Jekyll And Hyde: Two Faces Of Cannabinoid Signaling In Male And Female Fertility

Jacob Bell

New Member
Haibin Wang,
Sudhansu K. Dey and
Mauro Maccarrone

- Author Affiliations

Departments of Pediatrics, Cell and Developmental Biology, and Pharmacology (H.W., S.K.D.), Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biomedical Sciences (M.M.), University of Teramo, Teramo 64100, Italy; and Mondino-Tor Vergata Neuropharmacology Center (M.M.), University of Rome Tor Vergata, 00133 Rome, Italy

Address all correspondence and requests for reprints to: S. K. Dey, Division of Reproductive and Developmental Biology, Departments of Pediatrics and Cell & Developmental Biology and Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232. E-mail: sk.dey@vanderbilt.edu; or M. Maccarrone, Department of Biomedical Sciences, University of Teramo, 64100 Teramo, Italy. E-mail: mmaccarrone@unite.it


Next Section
Abstract

Mammalian reproduction is a complicated process designed to diversify and strengthen the genetic complement of the offspring and to safeguard regulatory systems at various steps for propagating procreation. An emerging concept in mammalian reproduction is the role of endocannabinoids, a group of endogenously produced lipid mediators, that bind to and activate cannabinoid receptors. Although adverse effects of cannabinoids on fertility have been implicated for years, the mechanisms by which they exert these effects were not clearly understood. With the identification of cannabinoid receptors, endocannabinoid ligands, their key synthetic and hydrolytic pathways, and the generation of mouse models missing cannabinoid receptors, a wealth of information on the significance of cannabinoid/endocannabinoid signaling in spermatogenesis, fertilization, preimplantation embryo development, implantation, and postimplantation embryonic growth has been generated. This review focuses on various aspects of the endocannabinoid system in male and female fertility. It is hoped that a deeper insight would lead to potential clinical applications of the endocannabinoid signaling as a target for correcting infertility and improving reproductive health in humans.

I. Lipid Signaling in Reproduction

II. The Endocannabinoid System

A. Introduction

B. Metabolic routes

C. Molecular targets and signaling pathways

III. Endocannabinoids and Male Fertility

A. Introduction

B. The endocannabinoid system in Sertoli cells

C. The endocannabinoid system in sperm

IV. Endocannabinoids and Female Fertility: Embryo Implantation

A. Introduction

B. Endocannabinoids and preimplantation embryo development

C. Endocannabinoids and oviductal embryo transport

D. Biphasic endocannabinoid sensor in blastocyst implantation

V. Endocannabinoids and Female Fertility: Immunoregulation

A. Th1/Th2 cytokines and fertility

B. The endocannabinoid system in lymphocytes of pregnant women

C. FAAH as a molecular integrator of fertility signals

VI. Endocannabinoids and Clinical Implications

VII. Conclusions and Future Direction

Previous SectionNext Section
I. Lipid Signaling in Reproduction

SEXUAL PROCREATION IS initiated by interactions between a sperm and an egg leading to fertilization (1, 2, 3, 4). The fertilized egg (embryo) undergoes several mitotic cell divisions, ultimately producing the blastocyst with two distinct cell types: the inner cell mass (ICM) and the trophectoderm (5, 6, 7, 8, 9). The nurturing of an offspring within the body and production of a live birth is an enduring task, requiring safeguard regulatory systems at various critical steps. Despite success in producing embryos and initiating embryonic development outside the womb by in vitro fertilization and embryo transfer, there is still a significant knowledge gap in understanding the mechanisms by which a successful pregnancy is achieved. A deeper insight into these processes will help to generate new ideas and concepts for improving fertility and pregnancy-associated health issues in humans. It is difficult to define the hierarchical landscape of the molecular pathways during human pregnancy, because of experimental difficulties and ethical restrictions on research with human embryos. It is hoped that experiments on mice and other animal models that bear certain reproductive similarities with humans combined with those feasible experiments in humans would generate meaningful information to address this critical issue. Over the past several years, molecular and genetic studies have provided evidence that lipid mediators serve as important signaling molecules in coordinating a series of events during early pregnancy.

Under pathophysiological conditions when a cell is activated in response to a stimulus, membrane phospholipids generate numerous lipid-signaling molecules, such as eicosanoids and lysophospholipids. Prostaglandins (PGs), one of the major group of eicosanoid lipid mediators, are generated from arachidonic acid (AA), which is released from membrane phospholipids by phospholipase (PL)A2. AA thus released is transformed by cyclooxygenases (COXs) to PGH, which is then converted to various PGs by specific PG synthases (10). Distinct expression profiles of cytosolic PLA2α (cPLA2α), COX-1, and COX-2 in the ovary and uterus at different stages of pregnancy implicate their differential functions (11, 12, 13, 14). In mice, COX-1-derived PGF2α, as a luteolytic hormone acting on the corpus luteum, is critical for the onset of parturition (15, 16, 17, 18), whereas PGI2 and PGE2 generated by COX-2 are essential for ovulation, fertilization, implantation, and decidualization (11, 13, 19, 20, 21, 22). The role of PG during pregnancy is further illustrated by poor fertility, resulting from deferred implantation, in mice lacking cPLA2α (12). Collectively, these studies in mice establish the importance of lipid signaling through the cPLA2α-COX axis during early pregnancy (23, 24). Observations of COX-2 expression in the periovulatory ovary and uterus during implantation as well as delayed follicular rupture and increased incidence of miscarriages upon pharmacological inhibition of COX further suggest that cPLA2α-COX-derived PG signaling is also operative in human ovulation and pregnancy maintenance (25, 26, 27, 28, 29). Moreover, PGE2 and PGF2α are also thought to play important roles in human parturition by directly acting on the myometrium (30, 31).

Another example of the critical role of lipid signaling in reproduction is the influence of lysophosphatidic acid (LPA), a small lipid molecule belonging to the lysophospholipid group. LPA influences a range of processes through its cell-surface G protein-coupled receptors, LPA1–4 (32). A recent study in mice shows that LPA3 is expressed in the uterine luminal epithelium, with peak expression occurring during the periimplantation period. Its expression overlaps with cPLA2α and COX-2 at the site of blastocyst implantation (33). More importantly, mice missing LPA3 exhibit remarkably similar defects as cPLA2α-deficient mice, such as deferred on-time implantation, retarded fetal development, embryo crowding, and sharing of one placenta by several embryos (12, 33). This study adds another genetic link between lipid signaling and female fertility. Restoration of on-time implantation in LPA3-deficient females by PG supplementation further suggests a fundamental interaction between LPA-LPA3 and cPLA2α-COX-2-PG signaling pathways (33). These findings establish a new concept that a short delay in the attachment of blastocysts to the uterine surface during early pregnancy adversely affects later developmental processes (24, 34, 35).

Increasing evidence points toward the pathophysiological significance of endocannabinoids, another group of bioactive lipid-signaling molecules, in both female and male fertility. These endogenous cannabinoid ligands mimic the action of natural cannabis compound Δ9-tetrahydrocannabinol (THC) in many aspects of central and peripheral functions, including reproductive events (23, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51). THC is thought to account for the majority of the reproductive hazards in marijuana users (Table 1⇓). For example, chronic marijuana use is associated with decreased plasma testosterone levels (52), reduced sperm counts, and impotency in men (52, 53, 54, 55, 56). In women, the chronic use of marijuana is often associated with fetal abnormalities and early pregnancy termination (57, 58, 59, 60, 61, 62, 63, 64, 65, 66). In addition, early studies indicate that embryotoxicity and specific teratological malformation in animal models are correlated with exposure to natural cannabis extracts during pregnancy (67, 68, 69, 70, 71, 72, 73). With the characterization of cannabinoid receptors and their ligands endocannabinoids, the key synthetic and hydrolytic pathways of endocannabinoids, and the generation of gene mutation mouse models, a large body of molecular, genetic, and physiological evidence has been generated that supports key roles of cannabinoid ligand-receptor signaling in spermatogenesis, fertilization, preimplantation embryo development, implantation, and postimplantation embryonic growth. This review focuses on the roles of the endocannabinoid system in male and female fertility. A better understanding of this field will help in developing strategies for potential clinical applications of the endocannabinoid-targeted drugs as the next generation of therapeutics to treat human infertility.
View this table:

In this window
In a new window

TABLE 1.

Comparative adverse impacts of marijuana usage on reproductive events
Previous SectionNext Section
II. The Endocannabinoid System
A. Introduction

Two main molecular targets of THC (Fig. 1⇓), the psychoactive component of Cannabis sativa, are brain-type (CB1) and spleen-type (CB2) cannabinoid receptors (74, 75, 76, 77, 78). Over the last few years, a number of endogenous ligands for CB receptors have been identified; they are collectively called endocannabinoids. They are amides, esters, and ethers of long-chain polyunsaturated fatty acids, isolated from brain and peripheral tissues (79, 80, 81, 82). Two arachidonate derivatives, N-arachidonoylethanolamine (AEA, know as anandamide) and 2-arachidonoylglycerol (2-AG) (Fig. 1⇓), are the endocannabinoids whose biological activity has been best characterized to date (75, 81, 83, 84, 85). Also 2-AG-ether (noladin ether), an ether-type endocannabinoid (Fig. 1⇓), is now included in the cohort of these lipid mediators (86), but its actual physiological relevance is still debatable (87). More recently, O-arachidonoylethanolamine (virodhamine), an “inverted AEA†(Fig. 1⇓), has been identified and shown to behave as a partial agonist and a full agonist for CB1 and CB2, respectively (88). In addition, N-oleoylethanolamine, N-palmitoylethanolamine, and N-stearoylethanolamine are considered endocannabinoid-like molecules and may have an “entourage effectâ€, i.e., they may potentiate the activity of AEA or 2-AG by inhibiting their degradation (82, 89, 90).
Fig. 1.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 1.

Chemical structures of natural and endogenous cannabinoids.
B. Metabolic routes
1. AEA synthesis and degradation.

It is now widely accepted that AEA is produced by a transacylase-phosphodiesterase-mediated synthesis, starting from the precursor N-arachidonoylphosphatidylethanolamine (NArPE). The latter compound originates from the transfer of AA from the sn-1 position of 1,2-sn-di-arachidonoyl- phosphatidylcholine to phosphatidylethanolamine, catalyzed by a calcium-dependent N-acyltransferase [trans-acylase (NAT)] (81, 91). NArPE is then cleaved by a recently characterized N-acylphosphatidylethanolamine (NAPE)-specific phospholipase D (NAPE-PLD), which belongs to the zinc metallo-hydrolase family of the β-lactamase fold (92) and releases AEA and phosphatidic acid. The biological activity of AEA at CB receptors is terminated by its removal from the extracellular space, which occurs through a two-step process: cellular uptake by a high-affinity transporter, followed by intracellular degradation by a fatty acid amide hydrolase (FAAH), N-arachidonoylethanolamine amidohydrolase (EC 3.5.1.4) (93, 94). Several properties of a selective AEA membrane transporter (AMT) have been characterized, although its molecular structure remains unknown (95, 96, 97, 98). In fact, there is controversy regarding the existence of endocannabinoid transporters, and the mechanism by which AEA is taken up by cells is currently being debated (99, 100). The uptake of AEA has the features of a facilitated transport; it is dependent on the concentration, time, and temperature, and independent of external Na+ ions or ATP hydrolysis. However, the molecular and genetic identity of AMT still remains unknown (100, 101). In particular, the relationship between AMT and FAAH is still under debate, because FAAH may not need a transporter to get in contact with AEA (102), and AMT perhaps exports, rather than imports, AEA across the plasma membrane (103). Increasing pharmacological, biochemical, and morphological evidence seems to favor for the existence of an AMT different from FAAH (96, 97). The development of new drugs able to inhibit AMT selectively without affecting FAAH corroborates this speculation (104). However, it has recently been suggested that AEA uptake is driven by nonprotein-mediated diffusion and is regulated by its degree of hydrolysis by FAAH in specific cell types (105). The target of some of the novel transport inhibitors recently developed seems not to be the membrane transporter, but rather FAAH or an uncharacterized intracellular component that delivers AEA to FAAH (105). Once taken up by cells, AEA is a substrate for FAAH that breaks the amide bond and releases AA and ethanolamine (106). Mammalian FAAH is a membrane-bound enzyme with a globular shape. It has 28 α-helices and 11β-sheets, which account for approximately 53 and 13% of the whole protein structure, respectively (102). This enzyme uses an unusual serine-serine-lysine (S241-S217-K142) catalytic triad (106).

Together with AEA and congeners, CB and non-CB receptors, NAT, NAPE-PLD, AMT, and FAAH, along with the enzymes that metabolize 2-AG (see Section II.B.2), constitute the “endocannabinoid system†(80, 82, 107, 108, 109, 110, 111). This system is schematically depicted in Fig. 2⇓.
Fig. 2.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 2.

The endocannabinoid system. The synthesis of AEA from membrane N-arachidonoylphosphatidylethanolamines is catalyzed by the sequential activity of NAT and NAPE-PLD, which releases AEA and phosphatidic acid. AEA is transported in both directions through the cell membrane by a selective AMT and, once taken up, is hydrolyzed by FAAH to ethanolamine (EtNH2) and AA. The main targets of AEA are CB1 and CB2 receptors (CBR), showing an extracellular binding site, and type-1 vanilloid receptors (TRPV1), showing an intracellular binding site. 2-AG is also released from membrane lipids through the activity of DAGL. 2-AG can also be hydrolyzed by FAAH or, more importantly, by MAGL, releasing glycerol and AA. The transport of 2-AG across the cell membrane may be mediated by AMT or a related transporter, and CBR (but not TRPV1) is the target of this endocannabinoid.
2. 2-AG synthesis and degradation.

2-AG acts as a potent and full agonist for both CB1 and CB2 and, like AEA, is not stored in intracellular compartments but is produced on demand. The biosynthetic pathway of 2-AG provides for rapid hydrolysis of inositol phospholipids by a specific PL, PLC; this enzyme generates diacylglycerol (DAG), which is converted to 2-AG by a sn-1-DAG lipase (85). Recently, two sn-1-specific DAG lipases (DAGL) responsible for 2-AG synthesis have been cloned by confronting human genome with Penicillium DAGL sequence (112). These two isoforms (α and β) have molecular masses of 120 and 70 kDa, respectively, with four transmembrane domains and are members of the serine-lipase family with serine and aspartic acid (S443-D495) participating in the catalytic triad. The α isoform is predominant in the adult brain, whereas the β isoform is expressed in developing brain (112).

The pharmacological effects of 2-AG depend on its life span in the extracellular space, which in turn is limited by a rapid transport through the membrane. It is proposed that the 2-AG membrane transporter is the same as AMT (113). In fact, 2-AG accumulation is reduced by an AMT inhibitor, AM404, and indirectly by high concentrations of AA (113). The effect of AM404 is due to the inhibition of AMT, but not due to FAAH activity, because the level of 2-AG remains unaltered in the presence of two strong FAAH inhibitors, URB597 and AM374 (114).

Once accumulated in the cell, 2-AG can be degraded by FAAH (115), but FAAH is not the only enzyme responsible for its metabolism. In fact, mice lacking FAAH are unable to metabolize AEA but can still hydrolyze 2-AG (116). An enzyme responsible for 2-AG degradation, monoacylglycerol lipase (MAGL), has been isolated from the porcine brain (115) and cloned and characterized in rat (117) and human brain (118). Rat brain MAGL is a 33-kDa protein and, unlike FAAH, is localized in the cytosol (117). MAGL and DAGL are members of the endocannabinoid system as shown in Fig. 2⇑.
C. Molecular targets and signaling pathways
1. Cannabinoid receptors.

AEA and 2-AG differentially activate cannabinoid receptors. CB1 are most abundant in the central nervous system but are present in peripheral tissues including the heart, uterus, embryo, testis, liver, small intestine, and peripheral cells like lymphocytes. CB2 are predominantly expressed in astrocytes, spleen, and immune cells (23, 74, 75, 76, 111, 119). Both CB1 and CB2 belong to the rhodopsin family of G protein-coupled seven trans-membrane spanning receptors. They show 44% overall identity, with 68% identity within the transmembrane regions, and are coupled mainly to the Gi/o family of G proteins (74). Signal transduction pathways regulated by CB-coupled Gi/o proteins include the inhibition of adenylyl cyclase (69, 77), regulation of ionic currents (inhibition of voltage-gated L, N, and P/Q-type Ca2+ channels as well as activation of K+ channels) (120, 121, 122, 123, 124, 125), the activation of focal adhesion kinase (126) and MAPK (125, 127, 128). In addition, activation of CB1 or inhibition of CB2 alters nitric oxide (NO) synthase (74, 119). However, a recent report shows that WIN55,212–2, an aminoalkylindole cannabinoid agonist, increases intracellular Ca++ via CB1 coupling to Gq/11 G proteins (129), suggesting diversity of CB1 signaling pathways. Furthermore, there is some evidence that endocannabinoids induce a biological activity via other CB receptors, like a purported CB3 (GPR55) receptor (130, 131, 132). GPR55 is an orphan G protein-coupled receptor that has low sequence homology (10–15%), compared to that of CB1 or CB2, and is expressed in the testis at approximately a 15-fold higher level than in the brain (130, 131). GPR55 does not appear to couple with Gi or Gs proteins, suggesting that it activates different signal transduction pathways from those executed by CB1 and CB2 (130, 131). Among the non-CB receptors, the type-1 vanilloid receptor [now called transient receptor potential vanilloid 1 (TRPV1)] has attracted great interest as a new molecular target of AEA.
2. Vanilloid receptors.

