You are on page 1of 8

Theriogenology 150 (2020) 280e287

Contents lists available at ScienceDirect

Theriogenology
journal homepage: www.theriojournal.com

Ovulation mechanism in South American Camelids: The active role of


b-NGF as the chemical signal eliciting ovulation in llamas and alpacas
Mauricio Silva a, b, Luis Paiva c, Marcelo H. Ratto c, *
a
Department of Veterinary Sciences and Public Health, Chile
b
Nucleus of Research on Agrifood Production, Universidad Cato lica de Temuco, Temuco, Chile
c
Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile

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

Article history: The ovulation-inducing effect of seminal plasma was first suggested in Bactrian camels over 30 years ago,
Received 21 January 2020 initiating a long search to identify the ‘ovulation-inducing factor’ (OIF) present in camelids semen.
Accepted 29 January 2020 During the last decade, primarily in llamas and alpacas, this molecule has been intensively studied
Available online 30 January 2020
characterizing its biological and chemical properties and ultimately identifying it as b-Nerve Growth
Factor (b-NGF). The high concentration of OIF/b-NGF in seminal plasma of llamas and alpacas, and the
Keywords:
striking effects of seminal fluid on ovarian function strongly support the notion of an endocrine mode of
Llama
action. Also, have challenged the dogma of mating induced ovulation in camelid species, questioning the
Alpaca
Ovulation
classical definition of reflex ovulators, which at the light of new evidence should be revised and updated.
Seminal plasma On the other hand, the presence of OIF/b-NGF and its ovulatory effect in camelids confirm the notion that
b-NGF seminal plasma is not only a transport and survival medium for sperm but also, a signaling agent tar-
geting female tissues after insemination, generating relevant physiological and reproductive conse-
quences. The presence of this molecule, conserved among induced as well as spontaneous ovulating
species, clearly suggests that the potential impacts of this reproductive feature extend beyond the
camelid species and may have broad implications in mammalian fertility. The aim of the present review
is to provide a brief summary of all research efforts undertaken to isolate and identify the ovulation
inducing factor present in the seminal plasma of camelids. Also to give an update of the current un-
derstanding of the mechanism of action of seminal b-NGF, at central and ovarian level; finally suggesting
possible brain targets for this molecule.
© 2020 Elsevier Inc. All rights reserved.

1. Introduction induced ovulators its occurrence is contingent upon copulatory


stimuli; hence, ovulation is not a regular cyclic event.
Mammalian species have been classified as either spontaneous There appears to be an evolutionary pattern or preference for
or induced ovulators based on the type of stimulus responsible for induced versus spontaneous ovulation among species. Some have
eliciting GnRH release from the mediobasal hypothalamus [1]. In suggested that induced ovulation is a primitive trait because of its
spontaneously ovulating species (e.g., human, sheep, cattle, horse, existence in primitive Orders like Rodentia, Lagomorpha and
pigs), release of GnRH from the hypothalamus is triggered when Insectivora [6e8] but the presence of induced ovulators in the
systemic estradiol concentrations exceed a certain threshold [2e5]. Order Artiodactyla (e.g., llamas, alpacas and camels) is not consis-
On the other hand, in reflex ovulators (e.g. rabbit, Bactrian camel, tent with this theory. While there appears to be no phylogenic
llama, alpaca, cat, ferret) neural signals from copulatory stimulation pattern to the ovulatory mechanism [9], it is believed that envi-
induce hypothalamic GnRH secretion, followed by the preovulatory ronmental factors or social cues influence the presence of induced
release of LH from the pituitary gland [1]. Similar to spontaneous ovulation. Most recent additions to the list of induced ovulators
ovulators, a surge in LH appears to be requisite for ovulation, but in belong to the Order Rodentia [9].
Classification of species as spontaneous or induced may be
overly strict since induced ovulators can occasionally ovulate
spontaneously and ovulation in those considered spontaneous
* Corresponding author.
E-mail address: marceloratto@uach.cl (M.H. Ratto).
ovulators can occasionally be induced or accelerated by copula.

https://doi.org/10.1016/j.theriogenology.2020.01.078
0093-691X/© 2020 Elsevier Inc. All rights reserved.
M. Silva et al. / Theriogenology 150 (2020) 280e287 281

