Canna~Fangled Abstracts

Cannabinoid Receptors and Signal Transduction

By June 27, 2013No Comments

pub med big

Cannabinoid Receptors and Signal Transduction

Allyn C. Howlett and Joong-Youn Shim.

pm1

The cannabinoid receptors are members of the rhodopsin-like family of 7-transmembrane spanning receptors that are believed to bind their agonist ligands within the central core formed by the interaction of the seven transmembrane helices. Cannabinoid receptors are associated with G proteins of the Gi/o family (Gi1,2 and 3, and Go1 and 2) via the intracellular loops of the protein. Two receptor subtypes have been characterized: CB1 receptors found predominantly in the nervous system and neuronal cells, and CB2 receptors found predominantly in immune tissue. An alternative receptor that is stimulated by anandamide has been pharmacologically described, and it is coupled to the Gi family of signal transduction proteins. Both CB1 and CB2 cannabinoid receptors inhibit adenylyl cyclase via Gi, and both subtypes stimulate p42/p44 mitogen-activated protein kinase (MAPK) activity. Only the CB1 receptor has been shown to signal via ion channels. CB1 and CB2 receptors influence other signal transduction pathways as well. The molecular mechanism for agonist activation of the cannabinoid receptors involves modification of the conformation of the third and fourth intracellular loops of the receptor such that associated G proteins can be activated.

CB1 Receptor and Signal Transduction Pathways

Gi/o-Mediated Regulation of Intracellular Cyclic AMP and Protein Kinase A (PKA)-Mediated Cellular Regulation

Inhibition of cyclic AMP production has been observed in brain tissue and neuronal cells expressing CB1 receptors (reviewed in refs. 1-4). Cannabinoid receptor agonists led to inhibition of cyclic AMP accumulation in host cell lines that have been made to express exogenous CB1 receptors.58 CB1-mediated inhibition of adenylyl cyclase was pertussis toxin-sensitive, indicating the requirement for Gi/o proteins.810 Anandamide, (R)-methanandamide and 2-arachidonoylglycerol, desacetyllevonantradol, WIN55212-2 and CP55940 behave as full agonists to inhibit adenylyl cyclase activity in N18TG2 membranes.1113 Inhibition of adenylyl cyclase only occurs in cells that express isoforms 5 and 6, and 1, 3 and 8.14 The expression of CB1 cannabinoid receptors in COS-7 cells evoked inhibition of cAMP accumulation in cells coexpressing those particular adenylyl cyclase isoforms, but not those coexpressing isoforms 2, 4, and 7.14
Ion channels in neurons can be phosphorylated by cyclic AMP-dependent protein kinase A (PKA), a pathway that is critical in modulating neuronal responses to ionotropic neurotransmitters.15 In rat hippocampal cells, intracellular cyclic AMP and PKA phosphorylated a potassium channel (A-current), leading to a negative shift in the voltage-dependence.16 CB1 receptors activated A-type potassium currents by decreasing intracellular cyclic AMP concentrations.16,17 The net reduction in phosphorylation of ion channel proteins would result in hyperpolarization of the axon terminals and a blunted response to depolarizing stimuli.15
Cellular changes in neuronal structure, which are necessary for synaptic plasticity, can also be modified by the cyclic AMP/PKA regulatory pathway.18,19 In a neuroblastoma cell model, CB1 receptor agonist HU210 induced neurite retraction.18 One mechanism for this type of regulation would be the cannabinoid-mediated modulation of either focal adhesion kinase (FAK), an enzyme important for integrating cytoskeletal changes with signal transduction events,20,21 or focal adhesion kinase-related nonkinase proteins.19 Anandamide, CP55940 and WIN55212-2 stimulated tyr-phosphorylation of FAK (pp125) in hippocampal slices. This response could be blocked with SR141716 and pertussis toxin demonstrating its mediation by CB1 receptors and Gi/o.21 The tyr-phosphorylation of FAK in brain slices20 and of the FAK-related nonkinase19 was reversed by 8-Br-cyclic AMP, and mimicked by PKA inhibitors, suggesting that Gi-mediated inhibition of adenylyl cyclase is integral to this pathway.
In neural progenitor cells or PC12 pheochromocytoma cells transfected with the CB1 receptor, anandamide, (R)-methanandamide and HU210 inhibited the neuronal differentiation and neurite extension in response to nerve growth factor (NGF), and this response was antagonized by the CB1 antagonist SR141716.22 NGF works via the Trk-A receptor to regulate rap1 and B-raf , producing a sustained activation of mitogen-activated protein kinase (p42/p44 MAPK) necessary for neurite outgrowth. CB1receptors could attenuate the sustained activation of p42/p44 MAPK.22 The observation that the effects of anandamide were reversed by forskolin or hormone-stimulated cyclic AMP production22 suggests that cannabinoid regulation of cyclic AMP and PKA may be intrinsic to modulation of the Trk-A receptor pathway.

Gi/o-Mediated Regulation of Phospholipase C ( PLC) Leading to Inositol Triphosphate (IP3) Release and Mobilization of Intracellular Ca2+

A transient increase in intracellular free Ca2+ was observed in undifferentiated N18TG2 neuroblastoma and NG108-15 neuroblastoma-glioma hybrid cells in response to 2-arachidonoylglycerol or 1(3)-arachidonoylglycerol.2325 HU210, CP55940, Δ9-THC, anandamide and (R)-methanandamide behaved as partial agonists.2326 The CB1 receptor was implicated because this response was blocked by SR141716.23,26 The transduction of this response by Gi/o proteins was inferred because the Ca2+transient was blocked by pertussis toxin. The response could be attenuated by a phospholipase C inhibitor, suggesting a mechanism for activation of phospholipase Cβ by Gα or Gβγ subunits, leading to IP3 release.23,25 CB1 cannabinoid receptors were shown to activate PLC in cultured cerebellar granule neurons, in which (R)-methanandamide, WIN55212-2 and HU210 augmented the Ca2+ signal in response to NMDA receptor stimulation or K+-depolarization.27 The response was antagonized by SR141716, pertussis toxin, and the PLC inhibitor U-73122.27 The source of the released Ca2+ was a caffeine-sensitive and IP3 receptor-sensitive pool. It should be noted that the regulation of PLC by cannabinoid receptors has not been well-documented in the literature. The release of IP3 or phosphatidic acid in response to anandamide or WIN55212-2 was not detected in CHO cells expressing recombinant CB1receptors under conditions in which other exogenously expressed receptors could activate PLC.28,29

