Content uploaded by Roger Hart
Author content
All content in this area was uploaded by Roger Hart on Feb 17, 2018
Content may be subject to copyright.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY:
ROLE OF THE ENVIRONMENT, MODERN
LIFESTYLE, AND GENETICS
Roger J. Hart
School of Women’s and Infants Health, University of Western Australia & Fertility Specialists of Western
Australia, Subiaco, Perth Western Australia
LHart RJ. Physiological Aspects of Female Fertility: Role of the Environment, Modern
Lifestyle, and Genetics. Physiol Rev 96: 873–909, 2016. Published June 1, 2016;
doi:10.1152/physrev.00023.2015.—Across the Western World there is an in-
creasing trend to postpone childbearing. Consequently, the negative influence of age on
oocyte quality may lead to a difficulty in conceiving for many couples. Furthermore,
lifestyle factors may exacerbate a couple’s difficulty in conceiving due mainly to the metabolic
influence of obesity; however, the negative impacts of low peripheral body fat, excessive exercise,
the increasing prevalence of sexually transmitted diseases, and smoking all have significant nega-
tive effects on fertility. Other factors that impede conception are the perceived increasing preva-
lence of the polycystic ovary syndrome, which is further exacerbated by obesity, and the presence
of uterine fibroids and endometriosis (a progressive pelvic inflammatory disorder) which are more
prevalent in older women. A tendency for an earlier sexual debut and to have more sexual partners
has led to an increase in sexually transmitted diseases. In addition, there are several genetic
influences that may limit the number of oocytes within the ovary; consequently, by postponing
attempts at childbearing, a limitation of oocyte number may become evident, whereas in previous
generations with earlier conception this potentially reduced reproductive life span did not manifest
in infertility. Environmental influences on reproduction are under increasing scrutiny. Although firm
evidence is lacking however, dioxin exposure may be linked to endometriosis, phthalate exposure
may influence ovarian reserve, and bisphenol A may interfere with oocyte development and matu-
ration. However, chemotherapy or radiotherapy is recognized to lead to ovarian damage and
predispose the woman to ovarian failure.
I. INTRODUCTION 873
II. PHYSIOLOGY 874
III. PATHOLOGICAL PROCESSES,... 878
IV. LIFESTYLE INFLUENCES... 888
V. ENVIRONMENTAL INFLUENCES... 893
VI. CONCLUSION 900
I. INTRODUCTION
The most powerful influence relating to a woman’s chance
of conceiving is her age. Female age has physiological and
genetic influences on conception, relating to a reduced ovar-
ian follicular pool, perturbations in ovulation, and an in-
crease in meiotic errors within the oocyte. Indeed, in some
instances, age could be considered a lifestyle decision; how-
ever, in most instances this is not the case, as with increasing
societal and professional pressures upon women childbear-
ing is increasingly postponed into the 30s, whereas in pre-
vious generations starting a family in the 20s was the norm.
This has resulted in the increasing recourse to fertility treat-
ment; indeed, 1 in 25 children in Australia are born as a
result of in vitro fertilization (IVF) treatment, and it is be-
lieved the figure reaches 1 in 7 for women over 37 years of
age (239), when the treatment is much less successful (FIG-
URE 1).
This delay in childbearing has provided a window of op-
portunity for various lifestyle, pathological, and genetic
perturbations to exert their influence further to reduce a
couple’s chance of conceiving. The lifestyle factors that
have a detrimental impact on reproduction relate mainly to
the metabolic influence of obesity; however, the negative
impacts of low peripheral body fat, excessive exercise, the
increasing prevalence of sexually transmitted diseases, and
smoking all have significant negative effects on female fe-
cundity at a population level.
Other factors that are believed to be exerting an increas-
ing negative influence upon female conception are the
greater prevalence of the polycystic ovary syndrome,
which is further exacerbated by obesity. Furthermore,
the incidence of uterine myomas (fibroids) and endome-
triosis (a progressive pelvic inflammatory disorder) are
more prevalent in older women. A tendency for an earlier
sexual debut and to have more sexual partners has led to
Physiol Rev 96: 873–909, 2016
Published June 1, 2016; doi:10.1152/physrev.00023.2015
8730031-9333/16 Copyright © 2016 the American Physiological Society
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
an increase in notifications of sexually transmitted dis-
eases, a well-established cause of infertility. In addition,
there are several genetic influences that may limit the
“ovarian reserve,” an expression of the total number of
oocytes within the ovary; consequently, by postponing
attempted childbearing a limitation in ovarian reserve
may become evident, whereas in previous generations
with earlier conception this propensity to a limited repro-
ductive lifespan was not revealed.
Environmental influences upon reproduction are under in-
creasing scrutiny; however, the firm evidence to date is lack-
ing. There are suggestions that dioxin exposure may be
linked to endometriosis and phthalate exposure may influ-
ence ovarian reserve, and bisphenol A may interfere with
oocyte development and maturation, although very good
evidence exists to relate the exposure to chemotherapy and
radiotherapy to gonadal damage.
The purpose of this review is to attempt to cover the sub-
stantial field of female fertility and to try to address the
influences of lifestyle and the environment on female infer-
tility, and to provide a limited insight into genetic influences
on female reproduction, from follicular development to im-
plantation and early pregnancy.
II. PHYSIOLOGY
A. Folliculogenesis
Follicular development originates in utero during the sec-
ond trimester of pregnancy by the rapid mitosis of the pri-
mordial germ cells into up to a maximum of ⬃6 million
oogonia at 20 wk of gestation; from this point onwards the
dominant activity is atresia. Indeed, there will only be 1
million germ cells surviving at birth (243). This finite re-
serve of primordial germ cells originates in the yolk sac
endoderm and migrates to the gonadal ridge, and upon
arrival enter the first meiotic division and become primary
oocytes, and constitute the “ovarian reserve” [follicles that
can subsequently be recruited for ovulation (20)]. These
germ cells are essential for the formation and maintenance
of the ovary, and in their absence the gonad degenerates
into cordlike structures (215, 243). These primary oocytes
are maintained at this arrested state of meiosis until the time
of the surge in luteinizing hormone (LH) that presages ovu-
lation. The oocyte is arrested in prophase I of meiosis by
high levels of cAMP (323). In addition to the germ cells, the
primordial follicle consists of somatic cells derived from the
primitive gonad which develop into the flattened granulosa,
theca, and interstitial cells. As described, this so-called fol-
licular reserve of 6-7 million oocytes at 20 wk of gestation
(20, 22) will continually deplete throughout the woman’s
life, the rate of which may be accelerated by factors such
as genetic influences such as Turner’s syndrome or Frag-
ile X premutation carrier status (67), virological expo-
sures such as the mumps virus (71), environmental expo-
sure such as treatment with chemotherapy or radiother-
apy (138), ovarian surgery (6), negative lifestyle factors
such as smoking (308), and also in relation to autoim-
mune causes (367).
With the use of rodent models, factors involved in the reg-
ulation of the recruitment of these primordial follicles into a
developing population of follicles for a menstrual cycle have
been explored. Once puberty commences, Kit ligand and
leukemia inhibitory factor (LIF) have been identified as sig-
nificant promoters of the follicular transition (210, 237,
323). Kit ligand is expressed on the granulosa cells of the
developing primordial follicle, with its receptor on the
membrane (oolema) of the primary oocyte, and LIF is se-
creted from the early granulosa cells. LIF secretion is be-
lieved to regulate the local signaling involved with primor-
dial follicle activation by promoting Kit ligand expression
(323). In addition, oocyte-derived factors bone-morpho-
genic protein 15 (BMP-15) and growth differentiation fac-
tor-9 (GDF-9) are involved in the promotion of primordial
0
5
10
15
20
25
30
< 22 23-24 25-26 27-28 29-30 31-32 33-34 35-36 37-38 39-40 41-42 43-44 ≥ 45
Percentage live birth rate
Age group (years)
FIGURE 1. Age-specific live delivery rates per initiated
autologous fresh cycle by two-year age groups, Australia
and New Zealand, 2013. The highest live delivery rates
were for women aged between their mid-20s to early-30s.
For women aged 45 or older, only one live delivery resulted
from every 80 initiated cycles compared with one live de-
livery from every four initiated cycles in women aged be-
tween 25 and 34. [From Macaldowie et al. (206), with
permission.]
ROGER J. HART
874 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
follicle maturation (70, 255); in addition, granulosa cell
anti-Müllerian hormone (AMH) secreted by the granulosa
cells of small antral follicles appears to act as a restraint (51,
184). Other paracrine signals involved in primordial follicle
activation include basic fibroblast growth factor (bFGF),
nobox and Foxo3, and insulin (176, 243). For a detailed
description of the local factors involved in follicle and
oocyte development, please refer to Sobinoff et al. (323) and
Kristensen et al. (184) (FIGURE 2). The flattened granulosa
cells become cuboidal during the transition into a primary
follicle under the influence of transcription factor Foxl2; the
oocyte increases in diameter and develops a zona pellucida.
This recruitment into primary follicles commences during
fetal life and continues until the menopause.
B. Ovulation
The development and recruitment of these primordial folli-
cles is regulated by paracrine and autocrine signals involv-
ing the transforming growth factor-

(TGF-

) superfamily,
which includes TGF-

, inhibins, activins, follistatins, bone
morphogenic proteins, growth factors, and AMH (184).
Then the follicle transitions to the gonadotrophin-depen-
dent antral follicle to the preovulatory stage; the whole
process takes ⬃6 months (129). The gradual maturational
change from the primordial follicle characterized by the
cuboidal granulosa cells, through the preantral stage (up to
0.2 mm in size), involves proliferation and maturational
changes and multi-layering within the granulosa and theca
cells, and the development of an antrum within the follicle.
With the development of the antrum, the follicle becomes
responsive to gonadotrophins, which takes several months
to complete (129, 243). The granulosa cell basal lamina is
traversed by many gap junctions that allow communication
and nutrition to surrounding granulosa cells and across the
zona pellucida to the oocyte. The innermost layers of gran-
ulosa cells become differentiated as cumulus cells. In con-
trast, the theca cells have a greater vascular supply and are
responsible for androgen synthesis, under LH stimulation.
This acts as the substrate for the granulosa cell synthesis of
estradiol, primarily under follicular stimulating hormone
(FSH) control, the so-called “two-cell two gonadotrophin
hypothesis” (21, 89). Both granulosa and theca cells secrete
growth factors that modulate follicle maturation; the acti-
vators include Kit ligand, FGF-2, KGF, LIF, BMPs, and
GDF-9, and the inhibitors include AMH, Foxl2, and
Foxo3A (for a detailed description, see Ref. 243). The pre-
antral follicle transition to the antral stage is modulated by
AMH and stimulated by activins and GDF-9, and then as
described the antral follicle becomes responsive to FSH
(243).
Gonadotrophin stimulation of the ovary requires integ-
rity of the hypothalamic-pituitary-ovary axis and appro-
priate peripheral signals that influence hypothalamic
function. Gonadotrophin releasing hormone (GnRH)
neurons arise alongside the olfactory nerves and migrate
during embryological development to the hypothalamus
and send neuronal projections from the arcuate nucleus
to the median eminence where they release GnRH into
the capillaries of the hypophysial-portal vessels (194).
The GnRH then binds to the GnRH receptor on the an-
terior pituitary initiating the synthesis and secretion of
LH and FSH (194). The control of hypothalamic secre-
tion of GnRH is complex, relying on a complex interplay
of ovarian feedback, primarily by inhibin B and estradiol
and neuroendocrine signals modulated by systemic met-
abolic signals. Kisspeptins have the strongest influence
over GnRH release via the kisspeptin receptor, coded for
by GPR54, and are also integral to the timing and onset
of puberty (232, 259; for detailed description, see Pinilla
et al., Ref. 259). The kisspeptin neurons also express the
FIGURE 2. Diagrammatic representation of primordial
follicle activation, oocyte maturation, and oocyte activa-
tion. A: the follicular activation of the dormant primordial
follicle occurs in response to cytokine growth factors
(e.g., Kit and LIF) and is characterized by oocyte growth
and granulosa cell differentiation/proliferation. B: oocyte
maturation occurs in response to the LH surge, resulting
in meiotic resumption and arrest at metaphase II. C:
oocyte activation occurs in ovulated oocytes after fertil-
ization, resulting in the completion of MII, extrusion of the
second polar body, and male/female pro-nuclear forma-
tion. Images are not to scale. [From Sobinoff et al. (323),
with permission from Oxford University Press.]
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
875Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
neuropeptides neurokinin B (NKB) and dynorphin which
modulate the GnRH secretion (123). Other neuropep-
tides integral to GnRH pulsatility are
␣
melanocyte-stim-
ulating hormone (
␣
MSH), produced by proopiomelano-
cortin and cocaine and amphetamine-regulated tran-
script (POMC/CART), and the agouti-related protein
(AgRP), an antagonist of
␣
MSH, produced by the neuro-
peptide Y (NPY)/AgRP neurons (for review, see Navarro
and Kaiser, Ref. 232). These neuropeptides are integral
in the coordination of the reproductive and metabolic
axes through their action in the hypothalamus (232); the
neuropeptides derived from POMC/CART neurons exert
a potent anorectic action, thus decreasing food intake
and body weight, whereas AgRP and NPY have the op-
posite (orexigenic) effect, inducing food intake. In addi-
tion, these neurons express receptors for leptin (secreted
by adipose tissue, anorexogenic, and a stimulus for
GnRH activity), insulin, and gherlin (secreted by the gut,
orexigenic, and a suppressor of GnRH activity). Further-
more, neuronal projections from the premammillary ven-
tral nucleus which can be stimulatory (glutamatergic) or
inhibitory (GABAergic) are responsive to peripheral lep-
tin signals, leading to a complex relation of reproductive
responses to metabolic signals (232).
After the antral stage of development, granulosa cell es-
tradiol production increases by granulosa cell prolifera-
tion, increased vascularity, and an increased supply of
theca cell androgens. Inhibin A secretion from larger fol-
licles increases, promoting androgen secretion from
theca cells, and there is a reduction of activin A secretion
from larger follicles. Activin A acts to inhibit androgen
secretion and promote oocyte developmental competence
(5). As preantral follicle development progresses, there is
an increase in FSH and LH receptor expression, aroma-
tase activity, and inhibin and progesterone production
(180).
The oocyte-derived BMPs and GDF-9 inhibit the premature
luteinization of the follicle, which commences after the re-
lease of the oocyte at ovulation. Inhibin A secretion via
pituitary feedback leads to a reduction in FSH secretion;
hence, larger, or dominant follicles, will have higher con-
centrations of FSH and LH receptors and will continue to
have higher aromatase activity and hence estradiol secre-
tion.
Prior to ovulation, FSH induces expression of LH and
progesterone receptors on the surface of the granulosa
cells. The transformation to luteinization of the domi-
nant follicle is initiated by the surge in LH and is often
preceded by a slight rise in serum progesterone. The pro-
cess of ovulation involves rapid expansion of the domi-
nant follicle (1–4 mm/day), and ovulation results when
tumor necrosis factor-
␣
(TNF-
␣
)-stimulated collagenase
weakens the apical surface epithelium of the ovary and
follicle rupture results (228). The corpus luteum is com-
posed of theca and granulosa cells, endothelial, immune
cells, macrophages, T and B lymphocytes, and fibroblast
cells and secretes up to 40 mg of progesterone per day, in
addition to estradiol and androgens (82, 83, 275). The
LH surge must last more than 24 h to initiate resumption
of oocyte meiosis and breakdown of the granulosa cell-
oocyte gap junctions as well as to promote luteinization
of the granulosa cells, ovulation, and the initiation of
corpus luteum function (82, 83). It is believed that stim-
ulation of the granulosa progesterone receptor is also a
prerequisite for ovulation (66). The granulosa-lutein
cells express the enzyme aromatase and produce estradiol
in addition to progesterone, and the theca-lutein cells
have P450C17 activity and generate the androgen pre-
cursors for granulosa cells (82), and are also believed to
secrete progesterone (83). Granulosa cell steroidogenic
acute regulatory protein (StAR) expression is substan-
tially increased around the time of ovulation, induced by
LH (171). StAR is essential for the movement of choles-
terol carried by low-density lipoprotein into the inner
membrane of the mitochondria where it becomes a sub-
strate for P450scc to commence steroidogenesis (171).
Over one-third of cells within the corpus luteum are en-
dothelial cells representing the significant degree of vas-
cularization the corpus luteum has undergone, induced
primarily through the expression of vascular epithelial
growth factor (VEGF). The immune cells are responsible
for the secretion of cytokines, primarily interleukin
(IL)-1

and TNF-
␣
, which modulate steroidogenesis
(82). Estradiol production by the granulosa-lutein cells is
stimulated by LH and insulin-like growth factor I (IGF-
I), not by FSH. In the absence of human chorionic gonad-
otrophin (hCG) secreted by a developing embryo or ex-
ogenously administered, there is a substantial reduction
of StAR expression (84), mirrored in the serum proges-
terone level and reductions in P450scc and 3

hydroxys-
teroid dehydrogenase (HSD) (responsible for conversion
of pregnenolone to progesterone) (83). Luteolysis leads
to reductions in progesterone, estradiol, and inhibin A
which through hypothalamic-pituitary-ovarian feedback
initiates a further wave of follicular recruitment via in-
creased GnRH pulses and the falling steroids precipitate
menstruation. The process of luteolysis is not well under-
stood, however, apoptosis is a significant feature, al-
though the percentage of cells with apoptotic markers is
low (358), and it involves breakdown of the extracellular
matrix by matrix metalloproteinases (159).
At the time of embryo implantation, trophoblast produc-
tion of hCG prevents the regression of the corpus luteum by
increasing StAR expression within the granulosa and theca-
lutein cells, increasing the vascular supply to the corpus
luteal cells and a reduction in luteolysis by inhibition of the
pro-apoptotic protein Bax and an increase in macrophages
ROGER J. HART
876 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
that are believed to be essential for the vascular support of
the corpus luteum (50, 83, 333).
C. Fertilization and Early Embryonic
Development
After release of the oocyte and cumulus cell complex from
the ovary to the fimbriae of the fallopian tube, the oocyte is
fertilized in the ampulla of the distal end of the fallopian
tube. The process of binding of the acrosomal membrane of
a sperm that has undergone the acrosome reaction and ca-
pacitation to the zona pellucida (a glycoprotein matrix sur-
rounding the oocyte) initiates release of cortical granules
within the oocyte. The binding of the sperm to the zona
pellucida precipitates a hardening of the zona preventing
polyspermic fertilization of the oocyte (the zona reaction),
intracellular calcium oscillations commence, and meiosis II
is completed by extrusion of the second polar body. In
addition, the sperm acrosome contains several lytic en-
zymes and zona pellucida binding proteins (241). The
mechanism of sperm-oocyte binding to the four zona pellu-
cida sperm binding proteins (ZP1 to 4) and the prevention
of polyspermic fertilization is still subject to debate (for
further discussion, see Clift and Schuh, Ref. 62), as from
animal studies there are believed to be several other sub-
stances involved in the sperm-oocyte fusion, such as sperm
ADAMs (consisting of a disintegrin and a metalloprotei-
nase) and their oocyte integrin ligands, and the sperm pro-
teins IZUMO1 and SPESP1 (98). The increase in calcium
within the oocyte is a trigger for the development of the
female pronucleus and the sperm DNA, which is tightly
packed with protamines, undergoes decondensation, and is
wrapped around nucelosomes and forms the male pronu-
cleus (62). In addition, global DNA demethylation occurs in
male pronucleus; this is active and rapid, and in the female
pronucleus this is passive and slower, and epigenetic repro-
gramming commences. Roughly 150 genes are considered
“imprinted” in that their methylation pattern (to suppress
the expression of a gene) is determined by the parent of
origin of the gene, and they retain their methylation pattern
(260). Mitochondria that originate in the sperm are de-
stroyed in early embryonic development, to prevent mito-
chondrial heteroplasmy (73).
Under the influence of the microtubules of the sperm aster
(a star-shaped structure derived from the centriole of the
mid-piece of the sperm), the pronuclei migrate towards the
center of the oocyte (296). Syngamy is the point at which
the pronuclei come together and break down completing
fertilization, and subsequently the centrioles duplicate and
migrate around the zygote nucleus to form opposite poles of
the first mitotic spindle and commence the first cleavage
(296). Early embryonic cleavage and development is regu-
lated by mRNA transcripts and proteins within the
ooplasm, and by cell division over the subsequent days with
activation of the embryonic genome develops to a blasto-
cyst of ⬃100 blastomeres (196).
D. Fallopian Tube Function
The fallopian tube is derived from the Müllerian duct and
averages 11 cm in length; however, it is made of four dis-
tinct regions with differing functional significance: the in-
fundibulum and fimbria (for oocyte capture), the ampullary
region (fertilization occurs at the junction with the isth-
mus), the isthmus, and interstitial portion of the fallopian
tube (regulating the release of the embryo into the endome-
trial cavity), and each region has differing secretions for the
nutrition of the early embryo and for capacitation and sus-
tenance of the sperm (294). The tube is responsible for the
transport and nutrition of the gametes and early embryo, by
muscular contractions and ciliary action (204). As de-
scribed, it is the site of fertilization and for the collection of
the released oocyte, and its secretory activity, cyclical mor-
phology, and contractility are related to the hormonal en-
vironment. In the early stages of embryo development, the
fallopian tube secretion is low in glucose and has relatively
high levels of pyruvate and lactate, the inverse of the uterine
environment (see TABLE 1), a fact that is mirrored in the
development of commercial embryo culture media for IVF
treatment to mimic the in vivo early embryo environment
by using sequential culture media as the embryo develop to
the blastocyst stage (191). For a detailed review of the phys-
iology of the fallopian tube, please refer to References 7, 16,
191.
E. Implantation
The endometrium is prepared for implantation under the
influence of estrogen and progesterone; however, only ap-
proximately half of all embryos that are generated will im-
plant and proceed to a successful on-going pregnancy (368).
The window of implantation is limited to a receptive win-
Table 1. Differences between fallopian tube and uterine
secretion for mammalian embryos
Component Oviduct Uterus
Glucose concentration, mM 0.5 3.15
Pyruvate concentration, mM 0.32 0.10
Lactate concentration, mM 10.5 5.2
Oxygen concentration, % 8 1.50
Carbon dioxide concentration, % 12 10
pH 7.5 7.1
Glycine concentration, mM 2.77 19.33
Alanine concentration, mM 0.5 1.24
Serine concentration, mM 0.32 0.8
From Lane and Gardner (191), with permission.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
877Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
dow between 7 and 10 days after ovulation when the endo-
metrium has undergone secretory morphological changes in
preparation for implantation with the development of api-
cal epithelial protrusions-pinopodes (236, 265) and is pro-
tected against blastocyst attachment until the appropriate
time by Muc-1 and facilitated by
␣
v