Indeed, recent pieces of evidence clearly indicate that the control of found that concentration of b-NGF is high in the seminal plasma of
the ovulatory process could be more complex that a simple copu- another induced ovulator, such as the rabbit [37e39]. Further
latory stimuli to induce ovulation, in the case of reflex ovulators. It studies, demonstrated that b-NGF represents approximately 50% of
has been well documented that an ovulation-inducing factor (OIF) total protein in the llama seminal plasma (12.4 mg/mL [23]), being
present in the seminal plasma of llamas, alpacas and Bactrian 10 times more concentrated than in bovine semen [34]. Also b-NGF
camels is responsible to induce ovulation in these species. The has been described as one of the most abundant seminal proteins in
factor has been isolated and purified from the llama and alpaca alpacas [28,29], accounting for 15e30% of total protein content of
seminal plasma and it has been identified as Nerve Growth Factor seminal fluid [28,40].
(b-NGF) [10e13].
As many reviews have recently been published [10e13] 3. Nerve Growth Factor (b-NGF): a neurotrophin beyond the
describing the role of OIF or b-NGF in the mechanism of ovulation neuronal survival and protection
in camelids, we believe that is relevant to present a retrospective
summary of all OIF/b-NGF studies that describe its effects on Neurotrophins (NTs) are proteins that promote differentiation,
ovarian function (i.e. ovulation and corpus luteum formation). In growth and survival of many populations of peripheral (PNS) and
addition, a brief update about the current knowledge of the pres- central nervous system (CNS) neurons during development and
ence, chemical identity and properties of OIF is given. Finally, the adulthood [41e44]. The NGF is the best known and most studied
potential brain targets for llama b-NGF as main sites to trigger member of NT family including brain-derived NTs 3 4/5, 6 and 7
GnRH secretion are also suggested. [45e47]. NGF is a neurotrophin conserved in species of sponta-
neous and induced ovulation and although its function was first
2. Unveiling the chemical induction of ovulation in camelids: recognized at the level of PNS and CNS, several subsequent studies
from OIF to b-NGF have documented its effects in non-neuronal tissues including also
the ovary [48e50]. High and low affinity NGF receptors, TrkA and
The identification of b-NGF in the semen of most camelids p75 respectively, are found in the human ovary as early as the fetal
species and the clarification of its striking signaling action on the stage [48]. In rodents, NGF allows early follicular development and
female genital tract, ending in ovulation-induction, was achieved ovulation [49,50] and ovary differentiation during development
after a long systematic search. Table 1 is intended to give a chro- [51].
nological summary of the most relevant research undertaken in Old Interestingly, roles for NGF have also been described in the
and New World Camelids that contributed to this fascinating endocrine [52], immune [53,54], female [55,56] and male repro-
discovery. ductive systems [57]. NGF and TrkA are expressed by granulosa cells
Several Chinese studies documented the first evidence for the in pre-antral and antral follicles [58] and appear to promote ovarian
presence of an ovulation-inducing factor (OIF) in the Bactrian camel development through initial differentiation of newly formed folli-
semen 40 years ago (Table 1). Based on the results of these studies, cles into gonadotropin responsive structures capable of ovulating
it was concluded that: a) a protein molecule present in the seminal [59], as well as playing a pivotal role during ovulation [56]. In vitro
plasma was responsible for eliciting ovulation rather than the studies conducted in mice suggested that NGF contributes to the
mechanical stimulation of copulation [14,15]; b) ovulation induced ovulatory process inducing the secretion of estradiol, progesterone,
by the systemic or intrauterine administration of the factor is PGE2 and proliferation of theca cells [49]. It has also been docu-
preceded by a preovulatory LH surge [16]; and c) the factor is a mented that NGF and its receptors are expressed in granulosa cells
novel protein chemically different to GnRH, LH, hCG, eCG and of antral follicles and in luteal cells of pigs and goats CL [60,61].
PGF2a [17]. However, all these studies failed to isolate, purify and Furthermore, it has been suggested that the preovulatory LH surge
identify this factor from the seminal plasma of Bactrian Camel. up regulates NGF and TrkA in the ovary [62,63], while NGF is
A study conducted years later in South American Camelids capable of regulating its own receptor in spontaneous ovulators
(SACs) [18], alpacas and llamas, also documented the presence of an [64].
OIF in the semen of these species because systemic administration Lastly, the discovery of this neurotrophin at high concentration
of homologous seminal plasma induced ovulation in 28 out of 30 in the semen of different spontaneous and induced ovulators,
treated females. Indeed, similar to the Bactria Camel studies, mainly rabbit and camelids, contributed to confirm the notion that
ovulation was preceded by a preovulatory surge of LH [18]. Since seminal plasma is not only a transport and survival medium for
then, llamas and alpacas have emerged as a powerful animal model sperm but also, a signaling agent targeting female tissues after
to test the presence of OIF using either homologous or heterologous insemination [65,66], generating physiological consequences as
seminal plasma [18e21], seminal fractions [22,23] or purified OIF relevant as ovulation induction.
[24e26] after intramuscular, intravenous or intrauterine adminis-
tration. We want to point out that most of the information from the 4. Ovarian and endocrine effects of b-NGF
initial studies presented in Table 1 refer to this protein molecule as
ovulation-inducing factor, OIF. This factor has now been identified Systemic administration of purified NGF from llama seminal
as the neurotrophin b-NGF in llamas, alpacas and Bactrian Camel plasma induces a higher and more sustained LH increase than that
[27e30], Additionally, during the last 10 years this factor has been observed after exogenous GnRH administration in female llamas
biochemical and functionally characterized [18e20,22e25]. Based [18,24e26]. A significant increase in LH plasma concentration is
on these studies, OIF has been described as a potent ovulatory observed within 60 min after NGF treatment [18,25,67], similar to
molecule in alpacas and llamas which also has a relevant luteo- that observed in females mated to an intact male [67]. Interestingly,
trophic effect in llamas [18,25,26,31,32], although, a similar luteo- NGF LH-releasing effect was completely inhibited in llamas pre-
trophic effect has not observed in alpacas [28,33]. treated with GnRH antagonist Cetrorelix [25], which competitively
The primary source of OIF/b-NGF in llamas is the prostate gland binds to GnRH receptors in the adenohypophysis. Therefore, it was
[34], the only accessory sex gland beside the bulbourethral gland in suggested that NGF elicits a preovulatory LH surge by a direct or
male llamas. It is well known that b-NGF concentration in the indirect action on GnRH hypothalamic neurons [25].
seminal plasma of llamas is much higher than that of spontaneous- Additionally, using an ovariectomized llama model it was
ovulating species (stallion and boar [35]; bull [36]). Also, it was demonstrated that the magnitude of LH secretion induced by NGF
282 M. Silva et al. / Theriogenology 150 (2020) 280e287

Table 1
Summary of main achievements in the discovery of ovulation-inducing factor (OIF/b-NGF) in the seminal plasma of camelids.

Old World Camelids: Bactrian camel (Camelus bactrianus)

Reference [Nº] Achievement

Chen et al., 1983 [14] - First report suggesting that seminal factors rather than mechanical stimulation of female genital tract are responsible for ovulation in
Bactrian camels.
Chen et al., 1985 [15] - Confirm that after vaginal artificial insemination (AI) seminal factors effectively induce ovulation in Bactrian camel.
- Suggest that ovulation-inducing factors (OIF) are present in seminal plasma and they are not related to sperm cells.
Xu et al., 1985 [16] - Show that as a consequence of vaginal AI with camel semen, circulating concentration of LH significantly rises after 4 h.
- Suggest that LH rise and consequently ovulation induction are evoked through a humoral pathway.
Zhao et al., 1990 [108] - Show that i.m. administration of camel seminal plasma induces ovulation in female camels.
- Suggest that OIF present in camel seminal plasma are proteins in nature.
- Suggest that OIF must reach the female pituitary gland via the blood stream to induce the LH surge.
Liu et al., 1995 [109] - Show that protein fractions isolated from camel seminal plasma can stimulate gonadotrophins secretion from in vitro cultures of rat, rabbit or
guinea pig pituitary cells.
Li et al., 2002 [110] - Demonstrate that i.m. administration of camel seminal plasma induce the release of LH in female camels.
Zhao et al., 2001 [106] - Describe the isolation of 5 protein fractions from Bactrian camel seminal plasma by DEAE-cellulose chromatography, one of which had LH-
releasing properties.
- Demonstrate that some of these seminal fractions can stimulate gonadotrophins secretion from in vitro cultures of rat pituitary cells.
- Demonstrate that i.m. administration of some of these seminal fractions induce the release of LH using female camels as an in vivo bioassay.
Pan et al., 2001 [17] - Demonstrate that OIF present in the seminal plasma of Bactrian camels is a novel protein different from GnRH, LH, hCG, eCG or PGF2a.
Li & Zhao 2004 [107] - Describe the isolation of 6 protein fractions from Bactrian camel seminal plasma by DEAE-cellulose chromatography, two of which had
in vitro and in vivo LH-releasing properties.
- Fail to purify the OIF present in the protein seminal fractions.
Druart et al., 2013 [29] - Identify Beta-Nerve Growth Factor (b-NGF) as the main protein in Bactrian camel seminal plasma.
Kumar et al., 2013 [30] - Describe the purification of b-NGF from Bactrian camel seminal plasma.

New World Camelids: Alpaca (Vicugna pacos)

Reference [Nº] Achievement

Paolicchi et al., 1999 [111] - First report suggesting the presence of a factor in alpaca seminal plasma that contributes to the secretion of LH and consequently to the
induction of ovulation in this species.
Adams et al., 2005 [18] - Report the existence of a potent OIF in the seminal plasma of alpacas and llamas capable of inducing a LH surge and ovulation.
- First evidence that OIF has also a luteotrophic effect in llamas.
Ratto et al., 2005 [19] - Suggest that OIF present in alpaca seminal plasma affects ovulation via a systemic rather than a local route.
- Demonstrate that curettage of endometrial mucosa facilitates the absorption of OIF.
- Demonstrate that ovulation in alpacas is not related to physical stimulation of the genital tract.
Ratto et al., 2006 [20] - Confirm the presence of an OIF in alpaca and llama seminal plasma.
- Suggest that OIF is a conserved molecule among mammals.
Kershaw-Young et al., - Identify the presence of b-NGF in high concentration in alpaca seminal plasma.
2012 [28] - Demonstrate that b-NGF is the seminal protein responsible for the induction of ovulation in alpacas.
Kershaw-Young et al., - Suggest that b-NGF is the seminal protein responsible for the induction of ovulation in alpacas.
2012 [40]
Druart et al., 2013 [29] - Confirm that b-NGF is the main protein in alpaca seminal plasma.
Stuart et al., 2014 [33] - Demonstrate that recombinant b-NGF at a dose of 1 or 0.1 mg is as effective as GnRH analogue (Busereline) for the induction of ovulation in
alpacas.
- Demonstrate that varying the dose of b-NGF does not affect timing of ovulation, size of corpus luteum (CL) or plasma progesterone
concentration in alpacas.

