The Emerging Role Of The Endocannabinoid System In Endocrine Regulation And Energy

Jacob Bell

New Member
The Emerging Role of the Endocannabinoid System in Endocrine Regulation and Energy Balance

Uberto Pagotto,
Giovanni Marsicano,
Daniela Cota,
Beat Lutz and
Renato Pasquali

- Author Affiliations

Endocrinology Unit, Department of Internal Medicine and Gastroenterology, and Center for Applied Biomedical Research (U.P., R.P.), Sant' Orsola-Malpighi Hospital, 40138 Bologna, Italy; Department of Physiological Chemistry (G.M., B.L.), Johannes Gutenberg-University Mainz, 55099 Mainz, Germany; and University of Cincinnati, Department of Psychiatry, Obesity Research Center, Genome Research Institute (D.C.), Cincinnati, Ohio 45237

Address all correspondence and requests for reprints to: Uberto Pagotto, M.D., Ph.D., Endocrinology Unit and Center for Applied Biomedical Research, Department of Internal Medicine and Gastroenterology, Sant' Orsola-Malpighi Hospital, Via Massarenti, 9, 40138 Bologna, Italy. E-mail: pagube@med.unibo.it


Next Section
Abstract

During the last few years, the endocannabinoid system has emerged as a highly relevant topic in the scientific community. Many different regulatory actions have been attributed to endocannabinoids, and their involvement in several pathophysiological conditions is under intense scrutiny. Cannabinoid receptors, named CB1 receptor and CB2 receptor, first discovered as the molecular targets of the psychotropic component of the plant Cannabis sativa, participate in the physiological modulation of many central and peripheral functions. CB2 receptor is mainly expressed in immune cells, whereas CB1 receptor is the most abundant G protein-coupled receptor expressed in the brain. CB1 receptor is expressed in the hypothalamus and the pituitary gland, and its activation is known to modulate all the endocrine hypothalamic-peripheral endocrine axes. An increasing amount of data highlights the role of the system in the stress response by influencing the hypothalamic-pituitary-adrenal axis and in the control of reproduction by modifying gonadotropin release, fertility, and sexual behavior.

The ability of the endocannabinoid system to control appetite, food intake, and energy balance has recently received great attention, particularly in the light of the different modes of action underlying these functions. The endocannabinoid system modulates rewarding properties of food by acting at specific mesolimbic areas in the brain. In the hypothalamus, CB1 receptor and endocannabinoids are integrated components of the networks controlling appetite and food intake. Interestingly, the endocannabinoid system was recently shown to control metabolic functions by acting on peripheral tissues, such as adipocytes, hepatocytes, the gastrointestinal tract, and, possibly, skeletal muscle. The relevance of the system is further strenghtened by the notion that drugs interfering with the activity of the endocannabinoid system are considered as promising candidates for the treatment of various diseases, including obesity.

I. Introduction

II. The Endocannabinoid System

A. Cannabinoid receptors

B. Endocannabinoids

C. Cannabinoid agonists

D. Cannabinoid type 1 receptor antagonists

III. Exogenous and Endogenous Cannabinoids and Their Role in Endocrine Regulation

A. Cannabinoids and the hypothalamic-pituitary-adrenal axis

B. The role of cannabinoids in GH secretion

C. Cannabinoids and the hypothalamic-pituitary-thyroid axis

D. The role of cannabinoids in prolactin secretion

E. The role of cannabinoids in the modulation of the hypothalamic-pituitary-gonadal axis and fertility

IV. Endocannabinoid System in the Modulation of Energy Balance

A. Animal studies before the discovery of endocannabinoids

B. Studies in humans with exogenous cannabinoids before the discovery of endocannabinoids

C. Endocannabinoid functions at mesolimbic level to regulate rewarding properties of food

D. The endocannabinoid system as a new hypothalamic player in the regulation of food intake

E. The peripheral effect of the endocannabinoid system in the modulation of metabolic functions

F. Oleoylethanolamide: a new anorectic fatty acid amide

V. Cannabinoid Receptor Antagonists as New Pharmacological Tools to Tackle Obesity and Obesity-Related Diseases

A. Emerging issues in the treatment of obesity and related diseases by cannabinoid antagonists

B. Clinical trial studies with rimonabant, the first CB1 receptor antagonist in clinical use to tackle obesity and obesity-related diseases

VI. Summary and Perspectives

Previous SectionNext Section
I. Introduction

THE FIRST STEPS in the discovery of the endocannabinoid system date back almost 4000 yr, when the therapeutic and psychotropic actions of the plant Cannabis sativa were first documented in India (1). Over the last 40 yr, after Gaoni and Mechoulam (2) purified the psychoactive component from hemp, a stunning amount of research has revealed the endocannabinoid system as a central modulatory system in animal physiology.

Elements of the endocannabinoid system comprise the cannabinoid receptors, the endogenous lipid ligands (endocannabinoids), and the machinery for their biosynthesis and metabolism (3, 4). Despite public concern related to the abuse of marijuana and its derivatives, the research on the endocannabinoid system has recently aroused enormous interest not only for the physiological functions, but also for the promising therapeutic potentials of drugs interfering with the activity of cannabinoid receptors. This review aims to provide an overview on the pivotal role of the endocannabinoid system in the modulation of the neuroendocrine and peripheral endocrine systems. Moreover, in the context of the recently proposed therapeutic applications of cannabinoid receptor antagonists in the treatment of obesity, the key role of the endocannabinoid system in the control of eating behavior, food intake, and energy metabolism will be discussed in the light of the recent data obtained from human and animal studies.
Previous SectionNext Section
II. The Endocannabinoid System

The large and widespread medical, religious, and recreational use of marijuana throughout the ages was apparently not sufficient to initiate careful and extensive research on cannabinoids until the last few decades of the 20th century. Conversely, the political antimarijuana attitude in the United States and the consequent prohibition in the 1930s did not help to encourage scientific interest on this topic. In the 1960s, the growing public concern regarding the potential negative healthy effects of cannabinoids associated with the exponential increase in its recreational use forced governmental institutions to invest resources to understand the modes of action of marijuana and the pathophysiological implications of its use in more detail. Cannabinoid research received a pivotal boost from the characterization of the chemical structure of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive constituent of marijuana (2). This finding paved the way to the understanding of marijuana's mechanisms of action and, many years later, to the cloning of the two receptor subtypes that are able to bind exogenous cannabinoids, named cannabinoid receptor type 1 (CB1 receptor) and type 2 (CB2 receptor), respectively, and to the identification of their endogenous ligands: the endocannabinoids (5, 6, 7, 8, 9). Cannabinoid receptors, endocannabinoids, and the machinery for their synthesis and degradation represent the elements of a novel endogenous signaling system (the so-called endocannabinoid system), which is involved in a plethora of physiological functions (3, 4). During the last few years, an overwhelming amount of data has been acquired to understand the biological roles of this system in more detail. However, many questions are still open, and promising new discoveries await us in the near future.

In general, the endocannabinoid system is involved in many different physiological functions, many of which relate to stress-recovery systems and to the maintenance of homeostatic balance (10). Among other functions, the endocannabinoid system is involved in neuroprotection (11, 12, 13), modulation of nociception (14), regulation of motor activity (15), and the control of certain phases of memory processing (16, 17, 18). In addition, the endocannabinoid system is involved in modulating the immune and inflammatory responses (19, 20, 21). It also influences the cardiovascular and respiratory systems by controlling heart rate, blood pressure, and bronchial functions (22). Finally, yet importantly, endocannabinoids are known to exert important antiproliferative actions in tumor cells (23). A full discussion of the plethora of functions of the endocannabinoid system in maintaining homeostasis is beyond the scope and space of the present review. However, the reviews cited in this article will further help to obtain a broad insight into the physiological roles of the endocannabinoid system.
A. Cannabinoid receptors

Two cannabinoid receptors have been identified and molecularly characterized so far, namely the seven transmembrane G protein-coupled cannabinoid receptor type 1 (CB1 receptor) (6) and type 2 (CB2 receptor) (7). CB1 receptor was originally described as the “brain type†cannabinoid receptor, because its levels of expression were high in the brain (24). However, recent studies attribute new sites of action of endocannabinoids to many peripheral organs through CB1 receptor activation. The generalization for CB1 receptor being the eminent “brain type†receptor is therefore no longer appropriate. Conversely, CB2 receptors are present almost exclusively in immune and blood cells, where they may participate in regulating immune responses (25). However, CB2 receptors also exert functions in nonimmune cells such as keratinocytes (26). Pharmacological evidence exists for the presence of other cannabinoid receptors, which, however, have not yet been cloned (27). The endocannabinoid anandamide is also able to bind to and activate vanilloid receptors, transient receptor potential vanilloid type 1 (28), and to inhibit TASK-1 K+ channels (29). Moreover, pharmacological studies indicate that still unidentified additional cannabinoid receptors might exist in the hippocampus, modulating the release of glutamate (30), and on endothelial cells (31). Two patents have been recently published claiming that a number of cannabinoid ligands also bind to GPR55, an orphan G protein-coupled receptor, suggesting that this receptor might represent a novel target of cannabinoid action (32). CB1 receptor, however, is the best characterized target of exogenous and endogenous cannabinoids in the modulation of neuroendocrine and metabolic responses, and this review will focus mainly on this receptor.
1. CB1 receptor expression in the brain.

Cannabinoid receptor distribution was studied by means of autoradiography of ligand-receptor binding on slide-mounted rat brain sections (24, 33), by in situ hybridization (ISH) (34, 35, 36), by autoradiography in human brain (37), by immunohistochemistry (IHC) (38, 39, 40, 41), and by agonist-stimulated [35S]GTPγS binding to slide-mounted sections (42, 43). Expression studies showed very early that CB1 receptor is one of the most abundant G protein-coupled receptors in the mammalian brain (24). CB1 receptors are widely expressed in the brain, including the olfactory bulb, cortical regions (neocortex, pyriform cortex, hippocampus, and amygdala), several parts of basal ganglia, thalamic and hypothalamic nuclei, cerebellar cortex, and brainstem nuclei. The levels of expression vary considerably among the various brain regions and neuronal subpopulations. For instance, agonist-mediated receptor binding revealed high densities of CB1 receptor protein in the cornu ammonis pyramidal cell layers of the hippocampus (24), which was later shown by IHC to be due to a dense plexus of immunoreactive fibers deriving from γ-aminobutyric acid (GABA)-ergic interneurons and surrounding the cell bodies of pyramidal cells, which appear per se to be devoid of CB1 receptor protein (38, 41, 44). However, pyramidal cells of the hippocampus and other cortical regions do express low but significant levels of CB1 receptor mRNA (34, 36), indicating the possibility that CB1 receptor protein in these cells is localized on distal projections and/or is expressed at low levels, which are below the limit of detectability with currently available immunohistochemical methods. A similar situation is present also in other cortical regions, such as the amygdala, neocortex, entorhinal cortex, and piriform cortex.

In subcortical regions, CB1 receptor is present at relatively high levels in the septal region (lateral and medial septum, and vertical and horizontal nuclei of the diagonal band). Lower levels of expression are present in hypothalamic regions, such as the medial and lateral preoptic nucleus, magnocellular preoptic nucleus, and paraventricular nucleus (PVN) (36). In the caudal hypothalamus, CB1 receptor is expressed in the premammillary nucleus. In the lateral hypothalamus, CB1 receptor is present in scattered cells (34, 36). In the PVN, CB1 receptor mRNA coexpresses with CRH mRNA (45). In the thalamus, CB1 receptor is present in the lateral habenula, reticular thalamic nucleus, and zona incerta. Midbrain dopaminergic neurons are generally considered to lack CB1 receptor expression. However, recent observations indicate that very low levels of CB1 receptor might be present in tyrosine hydroxylase-expressing neurons in the ventral tegmental area (VTA) (46) and in dopaminergic terminals in the striatum (47). In the hindbrain, apart from the molecular and granular layers of cerebellum expressing high levels of the receptor, CB1 receptor is present at low levels in some nuclei of the brain stem, such as the periaqueductal gray (34, 38). Functional mapping by agonist-stimulated [35S]GTPγS binding using different CB1 receptor agonists revealed that cannabinoid activation of G proteins occurs with the same regional distribution as the receptors (43, 48). However, in some regions, the ratio between the estimated amount of CB1 receptor and G protein activation is not always constant, thus indicating regional differences in receptor-coupling efficiencies (43). This is important to consider, because sometimes the endocannabinoid system seems to influence functions involving regions where the density of CB1 receptor is relatively low (e.g., modulation of food intake in the hypothalamic area). Therefore, the activity of cannabinoids on CB1 receptor cannot be predicted based solely on the relative receptor density, but other factors, such as receptor coupling efficiency, should be taken into account. For instance, by using conditional mutagenesis in mice, the relatively low levels of CB1 receptor expression in cortical pyramidal neurons were recently shown to play a central role in the endocannabinoid-mediated protection against excitotoxic seizures (12). In conclusion, CB1 receptor is widely expressed in the brain and is present at different levels in different neuronal subpopulation and brain regions, and there is apparently no strict correlation between levels of expression and receptor functionality.
2. CB1 receptor expression in the pituitary.

Early studies showed a scattered presence of CB1 receptor in both lobes of the rodent pituitary (33). Recent studies examined the distribution of CB1 receptor mRNA in the anterior pituitary lobe in more detail. In 1999, the abundant CB1 receptor presence in the rat adenohypophysis was associated with the ability of this gland to synthesize endocannabinoids (49). CB1 receptor was also shown to be present in prolactin (PRL)- and LH-secreting cells of the rat pituitary (50). CB1 receptor expression was also detected by means of double-immunofluorescence in the pituitary gland of Xenopus laevis, where the receptor was found in lactotrophs, gonadotrophs, and thyrotrophs (51). The expression of CB1 receptor in the human pituitary appears to be substantially different from the localization of the same receptor described in rodents and frogs (52). By using ISH and double IHC, CB1 receptor was localized in the majority of corticotrophs and somatotrophs of the normal human anterior lobe; only a small percentage of the PRL-secreting cells are positive for CB1 receptor, whereas no immunoreactivity was found in LH-, FSH-, or TSH-positive cells. The neural lobe is devoid of CB1 receptor immunoreactivity (52). Interestingly, folliculo-stellate cells are also positive for CB1 receptor, although functional data have not yet been associated with this expression (52). CB1 receptor was also found in human pituitary adenomas, such as ACTH-producing adenomas (which give rise to Cushing's syndrome), GH-producing tumors (leading to acromegaly), and in prolactinomas, whereas no CB1 receptor staining was found in so-called nonfunctioning pituitary adenomas, tumors expressing LH and/or FSH, and/or α-subunit being devoid of any hormonal staining (52). These data were confirmed by a study in which cDNA microarray analysis was used to compare gene expression pattern in pituitary adenomas vs. normal pituitary (53). Among other genes differentially expressed, ACTH- and GH-producing tumors express higher levels of CB1 receptor compared with the normal pituitary (53). Notably, the human normal anterior pituitary gland and pituitary tumors were shown to be capable of synthesizing endocannabinoids (52).

In rodents, CB1 receptor expression in the pituitary is under the influence of circulating sex hormones, as demonstrated by the ability of androgens and estrogens to up- and down-regulate CB1 receptor, respectively (49). In agreement with these findings, decreased CB1 receptor expression has been found in estrogen-induced pituitary hyperplasia in rats (49). Accordingly, in rats, the male pituitary displays higher levels of CB1 receptor mRNA than the female one (49). In contrast, the human pituitary does not show this gender difference (52).

Exogenous cannabinoids can modulate the expression of CB1 receptor in the pituitary. After a transient down-regulation of the receptor (first 1–3 d), chronic administration of CB1 receptor agonists is able to produce a consistent increase of CB1 receptor expression in the anterior pituitary lobe (after 14 d) (54). This finding seems to be in contrast with the level in the ventromedial hypothalamic nucleus, where CB1 receptor mRNA was down-regulated by chronic CB1 receptor agonist treatment (54).
3. CB1 receptor expression in the peripheral organs
a. CB1 receptor in the thyroid gland.

CB1 receptor expression during the late embryological stages of the rat thyroid was found to be very high (55), whereas lower but still detectable levels of CB1 receptor mRNA and protein were present in the adult rat gland distributed in both follicular and parafollicular cells as demonstrated by IHC (56).
b. CB1 receptor in the adrenal gland.

A faint signal for CB1 receptor was detected in the human adrenal glands by quantitative RT-PCR method (57). However, ISH or IHC studies are needed to clearly localize CB1 receptor in the different areas that make up the gland.
c. CB1 receptor in the peripheral organs involved in metabolic control.

In 2003, two independent groups found the presence of CB1 receptor in adipocytes of mice and humans (58, 59, 60). In both species, this expression is more evident in mature adipocytes than in preadipocytes (59, 60), indicating that the full cellular machinery of the fat cell is needed to exert cannabinoid action. Little is known about CB1 receptor expression in the muscle. Recently, the CB1 receptor antagonist SR141716 was shown to directly affect glucose uptake in the isolated soleus muscle of genetically obese mice (61). Consistently, CB1 receptor is present in the murine soleus muscle as shown by RT-PCR (Fig. 1⇓). Additional investigations are needed to fully understand the importance of this expression site.
Fig. 1.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 1.

Expression of CB1 mRNA in soleus muscle of mice. RT-PCR was performed using 1 μg of total RNA extracted by phenol-chloroform method from soleus muscle as shown in Ref.58 . β-actin and CB1 mRNA expression. Lane 1, Wild-type 12-wk-old mice undergoing standard diet; lane 2, CB1−/− littermate mice undergoing standard diet; lane 3, C56BL/6 mice used as control undergoing standard diet; lane 4, C56BL/6 under high-fat diet for 2 months; lane +, positive control (hypothalamus); and lane W, negative control (PCR blank). Note the increased CB1 signal in muscle derived from mice on high-fat diet in comparison to the muscle derived from mice on standard diet.

Recently, an elegant study by Kunos' group (62) localized CB1 receptor in the mouse liver. CB1 receptor mRNA was detected by ISH with strong labeling in Kupffer cells, whereas lower levels of expression were found in hepatocytes and endothelial cells. Interestingly, CB1 receptor expression was more prominent in hepatocytes surrounding the central veins. Human hepatic stellate cells also have been shown to express CB1 receptor (63).

At present, nothing is known about CB1 receptor in the exocrine and endocrine cells of the pancreas.
d. CB1 in the gastrointestinal tract.

The endocannabinoid system is present in the gastrointestinal tract where it modulates several functions, including motility, inflammation, and secretion (64). Interestingly, CB1 receptor is expressed in vagal nerve terminals innervating the gastrointestinal tract (64), which are involved in gut-brain signaling, modulating food intake. They express cholecystokinin (CCK) receptor type 1 whose activation is known to play a very important role in mediating satiety. Vagal neurons are known to express receptors for leptin and orexin-A (65, 66), whose ligands activate and reduce the anorectic effect of CCK on vagal afferent nerve discharge, respectively. Importantly, CB1 receptor is also present in these neurons, and its expression is decreased after feeding and enhanced in fasting conditions (67). CCK was shown to mediate the effect of food in down-regulating vagal CB1 receptor expression (67). CB1 receptor was also found in the fundus of the stomach, but the cellular localization is not yet known. However, a single SR141716 administration is able to reduce the levels of ghrelin (68), whose production takes place in the gastric endocrine (X-) cells (69).
e. CB1 receptor in the reproductive organs.

CB1 receptor has been known for a long time to be expressed in the testis (57, 70). In particular, it seems to be localized in Leydig cells (71), whereas Sertoli cells that are able to inactivate arachidonoyl ethanolamide (AEA) do not express CB1 receptor (72). Sea urchin sperms, an ideal model for studying fertilization processes, express cannabinoid binding sites (73). Human sperms possess functional binding sites for cannabinoids (74). Very recently, Rossato et al. (75) elegantly showed that CB1 receptor is present in the head and the middle piece of human sperm.

CB1 receptor is also expressed in the ovary (57), probably located in the granulosa cell layer where Δ9-THC was shown to inhibit cAMP accumulation (76). CB1 receptor is present in the mouse uterus (77) and in the human myometrium (78), and is associated with the relaxant effect of cannabinoid receptor agonists (78). Importantly, CB1 receptor is coexpressed with β-adrenergic receptors in the oviduct muscularis, where the endocannabinoid system regulates motility and embryo transport (79). Both CB1 and CB2 receptors are located in the mouse preimplantation embryos (80) as well as in all layers of human placenta; particularly high levels are detectable in the amniotic epithelium and in the maternal decidua layer (81).
4. Signal transduction of CB1 receptor.