TRPV1 is a six-trans-membrane spanning protein with intracellular N and C terminals and a pore-loop between the fifth and sixth transmembrane helices (133). TRPV1 is a ligand-gated and nonselective cationic channel that is activated by molecules derived from plants, such as capsaicin (the pungent component of “hot†red peppers) and resinferatoxin, and also by stimuli like heat and protons (134). In the last few years, a number of studies have suggested a physiological role for AEA as a TRPV1 agonist, leading to the concept that AEA, besides being an endocannabinoid, is also a true “endovanilloid†(135, 136). In contrast, 2-AG is unable to bind to and activate TRPV1 (134, 136). The interaction of AEA with TRPV1 occurs at a cytosolic binding site (135, 137), triggering activation of nonselective ion channels, activation of protein kinases, increased intracellular Ca++ concentration, mitochondrial uncoupling, and release of cytochrome c (138, 139). TRPV1 is expressed in peripheral sensory fibers (134) and in several nuclei of the central nervous system (140), suggesting the existence of central endogenous agonists for its activation. In this context, N-arachidonoyldopamine, an endogenous capsaicin-like substance (Fig. 1⇑), has been identified; it activates TRPV1 with high potency and is also a potent cannabimimetic compound (141). Collectively, there seems to be an overlap between the endogenous cannabinoid system and the vanilloid system.

Activation of the different molecular targets by AEA or 2-AG leads to several biological activities, some of which are summarized in Table 2⇓. It appears that virtually all central and peripheral systems in mammals are affected by endocannabinoids. Among the peripheral activities of AEA, the regulation of reproduction is an emerging interest (23, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49). The effects of AEA are under metabolic control, so that within a very narrow concentration range AEA regulates blastocyst function and implantation (125, 142, 143). A more detailed account of this regulation is described in the following sections.
View this table:

In this window
In a new window

TABLE 2.

Effects of endocannabinoids and congeners in the central nervous and peripheral systems
Previous SectionNext Section
III. Endocannabinoids and Male Fertility
A. Introduction

Although there is evidence that chronic administration of THC to animals induces male impotency (144) and reduces testosterone secretion, sperm production, motility, and viability as well as acrosome reaction and fertilization (51, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158), a role for the endocannabinoid system in male fertility is still largely unexplored. Recent reports show that N-acylethanolamines are present in human reproductive fluids at low nanomolar ranges (49), and that AEA influences human sperm functions (48, 146). For example, in vitro studies demonstrate that the AEA congener N-palmitoylethanolamine affects the time-course of capacitation of human spermatozoa by modulating their membrane properties (159, 160). Furthermore, the rat testis is able to synthesize AEA (161), and human seminal plasma contains AEA (49). The presence of CB1 in Leydig cells (162) and its association with testosterone secretion have also been observed in mice (163). There is evidence that THC alters Sertoli cell function (164), although the underlying molecular mechanism is not known. Because Sertoli cells are involved in the regulation of germ cell development by providing nutrients and hormonal signals needed for spermatogenesis, their ability to bind and degrade AEA seems important in controlling the spermatogenic output. Because AEA can serve as a proapoptotic factor (138), it may also be involved in the survival and death of Sertoli cells. In the same context, the possible interplay of AEA with FSH could be of interest, because FSH dramatically impacts fetal and early neonatal Sertoli cell proliferation and is critical for regulating spermatogenesis in adult males (165). These aspects of Sertoli cell biology with relation to endocannabinoid signaling are further elaborated below.
B. The endocannabinoid system in Sertoli cells

In mice, Sertoli cells have the biochemical machinery to bind and degrade AEA at various developmental stages (4 to 24 d) (166). For example, immature Sertoli cells express functional CB2 on their surface, but the receptor level does not fluctuate much during aging (166). Instead, FAAH activity declines age-dependently due to reduced transcription, and so does the uptake of AEA through AMT. A typical AMT was found to uptake AEA in Sertoli cells and, like AMT of other human peripheral cells, this uptake was significantly increased by NO donors (166). This effect might be relevant in vivo, because NO plays roles in regulating male fertility (167, 168). In particular, NO regulates the contribution of Sertoli cells to fertility and to inflammation-mediated infertility (169). A faster removal of AEA from the extracellular space might be the rationale for some effects of NO.

An interesting observation is that AEA can force Sertoli cells to undergo apoptosis and that this process is more evident upon aging (166). The proapoptotic effect of AEA is not mediated by CB1, CB2, or TRPV1. Instead, CB2 expressed by Sertoli cells have a protective role against the harmful effects of AEA, and so does FSH. In fact, FSH dose-dependently inhibits apoptosis, and this event is correlated with increased FAAH activity (166). The finding that the endocannabinoid system is operative in Sertoli cells opens up a new perspective to the understanding and treatment of male infertility. In particular, the observation that FAAH modulates the biological effects of AEA on Sertoli cells and that this FAAH-mediated control is under hormonal regulation constitutes a concept that AEA hydrolysis is an important check-point in human fertility.
C. The endocannabinoid system in sperm

AEA signaling is implicated in regulating sperm functions required for fertilization in invertebrates and mammals, including humans (48, 145, 146, 147, 148, 149, 150, 151, 152). It has been shown in sea urchin (Strongylocentrotus purpuratus) that sperm synthesizes AEA (170) and AEA binds to CB receptors and reduces fertilizing capacity of sperm (48, 146, 147, 148, 149, 150, 151, 152, 171). Unlike lower animals, ejaculated sperm from mammals must undergo functional maturation for fertilizing an egg. Sperm acquire fertilization competence because they reside in the female genital tract where, after a series of physiological changes, they become “capacitatedâ€, i.e., able to fertilize an egg (1, 2, 3). Capacitation consists of changes occurring at two sites: 1) on the sperm head that enables it to bind to the zona pellucida (ZP) and induces acrosome reaction; and 2) in the flagellum, where hyperactivated sperm motility is facilitated. The control of this crucial process involves modifications of intracellular ions (172, 173), plasma membrane fluidity (174), metabolism, and motility (175). However, the sequence of these changes and local regulatory mechanisms that allow capacitation to progress as sperm become closer to an oocyte still remains poorly understood. Recently, AEA has been shown to reduce human sperm motility by reducing mitochondrial activity (176). In addition, AEA inhibits capacitation-induced acrosome reaction, and its effects are prevented by the CB1 antagonist SR141716 (176). These data led to the suggestion that the activity of AEA on sperm function requires CB1 activation. We investigated whether sperm cells of boar (Sus scropha) are able to bind and metabolize AEA and whether this endocannabinoid modulates their function. Boar sperm biology in some aspects resembles that of humans, and boar spermatozoa are available in a greater amount than human spermatozoa. Thus, boar sperm were used as a model to fully characterize the endocannabinoid system (177, 178, 179).

Boar spermatozoa have the biochemical machinery to bind (CB1 and TRPV1), synthesize (NAPE-PLD), and degrade (AMT and FAAH) AEA (180). It was also shown that activation of CB1 by an AEA-stable analog, methanandamide, inhibits capacitation and hence the ability of sperm cells to react to ZP proteins with acrosome exocytosis through a cAMP-dependent pathway, although CB1 was ineffective on spontaneous acrosome reaction (180). It was also noted that once the capacitation is completed, AEA stabilizes the acrosome membranes by activating TRPV1, thus reducing spontaneous acrosome reaction. These results are schematically depicted in Fig. 3⇓.
Fig. 3.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 3.

The endocannabinoid system in sperm function. Binding of AEA to the extracellular site of CB1 (CB1R) leads to inhibition of sperm motility, capacitation, and ZP-induced acrosomal reaction (AR), without affecting spontaneous AR. In contrast, binding of a lipoxygenase-catalyzed oxygenation of arachidonylethanolamide to the intracellular site of vanilloid receptors (TRPV1) inhibits spontaneous AR. Sperm also possess the AMT, the AEA-synthesizing phospholipase D (NAPE-PLD), and the AEA-hydrolyzing FAAH. FAAH cleaves AEA into ethanolamine (EtNH2) and AA.

Taken together, sperm function seems to be regulated by endocannabinoids that exert a dual stage-dependent effect. On one hand, AEA, present in both seminal plasma and uterine fluids, may prevent premature capacitation in freshly ejaculated sperm via a CB1-mediated mechanism for traveling along the uterine tract without any fertilizing potential. Conversely, a few hours later when sperm have reached the oviduct (a condition that corresponds to in vitro capacitation), this inhibitory brake becomes less stringent. It is speculated that spermatozoa are exposed to a progressively reduced concentration of AEA in the proximal female genital tract (49), and sperm capacitation occurs as a consequence of release from CB1 inhibition. The observation that the endocannabinoid system is operative in sperm adds a new dimension to the intricate endocannabinoid network regulating mammalian fertility. Overall, these findings present new perspectives to the understanding and treatment of male fertility problems.
Previous SectionNext Section
IV. Endocannabinoids and Female Fertility: Embryo Implantation
A. Introduction

The onset of a new life begins with fertilization of a mature oocyte with capacitated sperm (1, 2, 3, 4). The one-cell fertilized zygote, now termed embryo, undergoes several mitotic cell divisions, eventually forming the blastocyst with two distinct cell populations, the ICM and a layer of trophectoderm cells surrounding the ICM (7, 8, 9). The embryo proper is derived exclusively from the ICM, whereas the placenta and extraembryonic membranes are generated from cells contributed by the trophectoderm (181, 182). A two-way interaction between the blastocyst and the maternal uterine luminal epithelium initiates the process of implantation. A considerable amount of early pregnancy loss occurs due to either preimplantation embryonic death or implantation failure resulting from asynchronous embryonic development and/or failure of the uterus to differentiate to the receptive stage (183, 184). Understanding the mechanism of preimplantation embryonic development and implantation in the uterus is a fundamental challenge to reproductive biologists with the goal of alleviating the problems of human infertility and ensuring the birth of quality offspring. Such knowledge will also help in developing novel contraceptive approaches to restrict rapidly growing world population.

Development of the preimplantation embryo to the blastocyst stage and differentiation of the uterus to the receptive stage are basic requirements for the initiation of implantation in all species (5, 6, 8). Although the precise sequence and details of the molecular interactions involved in these processes have not yet been defined, increasing evidence from gene expression and transgenic mouse studies during the last two decades shows that coordinated integration of a range of signaling pathways in paracrine, autocrine, and/or juxtacrine manners participates in embryo-uterine dialogue during implantation (5, 6, 8, 185, 186). Among these signaling molecules, endocannabinoid signaling has recently been highlighted as an important player in directing preimplantation embryo development and the timely homing of embryos into a receptive uterus for implantation.
B. Endocannabinoids and preimplantation embryo development

THC, the major psychoactive component in marijuana, has been shown to exert a wide array of adverse effects on human health, including reproduction (23, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49). With the identification of CB1 and CB2 (77, 78) and two major endocannabinoids, AEA and 2-AG (83, 84, 85), as ligands for these receptors in the early 1990s, we and others using mice as an animal model have been pursuing experiments to explore the pathophysiological significance of cannabinoid/endocannabinoid ligand-receptor signaling during early pregnancy.

In mice, only CB1 is expressed in the oviduct and uterus, whereas both CB1 and CB2 are expressed in preimplantation embryos (69, 187, 188, 189). CB1 mRNA is primarily detected from the four-cell stage through the blastocyst stage, whereas CB2 is present from the one-cell through the blastocyst stage (69). Autoradiographic binding sites of [3H]AEA are also evident from the one-cell through blastocyst stages. Interestingly, the majority of AEA binding sites are noted in outer cells of embryos at the eight-cell, morula, and blastocyst stages. Scatchard analysis of binding kinetics in d 4 blastocysts (d 1 = vaginal plug) showed that AEA binds to a single class of high-affinity receptors. The presence of CB1 mRNA and protein as detected by immunocytochemistry in preimplantation embryos correlates well with AEA binding sites (188, 190). Furthermore, blastocyst CB1 is biologically active, because both THC and AEA inhibit forskolin-stimulated cAMP formation, and this inhibition is prevented by pertussis toxin pretreatment (69, 187). The presence of biologically active CB1 in the blastocyst suggested that the mouse embryo is a potential target for both endocannabinoids and natural cannabinoids. In fact, synthetic (CP 55940, WIN 55212–2), natural (THC), or endogenous (AEA and 2-AG) cannabinoids arrest the development of two-cell embryos into blastocysts in culture (69, 191). A reduction in trophectodermal cell numbers is noted in those blastocysts that escape the developmental arrest in the presence of cannabinoid agonists (190). Furthermore, these adverse effects are reversed by simultaneous addition of selective antagonists to CB1 [SR 141716 (192)] with cannabinoid agonists (191), but not by a selective CB2 antagonist [SR 144528 (193)]. Recent observations of CB2 expression in early embryos and embryonic stem cells by microarray analysis (194), and the absence of its expression in trophoblast stem cells derived from preimplantation blastocysts (H. Wang and S. K. Dey, unpublished data) suggest that CB2 expression is restricted to blastocyst ICM cells. Collectively, these results suggest that cannabinoids mediate their actions on preimplantation embryos via CB1. The role of CB2 in the early embryo is yet to be defined.

With the availability of cannabinoid receptor knockout mice in the late 1990s (195, 196), the physiological relevance of cannabinoid receptor signaling during early embryo development was further examined. It was observed that CB1−/−, CB2−/−, or CB1−/−×CB2−/− double mutant embryos recovered from the oviduct on d 3 and from the uterus on d 4 of pregnancy show asynchronous development compared with wild-type embryos (188, 189). This impaired in vivo embryo development is rescued by mating CB mutant females with wild-type males, producing all heterozygous embryos in a mutant maternal environment. This indicates that embryonic CB receptors, but not maternal factors, direct early synchronous embryonic development (189). These studies also provide genetic evidence that CB1 and CB2 are critical for preimplantation embryo development, although asynchronous development of CB2−/− embryos still remains puzzling. Because CB2 is expressed in the embryonic stem cells, but not in trophectoderm-derived trophoblast stem cells, it is conceivable that CB2 plays a role in specifying pluripotent ICM cell lineage during blastocyst formation. To ascertain whether embryos deficient in cannabinoid receptors respond to endocannabinoids in vitro, two-cell wild-type or mutant embryos were cultured in the presence or absence of AEA. Although a comparable development of wild-type and mutant embryos was observed in the absence of AEA in culture, CB1−/− and CB1−/−×CB2−/− mutant embryos, but not CB2−/− or wild-type embryos, were resistant to the inhibitory action of AEA (188). This observation reinforces the tenet that CB1 is the functional receptor for ensuring normal embryo growth and differentiation to blastocysts. Collectively, these studies provide pharmacological, molecular, and genetic evidence that the preimplantation embryo is indeed a target for cannabinoid ligand-receptor signaling.
C. Endocannabinoids and oviductal embryo transport

During early pregnancy, another critical event occurring in parallel with preimplantation embryonic development is the timely transport of embryos from the oviduct into the uterus. In mice, embryos at the late morula or early blastocyst stage enter the uterus, where they develop and differentiate to achieve implantation competency, escape from the ZP, and implant into the receptive uterus. Thus, normal oviductal embryo transport is one of the prerequisites for on-time implantation in the uterus, whereas a dysfunctional regulation of this process resulting from oviductal embryo retention may increase the incidence of pregnancy failure or cause tubal pregnancy in humans.

During the course of our study over the past several years exploring the potential physiological roles of endocannabinoid signaling during early pregnancy, we have consistently observed that approximately 40% of CB1−/− mice show pregnancy loss (188, 189). Because these mutant mice have normal ovulation and fertilization when compared with wild-type mice (189), we initially thought that asynchronous embryo development could be the major cause of this pregnancy loss. We speculated that normal pregnancy in CB1−/− mice would be restored by mating mutant females with wild-type males to generate all heterozygous embryos with normal preimplantation growth. However, we still observed that about 40% of CB1−/− mothers did not yield any embryos in the uterus when examined in the midmorning on d 4 of pregnancy, suggesting that a maternal, but not embryonic, loss of CB1 is the determining cause for pregnancy failure.

