CB1 Receptors In The Preoptic Anterior Hypothalamus Regulate WIN 55212-2

Jacob Bell

New Member
CB1 Receptors in the Preoptic Anterior Hypothalamus Regulate WIN 55212-2 [(4,5-Dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo[3,2,1ij]quinolin-6-one]-Induced Hypothermia

S. M. Rawls,
Jose Cabassa,
Ellen B. Geller and
Martin W. Adler

- Author Affiliations

Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, Pennsylvania

Dr. Scott M. Rawls, Department of Pharmacology, Temple University School of Medicine, 3420 N. Broad Street, Philadelphia, PA 19140. E-mail: smrawls28@hotmail.com


Next Section
Abstract

The present study investigated the effect of the selective cannabinoid agonist, WIN 55212-2 [(4,5-dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo[3,2,1ij]quinolin-6-one], on body temperature. WIN 55212-2 (1, 2.5, 5, and 10 mg/kg, i.m.) induced hypothermia in a dose-dependent manner. The peak hypothermia occurred 60 to 180 min postinjection. Body temperature was still suppressed 5 h after the injection of the highest dose of WIN 55212-2. The selective CB1 antagonist, SR141716A [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride] (5 and 10 mg/kg, i.m.), blocked the WIN 55212-2-induced hypothermia, suggesting that CB1 receptor activation mediated the hypothermia. In contrast, the selective CB2antagonist, SR144528 [N-{(1S)-endo-1,3,3-trimethyl bicyclo heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide}] (5 mg/kg, i.m.), did not alter the WIN 55212-2-induced hypothermia. Neither SR141716A nor SR144528 alone altered body temperature. WIN 55212-2 (1—30 μg/μl) injected directly into the preoptic anterior hypothalamic nucleus (POAH) induced hypothermia in an immediate and dose-dependent fashion. The hypothermia produced by intra-POAH injection of WIN 55212-2 was brief, with body temperature returning to baseline 60 min postinjection. SR141716A (5 mg/kg, i.m.) abolished the hypothermia induced by intra-POAH injection of WIN 55212-2 (30 μg/μl), indicating that CB1 receptors in the POAH mediated the hypothermia. The present results confirm the idea that CB1 receptors mediate the hypothermic response to cannabinoid agonists. Moreover, the present data suggest that 1) the POAH is the central locus for thermoregulation, and 2) CB1receptors within the POAH are the primary mediators of cannabinoid-induced hypothermia.

Cannabis and its derivative compounds, collectively known as cannabinoids, produce an array of pharmacological symptoms in animals and humans (Chaperon and Thiebot, 1999), including hypothermia (Fitton and Pertwee, 1982; Ovadia et al., 1995). Two subtypes of receptors, CB1 and CB2, mediate cannabinoid-induced effects (Howlett, 1995; Felder et al., 1998). CB1 receptors are located primarily in the central nervous system and are thought to mediate the central effects of cannabinoids (Howlett, 1995). In contrast, CB2receptors are expressed almost exclusively by peripheral immune cells (Dragic et al., 1996).

The development of synthetic cannabinoid agonists has provided remarkable advances in cannabis research. One such ligand is the aminoalkylindole, (+)-WIN 55,212-2 [(4,5-dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo[3,2,1ij]quinolin-6-one] (WIN 55212-2), which displays high selectivity for cannabinoid receptors. Previous studies have demonstrated that WIN 55212-2 is highly potent and efficacious in vivo and in vitro. WIN 55212-2 prevents intravenous cocaine self-administration, increases tail-flick reflexes, exerts antihyperalgesic effects, and induces hypothermia in rats, indicating that WIN 55212-2 is pharmacologically active in vivo (Compton et al., 1992; Martin et al., 1998; Fox et al., 2001). WIN 55212-2 undergoes less nonspecific binding than classical cannabinoids and interacts negligibly with other neurotransmitter systems and ion channels (Jansen et al., 1992; Shire et al., 1996; Showalter et al., 1996). In contrast, Δ9-tetrahydrocannabinol (Δ9-THC) has been reported to produce hypothermia by interacting with other neurotransmitters, including serotonin (Davies and Graham, 1980). WIN 55212-2 is also more readily soluble than the extremely hydrophobic Δ9-THC. The actions of Δ9-THC in vivo have been investigated by suspending the Δ9-THC in solution using a suspending agent such as polyvinylpyrrolidone (Davies and Graham, 1980). In contrast, WIN 55212-2 is soluble in a Cremophor/saline vehicle, which has better dissolution characteristics than a suspension because it remains a homogenous solution even upon standing for extended periods of time. In addition, Cremophor/saline does not alter body temperature or require the use of ethanol, which has been commonly used to administer Δ9-THC (Compton et al., 1992, 1996; Aceto et al., 1995). This is crucial, since ethanol is a centrally acting depressant that may alter the neurochemical and behavioral effects of Δ9-THC. Thus, we chose WIN 55212-2 to induce hypothermia in the present study.