The signal transduction of cannabinoid receptors has been extensively described in many excellent reviews (3, 4, 25, 82, 83, 84, 85), and its detailed description is beyond the scope of the present article. It is important to note, however, that CB1 receptor activation might lead to the stimulation of different intracellular pathways, depending on the cell type involved and the experimental conditions. For instance, CB1 receptor, which normally inhibits adenylate cyclase, can also stimulate the cAMP pathway in particular conditions (86, 87). Moreover, recent results suggest the possibility of functional interactions of CB1 receptors with other receptors, for instance, with type 1 orexin receptors (88), 5HT2 serotonin receptors (89), and dopamine receptor type 2 (D2) (87). The possibility that such interactions depend on heterooligomerization processes might represent a very interesting novel aspect (87), which will expand the view of the pharmacology and physiology of the endocannabinoid system. These considerations should also be borne in mind to understand the roles of the endocannabinoid system in regulating the endocrine systems. Figure 2⇓ summarizes the best-described intracellular effects of CB1 receptor stimulation, including the regulation of the cAMP cascade, modulation of ion channels, stimulation of kinase pathways, and induction of immediate early genes.
Fig. 2.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 2.

Schematic representation of the main effects of CB1 on intracellular signaling cascades. Activation of CB1 leads to the stimulation of Gi/o proteins that, in turn, inhibits the adenylate cyclase-mediated conversion of ATP to cAMP. cAMP molecules can bind the regulatory subunits of protein kinase A (PKA) and cause the liberation of the catalytic subunits. Activated PKA can phosphorylate A-type potassium (K+A) channels, causing a decrease of the current. Given the negative effect of CB1 on adenylate cyclase, the final result is an activation of K+A channels. Gi/o activated by CB1 can also directly inhibit N- or P/Q-type Ca2+ channels and activate inwardly rectifying potassium (Kir) channels. These last two effects are controlled by protein kinase C (PKC), which, after activation, can phosphorylate CB1 in the third cytoplasmatic loop and uncouple the receptor from the ion channels. Activation of CB1 can also stimulate several intracellular kinases, such as focal adhesion kinase (FAK), phosphatidyl inositol-3-kinase (PI3-K) and its downstream effector protein kinase B (PKB)/AKT, ERKs, c-Jun N-terminal kinase (c-JNK), and p38 MAPK (p38). Stimulation of cytoplasmic kinases could also mediate the CB1-induced expression of the immediate early genes (IEG), such as the transcription factors c-fos, c-jun, and zif268, and the brain-derived neurotrophic factor (BDNF). Note that these events were described in different cellular systems and, therefore, they might not occur in the same cell types.
B. Endocannabinoids
1. Structure.

In 1992, the first endogenous cannabinoid, AEA, also called anandamide, was identified (8). Subsequently, a second endocannabinoid, 2-arachidonoyl glycerol (2-AG), was discovered (5, 9). Both these compounds are derivatives of arachidonic acid and are able to bind to CB1 and CB2 receptors, although with differences in affinities and activation efficacies (90). During the last few years, several other bioactive lipid mediators have been described; they appear to act, at least in part, through CB1 and/or CB2 receptors and confer specific pharmacological effects in vivo (91). Specifically, these compounds are 2-arachidonoyl-glyceryl-ether (noladin ether) (92), O-arachidonoyl-ethanolamine (virodhamine) (93), N-arachidonoyl-dopamine (94), and possibly oleamide (95). However, the endogenous function in physiological processes for all these latter compounds have not yet been established in detail and need further investigation (4). Furthermore, there are several additional putative lipid mediators that might have cannabimimetic actions, but whose exact mechanism of action is not known in detail (91). In some cases, their cannabimimetic effects may be partially attributed to interference with the endocannabinoid-inactivating enzymes (91). These lipids might, therefore, be able to enhance the activity of cannabinoid receptors by increasing the concentration of the endocannabinoids such as AEA and/or 2-AG.
2. Synthesis, release, uptake, and degradation of endocannabinoids: on demand activation of the endocannabinoid system.

Endocannabinoids are very lipophilic and thus cannot be stored in vesicles like other neurotransmitters. Consequently, the regulation of endocannabinoid signaling is tightly controlled by their synthesis, release, uptake, and degradation (3). Several different stimuli, including membrane depolarization and increased intracellular Ca2+ and/or receptor stimulation, can activate complex enzymatic machineries, which lead to the cleavage of membrane phospholipids and eventually to the synthesis of endocannabinoids. Importantly, different enzymes are involved in the synthesis of distinct endocannabinoids, indicating an independent involvement of endocannabinoids in different conditions. After synthesis, endocannabinoids can activate cannabinoid receptors, either after previous release into the extracellular space or directly moving within the cell membrane. Endocannabinoid signaling is limited by very efficient degradation processes, involving facilitated uptake from the extracellular space into the cell and enzymatic catabolism mediated by specific intracellular enzymes. The molecular nature of the carrier protein(s) involved in endocannabinoid uptake has not yet been elucidated. However, the enzymes able to degrade endocannabinoids are quite well characterized. They are fatty acid amide hydrolase (FAAH) for anandamide and related compounds (96) and monoglycerol lipase for 2-AG (97), although other enzymes might be partially involved in the degradation of this last compound (98). A detailed description of the biochemical mechanisms leading to the synthesis, release, uptake, and degradation of endocannabinoids is beyond the scope of the present article, and we refer the reader to several excellent and exhaustive reviews recently published on the subject (3, 4, 30, 82, 99, 100, 101). An interesting aspect of endocannabinoid activity is the rapid induction of their synthesis, receptor activation, and degradation (3, 102). The endocannabinoid system has thus been suggested to act on demand, with a tightly regulated spatial and temporal selectivity. The system exerts its modulatory actions only when and where it is needed. This fact poses an important distinction between the physiological functions of the endocannabinoid system (selective in time and space) and the pharmacological actions of exogenous cannabinoid receptor agonists, which lack such selectivity. In the context of endocrine regulation, it is interesting to mention here that hormonal stimulation with glucocorticoids can lead to the synthesis of endocannabinoids in the hypothalamus through rapid nongenomic mechanisms (103). It was also recently shown that phospholipase Cβ represents an intracellular coincidence detector of membrane depolarization and receptor stimulation leading to the synthesis and, possibly, the release of endocannabinoids in the hippocampus (104). These data reveal a novel mechanism for activation of the endocannabinoid system, which could also be involved in the regulation of endocrine systems. Concerning degradation of endocannabinoids, which represents an important regulatory aspect of the activity of the endocannabinoid system, it should also be mentioned that a recent study investigated whether endocytic processes are involved in the uptake of endocannabinoids and found that about half of the AEA uptake occurs via a caveola/lipid raft-related process (105).
3. Endocannabinoid-mediated inter- and intracellular signaling.

Several mechanisms underlying endocannabinoid-mediated signaling have been reported. 1) In the central nervous system (CNS), endocannabinoids can act as neurotransmitters transferring information from one neuron to the next. Here, postsynaptically released endocannabinoids travel to the presynaptic site where they activate CB1 receptors. They thus mediate a retrograde signal (30, 106, 107). The overall effect is a decrease in the release of neurotransmitters such as glutamate and GABA. This phenomenon is present in synaptic connections of many brain regions, thus representing an important modulatory mechanism of neuronal transmission. With respect to the aims of the present review, it is noteworthy that this function has also been shown in the VTA (108, 109), where the modulation of reward properties of food presumably occur, and in the hypothalamus, where endocannabinoids and CB1 receptor mediate the acute glucocorticoid-dependent depression of glutamatergic transmission (103). 2) Endocannabinoids can mediate an autocrine signaling that induces a self-inhibitory effect on neuronal activity. This was shown for GABAergic neurons in the cerebral cortex (110). 3) Endocannabinoids may act in a paracrine or autocrine manner, not involving synaptic transmission. This is presumably applicable for glial cells (111) and in nonneuronal cells such as the adipocytes and the hepatocytes. 4) Because endocannabinoids and CB1 receptor are also present within the cell, it cannot be excluded that endocannabinoids may act as intracellular signaling molecules. Importantly, AEA and 2-AG do not appear as interchangeable mediators. For instance, electrophysiological and biochemical evidence shows that 2-AG is mostly involved in retrograde control of synaptic activity in the VTA (109), or the hippocampus (112), whereas AEA appears to play an important role in other regions, such as the basal ganglia (113) and the amygdala (114).

In summary, endocannabinoids appear to be very versatile signaling mediators, involved in a broad spectrum of physiological regulatory processes.
C. Cannabinoid agonists
1. Plant-derived cannabinoids.

The isolation and characterization of the psychoactive component of C. sativa represented a challenging research task. This was due to the fact that the extracts from Cannabis plants contain more than 60 different, chemically closely related terpeno-phenols that are difficult to separate and purify. This prevented the isolation of pure crystals for determination of the structure. The breakthrough was achieved using improved column chromatography. As mentioned above, in the early 1960s, Gaoni and Mechoulam (2) succeeded in isolating and pharmacologically characterizing various plant-derived cannabinoids. In hemp, the major psychoactive compound is represented by Δ9-THC, whereas Δ8-tetrahydrocannabinol is only present in very low amounts. The majority of terpeno-phenols in hemp lack psychoactivity. They include cannabidiol, cannabinol, cannabigerol, and cannabichromene. Although psychoactive cannabinoids bind to and activate both CB1 and CB2 cannabinoid receptors, nonpsychoactive cannabinoids are also able to exert various pharmacological effects in vivo, although only at rather high concentrations and not by activation of CB1 or CB2 receptors. Cannabidiol has recently gained additional attention due to its anticonvulsive, neuroprotective, and antiemetic activities (115, 116, 117). The underlying mechanisms of actions of this plant-derived cannabinoid have not yet been elucidated.
2. Classification of exogenous and endogenous cannabinoids.

Based on structural features, plant-derived and synthetic cannabinoids are divided into different classes (25). In brief: 1) For “classic†cannabinoids, the main psychoactive constituent of Cannabis, Δ9-THC, encompasses tricyclic dibenzopyran compounds and serves as the lead structure. Δ9-THC is a partial agonist of CB1 and CB2 receptors. The synthetic derivative HU210 shows the highest potency among the known CB1 receptor agonists and also activates CB2 receptors (25). HU308, another synthetic Δ9-THC derivative, was found to be a selective CB2 receptor agonist (118). 2) So-called “nonclassic†cannabinoids are synthetic Δ9-THC derivatives that lack the dihydropyran ring. The most famous one is represented by CP-55,940, a potent and complete agonist of CB1 and CB2 receptors, synthesized by Pfizer. It was originally pivotal for the molecular identification of CB1 receptor (25). 3) Finally, aminoalkylindoles, represented by R-(+)-WIN-55,212-2, are compounds structurally unrelated to Δ9-THC but with strong cannabimimetic activities (25). They bind to both CB1 and CB2 receptors (25).

All endocannabinoids are structurally rather distinct from plant-derived and most synthetic cannabinoids. Prototypically, they belong to the eicosanoids, fatty acid derivatives containing a chain with 20 carbon atoms. The synthetic AEA derivative arachidonyl-2′-chloroethylamide represents a selective CB1 receptor agonist with very low activity on CB2 receptor (25).

The quest for specific ligands for either of the cannabinoid receptors represents an important research topic. In particular, if CB2 receptor is targeted with a specific agonist, with no activity on CB1 receptor, the psychotropic side effects of the agonist are avoided. This may be very relevant for alleviating peripheral pain where CB2 receptor is involved (26, 119). Further important progress may also be achieved by the development of cannabinoid receptor agonists that do not pass the blood-brain barrier. Such compounds would focus on the receptors in the periphery and would thus prevent undesirable side effects originating from the CNS.

Although not acting as ligands of cannabinoid receptors, inhibitors of cellular uptake of endocannabinoids, such as AM404 (120), VDM11 (121), and UCM707 (122) provide another interesting class of drugs interfering with the endocannabinoid system. Given the on demand nature of the synthesis and release of endocannabinoids, these drugs make it possible to induce a targeted increase in the concentration of endocannabinoids, likely reducing some of the undesirable side effects observed by using receptor agonists.
D. Cannabinoid type 1 receptor antagonists

Pharmacological investigations have placed emphasis on the generation of substances acting as specific antagonists of cannabinoid receptors. Among the increasing number of compounds sharing CB1 receptor antagonistic properties (123, 124), the compounds most characterized are SR141716 (125), SR14778 (126), AM251 (124), AM281 (127), LY320135 (128), and SLV319 (129). The CB1 receptor antagonists known so far are diarylpyrazoles, or aminoalkylindoles, or triazole derivatives. Diarylpyrazoles include SR141716, which is the first selective CB1 receptor antagonist reported. It was discovered approximately a decade ago, and it has been the compound most studied so far. Pharmacologically, SR141716 shows a Ki value of binding to rat brain synaptosome of 1.98 ± 0.36 nm (125). Few data on the metabolism and pharmacokinetics of SR141716 are available in humans (130). The dose of SR141716 that produced a 50% antagonism of agonist effect in the mouse was 0.23 mg/kg, and a dose of 3 mg/kg produces a long-lasting (18 h) blockade of the effect of WIN-55212–3 (131).

There are different possible mechanisms by which CB1 receptor antagonists produce their effects on the CB1 receptor (132). The ligands can be competitive antagonists of CB1 receptor activation by endogenously released endocannabinoids, or they can act as inverse agonists and modulating constitutive CB1 receptor activity by shifting it from an active “on†to an inactive “off†state (133). They may also act by CB1 receptor independent mechanisms (132). These mechanisms are not mutually exclusive.
Previous SectionNext Section
III. Exogenous and Endogenous Cannabinoids and Their Role in Endocrine Regulation

It has been known for a long time that exogenous cannabinoids are able to affect secretion of pituitary hormones, thus having a strong effect on peripheral target organ functions. Notably, in 1972 the first report of an induction of gynecomastia due to marijuana consumption led to a dramatic acceleration of studies on this topic (134). The hypothalamus is generally considered as the main site of cannabinoid action on neuroendocrine functions. This view is elegantly supported by a recent publication showing that endocannabinoids act as retrograde messengers activating CB1 receptors expressed at presynaptic glutamatergic terminals in the hypothalamus (103). The subsequent activation of the CB1 receptor signaling cascade leads to the inhibition of the release of the excitatory neurotransmitter glutamate onto the neuroendocrine cells of the PVN and the supraoptic nucleus (103). This leads to a general suppressive effect on neuroendocrine cells and a final inhibitory effect on neuroendocrine function.

However, it was recently proposed that the endocannabinoid system might control hormonal balance also through a direct effect at the level of the peripheral target organs. An overview of the cannabinoid actions on endocrine axes is given in Table 1⇓.
View this table:

In this window
In a new window

TABLE 1.

Overview of the cannabinoid action by CB1 activation in the various endocrine axes
A. Cannabinoids and the hypothalamic-pituitary-adrenal axis

Stimulation of the hypothalamic-pituitary-adrenal (HPA) axis is a crucial neuroendocrine response to stress. Psychological or physiological stressors are known to induce CRH production in the PVN of the hypothalamus, eventually leading to a release of this hypothalamic peptide onto the anterior pituitary gland. In turn, this leads to increased circulating levels of ACTH and, finally, to an increase of corticosteroids secreted by the adrenal gland.

Until a few years ago, the impact of the cannabinoids on the HPA axis was considered as an exception. Whereas the commonly accepted view attributes the cannabinoid system with a general inhibitory role on neuroendocrine functions, it was suggested that cannabinoids are, on the contrary, able to stimulate the HPA axis. In fact, many studies in animals point to a CB1 receptor-dependent (135) increase of circulating ACTH and glucocorticoid levels after pharmacological administration of plant-derived (136), synthetic (137, 138), or endogenous cannabinoid agonists (139, 140). In agreement with this, a simultaneous elevation of CRH in the PVN and of proopiomelanocortin in the anterior pituitary after chronic treatment (18 d) with the CB1 receptor agonist CP-55,940 was observed in rats (138). Cannabinoids were proposed to act exclusively at hypothalamic sites after the finding that Δ9-THC did not induce hyperactivation of the HPA axis in hypophysectomized rats (141), and that Δ9-THC or WIN 55,212-2 was unable to stimulate ACTH release from basal and CRH-stimulated dispersed pituitary cells or isolated pituitary slices, respectively (135, 142).

However, this concept was recently challenged by several reports showing a different function of endocannabinoids on the HPA axis. In fact, some studies showed that administration of the CB1 receptor antagonist SR141716 in rats is able to induce ACTH and corticosterone release and to produce anxiety-like behavior (143, 144). It is well known that this behavior represents part of the physiological response to stressful stimuli and is, indeed, associated with the hyperactivation of the HPA axis (145). Moreover, compounds able to increase endocannabinoid tone by inhibiting FAAH activity were recently proposed as treatment for anxiety-related disorders because they were shown to reduce restraint-induced corticosterone release (146) and to diminish the anxiety-like response in different tasks (147). In addition, mice lacking CB1 receptor (CB1−/−) are resistant to some actions of anxiolytic drugs (148). In support of the existence of a close interaction between the endocannabinoid system and CRH, it is important to mention that CB1 receptor and CRH mRNAs are coexpressed in PVN neurons, and that CB1−/− mice present increased CRH mRNA levels in this region, indicative of a possible basal alteration of the HPA axis activity due to the disruption of CB1 receptor signaling (58). Therefore, a novel view seems to attribute the endocannabinoid system with a critical inhibitory action on HPA functions. A recent elegant report by Patel et al. (146) shed light on this issue. The authors confirmed previous studies showing that systemic treatment with SR141716 is able to increase serum corticosterone concentrations in basal conditions; more importantly, they found that pretreatment of mice with the same CB1 receptor antagonist before acute restraint stress provokes a potentiation of the restraint-induced rise in serum corticosterone concentrations. In addition, endogenous cannabinoids and, in particular 2-AG, were found to be decreased after a short period of restraint stress, whereas a condition of prolonged stress was associated with an increase in 2-AG concentrations (146). Accordingly, they concluded that endocannabinoid signaling negatively modulates the stress-induced activation of the HPA axis, confirming the notion that a pharmacological increase in endocannabinoid signaling activity may constitute a novel approach to the treatment of anxiety-related disorders (146). These findings reinforce the general concept that the pharmacological administration of cannabinoids may lead to a completely different action when compared with the physiological functions of the endocannabinoid system as shown by experiments using CB1 receptor antagonist or CB1−/− mice.

