You are on page 1of 17

At the Cutting Edge

Neuroendocrinology 2018;107:400–416 Received: July 24, 2018


Accepted after revision: October 16, 2018
DOI: 10.1159/000494558 Published online: October 16, 2018

Sexual Behavior: From Hormonal


Regulation to Endocrine Disruption
Sakina Mhaouty-Kodja Lydie Naulé Daphné Capela
CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Sorbonne Université, Paris, France

Keywords the success of sexual reproduction, on which species sur-


Sexual behavior · Endocrine disruptor · Sex steroids · vival depends. Males and females adopt different behav-
Reproduction · Nervous system iors and postures during attraction and mating. Male ro-
dents readily express sexual behavior whenever a recep-
tive female is present. During the precopulatory or
Abstract appetitive phase, males engage in chemoinvestigation,
Sexual behavior constitutes a chain of behavioral responses displaying an olfactory preference for receptive females.
beginning with courtship and leading to copulation. These Males generate ultrasonic vocalizations in response to fe-
responses, which are exhibited in a sexually dimorphic man- males or their odors [1–3]. These vocalizations have char-
ner by the two partners, are tightly regulated by sex steroid acteristics in common with the songs of songbirds [4].
hormones as early as the perinatal period. Hormonal chang- The emission of courtship vocalizations conveys infor-
es or exposure to exogenous factors exhibiting hormone- mation about the male’s motivational state and helps to
mimetic activities, such as endocrine disrupting compounds attract the female partner. During the copulatory or con-
(EDC), can therefore interfere with their expression. Here we summatory phase, males mount, thrust, intromit. In male
review the experimental studies in rodents performed to ad- mice, mating ends with ejaculation, whereas male rats
dress the potential effects of exposure to EDC on sexual be- reach satiety after several ejaculations and do not then
havior and underlying mechanisms, with particular atten- copulate again for 1–3 days [5].
tion to molecules with estrogenic and/or anti-androgenic In female rodents, receptivity is constrained by
activities. © 2018 S. Karger AG, Basel threshold levels of estradiol (E2) following progesterone
secretion, and is, therefore, limited to the estrous phase
of the cycle corresponding to the ovulation period. The
female also participates actively in mating, through
Introduction three phases of sexual behavior: attractivity, proceptiv-
ity, and receptivity [6]. During attractivity, the female
Sexual reproduction in vertebrates requires sexually stimulates male behavior by emitting pheromones. The
differentiated behaviors to ensure attraction between female then adopts several proceptive behaviors (hops,
partners and mating. These behaviors are a key factor in darts, solicitations) in response to stimuli from the male

© 2018 S. Karger AG, Basel Sakina Mhaouty-Kodja


CNRS UMR 8246, INSERM U1130, Sorbonne Université
7, quai St Bernard, Bât A 3ème étage
E-Mail karger@karger.com
FR–75005 Paris (France)
www.karger.com/nen E-Mail sakina.mhaouty-kodja @ sorbonne-universite.fr
[7]. Finally, during the copulatory phase, the female responses. Projections are sent from the hypothalamic
adopts a receptive posture called lordosis while ap- paraventricular nucleus to the spinal centers that pro-
proached from behind for insemination by the courting mote penile erection and ejaculation, including the spinal
male [8]. nucleus of the bulbocavernosus, and the gastrin-releasing
Sexual behavior is tightly controlled by finely tuned peptide system.
neural processes, which begin during development and Perinatal testosterone secretion has organizational ef-
are tightly regulated by gonadal hormones. These pro- fects, resulting in structural, neurochemical and molecu-
cesses may, therefore, be highly sensitive to exposure to lar differences in circuitry between the sexes. Differences
endocrine disrupting compounds (EDC), defined by the in cell number and morphology or fiber density between
WHO and the Endocrine Society as exogenous chemical the sexes have frequently been described for the medial
substances or mixture of substances that alter the func- amygdala, the bed nucleus of the stria terminalis, and the
tions of the endocrine system. Exposure to EDC has been medial preoptic area [12]. For instance, the rat sexually
reported to alter several reproductive functions, includ- dimorphic nucleus (SDN) and corresponding cluster of
ing organ development, germ cell production, pubertal calbindin-immunoreactive neurons, both located in the
timing and other physiological processes required for medial preoptic area, contain more cells in males than in
fertility, as documented by both experimental and epide- females [13, 14]. Conversely, neurons expressing kiss-
miological studies [9]. Several studies have addressed the peptin and tyrosine hydroxylase in the anteroventral
effects of exposure to EDC on mating behavior. Here, we periventricular (AVPV) nucleus, a subdivision of the
review the experimental studies in rodents performed to preoptic area involved in the ovulatory surge of LH, are
address the potential effects of exposure to EDC on sex- more numerous in females than in males [15–17]. The
ual behavior. We focus on the underlying molecular and regulation of these neuronal populations, or of other sex-
neural processes affected by such exposure, paying par- ually dimorphic features, by perinatal testosterone can be
ticular interest to molecules with estrogenic and/or anti- mimicked by E2, because gonadal testosterone is aroma-
androgenic activities. A recent review by Gore et al. [10] tized into neural E2 by the aromatase cytochrome P450.
provided the general ethological background to sexual At the molecular level, testosterone and its neural me-
selection and reproductive competence in animal species tabolite E2 also trigger differences in gene expression be-
and their sensitivity to EDC. tween the sexes, through long-lasting changes including
epigenetic modifications [18].
The neural circuitry underlying behavior in females
Hormonal Regulation of Sexual Behavior and also includes the olfactory bulb, which transmits signals
Underlying Sexual Dimorphisms to the medial amygdala and then to the bed nucleus of
the stria terminalis, the preoptic area, and the ventrome-
Critical Periods of Hormonal Regulation dial hypothalamus, the principal facilitatory system for
Perinatal Period lordosis behavior. Systems inhibiting lordosis and in-
Since the pioneering work of Phoenix et al. [11], it has volving the lateral septum, the preoptic area, and the ar-
become clear that gonadal hormones play a key role in the cuate nucleus have also been reported. These nuclei also
sexual differentiation of mating behavior. This process receive projections from the medial amygdala and the
begins early, in the perinatal (late gestational and early bed nucleus of the stria terminalis. These inhibitory and
neonatal) period. In males, testosterone released from the facilitatory systems send projections to the periaque-
fetal and neonatal testes permanently potentiates male ductal gray regions through the ventromedial hypothal-
(masculinization) behavioral and anatomic characteris- amus and lateral septum, respectively. These projections
tics whilst inhibiting female (defeminization) character- relay the information to spinal motoneurons innervat-
istics in the neural circuitry underlying sexual behavior. ing the axial muscles involved in the lordosis posture
This circuitry is stimulated by pheromonal cues emitted [19]. During the prenatal and early postnatal periods,
by receptive females and transmitted from the main ol- these neural structures are protected from the masculin-
factory epithelium and vomeronasal organ to the main izing effects of sex steroids by the presence of
and accessory olfactory bulbs, respectively, and then to α-fetoprotein, which selectively sequesters E2 derived
the chemosensory responsive nuclei in the medial amyg- from the mother and male siblings since the fetal ovaries
dala, the bed nucleus of the stria terminalis, and the me- are inactive [20].
dial preoptic area, where they are processed in behavioral

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 401


DOI: 10.1159/000494558
Prepubertal/Pubertal Period pression of the inhibition exerted by the lateral septum
The pubertal period is characterized by the central ac- and inactivation of the β-endorphin system in the preop-
tivation of pulsatile GnRH secretion, which stimulates tic area. Several neuropeptides and neurotransmitters
the pituitary gonadotropin secretion required for sexual present in the ventromedial hypothalamus display differ-
maturation and fertility. During this period, testosterone ences between the sexes. For example, estrogen receptors
also exerts long-term effects on behavior to ensure the (ER), PR GABA, and enkephalin are all more abundant
maturation of processes initiated during the perinatal pe- in females than in males and are known to promote fe-
riod. Indeed, the prepubertal castration of male hamsters male sexual behavior [31].
has been shown to decrease sexual behavior (fewer
mountings and intromissions and longer times to ejacu- Mechanisms Underlying the Hormonal Regulation of
lation) and to shorten the time to lordosis relative to that Sexual Behavior
for males castrated after puberty [21]. Neural processes, Sex steroids regulate sexual behavior principally
such as cell proliferation, in sexually dimorphic regions through nuclear superfamily receptors. As the detailed
are influenced by gonadal hormones during the peripu- mechanisms underlying the expression of sexual behav-
bertal period [22, 23]. ior have been largely reviewed, this paragraph summa-
In females, α-fetoprotein levels decrease after birth rizes very briefly the genetic studies investigating the rel-
and the ovaries begin releasing E2 on postnatal day (PND) ative contribution of androgen (AR) and ER. Indeed, the
7. Postnatal/prepubertal E2 secretion plays an active role involvement of each of these receptors in this regulation
in the feminization of sexual behavior. Indeed, E2 admin- has been studied by ubiquitous gene invalidations. In
istration between PND15 and PND25 in aromatase males, data from the testicular feminization mutation and
knockout mice, which normally have highly impaired global AR knockout models, both of which result in a
lordosis behavior, partially restores this behavior [24]. feminine phenotype, have suggested that this receptor
During this postnatal period, ovarian E2 is also essential plays some kind of role in the expression of sexual behav-
for the establishment of the LH surge, which is synchro- ior in rats and mice [32]. Global ERα knockout mice are
nized with the receptivity period. In particular, prepuber- infertile and have impaired sexual behavior [33]. Global
tal E2 activates an increase in kisspeptin expression in the ERβ knockout males have normal sexual behavior [34],
rostral periventricular area of the third ventricle [25–27]. but mutant males derived from an ERβ knockout mouse
line completely devoid of ERβ transcripts [35] are infer-
Adulthood tile and display a mild impairment of sexual behavior
In adult males, gonadal testosterone acts on the male [36]. In the global ERα and ERβ knockout models, fe-
neural circuitry to stimulate sexual behavior. This activa- males are infertile, with much lower frequencies of lor­
tional effect of testosterone is transient by comparison to dosis and proceptive behaviors than wild-type females
the permanent organizational changes induced during [36, 37].
the developmental and pubertal periods. Male sexual The neural role of these receptors was dissected more
stimulation is reduced or inhibited by castration but can precisely by conditional gene invalidation, which has the
be restored by hormonal supplementation. Testosterone advantage of preserving peripheral receptor expression
and its neural metabolite E2 regulate the signaling path- and not interfering with gonadal and genital tract effects.
ways of neurotransmitters and neuropeptides playing an In males, the estrogenic and androgenic pathways seem
important role in displays of sexual behavior, such as oxy- to play complementary roles in the organization and ac-
tocin, dopamine, and glutamate. tivation of the neural circuitry underlying sexual behav-
Cyclic females mate only during the estrous phase and ior. The perinatal masculinization and defeminization of
are sexually inactive during the rest of the cycle [28]. The brain structures involve the estrogenic pathway, whereas
preovulatory surge of E2, which occurs during the proes- the neural AR is involved principally in the postnatal or-
trus phase, triggers not only an ovulatory surge of LH, but ganization of spinal nuclei; however, both pathways seem
also the expression of progesterone receptors (PR) in the to be involved in the adult activation of sexual behavior
ventromedial hypothalamus [29, 30]. Progesterone re- [32].
lease under the control of LH then induces female recep- In females, neural ERβ invalidation delays the onset of
tivity, which is perfectly synchronized with ovulation in puberty but has no effect on sexual behavior and fertility
such species [28]. The increase in E2 levels also relieves in either naïve or sexually experienced mutants [38]. This
constraints exerted by the inhibitory system through sup- suggests that the ovary may be the primary site of ERβ ac-

