). from December 2007 to June 2008. All procedures uti-
Of these, computerized, controlled-rate freezing machines lized in this experiment were approved by the University
(Johnson et al., 2000; Thurston et al., 2003) allow for opti- of Illinois Institutional Animal Care and Use Committee.
mal freezing rate control for maintaining sperm membrane Terminal line gilts (n = 123, 337 × C22, PIC North Amer-
integrity (Fiser et al., 1993; Hernandez et al., 2007b). In ica, Hendersonville, TN) were moved from a finishing barn
addition, sperm freezing in straws and bags allows for more into crates in a gestation building. All gilts were given
uniform freezing and thawing with the increased surface PG600 (400 IU eCG and 200 IU hCG, Intervet/Schering-
area when compared to pellets (Pursel and Johnson, 1976; Plough, Kenilworth, NJ) for initial induction of estrus. The
Bwanga et al., 1990; Eriksson and Rodriguez-Martinez, gilts selected for this experiment ranged from 165 to
2000a,b). Improvements in freezing media, such as addi- 198 days of age and weighed between 95 and 139 kg at
tion of cholesterol (Bailey et al., 2008), seminal plasma the time of induction. Estrus detection was performed
(Hernandez et al., 2007a; Okazaki et al., 2009b) or antiox- daily using the back-pressure test while providing fence-
idants (Grobfeld et al., 2008) have been reported to have line contact with a mature boar. Ten days following
potential for improving post-thaw sperm motility and treatment with PG600, regardless of estrus expression,
acrosome integrity. stage of estrous cycle in gilts was synchronized using a
Many early studies with frozen boar sperm resulted in synthetic progestagen, MATRIX (Altrenogest 2.2 mg/mL,
reduced post-thaw quality, and fertility following insem- Intervet/Schering-Plough, Kenilworth, NJ, USA) for 14 days
ination. The decreased sperm fertility may have resulted as a top-dress on a standard sow gestation diet. After
as a consequence of method of freezing while reduced 14 days of MATRIX feeding, in replicates 1–3, gilts were
AI fertility could be attributed to variation in number of checked for estrus once daily. In replicates 4–6, PG600
inseminations, number of sperm used, and less than opti- was given to all gilts 24 h following LMF to reduce vari-
mal AI timing. In studies, the insemination doses tested ation in timing of estrus and to aid in labor scheduling
were based on the number of motile or total sperm cells per and timing for AI. Beginning on the third day following
dose with estimates for motility averaging ∼30% (Einarsson LMF, estrus detection was performed at 12 h intervals.
et al., 1973; Larsson and Einarsson, 1976). In a review of ear- Transrectal real-time ultrasonography (Aloka 500 V, Tokyo,
lier research, farrowing rates remained at ∼55% and litter Japan) was initiated 12 h following onset of estrus and con-
sizes at ∼8 pigs/litter (Johnson, 1985). More recent studies tinued at 12 h intervals to observe ovaries and estimate
have used single or double inseminations with 5 to 6 × 109 time of ovulation (Knox et al., 2002). The total number of
total sperm with post-thaw motility reported at ∼40–60% large antral follicles >5.0 mm was counted and the images
(Roca et al., 2003; Okazaki et al., 2009a). Fertility following recorded in electronic format on DVD for playback confir-
AI in more recent trials has yielded pregnancy rates >60% mation of follicle size and number. Overall, 111 animals
with litter sizes >9 pigs per litter (Hofmo and Grevel, 2000; expressed estrus after LMF and were assigned to treat-
Eriksson et al., 2002). New technologies, such as intrauter- ment. At onset of estrus, gilts were randomly assigned in
ine and deep uterine AI, may allow for increased fertility a 3 × 2 factorial treatment design to receive 1 × 109 motile
when using reduced numbers of sperm (Roca et al., 2003; FTS (n = 19), 2 × 109 motile FTS (n = 19), 4 × 109 motile FTS
Rozeboom et al., 2004). Improved AI timing is also essential (n = 19) in a single AI at 32 h after onset of estrus, or 1 × 109
because frozen boar sperm has a reduced fertile lifespan motile FTS (n = 18), 2 × 109 motile FTS (n = 17), or 4 × 109
(Einarsson and Viring, 1973; Waberski et al., 1994) and motile FTS (n = 19) in each of the two AI at 24 and 32 h
must be deposited within 4 h before ovulation to result following onset of estrus. Gilts were randomly assigned to
K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266 261

each treatment in order of estrus detection so that all treat- insemination was sent to the University of Illinois in liquid
ments would receive sperm from the same boar. The order nitrogen dry vapor containers.
for boar use was random, but we selected each boar to be
represented across each treatment before the process was 2.3. Sperm thawing and insemination
repeated for next boar. The daily experimental procedures
involved semen thawing and dose preparation (0500 and To prepare each insemination dose, straws of frozen
1700 h), ultrasonography (0800 and 2000 h), followed by boar sperm were placed in a 50 ◦ C waterbath and agitated
estrous detection (0900 and 2100 h). for 20 s for thawing. The thawed sperm were diluted with
80 mL of Androhep CryoGuard Thaw extender (Minitube of
2.2. Semen collection, evaluation, and freezing America, Verona, WI) in a 26 ◦ C waterbath. Within 30 min of
thawing, intra-cervical insemination was performed using
Terminal line (L220), excellent health, crossbred boars standard spirette or foam tipped AI catheters (Minitube of
(PIC, n = 6) with a previous history of offspring production America, Verona, WI) in the presence of a boar. Because
were selected for use in this study. A total of 3 to 7 ejaculates all animals in this experiment received an insemination at
were collected for each boar and processed for freezing. 32 h following first expression of estrus, this AI was scored
Semen was collected at a commercial PIC stud, diluted for fluid loss during AI. Insemination scoring was based on
1:4 (v/v) with 37 ◦ C Androhep Plus (Minitube of Amer- categorical 1–3 scale, with a score of 1 having excessive vol-
ica, Verona, WI), cooled to 15 ◦ C and shipped overnight ume loss during AI, 2 having moderate loss, and 3 having
to the USDA-ARS National Center for Genetic Resources no volume loss during insemination.
Preservation, National Animal Germplasm Program in Fort
Collins, CO, USA. The samples were then centrifuged for 2.4. Pregnancy classification and reproductive tract
10 min at 800 × g and the supernatant aspirated and the processing
resulting pellet evaluated for sperm concentration using
a spectrophotometer (Purdy, 2008). Motility of the ejacu- Gilts were sacrificed between days 28 and 34 of ges-
late was determined by Computer Assisted Sperm Analysis tation and reproductive tracts collected. The tracts were
(CASA, Hamilton Thorne Research, Beverly MA, USA) as pre- identified and processed for pregnancy status (0 fetus = not
viously described (Purdy, 2008). pregnant, or 1 or more fetus = pregnant), number of nor-
The ejaculates used in this experiment were frozen mal fetuses, number of degenerative fetuses (those with
using the method of Pursel and Johnson (1975) as abnormal appearance), number of corpora lutea (CL), and
modified by Purdy (2008). Briefly, following centrifuga- presence of any abnormal structures associated with the
tion, sperm pellets were re-suspended in BF5 cooling ovaries or oviducts. Abnormalities were identified as fol-
extender (CE, 52 mM TES, 16.5 mM Tris[hydroxymethyl] licular or luteal cysts (>12.9 mm) or fluid-filled oviducts.
aminomethane, 178 mM glucose, 20% egg yolk; 325 mOsm)
to 750 × 106 sperm/mL. The sperm were then equilibrated 2.5. Statistical evaluation
at 5 ◦ C for 2.5 h and diluted to 500 × 106 sperm/mL by
drop-wise addition of the BF5 freezing extender (91.5% Data were evaluated in SAS (SAS Institute, Inc., Cary,
CE, 6% glycerol, 2.5% Equex Paste, Minitube of America; NC) using a mixed model ANOVA (PROC MIXED) for all
1450 mOsm (Pursel and Johnson, 1975). The sperm was continuous response variables and PROC GENMOD for
placed into 0.5 mL CBS straws (IMV Corporation, Min- binary response variables. The continuous response vari-
neapolis, MN, USA) and frozen using a programmable ables included age and weight of gilts, number of normal
freezer (Minidigicool UJ400, IMV USA) with the follow- fetuses, quality of insemination, number of degenerative
ing freeze curve: 5 to −8 ◦ C at 20◦ /min; −8 to −120 ◦ C at fetuses, and embryonic survival (total number of fetuses
69 ◦ C/min; −120 to −140 ◦ C at 20 ◦ C/min (Purdy, 2008). Fol- divided by number of CL at slaughter). The binary response
lowing the freezing cycle the straws were placed directly variables included: pregnancy at slaughter. The MIXED and
into liquid nitrogen for storage. GENMOD models included the main effects of dose, num-
Following ejaculate freezing, the numbers of motile ber of inseminations, the interaction of dose × number of
sperm/straw were estimated by thawing a random straw inseminations, boar, AI score, and replicate. Additional con-
and evaluating motility using CASA. Thawed sperm were tinuous variables such as number of large follicles at estrus
diluted 1:5 (v/v) in BTS and warmed to 37 ◦ C and evaluated and number of CL at slaughter were included as covari-
after 10 min. The CASA used the following variables which ates to test for linear, quadratic and cubic effects. To test
were preset by the manufacturer: 30 frames acquired, for the effects of interval from insemination to ovulation
frame rate of 60 Hz, minimum contrast of 55, minimum and an abnormal ovary at slaughter on pregnancy rate and
cell size of 5 pixels, VAP cutoff of 20 ␮m, progressive min- number of normal fetuses, one-way ANOVA procedures
imum VAP cutoff of 45 ␮m/s, VSL cutoff of 5 ␮m/s, static were performed using the MIXED and GENMOD proce-
head size of 0.53–4.45, and magnification of 1.89. A min- dures, respectively.
imum of seven fields and 1000 sperm were observed for To examine the effects of the procedures used for induc-
motility analysis. The percentage of motile sperm within tion and synchronization and use of PG600 following LMF,
each straw was used to determine the number of straws one-way ANOVA was performed using PROC MIXED with
needed to create doses with the proper number of motile replicate group (1–3 or 4–6) and expression of estrus (Yes
sperm. Each dose of FTS was comprised of each of 3–7 ejac- or No) following initial PG600 treatment as class variables
ulations from a single boar. Frozen sperm to be used for in the model. The response variables included the interval
262 K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266

from LMF to estrus, duration of estrus, interval from insem- The number of large follicles (>5 mm) counted during
ination to ovulation, numbers of follicles and CL, pregnancy ultrasound and then validated by counting from playback
rate and number of fetuses. All means comparisons were of the digital recording on a digital imaging system was
performed using the LSMEANS statement in SAS and dif- correlated (r = 0.90, P < 0.0001) and averaged 19.0 ± 0.7 fol-
ferences determined using the PDIFF option. In addition, licles, but were unrelated to the number of CL at slaughter
Pearson product moment–correlation analyses were per- (17.0 ± 0.3). The AI score at 32 h following estrus did not
formed using the PROC CORR function in SAS to determine differ among doses or number of inseminations (P > 0.20)
the simple correlation relationship between the number of and averaged 2.7 ± 0.1 (3.0 = no volume loss), with 73% of
follicles >5.0 mm counted using ultrasound with the final inseminations having an AI score = 3, while 24% had an AI
number of CL counted at slaughter. score = 2, and 3% had an AI score = 1. Across all replicates
at the time of slaughter, 7.2% of gilts had ovaries that were
3. Results classified as abnormal, with six gilts having cystic struc-
tures and two gilts showing fluid-filled oviducts.
3.1. Post-thaw sperm motility
3.3. Impact of dose and AI number on pregnancy rate
A total of 26 ejaculates were frozen from the six boars and litter size
for use in this experiment with post-thaw motility aver-
aging 38%. Each insemination dose included all ejaculates The average age (168.8 ± 1.2 days) and weight
from the same boar. The post-thaw motility for each boar (114.5 ± 1.0 kg) of gilts assigned to each treatment
averaged 38 ± 2.9% for boar A (three ejaculates), 36 ± 4.0% based on estrus were not different among treatments
for boar B (five ejaculates), 46 ± 3.5% for boar C (four ejac- (>0.20).
ulates), 32 ± 2.8% for boar D (seven ejaculates), 33 ± 6.4%
for boar E (four ejaculates), and 37 ± 3.8% for boar F (three 3.3.1. Pregnancy
ejaculates). Each 0.5 mL straw contained ∼94 × 106 motile Pregnancy rate at slaughter averaged 73.0 ± 4.2%. There
sperm cells. was no interaction of dose and number of AI and no effect
of dose or number of inseminations (Table 1). There was
3.2. Response to estrous induction and synchronization no effect of boar (Table 2), but interval from insemination
to ovulation influenced pregnancy rate (P < 0.05, Table 3).
The initial pubertal response to the estrus induction Increased pregnancy rates were observed when insemi-
treatment with PG600 was 66% with an interval of 100 ± 2 h nation occurred at 4 h before ovulation or up to 8 h after
(mean ± SE) from PG600 to estrus, with estrus lasting ovulation, compared to earlier or later intervals. While
42 ± 2 h. Expression of estrus following the pubertal induc- most additional sources of variation had no effect on preg-
tion with PG600 had no effect (P > 0.20) on expression of nancy, the presence of an abnormal ovary was associated
estrus following LMF, or on final pregnancy rate. Use of with only a 12.5% pregnancy rate (P < 0.0001).
PG600 following LMF reduced the interval from LMF to
estrus (P < 0.0001) in replicates 4–6 (130 ± 3 h) compared 3.3.2. Litter size
to replicates 1–3 (148 ± 3 h). Use of PG600 following LMF There was no significant interaction of dose and number
also reduced the duration of estrus (P < 0.0005) in repli- of inseminations and no effect of dose on numbers of nor-
cates 4–6 (42 ± 3 h) compared to replicates 1–3 (52 ± 3 h). mal fetuses (Table 1). However, number of inseminations
However, the interval from onset of estrus to ovulation tended to influence numbers of normal fetuses (P = 0.14)
was not influenced by use of PG600 (P > 0.20), and aver- with double inseminations resulting in greater numbers
aged 33 ± 1 h in replicates 1–3, and 34 ± 1 h replicates 4–6. of normal fetuses (11.5 ± 0.6) compared to single insem-
PG600 increased (P < 0.0001) the numbers of large follicles inations (10.2 ± 0.6). Boar influenced numbers of normal
at estrus in replicates 4–6 compared to replicates 1–3 (22 fetuses (P < 0.01) with boar A having the smallest and
compared to 16), but there was no effect of replicate on boar D the largest litter size (Table 2). Number of normal
number of CL at slaughter (17). There were no effects of fetuses increased as insemination occurred closer to ovula-
use of PG600 following LMF on pregnancy at slaughter or tion (P < 0.001, Table 3). Additional sources of variation had
on number of fetuses. no effect on normal fetus numbers. Number of degenerat-

Table 1
Least squares means for pregnancy and litter data in response to treatment with dose of motile, frozen-thawed sperm (1 to 4 × 109 ) and number of
inseminations (single or double). Data were collected from mature gilts between 28 and 34 days of gestation.

Motile sperm (×109 ) Number of AIa Pregnancy at slaughter (%)b Normal fetuses Degenerative fetuses Embryonic survival (%)

1 1 76.4 (14/19) 9.5 ± 1.1 0.2 ± 0.4 61.5 ± 5.8


1 2 70.4 (13/19) 11.3 ± 1.2 0.0 ± 0.4 65.6 ± 6.1
2 1 71.1 (13/19) 10.4 ± 1.2 0.4 ± 0.4 67.3 ± 6.0
2 2 79.2 (14/18) 12.6 ± 1.1 0.1 ± 0.4 71.0 ± 5.7
4 1 66.1 (12/17) 10.6 ± 1.2 0.7 ± 0.4 59.2 ± 6.5
4 2 87.3 (15/19) 10.7 ± 1.1 1.0 ± 0.4 69.5 ± 5.8
a
AI occurring at 32 h after detection of estrus for single AI and at 24 and 32 h for double AI.
b
Numbers in parentheses are proportions of gilts.
K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266 263

Table 2
Least squares means for pregnancy and litter data in response to the main effect of boar in GENMOD and MIXED models following insemination with 1 to
4 × 109 motile, frozen boar sperm in a single or double AI. Data were collected from mature gilts between 28 and 34 days of gestation.

Boar Pregnancy at slaughter (%) Normal fetuses Degenerative fetuses Embryonic survival (%)

A 78.6 (15/20) 7.2 ± 1.1z 0.4 ± 0.4 42.9 ± 6.1z


B 77.7 (14/18) 9.8 ± 1.1x 0.1 ± 0.4 74.1 ± 6.1xy
C 73.1 (13/19) 10.8 ± 1.1xy 0.1 ± 0.4 65.1 ± 6.2xy
D 71.4 (13/18) 13.8 ± 1.2y 0.5 ± 0.4 75.8 ± 6.6y
E 83.9 (15/18) 12.6 ± 1.1y 0.4 ± 0.4 74.9 ± 6.0xy
F 65.7 (11/18) 10.9 ± 1.3xy 1.1 ± 0.4 61.3 ± 6.8x
xyz
Different superscripts within a column indicate differences (P < 0.05). Numbers in parentheses are proportions of gilts.

Table 3
Least squares means resulting from GENMOD and MIXED models for pregnancy and litter data in response to estrus to ovulation interval following
insemination with 1 to 4 × 109 frozen boar sperm in a single or double AI. Data were collected from mature gilts between 28 and 34 days of gestation.

Estrus to ovulation (h)a Interval from last AI at 32 h Pregnancy at Normal fetuses Degenerative Embryonic survival (%)
to ovulationb slaughter (%) fetuses

12 +20 50.0 (2/4)xz 15.0 ± 3.0y 0.0 ± 1.0 75.5 ± 16.8y


24 +8 80.0 (27/35)y 12.1 ± 0.8xy 0.4 ± 0.4 76.0 ± 4.7y
36 −4 88.1 (37/43)y 11.1 ± 0.7xy 0.2 ± 0.3 64.0 ± 4.1y
48 −16 57.8 (11/19)x 6.0 ± 1.3z 0.1 ± 0.5 44.3 ± 7.2x
60 −28 0.0 (0/1)z – – –

Numbers within parentheses are proportions of gilts.


x,y,z
Different superscripts within column indicate differences (P < 0.05).
– No estimate.
a
Estrous detection and transrectal ultrasonography were conducted at 12 h intervals.
b
Use of ultrasonography allowed for observation of ovulation in 101 of 111 gilts.

ing fetuses/litter averaged 0.4 ± 0.2 and was not influenced Roca et al., 2006). In the present study, dose had no effect
by dose and number of inseminations. Embryonic survival on pregnancy or litter size. From a practical standpoint,
(65.5 ± 3.0%) was not influenced by dose and number of using the least amount of sperm would have the greatest
inseminations or other variables but was affected by boar advantages. Our test with 1 × 109 motile sperm in single
(P < 0.005, Table 2) and interval from insemination to ovu- or double AI resulted in pregnancy rates ≥70% and num-
lation (P < 0.005, Table 3). bers of normal fetuses between 9.5 and 11.3. Most of the
treatments resulted in acceptable fertility for use of frozen
4. Discussion boar sperm. Results in the present study are similar to
others where greater numbers of sperm were used and
The overall pregnancy rates (73%) and number of normal with comparable numbers of gilts (Eriksson and Rodriguez-
fetuses (10.8) observed in the present study were some- Martinez, 2000a), and also where greater numbers of sperm
what higher than expected. It is not clear whether the gilts and even larger numbers of sows were used (Eriksson et
in this study would have actually maintained pregnancy to al., 2002). The effect of sperm dose has been tested for both
term and whether the normal fetus numbers would survive liquid and frozen boar sperm. In some frozen boar sperm
to be liveborn pigs, but assessments for fetal and ovarian studies, altering dose had variable effects for pregnancy
measures for normality indicated there would be few lim- and litter size responses (Einarsson et al., 1973; Larsson
itations. The aim of this study was to demonstrate that et al., 1977; Johnson, 1985). Yet in certain studies with liq-
increasing the dose of motile, frozen-thawed sperm with uid semen (Watson and Behan, 2002) or with small dose,
double AI would result in acceptable pregnancy rates and or deep uterine insemination (DUI) of frozen sperm, the
litter sizes by compensating for reduced sperm quality and effects of dose were quite clear (Bolarín et al., 2006). The
variation in time of ovulation. There was no indication that reasons for the discrepancies among studies may reflect
dose of frozen sperm interacted with numbers of insemina- differences in any of the following variables: actual sperm
tions to compensate for poor sperm fertility or variation in dose (motile or total sperm used), number of insemina-
time of ovulation. However, double insemination tended to tions, use of single sire or pooled semen, boar fertility, AI
improve litter size compared to single insemination and as timing, and the reproductive maturity and management of
expected, interval from insemination to ovulation affected the animal used.
both pregnancy rate and litter size. Number of inseminations did not affect final pregnancy
Collectively it would appear that the technology for rate, but did tend to influence number of normal fetuses. It
frozen boar sperm use can be reliable for producing preg- was surprising that there were limited effects of number of
nancy rates at ∼70% and litter sizes of ∼10 pigs. Most inseminations, since a double AI system of frozen-thawed
studies testing fertility of frozen-thawed boar sperm uti- boar sperm had previously shown greater non-return rates
lize frozen sperm numbers equivalent to 2.5 to 3.0 × 109 and increased litter sizes in studies with fewer (Einarsson
motile cells or 5 to 6 × 109 total sperm with similar fer- et al., 1973) and even more animals tested (Reed, 1985).
tility outcomes shown in the present trial (Johnson, 1985; In contrast to the present study, it is possible that the ear-
264 K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266

lier methods of cryopreservation allowed for compensable In the present study, time of ovulation averaged 33 h. We
fertility in response to number of inseminations. How- observed that 34% of gilts had ovulations by 24 h, 42% by
ever, although not significant in the present study, at the 36 h, and 19% by 48 h. As a result, the majority of insemina-
greater doses, pregnancy rates were greater, and this is tions occurred close to ovulation, which is important since
similar to farrowing rate increases that have been noted frozen swine sperm has a shortened fertile lifespan in utero
with double AI compared to single AI in sows (Almlid et (Waberski et al., 1994). In the present study, interval from
al., 1987). Yet only a few studies have been conducted that estrus to ovulation affected pregnancy rate, litter size, and
have tested dose of frozen sperm with number of insem- embryonic survival similar to frozen sperm experiments
inations (Einarsson et al., 1973; Bathgate et al., 2008). In using DUI (Bolarín et al., 2006). These fertility measures
these studies, regardless of the AI method used (cervical or were greatest when gilts ovulated by 24 and 36 h. For gilts
DUI), results indicate that more inseminations at greater that ovulated by 48 h, pregnancy rate dropped to ∼58% and
doses yield greater fertility. litter size declined to 6.0 fetuses. Collectively, all studies
In the present experiment there was no clear choice show the importance of frozen sperm AI occurring close to
for optimizing final pregnancy establishment or litter size. the time of ovulation (Waberski et al., 1994; Wongtawan et
The greatest pregnancy rates were observed with 2 and al., 2006). Perhaps in the future, synchronization of ovula-
4 × 109 motile sperm with double AI, while the largest tion could be used to prevent delayed ovulation when using
litter sizes occurred with 1 and 2 × 109 motile sperm frozen sperm (Okazaki et al., 2009a).
in a double AI. To examine the fertility and sperm use Although not novel, it is still important to account for the
efficiency, fertility indices were created for total number fact that in this and other studies, boar affects the number
of normal pigs produced/treatment (final pregnancy rate of normal fetuses, and embryonic survival (Almlid et al.,
(×100) × number of normal fetuses) and numbers of motile 1987; Eriksson and Rodriguez-Martinez, 2000a). Because
sperm/piglet produced (total number of motile sperm used gilts in the present study were inseminated based on total
per treatment/total number of normal pigs produced per numbers of motile sperm, boar differences in this mea-
treatment). The fertility index revealed that double AI of 1, sure were not a factor in gilt fertility. However, variance
2, and 4 × 109 sperm produced more pigs (796, 998, and 934 in post-thaw motility was evident among boars and ejacu-
pigs) than single AI (726, 739, and 701 pigs, respectively). lates. Post-thaw motility averaged 38% and was lower than
The index for number of sperm used per pig produced indi- the 50% motility reported by (Eriksson et al., 2002; Roca et
cated that single insemination with 1 × 109 motile sperm al., 2003) while farrowing rates and litter sizes were sim-
was most efficient (0.14 × 109 sperm/pig). However, the ilar. However, poor motility does not always translate to
greatest number of pigs was produced when using 2 × 109 reduced fertility for liquid or frozen boar sperm (Flowers,
motile sperm in a double insemination. If equal weight is 1997; Eriksson et al., 2002). It should be noted that in some
given to both numbers of pigs produced and the numbers experiments, boars are selected based on post-thaw sperm
of sperm used for each pig produced, the double insemi- survival for use in frozen sperm fertility tests (Hernandez
nation at 1 and 2 × 109 motile cells allowed for the most et al., 2007a; Okazaki et al., 2009b). However, in the present
pigs produced with least amount of sperm used per pig study, boars were selected on previous history for commer-
(0.25 and 0.40 × 109 sperm/pig, respectively). For produc- cial litter production with liquid semen. As a result, we did
tion purposes a double insemination using 2 × 109 motile observe boar fertility differences similar to that reported
sperm would produce the greatest number of pigs consis- by (Flowers, 2002).
tently when using the least amount of frozen sperm. When While the present experiment focused on the primary
access to valuable genetics is limited by increased demand, fertility measures of pregnancy rate and number of nor-
a double insemination using 1 × 109 motile sperm would mal fetuses in early gestation, the timing of animal sacrifice
allow for acceptable pig production with minimal sperm allowed us to obtain data on numbers of degenerative
use. fetuses and embryonic survival. Both factors provided us
Compensating for variation in the time of ovulation was with an indication of the overall health of the litter. Addi-
a major objective of the present study and the reason why tional variables were also examined for their effects on
double insemination was included. Double insemination is fertility when using frozen boar sperm. Insemination tech-
standard practice for commercial pig production when uti- nique had no effect on fertility since the AI score was very
lizing liquid semen (Flowers and Esbenshade, 1993; Kemp good. This was important to test since the volume of the
et al., 1996). The timing system in the present study for the inseminate has been reported to affect sperm transport,
single and double AI following detection of estrus has been the sperm reservoir, and fertilization (Baker et al., 1968).
used in experiments with frozen sperm in gilts and sows Number of follicles at estrus and number of CL also did
with single (Johnson et al., 1982; Aalbers et al., 1985) and not influence litter size or measures of litter health. How-
double inseminations (Rodriguez-Martinez et al., 1996; ever, previous studies have suggested that ovulation rate
Eriksson and Rodriguez-Martinez, 2000a; Eriksson et al., could influence embryonic survival, pregnancy, and litter
2002). These published trials with gilts showed pregnancy size during early gestation (Town et al., 2005; Foxcroft et
rates of ∼60% (Rodriguez-Martinez et al., 1996; Eriksson al., 2007). Because follicle numbers and ovulation rate were
and Rodriguez-Martinez, 2000a), while the farrowing rates within normal limits, it is not surprising that we did not
for the sow trials averaged ≥70% (Eriksson et al., 2002). The observe any effect on fertility.
AI timing system in the present study was designed with In the present experiment prepubertal gilts induced
a predicted estrus to ovulation average of 36 h (Almeida with PG600 were used, with subsequent progestagen syn-
et al., 2000; Bortolozzo et al., 2005; Horsley et al., 2005). chronization to measure fertility from use of frozen boar
K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266 265