To determine the underlying cause for this pregnancy failure, we examined oviductal embryo transport in CB1−/−, CB2−/−, and CB1−/−×CB2−/− double mutant females. No embryos were found to be trapped in the oviducts in wild-type or CB2−/− mice, which was expected because only CB1 is expressed in the mouse oviduct and uterus. However, a substantial number of CB1−/− and CB1−/−×CB2−/− mice showed impaired oviductal transport with retention of embryos at the morula and blastocyst stages within the oviduct on d 4. These trapped embryos appeared morphologically normal and implanted upon transfer into d 4 pseudopregnant receptive uteri, suggesting that they remained implantation-competent. These results indicate that maternal expression of CB1 in the reproductive tracts plays a fundamental role in ensuring normal oviduct to uterine transport of embryos, and its deficiency results in embryo retention in the oviduct for an extended period, causing reduced fertility in CB1−/− mice. This observation was confirmed in our reciprocal embryo transfer experiments between CB mutant and wild-type mice. Indeed, only CB1−/− recipients show oviductal embryo retention and implantation failure, irrespective of the genotypes of donor embryos (189). We further observed that wild-type pregnant mice treated with a CB1-selective antagonist (SR141716) exhibit impaired embryo transit through the oviduct, but this defect did not occur in mice treated with vehicle or a CB2-selective antagonist (SR144528). Interestingly, wild-type females exposed to a stable AEA analog (methanandamide) or natural THC showed pregnancy loss with embryos retained in the oviduct (189). These observations demonstrate that aberrant cannabinoid signaling, either silenced or enhanced, impairs embryo transport. This suggests that there is an endocannabinoid tone mediated via CB1 in the oviduct that regulates normal embryo transport into the uterus for implantation. Collectively, these observations provide evidence that whereas embryonic CB1 primarily contributes to normal embryo development, oviductal CB1 directs timely oviductal transport of embryos. Although there is no evidence of ectopic pregnancy in animals other than humans, these findings may have clinical importance because embryo retention in the fallopian tube as a result of dysfunctional muscular contraction is one cause of ectopic pregnancy in women (197, 198). The ability of trapped blastocysts within CB1−/− oviducts to implant after transfer into wild-type pseudopregnant mouse uteri suggests that similar embryo retention in the fallopian tube would result in ectopic pregnancy in women.

Previous studies have established that the journey of the embryo from the oviductal isthmus into the uterus in rodents is aided by a wave of regulated contraction and relaxation of the oviduct muscularis. It is thought that the sympathetic neuronal circuitry, under the direction of ovarian hormones, coordinates the “closing and opening†of the sphincter at the isthmus-uterine junction, thereby regulating the timely passage of embryos from the oviduct into the uterus (199, 200). During pregnancy, rising progesterone (P4) levels from the newly formed corpus luteum decrease the turnover rates and thus the levels of noradrenaline, a ligand with higher affinity for the α-adrenergic receptor (AdR) than the β-AdR at the adrenergic nerve endings (201). In contrast, the sensitivity of the β-AdR is increased in the circular muscle of the oviduct isthmus under P4 dominance, causing muscle relaxation and facilitating embryo transport through the oviduct (199). Observations of embryo retention within the oviduct in wild-type females after exposure to an α1-AdR agonist phenylephrine and/or a β2-AdR antagonist butoxamine (189) led us to speculate that CB1-mediated endocannabinoid signaling is functionally coupled to adrenergic signaling to regulate oviductal motility conducive to embryo transport. Indeed, CB1 expression is colocalized with that of α1- and β2-AdRs in mouse oviductal muscularis at the isthmus region, and the β-AdR agonist isoproterenol restores normal embryo transport in CB1−/− mice (189). Experiments with in vitro [3H]noradrenaline release provided further evidence that genetic or pharmacological loss of oviductal CB1 increases noradrenaline release from the adrenergic nerve terminals, maintaining a smooth muscle contractile tone through α-AdR, and thereby impeding oviductal embryo transport. In contrast, exposure to excessive natural or synthetic cannabinoid ligands leads to predominant relaxation phase of the oviductal muscularis due to attenuated noradrenaline release, impairing embryo transport (189). Collectively, these findings reinforce the concept that aberrant cannabinoid signaling, arising from either silencing or amplification, impedes the highly coordinated oviductal smooth muscle contraction and relaxation critical to embryo transport during early pregnancy. The potential mechanism creating this endocannabinoid tone in the oviduct during early pregnancy remains to be explored. A differential regional expression of NAPE-PLD and FAAH with higher expression of NAPE-PLD in the isthmus and FAAH in the ampulla may contribute to generate an appropriate level of AEA conducive to preimplantation embryo growth and transportation (H. Wang and S. K. Dey, unpublished data).
D. Biphasic endocannabinoid sensor in blastocyst implantation

Upon a successful journey through the oviduct, the embryo at the late morula or early blastocyst stage encounters a new microenvironment in the uterus for its attainment of implantation competency. It is thought that blastocyst activation and uterine receptivity are two distinct events in the process of implantation (202). In mice, ovarian P4 and 17β-estradiol (E2) are the primary factors that coordinate these two events (5). Under P4 priming, the closure of the uterine lumen occurs and coincides with the escape of the blastocyst from the ZP, bringing the blastocyst trophectoderm in close contact with the uterine luminal epithelium (blastocyst apposition). Superimposition of the P4-primed uterus with preimplantation ovarian E2 secretion and its catechol metabolite, 4-hydroxy-17β-estradiol (4-OH-E2), produced from primary E2 in the uterus, differentially regulate uterine preparation and blastocyst activation, respectively. For example, the primary ovarian E2, via its interaction with nuclear E2 receptors, participates in the preparation of the P4-primed uterus to the receptive state in an endocrine manner, whereas its metabolite 4-OH-E2, mediates blastocyst activation for implantation in a paracrine manner (203). These coordinated actions of P4 and E2 set up the window of implantation. The first event in the process of implantation is the attachment of the blastocyst trophectoderm with the uterine luminal epithelium that occurs within a narrow time frame when an intimate two-way dialogue occurs between the implantation-competent blastocyst and the receptive uterus. In mice, this attachment reaction is initiated around 2400 h of d 4 of pregnancy (204). However, elimination of preimplantation E2 secretion by ovariectomy on the morning of d 4 results in implantation failure with blastocyst dormancy within the quiescent uterine lumen (205, 206). This condition is referred to as delayed implantation and can be maintained for many days by continued P4 treatment. However, implantation with blastocyst activation is rapidly initiated by a single injection of E2 in the P4-primed uterus (205, 206). This physiologically relevant delayed implantation model has been widely used to identify signaling pathways mediating embryo-uterine cross-talk during blastocyst implantation. Using normal pregnancy and delayed implantation model, we have elucidated a unique association of endocannabinoid-CB1 signaling in embryo-uterine interactions during implantation.

As stated earlier, natural, synthetic, or endogenous cannabinoids can dramatically inhibit preimplantation embryo development and blastocyst zona-hatching in culture (69, 191, 207). This observation correlates well with higher levels of AEA in the nonreceptive uterus (142, 188, 207). On the other hand, lower levels of AEA in the receptive uterus and at the implantation site suggest that regulated AEA levels are conducive to normal embryo development and implantation. There is evidence that cannabinoid effects are differentially executed, depending on the embryonic stage and cannabinoid levels in the uterine environment. Blastocysts exposed in culture to low levels of AEA exhibit accelerated trophoblast differentiation and outgrowth, whereas inhibition of trophoblast differentiation is observed at higher doses of AEA (143, 208), suggesting dual functions of AEA depending on its local concentration (143, 207). Thus, uterine AEA levels are critical in regulating the “window†of implantation by synchronizing trophoblast differentiation and uterine preparation to the receptive state.

To gain further insight into the underlying causes of these biphasic effects of AEA, the status of anandamine binding in preattachment and attachment-competent blastocysts immediately before implantation on d 4 of pregnancy was studied. Similar expression studies were also performed using dormant and estrogen-activated blastocysts. The results showed that normal blastocysts collected in the early morning of d 4 have higher levels of AEA binding, but this binding remarkably declines in blastocysts recovered on d 4 in late afternoon before the attachment reaction (188). These observations suggest that down-regulation of AEA binding to the blastocyst is important for achieving implantation competence. Similarly, dormant blastocysts also show increased levels of AEA binding sites, but this binding significantly decreases by 12 h after termination of dormancy by an E2 injection (188). The immunoreactive CB1 protein parallels AEA binding in dormant and activated blastocysts (125, 188). These results collectively suggest that coordinated down-regulation of blastocyst CB1 and uterine AEA levels in the receptive uterus are important for implantation. It is interesting to note that the peripheral AEA levels remain relatively low during implantation, whereas the levels increase before and during parturition in humans (209).

Increasing evidence suggests that the bioeffectiveness of AEA depends on its concentration in the extracellular space, which is regulated by its synthesis by NAPE-PLD, its transport across the plasma membrane, and its degradation by FAAH (92, 93, 94, 95, 96, 97, 98, 210). To further address the underlying mechanism by which differential uterine AEA levels are spatiotemporally established under different pregnancy status, we examined the expression profiles of NAPE-PLD and FAAH in the mouse uterus during early pregnancy. Correlating with higher levels of AEA in the nonreceptive uterus and interimplantation sites, higher levels of Nape-pld mRNA and NAPE-PLD activity were detected in these tissues compared with the implantation site and receptive uterus (142). It is interesting to note that the implanting blastocyst exerts an inhibitory effect on uterine Nape-pld expression (142). There is also evidence that blastocysts can up-regulate uterine FAAH activity by releasing a lipid “FAAH activator†(211). These observations suggest a potential role of the implanting embryo in regulating uterine AEA levels, perhaps to serve as a protective mechanism against exposure to detrimental levels of AEA. This is further confirmed by the observation of higher FAAH expression and activity in the implanting embryo (212, 213). Therefore, the differential and dynamic expression and activity of NAPE-PLD and FAAH in the embryo and uterus create optimal levels of AEA beneficial to blastocyst activation and uterine receptivity for implantation. This tight regulation of AEA synthesis and hydrolysis in the pregnant uterus further indicates that endocannabinoid ligand-receptor signaling plays an important role in implantation.

These studies clearly establish the concept that whereas lower levels of AEA and CB1 are beneficial for implantation, higher levels are detrimental. Using the delayed implantation mouse model, we have provided further evidence that AEA at low concentrations confers blastocyst competency to implantation via CB1 (125), whereas experimentally elevated natural or synthetic cannabinoid levels interfere with implantation. These findings are consistent with our previous observations of stimulation and inhibition of trophoblast growth at low and high AEA levels, respectively (143). To reveal the underlying mechanism of this biphasic AEA action in blastocyst implantation, we further explored the potential signaling pathways that are coupled with CB1 under different AEA concentrations. We found that AEA-induced stimulatory and inhibitory influences on blastocyst function and implantation are executed by different signal transduction pathways: the ERK and Ca++ signaling pathways. AEA at a low concentration activates ERK signaling in dormant blastocysts via CB1. In contrast, at higher AEA levels, it fails to achieve ERK activation, but instead inhibits Ca++ mobilization (125). This finding provided for the first time a potential “cannabinoid sensor†mechanism to influence crucial steps during early pregnancy. An association of spontaneous pregnancy loss with elevated peripheral AEA levels in women (214, 215) is consistent with the observations in mice (see Section V). These findings in mice and humans reinforce the concept that endocannabinoid signaling is at least one of the pathways determining the fate of embryo implantation. In this regard, there is evidence that activation of CB1 inhibits human decidualization and promotes apoptosis of decidual cells in vitro (216), thus adding a new role of endocannabinoids in human pregnancy. The possible physiological consequence of the different signaling pathway triggered by AEA through GPR55 (130, 131, 217) deserves further investigation. In addition, the pathophysiological impact of other endocannabinoids or “endocannabinoid-like†compounds (218) on various reproductive events warrants further investigation.

Taken together, these studies demonstrate that under normal physiological conditions, endocannabinoid signaling through CB1 is crucial to various female reproductive events that include development of embryos, their oviductal transport, and ultimately their homing and implantation in the receptive uterus; conversely, an aberration in endocannabinoid signaling, either silenced or enhanced, derails these processes (Fig. 4⇓). These observations add a new dimension to the concern that the adverse effects of maternal use of cannabinoids on offspring may be seeded very early in pregnancy. There is now evidence that defective implantation creates an adverse ripple effect during the subsequent course of pregnancy both in humans and mice (12, 33, 35). Therefore, our findings in mice raise a cautionary note for women of reproductive ages regarding chronic abuse or medicinal consumption of marijuana or other endocannabinoid system-oriented drugs. More importantly, they raise caution against the use of CB1 antagonists to treat obesity in humans.
Fig. 4.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 4.

Endocannabinoid signaling in blastocyst activation and implantation. Evidence suggests that regulated levels of endocannabinoids, primarily AEA, in the receptive uterus and CB1 in activated blastocysts, are beneficial for implantation, whereas higher levels are detrimental to this process. This biphasic role of AEA is further supported by findings that AEA within a very narrow range regulates blastocyst activation and implantation by differentially modulating ERK signaling and Ca2+ channel activity via CB1. Uterine AEA levels conducive to implantation are primarily regulated by the coordinated expression and activity of N-acylphosphatidylethanolamine-hydrolyzing PLD (NAPE-PLD) that generates AEA and by FAAH that degrades AEA in the uterus during early pregnancy. In addition, the implanting blastocyst down-regulates uterine NAPE-PLD expression, but enhances uterine FAAH activity via releasing a putative FAAH activator, thus contributing to rapid turnover of AEA at the implantation site. Ge, Glandular epithelium; IS, implantation site; INTER-IS, interimplantation site; Le, luminal epithelium; Myo, myometrium; S, stroma; Tr, trophectoderm.
Previous SectionNext Section
V. Endocannabinoids and Female Fertility: Immunoregulation
A. Th1/Th2 cytokines and fertility

There is evidence for the role of peripheral lymphocytes in embryo implantation and successful pregnancy in humans (219). In fact, normal gestation is based on an early immunological adaptation that involves peripheral T lymphocytes in pregnant women (40, 219, 220). These cells produce type 1 T-helper (Th1) and type 2 T-helper (Th2) cytokines, which have opposite effects on trophoblast growth, as schematically depicted in Fig. 5⇓. Th2 cytokines (IL-3, IL-4, and IL-10) favor blastocyst implantation and successful pregnancy by promoting trophoblast growth either directly or indirectly through the inhibition of natural killer (NK) cell activity and the stimulation of natural suppressor cells. Conversely, Th1 cytokines [IL-2, IL-12, and interferon-γ (INF-γ)] impair gestation by causing a direct damage to the trophoblast through stimulation of NK cells and secretion of TNF-α by macrophages. The latter cells play a role in this network, but several aspects of their contribution to the balance of Th1 and Th2 cytokines remain to be elucidated. The trophoblast stimulates the release of profertility Th2 cytokines from T lymphocytes (so-called “Th2 biasâ€) through IL-4, whereas the antifertility Th1 bias is signaled by IL-2. In addition, P4 plays a role in this network, and in fact it induces a Th2 bias by binding to the intracellular P4 receptor in T cells (219, 221). The resulting hormone-cytokine network is a key element at the fetal-maternal interface, and a defect in its integrity may result in fetal loss (219, 221, 222, 223, 224). Additionally, T lymphocytes produce leukemia inhibitory factor (LIF), which favors embryo implantation and survival (221, 222, 223, 224). IL-4 stimulates, whereas IL-2 inhibits LIF release (Fig. 5⇓). Clinical observations that women with unexplained recurrent abortions have reduced expression of LIF production suggest that this cytokine is indeed critical for implantation and pregnancy maintenance in humans (219, 220, 225). In this context, P4-induced Th2 bias has been found to stimulate LIF release from T lymphocytes (Fig. 5⇓), and IL-4 has been shown to mediate this P4 effect (219).
Fig. 5.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 5.

Th1/Th2 cytokines in human fertility. Th2 cytokines (IL-3, -4, and -10) are released by T cells and favor blastocyst implantation by promoting trophoblast growth through inhibition of NK cell activity. Conversely, Th1 cytokines (IL-2, IL-12, and INF-γ), also released by T cells, impair gestation by damaging the trophoblast through stimulation of NK cells. Macrophages contribute to NK activity by secreting TNFα. The trophoblast induces a Th2 bias through IL-4, whereas IL-2 stimulates the release of Th1 cytokines. Also, P4 induces the Th2 bias by binding to T lymphocytes. Finally, these cells release LIF under positive or negative control of IL-4 or IL-2, respectively.
B. The endocannabinoid system in lymphocytes of pregnant women

The FAAH activity is known to regulate the endogenous tone and the biological activity of AEA in vivo (226). In this respect, lymphocyte FAAH has been shown to influence pregnancy outcome by regulating AEA level at the fetal-maternal interface (40), which appears to interfere with the lymphocyte-dependent cytokine network. Thus, decreased activity and expression of FAAH in peripheral lymphocytes is associated with pregnancy loss and may serve as an early (<8 wk gestation) marker of human spontaneous abortion; AMT activity and cannabinoid binding are not altered (214, 227). Interestingly, defective FAAH in maternal lymphocytes is also associated with failure to achieve an ongoing pregnancy after in vitro fertilization and embryo transfer (215). Therefore, it seems that FAAH, but not AMT or cannabinoid receptors, is important in lymphocyte-mediated regulation of the hormone-cytokine network at the fetal-maternal interface in natural and medically-assisted gestation. In addition, analysis of the lymphocyte-endocannabinoid system during human ovulatory cycles has shown the highest FAAH activity and the lowest AEA concentrations on d 21 of the cycle (Table 3⇓), a period that temporally coincides with the putative window of uterine receptivity for implantation (228); binding to CB1 and activities of AMT and NAPE-PLD were similar in T cells at all stages of the ovulatory cycle (Table 3⇓).
View this table:

In this window
In a new window

TABLE 3.