402 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558
Table 1. The analyzed EDC presented by hormonal activity, uses and reference doses established by agencies when available

Source or use Reference doses

Estrogenic compounds
Bisphenol A (BPA) In polycarbonate plastic used in the manufacture of TDI = 0.004 mg/kg/day (EFSA, 2015)
food containers, dental resins, thermal papers TDI = 0.05 mg/kg/day (US EPA, 2012)
Diethylstilbestrol (DES) Used in cases of preterm labor. Banned in 1971 in the
USA and since 1975 in several European countries
Ethinyl estradiol (EE) Oral contraceptive treatment
Methoxychlor (MXC) Organochlorine insecticide banned since 2002 in EU ADI: 0.1 mg/kg/day (EU Pesticides
and 2003 in the USA. Still present in the environment Database)
and used for applications such as mosquito and malaria
control in developing countries
Nonylphenol (NP) Nonionic surfactant used in industrial, agricultural and TDI: 0.005 mg/kg/day (Danish EPA,
domestic applications such as soap, cosmetics, paints, 2000)
herbicides and pesticides, or plastic fabrication
Phytoestrogens
Coumestrol Present in plants (Alfalfa, soybeans…)
Ferutinin Present in roots of Ferula plants
Isoflavones Present in plants (lupin, soybeans, fava beans…) LOAEL of 35 mg/kg/day for isoflavones
(daidzein, genistein) with safety factor of 300 (Anses 2015)
Resveratrol (RVT) Present in grapes, blueberries, raspberries…
Anti-androgenic compounds
Phthalates
Dibutyl phthalate (DBP) Used in the manufacture of PVC plastics, adhesives, TDI: 0.01 mg/kg/day (EFSA, 2005)
inks DNEL: 0.0067 mg/kg/day (ECHA 2017)
Di-(2-ethylhexyl) adipate Used as a substitute for DEHP (marginal use): PVC,
(DEHA) food plastic, cosmetics
Di-(2-ethylexyl)phthalate Used in PVC, food containers, medical products, TDI: 0.050 mg/kg/day (EFSA, 2005)
(DEHP) automobile… NOAEL: 4.8 mg/kg/day, DNEL: 0.034 mg/
kg/day (ECHA 2017)
Diisononyl phthalate Used as a replacement for DEHP: PVC, paint, inks, Sum DIDP/DINP: 0.15 mg/kg/day
(DINP) toys, plastic, soles, automobile (EFSA, 2005)
Vinclozolin Fungicide widely used on fruits and vegetables to avoid ADI = 0.01 mg/kg/day (WHO, 1998)
rot and mildew NOAEL = 1.2 mg/kg/day (US EPA, 2003)
ADI = 0.005 mg/kg/day (EU Pesticides
Database)

ADI, acceptable daily intake dose; DNEL, derived no-effect level; LOAEL, lowest observed adverse effect level; NOAEL, no-observed
effect level; TDI, tolerable daily intake dose.

tion in these processes in adults. Further studies are re- Effects of EDC on Sexual Behavior
quired to address the behavioral phenotype of neural ERα
knockout mice, but this receptor seems to play a major We here describe the data collected for rodents up to
role in the E2-dependent regulation of reproductive pro- December 2017, with a special focus on EDC exhibiting
cesses, as demonstrated by the infertile phenotype in- estrogenic/anti-estrogenic or anti-androgenic activities
duced by neural mutations [27, 39, 40]. given the nature of signaling pathways involved in the

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 403


DOI: 10.1159/000494558
expression of sexual behavior. Table 1 lists the EDC ana- gens (coumestrol, genistein, resveratrol) nonylphenol
lyzed, with their estrogenic or anti-androgenic activities, and methoxychlor (Table 2).
uses and reference doses established by agencies when In rats, with the exception of high doses of NP, which
available. This list, which is far from exhaustive, includes did not affect male sexual behavior [43], changes in at
the most widely studied molecules of these categories. least one of the components of sexual behavior were re-
Furthermore, some molecules, such as bisphenol A (BPA) ported. Increases in the time to intromission and a larger
or methoxychlor, are listed as estrogenic compounds, but number of intromissions were reported for prenatal and
can actually display anti-androgenic activities or target postnatal exposure to BPA, respectively [44]. A dose of
the thyroid system as for BPA. The keywords used to 100 µg/kg/day of BPA decreased the intromission ratio
identify the studies concerned included the name of the without modifying the number of mounts, mating dura-
molecule of interest (e.g., BPA, phytoestrogen, phthal- tion or time to ejaculation in rats [42], and a decrease in
ate), and the terms “sexual behavior,” “endocrine disrup- time to intromission was associated with no change in the
tor,” “brain” or “nervous system.” Behavioral data are number of intromissions or the intromission ratio [45].
presented by period of exposure (developmental periods One interesting study reported different behavioral re-
including prenatal, postnatal and pre/pubertal develop- sponses as a function of BPA dose and sexual experience
ment, and adult exposure), type of activity of the com- [46]. Males exposed to phytoestrogens were reported to
pound (estrogenic or anti-androgenic), animal species have a lower frequency of mounting, intromission and
(rat, mouse), and year of publication, in Tables 2 and 3, ejaculation in a number of studies [42, 47–50], whereas 2
for males and females, respectively. When investigated in studies reported increases in these behaviors [41, 51]. In
the same studies, neuroanatomical analyses of sexually middle-aged males, chronic exposure to methoxychlor
dimorphic regions, neuropeptides or receptors involved has also been shown to decrease time to ejaculation [52].
in sexual behavior and hormonal measurements are in- The published neuroanatomical data are presented in on-
cluded in Tables 2 and 3. Such analyses are otherwise pre- line supplementary Table 1: 6 studies described changes
sented separately in online supplementary Tables 1 and 2 in the SDN or AVPV volume or cell number [47, 53–57],
(see www.karger.com/doi/10.1159/000494558 for all on- whereas 3 studies reported no effect [42, 58, 59]. In the
line suppl. material) for males and females, respectively. studies reporting effects of exposure on the organization
of sexually dimorphic populations, both similarities and
Effects of Developmental versus Adult Exposure to differences were observed, depending on the type of neu-
Estrogenic Compounds ronal cell analyzed. Indeed, increases in the number of
E2, estradiol benzoate or analogs, such as ethinyl estra- tyrosine hydroxylase cells or AVPV volume have been
diol (EE) or diethylstilbestrol (DES), have been used as described [47, 56, 57], suggesting demasculinization and/
positive controls in studies evaluating compounds with or feminization of this hypothalamic region. Opposite
potential estrogenic activity. The behavioral modifica- patterns have been observed, with either a decrease (res-
tions observed differed between studies, ranging from be- veratrol [47]; BPA [54]) or an increase in SDN volume or
havior inhibition with blocked ejaculation for estradiol calbindin cell number (genistein or nonylphenol [55];
benzoate in males [41] to unaltered behavior for DES BPA or genistein [57]; BPA [53]). Overall, such neuro-
[42], as shown in Table 2. These discrepancies probably anatomical measures alone are probably not sufficient or
reflect the different doses of E2 and its analogs used. This, enough sensitive to reflect EDC-induced effects on the
together with the use of only one dose of the positive con- expression of behavior.
trol in most studies, makes it difficult to draw any firm In mice, males exposed prenatally to methoxychlor
conclusions about the potential estrogenic effects of the displayed diminished sexual arousal, spending less time
EDC molecules studied. We will therefore discuss the re- close to a female partition [60]. Exposure to BPA or phy-
ported effects of exposure to BPA, phytoestrogens, non- toestrogen did not affect olfactory preference, time to the
ylphenol or methoxychlor without comparison to posi- initiation of certain behaviors or the frequency of behav-
tive controls. ioral events [61–64]. Decatanzaro et al. [61] observed
only a fewer number of intromissions in animals exposed
Effects of Exposure in Males to BPA in a high-phytoestrogen diet. In addition, male
Prenatal/Postnatal Exposure. Twelve studies in rats mice exposed to BPA displayed less lordosis behavior
and 7 in mice have investigated the effects of early expo- than the vehicle group when gonadectomized and primed
sure to estrogenic compounds, such as BPA, phytoestro- with E2 and progesterone, indicating a greater masculin-

404 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558
Table 2. Behavioral studies in males
Ref. Species Exposure period Route Doses Age at analyses Behavioral analyses Neuroanatomical and hormonal analyses
Developmental
Estrogenic compounds
44 Sprague- Prenatal: from Oral BPA: 40 µg/kg From PDN100 Prenatal exposure: normal preference for females Increased latency to genital sniffing
Dawley rat mating to weaning and intromission
Postnatal: pups from Postnatal exposure: increased number of intromissions
the vehicle group
fostered by BPA-
exposed mothers

42 Wistar rat GD0 to PND21 Oral BPA: 0.1 mg/L (30 µg/kg/day), 11–12 weeks No effect on the number of mounts, mating duration or latency to ejaculation Neither treatment affected the SDN-POA volume
1 mg/L (300 µg/kg/day) Diminution of intromission rate (number intromissions/number mounts ×100) at No effects on LH, FSH or testosterone levels
Trans-RVT: 5 mg/L (1,500 µg/kg/ BPA 0.1 and RVT
day)
DES: 50 µg/L (6.5 µg/kg/day)

Endocrine Disruption of Sexual Behavior


45 Sprague- PND23 to PND30 Oral BPA: 40 µg/kg From PND90 BPA exposure: no effect on the number and intromission ratio. Reduced latency to Reduced testosterone levels at PND37 and PND105 for
Dawley rat EE: 0.4 µg/kg intromission BPA, at PND 37 for EE
EE exposure: reduced duration of genital sniffing, increased number of intromission No effects on estradiol levels
and intromission ratio. Reduced latency to mount and intromit and increased
duration of the refractory period (time between ejaculation and subsequent
mounting)

46 Long- GD7 to PND14 Oral BPA: 5, 50, 500 µg/kg/day or 90–120 days Day 1: increased number of intromissions, reduced time between ejaculation and
Evans rat 5 mg/kg/day subsequent mounting, and reduced latency for the 5 mg group
Day 4: reduced number of intromissions in BPA–5 µg and BPA–50 µg. Increased
latency to intromission and ejaculation at BPA-50 µg, 500 µg and 5 mg. Increased
time between ejaculation and subsequent mounting at 50 µg; copulation more
efficient in controls and BPA–5 mg >BPA–50 µg and 5 mg >BPA–500 µg

65 Deer mice 2 weeks before Oral (diet) BPA: 50 mg/kg diet Since PND80 BPA-exposed males less attractive than controls in mate choice test
mating until
weaning

61 CF-1 mice GD10 to PND9 Oral BPA: 17.5, 175 or 1,750 µg/day in PND 90 No effect on latencies or number of mounts No effects on urinary levels of testosterone and estradiol
high phytoestrogen diet Fewer intromissions in BPA–17.5 and BPA–175 groups
Fewer ejaculation in BPA–17.5 group