3
and
␣
4

1
integrin
expression (134). Implantation is characterized by three
phases: apposition, adhesion, and invasion into the decidu-
alized endometrium. Decidualization describes the mor-
phological and functional changes occurring within the en-
dometrial stromal cells in response to progesterone in prep-
aration for embryo implantation. Apposition of the
blastocyst trophectoderm occurs with the inner cell mass
directed towards the epithelium. Factors that are believed to
be important in implantation and the appropriate endome-
trial changes are as follows: LIF, IL-1, colony stimulating
factor, L-selectin, Wnt, Hoxa10/11, heparin binding epider-
mal growth factor, and bone morphogenic protein 2, in
addition to cell adhesion molecules and glycoproteins, and
it is believed that signaling from the embryo is crucial to
trigger this process of endometrial decidualization (for a
detailed review, see Refs. 54, 85, 134). The trophoblast cells
then degrade the extracellular matrix and invade into the
endometrial epithelium, basal lamina, and the stroma and
placental formation commences.
III. PATHOLOGICAL PROCESSES, THEIR
IMPLICATIONS, AND THERAPEUTIC
OPTIONS
A. Folliculogenesis
1. Genetic
Premature ovarian insufficiency (POI) occurs in ⬃1% of
women and is defined as the cessation of menstrual cycles
under 40 years of age in the presence of an elevated serum
FSH measured on two separate occasions (128). The causes
may be genetic (107), environmental, infective (subsequent
to mumps infection), associated with autoimmune condi-
tions, metabolic [due to biochemical damage in the presence
of galactossaemia (174)], and subsequent to cancer therapy
(138) or surgery (230); however, in the majority of cases no
cause is determined (128). A positive family history exists in
10–15% of cases with the suggestion that inheritance is
autosomal dominant sex-linked or X-linked with incom-
plete penetrance (357). For a detailed review of the genetic
mutations associated with premature ovarian insufficiency,
please see Fortuno and Labarta (107). There are in excess of
20 genes on the X chromosome, particularly involving the
critical region of the short arm of the X chromosome be-
tween Xp21.1 and Xp22.1.22 and the long arm regions
Xq13.3-Xq21.1 and Xq26-Xqter (107), and well over 50
autosomal genes related to POI.
Possibly the most common genetic cause of POI is Turner
syndrome (347) characterized by the loss of all, or part of an
X chromosome, occurring in ⬃1 in 3,000 female births
(336). Approximately half due to X chromosome mono-
somy and the majority of the remainder due to mosaicism
(336). This is a condition with several phenotypic features
characterized by short stature, cardiac and renal abnormal-
ities, hypothyroidism, webbed-neck, and otological and
ophthalmological abnormalities are all common in child-
hood (336). The ovarian insufficiency commonly found in
this condition relates to disruption of the BMP15 gene locus
located at Xp11.2, within a critical region related to ovar-
ian failure (254, 391). Spontaneous puberty occurs in ap-
proximately one quarter of girls, more commonly in mosa-
ics; however, premature ovarian failure is universal.
Another common genetic cause of POI is related to the
fragile X mental retardation protein, occurring in 3–15% of
patients with POI (373). In this condition there is an expan-
sion of the CGG triplet repeats of the FMR1 gene at
Xq27.3, and in the presence of more than 200 repeats the
condition fragile X syndrome occurs. This is a severe form
of mental retardation and autism; however, in the presence
of the premutation of 55 and 200 triplet repeats, premature
ovarian failure results (373).
The fertility options for women with premature ovarian
insufficiency are essentially restricted to oocyte donation
treatment only. This is a reliable and effective treatment
provided the woman has been assessed as medically fit and
suitable for assisted reproduction and she has adequate
uterine and endometrial development, which may be lack-
ing in some women with POI (150).
This therefore raises the specter of attempts to preserve
fertility in women where it would be expected that there is
a premature depletion of oocytes; such as in Turner syn-
drome or galactossaemia for instance. Attempts have been
made by the freezing of ovarian cortical tissue for young
adolescent girls or by the collection of stimulated mature
oocytes as part of an IVF cycle for older girls; however, due
to the restricted oocyte number, these techniques are not
routine and have had to date limited success (150) but may
offer promise in the future (242). Furthermore, when a
woman with Turner syndrome commences fertility treat-
ment in view of the associated increased risk of cardiovas-
cular-related mortality in pregnancy, they should be cared
for under specialized care, and indeed, Turner syndrome
should be considered a relative contraindication to preg-
nancy (266).
B. Ovulation
1. Hypogonadotrophic hypogonadism
Insufficient ovarian stimulation with gonadotrophins LH
and FSH results in the condition of hypogonadotrophic
hypogonadism (HH), either due to insufficient hypotha-
ROGER J. HART
878 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
lamic GnRH stimulation of the pituitary or due to insuffi-
cient secretion due to pituitary compromise. After exclusion
of excessive exercise, extreme stress, or an eating disorder,
and after ensuring pituitary function, other than secretion
of LH and FSH, is normal, and pituitary imaging is normal
the condition is considered idiopathic HH (IHH) (194). The
most common cause of GnRH insufficiency is the failure of
migration of the GnRH secretory neurons to the forebrain
which may also result in olfactory disorder (Kallman’s syn-
drome); in the absence of olfactory, the condition is de-
scribed as normosmic IHH (354). The inheritance is either
X-linked (KAL genes), autosomal dominant or autosomal
recessive, and the condition is usually detected in adoles-
cence with a delay or absence of pubertal development, with
in excess of 20 genes implicated in this condition to date
(194). Kallman’s syndrome is characterized by HH and
anosmia and is most commonly caused by mutations of the
KAL-1, KAL-2, and KAL-6 and are all implicated in inter-
ference with the neuronal migration associated with HH
and anosmia, in addition to other abnormalities (354). Mu-
tations of the GnRH1 gene encoding GnRH are very rare;
however, mutations of the GnRHR gene (4q13.2–3) encod-
ing the GnRH receptor are more common and lead to vari-
able expression of the phenotype (354). The KISS1 gene
encodes kisspeptins which stimulates GnRH release, its re-
ceptor is coded for by the GPR54 gene, and hence muta-
tions of either gene will lead to variable expression of IHH
either in childhood or adulthood. In addition, the TAC3
gene codes for NK3R which also stimulates GnRH neurons,
mutations of which lead to perturbed GnRH secretion and
hence insufficient LH and FSH secretion. In addition, mu-
tations in the leptin gene, Ob, the LH and FSH

subunits,
LHB and FSHB, are rare but are associated with hypogo-
nadism. For a more detailed review of the genetics associ-
ated with IHH and Kallman’s syndrome, see Valdes-Socin
et al. (354) and Layman (194).
2. Hyperprolactinemia
Other central causes of HH can be caused by systemic dis-
ease, medication (such as opioids and psychotropic medica-
tion), hypothalamic or pituitary compression, or infiltra-
tion; however, the most common cause is probably hyper-
prolactinemia. Hyperprolactinemia may be caused by
physiological states such as pregnancy, breastfeeding,
stress, exercise, and some medications as well as patients
with chronic hypothyroidism. Kidney disease may predis-
pose a patient to hyperprolactinemia due to reduced clear-
ance and altered prolactin metabolism. As prolactin secre-
tion is suppressed by hypothalamic dopamine secretion,
interruption or compression of the pituitary stalk by a non-
prolactin-secreting pituitary tumor will lead to hyperpro-
lactinemia. Furthermore, prolactin secreting adenomas, ei-
ther a micro (⬍10 mm) or a macro adenoma (⬎10 mm in
size), lead to prolactin inhibition of gonadotrophin secre-
tion and anovulation (214).
3. Polycystic ovary syndrome
The polycystic ovary syndrome (PCOS) is a collection of
signs and symptoms related to ovarian dysfunction, found
within a phenotypically heterogeneous group of women. It
is classically described by the Rotterdam criteria (285) as a
syndrome consisting of two of three criteria related to in-
frequent or absent ovulation, a morphological description
of the ovaries by ultrasound assessment, and hyperandro-
genism. Other groups suggest that the excessive androgen
secretion is the most significant underlying pathology as this
is believed to lead to ovarian dysfunction and the longer
term metabolic consequences these women experience
(140), and they consequently adopt a more stringent defi-
nition of PCOS (19, 382).
The etiology of PCOS is unclear with putative causes
being a genetic predisposition modulated by hyperinsu-
linemia or the early life environment, as there is very
good evidence from animal models for a programming
effect of early life exposure to androgens (1). The pheno-
type of PCOS is modulated by the presence of obesity,
which also exacerbates the metabolic features of PCOS
(226). From genome-wide association studies (GWAS),
potential loci for PCOS have been identified at 9q22.32,
8p23.1, and 11p14.1 and single-nucleotide polymor-
phisms (SNPs) of the gonadotrophin receptors and the
androgen receptor (102), and several other SNPs have
been identified (18). A SNP at 11p14.1 was associated
with PCOS and elevated serum LH concentration, a fre-
quently observed feature in PCOS. Other genes believed
to be associated with the development of PCOS relate to
genes involved with insulin signaling and the epidermal
growth factor receptor (for review, see Ref. 18). There is
evidence from both human and animal studies that sup-
raphysiological maternal androgen levels may lead to
disordered folliculogenesis in female offspring with a
PCOS phenotype (1, 106, 281). Animal models of PCOS
have been generated with rodents (363), sheep (281), and
monkeys (1) by early life exposure to supraphysiological
androgens, leading to the development in the female of
offspring of hyperandrogenism, hyperinsulinemia, LH
hypersecretion, and ovulatory disorder. In humans, evi-
dence from early life exposure to hyperandrogenemia in
conditions such as congenital adrenal hyperplasia can
lead to the development of PCOS like features (24). How-
ever, the impact of variations in maternal androgens
within the normal physiological range is less well under-
stood. One study measured maternal circulating total
serum testosterone concentration at 18 wk of gestation
and demonstrated a significant positive association with
early follicular phase circulating AMH in female off-
spring in adolescence (147), but not PCOS per se (151).
Serum AMH is secreted from the granulosa cells within
preantral and small antral follicles and is elevated in both
adolescents (142) and women with a polycystic ovarian
morphology and PCOS (257). During normal pregnancy,
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
879Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
the fetus is protected from maternal androgens by pla-
cental aromatase. However, it is possible that placental
dysfunction may expose the fetus to higher concentra-
tions of androgens, or with the suppression of sex hor-
mone binding globulin by hyperinsulinemia the concen-
tration of free testosterone maybe increased. The associ-
ation of bisphenol A (BPA) as a potential environmental
cause of PCOS is discussed in section V.
It is believed that oocyte developmental competence and
the embryos resulting from fertilization are altered in
women with PCOS, compared with women without
PCOS (91). There are multiple serum and follicular fac-
tors that are reportedly altered in women with PCOS that
may be responsible for this poor embryonic development
and reduced implantation (summarized by Qiao and
Feng, see FIGURE 3 and TABLE 2), although it is not clear
whether this is associated with an increase in the rate of
embryo aneuploidy (268). Not only is the systemic and
follicular environment different in PCOS, the gene ex-
pression profile of oocytes derived from women with
PCOS are distinctly different (91). These genes relate to
signal transduction, transcription, RNA and DNA pro-
cessing, and the regulation of the cell cycle [summarized
by Dumesic and Abbott (91)]. Of particular importance
in the acquisition of oocyte developmental competence is
GDF-9 expression, which is reduced in the oocytes of
women with PCOS (342).
The ovulatory disorder is frequently exacerbated by hy-
perinsulinemia, which is present in well over 50% of
women with PCOS (225), and is further accentuated by
central obesity, and hence lifestyle modification pro-
grams should be the first intervention strategy (88). The
elevated endogenous serum insulin promotes ovarian an-
drogen secretion, via IGF-I receptor activation of theca
cell androgen secretion (317) and perturbs folliculogen-
esis (292). Second-line therapies consist of using the in-
sulin sensitizer metformin, which leads to a reduction in
androgen secretion, by effects on steroidogenic acute reg-
ulatory protein and 17

-hydroxylase, and inhibits FSH
and induces aromatase activity in granulosa cells (277).
Specifically metformin increases insulin sensitivity by de-
creasing gluconeogenesis and lipogenesis and enhancing
glucose uptake by the liver, skeletal muscle, and adipose
tissue (231). Other approaches are the use of the selective
estrogen receptor modulator clomiphene citrate which
leads to increased pituitary FSH secretion, exogenously
administered FSH itself on an incrementally increasing
regime according to response (311), and the use of aro-
matase inhibitors which lead to increased pituitary FSH
secretion by negative feedback in response to the reduced
estradiol production (155, 197, 220–222). A systematic
review of the pharmacological interventions for women
with PCOS was performed in 2011 and updated in 2015,
and the findings are listed below (9, 341) (see TABLE 3).
The purpose of these therapies is to induce monofollicu-
lar ovulation in the anovulatory woman, or to overcome
a subtle progesterone deficiency in the luteal phase of the
menstrual cycle, under strict ultrasound and serial serum
estradiol assessment to ensure single follicle development
and adequate endometrial thickness, and to prevent con-
ceiving a multiple gestation (23). This approach often
requires the initiation of ovulation when the dominant
follicle size has reached 18 mm, by the use of exogenously
administered hCG, as an LH substitute, due to the close
homology of the beta chains (37). However, women with
PCOS may ultimately be required to undergo IVF treat-
ment, either as they have been unsuccessful with the
treatment to date or they are required to embark on IVF
FIGURE 3. Intra- and extra-ovarian factors that are as-
sociated with the pathology PCOS that may negatively
influence oocyte and subsequent embryo quality. [From
Qiao and Feng (237), with permission from Oxford Uni-
versity Press.]
ROGER J. HART
880 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
treatment as either their fallopian tubes are compromised
or their partner has suboptimal semen parameters. If a
woman embarks on IVF treatment, she is at particular
risk of developing an idiosyncratic reaction called ovar-
ian hyperstimulation syndrome (OHSS) (267).
OHSS is triggered by the systemic release of inflamma-
tory cytokines, particularly VEGF which leads to endo-
thelial cell damage and increased vascular permeability
and the rapid development of ascites, and potentially
pleural and pericardial effusion (338). OHSS is a signif-
icant cause of morbidity and in Australia and New Zea-
land is reported to complicate 0.6% of IVF cycles (206).
Adjuvant therapies that have been demonstrated to sig-
nificantly reduce the incidence are the use of an GnRH
antagonist for pituitary downregulation (378), particu-
larly with the use of an GnRH agonist trigger (381), the
VEGF receptor blocker cabergoline (338), and by comb-
ing the ovarian stimulation with metformin administra-
tion (346). Other strategies include using low doses of
gonadotrophin drugs for stimulation or omitting com-
pletely (8), cancelling the IVF cycle prior to oocyte re-
trieval, omitting the gonadotrophin drugs for a few days–
“coasting” (75) and not proceeding to an embryo trans-
Table 2. Factors in serum and follicular fluid of patients with PCOS with an impact on the quality of the oocytes and embryos, oocyte
fertilization, and the outcome of pregnancy
Factors Serum Level
Follicular
Fluid
Level
Oocyte
Quality
Fertilization
Rate
Embryo
Quality
Pregnancy
Rate
Activin 22
AMH 111or 21or ⬃or 21or ⬃1or ⬃
Epidermal growth factor 1
Fibroblast growth factor 1or 21or 2⬃or 2⬃⬃
Follistatin 11
Brain-derived neurotrophic factor 1
Bone morphogenic protein-15 11 1 1
Estradiol 222 2 2
Follicular fluid meiosis-activating sterol 11 ⬃⬃
Growth differentiation factor-9 12
Homocysteine 112 2 2
Insulin-like growth factorsI&II 222
IGF binding protein 112
Interleukin-12 22 2 2
Interleukin-13 22 2 2
InhibinA&B 2or ⬃⬃
Corticotrophin releasing hormone 22
Leptin 112 2 22
Leukemia inhibitory factor 22 2 2 2
Malondialdehyde 112 2 2
Matrix metalloproteinase 2/9 1or ⬃
Nerve growth factor 1or 2
Renin 221 1 1
Resistin ⬃⬃⬃ ⬃ ⬃
Reactive oxygen species 12 2 2 2
Soluble Fas 222
sFas ligand 112
Superoxide dismutase 2or ⬃2or ⬃
Total antioxidant capacity 222 2 22
Testosterone 112
Tissue metalloproteinase1&2 2or ⬃
Tumor necrosis factor 112 2 22
Vascular endothelial growth factor 2or 12or 12 2or ⬃22
Visfatin 1⬃
1, Increases or positive impact; 2, decreases or negative impact; ⬃, similar; blank, no data. All data are as
compared with controls patients without PCOS. [Modified from Qiao and Feng (237), with permission from
Oxford University Press.]
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
881Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
fer (so-called “freeze-all” approach) (284). A further
innovation is to use a different stimulation approach
called in vitro maturation of oocytes (IVM) where no
final trigger for oocyte maturation is administered prior
to oocyte retrieval and oocyte maturation is performed in
the laboratory with some centers reporting similar preg-
nancy rates as their standard IVF approach (362) without
the risk of OHSS. The evidence for some of the therapeu-
tic interventions employed for women with OHSS is not
robust and hence for the prevention of OHSS clinicians
are advised to follow guidelines provided by con-
sensus statements after systematic review of the literature
(189).
The deranged metabolic environment frequently present in
women with PCOS is believed to lead to altered decidual
trophoblast invasion, placental development and endovas-
cular changes at the site of implantation (247), and in-
creased circulating markers of oxidative stress (229). Pro-
portionate to the degree of hyperinsulinemia and hyperan-
drogenemia, women with PCOS have abnormalities of
homocysteine metabolism (130), correctable with adequate
folate intake. Other systemic changes prevalent in women
with PCOS, influencing conception and miscarriage, are an
elevated serum plasminogen activator inhibitor-1 (124) and
an abnormal expression of some molecular markers with
the endometrium, including insulin-like growth factor
binding protein-1, glycodelin, homeobox protein (HOXA
10), and a endometrial progesterone resistance (53, 249,
295, 306). The consequences of this disturbed systemic and
endometrial environment is a reduced chance of concep-
tion, an increased risk of miscarriage, and in pregnancy a
predisposition to growth restriction, preeclampsia, and pre-
maturity (41, 87, 269).
C. Fallopian Tube Function
1. Disorders of ciliary action
The fallopian tube as a conduit for sperm and embryos
relies on effective cilial activity to perform these tasks. The
fallopian tube cilia can be affected by the environment,
mainly related to infection and inflammation; however, a
primary disorder of ciliary structure and function will also
lead to impaired tubal transport and a predisposition to
ectopic gestation implantation and subfertility. Primary cil-
iary dyskinesia (PCD) is associated with recurrent respira-
tory tract infections and potentially situs invertus. This is a
very heterogeneous condition as there exist many differing
structural and functional defects within the cilia, related to
as yet many unidentified genetic defects; however, over 20
genetic mutations related to axonemal-dynein function
have been identified (188) and in addition mutations within
the retinitis pigmentosa GTPase regulator on the X chro-
mosome have been found in men with this condition and
PCD. Due to the tissue specific expression of the multiple
genes responsible for PCD, not all women with the respi-
ratory phenotype of PCD have impaired fallopian tube
ciliary function, and spontaneous conceptions have been
reported (99).
Table 3. Pharmacological options for women with PCOS who are trying to conceive
Evidence Statement Level of Evidence
Clomiphene citrate should be the first-line pharmacological therapy to improve fertility outcomes in women
with PCOS and anovulatory infertility, with no other infertility factors.
A
The risk of multiple pregnancy is increased with clomiphene citrate use, and monitoring is recommended. PP
Metformin should be combined with clomiphene citrate to improve fertility outcomes rather than
persisting with further treatment with clomiphene citrate alone in women with PCOS who are
clomiphene citrate resistant, anovulatory, and infertile with no other infertility factors.
A
Metformin could be used alone to improve ovulation rate and pregnancy rate in women with PCOS who
are anovulatory, have a body mass index ⫽30 kg/m, and are infertile with no other infertility factors.
B
If one is considering using metformin alone to treat women with PCOS who are anovulatory, have a body
mass index ⫽30 kg/m, and are infertile with no other infertility factors, clomiphene citrate should be
added to improve fertility outcomes.
A
Gonadotrophins should be the second-line pharmacological therapy. B
Laparoscopic surgery in women who are overweight or obese is associated with both intraoperative and
postoperative risks.
PP
Letrozole, under caution, could be offered as a pharmacological treatment for ovulation induction
indicated for infertile anovulatory women with polycystic ovary syndrome with no other infertility factors.
A
A, body of evidence can be trusted to guide practice; B, body of evidence can be trusted to guide practice
in most situations; C, body of evidence provides some support for recommendation but care should be
taken in its application; PP, evidence not sought. A practice point has been made by the guideline
development group where important issues arose from discussion of evidence-based or clinical consensus
recommendations. Adapted from Evidence Based Guidelines for the Assessment and Management of
Polycystic Ovary Syndrome. Melbourne, Australia: Jean Hailes for Women’s Health on behalf of the PCOS
Australian Alliance, 2015.
ROGER J. HART
882 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
Cystic fibrosis (CF) is a condition characterized by ab-
normal mucus secretion due to being homozygous, or a
compound heterozygote, for one of the ⬎1,900 muta-
tions in the CF transmembrane regulator gene, limiting
chloride and bicarbonate secretion (3). In addition to the
associated symptoms of ovulatory disorder relating to
poor general health and low body fat levels, CF is asso-
ciated with female subfertility due to a direct effect on the
epithelial cells of the reproductive tract, although not
directly on cilial action. The thick cervical mucus impairs
sperm penetration, although it is believed that the effect
on the uterine cavity and the fallopian tube function is
less significant (3), although the influence on bicarbonate
metabolism may lead to problems with sperm capacita-
tion within the fallopian tube (55).
2. Inflammatory disorders: endometriosis and
infection
Inflammation within the fallopian tube due to extrinsic
infection, salpingitis, or salpingitis isthimica nodosa
(nodular thickening or scarring of the fallopian tubes
within the isthmic fallopian tube) may lead to an activa-
tion of an inflammatory cascade via the innate immune
system, within the tubal fluid and leading to fallopian
tube damage from the inflammatory response, and inhi-
bition of ciliary beat frequency (250). Salpingitis may
lead to distal occlusion of the fallopian tube and perma-
nent deciliation. The most common cause of salpingitis is
pelvic inflammatory disease (PID), due to sexually trans-
mitted infection. Hence, the risk factors for PID are mul-
tiple sexual partners, young age, smoking, and illicit
drugs (223). The most common infection agent is Chla-
mydia trachomatis (CT), and it is believed that genetic
polymorphisms within the Toll-like receptor genes may
increase the susceptibility to upper genital tract infection,
as not all women with cervical chlamydial infection will
have detectable infection within the endometrium and
fallopian tubes (223). CT appears to initiate a fallopian
tube inflammatory response via the innate immune in-
flammatory response and also an adaptive T-cell re-
sponse (76), and ongoing chlamydial infection is com-
mon (204), leading to continued influx of inflammatory
cells, damage to host epithelium, scarring, and ultimately
fibrosis and progressive tubal scarring. Furthermore, CT
causes a direct cytotoxic effect on the fallopian tube mu-
cosa (204). With increased screening of young women by
cervical swabs for CT, and commencement of prompt
treatment of early cervical infection in most Western
countries, it is proposed that this will lead to a reduction
in the incidence of PID, and consequently fallopian tube
damage (244). Other fallopian tube pathogenic organ-
isms include Neisseria gonorrhoea, which was tradition-
ally the most common responsible pathogen in Western
countries. In many instances the pelvic infection and the
resulting fallopian tube damage may be exacerbated by
the anaerobic bacterial agents often associated with bac-
terial vaginosis (223).
Endometriosis is another pathological pelvic inflammatory
process that is known to inhibit ciliary beat frequency
(203), and in severe disease leads to significant pelvic scar-
ring and pelvic anatomy distortion, and hence is associated
with doubling in the incidence of ectopic pregnancy (152),
in addition to the negative impact on endometrial receptiv-
ity (discussed later). PCOS has also been associated with a
doubling of the rate of ectopic pregnancies, although no
mechanism has been proposed (140).
3. Other gynecological conditions
Gross anatomical distortion of the fallopian tubes, either
due to extrinsic compression by large uterine fibroids and
ovarian cysts, or by mechanical distortion of the fallopian
tube itself by severe adhesions, fibrosis due to chronic in-
flammation may lead to compromise of the fallopian tube
contractility and potentially cilial function. The implica-
tions of fibroids for conception and the therapeutic options
for intervention have undergone systematic review (186),
and a summary of the most recent review of the literature is
listed in TABLE 4. The medical and surgical interventions for
women trying to conceive with endometriosis have under-
gone systematic review, and the overview published in 2014
(44) is summarized in TABLE 5.
Table 4. Royal Australian and New Zealand College of Obstetricians and Gynaecologists guidelines for the management of fibroids,
derived after systematic review of the literature
The majority of studies relate to the impact of fibroids on fertility outcomes of women undergoing IVF treatment
1) Subserosal fibroids (the majority of the fibroid is on the exterior of the uterus) do not appear to have an effect on fertility outcomes.
2) Intramural fibroids (within the muscle wall of the uterus) may be associated with reduced fertility and an increased miscarriage
rate; however, there is insufficient evidence to determine whether myomectomy will improve fertility outcomes.
3) Submucosal fibroids (a portion of the fibroid is within the endometrial cavity) are associated with reduced fertility and an increased
miscarriage rate; hysteroscopic resection of submucosal fibroids is likely to improve fertility outcomes. (Quality of studies is poor,
and further research is required.)
The size of the fibroid, the number, and their location within the uterus may impact on the utility of myomectomy.
Adapted from the RANZCOG guidelines, on behalf of the CREI Consensus Expert Panel on Trial evidence group.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
883Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
D. Embryonic Development
The human embryo is prone to chromosomal errors dur-
ing development. It is believed that at the blastocyst stage
(reached at the 5th or 6th day after fertilization) that
three-quarters of embryos of a 30-yr-old woman will be
normal; however, at 40 years of age, only 40% are nor-
mal (109). The centrosome, responsible for the subse-
quent spindle and microtubule development within the
embryo, is derived from the sperm, hence men with sig-
nificant impairment in spermatogenesis, and oligosper-
mia, may be responsible for higher rates of aneuploidy
(the gain or loss of whole chromosomes) within the sub-
sequent embryo (209, 339). However, the most common
cause of embryo aneuploidy is related to female age as the
oocyte has been in a stage of arrested meiotic develop-
ment in prophase since early fetal life, hence as a woman
ages and is exposed to reactive oxygen species within the
environment, there is a progressive loss of cohesion mol-
ecules that hold sister chromatids together, the incidence
of aneuploidy increases exponentially (114), particularly
the chiasmata proximal to the telomere (208). This is
exacerbated by deterioration in cytoplasmic mitochon-
dria and mRNA stores (369). This all leads to the subse-
quent substantial increase in the rate of miscarriage with
increasing age (FIGURE 4).
A significant initiator of aneuploidy which is independent
of maternal age is the lack of chromosomal recombina-
tion in the fetal stages of meiosis. To prevent chromo-
some missegregation, it is essential that at least one cross-
over (recombination) is formed by recombination of each
Table 5. Medical and surgical interventions for women with endometriosis seeking fertility treatment
Intervention Influence of Intervention
Number of
Studies
Grade Quality of
Evidence Assessment of Studies
Ovulation suppression versus
placebo (4)
There is no evidence of benefit in
the use of ovulation
suppression in subfertile
women with endometriosis
who wish to conceive.
11 (557
patients)
Low Lack of explanation for
allocation
concealment and
blinding.
Long downregulation of pituitary
with a GnRH agonist versus no
agonist (5)
The administration of GnRH
agonists for a period of 3–6
months prior to IVF in women
with endometriosis increases
the odds of clinical pregnancy
fourfold.
3 (165
patients)
Very low Included studies
lacked blinding and
explanation of
allocation
concealment. There
was some
imprecision.
Excisional versus ablative surgery
for endometriomata (3)
Excisional surgery for
endometriomata provides a
more favorable outcome than
drainage and ablation with
regard to subsequent
spontaneous pregnancy in
women. However, in women
who may subsequently
undergo fertility treatment,
insufficient evidence exists to
determine the favored surgical
approach.
2 (88
patients)
Low Included studies
lacked blinding and
there was some
imprecision.
Laparoscopic ablation or excision
versus diagnostic laparoscopy
(2)
Laparoscopic surgery for mild
and moderate endometriosis
was associated with a doubling
live birth rate than diagnostic
laparoscopy alone.
3 (528
patients)
Moderate Two studies did not
adequately describe
randomization; one
study was at high
risk of attrition bias.
Extract adapted from Brown and Farquhar (44).
50
45
40
35
30
25
20
15
10
5
0
Spontaneous abortion, %
2220 2624 3028 3432 36 4038 4442 46 5048 5452
Patient Age
FIGURE 4. Spontaneous abortion rates for assisted reproductive
technology pregnancies conceived with freshly fertilized embryos by
source of oocytes used and maternal age. The solid line indicates
pregnancies conceived with the patient’s oocytes, and the dashed
line indicates pregnancies conceived with donor oocytes. [From
Schieve et al. (298), with permission from Wolters Kluwer Health,
Inc.]
ROGER J. HART
884 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
pair of homologous chromosomes (for review, see Ma-
cLennan et al., Ref. 208). It is estimated that ⬃30% of
Down syndrome births are as a result of lack of recom-
bination between homologous chromosomes 21, as the
most common chromosomes prone to lack crossover
within fetal oocytes are chromosomes 21 and 22 (58).
Furthermore, the distance from the centromere that the
single cross-over are located also determines the risk of
aneuploidy, and again appears independent of maternal
age. Chiasmata close to the telomere are at greatest risk
of being lost as there exists less cohesion between the
chromosomes predisposing to missegregation.
Embryonic mosaicism is a common finding resulting from
abnormal chromosome segregation either within meiosis or
mitosis due to 1) a failure of the process whereby microtu-
bules pull the divided chromosomes towards their respec-
tive spindles prior to cytokinesis and cell division “non-
disjunction”; 2) “anaphase lag” whereby a chromatid is not
incorporated into the nucleus during mitosis, thus creating
two-cell lines, one monosomic for the chromosome and the
other disomic for the chromosome; and 3) chromosomal
gain by “endo-replication” (339). A further process that
leads to a mosaic embryo is uniparental disomy where an
embryo has two copies of either a maternally derived or
paternally derived chromosome, rather than one from each
(339). The earlier in development that mosaicism develops,
the more significant is the implication for the developing
embryo; however, it is possible that abnormal cells can be
forced away from the embryo, effectively being “selected
against” (330, 339).
The recent innovation of embryo morphokinetics using
time-lapse imaging systems for use with IVF laboratories
has enabled scientists to document abnormal embryonic
development (216). However, individual laboratories
will have differing culture systems, hence may have dif-
ferent findings, although the staging system proposed by
Meseguer is one of the most widely used (216). This
group demonstrated that the most predictive parameters
of embryo implantation potential were the time between
division to two cells and division to three cells, the time
between division to three cells and subsequent division to
four cells, and the time of division to five cells (114, 216).
They also demonstrated that the abnormal features of
uneven blastomere size at the two-cell stage and abrupt
cell division to three or more cells, and multi-nucleation
at the four-cell stage resulted in embryos that would not
implant. Incorporation of these features into a random-
ized controlled trial to improve IVF outcomes demon-
strated an increased on-going pregnancy rate and re-
duced miscarriage rate when embryos were selected using
this algorithm (287).
The abnormal embryo may also display an abnormal me-
tabolism that can be detected within the culture media
within the IVF laboratory, either by proteomics of the se-
cretome or by metabolomics to study the rate of consump-
tion of carbohydrates, oxygen, and amino within the cul-
ture media and consequently assist with improved embryo
selection during IVF treatment (114, 115).
E. Implantation
1. Systemic
It is generally believed that severe systemic illness, such as
sepsis or severe renal disease, will prevent embryonic im-
plantation, although infection is an unusual cause of early
pregnancy failure (315, 318). A comprehensive list of all
systemic conditions that have been demonstrated to have a
significant negative influence on embryonic implantation is
difficult to compile; however, conditions such as unstable
diabetes (64), subclinical hypothyroidism (356), periodon-
tal disease (144), and uncontrolled celiac disease (343) have
been demonstrated to reduce rates of conception, and it is
believed that low serum vitamin D (199) and active auto-
immune conditions (304) are also associated with a reduced
chance of conception, and strategies to control these condi-
tions may improve conception chances. Due to their high
prevalence and ease of correction of the abnormality, celiac
disease and subclinical hypothyroidism are discussed fur-
ther.
2. Celiac disease
In a population of women experiencing unexplained infer-
tility or recurrent miscarriage, celiac disease is five times
more prevalent than in the general population (343). A
meta-analysis of patients with celiac disease found that the
risk of miscarriage is 40% greater with increased risks in the
pregnancy for growth restriction and premature delivery,
and the effect is tempered if a gluten-free diet is observed
(343).
3. Subclinical hypothyroidism
Overt thyroid disorder must be appropriately managed
prior to conception; however, more subtle perturbations in
thyroid function are also associated with reproductive dis-
order. Approximately 1 in 25 women have subclinical hy-
pothyroidism, and thyroid antibodies are present in up to
one in eight of women (153). The presence of thyroid anti-
bodies in a woman with normal thyroid function is believed
to be associated with difficulty conceiving, recurrent im-
plantation failure of embryos, and early pregnancy loss,
potentially due to an unrecognized thyroid hormone defi-
ciency or due to a potential autoimmune cause (359). Treat-
ment of subclinical hypothyroidism is believed to poten-
tially improve embryo development and is recommended
for women prior to conception; however, whether to treat a
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
885Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
woman prior to conception, who is euthyroid in the pres-
ence of thyroid antibodies, is contentious (77).
4. Thrombophilia
It has been unclear whether an inherited thrombophilia
leading to microthrombi within the decidua are associ-
ated with implantation failure of the embryo as the inter-
villous spaces are not developed until 10 wk of gestation
(164). Although it is tempting to speculate that perturba-
tions in the clotting system may influence implantation
and early embryonic development, for example, factor
XII gene expression is increased in endometrial stromal
cells during in vitro decidualization, this is believed to
lead to an activation of the kallikrein-kininogen-kinin
system during the implantation of human embryos (175).
Furthermore, plasminogen activator inhibitor (PAI-1) is
a significant regulator of the thrombotic/fibrinolytic pro-
cess in early pregnancy and is elevated in insulin resistant
women with PCOS (17). Other changes within the clot-
ting process around the time of embryo implantation are
an increase in tissue factor, an activation of the extrinsic
coagulation cascade, and an increase in requirement for
methylation from folic acid. Hence, inherited abnormal-
ities of the clotting system such as the prothrombin gene
mutation, factor V Leiden, methyltetrahydrofolate re-
ductase (MTHFR), and protein S and C and anti-throm-
bin III would be expected to have a significant influence
on implantation and the early pregnancy (164). Although
the appropriate treatment is not clear, except for women
with the anti-cardiolipin syndrome, where appropriate
treatment must commence at conception is widely ac-
cepted (274).
Due to the effect of heparin on modulating endometrial
receptivity and potentially improving implantation, in
addition to its inhibition of factor Xa and thrombin, it
has been proposed as a treatment for women with im-
plantation failure undergoing IVF treatment (264). Hep-
arin reduces the expression of E-cadherin and promotes
trophoblast invasion and proliferation into the endome-
trial cells potentially improving implantation (95). A sys-
tematic review of women with recurrent implantation
failure undergoing IVF demonstrated that use of adjunct
low-molecular-weight heparin improved live birth rates,
and the rate of miscarriage was substantially reduced
compared with the control group (264). The authors ad-
vised caution with the interpretation of the results due to
the low numbers in the studies.
5. Natural killer cells
There has been substantial interest in the assessment of
blood and uterine natural killer cell populations in
women with poor embryo implantation (305). Evidence
would appear to suggest that women with unexplained
implantation failure may have an abnormal population
of natural killer (NK) cells in the blood and in the endo-
metrium in the mid-luteal phase of the menstrual cycle
(293), although strategies to improve the systemic and
endometrial environment to facilitate conception have
not been proven (305).
6. Endometrial and myometrial (endometriosis,
leiomyomas, hydrosalpines, PCOS, obesity,
endometrial polyps)
As evidenced by IVF success rates, embryonic implantation
potential is decreased in the presence of endometriosis (25),
leiomyomas (145), dilated fallopian tubes (hydrosalpines)
(332), and PCOS (346) and have been linked to reduced
endometrial expression of HOXA10 and HOXA11 (48). In
addition, women with endometriosis have reduced expres-
sions of endometrial integrin
␣
v