New World Camelids: Llama (Lama glama)

Reference Achievement

Adams et al., 2005 [18] - First report of the existence of a potent OIF in seminal plasma of alpacas and llamas capable of inducing a LH surge and ovulation.
- First evidence that OIF has also a luteotrophic effect in llamas.
Ratto et al., 2006 [20] - Confirm the presence of an OIF in alpaca and llama seminal plasma.
- Suggest that OIF is a conserved molecule among mammals.
Ratto et al., 2010 [22] - Demonstrate that seminal OIF is a protein resistant to heat and enzymatic digestion and has a molecular mass 30 kDa.
Ratto et al., 2011 [23] - Describe OIF as a 14 kDa seminal protein that elicits a LH surge, ovulation and the development of CL.
- Suggest an endocrine effect of OIF at hypothalamus or pituitary level.
Tanco et al., 2011 [24] - Demonstrate that OIF is found in high concentration in llama seminal plasma.
- Demonstrate that OIF presents a dose-dependent effect regarding induction of ovulation, CL diameter and plasma progesterone
concentration.
Bogle et al., 2011 [35] - Demonstrate that the mechanism for the biological response to OIF obtained from llama semen is also present in mice.
Bogle et al., 2012 [112] - Demonstrate that OIF induces LH secretion from in vitro cultured llama and bovine pituitary cells.
Ratto et al., 2012 [27] - Reveal that OIF present in llama seminal plasma is Beta-Nerve Growth Factor (b-NGF).
Silva et al., 2011 [37] - Show that rabbit seminal plasma induces a LH surge and ovulation in llamas but llama seminal plasma does not has the same effect in rabbit
does.
Silva et al., 2011 [25] - Demonstrate that GnRH antagonist (Cetrorelix) inhibits the preovulatory LH surge induced by OIF in llamas, strongly suggesting that LH
secretion is modulated by a direct or indirect effect of OIF on GnRH neurons in the hypothalamus.
Silva et al., 2012 [68] - Demonstrate that peripheral estradiol concentration partially modulates the effect of OIF on pituitary LH secretion; however, other(s) ovarian
factor(s) could also participate on this modulatory action in llamas.
Silva et al., 2014 [31] - Show that OIF induces larger CLs that secrete more progesterone when compared with GnRH in llamas.
Fernandez et al., 2014 - Demonstrate that b-NGF exerts a potent luteotrophic effect which is dependent of administration frequency.
[102]
Ulloa-Leal et al., 2014 [26] - Demonstrate that b-NGF exerts its luteotrophic effect by altering the secretion of pituitary LH and enhancing vascularization of pre-ovulatory
follicle and early CL.
M. Silva et al. / Theriogenology 150 (2020) 280e287 283

Table 1 (continued )

Old World Camelids: Bactrian camel (Camelus bactrianus)

Reference [Nº] Achievement

Silva et al., 2015 [114] - Show that regardless of route of administration (i.m., i.v. or intrauterine) b-NGF induces a LH surge, ovulation and the development of a
functional CL in female llamas.
- Demonstrate that the dose of b-NGF required by intrauterine route is higher than that of i.m. or i.v. routes.
Berland et al., 2016 [67] - Demonstrate that llamas can be induced to ovulate by seminal plasma b-NGF in the absence of copulation.
- Show that mechanical stimulation of copulation by itself is not able to induce ovulation in llamas.
- Demonstrate that promptly after copulation the concentration of b-NGF increases in general circulation in llamas.
Silva et al., 2017 [32] - Show that b-NGF increases CL vascularization and level of mRNA expression of steroidogenic enzymes during early stages of CL development
in llamas.
Silva et al., 2017 [113] - Suggest that the luteotrophic effect of b-NGF is influenced by administration route in llamas.
Bogle et al., 2018 [34] - Show that b-NGF is a common protein within the male accessory glands among camelids, bovids and cervids suggesting an important role in
the mechanism of ovulation in both induced and spontaneous ovulators.
Carrasco et al., 2018 [84] - Suggest that the ovulation-inducing effect of b-NGF in llamas is driven by an indirect interaction with GnRH neurons through hypothalamic
interneurons.
Valderrama et al., 2019 - Demonstrate that systemic administration of b-NGF purified from llama seminal plasma to female llamas induces a shift from estradiol to
[104] progesterone production in preovulatory follicles.
- Suggest that changes in gene expression patterns of steroidogenic enzymes observed in granulose cells of preovulatory follicles indicate an
ovarian local effect of b-NGF in this species.

is modulated by circulating concentration of estradiol [68], high- portal system that transport GnRH molecules to the anterior pitu-
lighting the role that the ovary has on the modulation of the hy- itary [76,77]. Several pieces of evidence indicate that the preovu-
pothalamus or pituitary response to NGF in this species. Moreover, latory LH secretion is commanded by a subpopulation of GnRH cells
the administration of exogenous estradiol to ovariectomized llamas located in the anterior hypothalamus in different species. Approx-
was able to partially restore the magnitude of pituitary LH secretion imately 40% of the GnRH cells located in the POA and anterior hy-
when compared to that observed in intact females, confirming that pothalamus express Fos protein, a widely used marker for neuron
plasma estradiol concentration affects pituitary LH secretion trig- activation, in the late proestrus preceding the LH surge in rats [78]
gered by NGF. and mice [79,80]. Similarly, in ferrets, also an induced ovulator,
In a recent llama study [67], it was undoubtedly concluded that mating induces the expression of Fos in 50% of the POA and anterior
the presence of b-NGF in semen is the pivotal factor responsible for hypothalamus GnRH cells, but also in the MBH [81,82].
inducing ovulation, while penile stimulation during mating had no Like in other species, the llama MBH and the anterior hypo-
effect. The results of this study showed that llamas mated with an thalamic area harvest the two largest populations of GnRH cells,
urethrostomized male did not ovulate (0/6), however ovulation rate exhibiting the former twice the number of this type of neurons
in llamas mated with an intact male or those given an intrauterine than that observed in the latter [83]. In this species, GnRH cells
infusion of seminal plasma was 83 (5/6) and 86% (6/7), respectively. were also detected in the POA and the diagonal band of Broca [83].
In addition, the results of this study indicated that intrauterine Interestingly, in llamas, the diagonal band of Broca displays the
administration of seminal plasma was sufficient to elicit the LH highest number of undifferentiated cells (i.e. not GnRH neurons)
surge, and that circulating LH concentration was positively corre- expressing the high-affinity b-NGF receptor TrkA, although this
lated with a rapid systemic increase in b-NGF observed during the expression is also high in the POA and anterior hypothalamus,
first hour after treatments (i.e. mating or intrauterine infusion of whereas the MBH exhibits the lowest number of cells expressing
seminal plasma). The results of these two complementary studies this receptor. The low-affinity b-NGF receptor p75 showed a similar
[67,68] suggest that b-NGF from the seminal plasma is effectively distribution pattern [84]. In the same llama study [84], expression
absorbed from the endometrium surface after copulation, rapidly of TrkA, but not p75, is reported to occur in a low number (less than
entering systemic circulation to elicit GnRH secretion from the 3%) of GnRH cells. Interestingly, according to Herbison [85], the
hypothalamus. recruitment of a small number of GnRH cells usually involved in
GnRH or LH surges (10% in sheep and 20% in mice, respectively) is
5. Potential brain targets of b-NGF eliciting ovulation sufficient to elicit ovulation. Hence, it could be that, as in other
species, the llama LH preovulatory release may be elicited by acti-
Once into the bloodstream, b-NGF could act at different pe- vation of GnRH cells located in the rostral hypothalamus. Whether
ripheral targets, but central targets (brain tissues) are restricted b-NGF is able to penetrate the brain in physiologically significant
because of the blood-brain barrier (BBB). Nevertheless, several amounts has not been determined yet.
evidences suggest that b-NGF may penetrate the brain or act in Interestingly, a recent mice study [86] supports the notion that,
brain areas lacking functional BBB [12,69] to stimulate the GnRH in llamas, b-NGF could modulate the activity of GnRH cells by
preovulatory surge. In mammals, GnRH cells are crucial for repro- activation of central receptors. In a male mice model of hypo-
duction, and yet their critical role is sustained by a remarkably low gonadotropic hypogonadism, intracerebroventricular administra-
number of cells [70,71] that are scattered in a bilateral continuum tion of b-NGF: a) increased plasma LH (and FSH) in vivo; b)
in the basal forebrain [72]. Two major hypothalamic areas are stimulated GnRH secretion from hypothalamic GT1-7 immortalized
known to harvest a large number of GnRH cells: (i) the mediobasal cells (expressing TrkA receptors) in vitro and c) reversed the
hypothalamus (MBH), and (ii) the preoptic area (POA) - anterior hypogonadism as increased gonadal weight and plasma testos-
hypothalamus [73]. Most of these cells display 2 long dendrites [74] terone. This evidence suggests that cellular and molecular mecha-
from which many axon-like projections emerge (so-called ‘den- nism of b-NGF signaling on GnRH systems are present in other
drons’) [75] directing to the median eminence (ME) where their species and are not sex-specific.
nerve endings are close to fenestrated vessels of the hypophyseal
284 M. Silva et al. / Theriogenology 150 (2020) 280e287