The anterior nucleus of the hypothalamus (POAH) is thought to play a major role in thermoregulation (Boulant, 1981; Xin et al., 1997). A large population of thermosensitive neurons in the POAH receives and integrates information from central and peripheral sensors. In turn, these intra-POAH neurons send the modulating signal to thermoregulatory effectors that maintain body temperature around a given point, or set-point temperature. The destruction or inactivation of the POAH disrupts thermoregulation.

Pharmacological evidence supports a role for the POAH in cannabinoid-induced hypothermia. Δ9-THC injected directly into the POAH produced dose-dependent hypothermia (Fitton and Pertwee, 1982). More recently, Ovadia et al. (1995)reported that HU-210, [(BaR)-trans-3-(1,1-dimethylheptyl)-6α,7,10,10α-tetrahydro-1-hydroxy-6,5-dimethyl-βH-dibenzo[b,d]pyran-9-methanol], a synthetic cannabinoid agonist, induced hypothermia after intra-POAH injection. Those studies indicate that cannabinoids act centrally, probably within the POAH, to produce hypothermia.

An accumulating body of evidence suggests that cannabinoid receptors are located in the POAH. In particular, cells expressing CB1 receptor mRNA are densely distributed in hypothalamic regions where autoradiographic binding studies have demonstrated dense CB1 receptor binding (Herkenham et al., 1991; Mailleux and Vanderhaeghen, 1992). Recently, CB1 receptor immunoreactivity was detected in the hypothalamus, with especially robust levels reported in the POAH (Moldrich and Wenger, 2000). Thus, it is likely that cells in the POAH express CB1 receptor protein.

Although these data indicate that the POAH is a central locus of cannabinoid-induced hypothermia, the lack of selective cannabinoid receptor antagonists has precluded the investigation of the role that cannabinoid receptor subtypes play in mediating the hypothermic response. The recent development of the selective CB1 antagonist, [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride (SR141716A), and the selective CB2antagonist, N-{(1S)-endo-1,3,3-trimethyl bicyclo heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide} (SR144528), has provided insight into the receptor-mediated mechanisms that underlie the physiological effects of cannabinoids (Rinaldi-Carmona et al., 1994, 1998; Wiley et al., 1995; Compton et al., 1996).

Thus, in the present study we used SR141716A and SR144528 to ascertain the roles of the POAH and of CB1 and CB2 receptors in cannabinoid-induced hypothermia. We investigated the 1) hypothermic effects of WIN 55212-2 after systemic or intra-POAH injection, 2) effects of SR141716A or SR144528 alone on hypothermia, and 3) effects of SR141716A or SR144528 on WIN 55212-2 hypothermia.
Previous SectionNext Section
Materials and Methods
Animals.

All animal use procedures were conducted in strict accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committee. Male Sprague-Dawley rats (Zivic-Miller, Pittsburgh, PA) weighing 250 to 350 g were used in the present study. Rats were housed three per cage for a minimum of 5 days before experimental use. They were fed rat chow and water ad libitum. The temperature and relative humidity of the animal room were kept at 22 ± 2°C and 50 ± 2%, respectively. Rats were maintained on a 12-h light/dark cycle.
Surgery and Cannula Implantation.

Rats were anesthetized with an intraperitoneal injection of a solution of ketamine hydrochloride (100 mg/kg) and acepromazine maleate (0.2 mg/kg) and placed into a stereotaxic apparatus. The skull was exposed and a 1-mm hole was bored dorsal to the POAH. The dura was carefully penetrated with a drill, leaving the adjacent meninges intact. A 21-gauge, stainless steel guide cannula (Small Parts Inc., Hialeah, FL) measuring 14 cm in length was implanted unilaterally into the POAH. Stereotaxic coordinates were as follows: 2 mm anterior to bregma, 1 mm lateral to the midline suture, and 7.5 mm ventral to the dural surface (Pellegrino and Cushman, 1967). The cannula was affixed to the skull with dental acrylic (Durelon, Seefeld, Germany) and stabilized by inserting self-tapping bond screws (Plastics One Inc., Roanoke, VA) adjacent to the cannula site. The cannula extended 6.5 mm above the dural surface. After solidification of the dental cement, a 25-gauge, 14-cm, stainless steel stylet (Small Parts Inc.) was inserted into the guide cannula to minimize blood coagulation in the cannula tract and prevent infection. After a recovery interval of at least 5 days, stylets were removed and drugs were injected into the POAH with a #10 μl airtight syringe (Hamilton Co., Reno, NV).
Drug Preparation and Administration.