Besides the hypothalamus, peripheral sites of action, such as pituitary and adrenal glands, could participate in the endocannabinoid modulation of the HPA functions. In cultured human ACTH-producing tumors, WIN 55,212-2 was found to be ineffective in influencing basal ACTH secretion. However, the simultaneous application of WIN 55,212-2 and CRH caused a synergistic action, which was abolished by SR141716, indicating that the activation of CB1 receptor might play a role during CRH-induced activation of ACTH-secreting cells (52). Therefore, in the corticotroph cells, an endocannabinoid tone could interfere with the normal regulation of the adenylate cyclase activity and, thus, with the secretion of ACTH. As mentioned above, a pending question regards CB1 receptor expression and endocannabinoid production at the level of cortical adrenal gland and their putative role in the secretive function of this gland. Further efforts are needed to solve this important issue. Interestingly, our recent unpublished studies indicate that CB1−/− have higher plasma levels of corticosterone but normal levels of ACTH, suggesting a putative regulation of adrenal activity by the endocannabinoid system (our unpublished results).
B. The role of cannabinoids in GH secretion

GH secretion is mainly stimulated by hypothalamic GHRH and by the recently discovered peptide ghrelin (69), whereas somatostatin is the most important negative regulator. Other neurotransmitters such as serotonin, dopamine, and catecholamines can affect GH secretion through modulation of GHRH release. Few data are available concerning the effects of marijuana on GH in humans. Four days of marijuana consumption were shown to inhibit the GH-counteracting response provoked by insulin-induced hypoglycemia (149). Δ9-THC and synthetic cannabinoids were shown to inhibit GH secretion in rodents (150, 151, 152). However, compared with other hormones, it is still questionable whether cannabinoids are able to decrease GH secretion acting exclusively at the hypothalamic level or whether they also directly influence GH pituitary output. Rettori et al. (153) observed that only intracerebroventricular Δ9-THC administration was able to reduce GH secretion, whereas no effect was observed in cultured rat pituitary cells. Interestingly, by incubating fragments of median eminence with Δ9-THC, a significant stimulation of basal somatostatin was found (154); this finding makes it possible to speculate that the inhibitory action of Δ9-THC on GH secretion could be mediated by somatostatinergic activation (154). Recent data point to a functional cross-talk between CB1 receptor and the ghrelinergic system. In fact, hyperphagia associated with intracerebroventricular administration of ghrelin is blocked by pretreating the rats with SR141716 (155). Unfortunately, no data have been provided concerning GH release in this experimental setting. Altogether, these data seem to indicate that the effect of exogenous cannabinoids on GH secretion is located at a suprapituitary level. However, the cannabinoid agonist WIN 55,212-2 inhibited GH secretion in human GH-producing adenomas in culture, and this effect was reversed by the specific CB1 receptor antagonist SR141716, suggesting that cannabinoids are able to directly influence basal GH secretion through CB1 receptor activation, at least in tumoral tissues (52). No data are available on the physiological modulation made by the endocannabinoid system on GH secretion.
C. Cannabinoids and the hypothalamic-pituitary-thyroid axis

Pioneer studies showed that marijuana is able to decrease TSH and thyroid hormones in rats (156, 157) and iodine accumulation in the isolated rat thyroid (158). The lack of changes in TRH secretion in the hypothalamus led the authors to conclude that the cannabinoid effect could be attributed to a direct action at the level of the pituitary or the thyroid gland (157). Recently, Porcella et al. (56) found a CB1 receptor-dependent decrease (30%) in both free T3 and free T4 4 h after the administration of the synthetic cannabinoid agonist WIN 55,212-2 in rats. TSH levels were unaffected, indicating that the thyroid gland itself may be the direct target of cannabinoid action (56). On the other hand, the lack of TSH changes may also be explained by an action of cannabinoids on the levels of thyroid binding protein or on the metabolism of thyroid hormones. More studies are needed to verify these hypotheses. Concerning the physiological roles of the endocannabinoid system, an inhibitory action on TRH neurons through a glucocorticoid-induced inhibition of glutamate transmission was recently proposed (103).
D. The role of cannabinoids in prolactin secretion

There is no general consensus regarding the effect of exogenous cannabinoids on PRL secretion. Early studies in rodents and primates favor an inhibitory role of cannabinoids on PRL release (153, 159, 160, 161, 162) through a CB1 receptor-mediated effect (163). Conversely, some reports showed that cannabinoids may also have either a stimulatory effect (164, 165) or no effect (166) on PRL release. As often occurs in the field of cannabinoids, this controversy may be largely due to the different experimental settings used. The conflicting data may also originate from the biphasic profile of PRL observed after Δ9-THC administration, with an initial increase followed by a marked decrease after time (167). In the same study, the antagonist SR141716 was only able to block the inhibitory effect, whereas no effect was seen toward the cannabinoid stimulatory effect on PRL (167). There is a general agreement that cannabinoid activation of the tuberoinfundibolar dopaminergic neurons controlling PRL secretion is the main mechanism responsible for the inhibition of this pituitary hormone (168, 169). When Δ9-THC was chronically administered to ovariectomized or hypophysectomized female rats or to dispersed pituitary cells in culture, no effect was seen on PRL release, suggesting that the inhibitory cannabinoid effect targets the CNS directly (161). Similar conclusions were drawn from similar models by other authors (153). Recently, exogenous AEA was shown to inhibit PRL release from male rats by acting on the CB1 receptor on dopaminergic neurons located in the medial basal hypothalamus (162). However, like other hormones, it has also been hypothesized that cannabinoids may also affect PRL secretion directly in the pituitary. Indeed, Δ9-THC was able to prevent estrogen-induced PRL secretion in vivo (170) and in vitro (170). The direct effect of cannabinoids at pituitary level was also confirmed by the demonstration that WIN 55,212-2 does not affect basal secretion, but inhibits vasoactive intestinal peptide- and TRH-stimulated PRL release in tumoral pituitary GH4C1 cells (171). WIN 55,212-2 was also able to inhibit PRL secretion in a single case of prolactinoma in culture (52). In conclusion, we can assume that the biphasic action on PRL secretion of exogenous cannabinoids is mediated by an initial activation of CB1 receptor located at the level of the pituitary and followed by a persistent inhibitory action mediated by the activation of the release of dopamine from hypothalamic structures.
E. The role of cannabinoids in modulation of the hypothalamic-pituitary-gonadal axis and fertility
1. In females.

While FSH secretion seems to be unaffected by administration of exogenous or endogenous cannabinoids (172), several pieces of evidence attribute cannabinoids with a strong ability to down-regulate blood LH levels (49, 165, 172, 173). This effect is due to a complete suppression of the secretory pulse of LH (174, 175). In monkeys, chronic administration (18 d) of Δ9-THC was shown to block estrogen and LH surges and the consequent elevation in progesterone (176). However, the same animals developed tolerance to the antireproductive effect of the drug after a few months of treatment (177). In women smoking a single marijuana cigarette with a fixed content of Δ9-THC, a decrease of LH was observed during the luteal phase, whereas no effect was seen on the same hormone in the follicular phase and in the postmenopausal state (178, 179). The sustained use of marijuana (at least four times per week) may cause alterations of the menstrual cycle, such as oligomenorrea; however, no changes were shown in hormonal parameters in a group of 13 pregnant women who continued to smoke marijuana during pregnancy (180). An excess of cannabinoids may also impair regular ovulation, not only acting at the hypothalamic level but also directly affecting ovarian granulosa layers (76).

A general consensus attributes the LH-inhibitory action of cannabinoids to a suprapituitary site of action. In fact, administration of gonadotropins or GnRH was able to induce ovulation or LH release, respectively, even in the presence of high levels of Δ9-THC (174, 175). However, a report showed that cannabinoids are not able to block the basal GnRH secretion from hypothalami in vitro (165). This last finding suggests that cannabinoids indirectly modify GnRH secretion by negatively modulating the activity of neurotransmitters known to facilitate GnRH secretion, such as norepinephrine (165) and glutamate (181), and by stimulating those modulators known to down-regulate GnRH secretion, such as dopamine (182), GABA (183), opioids (184), and CRH (185). The stimulatory effect of cannabinoids on dopaminergic neurons is well known (186), however their impact on the brain dopaminergic activity varies as a function of the gonadal status, as demonstrated by several lines of evidence (187). In particular, it has been shown that steroid hormone receptors mediate the well known Δ9-THC-facilitation on sexual behavior (188) exerted, as recently shown, by CB1 receptor activation (189). Moreover, in the same study Mani et al. (189) reported that an interaction between progesterone and dopamine receptor type 1 (D1) is required for Δ9-THC-facilitated sexual receptivity in female rats.

However, although pharmacological studies have helped to explain the relevant role of the cannabinoids in modulation of the hypothalamus-pituitary-gonadal axis and sexual behavior, it is not yet known how, where, and under what circumstances the endocannabinoids are produced to do so. The recent findings of fluctuation during the ovarian cycle of AEA in both hypothalamus and pituitary (49) allowed some authors to speculate that endocannabinoids may influence hormonal secretion and sexual behavior by directly targeting the CB1 receptor (190). Furthermore, an important production of endocannabinoids was found in the ovary, in particular at the time of ovulation, making it possible to hypothesize that the endocannabinoids may help to regulate follicular maturation and development of the ovary (74).

The uterus contains the highest level of AEA detected so far in mammalian tissues, and it is the only tissue where AEA is the main component (up to 95%) of N-acylethanolamides (191). This observation, together with the expression of CB1 receptors in preimplantation embryos (80), recently prompted strong efforts focused on the role of the endocannabinoid system during early pregnancy and in the modulation of embryo-uterine interactions. High levels of AEA adversely affect embryo development and implantation through CB1 receptor activation (192), whereas low levels of AEA promote embryonic growth and differentiation (193, 194, 195). It is therefore evident that the degradation of AEA by FAAH is a crucial enzymatic checkpoint in the control of reproduction. Notably, a strong inverse correlation was described between levels of FAAH activity in maternal peripheral blood mononuclear cells and spontaneous miscarriage in women (196). In addition, FAAH activity is lower, and consequently AEA higher, in patients who fail to achieve pregnancy during in vitro fertilization embryo transfer in comparison to patients who become pregnant (197). Furthermore, AEA levels in the mouse uterus are inversely related to uterine receptivity for implantation, being higher with uterine refractoriness to blastocyst implantation (191, 198, 199) and lower at implantation sites (194). We can therefore conclude that high levels of maternal AEA are detrimental to early placental and fetal development. In favor of this hypothesis, it was recently shown that high levels of FAAH are present in the cytotrophoblast, presumably to prevent the transfer of AEA from maternal blood to the embryo (200). A series of studies by Maccarrone et al. (72, 195) showed that the activity of FAAH is under the strict regulation of several hormones, such as progesterone, leptin, and FSH, very well-known modulators of fertility. Importantly, by using genetic or pharmacological blockade of the CB1 receptor, it was very recently demonstrated that an impairment in endocannabinoid signaling leads to a retention of a large number of embryos in the mouse oviduct, leading to pregnancy failure. This is due to a profound impairment of a coordinated oviductal smooth muscle contraction and relaxation (79). The authors propose that their findings may have strong implications for ectopic pregnancy in women because one major cause of tubal pregnancy is embryo retention in the fallopian tube (79). Consistently, both endogenous and exogenous cannabinoids exert a CB1 receptor-mediated relaxant effect, not only on the oviductal smooth muscle but also on the human pregnant myometrium, highlighting a possible role of endocannabinoids during human parturition and pregnancy (78). In fact, pregnancy also seems to be tightly controlled by the endocannabinoid system (200). In summary, all the steps starting with fertilization up to pregnancy seem to be tightly modulated by endocannabinoids, reinforcing the concept that the endocannabinoid system should be considered not only as a central neuromodulator but also as a physiological actor in a wider scenario.
2. In males.

Cannabinoids also were shown to decrease LH in males (201, 202). Although there is still no general consensus, chronic cannabinoid use in several species seems to decrease testosterone production (203) and secretion (201, 202), to suppress spermatogenesis, and to reduce the weight of testes and accessory reproductive organs (204). The important effects of cannabinoids on the gonadal system are mainly attributed to CB1 receptor activation, as demonstrated by using specific CB1 receptor agonists and antagonists (151, 205). Definitive confirmation was provided by a recent study showing that AEA injected ip is able to lower LH and testosterone in wild-type mice but not in CB1−/− mice (71). Interestingly, the testis is known to express CB1 receptor (70) and to synthesize endocannabinoids (206). The cannabinoid effect in down-regulating testosterone circulating levels may explain the reduced copulatory behavior in male rodents exposed to Δ9-THC (207).

The finding that male genital tract fluids contain significant concentrations of endocannabinoids (74) suggests that these lipid-signaling molecules may influence important processes controlling sperm/egg functions and gamete interactions. Studies with sea urchin gametes provided the first evidence that cannabinoids, in particular AEA, are able to directly inhibit achrosome reaction and sperm fertilization capacity (208). On the other hand, seminal plasma contains high amount of AEA, and this may contribute to maintaining sperms in a quiescent metabolic condition (74). The content of AEA decreases progressively in the uterus, oviduct, and follicular fluid, and this change in endocannabinoids may render sperms suitable for capacitation and fertilizing ability (74, 209). Furthermore, as shown in sea urchin, the eggs may have the capacity to release AEA after activation by the fertilizing sperm (210), inducing a CB1 receptor activation that might be able to prevent polyspermic fertilization by blocking the acrosome reaction in other sperm (209).

In humans, CB1 receptor activation by AEA was also shown to reduce sperm mobility by affecting mitochondrial activity, and to inhibit capacitation-induced acrosome reaction. Importantly, these effects are inhibited by the CB1 receptor antagonist SR141716 (75). It is therefore reasonable to hypothesize that AEA levels might be increased in different pathological conditions of the male reproductive tract. In these cases, the pharmacological blockade of the endocannabinoid system might be helpful in the treatment of some forms of male infertility (75).

In conclusion, it appears that the endocannabinoid system plays an important role in the regulation of the hypothalamus-pituitary-gonadal axis both in females and in males, and fertility may be affected by cannabinoid drugs. This evidence may represent an important issue in clinical endocrinological praxis. In the light of the widespread use of marijuana as a recreational drug among young people, subtle alterations of the gonadal hormonal profile or in fertility may therefore be attributed to a concomitant use of cannabis derivatives. On the other hand, the results of human epidemiological studies have not always been clear in confirming this negative impact (211), and more detailed research on this topic is needed in the future before drawing definitive conclusions.
Previous SectionNext Section
IV. Endocannabinoid System in the Modulation of Energy Balance

Two notions highlight the importance of the endocannabinoid system in the regulation of food intake and energy metabolism. The first is the finding of a high degree of evolutionary conservation of the role of this system in the regulation of feeding responses (212). The second is the observation that high levels of endocannabinoids in maternal milk are critically important for the initiation of the suckling response in newborns (213).
A. Animal studies before the discovery of endocannabinoids

Animal models are ideal tools for elucidating the putative mechanism(s) of cannabinoids in the control of energy metabolism. The studies performed in different species to test the orexigenic properties of Δ9-THC up to the discovery of endocannabinoids are summarized in Table 2⇓ (214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244). From a general point of view, one can say that rather contradictory results were obtained in these experiments. The ambiguous data could likely be attributed to differences in the animal model and in the experimental procedures used. Moreover, in early studies using marijuana extracts, comparisons between various experimental data sets are extremely difficult due to the variability of the activity of cannabis derivatives, the dosages, and the routes of administration. In general, early studies using low doses of cannabinoids showed a reliable increase in food intake. When doses of Δ9-THC above 10 mg/kg were used, a concomitant decrease in food intake was observed due to the confounding factors given by the sedative effect of the drug. Studies employing high amounts of Δ9-THC should thus be viewed with caution in terms of effects on appetite and body weight. This is also the reason why, in reviewing the studies published between 1965 and 1975, Abel reported an increased food intake after cannabinoid administration only in 3 of 25 experiments (245). In 1998, Williams et al. (246) provided a very convincing and well-performed experiment to characterize the orexigenic property of Δ9-THC. The authors maximized the ability to detect hyperphagia by adopting a prefed paradigm in which the animals were characterized by low baseline food intake before drug administration. In this experimental setting, Δ9-THC was given orally at increasing dosage before unrestricted access to a standard diet. The authors observed that the maximum effect of the drug (1.0 mg/kg) was far greater than previously reported results, showing a 4-fold increase in food consumption over 1 h. Importantly, this hyperphagic effect was largely attenuated by pretreatment with the CB1 receptor antagonist SR141716, strongly supporting the notion that CB1 receptor activation mediates the hyperphagic effect of Δ9-THC (247). In this experiment, it was also reported that at doses of Δ9-THC higher than 1.0 mg/kg, the rats become unable to overeat due to the presence of motoric and sedative side effects (246). These results strongly suggest that the anorectic effect of Δ9-THC shown by many previous reports was indirectly due to the sedated state induced by high doses of the drug.
View this table:

In this window
In a new window

TABLE 2.

Summary of the effects of exogenous cannabinoids on food intake
B. Studies in humans with exogenous cannabinoids before the discovery of endocannabinoids

Abel (245) also critically reviewed the studies aimed at proving the stimulating effect of cannabis on hunger in humans. However, the lack of scientific thoroughness of these earlier studies led Abel to conclude that the putative cannabis-induced hunger effect was still far from being proven (245). Greenberg et al. (248) were the first to systematically assess, under rigorous experimental conditions, the effect of a well-defined amount of Δ9-THC in terms of changes in feeding behavior and in body weight in humans. Both parameters increased after the first few days of the experiment. However, after this period, body weight continued to rise, averaging 2.3 kg across the whole 21-d period study, whereas a stabilization of energy intake was observed. This pioneer study already suggested that the ability of cannabinoids to stimulate hunger may vanish with time, whereas a possible metabolic effect of the drug may remain active longer (248). Nonetheless, later studies did not investigate the metabolic idea further, preferring to concentrate interest on the ability of cannabis to stimulate hyperphagia and overconsumption of highly palatable food at the central level. In 1986, Foltin et al. (249) noted a relevant increase in frequency and consumption of snack foods induced by marijuana only in the periods of social facilitation and environmental familiarity and not when the subjects were alone, indicating on the one hand a strong link between recreational use of the drug and its orexigenic properties and, on the other hand, the ability of marijuana to drive the tendency for palatable food. This hypothesis was further substantiated by the same group a few years later when increased total food intake particularly related to consumption of palatable food (sweet solid snacks) was observed as a main effect of smoked marijuana (250).

The stimulating effect of cannabinoids on appetite observed in healthy subjects promoted assessment of the efficacy of a cannabinoid treatment for clinical syndromes featuring loss of appetite or weight, such as cancer or AIDS-associated anorexia (251, 252, 253), or as adjuvant therapy to limit nausea and vomiting symptoms associated with most chemotherapeutic drugs (254). In 1985, the U.S. Food and Drug Administration officially approved the use of Δ9-THC (commercially named Dronabinol) for the treatment of chemotherapy-induced nausea and vomiting refractory to other drugs. In 1992, Dronabinol was approved for the treatment of patients with HIV-induced wasting syndrome. Recently, Dronabinol was also proposed as an orexigenic drug in patients suffering from Alzheimer's disease (255).

The most comprehensive data are those obtained when Dronabinol was administered in HIV patients with wasting syndrome (252, 256, 257, 258, 259). To varying degrees, the drug was able to mildly increase appetite and energy intake in all studies. However, a marked improvement in mood was also documented, raising the question of whether the positive effect in energy balance may derive from a specific action of cannabinoids in the brain areas controlling food intake or may be simply due to a generalized change in the sense of well-being. Intriguingly, in some reports, a significant gain was found in body fat mass associated with minimal changes in appetite rating and food intake (255, 258). At that time, this finding remained unexplained. However, with the current knowledge of CB1 receptor expression at the level of the adipose tissue (58, 59), we can hypothesize that the increase in fat mass of HIV patients was probably due to a direct lipogenic action of Δ9-THC. In this context, it is still unknown, and it would be of great relevance to investigate whether the administration of Dronabinol can improve the pathological changes in fat distribution induced by the concomitant retroviral therapy in patients with AIDS (260).
C. Endocannabinoid functions at mesolimbic level to regulate rewarding properties of food

After the finding of the hyperphagic effect of Δ9-THC mediated by CB1 receptor activation, Williams and Kirkham (261) reported that endocannabinoids were also able to stimulate hunger in a dose-dependent manner. The degree of overeating induced by 1 mg/kg AEA was only a 2-fold increase over a 3-h test, therefore less than that obtained with the same dosage of Δ9-THC. However, Δ9-THC-induced hyperphagia was restricted to the first hour of testing, whereas the AEA effect was evident later when the inhibitory effects of the prefeed started to wane (261). The authors speculated that administration of AEA may represent an amplification of endocannabinoid activity associated with the normal, episodic pattern of meal-taking in rats (261).

Importantly, the effect of AEA was completely blocked by pretreating the animals with SR141716, confirming the pivotal role of CB1 receptor activation in the hyperphagic effects of endocannabinoids (247, 262). Similar conclusions were derived from other studies in which AEA was able to exert an appetite-stimulating effect even at very low doses in mice (0.001 mg/kg) (263) and 2-AG was capable of promoting feeding behavior (264). These data therefore make it possible to attribute the endocannabinoid system with an important role in the processes underlying the motivation to obtain food. It is suggested that endocannabinoids gradually increase during intermeal intervals, reaching a critical level where motivation to eat is triggered. Accordingly, the longer the time since the last meal, the greater the activity in relevant endocannabinoid circuits, and consequently the higher the motivation to eat (265). The findings of increased levels of AEA and 2-AG in the fasting condition in the nucleus accumbens and a decline of 2-AG concomitant with the feeding state strongly support this hypothesis (264). Interestingly, unchanged levels of endocannabinoids were shown in the cerebellum, a region not involved in the control of feeding, further confirming the notion that endocannabinoids are produced in situ and on demand (264).

