Content uploaded by Anthony Taylor
Author content
All content in this area was uploaded by Anthony Taylor on Jul 25, 2018
Content may be subject to copyright.
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy
Invited critical review
Endocannabinoids and pregnancy
Anthony H. Taylor, Akwasi A. Amoako, Katerina Bambang, Tulay Karasu, Alpha Gebeh, Patricia M.W. Lam,
Timothy H. Marzcylo, Justin C. Konje ⁎
Endocannabinoid Research Group, Reproductive Sciences Section, University of Leicester, Leicester, United Kingdom
abstractarticle info
Article history:
Received 18 December 2009
Received in revised form 9 March 2010
Accepted 9 March 2010
Available online 17 March 2010
Keywords:
Anandamide
Endocannabinoids
Pregnancy
Reproduction
Acylethanolamides such as anandamide (AEA), and monoacylglycerols like 2-arachidonoylglycerol are
endocannabinoids that bind to cannabinoid, vanilloid and peroxisome proliferator-activated receptors. These
compounds, their various receptors, the purported membrane transporter(s), and related enzymes that
synthesize and degrade them are collectively referred to as the “endocannabinoid system (ECS)”. Poorly
defined cellular and molecular mechanisms control the biological actions of the ECS. Over the last decade
evidence has been emerging to suggest that the ECS plays a significant role in various aspects of human
reproduction. In this review, we summarize our current understanding of this role especially the
involvement of AEA and related ECS elements in regulating oogenesis, embryo oviductal transport,
blastocyst implantation, placental development and pregnancy outcomes, and sperm survival, motility,
capacitation and acrosome reaction. Additionally, the possibility that plasma and tissue AEA and other
cannabinoids may represent reliable diagnostic markers of natural and assisted reproduction and pregnancy
outcomes in women will be discussed.
© 2010 Elsevier B.V. All rights reserved.
Contents
1. Introduction . . . . . . . . . . .................................................. 921
2. The endocannabinoid system (ECS).................................................. 922
3. Endocannabinoids and gametogenesis . . . . . ........................................... 923
3.1. Oogenesis . . . . . . . . .................................................. 923
3.2. Spermatogenesis . . . . . .................................................. 924
4. Endocannabinoids, early embryo-blastocyst development and oviductal transport . . . . . . . ..................... 924
4.1. Endocannabinoids and early embryo development . . . . . . . . . . . . . . . . . . ..................... 924
4.2. Endocannabinoids and transport through the Oviduct . . . . . . . . . . . . . . . . . ..................... 924
5. Endocannabinoids and implantation ................................................. 925
6. Endocannabinoids in early pregnancy................................................. 925
7. Endocannabinoids in the maintenance of pregnancy and labour. . . . . . . . . . . . . . . . ..................... 925
8. Feto-placental changes in endocannabinoid levels ........................................... 926
9. Possible “immunocannabinoid”system actions in pregnancy . . . . . . . . . . . . . . . . . ..................... 926
10. Conclusions . . . . . . . . . . .................................................. 927
Conflicts of interest . . . . . . . . . . .................................................. 927
References . . .............................................................. 927
1. Introduction
Endocannabinoids are endogenous ligands that bind to the same
receptors as the most psychoactive agent in marijuana, delta-9-
tetrahydrocannabinol (Δ
9
-THC). The use of marijuana, an illicit drug
in the UK and USA [1,2], appears to be increasing [3,4]. Marijuana use
is associated with infertility in males and females, spontaneous
miscarriages, placental abruption, preterm birth, stillbirths [5–7] and
Clinica Chimica Acta 411 (2010) 921–930
⁎Corresponding author. Endocannabinoid Research Group, Reproductive Sciences
Section, Department of Cancer Studies and Molecular Medicine, University of Leicester,
Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester,
Leicestershire, LE2 7LX UnitedKingdom. Tel.: +44 116 252 5826;fax: + 44 116252 5824.
E-mail address: jck4@le.ac.uk (J.C. Konje).
0009-8981/$ –see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.cca.2010.03.012
Contents lists available at ScienceDirect
Clinica Chimica Acta
journal homepage: www.elsevier.com/locate/clinchim
Author's personal copy
delivery of low birth weight babies [8]. The fact that marijuana has
severe consequences on pregnancy, would suggest that the endoge-
nous ligands of the receptors are involved in the modulation of
reproduction. Recently, comprehensive studies by us and others have
indicated that endocannabinoids (especially the prototypical endo-
cannabinoid, N-acyl arachidonoylethanolamide [anandamide, AEA];
Fig. 1), are actively involved in the process of reproduction and
pregnancy. This review will examine the role of cannabinoids in
pregnancy, from gametogenesis through to birth with special focus
upon the effects of endocannabinoids and the endocannabinoid
system on these processes.
2. The endocannabinoid system (ECS)
The isolation of a specific cannabinoid receptor in the porcine brain
(termed CB
1
), that is responsible for mediating the pharmacological
effects of Δ
9
-THC [9], initiated the search for the endogenous ligands.
The first ligand identified that could imitate the pharmacology of Δ
9
-
THC at the CB
1
receptor was anandamide [10] (Fig. 1). Subsequently,
isolation of a second cannabinoid receptor (CB
2
)associatedwiththe
immune system [11] preceded the identification of a second major
endocannabinoid, 2-arachidonoylglycerol (2-AG) (Fig. 1).Both AEA and
2-AG have affinity for CB
1
and CB
2
[12], although with differentavidities.
More recently, other bioactive endocannabinoids have been identified
including O-arachidonoylethanolamine (virodhamine) [13],N-arachi-
donoyl dopamine [14] and most recently N-acyl taurines, such as N-
arachidonoyl taurine [15]. In addition, several endocannabinoid
congeners have been identified which have greatly reduced affinity
for CB
1
and CB
2
yet elicit endocannabinoid-like activities. These include
oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) (Fig. 1),
which are thought to elicit their endocannabinoid-like activities
through the so-called ‘entourage effect’, by either inhibition of
endocannabinoid catabolism [16], or reducing cellular uptake of AEA
thus leading to increased AEA concentrations [17,18]. Of all the possible
endocannabinoids, 2-AG and AEA are the most frequently studied, with
quantified levels in human tissues for 2-AG being consistently 10–100
times higher than those of AEA [19–21]. AEA, the entourage lipids OEA
and PEA, and to a lesser extent 2-AG have been quantified in human
biological fluids including plasma and serum, seminal plasma, cerebro-
spinal fluid, follicular fluid, oviductal fluid, amniotic fluid, ovarian cyst
fluid, milk and peritoneal fluid, but seem to be absent from urine and
saliva [22–28].
The ligands only comprise part of the endocannabinoid system;
the enzymes concerned with endocannabinoid synthesis and degra-
dation, a putative membrane transport system and the receptors
through which they elicit their physiological effects complete the
various components (Fig. 2). Endocannabinoids are synthesised on
demand from membrane phospholipids precursors and are not stored
[29].
In most tissues, the main rate-limiting step in AEA synthesis is the
conversion of N-arachidonoyl phosphatidylethanolamine (NAPE) into
AEA by a specific phospholipase D enzyme (NAPE-PLD) [30,31].
However, the continued synthesis of AEA in NAPE-PLD knockout mice
highlighted alternative pathways for the formation of AEA [32]. These
pathways involve either hydrolysis of NAPE by phospholipase C to
yield phosphoAEA which is dephosphorylated by a protein tyrosine
phosphatase [33] or sequential acyl hydrolysis of NAPE and lysoNAPE
by abhydrolase domain containing 4 (Abh4) to form glycerophospho-
NAPE, which is then hydrolysed to AEA by a phosphodiesterase [34].
The synthesis of other endocannabinoids, especially the monoacyl
glycerol, 2-AG, is less well-defined, but also thought to occur on
demand. For example, 2-AG is produced via diacylglycerol from
arachidonic acid-rich phospholipid precursors by the sequential
actions of phospholipase C and diacylglycerol lipase [35].
Degradation of AEA and 2-AG to free arachidonic acid is primarily
by the serine hydrolases fatty acid amide hydrolase (FAAH) [36] and
monoacylglycerol lipase (MAGL) [37–39], respectively. In addition to
hydrolysis, AEA has been shown to be metabolised by cyclooxygenase
2 to prostamides, ethanolamide derivatives of prostaglandins [40],
and by the cytochrome P450 isozyme CYP2D6 to several epoxide
metabolites [41], which are thought to have their own physiological
actions. Endocannabinoid degradation requires transport of the
endocannabinoids into cells and how these cross the plasma
membrane is still open to debate [42,43]. Current theories include
carrier protein-mediated transport [44], diffusion driven by metab-
olism by FAAH [45], diffusion driven by compartmentalization of
intracellular AEA [46] and endocytosis [47].
Endocannabinoids predominantly exert their effects through bind-
ing to the cannabinoid receptors (CB
1,
CB
2
and GPR55). These seven-
transmembrane G-protein coupled receptors differ significantly in their
distributions. Although CB
1
was initially localized in the nervous system
and CB
2
in the spleen,these receptors are now knownto be more widely
distributed throughoutthe body [48–58]. These receptors seem to signal
ligand occupancy by coupling inhibitory Gα
i/o
proteins which inhibit
adenylate cyclase activity, thus reducing intracellular cAMP concentra-
tions [59]. In addition, agonist binding to CB receptors also leads to
activation of MAP kinases, which may partly explain the extensive
pathophysiological roles ascribed to endocannabinoids such as cell cycle
progression, apoptosis, cell differentiation and oncogenesis [60,61].
Stimulation of CB receptors can also cause rapid, transient elevation of
intracellular Ca
2+
concentrations via phospholipase C, the inhibition of
voltage-gated Ca
2+
channelsand activation of K
+
channels[31].Butthis
may not be the only effects endocannabinoids exert, as they bind to and
activate other types of receptors. For example, the transient receptor
potential vanilloid type 1 (TRPV1), a ligand-gated non-selective cation
channel that can be activated by a number of stimuli including heat and
pH and is involved in the transmission and modulation of pain, also
binds endocannabinoids, especially AEA and N-arachidonoyl dopamine
[62]. Similarly, both endocannabinoids and entourage compounds bind
to and activate the G-protein coupled receptor GPR55, which is
considered a putative cannabinoid receptor [63] because compounds
Fig. 1. Chemical structures of the most psychoactive component of marijuana (Δ
9
-
tetrahydrocannabinol, the major endocannabinoids, and the “entourage compounds”.
922 A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
other than endocannabinoids also bind to this receptor. The so-called,
‘entourage endocannabinoids,’OEA and PEA, also bind to peroxisome
proliferator-activated receptors (PPAR). In particular, they activate the
PPARαisoform to modulate cell differentiation, lipid metabolism and
regulate food intake [64].