The cannabinoid agonist, WIN 55212-2, was obtained from Sigma-Aldrich (St. Louis, MO). The selective CB1 receptor antagonist, SR141716A, and the selective CB2 receptor antagonist, SR144528, were obtained from the National Institute on Drug Abuse (Rockville, MD). Cremophor EL, a derivative of castor oil and ethylene oxide, was purchased from Sigma-Aldrich. In all cases, the vehicle and control solution was 10% Cremophor/saline. All drugs were administered intramuscularly, except for the injection of WIN 55212-2 into the POAH. When injected into the POAH, WIN 55212-2 was injected in a volume of 1 μl. In the systemic experiments, WIN 55212-2 was injected into the right hind leg of each rat. Conversely, all other drugs were injected into the left hind leg.
Experimental Protocol.

Body temperature experiments were started between 9:00 and 10:00 AM. On the morning of the experiment, rats were placed in an environmental room which was maintained at a constant temperature of 21 ± 0.3°C and relative humidity of 52 ± 2%. After a 1-h acclimation interval, baseline temperature measurements were taken (Xin et al., 1997). A thermistor probe (YSI series 400; Yellow Springs Instrument Co., Yellow Springs, OH) was lubricated and inserted approximately 7 cm into the rectum. A digital thermometer (model 49 TA, YSI) was used to record body temperature. Rats were unrestrained during the temperature readings, with only the tail being held gently between two fingers. Body temperature was taken every 30 min during a 90-min baseline interval. After the baseline interval, either WIN 55212-2 (1, 2.5, 5, or 10 mg/kg, i.m.) or vehicle was injected. Body temperature was measured for 5 h.

To assess whether the WIN 55212-2-induced hypothermia was mediated via CB1 or CB2 receptors, either SR141716A or SR144528, respectively, was coadministered with WIN 55212-2. After a 90-min baseline interval, SR141716A (1—10 mg/kg, i.m.) or SR144528 (5, 10 mg/kg, i.m.) was injected. WIN 55212-2 (5 mg/kg, i.m.) was injected 30 min later. Body temperature was measured for 5 h.

The possibility that the POAH was the site of action of the WIN 55212-2-induced hypothermia was investigated in separate experiments. WIN 55212-2 (5- 30 μg/μl) or vehicle (10% Cremophor/saline) was injected into the POAH after a 90-min baseline period. Either 1 μl of 10% Cremophor/saline or 1 μl of WIN 55212-2 dissolved in 10% Cremophor/saline was injected into the POAH. Body temperature was measured for 5 h.

In separate experiments, it was determined whether intra-POAH CB1 receptors mediated the WIN 55212-2-induced hypothermia. After a 90-min baseline interval, either SR141716A (5 mg/kg, i.m.) or vehicle was injected. Thirty minutes later, either WIN 55212-2 (15 μg/μl) or vehicle was injected into the POAH. Body temperature was measured for 5 h.
Data and Histological Analysis.

To allow the rat to adapt to the technique, the first temperature reading was discarded in all experiments. Two consecutive body temperature readings were then recorded and averaged to establish a baseline temperature prior to drug injection. Data are presented as the mean ± S.E. of body temperature. Statistical analysis of differences between groups was determined by a one-way analysis of variance (ANOVA) followed by Tukey's post hoc test. A value of p ≤ 0.05 was considered to be statistically significant.

Cannula placement in the POAH was verified by injecting 1% Evans blue solution after the experiment according to standard procedures (Xin et al., 1997). Data from rats in which the probes were not located within the POAH were not included in the results.
Previous SectionNext Section
Results
Intramuscular Injection of WIN 55212-2 Produces Dose-Dependent Hypothermia.

The intramuscular injection of WIN 55212-2 at doses of 1 to 10 mg/kg produced hypothermia in a dose-dependent manner (Fig.1). Body temperature was measured 15 min after the injection of WIN 55212-2. The lowest dose did not produce a significant fall in body temperature. The hypothermia produced by a dose of 10 mg/kg was not greater than that produced by a dose of 5 mg/kg. Accordingly, a dose of 5 mg/kg was chosen for subsequent experiments. The fall in body temperature began 15 min after the injection of WIN 55212-2. The peak hypothermia (−3.4 ± 0.4°C) produced by 5 mg/kg WIN 55212-2 occurred 90 min postinjection. Temperatures recovered gradually and reached predrug levels 5 h postinjection.
Figure 1
View larger version:

In this page
In a new window

Figure 1

WIN 55212-2 (1—10 mg/kg, i.m.) produces hypothermia in a dose-dependent manner. WIN 55212-2 was injected at 0 min, as indicated by the arrow. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
CB1, but not CB2, Receptors Mediate WIN 55212-2-Induced Hypothermia.