63 C57BL/6 GD15 to PND 21 Oral BPA: 50 µg/kg/day, 5 mg/kg/day 8 weeks No effect on latency to mount, thrust, intromit, or ejaculate. Unaffected number of Unaffected number of calbindin neurons in the MPOA
mice (gavage) mounts, intromissions and thrusts and kisspeptin neurons in the AVPV
Reduced lordosis quotient at BPA–5 in gonadectomized males primed with estradiol No effect on testosterone levels
and progesterone

48 Rat PND0 to PND10 or Oral (diet) Coumestrol: 100 µg/g Reduced mount and ejaculation frequency; extended latency to mount and ejaculate No effect on testosterone levels
during PND21 of
lactational period

DOI: 10.1159/000494558
49 Sprague- PND0 to PND10 Oral (diet) Coumestrol 100 ppm PND 174–239 Reduced number of mounts (–70%) and ejaculations (–86%) No effect on testosterone levels at PND10, or at 7 or 11
Dawley rat months of age

51 Rat Neonatal Genistein: 20 µg Increased sexual activity


Genistein+benzopyrene 20 µg

50 Long- 2 weeks before Oral (diet) Genistein: 5 mg/kg diet (low) or 70 days Reduced percentage of mounts, intromissions and ejaculations at 5 mg and Reduced testosterone levels at 5 and 300 mg/kg
Evans rat mating until 300 mg/kg diet (high) percentage ejaculation at 300 mg

Neuroendocrinology 2018;107:400–416
weaning

47 Sprague- PND1 to PND22 Oral (water) Resveratrol: 5, 50 or 100 µM PND 132–140 Decreased mount frequency at all doses Reduced SDN-POA volume at all doses; increased AVPV
Dawley rat No effects on frequency, duration or latency of other behaviors (autogrooming and volume at 5 and 50 µM
anogenital investigation) Decreased testosterone levels for all resveratrol doses

41 Wistar rat PND1 to PND5 Subcutaneous Genistein:150 µg Sexual behavior: genistein reduced latency to intromission and ejaculation and
Coumestrol: 5 µg increased frequency of ejaculation; coumestrol induced opposite effects; EB: no
EB: 1 µg ejaculation, increased latency to mount and intromit
Partner preference: no preference of males exposed to coumestrol and genistein in
no-contact test; EB: preference for males; In contact test, genistein-exposed group
preferred receptive females by contrast to coumestrol- and EB-exposed groups

64 C57BL/6 2 weeks before Oral (diet) Genistein: 5 or 300 mg/kg of diet 70 days No effect on latency to mount and intromit, number or duration of mounts and No effect on testosterone levels (tendency to decrease at
mice mating until intromissions the higher dose)
weaning

62 CF-1 mice Gestation Oral (diet) Normal or phytoestrogen-free diet 43 days No effect on mounts or latency to plug

405
Table 2 (continued)

406
Ref. Species Exposure period Route Doses Age at analyses Behavioral analyses Neuroanatomical and hormonal analyses

101 Swiss mice GD10 to PND40 Oral EE: 0.1 µg/kg/day or 1 mg/kg/day 8 weeks Reduced latency to mount and intromit for EE–1 µg (2nd test) and 0.1 and 1 µg (3rd Increased number of GnRH neurons in the medial
(drinking) (F1, F2, F3 test) for F1–F4 generations; increased frequency of intromission for F2 generation septum at 0.1 µg; reduced GnRH area in the median
and F4) (2nd test) for the two doses and F4 at 0.1 µg and 3rd test for F1–F4 eminence at 0.1 and 1 µg. No effect on kisspeptin neurons
in the hypothalamus at F1, F3 and F4. Increased number
of calbindin neurons in the MPOA of males F1 at 1 µg
(F3, F4) at 0.1 and 1 µg
No effect on testosterone levels for all generations

43 Sprague- PND1 to PND5 Subcutaneous NP: 500 mg/kg PND84 Increased number of mounts for estradiol Unchanged levels of testosterone levels
Dawley rat Estradiol: 2 mg/kg No effect on the number of intromissions in NP or E2 groups

52 Long- PND21 to 11 Oral MXC:200–300–400 mg/kg/day 11 months of The latency to ejaculate was reduced at all doses Serum levels of LH, prolactin and testosterone were not
Evans rat months of age (gavage) age The latency to mount and number of intromissions were not affected affected; heterogeneity among group males were,
however, observed with infertile or sub-fertile males
exhibiting low testosterone levels

60 ICR mice GD5 to GD7 Subcutaneous MXC: 1 mg/0.05 ml (33 mg/kg) 4 months of Males of the MXC-treated group spent less time near the partition after a female in Female introduction in the cage increased plasma
age estrus was introduced into the cage by comparison to untreated or oil-treated groups; testosterone levels in controls, but not in MXC-treated
this suggested altered sexual arousal or motivation in the MXC-treated group groups

Anti-androgenic compounds
93 Long- GD14 to PND3 Oral Vinclozolin (VZ): 1,5, 3, 6 or 12 Reduced erection for all groups; no effect on latency to the first penile reflex;

DOI: 10.1159/000494558
Evans rat (gavage) mg/kg increased seminal emission for VZ–12 group

97 Sprague- E8 to E14 IP injection of Vinclozolin: 100 mg/kg/day F3 generation: Control females prefer control males in partner preference test
Dawley rat F0 females 3–4 months

96 Sprague- GD3 to PND21 per os DEHP: 0, 375, 750, or 1,500 mg/ PND 105 Many DEHP exposed males were sexually inactive in the presence of receptive control
Dawley rat kg/day females; sexual inactivity did not correlate with abnormal male reproductive organs

Neuroendocrinology 2018;107:400–416
98 Wistar rat GD6 to PND60 Oral DEHP: 0.015, 0.045, 0.135, 0.405, 110 days: No effects on behavior or fertility Increased testosterone levels at 0.045, 0.405 and 405 mg/
(gavage) 1.215 mg/kg/day or 5, 15, 45, 135, fertility kg/day (2-fold increase in the 405 group)
405 mg/kg/day 130 days:
sexual behavior

94 Sprague- GD1 to PND21 Oral DEHP: 20–100–500 mg/kg/day Adulthood 80% controls, 90% DEHP-20 and -100 and 60% DEHP-500 ejaculated; increased No effect on testosterone levels
Dawley rat (gavage) latency to intromission and intromission number

95 Wistar- GD15 to PND21 Oral (diet) DBP: 20, 200, 2,000 and 20–21weeks Reduced number of mounts and intromissions et 40 ppm DINP and 480 ppm DEHA; No effects on testosterone or estradiol, LH and FSH levels
Imamichi 10,000 ppm no effect of DBP. Reduced number of ejaculations at 200, 2,000 ppm DBP, 40 ppm
rat DINP: 40, 400, 4,000, 20,000 ppm DINP, 48 and 12,000 ppm DEHA; increased number of ejaculations at 10,000 DBP.
DEHA: 480, 2,400, 12,000 ppm Reduced interval post-ejaculation at 10,000 DBP

Adult
Estrogenic compounds
66 Rat Adulthood: 14 days Subcutaneous BPA: 1 mg/rat/day No effect on male sexual behavior Reduced testosterone levels and increased LH levels
of treatment

63 C57BL/6J 8–12 weeks of age Oral BPA: 50 µg/kg/day, 5 mg/kg/day 12 weeks Normal olfactory preference No effect on AR- and ERα-expressing neurons in the
mice (gavage) Increased latency to mount, thrust, intromit and ejaculate in naive and sexually MeA and BNST at 50 µg and 5 mg, but increased number
experienced males at BPA–50 in MPOA at 50 µg and 5 mg
Reduced number of intromissions at BPA–50 No effect on testosterone levels

68 Sprague- Adult: 7-10 days Ground corn- Adult Reduced number of mounts, intromissions, ejaculation, ejaculation latency and
Dawley rat cob bedding grooming frequency

67 Sprague- 7–12 weeks Oral THF-diol isomers from corncob Adulthood Increased latency to mount, intromit and ejaculate nonsignificant
Dawley rat (drinking) bedding: 2 µg/mL Reduced frequency of mount, intromission and grooming

69 Wistar rat 12 weeks of age for Subcutaneous Wet mesquite pod: 3.5 g/kg/day Tests at 10th, Increased latency to mount for Leucaena-, DAI- and E2-exposed groups Reduced testosterone levels at all treatments
50 days Wet plant (Leucaena leaves): 3.5 20th, 30th, Increased latency to intromit except for Leucaena-exposed group
g/kg/day 40th and 50th Increased latency to ejaculate for all groups with strong effect at 40th and 50th days of
Daidzein (DAI): 5 mg/kg/day days of test; increased number of mounts from 20th to 50th day
Genistein: 5 mg/kg/day treatment No effect on number of intromissions; diminished number of ejaculations per test
E2:2 30 µg/kg/day from 20th to 50th day

70 C57BL/6J 8 weeks Oral (diet) NP: 0.5, 5, 50 μg/kg/day 4 weeks later, Normal olfactory preference; increased emission of ultrasonic vocalizations at NP-5; Changes in the number of AR and ERα-immunoreactive
mice exposure increased number of mounts, intromissions and thrusts at NP-5; delayed ejaculation cells in the neural circuitry underlying sexual behavior
maintained No changes in testosterone levels, kisspeptin-
during analyses immunoreactivity in the POA and arcuate nucleus

Mhaouty-Kodja/Naulé/Capela
Anti-androgenic compounds
99 C57BL/6J 8 weeks Oral (diet) DEHP: 0.5, 5, 50 μg/kg/day 4 weeks later, Normal olfactory preference Unaffected colabelling of kisspeptin/AR or ERα in the
mice exposure Reduced emission of ultrasonic vocalizations and male attractiveness; delayed AVPV; reduced number of AR-immunoreactive
maintained initiation of mount and intromission behaviors 09:58 neurones in the POA, BNST and MeA; down-regulation
during Unaffected mating duration or number of mounts and thrusts of AR mRNAs levels in the POA; no effects on ERα
analyses Normal testosterone levels
AVPV, anteroventral periventricular nucleus; BNST, bed nucleus of stria terminalis; EB, estradiol benzoate; GD, gestational day; MeA, medial amygdala; MPOA, medial preoptic area; PND, postnatal day; POA, preoptic area; SDN, sexually dimorphic nucleus.
Table 3. Behavioral studies in females

Ref. Species Period of exposure Route Doses Age at analyses Behavioral analyses Neuroanatomical and hormonal analyses

Developmental
Estrogenic compounds
73 Sprague- GD11–PND20 Oral (gavage) BPA: 3.2–32–320 mg/kg/day PND10 for the neuroanatomical study No modification of lordosis behavior of OVX+E2/P adult BPA did not modify the volume of the SDN-POA at PND10
Dawley rat DES: 15 μg/kg/day 6 months of age for the behavioral analysis females DES increased SDN volume
(OVX+E2/P).