The endocannabinoid system in human lymphocytes during the ovulatory cycle
C. FAAH as a molecular integrator of fertility signals

FAAH expression in T lymphocytes has been shown to be regulated by Th1/Th2 cytokines: IL-4 and IL-10 enhance FAAH activity, whereas IL-2 and INF-γ attenuate its activity (227). In particular, IL-4, known to mediate favorable effects of P4 on pregnancy (221, 229), also partially mediates the effect of P4 on FAAH expression. Unlike FAAH, little alteration is noted for AMT, NAT, and NAPE-PLD activities and CB1 expression in intact lymphocytes by this steroid (227, 230). P4 also modulates the effects of THC on sexual receptivity (227). It appears that this effect of P4 occurs through increased level of the transcription factor Ikaros, which in turn increases FAAH gene expression by binding to a specific sequence in the promoter region (230). Profertility Th2 cytokines potentiate the activation of FAAH by P4, whereas antifertility Th1 cytokines have the opposite effect (227). Leptin is also an important regulator of fertility (231) and immune response (232), and leptin knockout (ob/ob−/−) mice are infertile (231). Leptin, too, enhances FAAH gene transcription through a signal transducer and activator of transcription 3-mediated up-regulation of the FAAH promoter (233). Leptin alone or synergistically with P4 reduces AEA levels in T cells, without affecting the other players in the endocannabinoid system (233). Overall, the up-regulation of lymphocyte FAAH by profertility signals strengthen the speculation that this enzyme affects human fertility by modulating the AEA levels. Also, uterine FAAH is regulated by sex hormones (234), but the implications and molecular details of this regulation are still elusive. Nonetheless, FAAH activity seems to be important for embryo-uterine cross-talk, because mouse blastocysts release a soluble “FAAH activatorâ€, which contributes to AEA disposal at the implantation site (211). The initial biochemical characterization of this activator shows that it is neutralized by lipase activity, whereas PLA2, PLC or PLD, DNAse I, or RNAse A are ineffective. Additionally, the FAAH activator does not resemble platelet-activating factor, leukotriene B4, or PGs known to be present in blastocysts (211); its molecular identity is under investigation. In addition, there is molecular evidence that uterine NAPE-PLD is a major player in regulating the levels of AEA in the uterus during early pregnancy (142). In fact, NAPE-PLD activity was higher at the interimplantation site and lower at the implantation site, and E2 and P4 down-regulated its expression (142). Thus, P4 seems to up-regulate lymphocyte FAAH activity, whereas it down-regulates uterine NAPE-PLD expression. This would suggest that regulated AEA levels are critical to successful implantation and pregnancy establishment. On a final note, it is also possible that interplay between endocannabinoids and eicosanoids (prostanoids, leukotrienes, or lipoxins) contributes to immunoregulation of fertility, but this speculation awaits experimental support.
Previous SectionNext Section
VI. Endocannabinoids and Clinical Implications

Implications of the endocannabinoid system working via cannabinoid and vanilloid receptors in many central and peripheral aspects of human pathophysiology have been proposed. We describe here the endocannabinoid signaling pathways that impact both male and female fertility with the hope of designing and developing endocannabinoid-oriented drugs for the treatment of infertility. In this respect, the biphasic roles of endocannabinoid signaling in fertilization, preimplantation embryo development, oviductal embryo transport, and implantation have major clinical implications (69, 125, 142, 143, 146, 148, 180, 187, 188, 189, 191, 207, 211, 212, 213, 215, 227, 230, 234, 235). The incidence of spontaneous abortion, the most common adverse outcome of pregnancy, associated with considerable sufferings and medical costs (236, 237), is increased by cigarette smoking and the use of illicit drugs (238, 239). It is interestingly to note that the early stages (< 8 wk) of spontaneous abortion in humans are associated with decreased activity and expression of FAAH in maternal T lymphocytes and increased blood levels of AEA. This correlation of down-regulation of FAAH in women who are prone to miscarriage appears specific, because other members of the endocannabinoid system are not affected (214, 215, 227). This suggests that high FAAH activity with low AEA levels are among the factors that are important for successful pregnancy and raises concerns about marijuana use by pregnant women.

We suggest that FAAH is an important player in coordinating hormone-cytokine-endocannabinoid networks critical to reproduction. Thus, the FAAH level may serve as a marker to monitor the early stages (<8 wk) of human gestation. Because low FAAH levels correlate with pregnancy failure, it is possible that drugs that enhance FAAH activity may improve human fertility. Regulating endocannabinoid levels via metabolic pathways could be an advantage over the use of CB receptor agonists and antagonists to eliminate their psychotropic effects (109, 240, 241, 242). However, the development and use of selective FAAH activators as therapeutics for fertility regulation may significantly reduce or eliminate AEA signaling via CB receptors and increase the incidence of reproductive disturbances (188, 189). Furthermore, FAAH regulates the levels of several other endogenous endocannabinoid-like compounds whose functions are not yet known, leaving open the question of the biological consequences of their enhanced degradation upon treatment with FAAH activators. Other enzymes that catalyze the hydrolysis of N-palmitoylethanolamine (243) and hydrolysis (117) and synthesis (112) of 2-AG have recently been identified. It remains to be seen how these pathways contribute to the overall endocannabinoid tone relevant to reproductive functions. In addition, the development and use of FAAH activators calls for attention to “the other side of the coin,†because there is effort to develop FAAH inhibitors (rather than activators) as novel therapeutic agents for the treatment of pain (226), neurodegenerative disorders (111), cancer (244, 245), and anxiety (246).
Previous SectionNext Section
VII. Conclusions and Future Direction

Mammalian reproduction is a complex process, involving spermatogenesis, oogenesis, fertilization, preimplantation embryo development, timely passage of embryos through the oviduct, and their implantation in the uterus, eventually establishing a functional placenta for successful pregnancy. Each step in this process requires spatiotemporally regulated various networks of endocrine, paracrine, juxtacrine, and autocrine modulators. Although previous and prevailing studies have placed much emphasis on the roles of cannabinoids/endocannabinoids on neuronal functions (109, 111, 247, 248, 249), there were sporadic reports of adverse effects of cannabinoids on pregnancy outcome, including retarded embryo development and pregnancy failure. Emerging evidence now points toward important roles of the endocannabinoid system in mammalian reproduction. In this review, we present molecular, genetic, physiological, and pharmacological evidence that cannabinoid/endocannabinoid signaling is functionally operative in both male and female reproductive events.

With respect to male reproductive functions, endocannabinoids show biphasic roles in regulating Sertoli cell apoptosis via differential receptor signaling mechanisms. Furthermore, we describe here a stage-dependent effect of AEA on sperm function in that it prevents premature capacitation via a CB1 to ensure normal transit of sperm through the uterus to oviduct. It is tempting to suggest that such an inhibitory brake may become less stringent when sperm reaches the oviduct, the site of fertilization. Activation of CB1 by AEA (extracellular signaling) leads to inhibition of sperm motility, capacitation, and ZP-induced but not spontaneous acrosome reaction. In contrast, binding of AEA to TRPV1 receptors (intracellular signaling) inhibits spontaneous acrosome reaction.

Upon fertilization, one-cell embryos initiate cell division and undergo preimplantation development. As described above, endocannabinoid signaling through CB1 is crucial to various female reproductive events that include development of embryos, their oviductal transport, and ultimately their homing and implantation in the receptive uterus; conversely, an aberration in endocannabinoid signaling, either silenced or enhanced, derails these processes. Furthermore, decreased activity and expression of FAAH in maternal T lymphocytes and resulting increased AEA levels are associated with spontaneous abortion in women. These findings add a new dimension to the concern that the adverse effects of maternal use of cannabinoids on offspring may be seeded very early in pregnancy, thus raising a cautionary note for women of reproductive ages regarding chronic abuse or medicinal consumption of marijuana or other endocannabinoid system-oriented drugs. More importantly, they raise caution against the use of CB1 antagonists to treat obesity in humans. Further in-depth investigation is warranted to better understand pathophysiological significance of the cannabinoid/endocannabinoid signaling pathway in mammalian reproduction.

Synchronous development of the preimplantation embryo to the blastocyst stage before implantation is one of the prerequisites for normal “on-time†implantation. We have observed that either silencing or amplifying CB signaling leads to asynchronous preimplantation embryo development (188, 189). However, the underlying molecular mechanisms remain unknown. Recently, global gene expression profiles during preimplantation mouse development analyzed by microarray techniques have generated a comprehensive data set covering nearly all mouse genes during early embryogenesis (194, 250, 251, 252, 253, 254, 255, 256). Therefore, strategies comparing global gene expression, proteomic, and/or lipidomic profiles between normal and defective embryos may help to identify novel genes and gene products regulated by endocannabinoid signaling in the periimplantation embryo development.

Furthermore, it is crucial to gather in-depth information on how the endocannabinoid signaling is differentially regulated in peripheral and central systems. It is especially an important concern for developing endocannabinoid system-oriented drugs for selectively targeting central or peripheral tissues to avoid adverse effects in unrelated tissue types. Therefore, more efforts should be directed to explore in detail the tissue- and cell-specific effects of endocannabinoid signaling using conditional transgenic mouse models. In fact, there is a recent study showing functional dissociation of FAAH between central and peripheral tissues using brain tissue-selective mutant mice (257). On the other hand, efforts should also be directed to elucidate the common link that encompasses the central and peripheral endocannabinoid signaling. For example, increasing evidence suggests that AEA-CB1 signaling exerts a regulatory role on hypothalamic neuronal activity for the pulsatile release of GnRH (GnRH) (258, 259, 260), a central hormone that controls reproductive performances in both males and females.

Although a wealth of knowledge on the roles of lipid mediators including endocannabinoids, LPA and PGs, and protein signaling molecules, such as growth factors, cytokines, homeotic genes, and transcription factors in embryo-uterine interactions during implantation, has been generated (5, 6, 8, 23, 36, 183), their hierarchical blueprint in directing uterine and embryonic functions during pregnancy remains to be deciphered. We need to understand whether these pathways function independently or in parallel, or converge to a common signaling pathway to establish a network of lipid-protein signaling cross-talk between the embryo and uterus that is necessary for implantation. In this respect, another area of attention in endocannabinoid research has evolved from the finding that the COX and lipoxygenase enzymes can use AEA and 2-AG as substrates to generate novel PGs and hydroxy-endocannabinoids (261, 262, 263, 264), suggesting a close link between endocannabinoids and eicosanoids (265). The potential function and signaling pathways of these metabolic products in reproductive events remain to be determined.
Previous SectionNext Section

Previous SectionNext Section
Acknowledgments

The authors thank Prof. A. Finazzi-Agrò (Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergataâ€) for continuing support and all colleagues who over the years gave their valuable contribution to the fertility studies. We also thank G. Bonelli for excellent production of the artwork and Susanne Tranguch for her critical reading of the manuscript. We apologize for unintended omission of any relevant references.
Previous SectionNext Section
Footnotes

Studies by the authors incorporated in this review were supported by National Institutes of Health Grants (DA06668, HD12304, and HD33994, to S.K.D.); Ministero dell'Istruzione, dell'Università e della Ricerca (COFIN 2002 and 2003, to M.M.); and Fondazione TERCAS (Research Program 2004, to M.M.). S.K.D. is a recipient of the Method to Extend Research in Time Awards from the National Institute of Child Health and Human Development and the National Institute on Drug Abuse. H.W. is a recipient of the Solvay/Mortola Research Award from the Society for Gynecologic Investigation.

The authors have nothing to declare.

First Published Online May 8, 2006

Abbreviations: AA, Arachidonic acid; AdR, adrenergic receptor; AEA, N-arachidonoylethanolamine (also known as anandamide); 2-AG, 2-arachidonoylglycerol; AMT, AEA membrane transporter; CB1, brain-type cannabinoid receptor; CB2, spleen-type cannabinoid receptor; COX, cyclooxygenase; cPLA2α, cytosolic PLA2α; DAG, diacylglycerol; DAGL, DAG lipase; E2, 17β-estradiol; FAAH, fatty acid amide hydrolase; ICM, inner cell mass; INF-γ, interferon-γ; LIF, leukemia inhibitory factor; LPA, lysophosphatidic acid; MAGL, monoacylglycerol lipase; NAPE, N-acylphosphatidylethanolamine; NAT, N-acyltransferase; NK, natural killer; NO, nitric oxide; P4, progesterone; PG, prostaglandin; PL, phospholipase; Th1, type 1 T-helper; Th2, type 2 T-helper; THC, Δ9-tetrahydrocannabinol; TRPV1, transient receptor potential vanilloid 1 (vanilloid receptor); ZP, zona pellucida.

Previous Section

References


Yanagimachi R 1994 Mammalian fertilization in the physiology of reproduction. In: Knobil E, Neill JD, eds. The physiology of reproduction. New York: Raven Press; 189–196

Evans JP, Florman HM 2002 The state of the union: the cell biology of fertilization. Nat Cell Biol 4 Suppl:s57–s63

Wassarman PM, Jovine L, Litscher ES 2001 A profile of fertilization in mammals. Nat Cell Biol 3:E59–E64

Primakoff P, Myles DG 2002 Penetration, adhesion, and fusion in mammalian sperm-egg interaction. Science 296:2183–2185
Abstract/FREE Full Text

Dey SK, Lim H, Das SK, Reese J, Paria BC, Daikoku T, Wang H 2004 Molecular cues to implantation. Endocr Rev 25:341–373
Abstract/FREE Full Text

Paria BC, Reese J, Das SK, Dey SK 2002 Deciphering the cross-talk of implantation: advances and challenges. Science 296:2185–2188
Abstract/FREE Full Text

Rossant J, Tam PP 2004 Emerging asymmetry and embryonic patterning in early mouse development. Dev Cell 7:155–164
CrossRefMedline

Wang H, Dey SK 2006 Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet 7:185–199
CrossRefMedline

Zernicka-Goetz M 2005 Developmental cell biology: cleavage pattern and emerging asymmetry of the mouse embryo. Nat Rev Mol Cell Biol 6:919–928
CrossRefMedline

Smith WL, DeWitt DL, Garavito RM 2000 Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 69:145–182
CrossRefMedline

Wang H, Ma WG, Tejada L, Zhang H, Morrow JD, Das SK, Dey SK 2004 Rescue of female infertility from the loss of cyclooxygenase-2 by compensatory up-regulation of cyclooxygenase-1 is a function of genetic makeup. J Biol Chem 279:10649–10658
Abstract/FREE Full Text

Song H, Lim H, Paria BC, Matsumoto H, Swift LL, Morrow J, Bonventre JV, Dey SK 2002 Cytosolic phospholipase A2α is crucial for ‘on-time' embryo implantation that directs subsequent development. Development 129:2879–2889
Medline

Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos JM, Dey SK 1997 Multiple female reproductive failures in cyclooxygenase 2-deficient mice. Cell 91:197–208
CrossRefMedline

Chakraborty I, Das SK, Wang J, Dey SK 1996 Developmental expression of the cyclo-oxygenase-1 and cyclo-oxygenase-2 genes in the peri-implantation mouse uterus and their differential regulation by the blastocyst and ovarian steroids. J Mol Endocrinol 16:107–122
Abstract/FREE Full Text

Reese J, Paria BC, Brown N, Zhao X, Morrow JD, Dey SK 2000 Coordinated regulation of fetal and maternal prostaglandins directs successful birth and postnatal adaptation in the mouse. Proc Natl Acad Sci USA 97:9759–9764
Abstract/FREE Full Text

Gross GA, Imamura T, Luedke C, Vogt SK, Olson LM, Nelson DM, Sadovsky Y, Muglia LJ 1998 Opposing actions of prostaglandins and oxytocin determine the onset of murine labor. Proc Natl Acad Sci USA 95:11875–11879
Abstract/FREE Full Text

Langenbach R, Morham SG, Tiano HF, Loftin CD, Ghanayem BI, Chulada PC, Mahler JF, Lee CA, Goulding EH, Kluckman KD, Kim HS, Smithies O 1995 Prostaglandin synthase 1 gene disruption in mice reduces arachidonic acid-induced inflammation and indomethacin-induced gastric ulceration. Cell 83:483–492
CrossRefMedline

Yu Y, Cheng Y, Fan J, Chen XS, Klein-Szanto A, Fitzgerald GA, Funk CD 2005 Differential impact of prostaglandin H synthase 1 knockdown on platelets and parturition. J Clin Invest 115:986–995
CrossRefMedline

Matsumoto H, Ma W, Smalley W, Trzaskos J, Breyer RM, Dey SK 2001 Diversification of cyclooxygenase-2-derived prostaglandins in ovulation and implantation. Biol Reprod 64:1557–1565
Abstract/FREE Full Text

Lim H, Gupta RA, Ma WG, Paria BC, Moller DE, Morrow JD, DuBois RN, Trzaskos JM, Dey SK 1999 Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARδ. Genes Dev 13:1561–1574
Abstract/FREE Full Text

Davis BJ, Lennard DE, Lee CA, Tiano HF, Morham SG, Wetsel WC, Langenbach R 1999 Anovulation in cyclooxygenase-2-deficient mice is restored by prostaglandin E2 and interleukin-1β. Endocrinology 140:2685–2695
Abstract/FREE Full Text

Segi E, Haraguchi K, Sugimoto Y, Tsuji M, Tsunekawa H, Tamba S, Tsuboi K, Tanaka S, Ichikawa A 2003 Expression of messenger RNA for prostaglandin E receptor subtypes EP4/EP2 and cyclooxygenase isozymes in mouse periovulatory follicles and oviducts during superovulation. Biol Reprod 68:804–811
Abstract/FREE Full Text

Wang H, Dey SK 2005 Lipid signaling in embryo implantation. Prostaglandins Other Lipid Mediat 77:84–102
CrossRefMedline