In separate experiments, we determined whether CB1 or CB2 receptors contributed to the WIN 55212-2-induced hypothermia. SR141716A or SR144528 was injected 30 min before WIN 55212-2. SR141716A abolished the hypothermia. SR141716A blocked the WIN 55212-2-induced hypothermia (Fig.2). The effect of SR141716A was dose-dependent. A dose of 1 mg/kg partially reversed the WIN 55212-2-induced hypothermia. Doses of 5 or 10 mg/kg SR141716A completely abolished the hypothermia produced by WIN 55212-2. In contrast to SR141716A, SR144528 (5 or 10 mg/kg) did not alter WIN 55212-2-evoked hypothermia (Fig. 3). These data suggest that WIN 55212-2 produces hypothermia by activating CB1 receptors.
Figure 2
View larger version:

In this page
In a new window

Figure 2

SR141716A (1—10 mg/kg, i.m.) blocks WIN 55212-2-induced (5 mg/kg, i.m.) hypothermia. WIN 55212-2 was injected at 0 min. SR141716A was injected 30 min prior to WIN 55212-2. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
Figure 3
View larger version:

In this page
In a new window

Figure 3

SR144528 (5 or 10 mg/kg, i.m.) does not alter WIN 55212-2-induced (5 mg/kg, i.m.) hypothermia. WIN 55212-2 was injected at 0 min. SR144528 was injected 30 min before WIN 55212-2. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
Endocannabinoid System Does Not Tonically Regulate Body Temperature.

To determine whether the endocannabinoid system tonically regulates body temperature, the effects of SR141716A or SR144528 on rectal temperature were measured. Neither SR141716A nor SR144528 significantly affected baseline temperatures (Fig.4). Moreover, SR141716A or SR144528 elicited no visible behavioral effects. The present data suggest that endogenous cannabinoids do not tonically affect body temperature.
Figure 4
View larger version:

In this page
In a new window

Figure 4

SR141716A (10 mg/kg, i.m.) or SR144528 (5 mg/kg, i.m.) does not affect baseline body temperature. SR141716A or SR144528 was injected at 0 min, as indicated by the arrow. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
Intra-POAH Injection of WIN 55212-2 Produces Hypothermia in a Dose-Dependent Manner.

To verify the central effect of WIN 55212-2 on the major thermoregulatory center, we injected WIN 55212-2 directly into the POAH. WIN 55212-2, at doses of 5 to 30 μg/μl, produced dose-dependent hypothermia (Fig. 5). The lowest dose did not alter body temperature. The highest dose, 30 μg/μl, produced a peak hypothermia of −1.3 ± 0.1°C 15 to 30 min postinjection. A dose of 15 μg/μl produced a similar transient hypothermic response, with peak hypothermia (−1.0 ± 0.1°C) occurring 30 to 45 min postinjection. Rectal temperatures returned to predrug levels by 60 min after the injection of 30 μg/μl WIN 55212-2.
Figure 5
View larger version:

In this page
In a new window

Figure 5

Intra-POAH injection of WIN 55212-2 (5—30 μg/μl) induces hypothermia in a dose-dependent manner. Either WIN 55212-2 or 10% Cremophor/saline was injected in a volume of 1 μl at 0 min, as indicated by the arrow. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
CB1 Receptors Mediate the Hypothermia Induced by the Intra-POAH Injection of WIN 55212-2.

To determine whether CB1 receptors in the POAH mediated WIN 55212-2-induced hypothermia, SR141716A was injected intramuscularly 30 min before the intra-POAH injection of WIN 55212-2. SR141716A (5 mg/kg) abolished WIN 55212-2-evoked hypothermia (Fig.6). The injection of SR141716A alone did not affect body temperature.
Figure 6
View larger version:

In this page
In a new window

Figure 6

SR141716A (5 mg/kg, i.m.) blocks WIN 55212-2-induced (15 μg/μl) hypothermia. Either 15 μg/μl WIN 55212-2 or 10% Cremophor/saline was injected into the POAH in a volume of 1 μl at 0 min. Either SR141716A or 10% Cremophor/saline was injected 30 min before WIN 55212-2. Data are expressed as the mean (±S.E.) of body temperature. n, number of rats. ΔTb, change in body temperature.
Previous SectionNext Section
Discussion

The acute administration of cannabinoid agonists produces hypothermia in rats, mice, and primates (Compton et al., 1992; Fan et al., 1994; Spina et al., 1998; Fox et al., 2001). Similarly, in the present study, the intramuscular injection of WIN 55212-2 induced hypothermia in rats. Three aspects of the present data correlate well with the previous studies (Compton et al., 1992; Spina et al., 1998;Fox et al., 2001). First, WIN 55212-2 elicited hypothermia in a dose-dependent manner, thus revealing a receptor-mediated mechanism. Second, the onset of action of WIN 55212-2-induced hypothermia was rapid, with significant hypothermia observed 15 min postinjection. Third, the duration of the hypothermic response evoked by WIN 55212-2 persisted for at least 3 h. In fact, the highest dose, 10 mg/kg, elicited hypothermia that was still present 5 h postinjection.