44 Sprague- Prenatal: Oral (pipette) BPA: 40 μg/kg Adult (PND100) No modification of agonistic behaviors. In proestrus
Dawley rat GD0–PND1 Intact with following of the cycle females, lordosis quotient and proceptive behaviors were
Postnatal: not modified
PND1–PND21 Increased lordosis frequency in BPA-treated proestrus
females

42 Wistar rat GD0–PND21 Oral BPA: 0.1 mg/L Adult (11-12 weeks, OVX+E2/P) BPA: No modification of lordosis quotient or rejection Neither treatment affected the SDN-POA volume
(30 µg/kg/day) – 1 mg/L score in BPA-treated females No effect on LH, FSH or prolactin levels
(300 µg/kg/day) DES and resveratrol: decreased lordosis quotient and Decreased estradiol levels by DES, but not BPA or RVT
DES: 50 µg/L (6.5 µg/kg/day) increased rejection score in DES-treated females
Trans-RVT: 5 mg/L
(1,500 µg/kg/day)

Endocrine Disruption of Sexual Behavior


71 Long- PND0–PND3 Subcutaneous BPA: 50 μg/kg–50 mg/kg Adult (OVX+E2/P) Lordosis quotient abolished in EB-treated females, No effect on hypothalamic GnRH activation
Evans rat PPT: 1 mg/kg unmodified in the other groups
EB: 25 μg

75 Wistar rat PND1–PND7 Subcutaneous BPA: 0.05–20 mg/kg Adult (PND100, No modification of lordosis behavior Reduced ERα expression in the medial preoptic and
OVX+E2/P) Reduced proceptive behaviors in BPA-treated females ventromedial nuclei
No modification of PR expression in these regions

74 Long- GD7–PND18 Oral (gavage) BPA: 2–20–200 μg/kg Adult (OVX+E2/P) Lordosis behavior unchanged following exposure to BPA
Evans rat EE: 15–50 μg/kg/day Abolished behavior by both doses of EE

46 Long- GD7–PND14 Oral (pipette) BPA: 5–50–500 μg/kg–5 mg/kg Adult (intact, behavior during behavioral No modification of sexual receptivity or proceptive
Evans rat estrous) behaviors

72 Long- PND0–PND3 Subcutaneous BPA: 50 μg/kg–50 mg/kg Adult (OVX+E2/P) Proceptive behavior abolished by EB-treated females, Increased number of oxytocin neurons within the
Evans rat PPT: 1 mg/kg unaffected in the other treatment groups paraventricular nucleus in all treatment groups
EB: 25 μg The number of ERα neurons reduced in the medial preoptic
area by EB and PPT, but not by BPA

76 C57BL6/J GD15–PND21 Oral (gavage) BPA: 0.05–5 mg/kg Adult (OVX+E2/P) Increased lordosis quotient in naive females of the BPA– No effect on the number of calbindin-positive cells
mice 0.05 group
No behavioral effects of BPA–5 Increased kisspeptin cell number in the preoptic periventricular
Ovariectomized females supplemented with testosterone nucleus of the rostral periventricular area of the third ventricle
showed comparable number of mounts and thrusts No modification of the kisspeptin fiber density in the arcuate
between vehicle and BPA-groups nucleus. No modification of the number of GnRH neurons in
the medial preoptic area
No modifications in ERα-immunoreactivity in the MeA, BNST
or POA
Increased estradiol levels during diestrus at BPA–0.05 No

DOI: 10.1159/000494558
significant effect on LH levels

51 Rat Neonatal Genistein: 20 mg Genistein significantly increased the lordosis quotient of


females

78 Wistar rats PND1–PND5 Subcutaneous Genistein: 1 mg Adult (OVX+E2) Reduced lordosis quotient in genistein-treated females
Daidzein: 1 mg compared to controls, but higher than that obtained for
17β-E2: 100 μg E2-treated group

Neuroendocrinology 2018;107:400–416
No modification of lordosis quotient in daidzein-treated
females

79 Wistar rat PND5 Subcutaneous Coumestrol: 1–3 mg Adult (OVX+E2) Decreased lordosis quotient of females treated with 3 mg
Genistein: 1 mg coumestrol or E2
E2: 1 mg No modification of lordosis quotient in females treated
with genistein and 1 mg coumestrol

47 Sprague- PND1–PND22 Daily drinking RVT: 5–50–100 μM Adult (OVX+E2/P) No modification of sexual receptivity or proceptive No modification of SDN-POA or AVPV volume
Dawley rat solution behaviors

80 C57BL6/J GD0– Diet Low and high phytoestrogen diet Adult (OVX+E2/P) Decreased lordosis behavior of the aromatase knockout
mice, adulthood females raised on a diet high in phytoestrogens compared
aromatase to both control and aromatase knockout females treated
knockout with a low phytoestrogens diet
mice

77 CF1 mice GD0– Diet Regular laboratory chow or PND43 Strong decrease of lordosis behavior in females fed with a

407
adulthood phytoestrogen-free diet diet deficient in phytoestrogens compared to animals fed
with a regular diet (containing phytoestrogens)
Table 3 (continued)

408
Ref. Species Period of exposure Route Doses Age at analyses Behavioral analyses Neuroanatomical and hormonal analyses

81 Sprague- PND1, PND3, Subcutaneous MXC: 1 or 2 mg/rat Adult PND75–90 E2 lowered the LQ; both E2 and the higher dose of MXC
Dawley rat PND5 E2: 10 μg/rat (OVX+E2/P) altered the average lordosis amplitude and increased
rejection behavior

Anti-androgenic compounds
95 Wistar- GD15–PND21 Diet DBP: 20–200–2,000–10,000 ppm Adult (day of proestrus) Decreased lordosis behavior in females perinatally No effect on testosterone, estradiol, LH or FSH levels
Imamichi DINP: 40–400–4,000–20,000 ppm exposed to DBP, DINP, DEHA at all the doses
rat DEHA: 480–2,400–12,000 ppm

Adult
Estrogenic compounds
82 Wistar rat Adult OVX Subcutaneous BPA: 10 mg (~40 mg/kg/day) Adult (OVX±P) Ovariectomized (OVX) rats that were primed with 10 mg Like estradiol, injection of BPA at 100 μg, 1,000 μg and 10 mg
(acute) E2: 10 μg (40 μg/kg/day) of BPA, followed by 1 mg of progesterone, displayed doses increased the number of progesterone immunoreactive
rejection behavior, but not lordosis unlike OVX rats cells in the preoptic area and the ventromedial hypothalamic
primed with E2 and progesterone nucleus

89 Long- Adult (5 days Diet Genistein: 13 ppm Adult (OVX+P)±E2 Decreased lordosis quotient in estrogen- and Isoflavone diet increased ERβ mRNA expression in the
Evans rat before testing) Daidzein: 33 ppm progesterone-primed females paraventricular nucleus, by contrast to estradiol which
decreased it

DOI: 10.1159/000494558
91 Long- Adult OVX Diet Genistein: 100–500 ppm Adult (OVX+P)±E2 No modification of lordosis quotient by genistein No effect of genistein treatment on oxytocin receptor density in
Evans rat treatment in females primed or not by E2 the ventromedial hypothalamic nucleus
Genistein treatment increased ERβ mRNA expression in the
paraventricular nucleus of the hypothalamus contrary to
17β-estradiol treatment which decreased it

68 Sprague- Adult OVX Ground Adult (OVX+E2/P) Strong decrease of lordosis behavior in females housed
Dawley rat corncob on a corncob bedding

Neuroendocrinology 2018;107:400–416
bedding

86 Sprague- Adult OVX Subcutaneous RVT: 10–100–1,000 μg Adult (OVX+P) RVT treatment did not induce lordosis behavior in Increased E2 levels at resveratrol 1,000 µg and 10 µg EB
Dawley rat (2 days) females

Adult intact: Daily drinking RVT: 100 mM Adult (OVX+E2/P) RVT delayed rejection behavior and lowered the
7 days water frequency of anogenital investigation of stimulus male
preexposure
before OVX

87 Long- Adult OVX Oral Diet: soy isoflavones including Adult (OVX+P 4–5 h before testing) When administered with P alone, diet or tamoxifen had
Evans rat Diet: 5 days (diet±soy) genistein and daidzein no effect
EB: 48 h before Sc (EB, EB: 10 μg/0.1 mL In females primed with both E2 and P, diet or tamoxifen
test tamoxifen) Tamoxifen: 5 mg capsule implant reduced LQ, and hopping and darting rate
Tamoxifen: Soy diet also reduced proceptive behavior
14 days before test

84 Sprague- Adult OVX Oral (gavage) Ferutinin: 0.5 mg/kg Adult (OVX+P)±E2 Ferutinin given alone induced a lordotic response in Ferutinin treatment increased hypothalamic ERα expression
Dawley rat (2, 3, and 4 weeks) ovariectomized rats but failed to affect proceptivity when administered alone, as estradiol did, but decreased the
In combination with estradiol, ferutinin reduced response to estradiol when administered in combination
estrogen-induced receptivity and proceptivity

85 Sprague- Adult OVX Oral (gavage) Ferutinin: 0.2–0.5 mg/kg Adult (OVX+P) Ferutinin given alone at both doses induced preference
Dawley rat (4 weeks) for males, increased lordosis quotient and proceptive
behavior in ovariectomized females like EB

90 Sprague- Adult OVX Oral (gavage) Humulus lupulus extract: Adult (OVX+E2/P) Humulus lupulus extract treatment induced preference
Dawley rat (acute) 5–10–25 mg/kg for males, increased proceptive behaviors at the highest
dose, without affecting lordosis quotient

83 Wistar rat Intact and Subcutaneous Mesquite pod extract (MPE): Adult (intact or OVX) Phytoestrogen and MPE treatment of intact females
OVX adult 4 g/kg decreased the percentage of females exhibiting lordosis
(30 days) Genistein: 1.6 mg/kg by comparison to control and E2 groups
Daidzein: 1.6 mg/kg Genistein and daidzein treatment of OVX females
E2: 40 μg/kg induced lordosis behavior, but lesser than in E2-treated
animals

88 Long- Adult OVX Intramuscular DES: 30–90 μg Adult (OVX+P) Treatment with 30 or 90 μg of DES activated lordosis

Mhaouty-Kodja/Naulé/Capela
Evans rat (27 days) injection (approximately 106–317 μg/kg behavior
respectively)

92 Long- Adult OVX Oral MXC: 200 mg/kg/day Adult (OVX±P) All P-injected rats displayed proceptive (darting) and MXC (400 mg/kg/day) did not affect serum levels of FSH and
Evans rat (24 days) lordosis behaviors by contrast to oil-injected animals LH in intact or OVX females