sperm. Gilt weight, age and initial estrus had no effect Taibl, B. Yantis, and D. Canaday for their excellent technical
on estrus synchronization or fertility outcomes. Despite assistance. We also extend our appreciation to the Univer-
an initial estrus induction response <70%, subsequent syn- sity of Illinois farm staff; R. Wischover, R. Alan, S. Hughes,
chronization with Matrix resulted in an estrous synchrony and D. Bidner for management of the animals used in this
response of >90% similar to other reports (Estienne et al., study.
2001). As a result, it is possible that insemination may have
occurred at pubertal estrus for ∼30% of gilts. Assessment
of our synchronized gilt model for fertility indicated nor- References
mal measures for duration of estrus, interval from estrus
to ovulation, numbers of large follicles at estrus, and num- Aalbers, J.G., Johnson, L.A., Aalbers-Smit, E.A., Rademaker, J.H.M., 1985.
bers of CL (Bracken et al., 2003; Horsley et al., 2005; Breen Motility acrosome morphology and fertilizing capacity of boar sper-
matozoa frozen in pellets and straws. In: Johnson, L.A., Larsson, K.
et al., 2006). The effect of PG600 on reducing the interval (Eds.), Deep Freezing Boar Semen. Proc. First Int. Conf. Deep Freezing
to estrus without influencing interval from estrus to ovula- of Boar Semen. Swedish University of Agricultural Sciences, Uppsala,
tion has been observed in weaned sows (Knox et al., 2002) Sweden, pp. 277–281.
Almeida, F.R., Novak, S., Foxcroft, G.R., 2000. The time of ovulation in
and suggests that the use of PG600 changes the time of ovu- relation to estrus duration in gilts. Theriogenology 53, 1389–1396.
lation from 63% of the way through estrus (replicates 1–3) Almlid, T., Hofmo, P.O., 1996. A brief review of frozen semen applica-
to 78% (replicates 4–6). Numbers of CL can be an important tion under Norwegian AI service conditions. Reprod. Dom. Anim. 31,
169–173.
component to consider when accounting for differences in Almlid, T., Stavne, S.E., Johnson, L.A., 1987. Fertility evaluation of the straw
fertility responses. To account for possible fertility effects, freezing technique for boar semen under practical artificial insemina-
we counted large follicles at estrus and correlated these to tion conditions. Reprod. Dom. Anim. 22, 193–202.
Bailey, J.L., Lessard, C., Jacques, J., Breque, C., Dobrinski, I., Zeng, W.,
digital recordings. Others have also examined this and cor-
Galantino-Homer, H.L., 2008. Cryopreservation of boar semen and its
related ultrasonographic measures with use of laparoscopy future importance to the industry. Theriogenology 70, 1251–1259.
(Bolarin et al., 2009). However, it is of interest to note Baker, R.D., Dziuk, P.J., Norton, H.W., 1968. Effect of volume of semen,
number of sperm and drugs on transport of sperm in artificially insem-
that follicle counts were not correlated with the number
inated gilts. J. Anim. Sci. 27, 88–93.
of CL at slaughter as has been reported previously (Soede Bathgate, R., Eriksson, B.M., Thomson, P.C., Maxwell, W.M., Evans, G., 2008.
et al., 1992). It is possible that this discrepancy might be Field fertility of frozen-thawed boar sperm at low doses using non-
explained by use of PG600 on follicle development or by surgical, deep uterine insemination. Anim. Reprod. Sci. 103, 323–335.
Bolarín, A., Roca, J., Rodríguez-Martínez, H., Hernández, M., Vázquez, J.M.,
the occurrence of follicle heterogeneity and the differen- Martínez, E.A., 2006. Dissimilarities in sows’ ovarian status at the
tial response to the LH surge for ovulation (Hunter and insemination time could explain differences in fertility between farms
Wiesak, 1990; Knox, 2005). Finally, in the present study, when frozen-thawed semen is used. Theriogenology 65, 669–680.
Bolarin, A., Vazquez, J.M., Parrilla, I., Vazquez, J.L., Martinez, E.A., Roca, J.,
it was noted that 7% of gilts had cystic ovaries or fluid- 2009. Validation of trans-rectal ultrasonography for counting preovu-
filled oviducts. In either case, this had a negative impact on latory follicles in weaned sows. Anim. Reprod. Sci. 113, 137–142.
pregnancy rate and number of normal fetuses. These occur- Bortolozzo, F.P., Uemoto, D.A., Bennemann, P.E., Pozzobon, M.C., Castagna,
C.D., Peixoto, C.H., Barioni Jr., W., Wentz, I., 2005. Influence of time of
rences were unrelated to use of PG600 and these problems insemination relative to ovulation and frequency of insemination on
have been observed in mature gilts and sows (Heinonen et gilt fertility. Theriogenology 64, 1956–1962.
al., 1998) with associated reduction in fertility (Waberski Bracken, C.J., Safranski, T.J., Cantley, T.C., Lucy, M.C., Lamberson, W.R.,
2003. Effect of time of ovulation and sperm concentration on fertil-
et al., 2000; Castagna et al., 2004).
ization rate in gilts. Theriogenology 60, 669–676.
Breen, S.M., Rodriguez-Zas, S.L., Knox, R.V., 2006. Effect of altering dose of
5. Conclusion P.G. 600 on reproductive performance responses in prepubertal gilts
and weaned sows. Anim. Reprod. Sci. 95, 316–323.
Bwanga, C.O., 1991. Cryopreservation of boar semen i: a literature review.
There was no interaction of dose and number of insem- Acta Vet. Scand. 32, 431–453.
inations when using frozen boar sperm on fertility. The Bwanga, C.O., De Braganca, M.M., Einarsson, S., Rodriguez-Martinez, H.,
results of this study indicate that double insemination with 1990. Cryopreservation of boar semen in mini- and maxi-straws. Zen-
tralbaltt fur veterinarmedizin reihe a 37, 651–658.
1 to 2 × 109 motile, frozen-thawed boar sperm could be Castagna, C.D., Peixoto, C.H., Bortolozzo, F.P., Wentz, I., Neto, G.B., Ruschel,
used to achieve acceptable pregnancy rates and litter sizes F., 2004. Ovarian cysts and their consequences on the reproductive
with the most efficient use of sperm. Use of single AI was performance of swine herds. Anim. Reprod. Sci. 81, 115–123.
Einarsson, S., Swensson, T., Viring, S., 1973. A field trial on the fertility of
less efficient for pig production while use of 4 × 109 motile deep-frozen boar spermatozoa. Nord. Vet. Med. 25, 372–376.
sperm in a double AI was less efficient for use of sperm. Einarsson, S., Viring, S., 1973. Distribution of frozen-thawed spermato-
Variation in interval from AI to ovulation and boar fertility zoa in the reproductive tract of gilts at different time intervals after
insemination. J. Reprod. Fertil. 32, 117–120.
accounted for significant fertility effects. Methods to con- Eriksson, B.M., Petersson, H., Rodriguez-Martinez, H., 2002. Field fertil-
trol ovulation time and maximize boar fertility would aid ity with exported boar semen frozen in the new flatpack container.
this technology. The induction and estrous synchronization Theriogenology 58, 1065–1079.
Eriksson, B.M., Rodriguez-Martinez, H., 2000a. Deep-freezing of boar
procedure for prepubertal gilts proved to be an effective
semen in plastic film ‘cochettes’. J. Vet. Med. 47, 89–97.
method for testing frozen sperm fertility. Eriksson, B.M., Rodriguez-Martinez, H., 2000b. Effect of freezing and thaw-
ing rates on the post-thaw viability of boar spermatozoa frozen in
FlatPacks and Maxi-straws. Anim. Reprod. Sci. 63, 205–220.
Acknowledgements
Estienne, M.J., Harper, A.F., Horsley, B.R., Estienne, C.E., Knight, J.W., 2001.
Effects of P.G. 600 on the onset of estrus and ovulation rate in gilts
We would like to thank the USDA for funding sup- treated with Regu-mate. J. Anim. Sci. 79, 2757–2761.
port, and PIC North America for providing boar semen and Fiser, P.S., Fairfull, R.W., Hansen, C., Painch, P.L., Shrestha, J.N.B., Underhill,
L., 1993. The effect of warming velocity on motility and acrosomal
Intervet/Schering-Plough for providing PG600 and MATRIX integrity of boar sperm as influenced by the rate of freezing and glyc-
for use in this study. We are also grateful to Dr. S. Breen, J. erol level. Mol. Reprod. Devel. 34, 190–195.
266 K.W. Spencer et al. / Animal Reproduction Science 121 (2010) 259–266

Flowers, W.L., 1997. Management of boars for efficient sperm production. Okazaki, T., Abe, S., Shimada, M., 2009a. Improved conception rates in sows
J. Reprod. Fertil. Suppl. 52, 67–78. inseminated with cryopreserved boar spermatozoa prepared with a
Flowers, W.L., 2002. Increasing fertilization rate of boars: Influence of more optimal combination of osmolality and glycerol in the freezing
number and quality of spermatozoa inseminated. J. Anim. Sci. 80, extender. Anim. Sci. J. 80, 121–129.
E47–53. Okazaki, T., Abe, S., Yoshida, S., Shimada, M., 2009b. Seminal plasma dam-
Flowers, W.L., Esbenshade, K.L., 1993. Optimizing management of natural ages sperm during cryopreservation, but its presence during thawing
and artificial matings in swine. J. Reprod. Fertil. Suppl. 48, 217–228. improves semen quality and conception rates in boars with poor post-
Foxcroft, G.R., Vinsky, M.D., Paradis, F., Tse, W.Y., Town, S.C., Putman, C.T., thaw semen quality. Theriogenology 71, 491–498.
Dyck, M.K., Dixon, W.T., 2007. Macroenvironment effects on oocytes Purdy, P.H., 2008. Ubiquitination and its influence in boar sperm physiol-
and embryos in swine. Theriogenology 68 (Suppl. 1), S30–39. ogy and cryopreservation. Theriogenology 70, 818–826.
Grobfeld, R., Sieg, B., Struckmann, C., Frenzel, A., Maxwell, W.M.C., Rath, D., Pursel, V.G., Johnson, L.A., 1975. Freezing of boar spermatozoa: fertilizing
2008. New aspects of boar semen freezing strategies. Theriogenology capacity with concentrated semen and a new thawing procedure. J.
70, 1225–1233. Anim. Sci. 40, 99–102.
Heinonen, M., Leppävuori, A., Pyörälä, S., 1998. Evaluation of reproduc- Pursel, V.G., Johnson, L.A., 1976. Frozen boar spermatozoa: methods of
tive failure of female pigs based on slaughterhouse material and herd thawing pellets. J. Anim. Sci. 42, 927–931.
record survey. Anim. Reprod. Sci. 52, 235–244. Reed, C.B., 1985. Current use of frozen boar semen—future need of frozen
Hernandez, M., Roca, J., Calvete, J.J., Sanz, L., Muino-Blanco, T., Cebrian- boar semen. In: Johnson, L., Larsson, K. (Eds.), Deep Freezing Boar
Perez, J.A., Vazquez, J.M., Martinez, E.A., 2007a. Cryosurvival and in Semen. Proc. First Int. Conf. Deep Freezing of Boar Semen. Swedish
vitro fertilizing capacity postthaw is improved when boar spermato- University of Agricultural Sciences, Uppsala, Sweden, pp. 225–237.
zoa are frozen in the presence of seminal plasma from good freezer Roca, J., Carvajal, G., Lucas, X., Vazquez, J.M., Martinez, E.A., 2003. Fertility
boars. J. Androl. 28, 689–697. of weaned sows after deep intrauterine insemination with a reduced
Hernandez, M., Roca, J., Gil, M.A., Vázquez, J.M., Martínez, E.A., 2007b. number of frozen-thawed spermatozoa. Theriogenology 60, 77–87.
Adjustments on the cryopreservation conditions reduce the incidence Roca, J., Vázquez, J.M., Gil, M.A., Cuello, C., Parrilla, I., Martínez, E.A.,
of boar ejaculates with poor sperm freezability. Theriogenology 67, 2006. Challenges in pig artificial insemination. Reprod. Dom. Anim.
1436–1445. 41, 43–53.
Hofmo, P.O., Grevel, I.S., 2000. Development and commercial use of frozen Rodriguez-Martinez, H., Eriksson, B., Lundeheim, N., 1996. Freezing boar
boar semen in Norway. In: Johnson, L.A., Gutherie, H.D. (Eds.), Boar semen in flat plastic bags membrane integrity and fertility. Reprod.
Semen Preservation IV. Allen Press Inc., Lawrence, KS, pp. 71–77. Dom. Anim. 31, 161–168.
Horsley, B.R., Estienne, M.J., Harper, A.F., Purcell, S.H., Baitis, H.K., Beal, Rozeboom, K.J., Reicks, D.L., Wilson, M.E., 2004. The reproductive per-
W.E., Knight, J.W., 2005. Effect of P.G. 600 on the timing of ovulation formance and factors affecting on-farm application of low-dose
in gilts treated with altrenogest. J. Anim. Sci. 83, 1690–1695. intrauterine deposit of semen in sows. J. Anim. Sci. 82, 2164–2168.
Hunter, M.G., Wiesak, T., 1990. Evidence for and implications of follicular Soede, N.M., Noordhuizen, J.P.T.M., Kemp, B., 1992. The duration of ovula-
heterogeneity in pigs. J. Reprod. Fertil. Suppl. 40, 163–177. tion in pigs, studied by transrectal ultrasonography, is not related to
Johnson, L.A., 1985. Fertility results when using frozen boar spermatozoa: early embryonic diversity. Theriogenology 38, 653–666.
1970 to 1985. In: Johnson, L.A., Larsson, K. (Eds.), Deep Freezing Boar Thurston, L.M., Holt, W.V., Watson, P.F., 2003. Post-thaw functional sta-
Semen. Proc. First Int. Conf. Deep Freezing of Boar Semen. Swedish tus of boar spermatozoa cryopreserved using three controlled rate
University of Agricultural Sciences, Uppsala, Sweden, pp. 192–222. freezers: a comparison. Theriogenology 60, 101–113.
Johnson, L.A., Aalbers, J.G., Arts, J.A.M., 1982. Use of boar spermatozoa Town, S.C., Patterson, J.L., Pereira, C.Z., Gourley, G., Foxcroft, G.R., 2005.
for artificial insemination II. Fertilizing Capacity of fresh and frozen Embryonic and fetal development in a commercial dam-line genotype.
spermatozoa in gilts inseminated either at a fixed time or according Anim. Reprod. Sci. 85, 301–316.
to Walsmeta readings. J. Anim. Sci. 54, 126–131. Waberski, D., Kunz-Schmidt, A., Borchardt Neto, G., Richter, L., Weitze, K.F.,
Johnson, L.A., Weitze, K.F., Fiser, P., Maxwell, W.M.C., 2000. Storage of boar 2000. Real-time ultrasound diagnosis of ovulation and ovarian cysts
semen. Anim. Reprod. Sci. 62, 143–172. in sows and its impact on artificial insemination efficiency. J. Anim.
Kemp, B., Soede, N.M., Hazeleger, W., Wetzels, C.C.H., 1996. The effects Sci. 77, 1-aj-8.
of a second insemination after ovulation—as determined with Waberski, D., Weitze, K.F., Gleumes, T., Schwarz, M., Willmen, T., Petzoldt,
ultrasonography—on fertilization rate and accessory sperm count in R., 1994. Effect of time of insemination relative to ovulation on fertility
sows. Reprod. Dom. Anim. 31, 297–298. with liquid and frozen boar semen. Theriogenology 42, 831–840.
Knox, R.V., 2005. Recruitment and selection of ovarian follicles for deter- Waterhouse, K.E., Hofmo, P.O., Tverdal, A., Miller Jr., R.R., 2006. Within
mination of ovulation rate in the pig. Dom. Anim. Endocrinol. 29, and between breed differences in freezing tolerance and plasma
385–397. membrane fatty acid composition of boar sperm. Reproduction 131,
Knox, R.V., Miller, G.M., Willenburg, K.L., Rodriguez-Zas, S.L., 2002. Effect of 887–894.
frequency of boar exposure and adjusted mating times on measures of Watson, P.F., Behan, J.R., 2002. Intrauterine insemination of sows with
reproductive performance in weaned sows. J. Anim. Sci. 80, 892–899. reduced sperm numbers: results of a commercially based field trial.
Larsson, K., 1978. Deep-freezing of boar semen. Cryobiology 15, 352–354. Theriogenology 57, 1683–1693.
Larsson, K., Einarsson, S., 1976. Influence of boars on the relationship Wongtawan, T., Saravia, F., Wallgren, M., Caballero, I., Rodríguez-Martínez,
between fertility and post thawing sperm quality of deep frozen boar H., 2006. Fertility after deep intra-uterine artificial insemination
spermatozoa. Acta Vet. Scand. 17, 74–82. of concentrated low-volume boar semen doses. Theriogenology 65,
Larsson, K., Einarsson, S., Swenson, T., 1977. The development of a practi- 773–787.
cable method for deepfreezing of boar spermatozoa. Nord. Vet. Med.
29, 113–118.
Animal Reproduction Science 121 (2010) 267–272

Contents lists available at ScienceDirect

Animal Reproduction Science


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

The role of sexual steroid hormones in the direct stimulation by


Kisspeptin-10 of the secretion of luteinizing hormone,
follicle-stimulating hormone and prolactin from bovine anterior
pituitary cells
A. Ahmed Ezzat a,b , H. Saito a , T. Sawada a , T. Yaegashi a , Y. Goto a , Y. Nakajima a ,
J. Jin a , T. Yamashita a , K.Sawai a , T. Hashizume a,∗
a
Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Japan
b
Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt

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

Article history: The aims of the present study were to clarify the effect of Kisspeptin-10 (Kp10) on the
Received 24 February 2010 secretion of luteinizing hormone (LH), follicle-stimulating hormone (FSH) and prolactin
Received in revised form 30 April 2010
(PRL) from bovine anterior pituitary (AP) cells and evaluate the ability of sex steroids to
Accepted 2 June 2010
enhance the sensitivity of gonadotropic and lactotropic cells to Kp10. AP cells prepared
Available online 11 June 2010
from 7-week-old male calves were incubated for 12 h with estradiol (E2 ; 10−8 M), proges-
terone (P4 ; 10−8 M), testosterone (T; 10−8 M), or vehicle only (control), and then for 2 h with
Keywords:
Kp10 (10−6 M). The amounts of LH, FSH and PRL released into the culture medium after the
Kisspeptin-10
Sex steroids 2-h incubation period were examined. Kp10 significantly stimulated the secretion of LH
LH from the AP cells treated with E2 and T (P < 0.05), but not from the P4 -treated cells. In con-
FSH trast, Kp10 had no effect on the secretion of FSH regardless of the steroid treatment. Kp10
PRL significantly stimulated the secretion of PRL (P < 0.05), the sexual steroid hormones having
Bovine no effect. The LH- or FSH-releasing response to gonadotropin-releasing hormone (GnRH;
10−8 M) and PRL-releasing response to thyrotropin-releasing hormone (TRH; 10−8 M) were
significantly greater than those to Kp10 (P < 0.05).
The present results suggest that E2 and T, but not P4 , enhance the sensitivity of
gonadotropic cells to the secretion of LH in response to Kp10. However, Kp10 had no stim-
ulatory effect on the secretion of FSH regardless of the effect of sex steroids. Kp10 directly
stimulates the secretion of PRL from the pituitary cells, and sex steroids do not enhance the
sensitivity of lactotropic cells to Kp10. Furthermore, the LH- and FSH-releasing effect and
the PRL-releasing effect of Kp10 are less potent than that of GnRH and TRH, respectively.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction coupled receptor (GPR54) (Kotani et al., 2001; Ohtaki et al.,


2001), play a significant role in the anterior pituitary (AP)
Kisspeptin is a neuropeptide hormone encoded by KiSS- gland as evidenced by (a) the secretion of kisspeptin in the
1 (Gottsch et al., 2009) in the hypothalamus. Studies have hypophyseal portal circulation (Smith et al., 2008), (b) the
found that kisspeptin and its functional ligand, G-protein- expression of GPR54 in human (Kotani et al., 2001; Ohtaki
et al., 2001), rat (Gutierrez-Pascual et al., 2007) and ovine
(Smith et al., 2008) pituitary cells, and (c) the presence
∗ Corresponding author. Tel.: +81 19 621 6161; fax: +81 19 621 6161. of kisspeptin-secreting cells in the AP gland of monkeys
E-mail address: hashi@iwate-u.ac.jp (T. Hashizume). (Ramaswamy et al., 2009). Furthermore, recent studies in

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.002
268 A.A. Ezzat et al. / Animal Reproduction Science 121 (2010) 267–272

vitro reported a direct effect of kisspeptin on gonadotropic, with phosphate-buffered saline (PBS). The volumes of PBS
i.e. luteinizing hormone (LH) (Suzuki et al., 2008), and non- added to the wells in control and treated groups were 27 ␮l
gonadotropic, i.e. prolactin (PRL) (Kadokawa et al., 2008), for E2 , 31 ␮l for P4 and 29 ␮l for T, respectively. Other wells
pituitary hormone release. Although hormone secretion by were left without steroid treatments and designed to be
the AP is controlled by hypothalamic peptides (Vankelecom treated with Kp10 without the effect of steroids. There
and Denef, 1997), sexual steroid hormones are also impli- were exactly four wells dedicated for each treatment and
cated in AP cell function. Estradiol (E2 ) is a key regulator each treatment has its separate control which is equal in
for the synthesis and secretion of pituitary reproduc- number. Twelve hours later, the cells were washed twice
tive hormones: LH, follicle-stimulating hormone (FSH) and with PBS, and the medium was replaced with DMEM alone
PRL (Sánchez-Criado et al., 2004). E2 has a direct effect (control), or DMEM containing 10−6 M of Kp10. The exper-
on pituitary gonadotropes and lactotropes (Scully et al., imental protocol for steroid treatments (Hashizume et al.,
1997). Progesterone (P4 ) (Fox et al., 1990) and testos- 2002) and the concentration of Kp10 (Kadokawa et al.,
terone (T) (Okada et al., 2003) receptors are expressed by 2008) was performed according to our previous study. After
gonadotropes. P4 exerts a direct modulatory effect on LH a 2-h incubation period, the medium was removed from
secretion (Krey and Kamel, 1990) and T regulates the syn- cells and stored at −30 ◦ C until the assays for hormones.
thesis of the LH␤ subunit in the AP (Burger et al., 2004;
Thackray et al., 2006). However, the role of sexual steroid 2.4. GnRH and TRH treatments
hormones and kisspeptin in the secretion of LH, FSH and
PRL has not been evaluated yet. In the present study, the non-steroid-treated AP cells
The present study aimed to clarify the direct effect of were also treated with GnRH or TRH. The plated cells were
Kisspeptin-10 (Kp10: a shorter variant of kisspeptin retain- cultured for 36 h, and then the cells were washed twice
ing full biological activity) on the secretion of LH, FSH, and with PBS. The medium was replaced with DMEM alone,
PRL from cultured bovine AP cells and evaluate the abil- DMEM containing 10−8 M of GnRH or TRH. After a 2-h incu-
ity of E2 , P4 , and T to enhance the sensitivity of pituitary bation period, the medium was removed from cells and
cells to Kp10. In the present study, the direct effects of stored at −30 ◦ C until the assays for hormones.
Kp10 on the secretion of the AP hormones were compared
to those of gonadotropin-releasing hormone (GnRH) and 2.5. Hormone assay
thyrotropin-releasing hormone (TRH).
Concentrations of bovine LH in media were measured
2. Materials and methods by a time-resolved fluoroimmunoassay procedure with
slight modifications to the system described previously
2.1. Peptides (Kaneko et al., 2002). The standard LH preparation and
the hormone for labeling europium were USDA-bLH-B-
Human Kp10 amide (amino acid sequence: 6. Concentrations of bovine FSH and PRL in media were
YNWNSFGLRF-NH2 ) was synthesized in our labora- measured by a double-antibody radioimmunoassay with
tory (Ezzat et al., 2009; Hashizume et al., 2010). The slight modifications (Hashizume et al., 1999). For FSH, the
peptide has been confirmed to stimulate the release of standard preparation and the hormone for iodination were
gonadotropins in goats (Hashizume et al., 2010) and cattle AFP5346D and AFP5318C, respectively. For PRL, the stan-
(Ezzat et al., 2009). GnRH and TRH were purchased from dard preparation and the hormone for iodination were
Peptide Institute Inc., Osaka, Japan. NIDDK oPRL-I-3. Assay sensitivities for LH, FSH and PRL
were 1.6, 0.2, and 1.0 ng/ml, respectively. All samples were
2.2. Adenohypophyseal cells and culture assayed in a single run. The intraassay coefficient of vari-
ation was 11.8% for LH, 7.5% for FSH and 5.8% for PRL. The
The pituitary glands were obtained from four male Hol- displacement curve for increasing volumes of the culture
stein calves (age: 7-week old). AP cells were enzymatically medium pool paralleled the standard curve of the LH, FSH
dispersed by a method described previously (Hashizume and PRL assays.
and Kasuya, 2009). Cell viability as determined by trypan
blue exclusion was greater than 90%. The dispersed cells 2.6. Statistical analysis
were suspended in Dulbecco’s modified Eagle’s medium
(DMEM; Gibco, Grand Island, NY, USA) containing 10% There were four wells per treatment and each exper-
horse serum (Gibco), 100 I.U./ml penicillin and 50 mg/ml iment was repeated three to four times with different
streptomycin. The cells (2.5 × 105 /ml) were then plated in pituitary glands. Hormone concentrations in the controls
Costar 24-well dishes and cultured at 37 ◦ C in a humidified were averaged and the mean value was set at 100% to trans-
atmosphere of 5% CO2 . form the data. Each hormone concentration was expressed
as a percentage of the control value. All data from the exper-
2.3. Sexual steroid hormones and Kp10 treatments iments were presented as the mean ± S.E.M. The statistical
significance of differences in the LH, FSH or PRL concen-
The plated cells were cultured for 24 h, and then E2 tration in response to 0 or 10−6 M of Kp10 between the
(10−8 M), P4 (10−8 M), T (10−8 M), or the respective vehi- steroid-treated and un-treated groups was evaluated by
cle only (controls) was added to the wells. Sexual steroid two-way ANOVA, and the Bonferroni test was used as a
hormones were dissolved in ethanol first, and then diluted post hoc test. Comparisons between the un-treated control
A.A. Ezzat et al. / Animal Reproduction Science 121 (2010) 267–272 269