3
and LIF, hypermethyl-
ation leading to silencing of the HOXA10 gene and endo-
metrial progesterone resistance (48) leading to a reduction
in the chance of conception which may potentially be im-
proved by surgical intervention (90) or by use of prolonged
downregulation with a GnRH analog prior to the initiation
of an IVF cycle (291). Similarly to integrin
␣
v

3
expression
normalizing with surgical intervention in the presence of
endometriosis (200), the surgical removal of hydrosalpin-
ges (salpingectomy) will restore the expression of integrin
␣
v

3
and LIF improving conception (332). Leiomyomas are
common benign smooth muscle tumors of the myome-
trium, if they distort the endometrial cavity, a submucosal
leiomyoma, implantation may be inhibited by a purely me-
chanical method. However, leiomyomas located in the
body of the uterus (myometrium), intramural leiomyomas,
may also mechanically inhibit conception implantation, but
overlying endometrial atrophy and an altered endometrial
environment has been demonstrated (48). Surgical inter-
vention is believed to improve implantation potential for
women with a submucosal leiomyoma; however, the case
for treating small intramural leiomyomas to improve con-
ception has yet to be proven (186). Due to the estrogenic
stimulation required for the growth of leiomyomas, it has
been speculated that their growth may be associated with
environmental estrogenic stimulation, such as bisphenol A
(BPA) (309); however, an observational study of the expo-
sure to BPA, phthalates, and five ultraviolet filters was un-
able to confirm any direct association (262).
The endometrium of women with PCOS is abnormal as it is
exposed to lower levels of serum progesterone in the luteal
phase, the consequences of ovulatory disorder prevalent in
women with PCOS. Furthermore, the endometrium is po-
tentially exposed to elevated levels of serum IGF-I and se-
rum androgens (140). Elevated IGF-I can lead to endome-
trial hyperplasia and a predisposition to endometrial malig-
nancy; however, in contrast, the high serum androgens,
often present in women with PCOS, can induce endometrial
atrophy and amenorrhea. In addition, there is an increase in
ROGER J. HART
886 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
estrogen receptor
␣
, an increase in 17

hydroxysteroid de-
hydrogenase type 1, and a reduction in type 2 (promoting a
greater local concentration of estradiol), in association with
a degree of progesterone resistance promoting endometrial
hyperplasia and leading to a decrease in fertility (306). In
women with PCOS, there is a decreased endometrial secre-
tory phase expression of selectins, integrin
␣
v

3
, and
HOXA10 reducing the implantation potential of an em-
bryo (306). A compounding factor for many women with
PCOS is obesity which has a synergistic effect to reduce the
chance of conception (29), leading to a greater perturbation
in endometrial gene expression (29), as demonstrated by the
association of an increase in BMI being associated with a
reduced chance of conception using the oocyte donor model
(81) [for a review, see Schulte et al. (302)]. However, life-
style interventions with diet and exercise will lead to alter-
ations in endometrial gene expression (348), although it is
not clear whether the improved conception rates noted due
to lifestyle interventions are due to an endometrial effect, an
oocyte effect, or a combination of both.
Common benign overgrowths of the endometrium known
as endometrial polyps may interfere with sperm transport
and embryo implantation to mechanically inhibit concep-
tion, but they may promote the abnormal expression of
markers of implantation; upon surgical removal of the en-
dometrial polyps, documented increases in endometrial se-
cretion of IGFBP-1, TNF-
␣
, and osteopontin have been
noted, assisting implantation (31).
7. Embryonic
With the advent of genetic testing of blastomeres from em-
bryos of women undergoing IVF treatment, it has become
evident that a significant cause of embryos failing to implant
is due to chromosomal rearrangements developing within
the embryo as described above (114). With the ability to
perform a low-resolution genome-wide survey of either sin-
gle blastomeres from a three-day-old embryo or by the
study of several cells from the trophectoderm of a five- or
six-day-old blastocyst, it has become evident that in addi-
tion to the common occurrence of aneuploidy within the
embryo, usually arising during meiosis, the embryo is pre-
disposed to segmental chromosomal imbalances (372)
which arise during programmed DNA breakage and repair
by homologous recombination during prophase I of meiosis
(360). These rearrangements may lead to a failure to de-
velop and implant, but also lead to phenotypic variability
and hence ultimately genome evolution [for a detailed de-
scription of the origin of chromosomal rearrangement, see
Voet et al. (360)]. Hence, in IVF programs the majority of
apparently morphologically normal embryos fail to implant
as aneuploidy is such a frequent occurrence, occurring more
frequently in an older woman, and a woman with a history
of failed embryonic implantation (371).
A less frequent cause of unsuccessful embryo development
and implantation, or implantation then subsequent early
pregnancy failure, is the prevalence of a chromosomal
translocation within either one of the couple trying to con-
ceive. The prevalence of a chromosomal translocation
within a couple with a history of failed implantation under-
going IVF treatment is 1.4%, substantially less than the
4.1% prevalence within couples with a history of recurrent
miscarriages (329), but significantly greater than the 0.2
and 0.3% prevalence in neonatal and general infertility pa-
tients, respectively (329). Chromosomal translocations
within a phenotypically normal prospective parent can be
either a balanced reciprocal translocation, whereby there is
an mutual exchange of genetic material from the distal end
of two different chromosomes, or a Roberstonian translo-
cation, wherein there is a fusion of the long arm of two of
the acrocentric chromosomes (chromosomes 13, 14, 15, 21,
and 22) and a resulting loss of the short arms. In both
instances, the gametes resulting from these chromosomes
can be “balanced” containing the appropriate amount of
genetic material, or “unbalanced” and contain an excess or
deficit of genetic material leading to miscarriage or a risk of
congenital malformations in the offspring.
F. Early Pregnancy Failure
The causes of early pregnancy failure overlap with the
causes of embryo implantation failure and hence are not
reiterated here. The discussion is limited to a description of
possible associations of genetic variations which may influ-
ence implantation and predispose to early pregnancy loss.
1. Genetic polymorphisms
Genetic polymorphisms associated with recurrent early
pregnancy loss suggest that genes regulating oxidative stress
may be involved (353). Single nucleotide polymorphisms of
genes associated with oxygen free radical metabolism,
ABCB1,COMT,GPX4, and OGG1, have been reported to
lead to a doubling of the risk of recurrent miscarriage (177).
In addition, polymorphism of genes that regulate the com-
plement cascade, such as membrane cofactor protein and
C4 binding protein, have a putative role in recurrent early
pregnancy loss (353). HLA-G, part of the major histocom-
patibility complex class I group, has been linked to the
success of IVF and is believed to have an immune modula-
tory role, and potentially lower expression of HLA-G is
associated with reduction in embryo implantation and early
pregnancy failure (353).
MTHFR is responsible for the synthesis of 5-methyltetra-
hydrofolate required to allow the conversion of homocys-
teine to methionine. Patients with the 677TT genotype are
reported to have up to a threefold increased risk of recur-
rent early pregnancy loss (233).
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
887Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
Due to the putative belief that an imbalance of the tolerant
[T helper cell1 (Th1)] over the prorejection [T helper cell 2
(Th2)] cytokines within the adaptive immune system for
women predisposed to reproductive failure these cytokines
have been studied. Of note, Th1 cytokines include IL-2 and
interferon-
␥
(INF
␥
), and Th2 include IL-4, IL-5, IL-6, IL-
10, and IL-13. Several haplotypes leading to a reduction in
serum IL-18 levels have been associated with a significant
increase in recurrent early pregnancy loss (4).
IV. LIFESTYLE INFLUENCES INFLUENCING
OVULATION, FALLOPIAN TUBE
FUNCTION, IMPLANTATION, AND
MISCARRIAGE
A. Delayed Childbearing
It is well established that oocyte quality deteriorates with
advancing reproductive age, in addition to an increase in
ovulatory disorder, a reduction in ovulatory frequency, an
impaired luteal phase and also premature recruitment of
follicles, all leading to reduced conception rates.
1. Ovulatory frequency
As a woman ages, in line with a progressive reduction in
follicle number, there is a progressive reduction in ovula-
tory frequency. This situation is also applicable to the
woman with premature ovarian insufficiency where there is
a reduction in functioning granulosa cells, leading to a re-
duction in secretion of inhibin B, further leading to a reduc-
tion in pituitary feedback there is an increase in late follic-
ular phase and then early follicular phase FSH secretion
(45, 46). This may lead to premature initiation of follicular
recruitment and early ovulation, or if there are no follicles
to recruit, a low rise in inhibin B and estradiol and a pro-
longed anovulatory menstrual cycle. Further follicular de-
pletion leads to an increase in the frequency of anovulatory
cycles. One study demonstrated that in the last 10 cycles
before the menopause, approximately only one-third of the
cycles will be ovulatory (190). The consequent impact on
the chances of conception are the fact that that with prema-
ture follicular recruitment the timing of intercourse may be
more difficult to plan and with impaired estradiol and pro-
gesterone secretion by the corpus luteum in the luteal phase,
“luteal phase insufficiency” (245), there will be a further
impediment to embryo implantation, often compounded by
a poorer quality oocyte.
2. Oocyte quality
The deterioration in oocyte quality is related to a predispo-
sition to the generation of aneuploid embryos caused by
chromosomal segregation errors and abnormal spindle de-
velopment, epigenetic modifications (120), and a deteriora-
tion of the oocyte cytoplasm (ooplasm), related to a de-
crease in mitochondria, a reduction in their quality and an
increase in oxidative stress within the ooplasm (2), which
can lead to promote free radical formation leading to the
modification of intracellular proteins, lipids, and nucleic
acids macromolecules (92) and alterations in mRNA (108).
Consequently, female fertility starts to rapidly decline in a
woman beyond 37 years of age, as manifest by the success
rates of IVF treatment (206) and also by measures of natu-
ral fertility (192). The causes of the age-related decline in
embryo quality are detailed below.
A)ANEUPLOIDY. As described previously as the woman’s age
increases, the chiasmata more proximal to the telomere be-
come more susceptible to missegregation as there is believed
to be less cohesion between the sister chromatids, due to an
age-related loss in cohesin and shugoshin cohesions pro-
teins, as they are produced in fetal life and deteriorate with
time and exposure to reactive oxygen species (ROS) (136,
208). Another reported peculiarity of the human oocyte is
the slow meiotic spindle formation compared with the
mouse, and consequent predisposition to spindle instability
and anaphase lag whereby due to slow segregation of the
chromosomes an aneuploid chromosomal constitution may
arise upon cytokinesis (154). Furthermore, a reduction in
supply of ATP from a progressive reduction in mitochon-
drial function will lead to a reduction in spindle formation,
microtubular activity, and polar body extrusion, promoting
aneuploidy.
B)OOCYTE MITOCHONDRIAL FUNCTION. Mitochondria are ma-
ternally inherited, and the oocyte has the largest number of
mitochondria (⬃200,000) and copies of mitochondrial
DNA (mtDNA) of any cell. They are essential ROS scaven-
gers and for the generation of ATP for the cellular processes
including cortical granule extrusion, polar body formation,
spindle formation, chromosome segregation, and cytokine-
sis. As the selection of mitochondria and mtDNA during
oocyte development is a random process, the possibility
arises for the amplification of mutated mtDNA during
oocyte maturation and mitochondrial expansion (32).
Upon fertilization mtDNA replication ceases, leading to a
progressive dilution of mtDNA within the blastomeres dur-
ing embryonic development, until blastulation when repli-
cation can resume. A reduced pool of mtDNA within the
oocyte has been associated with a reduced fertilization po-
tential (276) and also a reduced ovarian reserve (213). Fur-
thermore, ageing is associated with an adverse effect on the
mitochondria [reviewed by Schatten (297)], leading to an in
increase in mtDNA damage and mutations with mtDNA,
structural abnormalities with the mitochondria, a reduction
in ATP synthesis, and an increase in ROS production (57,
297). Putative methods to improve mitochondrial function
are the oral administration of CoQ10 (32), an electron
transporter involved in the transport of electrons within the
respiratory chain within the mitochondria (32), to assist
ROGER J. HART
888 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
mitochondrial function or even to introduce donor ooplasm
to “rejuvenate” an oocyte. In the area of embryonic screen-
ing within IVF cycles, it is now feasible to combine an
assessment of aneuploidy within the blastocyst with an as-
sessment of mtDNA content within blastomeres by mi-
croarray analysis (183).
C)EMBRYO METABOLISM. Evidence from animal studies sug-
gest that the metabolism of the developing embryo may
differ according to the age of the mother, as there were
measured differences in the embryo culture media of em-
bryos generated from older mice, compared with those gen-
erated from younger mice (212). If this is translated into
humans, this may lead to a reduced implantation potential
for such embryos independent of aneuploidy. In this murine
model, the offspring were growth restricted, which if repli-
cated in humans may have significant implications for the
child’s long-term health and development (212). To date,
there is limited data to suggest an adverse outcome for
children born from IVF (146). However, these children may
be predisposed to cardiometabolic disorder, which is also
associated with a fetus not reaching its growth potential
(157), which is an interesting observation in view of these
animal model findings, as IVF conceived children tend to be
born to a population of older mothers than naturally con-
ceived children (146).
D)EPIGENETIC ALTERATIONS. There is some evidence of an
alteration in DNA methylation patterns within oocytes de-
rived from older female animals (120), and there is one
report of an alteration of DNA methylation patterns within
metaphase II oocytes derived from women over 38 years of
age compared with women under 35 years of age (133), and
there are reports of a reduction in histone protein deacety-
lation in the metaphase I and II surplus oocytes derived
from older women (355), and an alteration in the expres-
sion of ubiquilin in metaphase II oocytes (131). Hence, the
consensus view is that oocytes derived from older women
may be at risk of epigenetic modification (120).
3. Fallopian tube function
The incidence of ectopic pregnancy increases as a woman
ages, although it is unclear whether this is directly related to
an age-related change in fallopian tube function (273).
B. Dietary Restriction and Over-exercise
As described earlier, an alteration of food intake can have
profound effects on the complex interplay of hormones re-
leased by the gastrointestinal system and neuropeptides in-
fluencing follicular development (97). It is well established
that calorie restriction and excessive exercise lead to a re-
duction in the frequency of ovulation, poor endometrial
development, amenorrhea, and subfertility, with even rec-
reational levels of activity potentially leading to abnormal-
ities of gonadotrophin secretion and ovulatory disorder
without inducing amenorrhea (79). This is due to a suppres-
sion of the hypothalamic-pituitary ovarian axis, due to a
reduction in the systemic stimulatory signals of GnRH re-
lease. As described previously, an alteration of food intake
can have profound effects on the complex interplay of hor-
mones released by the gastrointestinal system and neuro-
peptides influencing follicular development (97). Interest-
ingly, body weight in late adolescence is an important pre-
dictor of fecundity in later life, as women within the Nurses’
Health Study (117) who were underweight at 18 years of
age (BMI under 18.5 kg/m
2
) took an average 25% longer
than normal weight women at 18 years of age to conceive,
suggesting adolescence is a critical time for the program-
ming of the reproductive axis.
The negative consequences of exercise on the chance of
conception are demonstrated by an observational study of
women undergoing their first IVF cycle which showed that
women who exercised for four or more hours per week
were 40% less likely to have a live birth, twice as likely to
have implantation failure, and were more likely to miscarry
(227).
1. Leptin
Leptin is of particular importance in the regulation of the
reproductive axis, as evidenced by restoration of gonado-
trophin secretion with recombinant leptin administration
to over-exercising and underweight women leading to a
restoration of LH pulsatility, ovulation, and menstrual cy-
clicity (364). As adequate GnRH secretion is essential for
normal gonadotrophin secretion, a perturbation in GnRH
pulsatility may lead to impaired ovulation, inadequate
maintenance of a corpus luteum, and hence may predispose
a woman to infertility and early pregnancy loss (337), and
appropriate weight gain may lead to restoration of ovula-
tory cycles (111).
2. Insulin
Circulating concentrations of insulin are related to adipos-
ity, and concentrations are lower in amenorrheic and regu-
larly exercising women with functional hypothalamic
amenorrhea (FHA), and are associated with a reduction in
leptin secretion, although insulin does not have a direct
action on GnRH pulsatility as GnRH neurons do not ap-
pear to express insulin receptors.
3. Gherlin
Ghrelin secretion from the gastrointestinal tract is maximal
when the stomach is empty, it has an inhibitory effect on LH
secretion, and serum levels are greater in regularly exercis-
ing (78), underweight (113), women with functional hypo-
thalamic amenorrhea with abnormal eating habits (300)
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
889Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
leading to disordered ovulation as a consequence of the LH
suppression.
4. Protein YY
Protein YY (PYY) infusion in humans is an appetite sup-
pressor (28), and although animal studies suggest a degree
of sexual dimorphism in the response to PYY, it is believed
that in humans the action of PYY is suppressive on the
reproductive axis (65). Interestingly, in adolescent girls
with anorexia nervosa, the fasting concentrations of PYY
have been reported to be three times greater than those of
girls without anorexia, suggesting that PYY may play a role
in the suppression of appetite and the suppression of the
reproductive axis (219).
C. Stress
Due to central neuronal corticotrophin releasing hormone
(CRH) projection to GnRH cells and CRH-induced