SR141716A prevented the WIN 55212-2-evoked hypothermia, confirming that a CB1 receptor mechanism mediated the hypothermic effect. Blockade by SR141716A of cannabinoid-induced hypothermia in the present study is consistent with previous reports. For example, SR141716A blocked cannabinoid agonist-induced hypothermia in rats (McGregor et al., 1996; Nava et al., 2000; Fox et al., 2001) and mice (Wiley et al., 1995). Recent studies demonstrated that cannabinoid agonists were unable to evoke hypothermia in mice that lacked the CB1 receptor, providing still further evidence that CB1 receptors regulate cannabinoid-evoked hypothermia (Ledent et al., 1999; Zimmer et al., 1999). The present data and the fact that CB1 receptors are located primarily in the CNS confirm the idea that the hypothermic effect of cannabinoids is mediated centrally.

SR144528 did not affect WIN 55212-2-induced hypothermia, indicating that the hypothermia is insensitive to CB2receptor activation. These results suggest that a CB2 receptor mechanism does not play a role in mediating the hypothermic response to cannabinoids, underscoring the differing roles of CB1 and CB2 receptors in pharmacoregulation.

Neither SR141716A nor SR144528 by itself altered body temperature, suggesting that the endocannabinoid system does not tonically modulate body temperature. Previous studies have also demonstrated that cannabinoid antagonists do not affect thermoregulation (Compton et al., 1996; McGregor et al., 1996; Nava et al., 2000). This is unlike the opioid system, where the μ and κ receptors mediate hyperthermia and hypothermia, respectively, and are in tonic balance (Xin et al., 1997). The lack of cannabimimetic activity exhibited by SR141716A or SR144528 suggests that these antagonists bind selectively to CB1 and CB2 receptors, respectively, and that the recognition and activation of cannabinoid receptors are separable events. These data also argue against SR141716A and SR144528 acting as inverse agonists (MacLennan et al., 1998;Rinaldi-Carmona et al., 1998). Although it is unlikely that the endocannabinoid system tonically suppresses body temperature, other pharmacological endpoints, particularly inflammatory hyperalgesia and antinociception, are thought to be influenced by a cannabinergic tone (Calignano et al., 1998; Welch et al., 1998).

The injection of WIN 55212-2 into the POAH evoked dose-dependent hypothermia. The onset of WIN 55212-2-induced hypothermia was rapid, with the peak hypothermia occurring 15 to 45 min postinjection. The duration of hypothermia was brief. Our results provide further evidence that the POAH is a central locus of thermoregulation (Boulant, 1981;Xin et al., 1997) and indicate that the POAH plays a role in the regulation of cannabinoid-induced hypothermia (Fitton and Pertwee, 1982; Ovadia et al., 1995). The dose-dependent nature of the WIN 55212-2-induced hypothermia suggests the involvement of a receptor-sensitive mechanism.

SR141716A abolished the hypothermic response to the intra-POAH injection of WIN 55212-2. The present data are the first direct evidence that the activation of CB1 receptors in the POAH mediates cannabinoid-induced hypothermia. Prior to the discovery of cannabinoid receptors and development of cannabinoid antagonists, Fitton and Pertwee (1982) demonstrated that the injection of Δ9-THC into the POAH produced hypothermia in a dose-dependent manner (Fitton and Pertwee, 1982). In addition, Ovadia et al. (1995) demonstrated that the injection of HU-210, a synthetic cannabinoid agonist, into the POAH induced hypothermia. However, the authors did not determine whether the hypothermic response to HU-210 was dose-dependent or cannabinoid receptor-dependent. Because they did not attempt to reverse the HU-210-induced hypothermia with a cannabinoid receptor antagonist, the possibility exists that HU-210 acted through mechanisms other than cannabinoid receptor activation to produce hypothermia.

An accumulating body of evidence suggests that CB1 receptors are located in the POAH. CB1 receptor mRNA is expressed in hypothalamic nuclei where CB1 receptor binding has been demonstrated (Herkenham et al., 1991; Mailleux and Vanderhaeghen, 1992). Recently, CB1 receptor immunoreactivity has been detected in the hypothalamus (Pettit et al., 1998; Tsou et al., 1998), including the lateral hypothalamic area and the POAH (Moldrich and Wenger, 2000). The presence of CB1immunoreactivity indicates that neurons in the POAH express the CB1 receptor. It is still unclear whether the CB1 receptor-containing neurons are intrinsic or extrinsic to the POAH. Romero et al. (1998) suggested that cannabinoid receptor-containing neurons are all intrinsic to the hypothalamic nuclei, since hypothalamic deafferentation did not alter cannabinoid receptor binding. Although the mechanism that mediates WIN 55212-2-induced hypothermia is not entirely known, it is clear that CB1 receptors in the POAH play a major role.