failed to increase the concentration of LH in the culture


medium compared to the control (17.2 ± 2.6 ng/ml). In the
E2 -treated group, it significantly increased the concentra-
tion by 17.0% compared to the control (20.2 ± 2.6 ng/ml)
(P < 0.05). In the P4 -treated group, it did not increase the
concentration of LH in the culture medium compared to
the control (14.9 ± 2.4 ng/ml). In the T-treated group, it sig-
nificantly increased the concentration by 11.4% compared
to the control (12.2 ± 1.6 ng/ml) (P < 0.05). The percent
increase in the concentration of LH in T-treated group was
not significantly different from that in the E2 -treated group.
Fig. 1b shows the effect of Kp10 on the secretion of
FSH from cultured bovine AP cells, treated with steroids
or not. In the group not treated with a steroid, 10−6 M of
Kp10 failed to increase the concentration of FSH in the cul-
ture medium compared to the control (1.2 ± 0.1 ng/ml). In
the E2 -treated group, it significantly decreased the concen-
tration by 2.60% compared to the control (1.2 ± 0.1 ng/ml)
(P < 0.05). In the P4 -and T-treated groups, it did not increase
the FSH-concentration in the medium compared to each
respective control (1.2 ± 0.1 and 1.1 ± 0.1 ng/ml, respec-
tively).
Fig. 1c shows the effect of Kp10 on the secretion of
PRL from cultured bovine AP cells, treated with steroids
or not. In the group not treated with a steroid, 10−6 M
of Kp10 significantly increased the concentration of PRL
in the culture medium by 23.0% compared to the con-
trol (40.3 ± 4.5 ng/ml) (P < 0.05). In the E2 -treated group,
it significantly increased the concentration by 23.2% com-
pared to the control (39.3 ± 4.1 ng/ml) (P < 0.05). In the
P4 -treated group, Kp10 significantly increased the con-
centration of PRL by 20.2% compared to the control
(42.1 ± 5.5 ng/ml) (P < 0.05). In the T-treated group, it sig-
nificantly increased the concentration by 19.7% compared
to the control (42.7 ± 5.1 ng/ml) (P < 0.05). There were no
significant differences in the percent increase of PRL among
the non-steroid-treated group and each steroid-treated
Fig. 1. Effect of estradiol (E2 ; 10−8 M), progesterone (P4 ; 10−8 M) and group.
testosterone (T; 10−8 M) on Kisspeptin-10 (Kp10; 10−6 M)-stimulated
release of luteinizing hormone (LH) (a), follicle-stimulating hormone
3.2. Secretion of LH, FSH and PRL in response to GnRH or
(FSH) (b) and prolactin (PRL) (c) from cultured bovine anterior pituitary
(AP) cells. There were four treatments; Kp10 only without steroids, Kp10 TRH
proceeded by E2 , Kp10 proceeded by P4 and Kp10 proceeded by T. There
were four wells per treatment and the experiment was repeated three Fig. 2 shows the secretion of LH, FSH and PRL in response
to four times with cells obtained from different male calves. Each value to GnRH or TRH from cultured bovine AP cells. GnRH
represents the mean for 12 (LH) to 16 (FSH, PRL) wells ± S.E.M. *P < 0.05
vs. control (CTL).
significantly increased the LH (Fig. 2a) concentration by
104.1% compared to the control (22.7 ± 3.2 ng/ml) and this
response was significantly greater than that of E2 plus
group and group treated with 10−8 M of GnRH or TRH were Kp10- or T plus Kp10-treated group (P < 0.05). GnRH sig-
evaluated with Student’s t-test. The statistical significance nificantly increased the FSH (Fig. 2b) concentration by
of differences in all data was analyzed using GraphPad 178.0% compared to the control (2.1 ± 0.3 ng/ml) (P < 0.05).
Prism (GraphPad Software, San Diego, CA, USA). Results TRH significantly increased the PRL (Fig. 2c) concentration
were considered significant at the P < 0.05 level. by 117.0% compared to the control (41.1 ± 4.7 ng/ml) and
this response was significantly greater than that to Kp10-
3. Results treated group (P < 0.05).

3.1. Effects of sexual steroid hormones on the secretion of 4. Discussion


LH, FSH and PRL in response to Kp10
Recent studies have examined the direct effect of Kp10
Fig. 1a shows the effect of Kp10 on the secretion of LH on the pituitary release of LH (Suzuki et al., 2008) and PRL
from cultured bovine AP cells, treated with steroids or not. (Kadokawa et al., 2008). The present study is the first to
In the group not treated with a steroid, 10−6 M of Kp10 examine the direct effect of Kp10 on the release of FSH
270 A.A. Ezzat et al. / Animal Reproduction Science 121 (2010) 267–272

Work by a number of investigators using AP cell cul-


tures has shown that P4 exerts a direct modulatory effect
on LH secretion (Drouin and Labrie, 1981; Ortmann et al.,
1989; Krey and Kamel, 1990). P4 receptors were detected
on pituitary gonadotropes (Fox et al., 1990). P4 adminis-
tered for periods beyond 12 h inhibited the gonadotropin
release from cultured pituitary cells (Lesoon and Mahesh,
1992). Furthermore, luteal P4 was found to suppress the
secretion of LH in monkeys (O’Byrne et al., 1991). There-
fore, it was considered that Kp10 had no stimulatory effect
on LH secretion from the AP cells treated with P4 .
Androgen acts directly in the pituitary cell via its own
receptor rather than through prior conversion into estro-
gen (Belanger et al., 1980). Androgen receptors have been
detected in the pituitary gonadotropes (Okada et al., 2003).
T and dihydrotestosterone were reported to regulate LH-␤
mRNA levels in the AP gland (Burger et al., 2004; Thackray
et al., 2006). Androgens have a dual effect, inhibitory and
stimulatory, on the secretion of LH from the pituitary
gonadotropes in vitro (Turgeon and Waring, 1999). The
stimulatory action of T on the LH-secreting gonadotropes is
due to stimulation of LH-␤ mRNA expression (Yasin et al.,
1996) or modulation of Ca2+ signals (Ortmann et al., 1998).
Therefore, it was suggested that T enhanced the sensitiv-
ity of LH-secreting gonadotropes to Kp10 through these
pathways.
Regulation of FSH secretion is less clearly understood
(Lesoon and Mahesh, 1992). The secretion of FSH, like that
of LH, did not respond to Kp10 in the AP cells not treated
with steroids, and the sexual steroid hormones did not
enhance the FSH-secretory response to Kp10. In contrast to
LH, E2 significantly suppressed FSH secretion, but neither
P4 nor T had any effect on the FSH-secreting cells. A pre-
Fig. 2. Effect of gonadotropin-releasing hormone (GnRH; 10−8 M) or vious study in vivo found an inverse relationship between
thyrotropin-releasing hormone (TRH; 10−8 M) on the release of LH (a),
FSH and E2 (Robertson et al., 2009). E2 ’s inhibitory effect on
FSH (b) and PRL (c) from cultured bovine AP cells. There were four wells
per treatment and the experiment was repeated three times with cells the secretion of FSH is mediated through its ability to sup-
obtained from different male calves. Each value represents the mean for press the expression of the gene encoding activin (Nett et
12 wells ± S.E.M. *P < 0.05 vs. CTL. al., 2002). Our results support that E2 primarily acts on the
pituitary gonadotropes to inhibit FSH production (Ghosh
et al., 1996) and subsequently showed no response to the
in addition to LH and PRL from cultured bovine AP cells Kp10-releasing effect. It seems that the effect of P4 on the
and compare the characteristics of the secretion to those pituitary gonadotropes is the same for both LH and FSH,
of responses to GnRH and TRH. Furthermore, the present and T regulates the secretion of LH and FSH independently
study is the first to examine the role of sexual steroid hor- from its direct action on the pituitary cells.
mones in enhancing the sensitivity of gonadotropes and GnRH stimulated both the synthesis and release of the
lactotropes to Kp10. pituitary gonadotropins LH and FSH (Naor, 2009). This sup-
Although gonadotropes were suggested to express ports that LH and FSH secretion was greater in response to
GPR54 receptors (Smith et al., 2008), Kp10 alone failed to GnRH than to Kp10 in the present study. Our results show
stimulate the secretion of LH directly from the AP cells. Kp10 to be a less potent LH-releaser.
However, treatment with E2 or T but not P4 enhanced the Our findings revealed a direct effect of Kp10 on the
sensitivity of LH-releasing gonadotropes to Kp10. The E2 secretion of PRL from cultured bovine AP cells regardless of
receptor is expressed by 60–70% of pituitary cells (Keefer steroid treatments. The results were similar to those for GH
et al., 1976) and mostly by gonadotropes (Smith and (Ezzat et al., 2010); that is, GH was secreted by Kp10 regard-
Keefer, 1985). It increases LH-subunit gene expression via less of steroid treatments. E2 and T receptors have been
modulation of regulatory transcription factors (Kowase demonstrated in the pituitary tissue; however, there was
et al., 2007). Furthermore, a recent study found that rat no evidence for a direct effect of sex steroids on lactotropes
gonadotropes express both the GPR54 and KiSS-1 genes, (Shi et al., 1986). Sprangers et al. (1989) found that P4 recep-
and the latter expression is up-regulated by E2 (Richard et tors are not expressed in pituitary lactotropes and P4 had
al., 2008). Therefore, our results suggest that E2 regulates no direct effect on PRL secretion in monkey pituitary cell
an autocrine action of kisspeptin through the secretion of cultures. These findings support our results since we found
LH from the pituitary gonadotropes. no effect of sexual steroid hormones on the PRL-secreting
A.A. Ezzat et al. / Animal Reproduction Science 121 (2010) 267–272 271

lactotropes in response to Kp10. However, our results sug- secretion by kisspeptin-10 in female goats. Anim. Reprod. Sci. 118,
gest that kisspeptin plays an autocrine or paracrine role in 37–41.
Hashizume, T., Takahashi, Y., Numata, M., Sasaki, K., Ueno, K., Ohtsuki,
the pituitary tissue to regulate the function of lactotropes in K., Kawai, M., Ishi, A., 1999. Plasma profiles of growth hormone, pro-
cattle. In the present study, we examined the characteris- lactin and insulin-like growth factor-I during gestation, lactation and
tics of PRL secretion in response to Kp10 and TRH. However, neonatal period in goats. J. Reprod. Dev. 45, 273–281.
Kadokawa, H., Suzuki, S., Hashizume, T., 2008. Kisspeptin-10 stimulates
less PRL was secreted in response to Kp10 than to TRH. the secretion of growth hormone and prolactin directly from cultured
In summary, the present results suggest that E2 and T, bovine anterior pituitary cells. Anim. Reprod. Sci. 105, 404–408.
but not P4 , enhance the sensitivity of gonadotropic cells Kaneko, H., Noguchi, J., Kikuchi, K., Todoroki, J., Hasegawa, Y., 2002. Alter-
ations in peripheral concentrations of inhibin A in cattle studied
to Kp10-stimulated release of LH. However, Kp10 had no using a time-resolved immunofluorometric assay: relationship with
stimulatory effect on FSH secretion regardless of the effect estradiol and follicle-stimulating hormone in various reproductive
of sex steroids. Kp10 directly stimulates the release of PRL conditions. Biol. Reprod. 67, 38–45.
Keefer, D.A., Stumpf, W.E., Petrusz, P., 1976. Quantitative autoradiographic
from lactotropic cells and sex steroids do not enhance the
assessment of 3H-estradiol uptake in immunocytochemically charac-
sensitivity of lactotropic cells to Kp10. Furthermore, LH- terized pituitary cells. Cell Tissue Res. 166, 25–35.
and PRL-releasing effect of Kp10 was less potent than that Kotani, M., Detheux, M., Vandenbogaerde, A., Communi, D., Vander-
of GnRH and TRH, respectively. winden, J.M., Le Poul, E., Brezillion, S., Tyldesley, R., Suarez-Huerta,
N., Vandeput, F., Detheux, M., Blanpain, C., Schiffmann, S.N., Vas-
sart, G., Parmentier, M., 2001. The metastasis suppressor gene KiSS-1
Acknowledgements encodes kisspeptins, the natural ligands of the orphan G protein-
coupled receptor GPR54. J. Biol. Chem. 276, 34631–34636.
Kowase, T., Walsh, H.E., Darling, D.S., Shupnik, M.A., 2007. Estrogen
The authors thank Dr. E Kasuya (National Institute enhances gonadotropin-releasing hormone-stimulated transcription
of Agrobiological Science, Tsukuba, Japan) for providing of the luteinizing hormone subunit promoters via altered expression
bovine pituitary and Dr. A F Parlow (National Hormone and of stimulatory and suppressive transcription factors. Endocrinology
148, 6083–6091.
Peptide Program, Harbor-UCLA Medical Center, Torrance, Krey, L.C., Kamel, F., 1990. Progesterone modulation of gonadotropin
CA, USA) for providing the bovine FSH and ovine RIA kits. secretion by dispersed rat pituitary cells in culture. I. Basal and
This research was partly supported by a Grant-in-aid for gonadotropin-releasing hormone stimulated luteinizing hormone
release. Mol. Cell. Endocrinol. 68, 85–94.
Scientific Research (no. 21380169) from the Japan Society Lesoon, L.A., Mahesh, V.B., 1992. Stimulatory and inhibitory effects of
for the Promotion of Science to T. Hashizume. progesterone on FSH secretion by the anterior pituitary. J. Steroid
Biochem. Mol. Biol. 42, 479–491.
Naor, Z., 2009. Signaling by G-protein-coupled receptor (GPCR): studies
References
on the GnRH receptor. Front. Neuroendocrinol. 30, 10–29.
Nett, T.M., Turzillo, A.M., Baratta, M., Rispoli, L.A., 2002. Pituitary effects
Belanger, A., Lundell, L., Eneroth, P., Gustafsson, J.A., 1980. Andro- of steroid hormones on secretion of follicle-stimulating hormone and
gen metabolism in rat anterior pituitary cells in culture. Mol. Cell. luteinizing hormone. Domest. Anim. Endocrinol. 23, 33–42.
Endocrinol. 19, 175–181. O’Byrne, K.T., Thalabard, J.C., Grosser, P.M., Wilson, R.C., Williams, C.L.,
Burger, L.L., Haisenleder, D.J., Dalkin, A.C., Marshall, J.C., 2004. Regula- Chen, M.D., Ladendorf, D., Hotchkiss, J., Knobil, E., 1991. Radiotele-
tion of gonadotropin subunit gene transcription. J. Mol. Endocrinol. metric monitoring of hypothalamic gonadotropin-releasing hormone
33, 559–584. pulse generator activity throughout the menstrual cycle of the rhesus
Drouin, J., Labrie, F., 1981. Interaction between 17␤-estradiol and proges- monkey. Endocrinology 129, 1207–1214.
terone in the control of LH and FSH release in rat anterior pituitary Ohtaki, T., Shintani, Y., Honda, S., Matsumoto, H., Hori, A., Kanihashi, K.,
cell culture. Endocrinology 108, 52–57. Terao, Y., Kumano, S., Takatsu, Y., Masuda, Y., Ishibashi, Y., Watanabe,
Ezzat, A.A., Saito, H., Sawada, T., Yaegashi, T., Jin J, Sawai, K., Yamashita, T., T., Asada, M., Yamada, T., Suenaga, M., Kitada, C., Usuki, S., Kurokawa,
Hashizume, T., 2010. Sex steroid hormones do not enhance the direct T., Onda, H., Nishimura, O., Fujino, M., 2001. Metastatsis suppressor
stimulatory effect of kisspetin-10 on the secretion of growth hormone gene kiss-1 encodes peptide ligand of a G-protein coupled receptor.
from bovine anterior pituitary cells. Anim. Sci. J. (in press). Nature 411, 613–617.
Ezzat, A.A., Saito, H., Sawada, T., Yaegashi, T., Yamashita, T., Hirata, T.-I., Okada, Y., Fujii, Y., Moore Jr., J.P., Winters, S.J., 2003. Androgen receptors
Sawai, K., Hashizume, T., 2009. Characteristics of the stimulatory effect in gonadotrophs in pituitary cultures from adult male monkeys and
of Kisspeptin-10 on the secretion of luteinizing hormone, follicle- rats. Endocrinology 144, 267–273.
stimulating hormone and growth hormone in prepubertal male and Ortmann, O., Tomic, M., Weiss, J.M., Diedrich, K., Stojilkovic, S.S., 1998.
female cattle. J. Reprod. Dev. 55, 650–654. Dual action of androgen on calcium signaling and luteinizing hormone
Fox, S., Harlan, R.E., Shivers, B., Pfaff, D.W., 1990. Chemical characterization secretion in pituitary gonadotrophs. Cell Calcium 24, 223–231.
of neuroendocrine targets for progesterone in the female rat brain and Ortmann, O., Weise, H., Knuppen, R., Emons, G., 1989. Acute facilitory
pituitary. Neuroendocrinology 51, 276–283. action of progesterone on gonadotropin secretion of perifused rat
Ghosh, B.R., Wu, J.C., Strahl, B.D., Childs, G.V., Miller, W.L., 1996. Inhibin pituitary cells. Acta Endocr. (Copenh.) 121, 426–434.
and estradiol alter gonadotropes differentially in ovine pituitary cul- Ramaswamy, S., Gibbs, R.B., Plant, T.M., 2009. Studies of the locali-
tures: changing gonadotrope numbers and calcium responses to sation of Kisspeptin within the pituitary of the Rhesus Monkey
gonadotropin-releasing hormone. Endocrinology 137, 5144–5154. (Macaca mulatta) and the effect of Kisspeptin on the release of non-
Gottsch, M.L., Clifton, D.K., Steiner, R.A., 2009. From KISS1 to kisspeptins: gonadotropic pituitary hormones. J. Neuroendocrinol. 21, 795–804.
an historical perspective and suggested nomenclature. Peptides 30, Richard, N., Galmiche, G., Corvaisier, S., Caraty, A., Kottler, M.L., 2008.
4–9. KiSS-1 and GPR54 genes are co-expressed in rat gonadotrophs and
Gutierrez-Pascual, E., Martinez-Fuentes, A.J., Pinilla, L., Tena-Sempere, differentially regulated in vivo by oestradiol and gonadotrophin-
M., Malagon, M.M., Castano, J.P., 2007. Direct pituitary effects of releasing hormone. J. Neuroendocrinol. 20, 381–393.
kisspeptin: activation of gonadotrophs and somatotrophs and stim- Robertson, D.M., Hale, G.E., Jolley, D., Fraser, I.S., Hughes, C.L., Burger, H.G.,
ulation of luteinizing hormone and growth hormone secretion. J. 2009. Interrelationships between ovarian and pituitary hormones in
Neuroendocrinol. 19, 521–530. ovulatory menstrual cycles across reproductive age. J. Clin. Endocrinol.
Hashizume, T., Kasuya, E., 2009. Methodology for the study of the Metab. 94, 138–144.
hypothalamic-pituitary hormone secretion in cattle. Anim. Sci. J. 80, Scully, K.M., Gleiberman, A.S., Lindzey, J., Lubahn, D.B., Korach, K.S., Rosen-
1–11. feld, M.G., 1997. Role of estrogen receptor-a in the anterior pituitary
Hashizume, T., Kumahara, A., Fujino, M., Okada, K., 2002. Insulin- gland. Mol. Endocrinol. 11, 674–681.
like growth factor I enhances gonadotropin-releasing hormone- Shi, Y., Patterson, A.P., Sherins, R.J., 1986. Increased plasma and pituitary
stimulated luteinizing hormone release from bovine anterior pituitary prolactin concentrations in adult male rats with selective elevation of
cells. Anim. Reprod. Sci. 70, 13–21. FSH levels may be explained by reduced testosterone and increased
Hashizume, T., Saito, H., Sawada, T., Yaegashi, T., Ezzat, A.A., Sawai, K., estradiol production. J. Androl. 7, 105–111.
Yamashita, T., 2010. Characteristics of stimulation of gonadotropin
272 A.A. Ezzat et al. / Animal Reproduction Science 121 (2010) 267–272

Smith, J.T., Rao, A., Pereira, A., Caraty, A., Millar, R.P., Clarke, I.J., 2008. and action and on gonadotropin and prolactin secretion in the rat.
Kisspeptin is present in ovine hypophyseal portal blood but does Neuroendocrinology 79, 247–258.
not increase during the preovulatory luteinizing hormone surge: evi- Thackray, V.G., McGillivray, S.M., Mellon, P.L., 2006. Androgens, progestins
dence that gonadotropes are not direct targets of kisspeptin in vivo. and glucocorticoids induce follicle-stimulating hormone ␤-subunit
Endocrinology 149, 1951–1959. gene expression at the level of the gonadotrope. Mol. Endocrinol. 20,
Smith, P.F., Keefer, D.A., 1985. Estrogen uptake capacity of individual pitu- 2062–2079.
itary cell types from male and female rats one fourteen and fifty days Turgeon, J.L., Waring, D.W., 1999. Androgen modulation of luteinizing
after castration. Horm. Metab. Res. 17, 583–587. hormone secretion by female rat gonadotropes. Endocrinology 140,
Sprangers, S.A., Brenner, R.M., Bethea, C.L., 1989. Estrogen and progestin 1767–1774.
receptor immunocytochemistry in lactotropes versus gonadotropes Vankelecom, H., Denef, C., 1997. Paracrine communication in the anterior
of monkey pituitary cell cultures. Endocrinology 124, 1462–1470. pituitary as studied in reaggregate cell cultures. Microsc. Res. Tech.
Suzuki, S., Kadokawa, H., Hashizume, T., 2008. Direct Kisspeptin-10 stim- 39, 150–156.
ulation on luteinizing hormone secretion from bovine and porcine Yasin, M., Dalkin, A.C., Haisenleder, D.J., Marshall, J.C., 1996. Testosterone is
anterior pituitary cells. Anim. Reprod. Sci. 103, 360–365. required for gonadotropin-releasing hormone stimulation of luteiniz-
Sánchez-Criado, J.E., Martín De Las Mulas, J., Bellido, C., Tena-Sempere, ing hormone-beta messenger ribonucleic acid expression in female
M., Aguilar, R., Blanco, A., 2004. Biological role of pituitary estrogen rats. Endocrinology 137, 1265–1271.
receptors ERalpha and ERbeta on progesterone receptor expression
Animal Reproduction Science 121 (2010) 273–278

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Relationships between ovarian cysts and morphological


and hormonal state of ovarian cortex in sows
˛
K. Szulańczyk-Mencel, A. Rzasa, W. Bielas ∗
Department and Clinic of Reproduction, Faculty of Veterinary Medicine, University of Environmental and Life Sciences, Wrocław, Poland

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

Article history: The aim of this study was to evaluate the relationship between ovarian cysts and concen-
Received 25 November 2009 trations of ovarian steroid hormones: 17␤-estradiol (E2 ), progesterone (P4) , testosterone
Received in revised form 27 May 2010
(T), and androstendione (A4 ) both in blood plasma and in cysts and morphological state
Accepted 1 June 2010
of the ovarian cortex in sows. Females were divided into three groups: PCO (sows with
Available online 8 June 2010
polycystical ovaries), OO (sows with oligocystic ovaries) and control (sows without ovarian
cysts). The ovaries for evaluations were collected after slaughtering of 18 multiparous sows.
Keywords:
Between the PCO and OO animals, statistically significant differences in numbers of the fol-
Ovarian cyst
Steroid hormones licular cysts (FC) (8.6 vs. 1.5), follicular theca-lutein cysts (FTLC) (8.0 vs. 2.0), follicular lutein
Ovarian cortex cysts (FLC) (4.5 vs. 2.0) and corpus luteum cysts (CLC) (1.7 vs. 0.4) (P ≤ 0.01) were noted. In
Sow the PCO sows the most common kinds of cysts were FC and FTLC (8.6 and 8.0) whilst in OO
sows the cysts occurred on their ovaries on a similar level (FC – 1.6, FTLC – 2.0, FLC – 2.0).
Existence of more than 10 ovarian cysts in the sows significantly decreases the frequency of
physiological ovarian follicles (primary, growing and maturing) and significantly increases
the pathological process of atresia on all stages of ovarian follicles development (P ≤ 0.01).
The study did not reveal any effect of growing or decreasing number of ovarian cyst on con-
centrations of E2 and P4 in blood plasma of sows. Polycystical ovaries significantly decreased
concentrations of A4 but increased the concentration of T in blood plasma (P ≤ 0.01). The
general presence of ovarian cysts considerably positively correlated with concentrations of
E2 , T and A4 from cysts’ fluid, of all kinds of ovarian cysts and atresia of primary follicles (a
correlation coefficient r from 0.72 up to 0.97, P ≤ 0.05). The phenomenon of ovarian cysts
significantly negatively correlated with all generations of ovarian follicles (P ≤ 0.05).
© 2010 Elsevier B.V. All rights reserved.