AVPV, anteroventral periventricular nucleus; EB, estradiol benzoate; MPOA, medial preoptic area; OVX, ovariectomized; P, progesterone; PPT, ERα-selective agonist 4,40,400-(4-propyl-[1H]pyrazole-1,3,5-triyl)trisphenol; SDN, sexually dimorphic nucleus.
ization of behavior [63]. When given a choice between ference in mounting or thrusting behaviors relative to the
control and BPA-exposed partners, female mice pre- vehicle-treated group. Most neuroanatomical studies
ferred controls over BPA-exposed males [65]. BPA had have reported no effects on the SDN, corresponding cal-
no neuroanatomical effects on the number of calbindin- bindin cells [42, 53, 54, 59, 73, 76], and TH neurons [56].
immunoreactive neurons in the preoptic area or kiss- By contrast, an increase in the number of kisspeptin neu-
peptin cells in the AVPV [63]. rons in the AVPV was observed in the study reporting an
Thus, exposure to BPA or phytoestrogens induces increase in lordosis behavior following BPA exposure in
changes in the neural circuitry underlying sexual behav- mice [76]. Changes in the number of tyrosine hydroxylase
ior in rats. Little if any change was observed in mice, sug- neurons have also been reported in rats [54, 56]. These
gesting species differences, although more studies are re- data strongly suggest that BPA does not trigger masculin-
quired to confirm these observations. One key observa- ization in the female neural circuitry during the highly
tion is that most of these studies were performed at sensitive perinatal period, despite its lack of binding to
relatively low doses, equivalent to or lower than the refer- α-fetoprotein. Instead, it seems to potentiate estrogen-in-
ence doses established for these molecules. duced increases in sexual behavior and kisspeptin expres-
Adult Exposure. Exposure to BPA for 2 weeks has no sion, both of which occur later during the postnatal/pre-
effect on male rats [66]. By contrast, longer periods of ex- pubertal period in females.
posure of male mice to BPA increased the times to mount- By contrast to BPA, prenatal or postnatal exposure to
ing, intromission and ejaculation, and decreased the phytoestrogens mostly affected adult female behavior, in
numbers of intromissions and thrusts in both naïve and both rats and mice, as shown in Table 3 [47, 51, 62, 77–
sexually experienced male mice [63]. Similar differences 79]. Similar results have been reported for methoxychlor
between short- and long-term exposures were observed [80]. More behavioral and neuroanatomical analyses in
for corncob bedding and derived tetrahydrofuran-diols the same context would be required to determine wheth-
[67, 68]. Changes in behavior were also reported for rats er the observed alterations are linked to changes in the
subjected to long-term phytoestrogen exposure [69] and organization of sexually dimorphic populations. Indeed,
NP [70]. A non-monotonic dose response was observed no effects of resveratrol on SDN-preoptic area or AVPV
for NP since increased emission of ultrasonic vocaliza- volume were found in a study on female rats in which no
tions, reduced number of mounts, intromissions and change in behavior was detected [47]. Two other studies
thrust as well as delayed ejaculation were observed only reporting an absence of effect on these two hypothalamic
at the dose of 5 µg/kg/day but not at lower and higher regions did not assess female behaviors [56, 64].
doses. Despite the small number of studies performed, Adult Exposure. The acute exposure of ovariectomized
the results obtained all suggest that long-term exposure rats to BPA did not trigger lordosis behavior when fe-
in adults affects male sexual behavior. males were primed with progesterone only, or with pro-
gesterone plus E2, despite an increase in the number of
Effects of Exposure in Females progesterone-immunoreactive cells in the preoptic area
Prenatal/Postnatal Exposure. Female sexual behavior and ventromedial hypothalamus [81]. An analysis of 10
has mostly been investigated in rats (13 studies vs. only 3 sets of findings for adult exposure to phytoestrogens sug-
in mice). BPA exposure had no significant effect on lor- gested that behavioral responses differ between initial
dosis quotient or proceptive behavior in a number of hormonal environments. Hence, genistein and daidzein,
studies [42, 44, 46, 71–74]. However, pre- or postnatal and ferutinin administered to ovariectomized females
exposure to BPA decreased proceptive behavior in one primed only with progesterone either induced [82–84] or
study [75] and increased the frequency of lordosis in an- had no effect on female behaviors [85, 86]. In similar ex-
other [44]. In one study on mice, exposure to BPA at a perimental conditions, DES treatment induced lordosis
dose of 50 µg/kg/day resulted in a significant increase in behavior [87], whereas the anti-estrogen tamoxifen had
lordosis quotient in naïve but not sexually experienced no effect [86]. By contrast, as for tamoxifen [86], exposure
females, whereas a dose 100 times higher had no effect to genistein and daidzein, corncob bedding, ferutinin,
[76]. Olfactory preferences were unchanged, as BPA-ex- resveratrol or Humulus lupulus reduced lordosis behav-
posed females displayed normal levels of interest in che- ior in 5 studies in which females were either intact or
moinvestigating males relative to females [76]. Following ovariectomized and on E2 supplementation [68, 82, 83,
ovariectomy and testosterone treatment, females dis- 86, 88]. In 3 other studies, however, genistein, resveratrol
played no masculinization of their behavior, with no dif- and H. lupulus had no effect on lordosis behavior but al-

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 409


DOI: 10.1159/000494558
Fig. 1. Exposure to endocrine disrupting compounds (EDC) can interfere with the hormonal dependent regulation of the neural
alter sexual behavior through indirect (A) and/or direct pathways structures underlying sexual behavior in males and females. B Sim-
(B). A Kisspeptin neurons (green cells) of the preoptic area (POA) plified scheme of the male neural circuitry including the olfactory
and arcuate nucleus (ARC) activate synthesis and liberation of bulb (OB), medial amygdala (MeA), bed nucleus of stria termina-
GnRH, which stimulates gonadotropin hormone (LH, FSH) lib- lis (BNST), and preoptic area (POA). EDC can also directly target
eration, and consequently gonadal synthesis and liberation of sex these neural structures, to disrupt hormonal signaling pathways
steroid hormones. Estradiol and testosterone exert in turn a feed- necessary to elicit male sexual behavior.
back on this gonadotropic axis. Disruption of this axis by EDC may

tered other components of behavior, such as the frequen- 96]. Exposure to phthalates led to sexual inactivity [95],
cy of anogenital investigation or proceptive behavior [85, increases in the time to sexual behaviors and a decrease
89, 90]. In the absence of E2, exposure to methoxychlor in the frequency of these behaviors in rats [93, 94]. Only
also induced proceptive and lordosis behavior in females the study by Andrade et al. [97] reported an absence of
after progesterone injection [91]. These data suggest that effects for low or high doses of DEHP. One neuroana-
phytoestrogens and methoxychlor have estrogenic activ- tomical study reported no effect on SDN-preoptic area
ity in the absence of endogenous estrogens, but anti-es- volume of exposure to high doses of DINP (diisononyl
trogenic effects in the presence of endogenous or supple- phthalate) [58]. One recent study addressed the effects of
mented estrogens, particularly for phytoestrogens. adult exposure to low doses of DEHP on male sexual be-
havior. Such exposure decreased ultrasonic vocalizations,
Effects of Developmental or Adult Exposure to Anti- reduced female attraction and delayed the initiation of
Androgenic Compounds mating, without affecting olfactory preference for recep-
Five of the 6 studies addressing the developmental ef- tive females [98].
fects of vinclozolin and phthalates at relatively high doses Only one study has measured the effects of develop-
on components of sexual behavior in males reported be- mental exposure to phthalates on female sexual behavior.
havioral changes [92–96]. Diminished erections and in- It reported a decrease in the lordosis behavior of adult
creased seminal emission were observed, together with females in proestrus following exposure to DBP (dibutyl
reduced attractiveness, in rats exposed to vinclozolin [92, phthalate), DINP, and DEHA (di-[2-ethylhexyl] adipate)

410 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558
[94]. The effects of adult exposure to anti-androgenic havior does not, therefore, seem to be systematically
compounds have not been studied. linked to changes in hormone levels. However, these find-
More studies of developmental and adult exposure to ings do not rule out the possibility of transient changes in
anti-androgenic molecules are therefore required, in both hormone levels during the exposure period, particularly
males and females, particularly for doses close to the es- if exposure occurs during development, thereby inducing
timated environmental exposure. long-term changes to the neural processes or hormonal
signaling pathways underlying male behavior. Interest-
ingly, long-term changes in the levels of the sex steroid
Mode of Action receptors, ER and PR, have been reported to be induced
by prenatal and postnatal exposure to BPA, DES or me-
Endocrine disruption of sexual behavior may occur thoxychlor in the whole hypothalamus or more specifi-
through indirect or direct pathways (Fig. 1). The indirect cally hypothalamic areas (preoptic area, ventromedial hy-
pathway involves changes in the levels of gonadal hor- pothalamus), bed nucleus of stria terminalis or medial
mones, which then affect the organization or activation amygdala of rats [100–102], and mice [103, 104]. How-
of neural structures involved in the expression of behav- ever, no effects of such exposure were reported in 2 other
ior. The circulating levels of gonadal hormones may be mouse studies in the same brain areas [105, 106]. Chang-
modified by dysregulation of the hypothalamic GnRH es in total hypothalamic gene expression were observed
system and upstream regulators including kisspeptin for BPA and EE, or for neuropeptides involved in social
neurons and/or the disruption of pituitary function or behavior, such as AVP in the medial amygdala and lat-
gonadal steroidogenesis. Exposure to EDC can also di- eral septum and oxytocin in the hypothalamus [105, 107].
rectly affect the neural structures underlying sexual be- In females, sexual behavior is generally analyzed in
havior, by interfering with the neural synthesis of hor- ovariectomized animals supplemented with E2 and pro-
mones, such as the aromatization of testosterone into E2, gesterone. The behavioral changes observed under these
binding to and activation of sex steroid receptors, or the experimental conditions cannot be due to modifications
expression of these receptors. Modes of action involving of the hypothalamic-pituitary-gonadal axis. However, it
agonism or antagonism are more difficult to demonstrate is important to analyze hormone levels in intact females
in vivo. They can be suggested or supported by parallel in parallel, because they can provide information about
comparisons with the effects of positive controls, such as the integrity of the hypothalamic-pituitary-gonadal axis
estrogens or their analogs, or anti-androgens, such as flu- and its effects on the activation of behavior under physi-
tamide, although such comparisons may be limited for ological conditions. It is not possible to draw any firm
the reasons described in the section “Effects of Develop- conclusions from the small number of studies assessing
mental versus Adult Exposure to Estrogenic Com- hormone levels in intact females. An absence of effect [42,
pounds”. Analyses of the modes of action of various com- 105, 108] or increases in hormone levels have been re-
pounds are reported in Tables 2 and 3 when performed ported following exposure to BPA [76]. Lower levels of
in parallel to behavioral studies; otherwise, they are estradiol and LH were observed in rats exposed to me-
presented separately in online supplementary Tables 1 thoxychlor [109] or BPA [110], whereas phthalates had
and 2. no effect [94]. By contrast, changes in sex steroid receptor
levels have been reported in a larger number of studies.
Developmental Exposure Most of the studies on BPA presented in online supple-
Three of 12 studies on male rats reporting develop- mentary Table 2 dealt with the effects of exposure on ERα
mental effects of EDC on behaviors described a decrease and/or ERβ levels in neural structures underlying neuro-
in adult hormone levels following exposure to BPA [45], endocrine and behavioral reproductive functions such as
genistein [50], and resveratrol [47]. The other 9 studies the medial amygdala, the bed nucleus of stria terminalis,
found no effects on adult levels of testosterone, E2, and the preoptic area and its subdivisions, the ventromedial
gonadotropins [42, 43, 48, 49, 52, 93, 94]. In mice, no hypothalamus or the arcuate nucleus in rats and mice
change in hormone levels was observed following expo- [100, 101, 103, 105–109, 111–113]. This was probably
sure to BPA or EE [61, 63, 99]. Exposure to methoxychlor motivated by the facts that these receptors are the major
did not change basal hormone levels but prevented the mediators of estrogen-induced genomic regulation, and
increase in testosterone levels following exposure to fe- effects of EDC exposure on their neural levels can inter-
males [60]. The observed impairment of male sexual be- fere with of the expression of reproduction behaviors.