5.1. Kisspeptin neurons structure.


Finally, noradrenergic inputs arising from A2 cells of the caudal
Even though the role of GnRH neuroendocrine cells orches- brainstem are known to modulate the activity of preoptic GnRH
trating fertility remains undoubted, it is established that, in most cells at the time of the LH surge [100], and noradrenaline is thought
animal species, these cells lack the estrogen receptor alpha (ERa) to play a major role in the rabbit reflex ovulation [101], however, a
required to trigger the preovulatory GnRH surge by gonadal ste- recent study [67] has ruled out the contribution of neural pathways
roids in spontaneous ovulators [87]. In the last decades, kisspeptin arising from the reproductive tract on the llama ovulatory
neurons which express ERa have emerged as the main input con- mechanism.
trolling the activity of GnRH cells by two hypothalamic populations
located in the arcuate nucleus and the POA. So when estradiol 6. Luteotrophic effect of b-NGF
concentration increases, Kiss1 gene expression in cells of the
arcuate nucleus is downregulated, whereas it is upregulated in the The preovulatory LH surge is the key signal for inducing not only
POA eliciting the preovulatory GnRH surge [88]. In the induced ovulation but also the initial stages of luteinization, CL formation
ovulator musk shrew (Suncus murinus), the expression of Kiss1 and functioning. We have previously documented that seminal
seems to be controlled in a similar fashion as in spontaneous plasma b-NGF is a protein with a significant luteotrophic effect in
ovulators, but the copulatory stimulus is reported to be the final cue llamas [18,23e26]. From early stages of CL development, induced
that activates POA kiss1 cells by means of c-fos expression [89]; by intramuscular administration or by intrauterine infusion of b-
however, if this leads to activation of GnRH cells warrants further NGF, this gland consistently secreted more progesterone than those
investigation. Recently, kisspeptin has been proposed as the po- induced after GnRH treatment. In all these previous studies, there
tential mediator of the b-NGF-stimulated ovulation in camelids was a positive relationship between the magnitude of LH release,
[13]. Interestingly, in a preliminary llama experiment, the ovulatory and subsequent luteal function when females were treated with
effect of the i.v. administration of kisspeptin-10 (the short biolog- seminal plasma or purified seminal plasma b-NGF vs GnRH. These
ically active murine analogue) is interpreted as evidence for the pieces of evidence have provided a rationale for the hypothesis that
involvement of kisspeptin in camelids ovulatory pathway under the LH may mediate the luteotrophic effect of seminal plasma b-NGF. In
assumption that kisspeptin-10 acts on the brain [90]. Small pep- a recent llama study [26] the use of power doppler ultrasonography
tides, such as kisspeptin-10, are often believed to penetrate the demonstrated that the preovulatory follicle and the early CL has a
BBB, although other hypothalamic peptides of similar molecular greater vascularization in seminal plasma b-NGF- than in GnRH-
weights (~1000 g/mol), like oxytocin, do not, at least not in treated females, resulting in higher progesterone secretion.
neuroactive amounts [91]. In mice, i.v. kisspeptin-10 has failed to Indeed, further studies confirmed that luteotrophic effect of semi-
activate GnRH cells (as measured by Fos expression), and yet both nal plasma b-NGF was associated with enhanced tissue vasculari-
short and long kisspeptin forms stimulate LH release and depen- zation during the peri-ovulatory period and early stages of CL
dent effect [92,93], indicating that systemic kisspeptin acts outside development [26,102]. This could be related to an increase of the
the BBB (e.g. pituitary [94,95]; ME [96]). The detection of kisspeptin vascular endothelial growth factor (VEGF) which induces angio-
neurons in the llama brain and whether they express receptors for genesis by stimulating the proliferation of endothelial cells of
b-NGF (i.e. TrkA, p75) awaits to be shown to propose potential preexisting capillaries [103]. In addition, the increasing of CL
physiological roles. vascularization was confirmed in a previous llama study [32],
where females treated with seminal plasma b-NGF displayed a
5.2. Circumventricular organs (CVOs) greater CL vascularization area and up-regulation of mRNA
expression of p450SCC and StAR in luteal tissue, at Day 4 and 8 of
On the other hand, circumventricular organs (CVOs) are small luteal phase enhancing progesterone secretion. Recently, we [104]
neurohemal brain areas lacking functional BBB due to the presence found that mating and systemic administration of purified llama b-
of fenestrated capillaries allowing neurons to monitor and sense NGF induced a rapid shift from estradiol to progesterone produc-
compounds present in systemic circulation (i.e. sensory CVOs), and tion in the preovulatory follicle in llamas. In these females there
also the release of neuroendocrine products into the bloodstream was an up-regulation of the main genes related to progesterone
(i.e. secretory CVOs) [97]. Two CVOs are closely linked to the GnRH production resulting in an increase of P4/E2 ratio in the follicular
system and so are likely targets for circulating b-NGF in llamas: (i) fluid. Llamas mated or treated with b-NGF displayed significant
the organum vasculosum of the lamina terminalis (OVLT) which higher changes in gene expression patterns of steroidogenic en-
contain sensory neuronal elements, and (ii) the ME where GnRH zymes than that observed in GnRH-treated females, suggesting a
nerve endings release their product into the portal circulation. local effect of systemic b-NGF at ovarian level. In this sense, we
Consistent with the above, Loy [69] reported intense labeling of demonstrated that systemic concentration of NGF and LH increased
both CVOs following the i.v. administration of 125I-b-NFG in rats. during the first 3.5 h after administration of seminal plasma b-NGF
In mice, GnRH cells of the rostral POA extend their dendritic [67] which could be related to a local effect of seminal plasma b-
processes into the OVLT, and even some GnRH somas (located NGF at ovarian level stimulating progesterone production. In fact,
within 100 mm from OVLT) are reported to be exposed to com- the addition of purified llama b-NGF in the in vitro culture of llama
pounds present in the bloodstream; 85% of these neurons granulosa cells increased the mRNA relative expression of StAR and
expressed Fos in response to E2-induced LH surge, and locally VEGF resulting in higher progesterone secretion than that observed
applied kisspeptin to some of these cells induced excitatory re- in granulosa cells treated with LH after 48 h of in vitro culture [104].
sponses [98]. So, it might be that GnRH cells expressing TrkA re-
ceptors [83,84] are in contact with b-NGF reaching the OVLT 7. Final remarks
following copulation.
In the ME, b-NGF labeling has been reported to occur in cells of As demonstrated, the high correlation between the concentra-
this CVO in seminal plasma-treated llamas [12], and rats treated tion of b-NGF, in llama and alpaca semen, and its direct effects on
with an antibody against NGF showed blunted GnRH release from ovarian function support the notion of an endocrine mechanism
the ME in response to application of PGE2 in vitro [99], suggesting a controlling ovulation in these species, which is not as a response to
potential role for b-NGF modulating the GnRH release from this physical stimulation of the female genital tract itself. Thus, in these
M. Silva et al. / Theriogenology 150 (2020) 280e287 285