1. Introduction infertility, lesser conception rate, and behavioral changes


(Fitko et al., 1998b). Cysts appear to be an important risk
The occurrence of ovarian cysts may have detrimental factor in returning to estrus. These ovarian structures are
effect on sows health and reduce their reproductive per- present in approximately 10% of sows culled for fertility
formance. Some studies confirm the connections between problems (Ebbert and Bostedt, 1993; Castagna et al., 2004).
reproductive disorders and the presence of ovarian cysts However, little is known about the incidence of ovar-
found at slaughter (Castagna et al., 2004). COD (Cystic ian cysts in pig herds because they may also occur without
Ovarian Disease) in pigs appears frequently in multiparous leading to culling for reproductive problems. The presence
sows and offers no pathognomic symptoms except for an of cysts before insemination and conception might not
irregular or prolonged estrous cycle, permanent anoestrus, interfere with ovulation from other follicles but it could
decrease the number of normal viable eggs (Castagna et al.,
2004).
∗ Corresponding author. Tel.: +48 692 697 616; fax: +48 71 320 1006. Social stress may influence the delicate hormonal pat-
E-mail addresses: wbie@interia.pl, wbielas@up.wroc.pl (W. Bielas). tern around the time of ovulation and fertilization. It is

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.001
274 K. Szulańczyk-Mencel et al. / Animal Reproduction Science 121 (2010) 273–278

well known that stress can have a negative effect on the it is well known that this pathology is connected with
reproductive performance in animals. There is a cascade hormonal disturbances. It is complex to diagnose cysts
of hormonal events following the immediate endocrine trough serological methods because plasma concentrations
response to a stressor (Castagna et al., 2004; Prunier and of progesterone, estradiol and luteinizing hormone in this
Quesnel, 2000; Razdan et al., 2002). Stress is known to disturbance are similar in sows during diestrus (Castagna
be accompanied by elevated levels of ACTH (Close and et al., 2004).
Liptrap, 1975). ACTH is released by the anterior pituitary The aim of this study was to evaluate the relation-
gland in response to a stressful stimulus. ACTH acts on the ship between ovarian cysts and concentrations of ovarian
adrenal cortex causing it to produce and release cortisol steroid hormones: 17␤-estradiol (E2 ), progesterone (P4) ,
which affects reproductive organs and several hormonal testosterone (T), and androstendione (A4 ) both in blood
pathways (Brandt et al., 2007; Tsuma et al., 1996). A lack plasma and in cysts and morphological state of the ovarian
of luteinizing hormone (LH) that is necessary to induce cortex in sows.
ovulation has been suggested as the main cause of cystic
ovaries in the sow. Sows with weaning to estrus inter- 2. Material and methods
vals shorter than 3 days and a lactation length shorter
than 14 days have a greater probability of developing 2.1. Animals
ovarian cysts. It’s suggested that the cause of this state
is insufficient LH surge system so follicles fail to ovu- The experiment was performed with 18 purebred, 2–5
late and become cystic (Castagna et al., 2004; Knox et al., years old multiparous Rodone sows from a commercial pig-
2003). breeding farm located in the Region of Leszno, Poland. Sows
It was also shown that hypothyroidism promotes ovar- were fed with a commercial diet twice daily on the farm
ian cysts formation. The role of thyroid hormones in ovarian and had ad libitum access to water. Experimental females
function was evidenced by many authors (Fitko et al., derived from animals which were culled from the farm
1996). Progesterone of adrenal origin may serve, at least mainly due to fertility disturbances. Every culled sow was
in part, as a component in the etiology of this kind of infer- routinely checked for the presence of ovarian cysts before
tility (Brandt et al., 2007; Close and Liptrap, 1975; Madej et slaughtering, using transcutaneous ultrasound examina-
al., 2005; Scholten and Liptrap, 1978). Injections of proges- tion, according to Kauffold and Althouse (2007). Ovaries
terone during the follicular phase of the cycle also caused with ovarian cysts from the particular sow were taken into
development of ovarian cysts. Administration of proges- laboratory from slaughterhouse. After macroscopic, his-
tational compounds in order to synchronize estrus cycles tological and histometrical examination we distinguished
in pigs is known as reason resulting in a high incidence of several different types of ovarian cysts: follicular cysts (FC),
cystic ovaries (Scholten and Liptrap, 1978). Moreover some follicular theca-lutein cysts (FTLC), follicular lutein cysts
authors suggest that the presence of ovarian cysts in sows (FLC) and cysts of corpus luteum (CCL). Reduced granu-
is related to the seasons of the year (Castagna et al., 2004). losa and theca layer are characteristic for thin-walled FC
The hormonal interactions of the hypothalamic– in these structures. In FC cysts membrana granulosa is vis-
pituitary–ovarian axis are accountable for a normal repro- ibly thinner than in mature follicles. FTLC have an enlarged
duction in female pigs. Activation of the hypothalamic– internal zone of theca. The cells of tunica interna undergo
pituitary–adrenal axis may hamper the normal luteinization. In FLC the wall of follicle was luteinized. The
gonadotropin secretion and in consequence the ovar- CLC derived from corpora lutea that result from exces-
ian activity (Madej et al., 2005). Steroid hormones are sive central hemorrhage. Corpus luteum becomes over
best-known and best-characterized secretory products distended with blood. They contain clear fluid or altered
of the ovary. The ovarian steroids fulfill a number of blood. At the beginning cystic corpora lutea display the
important functions related to reproduction by endocrine same features as their non-cystic counterparts, but if the
mechanisms of action. The steroids are produced by luteal hemorrhage is excessive their involution is delayed and
or follicular cells of the ovary. They may act as hormones they reach pathological size often exceeding 3–4 cm in
on structures remote from the ovary and as local regu- diameter. 19 sows (7.98%) out of 238 females culled during
lators of ovarian activity. These hormones play roles of 15 months had ovarian cysts. 7 sows (2.94%) had poly-
paracrine/autocrine agents acting on or within the cells cystical ovaries whilst 12 females (5.04%) had oligocystic
in which they are produced (Schams and Berisha, 2002). ovaries. Allotment of a sow to the polycystical or to the
These hormones exert important effects on the correct oligocystic ovaries group depended on the number of cysts
functions of the ovary. The disturbances of endocrino- which had been discovered on its ovaries; to the oligocys-
logical mechanisms which regulate the physiology of tic ovaries group, up to 10 cysts, to the polycystic ovaries
reproductive organs may lead to infertility. Cystic ovarian group, when more than 10 cysts were present. The dis-
disease is connected with various hormonal interrelations criminating number between polycystical and oligocystic
between sex steroids (Fitko et al., 1998a). Because COD ovaries was 10 cysts on ovaries as was chosen according
is an endocrine disorder we can suspect that there are to what described by Ebbert and Bostedt (1993). Females
some correlations between the concentrations of steroid were randomly divided into three groups: PCO group – 6
hormones and the structural changes in structure of the sows with polycystical ovaries, OO group – 6 sows with
ovaries. oligocystic ovaries and – control group (6 sows with-
It is very difficult to diagnose the cystic ovarian dis- out ovarian cysts). OO sows with oligocystic ovaries were
ease in pigs on the basis of the hormonal profile. But selected from out of 12 sows with oligocystic ovaries whilst
K. Szulańczyk-Mencel et al. / Animal Reproduction Science 121 (2010) 273–278 275

Table 1
Effect of polycystical and oligocystic ovaries on structural components of ovarian cortex in sows (mean ± SD).

Sows with Sows with Sows without


polycystical oligocystic ovarian cysts
ovaries (n = 6) ovaries (n = 6) (n = 6) (control)

Primary ovarian follicles 46.16 ± 2.8C 71.66 ± 6.25B 96.50 ± 4.12A


Growing ovarian follicles 27.83 ± 2.7C 33.66 ± 2.60B 43.08 ± 2.93A
Maturing ovarian follicles 4.33 ± 1.15B 6.75 ± 0.96A 6.25 ± 0.96A
Corpora lutea 4.50 ± 1.08A 5.25 ± 0.96A 3.41 ± 0.79B
Corpora albicans 4.16 ± 1.11B 4.33 ± 1.43B 8.83 ± 1.11A
Atresia of primary ovarian Follicles 112.75 ± 3.19A 92.91 ± 4.14B 40.50 ± 5.0C
Atresia of growing ovarian Follicles 55.41 ± 2.35A 54.00 ± 2.92A 13.75 ± 2.41B
Atresia of maturing ovarian Follicles 16.16 ± 1.52A 14.08 ± 2.77A 3.83 ± 1.02B
Follicular cysts 8.66 ± 1.61A 1.58 ± 1.62A 0.50 ± 0.90B
Follicular theca-lutein cysts 8.00 ± 1.12A 2.00 ± 1.12B 0.16 ± 0.3C
Follicular lutein cysts 4.50 ± 1.62A 2.00 ± 0.95B 0.00 ± 0.0C
Corpus luteum cysts 1.75 ± 0.75A 0.41 ± 0.66B 0.00 ± 0.0C

Mean values marked with different capital letters, different at P ≤ 0.01.

control sows were selected from out of 219 animals with- 2.5. Histology and histometry
out ovarian cysts which were culled from the farm mainly
due to low fecundity, overweight, hoofs diseases and other The ovaries collected at the slaughterhouse were fixed
ailments. in 4% formalin for 48 h and embedded in paraffin. Then the
organs were cut at 6–7 ␮m, fixed on slides, dyed, deparaf-
finized and rehydrated in ethanol solutions. The slides were
2.2. Blood sampling stained with hematoxylin/eosin. The amount of ovarian fol-
licles of all generations, corpora lutea, and corpora albicans,
Peripheral blood was collected by a puncture of anterior atretic follicles and different kind of cysts were counted on
vena cava directly after the diagnosis of ovarian cysts in five sections from each ovary.
culled sows. The blood samples were centrifuged at 1500 g
for 10 min. Plasma samples were stored at −20 ◦ C until con- 2.6. Statistics
centrations of 17␤-estradiol, progesterone, testosterone,
and androstendione were determined. Treatment comparisons were made by analysis of vari-
ance procedures for a completely randomised design using
STATISTICA 8 statistical package. All the values were
2.3. Ovarian cysts fluid presented as mean ± SEM. Differences were considered sig-
nificant if the P value was less than 0.05 or 0.01. Differences
The samples of cysts fluid were collected post-mortem, between means were compared by the Duncan test when
frozen and stored at −20 ◦ C until the concentrations the analysis was done for three groups and by Tukey test
of hormones were determined. Cyst fluid was collected when the analysis was done for two groups. The Pearson’s
from random cysts from either polycystical or oligocystic correlations between analysed factors were calculated.
ovaries. The experimental protocol had previously been
reviewed and approved by the II Local Ethical Committee
for Experimentation with Animals, Wroclaw, Poland.
2.4. Hormone assays
3. Results
The concentration of 17␤-estradiol, progesterone,
testosterone, and androstendione in the blood plasma Table 1 presents the effect of polycystical and oligocys-
was determined by radioimmunoassay (Coat-A-Count, tic ovaries on structural components of the ovarian cortex
Diagnostic Products Corporation, Los Angeles, CA, USA) in sows. There were statistically significant differences in
according to the manufacturer’s instructions. numbers of FC (8.6 vs. 1.5), FTLC (8.0 vs. 2.0), FLC (4.5 vs.

Table 2
Effect of polycystical and oligocystic ovaries on concentrations of steroid hormones in blood plasma of sows (mean ± SD).

Sows with Sows with Sows without ovarian


polycystical oligocystic ovaries cysts (control) (n = 6)
ovaries (n = 6) (n = 6) (control)

17␤-estradiol pg/ml 5.19 ± 0.69 8.24 ± 5.29 6.47 ± 1.43


Progesterone ng/ml 22.05 ± 13.83 16.23 ± 14.05 12.34 ± 12.76
Testosterone ng/ml 0.04 ± 0.02 0.05 ± 0.02a 0.03 ± 0.02b
Androstendione ng/ml 0.06 ± 0.06b 0.14 ± 0.12 0.22 ± 0.08a

Mean values marked with different small letters, different at P ≤ 0.05.


276 K. Szulańczyk-Mencel et al. / Animal Reproduction Science 121 (2010) 273–278

Table 3
Concentrations of steroid hormones between polycystical and oligocystic ovaries in liquids derived from cysts of cystical sows (mean ± SD).

Steroid hormones Sows with polycystical ovaries (n = 6) Sows with oligocystic ovaries (n = 6)

17␤-estradiol pg/ml 1,195,839 ± 884,095A 10,714 ± 28,791B


Progesterone ng/ml 615,770 ± 801,750 926,508 ± 379,163
Testosterone ng/ml 3820 ± 3592A 448 ± 237B
Androstendione ng/ml 7272 ± 5072A 035 ± 1395B

Mean values marked with different capital letters, different at P ≤ 0.01.

2.0) and CLC (1.7 vs. 0.4) between the PCO and OO sows, centrations of steroid hormones in blood of the studied
respectively (P ≤ 0.01). An increasing number of ovarian sows (P > 0.05). The general presence of ovarian cysts sig-
cysts significantly decreases the frequency of physiologi- nificantly positively correlated both with concentrations of
cal ovarian follicles (primary, growing and maturing) and E2 , T and A 4 , derived from all kinds of cysts’ fluid, and with
significantly increases the pathological process of atresia all kinds of ovarian cysts and atresia of primary follicles (a
on all stages of ovarian follicles development (P ≤ 0.01). correlation coefficient r from 0.72 up to 0.97, P ≤ 0.05).
Table 2 presents the effect of polycystical and oligocys-
tic ovaries on concentrations of steroid hormones in blood 4. Discussion
plasma of sows. No effect of growing or decreasing num-
ber of ovarian cyst on concentrations of E2 and P4 in blood The ovarian cysts are an important reproductive disor-
plasma of sows was found (P > 0.01). Polycystical ovaries der in pigs (Ribadu et al., 2000). The formation of ovarian
significantly decreased concentrations of A4 but increased cysts is multifactorial. Many factors such as stress, nutri-
concentration of T in blood plasma (P ≤ 0.01). tion, and season are associated with the development
Table 3 compares concentrations of steroid hormones of ovarian cysts, although their exact relationship is not
between polycystical and oligocystic ovaries in liquids fully understood. Additionally, there are several limita-
derived from cysts of cystical sows. Cysts from polycys- tions in scientific studies on the spontaneous occurrences
tical ovaries produced significantly larger quantities of E2 , of ovarian cysts. The immediate reason of this condition
T and A4 than oligocystic ovaries, whilst P4 concentration is believed to be an alteration in gonadotropin secretion
was almost the same. (Peter and Liptrap, 1985); however the pathogenesis of
Table 4 presents correlations between the general ovarian cysts development is still unknown not only in
occurrence of ovarian cysts and the morphological and hor- the case of animals but also in women to. Perhaps, this is,
monal state of the cortex of ovary in sows. The phenomenon at least partially, a reason for ineffective therapy of this
of ovarian cysts significantly negatively correlated with all ailment.
generations of ovaries follicles (P ≤ 0.05). There were not We noted that the increase of the number of ovar-
any correlations between cysts phenomenon and the con- ian cysts was accompanied by elevated number of atretic
follicles, and on the other hand, this increase negatively
correlates with normal non-atretic ones. The atresia pro-
Table 4
cess was especially observed within primary follicles both
Correlations between general occurrence of ovarian cysts and morpho-
logical and hormonal state of cortex of ovary in sows (n = 12). in PCO and OO group of females. In the case of polycys-
tical ovaries we noted an advanced stage of degenerative
Studied factors of General occurrence of
changes in ovarian cortex which would have been a reason
sow ovaries ovarian cysts
functionality of persistent infertility. Our observations have demon-
Correlation strated that 7 females (7 out of 238) had 10 and more
coefficient (r) ovarian cysts on the ovaries. It appeared that this ail-
17␤-estradiol in blood −0.37 ment (polycystical ovaries) always caused reproductive
Progesterone in blood 0.26 disorders and reduced reproductive performance in these
Testosterone in blood −0.28 sows leading to culling them from herd. In the case of
Androstendione in blood −0.16
polycystical ovaries the corpora lutea or normal follicles
17␤-estradiol in cysts 0.80a
Progesterone in cysts −0.43 are often invisible in macroscopic investigation due to
Testosterone in cysts 0.73a a high number of large cysts present on the examined
Androstendione in cysts 0.75a ovary. Therefore, only precise histological and histomet-
Primary ovarian follicles −0.94a
rical investigations would allow evaluating the state of
Growing ovarian follicles −0.83a
Maturing ovarian follicles −0.72a
ovarian cortex in case of cystic ovarian degeneration. On
Corpora lutea −0.50 the contrary, the oligocystic ovaries are not accompanied
Corpora albicans −0.27 by intense unfavorable changes in ovarian cortex, and we
Atresia of primary ovarian follicles −0. 89a cannot exclude the possibility of ovulation and pregnancy
Atresia of growing ovarian follicles 0.27
in sows with this kind of cysts. 12 sows (12 out of 238
Atresia of maturing ovarian follicles 0.38
Follicular cysts 0.99a culled) exhibited only up to 10 cysts on ovaries. It was
Follicular theca-lutein cysts 0. 97a found in these and in the earlier studies that females
Follicular lutein cysts 0.76a with oligocystic ovaries were never culled from herd
Corpus luteum cysts 0.88a
by reproductive failures (Szulańczyk-Mencel and Bielas,
a
Correlation coefficient is highly significant, P ≤ 0.01. 2008).
K. Szulańczyk-Mencel et al. / Animal Reproduction Science 121 (2010) 273–278 277

We have to underline that the cystic ovary disorder trations of A4 , increased the concentration of T in blood
(COD) is a dynamic process, some of the cysts can disappear plasma and significantly negatively correlated with all gen-
spontaneously or undergo the process of degeneration, so erations of ovarian follicles. Therefore we should give more
we could suspect that the hormonal profile which accom- attention to these hormones in future researches. Elevated
panies this state is instable. It is very difficult to define levels of circulating androgens (hyperandrogenemia) and
for how long the cysts may have existed before diagno- clinical manifestation of androgen excess (hyperandro-
sis. 25–30% of the ascertained cysts disappeared when they genism) are the symptoms of polycystic ovarian syndrome
were evaluated by ultrasound examination until the 15th (PCOS) in women. Thus, experimental pigs may be a suit-
day of evaluation (Castagna et al., 2004). Lucy et al. (1999) able and useful model for the study of some aspects of
reported that approximately 1 week after the regression pathophysiology of PCOS in women and COD in female
of all the cysts on investigated ovaries the new cohort of animals (Ribadu et al., 2000).
follicles was formed on them. Correlations between various hormones or between
From the analysis of the hormonal profile in sows with hormones and the morphological state of ovaries have been
ovarian cysts in comparison to sows without cysts it may very rarely investigated up to this time. Therefore this work
be concluded that it is very difficult to ascertain the ovarian may provide valuable data concerning endocrine regula-
cysts presence on the basis of endocrinological investiga- tions, abnormalities in hormone secretion and dysfunction
tions. The estimation of hormone levels in the blood of of ovarian cortex in the case of ovarian cysts presence in
sows with naturally occurring cystic ovarian disease (COD) sows. Disturbed ovarian function caused serious deregu-
is rare and the results are very diverse. The concentration lations in the hormonal pattern of these animals. These
of estimated hormones in blood plasma showed a limited disturbances changed the hormonal profiles and elimi-
and incomplete parallelism (Ribadu et al., 2000). Moreover, nated the characteristic hormone fluctuations and peaks of
many results of ovarian steroid hormones were obtained secretion around the estrus period. Therefore, these facts
experimentally from sows with artificially induced ovarian should be taken into consideration when the hormonal
cysts. treatment of animals is undertaken (Fitko et al., 1998b).
In our studies the concentration of hormones in blood In conclusion some correlations between hormonal pro-
plasma of sows varied among groups as well as between file, state of ovarian cortex and cysts presence are now
individual animals. The differences between sows may documented and may stimulate further research in this
be due to individual, physiological variations to stres- field.
sors (Scholten and Liptrap, 1978). These differences may
result from the fact that the hormones produced by the
Acknowledgements
ovaries undergo various transformations (binding to spe-
cific proteins or receptors, conversion to other hormones,
This study was supported by grant KBN 7/14-W/2008/G.
elimination etc.) after their release into general blood circu-
lation. This is probably the reason why the data on hormone
concentration in blood plasma usually does not reflect hor- References
mone content in cystical fluid (Fitko et al., 1998a).
Brandt, Y., Lundeheim, N., Madej, A., Rodriguez-Martinez, H., Einarsson,
Results of this work do not confirm some details in the
S., 2007. Effects of ACTH injections during estrus on concentrations
findings of other authors investigating hormone levels in and patterns of progesterone, estradiol, LH, and inhibin ␣ and time of
COD in pigs. It is probably possible that this fact may be ovulation in the sow. Dom. Anim. Endocrinol. 32, 122–137.
Castagna, C.D., Peixoto, C.H., Bortolozzo, F.P., Wentz, I., Neto, G.B., Ruschel,
connected with the differences in degree of ovarian degen-
F., 2004. Ovarian cysts and their consequences on the reproductive
eration in females in our studies. The changes in steroids performance in swine herds. Anim. Reprod. Sci. 81, 115–123.
profile indicated that in sows with the presence of ovarian Close, R.W., Liptrap, R.M., 1975. Plasma progesterone levels in sows with
cysts some disturbances in hormonal metabolism exist. The induced cystic ovarian follicles. Res. Vet. Sci. 19, 28–34.
Ebbert, W., Bostedt, H., 1993. Cystic degeneration in porcine ovaries –
disturbances in ovarian steroid genesis in sows may also first communication: morphology of cystic ovaries, interpretation of
have resulted from a defect in aromatase complex which the results. Reprod. Dom. Anim. 28, 441–450.
caused an increased conversion of A4 to E1 and T to E2 in Fitko, R., Jana, B., Kucharski, J., 1998a. Correlations of hormone concen-
trations in blood plasma and their relations to ovarian structures in
peripheral fat tissue (Ribadu et al., 2000). hypo- and hyperthyroid cyst-bearing gilts. Pol J. Vet. Sci. 1, 11–15.
The data obtained in our studies revealed the distur- Fitko, R., Jana, B., Kucharski, J., Sobczak, J., 1998b. Changes in periestrous
bances in hormone production or secretion in examined hormonal profile in blood of gilts with different thyroid function and
ovary cysts. Pol J. Vet. Sci. 1, 3–9.
sows. We found some correlations between the hormone Fitko, R., Kucharski, J., Szlezyngier, B., Jana, B., 1996. The concentration
concentrations and the state of ovarian cortex in cystic of GnRH in hypothalamus, LH and FSH in pituitary, LH PRL and sex
ovaries in sows. The data evidenced different interrela- steroids in peripheral and ovarian venous plasma of hypo- and hyper-
thyroid, cysts-bearing gilts. Anim. Reprod. Sci. 45, 123–138.
tions between concentrations of hormones in the blood
Kauffold, J., Althouse, G.C., 2007. An update on the use of B-mode ultra-
plasma and cystical fluid and a number of various ovarian sonography in female pig reproduction. Theriogenology 67, 901–911.
structures. These abnormalities were probably the conse- Knox, R.V., Vatzias, G., Naber, C.H., Zimmerman, D.R., 2003. Plasma
gonadotropins and ovarian hormones during the estrous cycle in high
quences of a disturbed ovarian function connected with
compared to low ovulation rate gilts. J. Anim. Sci. 81, 249–260.
structural and functional changes in the ovaries. Lucy, M.C., Liu, J., Koenigsfeld, A.T., Cantley, T.C., Keisler, D.H., 1999.
We also noted some differences in the concentration of Ultrasonically-measured ovarian follicular development in weaned
testosterone and androstendione – hormones in which lev- sows. Biol. Reprod. 60 (Suppl. 1), 166–172.
Madej, A., Lang, A., Brandt, Y., Kindahl, H., Madsen, M.T., Einarsson, S., 2005.
els are not commonly measured in blood plasma of female Factors regulating ovarian function in pigs. Dom. Anim. Endocrinol. 29,
pigs. Polycystical ovaries significantly decreased concen- 347–361.
278 K. Szulańczyk-Mencel et al. / Animal Reproduction Science 121 (2010) 273–278

Peter, A.T., Liptrap, R.M., 1985. Plasma gonadotrophin levels in sows with Schams, D., Berisha, B., 2002. Steroids as a local regulators of ovarian
experimental cystic ovarian follicles. Br. Vet. J. 141, 288–296. activity in domestic animals. Dom. Anim. Endocrinol. 23, 53–65.
Prunier, A., Quesnel, H., 2000. Nutritional influences on the hormonal Scholten, J.A., Liptrap, R.M., 1978. A role of the adrenal cortex in the onset
control of reproduction in female pigs. Livestock Prod. Sci. 63, 1–16. of cystic ovarian follicles in the sow. Can. J. Comp. Med. 42, 525–533.
Razdan, P., Mwanza, A.M., Kindahl, H., Rodriguez-Martinez, H., Hul- Szulańczyk-Mencel, K., Bielas, W., 2008. Relationships between ovarian
ten, F., Einarsson, S., 2002. Effect of repeated ACTH-stimulation on cysts and changes in mucosa of the oviduct and uterus in sows. Bull.
early embryonic development and hormonal profiles in sows. Anim. Vet. Inst. Pulawy 52, 235–238.
Reprod. Sci. 70, 127–137. Tsuma, V.T., Einarsson, S., Madej, A., Kindahl, H., Lundeheim, N., Rojkit-
Ribadu, A.Y., Nakada, K., Moriyoshi, M., Zhang, W.C., Tanaka, Y., Nakao, T., tikhum, T., 1996. Endocrine changes during group housing of
2000. The role of Pulse frequency in ACTH-induced ovarian follicular primiparous sows in early pregnancy. Acta. Vet. Scand. 37, 481–490.
cysts in heifers. Anim. Reprod. Sci. 64, 21–31.
Animal Reproduction Science 121 (2010) 279–285

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Embryo transfer technique: Factors affecting the viability of the corpus


luteum in llamas
Virginia Trasorras a,∗ , María Graciela Chaves a , Deborah Neild a , Mariana Gambarotta b ,
Marcelo Aba c , Alicia Agüero a
a
Área de Teriogenología, Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos
Aires (UBA), Av. Chorroarín 280 (1427), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
b
Área de Bioestadística, Facultad de Ciencias Veterinarias, UBA, Argentina
c
Área de Endocrinología, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (U.N.C.P.B.A.), Campus
Universitario, Paraje Arroyo Seco s/n, Tandil, 7000 Buenos Aires, Argentina

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

Article history: The aim of the present study was to evaluate the effect of the embryo transfer (ET) maneu-
Received 24 November 2009 vers on plasma progesterone concentrations in recipient Lama glama females and the
Received in revised form 9 June 2010
relationship between the site the embryo was transferred to and corpus luteum (CL) local-
Accepted 14 June 2010
ization. Experiment I (effect of transcervical threading): adult non-pregnant, non-lactating
Available online 19 June 2010
llama females were randomly assigned into two groups: control group (without cervical
threading, n = 10) and group A (with cervical threading, n = 10). In both groups, CL activity
Keywords:
was evaluated through measurement of progesterone plasma concentrations. In group A,
Llama
Corpus luteum on Day 6 after inducing ovulation with buserelin, the cervix was threaded to evaluate the
Progesterone effect of the maneuver on CL viability. No significant differences were observed in mean
Embryo transfer progesterone concentrations between groups (P > 0.05). Experiment II (effect of depositing
Maternal recognition of pregnancy PBS): females (n = 66) were randomly assigned into six groups (n = 10 per group and con-
trol group: n = 6) to evaluate the effect of depositing PBS in different sites in the uterus in
relation to the localization of the CL: group ‘Left-Ipsilateral’: transcervical placing of PBS
in the left uterine horn (CL in left ovary); group ‘Left-Contralateral’: transcervical placing
of PBS in the left uterine horn (CL in right ovary); group ‘Right-Ipsilateral’: transcervical
placing of PBS in the right uterine horn (CL in right ovary); group ‘Body-Left’: transcervi-
cal placing of PBS in the uterine body (CL in left ovary); group ‘Body-Right’: transcervical
placing of PBS in the uterine body (CL in right ovary) and control group. Corpus luteum
activity was evaluated in all groups by measuring plasma progesterone concentrations.
On Day 6 post-buserelin, the corresponding maneuver was carried out according to the
group. No significant differences were found for the mean plasma progesterone concen-
trations between groups (P > 0.05). Experiment III (effect of ET on CL viability): females
(n = 22) were used as embryo donors and 50 females as recipients, in order to evaluate if
placing the embryo in different areas of the uterus influences CL viability. Recipients were
randomly divided into five groups, according to the place in the uterus where the ET was
conducted with respect to the ovary where ovulation occurred: group ‘Left-Ipsilateral’:

∗ Corresponding author. Tel.: +54 011 4524 8425; fax: +54 011 4524 8425.
E-mail addresses: vtrasorras@fvet.uba.ar, vickytrasorras@hotmail.com (V. Trasorras).