Shah BH, Catt KJ 2005 Roles of LPA3 and COX-2 in implantation. Trends Endocrinol Metab 16:397–399
CrossRefMedline

Pall M, Friden BE, Brannstrom M 2001 Induction of delayed follicular rupture in the human by the selective COX-2 inhibitor rofecoxib: a randomized double-blind study. Hum Reprod 16:1323–1328
Abstract/FREE Full Text

Tokuyama O, Nakamura Y, Muso A, Honda K, Ishiko O, Ogita S 2001 Expression and distribution of cyclooxygenase-2 in human periovulatory ovary. Int J Mol Med 8:603–606
Medline

Li DK, Liu L, Odouli R 2003 Exposure to non-steroidal anti-inflammatory drugs during pregnancy and risk of miscarriage: population based cohort study. BMJ 327:368
Abstract/FREE Full Text

Marions L, Danielsson KG 1999 Expression of cyclo-oxygenase in human endometrium during the implantation period. Mol Hum Reprod 5:961–965
Abstract/FREE Full Text

Critchley HO, Jones RL, Lea RG, Drudy TA, Kelly RW, Williams AR, Baird DT 1999 Role of inflammatory mediators in human endometrium during progesterone withdrawal and early pregnancy. J Clin Endocrinol Metab 84:240–248
Abstract/FREE Full Text

Challis JRG, Matthews SG, Gibb W, Lye SJ 2000 Endocrine and paracrine regulation of birth at term and preterm. Endocr Rev 21:514–550
Abstract/FREE Full Text

Slater DM, Zervou S, Thornton S 2002 Prostaglandins and prostanoid receptors in human pregnancy and parturition. J Soc Gynecol Investig 9:118–124

Ishii I, Fukushima N, Ye X, Chun J 2004 Lysophospholipid receptors: signaling and biology. Annu Rev Biochem 73:321–354
CrossRefMedline

Ye X, Hama K, Contos JJ, Anliker B, Inoue A, Skinner MK, Suzuki H, Amano T, Kennedy G, Arai H, Aoki J, Chun J 2005 LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature 435:104–108
CrossRefMedline

Dey SK 2005 Reproductive biology: fatty link to fertility. Nature 435:34–35
CrossRefMedline

Wilcox AJ, Baird DD, Weinberg CR 1999 Time of implantation of the conceptus and loss of pregnancy. N Engl J Med 340:1796–1799
CrossRefMedline

Paria BC, Dey SK 2000 Ligand-receptor signaling with endocannabinoids in preimplantation embryo development and implantation. Chem Phys Lipids 108:211–220
CrossRefMedline

Paria BC, Dey SK 2002 Embryonic cannabinoid receptors are targets for natural and endocannabinoids during early pregnancy. In: Onaivi ES, ed. Biology of marijuana: from gene to behavior. London: Taylor, Francis; 344–355

Paria BC, Wang H, Dey SK 2002 Endocannabinoid signaling in synchronizing embryo development and uterine receptivity for implantation. Chem Phys Lipids 121:201–210
CrossRefMedline

Maccarrone M, Falciglia K, Di Rienzo M, Finazzi-Agro A 2002 Endocannabinoids, hormone-cytokine networks and human fertility. Prostaglandins Leukot Essent Fatty Acids 66:309–317
CrossRefMedline

Maccarrone M, Finazzi-Agro A 2004 Anandamide hydrolase: a guardian angel of human reproduction? Trends Pharmacol Sci 25:353–357
CrossRefMedline

Maccarrone M, Wenger T 2005 Effects of cannabinoids on hypothalamic and reproductive function. Handb Exp Pharmacol 168:555–571
CrossRefMedline

Park B, McPartland JM, Glass M 2004 Cannabis, cannabinoids and reproduction. Prostaglandins Leukot Essent Fatty Acids 70:189–197
CrossRefMedline

Smith CG, Asch RH 1984 Acute, short-term, and chronic effects of marijuana on the female primate reproductive function. NIDA Res Monogr 44:82–96
Medline

Fride E, Shohami E 2002 The endocannabinoid system: function in survival of the embryo, the newborn and the neuron. Neuroreport 13:1833–1841
CrossRefMedline

Habayeb OM, Bell SC, Konje JC 2002 Endogenous cannabinoids: metabolism and their role in reproduction. Life Sci 70:1963–1977
CrossRefMedline

Wenger T, Toth BE, Juaneda C, Leonardelli J, Tramu G 1999 The effects of cannabinoids on the regulation of reproduction. Life Sci 65:695–701
CrossRefMedline

Bloch E, Thysen B, Morrill GA, Gardner E, Fujimoto G 1978 Effects of cannabinoids on reproduction and development. Vitam Horm 36:203–258
Medline

Schuel H, Burkman LJ 2005 A tale of two cells: endocannabinoid-signaling regulates functions of neurons and sperm. Biol Reprod 73:1078–1086
FREE Full Text

Schuel H, Burkman LJ, Lippes J, Crickard K, Forester E, Piomelli D, Giuffrida A 2002 N-Acylethanolamines in human reproductive fluids. Chem Phys Lipids 121:211–227
CrossRefMedline

Kuczkowski KM 2004 Marijuana in pregnancy. Ann Acad Med Singapore 33:336–339
Medline

Nahas GG, Frick HC, Lattimer JK, Latour C, Harvey D 2002 Pharmacokinetics of THC in brain and testis, male gametotoxicity and premature apoptosis of spermatozoa. Hum Psychopharmacol 17:103–113
CrossRefMedline

Kolodny RC, Masters WH, Kolodner RM, Toro G 1974 Depression of plasma testosterone levels after chronic intensive marihuana use. N Engl J Med 290:872–874
Medline

Hembree 3rd WC, Nahas GG, Zeidenberg P, Huang HF 1978 Changes in human spermatozoa associated with high dose marihuana smoking. Adv Biosci 22–23:429–439

Whan LB, West MC, McClure N, Lewis SE 2006 Effects of Δ-9-tetrahydrocannabinol, the primary psychoactive cannabinoid in marijuana, on human sperm function in vitro. Fertil Steril 85:653–660
CrossRefMedline

Hong CY, Chaput de Saintonge DM, Turner P, Fairbairn JW 1982 Comparison of the inhibitory action of Δ-9-tetrahydrocannabinol and petroleum spirit extract of herbal cannabis on human sperm motility. Hum Toxicol 1:151–154
Medline

Issidorides MR 1978 Observations in chronic hashish users: nuclear aberrations in blood and sperm and abnormal acrosomes in spermatozoa. Adv Biosci 22–23:377–388

Qazi QH, Mariano E, Milman DH, Beller E, Crombleholme W 1985 Abnormalities in offspring associated with prenatal marihuana exposure. Dev Pharmacol Ther 8:141–148
Medline

Frank DA, Bauchner H, Parker S, Huber AM, Kyei-Aboagye K, Cabral H, Zuckerman B 1990 Neonatal body proportionality and body composition after in utero exposure to cocaine and marijuana. J Pediatr 117:622–626
CrossRefMedline

Sherwood RA, Keating J, Kavvadia V, Greenough A, Peters TJ 1999 Substance misuse in early pregnancy and relationship to fetal outcome. Eur J Pediatr 158:488–492
CrossRefMedline

Zuckerman B, Frank DA, Hingson R, Amaro H, Levenson SM, Kayne H, Parker S, Vinci R, Aboagye K, Fried LE, et al 1989 Effects of maternal marijuana and cocaine use on fetal growth. N Engl J Med 320:762–768
Medline

Fried PA, Watkinson B, Willan A 1984 Marijuana use during pregnancy and decreased length of gestation. Am J Obstet Gynecol 150:23–27
Medline

Gibson GT, Baghurst PA, Colley DP 1983 Maternal alcohol, tobacco and cannabis consumption and the outcome of pregnancy. Aust N Z J Obstet Gynaecol 23:15–19
Medline

Day N, Sambamoorthi U, Taylor P, Richardson G, Robles N, Jhon Y, Scher M, Stoffer D, Cornelius M, Jasperse D 1991 Prenatal marijuana use and neonatal outcome. Neurotoxicol Teratol 13:329–334
CrossRefMedline

Hatch EE, Bracken MB 1986 Effect of marijuana use in pregnancy on fetal growth. Am J Epidemiol 124:986–993
Abstract/FREE Full Text

Hingson R, Alpert JJ, Day N, Dooling E, Kayne H, Morelock S, Oppenheimer E, Zuckerman B 1982 Effects of maternal drinking and marijuana use on fetal growth and development. Pediatrics 70:539–546
Abstract/FREE Full Text

Klonoff-Cohen HS, Natarajan L, Chen RV 2006 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 194:369–376
CrossRefMedline

Persaud TV, Ellington AC 1967 Cannabis in early pregnancy. Lancet 2:1306
Medline

Geber WF, Schramm LC 1969 Effect of marihuana extract on fetal hamsters and rabbits. Toxicol Appl Pharmacol 14:276–282
CrossRefMedline

Paria BC, Das SK, Dey SK 1995 The preimplantation mouse embryo is a target for cannabinoid ligand-receptor signaling. Proc Natl Acad Sci USA 92:9460–9464
Abstract/FREE Full Text

Persaud TV, Ellington AC 1968 The effects of Cannabis sativa L. (Ganja) on developing rat embryos–preliminary observations. West Indian Med J 17:232–234
Medline

Phillips RN, Turk RF, Forney RB 1971 Acute toxicity of Δ-9-tetrahydrocannabinol in rats and mice. Proc Soc Exp Biol Med 136:260–263
Abstract/FREE Full Text

Rosenkrantz H 1978 Effects of cannabis on fetal development of rodents. Adv Biosci 22–23:479–499

Rosenkrantz H, Grant RJ, Fleischman RW, Baker JR 1986 Marihuana-induced embryotoxicity in the rabbit. Fundam Appl Toxicol 7:236–243
CrossRefMedline

Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M, Mackie K, Martin BR, Mechoulam R, Pertwee RG 2002 International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev 54:161–202
Abstract/FREE Full Text

Pertwee RG, Ross RA 2002 Cannabinoid receptors and their ligands. Prostaglandins Leukot Essent Fatty Acids 66:101–121
CrossRefMedline

McAllister SD, Glass M 2002 CB(1) and CB(2) receptor-mediated signalling: a focus on endocannabinoids. Prostaglandins Leukot Essent Fatty Acids 66:161–171
CrossRefMedline

Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI 1990 Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346:561–564
CrossRefMedline

Munro S, Thomas KL, Abu-Shaar M 1993 Molecular characterization of a peripheral receptor for cannabinoids. Nature 365:61–65
CrossRefMedline

Mechoulam R, Panikashvili D, Shohami E 2002 Cannabinoids and brain injury: therapeutic implications. Trends Mol Med 8:58–61
CrossRefMedline

Maccarrone M, Finazzi-Agro A 2002 Endocannabinoids and their actions. Vitam Horm 65:225–255
Medline

Sugiura T, Kobayashi Y, Oka S, Waku K 2002 Biosynthesis and degradation of anandamide and 2-arachidonoylglycerol and their possible physiological significance. Prostaglandins Leukot Essent Fatty Acids 66:173–192
CrossRefMedline

De Petrocellis L, Cascio MG, Di Marzo V 2004 The endocannabinoid system: a general view and latest additions. Br J Pharmacol 141:765–774
CrossRefMedline

Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R 1992 Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258:1946–1949
Abstract/FREE Full Text

Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, Yamashita A, Waku K 1995 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun 215:89–97
CrossRefMedline

Stella N, Schweitzer P, Piomelli D 1997 A second endogenous cannabinoid that modulates long-term potentiation. Nature 388:773–778
CrossRefMedline

Hanus L, Abu-Lafi S, Fride E, Breuer A, Vogel Z, Shalev DE, Kustanovich I, Mechoulam R 2001 2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci USA 98:3662–3665
Abstract/FREE Full Text

Oka S, Tsuchie A, Tokumura A, Muramatsu M, Suhara Y, Takayama H, Waku K, Sugiura T 2003 Ether-linked analogue of 2-arachidonoylglycerol (noladin ether) was not detected in the brains of various mammalian species. J Neurochem 85:1374–1381
CrossRefMedline

Porter AC, Sauer JM, Knierman MD, Becker GW, Berna MJ, Bao J, Nomikos GG, Carter P, Bymaster FP, Leese AB, Felder CC 2002 Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther 301:1020–1024
Abstract/FREE Full Text

Ben-Shabat S, Fride E, Sheskin T, Tamiri T, Rhee MH, Vogel Z, Bisogno T, De Petrocellis L, Di Marzo V, Mechoulam R 1998 An entourage effect: inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur J Pharmacol 353:23–31
CrossRefMedline

Lambert DM, Di Marzo V 1999 The palmitoylethanolamide and oleamide enigmas: are these two fatty acid amides cannabimimetic? Curr Med Chem 6:757–773
Medline

Hansen HH, Hansen SH, Schousboe A, Hansen HS 2000 Determination of the phospholipid precursor of anandamide and other N-acylethanolamine phospholipids before and after sodium azide-induced toxicity in cultured neocortical neurons. J Neurochem 75:861–871
CrossRefMedline

Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N 2004 Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem 279:5298–5305
Abstract/FREE Full Text

Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB 1996 Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 384:83–87
CrossRefMedline

Giang DK, Cravatt BF 1997 Molecular characterization of human and mouse fatty acid amide hydrolases. Proc Natl Acad Sci USA 94:2238–2242
Abstract/FREE Full Text

Hillard CJ, Jarrahian A 2003 Cellular accumulation of anandamide: consensus and controversy. Br J Pharmacol 140:802–808
CrossRefMedline

Ligresti A, Morera E, Van Der Stelt M, Monory K, Lutz B, Ortar G, Di Marzo V 2004 Further evidence for the existence of a specific process for the membrane transport of anandamide. Biochem J 380:265–272
CrossRefMedline

Oddi S, Bari M, Battista N, Barsacchi D, Cozzani I, Maccarrone M 2005 Confocal microscopy and biochemical analysis reveal spatial and functional separation between anandamide uptake and hydrolysis in human keratinocytes. Cell Mol Life Sci 62:386–395
CrossRefMedline

Moore SA, Nomikos GG, Dickason-Chesterfield AK, Schober DA, Schaus JM, Ying BP, Xu YC, Phebus L, Simmons RM, Li D, Iyengar S, Felder CC 2005 Identification of a high-affinity binding site involved in the transport of endocannabinoids. Proc Natl Acad Sci USA 102:17852–17857
Abstract/FREE Full Text

Mechoulam R, Deutsch DG 2005 Toward an anandamide transporter. Proc Natl Acad Sci USA 102:17541–17542
FREE Full Text

Glaser ST, Kaczocha M, Deutsch DG 2005 Anandamide transport: a critical review. Life Sci 77:1584–1604
CrossRefMedline

Battista N, Gasperi V, Fezza F, Maccarrone M 2005 The anandamide membrane transporter and the therapeutic implications of its inhibition. Therapy 2:141–150
CrossRef

Bracey MH, Hanson MA, Masuda KR, Stevens RC, Cravatt BF 2002 Structural adaptations in a membrane enzyme that terminates endocannabinoid signaling. Science 298:1793–1796
Abstract/FREE Full Text

Maccarrone M, Bari M, Battista N, Finazzi-Agro A 2002 Estrogen stimulates arachidonoylethanolamide release from human endothelial cells and platelet activation. Blood 100:4040–4048
Abstract/FREE Full Text

Ortega-Gutierrez S 2005 Therapeutic perspectives of inhibitors of endocannabinoid degradation. Curr Drug Targets CNS Neurol Disord 4:697–707
CrossRefMedline

Kaczocha M, Hermann A, Glaser S, Bojesen I, Deutsch D 2006 Anandamide uptake is consistent with rate-limited diffusion and is regulated by the degree of its hydrolysis by FAAH. J Biol Chem 281:9066–9075
Abstract/FREE Full Text

McKinney MK, Cravatt BF 2005 Structure and function of fatty acid amide hydrolase. Annu Rev Biochem 74:411–432
CrossRefMedline

van der Stelt M, Di Marzo V 2003 The endocannabinoid system in the basal ganglia and in the mesolimbic reward system: implications for neurological and psychiatric disorders. Eur J Pharmacol 480:133–150
CrossRefMedline

Di Marzo V, Matias I 2005 Endocannabinoid control of food intake and energy balance. Nat Neurosci 8:585–589
CrossRefMedline

Di Marzo V, Bifulco M, De Petrocellis L 2004 The endocannabinoid system and its therapeutic exploitation. Nat Rev Drug Discov 3:771–784
CrossRefMedline

Piomelli D, Giuffrida A, Calignano A, Rodriguez de Fonseca F 2000 The endocannabinoid system as a target for therapeutic drugs. Trends Pharmacol Sci 21:218–224
CrossRefMedline

Piomelli D 2003 The molecular logic of endocannabinoid signalling. Nat Rev Neurosci 4:873–884
CrossRefMedline

Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A, Matias I, Schiano-Moriello A, Paul P, Williams EJ, Gangadharan U, Hobbs C, Di Marzo V, Doherty P 2003 Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J Cell Biol 163:463–468
Abstract/FREE Full Text

Beltramo M, Piomelli D 2000 Carrier-mediated transport and enzymatic hydrolysis of the endogenous cannabinoid 2-arachidonylglycerol. Neuroreport 11:1231–1235
Medline