With the advent of CB1 receptor-specific antagonists (Table 3⇓), it became clear that, even when injected alone, these compounds are able to modify ingestive behavior. An ip injection of SR141716 was found to significantly reduce sucrose or alcohol intake and craving in rodents (266, 267, 268) and in marmosets (269), leading to the hypothesis that the activation of the endocannabinoid system may alter the appetitive value of ingested substances. This idea is consistent with the evidence in favor of a facilitatory function of the endocannabinoid system on brain reward circuits (266, 269). Evidence therefore suggests that endocannabinoids bring forward the onset of eating in satiated animals and increase the incentive value of the food regardless of the quality of the macronutrients (“incentive hypothesisâ€) (270). Other findings, however, resembling the “marshmallow effect†in marijuana smokers (245), have been interpreted in terms of an endocannabinoid action toward a preference to eat highly palatable food (“orosensory reward hypothesisâ€) (271). In favor of this latter hypothesis, there are several reports indicating the ability of CB1 receptor blockade to decrease the rewarding properties of addictive drugs (186, 272, 273, 274). It is now clear that the endocannabinoid system participates in the modulation of “reward/reinforcement†circuitries and its manipulation is able to influence reward-related behaviors (275). The high expression of CB1 receptor in areas involved in reward constitutes a strong indication that the endocannabinoid system is directly involved in various physiological functions controlled in these brain regions, including feeding (43). The reward/reinforcement circuitry of the mammalian brain consists of a series of synaptically interconnected brain nuclei associated with the medial forebrain bundle, linking the VTA, the nucleus accumbens, and the ventral pallidum (275). This circuit is implicated in the pleasure produced by natural rewards, such as food, addictive drugs, and sex, and it is the neural substrate of drug addiction and addiction-related phenomena, such as craving and dysphoria induced by withdrawal (275). In such a framework, food intake acts on dopamine, opioid, serotonin, and noradrenaline neuronal fibers, which connect the hindbrain and midbrain to the hypothalamus to modulate the action of feeding and satiety factors (276).
View this table:

In this window
In a new window

TABLE 3.

Summary of the effect of CB1 antagonist treatment on food intake in different rodent models

The most relevant reward pathway is represented by the mesolimbic dopaminergic system. It has been shown that increased levels of extracellular dopamine and its metabolites are found within the nucleus accumbens after ingestion of highly palatable food (277). Moreover, administration of a dopamine D1 agonist reduces food intake (278). Both CB1 receptor and endocannabinoids were found in the rat limbic forebrain (279), in which colocalization with dopamine D1 and D2 and CB1 receptor were described (280). Psychoactive drugs such as marijuana, ethanol, and also pleasant stimuli or palatable food are known to induce the release of dopamine in specific brain regions (281). A correlation between limbic endocannabinoid/dopamine levels and craving for tasty food is thus presumed to occur (275). Verty et al. (282) recently substantiated the hypothesis of the existence of cannabinoid-dopamine interactions in feeding behavior, demonstrating that the dopamine D1 antagonist SCH 23390 attenuated feeding induced by Δ9-THC. The endocannabinoid system also provides retrograde control of synaptic transmission onto the VTA dopaminergic neurons, where the postsynaptic synthesis of endocannabinoids is under the control of somatodendritically released dopamine (108).

A relevant interplay also exists between the endocannabinoid system and the endogenous opioid peptides (283). Both systems are linked to central reward processes, and there is increasing evidence supporting an important functional cross-talk between the two systems, in relation to a wide range of physiological processes, including appetite. Several reports indicate that opioid receptor agonists increase food intake (284, 285, 286), whereas opioid antagonists induce anorectic effects (287). Gallate and McGregor (267) found that the facilitatory effects of a cannabinoid agonist on responding to palatable solutions were reversed not only by CB1 receptor antagonism but also by naloxone, an opioid receptor antagonist. The existence of cross-talk between the endocannabinoid and opioid systems in controlling food intake was also confirmed by several studies in which naloxone and SR141716 synergistically depress food intake at doses that do not alter food intake on their own (287, 288). However, a recent finding seems to localize the interaction between opioids and endocannabinoids involved in feeding behavior not at the mesolimbic system level but, preferentially, at the level of the PVN of the hypothalamus. In fact, SR141716 was able to attenuate morphine-induced feeding only when the opioid was directly injected in the PVN and not in the nucleus accumbens. According to this last finding, the endocannabinoid system appears to participate in the opioid-mediated enhancement of rewarding properties of food in the hypothalamus and not in the nucleus accumbens (286).

According to the involvement of serotonin in the control of feeding behavior (289), the interaction of the endocannabinoid system with the serotoninergic system has also been investigated. However, the administration of a CB1 receptor antagonist in rats combined with dexfenfluramine, an anorectic drug stimulating the release of serotonin, led to additional but not synergistic effects on reducing food intake, which is consistent with the hypothesis that the two pathways work via independent mechanisms of action (288). This notion is important, because it makes it possible to exclude a synergistic effect in a possible future combination of antiobesity drugs such as those inhibiting serotonin reuptake, like sibutramine (290) and CB1 receptor antagonists.
D. The endocannabinoid system as a new hypothalamic player in the regulation of food intake

A complex and redundant neuronal hypothalamic network provides high levels of adaptability of feeding behavior to various central and peripheral stimuli (291). Redundancy in appetite-stimulating signaling is conceivable in view of the vital importance of feeding for survival (291). Whereas defects in anorexigenic signaling pathways almost always lead to obesity, loss of orexigenic signals rarely results in a lean phenotype. An example of this redundancy in orexigenic hypothalamic signaling systems is provided by mice lacking neuropeptide Y (one of the most important appetite-stimulating neuropeptides) where compensatory mechanisms are likely to be activated (292). Signals coming from various peripheral organs, such as the liver, gastrointestinal tract, and adipose tissue, are conveyed mainly at the hypothalamic level to constantly inform the brain about the state of nutrition (291, 293). An example of such peripheral control is the adipocyte-derived hormone leptin, which acts on receptors located in the hypothalamus (291). A milestone in the identification of the endocannabinoid system as a new player in the regulation of food intake at hypothalamic level was the finding that leptin is a strong modulator of hypothalamic endocannabinoid levels (294). Di Marzo et al. showed that acute leptin treatment reduced AEA and 2-AG not only in the hypothalami of normal mice but also in mice lacking leptin signaling. They also described the defect in leptin signaling as being constitutively associated with elevated hypothalamic levels of endocannabinoids. In these animals, SR141716 was able to reduce food intake, confirming the anorectic properties of the compound (294). These findings suggest that, at least in genetically modified animal models, obesity is associated with a chronic hypothalamic overactivation of the endocannabinoid system, which may in turn explain the hyperphagic behavior of the animals having leptin signal impairment. However, before giving a general value to this assumption, the intrahypothalamic amount of endocannabinoid levels during the development of obesity in normal rodents eating a high-fat diet must be investigated. Nevertheless, endocannabinoids are variably produced in the hypothalamus of normal animals. In fact, 2-AG levels increase during acute fasting, decline as the animals are refed, and return to normal values in satiated animals (264, 295). However, a long period of diet restriction (12 d) was found to be associated with reduced levels of 2-AG in the hypothalamus (295). The authors interpreted these data observing that the decrease of 2-AG levels in mice after a prolonged diet may represent a general psychobehavioral strategy for intermittent starvation when food is scarce (295).

As mentioned above, the hypothalamus is not the cerebral area where the highest levels of CB1 receptor expression are found (24, 36, 38). However, studies using [35S]GTPγS binding indicated that the hypothalamic CB1 receptor coupling to G proteins is more efficient than in other cerebral areas known to be a site of high CB1 receptor expression, such as the hippocampus or the entopeduncular nucleus (43). On the other hand, it is also evident that CB1 receptors are present at a very high density in the brain compared with other receptors. Therefore, even regions with a relatively lower density of CB1 receptors, such as the hypothalamus, contain a significant number of receptors. Both these factors thus probably explain the ability of hypothalamic CB1 receptors to strongly affect the functions of this brain region. Interestingly, no changes in CB1 receptor expression have been shown at the level of hypothalamus after diet modification (296). The direct involvement of the hypothalamus in the modulation of food intake operated by endocannabinoids was also demonstrated by the significant hyperphagic effects of AEA directly administered into the ventromedial nucleus and by the inhibition of this effect obtained by the injection of SR141716 via the same route (297).

It was only during the last few years that the interaction of CB1 receptor and endocannabinoids in feeding-regulating pathways started to be elucidated in detail. The CB1 receptor is expressed in key hypothalamic peptidergic systems, such as those producing CRH in the PVN, cocaine-amphetamine-related transcript in the dorsomedial nucleus, and melanin-concentrating hormone and orexins in the lateral hypothalamus-perifornical area (58). Importantly, these data were recently confirmed by the demonstration that CB1 receptor activation strongly augments the orexin-A-stimulated intracellular pathway (88). CB1−/− mice also possess increased CRH and reduced cocaine-amphetamine-related transcript expression, indicating that the genetic impairment of the endocannabinoid system may affect the pattern of gene expression of peptides involved in the regulation of food intake (58). Conversely, the neuropeptide Y/agouti-related protein system in the arcuate nucleus does not seem to be directly targeted by endocannabinoid action (58, 294). This fact confirms that orexigenic pathways are less critical (or at least functionally more redundant) in the chronic maintenance of energy balance (298). Functional cross-talk between CB1 receptor and melanocortin receptor type 4 (MCR4) has been recently highlighted by the finding of the synergistic action of subanorectic doses of SR141716 and of a MCR4 agonist administered together (299). Furthermore, the same authors showed that the orexigenic impulse given by the administration of CB1 receptor agonists is not blocked by the costimulation with MCR4 agonists, whereas CB1 receptor antagonists are able to inhibit the stimulation of food intake induced by MCR4 antagonists. Consequently, the authors hypothesized that the melanocortin receptor signaling in the hypothalamic regulation of food intake is upstream of the activation of the endocannabinoid system (299).

The mechanism(s) of action of the endocannabinoids at hypothalamic synaptic level are still a matter of debate. Great progress has recently been made by the finding that postsynaptically released endocannabinoids acting at presynaptic CB1 receptors are able to decrease glutaminergic transmission onto CRH-producing neurons, resulting in an inhibition of CRH release (103). This release of endocannabinoids from the parvocellular neurons is stimulated by a nongenomic effect of glucocorticoids. Therefore, it is conceivable that the well-known regulation of food intake by glucocorticoids may partly derive from functional cross-talk with the endocannabinoid system (300). The same inhibitory mechanism mediated by glucocorticoids through an activation of the endocannabinoid system has also been proposed for other hormones and neuropeptides such as oxytocin and vasopressin (103). In this sense, we may speculate that the recently described interaction between endocannabinoid and the oxytocin system in modulating food intake (301) may derive from the same fast feedback mechanism mediated by nongenomic glucocorticoid inhibition.

Despite the dogma that neurons do not utilize fatty acids for energy, a growing body of evidence points to a critical role for both fatty acid production and utilization in regulating hypthalamic neurons that regulate food intake (302). In fact, inhibitors of fatty acid synthase are capable of greatly affecting appetite in an anorexigenic manner (303, 304). In such a scenario, it has recently been proposed that via CB1 receptors, endocannabinoids may modulate the fatty acid synthetic pathway in the hypothalamus, and the inhibition of the hypothalamic expression by rimonabant may explain the anorexigenic properties of cannabinoid antagonists (62).
E. The peripheral effect of the endocannabinoid system in the modulation of metabolic functions

Several lines of evidence are currently converging, indicating that the effects of CB1 receptor blockade on food intake and body weight are not limited to a central mode of action. An early report describing the effect of CB1 receptor blockade on changes in food intake and in body weight was, in this sense, highly predictive of a mechanism of action not limited to the mesolimibic or hypothalamic circuits. In fact, Colombo et al. (305) were the first to demonstrate, in lean rats fed with a standard diet, that the tolerance to the anorectic effects of two different doses of SR141716 (2.5 and 10 mg/kg) develops rather rapidly (5 d). Nevertheless, the body weight loss in SR141716-treated rats persisted for 14 d, well beyond the drug effect on food intake. At that time, the authors were not able to explain this body weight loss that was not related to a decrease in food intake, and they merely hypothesized a stimulatory action of SR141716 on the energy expenditure (305). However, in the last 2 yr, the use of CB1−/− mice has represented an important tool to substantiate further the hypothesis of an additional effect of endocannabinoids in peripheral organs. Indeed, the lack of CB1 receptor in mutant mice causes hypophagia and body fat reduction. Importantly, pair-feeding experiments showed that in young CB1−/− mice, the lean phenotype is predominantly caused by decreased caloric intake, whereas in adult CB1−/− mice metabolic factors appear to be the major cause of the lean phenotype. These experiments therefore suggested that the endocannabinoid system might regulate central food intake-related mechanisms at young ages, but that this function diminishes with age (58). These observations converge on the idea that additional peripheral food intake-independent metabolic functions may participate, or even predominate, in the control of energy balance exerted by the endocannabinoid system (58). Even more prominent differences in terms of body weight regulation are obtained when a high-fat diet is administered to adult CB1−/− mice and wild-type littermates. In contrast to wild-type littermates, CB1−/− mice do not display hyperphagia or reduction of their relative energy intake and were resistant to diet-induced obesity (DIO) (306). Importantly, the obesity-prone diet induced a significant increase of fasting glycemia in the two genotypes, but the sensitivity to insulin remained unchanged in CB1−/− mice, whereas it was significantly reduced in the wild-type animals (306).

The expression of CB1 receptor in adipocytes and the ability of SR141716 to block lipogenesis stimulated by cannabinoids represent a first important step forward in understanding the peripheral mechanisms of action of the endocannabinoid system in regulating metabolic processes (58). Moreover, the presence of CB1 receptor is increased in mature adipocytes compared with preadipocytes (59, 60), indicating that CB1 receptor activation is likely needed more for metabolic processes than for differentiation. Importantly, a recent study shed further light on the mechanisms of action of the endocannabinoid system on adipose tissue. By using SR141716 in DIO mice, Jbilo et al. (307) were able to reverse the phenotype of obese adipocytes at both macroscopic and genomic levels. They showed that a major restoration of white adipocyte morphology similar to lean animals occurred in adipocytes derived from obese animals after CB1 antagonist treatment. More importantly, they found that the major alterations in gene expression levels induced by obesity in white adipose tissue were mostly reversed in SR141716-treated obese mice. Importantly, the transcriptional patterns of treated obese mice were similar to those obtained in the CB1−/− mice fed with a high-fat diet, supporting a CB1 receptor-mediated process. Functional analysis of these modulations indicated that the reduction of adipose mass by the drug was due to enhanced lipolysis through the induction of enzymes of the β-oxidation and tricarboxylic acid cycle; increased energy expenditure, mainly through futile cycling (calcium and substrate); and a tight regulation of glucose homeostasis. In particular, in this last context the SR141716-induced increased expression of glucose transporter 4, the insulin-responsive glucose transporter, appears very important (307). This finding makes it possible to hypothesize that cannabinoid antagonists may also be attractive drugs in fighting diabetes. Altogether, these data confirmed that the endocannabinoid system has a major role in the regulation of energy metabolism in adipocytes. Importantly, CB1 receptor expression has been found to be higher in adipocytes derived from obese animals compared with lean controls (59). Similar to the finding of higher levels of endocannabinoids in the hypothalamus derived from obese animals, the overexpression of CB1 receptor in adipocytes of obese rats seems to confirm the notion that hyperactivity of the endocannabinoid system is associated with the obesity state. However, this up-regulation of CB1 receptor expression in fat pads derived from rodents has not been confirmed in adipocytes derived from sc fat of obese women (60); on the other hand, a partial limitation of this study is that CB1 receptors have not been measured in visceral fat tissue that is supposed to be more prone to the endocannabinoid action. Finally, the increase in levels of adiponectin in Zucker obese rats chronically treated with SR141716 in vivo (59) and in 3T3 F442A adipocytes acutely stimulated with the CB1 receptor antagonist in vitro (59) points to a close relationship between CB1 receptor blockade and the production of this antiatherogenic and antidiabetic adipocyte-derived protein (308). The quick and strong improvement of hyperinsulinemia detected after a very short-term treatment with SR141716 (4 d) in obese Zucker rats was also attributed to an increase in adiponectin (59). However, the well-known reduction in food intake and the consequent body weight loss displayed at the beginning of SR141716 treatment may be the most obvious explanation for the changes in adiponectin levels. The ability of long-term treatment with SR141716 to enhance the circulating levels of adiponectin was further confirmed in DIO mice (309).

In the last few years, several studies using different CB1 receptor antagonists confirmed the hypothesis that a potential peripheral mode of action of pharmacological CB1 receptor blockade may play a relevant role in the final weight loss effect. Ravinet-Trillou et al. (310) found that long-term (40 d) treatment with two different dosages of SR141716 (3 and 10 mg/kg, respectively) produces a marked acute hypophagia in DIO mice only in the first few days of treatment, followed by the development of tolerance to the anorectic effect of the drug. However, the effect on body weight was sustained until the end of the 5-wk experiment compared with DIO mice treated with the vehicle. The significant difference in weight of white adipose pads between SR141716- and vehicle-treated animals confirmed that weight loss was accompanied by a decrease in adipose tissue. Similar data showed a rapid tolerance to the anorectic action despite a sustained and prolonged effect on body fat loss also being obtained when obese Zucker rats were treated for 14 d with SR141716 (59). Importantly, another CB1 receptor antagonist, AM-251, produced similar effects in DIO mice (311). Very recently, Poirier et al. (309) monitored weight and metabolic marker changes in three groups of mice after establishing a condition of obesity by a 5-month high-fat diet. Two groups of animals were maintained on a high-fat diet, but one was treated for 10-wk with 10 mg/kg SR141716 and the other one with a vehicle. A third group received a dietary switch to standard food after the 5 months on a high-fat diet. SR141716 induced a weight loss of approximately of 78% in comparison to the weight of the animals receiving the vehicle. More importantly, the antiobesity effect of the drug was equivalent (both in terms of time course and maximum effect) to that achieved by switching obese mice to a normal diet (309). Again, the authors demonstrated that the anorectic effect of the CB1 receptor antagonist vanished with time because the energy intake in the SR141716-treated animals was equivalent to animals on a high-fat diet during the last 6 wk of the experiment and significantly greater than in the group receiving standard diet. Consistent with a previous report (310), the SR141716-induced weight loss was accompanied by normalization of leptin, insulin, and glucose levels (309). Notably, SR141716 also normalized triglycerides and low-density lipoprotein-cholesterol. Moreover, the high-density lipoprotein (HDL)-cholesterol/low-density lipoprotein-cholesterol ratio after SR141716 treatment was significantly higher than in the other two groups (309). Whether this effect on lipid metabolism is indirectly related to an elevation of adiponectin is still a matter of debate.

Shearman et al. (312) recently showed that a 9-d treatment of DIO mice with the CB1 receptor antagonist AM251 increases uncoupling protein (UCP)-1 and UCP-3 mRNA expression level in brown adipose tissue, suggesting that CB1 receptor blockade may contribute to increased thermogenesis. Moreover, Liu et al. (61) found that a 7-d treatment with SR141716 induces an increase in basal oxygen consumption compared with the vehicle in ob/ob mice. The authors were not able to identify the mechanism by which SR141716 treatment is able to affect energy expenditure. A start on clarifying the molecular mechanism by which treatment with SR141716 may favor thermogenesis has been made with the microarray experiment performed by Jbilo et al. (307). These data suggest that the cannabinoid antagonist treatment is able to stimulate the expression of genes favoring energy dissipation through mitochondrial heat production in brown adipose tissue (307). However, it should be mentioned that in vivo microdialysis studies showed that SR141716 increases noradrenaline outflow in rat anterior hypothalamus, suggesting a possible central stimulation of efferent sympathetic activity (313). Importantly, Liu et al. (61) also showed that a 7-d treatment of SR141716 induces a significant increase in glucose uptake in isolated soleus muscle. This activity might contribute to the improved hyperglycemia seen after SR141716 treatment in previous studies. As shown in Fig. 2⇑, we found that the soleus muscle derived from obese mice contains increased levels of CB1 receptor compared with lean controls, further confirming the hypothesis of a hyperactivity of the endocannabinoid system associated with a condition of obesity.

Hepatocytes, key players in the metabolic processes, were not considered as a target of endocannabinoid action for a long period of time. However, substantial amounts of 2-AG are present in the liver (1.15 nmol/g tissue), and this quantity is nearly double the amount detected in other peripheral organs (295). These observations suggested the idea that the liver might be a new target of endocannabinoid action. Very recently, Osei-Hyiaman et al. (62) strongly substantiated this hypothesis by a series of experiments in which they identified the liver as a primary site for endocannabinoid-mediated modulation of lipogenesis. In fact, probably via inhibition of adenylate cyclase, the cannabinoid agonist HU210 stimulates the expression of several genes involved in the de novo synthesis of fatty acids, such as lipogenic transcription factor SREBP-1c and its targets acetyl-CoA carboxylase-1 and fatty acid synthase. The inhibition of this lipogenic response by SR141716 and its absence in CB1−/− mice confirms the lipogenic role of CB1 receptors localized in hepatocytes. However, more importantly, the authors found that the marked increase in the basal rate of hepatic fatty acid synthesis as well as the development of hepatic steatosis observed after the administration of high-fat diet were blunted by SR141716 and absent in CB1 receptor knockout mice. High-fat diet also induces an increase in the number of CB1 receptors and in hepatic levels of AEA, strongly suggesting that the blockade of the endocannabinoid system plays an important protection against the pathological consequences of a fat diet in the liver (62). These data pave the way to hypothesize the clinical use of CB1 antagonists in preventing or reversing the development of fatty liver. Another recent report showed that cannabinoids inhibit AMP-activated protein kinase activity in the liver (314). A decrease of AMP-activated protein kinase activity is known to lead to increased storage of energy, particularly in the form of fat, in hepatocytes. This mechanism may contribute to explaining the role of endocannabinoids in promoting the development of hepatic steatosis. Based on the whole body result of these data, it has been hypothesized recently that the hepatic endocannabinoid system may represent a target for the treatment of nonalcoholic fatty liver disease (315).