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.004
280 V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285

ET in the left uterine horn (CL in left ovary); group ‘Left-Contralateral’: ET in the left uter-
ine horn (CL in right ovary); group ‘Right-Ipsilateral’: ET in the right uterine horn (CL in
right ovary); group ‘Body-Left’: ET in the uterine body (CL in left ovary) and group ‘Body-
Right’: ET in the uterine body (CL in right ovary). Corpus luteum activity was evaluated in
all groups by measuring plasma progesterone concentrations. Embryos were recovered by
flushing the uterus on Day 8 after the first mating of the donor and transcervical ET was
carried out in recipients 6 days after buserelin administration. Pregnancy rates were: group
‘Left-Ipsilateral’: 50%; group ‘Left-Contralateral’: 20%; group ‘Right-Ipsilateral’: 30%; group
‘Body-Left’ and ‘Body-Right’: 10%. No significant differences (P = 0.4728) were detected
between the pregnancy rates in the five groups. Threading the cervix and transcervical
placing of PBS either in the uterine horns or the body did not affect plasma progesterone
concentrations in the llama, indicating that the different embryo transfer maneuvers do
not interfere with CL viability. To improve pregnancy rates it could be suggested that ET in
the left uterine horn with an ipsilateral CL, is the most desirable option.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction the uterus and for MRP to maintain the CL viable in lla-
mas.
There have been minimal amounts of reproduction- Del Campo et al. (1996) found luteolytic action of the
focused research with South American Camelids (SAC). right uterine horn is solely local, while the luteolytic action
Complex reproductive characteristics of these species con- of the left uterine horn is on the CL located on the left
tribute to the lack of research: induced ovulation, short and right ovaries. This difference in luteolytic activity
half-life of the CL, different luteolytic activity between between uterine horns can be explained by the differ-
the two uterine horns, maternal recognition of pregnancy ent oviduct, ovarian and uterine vascular anatomy in the
(MRP) beginning before Day 10 after mating, pregnan- camelid female. The main vein originating from the left
cies only in left uterine horn, highly viscous semen with uterine horn crosses the midline and ends as a branch of the
low sperm concentration, etc. Nevertheless, some assisted right uterine horn vein. Therefore, vascular anatomy indi-
reproductive techniques (such as synchronization of time cates that much of the blood-flow of the left uterine horn
of ovarian follicular development, superstimulation and vein crosses over to the right side, thus being able to exert a
embryo transfer) indicate a greater increase in knowledge luteolytic control of the CL in the right ovary (Del Campo et
while others show less advances (artificial insemination, al., 1996). Although ovulations occur with equal frequency
in vitro fertilization and intracytoplasmic sperm injection) from both ovaries, the majority of pregnancies are localized
(Miragaya et al., 2006; Tibary et al., 2005) and certain basic in the left uterine horn (alpaca: 97.5 and 99.3% with a CL
reproductive physiology remains unclear such as MRP sig- in the right and left ovaries respectively, Fernández-Baca
naling. et al., 1973, 1979; llama: 100%, Sumar, 1988). When ovu-
In their natural habitat (Andean regions) SAC demon- lation occurs in the right ovary, the embryo must probably
strate a seasonal reproductive behavior, with mating and migrate from the right uterine horn to the left to be able to
parturitions occurring predominantly in the rainy season conduct MRP (Sumar and Leyva, 1979). These observations
when more food is available (November–March). In areas allow us to hypothesize that the success of ET could depend
at sea level (Buenos Aires, Entre Ríos, etc.) characterized on the site where the embryo is deposited in the uterus.
by a higher quality food supply, llamas are considered To date, the poor pregnancy rates obtained using tran-
non-seasonal because of follicular growth throughout the scervical ET are attributed to a high release of prostaglandin
year. Nevertheless in these areas during the same months, because of cervix manipulation, a weak signal from the
with greater ambient temperatures, fertilizing capacity embryo or a deficient embryo migration. In mares, the
in males is less (Giuliano et al., 2000, 2008). This short cervical threading involved in ET produces an increase in
reproductive period added to the long gestation of approx- endometrial PGF2␣ release, thus compromising CL viability
imately 11 months (Johnson, 1989; Leon et al., 1990), (Kask et al., 1997) but this has not yet been studied in SAC.
are partly responsible for the poor reproductive efficiency Therefore, the aim of the present study was to evaluate
observed in camelids. Application of reproductive biotech- the effect of uterine handling during ET on plasma proges-
nologies such as embryo transfer (ET) would allow an terone concentrations in recipient Lama glama females and
increase in the number of offspring from selected males the relationship between the place the embryo is deposited
and females and consequently increase the proportion of and CL localization, to improve the pregnancy rates using
genetically superior animals. This technique has spread ET.
widely as a practical production method in domestic ani-
mals (horses: Allen, 2005; cattle: Hasler, 2003). In llamas
there has been little success (Aller et al., 2002; Bourke 2. Materials and methods
et al., 1992; Taylor et al., 2000). The luteolytic release
of PGF2␣ starting on Day 7 or 8 post-mating and com- 2.1. Animals
pleted by Day 9 or 10 after mating, shortens the half-life
of the CL to 8 or 9 days (Aba et al., 1995, 2000) lim- Non-pregnant, non-lactating female llamas (n = 70)
iting the period necessary for transferring embryos to ranging between 4 and 8 years of age and with an average
V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285 281

body weight of 120 ± 22 kg were used in this study. Females pipette, covered with a sterile sheath, was inserted into the
were kept separated from males during the experiment vagina. Cervical threading was performed through tran-
and fed bales of hay and water ad libitum. The study was srectal manipulation. Blood samples were obtained once
conducted at the Faculty of Veterinary Sciences of the Uni- daily from both groups between Day −2 and 10 (Day 0:
versity of Buenos Aires, Buenos Aires, Argentina, situated induction of ovulation using buserelin).
34◦ 36 S and 58◦ 26 W, at sea level.
2.4.2. Experiment II
2.2. Ultrasonographic evaluation of ovarian dynamics The objective of experiment II was to evaluate the effect
and induction of ovulation of depositing PBS in different sites in the uterus in rela-
tion to CL localization and its relationship to CL lifespan.
Ovarian dynamics was monitored by transrectal pal- Females were randomly divided into five treatment groups
pation and ultrasonography (Berger LC 2010 plus with (n = 10) and one control group (n = 6). Treatment groups
a 5 MHz linear-array electronic transducer, Buenos Aires, were assigned as follows: group ‘Left-Ipsilateral’: transcer-
Argentina). When a dominant follicle (≥7 mm in diam- vical placing of PBS in the left uterine horn (CL in left
eter) was detected, a single dose of 8 ␮g of buserelin ovary); group ‘Left-Contralateral’: transcervical placing of
(GnRH synthetic analogue, Receptal® , Intervet, Buenos PBS in the left uterine horn (CL in right ovary); group
Aires, Argentina) was administered IV (day 0) for inducing ‘Right-Ipsilateral’: transcervical placing of PBS in the right
ovulation. Ovulation was confirmed on Day 2 using tran- uterine horn (CL in right ovary); group ‘Body-Left’: tran-
srectal ultrasonography based on the disappearance of the scervical placing of PBS in the uterine body (CL in left
dominant follicle and observation of a CL on Day 5. ovary); group ‘Body-Right’: transcervical placing of PBS in
the uterine body (CL in right ovary). Ovulation was induced
2.3. Hormone assay in all groups (treatment and control). On Day 6 after induc-
ing ovulation in treatment groups 0.25 ml of sterile D-PBS
Corpus luteum activity was evaluated in all groups (Gibco® , Grand Island, NY, USA) was deposited in the cor-
by measuring plasma progesterone concentrations using responding uterine site using an ET pipette. Control group
radioimmunoassay (RIA). Blood samples were obtained by females were not subject to any ET procedure. Blood sam-
jugular venipuncture using heparin as the anticoagulant. ples were obtained once daily in all groups from Day 5 to
The moment of extraction varied according to the exper- 12 after induction of ovulation with buserelin.
iment. Blood samples were immediately centrifuged at
3000 rpm for 20 min and the plasma separated and kept 2.4.3. Experiment III
at −20 ◦ C in plastic 5 ml tubes (Polistor® , Buenos Aires, The objective of this experiment was to evaluate
Argentina) until analysis. whether embryo transfer to different sites in the uterus
Progesterone was measured using a RIA kit (Diagnostic affects CL viability.
Products Corporation, Los Angeles, CA, USA) validated for
use in llama plasma (Bianchi et al., 2007). The sensitivity of 2.4.3.1. Treatment of donor llamas. The ovarian dynamics
the assay was 0.3 nmol l−1 and the intra-assay coefficient of all donor females (n = 22) was examined by transrec-
of variation was below 9% for concentrations between 0.4 tal ultrasonography and the absence of follicles bigger
and 128 nmol l−1 . All samples were measured in duplicates than 5 mm was confirmed before beginning the treatment.
and in one single assay for each experiment. A single IM dose (1000 IU) of eCG (Novormon® , Syntex,
Buenos Aires, Argentina) was administered (Trasorras et
2.4. Experimental procedure al., 2009) to stimulate multiple follicle growth. Daily ultra-
sonographic examinations of the ovaries were continued
2.4.1. Experiment I until two or more dominant follicles were found and at
The objective of this experiment was to evaluate the that moment, donor females were mated twice, 24 h apart
effect of transcervical threading on the viability of the CL. to increase the number of available spermatozoa, using dif-
Females were randomly separated into two groups: control ferent fertile L. glama males to minimize the male effect. In
group (n = 10) and group A (n = 10). In both groups, CL activ- addition, ovulation was assured in all donor llamas with a
ity after induction of ovulation was evaluated by measuring single IV dose of 8 ␮g of buserelin after the first mating.
plasma progesterone concentrations. In the control group,
females were not subject to any procedure. In group A, the 2.4.3.2. Treatment of recipient llamas. Synchronization of
maneuver of threading the cervix was conducted on Day recipient–donor females using a single dose of buserelin
6 after inducing ovulation to determine if plasma proges- in the presence of a dominant follicle was conducted 2
terone concentrations decrease because of PGF2␣ release days after inducing ovulation in the donor female. Recip-
during the process. The maneuvers were conducted with ient females (n = 50) were divided into five groups (n = 10
the female either standing or in sternal recumbency. The per group) according to the place in the uterus where the ET
animal was restrained in stocks, the tail was wrapped and was carried out and the ovary where ovulation occurred:
the rectum was emptied of feces. The perineum was then group ‘Left-Ipsilateral’: ET in the left uterine horn (CL in
scrubbed using an iodine solution, rinsed carefully with left ovary); group ‘Left-Contralateral’: ET in the left uter-
clean water and then dried. A lubricated gloved hand was ine horn (CL in right ovary); group ‘Right-Ipsilateral’: ET
inserted in the rectum to hold the cervix while an assistant in the right uterine horn (CL in right ovary); group ‘Body-
separated the vulva labia and an artificial insemination (AI) Left’: ET in the uterine body (CL in left ovary) and group
282 V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285

‘Body-Right’: ET in the uterine body (CL in right ovary).


Transcervical ET was performed on Day 6 after buserelin
administration (coinciding with the day of uterine flushing
of the donor female).
Blood samples were obtained once daily in all groups
from Day 5 to 12 after inducing ovulation with buserelin.
A progesterone profile, belonging to non-pregnant, non-
embryo transferred females (n = 6) was used (reference
profile) for comparison to the other groups.

2.4.3.3. Embryo recovery and transfer. The uterus of the


22 mated donor llamas were transcervically flushed for Fig. 1. Mean plasma progesterone concentrations in control group and
embryo recovery 8 days after the first mating. Aggressive group A (cervical threading). Arrows represent treatment days: Day
females received 0.2 mg/kg xylazine IV (Rompun® , Bayer, 0: buserelin; Day 6: cervical threading maneuver. Values represent
Buenos Aires, Argentina) before flushing. Collection was means ± SE.

conducted using a Foley catheter (12 or 16 Fr, according


to female size) and a stylet was inserted into the catheter 3. Results
to keep it from bending during recto-vaginal manipulation.
Uterine flushing was done by placing the catheter cuff 1 cm 3.1. Experiment I
cranial to the internal cervical os and inflating it with 5
or 10 ml of air (according to catheter gauge). The uterus Plasma progesterone concentrations in group A (with
was flushed 4–5 times with D-PBS supplemented with 1% cervical threading) and in control group (without cervical
fetal calf serum (FCS), using a total volume of 500 ml. After threading) were similar throughout the evaluation period
the flushing was finished, donor females received a single (Day −2 to 10 after buserelin), showing no significant dif-
IM dose of 250 ␮g of cloprostenol (Estrumate® , Schering- ferences between groups (P > 0.05) (Fig. 1).
Plough, Germany) to cause the lysis of all CLs in both
ovaries. 3.2. Experiment II
Embryos recovered were graded according to Tibary and
Anouassi (1997) and only grades I and II were used, consid- When evaluating the effect of depositing PBS in dif-
ering grade I: excellent quality embryo, size corresponds ferent sites of the uterus and its relationship to the
to the stage of collection in relation to ovulation; before localization of the CL, no significant differences were found
Day 8 should be perfectly spherical with smooth surface; between the mean plasma progesterone concentrations
and grade II: good embryo, same as grade I with some from groups ‘Left-Ipsilateral’, ‘Left-Contralateral’, ‘Right-
irregularities of the contour and very few protruded cells. Ipsilateral’, ‘Body-Left’, ‘Body-Right’ and the control group
These embryos were aspirated individually into 0.25 ml (P > 0.05) (Fig. 2).
straws (IMV® ET Straws, France), which were loaded into a
sheathed bovine/equine embryo transfer pipette (IMV® ET 3.3. Experiment III
Sheath, 21 , France). Then, embryos were transcervically
transferred into the uterus of the recipient llamas belong- After administration of 1000 IU of eCG, multiple folli-
ing to each group. cle growth required 5–7 days to reach dominant size. A
total of 67 embryos were recovered from the 22 llamas that
2.4.3.4. Pregnancy diagnosis. Pregnancy diagnosis was were flushed (recovery from each female was 3.04 ± 0.11
performed 13 days after the transfer by transrectal ultra- embryos, mean ± SE). Embryo size ranged from 0.3 to
sonographical visualization of the embryo vesicle and 3 1.2 mm in diameter and all of them were in the hatched
days later viability was confirmed by observing the heart-
beat.

2.5. Data analysis

In experiment I, plasma progesterone concentrations


from group A (with cervical threading) and control
group (without cervical threading) were analyzed using
a Student’s t test for comparing the means of each
group. In experiment II, ANOVA was used to evaluate
the means of groups ‘Left-Ipsilateral’, ‘Left-Contralateral’,
‘Right-Ipsitlateral’, ‘Body-Left’, ‘Body-Right’ and control
group. In experiment III, a chi-square test was performed Fig. 2. Mean plasma progesterone concentrations in group ‘Left-
Ipsilateral’, group ‘Left-Contralateral’, group ‘Right-Ipsilateral’, group
to compare pregnancy results. Statistical significance was
‘Body-Left’, group ‘Body-Right’ and control group. Day 6 after buserelin
set at P ≤ 0.05 and the statistical package used to analyze administration: PBS was deposited in the uterine site according to each
the data was InfoStat. group. Values correspond to means ± SE.
V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285 283

Fig. 3. Mean plasma progesterone concentration in non-pregnant and pregnant females from each ET group and non-transferred females (reference profile).
Values are means ± SE.

blastocyst stage (some embryos could be visualized with- in plasma progesterone concentrations was observed by
out optical instruments). This difference in embryo size is the time MRP was expected to occur in pregnant animals
probably due to the wide variability in occurrence of ovu- (between days 8 and 10) (Fig. 4).
lations in superstimulated animals.
When comparing the pregnancy rates in the dif- 4. Discussion
ferent groups no significant differences were detected
(P = 0.4728). Nevertheless, the ‘Left-Ipsilateral’ group To our knowledge this is the first comprehensive study
showed the highest pregnancy rate. The mean plasma pro- addressing the issue of the ET technique with regard to CL
gesterone concentrations in non-pregnant and pregnant viability in this species. The results hereby presented indi-
females of each ET group and those of the reference pro- cate that stimulation of the cervix during threading did not
file are presented in Fig. 3. Data for pregnancy rates are produce a decrease in plasma progesterone concentrations,
shown in Table 1. In non-pregnant females, a decrease as reflected by the lack of significant differences between

Table 1
Pregnancy rates in different groups according to ET in relationship to CL location.

Groups Left-Ipsilateral Left-Contralateral Right-Ipsilateral Body-Left Body-Right

Pregnancy rate % (pregnant females/n) 50 (5/10) 20 (2/10) 30 (3/10) 10 (1/10) 10 (1/10)


284 V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285

embryos carried out MRP in the left uterine horn where


they were deposited and then probably had to get a sig-
nal to the right horn. One possibility is migrating to the
right uterine horn to inhibit endometrial PGF2␣ release
locally and then return to the left uterine horn or elongat-
ing to establish pregnancy. In the dromedary it has been
suggested that migration is facilitated by the shortness of
the uterine body, the smallness of the right horn and the
increased number of mucosal folds in the endometrium of
the right horn (Musa, 1979). The time frame for this migra-
tion is short because camelid embryos have a rapid rate of
Fig. 4. Mean plasma progesterone concentrations in non-pregnant growth and rapidly start elongating (after day 9) and the
and pregnant females from all ET groups (group ‘Left-Ipsilateral’, uterus has a decreased tone and no contractions (Tibary
‘Left-Contralateral’, ‘Right-Ipsilateral’, ‘Body-Left’ and ‘Body-Right’) and and Anouassi, 1997). If migration is the way MRP is per-
non-transferred females (reference profile). Values are means ± SE.
formed, perhaps the lesser pregnancy rate in this group is
due to the fact that this double migration is not possible
the control group (without cervix manipulation) and group because the morphological changes of the embryo hinder
A (cervical threading). This observation is consistent with it.
results obtained in dromedaries (Skidmore et al., 2002). In group ‘Right-Ipsilateral’ 30% of the females were
Although these authors observed a short-lived increase in pregnant. In this case, embryos had to migrate from the
PGF2␣ produced during the passage of the ET pipette, this right uterine horn to the left one to establish pregnancy.
was not enough to harm the CL function or the pregnancy. This is physiologically normal because around 50.4% of
They suggest that the PGF2␣ release is either too short or pregnancies are supported by a right CL (Fernández-Baca
the amount is insufficient. et al., 1973, 1979). Tibary and Anouassi (1997) reported
Likewise, it was observed in the present study results from embryo transfer experiments showing that
that cervical threading together with transfer of ster- pregnancy is maintained with equal frequency whether
ile PBS to simulate the complete ET maneuver, did the recipient has a CL in the left or right ovary. They
not alter plasma progesterone concentrations in any of also observed that when embryos were deposited in the
the groups (‘Left-Ipsilateral’, ‘Left-Contralateral’, ‘Right- right uterine horn, they migrated to the left one after a
Ipsilateral’, ‘Body-Left’ and ‘Body-Right’) when compared few days and at ultrasound visualization on Days 15–18
to the control. Contrary to what occurs in the mare, where post-breeding, all embryos were located in the left uter-
inflammatory changes in the endometrium produce early ine horn. In the present study the ‘Right-Ipsilateral’ group
PG release (Koblischke et al., 2008), none of the females was expected to have similar results to the ‘Left-Ipsilateral’
in this experiment showed signs of inflammatory early (50% pregnancy rate) but this was not the case, although
release of PG after depositing PBS in the uterus, as the the number of animals in the study does not allow further
decrease in progesterone concentrations were similar to conclusions.
the control group (see Fig. 2). Therefore, it can be concluded The 10% pregnancy obtained in both groups where
that the ET technique as a whole (cervix manipulation and ET was performed in the uterine body was unex-
deposit of fluid in the uterus) is not responsible for the pected. Considering all the previous discussion, the
embryo loss occurring after transfer. percentage should have been higher in the “Body-Left”
Estimates are that more than 50% of embryo loss occurs group than the in the “Body-Right” group. Perhaps
during the first month of gestation (Fernández-Baca et al., proximity to the recently dilated cervix when the
1970) and this loss increases to 60–80% in the first 90 days embryos were deposited in the very short uterine body
(Alarcón et al., 1990) but the origin of these losses remains favors embryo loss through the cervix before MRP can
unknown. A possible cause for these losses is the failure of occur.
the right uterine horn to maintain pregnancy. In our study, Plasma progesterone concentrations in non-pregnant
the highest ET results were obtained when the embryo was females after ET were similar to that observed in females
deposited in the left uterine horn in the presence of a CL in belonging to the reference profile group, decreasing
the ipsilateral ovary (50%). Depositing the embryo in the between Days 8 and 10 after inducing ovulation; a similar
left horn is the only case where the embryo does not need decrease was observed in some of the groups of pregnant
to migrate to carry out MRP. Nevertheless, despite deposit- females (see Figs. 3 and 4). This coincides with the results
ing the embryo in the supposedly ideal uterine location, obtained by Aba et al. (1995), where pregnant females show
pregnancy rate is not as great as expected, therefore, there a slight decrease in plasma progesterone concentrations
would seem to be some other factor/s responsible for low between Days 8 and 18 but then recover and maintain sta-
embryo viability (for example, embryo defects, chromo- ble levels. This transitory fall in progesterone, which starts
some abnormalities, gamete ageing, etc.). Pregnancy rate at the same time as that observed in non-pregnant females,
decreases even more in the rest of the groups, where the would seem to indicate the presence of some factor that
embryo had to somehow get a signal to the other horn to inhibits luteolysis. Therefore MRP is a critical period that
maintain CL viability. determines the continuity or not of gestation. It is neces-
In group ‘Left-Contralateral’ 20% of the females were sary to further investigate the factor/s that allows embryos
pregnant. This case never happens physiologically, perhaps to continue their development.
V. Trasorras et al. / Animal Reproduction Science 121 (2010) 279–285 285