Hajos N, Kathuria S, Dinh T, Piomelli D, Freund TF 2004 Endocannabinoid transport tightly controls 2-arachidonoyl glycerol actions in the hippocampus: effects of low temperature and the transport inhibitor AM404. Eur J Neurosci 19:2991–2996
CrossRefMedline

Goparaju SK, Ueda N, Taniguchi K, Yamamoto S 1999 Enzymes of porcine brain hydrolyzing 2-arachidonoylglycerol, an endogenous ligand of cannabinoid receptors. Biochem Pharmacol 57:417–423
CrossRefMedline

Lichtman AH, Hawkins EG, Griffin G, Cravatt BF 2002 Pharmacological activity of fatty acid amides is regulated, but not mediated, by fatty acid amide hydrolase in vivo. J Pharmacol Exp Ther 302:73–79
Abstract/FREE Full Text

Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, Kathuria S, Piomelli D 2002 Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci USA 99:10819–10824
Abstract/FREE Full Text

Ho SY, Delgado L, Storch J 2002 Monoacylglycerol metabolism in human intestinal Caco-2 cells: evidence for metabolic compartmentation and hydrolysis. J Biol Chem 277:1816–1823
Abstract/FREE Full Text

Di Marzo V, De Petrocellis L, Fezza F, Ligresti A, Bisogno T 2002 Anandamide receptors. Prostaglandins Leukot Essent Fatty Acids 66:377–391
CrossRefMedline

Caulfield MP, Brown DA 1992 Cannabinoid receptor agonists inhibit Ca current in NG108–15 neuroblastoma cells via a pertussis toxin-sensitive mechanism. Br J Pharmacol 106:231–232
Medline

Mackie K, Hille B 1992 Cannabinoids inhibit N-type calcium channels in neuroblastoma-glioma cells. Proc Natl Acad Sci USA 89:3825–3829
Abstract/FREE Full Text

Mackie K, Lai Y, Westenbroek R, Mitchell R 1995 Cannabinoids activate an inwardly rectifying potassium conductance and inhibit Q-type calcium currents in AtT20 cells transfected with rat brain cannabinoid receptor. J Neurosci 15:6552–6561
Abstract/FREE Full Text

Henry DJ, Chavkin C 1995 Activation of inwardly rectifying potassium channels (GIRK1) by co-expressed rat brain cannabinoid receptors in Xenopus oocytes. Neurosci Lett 186:91–94
CrossRefMedline

Gebremedhin D, Lange AR, Campbell WB, Hillard CJ, Harder DR 1999 Cannabinoid CB1 receptor of cat cerebral arterial muscle functions to inhibit L-type Ca2+ channel current. Am J Physiol 276:H2085–H2093

Wang H, Matsumoto H, Guo Y, Paria BC, Roberts RL, Dey SK 2003 Differential G protein-coupled cannabinoid receptor signaling by anandamide directs blastocyst activation for implantation. Proc Natl Acad Sci USA 100:14914–14919
Abstract/FREE Full Text

Derkinderen P, Toutant M, Kadare G, Ledent C, Parmentier M, Girault JA 2001 Dual role of Fyn in the regulation of FAK+6,7 by cannabinoids in hippocampus. J Biol Chem 276:38289–38296
Abstract/FREE Full Text

Bouaboula M, Poinot-Chazel C, Bourrie B, Canat X, Calandra B, Rinaldi-Carmona M, Le Fur G, Casellas P 1995 Activation of mitogen-activated protein kinases by stimulation of the central cannabinoid receptor CB1. Biochem J 312:637–641
Medline

Bouaboula M, Poinot-Chazel C, Marchand J, Canat X, Bourrie B, Rinaldi-Carmona M, Calandra B, Le Fur G, Casellas P 1996 Signaling pathway associated with stimulation of CB2 peripheral cannabinoid receptor. Involvement of both mitogen-activated protein kinase and induction of Krox-24 expression. Eur J Biochem 237:704–711
Medline

Lauckner JE, Hille B, Mackie K 2005 The cannabinoid agonist WIN55,212–2 increases intracellular calcium via CB1 receptor coupling to Gq/11 G proteins. Proc Natl Acad Sci USA 102:19144–19149
Abstract/FREE Full Text

Baker D, Pryce G, Davies WL, Hiley CR 2006 In silico patent searching reveals a new cannabinoid receptor. Trends Pharmacol Sci 27:1–4
CrossRefMedline

Sawzdargo M, Nguyen T, Lee DK, Lynch KR, Cheng R, Heng HH, George SR, O'Dowd BF 1999 Identification and cloning of three novel human G protein-coupled receptor genes GPR52, PsiGPR53 and GPR55: GPR55 is extensively expressed in human brain. Brain Res Mol Brain Res 64:193–198
Medline

McPartland JM, Matias I, Di Marzo V, Glass M 2006 Evolutionary origins of the endocannabinoid system. Gene 370:64–74
CrossRefMedline

Jung J, Hwang SW, Kwak J, Lee SY, Kang CJ, Kim WB, Kim D, Oh U 1999 Capsaicin binds to the intracellular domain of the capsaicin-activated ion channel. J Neurosci 19:529–538
Abstract/FREE Full Text

Szallasi A, Blumberg PM 1999 Vanilloid (capsaicin) receptors and mechanisms. Pharmacol Rev 51:159–212
Abstract/FREE Full Text

De Petrocellis L, Bisogno T, Maccarrone M, Davis JB, Finazzi-Agro A, Di Marzo V 2001 The activity of anandamide at vanilloid VR1 receptors requires facilitated transport across the cell membrane and is limited by intracellular metabolism. J Biol Chem 276:12856–12863
Abstract/FREE Full Text

Van Der Stelt M, Di Marzo V 2004 Endovanilloids. Putative endogenous ligands of transient receptor potential vanilloid 1 channels. Eur J Biochem 271:1827–1834
Medline

Jordt SE, Julius D 2002 Molecular basis for species-specific sensitivity to “hot†chili peppers. Cell 108:421–430
CrossRefMedline

Maccarrone M, Finazzi-Agro A 2003 The endocannabinoid system, anandamide and the regulation of mammalian cell apoptosis. Cell Death Differ 10:946–955
CrossRefMedline

Yamaji K, Sarker KP, Kawahara K, Iino S, Yamakuchi M, Abeyama K, Hashiguchi T, Maruyama I 2003 Anandamide induces apoptosis in human endothelial cells: its regulation system and clinical implications. Thromb Haemost 89:875–884
Medline

Mezey E, Toth ZE, Cortright DN, Arzubi MK, Krause JE, Elde R, Guo A, Blumberg PM, Szallasi A 2000 Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci USA 97:3655–3660
Abstract/FREE Full Text

Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V 2002 An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci USA 99:8400–8405
Abstract/FREE Full Text

Guo Y, Wang H, Okamoto Y, Ueda N, Kingsley PJ, Marnett LJ, Schmid HH, Das SK, Dey SK 2005 N-Acylphosphatidylethanolamine-hydrolyzing phospholipase D is an important determinant of uterine anandamide levels during implantation. J Biol Chem 280:23429–23432
Abstract/FREE Full Text

Wang J, Paria BC, Dey SK, Armant DR 1999 Stage-specific excitation of cannabinoid receptor exhibits differential effects on mouse embryonic development. Biol Reprod 60:839–844
Abstract/FREE Full Text

Murphy LL, Gher J, Steger RW, Bartke A 1994 Effects of Δ9-tetrahydrocannabinol on copulatory behavior and neuroendocrine responses of male rats to female conspecifics. Pharmacol Biochem Behav 48:1011–1017
CrossRefMedline

Hall W, Solowij N 1998 Adverse effects of cannabis. Lancet 352:1611–1616
CrossRefMedline

Schuel H, Burkman LJ, Lippes J, Crickard K, Mahony MC, Giuffrida A, Picone RP, Makriyannis A 2002 Evidence that anandamide-signaling regulates human sperm functions required for fertilization. Mol Reprod Dev 63:376–387
CrossRefMedline

Schuel H, Chang MC, Berkery D, Schuel R, Zimmerman AM, Zimmerman S 1991 Cannabinoids inhibit fertilization in sea urchins by reducing the fertilizing capacity of sperm. Pharmacol Biochem Behav 40:609–615
CrossRefMedline

Schuel H, Goldstein E, Mechoulam R, Zimmerman AM, Zimmerman S 1994 Anandamide (arachidonylethanolamide), a brain cannabinoid receptor agonist, reduces sperm fertilizing capacity in sea urchins by inhibiting the acrosome reaction. Proc Natl Acad Sci USA 91:7678–7682
Abstract/FREE Full Text

Schuel H, Schuel R, Zimmerman AM, Zimmerman S 1987 Cannabinoids reduce fertility of sea urchin sperm. Biochem Cell Biol 65:130–136
Medline

Chang MC, Berkery D, Laychock SG, Schuel H 1991 Reduction of the fertilizing capacity of sea urchin sperm by cannabinoids derived from marihuana. III. Activation of phospholipase A2 in sperm homogenate by Δ9-tetrahydrocannabinol. Biochem Pharmacol 42:899–904
CrossRefMedline

Chang MC, Berkery D, Schuel R, Laychock SG, Zimmerman AM, Zimmerman S, Schuel H 1993 Evidence for a cannabinoid receptor in sea urchin sperm and its role in blockade of the acrosome reaction. Mol Reprod Dev 36:507–516
CrossRefMedline

Chang MC, Schuel H 1991 Reduction of the fertilizing capacity of sea urchin sperm by cannabinoids derived from marihuana. II. Ultrastructural changes associated with inhibition of the acrosome reaction. Mol Reprod Dev 29:60–71
CrossRefMedline

Zimmerman AM, Bruce WR, Zimmerman S 1979 Effects of cannabinoids on sperm morphology. Pharmacology 18:143–148
Medline

Dalterio S, Badr F, Bartke A, Mayfield D 1982 Cannabinoids in male mice: effects on fertility and spermatogenesis. Science 216:315–316
FREE Full Text

Dalterio S, Steger R, Peluso J, de Paolo L 1987 Acute Δ9-tetrahydrocannabinol exposure: effects on hypothalamic-pituitary-testicular activity in mice. Pharmacol Biochem Behav 26:533–537
CrossRefMedline

Dalterio SL, Bartke A, Mayfield D 1983 Cannabinoids stimulate and inhibit testosterone production in vitro and in vivo. Life Sci 32:605–612
CrossRefMedline

Dalterio S, Bartke A, Burstein S 1977 Cannabinoids inhibit testosterone secretion by mouse testes in vitro. Science 196:1472–1473
Abstract/FREE Full Text

Patra PB, Wadsworth RM 1990 Effect of the synthetic cannabinoid nabilone on spermatogenesis in mice. Experientia 46:852–854
CrossRefMedline

Ambrosini A, Zolese G, Wozniak M, Genga D, Boscaro M, Mantero F, Balercia G 2003 Idiopathic infertility: susceptibility of spermatozoa to in-vitro capacitation, in the presence and the absence of palmitylethanolamide (a homologue of anandamide), is strongly correlated with membrane polarity studied by Laurdan fluorescence. Mol Hum Reprod 9:381–388
Abstract/FREE Full Text

Ambrosini A, Zolese G, Ambrosi S, Bertoli E, Mantero F, Boscaro M, Balercia G 2005 Idiopathic infertility: effect of palmitoylethanolamide (a homologue of anandamide) on hyperactivated sperm cell motility and Ca2+ influx. J Androl 26:429–436
Abstract/FREE Full Text

Sugiura T, Kondo S, Sukagawa A, Tonegawa T, Nakane S, Yamashita A, Waku K 1996 Enzymatic synthesis of anandamide, an endogenous cannabinoid receptor ligand, through N-acylphosphatidylethanolamine pathway in testis: involvement of Ca(2+)-dependent transacylase and phosphodiesterase activities. Biochem Biophys Res Commun 218:113–117
CrossRefMedline

Gye MC, Kang HH, Kang HJ 2005 Expression of cannabinoid receptor 1 in mouse testes. Arch Androl 51:247–255
CrossRefMedline

Wenger T, Ledent C, Csernus V, Gerendai I 2001 The central cannabinoid receptor inactivation suppresses endocrine reproductive functions. Biochem Biophys Res Commun 284:363–368
CrossRefMedline

Newton SC, Murphy LL, Bartke A 1993 In vitro effects of psychoactive and non-psychoactive cannabinoids on immature rat Sertoli cell function. Life Sci 53:1429–1437
CrossRefMedline

Orth JM, McGuinness MP, Qiu J, Jester Jr WF, Li LH 1998 Use of in vitro systems to study male germ cell development in neonatal rats. Theriogenology 49:431–439

Maccarrone M, Cecconi S, Rossi G, Battista N, Pauselli R, Finazzi-Agro A 2003 Anandamide activity and degradation are regulated by early postnatal aging and follicle-stimulating hormone in mouse Sertoli cells. Endocrinology 144:20–28
Abstract/FREE Full Text

O'Bryan MK, Schlatt S, Gerdprasert O, Phillips DJ, de Kretser DM, Hedger MP 2000 Inducible nitric oxide synthase in the rat testis: evidence for potential roles in both normal function and inflammation-mediated infertility. Biol Reprod 63:1285–1293
Abstract/FREE Full Text

Herrero MB, de Lamirande E, Gagnon C 2001 Tyrosine nitration in human spermatozoa: a physiological function of peroxynitrite, the reaction product of nitric oxide and superoxide. Mol Hum Reprod 7:913–921
Abstract/FREE Full Text

Fujisawa M, Yamanaka K, Tanaka H, Tanaka H, Okada H, Arakawa S, Kamidono S 2001 Expression of endothelial nitric oxide synthase in the Sertoli cells of men with infertility of various causes. BJU Int 87:85–88
CrossRefMedline

Bisogno T, Ventriglia M, Milone A, Mosca M, Cimino G, Di Marzo V 1997 Occurrence and metabolism of anandamide and related acyl-ethanolamides in ovaries of the sea urchin Paracentrotus lividus. Biochim Biophys Acta 1345:338–348
Medline

Schuel H, Berkery D, Schuel R, Chang MC, Zimmerman AM, Zimmerman S 1991 Reduction of the fertilizing capacity of sea urchin sperm by cannabinoids derived from marihuana. I. Inhibition of the acrosome reaction induced by egg jelly. Mol Reprod Dev 29:51–59
CrossRefMedline

Fukami K, Yoshida M, Inoue T, Kurokawa M, Fissore RA, Yoshida N, Mikoshiba K, Takenawa T 2003 Phospholipase Cδ4 is required for Ca2+ mobilization essential for acrosome reaction in sperm. J Cell Biol 161:79–88
Abstract/FREE Full Text

Wennemuth G, Babcock DF, Hille B 2003 Calcium clearance mechanisms of mouse sperm. J Gen Physiol 122:115–128
Abstract/FREE Full Text

Gadella BM, Harrison RA 2000 The capacitating agent bicarbonate induces protein kinase A-dependent changes in phospholipid transbilayer behavior in the sperm plasma membrane. Development 127:2407–2420
Abstract

Suarez SS, Ho HC 2003 Hyperactivated motility in sperm. Reprod Domest Anim 38:119–124
CrossRefMedline

Rossato M, Ion Popa F, Ferigo M, Clari G, Foresta C 2005 Human sperm express cannabinoid receptor Cb1, the activation of which inhibits motility, acrosome reaction, and mitochondrial function. J Clin Endocrinol Metab 90:984–991
Abstract/FREE Full Text

Barboni B, Mattioli M, Seren E 1995 Influence of progesterone on boar sperm capacitation. J Endocrinol 144:13–18
Abstract/FREE Full Text

Mattioli M, Barboni B, Lucidi P, Seren E 1996 Identification of capacitation in boar spermatozoa by chlortetracycline staining. Theriogenology 45:373–377
CrossRefMedline

James PS, Hennessy C, Berge T, Jones R 2004 Compartmentalisation of the sperm plasma membrane: a FRAP, FLIP and SPFI analysis of putative diffusion barriers on the sperm head. J Cell Sci 117:6485–6495
Abstract/FREE Full Text

Maccarrone M, Barboni B, Paradisi A, Bernabo N, Gasperi V, Pistilli MG, Fezza F, Lucidi P, Mattioli M 2005 Characterization of the endocannabinoid system in boar spermatozoa and implications for sperm capacitation and acrosome reaction. J Cell Sci 118:4393–4404
Abstract/FREE Full Text

Rossant J 2004 Lineage development and polar asymmetries in the peri-implantation mouse blastocyst. Semin Cell Dev Biol 15:573–581
CrossRefMedline

Cross JC, Werb Z, Fisher SJ 1994 Implantation and the placenta: key pieces of the development puzzle. Science 266:1508–1518
Abstract/FREE Full Text

Norwitz ER, Schust DJ, Fisher SJ 2001 Implantation and the survival of early pregnancy. N Engl J Med 345:1400–1408
CrossRefMedline

Wilcox AJ, Weinberg CR, O'Connor JF, Baird DD, Schlatterer JP, Canfield RE, Armstrong EG, Nisula BC 1988 Incidence of early loss of pregnancy. N Engl J Med 319:189–194
Medline

Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, Yoshinaga K 2000 Embryo implantation. Dev Biol 223:217–237
CrossRefMedline