2 species ovulation is a direct consequence of b-NGF absorption select and control line gilts. J Anim Sci 1988;66:1462e74.
[5] Knobil E. The neuroendocrine control of the menstrual cycle. Recent Prog
from the genital mucosa after mating, its rapid increase and sys-
Horm Res 1980;36:53e88.
temic distribution to distant target tissues, the increase in hypo- [6] Jochle W. Current research in coitus-induced ovulation: a review. J Reprod
thalamic GnRH release, the surge in circulating concentrations of Fertil 1975;22:165e207.
LH, and finally a series of molecular, endocrine and morphological [7] Conaway CH. Ecological adaptation and mammalian reproduction. Biol
Reprod 1971;4:239e41.
changes at follicular level ending in the release of the oocyte and [8] Zarrow MX, Clark JH. Ovulation following vaginal stimulation in a sponta-
the development of a CL.The above clearly set the foundations for neous ovulator and its implications. J Endocrinol 1968;40:343e52.
the establishment of a new category of induced-ovulators. [9] Kauffman AS, Rissman EF. Neuroendocrine control of mating-induced
ovulation. In: Neill JD, editor. The physiology of reproduction. San Diego:
Although substantial advances have been made during the last Elsevier Academic Press; 2006. p. 2283e326.
decades to reveal the exact mechanism controlling ovulation in [10] Adams GP, Ratto MH. Ovulation-inducing factor in seminal plasma: a review.
llamas and alpacas, there is still missing information and several Anim Reprod Sci 2013;136:148e56.
[11] Adams GP, Ratto MH, Silva ME, Carrasco RA. Ovulation-inducing factor (OIF/
research gaps need to be addressed before full comprehension of NGF) in seminal plasma: a review and update. Reprod Domest Anim
this phenomena is acquired. According to the authors, the most 2016;51:4e17.
relevant research gaps still existing are: [12] Ratto MH, Berland M, Silva M, Adams GP. New insights of the role of b-NGF
in the ovulation mechanism of induced ovulating species. Reproduction
2019;157:R199e207.
a) The identification of brain targets from which b-NGF can [13] El Allali K, El Bousmaki N, Ainani H, Simonneaux V. Effect of the Camelid’s
start a neuronal chain reaction ending with the hypotha- seminal plasma ovulation-inducing factor/beta-NGF: a kisspeptin target
hypothesis. Front Vet Sci 2017;4:99.
lamic GnRH release. In this regard, the main questions still
[14] Chen BX, Yuen ZX, Pan GW, Huang YM, Gao YH. Studies on the ovulation
remaining unanswered are:i) how does b-NGF reaches GnRH mechanism on the Bactrian camel. II. The role of semen in induction of
neurons located inside de Blood-Brain Barrier (BBB)?, ii) is ovulation. Acta Vet Zootech Sin 1983;14:161e6.
there any possibility for b-NGF to affect target tissues outside [15] Chen BX, Yuen ZX, Pan GW. Semen-induced ovulation in the Bactrian camel
(Camelus bactrianus). J Reprod Fertil 1985;73:335e9.
the BBB to trigger its effect?, and iii) what are the compo- [16] Xu YS, Wang HY, Zeng GQ, Jiang GT, Gao YH. Hormone concentrations before
nents of the intermediary neuronal network translating and after semen-induced ovulation in the Bactrian camel (Camelus bac-
initial action of b-NGF on tissue target and its final effect on trianus). J Reprod Fertil 1985;74:341e6.
[17] Pan G, Chen Z, Liu X, Li D, Xie Q, Ling F, Fang L. Isolation and purification of
GnRH neurons? the ovulation-inducing factor from seminal plasma in the bactrian camel
b) The determination of a direct ovarian local effect of b-NGF on (Camelus bactrianus). Theriogenology 2001;55:1863e79.
follicular cell populations able to induce ovulation, or at least [18] Adams GP, Ratto MH, Huanca W, Singh J. Ovulation inducing factor in the
seminal plasma of alpacas and llamas. Biol Reprod 2005;73:452e7.
to complement its central action at hypothalamic- [19] Ratto MH, Wanca W, Singh J, Adams GP. Local versus systemic effect of
hypophyseal axis. The study of such a local effect using and ovulation-inducing factor in the seminal plasma of alpacas. Reprod Biol
in vivo model represents a great challenge since discrimi- Endocrinol 2005;3:29e44.
[20] Ratto MH, Wanca W, Singh J, Adams GP. Comparison of the effect of
nation of b-NGF actions at both levels (central vs ovarian) is ovulation-inducing factor (OIF) in the seminal plasma of llamas, alpacas and
extremely difficult in a confounding physiological environ- bulls. Theriogenology 2006;66:1102e6.
ment proper of this model. [21] Bogle OA, Ratto MH, Adams GP. Evidence for the conservation of the bio-
logical activity of ovulation-inducing factor in seminal plasma. Reproduction
c) The abundance of b-NGF in seminal plasma of llamas, alpacas
2011;142:277e83.
and camels is an extraordinary reproductive feature that [22] Ratto MH, Wanca W, Adams GP. Ovulation-inducing factor: a protein
probably has other implications besides ovulation induction component of llama seminal plasma. Reprod Biol Endocrinol 2010;8:44.
in these species. Thus, how the high concentration of this [23] Ratto MH, Delbaere LTJ, Leduc YA, Pierson RA, Adams GP. Biochemical
isolation and purification of ovulation-inducing factor (OIF) in seminal
seminal protein relates to male reproductive characteristics, plasma of llamas. Reprod Biol Endocrinol 2011;9:24.
such as sperm physiology or fertility; or how may affect other [24] Tanco VM, Ratto MH, Lazzarotto M, Adams GP. Dose-response of females
female reproductive traits, once it reaches the genital tract llamas to ovulation-inducing factor from seminal plasma. Biol Reprod
2011;85:452e6.
after copulation, are relevant subjects yet unexplored. [25] Silva ME, Smulders JP, Guerra M, Valderrama XP, Letelier C, Adams GP,
d) Although there is a direct relationship between both wild Ratto MH. Cetrorelix suppresses the preovulatory LH surge and ovulation
and domestic species of South American Camelids through induced by ovulation-inducing factor (OIF) present in llama seminal plasma.
Reprod Biol Endocrinol 2011;9:74.
the domestication process (i.e. llamas and alpacas evolved [26] Ulloa-Leal C, Bogle OA, Adams GP, Ratto MH. Luteotrophic effect of
from guanacos and vicugnas respectively), there is no infor- ovulation-inducing factor/nerve growth factor present in the seminal plasma
mation regarding the ovulation mechanism prevailing in of llamas. Theriogenology 2014;81:1101e7.
[27] Ratto MH, Leduc Y, Valderrama XP, van Straaten K, Delbaere L, Pierson R,
wild SACs, neither if the process of domestication altered Adams GP. The nerve of ovulation inducing factor. Proc Natl Acad Sci Unit
ovulation pattern present, as it has affected several other States Am 2012;109:15042e7.
reproductive characteristics in many animal species [105]. [28] Kershaw-Young CM, Druart X, Vaughan J, Maxwell WMC. b-Nerve growth
factor is a major component of alpaca seminal plasma and induces ovulation
in female alpacas. Reprod Fertil Dev 2012;24:1093e7.
Acknowledgements [29] Druart X, Rickard JP, Mactier S, Kohnke PL, Kershaw-Young CM, Bathgate R,
et al. Proteomic characterization and cross species comparison of mamma-
lian seminal plasma. J Proteomics 2013;91:13e22.
Several studies described in the present review were supported [30] Kumar S, Kumar-Sharma V, Singh S, Hariprasad GR, Mal G, Srinivasan A, et al.
by the Chilean Council of Science and Technology (CONICYT, Fon- Proteomic identification of camel seminal plasma: purification of b-nerve
decyt: 1160934, 11140396). growth factor. Anim Reprod Sci 2013;136:284e95.
[31] Silva M, Ulloa-Leal C, Norambuena C, Fern andez A, Adams GP, Ratto MH.
Ovulation-inducing factor (OIF/NGF) from seminal plasma origin enhances
References corpus luteum function in llamas regardless the preovulatory follicle diam-
eter. Anim Reprod Sci 2014;148:221e7.
[1] Bakker J, Baum MJ. Neuroendocrine regulation of GnRH release in induced [32] Silva M, Ulloa-Leal C, Valderrama XP, Bogle OA, Adams GP, Ratto MH. Nerve
ovulators. Front Neuroendocrinol 2000;21:220e62. Growth Factor from seminal plasma origin (spb-NGF) increases CL vascu-
[2] Turzillo AM, Nett TM. Regulations of the GnRH receptor gene expression in larization and level of mRNA expression of steroidogenic enzymes during the
sheep and cattle. J Reprod Fertil 1999;98:113e9. early stage of Corpus Luteum development in llamas. Theriogenology
[3] Jaffe RB, Keys Jr WR. Estradiol augmentation of pituitary responsiveness to 2017;103:69e75.
gonadotropin-releasing hormone in women. J Clin Endocrinol Metab [33] Stuart CC, Vaughan JL, Kershaw-Young CM, Wilkinson J, Bathgate R, de
1974;39:850e5. Graaf SP. Effects of varying doses of b-nerve growth factor on the timing of
[4] Kelly CR, Socha TE, Zimmerman DR. Characterization of gonadotrophic and ovulation, plasma progesterone concentration and corpus luteum size in
ovarian steroids hormones during the periovulatory period in high ovulating females alpacas (Vicugna pacos). Reprod Fertil Dev 2015;27:1181e6.
286 M. Silva et al. / Theriogenology 150 (2020) 280e287