Regulation of Ion Channels via βγ Subunits

Exogenously expressed CB1 receptors coupled to the inwardly rectifying potassium currents (Kir) in AtT-20 pituitary tumor cells in a pertussis toxin-sensitive manner, indicating that Gi/o proteins serve as transducers of the response.3032 Anandamide was a full agonist compared with WIN55212-2 in the Kircurrent activation in the AtT-20 cell model;30 however, it was a partial agonist in Xenopus laevis oocytes coexpressing the CB1 receptor and GIRK1 and GIRK4 channels.32
L-type Ca2+ channels were inhibited by anandamide and WIN55212-2 in cat brain arterial smooth muscle cells, which express mRNA for the CB1 receptor.33 The cannabinoid-evoked inhibition of L-type Ca2+currents was blocked by pertussis toxin and SR141716, and was pharmacologically correlated with vascular relaxation in cat cerebral arterial rings.33
The CB1 receptor and a Gi/o protein inhibited N-type voltage-gated Ca2+ channels in differentiated N18 neuroblastoma, NG108-15 neuroblastoma-glioma hybrid cells, and neuronal expression systems.3438Anandamide was a partial agonist compared with WIN55212-2 or CP55940.36 2-Arachidonoylglycerol (and anandamide as a partial agonist) inhibited the depolarization-evoked rise in intracellular Ca2+ as detected by fura-2 in differentiated NG108-15 cells.24
WIN55212-2 and anandamide both behaved as full agonists to inhibit Q-type Ca2+ currents in AtT-20 pituitary cells expressing recombinant CB1 receptors. This response was pertussis toxin-sensitive, implicating Gi/o proteins as transducers.30 Anandamide inhibited P/Q-type Ca2+ fluxes (i.e., blocked by T-agatoxin-IVa) as detected by fura-2 fluorescence in rat cortical and cerebellar brain slices.39 This response was blocked by SR141716 and pertussis toxin, indicating mediation by CB1 receptors and Gi/o proteins.

Signal Transduction via MAPK and Phosphatidyl Inositol-3-Kinase (PI3K)

MAPK (p38) was activated in CHO cells expressing recombinant CB1 receptors40 and in human vein endothelial cells possessing endogenous CB1 receptors.41 MAPK (p42/p44) was activated via CB1receptors in WI-38 fibroblasts, U373MG astrocytic cells, C6 glioma cells and primary astrocytes, and in host cells expressing recombinant CB1 receptors.4245 These effects were mediated by CB1 receptors and Gi/o proteins as evidenced by their attenuation by SR141716 and by pertussis toxin.
One mechanism for MAPK activation could involve a pathway in which CB1 receptor-mediated Gi/o activation leads to recruitment of PI3K, thereby intitiating a pathway that would result in tyrosine phosphorylation and activation of raf-1, and subsequent activation of p42/p44 MAPK. Evidence for this comes from studies in which CB1 receptor signaling via MAPK in several cell types was blocked by wortmannin or LY294002, which are inhibitors of PI3K.42,45,46 Δ9-THC, HU210 and CP55940 produced an SR141716-sensitive stimulation of protein kinase B (PKB/Akt) (isoform IB) in the human astrocytoma cell line U373MG and in CHO cells expressing recombinant CB1 receptors,46,47 a signal that usually follows PI3K-mediated phosphorylation of inositol phospholipids. Δ9-THC promoted PI3K and tyrosine phosphorylation of raf-1 and its translocation to the membrane in rat cortical astrocytes.44
MAPK activation can be linked to expression of immediate early genes, as has been demonstrated for krox-24 expression mediated by CB1 receptors in U373MG human astrocytoma cells.48 Cannabinoid receptor agonists utilized the Gi/o, PI3K and ras pathway to activate c-Jun N-terminal kinase (JNK1 and JNK2) in CHO cells expressing recombinant CB1 receptors.40 These transcription factors modulate the gene expression pattern of cells, thereby altering important cellular functions such as cell survival or differentiation.

Regulation of Nitric Oxide Synthase (NOS)

Anandamide has been reported to evoke nitric oxide (NO) production in several cell types, and this response could be blocked by SR141716 (see ref. 49 for review). In rat median eminence slices, NO production was stimulated by anandamide in a SR141716-sensitive manner.50 The presence of Ca2+-dependent constitutive NOS in N18 cell homogenates has been inferred from a cyclic GMP reporter assay.51 NO production was stimulated by anandamide in rat median eminence fragments50 and by anandamide or CP55940 in leech or muscle ganglia.52,53 Responses in these tissues were blocked by SR141716, implicating the involvement of a CB1-like receptor. Antagonism by (L)-N-arg-methyl ester (L-NAME) suggests that a signal transduction pathway must lead to regulation of NOS.50 A role for NO in mediating anandamide’s effects on neurotransmitter release is supported by the observation that both anandamide and the NO generating agent S-nitroso-N-acetyl-penicillamine could inhibit the release of preloaded radiolabelled dopamine from invertebrate ganglia.52
Anandamide or HU210 stimulated NO production in human saphenous vein,54 segments, cultured human arterial endothelial cells,55,56 cultured human umbilical vein endothelial cells57 and human monocytes.58 These responses were blocked by SR141716, implicating CB1 receptors. In cultured human arterial endothelial cells, NO generation was preceded by a rapid increase in the concentration of intracellular Ca2+,55,56 consistent with the stimulation of a Ca2+-regulated constitutive NOS. In saphenous vein endothelia, the generation of NO required Ca2+ in the perfusate, suggesting that an extracellular source of Ca2+ might be required for NOS activation.54 In human endothelial cells, generation of NO and peroxynitrite was associated with the activation of the anandamide transporter.57

CB2 Receptor and Signal Transduction Pathways

The inhibition of cyclic AMP production has been observed in human lymphocytes, mouse spleen cells expressing CB2 receptors, and in host cell lines expressing CB2 receptors.29,59,60 CB2 receptor-mediated inhibition of adenylyl cyclase was blocked by pertussis toxin, implicating Gi/o proteins as transducers.29 The inhibition of adenylyl cyclase by CB2 receptors probably occurs in only those cells that express adenylyl cyclase isoform families 5, 6 and 1, 3, 8.14 2-Arachidonoylglycerol was a full agonist, and anandamide and (R)-methanandamide were partial, low-efficacy agonists for the inhibition of forskolin-stimulated cyclic AMP accumulation in CHO cells expressing recombinant CB2receptors.29,5960
MAPK was activated by cannabinoid receptor agonists in cultured human promyelocytic HL60 cells possessing endogenous CB2 receptors, and in CHO cells expressing recombinant CB2 receptors.62However, cannabinoid agonists failed to activate PKB/Akt in HL60 cells, suggesting that a PI3K mechanism may not be regulated by CB2 receptors in this model.47 Krox-24 expression was stimulated via CB2 receptors in HL60 promyelocytes.62
A novel signal transduction pathway regulated by CB2 receptors appears to be directed by the synthesis of ceramide. Long-term (days) treatment with ceramide, which binds to and activates raf-1, led to the sustained activation of p42/p44 MAPK and the ensuing apoptosis in sensitive strains of rat C6 glioma cells.63 Cannabinoid agonists (Δ9-THC, CP55940, HU210, WIN55212-2 and endocannabinoids anandamide and 2-arachidonoylglycerol) could promote the antiproliferative effects in C6 cells by contributions from both CB1 and CB2 receptors,63,64 although in U373MG astrocytoma cells, stimulation of the CB1 receptor was protective against ceramide-induced apoptosis.46 The primary mechanism was related to the continued synthesis of ceramide over a period of days.63,65 The importance of the CB2receptors in regulation of serine palmitoyltransferase, the rate-limiting enzyme in ceramide synthesis, was demonstrated using enzyme inhibitors and the selective CB2 agonists JWH015 and JWH133 and antagonist SR144528.64,66 These data demonstrate the importance of ceramide as a cellular modulator, and suggest that the duration of the activation of p42/p44 MAPK is critical to the apoptotic response in transformed glioma cells.63
Several signal transduction pathways that have been characterized for the CB1 receptor have not been identified for the CB2 receptor. PLC activation in response to anandamide or WIN55212-2 was not able to be detected in CHO cells expressing recombinant CB2 receptors.7,28,29 The inhibition of Q-type Ca2+currents in AtT-20 cells expressing CB2 receptors could not be stimulated by WIN55212-2 or anandamide.29