3. Endocannabinoids and gametogenesis
3.1. Oogenesis
The recent demonstration of the endocannabinoid system in the
human ovary has attracted a growing interest into the exact role of the
endocannabinoids in mammalian folliculogenesis [65].CB
1
and CB
2
receptors have been localized in oocytes at all stages of maturation
and the AEA synthetic enzyme NAPE-PLD and the hydrolytic enzyme
FAAH have also been localized in the growing secondary and tertiary
follicles and corpora lutea and albicantes [65]. In addition, AEA has
been quantified in human follicular fluid [28] and its concentration
found to be strongly correlated to oocyte maturation and quality in
women undergoing infertility treatment [66]. The exact molecular
mechanism by which endocannabinoids modulate ovarian folliculo-
genesis remains speculative, but it is believed that they act through a
combination of central and peripheral mechanisms related to the
actions of gonadotrophins. Evidence for this comes from the now
dated studies showing that Δ
9
-THC exerted an adverse effect on
oocyte development and ovulation [67] in which Δ
9
-THC was shown
to interrupt the hypothalamic-pituitary-ovarian axis [68–70] and
suppress plasma follicle-stimulating hormone (FSH) and pre-ovula-
tory luteinizing hormone (LH) surge [71–73], causing an ovulation in
humans [74],rats[75], rabbits and rhesus monkeys [76].The
reduction in gonadotrophin production results in poor quality oocytes
being retrieved during in-vitro fertilization [77]. Since it is known that
cAMP accumulation is required for normal follicular development,
maturation and ovulation [78] and Δ
9
-THC acting through the CB
1
receptor normally has an inhibitory effect on cAMP levels, then the
inhibitory effect of Δ
9
-THC on folliculogenesis may be mediated via its
inhibition of adenylate cyclase, thereby reducing tissue concentra-
tions of cAMP, as has recently been shown in cultured rat granulosa
cells [79,80].
Other evidence also suggests that the endocannabinoid system,
which regulates energy balance by modulating appetite, food intake
and glucose metabolism [81–84] could interact with gametogenesis
through nutritional control. Obesity is commonly associated with
menstrual irregularities, chronic oligo-anovulation and infertility [85]
and regular ovulation is restored after simple management strategies
aimed at weight reduction leading to improved natural conception
[86,87]. Simultaneously, insulin resistance is a cardinal feature of
obesity-associated ovulatory dysfunction in the presence or absence
of polycystic ovary syndrome [88] and androgen hypersecretion, as a
result of hyperinsulinaemia, leads to altered ovarian physiology and
ovulatory dysfunction [89]. Activation of CB
1
receptors in pancreatic
beta cells by AEA induces glucose intolerance, insulin hypersecretion
and insulin resistance in rats [90,91], suggesting that activation of the
ECS in the pancreas might explain some forms of female infertility.
The use of CB
1
antagonists (such as Accomplia®; rimonabant) for the
treatment of obesity, and its metabolic consequences, has been shown
to be very effective in reducing body weight or controlling leptin
concentrations [92,93] although with some serious adverse side
effects that have led to its discontinuance [94]. It is therefore clear that
oogenesis could be disrupted through perturbations of the complex
interplay that exists between the endocannabinoids, leptin produc-
tion and obesity. For example, sustained stimulation of CB
1
in the
hypothalamus by AEA or 2-AG has been observed in leptin-deficient
mice, with elevation of hypothalamic levels of AEA and 2-AG which
Fig. 2. Components of the endocannabinoid system: The ligands 2-acylglycerol (2AG) and anandamide (AEA) are show as blue and red spheres respectively. The putative, but not yet
identified endocannabinoid membrane transporter (EMT) is shown as a bi-directional regulator. The enzymes responsible for the synthesis and degradation of AEA (N-acyl
transferase (NAT) and N-arachidonoyl phosphatidylethanolamine phospholipase D (NAPE-PLD) and fatty acid amide hydrolase (FAAH-1/2)) and of 2-AG (Di-acyl glycerol lipase
(DAG) and monoacyl glycerol lipase (MAGL)), convert these two endocannabinoids into arachidonic acid (AA; green spheres) and ethanolamine (silver spheres) or AA and glycerol
(yellow spheres), respectively. Cyclooxygenase 2 (COX-2) converts AEA into prostaglandin ethanolamines. Glycerol is acted upon by phospholipase C to produce diacylglycerols that
are reconverted into 2AG by DAGL. AEA once internalised may interact with the cytoplasmic domain of the transient vanilloid receptor type 1 (TRPV1) channel protein to alter
intracellular calcium levels. To aid clarity the possible signalling pathways of cannabinoid receptor (CBR) signalling have been omitted.
923A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
was reversed after leptin treatment [95]. Since FAAH is regulated in
some tissues by leptin [96–101] then these data are highly suggestive
of regulatory networks between leptin, FAAH and endocannabinoids
existing in the hypothalamus that are responsible for the exquisite
control of folliculogenesis. Nevertheless, this area of research is at an
early stage and further work is required to understand the complex
network of central and peripheral actions of endocannabinoids with
regard to ovarian function.
3.2. Spermatogenesis
Accumulating evidence suggest that endocannabinoids may also
modulate male reproductive function [102]. The first indication for
this comes from studies of chronic marijuana users, where transient
adverse reproductive toxicity occurs and Δ
9
-THC directly inhibits
sperm motility and viability in vitro [103]. In experimental animals,
Δ
9
-THC induces impotence in rats [104], decreases rat testicular
weight [105,106], decreases testosterone production which impairs
spermatogenesis and sperm motility [107,108] and in mice similarly
reduces the production of spermatozoa that tend to have abnormal
morphology [109].
The exact mechanism by which Δ
9
-THC inhibits spermatogenesis
remains speculative but may involve a complex cross-talk between
FSH, LH and cannabinoids [110].Δ
9
-THC inhibits the release of FSH
and LH from the pituitary gland and consequently [111–114] thereby
inhibits spermatogenesis and testosterone production. By contrast, 2-
AG and to a greater extent AEA, mimic Δ
9
-THC by inhibiting FSH, LH
and prolactin release from the pituitary gland [115–120] suggesting a
common mechanism of action in both the hypothalamus and
pituitary. CB
1
and CB
2
receptors have been demonstrated in
gonadotrophs within the hypothalamus [121] whilst the anterior
pituitary gland expresses CB
1
and TRPV1 receptors [115–117]
supporting a direct endocannabinoid effect on gonadotrophin
production in the male, similar to that observed in the female.
There is also some evidence of a direct effect of cannabinoids on
the testis [122–126]. Recent interest in the localization of the
endocannabinoid system within male reproductive tissues and
measurement of endocannabinoids in fluids of several mammalian
species has been the subject of intense interest, as well as provoking
controversy. For example, spermatogenic output depends on a
balance between cell proliferation and apoptosis and although,
functional CB
1
has been identified in rodent Sertoli and Leydig cells,
suggesting that endocannabinoids are produced by the testis and may
play a crucial role in spermatogenesis and testosterone production
[127,128], the predominant cannabinoid receptor found in mamma-
lian Sertoli cells is CB
2
, which has anti-apoptotic effects and counter-
acts the pro-apoptotic effects that occur through the TRPV1 receptor
(also present in the Sertoli cell), thereby protecting the germinal
epithelium and germ cells from apoptosis, promoting meiotic
progression [127] and ensuring increased spermatogenic output
[126,128–131]. The recent demonstration and cloning of testis-
specific functional isoforms of the CB
1
and CB
2
receptor that have
differential ligand binding properties [132,133] may help to resolve
some of these controversies.
4. Endocannabinoids, early embryo-blastocyst development and
oviductal transport
The oviduct is central to the process of human reproduction and
plays a critical role in oocyte fertilization and pre-implantation
embryo development during its passage to the uterus [134]. Events
occurring during oviductal transport must be co-ordinated such that
the blastocyst is ready for implantation when it arrives in the uterine
cavity and the latter is ready to receive the blastocyst, usually 6 days
post-fertilization [134]. These critical events are synchronised and
regulated by several endocrine, paracrine and autocrine factors
[96,135] of which the endocannabinoid system has been identified
as one of the key hormone–cytokine/receptor signalling systems
[79,136–140].
4.1. Endocannabinoids and early embryo development
In the mouse, FAAH is expressed at the 2-cell stage of the early pre-
implantation embryo [136,141,142] while NAPE-PLD, CB
1
,CB
2
and are
expressed at later stages. As development proceeds to the blastocyst
stage FAAH levels are significantly up regulated. In homozygous FAAH
knockout mice, embryos recovered from oviducts on day 3 of
pregnancy showed delayed and asynchronous development com-
pared to wild type mice [142] suggesting that low levels of AEA are
beneficial and high levels detrimental to blastocyst development
[136,141]. Furthermore, in vitro culture of mouse blastocysts with AEA
indicated that 7nM concentrations of AEA allowed blastocyst
hatching, and 28nM concentrations prevented blastocyst hatching
and induced apoptosis [138]. These data suggest that AEA synthesis by
NAPE-PLD and its degradation by FAAH in embryos and within
oviducts has to be well synchronised such that a locally appropriate
“anandamide tone”is created to permit normal embryo development
and oviductal transport [138]. Furthermore, studies with CB receptor
knockout mice suggest that CB receptor signalling is important in
oviductal transport and blastocyst development. For example, on day
4 of pregnancy, only 60–70% of CB
1
knockout, CB
2
knockout and CB
1
/
CB
2
double knockout embryos were blastocysts, whereas most of the
wild type embryos were blastocysts [143]. Data on the effects of
endocannabinoids on human embryo/blastocyst development are,
however, lacking. Nevertheless, the observation that low plasma
levels of AEA and high levels of FAAH in peripheral mononuclear cells
observed during the putative “implantation window”in humans
[66,144,145] suggests that the ECS also plays a similar role in human
embryo development.
4.2. Endocannabinoids and transport through the Oviduct
CB
1
signalling has been shown to play a crucial role in oviductal
transport of embryos. Studies on CB
1
,CB
2
and CB
1
/CB
2
double
knockout mice showed that only those lacking CB
1
protein had a
large number of embryos in the morula or blastocyst stage retained in
their oviducts on day 4 of pregnancy whereas CB2 knockout and wild
type mice had none retained, providing evidence that oviductal
transport is delayed in CB
1
deficient mice [140,143] and that CB
2
is
probably not involved in oviductal transport. This was confirmed by
studies using wild type mice treated with the CB
1
receptor antagonist
(SR141716) with retention of embryos in the oviducts but not when
the CB
2
antagonist (SR144528) was used [140].