Several aspects of the WIN 55212-2-evoked hypothermia were dependent on the injection route. The most striking differences were the duration and magnitude of the hypothermia. When injected intramuscularly, WIN 55212-2 produced a hypothermia that lasted for at least 3 h. However, the intra-POAH injection of WIN 55212-2 produced a brief hypothermia, with body temperature returning to baseline within 60 to 90 min. The administration route also affected the peak hypothermia. WIN 55212-2 produced maximal hypothermia 60 to 120 min after intramuscular injection. Not surprisingly, the intra-POAH injection of WIN 55212-2 produced peak hypothermia more rapidly, with the maximal reduction in body temperature occurring 30 min postinjection. In addition, the systemic injection of WIN 55212-2 produced a greater hypothermic response than intra-POAH injection.

It is unclear why the magnitude and duration of the WIN 55212-2-induced hypothermia are sensitive to the injection route. One explanation is that cannabinoid systems outside the POAH may play a role in mediating the hypothermia produced by the systemic, but not intra-POAH, injection of WIN 55212-2. Indeed, a high density of CB1receptors are located in extrahypothalamic sites where cannabinoid systems have been reported to interact with other neurotransmitter systems (Herkenham et al., 1991; Chaperon and Thiebot, 1999). Furthermore, an involvement of extrahypothalamic sites in cannabinoid-evoked hypothermia has been reported previously (Schmeling and Hosko, 1980; Fitton and Pertwee, 1982). Other neurotransmitters have also been reported to contribute to the hypothermic effects of systemically administered cannabinoids (Malone and Taylor, 1998;Chaperon and Thiebot, 1999; Nava et al., 2000). Thus, the POAH, as well as sites outside of the hypothalamus, appear to mediate cannabinoid-induced hypothermia.

Although structurally dissimilar, WIN 55212-2 and Δ9-THC appear to act at the same site and produce similar effects, such as catalepsy, hypothermia, antinociception, and hypolocomotion (Devane et al., 1988; Compton et al., 1992; Chaperon and Thiebot, 1999). Still, the lack of a consistent relationship between the potencies of these compounds across a spectrum of pharmacological endpoints, including hypothermia (Compton et al., 1992), does suggest that WIN 55212-2 and Δ9-THC may not reduce body temperature through identical mechanisms. In comparing rat data, we found that a dose of 1 mg/kg SR141716A, which prevented hypothermia evoked by 5 mg/kg Δ9-THC (Nava et al., 2000), did not completely block the hypothermic response to 5 mg/kg WIN 55212-2. Higher doses of SR141716A (2.5 and 5 mg/kg) were effective in our hands. Previous studies using mice have demonstrated that WIN 55212-2, in comparison with Δ9-THC, was 3-fold less potent in producing hypothermia, 3-fold more potent in producing antinociception, and 11-fold more potent in depressing spontaneous activity (Compton et al., 1992). Moreover, rat data suggest dissimilar mechanisms for discrimination and the behavioral disruption exemplified by response rate suppression. Therefore, receptor subtypes may exist for which WIN 55212-2 has different affinity or efficacy from Δ9-THC. In addition, actions on differing neuropeptide and/or neurotransmitter systems involved in thermoregulation cannot be ruled out. Differences in metabolism and disposition may also account for some of the dissimilar effects. It should be noted, however, that cross-tolerance develops to the hypothermic effects of WIN 55212-2 in Δ9-THC-treated mice (Pertwee et al., 1993; Fan et al., 1994).

In conclusion, the present experiments reveal that CB1 receptors in the POAH regulate WIN 55212-2-induced hypothermia. These data are the first demonstration that the blockade of intra-POAH CB1 receptors prevents the hypothermic effect of cannabinoids. These results also confirm that the POAH is a central locus of thermoregulation and provide further evidence that SR141716A and SR144528 are valuable tools for characterizing the pharmacological effects of cannabinoids in vivo.
Previous SectionNext Section
Acknowledgments

We thank Phyllis Beeton for assistance in histological analysis and Dr. Ronald J. Tallarida for assistance in statistical analysis.
Previous SectionNext Section
Footnotes

This study was supported by Grants DA 00376, DA 13429, and T32 DA 07237 from the National Institute on Drug Abuse.
Abbreviations:

WIN 55212-2
(4,5-dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo[3,2,1ij]quinolin-6-one
SR141716A
N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride
SR144528
N-{(1S)-endo-1,3,3-trimethyl bicyclo heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide}
POAH
preoptic anterior hypothalamic nucleus
Δ9-THC
Δ9-tetrahydrocannabinol

Received December 19, 2001.
Accepted February 25, 2002.