A considerable amount of evidence suggests that the endocannabinoid system may regulate food intake by also acting in the gastrointestinal tract. Importantly, the concentration of AEA in intestinal tissue increases during food deprivation in rats, reaching levels that are 3-fold greater than those needed to halve maximally activated CB1 receptor and 7-fold higher than the amount detected after refeeding. This surge in AEA levels may, together with the increase in the CNS, be another hunger signal to promote feeding (316). In general, we can conclude that through multiple interactions, endocannabinoids may modulate food intake also at the level of the gastrointestinal tract.
F. Oleoylethanolamide: a new anorectic fatty acid amide

Another endogenous lipid, a monounsaturated fatty acid ethanolamide, named oleoylethanolamide (OEA), was recently proposed as an important modulator of food intake (317). OEA is an analog of AEA, but the activation of any of the known cannabinoid receptors cannot explain its pharmacological effects. Recently, Piomelli's group elucidated that its action is through an activation of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)-α (318).

Peripheral administration of OEA causes a potent and persistent decrease in food intake, but this compound is completely ineffective when administered centrally (316, 317, 318). OEA-induced anorexia is not caused by nonspecific behavioral effects, because no aversion or illnesses have been reported after the peripheral administration of the compound (319). Interestingly, similar to the effects described after the administration of capsaicin (vanilloid type 1 receptor agonist) and of the PPAR-α agonist Wy-14643, a short-term reduction in heat expenditure and locomotor activity has been observed after the peripheral administration of OEA (319). However, the mechanisms underlying the reduction in motor activity remain unclear (319). OEA not only acts as a satiety signal, but also reduces body weight gain and serum lipid levels in genetically obese rats and in DIO mice (318). Through the direct activation of PPAR-α, OEA may stimulate lipolysis and fatty acid oxidation (320). However, when administered orally (321), its tissue distribution is mainly at gastrointestinal levels rather than in other visceral organs controlling metabolism, supporting the hypothesis that OEA acts on PPAR-α present in the initial segment of the gastrointestinal tract, such as stomach, duodenum, and jejunum. Importantly, these data were recently independently confirmed by another group (322). In conclusion, OEA is a new orally active anorectic agent that may possess potential as a new antiobesity drug.
Previous SectionNext Section
V. Cannabinoid Receptor Antagonists as New Pharmacological Tools to Tackle Obesity and Obesity-Related Diseases
A. Emerging issues in the treatment of obesity and related diseases by cannabinoid antagonists

The whole body of data mentioned above highlights the role of the endocannabinoid system in feeding and energy balance regulation. Indeed, it was reasonable to hypothesize a therapeutic role for cannabinoid antagonists in the treatment of obesity. SR141716, also named rimonabant (commercialized as Acomplia), is now undergoing multicenter randomized, double-blind phase III trials to assess the effects on weight loss in obese patients with or without comorbidities with dyslipidemia and with type 2 diabetes (323). Moreover, the multitude of patents filed over the last few years claiming the synthesis of novel CB1 receptor antagonists reflects the intense competition in this area (123). Other compounds are under development, such as SLV-319 (Solvay, Weesp, The Netherlands) (129), which is undergoing phase I trials (323). However, at present, little is known about the results of these trials.
B. Clinical trial studies with rimonabant, the first CB1 receptor antagonist in clinical use to tackle obesity and obesity-related diseases

The CB1 receptor antagonist rimonabant was initially tested in humans not as an antiobesity drug but for its potential ability to reduce subjective intoxication and tachycardia in healthy subjects with a history of marijuana use or as an antipsychotic agent in schizophrenic patients. The first study showed that rimonabant was well tolerated by the participants even at a 90-mg dose (single oral dose). A significant dose-dependent blockade of marijuana effects was shown. However, the ability to reduce the intoxication induced by marijuana was very mild (130).

The results derived from the clinical trial in which rimonabant was tested to treat schizophrenia and schizoaffective disorders were not very satisfactory, because the effects of the drug in ameliorating clinical symptoms were not different from those obtained by placebo (324). However, in this trial, rimonabant treatment at 20 mg/d dosage was very well tolerated.

Bearing in mind the function of the endocannabinoid system in the mesolimbic rewarding system, rimonabant is also undergoing clinical trials as an aid to preventing the relapse of smoking cessation (323). Preliminary data from the STRATUS-US trial (smoking cessation in smokers motivated to quit) were recently presented at the 53rd Annual Scientific Session of the American College of Cardiology. The clinical study enrolled 787 smokers who received rimonabant at a dose of 5 or 20 mg or a placebo in a randomized fashion. The clinical trial lasted 10 wk, and the smokers were permitted to smoke during the first 2 wk but were asked to abstain from smoking after this period. The quit rate for subjects in the 20-mg rimonabant group was double that of the placebo group. In particular, the smokers characterized by overweight and obesity showed a relevant reduction in weight gain over the 10-wk treatment (325).

The most promising data seem to derive from rimonabant as a treatment for obesity. A phase II, 4-month, double blind, placebo-controlled study examined the effect of three different dosages of rimonabant (5, 10, or 20 mg/d) in obese patients with a body mass index between 30 and 40 kg/m2. Patients taking the 20-mg dose reported a weight loss of 4.4 kg in comparison to the 1.1-kg average in the placebo group. No significant adverse effects were noted. At the end of the treatment, weight loss was not maintained. However, the rebound in weight did not reach the pretreatment values (323). Another phase II, 7-d treatment, double-blind, placebo-controlled study was performed to evaluate hunger, calorie and fat intake. All these parameters were significantly reduced at the end of the short treatment, and the resulting average loss in body weight was 0.72 kg. The drug showed a good safety profile (323).

A large phase III trial named as RIO (rimonabant in obesity) was initiated in August 2001 including more than 6600 overweight or obese patients (323). All studies have already been concluded, and some of them are already reported in the literature (326, 327). Two of these studies, named RIO-North America and RIO-Europe, recruited obese and overweight patients with or without comorbidities who were treated for 2 yr with 5 or 20 mg rimonabant vs. placebo. The primary endpoints of the RIO-North America study were the absolute change in weight from baseline to 1 yr and the prevention of weight regain after rerandomization (second year), whereas the main endpoint of the RIO-Europe study was the assessment of weight reduction by using the same dosages. Secondary endpoints of both studies were the number of weight responders and the changes in waist circumference, metabolic and lipid parameters, and the number of patients affected by the metabolic syndrome as defined by National Cholesterol Education Program's Adult Treatment Program III (NCEP-ATP III) criteria (328). RIO-Lipids and RIO-Diabetes are the other two clinical trials with rimonabant aimed at investigating the amelioration, after treatment with the CB1 receptor antagonist, of specific comorbidity factors associated with obesity or overweight such as hyperlipidemia and diabetes. In the RIO-Lipids study, presented by the American College of Cardiology in New Orleans in March 2004, 1036 obese patients characterized by lipid profile alterations and body mass index of 27–40 kg/m2 were randomized to double-blind treatment with either placebo or rimonabant 5 or 20 mg/d (326). All patients were required to follow a reduced calorie diet. After 1 yr of therapy, patients in the 20-mg dose group showed a loss of 8.8 kg compared with the 2-kg reduction in the patients treated with placebo. Rimonabant was associated with an important and significant reduction in waist circumference, tryglicerides, and C reactive protein, whereas a significant increase in HDL-cholesterol was found in the 20-mg treatment group compared with the group of patients undergoing placebo treatment. Forty-three percent of patients in the 20-mg treatment cohort lost more than 10% of their initial body weight compared with the 10.3% observed in the placebo group. The number of patients in the 20-mg rimonabant group classified as having metabolic syndrome (according to NCEP-ATP III criteria) decreased from 52.9 to 25.8% after 1 yr. Rimonabant was generally well tolerated, and the most frequently reported side effects were gastrointestinal and upper respiratory tract symptoms (326).

Similar data have been obtained by the ad interim analysis of the first year treatment in the RIO-Europe study (327, 329). More than 67% of patients who completed treatment with 20 mg rimonabant achieved 5% or more weight loss, whereas 39% achieved 10% or more weight loss. The pattern of weight loss appeared to be sustained for up to 36–40 wk. A concomitant reduction in waist circumference of about 9 cm was observed in patients treated with 20 mg rimonabant. A significant improvement of lipid and glycemic profile was also observed in this study in patients with 20 mg rimonabant, with a significant increase in HDL-cholesterol (22% vs. 14% in placebo-treated patients) and a concomitant reduction of triglycerides (6.8% vs. an increase of 8.3% in placebo-treated patients). As expected by studies in the animals described above, the study of Van Gaal et al. (327) demonstrated that rimonabant adds a further important and significant weight-independent effect on lipid parameters to the positive effects derived from weight loss and waist reduction. In fact, as determined by statistical analysis, the effect of 20 mg rimonabant on both HDL-cholesterol and tryglicerides at 12 months has been shown to be partly independent of weight loss, being 60% of the increase in HDL-cholesterol and 45% of the reduction in trygliceride accounted for by weight loss, and the remainder due to reasons not related to body weight changes (327). Although Van Gaal et al. (327) proposed that a rise in adiponectin might be responsible for these relevant positive changes in lipid profile, other mechanisms might enter into play. Full understanding of these still unknown modes of action is urgently needed to better characterize the ideal phenotype of obese patients to be targeted with CB1 receptor antagonist drugs.

Rimonabant treatment was well tolerated, and the most common adverse events experienced with 20 mg rimonabant were gastrointestinal symptoms such as nausea and diarrhea and mood disorders such as anxiety and depression. However, the effects were found to be mild, and the discontinuation rate due to these events was similar between patients taking 20 mg rimonabant or placebo. The genesis of these adverse events might be explained by bearing in mind that, as explained above, CB1 receptor plays a role in gastrointestinal motility and in HPA axis activation. Nausea and diarrhea on the one hand and anxiety and depression on the other hand might be due to CB1 receptor pharmacological blockade.

Concerning studies in humans, a very recent report (330) confirms, on a genetic basis, the possible association between the chronic pathological overactivation of the endocannabinoid system and the development of obesity. In fact, in a large cohort of Caucasian and black subjects, overweight and obesity have been found to be associated with a polymorphism in FAAH. This genetic variant predicts a substitution of threonine for a highly conserved proline residue (P129T). It has been observed that patients carrying this polymorphism may have approximately half the enzymatic activity of FAAH. This may lead to a reduced inactivation of AEA and, eventually, to an inappropriate chronic increase of endocannabinoid tone (330). In such a context, a recent work (60) showed increased circulating levels of AEA and 2-AG in obese women when compared with a lean control group. Moreover, in the same study, a marked down-regulation of FAAH gene expression in adipose tissue of obese women has been found, suggesting that the increased endocannabinoid levels may be secondary to decreased enzymatic degradation (60).
Previous SectionNext Section
VI. Summary and Perspectives

A number of studies show that the endocannabinoid system profoundly influences both hormone secretion and metabolic processes. Animal models have represented the ideal tool for advancing the understanding of the mechanisms of these functions. However, the data derived from early studies were not always straightforward in the conclusions. The contradictory results had to be largely attributed to the heterogeneous variety of substances, dosages, and routes of administration used in each experimental model. Studies in humans with marijuana or Δ9-THC were even more contradictory in their conclusions, because no standardization of dose was used and no stringent criteria (i.e., randomization) of patient recruitment were defined in nearly all the experimental models.

However, the generation of CB1−/− mice and the introduction of CB1 receptor antagonists initially in animal models and later in humans provided a remarkable stimulus to better characterize the functions of the endocannabinoid system in the regulation of hormone secretion and metabolic processes (Fig. 3⇓).
Fig. 3.
View larger version:

In this page
In a new window

Download as PowerPoint Slide

Fig. 3.

Actions of CB1 antagonists on the target organs involved in food intake and metabolic control. Schematic drawing illustrating the main sites of action of CB1 antagonists in the control of energy balance

As a general conclusion, the endocannabinoid system appears to play a very important regulatory role in the secretion of hormones related to reproductive functions and to stress responses. These observations have led to some important clinical considerations. High levels of endocannabinoids seem to negatively affect reproduction by acting at different sites. It is therefore possible to speculate about a clinical use of CB1 receptor antagonists to ameliorate gonadotropin pulsatility or to improve fertilization capability. On the other hand, endocannabinoids are important modulators in the physiological response of the HPA axis during repetitive stress conditions and in pathological conditions, such as anxiety, phobias, depression, and posttraumatic stress disorders (16, 147). Moreover, the endocannabinoid system has been proposed as playing an important role in protection against neurotoxicity and, possibly, certain forms of epilepsy (115, 331, 332). Drugs presumed to increase endocannabinoid tone are therefore currently proposed as a new therapeutical frontier to treat anxiety-related disorders and neurodegenerative diseases (82). The use of drugs acting as antagonists of CB1 receptor should thus be carefully monitored when administered, for instance, to patients with anxiety traits, epilepsy, or neurodegenerative disorders.

The anecdotes regarding the orexigenic properties of marijuana have nowadays been substantiated by an impressive number of reports that make it possible to definitively include cannabinoids in the large family of orexigenic signals. This large body of data provided the basis to establish a novel approach to tackle obesity and related disorders by means, as strongly suggested by the clinical trials with rimonabant, of a CB1 receptor antagonist.

During the last few years, it has become evident that multiple mechanisms of action, not solely limited to the CNS, are involved in the endocannabinoid-mediated control of food intake and energy balance. The full understanding of these modes of action may lead to the identification of the particular types of obesity where treatment with CB1 receptor antagonists work most efficiently. The potential clinical use of rimonabant will also help us to clarify how the endocannabinoid system affects the physiological functions and the pathological diseases related to hormonal secretion and energy balance.
Previous SectionNext Section
Note Added in Proof

After this manuscript was accepted, the complete RIO-Lipids study was published (see Refs.326 and 333).
Previous SectionNext Section

Previous SectionNext Section
Footnotes

This work was supported by Grant LSHM-CT-2003-503041 from the Sixth European Community Program (to U.P.).

First Published Online November 23, 2005

Abbreviations: AEA, Arachidonoyl ethanolamide; 2-AG, 2-arachidonoyl-glycerol; CB1−/−, mice lacking cannabinoid receptor type 1; CB1 receptor, cannabinoid receptor type 1; CB2 receptor, cannabinoid receptor type 2; CCK, cholecystokinin; CNS, central nervous system; D1, dopamine receptor type 1; D2, dopamine receptor type 2; DIO, diet-induced obesity; FAAH, fatty acid amide hydrolase; GABA, γ-aminobutyric acid; HDL, high-density lipoprotein; HPA, hypothalamus-pituitary-adrenal; IHC, immunohistochemistry; ISH, in situ hybridization; MCR4, melanocortin receptor type 4; OEA, oleoylethanolamide; PPAR, peroxisome proliferator-activated receptor; PRL, prolactin; PVN, paraventricular nucleus; RIO, rimonabant in obesity; Δ9-THC, Δ9-tetrahydrocannabinol; UCP, uncoupling protein; VTA, ventral tegmental area.

Previous Section

References


Peters H, Nahas GG 1999 A brief history of four millennia (B.C. 2000-A.D. 1974). In: Nahas GG, Sutin KM, Harvey D, Agurell S, eds. Marihuana and medicine. Totowa, NJ: Humana Press; 3–7

Gaoni Y, Mechoulam R 1964 Isolation, structure and partial synthesis of an active constituent of hashish. J Am Chem Soc 86:1646–1647
CrossRef

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

Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, Gopher A, Almog S, Martin BR, Compton DR, Pertwee RG, Griffin G, Bayewitch M, Barg J, Vogel Z 1995 Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol 50:83–90
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

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

Di Marzo V, Melck D, Bisogno T, De Petrocellis L 1998 Endocannabinoids: endogenous cannabinoid receptor ligands with neuromodulatory action. Trends Neurosci 21:521–528
CrossRefMedline

Panikashvili D, Simeonidou C, Ben-Shabat S, Hanus L, Breuer A, Mechoulam R, Shohami E 2001 An endogenous cannabinoid (2-AG) is neuroprotective after brain injury. Nature 413:527–531
CrossRefMedline

Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, Azad SC, Cascio MG, Gutierrez SO, van der Stelt M., Lopez-Rodriguez ML, Casanova E, Schutz G, Zieglgansberger W, Di Marzo V, Behl C, Lutz B 2003 CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science 302:84–88
Abstract/FREE Full Text

Panikashvili D, Mechoulam R, Beni SM, Alexandrovich A, Shohami E 2005 CB1 cannabinoid receptors are involved in neuroprotection via NF-κB inhibition. J Cereb Blood Flow Metab 25:477–484
CrossRefMedline

Cravatt BF, Lichtman AH 2004 The endogenous cannabinoid system and its role in nociceptive behavior. J Neurobiol 61:149–160
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

Wotjak CT 2005 Role of endogenous cannabinoids in cognition and emotionality. Mini Rev Med Chem 5:659–670
CrossRefMedline

Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, Hermann H, Tang J, Hofmann C, Zieglgansberger W, Di Marzo V, Lutz B 2002 The endogenous cannabinoid system controls extinction of aversive memories. Nature 418:530–534
CrossRefMedline

Varvel SA, Lichtman AH 2002 Evaluation of CB1 receptor knockout mice in the Morris water maze. J Pharmacol Exp Ther 301:915–924
Abstract/FREE Full Text

Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, Mackie K, Stella N 2003 Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci 23:1398–1405
Abstract/FREE Full Text

Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, Friedman H 2003 The cannabinoid system and immune modulation. J Leukoc Biol 74:486–496
Abstract/FREE Full Text

Massa F, Marsicano G, Hermann H, Cannich A, Monory K, Cravatt BF, Ferri GL, Sibaev A, Storr M, Lutz B 2004 The endogenous cannabinoid system protects against colonic inflammation. J Clin Invest 113:1202–1209
CrossRefMedline

Mendizabal VE, Adler-Graschinsky E 2003 Cannabinoid system as a potential target for drug development in the treatment of cardiovascular disease. Curr Vasc Pharmacol 1:301–313
CrossRefMedline

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

Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, De Costa BR, Rice KC 1990 Cannabinoid receptor localization in brain. Proc Natl Acad Sci USA 87:1932–1936
Abstract/FREE Full Text

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

Ibrahim MM, Porreca F, Lai J, Albrecht PJ, Rice FL, Khodorova A, Davar G, Makriyannis A, Vanderah TW, Mata HP, Malan Jr TP 2005 CB2 cannabinoid receptor activation produces antinociception by stimulating peripheral release of endogenous opioids. Proc Natl Acad Sci USA 102:3093–3098
Abstract/FREE Full Text

Begg M, Pacher P, Batkai S, Osei-Hyiaman D, Offertaler L, Mo FM, Liu J, Kunos G 2005 Evidence for novel cannabinoid receptors. Pharmacol Ther 106:133–145
CrossRefMedline

van der Stelt M, Trevisani M, Vellani V, De Petrocellis L, Schiano-Moriello A, Campi B, McNaughton P, Geppetti P, Di Marzo V 2005 Anandamide acts as an intracellular messenger amplifying Ca(2+) influx via TRPV1 channels. EMBO J 24:3026–3037
CrossRefMedline

Maingret F, Patel AJ, Lazdunski M, Honore E 2001 The endocannabinoid anandamide is a direct and selective blocker of the background K(+) channel TASK-1. EMBO J 20:47–54
CrossRefMedline

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

Mo FM, Offertaler L, Kunos G 2004 Atypical cannabinoid stimulates endothelial cell migration via a Gi/Go-coupled receptor distinct from CB1, CB2 or EDG-1. Eur J Pharmacol 489:21–27
CrossRefMedline

Baker D, Pryce G, Davis WL, Hiley CR, In silico patent searching reveals a new cannabinoid receptor. Trends Pharmacol Sci, in press