The mechanism whereby endocannabinoids might affect normal
transport is currently unclear. Transport of embryos through the
oviduct is aided by a peristaltic wave of oviductal smooth muscle
movement regulated by the sympathetic nervous system [146]. The
demonstration of CB
1
co-localization with adrenergic receptors in
rodent oviduct, suggests that endocannabinoids could affect cate-
cholamine function [140]. Indeed, excess sympathetic stimulation has
been shown to lead to increased smooth muscle contraction and
impaired embryo transport at the isthmus-uterine junction [140]. The
overstimulation of the oviductal smooth muscle with potassium
chloride led to a 33% overflow of radio-labelled noradrenaline into the
oviducts of CB
1
knockout mice, an effect that was significantly higher
compared to that in CB
2
knockout and wild-type oviducts [140],
suggesting that CB
1
signalling negatively regulates neuronal nor-
adrenaline release to directly modulate tubal motility. The authors
suggest that aberrations in the endocannabinoid system could
therefore form a mechanistic basis for ectopic pregnancy, where
there is no obvious tubal damage.
924 A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
In humans, CB
1
has been localized in the Fallopian tube smooth
muscle wall and CB
1
transcript levels were found to be significantly
lower in the Fallopian tubes of patients with ectopic pregnancy
compared to non pregnant controls [147]. Furthermore, that study
examined 2 polymorphisms of the CB
1
gene and found a possible
association between the 1359G/A polymorphism and ectopic preg-
nancy. The small numbers involved make a definitive association
difficult, but if larger studies support a possible genetic predisposition
to ectopic pregnancy due to CB
1
dysfunction, then genetic screening of
women could lead to the identification or directed therapies for
women at risk of this life-threatening condition [147].
5. Endocannabinoids and implantation
It is now acknowledged that the endocannabinoid system is
critical in the process of implantation [66,79,110,139,147–154].
Successful implantation depends on an accurate interaction between
the endometrium and the implanting blastocyst. Firstly, it requires a
receptive uterine environment and secondly, an activated blastocyst,
capable of invasion and subsequent implantation [155–157].
Implantation occurs about six to seven days after fertilization and
in order for the process to begin, the blastocyst needs to emerge from
the zona pellucida (‘blastocyst hatching’) thereby exposing a
trophoblast capable of invasion [152,155], followed by apposition
with the endometrium, adhesion and finally, trophoblast invasion
[155]. Animal studies have established that AEA regulates the
‘window’during which the uterus is receptive [29,79,139,157].As
the blastocyst enters the uterus, it is differentially affected by local
physiological AEA concentrations. At low concentrations, AEA acti-
vates ERK signalling in dormant mouse blastocysts, whereas high
levels of AEA inhibit calcium mobilisation [141]. Furthermore, low
AEA levels promote blastocyst growth. Studies by Paria et al.have
shown that trophoblasts exposed to low levels of endocannabinoids
show accelerated growth and differentiation as well as increased rates
of zona hatching [137,158,159]. These effects are mediated via CB
1
receptors. Trophoblasts also expresses CB
2
receptors whose function
in this tissue is currently unknown [158,160]. At the same time, lower
AEA levels and elevated levels of FAAH occur in the uterus at
implantation sites [159] when compared to the inter-implantation
sites, which have much higher levels of AEA and lower levels of FAAH.
There is indirect evidence that during apposition, the mouse
blastocyst produces an unidentified lipid [161] that induces the
changes in FAAH and NAPE-PLD enzyme expression conducive for
attachment and initial invasion and thus the changes in localized AEA
concentrations at the implantation site [157,162]. Evidence that local
AEA concentrations are critical to implantation comes from studies
demonstrating that embryos exposed to high levels of AEA in the
uterus suffer embryotoxicity, reduced trophoblast proliferation and
implantation failure [158,160,162].
Similarly, women in an in vitro fertilization programme who have
successful implantation, have low serum AEA levels associated with
elevated levels of FAAH in their peripheral lymphocytes at 6 weeks
gestation [163,164]. The process is likely being controlled by the
expression and activity of lymphocytic FAAH. Indeed, it has been
suggested that the decreased activity and expression of FAAH in
peripheral lymphocytes could be used as an early marker for first
trimester miscarriage [164]. These data are mirrored by the studies of
Trabucco et al.who showed that placental tissue from women
following spontaneous miscarriage had very low levels of FAAH and
increased expression of the CB
1
receptor [151]. These findings are
corroborated by observations that higher plasma AEA levels in women
with threatened miscarriage are associated with a subsequent
spontaneous miscarriage [165].
Plasma AEA levels fluctuate during the menstrual cycle
[23,24,144,145]. Recently our group showed that plasma AEA levels
declined during the implantation window in women with a successful
pregnancy after IVF treatment and in women with normal menstrual
cycles [66,144,145]. During this period, FAAH has been shown to have
the highest activity, whereas there is no change in CB
1
binding, or
AMT and NAPE-PLD activity [145]. Interestingly, FAAH and proges-
terone show the same fluctuations during the menstrual cycle [145].
This is confirmed by the finding that progesterone up-regulates the
FAAH gene [166,167], but contradicted by the finding that serum
progesterone was not correlated with plasma AEA levels during the
menstrual cycle or in women with positive implantation in an IVF
treatment programme [144]. There were, however, positive correla-
tions between the levels of 17β-estradiol, FSH and LH, suggesting that
implantation could be regulated by changes in the levels of these
hormones and their influence of AEA levels in the uterus [144].
Research so far indicates that the endocannabinoid system is an
important regulator for implantation and successful pregnancy.
Endocannabinoid signalling is under tight control and any disruption
of this balance may lead to reproductive failure. The delicate
intricacies related to how endocannabinoids regulate implantation
and whether this is a direct influence or via T lymphocyte function
remains to be elucidated.
6. Endocannabinoids in early pregnancy
Although Δ
9
-THC is known to cross the placenta and affect human
pregnancies, it was not known if the effect was via actions on
trophoblast activity. Recently, however, we demonstrated that Δ
9
-
THC acts directly on the human BeWo cytotrophoblast cell line to
inhibit cell growth and the transcription of genes involved in growth
and apoptosis [168], via the relatively unstudied CB
2
receptor [169].
Indeed, using human cell line models of the 1st and 3rd trimester
trophoblast and the pregnant rat model, we have shown that in
human cells AEA inhibits cell proliferation via the CB
2
receptor by the
differential modulation of histone deactylase 3 (HDAC3) expres-
sion [170]. AEA at concentrations in excess of 15 μM stimulate HDAC3
expression in first trimester trophoblast cells and inhibit HDAC3
expression in third trimester trophoblasts. These data support the
observations that Δ
9
-THC has differential effects on the first and third
trimester trophoblast [171,172], and that differential CB receptor
expression in the rat placenta occurs during gestation [173]. Indeed,
treatment of rats with varying doses of AEA in excess of 10 μM induces
trophoblast cell apoptosis via the CB
1
receptor [173]. These data
suggest that there may be subtle differences in the actions of exo- and
endo-cannabinoids in the placenta in the human and rat and that a
complex interplay between AEA concentrations and implantation of
the blastocyst and subsequent placentation is important during early
pregnancy. Indeed, in studies of IVF-ET women, where the introduc-
tion of the embryo can precisely be timed and implantation can be
closely followed [66], plasma AEA levels initially increase, presumably
to aid blastocyst hatching and implantation, but then subsequently
fall, presumably to prevent trophoblast apoptosis and so aid retention
of the developing embryo. It therefore seems that a critical balance in
endocannabinoid levels, which has been shown to be vital to
successful pregnancy, is required for early trophoblast development
and subsequently that of the placenta.
7. Endocannabinoids in the maintenance of pregnancy and labour
The fact that the use of marijuana is associated with preterm
labour, prolonged pregnancy, fetal growth restriction, stillbirth and
placental abruption suggest that the endocannabinoid system is
involved in late pregnancy and labour. We have quantified AEA levels
during pregnancy and found that the levels are low in the first
trimester (similar to the levels measured in the luteal phase of the
menstrual cycle) and are maintained at these low levels until the
beginning of the 3rd trimester when they start to rise. They then rise
by almost 2- to 4-fold with the onset of labour (Table 1;[23,24,174]).
925A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
In the rat, CB1, CB
2
and TRPV1 expression fluctuates in the decidua
in a predictable pattern from a high level on days 10–12 of pregnancy
to a lower level towards the end of pregnancy [173]. Simultaneously,
plasma AEA levels are not significantly altered, but local tissue levels
of AEA and PEA, but not OEA are increased at two points through
pregnancy; on day 10, when the placenta and decidua regress to make
room for the developing embryo and on day 19 when the activation of
the myometrium is initiated in preparation for parturition on day 21
[175]. These observations suggest that local production of endocan-
nabinoids may be associated with the apoptosis-induced regression of
the decidua in the rat and the preparation of parturition events. These
rodent data together with the aforementioned increase of plasma AEA
levels indicated in the 3rd trimester of pregnancy suggest that plasma
AEA levels could be elevated in women with incipient preterm labour,
preterm premature rupture of membranes or cervical insufficiency,
obstetrical problems that are almost impossible to predict. Data
emerging from our laboratory support this suggestion, as women at
high risk of preterm birth, or presenting with abdominal pain
consistent with the Braxton Hicks myometrial contractions indicative
of impending preterm labour, that subsequently went on to deliver
preterm have higher levels of plasma AEA than those that continue to
term [174]. Indeed, those who delivered quicker (less than 6 days)
have even higher plasma AEA levels than those that took longer to
deliver but still delivered prematurely. These data are consistent with
studies performed in the CB
1
knockout mouse, in which delivery
occurs preterm (i.e. 1 day early) and could be prevented by changing
cortisol and progesterone levels, suggesting that the entire endocan-
nabinoid system, (i.e. not only endocannabinoid levels), is involved in
the timing of parturition [176]. The alternative observation that other
endocannabinoids are present in the uterus at this important time
during pregnancy, and the apparent increase in plasma AEA levels in
women in labour, even if they had been induced [177], suggest that
the ‘entourage effect’is in action and that FAAH activity is maximised
at this point and that FAAH is dealing with the other endocannabi-
noids that exist at higher levels than AEA [19–21,28]. This might
explain the apparent discrepancy between increasing lymphocyte
numbers during the latter stages of pregnancy [178] and the increased
levels of plasma AEA during labour. The obvious extension of these
observations is that CB
1
receptor mutations [147] and CB
1
signalling
or FAAH mutations or activities may be important in the timing of
birth and hence an important area of study for perinatologists. In our
opinion, this understudied area of endocannabinoid research needs
urgent investigation.