Endothelial Anandamide Receptor: Pharmacology and Signal Transduction Pathways

A distinct non-CB1/non-CB2 receptor for anandamide has recently been characterized in endothelial cells.6760 Vasorelaxation is the physiological response regulated by this receptor, and this was demonstrated in a rodent mesenteric vascular preparation6771 as well as a rabbit aortic ring preparation.68 The response required endothelial cell-intact preparations, suggesting an endothelial location for the receptor.6768 This notion has subsequently been confirmed by signal transduction studies in endothelial cells.6972 The observation that vasorelaxation could be reproduced in preparations from CB1/CB2 (-/-) transgenic mice is evidence that these genetically defined receptors are not responsible for this biological response.67 The pharmacology of the response is distinct from that of either CB1 or CB2 receptors. The agonists that evoked vasodilation include anandamide, (R)-methanandamide and abnormal cannabidiol. It is noteworthy that potent cannabinoid receptor agonists including Δ9-THC, CP55940, WIN55212-2 and HU210 failed to elicit this response.6769,73 Although the CB1 antagonist SR141716 could competitively antagonize the vasorelaxation response, higher concentrations were required than would be expected for a CB1 receptor-mediated response.67,68Cannabidiol, which exhibits particularly poor efficacy as a CB1 receptor agonist, antagonized the anandamide-mediated vasorelaxation.67,74 Subsequently, a novel antagonist for this receptor, O-1918, a congener of cannabidiol, was developed and demonstrated to behave as a competitive antagonist against anandamide and abnormal cannabidiol in the vasorelaxation response.69 In intact anesthetized mice, O-1918 blocked the hypotensive action of abnormal cannabidiol but not that of the cannabinoid receptor agonist HU210.69 Neither abnormal cannabidiol nor O-1918 were able to compete for the binding of [3H]CP55940 to the CB1 or CB2 receptor, confirming that these ligands are not acting via these receptors.
The signal transduction pathway by which this endothelial anandamide receptor mediates its response has been examined. Both (R)-methanandamide-mediated vasorelaxation of aortic rings and abnormal cannabidiol-mediated vasorelaxation of rat mesenteric artery segments were pertussis toxin-sensitive, indicating that the receptor is coupled to the Gi/o family of signal transducing proteins.68,69 The vasorelaxation in the rabbit aortic ring preparation was blocked by the NOS inhibitor L-NAME, indicating that NO production is an intrinsic part of the pathway.68 However, this appeared not to be the case in the rat mesenteric artery preparation69 or perfused mesenteric bed preparations.67,75 The vasorelaxation response to anandamide in coronary or mesenteric artery preparations and to abnormal cannabidiol in rat mesenteric artery segments was blocked by charybdotoxin.69,7577 This implicates the involvement of either a delayed rectifier K+ channel or a large conductance calcium-activated K+ (BKCa) channel. Further analysis of the signal transduction pathways was performed in cultured endothelial cells. In human umbilical vein endothelial cells (HUVEC), abnormal cannabidiol could evoke the phosphorylation of p42/44 MAPK and PKB/Akt. The latter could be blocked by a PI3K inhibitor (LY294002 or wortmannin), indicating the mediation of this response via PI3K.69 In cultured rabbit aortic endothelial cells, (R)-methanandamide elicited NO production, which was blocked by pertussis toxin and LY294002, indicating the involvement of Gi/o proteins and the PI3K pathway, respectively.72 Thus, the endothelial anandamide receptor presents distinct pharmacological and signal transduction properties compared with the two characterized cannabinoid receptors.

Mechanism of Receptor-Mediated G Protein Coupling

Cannabinoid receptors are 7-transmembrane spanning receptors associated with intracellular G proteins (fig. 1). When the CB1 receptor-G protein complex is solubilized using the mild nonionic detergent CHAPS, the protein-protein interactions remain stable even as the lipids surrounding the proteins are supplanted by the detergent.78,79 The CB1 receptor must remain precoupled with G proteins even in the absence of an exogenously added agonist, inasmuch as it is unlikely that any endogenous agonist can remain bound to the receptor during the solubilization and immunoprecipitation processes.

Figure 1. The rat CB1 receptor in two-dimensional representation.

Figure 1

The rat CB1 receptor in two-dimensional representation. Three extracellular (E1, E2, and E3) and intracellular (C1, C2, and C3) regions are represented. The most conserved residues within each helix is indicated by an arrow (see ref. ). The palmitoylation(more…)