-en-
dorphin inhibition of GnRH secretion, stress may exert a
modulating effect on subsequent pituitary LH and FSH pul-
satility (33, 60), which may be overcome by modified be-
havior restoring ovulation (35). Higher daily reported stress
levels in a cohort of normal healthy women were associated
with a reduction in serum estradiol, LH, and luteal phase
progesterone concentrations as well as a predisposition to
anovulation (299). Furthermore, while it is known that an
elevated serum cortisol concentration is related to FHA,
women with FHA who resume ovulation have serum corti-
sol concentrations similar to eumenorrheic women, sug-
gesting that by reducing stress levels by therapeutic inter-
ventions normal ovulation may return (34). The Nurses’
Health Study demonstrated that working longer hours
(over 40 h/wk) and also lifting heavy loads were associated
with increased time to conceive, suggesting a relation to
tiredness or stress may impact upon conception (116).
D. Obesity
Women who are overweight are less likely to ovulate (240)
and spontaneously conceive (256), and upon conception,
they have an increased risk of miscarriage and are predis-
posed to an adverse pregnancy outcome (15). Obesity ap-
pears to alter the follicular fluid environment (282), the
ooplasm of the oocyte, and leads to a reduction in fertiliza-
tion and impairs embryonic development (92). Evidence
derived from mouse models would suggest that obesity
leads to slower growth and delayed maturation of the
oocyte, epigenetic modifications, increased granulosa cell
apoptosis, and mitochondrial dysfunction within the
oocyte (92, 327, 380). Hence, maternal obesity may exert
germ-line effects by affecting oocyte quality and the meth-
ylation of imprinted genes, and in the mouse model, this has
led to altered methylation patterns within metabolism-re-
lated genes in the oocytes and the liver of the offspring of
obese mice (72, 118). From mouse studies it has been dem-
onstrated that maternal obesity leads to a deterioration in
blastocyst development compared with matched controls;
this effect can be overcome by intervention (218). To help
to determine whether there is an element of harm inflicted
on the oocyte or embryo by exposure to metabolic derange-
ment rather than the intrauterine environment, in an ele-
gant series of experiments, embryos were transferred from
diabetic mice (377) or mice that had been fed a high-fat diet
(202). In this study the embryos that were transferred from
the diabetic mice were more prone to develop intrauterine
growth restriction and congenital malformations, and
when the oocytes from the high-fat diet fed mice were ana-
lyzed, there was a greater incidence of oocyte aneuploidy
and mitochondrial damage (202, 377). A retrospective hu-
man study also demonstrated phenotypic differences in
oocyte morphology, embryonic metabolism, and the rate of
embryonic development in embryos derived from over-
weight women (195).
Population data suggest that obesity is associated with ad-
verse obstetric outcomes such as preeclampsia, gestational
diabetes, premature delivery and stillbirth (303), and con-
genital malformations in the offspring (331). Although it is
believed that an improvement in obstetric and neonatal out-
comes may be achieved by weight loss (13), not all studies
agree (135). The use of bariatric surgery for obesity treat-
ment is associated with a reduced rate of gestational diabe-
tes and large-for-gestational-age infants. However, it is as-
sociated with an increase in the incidence of small-for-ges-
tational-age infants, a shorter pregnancy, and potentially an
increased risk of stillbirth or neonatal death. Despite the
potential concern for nutritional deficiencies after bariatric
surgery, a recent study reported that there was no signifi-
cant effect of bariatric surgery on the overall risk of con-
genital malformations (168); however, a delay in attempt-
ing to conceive for at least a year post surgery is a recom-
mendation (132).
In the Nurses’ Health Study (117), every 5 kg increase in
body weight from the age of 18 years of age was associated
with a 5% increase in the median time to conceive, but
women who gained more than 20 kg since 18 years of age
took on average an extra 1.4 months to conceive, compared
with women who maintained their weight. Interestingly, of
particular detriment to conception was being underweight
at 18 years of age but overweight at the time of attempt to
conceive, where this led to a doubling of the time to con-
ception (117), similar to data derived from the Danish Birth
Cohort (272).
Several studies demonstrate the benefit of weight loss as
part of a structured weight loss program upon the chance of
conception for overweight women, and also as part of their
IVF treatment (61, 314). In a systematic review of 11 stud-
ROGER J. HART
890 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
ies that met the search criteria weight loss on the outcome of
subsequent IVF treatment, losing weight by either diet and
lifestyle changes (7 studies), nonsurgical medical interven-
tions (1 study), or bariatric surgery (2 studies) led to a
significant increase in the natural conception rate, an in-
crease in the number of embryos available for transfer as
well as the subsequent pregnancy rate, and a decrease in the
miscarriage rate (314). Due to the difficulty in completing
such studies, the overall quality of the studies was reported
as weak, although all interventions led to significant im-
provements in pregnancy or live birth rates in overweight or
obese women, with several studies reporting an improve-
ment in spontaneous pregnancy rates. Due to the heteroge-
neity within the studies, a quantification of the benefit was
not possible; however, the greatest improvements in out-
comes was reported for women embarking on a multidisci-
plinary structured program of dietary and lifestyle interven-
tion (314).
Of particular importance in relation to obesity is that
women with PCOS, which is characterized by hyperinsulin-
emia and hyperandrogenism (231), are particularly predis-
posed to weight gain (340), exacerbating their risk of ob-
stetric, perinatal, and neonatal complications in addition to
an increase in congenital malformations (87). In general,
women with PCOS are at a greater risk of miscarriage than
women without features of PCOS; in addition, they are at
greater risk of gestational diabetes, preeclampsia, and pre-
mature labor in pregnancy, and the perinatal mortality rate
is greater for infants born to women with PCOS (87, 139).
In later life, women with PCOS were twice as likely to have
a non-injury-related hospital admissions, three times more
likely to develop type II diabetes, four times more likely to
be obese, and were significantly more likely to have cere-
brovascular and cardiovascular disease, suffer a thrombo-
embolic event, and have a hospital admission for a mental
health condition than a woman without a diagnosis of
PCOS (139). Furthermore, as it is believed hyperinsulin-
emia is present in 75% of lean women with PCOS and 95%
of overweight women with PCOS (328), the reproductive,
metabolic, and psychological consequences of PCOS are
further exacerbated by weight gain (231).
Overweight women take longer to conceive through natural
conception (166), and overweight women who require IVF
treatment to conceive are less likely to conceive and more
likely to miscarry than women of normal weight (279). The
ideal way to determine whether the negative influence of
obesity on conception is related to a negative influence on
the oocyte or to the endometrium is to use the egg donation
model. Whilst one large retrospective study of young nor-
mal weight oocyte donors demonstrated a negative trend
upon conception related to an increasing BMI amongst re-
cipients (30), a recent systematic review of six data sources
demonstrated that there was no negative influence of obe-
sity upon implantation, clinical pregnancy, miscarriage
rate, and live birth (169). This may suggest the primary
influence may relate more to oocyte quality rather than an
endometrial effect. To study this thoroughly, a study con-
trolling for the body mass index of the donor is required.
The effect of obesity on the endometrium is to alter the
expression of over 150 endometrial genes, primarily related
to transcription and biosynthesis, to reduce the chance of
conception (29). The benefit of preconception intervention
was demonstrated in an RCT of a preconception structured
lifestyle modification program for women with PCOS, in-
cluding a combination of calorie restriction, weight loss
medication, and exercise, that led to a significant improve-
ment in metabolic features, ovulation rates, and trend to-
wards an increased live birth rate (198).
Obesity is associated with an increased risk of miscarriage
in women undergoing IVF treatment (279), although as
described above this may primarily be due to an oocyte
effect, rather than an endometrial effect. However, in over-
weight women with PCOS, it is believed that integral to the
endometrial defect is insulin resistance leading to decreased
expression of
␣
v

3 integrin, HOXA10, and IGFBP-1 and
increased androgen receptor expression (302), leading to an
increased risk of miscarriage [for review, see Schulte et al.
(302)]. Consequently, therapies to address the insulin resis-
tance such as weight loss (61), and use of the insulin sensi-
tizer metformin (248), have been employed to reduce the
risk of miscarriage. The practice guidelines from the system-
atic review of the nonpharmacological interventions for
women with PCOS was performed in 2011 and updated in
2015 and the finding are listed in TABLE 6 (9, 341).
E. Cigarette Smoking
Cigarette smoking has a profound effect on fertility for both
the male and the female (80, 270); despite this, many
women continue to smoke while attempting to conceive and
even when undergoing IVF treatment (127). In one study, if
both partners smoked, the woman’s chance of conceiving
was one-fifth of those couples where neither partners
smoked; despite this, the authors cite data that 20% of
women in the United States are believed to smoke in the 3
months prior to starting to try to conceive (263). For a
cigarette smoking woman, all aspects of fertility are im-
paired (80), as the smoke contains heavy metals, polycylic
hydrocarbons, nitrosamines, and aromatic amines (80, 86)
[for review, see Dechanet et al. (80) and Camlin et al. (49)].
1. Ovarian function
In animal models cigarette smoking leads to a depletion of
primordial follicles (49), further loss of follicles throughout
folliculogenesis, impaired cumulus oocyte complex expan-
sion (361) and impaired granulosa cell function, granulosa
cell steroid production, impaired oocyte meiosis (49), and a
predisposition to aneuploidy within the oocyte (201). It is
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
891Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
unclear how these translate for the woman that smokes
cigarettes while attempting to conceive; however, it is
known that smoking leads to a reduction in ovarian reserve
and an advancement of the age of menopause for the
woman (167, 301), potentially due to the influence of poly-
cyclic hydrocarbons which are activated into more toxic
metabolites by the liver (49, 271). Further human studies
have demonstrated that oocytes derived from cigarette
smoking women undergoing IVF treatment have a greater
number of immature oocytes (385), an increased thickness
of the zona pellucida (310), with follicles containing higher
level of markers of oxidative stress within the follicular fluid
(252), and within the cumulus (316).
Women embarking on an IVF cycle have a reduced ovarian
response if they had been recent cigarette smokers (112).
With the use of the smoking mouse model, the ovaries of
cigarette smoke-exposed mice were smaller, contained
fewer primordial and primary follicles, and resulted in
fewer oocytes at the time of ovulation (321).
2. Fallopian tube function
Smoking is the main lifestyle and environmental agent that
negatively influences fallopian tube function. Rhesus mon-
key and mouse studies suggest that smoking exposure re-
duces fallopian tube blood flow (224) and smooth muscle
contraction (234), impairs fimbrial oocyte collection (181)
and tubal motility, and reduces the number of ciliated cells
within the fallopian tube and their beat frequency [reviewed
by Shao et al. (307)]. Human studies of fallopian tube func-
tion are challenging to perform; however, it is believed that
there is a close similarity between the rodent and human
fallopian tube physiology assisting with our understanding
of fallopian tube physiology (307). However, it is still un-
clear what is the cause of an ectopic embryo implantation,
as it may be mechanical due to obstruction, due to a change
in fallopian tube histology, or an abnormal expression of
hormone receptors within the tube (307). Indeed, it is be-
lieved that smoking more than 20 cigarettes a day increases
the risk of a fallopian tube ectopic gestation fourfold (43,
100). This epidemiological evidence, combined with the
many animal studies, of the influence of cigarette smok-
ing on fallopian tube function suggests that the agents, or
their metabolites, within cigarette smoke are a substan-
tial contributing cause of female infertility and ectopic
gestation (80).
3. Fertilization and embryonic development
The effects of cigarette smoking exposure on the oocyte
of women undergoing IVF treatment appears to consist
of the development of a thicker zona pellucida compared
with nonsmokers (310), a potential cause of the reported
reduction in fertilization rate with IVF (185), and de-
layed embryo morphokinetic cleavage events in couples
undergoing ICSI treatment (110), which as previously
discussed may reduce embryo implantation. Further-
more, the oocyte derived from a cigarette smoking
woman may have delayed maturation and be at risk of
meiotic errors (385), leading to an increased risk of the
development of an aneuploid embryo (384). Rodent
studies also suggest slower rates of embryonic develop-
ment with reduced cell numbers and implantation rates
upon exposure to cigarette smoke (224).
Table 6. Lifestyle management for women with PCOS who are trying to conceive: recommendations after systematic review
Evidence Statement Level of Evidence
Lifestyle management, including diet and exercise programs, should be used throughout the life span
in women PCOS to optimize health generally and to alleviate PCOS clinical severity including
infertility.
C
In women with PCOS and body mass index ⫽30 kg/m with due consideration given to age-related
infertility, intensive (frequent multidisciplinary contact) lifestyle modification alone (and not in
combination with pharmacological ovulation induction therapy) should be the first-line therapy for
3–6 months to determine if ovulation is induced.
C
Pharmacological ovulation induction should not be recommended for first-line therapy in women with
PCOS who are morbidly obese (body mass index ⫽35 kg/m) until appropriate weight loss has
occurred either through diet, exercise, bariatric surgery, or other appropriate means.
C
Pharmacological ovulation induction could be second-line therapy, after intensive lifestyle modification
has been undertaken.
C
Morbid obesity increases risks during pregnancy and should be regarded as a relative
contraindication to assisted fertility.
PP
Psychological factors should be considered and managed in infertile women with PCOS, to optimize
engagement and adherence with lifestyle interventions.
PP
C, body of evidence provides some support for recommendation, but care should be taken in its application; PP,
evidence not sought. A practice point has been made by the guideline development group where important
issues arose from discussion of evidence-based or clinical consensus recommendations. Adapted from
Evidence Based Guidelines for the Assessment and Management of Polycystic Ovary Syndrome. Melbourne,
Australia: Jean Hailes for Women’s Health on behalf of the PCOS Australian Alliance, 2015.
ROGER J. HART
892 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
4. Implantation
It is widely believed that cigarette smoking has a detrimen-
tal effect on implantation, as the recipient in an oocyte
donation program will have less chance of conceiving if she
smokes, proportionate to the amount of cigarettes smoked
(319). With the use of animal models and cultures of human
endometrial cells, the effect of inhalation of cigarette smoke
upon the endometrium can be analyzed (80). It appears that
the influence of cigarette smoke may lead to anti-estrogenic
effects on the developing endometrium, altered angiogene-
sis within the endometrium, influence on trophoblast inva-
sion, and altered gene expression within the endometrium
(80). The actual influences are determined by the individual
compounds and the dose to which the endometrium is ex-
posed. Human placental trophoblastic cell lines exposed to
compounds within cigarette smoke have been reported to
lead to altered expression of epidermal growth factor (387),
matrix metalloproteases (121), alterations of hCG secretion
(40), L-selectin expression (383), in addition to increased
apoptosis, altered cellular architecture, and reduced cy-
totrophoblast invasion (80, 122). Cigarette smoking
women are exposed to high levels of cadmium and lead, and
these metals have been found within the endometrium of
cigarette smokers, correlating with the number of cigarettes
smoked and the duration of smoking (289).
5. Miscarriage
Smoking is believed to increase the risk of miscarriage by
⬃1% per cigarette smoked (258), with the overall adjusted
relative risk of miscarriage being 1.23, with a nonsignificant
increased risk of miscarriage associated with passive smok-
ing (258). The cause of the increased miscarriage risk is
likely to be multifactorial. One large study that analyzed the
effect of cigarette smoking on the miscarriage of chromo-
somally normal conceptions demonstrated a tendency to-
wards an increased likelihood of miscarriage in this group
(179), suggesting an endometrial effect in addition to the
potential for an increased risk of aneuploidy described
above (384).
F. Periodontal Disease
There is increasing evidence of an association of poor oral
health with a number of clinically important medical con-
ditions (370). Periodontal disease is a chronic infectious and
inflammatory disease of the gums and supporting tissues
and has been associated with cardiovascular disease, type 2
diabetes, respiratory disease, kidney disease, and adverse
pregnancy outcomes (105, 235, 312, 313, 370). It is be-
lieved that up to 10% of the population have severe peri-
odontal disease. Several studies suggest that successful
treatment of periodontal disease may alter or modify in-
flammatory markers (74) and improve endothelial and vas-
cular function after therapy (345), and it is believed to be a
modifiable factor that may lead to improvements in long-
term health (235, 344). An observational study of the prev-
alence of periodontal disease in pregnant women suggested
that periodontal disease may also be a factor limiting a
woman’s time to conceive (143). The mechanism proposed
was that it caused a low-grade inflammation reducing em-
bryo implantation, as pregnant woman with periodontal
disease took, on average, an extra 2 mo to conceive, a
negative influence on conception of the same order of mag-
nitude as obesity.
In summary, the main lifestyle factors that may have a neg-
ative impact on a woman’s fertility are smoking and obe-
sity. Fortunately, evidence suggests that smoking cessation
will improve the chance of conception, and embarking on a
supervised weight loss program will improve the chances of
conceiving for the overweight woman. Unfortunately, the
age-related decline in oocyte quality for a woman leads
many women to seek fertility treatment using an oocyte
donor. Potential future interventions of ovarian rejuvena-
tion may offer some of these women in the future alterna-
tive options.
V. ENVIRONMENTAL INFLUENCES
AFFECTING
It is suspected that potentially the most significant of the
common environmental influences that may impact upon
female fertility relate to endocrine disrupting chemicals
(EDCs). EDCs are ubiquitous chemicals that interfere with
hormone action, and almost all pregnant women in the
United States have measurable quantities within their bod-
ies (375). Evidence derived from environmental disasters
where toxic chemical have leaked into the environment has
demonstrated that permanent changes can occur to the en-
docrine system and confirms that environmental chemicals
can act as EDCs. However, for many years there has been
substantial controversy around the subject of environmen-
tal EDCs at lower levels of environmental exposure, as
many stakeholders have been involved in investigating the
subject, often with potential for bias, for example, the
chemical industry has a vested interest in proving safety,
and in contrast the media have at times generated sensa-
tional reports. The Endocrine Society published an updated
systematic review in 2015 that endeavored to analyze the
literature in a very balanced manner with regard to any
potential conflict of interest of the investigators, any ten-
dency towards negative or positive findings bias (depending
on the source of funding for the research), and being mind-
ful of the extrapolation of animal studies to the human
situation (125).
EDCs have numerous potential mechanisms of actions in
addition to traditional receptor activation and generally do
not have a predictable dose-response curve, and the same
chemical may have agonistic or antagonistic properties at
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
893Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
different concentrations and at different sites. Furthermore,
they are frequently additive with other EDCs, of which
there are believed to be in excess of 800 chemicals, and also
they may have several active metabolites, and the effect
of the EDC may depend on the timing of exposure (26,
392). Consequently, it is often difficult to determine the
properties of many substances with potential endocrine dis-
rupting actions. EDCs interfere with the action of hormones
either at a hormone receptor or they may alter the number
of hormone receptors within a cell, and if these influences
happen at a critical stage of development, the changes may
be permanent. Due to single-nucleotide polymorphisms,
some individuals may be more susceptible than others. In
addition, it is possible that some EDCs may cause epigenetic
changes leading to a transgenerational effect by DNA meth-
ylation, histone modification, and influence on micro-RNA
expression (125). A further complicating factor when ana-
lyzing the effects of EDCs is that the EDC may act on the
receptors of several hormones, and depending on the levels
of exposure studied, the outcome may vary as each organ
may have a different threshold for disruption (125). To
study the endocrine disrupting nature of a substance in the
United States, the Endocrine Disruptor Screening Program
employs two series of assays (Tox21 and ToxCast) to study
thousands of chemicals and their signaling pathways.
Animal studies offer several advantages when studying the
effects of EDCs as many biological processes are conserved
across several species; furthermore, they enable the investi-
gator to control the dosing of exposure, the timing of expo-
sure, limit any interference from other chemicals and con-
trol for many variables, and potentially perform the study
over a relatively short period of time. However, the limita-
tions of using animals as experimental subjects are that the
action in the animal may differ from their actions on hu-
mans. Many EDCs have additive effects, and this may am-
plify the effect of the particular chemical studied which may
not be evident in an animal study. In addition, replicating
the “human timing of exposure” or “window of vulnera-
bility” in animal studies can be difficult, and in animal
studies it may be required to use higher doses of an EDC for
a shorter period of time, rather than prolonged low level of
exposure as is often found in the human situation (374).
Therefore, it is believed that observational human studies
may provide useful epidemiological data; however, chal-
lenges that may be faced are that the particular EDC expo-
sure may be difficult to identify and quantify and may be
reliant on human recall, plus eliminating confounding fac-
tors may be difficult in a long-term epidemiological study
(374).
The main endocrine disrupting chemicals are BPA, the
phthalates and their esters, the pesticide Atrazine, and the
polychlorinated biphenyls (PCBs) and DDT/DDE. They are
briefly described below, and their actions are described in
greater detail where they influence fertility.
1) BPA is a synthetic chemical widely used in the manufac-
ture of plastics and resins for many years, and the main
route of exposure is within the diet and through transder-
mal contact. In many countries its use in the manufacture of
plastic bottles has been phased out. However, BPA along
with phthalates are frequently measurable within urine
(182), serum (141), follicular fluid (187), umbilical cord
blood (162), and the amniotic fluid (162). Of particular
concern was the finding that the amniotic fluid concentra-
tions of BPA were five times greater than serum, presumably
due to fetal renal clearance (162). The methods of action of
BPA are that it can bind to the nuclear receptors for ER
␣
and ER

, with differing affinities for each which may lead
to differing agonist/antagonist responses, although it is be-
lieved that the majority of its action is through other mech-
anisms, such as through intracellular signal transduction
pathways independent of the nuclear hormone receptors,
modification of cytochrome P-450 enzyme expression
and activity, alterations in the level and activity of sex
hormone binding globulin, and epigenetic modulation by
the silencing of promoters by methylation [for review, see
Wetherill et al. (365)]. In addition, BPA is believed to
impair mitochondrial function and promote oxidative
stress in high doses in rat studies (178). It is reported that
more BPA is produced than any other chemical with ⬃15
billion pounds produced in 2013 (125). Interestingly, the
United States Environmental Protection Agency safety
level is set at 50
g·kg
⫺1
·day
⫺1
, whereas the European
Food Safety Authority tolerable daily intake is 4
g·kg
⫺1
·day
⫺1
. However, it is reported in the The Endo-
crine Society’s Second Scientific Statement on Endocrine-
Disrupting Chemicals that BPA is believed to have effects
at or below these levels (125).
2) Phthalates and their esters are plasticizers to provide
flexibility to materials and are present widely within the
home and hospital environment and are also used in per-
sonal care products, such as cosmetics. As they are nonco-
valently bound, they leach into the environment and they
are detectable and hence they are ubiquitous within the
environment (125).
3) Many pesticides have been suspected of being EDCs.
Atrazine is a widely employed herbicide used in commercial
crop growing in the United States, is persistent with its
metabolites in ground and surface water for many years,
and is detectable within drinking water (125).
4) PCBs were banned in 1979; however, they bioaccumu-
late within the environment and are stored in body fat, and
some PCBs have thyroidogenic, estrogenic, and antiandro-
genic actions (125).
5) DDT is an insecticide with a long half-life which was
widely used prior to being banned in the United States in
1972 due to being linked to several cancers, and as with
ROGER J. HART
894 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
PCBs, DDT is persistent within the environment due to its
long half-life and bioacumulates in fat (125). The conse-
quences of substances that are stored in fat tissue are often
difficult to determine, as the measurable levels in the blood
are low, however, the substances may be released rapidly
within the body during periods of weight loss.
The effects of EDCs on reproduction has recently under-
gone an updated systematic review by the Endocrine Society
and published in October 2015, and several summary state-
ments are derived from this review (125).
A. Folliculogenesis and Ovulation
1. Premature ovarian insufficiency
Environmental causes of premature ovarian insufficiency
(POI) are not well defined, other than being related to che-
motherapy and radiotherapy; however, the potential for in
utero or other early life exposure to gonadotoxic chemicals
to lead to POI in later life is a possibility, although no
common environmental chemicals or EDCs have been dem-
onstrated to conclusively lead to POI (36, 125, 141). Al-
though there is evidence from a series of studies performed
on rats that a mixture of estrogenic EDCs (phthalates, pes-
ticides, chemical ultraviolet filters, bisphenol A, parabens),
exposed from day 7 of gestation to 22 days postnatally, may
lead to a reduction in the frequency of estrous in the rats,
there was no significant difference in ovarian weight or
follicle count in this short study (163).
2. Influences on follicular development and ovulation
A)ENDOCRINE DISRUPTORS.I) BPA. With regard to folliculo-
genesis, animal studies suggest that maternal BPA exposure
may lead to disorders of meiosis in the female offspring,
increasing the risk of oocyte aneuploidy and abnormal pri-
mordial follicle development and progression through fol-
liculogenesis (160, 386).
The multiple purported effects of BPA during early stages of
oogenesis and the final stages of maturation (metaphase I
and metaphase II) from human and animal studies are dem-
onstrated in FIGURE 5.
Studies using culture of murine ovarian tissue demonstrate
that, at exposure levels found in Chinese children, BPA may
lead to a premature activation of folliculogenesis ultimately
leading to a reduced follicular pool (388). Similar studies
have been performed in vivo using mice, rats, sheep, and
monkeys (see TABLE 7); however, there is limited data avail-
able on the effects of BPA on ovarian development and early
folliculogenesis in humans (125). Animal studies suggest
that early life exposure to BPA at higher doses appears to
accelerate the follicular development leading to cystic folli-
cles and reducing the antral follicle pool, but at lower doses
decreases all follicle stages both in in vivo animal studies
and in cultures of ovarian cells [summarized by Gore et al.
(125)].
The negative influence of BPA upon follicular growth is
believed to have several mechanisms: interference with the
aryl hydrocarbon receptor, alteration of cell cycle regula-
tors, and altered steroidogenesis (253). In animal studies
BPA-induced follicular atresia is associated with increased
markers of apoptosis (253). These animal studies appear to
be corroborated in human studies of the antral follicle count
of 209 women undergoing a fertility assessment, where
there was an association of a higher urinary BPA concen-
tration with a lower antral follicle count (326).
Both BPA and phthalates are believed to have a modulat-
ing effect on hypothalamic-pituitary action in animal
studies (52), and it is hypothesized that BPA may be
FIGURE 5. The effects of bisphenol A during early stages of oogenesis and the final stages of maturation
(metaphase I and metaphase II). [From Machtinger and Orbvieto (207), with permission from Elsevier.]
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
895Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
associated with a premature activation of the hypotha-
lamic-pituitary-ovarian axis and be related to premature
pubertal development (52).
Depending on the dose and the species studied, BPA is
believed to have several points of action to interfere with
steroidogenesis (125). Associations of higher serum BPA
have been linked with a lower estradiol response in
women undergoing IVF treatment, suggesting a negative
impact on ovarian steroidogenesis (39, 94), and in-
creased urinary concentrations of BPA have been associ-
ated with lower antral follicle counts in women undergo-
ing IVF treatment (326). Although, seemingly in con-
trast, BPA exposure has also been associated with PCOS
(1, 26, 172, 288); however, it is unclear whether this is a
causal relationship as higher serum androgen concentra-
tions are associated with delayed clearance of BPA (52),
as the finding of an increased follicular count is in direct
contrast to Souter et al. (326).
With regard to PCOS, BPA is probably the most studied
of the environmental endocrine disrupters. BPA has been
demonstrated in vitro to increase in adipocyte differenti-
ation in human and mice cell culture (42), stimulate rat
ovarian theca cells to synthesize testosterone (390), and
induce insulin resistance in offspring of rats exposed in
pregnancy (325). How these in vitro studies and the use
of animal models relate to human programming of the
disease is unclear, particularly as the “window” of expo-
sure that may program PCOS is unknown due to the
difficulties in studying transgenerational effects in hu-
mans. However, it is believed that the most likely vulner-
able period of exposure is in utero and the first few years
of life (26). The reasoning proposes that animal studies of
Table 7. Effects on oocytes of prenatal and postnatal BPA exposure
Study Sample Dose
Route of
Administration Time of Exposure Results
Zhang et al. (386) Pregnant mice 0.02, 0.04, 0.08
mg
.
kg body
wt
⫺1.
day
⫺1
Oral route 12.5–18.5 day
post-coital
Inhibition of meiotic progression
to prophase I in 0.08 BPA-
treated group; decreased
mRNA expression of specific
meiotic genes; inhibition of
germ cell cyst breakdown
Hunt et al. (160) Pregnant rhesus
monkeys
400
g
.
kg body
wt
⫺1.
day
⫺1
Tubing implants GD 50–100, GD 100
to term
Disturbances in prophase
events; increase in MOFs
Susiarjo et al. (335) Pregnant mice,
offspring
400 ng/day Pellets
releasing
BPA
GD 11.5–17.5 Aberrant meiotic prophase;
increased aneuploidy in eggs
and embryos from adult
females
Rivera et al. (280) Lambs 50
g
.
kg
⫺1.
day
⫺1
Subcutaneous
injections
PND 1–14 Decreased ovarian weight;
increased primordial-to-
primary follicle transition;
increased incidence of MOFs;
increased number of small
antral atretic follicles
associated with higher p27
expression
Karavan et al. (173) Mice 10, 100
g/day Subcutaneous
injections
PND 1–4 Increased incidence of MOFs;
increased total number
oocytes; increased
percentage of primordial
follicles
Rodriguez et al. (283) Rats 0.05, 20
mg
.
kg
⫺1.
day
⫺1
Subcutaneous
injections
PND 1–7 In BPA 20 group stimulation of
neonatal initial follicle
recruitment; p27 and ER
␣
increased expression;
increased proliferation rate of
granulosa cells
Chao et al. (56) Mice 20, 40
g/kg Subcutaneous
injections
PND 7–14, PND 5–20
(every 5 days)
Decreased methylation pattern
of two maternal imprinted
genes; upregulated mRNA
expression of ER
␣
; decreased
primordial follicle number but
increased primary,
secondary, and antral follicle
number; abnormal spindle
assembling in meiosis
From Caserta et al. (52).
ROGER J. HART
896 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
exogenous androgen exposure and human evidence of in
utero growth restriction with subsequent catch-up
growth, leading to the subsequent development of fea-
tures of PCOS and the metabolic syndrome, suggest that
a period of vulnerability exists at this time to individuals
who may have a genetic predisposition to PCOS (26,
246). Indeed, with the use of both high- and low-dose
exposures to BPA in early postnatal life, a rodent model
of PCOS has been developed (103). From animal studies
of BPA exposure at levels of exposure found in humans,
animals tend to accrue body fat, leading to metabolic
derangement, and develop impaired ovarian function
(26), although human data are not clear (125). It is im-
portant to note that a significant vehicle of BPA exposure
is transdermal; hence, larger individuals will tend to have
greater exposure leading to an assumption of an associ-
ation with obesity, rather than potentially being an arte-
fact of an increased surface area. It is also important to
note for substances with a high degree of transdermal
absorption that since an infant has the greatest surface
area-to-volume ratio, it will receive a greater exposure
relative to height than an adult. No epidemiological stud-
ies of early life BPA exposure into adulthood have yet
been performed, although studies suggest serum BPA
concentrations are increased in the presence of PCOS in
adulthood after controlling for BMI (172), as serum BPA
correlates to BMI; however, this association does not
demonstrate causality as, for example, the excretion of
BPA may be impaired in PCOS and hence its concentra-
tion would be artificially elevated.
II) Phthalates. There is limited information on the effects of
phthalates on follicular development. Animal studies sug-
gest that DEHP, MEHP, and DBP induce follicular arrest
and atresia in cultured rat and mice cells [summarized by
Gore et al. (125)]. Limited human data exist, although by
measuring the metabolites of phthalates in stored maternal
blood, our group demonstrated a trend toward an earlier
menarche in Western Australian girls, a degree of hypogo-
nadism with a reduction in early follicular phase serum FSH
associated with the phthalate metabolite MEHP (141). Of
particular concern was the finding that maternal exposure
to MEP, widely found in cosmetics, was associated with a
reduction in their daughters’ serum AMH in adolescence, a
marker of antral follicles (ovarian reserve) and granulosa
cell function (141). In addition, higher MEP exposure was
associated with a reduced prevalence of PCOS in adoles-
cence and reduced antral follicle count (141). A similar
finding was observed in a small study of patients with PCOS
where lower urine concentrations of mBP and mBzP were
detected in women with PCOS (352). Several in vivo and in
vitro studies have been performed on animals with phtha-
late exposure, and exposure, particularly to DEHP and
MEHP, appears to impair estradiol, progesterone, andro-
stenedione, and testosterone production [summarized by
Gore et al. (125)].
III) Pesticides. Methoxyclor (MXC) is a pesticide and an
insecticide; hence, it is present in food and water and has
been used as a replacement for dichlorodiphenyltrichloro-
ethane (DDT), and it has been reported to inhibit the re-
sponse to ovarian stimulation in mice (96), and also induces
apoptosis in baboon antral follicles by activation of the
aryl-hydrocarbon and estrogen receptor and by the activa-
tion of apoptotic cascade and via oxidative stress [summa-
rized by Gore et al. (125)]. In mice, it inhibits steroidogen-
esis at multiple points in the steroidogenic pathway, leading
to a reduced antral follicle production of estradiol, testos-
terone, and androstenedione (27). Some of the other pesti-
cides that have been studied in animals and are suspected to
have an adverse effect upon folliculogenesis include endo-
sulfan, malathion, cypermethrin, carbamate, imidacloprid,
fenvalerate, trifluralin, bifenthrin, and diurin, and it is sug-
gested that pesticides may alter gene expression, impair fol-
licle growth, induce atresia, and reduce oocyte quality and
impair steroidogenesis [summarized by Gore et al. (125)].
Atrazine has been associated with menstrual cycle length
irregularity, as women who drunk more than 2 cups of
unfiltered Illinois water (where atrazine is widely used and
detectable in the drinking water) were up to six times as
likely to have menstrual irregularity as women who did not
drink the water (69), suggesting a disruption of the ovula-
tory menstrual cycle.
IV) Persistent environmental contaminants. 2,3,7,8-Tetra-
chlorodibenzo-p-dioxin (TCDD) is a dioxin and a wide-
spread persistent environmental pollutant. It has inhibitory
influences upon pubertal timing, ovarian steroidogenesis,
folliculogenesis, and ovulation in several species (165, 253),
although human data on the effect of TCDD upon repro-
duction is limited. In mice studies, TCDD and PCBs reduced
ovarian weight and induced follicular atresia (261). Al-
though, in contrast, a small observational study reported
that PCOS subjects, where antral follicle counts are usually
increased, had higher serum concentrations of some persis-
tent environmental pollutants, perfluorooctanoate and per-
fluorooctane sulfonate (352); however, associations do not
demonstrate causality.
V) Phytoestrogens. Genistein is a phytoestrogen present in
soy, lentils, and chickpeas and appears to either promote or
inhibit follicular development on the rat ovary, depending
on the dose, strain, and age to promote, and influence sex
steroid production within the follicle and activate apoptosis
(253).
For a detailed description of the effect of environmental
exposures in ovarian function, readers are directed to Bhat-
tacharya and Keating (36) and Patel et al. (253). Trichloro-
ethylene (TCE) is commonly found in adhesives and lubri-
cants. Both TCE and its metabolites led to a reduction in
fertilization of mouse oocyte (68). Exposure to 7,12-dim-
ethylbenz[a]anthracene (DMBA), found in cigarette smoke
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
897Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
and car exhaust fumes, leads to a loss of mouse or rat
follicles at all stages of development (161), and exposure to
DMBA led to altered expression of genes regulating follic-
ular development (322). A common by-product of pesti-
cide, rubber, plastic, and flame retardant manufacture is
4-vinylcyclohexene (VCH). In animal studies, VCH de-
stroys primordial and small primary follicles leading to
ovarian failure (156).
B)HEAVY METALS. Heavy metals are resistant to degradation
and hence may persist in the environment for many years.
Potentially their concentration may be amplified by bioac-
cumulation within the food chain, particularly individuals
with high fish intake which may be at risk of exposure to
mercury. Some of these metals are essential for life in low
concentrations, but highly toxic in higher concentrations,
such as copper, chromium, manganese, and zinc, although
cadmium, lead, mercury, and the metalloid arsenic are non-
essential and are toxic (290). Women are exposed to these
chemicals through inhalation, drinking contaminated wa-
ter, or eating food contaminated by exposure or by bio-
accumulation. With the use of mercury-based dental amal-
gams, dental technicians may have greater exposure than
the general population as measured by urinary excretion of
the metal and are potentially at risk of a dose-dependent
reduction in fecundability for a woman actively trying to
conceive (286), as are women who have a high fish intake in
their diet (11). However, the evidence for normal environ-
mental mercury exposure interfering with ovulation is lim-
ited (59). Despite this, it is advised that women trying to
conceive should avoid processed and “fast food” and min-
imize exposure to mercury until completion of breast feed-
ing by avoiding large fish such as shark, king mackerel, and
tilefish (238).
Evidence exists for some heavy metals to have a potential
epigenetic modification influence on various cultured cell
lines (290), influencing DNA methylation leading to gene
inactivation loss of acetylation and increasing histone
methylation (12) as well as activating apoptosis, and ar-
senic is known to disrupt the cell cycle (12, 104). In
women undergoing IVF treatment, higher mercury expo-
sure has been associated with altered methylation pat-
terns within CpG sites within whole blood, and cadmium
exposure has been linked to altered methylation patterns
in Andean women from Argentina, suggesting that these
laboratory-observed influences on epigenetic modifica-
tions may occur at concentrations present within the en-
vironment.
A major source of exposure to heavy metals is derived from
cigarette smoking, as the tobacco plant accumulates cad-
mium and lead from the soil and the serum concentrations
of these heavy metals have been correlated to the brand and
the number of cigarettes smoked (14).
B. Fallopian Tube Function and Inflammatory
Disorders
1. Endocrine disrupting chemicals
Due to the estrogen responsive nature of the development of
endometriosis, researchers have extensively studied the as-
sociation of endometriosis and endocrine disrupting chem-
icals. Dioxin exposure (47, 324) and exposure to the a
phthalate BzBP metabolite and its metabolites MBz, P and
the DEP metabolite MEP may be associated with increased
risk of endometriosis (63), although BPA exposure was not
linked in an epidemiological study of women with endome-
triosis (350). One large cross-sectional study from the
United States demonstrated an association of MBP with an
increased risk of endometriosis (366), and another did not
confirm the associations of phthalate exposure with endo-
metriosis and demonstrated that MEHP had an inverse as-
sociation with the presence of endometriosis (351). Serum
levels of the organochlorine pesticide