It is not clear what elevated plasma AEA levels mean in relation to
parturition. Three main tissue-specific events occur during the
parturition process; myometrial activation, cervical ripening and
fetal membrane rupture. The actions of endocannabinoids in
myometrial activation, cervical ripening and fetal membrane are
unclear due to the paucity of data. Studies on oxytocin-stimulated
myometrial muscle strips demonstrated a relaxant effect of both Δ
9
-
THC and AEA through CB
1
[179]. Recently, we demonstrated that AEA
increased the production of phosphorylated ERK in primary human
myometrial cells and a human myometrial cell line, without changes
in intracellular calcium levels [180]. Such changes in the myometrial
cell should result in myometrial relaxation as was suggested by
Dennedy and colleagues [179]. The data suggest that the myometrium
is a possible endocannabinoid target during the latter stages of
pregnancy. The fetal membranes and placenta at term are also obvious
targets as these tissues express the cannabinoid receptors [181] and
contain significant amounts of AEA (Table 1).
8. Feto-placental changes in endocannabinoid levels
Levels of FAAH in both the human placenta and the maternal
circulation increase towards the end of the first trimester of
pregnancy, before declining by the early second trimester [163,182].
Furthermore, high levels of FAAH have been observed in the villous
cytotrophoblast [183]. Its expression in the syncytiotrophoblast
suggest that FAAH in these cells help prevent the transfer of AEA
from maternal blood [182], which, to some degree, has been
suggested as a protective mechanism for the developing fetus [182].
Previous studies have suggested that circulating FAAH and AEA levels
may be critical to the outcome of early pregnancy [163,164]. As stated
earlier, decreased expression and activity of FAAH in peripheral blood
lymphocytes maybe an early marker of spontaneous miscarriage
[163]. The importance of FAAH in early placental development has
been supported by a recent study that demonstrated the expression of
FAAH and CB
2
(rather than CB
1
) receptors in the human first trimester
placenta. This study also provided evidence that the endocannabinoid
regulation within placental tissue is independent of the maternal
immune system [182]. We recently demonstrated the presence of
both CB
1
and CB
2
receptors and FAAH immunoreactive protein in first
trimester trophoblast which was supported by the presence of
transcripts for these genes in the tissue [183]. The apparent
discrepancy between these two studies could be related to the
antibodies used, as has been documented [184]. Nevertheless,
cannabinoid receptors are expressed by placental tissue from the
early first trimester right through to term [181–183]. Interestingly,
levels of AEA fall progressively during pregnancy [23,24], supporting
other evidence that low systemic levels are required for normal
pregnancy progression [163]. Therefore, exposure to the exocanna-
binoid Δ
9
-THC could lead to inappropriate activation of the CB
mediated pathways in the placental trophoblast.
9. Possible “immunocannabinoid”system actions in pregnancy
Although the involvement of the endocannabinoid system in
reproduction may be direct, it may also be through modulation of the
immune system. Despite a great interest in the role of cannabinoids
within the immune system [57,185–189], the possible relationship
between endocannabinoids and immunity, especially in relation to
reproductive events, remains to be clearly determined. For successful
implantation and maintenance of pregnancy, the embryonic/fetal
allograft [190,191] has to be tolerated by the maternal immune
Table 1
Anandamide concentrations in different human biomatrices related to reproduction
and the reproductive tract.
Biomatrix/Tissue AEA (nM)
1
Ref
Seminal Plasma
3
12.1 [28]
Follicular fluid Immature oocyte 0.36–1.96 [65]
Mature oocyte 0.25–2.78 [65]
Oviductal fluid
3
10.7 [28]
Endometrium ?
Myometrium ?
Cervix ?
Vagina ?
Hypothalamus
2
86.3–87.6 [21]
Pituitary
2
?
Plasma (menstrual cycle) Follicular Phase 1.45–1.68 [23,24,143]
Ovulation 1.26–4.04 [143]
Luteal Phase 0.77–0.87 [23,24,143]
Plasma (pregnancy) 1st trimester 0.89–0.91 [23,24]
2nd trimester 0.44–0.91 [23,24]
3rd trimester 0.44–1.14 [23,24]
Term not in labour 0.68–1.27 [23,24]
Term in active labour 2.30–2.50 [23,24]
Placenta
2
0.98–4.54 [215]
Fetal Membranes
2
0.42–2.83 [215]
Amniotic fluid 0.01–0.18 [26,215]
Umbilical artery plasma 0.32–1.53 [26,215]
Umbilical vein plasma 0.38–1.84 [26,215]
Maternal Plasma at term 0.59–1.91 [26,215]
1
AEA = anandamide;
2
in pmol/g wet weight;
3
only a single human study performed;
? = data unknown.
926 A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
system through mechanisms that might involve the endocannabinoid
system [169]. For example, arachidonic acid (AA), a local immuno-
modulatory molecule [192], is released from cultured mouse
macrophage cells by Δ
9
-THC and AEA through a CB
2
-dependent
mechanism and by nitric oxide (without raising AEA levels) indicating
that both CB receptor-dependent and -independent mechanisms are
involved in this possible immunomodulatory pathway [193,194].
Maccarrone et al. [195] also demonstrated that bacterial lipopolysac-
charide down regulates FAAH expression in human peripheral
lymphyocytes, an effect that was not influence by AEA-induced
activation of the cannabinoid receptors, whilst mast cells absorb AEA
in a saturable process that activates intracellular FAAH and AEA
degradation [196]. Additionally, the physiological protection of the
fetus from rejection is believed to be dependent on a type 2 T-helper
(Th2) cell immune response at the fetal–maternal interface [197]. The
initial state of pregnancy is nevertheless characterised by immuno-
suppression after an early inflammatory state during implantation
[198]. Indeed, the demonstration that monocytes [199] and leuko-
cyte/macrophages [200,201] numbers increase towards the end of
pregnancy suggest an inflammation–immunosuppression–inflamma-
tion phenotype during pregnancy [202]. The production of plasma
AEA mimics this inflammation–immunosuppression–inflammation
pattern during implantation, the 2nd and 3rd trimesters of pregnancy
and just before labour [23,24,66]. Such data suggest that immune cells
may take part in regulating the peripheral endocannabinoid system or
local endocannabinoid homeostasis and vice versa.
Many studies have demonstrated that, in the main, endocannabi-
noids suppress the production of inflammatory cytokines in innate
and adaptive immune responses both in animal models and in human
cell cultures [186,203,204]. Immunosuppression is one of the most
important steps involved in early pregnancy [205] and evidence of a
reduction in interleukin-1β, -10, and -12 levels in animal models of
inflammatory disease [206–208] suggests that similar reductions in
the levels of these cytokines in reproductive tissues may occur in
response to local endocannabinoid production, although this is yet to
be directly determined. However, cannabinoids have also been
demonstrated to increase the production of IL-1, -4, -6 and -10
cytokines either in the presence of bacteria [204,207,209] or alone
[210,211], suggesting that in vivo, cannabinoids may either suppress
or enhance the production of pro-inflammatory agents, depending on
either the nature of the pro-inflammatory stimulus or the type of
cannabinoid present. These changes may arise from the predominant
effect of cannabinoids on helper T-cell (Th) biasing [186,203] whereby
Th-1 cell activity is suppressed and Th-2 cell activity is increased
[208,212]. This may change the levels of leukaemia inhibitory factor,
which is known to be essential for successful invasion and migration
during placental development [213,214].
Taken together these data suggest that endocannabinoids have a
key role, not only on the cells and tissues of the fetal–maternal
interface but also on their interaction with the innate and adaptive
immune system. This under-explored area of possible interaction
between the immunocannabinoid system [187] and reproduction is
likely to be a focus of intense research in the near future.
10. Conclusions
The endocannabinoid system (ECS) is involved in various aspects
of human reproduction. It appears to be an important cog in the
complex mechanisms (wheel) of gametogenesis. With respect to
spermatogenesis, 2-AG appears to promote meiotic progression in the
germ cell by activating type-2 cannabinoid receptors [126]. Levels of
AEA also appear to be related to follicular size and quality of the ova
produced during oogenesis. This system is involved in the regulation
of fertilization through actions that result in spermatozoa activation
and thereafter influence hatching of the embryo, blastocyst matura-
tion, oviductal transport, implantation and early pregnancy mainte-
nance. Changes in plasma levels of some of the cannabinoid ligands
(e.g. anandamide) provide evidence for a tight regulation of early
pregnancy by this group of compounds. Changes in pregnancy and
labour also suggest an involvement in labour onset and maintenance.
Although this review has highlighted areas where the evidence is
coherent and strong, so many questions remain unanswered and only
through collaborative and international studies will these be
adequately addressed. Such collaboration may initially focus on the
changes of the cannabinoid receptors, FAAH, NAPE-PLD, and AEA
which have been thoroughly investigated in animals and partly in
humans, to determine the translational potential of the ECS in
establishing and maintaining a healthy pregnancy.
Conflicts of interest
The authors affirm that there are no conflicts of interest.
References
[1] Hutchings DE, Dow-Edwards D. Animal models of opiate, cocaine, and cannabis
use. Clin Perinatol 1991;18:1–21.
[2] Parliamentary Office of Science and Technology UK, Common illegal drugs and
their effects: Cannabis, Ecstacy, Amphetamines and LSD. London: House of
Commons; 1996.
[3] King JC. Substance abuse in pregnancy. A bigger problem than you think. Postgrad
Med 1997;102:135–50.
[4] Knight EM, James H, Edwards CH, et al. Relationships of serum illicit drug
concentrations during pregnancy to maternal nutritional status. J Nutr 1994;124:
973S–80S.
[5] Conner CS. Marijuana and alcohol use in pregnancy. Drug Intell Clin Pharm
1984;18:233–4.
[6] Felder CC, Glass M. Cannabinoid receptors and their endogenous agonists. Annu
Rev Pharmacol 1998;38:179–200.
[7] Hatch EE, Bracken MB. Effect of marijuana use in pregnancy on fetal growth. Am J
Epidemiol 1986;124:986–93.
[8] Zuckerman B, Frank DA, Hingson R, et al. Effects of maternal marijuana and
cocaine use on fetal growth. N Engl J Med 1989;320:762–8.
[9] Devane WA, Dysarz 3rd FA, Johnson MR, Melvin LS, Howlett AC. Determination
and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol
1988;34:605–13.
[10] Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent
that binds to the cannabinoid receptor. Science 1992;258:1946–9.
[11] Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral
receptor for cannabinoids. Nature 1993;365:61–5.