When the CB1 receptor was immunoprecipitated from CHAPS-solubilized rat brain or N18TG2 neuroblastoma cell membranes, specific Gα proteins associated with the CB1 receptor were found to be in the same proportions as those found in particular brain regions or cell types.79,80 For example, Gαo was extremely abundant in rat brain membranes, but barely detectable in N18TG2 membranes, and thus CB1 receptors from rat brain coimmunoprecipitated with an abundance of Gαo. In contrast, Gαi1, Gαi2 and Gαi3 were much more abundant in N18TG2 membranes than in rat brain. These CB1 receptor-Gα complexes represent a functional association in dynamic equilibrium both in CHAPS as well as in intact membranes, as demonstrated by the finding that pertussis toxin treatment disrupted the receptor-Gα association.79,81 The observation that many different signal transduction pathways are possible suggests that the cell-type specific G proteins and the stoichiometry of these various Gα and Gβγ dimers with the receptor are factors in determining which signal transduction pathways are operative.
The signal transduction response is inititiated by the binding of agonist ligands to the receptor as it exists in a receptor-G protein complex. Moieties on classical and nonclassical cannabinoid ligands that are important for binding to the CB1 cannabinoid receptor have been identified by structure-activity relationship (SAR) studies (see reviews refs. 2,4,82,83 for original references). It was proposed that lys192 in TM3 might be a key residue for binding because this residue in the cannabinoid receptor is homologous to key amino acids important for agonist binding to rhodopsin and the aminergic neurotransmitter receptors.84,85 When tested by site-directed mutagenesis studies, lys192 was critical for binding of [3H]CP55940 but not [3H]WIN55212-2, and the inhibition of cyclic AMP production in response to CP55940, HU210 and anandamide was attenuated.84,85 The similarly charged arg could substitute, but not gln or glt,85 lending credence to the hypothesis that lys192 may serve as a hydrogen bond donor to the oxygen of the phenolic hydroxyl known to be critical from classical and nonclassical cannabinoid SAR studies.86,87
SAR studies for CB2 agonists have not been as extensive as for CB1 receptor ligands (see ref. 2 for review). Ligands having notable selectivity for the CB2 receptor include AAI derivatives and cannabinoid structures that lack the phenolic hydroxyl or possess hydroxymethyl groups at that position.29,8892Studies of CB1/2(TM3) mutant receptors have suggested that the TM3 ser in CB2 receptors may contribute to greater affinity for AAI ligands than the gly in the comparable position in CB1 receptors.93
The domains of the CB1 receptor that interact with G proteins are on the cytoplasmic surface. Peptides representing defined regions of the C3 loop and the juxtamembrane C-terminal region were used to mimic the intracellular domain(s) of the CB1 receptor.94 The juxtamembrane C-terminal peptide promoted G protein activation in rat brain membranes and the inhibition of hormone- or forskolin-stimulated adenylyl cyclase in N18TG2 membranes, so it is believed that this peptide mimics the receptor domain that recognizes and activates G proteins.95 The requirement for G proteins in the inhibition of adenylyl cyclase by this peptide mimic was demonstrated by its sensitivity to pertussis toxin.8195 The juxtamembrane C-terminal peptide competed for the interaction between the CB1receptor with Gαo or Gαi379 in solubilized brain preparations, and the CB1 receptor with Gαi3 in solubilized N18TG2 membrane extracts.80 However, this peptide failed to disrupt the CB1 receptor interaction with Gαi1 or Gαi2 in rat brain or N18TG2 cell membranes. These data strongly suggest that the juxtamembrane C-terminal domain interacts with Gαo or Gαi3 proteins, and therefore would direct signal transduction pathways mediated by these G protein subtypes. The C-terminal juxtamembrane peptide from the CB2 receptor failed to compete for Gαo or Gαi,79,80 suggesting that this domain does not perform the same function in the CB2 receptor as it does in the CB1 receptor.
Under environmental conditions that would mimic the anionic phospholipid headgroups of the lipid bilayer or a negative charge-patch on the Gα surface, the C-terminal juxtamembrane peptide is able to form a helical structure.95 It can be hypothesized that changes in TM7 that might alter the positions of the cationic residues in the C-terminal domain may promote activation of these G proteins. There is no evidence to suggest that the C-terminal distal to this region is required for Go-mediated responses, because truncation beyond this domain fails to alter the coupling of the CB1 receptor to inward rectifier K+ currents.96
Palmitoylation of a cys residue anchors the C-terminal domain to the plasma membrane distal to the putative helical intracellular domain.97 Mutants of the receptor that are truncated two residues but not 23 residues beyond this cys, resulted in a loss of desensitization, suggesting that critical sites of phosphorylation facilitated by G protein receptor kinase-3 and subsequent association with β-arrestin2 occur within this region.96 Studies of site-mutations of ser426 and ser430 indicated that these residues are required for desensitization, but not internalization.96 Internalization of the CB1 receptor involved the distal region 14 residues from the C-terminal (val460-thr-met-ser463), and apparently did not require coupling with functional Gi proteins.98
The third intracellular loop (C3) is also important for directing signal transduction through G proteins. The combination of three peptides (N-terminal side, middle or C-terminal side) comprising the C3 of the receptor was able to disrupt the CB1 receptor association with Gαi1 or Gαi2 in both rat brain and N18TG2 membrane extracts,79,80 whereas the C3 peptides were unable to compete with Gαo in rat brain extracts or Gαi3 in N18TG2 membrane extracts. None of the C3 peptides alone were able to disrupt the CB1 receptor-Gα interaction. However, the combination of middle plus C-terminal side was capable of dissociating the CB1 receptor association with Gαi1 or Gαi2, whereas the combination of N-terminal side plus middle failed to do so. Thus, the C3 (C-terminal side) domain interacts with Gαi1 or Gαi2, but not Gαi3 or Gαo. The C3 peptides activated G proteins in rat brain membranes but to a much lesser extent than did the juxtamembrane C-terminal peptide.94 One explanation is that the G protein that is highly predominant in brain is Go. A 9- amino acid C-terminal side peptide, predicted by homology with rhodopsin to be embedded within the membrane-cytosol interface, was able to sustain the GTPase activity of a pure preparation of Gαi1.99
In an SDS micelle environment, thought to mimic the lipid bilayer and phospholipid headgroup region, the structure of a peptide encompassing the C3 loop was shown by NMR analysis to be helical for the N-terminal third of the intracellular domain extending beyond TM5.100 Ulfers and colleagues100speculate that a highly hydrophobic sequence of three ile residues at the terminal end of this helical domain may anchor this region to the membrane. At the juncture between the helical region and the triple-ile hydrophobic domain resides a ser317, which has the potential to be phosphorylated by PKC.101Although phosphorylation of ser317 prevented the cannabinoid agonist-induced regulation of ion channels, mutation of ser317 to ala did not.101 Thus, negative charge in this region must alter the structural properties. The middle third of C3 has limited structure according to the NMR analyses; however, the NMR suggests that a turn may occur at an intracellular gly residue.101
The C3 was shown to be helical beginning within the C-terminal third approximately two turns proximal to the position that is predicted (by homology with rhodopsin102) to enter the lipid bilayer as TM6.100 NMR analysis of a peptide representing this C-terminal region indicated that this peptide is also helical in the presence of Gαi1.99 Within this domain, approximately two helical coils embedded in the membrane, exists a leu-ala-lys-thr sequence that is noteworthy because the leu-ala residues are reversed in the Gs-coupled β-adrenergic receptor. 103 Reversal of this leu-ala sequence in the CB1 receptor resulted in a loss of coupling to Gi thereby attenuating inhibition of cyclic AMP production.103 Ulfers and colleagues100 speculated that this region may be oriented along the plasma membrane surface as the result of TM6 being bent at an intracellular pro residue. Alternatively, this part of the coil may be exposed upon activation of the receptor. The ala-leu sequence may not form the appropriate helical structure.99 A stimulation of cyclic AMP production was observed when cells bearing the mutant CB1receptor ala-leu sequence were treated with pertussis toxin to block interaction of Gi proteins with the receptor. This suggests that in the absence of functional Gi proteins, the mutant CB1 receptor was able to couple to Gs as might be predicted from the sequence homology with the Gs-coupling receptor.103
It is possible to speculate on how binding of the CB1 receptor agonists within the receptor binding pocket might activate G proteins, based upon analogy with what has been reported for other G protein coupled receptors. One mechanisms has been proposed by which the TM2 and TM7 helices might interact via a hydrogen bonding association between asp163 in TM2 and asn394 in TM7.104,105 In the proposed activation scheme, ligand binding would disrupt this association and thereby promote movement in TM7 and the juxtamembrane C-terminal domain. This TM2 asp163 is the putative site of the regulation of receptor-G protein coupling by Na+ ions and has been implicated in the modulation of signal transduction (see ref. 78 for data and original literature). When expressed in AtT20 host cells, CB1 receptors having a mutation in TM2 asp163 exhibited a disruption of signal transduction through the inwardly rectifying K+ channel, but other signal transduction pathways (cyclic AMP inhibition, MAPK activation, Ca2+ currents) remained unaffected.105 Similarly, expression of CB1 receptors having a mutation of TM2 asp163 exhibited an attenuation of cannabinoid-stimulated inwardly rectifying K+currents due to GIRK1/4 channel proteins that were coexpressed in Xenopus oocytes.32 In contrast to findings in the AtT20 expression system, CB1 receptors having this TM2 asp163 mutation that were expressed in HEK293 cells were able to attenuate the inhibition of cyclic AMP production,106suggesting that cell-type differences may be responsible for the discrepancy. The original hypothesis that a TM2 to TM7 interaction my be guiding signal transduction is not well supported by experimental evidence because a reciprocal mutation (TM2 asp164asn plus TM7 asn394asp) failed to “rescue” the asp163 mutation as would have been predicted if these two amino acids were associated by hydrogen bonding.105 Thus, it is known that the TM2 asp163 is important for conferring a signal to G proteins, but there is insufficient evidence to propose a mechanism.
Another proposed mechanism for conferring a signal to G proteins is via a TM3 to TM6 interaction that could modulate protein conformation in the IC3 domain. Barnett-Norris and colleagues107 have proposed an interaction between TM3 arg214 in the highly conserved asp-arg-tyr-x-x-ile sequence of rhodopsin-like G protein-coupled receptors and the TM6 asp338 at the membrane interface. The TM6 possesses a pro kink, allowing for the possibility that movement in that helix could be facilitated by a ligand binding interaction. Based upon helical chain conformation models, Barnett-Norris and colleagues107 have proposed that a TM6 hydrophobic interaction with the alkyl side chain of the cannabinoid agonists acts as the trigger to move TM6. The CB1 homology model of Shim and colleagues,102 based upon the structure of rhodopsin, also depicts hydrophobic interactions with cannabinoid agonists at the TM6; however, different amino acid residues were involved. The motion of the TM6 could alter the conformation of the C-terminal side of IC3 to promote G protein activation.