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 411


DOI: 10.1159/000494558
The differences observed in the effects induced may re- Summary and Conclusions
flect differences in the doses of BPA used, the period of
exposure, and the area of the brain analyzed. The few Several observations can be made based on the data
studies to have assessed ER expression in parallel to be- concerning the impact of EDC on sexual behavior re-
haviors reported lower levels of ERα expression in the viewed here. In general, sexual behavior appears to be
preoptic and ventromedial nuclei of females with low lev- highly sensitive to EDC, and can be added to the end-
els of proceptive behavior [75], or no change in the num- points generally used in assessments of the risks of ex-
ber of ERα-immunoreactive neurons in the medial amyg- posure to EDC and their potential impact on reproduc-
dala, bed nucleus of stria terminalis and preoptic area of tion. Like other reproductive endpoints, the behavioral
animals with normal or higher levels of proceptive behav- effects induced by exposure to EDC depend on the pe-
ior [72, 76]. Exposure to methoxychlor has also been riod of exposure. The developmental and pubertal stag-
shown to trigger changes in ER and PR levels in the pre- es are particularly vulnerable due to the organizational
optic area [102, 110]. Some studies pointed out the in- effects of hormones during these periods, but exposure
volvement of long-lasting epigenetic modifications in in adults may also have effects, the doses used in several
such changes. Epigenetic gene regulation includes DNA studies having elicited effects at doses below the refer-
methylation at CpG sites, post-translational histone ence dose. Effects may also depend on the sex of the an-
modifications such as methylation or acetylation, and imals.
microRNAs. A previous study showed that changes in- The amounts of data published differ considerably be-
duced by BPA exposure in the expression levels of ERα in tween EDC compounds. The most frequently investigat-
the hypothalamus are associated with modifications in ed compounds are estrogen-like molecules, such as BPA
DNA methylation in the promoter region of this gene and phytoestrogens. However, published studies have not
[103]. Similar observations were made in the preoptic addressed all possible periods of exposure in both sexes.
area of females exposed to methoxychlor [110]. These Developmental exposure to BPA or phytoestrogens has
findings are of particular interest given the role of ER in been extensively studied in both males and females, but
the expression of female sexual behavior. Only a few stud- adult exposure to phytoestrogens has been studied only
ies have monitored the effects of EDC exposure on other in females. Much more studies are required for other es-
targets, such as neuropeptides essential for the expression trogenic or anti-androgenic compounds, for which fewer
of reproductive behaviors [107, 114]. data are available, regardless of the exposure period con-
sidered.
Adult Exposure Another interesting observation is that changes in the
In male rats, changes in male sexual behavior follow- level of expression of sex steroid receptors (ER, AR, PR)
ing exposure to phytoestrogens were associated with low- have been widely documented in both males and females.
er levels of testosterone [69]. In mice, the impairment of This finding strongly suggests that EDC may act, at least
sexual behavior following chronic adult exposure to BPA, partly, through changes in the neural signaling pathways
NP or DEHP was associated with unchanged hormonal underlying sexual behavior, either directly through epi-
levels and integrity of the hypothalamic-pituitary-gonad- genetic modifications to the ERα promoter, as reported
al axis, together with changes in the level of sex steroid for developmental exposure to BPA [103], or through as
receptor expression [63, 70, 98]. In particular, DEHP yet unidentified pathways, as for the down-regulation of
down-regulated AR protein and mRNA levels in the neu- AR induced by the exposure of adults to DEHP [98].
ral circuitry involved in sexual behavior [98], while both Finally, although caution is required when extrapolat-
the numbers of AR and ERα-immunoreactive cells were ing findings from rodents to other species, these altered
affected by NP exposure [70]. processes may be of considerable relevance in both hu-
The exposure of adult females to resveratrol or BPA mans and wildlife. The regulation of libido and erectile
increased E2 levels in both rats and mice [85, 115]. Inter- function or reproductive behaviors by sex steroid hor-
estingly, regardless of its behavioral effects, exposure to mones at the neural level is highly conserved across spe-
phytoestrogens or BPA increased the levels of ERα, ERβ cies. In this context, human studies have reported an as-
or PR expression in the preoptic area, the ventromedial sociation between a decrease in sexual activity and expo-
hypothalamus or the paraventricular nucleus [81, 83, 85, sure to environmental doses of DEHP and BPA [117,
88, 90, 116]. 118], but the mechanisms underlying these effects remain
unclear.

412 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558
References
  1 Bean NJ. Olfactory and vomeronasal media- 17 Simerly RB, Swanson LW, Gorski RA. The 30 Shughrue PJ, Lane MV, Merchenthaler I.
tion of ultrasonic vocalizations in male mice. distribution of monoaminergic cells and fi- Comparative distribution of estrogen recep-
Physiol Behav. 1982 Jan;28(1):31–7. bers in a periventricular preoptic nucleus in- tor-alpha and -beta mRNA in the rat central
  2 Dizinno G, Whitney G. Androgen influence volved in the control of gonadotropin release: nervous system. J Comp Neurol. 1997 Dec;
on male mouse ultrasounds during court- immunohistochemical evidence for a dopa- 388(4):507–25.
ship. Horm Behav. 1977 Apr;8(2):188–92. minergic sexual dimorphism. Brain Res. 1985 31 Micevych PE, Meisel RL. Integrating Neural
  3 Nyby J, Wysocki CJ, Whitney G, Dizinno G. Mar;330(1):55–64. Circuits Controlling Female Sexual Behavior.
Pheromonal regulation of male mouse ultra- 18 Nugent BM, Wright CL, Shetty AC, Hodes Front Syst Neurosci. 2017 Jun;11:42.
sonic courtship (Mus musculus). Anim Be- GE, Lenz KM, Mahurkar A, et al. Brain femi- 32 Mhaouty-Kodja S. Role of the androgen re-
hav. 1977 May;25(2):333–41. nization requires active repression of mascu- ceptor in the central nervous system. Mol
  4 Holy TE, Guo Z. Ultrasonic songs of male linization via DNA methylation. Nat Neuro- Cell Endocrinol. 2018 Apr;465:103–12.
mice. PLoS Biol. 2005 Dec;3(12):e386. sci. 2015 May;18(5):690–7. 33 Ogawa S, Lubahn DB, Korach KS, Pfaff DW.
  5 Hull EM, Dominguez JM. Sexual behavior in 19 Kow LM, Pfaff DW. Physiology of somato- Behavioral effects of estrogen receptor gene
male rodents. Horm Behav. 2007 Jun; 52(1): sensory and estrogenic control over the lor- disruption in male mice. Proc Natl Acad Sci
45–55. dosis reflex. Exp Brain Res. 1981;Suppl 3: USA. 1997 Feb;94(4):1476–81.
 6 Beach FA. Sexual attractivity, proceptivity, 262–73. 34 Ogawa S, Chan J, Chester AE, Gustafsson JA,
and receptivity in female mammals. Horm 20 Bakker J, De Mees C, Douhard Q, Balthazart Korach KS, Pfaff DW. Survival of reproduc-
Behav. 1976 Mar;7(1):105–38. J, Gabant P, Szpirer J, et al. Alpha-fetoprotein tive behaviors in estrogen receptor beta gene-
  7 Erskine MS. Solicitation behavior in the es- protects the developing female mouse brain deficient (betaERKO) male and female mice.
trous female rat: a review. Horm Behav. 1989 from masculinization and defeminization Proc Natl Acad Sci USA. 1999 Oct; 96(22):
Dec;23(4):473–502. by estrogens. Nat Neurosci. 2006 Feb; 9(2): 12887–92.
  8 Harlan RE, Shivers BD, Pfaff DW. Lordosis as 220–6. 35 Antal MC, Krust A, Chambon P, Mark M.
a sexually dimorphic neural function. Prog 21 Schulz KM, Richardson HN, Zehr JL, Osetek Sterility and absence of histopathological de-
Brain Res. 1984;61:239–55. AJ, Menard TA, Sisk CL. Gonadal hormones fects in nonreproductive organs of a mouse
  9 World Health Organization. State of the sci- masculinize and defeminize reproductive be- ERbeta-null mutant. Proc Natl Acad Sci USA.
ence of endocrine disrupting chemicals. 2012 haviors during puberty in the male Syrian 2008 Feb;105(7):2433–8.
[cited 2016 Oct 24]. Available from: http:// hamster. Horm Behav. 2004 Apr; 45(4): 36 Antal MC, Petit-Demoulière B, Meziane H,
www.who.int/ceh/publications/endocrine/ 242–9. Chambon P, Krust A. Estrogen dependent
en/. 22 Ahmed EI, Zehr JL, Schulz KM, Lorenz BH, activation function of ERβ is essential for the
10 Gore AC, Holley AM, Crews D. Mate choice, DonCarlos LL, Sisk CL. Pubertal hormones sexual behavior of mouse females. Proc Natl
sexual selection, and endocrine-disrupting modulate the addition of new cells to sexu- Acad Sci USA. 2012 Nov;109(48):19822–7.
chemicals. Horm Behav. 2018 May;101:3–12. ally dimorphic brain regions. Nat Neurosci. 37 Ogawa S, Eng V, Taylor J, Lubahn DB, Ko-
11 Phoenix CH, Goy RW, Gerall AA, Young 2008 Sep;11(9):995–7. rach KS, Pfaff DW. Roles of estrogen recep-
WC. Organizing action of prenatally admin- 23 De Lorme KC, Schulz KM, Salas-Ramirez tor-alpha gene expression in reproduction-
istered testosterone propionate on the tissues KY, Sisk CL. Pubertal testosterone organizes related behaviors in female mice. Endocri-
mediating mating behavior in the female regional volume and neuronal number with- nology. 1998 Dec;139(12):5070–81.
guinea pig. Endocrinology. 1959 Sep; 65(3): in the medial amygdala of adult male Syrian 38 Naulé L, Robert V, Parmentier C, Martini M,
369–82. hamsters. Brain Res. 2012 Jun;1460:33–40. Keller M, Cohen-Solal M, et al. Delayed pu-
12 Morris JA, Jordan CL, Breedlove SM. Sexual 24 Brock O, Baum MJ, Bakker J. The develop- bertal onset and prepubertal Kiss1 expression
differentiation of the vertebrate nervous sys- ment of female sexual behavior requires pre- in female mice lacking central oestrogen re-
tem. Nat Neurosci. 2004 Oct;7(10):1034–9. pubertal estradiol. J Neurosci. 2011 Apr; ceptor beta. Hum Mol Genet. 2015 Dec;
13 Gilmore RF, Varnum MM, Forger NG. Ef- 31(15):5574–8. 24(25):7326–38.
fects of blocking developmental cell death on 25 Brock O, Bakker J. The two kisspeptin neuro- 39 Cheong RY, Porteous R, Chambon P,
sexually dimorphic calbindin cell groups in nal populations are differentially organized Abrahám I, Herbison AE. Effects of neuron-
the preoptic area and bed nucleus of the stria and activated by estradiol in mice. Endocri- specific estrogen receptor (ER) α and ERβ de-
terminalis. Biol Sex Differ. 2012 Feb;3(1):5. nology. 2013 Aug;154(8):2739–49. letion on the acute estrogen negative feed-
14 Orikasa C, Sakuma Y. Estrogen configures 26 Clarkson J, Boon WC, Simpson ER, Herbison back mechanism in adult female mice. Endo-
sexual dimorphism in the preoptic area of AE. Postnatal development of an estradiol- crinology. 2014 Apr;155(4):1418–27.
C57BL/6J and ddN strains of mice. J Comp kisspeptin positive feedback mechanism im- 40 Wintermantel TM, Campbell RE, Porteous
Neurol. 2010 Sep;518(17):3618–29. plicated in puberty onset. Endocrinology. R, Bock D, Gröne HJ, Todman MG, et al. Def-
15 Clarkson J, Herbison AE. Postnatal develop- 2009 Jul;150(7):3214–20. inition of estrogen receptor pathway critical
ment of kisspeptin neurons in mouse hypo- 27 Mayer C, Acosta-Martinez M, Dubois SL, for estrogen positive feedback to gonadotro-
thalamus; sexual dimorphism and projec- Wolfe A, Radovick S, Boehm U, et al. Timing pin-releasing hormone neurons and fertility.
tions to gonadotropin-releasing hormone and completion of puberty in female mice de- Neuron. 2006 Oct;52(2):271–80.
neurons. Endocrinology. 2006 Dec; 147(12): pend on estrogen receptor alpha-signaling in 41 Morales-Otal A, Ferreira-Nuño A, Olayo-
5817–25. kisspeptin neurons. Proc Natl Acad Sci USA. Lortia J, Barrios-González J, Tarragó-Castel-
16 Kauffman AS, Gottsch ML, Roa J, Byquist 2010 Dec;107(52):22693–8. lanos R. Effects of neonatal treatment with
AC, Crown A, Clifton DK, et al. Sexual dif- 28 Powers JB. Hormonal control of sexual re- two phytoestrogens on male rat sexual behav-
ferentiation of Kiss1 gene expression in the ceptivity during the estrous cycle of the rat. ior and partner preference. Behav Pharma-
brain of the rat. Endocrinology. 2007 Apr; Physiol Behav. 1970 Aug;5(8):831–5. col. 2016 Oct;27(7):570–8.
148(4):1774–83. 29 Parsons B, Rainbow TC, Pfaff DW, McEwen
BS. Oestradiol, sexual receptivity and cytosol
progestin receptors in rat hypothalamus. Na-
ture. 1981 Jul;292(5818):58–9.