Red-Horse K, Zhou Y, Genbacev O, Prakobphol A, Foulk R, McMaster M, Fisher SJ 2004 Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface. J Clin Invest 114:744–754
CrossRefMedline

Das SK, Paria BC, Chakraborty I, Dey SK 1995 Cannabinoid ligand-receptor signaling in the mouse uterus. Proc Natl Acad Sci USA 92:4332–4336
Abstract/FREE Full Text

Paria BC, Song H, Wang X, Schmid PC, Krebsbach RJ, Schmid HH, Bonner TI, Zimmer A, Dey SK 2001 Dysregulated cannabinoid signaling disrupts uterine receptivity for embryo implantation. J Biol Chem 276:20523–20528
Abstract/FREE Full Text

Wang H, Guo Y, Wang D, Kingsley PJ, Marnett LJ, Das SK, DuBois RN, Dey SK 2004 Aberrant cannabinoid signaling impairs oviductal transport of embryos. Nat Med 10:1074–1080
CrossRefMedline

Yang ZM, Paria BC, Dey SK 1996 Activation of brain-type cannabinoid receptors interferes with preimplantation mouse embryo development. Biol Reprod 55:756–761
Abstract

Paria BC, Ma W, Andrenyak DM, Schmid PC, Schmid HH, Moody DE, Deng H, Makriyannis A, Dey SK 1998 Effects of cannabinoids on preimplantation mouse embryo development and implantation are mediated by brain-type cannabinoid receptors. Biol Reprod 58:1490–1495
Abstract/FREE Full Text

Rinaldi-Carmona M, Barth F, Heaulme M, Shire D, Calandra B, Congy C, Martinez S, Maruani J, Neliat G, Caput D, Ferrarab P, Soubriea P, Brelierea JC, Le Fur GL 1994 SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett 350:240–244

Rinaldi-Carmona M, Barth F, Millan J, Derocq JM, Casellas P, Congy C, Oustric D, Sarran M, Bouaboula M, Calandra B, Portier M, Shire D, Breliere JC, Le Fur GL 1998 SR 144528, the first potent and selective antagonist of the CB2 cannabinoid receptor. J Pharmacol Exp Ther 284:644–650
Abstract/FREE Full Text

Sharov AA, Piao Y, Matoba R, Dudekula DB, Qian Y, VanBuren V, Falco G, Martin PR, Stagg CA, Bassey UC, Wang Y, Carter MG, Hamatani T, Aiba K, Akutsu H, Sharova L, Tanaka TS, Kimber WL, Yoshikawa T, Jaradat SA, Pantano S, Nagaraja R, Boheler KR, Taub D, Hodes RJ, Longo DL, Schlessinger D, Keller J, Klotz E, Kelsoe G, Umezawa A, Vescovi AL, Rossant J, Kunath T, Hogan BL, Curci A, D'Urso M, Kelso J, Hide W, Ko MS 2003 Transcriptome analysis of mouse stem cells and early embryos. PLoS Biol 1:E74

Zimmer A, Zimmer AM, Hohmann AG, Herkenham M, Bonner TI 1999 Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proc Natl Acad Sci USA 96:5780–5785
Abstract/FREE Full Text

Jarai Z, Wagner JA, Varga K, Lake KD, Compton DR, Martin BR, Zimmer AM, Bonner TI, Buckley NE, Mezey E, Razdan RK, Zimmer A, Kunos G 1999 Cannabinoid-induced mesenteric vasodilation through an endothelial site distinct from CB1 or CB2 receptors. Proc Natl Acad Sci USA 96:14136–14141
Abstract/FREE Full Text

Farquhar CM 2005 Ectopic pregnancy. Lancet 366:583–591
CrossRefMedline

Pisarska MD, Carson SA, Buster JE 1998 Ectopic pregnancy. Lancet 351:1115–1120
CrossRefMedline

Heilman RD, Reo RR, Hahn DW 1976 Changes in the sensitivity of adrenergic receptors in the oviduct during early gestation in the rabbit. Fertil Steril 27:426–430
Medline

Howe GR, Black DL 1973 Autonomic nervous system and oviduct function in the rabbit. I. Hormones and contraction. J Reprod Fertil 33:425–430
Abstract/FREE Full Text

Kennedy DR, Marshall JM 1977 Effect of adrenergic nerve stimulation on the rabbit oviduct: correlation with norepinephrine content and turnover rate. Biol Reprod 16:200–211
Abstract

Paria BC, Huet-Hudson YM, Dey SK 1993 Blastocyst's state of activity determines the “window†of implantation in the receptive mouse uterus. Proc Natl Acad Sci USA 90:10159–10162
Abstract/FREE Full Text

Paria BC, Lim H, Wang XN, Liehr J, Das SK, Dey SK 1998 Coordination of differential effects of primary estrogen and catecholestrogen on two distinct targets mediates embryo implantation in the mouse. Endocrinology 139:5235–5246
Abstract/FREE Full Text

Das SK, Wang XN, Paria BC, Damm D, Abraham JA, Klagsbrun M, Andrews GK, Dey SK 1994 Heparin-binding EGF-like growth factor gene is induced in the mouse uterus temporally by the blastocyst solely at the site of its apposition: a possible ligand for interaction with blastocyst EGF-receptor in implantation. Development 120:1071–1083
Abstract

Yoshinaga K, Adams CE 1966 Delayed implantation in the spayed, progesterone treated adult mouse. J Reprod Fertil 12:593–595
Abstract/FREE Full Text

McLaren 1971 A Blastocysts in the mouse uterus: the effect of ovariectomy, progesterone and oestrogen. J Endocrinol 50:515–526
Abstract/FREE Full Text

Schmid PC, Paria BC, Krebsbach RJ, Schmid HH, Dey SK 1997 Changes in anandamide levels in mouse uterus are associated with uterine receptivity for embryo implantation. Proc Natl Acad Sci USA 94:4188–4192
Abstract/FREE Full Text

Liu WM, Duan EK, Cao YJ 2002 Effects of anandamide on embryo implantation in the mouse. Life Sci 71:1623–1632
CrossRefMedline

Habayeb OM, Taylor AH, Evans MD, Cooke MS, Taylor DJ, Bell SC, Konje JC 2004 Plasma levels of the endocannabinoid anandamide in women—a potential role in pregnancy maintenance and labor? J Clin Endocrinol Metab 89:5482–5487
Abstract/FREE Full Text

Di Marzo V, Fontana A, Cadas H, Schinelli S, Cimino G, Schwartz JC, Piomelli D 1994 Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 372:686–691
CrossRefMedline

Maccarrone M, DeFelici M, Klinger FG, Battista N, Fezza F, Dainese E, Siracusa G, Finazzi-Agro A 2004 Mouse blastocysts release a lipid which activates anandamide hydrolase in intact uterus. Mol Hum Reprod 10:215–221
Abstract/FREE Full Text

Paria BC, Deutsch DD, Dey SK 1996 The uterus is a potential site for anandamide synthesis and hydrolysis: differential profiles of anandamide synthase and hydrolase activities in the mouse uterus during the periimplantation period. Mol Reprod Dev 45:183–192
CrossRefMedline

Paria BC, Zhao X, Wang J, Das SK, Dey SK 1999 Fatty-acid amide hydrolase is expressed in the mouse uterus and embryo during the periimplantation period. Biol Reprod 60:1151–1157
Abstract/FREE Full Text

Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C, Finazzi-Agro A 2000 Relation between decreased anandamide hydrolase concentrations in human lymphocytes and miscarriage. Lancet 355:1326–1329
CrossRefMedline

Maccarrone M, Bisogno T, Valensise H, Lazzarin N, Fezza F, Manna C, Di Marzo V, Finazzi-Agro A 2002 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 8:188–195
Abstract/FREE Full Text

Kessler CA, Moghadam KK, Schroeder JK, Buckley AR, Brar AK, Handwerger S 2005 Cannabinoid receptor I activation markedly inhibits human decidualization. Mol Cell Endocrinol 229:65–74
CrossRefMedline

Breivogel CS, Griffin G, Di Marzo V, Martin BR 2001 Evidence for a new G protein-coupled cannabinoid receptor in mouse brain. Mol Pharmacol 60:155–163
Abstract/FREE Full Text

Bari M, Battista N, Fezza F, Gasperi V, Maccarrone M 2006 New insights into endocannabinoid degradation and its therapeutic potential. Mini Rev Med Chem 6:257–268
CrossRefMedline

Piccinni MP, Beloni L, Livi C, Maggi E, Scarselli G, Romagnani S 1998 Defective production of both leukemia inhibitory factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat Med 4:1020–1024
CrossRefMedline

Sharkey A 1998 Cytokines and implantation. Rev Reprod 3:52–61
Abstract

Szekeres-Bartho J, Faust Z, Varga P, Szereday L, Kelemen K 1996 The immunological pregnancy protective effect of progesterone is manifested via controlling cytokine production. Am J Reprod Immunol 35:348–351
Medline

Chaouat G, Menu E, Delage G, Moreau JF, Khrishnan L, Hui L, Meliani AA, Martal J, Raghupathy R, Lelaidier C, Bertrand C, Freitas S, Hambartsumian E, Wegmann TG, Frydman R 1995 Immuno-endocrine interactions in early pregnancy. Hum Reprod 10(Suppl 2):55–59

Stewart CL, Cullinan EB 1997 Preimplantation development of the mammalian embryo and its regulation by growth factors. Dev Genet 21:91–101
CrossRefMedline

Duval D, Reinhardt B, Kedinger C, Boeuf H 2000 Role of suppressors of cytokine signaling (Socs) in leukemia inhibitory factor (LIF)-dependent embryonic stem cell survival. FASEB J 14:1577–1584
Abstract/FREE Full Text

Taupin JL, Minvielle S, Thèze J, Jacques Y, Moreau JF 1999 The interleukin-6 family of cytokines and their receptors. In: Thèze J, ed. The cytokine network and immune functions. New York: Oxford University Press; 31–44

Cravatt BF, Lichtman AH 2003 Fatty acid amide hydrolase: an emerging therapeutic target in the endocannabinoid system. Curr Opin Chem Biol 7:469–475
CrossRefMedline

Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C, Finazzi-Agro A 2001 Progesterone up-regulates anandamide hydrolase in human lymphocytes: role of cytokines and implications for fertility. J Immunol 166:7183–7189
Abstract/FREE Full Text

Lazzarin N, Valensise H, Bari M, Ubaldi F, Battista N, Finazzi-Agro A, Maccarrone M 2004 Fluctuations of fatty acid amide hydrolase and anandamide levels during the human ovulatory cycle. Gynecol Endocrinol 18:212–218
CrossRefMedline

Piccinni MP, Romagnani S 1996 Regulation of fetal allograft survival by a hormone-controlled Th1- and Th2-type cytokines. Immunol Res 15:141–150
Medline

Maccarrone M, Bari M, Di Rienzo M, Finazzi-Agro A, Rossi A 2003 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 278:32726–32732
Abstract/FREE Full Text

Ahima RS, Flier JS2000 Leptin. Annu Rev Physiol 62:413–437

Matarese G, La Cava A, Sanna V, Lord GM, Lechler RI, Fontana S, Zappacosta S 2002 Balancing susceptibility to infection and autoimmunity: a role for leptin? Trends Immunol 23:182–187
CrossRefMedline

Maccarrone M, Di Rienzo M, Finazzi-Agro A, Rossi A 2003 Leptin activates the anandamide hydrolase promoter in human T lymphocytes through STAT3. J Biol Chem 278:13318–13324
Abstract/FREE Full Text

MacCarrone M, De Felici M, Bari M, Klinger F, Siracusa G, Finazzi-Agro A 2000 Down-regulation of anandamide hydrolase in mouse uterus by sex hormones. Eur J Biochem 267:2991–2997
Medline

Maccarrone M, Fride E, Bisogno T, Bari M, Cascio MG, Battista N, Finazzi Agro A, Suris R, Mechoulam R, Di Marzo V 2005 Up-regulation of the endocannabinoid system in the uterus of leptin knockout (ob/ob) mice and implications for fertility. Mol Hum Reprod 11:21–28
Abstract/FREE Full Text

Kline J, Stein Z, Susser ME 1989 Conception to birth: epidemiology of prenatal development. New York: Oxford University Press

Sozio J, Ness RB 1998 Chlamydial lower genital tract infection and spontaneous abortion. Infect Dis Obstet Gynecol 6:8–12
CrossRefMedline

Walsh RA 1994 Effects of maternal smoking on adverse pregnancy outcomes: examination of the criteria of causation. Hum Biol 66:1059–1092
Medline

Ness RB, Grisso JA, Hirschinger N, Markovic N, Shaw LM, Day NL, Kline J 1999 Cocaine and tobacco use and the risk of spontaneous abortion. N Engl J Med 340:333–339
CrossRefMedline

Iversen L 2003 Cannabis and the brain. Brain 126:1252–1270
Abstract/FREE Full Text

Di Carlo G, Izzo AA 2003 Cannabinoids for gastrointestinal diseases: potential therapeutic applications. Expert Opin Investig Drugs 12:39–49
Medline

Fowler CJ 2004 Possible involvement of the endocannabinoid system in the actions of three clinically used drugs. Trends Pharmacol Sci 25:59–61
CrossRefMedline

Tsuboi K, Sun YX, Okamoto Y, Araki N, Tonai T, Ueda N 2005 Molecular characterization of N-acylethanolamine-hydrolyzing acid amidase, a novel member of the choloylglycine hydrolase family with structural and functional similarity to acid ceramidase. J Biol Chem 280:11082–11092
Abstract/FREE Full Text

Bifulco M, Di Marzo V 2002 Targeting the endocannabinoid system in cancer therapy: a call for further research. Nat Med 8:547–550
CrossRefMedline

Guzman M 2003 Cannabinoids: potential anticancer agents. Nat Rev Cancer 3:745–755
CrossRefMedline

Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, Mor M, Tarzia G, La Rana G, Calignano A, Giustino A, Tattoli M, Palmery M, Cuomo V, Piomelli D 2003 Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med 9:76–81
CrossRefMedline

Freund TF, Katona I, Piomelli D 2003 Role of endogenous cannabinoids in synaptic signaling. Physiol Rev 83:1017–1066
Abstract/FREE Full Text

van der Stelt M, Veldhuis WB, Maccarrone M, Bar PR, Nicolay K, Veldink GA, Di Marzo V, Vliegenthart JF 2002 Acute neuronal injury, excitotoxicity, and the endocannabinoid system. Mol Neurobiol 26:317–346
CrossRefMedline

Wilson RI, Nicoll RA 2002 Endocannabinoid signaling in the brain. Science 296:678–682
Abstract/FREE Full Text

Hamatani T, Carter MG, Sharov AA, Ko MS 2004 Dynamics of global gene expression changes during mouse preimplantation development. Dev Cell 6:117–131
CrossRefMedline

Hamatani T, Daikoku T, Wang H, Matsumoto H, Carter MG, Ko MS, Dey SK 2004 Global gene expression analysis identifies molecular pathways distinguishing blastocyst dormancy and activation. Proc Natl Acad Sci USA 101:10326–10331
Abstract/FREE Full Text

Ko MS, Kitchen JR, Wang X, Threat TA, Wang X, Hasegawa A, Sun T, Grahovac MJ, Kargul GJ, Lim MK, Cui Y, Sano Y, Tanaka T, Liang Y, Mason S, Paonessa PD, Sauls AD, DePalma GE, Sharara R, Rowe LB, Eppig J, Morrell C, Doi H 2000 Large-scale cDNA analysis reveals phased gene expression patterns during preimplantation mouse development. Development 127:1737–1749
Abstract

Carter MG, Hamatani T, Sharov AA, Carmack CE, Qian Y, Aiba K, Ko NT, Dudekula DB, Brzoska PM, Hwang SS, Ko MS 2003 In situ-synthesized novel microarray optimized for mouse stem cell and early developmental expression profiling. Genome Res 13:1011–1021
Abstract/FREE Full Text

Carter MG, Piao Y, Dudekula DB, Qian Y, VanBuren V, Sharov AA, Tanaka TS, Martin PR, Bassey UC, Stagg CA, Aiba K, Hamatani T, Matoba R, Kargul GJ, Ko MS 2003 The NIA cDNA project in mouse stem cells and early embryos. C R Biol 326:931–940
Medline

Wang QT, Piotrowska K, Ciemerych MA, Milenkovic L, Scott MP, Davis RW, Zernicka-Goetz M 2004 A genome-wide study of gene activity reveals developmental signaling pathways in the preimplantation mouse embryo. Dev Cell 6:133–144
CrossRefMedline

Zeng F, Baldwin DA, Schultz RM 2004 Transcript profiling during preimplantation mouse development. Dev Biol 272:483–496
CrossRefMedline

Cravatt BF, Saghatelian A, Hawkins EG, Clement AB, Bracey MH, Lichtman AH 2004 Functional disassociation of the central and peripheral fatty acid amide signaling systems. Proc Natl Acad Sci USA 101:10821–10826
Abstract/FREE Full Text

Fernandez-Solari J, Scorticati C, Mohn C, De Laurentiis A, Billi S, Franchi A, McCann SM, Rettori V 2004 Alcohol inhibits luteinizing hormone-releasing hormone release by activating the endocannabinoid system. Proc Natl Acad Sci USA 101:3264–3268
Abstract/FREE Full Text