[34] Bogle OA, Carrasco RA, Ratto MH, Singh J, Adams GP. Source and localization TrkA, p75 and inhibin alpha-subunit in interstitial cells of golden hamsters
of ovulation-inducing factor/nerve growth factor in male reproductive tis- treated with hCG. J Reprod Dev 2009;55:622e8.
sues among mammalian species. Biol Reprod 2018;99:1194e204. [64] Sofroniew MV, Howe CL, Mobley WC. Nerve growth factor signaling, neu-
[35] Bogle O, Ratto MH, Adams GP. Evidence for the conservation of biological roprotection, and neural repair. Annu Rev Neurosci 2001;24:1217e81.
activity of ovulation-inducing factor (OIF) in the seminal plasma. Repro- [65] Robertson SA. Seminal fluid signaling in the female reproductive tract: les-
duction 2011;142:277e83. sons from rodents and pigs. J Anim Sci 2007;85(Suppl):36e44.
[36] Ratto MH, Huanca W, Singh J, Adams GP. Comparison of the effect of [66] Robertson SA, Sharkey DJ. The role of semen in the induction of maternal
ovulation-inducing factor (OIF) in seminal plasma of llamas, alpacas, and immune tolerance to pregnancy. Semin Inmunol 2001;13:243e54.
bulls. Theriogenology 2006;66:1102e6. [67] Berland MA, Ulloa-Leal C, Barría M, Wright H, Dissen GA, Silva ME, et al.
[37] Silva M, Nin~ o A, Guerra M, Letelier C, Valderrama XP, Adams GP, Ratto MH. Is Seminal plasma induces ovulation in llamas in the absence of a copulatory
an ovulation-inducing factor (OIF) present in the seminal plasma of rabbits? stimulus: role of nerve growth factor as an ovulation-inducing factor.
Anim Reprod Sci 2011;127:213e21. Endocrinology 2016;157:3224e32.
[38] Maranesi M, Zerani M, Leonardi L, Pistilli A, Arruda-Alencar J, Stabile AM, [68] Silva ME, Recabarren MP, Recabarrren SE, Adams GP. Ratto MH Ovarian
et al. Gene expression and localization of NGF and its cognate receptors estradiol modulates the stimulatory effect of ovulation-inducing factor (OIF)
NTRK1 and NGFR in the sex organs of male rabbits. Reprod Domest Anim on pituitary LH secretion in llamas. Theriogenology 2012;77:1873e82.
2015;50:918e25. [69] Loy R, Taglialatela G, Angelucci L, Heyer D, Perez-Polo R. Regional CNS uptake
[39] Maranesi M, Petrucci L, Leonardi L, Piro F, García-Rebollar P, Millan P, et al. of blood-borne nerve growth factor. J Neurosci Res 1994;39:339e46.
New Insights on a NGF-mediated pathway to induce ovulation in rabbits [70] Casoni F, Malone SA, Belle M, Luzzati F, Collier F, Allet C, et al. Development
(Oryctolagus cuniculus). Biol Reprod 2018;98:634e43. of the neurons controlling fertility in humans: new insights from 3D imaging
[40] Kershaw-Young CM, Maxwell WMC. Seminal plasma components in Cam- and transparent fetal brains. Development 2016;143:3969e81.
elids and comparisons with other species. Reprod Domest Anim [71] Wray S, Grant P, Gainer H. Evidence that cells expressing luteinizing
2012;47(Suppl 4):369e75. hormone-releasing hormone mRNA in the mouse are derived from pro-
[41] Korsching S, Auburger G, Heumann R, Scott J, Thoenen H. Levels of nerve genitor cells in the olfactory placode. Proc Natl Acad Sci Unit States Am
growth factor and its mRNA in the central nervous system of the rat correlate 1989;86:8132e6.
with cholinergic innveration. EMBO J 1985;4:1389e93. [72] Wray S. From nose to brain: development of gonadotrophin-releasing hor-
[42] Korsching S, Thoenen H. Treatment with 6-hydroxydopamine and colchicine mone-1 neurones. J Neuroendocrinol 2010;22:743e53.
decreases nerve growth factor levels in sympathetic ganglia and increases [73] Herbison AE. Physiology of the adult gonadotropin-releasing hormone
them in the corresponding target tissues. J Neurosci 1985:1058e61. neuronal network. In: Plant TM, Zeleznik AJ, editors. Knobil and Neill’s
[43] Levi-Montalcini R. The nerve growth factor: thirty-five years later. EMBO J physiology of reproduction. San Diego: Academic Press; 2015. p. 399e467.
1987;6:1145e54. [74] Campbell RE, Han SK, Herbison AE. Biocytin filling of adult gonadotropin-
[44] Lewin GR, Barde YA. Physiology of the neurotrophins. Annu Rev Neurosci releasing hormone neurons in situ reveals extensive, spiny, dendritic pro-
1996;19:289e317. cesses. Endocrinology 2005;146:1163e9.
[45] Go€ tz R, Ko
€ster RW, Winkler C, Raulf F, Lottspeich F, Schartl M, et al. Neu- [75] Herde MK, Iremonger KJ, Constantin S, Herbison AE. GnRH neurons elaborate
rotrophin-6 is a new member of the nerve growth factor family. Nature a long-range projection with shared axonal and dendritic functions.
1994;372:266e9. J Neurosci 2013;33:12689e97.
[46] Lai KO, Fu WY, Ip FCF, IP NY. Cloning and expression of a novel neurotrophin, [76] Silverman AJ, Jhamandas J, Renaud LP. Localization of luteinizing hormone-
NT-7. Mol Cell Neurosci 1998;11:64e76. releasing hormone (LHRH) neurons that project to the median eminence.
[47] Nilsson AS, Fainzilber M, Falck P, Ibanez CF. Neurotrophin-7: a novel member J Neurosci 1987;7:2312e9.
of the neurotrophin family from the zebrafish. FEBS Lett 1998;424:285e90. [77] Goldsmith PC, Thind KK, Song T, Kim EJ, Boggant JE. Location of the neuro-
[48] Anderson RA, Robinson LL, Brooks J, Spears N. Neurotrophins and their re- endocrine gonadotropin-releasing hormone neurons in the monkey hypo-
ceptors are expressed in the human fetal ovary. J Clin Endocrinol Metab thalamus by retrograde tracing and immunostaining. J Neuroendocrinol
2002;87:890e7. 1990;2:157e68.