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 413


DOI: 10.1159/000494558
42 Kubo K, Arai O, Omura M, Watanabe R, 55 Scallet AC, Divine RL, Newbold RR, Delclos 66 Tohei A, Suda S, Taya K, Hashimoto T, Kogo
Ogata R, Aou S. Low dose effects of bisphenol KB. Increased volume of the calbindin D28k- H. Bisphenol A inhibits testicular functions
A on sexual differentiation of the brain and labeled sexually dimorphic hypothalamus in and increases luteinizing hormone secretion
behavior in rats. Neurosci Res. 2003 Mar; genistein and nonylphenol-treated male rats. in adult male rats. Exp Biol Med (Maywood).
45(3):345–56. Toxicol Sci. 2004 Dec;82(2):570–6. 2001 Mar;226(3):216–21.
43 Nagao T, Saito Y, Usumi K, Nakagomi M, 56 Patisaul HB, Fortino AE, Polston EK. Neona- 67 Mani SK, Reyna AM, Alejandro MA, Crowley
Yoshimura S, Ono H. Disruption of the re- tal genistein or bisphenol-A exposure alters J, Markaverich BM. Disruption of male sexual
productive system and reproductive perfor- sexual differentiation of the AVPV. Neuro- behavior in rats by tetrahydrofurandiols
mance by administration of nonylphenol to toxicol Teratol. 2006 Jan-Feb;28(1):111–8. (THF-diols). Steroids. 2005 Oct;70(11):750–4.
newborn rats. Hum Exp Toxicol. 2000 May; 57 Patisaul HB, Fortino AE, Polston EK. Differ- 68 Markaverich B, Mani S, Alejandro MA,
19(5):284–96. ential disruption of nuclear volume and neu- Mitchell A, Markaverich D, Brown T, et al. A
44 Farabollini F, Porrini S, Della Seta D, Bianchi ronal phenotype in the preoptic area by neo- novel endocrine-disrupting agent in corn
F, Dessì-Fulgheri F. Effects of perinatal expo- natal exposure to genistein and bisphenol-A. with mitogenic activity in human breast and
sure to bisphenol A on sociosexual behavior Neurotoxicology. 2007 Jan;28(1):1–12. prostatic cancer cells. Environ Health Per-
of female and male rats. Environ Health Per- 58 Masutomi N, Shibutani M, Takagi H, Uneya- spect. 2002 Feb;110(2):169–77.
spect. 2002 Jun;110 Suppl 3:409–14. ma C, Takahashi N, Hirose M. Impact of di- 69 Retana-Márquez S, Juárez-Rojas L, Hernán-
45 Della Seta D, Minder I, Belloni V, Aloisi AM, etary exposure to methoxychlor, genistein, or dez A, Romero C, López G, Miranda L, et al.
Dessì-Fulgheri F, Farabollini F. Pubertal ex- diisononyl phthalate during the perinatal pe- Comparison of the effects of mesquite pod
posure to estrogenic chemicals affects behav- riod on the development of the rat endocrine/ and Leucaena extracts with phytoestrogens
ior in juvenile and adult male rats. Horm Be- reproductive systems in later life. Toxicology. on the reproductive physiology and sexual
hav. 2006 Aug;50(2):301–7. 2003 Nov;192(2-3):149–70. behavior in the male rat. Physiol Behav. 2016
46 Jones BA, Shimell JJ, Watson NV. Pre- and 59 Takagi H, Shibutani M, Masutomi N, Uneya- Oct;164(Pt A):1–10.
postnatal bisphenol A treatment results in ma C, Takahashi N, Mitsumori K, et al. Lack 70 Capela D, Dombret C, Poissenot K, Poignant
persistent deficits in the sexual behavior of of maternal dietary exposure effects of bi- M, Malbert-Colas A, Franceschini I, et al.
male rats, but not female rats, in adulthood. sphenol A and nonylphenol during the criti- Adult male mice exposure to nonylphenol al-
Horm Behav. 2011 Feb;59(2):246–51. cal period for brain sexual differentiation on ters courtship vocalizations and mating. Sci
47 Henry LA, Witt DM. Effects of neonatal res- the reproductive/endocrine systems in later Rep. 2018 Feb;8(1):2988.
veratrol exposure on adult male and female life. Arch Toxicol. 2004 Feb;78(2):97–105. 71 Adewale HB, Jefferson WN, Newbold RR,
reproductive physiology and behavior. Dev 60 Amstislavsky SY, Amstislavskaya TG, Ero- Patisaul HB. Neonatal bisphenol-a exposure
Neurosci. 2006;28(3):186–95. schenko VP. Methoxychlor given in the peri- alters rat reproductive development and
48 Whitten PL, Lewis C, Russell E, Naftolin F. implantation period blocks sexual arousal in ovarian morphology without impairing acti-
Phytoestrogen influences on the development male mice. Reprod Toxicol. 1999 Sep-Oct; vation of gonadotropin-releasing hormone
of behavior and gonadotropin function. Proc 13(5):405–11. neurons. Biol Reprod. 2009 Oct;81(4):690–9.
Soc Exp Biol Med. 1995 Jan;208(1):82–6. 61 Decatanzaro D, Berger RG, Guzzo AC, Thor- 72 Adewale HB, Todd KL, Mickens JA, Patisaul
49 Whitten PL, Patisaul HB, Young LJ. Neu- pe JB, Khan A. Perturbation of male sexual HB. The impact of neonatal bisphenol-A ex-
robehavioral actions of coumestrol and re- behavior in mice (Mus musculus) within a posure on sexually dimorphic hypothalamic
lated isoflavonoids in rodents. Neurotoxicol discrete range of perinatal bisphenol-A doses nuclei in the female rat. Neurotoxicology.
Teratol. 2002 Jan-Feb;24(1):47–54. in the context of a high- or low-phytoestro- 2011 Jan;32(1):38–49.
50 Wisniewski AB, Klein SL, Lakshmanan Y, gen diet. Food Chem Toxicol. 2013 May; 55: 73 Kwon S, Stedman DB, Elswick BA, Cattley
Gearhart JP. Exposure to genistein during 164–71. RC, Welsch F. Pubertal development and re-
gestation and lactation demasculinizes the 62 Khan A, Bellefontaine N, deCatanzaro D. productive functions of Crl:CD BR Sprague-
reproductive system in rats. J Urol. 2003 Apr; Onset of sexual maturation in female mice as Dawley rats exposed to bisphenol A during
169(4):1582–6. measured in behavior and fertility: interac- prenatal and postnatal development. Toxicol
51 Csaba G, Karabélyos C. Effect of single neo- tions of exposure to males, phytoestrogen Sci. 2000 Jun;55(2):399–406.
natal treatment with the soy bean phy- content of diet, and ano-genital distance. 74 Ryan BC, Hotchkiss AK, Crofton KM, Gray
tosteroid, genistein on the sexual behavior of Physiol Behav. 2008 Feb;93(3):588–94. LE Jr. In utero and lactational exposure to bi-
adult rats. Acta Physiol Hung. 2002; 89(4): 63 Picot M, Naulé L, Marie-Luce C, Martini M, sphenol A, in contrast to ethinyl estradiol,
463–70. Raskin K, Grange-Messent V, et al. Vulnera- does not alter sexually dimorphic behavior,
52 Gray LE Jr, Ostby J, Cooper RL, Kelce WR. bility of the neural circuitry underlying sexu- puberty, fertility, and anatomy of female LE
The estrogenic and antiandrogenic pesticide al behavior to chronic adult exposure to oral rats. Toxicol Sci. 2010 Mar;114(1):133–48.
methoxychlor alters the reproductive tract bisphenol a in male mice. Endocrinology. 75 Monje L, Varayoud J, Muñoz-de-Toro M,
and behavior without affecting pituitary size 2014 Feb;155(2):502–12. Luque EH, Ramos JG. Neonatal exposure to
or LH and prolactin secretion in male rats. 64 Wisniewski AB, Cernetich A, Gearhart JP, bisphenol A alters estrogen-dependent
Toxicol Ind Health. 1999 Jan-Mar; 15(1-2): Klein SL. Perinatal exposure to genistein al- mechanisms governing sexual behavior in
37–47. ters reproductive development and aggres- the adult female rat. Reprod Toxicol. 2009
53 He Z, Paule MG, Ferguson SA. Low oral dos- sive behavior in male mice. Physiol Behav. Dec;28(4):435–42.
es of bisphenol A increase volume of the sex- 2005 Feb;84(2):327–34. 76 Naulé L, Picot M, Martini M, Parmentier C,
ually dimorphic nucleus of the preoptic area 65 Jašarević E, Sieli PT, Twellman EE, Welsh TH Hardin-Pouzet H, Keller M, et al. Neuroen-
in male, but not female, rats at postnatal day Jr, Schachtman TR, Roberts RM, et al. Dis- docrine and behavioral effects of maternal
21. Neurotoxicol Teratol. 2012 May-Jun; ruption of adult expression of sexually select- exposure to oral bisphenol A in female mice.
34(3):331–7. ed traits by developmental exposure to bi- J Endocrinol. 2014 Feb;220(3):375–88.
54 McCaffrey KA, Jones B, Mabrey N, Weiss B, sphenol A. Proc Natl Acad Sci USA. 2011 Jul; 77 Kouki T, Kishitake M, Okamoto M, Oosuka
Swan SH, Patisaul HB. Sex specific impact of 108(28):11715–20. I, Takebe M, Yamanouchi K. Effects of neo-
perinatal bisphenol A (BPA) exposure over a natal treatment with phytoestrogens, genis-
range of orally administered doses on rat hy- tein and daidzein, on sex difference in female
pothalamic sexual differentiation. Neurotox- rat brain function: estrous cycle and lordosis.
icology. 2013 May;36:55–62. Horm Behav. 2003 Aug;44(2):140–5.