Gammon CM, Freeman Jr GM, Xie W, Petersen SL, Wetsel WC 2005 Regulation of gonadotropin-releasing hormone secretion by cannabinoids. Endocrinology 146:4491–4499
Abstract/FREE Full Text

Scorticati C, Fernandez-Solari J, De Laurentiis A, Mohn C, Prestifilippo JP, Lasaga M, Seilicovich A, Billi S, Franchi A, McCann SM, Rettori V 2004 The inhibitory effect of anandamide on luteinizing hormone-releasing hormone secretion is reversed by estrogen. Proc Natl Acad Sci USA 101:11891–11896
Abstract/FREE Full Text

Kozak KR, Crews BC, Morrow JD, Wang LH, Ma YH, Weinander R, Jakobsson PJ, Marnett LJ 2002 Metabolism of the endocannabinoids, 2-arachidonylglycerol and anandamide, into prostaglandin, thromboxane, and prostacyclin glycerol esters and ethanolamides. J Biol Chem 277:44877–44885
Abstract/FREE Full Text

Yu M, Ives D, Ramesha CS 1997 Synthesis of prostaglandin E2 ethanolamide from anandamide by cyclooxygenase-2. J Biol Chem 272:21181–21186
Abstract/FREE Full Text

van der Stelt M, van Kuik JA, Bari M, van Zadelhoff G, Leeflang BR, Veldink GA, Finazzi-Agro A, Vliegenthart JF, Maccarrone M 2002 Oxygenated metabolites of anandamide and 2-arachidonoylglycerol: conformational analysis and interaction with cannabinoid receptors, membrane transporter, and fatty acid amide hydrolase. J Med Chem 45:3709–3720
CrossRefMedline

Ueda N, Yamamoto K, Yamamoto S, Tokunaga T, Shirakawa E, Shinkai H, Ogawa M, Sato T, Kudo I, Inoue K, Takizawa H, Nagano T, Hirobe M, Matsuki N, Saito H 1995 Lipoxygenase-catalyzed oxygenation of arachidonylethanolamide, a cannabinoid receptor agonist. Biochim Biophys Acta 1254:127–134
Medline

Maccarrone M, Salvati S, Bari M, Finazzi A 2000 Anandamide and 2-arachidonoylglycerol inhibit fatty acid amide hydrolase by activating the lipoxygenase pathway of the arachidonate cascade. Biochem Biophys Res Commun 278:576–583
CrossRefMedline

Dalterio S, Bartke A, Roberson C, Watson D, Burstein S 1978 Direct and pituitary-mediated effects of Δ9-THC and cannabinol on the testis. Pharmacol Biochem Behav 8:673–678
CrossRefMedline

Wenger T, Rettori V, Snyder GD, Dalterio S, McCann SM 1987 Effects of Δ-9-tetrahydrocannabinol on the hypothalamic-pituitary control of luteinizing hormone and follicle-stimulating hormone secretion in adult male rats. Neuroendocrinology 46:488–493
Medline

Chakravarty I, Sheth PR, Sheth AR, Ghosh JJ 1982 Δ-9-Tetrahydrocannabinol: its effect on hypothalamo-pituitary system in male rats. Arch Androl 8:25–27
Medline

Murphy LL, Adrian BA, Kohli M 1999 Inhibition of luteinizing hormone secretion by Δ9-tetrahydrocannabinol in the ovariectomized rat: effect of pretreatment with neurotransmitter or neuropeptide receptor antagonists. Steroids 64:664–671
CrossRefMedline

Steger RW, Murphy LL, Bartke A, Smith MS 1990 Effects of psychoactive and nonpsychoactive cannabinoids on the hypothalamic-pituitary axis of the adult male rat. Pharmacol Biochem Behav 37:299–302
CrossRefMedline

Marks BH 1973 Δ1-Tetrahydrocannabinol and luteinizing hormone secretion. Prog Brain Res 39:331–338
Medline

Symons AM, Teale JD, Marks V 1976 Proceedings: Effect of Δ9-tetrahydrocannabinol on the hypothalamic-pituitary-gonadal system in the maturing male rat. J Endocrinol 68:43P–44P
Abstract/FREE Full Text

Collu R, Letarte J, Leboeuf F, Ducharme JR 1975 Endocrine effects of chronic administration of psychoactive drugs to prepuberal male rats. I: Δ9-tetrahydrocannabinol. Life Sci 16:533–542
Medline

Smith CG, Moore CE, Besch NF, Besch PK 1976 The effect of marihuana Δ-9-tetrahydrocannabinol on the secretion of sex hormones in the mature male rhesus monkey. Clin Chem 22:1184

Kramer J, Ben-David M 1974 Suppression of prolactin secretion by acute administration of Δ9-THC in rats. Proc Soc Exp Biol Med 147:482–484
Abstract/FREE Full Text

Dalterio SL, Michael SD, Macmillan BT, Bartke A 1981 Differential effects of cannabinoid exposure and stress on plasma prolactin, growth hormone and corticosterone levels in male mice. Life Sci 28:761–766
CrossRefMedline

Asch RH, Smith CG, Siler-Khodr TM, Pauerstein CJ 1979 Acute decreases in serum prolactin concentrations caused by Δ 9-tetrahydrocannabinol in nonhuman primates. Fertil Steril 32:571–575
Medline

List A, Nazar B, Nyquist S, Harclerode J 1977 The effects of Δ9-tetrahydrocannabinol and cannabidiol on the metabolism of gonadal steroids in the rat. Drug Metab Dispos 5:268–272
Abstract

Harclerode J, Nyquist SE, Nazar B, Lowe D 1978 Effects of cannabis on sex hormones and testicular enzymes of the rodent. Adv Biosci 22-23:395–405

Rosenkrantz H, Esber HJ 1980 Cannabinoid-induced hormone changes in monkeys and rats. J Toxicol Environ Health 6:297–313
Medline

Fujimoto GI, Morrill GA, O'Connell ME, Kostellow AB, Retura G 1982 Effects of cannabinoids given orally and reduced appetite on the male rat reproductive system. Pharmacology 24:303–313
Medline

Thompson GR, Fleischman RW, Rosenkrantz H, Braude MC 1974 Oral and intravenous toxicity of Δ9-tetrahydrocannabinol in rhesus monkeys. Toxicol Appl Pharmacol 27:648–665
CrossRefMedline

Maskarinec MP, Shipley G, Novotny M, Brown DJ, Forney RB 1978 Different effects of synthetic Δ9-tetrahydrocannabinol and cannabis extract on steroid metabolism in male rats. Experientia 34:88–89
CrossRefMedline

Rosenkrantz H, Fleischman RW, Grant RJ 1981 Toxicity of short-term administration of cannabinoids to rhesus monkeys. Toxicol Appl Pharmacol 58:118–131
CrossRefMedline

Dixit VP, Gupta CL, Agrawal M 1977 Testicular degeneration and necrosis induced by chronic administration of cannabis extract in dogs. Endokrinologie 69:299–305
Medline

Rosenkrantz H, Hayden DW 1979 Acute and subacute inhalation toxicity of Turkish marihuana, cannabichromene, and cannabidiol in rats. Toxicol Appl Pharmacol 48:375–386
CrossRefMedline

Dixit VP, Jain HC, Verma OP, Sharma AN 1977 Effects of cannabis extract on the testicular function of the toad Bufo andersonii Boulenger. Indian J Exp Biol 15:555–557
Medline

Dixit VP, Sharma VN, Lohiya NK 1974 The effect of chronically administered cannabis extract on the testicular function of mice. Eur J Pharmacol 26:111–114
CrossRefMedline

Vyas DK, Singh R 1976 Effect of cannabis, opium on the testis of the pigeon Columba livia Gmelin. Indian J Exp Biol 14:22–25
Medline

Okey AB, Truant GS 1975 Cannabis demasculinizes rats but is not estrogenic. Life Sci 17:1113–1117
CrossRefMedline

Zimmerman AM, Zimmerman S, Raj AY 1978 Effects of cannabinoids on spermatogenesis in mice. Adv Biosci 22-23:407–418

Huang HF, Nahas GG, Hembree 3rd WC 1978 Effects of marihuana inhalation on spermatogenesis of the rat. Adv Biosci 22-23:419–427

Tilak SK, Zimmerman AM 1984 Effects of cannabinoids on macromolecular synthesis in isolated spermatogenic cells. Pharmacology 29:343–350
CrossRefMedline

Merari A, Barak A, Plaves M 1973 Effects of 1(2) -tetrahydrocannabinol on copulation in the male rat. Psychopharmacologia 28:243–246
CrossRefMedline

Corcoran ME, Amit Z, Malsbury CW, Daykin S 1974 Reduction in copulatory behavior of male rats following hashish injections. Res Commun Chem Pathol Pharmacol 7:779–782
Medline

Cone EJ, Johnson RE, Moore JD, Roache JD 1986 Acute effects of smoking marijuana on hormones, subjective effects and performance in male human subjects. Pharmacol Biochem Behav 24:1749–1754
CrossRefMedline

Vescovi PP, Pedrazzoni M, Michelini M, Maninetti L, Bernardelli F, Passeri M 1992 Chronic effects of marihuana smoking on luteinizing hormone, follicle-stimulating hormone and prolactin levels in human males. Drug Alcohol Depend 30:59–63
CrossRefMedline

Harmon J, Aliapoulios MA 1972 Gynecomastia in marihuana users. N Engl J Med 287:936
Medline

Smith CG, Besch NF, Smith RG, Besch PK 1979 Effect of tetrahydrocannabinol on the hypothalamic-pituitary axis in the ovariectomized rhesus monkey. Fertil Steril 31:335–339
Medline

Ayalon D, Nir I, Cordova T, Bauminger S, Puder M, Naor Z, Kashi R, Zor U, Harell A, Lindner HR 1977 Acute effect of Δ1-tetrahydrocannabinol on the hypothalamo-pituitary-ovarian axis in the rat. Neuroendocrinology 23:31–42
Medline

Dalterio SL, Mayfield DL, Bartke A 1983 Effects of Δ 9-THC on plasma hormone levels in female mice. Subst Alcohol Actions Misuse 4:339–345
Medline

Nir I, Ayalon D, Tsafriri A, Cordova T, Lindner HR 1973 Letter: Suppression of the cyclic surge of luteinizing hormone secretion and of ovulation in the rat by Δ 1-tetrahydrocannabinol. Nature 243:470–471
CrossRefMedline

Tyrey L 1978 Δ-9-Tetrahydrocannabinol suppression of episodic luteinizing hormone secretion in the ovariectomized rat. Endocrinology 102:1808–1814
Abstract/FREE Full Text

Smith CG, Smith MT, Besch NF, Smith RG, Asch RH 1978 Effect of Δ 9-tetrahydrocannabinol (THC) on female reproductive function. Adv Biosci 22-23:449–467

Jackson AL, Murphy LL 1997 Role of the hypothalamic-pituitary-adrenal axis in the suppression of luteinizing hormone release by Δ-9-tetrahydrocannabinol. Neuroendocrinology 65:446–452
Medline

Chakravarty I, Sheth AR, Ghosh JJ 1975 Effect of acute Δ9-tetrahydrocannabinol treatment on serum luteinizing hormone and prolactin levels in adult female rats. Fertil Steril 26:947–948
Medline

Tyrey L 1992 Δ-9-Tetrahydrocannabinol attenuates luteinizing hormone release induced by electrochemical stimulation of the medial preoptic area. Biol Reprod 47:262–267
Abstract

Besch NF, Smith CG, Besch PK, Kaufman RH 1977 The effect of marihuana (Δ-9-tetrahydrocannabinol) on the secretion of luteinizing hormone in the ovariectomized rhesus monkey. Am J Obstet Gynecol 128:635–642
Medline

Hughes Jr CL, Everett JW, Tyrey L 1981 Δ 9-Tetrahydrocannabinol suppression of prolactin secretion in the rat: lack of direct pituitary effect. Endocrinology 109:876–880
Abstract/FREE Full Text

Chakravarty I, Shah PG, Sheth AR, Ghosh JJ 1979 Mode of action of Δ-9-tetrahydrocannabinol on hypothalamo-pituitary function in adult female rats. J Reprod Fertil 57:113–115
Abstract/FREE Full Text

Almirez RG, Smith CG, Asch RH 1983 The effects of marijuana extract and Δ 9-tetrahydrocannabinol on luteal function in the rhesus monkey. Fertil Steril 39:212–217
Medline

Kostellow AB, Ziegler D, Kunar J, Fujimoto GI, Morrill GA 1980 Effect of cannabinoids on estrous cycle, ovulation and reproductive capacity of female A/J mice. Pharmacology 21:68–75
Medline

Asch RH, Smith CG, Siler-Khodr TM, Pauerstein CJ 1981 Effects of Δ 9-tetrahydrocannabinol during the follicular phase of the rhesus monkey (Macaca mulatta). J Clin Endocrinol Metab 52:50–55
Abstract/FREE Full Text

Asch RH, Fernandez EO, Smith CG, Pauerstein CJ 1979 Precoital single doses of Δ9-tetrahydrocannabinol block ovulation in the rabbit. Fertil Steril 31:331–334
Medline

Dixit VP, Arya M, Lohiya NK 1975 The effect of chronically administered cannabis extract on the female genital tract of mice and rats. Endokrinologie 66:365–368
Medline

Wenger T, Croix D, Tramu G 1988 The effect of chronic prepubertal administration of marihuana (Δ-9-tetrahydrocannabinol) on the onset of puberty and the postpubertal reproductive functions in female rats. Biol Reprod 39:540–545
Abstract

Field E, Tyrey L 1984 Delayed sexual maturation in the female rat during chronic exposure to Δ-9-tetrahydrocannabinol. Life Sci 35:1725–1730
CrossRefMedline

Mani SK, Mitchell A, O'Malley BW 2001 Progesterone receptor and dopamine receptors are required in Δ 9-tetrahydrocannabinol modulation of sexual receptivity in female rats. Proc Natl Acad Sci USA 98:1249–1254
Abstract/FREE Full Text

Gordon JH, Bromley BL, Gorski RA, Zimmermann E 1978 Δ9-Tetrahydrocannabinol enhancement of lordosis behavior in estrogen treated female rats. Pharmacol Biochem Behav 8:603–608
CrossRefMedline

Turley Jr WA, Floody OR 1981 Δ-9-Tetrahydrocannabinol stimulates receptive and proceptive sexual behaviors in female hamsters. Pharmacol Biochem Behav 14:745–747
CrossRefMedline

Asch RH, Smith CG 1986 Effects of Δ 9-THC, the principal psychoactive component of marijuana, during pregnancy in the rhesus monkey. J Reprod Med 31:1071–1081
Medline

Sassenrath EN, Chapman LF, Goo GP 1978 Reproduction in rhesus monkeys chronically exposed to Δ-9-tetrahydrocannabinol. Adv Biosci 22-23:501–512

Abel EL, Tan SE 1987 Effects of Δ-9-tetrahydrocannabinol, chlordiazepoxide (Librium), and their combination on pregnancy and offspring in mice [correction of rats]. Reprod Toxicol 1:37–40
CrossRefMedline

Harbison RD, Mantilla-Plata B 1972 Prenatal toxicity, maternal distribution and placental transfer of tetrahydrocannabinol. J Pharmacol Exp Ther 180:446–453
Abstract/FREE Full Text

Abel EL, Tan SE, Subramanian M 1987 Effects of Δ 9-tetrahydrocannabinol, phenobarbital, and their combination on pregnancy and offspring in rats. Teratology 36:193–198
CrossRefMedline

Wenger T, Fragkakis G, Giannikou P, Probonas K, Yiannikakis N 1997 Effects of anandamide on gestation in pregnant rats. Life Sci 60:2361–2371
CrossRefMedline

Tyrey L, Murphy LL 1988 Inhibition of suckling-induced milk ejections in the lactating rat by Δ 9-tetrahydrocannabinol. Endocrinology 123:469–472
Abstract/FREE Full Text

Mendelson JH, Mello NK, Ellingboe J, Skupny AS, Lex BW, Griffin M 1986 Marihuana smoking suppresses luteinizing hormone in women. J Pharmacol Exp Ther 237:862–866
Abstract/FREE Full Text

Mendelson JH, Mello NK 1984 Effects of marijuana on neuroendocrine hormones in human males and females. NIDA Res Monogr 44:97–114
Medline

Mendelson JH, Mello NK, Ellingboe J 1985 Acute effects of marihuana smoking on prolactin levels in human females. J Pharmacol Exp Ther 232:220–222
Abstract/FREE Full Text

Bauman J 1980 Marijuana and the female reproductive system. In: Health consequence of marijuana use. Washington DC: U.S. Government Printing Office; 85–97

Greenland S, Staisch KJ, Brown N, Gross SJ 1982 Effects of marijuana on human pregnancy, labor, and delivery. Neurobehav Toxicol Teratol 4:447–450
Medline

Greenland S, Staisch KJ, Brown N, Gross SJ 1982 The effects of marijuana use during pregnancy. I. A preliminary epidemiologic study. Am J Obstet Gynecol 143:408–413
Medline

Greenland S, Richwald GA, Honda GD 1983 The effects of marijuana use during pregnancy. II. A study in a low-risk home-delivery population. Drug Alcohol Depend 11:359–366


Source: Jekyll and Hyde: Two Faces of Cannabinoid Signaling in Male and Female Fertility
 
Back
Top Bottom