Summary and Future Directions

Cannabinoid receptors are G protein coupled receptors, of which, two receptor types have been characterized: CB1 receptors found predominantly in the brain and neuronal cells, and CB2 receptors found predominantly in immune tissue. An alternative receptor that is stimulated by anandamide has been pharmacologically described, and it can be speculated that additional receptors may be described and cloned in the future. It is possible that receptors that respond to endocannabinoid ligands but not classical cannabinoid or aminoalkylindole agonists may have to be named to describe more accurately the endogenous compounds that stimulate those receptors in physiological situations.
Cannabinoid receptors are coupled to the Gi family of signal transduction proteins. Both CB1 and CB2receptor types inhibit adenylyl cyclase via Gi, and both types stimulate MAPK activity. Only the CB1receptor has been shown to signal via ion channels. These signal transduction events can undergo complex regulation between pathways, as evidenced by the PKA phosphorylation of ion channels and MAPK pathway proteins. Ultimately, these pathways regulate important cellular functions necessary for synaptic plasticity and long-term changes in neuronal function. Thus, endocannabinoid agonists may be critical in the formation and maintenance of interneuronal communication.
The molecular mechanism for agonist activation of the cannabinoid receptors involves binding of agonist ligands within the central core formed by the interaction of the seven transmembrane helices and subsequent modification of the conformation of the third and fourth intracellular loops of the receptor such that associated G proteins can be activated. Mutation, peptide and modeling studies should provide greater insight into the nature of these mechanisms, so that novel drugs can be designed that confer greater efficacy to selected G proteins. This should promote greater selectivity for signal transduction pathways that are intrinsic to the cellular mechanisms important for therapeutic benefits.

Go to:

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].
Austin (TX): Landes Bioscience; 2000-.