414 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558
78 Kouki T, Okamoto M, Wada S, Kishitake M,   91 Gray LE Jr, Ostby JS, Ferrell JM, Sigmon ER, 102 Takagi H, Shibutani M, Lee KY, Masutomi
Yamanouchi K. Suppressive effect of neona- Goldman JM. Methoxychlor induces estro- N, Fujita H, Inoue K, et al. Impact of mater-
tal treatment with a phytoestrogen, coumes- gen-like alterations of behavior and the re- nal dietary exposure to endocrine-acting
trol, on lordosis and estrous cycle in female productive tract in the female rat and ham- chemicals on progesterone receptor expres-
rats. Brain Res Bull. 2005 Jan;64(5):449–54. ster: effects on sex behavior, running wheel sion in microdissected hypothalamic medi-
79 Kudwa AE, Boon WC, Simpson ER, Handa activity, and uterine morphology. Toxicol al preoptic areas of rat offspring. Toxicol
RJ, Rissman EF. Dietary phytoestrogens Appl Pharmacol. 1988 Dec;96(3):525–40. Appl Pharmacol. 2005 Oct;208(2):127–36.
dampen female sexual behavior in mice with   92 Colbert NK, Pelletier NC, Cote JM, Concan- 103 Kundakovic M, Gudsnuk K, Franks B, Ma-
a disrupted aromatase enzyme gene. Behav non JB, Jurdak NA, Minott SB, et al. Perina- drid J, Miller RL, Perera FP, et al. Sex-spe-
Neurosci. 2007 Apr;121(2):356–61. tal exposure to low levels of the environ- cific epigenetic disruption and behavioral
80 Bertolasio J, Fyfe S, Snyder BW, Davis AM. mental antiandrogen vinclozolin alters sex- changes following low-dose in utero bisphe-
Neonatal injections of methoxychlor de- differentiated social play and sexual nol A exposure. Proc Natl Acad Sci USA.
crease adult rat female reproductive behav- behaviors in the rat. Environ Health Per- 2013 Jun;110(24):9956–61.
ior. Neurotoxicology. 2011 Dec; 32(6): 809– spect. 2005 Jun;113(6):700–7. 104 Tanaka M, Ohtani-Kaneko R, Yokosuka M,
13.  93 Dalsenter PR, Santana GM, Grande SW, Watanabe C. Low-dose perinatal diethyl-
81 Funabashi T, Sano A, Mitsushima D, Kimura Andrade AJ, Araújo SL. Phthalate affect the stilbestrol exposure affected behaviors and
F. Bisphenol A increases progesterone recep- reproductive function and sexual behavior hypothalamic estrogen receptor-alpha-pos-
tor immunoreactivity in the hypothalamus in of male Wistar rats. Hum Exp Toxicol. 2006 itive cells in the mouse. Neurotoxicol Tera-
a dose-dependent manner and affects sexual Jun;25(6):297–303. tol. 2004 Mar-Apr;26(2):261–9.
behaviour in adult ovariectomized rats. J   94 Lee HC, Yamanouchi K, Nishihara M. Ef- 105 Goldsby JA, Wolstenholme JT, Rissman EF.
Neuroendocrinol. 2003 Feb;15(2):134–40. fects of perinatal exposure to phthalate/adi- Multi- and transgenerational consequences
82 Retana-Márquez S, Aguirre FG, Alcántara M, pate esters on hypothalamic gene expres- of bisphenol A on sexually dimorphic cell
García-Díaz E, Muñoz-Gutiérrez M, Artea- sion and sexual behavior in rats. J Reprod populations in mouse brain. Endocrinolo-
ga-Silva M, et al. Mesquite pod extract modi- Dev. 2006 Jun;52(3):343–52. gy. 2017 Jan;158(1):21–30.
fies the reproductive physiology and behav-   95 Moore RW, Rudy TA, Lin TM, Ko K, Peter- 106 Yu CJ, Fang QQ, Tai FD. Pubertal BPA ex-
ior of the female rat. Horm Behav. 2012 Apr; son RE. Abnormalities of sexual develop- posure changes central ERα levels in female
61(4):549–58. ment in male rats with in utero and lacta- mice. Environ Toxicol Pharmacol. 2015
83 Zanoli P, Zavatti M, Geminiani E, Corsi L, tional exposure to the antiandrogenic plas- Sep;40(2):606–14.
Baraldi M. The phytoestrogen ferutinin af- ticizer Di(2-ethylhexyl) phthalate. Environ 107 Arambula SE, Belcher SM, Planchart A,
fects female sexual behavior modulating Health Perspect. 2001 Mar;109(3):229–37. Turner SD, Patisaul HB. Impact of low dose
ERalpha expression in the hypothalamus. Be-   96 Skinner MK, Savenkova MI, Zhang B, Gore oral exposure to bisphenol A (BPA) on the
hav Brain Res. 2009 May;199(2):283–7. AC, Crews D. Gene bionetworks involved in neonatal rat Hypothalamic and hippocam-
84 Zavatti M, Benelli A, Montanari C, Zanoli P. the epigenetic transgenerational inheri- pal transcriptome: A CLARITY-BPA con-
The phytoestrogen ferutinin improves sexual tance of altered mate preference: environ- sortium study. Endocrinology. 2016 Oct;
behavior in ovariectomized rats. Phytomedi- mental epigenetics and evolutionary biolo- 157(10):3856–72.
cine. 2009 Jun;16(6-7):547–54. gy. BMC Genomics. 2014 May;15(1):377. 108 Ceccarelli I, Della Seta D, Fiorenzani P, Far-
85 Henry LA, Witt DM. Resveratrol: phytoes-   97 Andrade AJ, Grande SW, Talsness CE, Ger- abollini F, Aloisi AM. Estrogenic chemicals
trogen effects on reproductive physiology icke C, Grote K, Golombiewski A, et al. A at puberty change ERalpha in the hypothal-
and behavior in female rats. Horm Behav. dose response study following in utero and amus of male and female rats. Neurotoxicol
2002 Mar;41(2):220–8. lactational exposure to di-(2-ethylhexyl) Teratol. 2007 Jan-Feb;29(1):108–15.
86 Patisaul HB, Luskin JR, Wilson ME. A soy phthalate (DEHP): reproductive effects on 109 Monje L, Varayoud J, Muñoz-de-Toro M,
supplement and tamoxifen inhibit sexual be- adult male offspring rats. Toxicology. 2006 Luque EH, Ramos JG. Exposure of neonatal
havior in female rats. Horm Behav. 2004 Apr; Nov;228(1):85–97. female rats to bisphenol A disrupts hypo-
45(4):270–7.  98 Dombret C, Capela D, Poissenot K, Par- thalamic LHRH pre-mRNA processing and
87 Doering CH. Diethylstilbestrol facilitates mentier C, Bergsten E, Pionneau C, et al. estrogen receptor alpha expression in nuclei
masculine and feminine copulatory behavior Neural mechanisms underlying disruption controlling estrous cyclicity. Reprod Toxi-
in female rats. Horm Behav. 1982 Dec;16(4): of male courtship behavior by adult expo- col. 2010 Dec;30(4):625–34.
462–74. sure to di-(2-ethylhexyl)phthalate in mice. 110 Gore AC, Walker DM, Zama AM, Armenti
88 Patisaul HB, Dindo M, Whitten PL, Young Environ Health Perspect. 2017 Sep 1;125(9): AE, Uzumcu M. Early life exposure to endo-
LJ. Soy isoflavone supplements antagonize 097001. crine-disrupting chemicals causes lifelong
reproductive behavior and estrogen receptor   99 Derouiche L, Keller M, Duittoz AH, Pillon molecular reprogramming of the hypothal-
alpha- and beta-dependent gene expression D. Developmental exposure to Ethinylestra- amus and premature reproductive aging.
in the brain. Endocrinology. 2001 Jul;142(7): diol affects transgenerationally sexual be- Mol Endocrinol. 2011 Dec;25(12):2157–68.
2946–52. havior and neuroendocrine networks in 111 Cao J, Mickens JA, McCaffrey KA, Leyrer
89 Di Viesti V, Carnevale G, Zavatti M, Benelli male mice. Sci Rep. 2015 Dec;5(1):17457. SM, Patisaul HB. Neonatal Bisphenol A ex-
A, Zanoli P. Increased sexual motivation in 100 Cao J, Rebuli ME, Rogers J, Todd KL, Ley- posure alters sexually dimorphic gene ex-
female rats treated with Humulus lupulus L. rer SM, Ferguson SA, et al. Prenatal bisphe- pression in the postnatal rat hypothalamus.
extract. J Ethnopharmacol. 2011 Mar;134(2): nol A exposure alters sex-specific estrogen Neurotoxicology. 2012 Jan;33(1):23–36.
514–7. receptor expression in the neonatal rat hy- 112 Monje L, Varayoud J, Luque EH, Ramos JG.
90 Patisaul HB, Melby M, Whitten PL, Young pothalamus and amygdala. Toxicol Sci. Neonatal exposure to bisphenol A modifies
LJ. Genistein affects ER beta- but not ER al- 2013 May;133(1):157–73. the abundance of estrogen receptor alpha
pha-dependent gene expression in the hypo- 101 Cao J, Joyner L, Mickens JA, Leyrer SM, transcripts with alternative 5′-untranslated
thalamus. Endocrinology. 2002 Jun; 143(6): Patisaul HB. Sex-specific Esr2 mRNA ex- regions in the female rat preoptic area. J En-
2189–97. pression in the rat hypothalamus and amyg- docrinol. 2007 Jul;194(1):201–12.
dala is altered by neonatal bisphenol A ex-
posure. Reproduction. 2014 Mar; 147(4):
537–54.

Endocrine Disruption of Sexual Behavior Neuroendocrinology 2018;107:400–416 415


DOI: 10.1159/000494558
113 Rebuli ME, Cao J, Sluzas E, Delclos KB, 115 Wang X, Chang F, Bai Y, Chen F, Zhang J, 117 Barrett ES, Parlett LE, Wang C, Drobnis EZ,
Camacho L, Lewis SM, et al. Investigation of Chen L. Bisphenol A enhances kisspeptin Redmon JB, Swan SH. Environmental expo-
the effects of subchronic low dose oral expo- neurons in anteroventral periventricular sure to di-2-ethylhexyl phthalate is associ-
sure to bisphenol A (BPA) and ethinyl estra- nucleus of female mice. J Endocrinol. 2014 ated with low interest in sexual activity in
diol (EE) on estrogen receptor expression in Apr;221(2):201–13. premenopausal women. Horm Behav. 2014
the juvenile and adult female rat hypothala- 116 Patisaul HB, Whitten PL, Young LJ. Regula- Nov;66(5):787–92.
mus. Toxicol Sci. 2014 Jul;140(1):190–203. tion of estrogen receptor beta mRNA in the 118 Li DK, Zhou Z, Miao M, He Y, Qing D, Wu
114 Roepke TA, Yang JA, Yasrebi A, Mamounis brain: opposite effects of 17beta-estradiol T, et al. Relationship between urine bisphe-
KJ, Oruc E, Zama AM, et al. Regulation of and the phytoestrogen, coumestrol. Brain nol-A level and declining male sexual func-
arcuate genes by developmental exposures to Res Mol Brain Res. 1999 Apr;67(1):165–71. tion. J Androl. 2010 Sep-Oct;31(5):500–6.
endocrine-disrupting compounds in female
rats. Reprod Toxicol. 2016 Jul;62:18–26.

416 Neuroendocrinology 2018;107:400–416 Mhaouty-Kodja/Naulé/Capela


DOI: 10.1159/000494558

You might also like