[12] Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-
monoglyceride, present in canine gut, that binds to cannabinoid receptors.
Biochem Pharmacol 1995;50:83–90.
[13] Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel
endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J
Pharmacol Exp Ther 2002;301:1020–4.
[14] Bisogno T, Melck D, Bobrov MY, et al. N-acyl-dopamines: novel synthetic CB(1)
cannabinoid-receptor ligands and inhibitors of anandamide inactivation with
cannabimimetic activity in vitro and in vivo. Biochem J 2000;351:817–24.
[15] Saghatelian A, Trauger SA, Want EJ, Hawkins EG, Siuzdak G, Cravatt BF.
Assignment of endogenous substrates to enzymes by global metabolite profiling.
Biochemistry 2004;43:14332–9.
[16] Katayama K, Ueda N, Kurahashi Y, Suzuki H, Yamamoto S, Kato I. Distribution of
anandamide amidohydrolase in rat tissues with special reference to small
intestine. Biochim Biophys Acta 1997;1347:212–8.
[17] Ben-Shabat S, Fride E, Sheskin T, et al. An entourage effect: inactive endogenous
fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity.
Eur J Pharmacol 1998;353:23–31.
[18] Hillard CJ, Edgemond WS, Jarrahian A, Campbell WB. Accumulati on of N-
arachidonoylethanolamine (anandamide) into cerebellar granule cells occurs via
facilitated diffusion. J Neurochem 1997;69:631–8.
[19] Chen J, Matias I, Dinh T, et al. Finding of endocannabinoids in human eye tissues:
implications for glaucoma. Biochem Biophys Res Commun 2005;330:1062–7.
[20] Ligresti A, Bisogno T, Matias I, et al. Possible endocannabinoid control of
colorectal cancer growth. Gastroenterology 2003;125:677–87.
[21] Palkovits M, Harvey-White J, Liu J, et al. Regional distribution and effects of
postmortal delay on endocannabinoid content of the human brain. Neuroscience
2008;152:1032–9.
[22] De Marchi N, De Petrocellis L, Orlando P, Daniele F, Fezza F, Di Marzo V.
Endocannabinoid signalling in the blood of patients with schizophrenia. Lipids
Health Dis 2003;5.
[23] Habayeb OM, Taylor AH, Evans MD, et al. Plasma levels of the endocannabinoid
anandamide in women —a potential role in pregnancy maintenance and labor? J
Clin Endocrinol Metab 2004;89:5482–7.
[24] Lam PM, Marczylo TH, El-Talatini M, et al. Ultra performance liquid chromatog-
raphy tandem mass spectrometry method for the measurement of anandamide
in human plasma. Anal Biochem 2008;380:195–201.
927A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
[25] Leweke FM, Giuffrida A, Koethe D, et al. Anandamide levels in cerebrospinal fluid
of first-episode schizophrenic patients: impact of cannabis use. Schizophr Res
2007;94:29–36.
[26] Marczylo TH, Lam PM, Nallendran V, Taylor AH, Konje JC. A solid-phase method
for the extraction and measurement of anandamide from multiple human
biomatrices. Anal Biochem 2009;384:106–13.
[27] Schreiber D, Harlfinger S, Nolden BM, et al. Determination of anandamide and
other fatty acyl ethanolamides in human serum by electrospray tandem mass
spectrometry. Anal Biochem 2007;361:162–8.
[28] Schuel H, Burkman LJ, Lippes J, et al. N-Acylethanolamines in human
reproductive fluids. Chem Phys Lipids 2002;121:211–27.
[29] Habayeb OM, Bell SC, Konje JC. Endogenous cannabinoids: metabolism and their
role in reproduction. Life Sci 2002;70:1963–77.
[30] Di Marzo V, Fontana A, Cadas H, et al. Formation and inactivation of endogenous
cannabinoid anandamide in central neurons. Nature 1994;372:686–91.
[31] Sugiura T, Kodaka T, Kondo S, et al. 2-Arachidonoylglycerol, a putative
endogenous cannabinoid receptor ligand, induces rapid, transient elevation of
intracellular free Ca
2+
in neurobla stoma × glioma hybrid NG108-15 ce lls.
Biochem Biophys Res Commun 1996;229:58–64.
[32] Leung D, Saghatelian A, Simon GM, Cravatt BF. Inactivation of N-acyl
phosphatidylethanolamine phospholipase D reveals multiple mechanisms for
the biosynthesis of endocannabinoids. Biochemistry 2006;45:4720–6.
[33] Liu J, Wang L, Harvey-White J, et al. A biosynthetic pathway for anandamide. Proc
Natl Acad Sci USA 2006;103:13345–50.
[34] Simon GM, Cravatt BF. Endocannabinoid biosynthesis proceeding through
glycerophospho-N-acyl ethanolamine and a role for alpha/beta-hydrolase 4 in
this pathway. J Biol Chem 2006;281:26465–72.
[35] Prescott SM, Majerus PW. Characterization of 1, 2-diacylglycerol hydrolysis in
human platelets. Demonstration of an arachidonoyl-monoacylglycerol interme-
diate. J Biol Chem 1983;258:764–9.
[36] Schmid PC, Zuzarte-Augustin ML, Schmid HH. Properties of rat liver N-
acylethanolamine amidohydrolase. J Biol Chem 1985;260:14145–9.
[37] Di Marzo V, Bisogno T, Sugiura T, Melck D, De Petrocellis L. The novel endogenous
cannabinoid 2-arachidonoylglycerol is inactivated by neuronal- and basophil-
like cells: connections with anandamide. Biochem J 1998;331:15–9.
[38] Goparaju SK, Ueda N, Taniguchi K, Yamamoto S. Enzymes of porcine brain
hydrolyzing 2-arac hidonoylglycerol, an endogenous ligand of cannabinoid
receptors. Biochem Pharmacol 1999;57:417–23.
[39] Goparaju SK, Ueda N, Yamaguchi H, Yamamoto S. Anandamide amidohydrolase
reacting with 2-arachidonoylglycerol, another cannabinoid receptor ligand. FEBS
Lett 1998;422:69–73.
[40] Yu M, Ives D, Ramesha CS. Synthesis of prostaglandin E2 ethanolamide from
anandamide by cyclooxygenase-2. J Biol Chem 1997;272:21181–6.
[41] Snider NT, Sikora MJ, Sridar C, Feuerstein TJ, Rae JM, Hollenberg PF. The
endocannabinoid anandamide is a substrate for the human polymorphic
cytochrome P450 2D6. J Pharmacol Exp Ther 2008;327:538–45.
[42] Glaser SA, Abumrad NA, Fatade F, Kaczocha M, Studholme KM, Deutsch DG.
Evidence against the presence of anandamide transporter. Proc Natl Acad Sci USA
2003;100:4269–74.
[43] Lopez-Rodriguez ML, Viso A, Ortega-Gutierrez S, et al. Design, synthesis, and
biological evaluation of new inhibitors of the endocannabinoid uptake:
comparison with effects on fatty acid amidohydrolase. J Med Chem 2003;46:
1512–22.
[44] Moore SA, Nomikos GG, Dickason-Chesterfield AK, et al. Identification of a high-
affinity binding site involved in the transport of endocannabinoids. Proc Natl
Acad Sci USA 2005;102:17852–7.
[45] Day TA, Rakhshan F, Deutsch DG, Barker EL. Role of fatty acid amide hydrolase in
the transport of the endogenous cannabinoid anandamide. Mol Pharmacol
2001;59:1369–75.
[46] Hillard CJ, Jarrahian A. Cellular accumulation of anandamide: consensus and
controversy. Br J Pharmacol 2003;140:802–8.
[47] McFarland MJ, Porter AC, Rakhshan FR, Rawat DS, Gibbs RA, Barker EL. A role for
caveolae/lipid rafts in the uptake and recycling of the endogenous cannabinoid
anandamide. J Biol Chem 2004;279:41991–7.
[48] Akbas F, Gasteyger C, Sjodin A, Astrup A, Larsen TM. A critical review of the
cannabinoid receptor as a drug target for obesity management. Obes Rev
2009;10:58–67.
[49] Biro T, Toth BI, Hasko G, Paus R, Pacher P. The endocannabinoid system of the skin
in health and disease: novel perspectives and therapeutic opportunities. Trends
Pharmacol Sci 2009;30:411–20.
[50] Fioramonti J, Bueno L. Role of cannabinoid receptors in the control of
gastrointestinal motility and perception. Expert Rev Gastroenterol Hepatol
2008;2:385–97.
[51] Idris AI. Role of cannabinoid receptors in bone disorders: Alternatives for
treatment. Drug News Perspect 2008;21:533–40.
[52] Ligresti A, Petrosino S, Di Marzo V. From endocannabinoid profiling to
‘endocannabinoid therapeutics’. Curr Opin Chem Biol 2009;13:321–31.
[53] Lopez-Miranda V, Herradon E, Martin MI. Vasorelaxation caused by cannabi-
noids: mechanisms in different vascular beds. Curr Vasc Pharmacol 2008;6:
335–46.
[54] Maccarrone M. Endocannabinoids and reproductive endocrinology. Curr Opin
Investig Drugs 2009;10:305–10.
[55] Mallat A, Lotersztajn S. Cannabinoid receptors as therapeutic targets in the
management of liver diseases. Drug News Perspect 2008;21:363–8.
[56] Pacher P, Steffens S. The emerging role of the endocanna binoid system in
cardiovascular disease. Semin Immunopathol 2009;31:63–77.
[57] Pandey R, Mousawy K, Nagarkatti M, Nagarkatti P. Endocannabinoids and
immune regulation. Pharmacol Res 2009;60:85–92.
[58] Schneider M. Cannabis use in pregnancy and early life and its consequences:
animal models. Eur Arch Psychiatry Clin Neurosci 2009;259:383–93.
[59] Pertwee RG. Pharmacology of cannabinoid receptor ligands. Curr Med Chem
1999;6:635–64.
[60] Bouaboula M, Poinot-Chazel C, Bourrie B, et al. Activation of mitogen-activated
protein kinases by stimulation of the central cannabinoid receptor CB1. Biochem J
1995;312:637–41.
[61] Chen Z, Gibson TB, Robinson F, et al. MAP kinases. Chem Rev 2001;101:2449–76.
[62] Huang SM, Bisogno T, Trevisani M, et al. An endogenous capsaicin-like substance
with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl
Acad Sci USA 2002;99:8400–5.
[63] Pertwee RG. GPR55: a new member of the cannabinoid receptor clan? Br J
Pharmacol 2007;152:984–6.