-hexachlorocyclo-
hexane have been associated with endometriosis in a cohort
of women undergoing surgical exploration for the disease
(349), and dioxin and dioxin-like compounds have for
many years been believed to be associated with the devel-
opment of endometriosis. However, the literature is con-
flicting (320), although a small study that performed anal-
ysis of adipose tissue at the time of laparoscopic surgery
demonstrated an association between adipose concentra-
tions of dioxin and PCBs and the presence of endometriosis
at the time of surgery (211).
C. Embryonic Development
1. Teratogens
While not the focus of this fertility review, it is important to
mention teratogens. Teratogens are agents that an individ-
ual may be administered or self-administered, or be exposed
to in pregnancy, or around the time of conception, that may
cause a structural or functional defect to the fetus (101).
Hence, the most common preconception teratogen is prob-
ably being overweight or obese, as being overweight leads
to an increase in the incidence of neural tube defects as well
as cardiac and oro-facial abnormalities for the offspring
(101). Furthermore, the use of fertility drugs themselves and
IVF have been linked to an increase in congenital abnormal-
ities in the children born (137). Consequently, it is essential
that a treating physician aims to ensure a woman trying to
conceive is as healthy as possible at the time of conception
and she should aim to keep the use of over-the-counter and
prescribed medications in pregnancy to a minimum.
In addition to medication exposure, teratogens can consist
of infections, such as syphilis, rubella, and cytomegalovirus
exposure. They may be caused by metabolic disturbance,
such as diabetes, and may be a physical insult such as ra-
ROGER J. HART
898 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
diotherapy or exposure to a chemotherapeutic agent. How-
ever, the most common teratogens are physician-prescribed
medications, as up to two-thirds of women in the United
States are prescribed drugs in pregnancy (251). Further-
more, as the most critical window of exposure is the first
trimester, often before the woman recognizes she is preg-
nant, it is imperative that a doctor is vigilant to this factor in
treating a woman of reproductive age and prescribes med-
ication sparingly and also manages chronic medical condi-
tions such as thyroid disorder and diabetes in an optimal
manner to reduce the fetal exposure to teratogenic medica-
tion and also to the harm of an unstable systemic illness.
The Federal Drug Administration describes drugs accord-
ing to their teratogenicity by ascribing a category (A, B, C,
D, and X), and this is readily available to clinicians for their
reference.
2. Pollutants
Organochlorine pollutants, such as polychlorinated biphe-
nyls and DDT, are persistent within the environment and
within the body and have been speculated to be associated
with an increased time to conceive for women, although this
has not been verified (170). Furthermore, review of the
literature did not demonstrate any association with oocyte
quality, fertilization rate, embryo development, or ulti-
mately the pregnancy rate for women embarking on IVF
treatment (170). Human and animal studies demonstrated
impaired steroidogenesis in granulosa cells exposed to BPA
(52), and this is borne out by women with higher serum
BPA concentrations undergoing IVF treatment whereby
they have a lower peak estradiol concentration and reduced
oocyte yield (39, 94). A recent study of hair mercury con-
centrations, a marker of dietary fish intake, in women un-
dergoing IVF treatment did not find a relationship between
mercury levels and ovarian responsive to stimulation,
oocyte fertilization rate, embryo development, and preg-
nancy rates (376).
D. Implantation
1. Endocrine disrupting chemicals
An environmental exposure assessment of 501 couples try-
ing to conceive in the United States did not determine a
relationship between exposures to BPA and 14 phthalate
metabolites in the urine on the length of time it took a
couple to conceive, although this does not suggest that any
exposures were not in some way influencing early embry-
onic development.
A)BPA. In animal studies, BPA exposure has led to a signif-
icant reduction in embryo implantation (379). Exposure to
BPA is believed to lead to a downregulation of HOXA10
expression (217), with IVF embryo implantation failure be-
ing more common in women with higher urinary BPA levels
(93). A United States study of women with unexplained
miscarriage demonstrated that those with a serum conju-
gated BPA concentration in the highest quartile were almost
twice as likely to miscarry as those women in the lowest
quartile. The authors hypothesized that the cause was due
to a negative influence of BPA upon the endometrium or
early placentation (193), and BPA exposure has also been
associated with a predisposition to with recurrent miscar-
riage (334, 389).
B)PHTHALATES. There are conflicting reports on the associa-
tion of urinary and follicular fluid levels of phthalates and
conception, although DEHP exposure has been associated
with increased time to conception and miscarriage in mice
studies [summarized by Gore et al. (125)].
C)PESTICIDES. Exposures to higher levels of the pesticides
DDT and DDE have been associated with subfertility and
increased risk of miscarriage in observational studies from
countries with a high environmental exposure [summarized
by Gore et al. (125)].
2. Heavy metals
Mercury does not appear to be teratogenic within the con-
centrations expected due to occupational exposure (149),
and in a study of women undergoing IVF treatment, total
hair mercury was measured and the concentration did not
correlate with any IVF treatment outcome, response to
ovarian stimulation, oocyte fertilization, embryonic devel-
opment, pregnancy rate, or live birth rate when controlled
for confounders (376).
3. Other environmental exposures
Flight attendants and health workers using ionizing radia-
tion may be at increased risk of exposure to radiation
through their work (10, 38), which may have a detrimental
impact on the developing embryo and potentially increase
the risk of miscarriage; hence, employers should develop
guidelines for the occupational exposure to ionizing radia-
tion for pregnant women.
There is also the possibility that environmental exposure to
excessive noise may have detrimental effects upon and im-
plantation and increase the risk of miscarriage (278).
In summary, there is extensive evidence derived from ani-
mal studies of a negative influence of environmental chem-
icals on many aspects of female fertility: follicular number,
ovulation, meiosis, and embryo implantation; however, the
evidence of such negative associations in humans is often
lacking and contradictory. Further epidemiological studies
may assist in the clarification of these associations. How-
ever, the inherent difficulty with human studies lies with the
varying human exposures, often over many years of poten-
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
899Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
tial exposure, potential intergenerational influences and the
multiple confounding variables that are present when
studying fertility in a population.
VI. CONCLUSION
This review aimed to provide an overview of the processes
involved in conception, embryo implantation, and embry-
onic development. It provided an insight into the patholog-
ical conditions that may impair these processes and result in
a couple having difficulty conceiving and discussed revers-
ible lifestyle and environmental factors that may impact on
conception.
ACKNOWLEDGMENTS
Address for reprint requests and other correspondence: R. J.
Hart, School of Women’s and Infants Health, The Univer-
sity of Western Australia, King Edward Memorial Hospital,
374 Bagot Rd., Subiaco, Perth Western Australia 6008
(e-mail: roger.hart@uwa.edu.au).
DISCLOSURES
The author is the Medical Director of the IVF unit Fertility
Specialists of Western Australia and is a shareholder in
Western IVF.
REFERENCES
1. Abbott DH, Barnett DK, Bruns CM, Dumesic DA. Androgen excess fetal program-
ming of female reproduction: a developmental aetiology for polycystic ovary syn-
drome? Hum Reprod Update 11: 357–374, 2005.
2. Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of
oxidative stress on female reproduction: a review. Reprod Biol Endocrinol 10: 49, 2012.
3. Ahmad A, Ahmed A, Patrizio P. Cystic fibrosis and fertility. Curr Opin Obstet Gynecol
25: 167–172, 2013.
4. Al-Khateeb GM, Sater MS, Finan RR, Mustafa FE, Al-Busaidi AS, Al-Sulaiti MA, Almawi
WY. Analysis of interleukin-18 promoter polymorphisms and changes in interleu-
kin-18 serum levels underscores the involvement of interleukin-18 in recurrent spon-
taneous miscarriage. Fertil Steril 96: 921–926, 2011.
5. Alak BM, Coskun S, Friedman CI, Kennard EA, Kim MH, Seifer DB. Activin A stimu-
lates meiotic maturation of human oocytes and modulates granulosa cell steroidogen-
esis in vitro. Fertil Steril 70: 1126 –1130, 1998.
6. Alborzi S, Keramati P, Younesi M, Samsami A, Dadras N. The impact of laparoscopic
cystectomy on ovarian reserve in patients with unilateral and bilateral endometri-
omas. Fertil Steril 101: 427– 434, 2014.
7. Allahbadia GN, Saradogan E, Djahanbakhch O. The Fallopian Tube. Kent, UK: Ashnan,
2009.
8. Allersma T, Farquhar C, Cantineau AE. Natural cycle in vitro fertilisation (IVF) for
subfertile couples. Cochrane Database Syst Rev 8: CD010550, 2013.
9. Alliance JH, Hobot PA. Evidence Based Guidelines for the Assessment and Management
of Polycystic Ovary Syndrome. Melbourne: Jean Hailes for Women’s Health on behalf of
the PCOS Australian Alliance, 2015.
10. Anderson JL, Mertens CJ, Grajewski B, Luo L, Tseng CY, Cassinelli RT 2nd. Flight
attendant radiation dose from solar particle events. Aviation Space Environ Med 85:
828 – 832, 2014.
11. Arakawa C, Yoshinaga J, Mizumoto Y, Abe M. Preliminary study on measurement of
human fecundity applicability of time to pregnancy with Japanese subjects. Jpn J Public
Health 50: 414 – 419, 2003.
12. Arita A, Costa M. Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and
cadmium. Metallomics 1: 222–228, 2009.
13. Artal R, Catanzaro RB, Gavard JA, Mostello DJ, Friganza JC. A lifestyle intervention of
weight-gain restriction: diet and exercise in obese women with gestational diabetes
mellitus. Appl Physiol Nutr Metab 32: 596 – 601, 2007.
14. Ashraf MW. Levels of heavy metals in popular cigarette brands and exposure to these
metals via smoking. Scientific World J 2012: 729430, 2012.
15. Aune D, Saugstad OD, Henriksen T, Tonstad S. Maternal body mass index and the risk
of fetal death, stillbirth, and infant death: a systematic review and meta-analysis. JAMA
311: 1536 –1546, 2014.
16. Aviles M, Gutierrez-Adan A, Coy P. Oviductal secretions: will they be key factors for
the future ARTs? Mol Hum Reprod 16: 896 –906, 2010.
17. Aziz M, Sidelmann JJ, Faber J, Wissing MM, Naver KV, Mikkelsen AL, Nilas L, Skouby
SO. Polycystic ovary syndrome: cardiovascular risk factors according to specific phe-
notypes. Acta Obstet Gynecol Scand 94: 1082–1089, 2015.
18. Azziz R. PCOS in 2015: New insights into the genetics of polycystic ovary syndrome.
Nature Rev Endocrinol 12: 74 –75, 2016.
19. Azziz R, Carmina E, Dewailly D, Diamanti-Kandarakis E, Escobar-Morreale HF,
Futterweit W, Janssen OE, Legro RS, Norman RJ, Taylor AE, Witchel SF, Task
Force on the Phenotype of the Polycystic Ovary Syndrome of The Androgen
Excess and PCOS Society. The Androgen Excess and PCOS Society criteria for the
polycystic ovary syndrome: the complete task force report. Fertil Steril 91: 456 –
488, 2009.
20. Baerwald AR, Adams GP, Pierson RA. Ovarian antral folliculogenesis during the hu-
man menstrual cycle: a review. Hum Reprod Update 18: 73–91, 2012.
21. Baird DT. Evidence in vivo for the two-cell hypothesis of oestrogen synthesis by the
sheep Graafian follicle. J Reprod Fertil 50: 183–185, 1977.
22. Baker TG. A quantitative and cytological study of germ cells in human ovaries. Proc R
Soc Lond Ser B 158: 417– 433, 1963.
23. Balen AH. Ovulation induction in the management of anovulatory polycystic ovary
syndrome. Mol Cell Endocrinol 373: 77– 82, 2013.
24. Barnes RB, Rosenfield RL, Ehrmann DA, Cara JF, Cuttler L, Levitsky LL, Rosenthal IM.
Ovarian hyperandrogynism as a result of congenital adrenal virilizing disorders: evi-
dence for perinatal masculinization of neuroendocrine function in women. J Clin
Endocrinol Metab 79: 1328 –1333, 1994.
25. Barnhart K, Dunsmoor-Su R, Coutifaris C. Effect of endometriosis on in vitro fertil-
ization. Fertil Steril 77: 1148 –1155, 2002.
26. Barrett ES, Sobolewski M. Polycystic ovary syndrome: do endocrine-disrupting chem-
icals play a role? Semin Reprod Med 32: 166 –176, 2014.
27. Basavarajappa MS, Craig ZR, Hernandez-Ochoa I, Paulose T, Leslie TC, Flaws JA.
Methoxychlor reduces estradiol levels by altering steroidogenesis and metabolism in
mouse antral follicles in vitro. Toxicol Appl Pharmacol 253: 161–169, 2011.
28. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM,
Brynes AE, Low MJ, Ghatei MA, Cone RD, Bloom SR. Gut hormone PYY(3–36)
physiologically inhibits food intake. Nature 418: 650 – 654, 2002.
29. Bellver J, Martinez-Conejero JA, Labarta E, Alama P, Melo MA, Remohi J, Pellicer A,
Horcajadas JA. Endometrial gene expression in the window of implantation is altered
in obese women especially in association with polycystic ovary syndrome. Fertil Steril
95: 2335–2341, 2011.
30. Bellver J, Melo MA, Bosch E, Serra V, Remohi J, Pellicer A. Obesity and poor repro-
ductive outcome: the potential role of the endometrium. Fertil Steril 88: 446– 451,
2007.
31. Ben-Nagi J, Miell J, Yazbek J, Holland T, Jurkovic D. The effect of hysteroscopic
polypectomy on the concentrations of endometrial implantation factors in uterine
flushings. Reprod Biomed Online 19: 737–744, 2009.
ROGER J. HART
900 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
32. Bentov Y, Yavorska T, Esfandiari N, Jurisicova A, Casper RF. The contribution of
mitochondrial function to reproductive aging. J Assist Reprod Genet 28: 773–783,
2011.
33. Berga S, Naftolin F. Neuroendocrine control of ovulation. Gynecol Endocrinol 28 Suppl
1: 9 –13, 2012.
34. Berga SL, Daniels TL, Giles DE. Women with functional hypothalamic amenorrhea
but not other forms of anovulation display amplified cortisol concentrations. Fertil
Steril 67: 1024 –1030, 1997.
35. Berga SL, Marcus MD, Loucks TL, Hlastala S, Ringham R, Krohn MA. Recovery of
ovarian activity in women with functional hypothalamic amenorrhea who were
treated with cognitive behavior therapy. Fertil Steril 80: 976 –981, 2003.
36. Bhattacharya P, Keating AF. Impact of environmental exposures on ovarian function
and role of xenobiotic metabolism during ovotoxicity. Toxicol Appl Pharmacol 261:
227–235, 2012.
37. Bhattacharya S, Johnson N, Tijani HA, Hart R, Pandey S, Gibreel AF. Female infertility.
BMJ Clin Evid pii: 0819, 2010.
38. Birnie AM, Keoghane SR. Radiation exposure to a pregnant urological surgeon: what
is safe? BJU Int 115: 683– 685, 2015.
39. Bloom MS, Kim D, Vom Saal FS, Taylor JA, Cheng G, Lamb JD, Fujimoto VY. Bisphenol
A exposure reduces the estradiol response to gonadotropin stimulation during in vitro
fertilization. Fertil Steril 96: 672– 677, 2011.
40. Boadi WY, Shurtz-Swirski R, Barnea ER, Urbach J, Brandes JM, Philo E, Yannai S.
Secretion of human chorionic gonadotropin in superfused young placental tissue ex-
posed to cadmium. Arch Toxicol 66: 95–99, 1992.
41. Boomsma CM, Eijkemans MJ, Hughes EG, Visser GH, Fauser BC, Macklon NS. A
meta-analysis of pregnancy outcomes in women with polycystic ovary syndrome.
Hum Reprod Update 12: 673– 683, 2006.
42. Boucher JG, Boudreau A, Ahmed S, Atlas E. In vitro effects of bisphenol A beta-D-
glucuronide (BPA-G) on adipogenesis in human and murine preadipocytes. Environ
Health Perspect 123: 1287–1293, 2015.
43. Bouyer J, Coste J, Shojaei T, Pouly JL, Fernandez H, Gerbaud L, Job-Spira N. Risk
factors for ectopic pregnancy: a comprehensive analysis based on a large case-control,
population-based study in France. Am J Epidemiol 157: 185–194, 2003.
44. Brown J, Farquhar C. Endometriosis: an overview of Cochrane Reviews. Cochrane
Database Syst Rev 3: CD009590, 2014.
45. Burger HG, Cahir N, Robertson DM, Groome NP, Dudley E, Green A, Dennerstein
L. Serum inhibins A and B fall differentially as FSH rises in perimenopausal women. Clin
Endocrinol 48: 809 – 813, 1998.
46. Burger HG, Hale GE, Dennerstein L, Robertson DM. Cycle and hormone changes
during perimenopause: the key role of ovarian function. Menopause 15: 603– 612,
2008.
47. Cai LY, Izumi S, Suzuki T, Goya K, Nakamura E, Sugiyama T, Kobayashi H. Dioxins in
ascites and serum of women with endometriosis: a pilot study. Hum Reprod 26:
117–126, 2011.
48. Cakmak H, Taylor HS. Implantation failure: molecular mechanisms and clinical treat-
ment. Hum Reprod Update 17: 242–253, 2011.
49. Camlin NJ, McLaughlin EA, Holt JE. Through the smoke: use of in vivo and in vitro
cigarette smoking models to elucidate its effect on female fertility. Toxicol Appl Phar-
macol 281: 266 –275, 2014.
50. Care AS, Diener KR, Jasper MJ, Brown HM, Ingman WV, Robertson SA. Macrophages
regulate corpus luteum development during embryo implantation in mice. J Clin Invest
123: 3472–3487, 2013.
51. Carlsson IB, Scott JE, Visser JA, Ritvos O, Themmen AP, Hovatta O. Anti-Mullerian
hormone inhibits initiation of growth of human primordial ovarian follicles in vitro.
Hum Reprod 21: 2223–2227, 2006.
52. Caserta D, Di Segni N, Mallozzi M, Giovanale V, Mantovani A, Marci R, Moscarini M.
Bisphenol A and the female reproductive tract: an overview of recent laboratory
evidence and epidemiological studies. Reprod Biol Endocrinol 12: 37, 2014.
53. Cermik D, Selam B, Taylor HS. Regulation of HOXA-10 expression by testosterone in
vitro and in the endometrium of patients with polycystic ovary syndrome. J Clin
Endocrinol Metab 88: 238 –243, 2003.
54. Cha J, Sun X, Dey SK. Mechanisms of implantation: strategies for successful preg-
nancy. Nature Med 18: 1754 –1767, 2012.
55. Chan HC, Shi QX, Zhou CX, Wang XF, Xu WM, Chen WY, Chen AJ, Ni Y, Yuan YY.
Critical role of CFTR in uterine bicarbonate secretion and the fertilizing capacity of
sperm. Mol Cell Endocrinol 250: 106 –113, 2006.
56. Chao HH, Zhang XF, Chen B, Pan B, Zhang LJ, Li L, Sun XF, Shi QH, Shen W.
Bisphenol A exposure modifies methylation of imprinted genes in mouse oocytes via
the estrogen receptor signaling pathway. Histochem Cell Biol 137: 249 –259, 2012.
57. Chappel S. The role of mitochondria from mature oocyte to viable blastocyst. Obstet
Gynecol Int 2013: 183024, 2013.
58. Cheng EY, Hunt PA, Naluai-Cecchini TA, Fligner CL, Fujimoto VY, Pasternack TL,
Schwartz JM, Steinauer JE, Woodruff TJ, Cherry SM, Hansen TA, Vallente RU, Bro-
man KW, Hassold TJ. Meiotic recombination in human oocytes. PLoS Genet 5:
e1000661, 2009.
59. Choy CM, Lam CW, Cheung LT, Briton-Jones CM, Cheung LP, Haines CJ. Infertility,
blood mercury concentrations and dietary seafood consumption: a case-control
study. BJOG 109: 1121–1125, 2002.
60. Chrousos GP, Torpy DJ, Gold PW. Interactions between the hypothalamic-pituitary-
adrenal axis and the female reproductive system: clinical implications. Ann Intern Med
129: 229 –240, 1998.
61. Clark AM, Thornley B, Tomlinson L, Galletley C, Norman RJ. Weight loss in obese
infertile women results in improvement in reproductive outcome for all forms of
fertility treatment. Hum Reprod 13: 1502–1505, 1998.
62. Clift D, Schuh M. Restarting life: fertilization and the transition from meiosis to mitosis.
Nature Rev Mol Cell Biol 14: 549 –562, 2013.
63. Cobellis L, Latini G, De Felice C, Razzi S, Paris I, Ruggieri F, Mazzeo P, Petraglia F.
High plasma concentrations of di-(2-ethylhexyl)-phthalate in women with endome-
triosis. Hum Reprod 18: 1512–1515, 2003.
64. Codner E, Merino PM, Tena-Sempere M. Female reproduction and type 1 diabetes:
from mechanisms to clinical findings. Hum Reprod Update 18: 568 –585, 2012.
65. Comninos AN, Jayasena CN, Dhillo WS. The relationship between gut and adipose
hormones, and reproduction. Hum Reprod Update 20: 153–174, 2014.
66. Conneely OM, Mulac-Jericevic B, DeMayo F, Lydon JP, O’Malley BW. Reproductive
functions of progesterone receptors. Recent Prog Horm Res 57: 339 –355, 2002.
67. Cordts EB, Christofolini DM, Dos Santos AA, Bianco B, Barbosa CP. Genetic aspects
of premature ovarian failure: a literature review. Arch Gynecol Obstet 283: 635– 643,
2011.
68. Cosby NC, Dukelow WR. Toxicology of maternally ingested trichloroethylene (TCE)
on embryonal and fetal development in mice and of TCE metabolites on in vitro
fertilization. Fundam Appl Toxicol 19: 268 –274, 1992.
69. Cragin LA, Kesner JS, Bachand AM, Barr DB, Meadows JW, Krieg EF, Reif JS. Men-
strual cycle characteristics and reproductive hormone levels in women exposed to
atrazine in drinking water. Environ Res 111: 1293–1301, 2011.
70. Craig J, Orisaka M, Wang H, Orisaka S, Thompson W, Zhu C, Kotsuji F, Tsang BK.
Gonadotropin and intra-ovarian signals regulating follicle development and atresia: the
delicate balance between life and death. Front Biosci 12: 3628 –3639, 2007.
71. Cramer DW, Welch WR, Cassells S, Scully RE. Mumps, menarche, menopause, and
ovarian cancer. Am J Obstet Gynecol 147: 1– 6, 1983.