5. Conclusions Fernández-Baca, S., Sumar, J., Novoa, C., Leyva, V., 1973. Relación
entre la ubicación del cuerpo lúteo y la localización del
embrión en la alpaca. Revista de Investigaciones Pecuarias 2,
To our knowledge this is the first study to demonstrate 131–135.
that threading the cervix and transcervical placing of PBS Giuliano, S., Agüero, A., Spirito, S., Chaves, G., Boquet, M., Rutter, B., Fer-
in either the uterine horns or uterine body did not decrease rari, M., 2000. Heat stress effect on sperm morphology in Lama glama.
Biocell 24, 265.
plasma progesterone concentrations in llamas. Therefore, Giuliano, S., Director, A., Gambarotta, M., Trasorras, V., Miragaya, M.,
the different ET maneuvers do not interfere with CL via- 2008. Collection method, season and individual variation on semi-
bility. To improve pregnancy rates it would seem that ET nal characteristics in the llama (Lama glama). Anim. Reprod. Sci. 104,
359–369.
in the left uterine horn with an ipsilateral CL, is the most Hasler, J.F., 2003. The current status and future of commercial embryo
desirable option. transfer in cattle. Anim. Reprod. Sci. 79, 245–264.
Johnson, L.W., 1989. Llama reproduction. In: Johnson, L.W. (Ed.), The Vet-
erinary Clinics of North America, Llama Medicine. WB Saunders Co.,
Acknowledgements
Philadelphia, pp. 159–182.
Kask, K., Odensvik, K., Kindahl, H., 1997. Prostaglandin F2 alpha release
This research was supported by grants from the Uni- associated with an embryo transfer procedure in the mare. Equine
versity of Buenos Aires (UBACyT V018) and the Agencia Vet. J. 29, 286–289.
Koblischke, P., Kindahl, H., Budik, S., Aurich, J., Palm, F., Walter, I.,
Nacional de Promoción Científica y Tecnológica (PICT 2003- Kolodziejek, J., Nowotny, N., Hoppen, H.O., Aurich, C., 2008. Embryo
08-14231). The authors thank Ignacio Videla Dorna from transfer induces a subclinical endometritis in recipient mares which
Syntex Laboratories for donating the Novormon® for this can be prevented by treatment with non-steroid anti-inflammatory
drugs. Theriogenology 70, 1147–1158.
study. Leon, J.B., Smith, B.B., Timm, K.I., LeCren, G., 1990. Endocrine changes dur-
ing pregnancy, parturition and the early post-partum period in the
References llama (Lama glama). J. Reprod. Fertil. 88, 503–511.
Miragaya, M.H., Chaves, M.G., Agüero, A., 2006. Reproductive biotehnol-
ogy in South American camelids. Small Ruminant Res. 61,
Aba, M.A., Forsberg, M., Kindhal, H., Sumar, J., Edqvist, L.E., 1995. Endocrine
299–310.
changes after mating in pregnant and non-pregnant llamas and
Musa, B.E., 1979. Reproductive patterns in the female camel (Camelus
alpacas. Acta Vet. Scand. 36, 489–498.
dromedarius). In: Workshop in Camels, Karthoum, Sudan. Intl. Fnd.
Aba, M.A., Kindhal, H., Forsberg, M., Quiroga, M., Auza, N., 2000. Levels of
For. Sci., pp. 279–284.
progesterone and changes in prostaglandin F2␣ release during luteol-
Skidmore, J.A., Billah, M., Allen, W.R., 2002. Investigation of factors
ysis and early pregnancy in llamas and the effect of treatment with
affecting pregnancy rate after embryo transfer in dromedary camels.
flunixin meglumine. Anim. Reprod. Sci. 59 (1–2), 87–97.
Reprod. Fertil. Dev. 14, 109–116.
Alarcón, V., Sumar, J., Riera, G.S., Foote, W.C., 1990. Comparison of three
Sumar, J., 1988. Removal of the ovaries or ablation of the corpus luteum
methods of pregnancy diagnosis in alpacas and llamas. Theriogenol-
and its effect on the maintenance of gestation in the alpaca and llama.
ogy 34, 1119–1127.
Acta Vet. Scand. Suppl. 83, 133–141.
Allen, W.R., 2005. The development and application of the modern repro-
Sumar, J., Leyva, V., 1979. Relación entre la ubicación del cuerpo lúteo y
ductive technologies to horse breeding. Reprod. Domest. Anim. 40,
la localización del embrión en la llama (Lama glama). Res. Proyec-
310–329.
tos Investigación, Univ. Nacional Mayor de San Marcos, Período
Aller, J.F., Rebuffi, G.E., Cancino, A.K., Alberio, R.H., 2002. Successful transfer
1975–1979, Lima, p. 124.
of vitrified llama (Lama glama) embryos. Anim. Reprod. Sci. 73 (1–2),
Taylor, S., Taylor, P.J., James, A.N., Godke, R.A., 2000. Successful commer-
121–127.
cial embryo transfer in the llama (Lama glama). Theriogenology 53,
Bianchi, C.P., Meikle, A., Sartore, I., González, F., Aba, M.A., 2007. Uter-
344.
ine estrogen receptor alpha and progesterone receptor during the
Tibary, A., Anouassi, A., 1997. Theriogenology in camelidae. Anatomy,
follicular and luteal phase in llamas. Anim. Reprod. Sci. 99, 117–126.
physiology, pathology and artificial breeding. In: Actes Éditions, Insti-
Bourke, D.A., Adam, C.L., Kyle, C.E., Young, P., McEvoy, T.G., 1992. Super-
tut Agronomique et Vetérinaire Hassan II (Maroc) (Eds.), Reproductive
ovulation and embryo transfer in the llama. In: Allen, W.R., Higgins,
Physiology in Female Camelidae. Abu Dhabi Printing and Publish-
A.J., Maythew, I.G., Snow, D., Wade, J.F. (Eds.), Proceedings of the First
ing Company Institut Agronomique et Vetérinaire Hassan II (Maroc),
International Camel Conference on R&W. Publications Newmarket
United Arab Emirates, p. 438.
Ltd, pp. 183–185.
Tibary, A., Anouassi, A., Khatir, H., 2005. Update on reproductive
Del Campo, M.R., Del Campo, C.H., Ginther, O.J., 1996. Vascular provisions
biotechnologies in small ruminants and camelids. Theriogenology 64,
for a local utero-ovarian cross-over pathway in New World Camelids.
618–638.
Theriogenology 46, 983–991.
Trasorras, V.L., Chaves, M.G., Miragaya, M.H., Pinto, M., Rutter, B., Flores,
Fernández-Baca, S., Hansel, W., Novoa, C., 1970. Embryonic mortality in
M., Agüero, A., 2009. Effect of eCG superstimulation and buserelin on
the alpaca. Biol. Reprod. 3, 243–251.
cumulus-oocyte complexes recovery and maturation in llamas (Lama
Fernández-Baca, S., Hansel, W., Saatman, R., Sumar, J., Novoa, C., 1979. Dif-
glama). Reprod. Domest. Anim. 44 (3), 359–364.
ferential luteolytic effects of right and left uterine horns in the alpaca.
Biol. Reprod. 20, 586–595.
Animal Reproduction Science 121 (2010) 286–293

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Differential expression of estrogen receptor alpha gene in the ampullae


and isthmus regions of the rabbit oviduct during early pregnancy
Noemi Baranda-Avila a , M. Enrique Cardoso-Rangel b , Marco Cerbón a,∗ ,
Ignacio Camacho-Arroyo a , C. Adriana Mendoza-Rodríguez a ,
Héctor Villaseñor-Gaona b , Santiago R. Anzaldúa-Arce b
a
Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, D.F., Mexico
b
Facultad de Medicina Veterinaria y Zootecnia, Departamento de Morfología, Universidad Nacional Autónoma de México, D.F., Mexico

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

Article history: We characterized the expression pattern of estrogen receptor ␣ (ER␣) gene in two regions
Received 26 March 2010 of the oviduct, ampullae and isthmus, of the rabbit (Oryctolagus cuniculus) during early
Received in revised form 11 June 2010
pregnancy (1–4 days) by RT-PCR and immunohistochemistry. In both regions of the oviduct,
Accepted 5 July 2010
ER␣ mRNA was increased (P < 0.01) in pregnant rabbits as compared with non-pregnant
Available online 13 July 2010
animals (NG). In the ampullae, the greatest amount of ER␣ mRNA was detected on the
third day of pregnancy (1.01 ± 0.02 relative amount), followed by a decrease on Day 4. In the
Keywords:
isthmus, an increase in ER␣ mRNA was observed during the first 4 days of pregnancy, with
Oviduct
Estrogen receptor ␣ the greatest amount on Day 3 (1.49 ± 0.3 relative amount). A marked ER␣ immunostaining
Rabbit was detected in epithelial, stromal and smooth muscle cells of the ampullae on the second
Early pregnancy and third days of pregnancy as compared with the NG group. In contrast, a significant
increase in ER␣ immunostaining was observed on the first and second days of pregnancy
with a reduction on the third and fourth days in the epithelial, stromal and smooth muscle
cells of the isthmus. The overall results suggest there is differential expression pattern for
the ER␣ gene in the ampullae and isthmus during early pregnancy of the rabbit, and that
these variations are related to specific functions of ER␣ in the reproductive tract during
early pregnancy.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction (AMP), isthmus (IST) and uterotubal junction (UTJ). The UTJ
provides a barrier to infectious microbes that might enter
The mammalian oviduct is a key tissue in rabbit repro- the oviduct from the uterus. It also regulates sperm entry to
duction because it is involved in the transport and stability the oviduct. The IST serves as a sperm storage organ and the
of sperm, fertilization and early embryo development AMP provides an environment conducive to fertilization
(Hamner, 1973; Pauerstein and Eddy, 1979; Roblero and and early embryonic development. The INF is responsible
Garavagno, 1979; Killian, 2004). The rabbit oviduct is a for picking up the oocyte cumulus complex following ovu-
coiled structure composed of four different regions, each lation and moving it into the AMP where the fertilization
with different functions: infundibulum (INF), ampullae occurs (Harper, 1994; Talbot et al., 1999; Suarez, 2008). The
oviduct has at least three types of cells: epithelial, stromal
and smooth muscle cells. Selective modifications in the his-
tomorphology of the rabbit oviduct during early pregnancy
∗ Corresponding author at: Facultad de Química, Universidad Nacional
have been reported (Anzaldúa et al., 2002).
Autónoma de México, Ciudad Universitaria, Coyoacán 04510, México, D.F.,
Mexico. Tel.: +52 55 5622 3820; fax: +52 55 5622 3820. Estrogens participate in the regulation of the struc-
E-mail address: mcerbon85@yahoo.com.mx (M. Cerbón). ture and function of the oviduct. Estrogen actions in target

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.07.003
N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293 287

tissues are mainly mediated by its interaction with the two tobarbital injection (90 mg/kg) after deep anesthesia with
intracellular estrogen receptor (ER) subtypes, denoted ER␣ ketamine (Rhóne, Mérieux, Qro. México) on Days 1–4 of
and ER␤ (Nilsson et al., 2001; Diel, 2002; Edwards, 2005). pregnancy. Pregnancy was confirmed by the observation
Both ERs are widely distributed throughout different tis- of zygotes and morula under a stereoscopic microscope
sues with a distinct gene expression pattern in a variety after washing the oviduct with PBS. The experiments were
of tissues. In the oviduct of rats, mice and rabbits, ER␣ is conducted following the Mexican Law for the Protection of
the predominant subtype (Saunders et al., 1997; Mowa and Animals (México), and NOM-062-200-1999 (Aluja, 2002).
Iwanaga, 2000a,b; Couse et al., 1997; Sar and Welsch, 1999;
Jefferson et al., 2000). Evaluation of the gene expression 2.2. Total RNA extraction and RT-PCR
and localization of ER␣ is fundamental for understanding
estrogen functions in reproductive tissues. In the uterus Total RNA was isolated from two regions of the oviduct,
and vagina, ER␣ gene expression during pregnancy has AMP and IST, with the single-step method based on guani-
been well documented (Iwai et al., 1991; Hegele-Hartung dine isothiocyanate/phenol/chloroform extraction using
et al., 1992; Spencer and Bazer, 1995; Murata et al., 2003; TRIzol reagent (Gibco-BRL, Inc.) (Chomczynski and Sacchi,
Asakawa et al., 2008). However, the expression and regula- 1987). RNA concentration was determined by absorbance
tion of this receptor gene in the oviduct of mammals have at 260 nm and its integrity was verified by electrophore-
been scarcely studied. sis on 1.1% denaturing agarose gels in the presence of
ER␣ gene regulation is under tissue-specific hormonal 2.2 M formaldehyde. Total RNA was reverse transcribed to
control, e.g., in most mammals, cyclical estradiol (E2 ) con- synthesize single strand cDNA. Briefly, 4 ␮g of total RNA
centrations regulate ER␣ gene expression in the uterus were incubated at 37 ◦ C for 1 h with 800 units of M-ML
and oviduct (Nephew et al., 2000; Slayden and Brenner, reverse transcriptase (Gibco-BRL, Inc.) in 20 ␮L reaction
2004; Bläuer et al., 2005). Previous studies have reported volume containing 50 mM Tris–HCl (pH 8.3), 75 mM KCl,
the nuclear expression of ER␣ gene in epithelial, stromal 3 mM MgCl2 , 10 mM DTT, 0.5 mM for each dNP and 2.5 ␮M
and smooth muscle cells of the rat oviduct (Pelletier et al., oligo d-T (Gibco-BRL, Inc.) (Camacho-Arroyo et al., 1996).
2000). During pregnancy, ER␣ immunostaining increases Ten microliters of reverse transcriptase (RT) reaction were
in all cell types of all the rat oviduct regions (Okada subjected to PCR to simultaneously amplify ER␣ and glyc-
et al., 2003) and the administration of E2 increases the eraldehyde 3-phosphate dehydrogenase (GAPDH) gene
immunoreactivity of ER␣ in the AMP and the IST of preg- which was used as an internal control. The sequences of the
nant rats (Orihuela et al., 2009). specific primers for ER␣ amplification segment (from +151
In the rabbit, interactions of E2 with different segments to +304 relative to the start site of transcription of exon 1)
of the oviduct during early pregnancy have been analyzed (Kaluz et al., 1997) were 5 -[GCGTATGAGTTCAACGCCG]-3
by hormonal binding assays. The amount of nuclear binding in the sense primer and 5 -[GGCTCGGAGACACGCTGTT]-3
in the IST reaches its maximum at 48 h post-coitum (p.c.), in the antisense. The PCR reaction (50 ␮L) included 10 ␮L
whereas in the AMP it is detected at 72 h p.c. (El-Banna of previously synthesized cDNA, 20 mM Tris–HCl (pH 8.3),
and Sacher, 1977; Puri and Roy, 1981). ER␣ gene expres- 50 mM KCl, 1 mM MgCl2 , 0.2 mM of each dNTP, 0.5 ␮M of
sion has been described in both the AMP and the IST of the each primer, and 2.5 units of Taq DNA polymerase. Negative
oviduct and in the uterus of pseudopregnant rabbits. ER␣ controls without RNA and with non-retrotranscribed RNA
gene expression was increased in the AMP and myosalpinx were included in all experiments. After an initial denatu-
of the IST with a decrease in the uterus (Karbowski et al., ration step at 95 ◦ C for 5 min, PCR reaction was performed
1992). However, the expression and anatomical distribu- for 25 cycles. The cycle profile for ER␣ and GAPDH genes
tion of ER␣ gene expression during early pregnancy before amplification was: 95 ◦ C, 1 min; 60 ◦ C, 1 min; 72 ◦ C, 1 min.
implantation (1–4 days) in the oviduct of the rabbit has not A final extension cycle was performed at 72 ◦ C for 5 min.
been described. To contribute to the understanding of the The number of cycles, was within the exponential phase of
molecular mechanisms involved in E2 actions during the the amplification process. All PCR products were studied
pre-implantation period in the oviduct, we investigated the and analyzed together throughout the experiments. PCR
expression pattern of the ER␣ gene in the AMP and the IST products (25 ␮L) were separated on 2% agarose gel and
of the oviduct of rabbits during the first 4 days of pregnancy. stained with ethidium bromide. The image was captured
under a UV transilluminator with Type 665 negative films
2. Materials and methods (Polaroid Co., Cambridge, MA, USA). The intensity of ER␣
or GAPDH bands was quantified by densitometry using a
2.1. Animals Scanjet 3C apparatus (Hewlett Packard). Amount of ER␣
gene expression was normalized to that of GAPDH.
Fifteen adult virgin female New Zealand white rabbits
(Oryctolagus cuniculus) (Harlan, Mexico) (3.5–4.5 kg) were 2.3. Immunohistochemistry
used throughout the study. Animals were housed in indi-
vidual cages with controlled temperature (23 ± 2◦ C) under The endometrium, AMP and the IST of the rabbit were
a 12:12 h light:dark cycle with food and water available ad immediately dissected and fixed in 15% picric acid and
libitum. A group of animals (n = 12) was mated twice on the 4% paraformaldehyde in phosphate buffer saline (PBS) for
same day with two experienced bucks. The day of mating 8 h. Tissues were dehydrated through a series of increasing
was designated as Day 0. Non-pregnant animals (NG) (n = 3) ethanol concentrations and finally cleared with xylene. Tis-
were used as controls. Animals were euthanized by i.v. pen- sues were then embedded in paraplast (OxfordLabware, St.
288 N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293

Louis, MO). Serial tissue sections were cut at 5 ␮M thickness


and mounted on poly-l-lysine (Sigma) coated slides. Slides
were deparaffinized, rehydrated through graded concen-
trations of alcohol to distilled water, transferred to sodium
citrate buffer (pH 6.0), and heated two times for 10 min
in a microwave oven set at 800 W. Slides were cooled
between microwave irradiation for 5 min. After this pro-
cedure, slides were washed twice with 10 mM PBS, pH
7.4. Endogenous peroxidase was blocked by incubation
with 3% hydrogen peroxide in PBS for 30 min. After rins-
ing in PBS, slides were incubated in 0.5% Triton X-100,
and non-specific immunoglobulin binding was blocked by
incubating sections in 5% BSA for 30 min. Sections were
incubated overnight with monoclonal primary antibody
against ER␣ at a 1:100 dilution (6F11; Novocastra Labo-
ratories Ltd, Newcastle upon Tyne, UK) in a humidified
chamber at 4 ◦ C. For negative control, primary antibody was
omitted. After washing with PBS, sections were incubated
for 2 h at room temperature with biotin-conjugated goat
anti-rabbit IgG antibody (Santa Cruz Biotechnology, Inc.,
Santa Cruz, CA) at a 1:100 dilution and then washed with
PBS to remove unbound secondary antibody, and incubated
for 1 h at room temperature in peroxidase-conjugated
avidin–biotin reagent (Santa Cruz Biotechnology, Inc.,
Santa Cruz, CA). The slides were washed with PBS, devel-
oped with DAB substrate, and counterstained with Mayer’s
hematoxylin.
The number of immunopositive nuclei and staining
intensity in the epithelium and stroma were determined
using Metamorph Imaging System (Westchester, PA, USA).
All the nuclei from the epithelium, stoma and smooth mus- Fig. 1. Densitometric analysis of ER␣ mRNA gene expression in the
cle were analyzed in each section. Three sections by animal ampullae and isthmus of the rabbit oviduct during early pregnancy. RT-
(n = 3 animals per day of pregnancy) were analyzed. The PCR was performed using total RNA isolated from (A) ampullae and (B)
staining intensity of the cell nuclei was assigned the follow- isthmus of non-pregnant rabbits (NG) and the first 4 days of the preg-
nancy (D-1 to D-4). ER␣ gene expression was corrected for that of GAPDH.
ing scores: 0, absent; 1, weak; 2, moderate and 3, intense.
Results are expressed as the mean ± SEM (n = 3). In (A) *P < 0.01 compared
The histologic score (HSCORE) was calculated as follows: to all other groups, **P < 0.05 compared to all other groups, ***P < 0.05
HSCORE = ˙Pi (i + 1), where i = 1, 2 or 3, and Pi is the per- compared with D-1 and D-2. In (B) *P < 0.01 compared to all other groups.
centage of each intensity, from 0 to 100% (Lessey et al.,
1988). of pregnancy with a reduction in ER␣ gene expression on
the fourth day (0.75 ± 0.04 relative amount) (Fig. 1A). In the
2.4. Statistical analysis IST, relative amount of ER␣ mRNA was increased (P < 0.01)
during the first 4 days of pregnancy as compared with NG
RT-PCR and immunohistochemistry data were analyzed rabbits. The greatest amounts of ER␣ gene expression were
using one-way analysis of variance (ANOVA) followed by detected on the third day (1.49 ± 0.3 relative expression)
Student–Newman–Keuls or Tukey’s multiple comparison and were maintained until the fourth day of pregnancy
test, respectively. The Prism 2.01 program (Graph Pad, San (1.2 ± 0.4 relative expression) (Fig. 1B).
Diego, CA) was used for calculating probability values. ER␣ protein was detected by immunohistochem-
istry in the rabbit oviduct of pregnant rabbits. ER␣
3. Results immunopositive cells were observed in secretory and
ciliated cells in both regions (AMP and IST) of rab-
In all RT-PCR amplifications, single bands of 154 and bit oviduct. ER␣ immunopositive nuclei in epithelial,
454 bp, corresponding to the expected size fragments of stromal and smooth muscle cells of rabbit AMP were
ER␣ and GAPDH genes, respectively, were obtained. No undectectable in NG rabbits and on the first day of preg-
bands were observed in the negative controls. The identity nancy (Figs. 2A and B, 3A and B, and 6A, C, and E).
of each PCR product was confirmed by nucleotide sequenc- An increase in ER␣ immunoreactivity was observed in
ing (data not shown). all cell types in AMP on the second day of pregnancy
Relative amount of ER␣ mRNA in the AMP was detected (Figs. 2C, 3C, and 6A, C, and E), the highest ER␣ HSCORE
by RT-PCR. An increase (P < 0.01) of ER␣ gene expression in all cell types studied was observed on the third day of
was observed during the first 4 days of pregnancy as com- pregnancy (Figs. 2D, 3D, and 6A, C, and E), with a decrease
pared with NG rabbits. The greatest content of ER␣ mRNA on the fourth day (Figs. 2E, 3E, and 6A, C, and E). In the
(1.01 ± 0.02 relative amount) was observed on the third day rabbit uterus, used as a positive control tissue, ER␣ was
N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293 289

Fig. 2. Nuclear localization of ER␣ in epithelial and stromal cells of the rab- Fig. 3. Immunohistochemical localization of ER␣ protein in smooth mus-
bit ampullae during early pregnancy. Ampullae from an NG animal (A), on cle cells of the rabbit ampullae during early pregnancy. Ampullae from an
Days 1 (B), 2 (C), 3 (D) and 4 of pregnancy (E). Positive (F) and negative (G) NG animal (A), on Days 1 (B), 2 (C), 3 (D) and 4 of pregnancy (E). Positive (F)
controls from rabbit endometrium. Arrows indicate ER␣ immunopositive and negative (G) controls from rabbit endometrium. Arrows indicate ER␣
cells. Epithelium (E) and stroma (S). Bar = 100 ␮m. immunopositive cells. Smooth muscle (M) and stroma (S). Bar = 100 ␮m.

4. Discussion
detected in the nuclei of epithelia, stroma and muscle layers
(Figs. 2F and 3F). No staining was observed in the negative Although ER␣ gene expression in female reproductive
controls of rabbit uterus, where the primary antibody was tissues has been extensively studied, the expression and
omitted (Figs. 2G and 3G). localization of this receptor in the rabbit oviduct dur-
In the IST, a significant increase in the ER␣ ing early pregnancy remained unclear. The present study
HSCORE of epithelial, stromal and smooth muscle demonstrates that ER␣ gene is expressed in the rabbit
cells was observed on the first 2 days of pregnancy oviduct during early pregnancy in a tissue-dependent man-
(Figs. 4B, C, 5B, C and 6B, D, and F) as compared with ner. Differences in the time of induction of ER␣ gene
NG rabbits (Figs. 4A, 5A and 6B, D, and F), but a decrease expression were observed between AMP and IST. In addi-
(P < 0.05) in the ER␣ HSCORE was observed on the third tion, this receptor was found in ciliated and secretory cells
and fourth days of pregnancy in all cell types of the IST of the rabbit oviduct.
(Figs. 4D, E, 5D, E, and 6B, D, and F). Positive and negative In the present study, a progressive increase in relative
immunostaining controls were similar to those found in amounts of ER␣ mRNA during the 4 days of pregnancy was
AMP (Figs. 4F, G, and 5F and G). observed in the AMP and IST of the rabbit oviduct (Fig. 1).
290 N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293

Fig. 4. Nuclear localization of ER␣ in epithelial and stromal cells of the Fig. 5. Immunohistochemical localization of ER␣ protein in smooth mus-
rabbit isthmus during early pregnancy. Isthmus from an NG animal (A), on cle cells of the rabbit isthmus during early pregnancy. Isthmus from an NG
Days 1 (B), 2 (C), 3 (D) and 4 of pregnancy (E). Positive (F) and negative (G) animal (A), on Days 1 (B), 2 (C), 3 (D) and 4 of pregnancy (E). Positive (F)
controls from rabbit endometrium. Arrows indicate ER␣ immunopositive and negative (G) controls from rabbit endometrium. Arrows indicate ER␣
cells. Epithelium (E) and stroma (S). Bar = 100 ␮m. immunopositive cells. Smooth muscle (M) and stroma (S). Bar = 100 ␮m.