References

1.
Howlett AC. Pharmacology of cannabinoid receptors. Annu Rev Pharmacol Toxicol. 1995;35:607–634. [PubMed]
2.
Howlett AC, Barth F, Bonner TI. et al. International union of pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev. 2002;54(2):161–202. [PubMed]
3.
Pertwee RG. Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther.1997;74(2):129–180. [PubMed]
4.
Pertwee RG. Pharmacology of cannabinoid receptor ligands. Curr Med Chem. 1999;6(8):635–664.[PubMed]
5.
Matsuda LA, Lolait SJ, Brownstein MJ. et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346(6284):561–564. [PubMed]
6.
Gerard C, Mollereau C, Vassart G. et al. Nucleotide sequence of a human cannabinoid receptor cDNA. Nucleic Acids Res. 1990;18(23):7142. [PMC free article] [PubMed]
7.
Felder CC, Briley EM, Axelrod J. et al. Anandamide, an endogenous cannabimimetic eicosanoid, binds to the cloned human cannabinoid receptor and stimulates receptor-mediated signal transduction. Proc Natl Acad Sci USA. 1993;90(16):7656–7660. [PMC free article] [PubMed]
8.
Vogel Z, Barg J, Levy R. et al. Anandamide, a brain endogenous compound, interacts specifically with cannabinoid receptors and inhibits adenylate cyclase. J Neurochem. 1993;61(1):352–355.[PubMed]
9.
Howlett AC, Qualy JM, Khachatrian LL. Involvement of Gi in the inhibition of adenylate cyclase by cannabimimetic drugs. Mol Pharmacol. 1986;29(3):307–313. [PubMed]
10.
Pacheco MA, Ward SJ, Childers SR. Identification of cannabinoid receptors in cultures of rat cerebellar granule cells. Brain Res. 1993;603(1):102–110. [PubMed]
11.
Howlett AC, Mukhopadhyay S. Cellular signal transduction by anandamide and 2-arachidonoylglycerol. Chem Phys Lipids. 2000;108(1-2):53–70. [PubMed]
12.
Pinto JC, Potie F, Rice KC. et al. Cannabinoid receptor binding and agonist activity of amides and esters of arachidonic acid. Mol Pharmacol. 1994;46(3):516–522. [PubMed]
13.
Childers SR, Sexton T, Roy MB. Effects of anandamide on cannabinoid receptors in rat brain membranes. Biochem Pharmacol. 1994;47(4):711–715. [PubMed]
14.
Rhee MH, Bayewitch M, Avidor-Reiss T. et al. Cannabinoid receptor activation differentially regulates the various adenylyl cyclase isozymes. J Neurochem. 1998;71(4):1525–1534. [PubMed]
15.
Childers SR, Deadwyler SA. Role of cyclic AMP in the actions of cannabinoid receptors. Biochem Pharmacol. 1996;52(6):819–827. [PubMed]
16.
Deadwyler SA, Hampson RE, Mu J. et al. Cannabinoids modulate voltage sensitive potassium A-current in hippocampal neurons via a cAMP-dependent process. J Pharmacol Exp Ther.1995;273(2):734–743. [PubMed]
17.
Hampson RE, Evans GJ, Mu J. et al. Role of cyclic AMP dependent protein kinase in cannabinoid receptor modulation of potassium “A-current” in cultured rat hippocampal neurons. Life Sci.1995;56(23-24):2081–2088. [PubMed]
18.
Zhou D, Song ZH. CB1 cannabinoid receptor-mediated neurite remodeling in mouse neuroblastoma N1E-115 cells. J Neurosci Res. 2001;65(4):346–353. [PubMed]
19.
Zhou D, Song ZH. CB1 cannabinoid receptor-mediated tyrosine phosphorylation of focal adhesion kinase-related nonkinase. FEBS Lett. 2002;525(1-3):164–168. [PubMed]
20.
Derkinderen P, Toutant M, Burgaya F. et al. Regulation of a neuronal form of focal adhesion kinase by anandamide. Science. 1996;273(5282):1719–1722. [PubMed]
21.
Derkinderen P, Toutant M, Kadare G. et al. Dual role of Fyn in the regulation of FAK+6,7 by cannabinoids in hippocampus. J Biol Chem. 2001;276(41):38289–38296. [PubMed]
22.
Rueda D, Navarro B, Martinez-Serrano A. et al. The endocannabinoid anandamide inhibits neuronal progenitor cell differentiation through attenuation of the Rap1/B-Raf/ERK pathway. J Biol Chem.2002;277(48):46645–46650. [PubMed]
23.
Sugiura T, Kodaka T, Kondo S. et al. 2-Arachidonoylglycerol, a putative endogenous cannabinoid receptor ligand, induces rapid, transient elevation of intracellular free Ca2+ in neuroblastoma x glioma hybrid NG108-15 cells. Biochem Biophys Res Commun. 1996;229(1):58–64. [PubMed]
24.
Sugiura T, Kodaka T, Kondo S. et al. Inhibition by 2-arachidonoylglycerol, a novel type of possible neuromodulator, of the depolarization-induced increase in intracellular free calcium in neuroblastoma x glioma hybrid NG108-15 cells. Biochem Biophys Res Commun. 1997;233(1):207–210. [PubMed]
25.
Sugiura T, Kodaka T, Kondo S. et al. Is the cannabinoid CB1 receptor a 2-arachidonoylglycerol receptor? Structural requirements for triggering a Ca2+ transient in NG108-15 cells. J Biochem (Tokyo). 1997;122(4):890–895. [PubMed]
26.
Sugiura T, Kodaka T, Nakane S. et al. Evidence that the cannabinoid CB1 receptor is a 2-arachidonoylglycerol receptor. Structureactivity relationship of 2-arachidonoylglycerol, ether-linked analogues, and related compounds. J Biol Chem. 1999;274(5):2794–2801. [PubMed]
27.
Netzeband JG, Conroy SM, Parsons KL. et al. Cannabinoids enhance NMDA-elicited Ca2+ signals in cerebellar granule neurons in culture. J Neurosci. 1999;19(20):8765–8777. [PubMed]
28.
Felder CC, Veluz JS, Williams HL. et al. Cannabinoid agonists stimulate both receptor- and nonreceptor-mediated signal transduction pathways in cells transfected with and expressing cannabinoid receptor clones. Mol Pharmacol. 1992;42(5):838–845. [PubMed]
29.
Felder CC, Joyce KE, Briley EM. et al. Comparison of the pharmacology and signal transduction of the human cann
abinoid CB1 and CB2 receptors. Mol Pharmacol. 1995;48(3):443–450. [PubMed]
30.
Mackie K, Lai Y, Westenbroek R. et al. Cannabinoids activate an inwardly rectifying potassium conductance and inhibit Q-type calcium currents in AtT20 cells transfected with rat brain cannabinoid receptor. J Neurosci. 1995;15(10):6552–6561. [PubMed]
31.
Henry DJ, Chavkin C. Activation of inwardly rectifying potassium channels (GIRK1) by coexpressed rat brain cannabinoid receptors in Xenopus oocytes. Neurosci Lett. 1995;186(2-3):91–94. [PubMed]
32.
McAllister SD, Griffin G, Satin LS. et al. Cannabinoid receptors can activate and inhibit G protein-coupled inwardly rectifying potassium channels in a xenopus oocyte expression system. J Pharmacol Exp Ther. 1999;291(2):618–626. [PubMed]
33.
Gebremedhin D, Lange AR, Campbell WB. et al. Cannabinoid CB1 receptor of cat cerebral arterial muscle functions to inhibit L-type Ca2+ channel current. Am J Physiol. 1999;276(6 Pt 2):H2085–H2093. [PubMed]
34.
Caulfield MP, Brown DA. Cannabinoid receptor agonists inhibit Ca current in NG108-15 neuroblastoma cells via a pertussis toxin-sensitive mechanism. Br J Pharmacol. 1992;106(2):231–232. [PMC free article] [PubMed]
35.
Mackie K, Hille B. Cannabinoids inhibit N-type calcium channels in neuroblastoma-glioma cells.Proc Natl Acad Sci USA. 1992;89(9):3825–3829. [PMC free article] [PubMed]