72. Crujeiras AB, Casanueva FF. Obesity and the reproductive system disorders: epige-
netics as a potential bridge. Hum Reprod Update 21: 249 –261, 2015.
73. Cummins JM. Fertilization and elimination of the paternal mitochondrial genome.
Hum Reprod 15 Suppl 2: 92–101, 2000.
74. D’Aiuto F, Parkar M, Andreou G, Suvan J, Brett PM, Ready D, Tonetti MS. Periodon-
titis and systemic inflammation: control of the local infection is associated with a
reduction in serum inflammatory markers. J Dent Res 83: 156 –160, 2004.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
901Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
75. D’Angelo A, Brown J, Amso NN. Coasting (withholding gonadotrophins) for prevent-
ing ovarian hyperstimulation syndrome. Cochrane Database Syst Rev CD002811, 2011.
76. Darville T, Hiltke TJ. Pathogenesis of genital tract disease due to Chlamydia trachoma-
tis.J Infectious Dis 201 Suppl 2: S114 –125, 2010.
77. De Groot L, Abalovich M, Alexander EK, Amino N, Barbour L, Cobin RH, Eastman CJ,
Lazarus JH, Luton D, Mandel SJ, Mestman J, Rovet J, Sullivan S. Management of thyroid
dysfunction during pregnancy and postpartum: an Endocrine Society clinical practice
guideline. J Clin Endocrinol Metab 97: 2543–2565, 2012.
78. De Souza MJ, Leidy HJ, O’Donnell E, Lasley B, Williams NI. Fasting ghrelin levels in
physically active women: relationship with menstrual disturbances and metabolic
hormones. J Clin Endocrinol Metab 89: 3536 –3542, 2004.
79. De Souza MJ, Miller BE, Loucks AB, Luciano AA, Pescatello LS, Campbell CG, Lasley
BL. High frequency of luteal phase deficiency and anovulation in recreational women
runners: blunted elevation in follicle-stimulating hormone observed during luteal-
follicular transition. J Clin Endocrinol Metab 83: 4220 – 4232, 1998.
80. Dechanet C, Anahory T, Mathieu Daude JC, Quantin X, Reyftmann L, Hamamah S,
Hedon B, Dechaud H. Effects of cigarette smoking on reproduction. Hum Reprod
Update 17: 76 –95, 2011.
81. Dessolle L, Darai E, Cornet D, Rouzier R, Coutant C, Mandelbaum J, Antoine JM.
Determinants of pregnancy rate in the donor oocyte model: a multivariate analysis of
450 frozen-thawed embryo transfers. Hum Reprod 24: 3082–3089, 2009.
82. Devoto L, Fuentes A, Kohen P, Cespedes P, Palomino A, Pommer R, Munoz A,
Strauss JF 3rd. The human corpus luteum: life cycle and function in natural cycles.
Fertil Steril 92: 1067–1079, 2009.
83. Devoto L, Kohen P, Munoz A, Strauss JF 3rd. Human corpus luteum physiology and
the luteal-phase dysfunction associated with ovarian stimulation. Reprod Biomed On-
line 18 Suppl 2: 19 –24, 2009.
84. Devoto L, Vega M, Kohen P, Castro O, Carvallo P, Palomino A. Molecular regulation
of progesterone secretion by the human corpus luteum throughout the menstrual
cycle. J Reprod Immunol 55: 11–20, 2002.
85. Dey SK, Lim H, Das SK, Reese J, Paria BC, Daikoku T, Wang H. Molecular cues to
implantation. Endocr Rev 25: 341–373, 2004.
86. Ding YS, Trommel JS, Yan XJ, Ashley D, Watson CH. Determination of 14 polycyclic
aromatic hydrocarbons in mainstream smoke from domestic cigarettes. Environ Sci
Technol 39: 471– 478, 2005.
87. Doherty DA, Newnham JP, Bower C, Hart R. Implications of polycystic ovary syn-
drome for pregnancy and for the health of offspring. Obstet Gynecol 125: 1397–1406,
2015.
88. Domecq JP, Prutsky G, Mullan RJ, Hazem A, Sundaresh V, Elamin MB, Phung OJ,
Wang A, Hoeger K, Pasquali R, Erwin P, Bodde A, Montori VM, Murad MH. Lifestyle
modification programs in polycystic ovary syndrome: systematic review and meta-
analysis. J Clin Endocrinol Metab 98: 4655– 4663, 2013.
89. Dorrington JH, Moon YS, Armstrong DT. Estradiol-17beta biosynthesis in cultured
granulosa cells from hypophysectomized immature rats; stimulation by follicle-stim-
ulating hormone. Endocrinology 97: 1328 –1331, 1975.
90. Duffy JM, Arambage K, Correa FJ, Olive D, Farquhar C, Garry R, Barlow DH, Jacob-
son TZ. Laparoscopic surgery for endometriosis. Cochrane Database Syst Rev 4:
CD011031, 2014.
91. Dumesic DA, Abbott DH. Implications of polycystic ovary syndrome on oocyte de-
velopment. Semin Reprod Med 26: 53– 61, 2008.
92. Dumesic DA, Meldrum DR, Katz-Jaffe MG, Krisher RL, Schoolcraft WB. Oocyte
environment: follicular fluid and cumulus cells are critical for oocyte health. Fertil Steril
103: 303–316, 2015.
93. Ehrlich S, Williams PL, Missmer SA, Flaws JA, Berry KF, Calafat AM, Ye X, Petrozza JC,
Wright D, Hauser R. Urinary bisphenol A concentrations and implantation failure
among women undergoing in vitro fertilization. Environ Health Perspect 120: 978 –983,
2012.
94. Ehrlich S, Williams PL, Missmer SA, Flaws JA, Ye X, Calafat AM, Petrozza JC, Wright
D, Hauser R. Urinary bisphenol A concentrations and early reproductive health out-
comes among women undergoing IVF. Hum Reprod 27: 3583–3592, 2012.
95. Erden O, Imir A, Guvenal T, Muslehiddinoglu A, Arici S, Cetin M, Cetin A. Investiga-
tion of the effects of heparin and low molecular weight heparin on E-cadherin and
laminin expression in rat pregnancy by immunohistochemistry. Hum Reprod 21:
3014 –3018, 2006.
96. Eroschenko VP, Swartz WJ, Ford LC. Decreased superovulation in adult mice follow-
ing neonatal exposures to technical methoxychlor. Reprod Toxicol 11: 807– 814, 1997.
97. Evans JJ, Anderson GM. Balancing ovulation and anovulation: integration of the repro-
ductive and energy balance axes by neuropeptides. Hum Reprod Update 18: 313–332,
2012.
98. Evans JP. Sperm-egg interaction. Annu Rev Physiol 74: 477–502, 2012.
99. Ezzati M. The role of dynein and its mutations in the ciliary activity of human fallopian
tubes. In: The Fallopian Tube, edited by Allahbadia GN, Saradogan E, and Djahan-
bakhch O. Kent, UK: Anshan, 2009.
100. Ezzati M, Djahanbakhch O, Arian S, Carr BR. Tubal transport of gametes and em-
bryos: a review of physiology and pathophysiology. J Assist Reprod Genet 31: 1337–
1347, 2014.
101. Feldkamp ML, Botto LD, Carey JC. Reflections on the etiology of structural birth
defects: established teratogens and risk factors. Birth Defects Res A Clin Mol Teratol
103: 652– 655, 2015.
102. Ferk P, Pohar Perme M, Teran N, Gersak K. Androgen receptor gene (CAG)(n)
polymorphism in patients with polycystic ovary syndrome. Fertil Steril 90: 860 – 863,
2008.
103. Fernandez M, Bourguignon N, Lux-Lantos V, Libertun C. Neonatal exposure to bis-
phenol a and reproductive and endocrine alterations resembling the polycystic ovar-
ian syndrome in adult rats. Environ Health Perspect 118: 1217–1222, 2010.
104. Florea AM, Yamoah EN, Dopp E. Intracellular calcium disturbances induced by arse-
nic and its methylated derivatives in relation to genomic damage and apoptosis induc-
tion. Environ Health Perspect 113: 659 – 664, 2005.
105. Ford PJ, Raphael SL, Cullinan MP, Jenkins AJ, West MJ, Seymour GJ. Why should a
doctor be interested in oral disease? Expert Rev Cardiovasc Ther 8: 1483–1493, 2010.
106. Forsdike RA, Hardy K, Bull L, Stark J, Webber LJ, Stubbs S, Robinson JE, Franks S.
Disordered follicle development in ovaries of prenatally androgenized ewes. J Endo-
crinol 192: 421– 428, 2007.
107. Fortuno C, Labarta E. Genetics of primary ovarian insufficiency: a review. J Assist
Reprod Genet 31: 1573–1585, 2014.
108. Fragouli E, Bianchi V, Patrizio P, Obradors A, Huang Z, Borini A, Delhanty JD, Wells
D. Transcriptomic profiling of human oocytes: association of meiotic aneuploidy and
altered oocyte gene expression. Mol Hum Reprod 16: 570 –582, 2010.
109. Franasiak JM, Forman EJ, Hong KH, Werner MD, Upham KM, Treff NR, Scott RT Jr.
The nature of aneuploidy with increasing age of the female partner: a review of 15,169
consecutive trophectoderm biopsies evaluated with comprehensive chromosomal
screening. Fertil Steril 101: 656 – 663, 2014.
110. Freour T, Dessolle L, Lammers J, Lattes S, Barriere P. Comparison of embryo mor-
phokinetics after in vitro fertilization-intracytoplasmic sperm injection in smoking and
nonsmoking women. Fertil Steril 99: 1944 –1950, 2013.
111. Frisch RE, McArthur JW. Menstrual cycles: fatness as a determinant of minimum
weight for height necessary for their maintenance or onset. Science 185: 949–951,
1974.
112. Fuentes A, Munoz A, Barnhart K, Arguello B, Diaz M, Pommer R. Recent cigarette
smoking and assisted reproductive technologies outcome. Fertil Steril 93: 89 –95,
2010.
113. Galusca B, Prevost G, Germain N, Dubuc I, Ling Y, Anouar Y, Estour B, Chartrel N.
Neuropeptide Y and alpha-MSH circadian levels in two populations with low body
weight: anorexia nervosa and constitutional thinness. PLoS One 10: e0122040, 2015.
114. Gardner DK, Meseguer M, Rubio C, Treff NR. Diagnosis of human preimplantation
embryo viability. Hum Reprod Update 21: 727–747, 2015.
115. Gardner DK, Wale PL, Collins R, Lane M. Glucose consumption of single post-com-
paction human embryos is predictive of embryo sex and live birth outcome. Hum
Reprod 26: 1981–1986, 2011.
ROGER J. HART
902 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
116. Gaskins AJ, Rich-Edwards JW, Lawson CC, Schernhammer ES, Missmer SA, Chavarro
JE. Work schedule and physical factors in relation to fecundity in nurses. Occup Environ
Med 72: 777–783, 2015.
117. Gaskins AJ, Rich-Edwards JW, Missmer SA, Rosner B, Chavarro JE. Association of
fecundity with changes in adult female weight. Obstet Gynecol 126: 850 – 858, 2015.
118. Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, Hou Y, Han ZM, Schatten H, Sun
QY. DNA methylation in oocytes and liver of female mice and their offspring: effects
of high-fat-diet-induced obesity. Environ Health Perspect 122: 159 –164, 2014.
120. Ge ZJ, Schatten H, Zhang CL, Sun QY. Oocyte ageing and epigenetics. Reproduction
149: R103–114, 2015.
121. Genbacev O, Bass KE, Joslin RJ, Fisher SJ. Maternal smoking inhibits early human
cytotrophoblast differentiation. Reprod Toxicol 9: 245–255, 1995.
122. Genbacev O, McMaster MT, Lazic J, Nedeljkovic S, Cvetkovic M, Joslin R, Fisher SJ.
Concordant in situ and in vitro data show that maternal cigarette smoking negatively
regulates placental cytotrophoblast passage through the cell cycle. Reprod Toxicol 14:
495–506, 2000.
123. George JT, Seminara SB. Kisspeptin and the hypothalamic control of reproduction:
lessons from the human. Endocrinology 153: 5130 –5136, 2012.
124. Glueck CJ, Wang P, Fontaine RN, Sieve-Smith L, Tracy T, Moore SK. Plasminogen
activator inhibitor activity: an independent risk factor for the high miscarriage rate
during pregnancy in women with polycystic ovary syndrome. Metab Clin Exp 48:
1589 –1595, 1999.
125. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT.
EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupt-
ing Chemicals. Endocr Rev 36: E1–E150, 2015.
126. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT.
Executive Summary to EDC-2: The Endocrine Society’s Second Scientific Statement
on Endocrine-Disrupting Chemicals. Endocr Rev 36: 593– 602, 2015.
127. Gormack AA, Peek JC, Derraik JG, Gluckman PD, Young NL, Cutfield WS. Many
women undergoing fertility treatment make poor lifestyle choices that may affect
treatment outcome. Hum Reprod 30: 1617–1624, 2015.
128. Goswami D, Conway GS. Premature ovarian failure. Hum Reprod Update 11: 391–
410, 2005.
129. Gougeon A. Dynamics of follicular growth in the human: a model from preliminary
results. Hum Reprod 1: 81– 87, 1986.
130. Grodnitskaya EE, Kurtser MA. Homocysteine metabolism in polycystic ovary syn-
drome. Gynecol Endocrinol 28: 186 –189, 2012.
131. Grondahl ML, Yding Andersen C, Bogstad J, Nielsen FC, Meinertz H, Borup R. Gene
expression profiles of single human mature oocytes in relation to age. Hum Reprod 25:
957–968, 2010.
132. Guelinckx I, Devlieger R, Vansant G. Reproductive outcome after bariatric surgery: a
critical review. Hum Reprod Update 15: 189 –201, 2009.
133. Guglielmino MR, Santonocito M, Vento M, Ragusa M, Barbagallo D, Borzi P, Casciano
I, Banelli B, Barbieri O, Astigiano S, Scollo P, Romani M, Purrello M, Di Pietro C.
TAp73 is downregulated in oocytes from women of advanced reproductive age. Cell
Cycle 10: 3253–3256, 2011.
134. Halasz M, Szekeres-Bartho J. The role of progesterone in implantation and tropho-
blast invasion. J Reprod Immunol 97: 43–50, 2013.
135. Halse RE, Wallman KE, Dimmock JA, Newnham JP, Guelfi KJ. Home-based exercise
improves fitness and exercise attitude and intention in women with GDM. Med Sci
Sports Exerc 47: 1698 –1704, 2015.
136. Handyside AH. Molecular origin of female meiotic aneuploidies. Biochim Biophys Acta
1822: 1913–1920, 2012.
137. Hansen M, Bower C, Milne E, de Klerk N, Kurinczuk JJ. Assisted reproductive tech-
nologies and the risk of birth defects–a systematic review. Hum Reprod 20: 328 –338,
2005.
138. Hart R. Preservation of fertility in adults and children diagnosed with cancer. BMJ 337:
a2045, 2008.
139. Hart R, Doherty DA. The potential implications of a PCOS diagnosis on a woman’s
long-term health using data linkage. J Clin Endocrinol Metab 100: 911–919, 2015.
140. Hart R, Doherty DA. The potential implications of a PCOS diagnosis on a woman’s
long-term health using data linkage. J Clin Endocrinol Metab jc20143886, 2014.
141. Hart R, Doherty DA, Frederiksen H, Keelan JA, Hickey M, Sloboda D, Pennell CE,
Newnham JP, Skakkebaek NE, Main KM. The influence of antenatal exposure to
phthalates on subsequent female reproductive development in adolescence: a pilot
study. Reproduction 147: 379 –390, 2014.
142. Hart R, Doherty DA, Norman RJ, Franks S, Dickinson JE, Hickey M, Sloboda DM.
Serum antimullerian hormone (AMH) levels are elevated in adolescent girls with
polycystic ovaries and the polycystic ovarian syndrome (PCOS). Fertil Steril 94: 1118 –
1121, 2010.
143. Hart R, Doherty DA, Pennell CE, Newnham IA, Newnham JP. Periodontal disease: a
potential modifiable risk factor limiting conception. Hum Reprod 27: 1332–1342,
2012.
144. Hart R, Doherty DA, Pennell CE, Newnham IA, Newnham JP. Periodontal disease: a
potential modifiable risk factor limiting conception. Hum Reprod 27: 1332–1342,
2012.
145. Hart R, Khalaf Y, Yeong CT, Seed P, Taylor A, Braude P. A prospective controlled
study of the effect of intramural uterine fibroids on the outcome of assisted concep-
tion. Hum Reprod 16: 2411–2417, 2001.
146. Hart R, Norman RJ. The longer-term health outcomes for children born as a result of
IVF treatment. Part I–General health outcomes. Hum Reprod Update 19: 232–243,
2013.
147. Hart R, Sloboda DM, Doherty DA, Norman RJ, Atkinson HC, Newnham JP, Dickinson
JE, Hickey M. Circulating maternal testosterone concentrations at 18 weeks of ges-
tation predict circulating levels of antimullerian hormone in adolescence: a prospec-
tive cohort study. Fertil Steril 94: 1544 –1547, 2010.
148. Hart RJ, Hickey M, Maouris P, Buckett W. Excisional surgery versus ablative surgery
for ovarian endometriomata. Cochrane Database Syst Rev CD004992, 2008.
149. Heggland I, Irgens A, Tollanes M, Romundstad P, Syversen T, Svendsen K, Melo I, Hilt
B. Pregnancy outcomes among female dental personnel–a registry-based retrospec-
tive cohort study. Scand J Work Environ Health 37: 539 –546, 2011.
150. Hewitt JK, Jayasinghe Y, Amor DJ, Gillam LH, Warne GL, Grover S, Zacharin MR.
Fertility in Turner syndrome. Clin Endocrinol 79: 606 – 614, 2013.
151. Hickey M, Sloboda DM, Atkinson HC, Doherty DA, Franks S, Norman RJ, Newnham
JP, Hart R. The relationship between maternal and umbilical cord androgen levels and
polycystic ovary syndrome in adolescence: a prospective cohort study. J Clin Endocri-
nol Metab 94: 3714 –3720, 2009.
152. Hjordt Hansen MV, Dalsgaard T, Hartwell D, Skovlund CW, Lidegaard O. Reproduc-
tive prognosis in endometriosis. A national cohort study. Acta Obstet Gynecol Scand
93: 483– 489, 2014.
153. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA,
Braverman LE. Serum TSH, T(4), and thyroid antibodies in the United States popu-
lation (1988 to 1994): National Health and Nutrition Examination Survey (NHANES
III). J Clin Endocrinol Metab 87: 489 – 499, 2002.
154. Holubcova Z, Blayney M, Elder K, Schuh M. Human oocytes. Error-prone chromo-
some-mediated spindle assembly favors chromosome segregation defects in human
oocytes. Science 348: 1143–1147, 2015.
155. Homburg R, Hendriks ML, Konig TE, Anderson RA, Balen AH, Brincat M, Child T,
Davies M, D’Hooghe T, Martinez A, Rajkhowa M, Rueda-Saenz R, Hompes P, Lam-
balk CB. Clomifene citrate or low-dose FSH for the first-line treatment of infertile
women with anovulation associated with polycystic ovary syndrome: a prospective
randomized multinational study. Hum Reprod 27: 468 – 473, 2012.
156. Hooser SB, Douds DP, DeMerell DG, Hoyer PB, Sipes IG. Long-term ovarian and
gonadotropin changes in mice exposed to 4-vinylcyclohexene. Reprod Toxicol 8: 315–
323, 1994.
157. Huang RC, Mori TA, Beilin LJ. Early life programming of cardiometabolic disease in the
Western Australian pregnancy cohort (Raine) study. Clin Exp Pharmacol Physiol 39:
973–978, 2012.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
903Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
158. Hughes E, Brown J, Collins JJ, Farquhar C, Fedorkow DM, Vandekerckhove P. Ovu-
lation suppression for endometriosis. Cochrane Database Syst Rev CD000155, 2007.
159. Hulboy DL, Rudolph LA, Matrisian LM. Matrix metalloproteinases as mediators of
reproductive function. Mol Hum Reprod 3: 27– 45, 1997.
160. Hunt PA, Lawson C, Gieske M, Murdoch B, Smith H, Marre A, Hassold T, VandeVoort
CA. Bisphenol A alters early oogenesis and follicle formation in the fetal ovary of the
rhesus monkey. Proc Natl Acad Sci USA 109: 17525–17530, 2012.
161. Igawa Y, Keating AF, Rajapaksa KS, Sipes IG, Hoyer PB. Evaluation of ovotoxicity
induced by 7, 12-dimethylbenz[a]anthracene and its 3,4-diol metabolite utilizing a rat
in vitro ovarian culture system. Toxicol Appl Pharmacol 234: 361–369, 2009.
162. Ikezuki Y, Tsutsumi O, Takai Y, Kamei Y, Taketani Y. Determination of bisphenol A
concentrations in human biological fluids reveals significant early prenatal exposure.
Hum Reprod 17: 2839 –2841, 2002.
163. Isling LK, Boberg J, Jacobsen PR, Mandrup KR, Axelstad M, Christiansen S, Vinggaard
AM, Taxvig C, Kortenkamp A, Hass U. Late-life effects on rat reproductive system
after developmental exposure to mixtures of endocrine disrupters. Reproduction 147:
465– 476, 2014.
164. Ivanov P, Tsvyatkovska T, Konova E, Komsa-Penkova R. Inherited thrombophilia and
IVF failure: the impact of coagulation disorders on implantation process. Am J Reprod
Immunol 68: 189 –198, 2012.
165. Jablonska O, Shi Z, Valdez KE, Ting AY, Petroff BK. Temporal and anatomical sensi-
tivities to the aryl hydrocarbon receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin
leading to premature acyclicity with age in rats. Int J Androl 33: 405– 412, 2010.
166. Jensen TK, Scheike T, Keiding N, Schaumburg I, Grandjean P. Fecundability in relation
to body mass and menstrual cycle patterns. Epidemiology 10: 422– 428, 1999.
167. Jick H, Porter J. Relation between smoking and age of natural menopause. Report
from the Boston Collaborative Drug Surveillance Program, Boston University Medical
Center. Lancet 1: 1354 –1355, 1977.
168. Johansson K, Cnattingius S, Naslund I, Roos N, Trolle Lagerros Y, Granath F, Stepha-
nsson O, Neovius M. Outcomes of pregnancy after bariatric surgery. N Engl J Med
372: 814 – 824, 2015.
169. Jungheim ES, Schon SB, Schulte MB, DeUgarte DA, Fowler SA, Tuuli MG. IVF out-
comes in obese donor oocyte recipients: a systematic review and meta-analysis. Hum
Reprod 28: 2720 –2727, 2013.
170. Kadhel P, Monnier P, Boucoiran I, Chaillet N, Fraser WD. Organochlorine pollutants
and female fertility: a systematic review focusing on in vitro fertilization studies. Reprod
Sci 19: 1246 –1259, 2012.
171. Kallen CB, Arakane F, Christenson LK, Watari H, Devoto L, Strauss JF 3rd. Unveiling
the mechanism of action and regulation of the steroidogenic acute regulatory protein.
Mol Cell Endocrinol 145: 39 – 45, 1998.
172. Kandaraki E, Chatzigeorgiou A, Livadas S, Palioura E, Economou F, Koutsilieris M,
Palimeri S, Panidis D, Diamanti-Kandarakis E. Endocrine disruptors and polycystic
ovary syndrome (PCOS): elevated serum levels of bisphenol A in women with PCOS.
J Clin Endocrinol Metab 96: E480 – 484, 2011.
173. Karavan JR, Pepling ME. Effects of estrogenic compounds on neonatal oocyte devel-
opment. Reprod Toxicol 34: 51–56, 2012.
174. Kaufman F, Kogut MD, Donnell GN, Koch H, Goebelsmann U. Ovarian failure in
galactosaemia. Lancet 2: 737–738, 1979.
175. Kawato H, Tabata T, Minoura H, Murabayashi N, Ma N, Wang DF, Sagawa N. Factor
XII gene expression in endometrial stromal cells during decidualisation. Reprod Fertil
Dev 21: 840 – 847, 2009.
176. Kezele PR, Nilsson EE, Skinner MK. Insulin but not insulin-like growth factor-1 pro-
motes the primordial to primary follicle transition. Mol Cell Endocrinol 192: 37–43,
2002.
177. Khadzhieva MB, Lutcenko NN, Volodin IV, Morozova KV, Salnikova LE. Association
of oxidative stress-related genes with idiopathic recurrent miscarriage. Free Radical
Res 48: 534 –541, 2014.
178. Khan S, Beigh S, Chaudhari BP, Sharma S, Aliul Hasan Abdi S, Ahmad S, Ahmad F,
Parvez S, Raisuddin S. Mitochondrial dysfunction induced by Bisphenol A is a factor of
its hepatotoxicity in rats. Environ Toxicol toxicology 2015 doi: 10.1002/tox.22193.
179. Kline J, Levin B, Kinney A, Stein Z, Susser M, Warburton D. Cigarette smoking and
spontaneous abortion of known karyotype. Precise data but uncertain inferences. Am