Herkenham M, Lynn AB, Johnson MR, Melvin LS, De Costa BR, Rice KC 1991 Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci 11:563–583
Abstract

Matsuda LA, Bonner TI, Lolait SJ 1993 Localization of cannabinoid receptor mRNA in rat brain. J Comp Neurol 327:535–550
CrossRefMedline

Westlake TM, Howlett AC, Bonner TI, Matsuda LA, Herkenham M 1994 Cannabinoid receptor binding and messenger RNA expression in human brain: an in vitro receptor autoradiography and in situ hybridization histochemistry study of normal aged and Alzheimer's brains. Neuroscience 63:637–652
CrossRefMedline

Marsicano G, Lutz B 1999 Expression of the cannabinoid receptor CB1 in distinct neuronal subpopulations in the adult mouse forebrain. Eur J Neurosci 11:4213–4225
CrossRefMedline

Glass M, Dragunow M, Faull RL 1997 Cannabinoid receptors in the human brain: a detailed anatomical and quantitative autoradiographic study in the fetal, neonatal and adult human brain. Neuroscience 77:299–318
CrossRefMedline

Tsou K, Brown S, Sanudo-Pena MC, Mackie K, Walker JM 1998 Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience 83:393–411
CrossRefMedline

Pettit DA, Harrison MP, Olson JM, Spencer RF, Cabral GA 1998 Immunohistochemical localization of the neural cannabinoid receptor in rat brain. J Neurosci Res 51:391–402
CrossRefMedline

Egertova M, Cravatt BF, Elphick MR 2003 Comparative analysis of fatty acid amide hydrolase and CB1 cannabinoid receptor expression in the mouse brain: evidence of a widespread role for fatty acid amide hydrolase in regulation of endocannabinoid signaling. Neuroscience 119:481–496
CrossRefMedline

Katona I, Sperlagh B, Sik A, Kafalvi A, Vizi ES, Mackie K, Freund TF 1999 Presynaptically located CB1 cannabinoid receptors regulate GABA release from axon terminals of specific hippocampal interneurons. J Neurosci 19:4544–4558
Abstract/FREE Full Text

Sim LJ, Selley DE, Childers SR 1995 In vitro autoradiography of receptor-activated G proteins in rat brain by agonist-stimulated guanylyl 5′-[γ-[35S]thio]-triphosphate binding. Proc Natl Acad Sci USA 92:7242–7246
Abstract/FREE Full Text

Breivogel CS, Childers SR 1998 The functional neuroanatomy of brain cannabinoid receptors. Neurobiol Dis 5:417–431
CrossRefMedline

Katona I, Sperlagh B, Magloczky Z, Santha E, Kofalvi A, Czirjak S, Mackie K, Vizi ES, Freund TF 2000 GABAergic interneurons are the targets of cannabinoid actions in the human hippocampus. Neuroscience 100:797–804
CrossRefMedline

Cota D, Marsicano G, Lutz B, Vicennati V, Stalla GK, Pasquali R, Pagotto U 2003 Endogenous cannabinoid system as a modulator of food intake. Int J Obes Relat Metab Disord 27:289–301
CrossRefMedline

Wenger T, Moldrich G, Furst S 2003 Neuromorphological background of cannabis addiction. Brain Res Bull 61:125–128
CrossRefMedline

Kofalvi A, Rodrigues RJ, Ledent C, Mackie K, Vizi ES, Cunha RA, Sperlagh B 2005 Involvement of cannabinoid receptors in the regulation of neurotransmitter release in the rodent striatum: a combined immunochemical and pharmacological analysis. J Neurosci 25:2874–2884
Abstract/FREE Full Text

Breivogel CS, Selley DE, Childers SR 1998 Cannabinoid receptor agonist efficacy for stimulating [35S]GTPγS binding to rat cerebellar membranes correlates with agonist-induced decreases in GDP affinity. J Biol Chem 273:16865–16873
Abstract/FREE Full Text

Gonzalez S, Bisogno T, Wenger T, Manzanares J, Milone A, Berrendero F, Di Marzo V, Ramos JA, Fernandez-Ruiz JJ 2000 Sex steroid influence on cannabinoid CB1 receptor mRNA and endocannabinoid levels in the anterior pituitary gland. Biochem Biophys Res Commun 270:260–266
CrossRefMedline

Wenger T, Fernandez-Ruiz JJ, Ramos JA 1999 Immunocytochemical demonstration of CB1 cannabinoid receptors in the anterior lobe of the pituitary gland. J Neuroendocrinol 11:873–878
CrossRefMedline

Cesa R, Guastalla A, Cottone E, Mackie K, Beltramo M, Franzoni MF 2002 Relationships between CB1 cannabinoid receptors and pituitary endocrine cells in Xenopus laevis: an immunohistochemical study. Gen Comp Endocrinol 125:17–24
CrossRefMedline

Pagotto U, Marsicano G, Fezza F, Theodoropoulou M, Gruebler Y, Stalla J, Arzberger T, Milone A, Losa M, Di Marzo V, Lutz B, Stalla GK 2001 Normal human pituitary gland and pituitary adenomas express cannabinoid receptor type 1 and synthesize endogenous cannabinoids: first evidence for a direct role of cannabinoids on hormone modulation at the human pituitary level. J Clin Endocrinol Metab 86:2687–2696
Abstract/FREE Full Text

Evans CO, Young AN, Brown MR, Brat DJ, Parks JS, Neish AS, Oyesiku NM 2001 Novel patterns of gene expression in pituitary adenomas identified by complementary deoxyribonucleic acid microarrays and quantitative reverse transcription-polymerase chain reaction. J Clin Endocrinol Metab 86:3097–3107
Abstract/FREE Full Text

Gonzalez S, Manzanares J, Berrendero F, Wenger T, Corchero J, Bisogno T, Romero J, Fuentes JA, Di Marzo V, Ramos JA, Fernandez-Ruiz JJ 1999 Identification of endocannabinoids and cannabinoid CB1 receptor mRNA in the pituitary gland. Neuroendocrinology 70:137–145
CrossRefMedline

Buckley NE, Hansson S, Harta G, Mezey E 1998 Expression of the CB1 and CB2 receptor messenger RNAs during embryonic development in the rat. Neuroscience 82:1131–1149
CrossRefMedline

Porcella A, Marchese G, Casu MA, Rocchitta A, Lai ML, Gessa GL, Pani L 2002 Evidence for functional CB1 cannabinoid receptor expressed in the rat thyroid. Eur J Endocrinol 147:255–261
Abstract

Galiegue S, Mary S, Marchand J, Dussossoy D, Carriere D, Carayon P, Bouaboula M, Shire D, Le Fur G, Casellas P 1995 Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem 232:54–61
Medline

Cota D, Marsicano G, Tschoep M, Gruebler Y, Flachskamm C, Schubert M, Auer D, Yassouridis A, Thone-Reineke C, Ortmann S, Tomassoni F, Cervino C, Nisoli E, Linthorst AC, Pasquali R, Lutz B, Stalla GK, Pagotto U 2003 The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J Clin Invest 112:423–431
CrossRefMedline

Bensaid M, Gary-Bobo M, Esclangon A, Maffrand JP, Le Fur G, Oury-Donat F, Soubrié P 2003 The cannabinoid CB1 receptor antagonist SR141716 increases Acrp30 mRNA expression in adipose tissue of obese fa/fa rats and in cultured adipocyte cells. Mol Pharmacol 63:908–914
Abstract/FREE Full Text

Engeli S, Bohnke J, Feldpausch M, Gorzelniak K, Janke J, Batkai S, Pacher P, Harvey-White J, Luft FC, Sharma AM, Jordan J 2005 Activation of the peripheral endocannabinoid system in human obesity. Diabetes 54:2838–2843
Abstract/FREE Full Text

Liu YL, Connoley IP, Wilson CA, Stock MJ 2005 Effects of the cannabinoid CB1 receptor antagonist SR141716 on oxygen consumption and soleus muscle glucose uptake in Lep(ob)/Lep(ob) mice. Int J Obes Relat Metab Disord 29:183–187
CrossRefMedline

Osei-Hyiaman D, DePetrillo M, Pacher P, Liu J, Radaeva S, Batkai S, Harvey-White J, Mackie K, Offertaler L, Wang L, Kunos G 2005 Endocannabinoid activation at hepatic CB1 receptors stimulates fatty acid synthesis and contributes to diet-induced obesity. J Clin Invest 115:1298–1305
CrossRefMedline

Siegmund SV, Uchinami H, Osawa Y, Brenner DA, Schwabe RF 2005 Anandamide induces necrosis in primary hepatic stellate cells. Hepatology 41:1085–1095
CrossRefMedline

Pertwee RG 2001 Cannabinoids and the gastrointestinal tract. Gut 48:859–867
Abstract/FREE Full Text

Buyse M, Ovesjo ML, Goiot H, Guilmeau S, Peranzi G, Moizo L, Walker F, Lewin MJ, Meister B, Bado A 2001 Expression and regulation of leptin receptor proteins in afferent and efferent neurons of the vagus nerve. Eur J Neurosci 14:64–72
CrossRefMedline

Burdyga G, Lal S, Spiller D, Jiang W, Thompson D, Attwood S, Saeed S, Grundy D, Varro A, Dimaline R, Dockray GJ 2003 Localization of orexin-1 receptors to vagal afferent neurons in the rat and humans. Gastroenterology 124:129–139
CrossRefMedline

Burdyga G, Lal S, Varro A, Dimaline R, Thompson DG, Dockray GJ 2004 Expression of cannabinoid CB1 receptors by vagal afferent neurons is inhibited by cholecystokinin. J Neurosci 24:2708–2715
Abstract/FREE Full Text

Cani PD, Montoya ML, Neyrinck AM, Delzenne NM, Lambert DM 2004 Potential modulation of plasma ghrelin and glucagon-like peptide-1 by anorexigenic cannabinoid compounds, SR141716A (rimonabant) and oleoylethanolamide. Br J Nutr 92:757–761
CrossRefMedline

van der Lely AJ, Tschoep M, Heiman ML, Ghigo E 2004 Biological, physiological, pathophysiological, and pharmacological aspects of ghrelin. Endocr Rev 25:426–457
Abstract/FREE Full Text

Gerard CM, Mollereau C, Vassart G, Parmentier M 1991 Molecular cloning of a human cannabinoid receptor which is also expressed in testis. Biochem J 279:129–134
Medline

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

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

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

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

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

Treinen KA, Sneeden JL, Heindel JJ 1993 Specific inhibition of FSH-stimulated cAMP accumulation by Δ9-tetrahydrocannabinol in cultured rat granulosa cells. Toxicol Appl Pharmacol 118:53–57
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

Dennedy MC, Friel AM, Houlihan DD, Broderick VM, Smith T, Morrison JJ 2004 Cannabinoids and the human uterus during pregnancy. Am J Obstet Gynecol 190:2–9
CrossRefMedline

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

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

Park B, Gibbons HM, Mitchell MD, Glass M 2003 Identification of the CB1 cannabinoid receptor and fatty acid amide hydrolase (FAAH) in the human placenta. Placenta 24:990–995
CrossRefMedline

Mackie K 19 August 2005 Cannabinoid receptors as therapeutic targets. Annu Rev Pharmacol Toxicol 10.1146/annurev.pharmtox.46.120604.141254

Howlett AC 2004 Efficacy in CB1 receptor-mediated signal transduction. Br J Pharmacol 142:1209–1218
CrossRefMedline

Diaz-Laviada I, Ruiz-Llorente L 2005 Signal transduction activated by cannabinoid receptors. Mini Rev Med Chem 5:619–630
CrossRefMedline

Fowler CJ, Holt S, Nilsson O, Jonsson KO, Tiger G, Jacobsson SO 2005 The endocannabinoid signaling system: pharmacological and therapeutic aspects. Pharmacol Biochem Behav 81:248–262
CrossRefMedline

Glass M, Felder CC 1997 Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors augments cAMP accumulation in striatal neurons: evidence for a Gs linkage to the CB1 receptor. J Neurosci 17:5327–5333
Abstract/FREE Full Text

Kearn CS, Blake-Palmer K, Daniel E, Mackie K, Glass M 2005 Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: a mechanism for receptor crosstalk? Mol Pharmacol 67:1697–1704
Abstract/FREE Full Text

Hilairet S, Bouaboula M, Carriere D, Le Fur G, Casellas P 2003 Hypersensitization of the orexin 1 receptor by the CB1 receptor: evidence for cross-talk blocked by the specific CB1 antagonist, SR141716. J Biol Chem 278:23731–23737
Abstract/FREE Full Text

Devlin MG, Christopoulos A 2002 Modulation of cannabinoid agonist binding by 5-HT in the rat cerebellum. J Neurochem 80:1095–1102
CrossRefMedline

Howlett AC 2002 The cannabinoid receptors. Prostaglandins Other Lipid Mediat 68- 69:619–631

Bradshaw HB, Walker JM 2005 The expanding field of cannabimimetic and related lipid mediators. Br J Pharmacol 144:459–465
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

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

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

Leggett JD, Aspley S, Beckett SR, D'Antona AM, Kendall DA, Kendall DA 2004 Oleamide is a selective endogenous agonist of rat and human CB1 cannabinoid receptors. Br J Pharmacol 141:253–262
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

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

Dinh TP, Kathuria S, Piomelli D 2004 RNA interference suggests a primary role for monoacylglycerol lipase in the degradation of the endocannabinoid 2-arachidonoylglycerol. Mol Pharmacol 66:1260–1264
Abstract/FREE Full Text

Barnett-Norris J, Lynch D, Reggio PH 2005 Lipids, lipid rafts and caveolae: their importance for GPCR signaling and their centrality to the endocannabinoid system. Life Sci 77:1625–1639
CrossRefMedline

McFarland MJ, Barker EL 2004 Anandamide transport. Pharmacol Ther 104:117–135
CrossRefMedline

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

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

Di S, Malcher-Lopes R, Halmos KC, Tasker JG 2003 Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J Neurosci 23:4850–4857
Abstract/FREE Full Text

Hashimotodani Y, Ohno-Shosaku T, Tsubokawa H, Ogata H, Emoto K, Maejima T, Araishi K, Shin HS, Kano M 2005 Phospholipase C b serves as a coincidence detector through its Ca2+ dependency for triggering retrograde endocannabinoid signal. Neuron 45:257–268
CrossRefMedline

McFarland MJ, Porter AC, Rakhshan FR, Rawat DS, Gibbs RA, Barker EL 2004 A role for caveolae/lipid rafts in the uptake and recycling of the endogenous cannabinoid anandamide. J Biol Chem 279:41991–41997
Abstract/FREE Full Text

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

Alger BE 2002 Retrograde signaling in the regulation of synaptic transmission: focus on endocannabinoids. Prog Neurobiol 68:247–286
CrossRefMedline

Melis M, Pistis M, Perra S, Muntoni AL, Pillolla G, Gessa GL 2004 Endocannabinoids mediate presynaptic inhibition of glutamatergic transmission in rat ventral tegmental area dopamine neurons through activation of CB1 receptors. J Neurosci 24:53–62
Abstract/FREE Full Text

Melis M, Perra S, Muntoni AL, Pillolla G, Lutz B, Marsicano G, Di Marzo V, Gessa GL, Pistis M 2004 Prefrontal cortex stimulation induces 2-arachidonoyl-glycerol-mediated suppression of excitation in dopamine neurons. J Neurosci 24:10707–10715
Abstract/FREE Full Text

Bacci A, Huguenard JR, Prince DA 2004 Long-lasting self-inhibition of neocortical interneurons mediated by endocannabinoids. Nature 431:312–316
CrossRefMedline

Stella N 2004 Cannabinoid signaling in glial cells. Glia 48:267–277
CrossRefMedline

Chevaleyre V, Castillo PE 2004 Endocannabinoid-mediated metaplasticity in the hippocampus. Neuron 43:871–881
CrossRefMedline

Gerdeman G, Lovinger DM 2001 CB1 cannabinoid receptor inhibits synaptic release of glutamate in rat dorsolateral striatum. J Neurophysiol 85:468–471
Abstract/FREE Full Text

Azad SC, Monory K, Marsicano G, Cravatt BF, Lutz B, Zieglgansberger W, Rammes G 2004 Circuitry for associative plasticity in the amygdala involves endocannabinoid signaling. J Neurosci 24:9953–9961
Abstract/FREE Full Text

Mechoulam R, Spatz M, Shohami E 2002 Endocannabinoids and neuroprotection. Sci STKE 2002:RE5

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

Parker LA, Mechoulam R, Schlievert C 2002 Cannabidiol, a non-psychoactive component of cannabis and its synthetic dimethylheptyl homolog suppress nausea in an experimental model with rats. Neuroreport 13:567–570
CrossRefMedline

Hanus L, Breuer A, Tchilibon S, Shiloah S, Goldenberg D, Horowitz M, Pertwee RG, Ross RA, Mechoulam R, Fride E 1999 HU-308: a specific agonist for CB2, a peripheral cannabinoid receptor. Proc Natl Acad Sci USA 96:14228–14233
Abstract/FREE Full Text

Calignano A, La Rana G, Giuffrida A, Piomelli D 1998 Control of pain initiation by endogenous cannabinoids. Nature 394:277–281
CrossRefMedline

Beltramo M, Stella N, Calignano A, Lin SY, Makriyannis A, Piomelli D 1997 Functional role of high-affinity anandamide transport, as revealed by selective inhibition. Science 277:1094–1097
Abstract/FREE Full Text

De Petrocellis L, Bisogno T, Davis JB, Pertwee RG, Di Marzo V 2000 Overlap between the ligand recognition properties of the anandamide transporter and the VR1 vanilloid receptor: inhibitors of anandamide uptake with negligible capsaicin-like activity. FEBS Lett 483:52–56

de Lago E, Fernandez-Ruiz JJ, Ortega-Gutierrez S, Viso A, Lopez-Rodriguez ML, Ramos JA 2002 UCM707, a potent and selective inhibitor of endocannabinoid uptake, potentiates hypokinetic and antinociceptive effects of anandamide. Eur J Pharmacol 449:99–103
CrossRefMedline

Lange JH, Kruse CG 2004 Recent advances in CB1 cannabinoid receptor antagonists. Curr Opin Drug Discov Devel 7:498–506
Medline

Gatley SJ, Gifford AN, Volkow ND, Lan R, Makriyannis A 1996 123I-labeled AM251: a radioiodinated ligand which binds in vivo to mouse brain cannabinoid CB1 receptors. Eur J Pharmacol 307:331–338
CrossRefMedline

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

Rinaldi-Carmona M, Barth F, Congy C, Martinez S, Oustric D, Perio A, Poncelet M, Maruani J, Arnone M, Finance O, Soubrié P, Le Fur G 2004 SR147778 [5-(4-bromophenyl)-1-(2,4-dichlorophenyl)-4-ethyl-N-(1-piperidinyl)-1H-pyrazole-3-carboxamide], a new potent and selective antagonist of the CB1 cannabinoid receptor: biochemical and pharmacological characterization. J Pharmacol Exp Ther 310:905–914
Abstract/FREE Full Text

Lan R, Gatley J, Lu Q, Fan P, Fernando SR, Volkow ND, Pertwee R, Makriyannis A 1999 Design and synthesis of the CB1 selective cannabinoid antagonist AM281: a potential human SPECT ligand. AAPS PharmSci 1:E4

Felder CC, Joyce KE, Briley EM, Glass M, Mackie KP, Fahey KJ, Cullinan GJ, Hunden DC, Johnson DW, Chaney MO, Koppel GA, Brownstein M 1998 LY320135, a novel cannabinoid CB1 receptor antagonist, unmasks coupling of the CB1 receptor to stimulation of cAMP accumulation. J Pharmacol Exp Ther 284:291–297
Abstract/FREE Full Text

Lange JH, Coolen HK, van Stuivenberg HH, Dijksman JA, Herremans AH, Ronken E, Keizer HG, Tipker K, McCreary AC, Veerman W, Wals HC, Stork B, Verveer PC, den Hartog AP, de Jong NM, Adolfs TJ, Hoogendoorn J, Kruse CG 2004 Synthesis, biological properties, and molecular modeling investigations of novel 3,4-diarylpyrazolines as potent and selective CB1 cannabinoid receptor antagonists. J Med Chem 47:627–643
CrossRefMedline

Huestis MA, Gorelick DA, Heishman SJ, Preston KL, Nelson RA, Moolchan ET, Frank RA 2001 Blockade of effects of smoked marijuana by the CB1-selective cannabinoid receptor antagonist SR141716. Arch Gen Psychiatry 58:322–328
Abstract/FREE Full Text