The American Society for Pharmacology and Experimental Therapeutics

Previous Section

References


Aceto MD,
Scates SM,
Lowe JA,
Martin BR
(1995) Cannabinoid precipitated withdrawal by the selective cannabinoid receptor antagonist, SR 141716A. Eur J Pharmacol 282:R1—R2.
CrossRefMedline

Boulant JA
(1981) Hypothalamic mechanisms in thermoregulation. Fed Proc 40:2843—2850.
Medline

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

Chaperon F,
Thiebot MH
(1999) Behavioral effects of cannabinoid agents in animals. Crit Rev Neurobiol 13:243—281.
Medline

Compton DR,
Aceto M,
Lowe J,
Martin BR
(1996) In vivo characterization of a specific cannabinoid receptor antagonist (SR141716A): inhibition of delta 9-tetrahydrocannabinol-induced responses and apparent agonist activity. J Pharmacol Exp Ther 277:586—594.
Abstract/FREE Full Text

Compton DR,
Gold LH,
Ward SJ,
Balster RL,
Martin BR
(1992) Aminoalkylindole analogs: cannabimimetic activity of a class of compounds structurally distinct from delta 9-tetrahydrocannabinol. J Pharmacol Exp Ther 263:1118—1126.
Abstract/FREE Full Text

Davies JA,
Graham JD
(1980) The mechanism of action of delta 9-tetrahydrocannabinol on body temperature in mice. Psychopharmacology 69:299—305.
CrossRefMedline

Devane WA,
Dysarz FA, 3rd,
Johnson MR,
Melvin LS,
Howlett AC
(1988) Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 34:605—613.
Abstract

Dragic T,
Litwin V,
Allaway GP,
Martin SR,
Huang YX,
Nagashima KA,
Cayanan C,
Maddon PJ,
Koup RA,
Moore JP,
Paxton WA
(1996) HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature (Lond) 381:667—673.
CrossRefMedline

Fan F,
Compton DR,
Ward S,
Melvin L,
Martin BR
(1994) Development of cross-tolerance between delta 9-tetrahydrocannabinol, CP 55,940 and WIN 55,212. J Pharmacol Exp Ther 271:1383—1390.
Abstract/FREE Full Text

Felder CC,
Joyce KE,
Briley EM,
Glass M,
Mackie KP,
Fahey KJ,
Cullinan GJ,
Hunden DC,
Johnson DW,
Chaney MO,
et al.
(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

Fitton AG,
Pertwee RG
(1982) Changes in body temperature and oxygen consumption rate of conscious mice produced by intrahypothalamic and intracerebroventricular injections of delta 9-tetahyrocannabinol. Br J Pharmacol 75:409—414.
Medline

Fox A,
Kesingland A,
Gentry C,
McNair K,
Patel S,
Urban L,
James I
(2001) The role of central and peripheral cannabinoid1 receptors in the antihyperalgesic activity of cannabinoids in a model of neuropathic pain. Pain 92:91—100.
CrossRefMedline

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

Howlett AC
(1995) Pharmacology of cannabinoid receptors. Annu Rev Pharmacol Toxicol 35:607—634.
CrossRefMedline

Jansen EM,
Haycock DA,
Ward SJ,
Seybold VS
(1992) Distribution of cannabinoid receptors in rat brain determined with aminoalkylindoles. Brain Res 575:93—102.
CrossRefMedline

Ledent C,
Valverde O,
Cossu G,
Petitet F,
Aubert J-F,
Beslot F,
Böhme GA,
Imperato A,
Pedrazzini T,
Roques BP,
et al.
(1999) Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science (Wash DC) 283:401—404.
Abstract/FREE Full Text

MacLennan SJ,
Reynen PH,
Kwan J,
Bonhaus DW
(1998) Evidence for inverse agonism of SR141716A at human recombinant cannabinoid CB1 and CB2 receptors. Br J Pharmacol 124:619—622.
CrossRefMedline

Mailleux P,
Vanderhaeghen J-J
(1992) Distribution of neuronal cannabinoid receptor in the adult rat brain: a comparative receptor binding radioautography and in situ hybridization histochemistry. Neuroscience 48:655—668.
CrossRefMedline

Malone DT,
Taylor DA
(1998) Modulation of delta9-tetrahydrocannabinol-induced hypothermia by fluoxetine in the rat. Br J Pharmacol 124:1419—1424.
CrossRefMedline