J Epidemiol 141: 417– 427, 1995.
180. Knight PG, Satchell L, Glister C. Intra-ovarian roles of activins and inhibins. Mol Cell
Endocrinol 359: 53– 65, 2012.
181. Knoll M, Talbot P. Cigarette smoke inhibits oocyte cumulus complex pick-up by the
oviduct in vitro independent of ciliary beat frequency. Reprod Toxicol 12: 57– 68, 1998.
182. Koch HM, Calafat AM. Human body burdens of chemicals used in plastic manufacture.
Philos Trans R Soc Lond B Biol Sci 364: 2063–2078, 2009.
183. Konstantinidis M, Alfarawati S, Hurd D, Paolucci M, Shovelton J, Fragouli E, Wells D.
Simultaneous assessment of aneuploidy, polymorphisms, and mitochondrial DNA
content in human polar bodies and embryos with the use of a novel microarray
platform. Fertil Steril 102: 1385–1392, 2014.
184. Kristensen SG, Andersen K, Clement CA, Franks S, Hardy K, Andersen CY. Expres-
sion of TGF-beta superfamily growth factors, their receptors, the associated SMADs
and antagonists in five isolated size-matched populations of pre-antral follicles from
normal human ovaries. Mol Hum Reprod 20: 293–308, 2014.
185. Krog M, Prior M, Carlsen E, Loft A, Forman J, Pinborg A, Andersen AN. Fertilization
failure after IVF in 304 couples–a case-control study on predictors and long-term
prognosis. Eur J Obstet Gynecol Reprod Biol 184: 32–37, 2015.
186. Kroon B, Johnson N, Chapman M, Yazdani A, Hart R, Australasian CCEPoTeg. Fi-
broids in infertility– consensus statement from ACCEPT (Australasian CREI Consen-
sus Expert Panel on Trial evidence). AustNZJObstet Gynaecol 51: 289 –295, 2011.
187. Krotz SP, Carson SA, Tomey C, Buster JE. Phthalates and bisphenol do not accumu-
late in human follicular fluid. J Assist Reprod Genet 29: 773–777, 2012.
188. Kurkowiak M, Zietkiewicz E, Witt M. Recent advances in primary ciliary dyskinesia
genetics. J Med Genet 52: 1–9, 2015.
189. Kwik M, Karia S, Boothroyd C. RANZCOG CREI Consensus Statement on treatment
of Ovarian Hyperstimulation Syndrome. AustNZJObstet Gynaecol 55: 413– 419,
2015.
190. Landgren BM, Collins A, Csemiczky G, Burger HG, Baksheev L, Robertson DM.
Menopause transition: Annual changes in serum hormonal patterns over the men-
strual cycle in women during a nine-year period prior to menopause. J Clin Endocrinol
Metab 89: 2763–2769, 2004.
191. Lane M, Gardner DK. Embryo culture medium: which is the best? Best Pract Res Clin
Obstet Gynaecol 21: 83–100, 2007.
192. Larsen U, Vaupel JW. Hutterite fecundability by age and parity: strategies for frailty
modeling of event histories. Demography 30: 81–102, 1993.
193. Lathi RB, Liebert CA, Brookfield KF, Taylor JA, vom Saal FS, Fujimoto VY, Baker VL.
Conjugated bisphenol A in maternal serum in relation to miscarriage risk. Fertil Steril
102: 123–128, 2014.
194. Layman LC. The genetic basis of female reproductive disorders: etiology and clinical
testing. Mol Cell Endocrinol 370: 138 –148, 2013.
195. Leary C, Leese HJ, Sturmey RG. Human embryos from overweight and obese women
display phenotypic and metabolic abnormalities. Hum Reprod 30: 122–132, 2015.
196. Lechniak D, Pers-Kamczyc E, Pawlak P. Timing of the first zygotic cleavage as a
marker of developmental potential of mammalian embryos. Reprod Biol 8: 23– 42,
2008.
197. Legro RS, Brzyski RG, Diamond MP, Coutifaris C, Schlaff WD, Casson P, Christman
GM, Huang H, Yan Q, Alvero R, Haisenleder DJ, Barnhart KT, Bates GW, Usadi R,
Lucidi S, Baker V, Trussell JC, Krawetz SA, Snyder P, Ohl D, Santoro N, Eisenberg E,
Zhang H, Network NRM. Letrozole versus clomiphene for infertility in the polycystic
ovary syndrome. N Engl J Med 371: 119 –129, 2014.
198. Legro RS, Dodson WC, Kris-Etherton PM, Kunselman AR, Stetter CM, Williams NI,
Gnatuk CL, Estes SJ, Fleming J, Allison KC, Sarwer DB, Coutifaris C, Dokras A.
Randomized controlled trial of preconception interventions in infertile women with
polycystic ovary syndrome. J Clin Endocrinol Metab 100: 4048 – 4058, 2015.
199. Lerchbaum E, Rabe T. Vitamin D and female fertility. Curr Opin Obstet Gynecol 26:
145–150, 2014.
ROGER J. HART
904 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
200. Lessey BA, Young SL. Integrins and other cell adhesion molecules in endometrium and
endometriosis. Semin Reprod Endocrinol 15: 291–299, 1997.
201. Liu Y, Li GP, Sessions BR, Rickords LF, White KL, Bunch TD. Nicotine induces
multinuclear formation and causes aberrant embryonic development in bovine. Mol
Reprod Dev 75: 801– 809, 2008.
202. Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T, Moley KH.
High fat diet induced developmental defects in the mouse: oocyte meiotic aneuploidy
and fetal growth retardation/brain defects. PLoS One 7: e49217, 2012.
203. Lyons RA, Djahanbakhch O, Saridogan E, Naftalin AA, Mahmood T, Weekes A, Che-
noy R. Peritoneal fluid, endometriosis, and ciliary beat frequency in the human fallo-
pian tube. Lancet 360: 1221–1222, 2002.
204. Lyons RA, Saridogan E, Djahanbakhch O. The reproductive significance of human
Fallopian tube cilia. Hum Reprod Update 12: 363–372, 2006.
205. Macaldowie A, Lee E, Chambers G. Assisted Reproductive Technology in Australia and
New Zealand 2013. Sydney: National Perinatal Epidemiology and Statistics Unit, Univ.
of New South Wales, 2015.
206. Macaldowie A, Wang Y, Chambers G, Sullivan E. Assisted Reproductive Technology in
Australia and New Zealand 2011. Sydney: Univ. of New South Wales, 2013.
207. Machtinger R, Orvieto R. Bisphenol A, oocyte maturation, implantation, and IVF
outcome: review of animal and human data. Reprod Biomed Online 29: 404 – 410, 2014.
208. MacLennan M, Crichton JH, Playfoot CJ, Adams IR. Oocyte development, meiosis and
aneuploidy. Semin Cell Dev Biol 45: 68 –76, 2015.
209. Magli MC, Gianaroli L, Ferraretti AP, Gordts S, Fredericks V, Crippa A. Paternal
contribution to aneuploidy in preimplantation embryos. Reprod Biomed Online 18:
536 –542, 2009.
210. Manova K, Huang EJ, Angeles M, De Leon V, Sanchez S, Pronovost SM, Besmer P,
Bachvarova RF. The expression pattern of the c-kit ligand in gonads of mice supports
a role for the c-kit receptor in oocyte growth and in proliferation of spermatogonia.
Dev Biol 157: 85–99, 1993.
211. Martinez-Zamora MA, Mattioli L, Parera J, Abad E, Coloma JL, van Babel B, Galceran
MT, Balasch J, Carmona F. Increased levels of dioxin-like substances in adipose tissue
in patients with deep infiltrating endometriosis. Hum Reprod 30: 1059 –1068, 2015.
212. Master JS, Thouas GA, Harvey AJ, Sheedy JR, Hannan NJ, Gardner DK, Wlodek ME.
Low female birth weight and advanced maternal age programme alterations in next-
generation blastocyst development. Reproduction 149: 497–510, 2015.
213. May-Panloup P, Chretien MF, Jacques C, Vasseur C, Malthiery Y, Reynier P. Low
oocyte mitochondrial DNA content in ovarian insufficiency. Hum Reprod 20: 593–597,
2005.
214. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA,
Wass JA, Endocrine S. Diagnosis and treatment of hyperprolactinemia: an Endocrine
Society clinical practice guideline. J Clin Endocrinol Metab 96: 273–288, 2011.
215. Merchant-Larios H, Centeno B. Morphogenesis of the ovary from the sterile W/Wv
mouse. Prog Clin Biol Res 59B: 383–392, 1981.
216. Meseguer M, Herrero J, Tejera A, Hilligsoe KM, Ramsing NB, Remohi J. The use of
morphokinetics as a predictor of embryo implantation. Hum Reprod 26: 2658 –2671,
2011.
217. Milesi MM, Alarcon R, Ramos JG, Munoz-de-Toro M, Luque EH, Varayoud J. Neonatal
exposure to low doses of endosulfan induces implantation failure and disrupts uterine
functional differentiation at the pre-implantation period in rats. Mol Cell Endocrinol
401: 248 –259, 2015.
218. Minge CE, Bennett BD, Norman RJ, Robker RL. Peroxisome proliferator-activated
receptor-gamma agonist rosiglitazone reverses the adverse effects of diet-induced
obesity on oocyte quality. Endocrinology 149: 2646 –2656, 2008.
219. Misra M, Miller KK, Tsai P, Gallagher K, Lin A, Lee N, Herzog DB, Klibanski A.
Elevated peptide YY levels in adolescent girls with anorexia nervosa. J Clin Endocrinol
Metab 91: 1027–1033, 2006.
220. Misso ML, Costello MF, Garrubba M, Wong J, Hart R, Rombauts L, Melder AM,
Norman RJ, Teede HJ. Metformin versus clomiphene citrate for infertility in non-
obese women with polycystic ovary syndrome: a systematic review and meta-analy-
sis. Hum Reprod Update 19: 2–11, 2013.
221. Misso ML, Teede HJ, Hart R, Wong J, Rombauts L, Melder AM, Norman RJ, Costello
MF. Status of clomiphene citrate and metformin for infertility in PCOS. Trends Endo-
crinol Metab 23: 533–543, 2012.
222. Misso ML, Wong JL, Teede HJ, Hart R, Rombauts L, Melder AM, Norman RJ, Costello
MF. Aromatase inhibitors for PCOS: a systematic review and meta-analysis. Hum
Reprod Update 18: 301–312, 2012.
223. Mitchell C, Prabhu M. Pelvic inflammatory disease: current concepts in pathogenesis,
diagnosis and treatment. Infect Dis Clinic N Am 27: 793– 809, 2013.
224. Mitchell JA, Hammer RE. Effects of nicotine on oviducal blood flow and embryo
development in the rat. J Reprod Fertil 74: 71–76, 1985.
225. Moran L, Teede H. Metabolic features of the reproductive phenotypes of polycystic
ovary syndrome. Hum Reprod Update 15: 477– 488, 2009.
226. Moran LJ, Misso ML, Wild RA, Norman RJ. Impaired glucose tolerance, type 2 diabetes
and metabolic syndrome in polycystic ovary syndrome: a systematic review and meta-
analysis. Hum Reprod Update 16: 347–363, 2010.
227. Morris SN, Missmer SA, Cramer DW, Powers RD, McShane PM, Hornstein MD.
Effects of lifetime exercise on the outcome of in vitro fertilization. Obstet Gynecol 108:
938 –945, 2006.
228. Murdoch WJ. Proteolytic and cellular death mechanisms in ovulatory ovarian rupture.
Biol Signals Receptors 9: 102–114, 2000.
229. Murri M, Luque-Ramirez M, Insenser M, Ojeda-Ojeda M, Escobar-Morreale HF.
Circulating markers of oxidative stress and polycystic ovary syndrome (PCOS): a
systematic review and meta-analysis. Hum Reprod Update 19: 268 –288, 2013.
230. Muzii L, Di Tucci C, Di Feliciantonio M, Marchetti C, Perniola G, Panici PB. The effect
of surgery for endometrioma on ovarian reserve evaluated by antral follicle count: a
systematic review and meta-analysis. Hum Reprod 29: 2190 –2198, 2014.
231. Naderpoor N, Shorakae S, de Courten B, Misso ML, Moran LJ, Teede HJ. Metformin
and lifestyle modification in polycystic ovary syndrome: systematic review and meta-
analysis. Hum Reprod Update 21: 560 –574, 2015.
232. Navarro VM, Kaiser UB. Metabolic influences on neuroendocrine regulation of re-
production. Curr Opin Endocrinol Diabetes Obesity 20: 335–341, 2013.
233. Nelen WL, Steegers EA, Eskes TK, Blom HJ. Genetic risk factor for unexplained
recurrent early pregnancy loss. Lancet 350: 861, 1997.
234. Neri A, Marcus SL. Effect of nicotine on the motility of the oviducts in the rhesus
monkey: a preliminary report. J Reprod Fertil 31: 91–97, 1972.
235. Newnham JP, Newnham IA, Ball CM, Wright M, Pennell CE, Swain J, Doherty DA.
Treatment of periodontal disease during pregnancy: a randomized controlled trial.
Obstet Gynecol 114: 1239 –1248, 2009.
236. Nikas G, Psychoyos A. Uterine pinopodes in peri-implantation human endometrium.
Clinical relevance. Ann NY Acad Sci 816: 129 –142, 1997.
237. Nilsson EE, Kezele P, Skinner MK. Leukemia inhibitory factor (LIF) promotes the
primordial to primary follicle transition in rat ovaries. Mol Cell Endocrinol 188: 65–73,
2002.
238. No ACO. Exposure to toxic environmental agents. Obstet Gynecol 122: 931–935,
2013.
239. Norman RJ. The power of one and its cost. Med J Aust 195: 564 –565, 2011.
240. Norman RJ, Clark AM. Obesity and reproductive disorders: a review. Reprod Fertil
Dev 10: 55– 63, 1998.
241. Okabe M. The cell biology of mammalian fertilization. Development 140: 4471– 4479,
2013.
242. Oktay K, Bedoschi G. Oocyte cryopreservation for fertility preservation in postpu-
bertal female children at risk for premature ovarian failure due to accelerated follicle
loss in turner syndrome or cancer treatments. J Pediatr Adolescent Gynecol 27: 342–
346, 2014.
243. Oktem O, Urman B. Understanding follicle growth in vivo. Hum Reprod 25: 2944–
2954, 2010.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
905Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
244. Owusu-Edusei K Jr, Bohm MK, Chesson HW, Kent CK. Chlamydia screening and
pelvic inflammatory disease: Insights from exploratory time-series analyses. Am J
Prevent Med 38: 652– 657, 2010.
245. Pal L, Zhang K, Zeitlian G, Santoro N. Characterizing the reproductive hormone
milieu in infertile women with diminished ovarian reserve. Fertil Steril 93: 1074 –1079,
2010.
246. Palioura E, Diamanti-Kandarakis E. Polycystic ovary syndrome (PCOS) and endocrine
disrupting chemicals (EDCs). Rev Endocr Metab Disorders. In press.
247. Palomba S, Falbo A, Chiossi G, Tolino A, Tucci L, La Sala GB, Zullo F. Early trophoblast
invasion and placentation in women with different PCOS phenotypes. Reprod Biomed
Online 29: 370 –381, 2014.
248. Palomba S, Falbo A, La Sala GB. Effects of metformin in women with polycystic ovary
syndrome treated with gonadotrophins for in vitro fertilisation and intracytoplasmic
sperm injection cycles: a systematic review and meta-analysis of randomised con-
trolled trials. BJOG 120: 267–276, 2013.
249. Palomba S, Russo T, Falbo A, Di Cello A, Amendola G, Mazza R, Tolino A, Zullo F,
Tucci L, La Sala GB. Decidual endovascular trophoblast invasion in women with
polycystic ovary syndrome: an experimental case-control study. J Clin Endocrinol
Metab 97: 2441–2449, 2012.
250. Papathanasiou A, Djahanbakhch O, Saridogan E, Lyons RA. The effect of interleukin-6
on ciliary beat frequency in the human fallopian tube. Fertil Steril 90: 391–394, 2008.
251. Parisi MA, Spong CY, Zajicek A, Guttmacher AE. We don’t know what we don’t
study: the case for research on medication effects in pregnancy. Am J Med Genet C Med
Genet 157C: 247–250, 2011.
252. Paszkowski T, Clarke RN, Hornstein MD. Smoking induces oxidative stress inside the
Graafian follicle. Hum Reprod 17: 921–925, 2002.
253. Patel S, Zhou C, Rattan S, Flaws JA. The Effects of Endocrine Disrupting Chemicals on
the Ovary. Biol Reprod 93: 20, 2015.
254. Persani L, Rossetti R, Cacciatore C, Fabre S. Genetic defects of ovarian TGF-beta-like
factors and premature ovarian failure. J Endocrinol Invest 34: 244 –251, 2011.
255. Persani L, Rossetti R, Di Pasquale E, Cacciatore C, Fabre S. The fundamental role of
bone morphogenetic protein 15 in ovarian function and its involvement in female
fertility disorders. Hum Reprod Update 20: 869 – 883, 2014.
256. Petersen GL, Schmidt L, Pinborg A, Kamper-Jorgensen M. The influence of female and
male body mass index on live births after assisted reproductive technology treatment:
a nationwide register-based cohort study. Fertil Steril 99: 1654 –1662, 2013.
257. Pigny P, Merlen E, Robert Y, Cortet-Rudelli C, Decanter C, Jonard S, Dewailly D.
Elevated serum level of anti-mullerian hormone in patients with polycystic ovary
syndrome: relationship to the ovarian follicle excess and to the follicular arrest. J Clin
Endocrinol Metab 88: 5957–5962, 2003.
258. Pineles BL, Park E, Samet JM. Systematic review and meta-analysis of miscarriage and
maternal exposure to tobacco smoke during pregnancy. Am J Epidemiol 179: 807–
823, 2014.
259. Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and repro-
duction: physiological roles and regulatory mechanisms. Physiol Rev 92: 1235–1316,
2012.
260. Plasschaert RN, Bartolomei MS. Genomic imprinting in development, growth, behav-
ior and stem cells. Development 141: 1805–1813, 2014.
261. Pocar P, Fiandanese N, Secchi C, Berrini A, Fischer B, Schmidt JS, Schaedlich K, Rhind
SM, Zhang Z, Borromeo V. Effects of polychlorinated biphenyls in CD-1 mice: repro-
ductive toxicity and intergenerational transmission. Toxicol Sci 126: 213–226, 2012.
262. Pollack AZ, Buck Louis GM, Chen Z, Sun L, Trabert B, Guo Y, Kannan K. Bisphenol A,
benzophenone-type ultraviolet filters, and phthalates in relation to uterine leiomy-
oma. Environ Res 137: 101–107, 2015.
263. Polotsky AJ, Allshouse AA, Casson PR, Coutifaris C, Diamond MP, Christman GM,
Schlaff WD, Alvero R, Trussell JC, Krawetz SA, Santoro N, Eisenberg E, Zhang H,
Legro RS. Impact of male and female weight, smoking, and intercourse frequency on
live birth in women with polycystic ovary syndrome. J Clin Endocrinol Metab 100:
2405–2412, 2015.
264. Potdar N, Gelbaya TA, Konje JC, Nardo LG. Adjunct low-molecular-weight heparin
to improve live birth rate after recurrent implantation failure: a systematic review and
meta-analysis. Hum Reprod Update 19: 674 – 684, 2013.
265. Potts M, Psychoyos A. Evolution of the ultrastructure of the ovoendometrial connec-
tions under the influence of estrogen in the rat during experimental retardation of
nidation. Comptes Rendus hebdomadaires des seances de l’Academie des sciences Serie D:
Sciences naturelles 264: 370 –373, 1967.
266. Practice Committee of American Society For Reproductive Medicine. Increased ma-
ternal cardiovascular mortality associated with pregnancy in women with Turner
syndrome. Fertil Steril 97: 282–284, 2012.
267. Practice Committee of American Society for Reproductive Medicine. Ovarian hyper-
stimulation syndrome. Fertil Steril 90: S188 –193, 2008.
268. Qiao J, Feng HL. Extra- and intra-ovarian factors in polycystic ovary syndrome: impact
on oocyte maturation and embryo developmental competence. Hum Reprod Update
17: 17–33, 2011.
269. Qin JZ, Pang LH, Li MJ, Fan XJ, Huang RD, Chen HY. Obstetric complications in
women with polycystic ovary syndrome: a systematic review and meta-analysis. Re-
prod Biol Endocrinol 11: 56, 2013.
270. Radin RG, Hatch EE, Rothman KJ, Mikkelsen EM, Sorensen HT, Riis AH, Wise LA.
Active and passive smoking and fecundability in Danish pregnancy planners. Fertil Steril
102: 183–191, 2014.
271. Ramesh A, Archibong AE, Niaz MS. Ovarian susceptibility to benzo[a]pyrene: tissue
burden of metabolites and DNA adducts in F-344 rats. J Toxicol Environ Health A 73:
1611–1625, 2010.
272. Ramlau-Hansen CH, Thulstrup AM, Nohr EA, Bonde JP, Sorensen TI, Olsen J. Sub-
fecundity in overweight and obese couples. Hum Reprod 22: 1634 –1637, 2007.
273. Rana P, Kazmi I, Singh R, Afzal M, Al-Abbasi FA, Aseeri A, Singh R, Khan R, Anwar F.
Ectopic pregnancy: a review. Arch Gynecol Obstet 288: 747–757, 2013.
274. Regan L, Rai R. Thrombophilia and pregnancy loss. J Reprod Immunol 55: 163–180,
2002.
275. Retamales I, Carrasco I, Troncoso JL, Las Heras J, Devoto L, Vega M. Morpho-
functional study of human luteal cell subpopulations. Hum Reprod 9: 591–596, 1994.
276. Reynier P, May-Panloup P, Chretien MF, Morgan CJ, Jean M, Savagner F, Barriere P,
Malthiery Y. Mitochondrial DNA content affects the fertilizability of human oocytes.
Mol Hum Reprod 7: 425– 429, 2001.
277. Rice S, Elia A, Jawad Z, Pellatt L, Mason HD. Metformin inhibits follicle-stimulating
hormone (FSH) action in human granulosa cells: relevance to polycystic ovary syn-
drome. J Clin Endocrinol Metab 98: E1491–1500, 2013.
278. Ristovska G, Laszlo HE, Hansell AL. Reproductive outcomes associated with noise
exposure: a systematic review of the literature. Int J Environ Res Public Health 11:
7931–7952, 2014.
279. Rittenberg V, Seshadri S, Sunkara SK, Sobaleva S, Oteng-Ntim E, El-Toukhy T. Effect
of body mass index on IVF treatment outcome: an updated systematic review and
meta-analysis. Reprod Biomed Online 23: 421– 439, 2011.
280. Rivera OE, Varayoud J, Rodriguez HA, Munoz-de-Toro M, Luque EH. Neonatal ex-
posure to bisphenol A or diethylstilbestrol alters the ovarian follicular dynamics in the
lamb. Reprod Toxicol 32: 304 –312, 2011.
281. Robinson JE, Birch RA, Foster DL, Padmanabhan V. Prenatal exposure of the ovine
fetus to androgens sexually differentiates the steroid feedback mechanisms that con-
trol gonadotropin releasing hormone secretion and disrupts ovarian cycles. Arch Sex
Behav 31: 35– 41, 2002.
282. Robker RL, Akison LK, Bennett BD, Thrupp PN, Chura LR, Russell DL, Lane M,
Norman RJ. Obese women exhibit differences in ovarian metabolites, hormones, and
gene expression compared with moderate-weight women. J Clin Endocrinol Metab 94:
1533–1540, 2009.
283. Rodriguez HA, Santambrosio N, Santamaria CG, Munoz-de-Toro M, Luque EH. Neo-
natal exposure to bisphenol A reduces the pool of primordial follicles in the rat ovary.
Reprod Toxicol 30: 550 –557, 2010.
284. Roque M. Freeze-all policy: is it time for that? J Assisted Reprod Genet 32: 171–176,
2015.
ROGER J. HART
906 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
285. Rotterdam EASPCWG. Revised 2003 consensus on diagnostic criteria and long-term
health risks related to polycystic ovary syndrome. Fertil Steril 81: 19 –25, 2004.
286. Rowland AS, Baird DD, Weinberg CR, Shore DL, Shy CM, Wilcox AJ. The effect of
occupational exposure to mercury vapour on the fertility of female dental assistants.
Occup Environ Med 51: 28 –34, 1994.
287. Rubio I, Galan A, Larreategui Z, Ayerdi F, Bellver J, Herrero J, Meseguer M. Clinical
validation of embryo culture and selection by morphokinetic analysis: a randomized,
controlled trial of the EmbryoScope. Fertil Steril 102: 1287–1294, 2014.
288. Rutkowska A, Rachon D. Bisphenol A (BPA) and its potential role in the pathogenesis
of the polycystic ovary syndrome (PCOS). Gynecol Endocrinol 30: 260 –265, 2014.
289. Rzymski P, Rzymski P, Tomczyk K, Niedzielski P, Jakubowski K, Poniedzialek B, Opala
T. Metal status in human endometrium: relation to cigarette smoking and histological
lesions. Environ Res 132: 328 –333, 2014.