[64] O'Sullivan SE. Cannabinoids go nuclear: evidence for activation of peroxisome
proliferator-activated receptors. Br J Pharmacol 2007;152:576–82.
[65] El-Talatini MR, Taylor AH, Elson JC, Brown L, Davidson AC, Konje JC. Localisation
and function of the endocannabinoid system in the human ovary. PLoS One
2009;4:e4579.
[66] El-Talatini MR, Taylor AH, Konje JC. Fluctuation in anandamide levels from
ovulation to early pregnancy in in-vitro fertilization-embryo transfer women,
and its hormonal regulation. Hum Reprod 2009;24:1989–98.
[67] Abel EL. Marihuana and sex: a critical survey. Drug Alcohol Depend 1981;8:1–22.
[68] Asch RH, Smith CG, Siler-Khodr TM, Pauerstein CJ. Effects of delta 9-
tetrahydrocannabinol administration on gonadal steroidogenic activity in vivo.
Fertil Steril 1979;32:576–82.
[69] Ayalon D, Nir I, Cordova T, et al. Acute effect of delta-1-tetrahydrocannabinol on
hypthalamo-pituitary-ovarian axis in rat. Neuroendocrinology 1977;23:31–42.
[70] Smith RG, Besch NF, Besch PK, Smith CG. Inhibition of gonadotropin by delta9-
tetrahydrocannabinol: mediation by steroid receptors? Science 1979;204:325–7.
[71] Adashi EY, Jones PB, Hsueh AJ. Direct antigonadal activity of cannabinoids:
suppression of rat granulosa cell functions. Am J Physiol 1983;244:E177–85.
[72] Mendelson JH, Mello NK, Ellingboe J, Skupny AS, Lex BW, Griffin M. Marihuana
smoking suppresses luteinizing hormone in women. J Pharmacol Exp Ther
1986;237:862–6.
[73] Tyrey L. delta 9-tetrahydrocannabinol: a potent inhibitor of episodic luteinizing
hormone secretion. J Pharmacol Exp Ther 1980;213:306–8.
[74] Mueller BA, Daling JR, Weiss NS, Moore DE. Recreational drug use and the risk of
primary infertility. Epidemiology 1990;1:195–200.
[75] Nir I, Ayalon D, Tsafriri A, Cordova T, Lindner HR. Suppression of the cyclic surge
of luteinizing hormone secretion and of ovulation in the rat by delta-1-
tetrahydrocannabinol. Nature 1973;243:470–1.
[76] Smith CG, Besch NF, Smith RG, Besch PK. Effect of tetrahydrocannabinol on the
hypothalamic-pituitary axis in the ovariectomized rhesus monkey. Fertil Steril
1979;31:335–9.
[77] Klonoff-Cohen HS, Natarajan L, Chen RV. A prospective study of the effects of
female and male marijuana use on in vitro fertilization (IVF) and gamete
intrafallopian transfer (GIFT) outcomes. Am J Obstet Gynecol 2006;194:369–76.
[78] Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination
through the cumulus complex. Hum Reprod Update 2007;13:289–312.
[79] Taylor AH, Ang C, Bell SC, Konje JC. The role of the endocannabinoid system in
gametogenesis, implantation and early pregnancy. Hum Reprod Update 2007;13:
501–13.
[80] Treinen KA, Sneeden JL, Heindel JJ. Specific inhibition of FSH-stimulated cAMP
accumulation by delta 9-tetrahydrocannabinol in cultured rat granulosa cells.
Toxicol Appl Pharmacol 1993;118:53–7.
[81] Bellocchio L, Vicennati V, Cervino C, Pasquali R, Pagotto U. The endocannabinoid
system in the regulation of cardiometabolic risk factors. Am J Cardiol 2007;100:
7P–17P.
[82] Bluher M, Engeli S, Kloting N, et al. Dysregulation of the peripheral and adipose
tissue endocannabinoid system in human abdominal obesity. Diabetes 2006;55:
3053–60.
[83] Cota D, Marsicano G, Lutz B, et al. Endogenous cannabinoid system as a
modulator of food intake. Int J Obes Relat Metab Disord 2003;27:289–301.
[84] Nogueiras R, Diaz-Arteaga A, Lockie SH, et al. The endocannabinoid system: role
in glucose and energy metabolism. Pharmacol Res 2009;60:93–8.
[85] Pasquali R, Patton L, Gambineri A. Obesity and infertility. Curr Opin Endocrinol
Diabetes Obes 2007;14:482–7.
[86] Wilkes S, Murdoch A. Obesity and female fertility: a primary care perspective. J
Fam Plann Reprod Health Care 2009;35:181–5.
[87] Zain MM, Norman RJ. Impact of obesity on female fertility and fertilit y treatment.
Women's Health 2008;4:183–94.
[88] Diamanti-Kandarakis E. Insulin resistance in PCOS. Endocrine 2006;30:13–7.
[89] Pasquali R, Gambineri A. Metabolic effects of obesity on reproduction. Reprod
Biomed Online 2006;12:542–51.
[90] Ahren B. Islet G protein-coupled receptors as potential targets for treatment of
type 2 diabetes. Nat Rev Drug Discov 2009;8:369–85.
[91] Bermudez-Siva FJ, Serrano A, Diaz-Molina FJ, et al. Activation of cannabinoid CB1
receptors induces glucose intolerance in rats. Eur J Pharmacol 2006;531:282–4.
[92] Cota D, Genghini S, Pasquali R, Pagotto U. Antagonizing the cannabinoid receptor
type 1: a dual way to fight obesity. J Endocrinol Invest 2003;26:1041–4.
[93] Di Marzo V, Despres JP. CB1 antagonists for obesity —what lessons have we
learned from rimonabant? Nat Rev Endocrinol 2009;5:633–8.
[94] Jones D. End of the line for cannabinoid receptor 1 as an anti-obesity target? Nat
Rev Drug Discov 2008;7:961–2.
928 A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
[95] Di Marzo V, Goparaju SK, Wang L, et al. Leptin-regulated endocannabinoids are
involved in maintaining food intake. Nature 2001;410:822–5.
[96] Battista N, Pasquariello N, Di Tommaso M, Maccarrone M. Interplay between
endocannabinoids, steroids and cytokines in the control of human reproduction. J
Neuroendocrinol 2008;20(Suppl 1):82–9.
[97] Gasperi V, Fezza F, Spagnuolo P, Pasquariello N, Maccarrone M. Further insights
into the regulation of human FAAH by progesterone and leptin: Implications for
endogenous levels of anandamide and apoptosis of immune and neuronal cells.
NeuroToxicology 2005;26:811–7.
[98] Maccarrone M, Bari M, Di Rienzo M, Finazzi-Agro A, Rossi A. Progesterone
activates fatty acid amide hydrolase (FAAH) promoter in human T lymphocytes
through the transcription factor Ikaros: evidence for a synergistic effect of leptin.
J Biol Chem 2003;278:32726–32.
[99] Maccarrone M, Di Rienzo M, Finazzi-Agro A, Rossi A. Leptin activates the
anandamide hydrolase promoter in human T lymphocytes through STAT3. J Biol
Chem 2003;278:13318–24.
[100] Maccarrone M, Ga speri V, Fe zza F, Finazzi-Agro A, Rossi A. Differential
regulation of fatty acid amide hydrolase promoter in human immune cells
and neuronal cells by leptin and progesterone. Eur J Biochem 2004;271:
4666–76.
[101] Pagano C, Rossato M, Vettor R. Endocannabinoids, adipose tissue and lipid
metabolism. J Neuroendocrinol 2008;20:124–9.
[102] Lewis SE, Maccarrone M. Endocannabinoids, sperm biology and human fertility.
Pharmacol Res 2009;60:126–31.
[103] Whan LB, West MCL, McClure N, Lewis SEM. Effects of delta-9-tetrahydrocan-
nabinol, the primary psychoactive cannabinoid in marijuana, on human sperm
function in vitro. Fertil Steril 2006;85:653–60.
[104] Murphy LL, Gher J, Steger RW, Bartke A. Effects of delta(9)-tetrahydrocannabinol
on copulatory-behaviour and neuroendocrine responses of male-rats to female
conspecifics. Pharmacol Biochem Behav 1994;48:1011–7.
[105] Harclerode J, Nyquist SE, Nazar B, Lowe D. Effects of cannabis on sex hormones
and testicular enzymes of the rodent. Adv Biosci 1978;22–23:395–405.
[106] Schwarz S, Harclerode J, Nyquist SE. Effects of delta9-tetrahydrocannabinol
administration on marker proteins of rat testicular cells. Life Sci 1978;22:7–13.
[107] Huang HFS, Nahas GG, Hembree WC. Morphological changes of spermatozoa
during marihuana induced depression of human spermatogenesis. Fed Proc
1978;37:739.
[108] Kolodny RC, Masters WH, Kolodner RM, Toro G. Depression of plasma testosterone
levels after chronic intensive marihuana use. New Eng J Med 1974;290:872–4.
[109] Zimmerman AM, Zimmerman S, Raj AY. Effects of cannabinoids on spermato-
genesis in mice. Adv Biosci 1978;22–23:407–18.
[110] Maccarrone M. Endocannabinoids: friends and foes of reproduction. Prog Lipid
Res 2009;48:344–54.
[111] Dalterio S, Bartke A, Burstein S. Cannabinoids inhibit testosterone secretion by
mouse testes in vitro. Science 1977;196:1472–3.
[112] Maccarrone M, Wenger T. Effec ts of cannabinoids on hypothalamic and
reproductive function. Handb Exp Pharmacol 2005:555–71.
[113] Wenger T, Croix D, Tramu G, Leonardelli J. Marihuana and reproduction-effects
on puberty and pregnancy in female rats —experimantal results. Annales
D'Endocrinol (Paris) 1992;53:37–43.
[114] Wenger T, Rettori V, Snyder GD, Dalterio S, McCann SM. Effects of delta-9-
tetrahydrocannabinol on the hypothalamic-pituitary control of luteinizing
hormone and follicle-stimulating hormone secretion in adult male rats.
Neuroendocrinology 1987;46:488–93.
[115] Gonzalez S, Bisogno T, Wenger T, et al. Sex steroid influence on cannabinoid CB1
receptor mRNA and endocannabinoid levels in the anterior pituitary gland.
Biochem Biophys Res Commun 2000;270:260–6.
[116] Gonzalez S, Manzanares J, Berrendero F, et al. Identification of endocannabinoids
and cannabinoid CB(1) receptor mRNA in the pituitary gland. Neuroendocrinol-
ogy 1999;70:137–45.