Martin WJ,
Tsou K,
Walker JM
(1998) Cannabinoid receptor-mediated inhibition of the rat tail flick reflex after microinjection into the rostral ventromedial medulla. Neurosci Lett 242:33—36.
CrossRefMedline

McGregor IS,
Issakidis CN,
Prior G
(1996) Aversive effects of the synthetic cannabinoid CP 55,940 in rats. Pharmacol Biochem Behav 53:657—664.
CrossRefMedline

Moldrich G,
Wenger T
(2000) Localization of the CB1 cannabinoid receptor in the rat brain. An immunohistochemical study. Peptides 21:1735—1742.
CrossRefMedline

Nava F,
Carta G,
Gessa GL
(2000) Permissive role of dopamine D(2) receptors in the hypothermia induced by delta(9)-tetrahydrocannabinol in rats. Pharmacol Biochem Behav 66:183—187.
CrossRefMedline

Ovadia H,
Wohlman A,
Mechoulam R,
Weidenfeld J
(1995) Characterization of the hypothermic effect of the synthetic cannabinoid HU-210 in the rat. Relation to the adrenergic system and endogenous pyrogens. Neuropharmacology 34:175—180.
CrossRefMedline

Pellegrino LJ,
Cushman AJ
(1967) A Stereotaxic Atlas of the Rat Brain. (Appleton-Century-Crofts, New York).

Pertwee RG,
Stevenson LA,
Griffin G
(1993) Cross-tolerance between delta 9-tetrahydrocannabinol and the cannabimimetic agents, CP 55,940, WIN 55,212-2 and anandamide. Br J Pharmacol 110:1483—1490.
Medline

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

Rinaldi-Carmona M,
Barth F,
Héaulme M,
Shire D,
Calandra B,
Cogny C,
Martinez S,
Maruani J,
Néliat G,
Caput D,
et al.
(1994) SR141617A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett 350:240—244.
CrossRefMedline

Rinaldi-Carmona M,
Barth F,
Millan J,
Derocq JM,
Casellas P,
Congy C,
Oustric D,
Sarran M,
Bouaboula M,
Calandra B,
et al.
(1998) SR 144528, the first potent and selective antagonist of the CB2 cannabinoid receptor. J Pharmacol Exp Ther 284:644—650.
Abstract/FREE Full Text

Romero J,
Wenger T,
de Miguel R,
Ramos JA,
Fernández-Ruiz JJ
(1998) Cannabinoid receptor binding did not vary in several hypothalamic nuclei after hypothalamic deafferentation. Life Sci 63:351—356.
CrossRefMedline

Schmeling WT,
Hosko MJ
(1980) Hypothermic effects of intraventricular and intravenous administration of cannabinoids in intact and brainstem transected cats. Neuropharmacology 19:567—573.
CrossRefMedline

Shire D,
Calandra B,
Rinaldi-Carmona M,
Oustric D,
Pessegue B,
Bonnin-Cabanne O,
LeFur G,
Caput D,
Ferrara P
(1996) Molecular cloning, expression and function of the murine CB2 peripheral cannabinoid receptor. Biochim Biophys Acta 1307:132—136.
Medline

Showalter VM,
Compton DR,
Martin BR,
Abood ME
(1996) Evaluation of binding in a transfected cell line expressing a peripheral cannabinoid receptor (CB2): identification of cannabinoid receptor subtype selective ligands. J Pharmacol Exp Ther 278:989—999.
Abstract/FREE Full Text

Spina E,
Trovati A,
Parolaro D,
Giagnoni G
(1998) A role of nitric oxide in WIN 55,212-2 tolerance in mice. Eur J Pharmacol 343:157—163.
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

Welch SP,
Huffman JW,
Lowe J
(1998) Differential blockade of the antinociceptive effects of centrally administered cannabinoids by SR141716A. J Pharmacol Exp Ther 286:1301—1308.
Abstract/FREE Full Text

Wiley JL,
Barrett RL,
Lowe J,
Balster RL,
Martin BR
(1995) Discriminative stimulus effects of CP 55,940 and structurally dissimilar cannabinoids in rats. Neuropharmacology 34:669—676.
CrossRefMedline

Xin L,
Geller EB,
Adler MW
(1997) Body temperature and analgesic effects of selective mu and kappa opioid receptor agonists microdialyzed into rat brain. J Pharmacol Exp Ther 281:499—507.
Abstract/FREE Full Text

Zimmer A,
Zimmer AM,
Hohmann AG,
Herkenham M,
Bonner TI
(1999) Increased mortality, hypoactivity and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proc Natl Acad Sci USA 96:5780—5785.


Source: CB1 Receptors in the Preoptic Anterior Hypothalamus Regulate WIN 55212-2 [(4,5-Dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)-6H-pyrrolo[3,2,1ij]quinolin-6-one]-Induced Hypothermia
 
Back
Top Bottom