[49] Dissen GA, Parrot JA, Skinner MK, Hill DF, Costa ME, Ojeda SR. Direct effects [78] Halasz B, Pupp L. Hormone secretion of the anterior pituitary gland after
of nerve growth factor on thecal cells from antral ovarian follicles. Endo- physical interruption of all nervous pathways to the hypophysiotrophic area.
crinology 2000;141:4736e50. Endocrinology 1965;77:553e62.
[50] Lara HE, Hill DF, Katz KH, Ojeda SR. The gene encoding nerve growth factor is [79] Lee WS, Smith MS, Hoffman GE. Luteinizing hormone-releasing hormone
expressed in the inmature rat ovary: effect of denervation and hormonal neurons express Fos protein during the proestrous surge of luteinizing
treatment. Endocrinology 1990;126:357e63. hormone. Proc Natl Acad Sci Unit States Am 1990;87:5163e7.
[51] Romero C, Paredes A, Dissen GA, Ojeda SR. Nerve Growth factor induces the [80] Constantin S, Piet R, Iremonger K, Hwa Yeo S, Clarkson J, Porteous R, et al.
expression of functional FSH receptors in newly formed follicles of the rat GnRH neuron firing and response to GABA in vitro depend on acute brain
ovary. Endocrinology 2002;143:1485e94. slice thickness and orientation. Endocrinology 2012;153:3758e69.
[52] Taglialatela G, Angelucci L, Scaccianoce S, Foreman YJ, Perez-Polo IR. Nerve [81] Chan H, Prescott M, Ong Z, Herde MK, Herbison AE, Campbell RE. Dendritic
growth factor modulates the activation of the hypothalamo-pituitary- spine plasticity in gonadatropin-releasing hormone (GnRH) neurons acti-
adrenocortical axis during the stress response. Endocrinology 1991;129: vated at the time of the preovulatory surge. Endocrinology 2011;152:
2212e8. 4906e14.
[53] Otten U, Ehrhard P, Peck R. Nerve growth factor induces growth and dif- [82] Lambert GM, Rubin BS, Baum MJ. Sex difference in the effect of mating on c-
ferentiation of human B lymphocytes. Proc Natl Acad Sci Unit States Am fos expression in luteinizing hormone-releasing hormone neurons of the
1989;86:10059e63. ferret forebrain. Endocrinology 1992;131:1473e80.
[54] De Simone R, Alleva E, Tirassa P, Aloe L. Nerve growth factor released into the [83] Carrasco RA, Singh J, Adams GP. Distribution and morphology of
bloodstream following intraspecific fighting induces mast cell degranulation gonadotropin-releasing hormone neurons in the hypothalamus of an
in adult male mice. Brain Behav Immun 1990;4:74e81. induced ovulator - the llama (Lama glama). Gen Comp Endocrinol 2018;263:
[55] Julio-Pieper M, Lara H, Bravo J, Romero C. Effects of nerve growth factor 43e50.
(NGF) on blood vessels area and expression of the angiogenic factors VEGF [84] Carrasco RA, Singh J, Adams GP. The relationship between gonadotropin
and TGFbeta1 in the rat ovary. Reprod Biol Endocrinol 2006;4:57. releasing hormone and ovulation inducing factor/nerve growth factor re-
[56] Dissen GA, Mayerhofer A, Ojeda SR. Participation of nerve growth factor in ceptors in the hypothalamus of the llama. Reprod Biol Endocrinol 2018;16:
the regulation of ovarian function. Zygote 1996;4:309e12. 83.
[57] Harper GP, Glanville RW, Thoenen H. The purification of nerve growth factor [85] Herbison AE. Control of puberty onset and fertility by gonadotropin-
from bovine seminal plasma. Biochemical characterization and partial amino releasing hormone neurons. Nat Rev Endocrinol 2016;12:452e66.
acid sequence. J Biol Chem 1982;257:8541e8. [86] Luo J, Yang Y, Zhang T, Su Z, Yu D, Lin Q, et al. Nasal delivery of nerve growth
[58] Fraser HM, Wulff C. Angiogenesis in the primate ovary. Reprod Fertil Dev factor rescue hypogonadism by up-regulating GnRH and testosterone in
2001;13:557e66. aging male mice. EBioMedicine 2018;35:295e306.
[59] Dissen GA, Romero C, Paredes A, Ojeda S. Neurotrophic control of ovarian [87] Wintermantel TM, Campbell RE, Porteous R, Bock D, Grone HJ, Todman MG,
development. Microsc Res Tech 2002;59:509e15. et al. Definition of estrogen receptor pathway critical for estrogen positive
[60] Jana B, Koszykowska M, Czarzasta J. Expression of nerve growth factor and feedback to gonadotropin-releasing hormone neurons and fertility. Neuron
its receptors, TrkA and p75, in porcine ovaries. J Reprod Dev 2011;57: 2006;52:271e80.
468e74. [88] Uenoyama Y, Inoue N, Nakamura S, Tsukamura H. Central mechanism con-
[61] Ren LQ, Medan MS, Weng Q, Jin W, Li CM, Watanabe G, et al. Immunoloc- trolling pubertal onset in mammals: a triggering role of kisspeptin. Front
alization of nerve growth factor (NGF) and its receptors (TrkA and Endocrinol 2019;10:312.
p75LNGFR) in the reproductive organs of Shiba goats. J Reprod Dev 2005;51: [89] Inoue N, Sasagawa K, Ikai K, Sasaki Y, Tomikawa J, Oishi S, et al. Kisspeptin
399e404. neurons mediate reflex ovulation in the musk shrew (Suncus murinus). Proc
[62] Shi Z, Jin W, Watanabe G, Suzuki AK, Takahashi S, Taya K. Expression of nerve Natl Acad Sci Unit States Am 2011;108:17527e32.
growth factor (NGF) and its receptors TrkA and p75 in ovaries of the cyclic [90] Carrasco RA, Leonardi CE, Hutt KD, Singh J, Adams GP. 4 Induction of
golden hamster (Mesocricetus auratus) and the regulation of their produc- ovulation by kisspeptin in llamas. Reprod Fertil Dev 2019;31:127.
tion by luteinizing hormone. J Reprod Dev 2004;50:605e11. [91] Leng G, Ludwig M. Intranasal oxytocin: myths and delusions. Biol Psychiatr
[63] Weng Q, Shi Z, Tukada J, Watanabe G, Taya K. Immunodetection of NGF, 2016;79:243e50.
M. Silva et al. / Theriogenology 150 (2020) 280e287 287