In many mammals, E2 concentrations regulate ER␣ gene


expression in the uterus and oviduct. Both up and down Results of the present study are consistent with previous
regulation of ER␣ mRNA by E2 have been demonstrated. Up studies that showed the presence of ER␣ in secretory and
regulation of ER␣ mRNA in the rat oviduct and uterus was ciliated cells of endosalpinx and myosalpinx of pseudo-
observed from day 1 to day 3 of pregnancy when E2 concen- pregnant rabbits (Karbowski et al., 1992). E2 administration
trations were greater (Wu et al., 1996; Ing and Ott, 1999; induces ciliogenesis in the oviduct of ovariectomized rats
Okada et al., 2003). In line with this observation, the great- and rabbits and accelerates ovum transport in pregnant
est relative amount of ER␣ mRNA occurred in both regions animals (Banik and Pincus, 1964; Fuentealba et al., 1988).
(AMP and IST) on the third day of pregnancy when a signif- In addition, in pregnant rats, ovariectomy or hypophy-
icant increase of E2 concentrations is observed (Browning sectomy, and treatment with the aromatase inhibitor
et al., 1980; Hegele-Hartung et al., 1992). 4-hydroxyandrostenedione, caused delayed ovum trans-
The cilia of the oviduct are believed to play a critical port due to a reduction of estrogen production (Wu et al.,
role in ovum transport to the uterus in pregnant animals 1971; Forcelledo and Croxatto, 1986, 1988). Our group pre-
(Halbert et al., 1989). Importantly, in the present study viously reported the presence of PR immunopositive cells
ER␣ immunopositive cells in secretory and ciliated cells in secretory and ciliated cells in the AMP and IST of the
in the AMP and IST of the rabbit oviduct were detected. rabbit oviduct (Anzaldúa et al., 2007). Thus, both E2 and P4
N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293 291

Fig. 6. ER␣ protein in the ampullae and isthmus of the rabbit during early pregnancy. Immunohistochemistry was performed in the ampullae and isthmus
of NG and pregnant rabbits during the first 4 days of pregnancy. The histologic score (HSCORE), was calculated as described in Material and Methods. Results
are expressed as the mean ± SEM. In (A–F) *P < 0.05 compared with all other groups without *. In (C and E) **P < 0.005 compared with all other groups.

may participate in ovum transport by regulating oviductal later, beginning on the second day and reaching a max-
ciliogenesis in rabbits. imum on the third day of pregnancy (Fig. 6A, C and E).
Interestingly, some differences were observed between These differences in the time of induction of nuclear ER␣
the AMP and IST in ER␣ gene expression during early preg- gene expression between AMP and IST could reflect a spe-
nancy. In the IST, the highest relative amounts of ER␣ cific contribution of these regions to signals provided by
protein (HSCORE) were observed on the first and second E2 to regulate egg transport. Morphological changes in the
days of pregnancy (Fig. 6B, D and F). In contrast, in the oviduct during early pregnancy, such as height and pro-
AMP, ER␣ gene expression was detected several hours portion of PAS+ and PAS− cells in different regions of the
292 N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293

rabbit oviduct, could be related to the changes observed in References


ER␣ expression in the AMP and IST (Anzaldúa et al., 2002).
In the rabbit reproductive tract, the content and Aluja, A.S., 2002. Animales de Laboratorio y la Norma Oficial Mexicana
(NOM-062-ZOO-1999). Gac. Méd. Méx. 138, 295–298.
immunolocalization of ER␣ have been studied using Asakawa, M.G., Goldschmidt, M.H., Une, Y., Nomura, Y., 2008. The
hormonal binding assays and during induced pseudopreg- immunohistochemical evaluation of estrogen receptor-␣ and
nancy by human chorionic gonadotropin (hCG). According progesterone receptors of normal. Hyperplastic, and neoplastic
endometrium in 88 pet rabbits. Vet. Pathol. 45, 217–225.
to results of the present study, in the AMP and IST, E2 bind- Anzaldúa, S.R., Camacho-Arroyo, I., Cerbón, M.A., 2002. Histomorpho-
ing reaches a peak at 72 and 48 h p.c., respectively (Puri and logical changes in the oviduct epithelium of the rabbit during early
Roy, 1981). In addition, and in agreement with our results, pregnancy. Anat. Histol. Embryol. 31, 308–312.
Anzaldúa, S.R., Camacho-Arroyo, I., Reyna-Neyra, A., Pérez-Martínez, M.,
Karbowski’s group (1992) reported there was a significant
Cerbón, M., 2007. Regional differences in expression of progesterone
increase in ER␣ immunoreactive score 24 h after hCG treat- receptor in oviduct and uterus of rabbit during early pregnancy. Comp.
ment in the IST and 48 h post-hCG treatment in the AMP Biochem. Physiol. A Mol. Integr. Physiol. 147, 685–690.
Banik, U.K., Pincus, G., 1964. Estrogens and transport of ova in the rat. Proc.
(Karbowski et al., 1992).
Soc. Exp. Biol. Med. 116, 1032–1034.
In contrast with results from the present study, Bläuer, M., Heinonen, P.K., Martikainen, P.M., Tomás, E., Ylikomi, T., 2005.
Karbowski et al. (1992) observed an increase of ER␣ A novel organotypic culture model for normal human endometrium:
immunoreactive score in the IST 72 h post-hCG injection regulation of epithelial cell proliferation by estradiol and medroxypro-
gesterone acetate. Hum. Reprod. 20, 864–871.
when we observed a significant decrease in ER␣ HSCORE Browning, J.Y., Keyes, P.L., Wolf, R.C., 1980. Comparison of serum pro-
(Fig. 6B, D and F). Although E2 and P4 plasma concentra- gesterone, 20 alpha-dihydroprogesterone, and estradiol-17 beta in
tions are similar during pregnancy and pseudopregnancy pregnant and pseudopregnant rabbits: evidence for postimplantation
recognition of pregnancy. Biol. Reprod. 23, 1014–1019.
(Challis et al., 1973; Browning et al., 1980; Hegele-Hartung Camacho-Arroyo, I., Pasapera, A.M., Cerbón, M.A., 1996. Regulation of pro-
et al., 1992), a different ER␣ gene expression pattern was gesterone receptor gene expression by sex steroid hormones in the
observed in the IST and AMP between the pseudopreg- hypothalamus and cerebral cortex of the rabbit. Neurosci. Lett. 214,
25–28.
nant and pregnant animals. This suggests that ER␣ gene Casan, E.M., Raga, F., Bonilla-Musoles, F., Polan, M.L., 2000. Human
expression not only depends on P4 and E2 concentra- oviductal gonadotropin-releasing hormone: possible implications in
tions, but also on other local or paracrine factors such fertilization, early embryonic development, and implantation. J. Clin.
Endocrinol. Metab. 85, 1377–1381.
as gonadotropin-releasing hormone (GnRH) or androgens
Challis, J.R., Davies, J., Ryan, K.J., 1973. The concentrations of progesterone,
and their receptors (GnRHR and AR, respectively). Recently, estrone and estradiol-17 beta in the plasma of pregnant rabbits.
a direct inhibitory effect of the ER␣/AR heterodimer on Endocrinology 93, 971–976.
Chomczynski, P., Sacchi, N., 1987. Single-step method of RNA isolation
both ER␣ and AR transactivational properties has been
by acid guanidinium thyocyanate–phenol–chlorophorm extraction.
reported (Panet-Raymond et al., 2000). Also, expression of Anal. Biochem. 162, 156–159.
GnRHR gene in the oviduct during pregnancy was reported Couse, J.F., Lindzey, J., Grandien, K., Gustafsson, J.Å., Korach, K.S., 1997.
(Casan et al., 2000; Sengupta and Sridaran, 2008). Oviduc- Tissue distribution and quantitative analysis of estrogen receptor-
␣ (ER␣) and estrogen receptor-␤ (ER␤) messenger ribonucleic acid
tal GnRH is predicted to be involved in autocrine/paracrine in the wild-type and ER␣-knockout mouse. Endocrinology 138,
regulation of functions like fertilization, early embryonic 4613–4621.
development, and implantation in human (Casan et al., Diel, P., 2002. Tissue-specific estrogenic response and molecular mecha-
nisms. Toxicol. Lett. 127, 217–224.
2000). The role of these oviductal receptors during early Edwards, D.P., 2005. Regulation of signal transduction pathways by estro-
pregnancy requires further investigations. gen and progesterone. Annu. Rev. Physiol. 67, 335–376.
In the present study, the greatest relative amount of El-Banna, A.A., Sacher, B., 1977. A study on steroid hormone receptors in
the rabbit oviduct and uterus during first few days after coitus and
ER␣ protein was detected in the AMP (on day 3) (Fig. 6A, during egg transport. Biol. Reprod. 17, 1–8.
C and E) and the IST (on days 1 and 2) (Fig. 6B, D and Forcelledo, M.L., Croxatto, H.B., 1986. Ovum transport in cyclic
F) of rabbit oviduct during early pregnancy, when the rats rendered hypo-oestrogenic by treatment with 4-hydroxy-4-
androstene-3,17-dione. J. Reprod. Fertil. 76, 325–330.
least PR gene expression was previously observed in these
Forcelledo, M.L., Croxatto, H.B., 1988. Effects of 4-
oviduct regions (Anzaldúa et al., 2007). This differential hydroxyandrostenedione and exogenous testosterone on blood
gene expression patterns between ER␣ and PR may be concentrations of oestradiol and oviductal embryo transport in the
rat. J. Endocrinol. 118, 93–100.
related to the hormonal mileu observed in rabbits during
Fuentealba, B., Nieto, M., Croxatto, H.B., 1988. Progesterone abbreviates
early pregnancy (Challis et al., 1973). the nuclear retention of estrogen receptor in the rat oviduct and coun-
The differences observed in the distribution and expres- teracts estrogen action on egg transport. Biol. Reprod. 38, 63–69.
sion of the ER␣ gene in the AMP and IST during early Halbert, S.A., Becker, D.R., Szal, S.E., 1989. Ovum transport in the rat
oviductal ampulla in the absence of muscle contractility. Biol. Reprod.
pregnancy and before implantation in the rabbit, suggest 40, 1131–1136.
that there are different regulatory mechanisms for the Hamner, C.E., 1973. Oviductal fluid—composition and physiology. In:
expression of steroid receptor genes in distinct regions of Greep, R., Astwood, E.B. (Eds.), Handbook of Physiology, vol. 2.
Female Reproductive Physiology. Waverly, Baltimore, MD, pp. 141–
the rabbit reproductive tract that could reflect specific con- 151.
tributions of these regions to the signals provided by E2 to Harper, M.J.K., 1994. In: Knobil, E., Niel, J.D. (Eds.), The Physiology of Repro-
regulate egg transport. duction, 2nd ed. Raven Press, New York.
Hegele-Hartung, C., Chwalisz, K., Beier, H.M., 1992. Distribution of
estrogen and progesterone receptors in the uterus: an immunohis-
tochemical study in the immature and adult pseudopregnant rabbit.
Histochemistry 97, 39–50.
Acknowledgements Ing, N.H., Ott, T.L., 1999. Estradiol up-regulates estrogen receptor-alpha
messenger ribonucleic acid in sheep endometrium by increasing its
stability. Biol. Reprod. 60, 134–139.
This research was supported in part by grant from Iwai, T., Fujii, S., Nanbu, Y., Nonogaki, H., Konishi, I., Mori, T., Okamura,
PAPIIT-IN207207 and CONACyT (AC-89544 and 80338). H., 1991. Effect of human chorionic gonadotropin on the expression
N. Baranda-Avila et al. / Animal Reproduction Science 121 (2010) 286–293 293

of progesterone receptors and estrogen receptors in rabbit ovarian Panet-Raymond, V., Gottlieb, B., Beitel, L.K., Pinsky, L., Trifiro, M.A.,
granulosa cells and the uterus. Endocrinology 129, 1840–1848. 2000. Interactions between androgen and estrogen receptors and the
Jefferson, W.N., Couse, J.F., Banks, E.P., Korach, K.S., Newbold, R.R., 2000. effects on their transactivational properties. Mol. Cell. Endocrinol. 167,
Expression of estrogen receptor ␤ is developmentally regulated in 139–150.
reproductive tissues of male and female mice. Biol. Reprod. 62, Pauerstein, C.J., Eddy, C.A., 1979. The role of the oviduct in reproduction;
310–317. our knowledge and our ignorance. J. Reprod. Fertil. 55, 223–229.
Kaluz, S., Kaluzova, M., Flint, A.P.F., 1997. Sequencing variability in the A/B Pelletier, G., Labrie, C., Labrie, F., 2000. Localization of oestrogen receptor
region of the estrogen receptor. Anim. Biotechnol. 8, 221–226. ␣, oestrogen receptor ␤ and androgen receptors in the rat reproduc-
Karbowski, B., Vollmer, E., Schneider, P.G., 1992. Steroid receptors in the tive organs. J. Endocrinol. 165, 359–370.
fallopian tube—morphological and functional investigation in rabbits. Puri, R.K., Roy, S.K., 1981. Estradiol binding in different parts of the rabbit
Prog. Histochem. Cytochem. 26, 140–144. oviduct during egg transport. Endokrinologie 78, 12–20.
Killian, G.J., 2004. Evidence for the role of oviduct secretions in sperm Roblero, L.S., Garavagno, A.C., 1979. Effect of oestradiol-17␤ and pro-
function, fertilization and embryo development. Anim. Reprod. Sci. gesterone on oviductal transport and early development of mouse
82–83, 141–153. embryos. J. Reprod. Fertil. 57, 91–95.
Lessey, B.A., Killam, A.P., Metzger, D.A., Haney, A.F., Greene, G.L., McCarty Sar, M., Welsch, F., 1999. Differential expression of estrogen receptor-␣
Jr., K.S., 1988. Immunohistochemical analysis of human uterine estro- and estrogen receptor-␤ in the rat ovary. Endocrinology 140, 963–971.
gen and progesterone receptors throughout the menstrual cycle. J. Saunders, P.T., Maguire, S.M., Gaughan, J., Millar, M.R., 1997. Expression
Clin. Endocrinol. Metab. 67, 334–340. of oestrogen receptor beta (ER␤) in multiple rat tissues visualised by
Mowa, C.N., Iwanaga, T., 2000a. Differential distribution of oestrogen immunohistochemistry. J. Endocrinol. 154, R13–R16.
receptor-␣ and -␤ mRNAs in the female reproductive organ of rats Sengupta, A., Sridaran, R., 2008. Expression and localization of
as revealed by in situ hybridization. J. Endocrinol. 165, 59–66. gonadotropin-releasing hormone receptor in the rat oviduct during
Mowa, C.N., Iwanaga, T., 2000b. Developmental changes of the oestrogen pregnancy. J. Histochem. Cytochem. 56, 25–31.
receptor-␣ and -␤ mRNAs in the female reproductive organ of the Slayden, O.D., Brenner, R.M., 2004. Hormonal regulation and localization
rat—an analysis by in situ hybridization. J. Endocrinol. 167, 363–369. of estrogen, progestin and androgen receptors in the endometrium
Murata, T., Narita, K., Honda, K., Matsukawa, S., Higuchi, T., 2003. Differen- of nonhuman primates: effects of progesterone receptor antagonists.
tial regulation of estrogen receptor alpha and beta mRNAs in the rat Arch. Histol. Cytol. 67, 393–409.
uterus during pregnancy and labor: possible involvement of estrogen Spencer, T.E., Bazer, F.W., 1995. Temporal and spatial alterations in uterine
receptors in oxytocin receptor regulation. Endocr. J. 50, 579–587. estrogen receptor and progesterone receptor gene expression during
Nephew, K.P., Long, X., Osborne, E., Burke, K.E., Ahluwalia, A., Bigbsby, R.M., the estrous cycle and early pregnancy in the ewe. Biol. Reprod. 53,
2000. Effect of estradiol on estrogen receptor expression in rat uterine 1527–1543.
cell types. Biol. Reprod. 62, 168–177. Suarez, S.S., 2008. Regulation of sperm storage and movement in the mam-
Nilsson, S., Mäkelä, S., Treuter, E., Tujague, M., Thomsen, J., Andersson, G., malian oviduct. Int. J. Dev. Biol. 52, 455–462.
Enmark, E., Pettersson, K., Warner, M., Gustafsson, J.A., 2001. Mecha- Talbot, P., Geiske, C., Knoll, M., 1999. Oocyte pick-up by the mammalian
nisms of estrogen action. Physiol. Rev. 81, 1535–1565. oviduct. Mol. Biol. Cell 10, 5–8.
Okada, A., Ohta, Y., Inoue, S., Hiroi, H., Muramatsu, M., Iguchi, T., 2003. Wu, J.T., Dickmann, Z., Johnson, D.C., 1971. Effects of ovariectomy or
Expression of estrogen, progesterone and androgen receptors in the hypophysectomy on day one of pregnancy on development and trans-
oviduct of developing, cycling and pre-implantation rats. J. Mol. port of fertilized rat eggs. J. Endocrinol. 49, 507–513.
Endocrinol. 30, 301–315. Wu, W.X., Owiny, J., Zhang, Q., Ma, X.H., Nathanielsz, P.W., 1996. Regu-
Orihuela, A.P., Zuñiga, M.L., Rios, M., Parada-Bustamante, A., Sierralta, lation of the estrogen receptor and its messenger ribonucleic acid in
D.W., Velásquez, A.L., Croxatto, B.H., 2009. Mating changes the sub- the ovariectomized sheep myometrium and endometrium: the role
cellular distribution and the functionality of estrogen receptors in the of estradiol and progesterone. Biol. Reprod. 55, 762–768.
rat oviduct. Reprod. Biol. Endocrinol. 7, 139.
Animal Reproduction Science 121 (2010) 294–300

Contents lists available at ScienceDirect

Animal Reproduction Science


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

Body reserves and ovarian performance in primiparous lactating rabbit


does submitted to early weaning as a strategy to decrease
energy deficit
O.G. Sakr a,b , R.M. García-García c , M. Arias-Álvarez c , P. Millán c ,
P.L. Lorenzo c , P.G. Rebollar a,∗
a
Department of Animal Production, Polytechnic University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
b
Department of Animal Production, Faculty of Agriculture, Cairo University,12613 Giza, Egypt
c
Department of Physiology (Animal Physiology), Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain

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

Article history: The aim of this work was to study the effect on body composition, serum metabolic
Received 26 April 2010 parameters and ovarian status of early weaning at 25 Days post-partum (dpp) as a
Received in revised form 22 June 2010
strategy to decrease energy deficit of primiparous lactating rabbit does prior to insem-
Accepted 28 June 2010
ination at 32 dpp following an extensive rhythm. A total of 34 primiparous lactating
Available online 6 August 2010
rabbit does were used and distributed in three groups: 10 lactating does euthanized
at 25 dpp (group L25), 13 does weaned at 25 dpp and euthanized at 32 dpp (group
Keywords:
NL32), and 11 non weaned lactating does euthanized at 32 dpp (group L32). No sig-
Body composition
Weaning nificant differences were observed in live body weight, ovary weight, serum NEFA
Oocyte and total protein concentration among groups. Although NL32 does had a low feed
Follicle intake (122 ± 23.5 g/Day; P < 0.001), their estimated lipids (16.9 ± 1.09%, P < 0.008), pro-
PRL-R tein (19.7 ± 0.07%, P < 0.0001), and energy (1147 ± 42.7 MJ/kg, P < 0.006) body contents were
Rabbit higher and their serum glucose concentrations (158 ± 24.5 mg/dl, P < 0.04) were lower com-
pared to L25 does (11.9 ± 1.3%, 18.5 ± 0.08%, 942 ± 51.3 MJ/kg and 212 ± 27.9 mg/dl) and L32
does (13.4 ± 1.03%, 18.5 ± 0.1%, 993 ± 40.4 MJ/kg and 259 ± 29.5 mg/dl), respectively. In the
ovarian surface of L25 does a lower number of follicles ≥1 mm was observed compared to
NL32 and L32 groups (12.7 ± 1.5 vs. 18.0 ± 1.45 and 17.6 ±1.67; P < 0.05). Follicular pop-
ulation in the histological ovarian sections and immunolocalization of prolactin receptor
were similar between groups. In group L25, both nuclear maturation of oocytes in terms of
Metaphase II rate (67.0 vs. 79.7 and 78.3%; P < 0.05) and cytoplasmic maturation measured
by percentage of cortical granules (CG) totally or partially migrated in oocytes were signifi-
cantly lower than in groups NL32 and L32 (16.0 vs. 38.3 and 60.0%; P < 0.05). Consequently,
a higher rate of oocytes with non-migrated CGs was found in group L25 than in groups
NL32 and L32 (76.0 vs. 46.8 and 33.3%; P < 0.05). In conclusion, even though early weaning
at 25 dpp seemed to improve body energy stores of primiparous does, this fact was not well
reflected on the ovarian status at 32 dpp, which was similar regardless of weaning time and
it could be performed later.
© 2010 Elsevier B.V. All rights reserved.

∗ Corresponding author. Tel.: +34 914 524 868; fax: +34 915 499 763.
E-mail address: pilar.grebollar@upm.es (P.G. Rebollar).

0378-4320/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.anireprosci.2010.06.008
O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300 295

1. Introduction 2. Materials and methods

In commercial rabbit farms, litter weaning usually 2.1. Animals


occurs from Days 28 to 35 of age. However, primi-
parous does cannot completely meet the high nutritional A total of 34 primiparous lactating Californian × New
demands of lactation and usually show an energy deficit Zealand White rabbit does were used for this study. Ani-
(Cervera et al., 1993; Fortun-Lamothe, 1998; Parigi Bini mals were housed at the experimental farm in the Animal
and Xiccato, 1998; Xiccato et al., 2004) which reduces Production Department, Polytechnic University of Madrid
their body fat depots and is partly responsible for their (Spain), in individual metal cages (50 cm × 70 cm × 32 cm)
low reproductive performance (Castellini et al., 2006). It with a closeable nest box, under a constant photoperiod of
has been reported that an extensive reproductive rhythm 16 h light per Day, a temperature of 18–22 ◦ C, and a rela-
can be adopted in these animals to improve their nutri- tive humidity of 60–75% maintained by a forced ventilation
tional status, fertility, length of reproductive activity and system. All females were fed ad libitum a commercial diet
welfare (Castellini et al., 2003; Dal Bosco et al., 2003; (NANTA S.A., Tres Cantos, Madrid, Spain) containing 16%
Feugier and Fortun-Lamothe, 2006). In a recent work, it crude protein, 15.7% crude fiber and 2.5% fat; digestible
has been shown that insemination of primiparous does at energy of the diet was 2700 kcal/kg. Animals had free access
32 Days post-partum (dpp), only 4 Days after weaning, to water. All experimental procedures used in this research
improved estimated body composition and energy con- were approved by the Animal Ethics Committee of the
tent, serum leptin and protein concentrations, health of Polytechnic University of Madrid and were in compliance
follicular populations in the ovary, oocyte quality and, as with the Spanish guidelines for care and use of animals in
a consequence, conception rate and prolificacy compared research (BOE, 2005).
to lactating does inseminated at 11 dpp (Arias-Álvarez et
al., 2007). 2.2. Experimental design
Also, in primiparous does, early weaning between 21
and 25 dpp seems to be a feasible practice to improve their Primiparous lactating does were equalized at eight
poor body condition before artificial insemination time. pups one day after parturition, keeping this number con-
Thus, Feugier and Fortun-Lamothe (2006) observed that stant throughout lactation and dead pups being replaced
early weaning at 23 dpp significantly reduced dissectible with others of a similar weight and age provided from
fat mobilisation of does compared to weaning at 35 dpp. nurse does. Thirty-four lactating does were randomly
Nevertheless, in both cases, reproductive performance is distributed in three experimental groups: 10 lactating
not affected when females are inseminated at 25 dpp. In does were euthanized on Day 25 pp (group L25), 13
turn, Xiccato et al. (2005) have shown that early-weaned non-lactating does were weaned on Day 25 pp and
does (at 21 or 25 dpp) display a substantial decrease in euthanized on Day 32 pp (group NL32), and 11 lactat-
food intake in the post-weaning period, reducing the daily ing does were euthanized on Day 32 pp (group L32).
energy surplus and delaying the complete restoration of Live body weight (LBW), estimated body composition
their reserves. In these previous studies, corpora lutea and and serum metabolic parameters (glucose, non-esterified
foetuses were counted, and, when does were inseminated fatty acids (NEFA) and total protein) were measured
at 25 dpp, the number of corpora lutea and embryonic or before euthanasia in all groups. Also, solid feed intake
fetal mortality were not affected by the age of weaning. of does from parturition until 25 dpp, as well as solid
To our knowledge, no studies have been done to assess feed intake of does and their litters between 25 and
the consequences of early weaning on ovarian follicle and 32 dpp were determined. The solid feed intake of
oocyte features in extensively-reared primiparous rabbit young rabbits from parturition to Day 25 was consid-
does. In this sense, it is well known that a good ovarian ered null. Nevertheless, feed intake of L32 does from
status leads to reproductive success. Day 25 pp to 32 pp was calculated considering that
Additionally, in previous works, high prolactin (PRL) kits, apart from milk, fed a mean of 30 g of solid feed
concentrations were observed during lactation in rabbit from Day 25 to 32 of age according to Gidenne and
does (Ubilla et al., 2000). This hormone is the main respon- Lebas (2006). Euthanasia was performed with 30 mg/kg
sible for the negative effect of lactation on reproductive of intravenous pentobarbital sodium (Dolethal, Veto-
function (McNeilly et al., 1982). Kermabon et al. (1994) quinol, Alcobendas, Spain). To further determine ovarian
indicated that receptive behavior is correlated with signif- features, both ovaries were recovered by laparotomy
icantly more follicles higher than 1 mm of diameter and after euthanasia. One of them was preserved for his-
an increase in the concentration of PRL receptors on the tological and immunohistochemical studies (follicular
rabbit ovary. Early weaning could decrease the possible categorization and immunolocalization of prolactin recep-
inhibitory action of this hormone in follicular development tor), and the other one was used for in vitro maturation
and oocyte quality as described in several species (rabbit: assessment.
Yoshimura et al., 1989; bovine: Torner et al., 2001).
On the basis of the findings stated above, this study was
conducted to assess the influence of early weaning at 25 2.3. Analytical methods
dpp on estimated body composition, serum metabolic and
ovarian parameters of primiparous lactating does when Unless otherwise stated, all chemicals were purchased
they are reared in an extensive reproductive rhythm. from Sigma Chemical Company (St. Louis, MO, USA).
296 O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300

2.3.1. Body composition atmosphere of 5% CO2 in air with maximum humidity. The
Body composition was estimated by means of bioelec- maturation medium was prepared according to Lorenzo
trical impedance analysis (BIA) using a four-terminal body et al. (1997). After the maturation period, COCs were
composition analyser (Model Quantum II, RJL Systems, processed for the confocal study as previously reported
Detroit, MI, USA) which reports reactance and resistance by Arias-Álvarez et al. (2007). Nuclear maturation was
between two sets of two electrodes. In our case, we used measured in terms of metaphase II rate. Cortical granule
standard 21-gauge needles (Terumo Europe N.V. 3001 Leu- distribution was classified as follows: migrated (CGs were
ven, Belgium) as electrodes inserted into the skin of the adjacent to the plasma membrane, showing they were
animal. One pair of needles was held 1 cm apart in the cytoplasmically matured) or partially migrated (CGs were
neck and the other one in the rump. These components spread throughout the cortical area showing they had ini-
have been correlated to BIA measurements through regres- tiated their cytoplasmic maturation), non-migrated (CGs
sion equations built on multiple measurements of rabbit were distributed throughout the cytoplasm, as they did not
does in different physiological status (pregnant, lactating, show cytoplasmic maturation) and abnormally migrated
non pregnant, non lactating) and validated according to (anomalous distribution of CGs compatible with poor qual-
Nicodemus et al. (2009) and Pereda (2010). ity or degenerated oocytes).
To determine PRL receptor on ovarian surface a proto-
2.3.2. Serum parameters col was used according to that described by Arias-Álvarez
Blood samples were collected from the margin ear vein et al. (2009). The primary antibody against prolactin
into non-heparinized tubes at 9:00 h a.m. to avoid circa- receptor (1:75, Affinity Bioreagents, Golden, CO, USA)
dian variations and considering that over 60% of the solid used in this study specifically recognized long and short
feed is consumed by the rabbit in the dark period (Gidenne forms of rabbit prolactin receptor (PRL-R). After detec-
and Lebas, 2006). Serum was obtained after centrifugation tion with diaminobenzidine, sections were counterstained
at 1200 × g for 10 min at 4 ◦ C and stored at −32 ◦ C until with hematoxylin and photographed under the light micro-
analysed. scope (Olympus BX40). A subjective image analysis was
Serum non-esterified fatty acids (NEFA) determinations performed to estimate the immunoreactivity of PRL-R in
were performed in duplicate samples using a two-reaction different cell types by an observer unaware of which group
enzymatic-based colorimetric assay from Wako Chemi- of animals was examining. At least five fields of each ovary
cals GmbH Specialty Chemicals (Neuss, Germany). The sample were examined at 400× magnification to evalu-
method is linear over the range from 0.01 to 4.0 mEq/l. ate PRL-R immunostaining. Each cell compartment was
Serum glucose was determined in duplicate samples using given an immunohistochemical score, based on the stain-
the GOD-PAP method from RANDOX Laboratories Ltd. ing intensity (0–3: 0 = negative, 1 = weak, 2 = intermediate,
(Crumlin, United Kingdom), according to Tietz (1995). This 3 = strong) according to Picazo et al. (2004) and García-
method is linear up to a glucose concentration of 400 mg/dl. Palencia et al. (2007).
Serum total protein was determined by the Biuret method
from RANDOX Laboratories Ltd. (Crumlin, United King- 2.4. Statistical analysis
dom), according to Tietz (1995). This method is linear up
to 13 g/dl. Trial data were analysed using the Statistical Analysis
Systems (SAS/STAT, 1999–2001). Mean differences of LBW,
2.3.3. Ovarian parameters estimated body composition, serum metabolic parame-
Ovary weight, number of follicles ≥1 mm on the ovar- ters, feed intake, ovary weight, number of haemorrhagic
ian surface and hemorrhagic follicles were first recorded. follicles, follicles more than 1 mm diameter, follicular cat-
Then, one ovary from each animal was placed into a 4%- egorization and staining intensity of PRL-R were analyzed
buffered neutral paraformaldehyde solution (pH 7.2–7.4). with a GLM procedure by a one-way analysis of variance
All samples were gradually dehydrated with increasing (three differentiated groups L25, NL32 and L32, represent-
concentrations of ethyl alcohol, embedded in paraffin, ing two post-partum times: 25 and 32 dpp). Means were
prepared by sectioning at 5 ␮m, and stained with hema- compared using a protected Student’s t-test. In order to
toxylin/eosin. In order to study follicle population, a pair compare nuclear maturation and CG migration index of in
of histological sections of each half ovary was examined vitro-matured oocytes among experimental groups a Chi-
at light microscope (Olympus BX40, Hamburg, Germany). square test (proc CATMOD) was carried out. All the results
Rabbit ovarian follicles were categorized into three spe- are expressed as mean ± S.E.M., and statistical significance
cific development stages related to the number of layers of was accepted for P < 0.05.
granulosa cells according to Rebollar et al. (2008), namely,
primary, preantral and antral follicles. 3. Results
The remaining ovaries were placed in PBS at 37 ◦ C.
Cumulus-oocyte complexes (COCs) were obtained from 3.1. Live body weight, estimated body composition, feed
ovarian follicles more than 1 mm diameter by aspiration intake and serum metabolic parameters
with a 25-gauge needle under a stereoscopic microscope.
A total of 317 cumulus-oocyte complexes (group L25, Early weaning did not affect LBW of does, which was
n = 116; group NL32, n = 126; group L32, n = 75) with com- in average 3.82 ± 1.15 kg. In group NL32, the estimated
pact cumulus cells were washed and placed in 500 ␮l of water content was lower (P < 0.001), and estimated lipids
maturation medium and cultured for 16 h at 38 ◦ C under an (P < 0.01), protein (P < 0.001), and energy (P < 0.01) body
O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300 297

Table 1
Live body weight, estimated body composition, feed intake and serum parameters in primiparous lactating does on Day 25 pp (L25), on Day 32
pp (L32) and in early-weaned, non-lactating does on Day 32 pp (NL32).