36.
Mackie K, Devane WA, Hille B. Anandamide, an endogenous cannabinoid, inhibits calcium currents as a partial agonist in N18 neuroblastoma cells. Mol Pharmacol. 1993;44(3):498–503. [PubMed]
37.
Priller J, Briley EM, Mansouri J. et al. Mead ethanolamide, a novel eicosanoid, is an agonist for the central (CB1) and peripheral (CB2) cannabinoid receptors. Mol Pharmacol. 1995;48(2):288–292.[PubMed]
38.
Pan X, Ikeda SR, Lewis DL. Rat brain cannabinoid receptor modulates N-type Ca2+ channels in a neuronal expression system. Mol Pharmacol. 1996;49(4):707–714. [PubMed]
39.
Hampson AJ, Bornheim LM, Scanziani M. et al. Dual effects of anandamide on NMDA receptor-mediated responses and neurotransmission. J Neurochem. 1998;70(2):671–676. [PubMed]
40.
Rueda D, Galve-Roperh I, Haro A. et al. The CB(1) cannabinoid receptor is coupled to the activation of c-Jun N-terminal kinase. Mol Pharmacol. 2000;58(4):814–820. [PubMed]
41.
Liu J, Gao B, Mirshahi F. et al. Functional CB1 cannabinoid receptors in human vascular endothelial cells. Biochem J. 2000;346(Pt 3):835–840. [PMC free article] [PubMed]
42.
Bouaboula M, Poinot-Chazel C, Bourrie B. et al. Activation of mitogen-activated protein kinases by stimulation of the central cannabinoid receptor CB1. Biochem J. 1995;312(Pt 2):637–641. [PMC free article] [PubMed]
43.
Guzman M, Sanchez C. Effects of cannabinoids on energy metabolism. Life Sci. 1999;65(6-7):657–664. [PubMed]
44.
Sanchez C, Galve-Roperh I, Rueda D. et al. Involvement of sphingomyelin hydrolysis and the mitogen-activated protein kinase cascade in the Delta9-tetrahydrocannabinol-induced stimulation of glucose metabolism in primary astrocytes. Mol Pharmacol. 1998;54(5):834–843. [PubMed]
45.
Wartmann M, Campbell D, Subramanian A. et al. The MAP kinase signal transduction pathway is activated by the endogenous cannabinoid anandamide. FEBS Lett. 1995;359(2-3):133–136.[PubMed]
46.
Galve-Roperh I, Rueda D, Gomez dP. et al. Mechanism of extracellular signal-regulated kinase activation by the CB(1) cannabinoid receptor. Mol Pharmacol. 2002;62(6):1385–1392. [PubMed]
47.
Gomez dP, Velasco G, Guzman M. The CB1 cannabinoid receptor is coupled to the activation of protein kinase B/Akt. Biochem J. 2000;347(Pt 2):369–373. [PMC free article] [PubMed]
48.
Bouaboula M, Bourrie B, Rinaldi-Carmona M. et al. Stimulation of cannabinoid receptor CB1induces krox-24 expression in human astrocytoma cells. J Biol Chem. 1995;270(23):13973–13980.[PubMed]
49.
Fimiani C, Liberty T, Aquirre AJ. et al. Opiate, cannabinoid, and eicosanoid signaling converges on common intracellular pathways nitric oxide coupling. Prostaglandins Other Lipid Mediat.1999;57(1):23–34. [PubMed]
50.
Prevot V, Rialas CM, Croix D. et al. Morphine and anandamide coupling to nitric oxide stimulates GnRH and CRF release from rat median eminence: Neurovascular regulation. Brain Res.1998;790(1-2):236–244. [PubMed]
51.
Simmons ML, Murphy S. Induction of nitric oxide synthase in glial cells. J Neurochem.1992;59(3):897–905. [PubMed]
52.
Stefano GB, Salzet B, Rialas CM. et al. Morphine- and anandamide-stimulated nitric oxide production inhibits presynaptic dopamine release. Brain Res. 1997;763(1):63–68. [PubMed]
53.
Stefano GB, Salzet B, Salzet M. Identification and characterization of the leech CNS cannabinoid receptor: Coupling to nitric oxide release. Brain Res. 1997;753(2):219–224. [PubMed]
54.
Stefano GB, Salzet M, Magazine HI. et al. Antagonism of LPS and IFN-gamma induction of iNOS in human saphenous vein endothelium by morphine and anandamide by nitric oxide inhibition of adenylate cyclase. J Cardiovasc Pharmacol. 1998;31(6):813–820. [PubMed]
55.
Fimiani C, Mattocks D, Cavani F. et al. Morphine and anandamide stimulate intracellular calcium transients in human arterial endothelial cells: Coupling to nitric oxide release. Cell Signal.1999;11(3):189–193. [PubMed]
56.
Mombouli JV, Schaeffer G, Holzmann S. et al. Anandamide-induc
ed mobilization of cytosolic Ca2+in endothelial cells. Br J Pharmacol. 1999;126(7):1593–1600. [PMC free article] [PubMed]
57.
Maccarrone M, Bari M, Lorenzon T. et al. Anandamide uptake by human endothelial cells and its regulation by nitric oxide. J Biol Chem. 2000;275(18):13484–13492. [PubMed]
58.
Stefano GB, Liu Y, Goligorsky MS. Cannabinoid receptors are coupled to nitric oxide release in invertebrate immunocytes, microglia, and human monocytes. J Biol Chem. 1996;271(32):19238–19242. [PubMed]
59.
Gonsiorek W, Lunn C, Fan X. et al. Endocannabinoid 2-arachidonyl glycerol is a full agonist through human type 2 cannabinoid receptor: Antagonism by anandamide. Mol Pharmacol. 2000;57(5):1045–1050. [PubMed]
60.
Slipetz DM, O’Neill GP, Favreau L. et al. Activation of the human peripheral cannabinoid receptor results in inhibition of adenylyl cyclase. Mol Pharmacol. 1995;48(2):352–361. [PubMed]
61.
Bayewitch M, Rhee MH, Avidor-Reiss T. et al. (-)-Delta9-tetrahydrocannabinol antagonizes the peripheral cannabinoid receptor-mediated inhibition of adenylyl cyclase. J Biol Chem.1996;271(17):9902–9905. [PubMed]
62.
Bouaboula M, Poinot-Chazel C, Marchand J. et al. Signaling pathway associated with stimulation of CB2 peripheral cannabinoid receptor. Involvement of both mitogen-activated protein kinase and induction of Krox-24 expression. Eur J Biochem. 1996;237(3):704–711. [PubMed]
63.
Galve-Roperh I, Sanchez C, Cortes ML. et al. Anti-tumoral action of cannabinoids: Involvement of sustained ceramide accumulation and extracellular signal-regulated kinase activation. Nat Med.2000;6(3):313–319. [PubMed]
64.
Jacobsson SO, Wallin T, Fowler CJ. Inhibition of rat C6 glioma cell proliferation by endogenous and synthetic cannabinoids. Relative involvement of cannabinoid and vanilloid receptors. J Pharmacol Exp Ther. 2001;299(3):951–959. [PubMed]
65.
Guzman M, Galve-Roperh I, Sanchez C. Ceramide: A new second messenger of cannabinoid action. Trends Pharmacol Sci. 2001;22(1):19–22. [PubMed]
66.
Sanchez C, de CeballosML, del Pulgar TG. et al. Inhibition of glioma growth in vivo by selective activation of the CB(2) cannabinoid receptor. Cancer Res. 2001;61(15):5784–5789. [PubMed]
67.
Jarai Z, Wagner JA, Varga K. et al. Cannabinoid-induced mesenteric vasodilation through an endothelial site distinct from CB1 or CB2 receptors. Proc Natl Acad Sci USA. 1999;96(24):14136–14141. [PMC free article] [PubMed]
68.
Mukhopadhyay S, Chapnick BM, Howlett AC. Anandamide-induced vasorelaxation in rabbit aortic rings has two components: G protein dependent and independent. Am J Physiol Heart Circ Physiol.2002;282(6):H2046–H2054. [PubMed]
69.