Dutta AK, Sard H, Ryan W, Razdan RK, Compton DR, Martin BR 1994 The synthesis and pharmacological evaluation of the cannabinoid antagonist SR141716A. Med Chem Res 5:54–62

Pertwee RG 2005 Inverse agonism and neutral antagonism at cannabinoid CB1 receptors. Life Sci 76:1307–1324
CrossRefMedline

Bouaboula M, Perrachon S, Milligan L, Canat X, Rinaldi-Carmona M, Portier M, Barth F, Calandra B, Pecceu F, Lupker J, Maffrand JP, Le Fur G, Casellas P 1997 A selective inverse agonist for central cannabinoid receptor inhibits mitogen-activated protein kinase activation stimulated by insulin or insulin-like growth factor 1. Evidence for a new model of receptor/ligand interactions. J Biol Chem 272:22330–22339
Abstract/FREE Full Text

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

Murphy LL, Munoz RM, Adrian BA, Villanua MA 1998 Function of cannabinoid receptors in the neuroendocrine regulation of hormone secretion. Neurobiol Dis 5:432–446
CrossRefMedline

Dewey WL, Peng TC, Harris LS 1970 The effect of 1-trans-Δ9-tetrahydrocannabinol on the hypothalamo-hypophyseal-adrenal axis of rats. Eur J Pharmacol 12:382–384
CrossRefMedline

Rodriguez de Fonseca F, Rubio P, Menzaghi F, Merlo-Pich E, Rivier J, Koob GF, Navarro M 1996 Corticotropin-releasing factor (CRF) antagonist [D-Phe12, Nle21,38,C α MeLeu37]CRF attenuates the acute actions of the highly potent cannabinoid receptor agonist HU-210 on defensive-withdrawal behavior in rats. J Pharmacol Exp Ther 276:56–64
Abstract/FREE Full Text

Corchero J, Fuentes JA, Manzanares J 1999 Chronic treatment with CP-55,940 regulates corticotropin releasing factor and proopiomelanocortin gene expression in the hypothalamus and pituitary gland of the rat. Life Sci 64:905–911
CrossRefMedline

Weidenfeld J, Feldman S, Mechoulam R 1994 Effect of the brain constituent anandamide, a cannabinoid receptor agonist, on the hypothalamo-pituitary-adrenal axis in the rat. Neuroendocrinology 59:110–112
Medline

Wenger T, Jamali KA, Juaneda C, Leonardelli J, Tramu G 1997 Arachidonyl ethanolamide (anandamide) activates the parvocellular part of hypothalamic paraventricular nucleus. Biochem Biophys Res Commun 237:724–728
CrossRefMedline

Kubena RK, Barry HI 1970 Interactions of D-tetrahydrocannabinol with barbiturates and methamphetamine. J Pharmacol Exp Ther 173:94–100
Abstract/FREE Full Text

Barna I, Zelena D, Arszovszki AC, Ledent C 2004 The role of endogenous cannabinoids in the hypothalamo-pituitary-adrenal axis regulation: in vivo and in vitro studies in CB1 receptor knockout mice. Life Sci 75:2959–2970
CrossRefMedline

Navarro M, Hernandez E, Munoz RM, Del Arco I, Villanua MA, Carrera MR, Rodriguez de Fonseca F 1997 Acute administration of the CB1 cannabinoid receptor antagonist SR 141716A induces anxiety-like responses in the rat. Neuroreport 8:491–496
Medline

Arevalo C, de Miguel R, Hernandez-Tristan R 2001 Cannabinoid effects on anxiety-related behaviours and hypothalamic neurotransmitters. Pharmacol Biochem Behav 70:123–131
CrossRefMedline

De Kloet ER 2003 Hormones, brain and stress. Endocr Regul 37:51–68
Medline

Patel S, Roelke CT, Rademacher DJ, Cullinan WE, Hillard CJ 2004 Endocannabinoid signaling negatively modulates stress-induced activation of the hypothalamic-pituitary-adrenal axis. Endocrinology 145:5431–5438
Abstract/FREE Full Text

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

Uriguen L, Perez-Rial S, Ledent C, Palomo T, Manzanares J 2004 Impaired action of anxiolytic drugs in mice deficient in cannabinoid CB1 receptors. Neuropharmacology 46:966–973
CrossRefMedline

Benowitz NL, Jones RT, Lerner CB 1976 Depression of growth hormone and cortisol response to insulin-induced hypoglycemia after prolonged oral Δ9-tetrahydrocannabinol administration in man. J Clin Endocrinol Metab 42:938–941
Abstract/FREE Full Text

Kokka N, Garcia JF 1974 Effects of Δ9-THC on growth hormone and ACTH secretion in rats. Life Sci 15:329–338
CrossRefMedline

Martin-Calderon JL, Munoz RM, Villanua MA, Del Arco I, Moreno JL, Rodriguez de Fonseca F, Navarro M 1998 Characterization of the acute endocrine actions of (−)-11-hydroxy-D8-tetrahydrocannabinol-dimethylheptyl (HU-210), a potent synthetic cannabinoid in rats. Eur J Pharmacol 344:77–86
CrossRefMedline

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

Rettori V, Wenger T, Snyder G, Dalterio S, McCann SM 1988 Hypothalamic action of Δ9-tetrahydrocannabinol to inhibit the release of prolactin and growth hormone in the rat. Neuroendocrinology 47:498–503
Medline

Rettori V, Aguila MC, Gimeno MF, Franchi AM, McCann SM 1990 In vitro effect of Δ9-tetrahydrocannabinol to stimulate somatostatin release and block that of luteinizing hormone-releasing hormone by suppression of the release of prostaglandin E2. Proc Natl Acad Sci USA 87:10063–10066
Abstract/FREE Full Text

Tucci SA, Rogers EK, Korbonits M, Kirkham TC 2004 The cannabinoid CB1 receptor antagonist SR141716 blocks the orexigenic effects of intrahypothalamic ghrelin. Br J Pharmacol 143:520–523
CrossRefMedline

Lomax P 1970 The effect of marihuana on pituitary-thyroid activity in the rat. Agents Actions 1:252–257
CrossRefMedline

Hillard CJ, Farber NE, Hagen TC, Bloom AS 1984 The effects of Δ9-tetrahydrocannabinol on serum thyrotropin levels in the rat. Pharmacol Biochem Behav 20:547–550
CrossRefMedline

Miras CJ 1965 Some aspects of cannabis action. In: Wolstenhome GE, Knight J, eds. Hashish: its chemistry and pharmacology. Boston: Brown; 37–52

Kramer J, Ben-David M 1978 Prolactin suppression by (−) Δ9-tetrahydrocannabinol (THC): involvement of serotonergic and dopaminergic pathways. Endocrinology 103:452–457
Abstract/FREE Full Text

Asch RH, Smith CG, Siler-Khodr TM, Pauerstein CJ 1979 Acute decreases in serum prolactin concentrations caused by Δ9-tetrahydrocannabinol in nonhuman primates. Fertil Steril 32:571–575
Medline

Hughes Jr CL, Everett JW, Tyrey L 1981 Δ9-Tetrahydrocannabinol suppression of prolactin secretion in the rat: lack of direct pituitary effect. Endocrinology 109:876–880
Abstract/FREE Full Text

Scorticati C, Mohn C, De Laurentiis A, Vissio P, Fernandez SJ, Seilicovich A, McCann SM, Rettori V 2003 The effect of anandamide on prolactin secretion is modulated by estrogen. Proc Natl Acad Sci USA 100:2134–2139
Abstract/FREE Full Text

Rodriguez de Fonseca F, Wenger T, Navarro M, Murphy LL 1999 Effects of Δ9-THC on VIP-induced prolactin secretion in anterior pituitary cultures: evidence for the presence of functional cannabinoid CB1 receptors in pituitary cells. Brain Res 841:114–122
CrossRefMedline

Daley JD, Branda LA, Rosenfeld J, Younglai EV 1974 Increase of serum prolactin in male rats by (−)-trans-Δ9-tetrahydrocannabinol. J Endocrinol 63:415–416
Abstract/FREE Full Text

Murphy LL, Steger RW, Smith MS, Bartke A 1990 Effects of Δ9-tetrahydrocannabinol, cannabinol and cannabidiol, alone and in combinations, on luteinizing hormone and prolactin release and on hypothalamic neurotransmitters in the male rat. Neuroendocrinology 52:316–321
CrossRefMedline

Murphy LL, Newton SC, Dhali J, Chavez D 1991 Evidence for a direct anterior pituitary site of Δ9-tetrahydrocannabinol action. Pharmacol Biochem Behav 40:603–607
CrossRefMedline

Fernandez-Ruiz JJ, Munoz RM, Romero J, Villanua MA, Makriyannis A, Ramos JA 1997 Time course of the effects of different cannabimimetics on prolactin and gonadotrophin secretion: evidence for the presence of CB1 receptors in hypothalamic structures and their involvement in the effects of cannabimimetics. Biochem Pharmacol 53:1919–1927
CrossRefMedline

Rodriguez de Fonseca F, Cebeira M, Fernandez-Ruiz JJ, Navarro M, Ramos JA 1991 Effects of pre- and perinatal exposure to hashish extracts on the ontogeny of brain dopaminergic neurons. Neuroscience 43:713–723
CrossRefMedline

Bonnin A, Ramos JA, Rodriguez de Fonseca F, Cebeira M, Fernandez-Ruiz JJ 1993 Acute effects of Δ9-tetrahydrocannabinol on tuberoinfundibular dopamine activity, anterior pituitary sensitivity to dopamine and prolactin release vary as a function of estrous cycle. Neuroendocrinology 58:280–286
Medline

Murphy LL, Rodriguez de Fonseca F, Steger RW 1991 Δ9-tetrahydrocannabinol antagonism of the anterior pituitary response to estradiol in immature female rats. Steroids 56:97–102
CrossRefMedline

Ho BY, Stadnicka A, Prather PL, Buckley AR, Current LL, Bosnjak ZJ, Kwok WM 2000 Cannabinoid CB1 receptor-mediated inhibition of prolactin release and signaling mechanisms in GH4C1 cells. Endocrinology 141:1675–1685
Abstract/FREE Full Text

Wenger T, Toth BE, Martin BR 1995 Effects of anandamide (endogen cannabinoid) on anterior pituitary hormone secretion in adult ovariectomized rats. Life Sci 56:2057–2063
CrossRefMedline

Besch NF, Smith CG, Besch PK, Kaufman RH 1977 The effect of marihuana (Δ9-tetrahydrocannabinol) on the secretion of luteinizing hormone in the ovariectomized rhesus monkey. Am J Obstet Gynecol 128:635–642
Medline

Tyrey L 1978 Δ9-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 Δ9-tetrahydrocannabinol (THC) on female reproductive function. Adv Biosci 22- 23:449–467

Asch RH, Smith CG, Siler-Khodr TM, Pauerstein CJ 1981 Effects of Δ9-tetrahydrocannabinol during the follicular phase of the rhesus monkey (Macaca mulatta). J Clin Endocrinol Metab 52:50–55
Abstract/FREE Full Text

Smith CG, Almirez RG, Berenberg J, Asch RH 1983 Tolerance develops to the disruptive effects of Δ9-tetrahydrocannabinol on primate menstrual cycle. Science 219:1453–1455
Abstract/FREE Full Text

Mendelson JH, Cristofaro P, Ellingboe J, Benedikt R, Mello NK 1985 Acute effects of marihuana on luteinizing hormone in menopausal women. Pharmacol Biochem Behav 23:765–768
CrossRefMedline

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 1999 Marihuana effects on pituitary and gonadal hormones in women. In: Nahas GG, Sutin KM, Harvey D, Agurell S, eds. Marihuana and medicine. Totowa, NJ: Humana Press; 385–392

Shen M, Piser TM, Seybold VS, Thayer SA 1996 Cannabinoid receptor agonists inhibit glutamatergic synaptic transmission in rat hippocampal cultures. J Neurosci 16:4322–4334
Abstract/FREE Full Text

Rodriguez de Fonseca F, Fernandez-Ruiz JJ, Murphy LL, Cebeira M, Steger RW, Bartke A, Ramos JA 1992 Acute effects of Δ9-tetrahydrocannabinol on dopaminergic activity in several rat brain areas. Pharmacol Biochem Behav 42:269–275
CrossRefMedline

de Miguel R, Romero J, Munoz RM, Garcia-Gil L, Gonzalez S, Villanua MA, Makriyannis A, Ramos JA, Fernandez-Ruiz JJ 1998 Effects of cannabinoids on prolactin and gonadotrophin secretion: involvement of changes in hypothalamic γ-aminobutyric acid (GABA) inputs. Biochem Pharmacol 56:1331–1338
CrossRefMedline

Kumar MS, Simpkins JW 1983 Naloxone blocks the effects of Δ9-tetrahydrocannabinol on serum luteinizing hormone and prolactin in rats. Subst Alcohol Actions Misuse 4:347–353
Medline

Jackson AL, Murphy LL 1997 Role of the hypothalamic-pituitary-adrenal axis in the suppression of luteinizing hormone release by Δ9-tetrahydrocannabinol. Neuroendocrinology 65:446–452
Medline

Maldonado R 2002 Study of cannabinoid dependence in animals. Pharmacol Ther 95:153–164
CrossRefMedline

Rodriguez de Fonseca F, Cebeira M, Ramos JA, Martin M, Fernandez-Ruiz JJ 1994 Cannabinoid receptors in rat brain areas: sexual differences, fluctuations during estrous cycle and changes after gonadectomy and sex steroid replacement. Life Sci 54:159–170
CrossRefMedline

Turley Jr WA, Floody OR 1981 Δ9-Tetrahydrocannabinol stimulates receptive and proceptive sexual behaviors in female hamsters. Pharmacol Biochem Behav 14:745–747
CrossRefMedline

Mani SK, Mitchell A, O'Malley BW 2001 Progesterone receptor and dopamine receptors are required in Δ9-tetrahydrocannabinol modulation of sexual receptivity in female rats. Proc Natl Acad Sci USA 98:1249–1254
Abstract/FREE Full Text

Stella N 2001 How might cannabinoids influence sexual behavior? Proc Natl Acad Sci USA 98:793–795
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

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

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

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

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

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, 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

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

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

Helliwell RJ, Chamley LW, Blake-Palmer K, Mitchell MD, Wu J, Kearn CS, Glass M 2004 Characterization of the endocannabinoid system in early human pregnancy. J Clin Endocrinol Metab 89:5168–5174
Abstract/FREE Full Text

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

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

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

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

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

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

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

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

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

De Petrocellis L, Melck D, Bisogno T, Milone A, Di Marzo V 1999 Finding of the endocannabinoid signalling system in Hydra, a very primitive organism: possible role in the feeding response. Neuroscience 92:377–387
CrossRefMedline

Fride E, Ginzburg Y, Breuer A, Bisogno T, Di Marzo V, Mechoulam R 2001 Critical role of the endogenous cannabinoid system in mouse pup suckling and growth. Eur J Pharmacol 419:207–214
CrossRefMedline

Abel EL, McMillan DE, Harris LS 1972 Tolerance to the behavioral and hypothermic effects of 1–9-tetrahydrocannabinol in neonatal chicks. Experientia 28:1188–1189
CrossRefMedline

Dewey WL, Jenkins J, O'Rourke T, Harris LS 1972 The effects of chronic administration of trans-9-tetrahydrocannabinol on behavior and the cardiovascular system of dogs. Arch Int Pharmacodyn Ther 198:118–131
Medline

Huy ND, Gailis L, Roy PE 1974 Effects on dogs of 3 months of inhalation of marijuana and tobacco smoke. Union Med Can 103:65–71
Medline

Thompson GR, Rosenkrantz H, Schaeppi UH, Braude MC 1973 Comparison of acute oral toxicity of cannabinoids in rats, dogs and monkeys. Toxicol Appl Pharmacol 25:363–372
CrossRefMedline

Vaupel DB, Morton EC 1982 Anorexia and hyperphagia produced by five pharmacologic classes of hallucinogens. Pharmacol Biochem Behav 17:539–545
CrossRefMedline

Huy ND, Gailis L, Cote G, Roy PE 1975 Effects of chronic administration of Δ9-transtetrahydrocannabinol (Δ9-THC) in guinea-pigs. Int J Clin Pharmacol Biopharm 12:284–289
Medline

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

Scheckel CL, Boff E, Dahlen P, Smart T 1968 Behavioral effects in monkeys of racemates of two biologically active marijuana constituents. Science 160:1467–1469
Abstract/FREE Full Text

Grunfeld Y, Edery H 1969 Psychopharmacological activity of the active constituents of hashish and some related cannabinoids. Psychopharmacologia 14:200–210
CrossRefMedline

McMillan DE, Harris LS, Frankenheim JM, Kennedy JS 1970 I-Δ9-Trans-tetrahydrocannabinol in pigeons: tolerance to the behavioral effects. Science 169:501–503
Abstract/FREE Full Text

Banerjee BN, Sofia RD, Erikson D, Ivins NJ 1976 Toxicity of Δ9-transtetrahydrocannabinol (THC) administered subcutaneously for 13 days to female rabbits. J Toxicol Environ Health 1:769–776
Medline

Carlini EA, Kramer C 1965 Effects of Cannabis sativa (marihuana) on maze performance of the rat. Psychopharmacologia 7:175–181
CrossRefMedline

Borgen LA, Davis WM, Pace HB 1971 Effects of synthetic 9 -tetrahydrocannabinol on pregnancy and offspring in the rat. Toxicol Appl Pharmacol 20:480–486
CrossRefMedline

Gonzalez SC, Karniol IG, Carlini EL 1972 Effects of Cannabis sativa extract on conditioned fear. Behav Biol 7:83–94
CrossRefMedline

Rating D, Broermann I, Honecker H, Kluwe S, Coper H 1972 Effect of subchronic treatment with (−)8-trans-tetrahydrocannabinol (8-THC) on food intake, body temperature, hexobarbital sleeping time and hexobarbital elimination in rats. Psychopharmacologia 27:349–357
Medline

Graham JD, Li DM 1973 Cardiovascular and respiratory effects of cannabis in cat and rat. Br J Pharmacol 49:1–10
Medline

Nahas GG, Schwartz IW, Adamec J, Manger WM 1973 Tolerance of Δ9-tetrahydrocannabinol in the spontaneously hypertensive rat. Proc Soc Exp Biol Med 142:58–60
Abstract/FREE Full Text

Fernandes M, Schabarek A, Coper H, Hill R 1974 Modification of Δ9-tetrahydrocannabinol actions by cannabinol and cannabidiol in the rat. Psychopharmacologia 38:329–338
Medline

Gluck JP, Ferraro DP 1974 Effects of Δ9-THC on food and water intake of deprivation experienced rats. Behav Biol 11:395–401
CrossRefMedline

Sofia RD, Barry III H 1974 Acute and chronic effects of Δ9-tetrahydrocannabinol on food intake by rats. Psychopharmacologia 39:213–222
Medline

Johansson JO, Jarbe TU, Henriksson BG 1975 Acute and subchronic influences of tetrahydrocannabinols on water and food intake, body weight, and temperature in rats. TIT J Life Sci 5:17–27
Medline

Sofia RD, Knobloch LC 1976 Comparative effects of various naturally occurring cannabinoids on food, sucrose and water consumption by rats. Pharmacol Biochem Behav 4:591–599
CrossRefMedline

Drewnowski A, Grinker JA 1978 Temporal effects of Δ9-tetrahydrocannabinol on feeding patterns and activity of obese and lean Zucker rats. Behav Biol 23:112–117
CrossRefMedline

Drewnowski A, Grinker JA 1978 Food and water intake, meal patterns and activity of obese and lean Zucker rats following chronic and acute treatment with Δ9-tetrahydrocannabinol. Pharmacol Biochem Behav 9:619–630
CrossRefMedline

Anderson-Baker WC, McLaughlin CL, Baile CA 1979 Oral and hypothalamic injections of barbiturates, benzodiazepines and cannabinoids and food intake in rats. Pharmacol Biochem Behav 11:487–491
CrossRefMedline

Miczek KA, Dixit BN 1980 Behavioral and biochemical effects of chronic Δ9-tetrahydrocannabinol in rats. Psychopharmacology (Berl) 67:195–202
CrossRefMedline

Milano WC, Wild KD, Hui YZ, Hubbell CL, Reid LD 1988 PCP, THC, ethanol, and morphine and consumption of palatable solutions. Pharmacol Biochem Behav 31:893–897
CrossRefMedline

Trojniar W, Wise RA 1991 Facilitory effect of Δ9-tetrahydrocannabinol on hypothalamically induced feeding. Psychopharmacology (Berl) 103:172–176
CrossRefMedline