Day 25 pp Day 32 pp

L25 NL32 L32

No. of does 10 13 11
LBW (kg) 3.86 ± 0.14 3.91 ± 0.94 3.68 ± 1.13
Water (%) 65.9 ± 1.06A 61.5 ± 0.89B 66.9 ± 1.07A
Ash (%) 3.09 ± 0.05 3.04 ± 0.04 3.14 ± 0.04
Lipids (%) 13.4 ± 1.03b 16.9± 0.09a 11.9 ± 1.31b
Proteins (%) 18.5 ± 0.10B 19.7 ± 0.07A 18.5 ± 0.08B
Energy (MJ/kg) 993 ± 40.4b 1147 ± 2.7a 942 ± 51.3b

Feed intake (g/Day)


Parturition to 25 Days pp 345 ± 19.6 357 ± 17.2 359 ± 17.9
25 to 32 Days pp* – 122 ± 23.5B 402 ± 26.7A

Serum concentrations
NEFA (mmol/L) 0.26 ± 0.04 0.23± 0.03 0.19 ± 0.04
Glucose (mg/dl) 212 ± 27.9ab 158 ± 24.5b 259 ± 29.5a
Total protein (g/dl) 7.60 ± 1.26 7.40 ± 1.10 6.50 ± 1.33

pp: post-partum. LBW: live body weight. NEFA: non-esterified fatty acids.
A, B: P < 0.001; a, b: P < 0.01 in the same row.
*
Calculated considering litter size of each doe (8 kits) and a mean dry feed intake of kits of 30 g/d.

contents were higher than in the two other experimental haemorrhagic follicles between experimental groups. Nev-
groups (Table 1). Feed intake from parturition to 25 dpp ertheless, L25 does showed a lower number of follicles
was similar in all groups, but just as we expected, group ≥1 mm in the ovarian surface per ovary than groups NL32
L32 had a higher feed intake from 25 to 32 dpp compared and L32 (P < 0.05).
to group NL32 (P < 0.0001). Follicular population observed in the histological sec-
Serum NEFA and total protein concentrations did not tions was not affected by early weaning, and similar means
show significant differences between experimental groups, of primary, preantral and antral follicles in the ovarian sec-
but serum glucose concentration was lower in group NL32 tions were found (11.4 ± 1.71, 13.7 ± 1.84, and 13.3 ± 1.12
than in group L32 (P < 0.04), being intermediate in group follicles per ovary, respectively; P > 0.05).
L25. PRL-R cytoplasmic staining was observed in all ovarian
compartments studied (granulose cells of all categorized
3.2. Features of ovarian follicular population follicles and also in corpora lutea (CL) and stromal cells),
but not in theca cells, as shown in Fig. 1. No staining was
As shown in Table 2, no significant differences detected in the negative controls. No statistical differences
were found in average ovary weight in the number of were found in the staining intensity of PRL-R in different

Table 2
Ovarian status in primiparous lactating does and in vitro oocyte maturation from primiparous lactating does on Day 25 pp (L25), on Day 32 pp (L32), and
in early-weaned, non-lactating does on Day 32 pp (NL32). Nuclear and cytoplasmic maturation were measured in terms of metaphase II rate (MII) and
cortical granule (CG) migration.

Day 25 pp Day 32 pp

L25 NL32 L32

No. of does 10 13 11
Ovary Weight (mg)
Right 347 ± 37.4 329 ± 34.1 295 ± 39.4
Left 331 ± 32.9 323 ± 30.1 286 ± 34.7

Follicles (n)
>1mm 12.7 ± 1.58b 18.0 ± 1.45a 17.6 ± 1.67a
Haemorrhagic 1.30 ± 0.85 1.00 ± 0.77 0.90 ± 0.89

Mean of follicular population/ovary (n)


Antral 9.88 ± 2.17 15.7 ± 2.05 13.3 ± 1.70
Preantral 12.9 ± 3.6 19.3 ± 3.36 10.4 ± 2.80
Primary 10.8 ± 3.32 13.4 ± 3.13 10.4 ± 2.61

In vitro oocyte maturation


MII rate (%) 67.0b 79.7a 78.3ab
Migrated or partially migrated CGs (%) 16.0b 38.3a 60.0a
Non-migrated CGs (%) 76.0a 46.8b 33.3b
Abnormally migrated CGs (%) 8.0 14.9 3.3

pp: post-partum. a,b P < 0.05 in the same row.


298 O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300

Fig. 1. Immunohistochemical localization of PRL-R in ovaries of does early weaned at 25 days post-partum (group L25 and group NL 32) or non weaned
(group L32) in the different sized follicles and corpus luteum. (A) Negative control (20×); (B) primary follicles (PF) (40×) with positive (brown) granulose
cells and oocytes, surrounded by positive stromal cells (SC); (C) antral follicle (AF) showing strong immunoreactivity to PRL-R in granulosa cells, and negative
theca cells (20×); (D) Corpus luteum (CL) (20×) showing positive immunostaining in all luteal cells. Given that the pattern and intensity of localizations
did not change between ovaries of the three experimental groups, only representative images of follicles and corpus luteum are shown (A–D). Scale bar:
100 ␮m.

size follicles and CL in group L25 compared to groups L32 2009). The greatest interest in early weaning techniques
and NL32. lies in the possibility of reducing doe body energy deficit.
In this sense, estimated body composition and energy
3.3. In vitro oocyte maturation changes observed in this work have confirmed previous
results found on primiparous lactating rabbit does (Xiccato
As shown in Table 2, Metaphase II rate observed in et al., 2005; Fortun-Lamothe and Prunier, 1999; Feugier
oocytes from non-lactating does was significantly higher and Fortun-Lamothe, 2006; Castellini et al., 2006). A high
(P < 0.05) than in oocytes from lactating does at 25 dpp energy supply is required during the final lactation period,
(L25), but similar to lactating rabbits at 32 dpp. Percent- since milk dry matter and lipid concentrations increase
age of cytoplasmic maturation was significantly different from Day 20 onwards when production begins falling
between groups (P < 0.05). In group NL32, a significantly (Lebas, 1971, 1972; Pascual et al., 1999). Using ultrasounds,
higher rate of oocytes with migrated or partially migrated Pascual et al. (2002) observed a decrease of around 0.2 mm
CGs was found with respect to L25 (P < 0.05), but similar to in peri-renal fat thickness during the first three weeks of
L32 (P = 0.06). Oocytes with no migrated CGs were higher lactation, rising to a loss of 0.9 mm in the last week. Simi-
for group L25 than for the two other groups (P < 0.05). No lar results were obtained by Fortun-Lamothe et al. (2002),
significant differences were found among groups regarding who found a decrease of 5 MJ in the total energy content
abnormal migration of CGs. of primiparous does during lactation, or Fortun-Lamothe
and Prunier (1999), who reported a weight loss (−97 g
4. Discussion per week) after the first two weeks post-partum. In our
study, focused on the last week of lactation, primiparous
Early weaning is one factor to settle an extensive repro-
lactating does at 32 dpp maintained similar body energy
ductive rhythm. From the ovarian and energy points of
reserves than lactating does at 25 dpp. Nevertheless, wean-
view, the present study will clarify some aspects of this
ing at 25 dpp increased energy reserves; non-lactating
critical reproductive period.
animals had 154 and 205 MJ/kg more body energy con-
tent than lactating does at 25 and 32 dpp, respectively.
4.1. Live body weight, estimated body composition, feed Although feed intake was lower in weaned does, decreased
intake and serum metabolic parameters milk production for seven days was enough to improve
slightly their energy reserves. Consequently, body lipids
LBW of does on Day 25 pp was similar to that observed and protein content were also improved in non-lactating
in primiparous does in previous studies (Rebollar et al., does.
O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300 299

Previous studies observed that weaning age of litters Glucose, insulin and leptin are the most likely signals
strongly influence the evolution of the body condition of informing the hypothalamus of the animal metabolic sta-
females during their reproductive cycles. Early weaning (23 tus (Diskin et al., 2003). In our work, follicular growth
Days of age, Feugier and Fortun-Lamothe, 2006; 21 Days took place irrespective of glucose concentrations availabil-
of age, Xiccato et al., 2005) reduces fat loss and energy ity because lactating and non-lactating does at 32 dpp had
deficit with respect to weaning performed later (33 Days different serum glucose concentrations.
of age, Feugier and Fortun-Lamothe, 2006, or 25 Days of
age, Xiccato et al., 2005, respectively). This improvement 4.3. In vitro oocyte maturation
on body condition is observed even though the feed intake
of does is voluntarily reduced, according to our results. Oocyte maturation implies both nuclear and cytoplas-
This is due to the chemostatic mechanism of appetite mic maturation including CG migration (Edwards, 1965;
regulation according to which, after litter weaning, does Yanagimachi, 1994). A recent report in rabbit (Arias-
quickly decrease their feed intake by 35–45% of the lacta- Álvarez et al., 2009) clearly shows that conditions related
tion level in just a week (Xiccato et al., 2005). The lower to early lactation and negative energy balance adversely
serum glucose concentrations observed in non-lactating affect oocyte quality and endocrine parameters, effects
does on Day 32 pp compared to lactating ones could be which are especially pronounced after the first parturi-
explained by the substantial decrease in feed intake dur- tion. In the current study, on Day 32 pp oocytes matured
ing the dry period after weaning, reducing the daily energy in vitro from ovaries of lactating or non-lactating females
intake as Xiccato et al. (2004) demonstrated. When serum showed higher nuclear and cytoplasmic maturation rates
glucose concentrations fall, NEFA must be released into than oocytes of lactating does on Day 25 pp. Addition-
the blood stream (Brecchia et al., 2006). In fact, around ally, the percentage of cytoplasmic immature oocytes, with
parturition, elevated concentrations of NEFA have been non-migrated CGs, was also higher in does on Day 25 of
previously described due to mobilization of body reserves lactation. These results confirm the previously described
and reduced feed intake at the end of pregnancy (Arias- progressive improvement of reproductive performance
Álvarez et al., 2009). Nevertheless, neither lactating does during lactation (Ubilla and Rebollar, 1995; Feugier and
at 25 dpp nor weaned ones at 32 dpp showed differences Fortun-Lamothe, 2006).
in NEFA concentrations. In that sense, Arias-Álvarez et al. Prolactin effects are related to the presence of its
(2009) did not observe differences among does on Day 11 receptor. Prolactin concentration fluctuates according
pp and on Day 32 pp. As regards both serum parameters, to lactation stage (Durand and Djiane, 1977), and it acts
low concentrations of glucose are not enough to evoke directly on rabbit follicles, modulating follicular health
any meaningful effect on the evolution of NEFA concentra- and development (Ben-Jonathan et al., 1996). In this study,
tions in lactating rabbit does. In fact, Xiccato et al. (2005) PRL-R appeared in different ovarian localizations, includ-
observed a high individual variability in NEFA concentra- ing granulosa cells and antral follicle oocytes, matching
tions of does under different reproductive rhythms and previous results (Kermabon et al., 1994; García-García et
weaning ages. al., 2007). However, we found no significant differences
in the cellular localization of prolactin receptor among
4.2. Features of ovarian follicular population groups. Matching our results, previous studies showed
no differences between lactating and non-lactating rab-
Ovarian features were similar in lactating and non- bit does (Kermabon et al., 1994; Arias-Álvarez et al.,
lactating does at 32 dpp. Only the post-partum times 2009). Some works have shown that localization of PRL-R
considered (25 or 32 dpp) had a significant effect in exhibits remarkable changes throughout the oestrus cycle
mean preovulatory follicles, since they were lower on in ewe (Picazo et al., 2004), which is closely regulated by
Day 25 pp than on Day 32 pp. The reduction in the hormones and/or local factors. In our work, rabbits are
number of preovulatory follicles observed in group L25 probably having the physiological follicular wave on Days
is consistent to their lower estimated body composition, 23–30, pp, as is suggested by Ubilla and Rebollar (1995), so
which corresponds to the negative energy balance and time of oestrus cycle and endocrine environment, including
the endocrine changes expected in these animals, changes distribution of PRL receptor could be similar for all groups.
affecting in turn follicular growth (Diskin et al., 2003).
The distribution or evolution of follicular waves in the 5. Conclusions
rabbit ovary is not well described. These animals con-
tinuously produce mature follicles which become atretic Early weaning at 25 dpp seems to improve body energy
if ovulation is not stimulated by coitus (Kranzfelder et stores of primiparous does, but this fact is not well reflected
al., 1984). Ubilla and Rebollar (1995) suggested the pres- on their ovarian status, which is better on Day 32 pp than
ence of a follicular growth wave between Days 23 and on Day 25 pp, regardless of weaning time and it could be
30 of the post-partum period in non-pregnant lactating performed later.
rabbits. This wave coincided with an increase in plasma
oestradiol levels found on Days 23–30 after parturition Acknowledgements
and with a rise of sexual receptivity. Current results
show the existence of this physiological follicular wave This research was supported by the CICYT project
irrespective of whether rabbit does were lactating or (AGL08-022283) and the Community of Madrid
not. (S2009/AGR1704).
300 O.G. Sakr et al. / Animal Reproduction Science 121 (2010) 294–300

References Lebas, F., 1971. Composition chimique du lait de lapine, évolution au cours
de la traite et en fonction du stade de lactation. Annales de Zootechnie
Arias-Álvarez, M., López-Béjar, M., Rebollar, P.G., García-García, R.M., 20, 185–191.
Lorenzo, P.L., 2007. Nuclear and cytoplasmic patterns of rabbit IVM Lebas, F., 1972. Effet de la simultanéité de la lactation et de la gestation sur
oocytes. Investigación Técnica Económica Agraria 28, 42–44. les performances latières chez la lapine. Ann. Zootech. 21, 129–191.
Arias-Álvarez, M., García-García, R.M., Rebollar, P.G., Revuelta, L., Millán, Lorenzo, P.L., Illera, J.C., Silván, G., Munro, C.J., Illera, M.J., Illera, M.,
P., Lorenzo, P.L., 2009. Influence of metabolic status on oocyte qual- 1997. Steroid-level response to insulin-like growth factor-1 in oocytes
ity and follicular characteristics at different post-partum periods in matured in vitro. J. Reprod. Immunol. 35, 11–29.
primiparous rabbit does. Theriogenology 72, 612–623. McNeilly, A.S., Glasier, A., Jonassen, J., Howie, P.W., 1982. Evidence for
Ben-Jonathan, N., Mershon, J.L., Allen, D.L., Steinmetz, R.W., 1996. Extra- direct inhibition of ovarian function by prolactin. J. Reprod. Fertil. 65,
pituitary prolactin: distribution, regulation, functions, clinical aspects. 559–569.
Endocr. Rev. 17, 639–669. Nicodemus, N., Pereda, N., Romero, C., Rebollar, P.G., 2009. Évaluation de
BOE. Boletín Oficial del Estado, 2005. Real Decreto 1201/2005. Sobre pro- la technique d’impedance bioélectrique (IBE) pour estimer la compo-
tección de los animales utilizados para experimentación y otros fines sition corporelle de lapines reproductrices. In: Bolet, G. (Ed.), 13 èmes
científicos. BOE 252, 34367–34391. Journées de la Recherche Cunicole. 17–18 novembre 2009, LeMans,
Brecchia, G., Bonanno, A., Galeati, G., Federici, G., Maranesi, M., Gobbetti, France, pp. 109–112.
A., Zerani, M., Boiti, C., 2006. Hormonal and metabolic adaptation to Pascual, J.J., Cervera, C., Blas, E., Fernández-Carmona, J., 1999. Effect of
fasting: effects on the hipotalamic–pituitary–ovarian axis and repro- high fat diets on the performance, milk yield and milk composition of
ductive performance of rabbit does. Domest. Anim. Endocrin. 31, multiparous rabbit does. Anim. Sci. 68, 151–162.
105–122. Pascual, J.J., Motta, W., Cervera, C., Quevedo, F., Blas, E., Fernández-
Castellini, C., Dal Bosco, A., Mugnai, C., 2003. Comparison of different Carmona, J., 2002. Effect of dietary energy source on the performance
reproductive protocols for rabbit doe: effect of litter size and re- and peri-renal fat thickness evolution of primiparous rabbit does.
mating interval. Livest. Prod. Sci. 83, 131–139. Anim. Sci. 75, 267–279.
Castellini, C., Dal Bosco, A., Cardinali, C., 2006. Long term effect of post- Parigi Bini, R., Xiccato, G., 1998. Energy metabolism and requirements.
weaning rhythm on the body fat and performance of rabbit doe. In: De Blas, J.C., Wiseman, J. (Eds.), The Nutrition of the Rabbit. CABI
Reprod. Nutr. Dev. 46, 195–204. Publishing, Wallingford, UK, pp. 103–131.
Cervera, C.J., Fernández-Carmona, J., Viudes, P., Blas, E., 1993. Effect of Pereda, N., 2010. Evaluación de la técnica del Análisis de Impedancia
remating interval and diet on the performance of female rabbits and Bioeléctrica para predecir la composición corporal: aplicación en
their litters. Anim. Prod. 56, 399–405. conejas sometidas a diferentes sistemas de alimentación durante la
Dal Bosco, A., Castellini, C., Cardinali, R., 2003. Evaluation of body condi- recría. In: Tesis Doctoral. Universidad Politécnica de Madrid, Madrid,
tion score in pregnant rabbit does by ultrasound scanner. In: Atti XV Spain, pp. 194.
Congresso. ASPA, Parma, pp. 480–482. Picazo, R.A., García Ruiz, J.P., Santiago Moreno, J., González de Bulnes,
Diskin, M.G., Mackey, D.R., Roche, J.F., Sreenan, J.M., 2003. Effects of nutri- A., Muñoz, J., Silván, G., Lorenzo, P.L., Illera, J.C., 2004. Cellular local-
tion and metabolic status on circulating hormones and ovarian follicle ization and changes in expression of prolactin receptor isoforms in
development in cattle. Anim. Reprod. Sci. 78, 345–370. sheep ovary throughout the estrous cycle. Reproduction 128, 545–
Durand, P., Djiane, J., 1977. Lactogenic activity in the serum of rabbits 553.
during pregnancy and lactation. Endocrinology 75, 33–42. Rebollar, P.G., Bonanno, A., Di Grigoli, A., Tornambè, G., Lorenzo, P.L., 2008.
Edwards, R.G., 1965. Maturation in vitro of mouse, sheep, cow, pig, rhesus Endocrine and ovarian response after a 2-Day controlled suckling
monkey and human ovarian oocytes. Nature 208, 349–351. and eCG treatment in lactating rabbit does. Anim. Reprod. Sci. 104,
Feugier, A., Fortun-Lamothe, L., 2006. Extensive reproductive rhythm and 316–328.
early weaning improve body condition and fertility of rabbit does. Rebollar, P.G., Pérez-Cabal, M.A., Pereda, M.A., Lorenzo, P.L., Arias-Álvarez,
Anim. Res. 55, 459–470. M., García-Rebollar, P., 2009. Effects of parity order and reproductive
Fortun-Lamothe, L., 1998. Effects of pre-mating energy intake on repro- management on the efficiency of rabbit productive systems. Livest.
ductive performance of rabbit does. Anim. Sci. 66, 263–269. Sci. 123, 107–115.
Fortun-Lamothe, L., Prunier, A., 1999. Effects of lactation, energetic deficit SAS/STAT, 1999–2001. SAS 7. In: STAT User’s Guide (Release 8.2). SAS Inst.
and remating interval or reproductive performance of primiparous Inc., Cary, NC.
rabbit does. Anim. Rep. Sci. 55, 289–298. Tietz, N.W., 1995. Clinical Guide to Laboratory Tests, 3rd Ed. W.B. Saunders
Fortun-Lamothe, L., Lamboley-Gaüzère, B., Bannelier, C., 2002. Prediction Co., Philadelphia, pp. 518–519.
of body composition in rabbit female using total body electrical con- Torner, H., Kubelka, M., Heleil, B., Tomek, W., Aim, H., Kuzmina, T., Guiard,
ductivity (TOBEC). Livest. Prod. Sci. 78, 132–142. V., 2001. Dynamics of meiosis and protein kinase activities in bovine
García-García, R.M., Arias-Álvarez, M., García-Palencia, P., Revuelta, L., oocytes correlated to prolactin treatment and follicle size. Theri-
Sánchez-Maldonado, B., Rebollar, P.G., Lorenzo, P.L., 2007. Local- ogenology 55, 885–899.
ización del receptor de prolactina en el ovario de conejas en diferentes Ubilla, E., Rebollar, P.G., 1995. Influence of the postpartum day on
estados fisiológicos. II Congreso Ibérico de Cunicultura, IV Jornadas plasma oestradiol-17␤ levels, sexual behavior and fertility rate
Internacionales de Cunicultura y XXXII Symposium de Cunicultura. in artificially inseminated lactating rabbits. Anim. Reprod. Sci. 38,
5,6 junio. Ed. Asociación Española de Cunicultura, 41–46. Vila Real 337–344.
(Portugal). Ubilla, E., Rebollar, P.G., Pazo, D., Esquifino, A., Alvariño, J.M., 2000. Effects
García-Palencia, P., Sánchez, M.A., Nieto, A., Vilar, M.P., González, M., of doe litter separation on endocrinological and productivity variables
Veiga-López, A., González-Bulnes, A., Flores, J.M., 2007. Sex steroid in lactating rabbits. Livest. Sci. 67, 67–74.
receptor expression in the oviduct and uterus of sheep with estrus Xiccato, G., Trocino, A., Sartori, A., Queaque, P.I., 2004. Effect of parity order
synchronized with progestagen or prostaglandin analogues. Anim. and litter weaning age on the performance and body energy balance
Reprod. Sci. 97, 25–35. of rabbit does. Livest. Prod. Sci. 16, 239–251.
Gidenne, T., Lebas, F., 2006. Feeding behaviour in rabbits. In: Bels, V. (Ed.), Xiccato, G., Trocino, A., Boiti, C., Brecchia, G., 2005. Reproductive rhythm
Feeding in Domestic Vertebrates. From Structure to Behaviour. CABI and litter weaning age as they affect rabbit doe performance and body
Publishing, Wallingford, UK, pp. 179–209 (Chapter 11). energy balance. Anim. Sci. 81, 289–296.
Kermabon, A.Y., Belair, L., Theau-Clement, M., Salesse, R., Djiane, J., 1994. Yanagimachi, R., 1994. Mammalian fertilization. In: Knobil, E.,
Effects of anoestrus and bromocryptine treatment on the expression Neill, J.D. (Eds.), The Physiology of Reproduction. Raven Press,
of prolactin and LH receptors in the rabbit ovary during lactation. J. pp. 189–279.
Reprod. Fertil. 102, 131–138. Yoshimura, Y., Hosoi, Y., Irritan, A., Nakamura, Y., Atlas, S.J., Wal-
Kranzfelder, D., Korr, H., Mestwerdt, W., Maurer-Schultze, B., 1984. Follicle lach, E.E., 1989. Developmental potential of rabbit oocyte matured
growth in the ovary of the rabbit after ovulation-inducing application in vitro: the possible contribution of prolactin. Biol. Reprod. 41,
of human gonadotrophin. Cell Tissue Res. 238, 611–620. 26–33.

You might also like