[117] Olah M, Milloh H, Wenger T. The role of endocannabinoids in the regulation of
luteinizing hormone and prolactin release —differences between the effects of
AEA and 2AG. Mol Cell Endocrinol 2008;286:S36–40.
[118] Rodriguez de Fonseca F, Wenger T, Navarro M, Murphy LL. Effects of delta9-THC
on VIP-induced prolactin secretion in anterior pituitary cultures: evidence for the
presence of functional cannabinoid CB1 receptors in pituitary cells. Brain Res
1999;841:114–22.
[119] Wenger T, Fernandez-Ruiz JJ, Ramos JA. Immunocytochemical demonstration of
CB1 cannabinoid receptors in the anterior lobe of the pit uitary gland. J
Neuroendocrinol 1999;11:873–8.
[120] Wenger T, Toth BE, Juaneda C, Leonardelli J, Tramu G. The effects of cannabinoids
on the regulation of reproduction. Life Sci 1999;65:695–701.
[121] Gammon CM, Freeman GM, Xie WH, Petersen SL, Wetsel WC. Regulation of
gonadotropin-releasing hormone secre tion by cannabinoids. Endocrinology
2005;146:4491–9.
[122] Cacciola G, Chioccarelli T, Mackie K, et al. Expression of type-1 cannabinoid
receptor during rat postnatal testicular development: Possible involvement in
adult Leydig cell differentiation. Biol Reprod 2008;79:758–65.
[123] Cacciola G, Chioccarelli T, Ricci G, et al. The endocannabinoid system in
vertebrate male reproduction: a comparative overview. Mol Cell Endocrinol
2008;286:S24–30.
[124] Fasano S, Meccariello R, Cobellis G, et al. The endocannabinoid system: an ancient
signaling involved in the control of male fertility. Ann NY Acad Sci 2009;1163:
112–24.
[125] Ricci G, Cacciola G, Altucci L, et al. Endocannabinoid control of sperm motility:
the role of epididymus. Gen Comp Endocrinol 2007;153:320–2.
[126] Rossi G, Gasperi V, Paro R, Barsacchi D, Cecconi S, Maccarrone M. Follicle-
stimulating hormone activates fatty acid amide hydrolase by protein kinase A
and aromatase-dependent pathways in mouse primary sertoli cells. Endocrinol-
ogy 2007;148:1431–9.
[127] Grimaldi P, Orlando P, Di Siena S, et al. The endocannabinoid system and pivotal
role of the CB2 receptor in mouse spermatogenesis. Proc Natl Acad Sci USA
2009;106:11131–6.
[128] Maccarrone M, Cecconi S, Rossi G, Battista N, Pauselli R, Finazzi-Agro A.
Anandamide activity and degradation are regulated by early postnatal aging
and follicle-stimulating hormone in mouse sertoli cells. Endocrinology
2003;144:20–8.
[129] Maccarrone M, Bari M, Lorenzon T, Bisogno T, Di Marzo V, Finazzi-Agro A.
Anandamide uptake by human endothelial cells and its regulation by nitric
oxide. J Biol Chem 2000;275:13484–92.
[130] Maccarrone M, Finazzi-Agro A. The endocannabinoid system, anandamide and
the regulation of mammalian cell apoptosis. Cell Death Diff 2003;10:946–55.
[131] Maccarrone M, Lorenzon T, Bari M, Melino G, Finazzi-Agro A. Anandamide
induces apoptosis in human cells via vanilloid receptors. Evidence for a
protective role of cannabinoid receptors. J Biol Chem 2000;275:31938–45.
[132] Liu QR, Pan CH, Hishimoto A, et al. Species differences in cannabinoid receptor 2
(CNR2 gene): Identification of novel human and rodent CB2 isoforms, differential
tissue expression and regulation by cannabinoid receptor ligands. Genes, Brain
and Behavior 2009;8:519–30.
[133] Rinaldi-Carmona M, Calandra B, Shire D, et al. Characterization of two cloned
human CB1 cannabinoid receptor isoforms. J Pharmacol Exp Ther 1996;278:
871–8.
[134] Sadler TW, Langman J. Langman's medical embryology. 11th ed. Baltimore: Md.:
Lippincott William & Wilkins; 2009.
[135] Guzeloglu-Kayisli O, Kayisli UA, Taylor HS. The role of growth factors and
cytokines during implantation: endocrine and paracrine interactions. Semin
Reprod Med 2009;27:62–79.
[136] Paria BC, Das SK, Dey SK. The preimplantation mouse embryo is a target for
cannabinoid ligand-receptor signaling. Proc Natl Acad Sci USA 1995;92:9460–4.
[137] Paria BC, Ma W, Andrenyak DM, et al. Effects of cannabinoids on preimplantation
mouse embryo development and implantation are mediated by brain-type
cannabinoid receptors. Biol Reprod 1998;58:1490–5.
[138] Schuel H. Tuning the oviduct to the anandamide tone. J Clin Invest 2006;116:
2087–90.
[139] Sun X, Dey SK. Aspects of endocannabinoid signaling in periimplantation biology.
Mol Cell Endocrinol 2008;286:S3–S11.
[140] Wang H, Guo Y, Wang D, et al. Aberrant cannabinoid signaling impairs oviductal
transport of embryos. Nat Med 2004;10:1074–80.
[141] Wang H, Matsumoto H, Guo Y, Paria BC, Roberts RL, Dey SK. Differential G
protein-coupled cannabinoid receptor signaling by anandamide directs blasto-
cyst activation for implantation. Proc Natl Acad Sci USA 2003;100:14914–9.
[142] Wang H, Xie H, Guo Y, et al. Fatty acid amide hydrolase deficiency limits early
pregnancy events. J Clin Invest 2006;116:2122–31.
[143] Paria BC, Song H, Wang X, et al. Dysregulated cannabinoid signaling disrupts
uterine receptivity for embryo implantation. J Biol Chem 2001;276:20523–8.
[144] El-Talatini MR, Taylor AH, Konje JC. The relationship between plasma levels of
the endocannabinoid, anandamide, sex steroids, and gonadotrophins during the
menstrual cycle. Fertil Steril 2009; In Press, doi:10.1016/j.fertnStert.1008.12.033.
[145] Lazzarin N, Valensise H, Bari M, et al. Fluctuations of fatty acid amide hydrolase
and anandamide levels during the human ovulatory cycle. Gynecol Endocrinol
2004;18:212–8.
[146] Heilman RD, Reo RR, Hahn DW. Changes in the sensitivity of adrenergic receptors
in the oviduct during early gestation in the rabbit. Fertil Steril 1976;27:426–30.
[147] Horne AW, Phillips 3rd JA, Kane N, et al. CB1 expression is attenuated in Fallopian
tube and decidua of women with ectopic pregnancy. PLoS One 2008;3:e3969.
[148] Battista N, Bari M, Rapino C, Trasatti F, D'Agostino A, Maccarrone M. Regulation of
female fertility by the endocannabinoid system. Hum Fertil 2007;10:207–16.
[149] Mitchell MD, Sato TA, Wang A, Keelan JA, Ponnampalam AP, Glass M.
Cannabinoids stimulate prostaglandin production by human gestational tissues
through a tissue- and CB1-receptor-specific mechanism. Am J Physiol Endocrinol
Metab 2008;294:E352–6.
[150] Ribeiro ML, Vercelli CA, Sordelli MS, et al. 17beta-oestradiol and progesterone
regulate anandamide synthesis in the rat uterus. Reprod Biomed Online 2009;18:
209–18.
[151] Trabucco E, Acone G, Marenna A, et al. Endocannabinoid system in first trimester
placenta: low FAAH and high CB1 expression characterize spontaneous
miscarriage. Placenta 2009;30:516–22.
[152] TranguchS, Daikoku T, GuoY, Wang H, Dey SK.Molecular complexityin establishing
uterine receptivity and implantation. Cell Mol Life Sci 2005;62:1964–73.
[153] Vercelli CA, Aisemberg J, Billi S, Wolfson ML, Franchi AM. Endocannabinoid
system and nitric oxide are involved in the deleterious effects of lipopolysac-
charide on murine decidua. Placenta 2009;30:579–84.
[154] Wang H, Xie H, Sun X, et al. Differential regulation of endocannabinoid synthesis
and degradation in the uterus during embryo implantation. Prostaglandins Other
Lipid Mediat 2007;83:62–74.
[155] Norwitz ER, Schust DJ, Fisher SJ. Implantation and the survival of early
pregnancy. New Engl J Med 2001;345:1400–8.
[156] Paria BC, Wang H, Dey SK. Endocannabinoid signaling in synchronizing embryo
development and ut erine receptivity for implantation. Chem Phys Lipids
2002;121:201–10.
[157] Wang H, Dey SK, Maccarrone M. Jekyll and hyde: two faces of cannabinoid
signaling in male and female fertility. Endocr Rev 2006;27:427–48.
929A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930
Author's personal copy
[158] Paria BC, Dey SK. Ligand-receptor signaling with endocannabinoids in preim-
plantation embryo development and implantation. Chem Phys Lipids 2000;108:
211–20.
[159] Schmid PC, Paria BC, Krebsbach RJ, Schmid HH, Dey SK. Changes in anandamide
levels in mouse uterus are associated with uterine receptivity for embryo
implantation. Proc Natl Acad Sci USA 1997;94:4188–92.
[160] Yang ZM, Paria BC, Dey SK. Activation of brain-type cannabinoid receptors
interferes with preimplantation mouse embryo development. Biol Reprod
1996;55:756–61.
[161] Maccarrone M, DeFelici M, Klinger FG, et al. Mouse blastocyts release a lipid
which activates anandamide hydrolase in intact uterus. Mol Hum Reprod
2004;10:215–21.
[162] Wang H, Dey SK. Lipid signaling in embryo implantation. Prostaglandins Other
Lipid Mediat 2005;77:84–102.
[163] Maccarrone M, Bisogno T, Valensise H, et al. Low fatty acid amide hydrolase and
high anandamide levels are associated with failure to achieve an ongoing
pregnancy after IVF and embryo transfer. Mol Hum Reprod 2002;8:188–95.
[164] Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C, Finazzi-Agro A.
Relation between decreased anandamide hydrolase concentrations in human
lymphocytes and miscarriage. Lancet 2000;355:1326–9.
[165] Habayeb OM, Taylor AH, Finney M, Evans MD, Konje JC. Plasma anandamide
concentration and pregnancy outcome in women with threatened miscarriage. J
Am Med Assoc 2008;299:1135–6.
[166] Maccarrone M, Bari M, Di Rienzo M, Finazzi-Agro A, Rossi A. Progesterone
activates fatty acid amide hydrolase (FAAH) promoter in human T lymphocytes
through the transcription factor Ikaros. Evidence for a synergistic effect of leptin.