290. Rzymski P, Tomczyk K, Rzymski P, Poniedzialek B, Opala T, Wilczak M. Impact of
heavy metals on the female reproductive system. Ann Agricultural Environmental Med
22: 259 –264, 2015.
291. Sallam HN, Garcia-Velasco JA, Dias S, Arici A. Long-term pituitary down-regulation
before in vitro fertilization (IVF) for women with endometriosis. Cochrane Database
Syst Rev CD004635, 2006.
292. Sander VA, Hapon MB, Sicaro L, Lombardi EP, Jahn GA, Motta AB. Alterations of
folliculogenesis in women with polycystic ovary syndrome. J Steroid Biochem Mol Biol
124: 58 – 64, 2011.
293. Santillan I, Lozano I, Illan J, Verdu V, Coca S, Bajo-Arenas JM, Martinez F. Where and
when should natural killer cells be tested in women with repeated implantation
failure? J Reprod Immunol 108: 142–148, 2015.
294. Saradogan E, Djahanbakhch O. Hormonal control of the fallopian tube. In: The Fal-
lopain Tube, edited by Allahbadia GN, Saradogan E, and Djahanbakhch O. Kent:
Anshan, 2009.
295. Savaris RF, Groll JM, Young SL, DeMayo FJ, Jeong JW, Hamilton AE, Giudice LC,
Lessey BA. Progesterone resistance in PCOS endometrium: a microarray analysis in
clomiphene citrate-treated and artificial menstrual cycles. J Clin Endocrinol Metab 96:
1737–1746, 2011.
296. Schatten H, Sun QY. The role of centrosomes in mammalian fertilization and its
significance for ICSI. Mol Hum Reprod 15: 531–538, 2009.
297. Schatten H, Sun QY, Prather R. The impact of mitochondrial function/dysfunction on
IVF and new treatment possibilities for infertility. Reprod Biol Endocrinol 12: 111, 2014.
298. Schieve LA, Tatham L, Peterson HB, Toner J, Jeng G. Spontaneous abortion among
pregnancies conceived using assisted reproductive technology in the United States.
Obstet Gynecol 101: 959 –967, 2003.
299. Schliep KC, Mumford SL, Vladutiu CJ, Ahrens KA, Perkins NJ, Sjaarda LA, Kissell KA,
Prasad A, Wactawski-Wende J, Schisterman EF. Perceived stress, reproductive hor-
mones, and ovulatory function: a prospective cohort study. Epidemiology 26: 177–184,
2015.
300. Schneider LF, Warren MP. Functional hypothalamic amenorrhea is associated with
elevated ghrelin and disordered eating. Fertil Steril 86: 1744 –1749, 2006.
301. Schoenaker DA, Jackson CA, Rowlands JV, Mishra GD. Socioeconomic position, life-
style factors and age at natural menopause: a systematic review and meta-analyses of
studies across six continents. Int J Epidemiol 43: 1542–1562, 2014.
302. Schulte MM, Tsai JH, Moley KH. Obesity and PCOS: the effect of metabolic derange-
ments on endometrial receptivity at the time of implantation. Reprod Sci 22: 6–14,
2015.
303. Schummers L, Hutcheon JA, Bodnar LM, Lieberman E, Himes KP. Risk of adverse
pregnancy outcomes by prepregnancy body mass index: a population-based study to
inform prepregnancy weight loss counseling. Obstet Gynecol 125: 133–143, 2015.
304. Sen A, Kushnir VA, Barad DH, Gleicher N. Endocrine autoimmune diseases and
female infertility. Nature Rev Endocrinol 10: 37–50, 2014.
305. Seshadri S, Sunkara SK. Natural killer cells in female infertility and recurrent miscar-
riage: a systematic review and meta-analysis. Hum Reprod Update 20: 429 – 438, 2014.
306. Shang K, Jia X, Qiao J, Kang J, Guan Y. Endometrial abnormality in women with
polycystic ovary syndrome. Reprod Sci 19: 674 – 683, 2012.
307. Shao R, Zou S, Wang X, Feng Y, Brannstrom M, Stener-Victorin E, Billig H. Revealing
the hidden mechanisms of smoke-induced fallopian tubal implantation. Biol Reprod 86:
131, 2012.
308. Sharara FI, Beatse SN, Leonardi MR, Navot D, Scott RT Jr. Cigarette smoking accel-
erates the development of diminished ovarian reserve as evidenced by the clomi-
phene citrate challenge test. Fertil Steril 62: 257–262, 1994.
309. Shen Y, Ren ML, Feng X, Cai YL, Gao YX, Xu Q. An evidence in vitro for the influence
of bisphenol A on uterine leiomyoma. Eur J Obstet Gynecol Reprod Biol 178: 80– 83,
2014.
310. Shiloh H, Lahav-Baratz S, Koifman M, Ishai D, Bidder D, Weiner-Meganzi Z, Dirnfeld
M. The impact of cigarette smoking on zona pellucida thickness of oocytes and em-
bryos prior to transfer into the uterine cavity. Hum Reprod 19: 157–159, 2004.
311. Shoham Z, Patel A, Jacobs HS. Polycystic ovarian syndrome: safety and effectiveness
of stepwise and low-dose administration of purified follicle-stimulating hormone.
Fertil Steril 55: 1051–1056, 1991.
312. Shub A, Wong C, Jennings B, Swain JR, Newnham JP. Maternal periodontal disease and
perinatal mortality. AustNZJObstet Gynaecol 49: 130 –136, 2009.
313. Shultis WA, Weil EJ, Looker HC, Curtis JM, Shlossman M, Genco RJ, Knowler WC,
Nelson RG. Effect of periodontitis on overt nephropathy and end-stage renal disease
in type 2 diabetes. Diabetes Care 30: 306 –311, 2007.
314. Sim KA, Partridge SR, Sainsbury A. Does weight loss in overweight or obese women
improve fertility treatment outcomes? A systematic review. Obes Rev 15: 839– 850,
2014.
315. Simpson JL, Mills JL, Kim H, Holmes LB, Lee J, Metzger B, Knopp R, Jovanovic-
Peterson L, Aarons J, Conley M. Infectious processes: an infrequent cause of first
trimester spontaneous abortions. Human Reprod 11: 668 – 672, 1996.
316. Sinko I, Morocz M, Zadori J, Kokavszky K, Rasko I. Effect of cigarette smoking on
DNA damage of human cumulus cells analyzed by comet assay. Reprod Toxicol 20:
65–71, 2005.
317. Sivalingam VN, Myers J, Nicholas S, Balen AH, Crosbie EJ. Metformin in reproductive
health, pregnancy and gynaecological cancer: established and emerging indications.
Hum Reprod Update 20: 2853– 867, 2014.
318. Smyth A, Radovic M, Garovic Women VD. Kidney disease, pregnancy. Adv Chronic
Kidney Dis disease 20: 402– 410, 2013.
319. Soares SR, Simon C, Remohi J, Pellicer A. Cigarette smoking affects uterine recep-
tiveness. Hum Reprod 22: 543–547, 2007.
320. Soave I, Caserta D, Wenger JM, Dessole S, Perino A, Marci R. Environment and
Endometriosis: a toxic relationship. Eur Rev Med Pharmacol Sci 19: 1964 –1972, 2015.
321. Sobinoff AP, Beckett EL, Jarnicki AG, Sutherland JM, McCluskey A, Hansbro PM,
McLaughlin EA. Scrambled and fried: cigarette smoke exposure causes antral follicle
destruction and oocyte dysfunction through oxidative stress. Toxicol Appl Pharmacol
271: 156 –167, 2013.
322. Sobinoff AP, Mahony M, Nixon B, Roman SD, McLaughlin EA. Understanding the
Villain: DMBA-induced preantral ovotoxicity involves selective follicular destruction
and primordial follicle activation through PI3K/Akt and mTOR signaling. Toxicol Sci
123: 563–575, 2011.
323. Sobinoff AP, Sutherland JM, McLaughlin EA. Intracellular signalling during female ga-
metogenesis. Mol Hum Reprod 19: 265–278, 2013.
324. Sofo V, Gotte M, Lagana AS, Salmeri FM, Triolo O, Sturlese E, Retto G, Alfa M,
Granese R, Abrao MS. Correlation between dioxin and endometriosis: an epigenetic
route to unravel the pathogenesis of the disease. Arch Gynecol Obstet 292: 973–986,
2015.
325. Song S, Zhang L, Zhang H, Wei W, Jia L. Perinatal BPA exposure induces hyperglyce-
mia, oxidative stress and decreased adiponectin production in later life of male rat
offspring. Int J Environ Res Public Health 11: 3728 –3742, 2014.
326. Souter I, Smith KW, Dimitriadis I, Ehrlich S, Williams PL, Calafat AM, Hauser R. The
association of bisphenol-A urinary concentrations with antral follicle counts and other
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
907Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
measures of ovarian reserve in women undergoing infertility treatments. Reprod Toxi-
col 42: 224 –231, 2013.
327. Steegers-Theunissen RP, Twigt J, Pestinger V, Sinclair KD. The periconceptional
period, reproduction and long-term health of offspring: the importance of one-carbon
metabolism. Hum Reprod Update 19: 640 – 655, 2013.
328. Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, Teede HJ.
Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycae-
mic-hyperinsulaemic clamp. Hum Reprod 28: 777–784, 2013.
329. Stern C, Pertile M, Norris H, Hale L, Baker HW. Chromosome translocations in
couples with in-vitro fertilization implantation failure. Hum Reprod 14: 2097–2101,
1999.
330. Stetten G, Escallon CS, South ST, McMichael JL, Saul DO, Blakemore KJ. Reevaluating
confined placental mosaicism. Am J Med Genet A 131: 232–239, 2004.
331. Stothard KJ, Tennant PW, Bell R, Rankin J. Maternal overweight and obesity and the
risk of congenital anomalies: a systematic review and meta-analysis. JAMA 301: 636 –
650, 2009.
332. Strandell A, Lindhard A, Waldenstrom U, Thorburn J, Janson PO, Hamberger L.
Hydrosalpinx and IVF outcome: a prospective, randomized multicentre trial in Scan-
dinavia on salpingectomy prior to IVF. Hum Reprod 14: 2762–2769, 1999.
333. Sugino N, Suzuki T, Kashida S, Karube A, Takiguchi S, Kato H. Expression of Bcl-2 and
Bax in the human corpus luteum during the menstrual cycle and in early pregnancy:
regulation by human chorionic gonadotropin. J Clin Endocrinol Metab 85: 4379 – 4386,
2000.
334. Sugiura-Ogasawara M, Ozaki Y, Sonta S, Makino T, Suzumori K. Exposure to bisphe-
nol A is associated with recurrent miscarriage. Hum Reprod 20: 2325–2329, 2005.
335. Susiarjo M, Hassold TJ, Freeman E, Hunt PA. Bisphenol A exposure in utero disrupts
early oogenesis in the mouse. PLoS Genet 3: e5, 2007.
336. Sybert VP, McCauley E. Turner’s syndrome. N Engl J Med 351: 1227–1238, 2004.
337. Szekeres-Bartho J, Balasch J. Progestagen therapy for recurrent miscarriage. Hum
Reprod Update 14: 27–35, 2008.
338. Tang H, Hunter T, Hu Y, Zhai SD, Sheng X, Hart RJ. Cabergoline for preventing
ovarian hyperstimulation syndrome. Cochrane Database Syst Rev 2: CD008605, 2012.
339. Taylor TH, Gitlin SA, Patrick JL, Crain JL, Wilson JM, Griffin DK. The origin, mecha-
nisms, incidence and clinical consequences of chromosomal mosaicism in humans.
Hum Reprod Update 20: 571–581, 2014.
340. Teede HJ, Joham AE, Paul E, Moran LJ, Loxton D, Jolley D, Lombard C. Longitudinal
weight gain in women identified with polycystic ovary syndrome: results of an obser-
vational study in young women. Obesity 21: 1526 –1532, 2013.
341. Teede HJ, Misso ML, Deeks AA, Moran LJ, Stuckey BG, Wong JL, Norman RJ, Costello
MF. Assessment and management of polycystic ovary syndrome: summary of an
evidence-based guideline. Med J Aust 195: S65–112, 2011.
342. Teixeira Filho FL, Baracat EC, Lee TH, Suh CS, Matsui M, Chang RJ, Shimasaki S,
Erickson GF. Aberrant expression of growth differentiation factor-9 in oocytes of
women with polycystic ovary syndrome. J Clin Endocrinol Metab 87: 1337–1344, 2002.
343. Tersigni C, Castellani R, de Waure C, Fattorossi A, De Spirito M, Gasbarrini A,
Scambia G, Di Simone N. Celiac disease and reproductive disorders: meta-analysis of
epidemiologic associations and potential pathogenic mechanisms. Hum Reprod Update
20: 582–593, 2014.
344. Teughels W, Dekeyser C, Van Essche M, Quirynen M. One-stage, full-mouth disin-
fection: fiction or reality? Periodontol 2000 50: 39 –51, 2009.
345. Tonetti MS, D’Aiuto F, Nibali L, Donald A, Storry C, Parkar M, Suvan J, Hingorani AD,
Vallance P, Deanfield J. Treatment of periodontitis and endothelial function. N Engl J
Med 356: 911–920, 2007.
346. Tso LO, Costello MF, Albuquerque LE, Andriolo RB, Macedo CR. Metformin treat-
ment before and during IVF or ICSI in women with polycystic ovary syndrome.
Cochrane Database Syst Rev 11: CD006105, 2014.
347. Turner HH. A syndrome of infantilism, congenital webbed neck, and cubitus valgus.
Endocrinology 23: 566 –574, 1938.
348. Ujvari U, Hulchiy M, Calaby A, Nybacka A, Bystrom B, Hirschberg AL. Lifestyle
intervention up-regulates gene and protein levels of molecules involved in insulin
signaling in the endometrium of overweight/obese women with polycystic ovary
syndrome. Hum Reprod 29: 1526 –1535, 2014.
349. Upson K, De Roos AJ, Thompson ML, Sathyanarayana S, Scholes D, Barr DB, Holt VL.
Organochlorine pesticides and risk of endometriosis: findings from a population-
based case-control study. Environ Health Perspect 121: 1319 –1324, 2013.
350. Upson K, Sathyanarayana S, De Roos AJ, Koch HM, Scholes D, Holt VL. A population-
based case-control study of urinary bisphenol A concentrations and risk of endome-
triosis. Hum Reprod 29: 2457–2464, 2014.
351. Upson K, Sathyanarayana S, De Roos AJ, Thompson ML, Scholes D, Dills R, Holt VL.
Phthalates and risk of endometriosis. Environ Res 126: 91–97, 2013.
352. Vagi SJ, Azziz-Baumgartner E, Sjodin A, Calafat AM, Dumesic D, Gonzalez L, Kato K,
Silva MJ, Ye X, Azziz R. Exploring the potential association between brominated
diphenyl ethers, polychlorinated biphenyls, organochlorine pesticides, perfluorinated
compounds, phthalates, and bisphenol A in polycystic ovary syndrome: a case-control
study. BMC Endocr Disorders 14: 86, 2014.
353. Vaiman D. Genetic regulation of recurrent spontaneous abortion in humans. Biomed J
38: 11–24, 2015.
354. Valdes-Socin H, Rubio Almanza M, Tome Fernandez-Ladreda M, Debray FG, Bours
V, Beckers A. Reproduction, smell, and neurodevelopmental disorders: genetic de-
fects in different hypogonadotropic hypogonadal syndromes. Front Endocrinol 5: 109,
2014.
355. Van den Berg IM, Eleveld C, van der Hoeven M, Birnie E, Steegers EA, Galjaard RJ,
Laven JS, van Doorninck JH. Defective deacetylation of histone 4 K12 in human
oocytes is associated with advanced maternal age and chromosome misalignment.
Hum Reprod 26: 1181–1190, 2011.
356. Van den Boogaard E, Vissenberg R, Land JA, van Wely M, van der Post JA, Goddijn M,
Bisschop PH. Significance of (sub)clinical thyroid dysfunction and thyroid autoimmu-
nity before conception and in early pregnancy: a systematic review. Hum Reprod
Update 17: 605– 619, 2011.
357. Van Kasteren YM, Hundscheid RD, Smits AP, Cremers FP, van Zonneveld P, Braat
DD. Familial idiopathic premature ovarian failure: an overrated and underestimated
genetic disease? Hum Reprod 14: 2455–2459, 1999.
358. Vega M, Urrutia L, Iniguez G, Gabler F, Devoto L, Johnson MC. Nitric oxide induces
apoptosis in the human corpus luteum in vitro. Mol Hum Reprod 6: 681– 687, 2000.
359. Vissenberg R, Manders VD, Mastenbroek S, Fliers E, Afink GB, Ris-Stalpers C, Goddijn
M, Bisschop PH. Pathophysiological aspects of thyroid hormone disorders/thyroid
peroxidase autoantibodies and reproduction. Hum Reprod Update 21: 378 –387, 2015.
360. Voet T, Vanneste E, Vermeesch JR. The human cleavage stage embryo is a cradle of
chromosomal rearrangements. Cytogenet Genome Res 133: 160 –168, 2011.
361. Vrsanska S, Nagyova E, Mlynarcikova A, Fickova M, Kolena J. Components of ciga-
rette smoke inhibit expansion of oocyte-cumulus complexes from porcine follicles.
Physiol Res Acad Sci Bohem 52: 383–387, 2003.
362. Walls ML, Hunter T, Ryan JP, Keelan JA, Nathan E, Hart RJ. In vitro maturation as an
alternative to standard in vitro fertilization for patients diagnosed with polycystic
ovaries: a comparative analysis of fresh, frozen and cumulative cycle outcomes. Hum
Reprod 30: 88 –96, 2014.
363. Walters KA, Allan CM, Handelsman DJ. Rodent models for human polycystic ovary
syndrome. Biol Reprod 86: 149, 141-112, 2012.
364. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS.
Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med
351: 987–997, 2004.
365. Wetherill YB, Akingbemi BT, Kanno J, McLachlan JA, Nadal A, Sonnenschein C, Wat-
son CS, Zoeller RT, Belcher SM. In vitro molecular mechanisms of bisphenol A action.
Reprod Toxicol 24: 178 –198, 2007.
366. Weuve J, Hauser R, Calafat AM, Missmer SA, Wise LA. Association of exposure to
phthalates with endometriosis and uterine leiomyomata: findings from NHANES,
1999 –2004. Environ Health Perspect 118: 825– 832, 2010.
ROGER J. HART
908 Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.
367. Wheatcroft NJ, Salt C, Milford-Ward A, Cooke ID, Weetman AP. Identification of
ovarian antibodies by immunofluorescence, enzyme-linked immunosorbent assay or
immunoblotting in premature ovarian failure. Hum Reprod 12: 2617–2622, 1997.
368. Wilcox AJ, Weinberg CR, O’Connor JF, Baird DD, Schlatterer JP, Canfield RE, Arm-
strong EG, Nisula BC. Incidence of early loss of pregnancy. N Engl J Med 319: 189–
194, 1988.
369. Wilding M. Can we define maternal age as a genetic disease? Facts Views Vision in
ObGyn 6: 105–108, 2014.
370. Williams RC, Barnett AH, Claffey N, Davis M, Gadsby R, Kellett M, Lip GY, Thackray
S. The potential impact of periodontal disease on general health: a consensus view.
Curr Med Res Opin 24: 1635–1643, 2008.
371. Wilton L. Preimplantation genetic diagnosis and chromosome analysis of blastomeres
using comparative genomic hybridization. Hum Reprod Update 11: 33– 41, 2005.
372. Wilton L, Voullaire L, Sargeant P, Williamson R, McBain J. Preimplantation aneuploidy
screening using comparative genomic hybridization or fluorescence in situ hybridiza-
tion of embryos from patients with recurrent implantation failure. Fertil Steril 80:
860 – 868, 2003.
373. Wittenberger MD, Hagerman RJ, Sherman SL, McConkie-Rosell A, Welt CK, Rebar
RW, Corrigan EC, Simpson JL, Nelson LM. The FMR1 premutation and reproduction.
Fertil Steril 87: 456 – 465, 2007.
374. Woodruff TJ. Bridging epidemiology and model organisms to increase understanding
of endocrine disrupting chemicals and human health effects. J Steroid Biochem Mol Biol
127: 108 –117, 2011.
375. Woodruff TJ, Zota AR, Schwartz JM. Environmental chemicals in pregnant women in the
United States: NHANES 2003–2004. Environ Health Perspect 119: 878 –885, 2011.
376. Wright DL, Afeiche MC, Ehrlich S, Smith K, Williams PL, Chavarro JE, Batsis M, Toth
TL, Hauser R. Hair mercury concentrations and in vitro fertilization (IVF) outcomes
among women from a fertility clinic. Reprod Toxicol 51: 125–132, 2015.
377. Wyman A, Pinto AB, Sheridan R, Moley KH. One-cell zygote transfer from diabetic to
nondiabetic mouse results in congenital malformations and growth retardation in
offspring. Endocrinology 149: 466 – 469, 2008.
378. Xiao JS, Su CM, Zeng XT. Comparisons of GnRH Antagonist versus GnRH Agonist
Protocol in Supposed Normal Ovarian Responders Undergoing IVF: A Systematic
Review and Meta-Analysis. PloS One 9: e106854, 2014.
379. Xiao S, Diao H, Smith MA, Song X, Ye X. Preimplantation exposure to bisphenol A
(BPA) affects embryo transport, preimplantation embryo development, and uterine
receptivity in mice. Reprod Toxicol 32: 434 – 441, 2011.
380. Yang X, Wu LL, Chura LR, Liang X, Lane M, Norman RJ, Robker RL. Exposure to
lipid-rich follicular fluid is associated with endoplasmic reticulum stress and impaired
oocyte maturation in cumulus-oocyte complexes. Fertil Steril 97: 1438 –1443, 2012.
381. Youssef MA, Van der Veen F, Al-Inany HG, Mochtar MH, Griesinger G, Nagi Mohesen
M, Aboulfoutouh I, van Wely M. Gonadotropin-releasing hormone agonist versus
HCG for oocyte triggering in antagonist-assisted reproductive technology. Cochrane
Database Syst Rev 10: CD008046, 2014.
382. Zawadski JK, Dunaif A. Diagnostic criteria for polycystic ovary syndrome; towards a
rational approach. In: Polycystic Ovary Syndrome. Boston: Blackwell Scientific, 1992, p.
377–384.
383. Zdravkovic T, Genbacev O, Prakobphol A, Cvetkovic M, Schanz A, McMaster M,
Fisher SJ. Nicotine downregulates the L-selectin system that mediates cytotropho-
blast emigration from cell columns and attachment to the uterine wall. Reprod Toxicol
22: 69 –76, 2006.
384. Zenzes MT. Smoking and reproduction: gene damage to human gametes and em-
bryos. Hum Reprod Update 6: 122–131, 2000.
385. Zenzes MT, Wang P, Casper RF. Cigarette smoking may affect meiotic maturation of
human oocytes. Hum Reprod 10: 3213–3217, 1995.
386. Zhang HQ, Zhang XF, Zhang LJ, Chao HH, Pan B, Feng YM, Li L, Sun XF, Shen W.
Fetal exposure to bisphenol A affects the primordial follicle formation by inhibiting
the meiotic progression of oocytes. Mol Biol Reports 39: 5651–5657, 2012.
387. Zhang L, Shiverick KT. Benzo(a)pyrene, but not 2,3,7,8-tetrachlorodibenzo-p-
dioxin, alters cell proliferation and c-myc and growth factor expression in human
placental choriocarcinoma JEG-3 cells. Biochem Biophys Res Commun 231: 117–
120, 1997.
388. Zhao Q, Ma Y, Sun NX, Ye C, Zhang Q, Sun SH, Xu C, Wang F, Li W. Exposure to
bisphenol A at physiological concentrations observed in Chinese children promotes
primordial follicle growth through the PI3K/Akt pathway in an ovarian culture system.
Toxicol In Vitro 28: 1424 –1429, 2014.
389. Zheng YM, Wang Y, Zhao J, Dai YH, Luo XM, Shen ZJ, Chen X, Yuan W, Shen YP.
Association between serum bisphenol-A and recurrent spontaneous abortion: a 1:2
case-control study, China. Zhonghua liu xing bing xue za zhi ⫽Zhonghua liuxingbingxue
zazhi 33: 841– 845, 2012.
390. Zhou W, Liu J, Liao L, Han S, Liu J. Effect of bisphenol A on steroid hormone produc-
tion in rat ovarian theca-interstitial and granulosa cells. Mol Cell Endocrinol 283: 12–18,
2008.
391. Zinn AR, Tonk VS, Chen Z, Flejter WL, Gardner HA, Guerra R, Kushner H, Schwartz
S, Sybert VP, Van Dyke DL, Ross JL. Evidence for a Turner syndrome locus or loci at
Xp11.2-p221 Am J Hum Genet 63: 1757–1766, 1998.
392. Zoeller RT, Brown TR, Doan LL, Gore AC, Skakkebaek NE, Soto AM, Woodruff
TJ, Vom Saal FS. Endocrine-disrupting chemicals and public health protection: a
statement of principles from The Endocrine Society. Endocrinology 153: 4097–
4110, 2012.
PHYSIOLOGICAL ASPECTS OF FEMALE FERTILITY
909Physiol Rev •VOL 96 •JULY 2016 •www.prv.org
Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (176.116.019.134) on February 16, 2018.
Copyright © 2016 American Physiological Society. All rights reserved.