McLaughlin CL, Baile CA, Bender PE 1979 Cannabinols and feeding in sheep. Psychopharmacology (Berl) 64:321–323
CrossRefMedline

Van Den Broek GW, Robertson J, Keim DA, Baile CA 1979 Feeding and depression of abomasal secretion in sheep elicited by elfazepam and 9-aza-cannabinol. Pharmacol Biochem Behav 11:51–56
CrossRefMedline

Rosenkrantz H, Braude M 1974 Acute, subacute and 23-day chronic marihuana inhalation toxicities in the rat. Toxicol Appl Pharmacol 28:428–441
CrossRefMedline

Abel EL 1975 Cannabis: effects on hunger and thirst. Behav Biol 15:255–281
CrossRefMedline

Williams CM, Rogers PJ, Kirkham TC 1998 Hyperphagia in pre-fed rats following oral Δ9-THC. Physiol Behav 65:343–346
CrossRefMedline

Williams CM, Kirkham TC 2002 Reversal of Δ9-THC hyperphagia by SR141716 and naloxone but not dexfenfluramine. Pharmacol Biochem Behav 71:333–340
CrossRefMedline

Greenberg I, Kuehnle J, Mendelson JH, Bernstein JG 1976 Effects of marihuana use on body weight and caloric intake in humans. Psychopharmacology (Berl) 49:79–84
CrossRefMedline

Foltin RW, Brady JV, Fischman MW 1986 Behavioral analysis of marijuana effects on food intake in humans. Pharmacol Biochem Behav 25:577–582
CrossRefMedline

Foltin RW, Brady JV, Fischman MW, Emurian CS, Dominitz J 1987 Effects of smoked marijuana on social interaction in small groups. Drug Alcohol Depend 20:87–93
CrossRefMedline

Cat LK, Coleman RL 1994 Treatment for HIV wasting syndrome. Ann Pharmacother 28:595–597
Medline

Beal JE, Olson R, Lefkowitz L, Laubenstein L, Bellman P, Yangco B, Morales JO, Murphy R, Powderly W, Plasse TF, Mosdell KW, Shepard KV 1997 Long-term efficacy and safety of dronabinol for acquired immunodeficiency syndrome-associated anorexia. J Pain Symptom Manage 14:7–14
CrossRefMedline

Gorter RW 1999 Cancer cachexia and cannabinoids. Forsch Komplementarmed 6(Suppl 3):21–22

Sallan SE, Zinberg NE, Frei EI 1975 Antiemetic effect of Δ9-tetrahydrocannabinol in patients receiving cancer chemotherapy. N Engl J Med 293:795–797
Medline

Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ 1997 Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer's disease. Int J Geriatr Psychiatry 12:913–919
CrossRefMedline

Plasse TF, Gorter RW, Krasnow SH, Lane M, Shepard KV, Wadleigh RG 1991 Recent clinical experience with dronabinol. Pharmacol Biochem Behav 40:695–700
CrossRefMedline

Gorter R, Seefried M, Volberding P 1992 Dronabinol effects on weight in patients with HIV infection. AIDS 6:127
Medline

Struwe M, Kaempfer SH, Geiger CJ, Pavia AT, Plasse TF, Shepard KV, Ries K, Evans TG 1993 Effect of dronabinol on nutritional status in HIV infection. Ann Pharmacother 27:827–831
Abstract

Beal JE, Olson R, Laubenstein L, Morales JO, Bellman P, Yangco B, Lefkowitz L, Plasse TF, Shepard KV 1995 Dronabinol as a treatment for anorexia associated with weight loss in patients with AIDS. J Pain Symptom Manage 10:89–97
CrossRefMedline

Grinspoon S, Carr A 2005 Cardiovascular risk and body-fat abnormalities in HIV-infected adults. N Engl J Med 352:48–62
CrossRefMedline

Williams CM, Kirkham TC 1999 Anandamide induces overeating: mediation by central cannabinoid (CB1) receptors. Psychopharmacology (Berl) 143:315–317
CrossRefMedline

Kirkham TC, Williams CM 2001 Endogenous cannabinoids and appetite. Nutr Res Reviews 14:65–86

Hao S, Avraham Y, Mechoulam R, Berry EM 2000 Low dose anandamide affects food intake, cognitive function, neurotransmitter and corticosterone levels in diet-restricted mice. Eur J Pharmacol 392:147–156
CrossRefMedline

Kirkham TC, Williams CM, Fezza F, Di Marzo V 2002 Endocannabinoid levels in rat limbic forebrain and hypothalamus in relation to fasting, feeding and satiation: stimulation of eating by 2-arachidonoyl glycerol. Br J Pharmacol 136:550–557
CrossRefMedline

Williams CM, Kirkham TC 2002 Observational analysis of feeding induced by Δ9-THC and anandamide. Physiol Behav 76:241–250
CrossRefMedline

Arnone M, Maruani J, Chaperon F, Thiebot MH, Poncelet M, Soubrié P, Le Fur G 1997 Selective inhibition of sucrose and ethanol intake by SR 141716, an antagonist of central cannabinoid (CB1) receptors. Psychopharmacology (Berl) 132:104–106
CrossRefMedline

Gallate JE, McGregor IS 1999 The motivation for beer in rats: effects of ritanserin, naloxone and SR 141716. Psychopharmacology (Berl) 142:302–308
CrossRefMedline

Freedland CS, Poston JS, Porrino LJ 2000 Effects of SR141716A, a central cannabinoid receptor antagonist, on food-maintained responding. Pharmacol Biochem Behav 67:265–270
CrossRefMedline

Simiand J, Keane M, Keane PE, Soubrié P 1998 SR 141716, a CB1 cannabinoid receptor antagonist, selectively reduces sweet food intake in marmoset. Behav Pharmacol 9:179–181
Medline

Verty AN, McGregor IS, Mallet PE 2004 Consumption of high carbohydrate, high fat, and normal chow is equally suppressed by a cannabinoid receptor antagonist in non-deprived rats. Neurosci Lett 354:217–220
CrossRefMedline

Higgs S, Williams CM, Kirkham TC 2003 Cannabinoid influences on palatability: microstructural analysis of sucrose drinking after Δ9-tetrahydrocannabinol, anandamide, 2-arachidonoyl glycerol and SR141716. Psychopharmacology (Berl) 165:370–377
Medline

Chaperon F, Soubrié P, Puech AJ, Thiebot MH 1998 Involvement of central cannabinoid (CB1) receptors in the establishment of place conditioning in rats. Psychopharmacology (Berl) 135:324–332
CrossRefMedline

Ledent C, Valverde O, Cossu G, Petitet F, Aubert JF, Beslot F, Bohme GA, Imperato A, Pedrazzini T, Roques BP, Vassart G, Fratta W, Parmentier M 1999 Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science 283:401–404
Abstract/FREE Full Text

Arnold JC, Hunt GE, McGregor IS 2001 Effects of the cannabinoid receptor agonist CP55,940 and the cannabinoid receptor antagonist SR 141716 on intracranial self-stimulation in Lewis rats. Life Sci 70:97–108
CrossRefMedline

Gardner EL, Vorel SR 1998 Cannabinoid transmission and reward-related events. Neurobiol Dis 5:502–533
CrossRefMedline

Spanagel R, Weiss F 1999 The dopamine hypothesis of reward: past and current status. Trends Neurosci 22:521–527
CrossRefMedline

Hernandez L, Hoebel BG 1988 Food reward and cocaine increase extracellular dopamine in the nucleus accumbens as measured by microdialysis. Life Sci 42:1705–1712
CrossRefMedline

Rusk IN, Cooper SJ 1989 The selective dopamine D1 receptor agonist SK&F 38393: its effects on palatability- and deprivation-induced feeding, and operant responding for food. Pharmacol Biochem Behav 34:17–22
CrossRefMedline

Bisogno T, Berrendero F, Ambrosino G, Cebeira M, Ramos JA, Fernandez-Ruiz JJ, Di Marzo V 1999 Brain regional distribution of endocannabinoids: implications for their biosynthesis and biological function. Biochem Biophys Res Commun 256:377–380
CrossRefMedline

Hermann H, Marsicano G, Lutz B 2002 Coexpression of the cannabinoid receptor type 1 with dopamine and serotonin receptors in distinct neuronal subpopulations of the adult mouse forebrain. Neuroscience 109:451–460
CrossRefMedline

Berridge KC, Robinson TE 1998 What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Res Brain Res Rev 28:309–369
CrossRefMedline

Verty AN, McGregor IS, Mallet PE 2004 The dopamine receptor antagonist SCH 23390 attenuates feeding induced by Δ9-tetrahydrocannabinol. Brain Res 1020:188–195
CrossRefMedline

Corchero J, Manzanares J, Fuentes JA 2004 Cannabinoid/opioid crosstalk in the central nervous system. Crit Rev Neurobiol 16:159–172
CrossRefMedline

Bakshi VP, Kelley AE 1993 Feeding induced by opioid stimulation of the ventral striatum: role of opiate receptor subtypes. J Pharmacol Exp Ther 265:1253–1260
Abstract/FREE Full Text

Jackson HC, Sewell RD 1985 Are d-opioid receptors involved in the regulation of food and water intake? Neuropharmacology 24:885–888
CrossRefMedline

Verty AN, Singh ME, McGregor IS, Mallet PE 2003 The cannabinoid receptor antagonist SR 141716 attenuates overfeeding induced by systemic or intracranial morphine. Psychopharmacology (Berl) 168:314–323
CrossRefMedline

Kirkham TC, Williams CM 2001 Synergistic effects of opioid and cannabinoid antagonists on food intake. Psychopharmacology (Berl) 153:267–270
CrossRefMedline

Rowland NE, Mukherjee M, Robertson K 2001 Effects of the cannabinoid receptor antagonist SR 141716, alone and in combination with dexfenfluramine or naloxone, on food intake in rats. Psychopharmacology (Berl) 159:111–116
CrossRefMedline

Vickers SP, Dourish CT, Kennett GA 2001 Evidence that hypophagia induced by d-fenfluramine and d-norfenfluramine in the rat is mediated by 5-HT2C receptors. Neuropharmacology 41:200–209
CrossRefMedline

Vettor R, Serra R, Fabris R, Pagano C, Federspil G 2005 Effect of sibutramine on weight management and metabolic control in type 2 diabetes: a meta-analysis of clinical studies. Diabetes Care 28:942–949
Abstract/FREE Full Text

Flier JS 2004 Obesity wars: molecular progress confronts an expanding epidemic. Cell 116:337–350
CrossRefMedline

Erickson JC, Clegg KE, Palmiter RD 1996 Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381:415–421
CrossRefMedline

Wynne K, Stanley S, McGowan B, Bloom S 2005 Appetite control. J Endocrinol 184:291–318
Abstract/FREE Full Text

Di Marzo V, Goparaju SK, Wang L, Liu J, Batkai S, Jarai Z, Fezza F, Miura GI, Palmiter RD, Sugiura T, Kunos G 2001 Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 410:822–825
CrossRefMedline

Hanus L, Avraham Y, Ben-Shushan D, Zolotarev O, Berry EM, Mechoulam R 2003 Short-term fasting and prolonged semistarvation have opposite effects on 2-AG levels in mouse brain. Brain Res 983:144–151
CrossRefMedline

Harrold JA, Elliott JC, King PJ, Widdowson PS, Williams G 2002 Down-regulation of cannabinoid-1 (CB-1) receptors in specific extrahypothalamic regions of rats with dietary obesity: a role for endogenous cannabinoids in driving appetite for palatable food? Brain Res 952:232–238
CrossRefMedline

Jamshidi N, Taylor DA 2001 Anandamide administration into the ventromedial hypothalamus stimulates appetite in rats. Br J Pharmacol 134:1151–1154
CrossRefMedline

Horvath TL 2003 Endocannabinoids and the regulation of body fat: the smoke is clearing. J Clin Invest 112:323–326
CrossRefMedline

Verty AN, McFarlane JR, McGregor IS, Mallet PE 2004 Evidence for an interaction between CB1 cannabinoid and melanocortin MCR-4 receptors in regulating food intake. Endocrinology 145:3224–3231
CrossRefMedline

Dallman MF, Akana SF, Strack AM, Scribner KS, Pecoraro N, La Fleur SE, Houshyar H, Gomez F 2004 Chronic stress-induced effects of corticosterone on brain: direct and indirect. Ann NY Acad Sci 1018:141–150
CrossRefMedline

Verty AN, McFarlane JR, McGregor IS, Mallet PE 2004 Evidence for an interaction between CB1 cannabinoid and oxytocin receptors in food and water intake. Neuropharmacology 47:593–603
CrossRefMedline

Seeley RJ, York DA 2005 Fuel sensing and the central nervous system (CNS): implications for the regulation of energy balance and the treatment for obesity. Obes Rev 6:259–265
CrossRefMedline

Loftus TM, Jaworsky DE, Frehywot GL, Townsend CA, Ronnett GV, Lane MD, Kuhajda FP 2000 Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288:2379–2381
Abstract/FREE Full Text

Kim EK, Miller I, Landree LE, Borisy-Rudin FF, Brown P, Tihan T, Townsend CA, Witters LA, Moran TH, Kuhajda FP, Ronnett GV 2002 Expression of FAS within hypothalamic neurons: a model for decreased food intake after C75 treatment. Am J Physiol Endocrinol Metab 283:E867–E879

Colombo G, Agabio R, Diaz G, Lobina C, Reali R, Gessa GL 1998 Appetite suppression and weight loss after the cannabinoid antagonist SR 141716. Life Sci 63:L113–L117

Ravinet-Trillou C, Delgorge C, Menet C, Arnone M, Soubrié P 2004 CB1 cannabinoid receptor knockout in mice leads to leanness, resistance to diet-induced obesity and enhanced leptin sensitivity. Int J Obes Relat Metab Disord 28:640–648
CrossRefMedline

Jbilo O, Ravinet-Trillou C, Arnone M, Buisson I, Bribes E, Peleraux A, Penarier G, Soubrié P, Le Fur G, Galiegue S, Casellas P 2005 The CB1 receptor antagonist rimonabant reverses the diet-induced obesity phenotype through the regulation of lipolysis and energy balance. FASEB J 19:1567–1569
Abstract/FREE Full Text

Chandran M, Phillips SA, Ciaraldi T, Henry RR 2003 Adiponectin: more than just another fat cell hormone? Diabetes Care 26:2442–2450
FREE Full Text

Poirier B, Bidouard JP, Cadrouvele C, Marniquet X, Staels B, O'Connor SE, Janiak P, Herbert JM 2005 The anti-obesity effect of rimonabant is associated with an improved serum lipid profile. Diabetes Obes Metab 7:65–72
CrossRefMedline

Ravinet-Trillou C, Arnone M, Delgorge C, Gonalons N, Keane P, Maffrand JP, Soubrié P 2003 Anti-obesity effect of SR141716, a CB1 receptor antagonist, in diet-induced obese mice. Am J Physiol Regul Integr Comp Physiol 284:R345–R353

Hildebrandt AL, Kelly-Sullivan DM, Black SC 2003 Antiobesity effects of chronic cannabinoid CB1 receptor antagonist treatment in diet-induced obese mice. Eur J Pharmacol 462:125–132
CrossRefMedline

Shearman LP, Zhou D, Rosko KM, Camacho RE, Stribling D, MacIntyre D, Strack AM, Multiple day CB1R inverse agonist effects on hypothalamic gene expression and energy balance in diet-induced obese mice. Proc 34th Annual Meeting of the Society for Neuroscience, San Diego, CA, 2004 (Abstract 76.4)

Tzavara ET, Perry KW, Rodriguez DE, Bymaster FP, Nomikos GG 2001 The cannabinoid CB1 receptor antagonist SR141716A increases norepinephrine outflow in the rat anterior hypothalamus. Eur J Pharmacol 426:R3–R4

Kola B, Hubina E, Tucci SA, Kirkham TC, Garcia EA, Mitchell SE, Williams LM, Hawley SA, Hardie DG, Grossman AB, Korbonits M 2005 Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. J Biol Chem 280:25196–25201
Abstract/FREE Full Text

Schwabe RF 2005 Endocannabinoids promote hepatic lipogenesis and steatosis through CB1 receptors. Hepatology 42:959–961
CrossRef

Gomez R, Navarro M, Ferrer B, Trigo JM, Bilbao A, Del Arco I, Cippitelli A, Nava F, Piomelli D, Rodriguez de Fonseca F 2002 A peripheral mechanism for CB1 cannabinoid receptor-dependent modulation of feeding. J Neurosci 22:9612–9617
Abstract/FREE Full Text

Rodriguez de Fonseca F, Navarro M, Gomez R, Escuredo L, Nava F, Fu J, Murillo-Rodriguez E, Giuffrida A, LoVerme J, Gaetani S, Kathuria S, Gall C, Piomelli D 2001 An anorexic lipid mediator regulated by feeding. Nature 414:209–212
CrossRefMedline

Fu J, Gaetani S, Oveisi F, LoVerme J, Serrano A, Rodriguez de Fonseca F, Rosengarth A, Luecke H, Di Giacomo B, Tarzia G, Piomelli D 2003 Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-α. Nature 425:90–93
CrossRefMedline

Proulx K, Cota D, Castaneda TR, Tschoep M, D'Alessio DA, Tso P, Woods SC, Seeley RJ 2005 Mechanisms of oleoylethanolamide-induced changes in feeding behavior and motor activity. Am J Physiol Regul Integr Comp Physiol 289:R729–R737

Guzman M, LoVerme J, Fu J, Oveisi F, Blazquez C, Piomelli D 2004 Oleoylethanolamide stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activated receptor α (PPAR-α). J Biol Chem 279:27849–27854
Abstract/FREE Full Text

Oveisi F, Gaetani S, Eng KT, Piomelli D 2004 Oleoylethanolamide inhibits food intake in free-feeding rats after oral administration. Pharmacol Res 49:461–466
CrossRefMedline

Nielsen MJ, Petersen G, Astrup A, Hansen HS 2004 Food intake is inhibited by oral oleoylethanolamide. J Lipid Res 45:1027–1029
Abstract/FREE Full Text

Fernandez JR, Allison DB 2004 Rimonabant Sanofi-Synthelabo. Curr Opin Investig Drugs 5:430–435
Medline

Meltzer HY, Arvanitis L, Bauer D, Rein W 2004 Placebo-controlled evaluation of four novel compounds for the treatment of schizophrenia and schizoaffective disorder. Am J Psychiatry 161:975–984
Abstract/FREE Full Text

Anthenelli RM, Despres JP, Effects of rimonabant in the reduction of major cardiovascular risk factors. Results from the STRATUS-US trial (smoking cessation in smokers motivated to quit). Proc Annual Meeting of the American College of Cardiology, New Orleans, LA, 2004

Cleland JG, Ghosh J, Freemantle N, Kaye GC, Nasir M, Clark AL, Coletta AP 2004 Clinical trials update and cumulative meta-analyses from the American College of Cardiology: WATCH, SCD-HeFT, DINAMIT, CASINO, INSPIRE, STRATUS-US, RIO-Lipids and cardiac resynchronisation therapy in heart failure. Eur J Heart Fail 6:501–508
Abstract/FREE Full Text

Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rossner S, for the RIO-Europe Study Group 2005 Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet 365:1389–1397
CrossRefMedline

2002 Third Report of the National Cholesterol Educational Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (Adult Treatment Panel III) final report. Circulation 106:3143–3421

Pagotto U, Pasquali R 2005 Fighting obesity and associated risk factors by antagonising cannabinoid type 1 receptors. Lancet 365:1363–1364
CrossRefMedline

Sipe JC, Waalen J, Gerber A, Beutler E 2005 Overweight and obesity associated with a missense polymorphism in fatty acid amide hydrolase (FAAH). Int J Obes Relat Metab Disord 29:755–759
CrossRefMedline

Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Huffman JW, Layward L 2000 Cannabinoids control spasticity and tremor in a multiple sclerosis model. Nature 404:84–87
CrossRefMedline

Parmentier-Batteur S, Jin K, Mao XO, Xie L, Greenberg DA 2002 Increased severity of stroke in CB1 cannabinoid receptor knock-out mice. J Neurosci 22:9771–9775
Abstract/FREE Full Text

Despres JP, Golay A, Sjostrum L, for the Rimonabant in Obesity-Lipids Study Group 2005 Effects of rimonabant on body weight and the metabolic syndrome in overweight patients. N Engl J Med 353:2121–2134


Source: The Emerging Role of the Endocannabinoid System in Endocrine Regulation and Energy Balance
 
Back
Top Bottom