J Biol Chem 2003;278:32726–32.
[167] Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C, Finazzi-Agro A.
Progesterone up-regulates anandamide hydrolase in human lymphocytes: role
of cytokines and implications for fertility. J Immunol 2001;166:7183–9.
[168] Khare M, Taylor AH, Konje JC, Bell SC. Delta9-tetrahydrocannabinol inhibits
cytotrophoblast cell proliferation and modulates gene transcription. Mol Hum
Reprod 2006;12:321–33.
[169] Maccarrone M. CB2 receptors in reproduction. Br J Pharmacol 2008;153:189–98.
[170] Taylor AH, Abbas MA, Bell SC, Takeda S, Taylor DJ, Konje JC. The inhibition of 1st
and 3rd trimester trophoblast cell line proliferation by the endocannabinoid,
anandamide, is characterised by differential expression of histone deacetylase 3.
Reprod Sci 2009;16:165A.
[171] Fisher SE, Atkinson M, Chang B. Effect of delta-9-tetrahydrocannabinol on the in
vitro uptake of alpha-amino isobutyric acid by term human placental slices.
Pediatr Res 1987;21:104–7.
[172] Kenney SP, Kekuda R, Prasad PD, Leibach FH, Devoe LD, Ganapathy V.
Cannabinoid recep tors and their role in the regulation of the serotonin
transporter in human placenta. Am J Obstet Gynecol 1999;181:491–7.
[173] Fonesca BM, Correia-da-Silva G, Taylor AH, Konje JC, Bell SC, Teixeira NA. Spatio-
temporal expression patterns of anandamide-binding receptors in rat implan-
tation sites: evidence for a role of the endocannabinoid system during the period
of placental development. Am J Obstet Gynecol 2009;7:121.
[174] Nallendran V, Lam PM, McParland PC, Taylor AH, Konje JC. Prediction of preterm
labour among asymptomatic high risk patients using plasma anandamide levels.
Reprod Sci 2009;16:220A.
[175] Fonseca BM, Correia-da-Silva G, Teixeira NA. Anandamide-induced cell death:
dual effects in primary rat decidual cell cultures. Placenta 2009;30:686–92.
[176] Wang H, Xie H, Dey SK. Loss of cannabinoid receptor CB1 induces preterm birth.
PLoS ONE 2008;3:e3320.
[177] Nallendran V, Taylor AH, Lam PWM, Bell SC, Taylor DJ, Konje JC. Plasma
anandamide levels increase during labour induction and appear to delay labour
progression. Reprod Sci 2008;15:109A–10A.
[178] Ong KS, Grieco MH, Goel Z. Increased ‘T lymphocyte’bearing Fc receptors for IgG
in pregnancy. Int Arch Allergy Appl Immunol 1983;70:220–4.
[179] Dennedy MC, Friel AM, Houlihan DD, Broderick VM, Smith T, Morrison JJ.
Cannabinoids and the human uterus during pregnancy. Am J Obstet Gynecol
2004;190:2–9.
[180] Brighton PJ, McDonald J, Taylor AH, et al. Characterization of anandamide-
stimulated cannabinoid receptor signaling in human ULTR myometrial smooth
muscle cells. Mol Endocrinol 2009;23:1415–27.
[181] Park B, Gibbons HM, Mitchell MD, Glass M. Identification of the CB1 cannabinoid
receptor and fatty acid amide hydrolase (FAAH) in the human placenta. Placenta
2003;24:473–8.
[182] Helliwell RJ, Chamley LW, Blake-Palmer K, et al. Characterization of the
endocannabinoid system in early human pregnancy. J Clin Endocrnol Metab
2004;89:5168–74.
[183] Habayeb OM, Taylor AH, Bell SC, Taylor DJ, Konje JC. Expression of the
endocannabinoid system in human first trimester placenta and its role in
trophoblast proliferation. Endocrinology 2008;149:5052–60.
[184] Grimsey NL, Goodfellow CE, Scotter EL, Dowie MJ, Glass M, Graham ES. Specific
detection of CB1 receptors; cannabinoid CB1 receptor antibodies are not all
created equal! J Neurosci Methods 2008;171:78–86.
[185] Baker D, Jackson SJ, Pryce G. Cannabinoid control of neuroinflammation related
to multiple sclerosis. Br J Pharmacol 2007;152:649–54.
[186] Berdyshev EV. Cannabinoid receptors and the regulation of immune response.
Chem Phys Lipids 2000;108:169–90.
[187] Klein TW, Newton C, Larsen K, et al. The cannabinoid system and immune
modulation. J Leukoc Biol 2003;74:486–96.
[188] Parolaro D, Massi P, Rubino T, Monti E. Endocannabinoids in the immune system
and cancer. Prostaglandins Leukot Essent Fatty Acids 2002;66:319–32.
[189] Ullrich O, Merker K, Timm J, Tauber S. Immune control by endocannabinoids —
new mechanisms of neuroprotection? J Neuroimmunol 2007;184:127–35.
[190] Medawar PB. Some immunological and endocrinological problems raised by the
evolution of vivaparity in vertebrates. Symp Soc Exp Biol 1953;7:320–8.
[191] van Nieuwenhoven AL Veenstra, Heineman MJ, Faas MM. The immunology of
successful pregnancy. Hum Reprod Update 2003;9:347–57.
[192] Noverr MC, Erb-Downward JR, Huffnagle GB. Production of eicosanoids and other
oxylipins by pathogenic eukaryotic microbes. Clin Microbiol Rev 2003;16:
517–33.
[193] Kaplan BL, Rockwell CE, Kaminski NE. Evidence for cannabinoid receptor-
dependent and -independent mechanisms of action in leukocytes. J Pharmacol
Exp Ther 2003;306:1077–85.
[194] Pestonjamasp VK, Burstein SH. Anandamide synthesis is induced by arachido-
nate mobilizing agonists in cells of the immune system. Biochim Biophys Acta
1998;1394:249–60.
[195] Maccarrone M, De Petrocellis L, Bari M, et al. Lipopolysaccharide downregulates
fatty acid amide hydrolase expression and increases anandamide levels in
human peripheral lymphocytes. Arch Biochem Biophys 2001;393:321–8.
[196] Maccarrone M, Fiorucci L, Erba F, Bari M, Finazzi-Agro A, Ascoli F. Human mast
cells take up and hydrolyze anandamide under the control of 5-lipoxygenase and
do not express cannabinoid receptors. FEBS Lett 2000;468:176–80.
[197] Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine interactions
in the maternal–fetal relationship: is successful pregnancy a TH2 phenomenon?
Immunol Today 1993;14:353–6.
[198] Chaouat G, Menu E, Delage G, et al. Immuno-endocrine interactions in early
pregnancy. Hum Reprod 1995;10(Suppl 2):55–9.
[199] Luppi P, Haluszczak C, Betters D, Richard CA, Trucco M, DeLoia JA. Monocytes are
progressively activated in the circulation of pregnant women. J Leukoc Biol
2002;72:874–84.
[200] Mor G, Abrahams VM. Potential role of macrophages as immunoregulators of
pregnancy. Reprod Biol Endocrinol 2003;1:119.
[201] Pitkin RM, Witte DL. Platelet and leukocyte counts in pregnancy. J Am Med Assoc
1979;242:2696–8.
[202] Aris A, Lambert F, Bessette P, Moutquin JM. Maternal circulating interferon-
gamma and interleukin-6 as biomarkers of Th1/Th2 immune status throughout
pregnancy. J Obstet Gynaecol Res 2008;34:7–11.
[203] Klein TW, Newton C, Friedman H. Cannabinoid receptors and immunity.
Immunol Today 1998;19:373–81.
[204] Klein TW, Newton CA, Nakachi N, Friedman H. Delta 9-tetrahydrocanna binol
treatment suppresses immunity and early IFN-gamma, IL-12, and IL-12 receptor
beta 2 responses to Legionella pneumophila infection. J Immunol 2000;164:
6461–6.
[205] Norwitz ER, Schust DJ, Fisher SJ. Implantation and the survival of early
pregnancy. New Engl J Med 2001;345:1400–8.
[206] Newton CA, Klein TW, Friedman H. Secondary immunity to Legionella
pneumophila and Th1 activity are suppressed by delta-9-tetrahydrocannabinol
injection. Infect Immun 1994;62:4015–20.
[207] Smith SR, Terminelli C, Denhardt G. Modulation of cytokine responses in
Corynebacterium parvum-primed endotoxemic mice by centrally administered
cannabinoid ligands. Eur J Pharmacol 2001;425:73–83.
[208] Klein TW. Cannabinoid-based drugs as anti-inflammatory therapeutics. Nature
Rev Immunol 2005;5:400–11.
[209] Klein TW, Newton C, Widen R, Friedman H. Delta 9-tetrahydrocannabinol
injection induces cytokine-mediated mortality of mice infected with Legionella
pneumophila. J Pharmacol Exp Ther 1993;267:635–40.
[210] Derocq JM, Jbilo O, Bouaboula M, Segui M, Clere C, Casellas P. Genomic and
functional changes induced by the activation of the peripheral cannabinoid
receptor CB2 in the promyelocytic cells HL-60. Possible involvement of the CB2
receptor in cell differentiation. J Biol Chem 2000;275:15621–8.
[211] Kishimoto S, Kobayashi Y, Oka S, Gokoh M, Waku K, Sugiura T. 2-Arachido-
noylglycerol, an endogenous cannabinoid receptor ligand, induces accelerated
production of chemokines in HL-60 cells. J Biochem (Tokyo) 2004;135:517–24.
[212] Klein TW, Newton C, Larsen K, et al. Cannabinoid receptors and T helper cells. J
Neuroimmunol 2004;147:91–4.
[213] Sharkey AM,King A, Clark DE, et al.Localization of leukemia inhibitoryfactor and its
receptor in human placenta throughout pregnancy. Biol Reprod 1999;60:355–64.
[214] Tapia A, Salamonsen LA, Manuelpillai U, Dimitriadis E. Leukemia inhibitory factor
promotes human first trimester extravillous trophoblast adhesion to extracel-
lular matrix and secretion of tissue inhibitor of metalloproteinases-1 and -2.
Hum Reprod 2008;23:1724–32.
[215] Marczylo TH, Lam PM, Amoako AA, Konje JC. Anandamide levels in human
female reproductive tissues: Solid-phase extraction and measurement by ultra
performance liquid chromatography tandem mass spectrometry. Anal Biochem
2010;400:155–62
930 A.H. Taylor et al. / Clinica Chimica Acta 411 (2010) 921–930