[92] Mikkelsen JD, Bentsen AH, Ansel L, Simonneaux V, Juul A. Comparison of the [104] Valderrama XP, Goicochea JF, Silva ME, Ratto MH. The effect of seminal
effects of peripherally administered kisspeptins. Regul Pept 2009;152: plasma b-NGF on follicular fluid hormone concentration and gene expres-
95e100. sion of steroidogenic enzymes in llama granulosa cells. Reprod Biol Endo-
[93] d’Anglemont de Tassigny X, Jayasena CN, Murphy KG, Dhillo WS, crinol 2019;17:60.
Colledge WH. Correction: mechanistic insights into the more potent effect of [105] Setchell BP. Domestication and reproduction. Anim Reprod Sci 1992;28:
KP-54 compared to KP-10 in vivo. PloS One 2018;13:e0192014. 195e202.
[94] Suzuki S, Kadokawa H, Hashizume T. Direct kisspeptin-10 stimulation on [106] Zhao XX, Li XL, Chen BX. Isolation of ovulation-inducing factors in the
luteinizing hormone secretion from bovine and porcine anterior pituitary seminal plasma of Bactrian camels (Camelus bactrianus) by DEAE-cellulose
cells. Anim Reprod Sci 2008;103:360e5. chromatography. Reprod Domest Anim 2001;36:177e81.
[95] Gutierrez-Pascual E, Martinez-Fuentes AJ, Pinilla L, Tena-Sempere M, [107] Li X, Zhao X. Separation and purification of ovulation-inducing factors in the
Malagon MM, Castano JP. Direct pituitary effects of kisspeptin: activation of seminal plasma of the Bactrian Camel (Camelus bactrianus). Vet Res Com-
gonadotrophs and somatotrophs and stimulation of luteinising hormone and mun 2004;28:235e45.
growth hormone secretion. J Neuroendocrinol 2007;19:521e30. [108] Zhao XX, Pan GW, Huang YM. Studies on the ovulation inducing factors in
[96] d’Anglemont de Tassigny X, Fagg LA, Carlton MB, Colledge WH. Kisspeptin the seminal plasma of Bactrian camel. In: Saint-Martin G, editor. Procceed-
can stimulate gonadotropin-releasing hormone (GnRH) release by a direct ings of the Workshop: is it possible to improve the reproductive perfor-
action at GnRH nerve terminals. Endocrinology 2008;149:3926e32. mance of the camel? Paris: CIRAD-EMVT; 1990. p. 197e208.
[97] Miyata S. New aspects in fenestrated capillary and tissue dynamics in the [109] Liu XT, Pan GW, Li D, Zhao QZ, Xie QG, Ling FX, et al. Separation of active
sensory circumventricular organs of adult brains. Front Neurosci 2015;9:390. peptides from the seminal plasma of camels (Camelus bactrianus). Chinese
[98] Herde MK, Geist K, Campbell RE, Herbison AE. Gonadotropin-releasing Vet Sci Tech 1995;25:7e9.
hormone neurons extend complex highly branched dendritic trees outside [110] Li XL, Zhang Y, Chen BX, Zhao XX. The concentrations of LH, FSH, Oestradiol-
the blood-brain barrier. Endocrinology 2011;152:3832e41. 17B and Progesterone in blood plasma of the Bactrian Camel (Camelus
[99] Lara HE, McDonald JK, Ojeda SR. Involvement of nerve growth factor in fe- bactrianus) before and after intramuscular injection of seminal plasma. Vet
male sexual development. Endocrinology 1990;126:364e75. Res Commun 2002;26:571e6.
[100] Herbison AE. Noradrenergic regulation of cyclic GnRH secretion. Rev Reprod [111] Paolicchi F, Urquieta B, Del Valle L, Bustos-Obregon E. Biological activity of
1997;2:1e6. the seminal plasma of alpacas: stimulus for the production of LH by pituitary
[101] Kaynard AH, Pau KY, Hess DL, Spies HG. Gonadotropin-releasing hormone cells. Anim Reprod Sci 1999;54:203e10.
and norepinephrine release from the rabbit mediobasal and anterior hypo- [112] Bogle OA, Ratto MH, Adams GP. Ovulation-inducing factor (OIF) induces LH
thalamus during the mating-induced luteinizing hormone surge. Endocri- secretion from pituitary cells. Anim Reprod Sci 2012;133:117e22.
nology 1990;127:1176e85. [113] Silva M, Urra F, Ulloa-Leal C, Ratto MH. A comparative study of the effects of
[102] Fernandez A, Ulloa-Leal C, Silva M, Normabuena C, Adams GP, Guerra M, intramuscular administration of gonadorelin, mating and intrauterine infu-
Ratto MH. The effect of repeated administration of llama ovulation-inducing sion of either raw seminal plasma or seminal plasma purified b-NGF on
factor (OIF/NGF) during the periovulatory period on corpus luteum devel- luteal development in llamas. Reprod Domest Anim 2017;52:625e31.
opment and function in llamas. Anim Reprod Sci 2014;149:345e52. [114] Silva M, Fernandez A, Ulloa-Leal C, Adams GP, Berland MA, Ratto MH. LH
[103] Duorak HF, Nagy JA, Feng D, Brown LF, Dvorak AM. Vascular permeability release and ovulatory response after intramuscular, intravenous, and intra-
factor/vascular endothelial growth factor and the significance of microvas- uterine administration of b-nerve growth factor of seminal plasma origin in
cular hyperpermeability in angiogenesis. Curr Microbio Immuno 1999;237: female llamas. Theriogenology 2015;84:1096e102.
97e132.

You might also like