Offertaler L, Mo FM, Batkai S. et al. Selective ligands and cellular effectors of a G protein-coupled endothelial cannabinoid receptor. Mol Pharmacol. 2003;63(3):699–705. [PubMed]
70.
Wagner JA, Varga K, Jarai Z. et al. Mesenteric vasodilation mediated by endothelial anandamide receptors. Hypertension. 1999;33(1 Pt 2):429–434. [PubMed]
71.
Wagner JA, Varga K, Jarai Z. et al. Mesenteric vasodilation mediated by endothelial anandamide receptors. Hypertension. 1999;33(1 Pt 2):429–434. [PubMed]
72.
Mukhopadhyay S, McCollum L, Howlett AC. Anandamide-mediated NO production in endothelial cells is regulated by Gi proteins: Further proof of existence of a novel nonCB1 anandamide receptor in vasculature. FASEB J. 2003;16:A111.
73.
Wagner JA, Varga K, Jarai Z. et al. Mesenteric vasodilation mediated by endothelial anandamide receptors. Hypertension. 1999;33(1 Pt 2):429–434. [PubMed]
74.
Wagner JA, Varga K, Jarai Z. et al. Mesenteric vasodilation mediated by endothelial anandamide receptors. Hypertension. 1999;33(1 Pt 2):429–434. [PubMed]
75.
White R, Hiley CR. A comparison of EDHF-mediated and anandamide-induced relaxations in the rat isolated mesenteric artery. Br J Pharmacol. 1997;122(8):1573–1584. [PMC free article] [PubMed]
76.
Grainger J, Boachie-Ansah G. Anandamide-induced relaxation of sheep coronary arteries: The role of the vascular endothelium, arachidonic acid metabolites and potassium channels. Br J Pharmacol. 2001;134(5):1003–1012. [PMC free article] [PubMed]
77.
White R, Ho WS, Bottrill FE. et al. Mechanisms of anandamide-induced vasorelaxation in rat isolated coronary arteries. Br J Pharmacol. 2001;134(4):921–929. [PMC free article] [PubMed]
78.
Houston DB, Howlett AC. Differential receptor-G-protein coupling evoked by dissimilar cannabinoid receptor agonists. Cell Signal. 1998;10(9):667–674. [PubMed]
79.
Mukhopadhyay S, McIntosh HH, Houston DB. et al. The CB(1) cannabinoid receptor juxtamembrane C-terminal peptide confers activation to specific G proteins in brain. Mol Pharmacol. 2000;57(1):162–170. [PubMed]
80.
Mukhopadhyay S, Howlett AC. CB1 receptor-G protein association. Subtype selectivity is determined by distinct intracellular domains. Eur J Biochem. 2001;268(3):499–505. [PubMed]
81.
Howlett AC, Mukhopadhyay S, Shim JY. et al. Signal transduction of eicosanoid CB1 receptor ligands. Life Sci. 1999;65(6-7):617–625. [PubMed]
82.
Howlett AC, Berglund B, Melvin LS. Cannabinoid receptor agonists and antagonists. Curr Pharm Des. 1995;1:343–354.
83.
Razdan RK. Structureactivity relationships in cannabinoids. Pharmacol Rev. 1986;38(2):75–149.[PubMed]
84.
Song ZH, Bonner TI. A lysine residue of the cannabinoid receptor is critical for receptor recognition by several
agonists but not WIN55212-2. Mol Pharmacol. 1996;49(5):891–896. [PubMed]
85.
Chin CN, Lucas-Lenard J, Abadji V. et al. Ligand binding and modulation of cyclic AMP levels depend on the chemical nature of residue 192 of the human cannabinoid receptor 1. J Neurochem.1998;70(1):366–373. [PubMed]
86.
Reggio PH. Molecular determinants for cannabinoid activity: Refinement of a molecular reactivity template. NIDA Res Monogr. 1987;79:82–95. [PubMed]
87.
Semus SF, Martin BR. A computergraphic investigation into the pharmacological role of the THC-cannabinoid phenolic moiety. Life Sci. 1990;46(24):1781–1785. [PubMed]
88.
Showalter VM, Compton DR, Martin BR. et al. 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. 1996;278(3):989–999. [PubMed]
89.
Gareau Y, Dufresne C, Gallant M. et al. Structure activity relationships of tetrahydrocannabinol analogues on human cannabinoid receptors. Bioorg Med Chem Lett. 1996;6:189–194.
90.
Huffman JW, Yu S, Showalter V. et al. Synthesis and pharmacology of a very potent cannabinoid lacking a phenolic hydroxyl with high affinity for the CB2 receptor. J Med Chem. 1996;39(20):3875–3877. [PubMed]
91.
Huffman JW, Liddle J, Yu S. et al. 3-(1′,1′-Dimethylbutyl)-1-deoxy-delta8-THC and related compounds: Synthesis of selective ligands for the CB2 receptor. Bioorg Med Chem.1999;7(12):2905–2914. [PubMed]
92.
Hanus L, Breuer A, Tchilibon S. et al. HU-308: A specific agonist for CB(2), a peripheral cannabinoid receptor. Proc Natl Acad Sci USA. 1999;96(25):14228–14233. [PMC free article] [PubMed]
93.
Chin CN, Murphy JW, Huffman JW. et al. The third transmembrane helix of the cannabinoid receptor plays a role in the selectivity of aminoalkylindoles for CB2, peripheral cannabinoid receptor. J Pharmacol Exp Ther. 1999;291(2):837–844. [PubMed]
94.
Howlett AC, Song C, Berglund BA. et al. Characterization of CB1 cannabinoid receptors using receptor peptide fragments and site-directed antibodies. Mol Pharmacol. 1998;53(3):504–510.[PubMed]
95.
Mukhopadhyay S, Cowsik SM, Lynn AM. et al. Regulation of Gi by the CB1 cannabinoid receptor C-terminal juxtamembrane region: Structural requirements determined by peptide analysis.Biochemistry. 1999;38(11):3447–3455. [PubMed]
96.
Jin W, Brown S, Roche JP. et al. Distinct domains of the CB1 cannabinoid receptor mediate desensitization and internalization. J Neurosci. 1999;19(10):3773–3780. [PubMed]
97.
Mukhopadhyay S, Sandiford S, Howlett AC. Palmitoylation regulates CB1 cannabinoid receptor-G protein interaction. Proc XIV World Congress Pharmacol. 2002
98.
Hsieh C, Brown S, Derleth C. et al. Internalization and recycling of the CB1 cannabinoid receptor. J Neurochem. 1999;73(2):493–501. [PubMed]
99.
Ulfers AL, McMurry JL, Miller A. et al. Cannabinoid receptor-G protein interactions: G(alphai1)-bound structures of IC3 and a mutant with altered G protein specificity. Protein Sci.2002;11(10):2526–2531. [PMC free article] [PubMed]
100.
Ulfers AL, McMurry JL, Kendall DA. et al. Structure of the third intracellular loop of the human cannabinoid 1 receptor. Biochemistry. 2002;41(38):11344–11350. [PubMed]
101.
Garcia DE, Brown S, Hille B. et al. Protein kinase C disrupts cannabinoid actions by phosphorylation of the CB1 cannabinoid receptor. J Neurosci. 1998;18(8):2834–2841. [PubMed]
102.
Shim JY, Welsh WJ, Howlett AC. Homology model of the CB1 cannabinoid receptor: Sites critical for nonclassical cannabinoid agonist interaction. Biopolymers. 2003;71:169–189. [PubMed]
103.
Abadji V, Lucas-Lenard JM, Chin C. et al. Involvement of the carboxyl terminus of the third intracellular loop of the cannabinoid CB1 receptor in constitutive activation of Gs. J Neurochem.1999;72(5):2032–2038. [PubMed]
104.
Howlett AC. The CB1 cannabinoid receptor in the brain. Neurobiol Dis. 1998;5(6 Pt B):405–416.[PubMed]
105.
Roche JP, Bounds S, Brown S. et al. A mutation in the second transmembrane region of the CB1receptor selectively disrupts G protein signaling and prevents receptor internalization. Mol Pharmacol. 1999;56(3):611–618. [PubMed]
106.
Tao Q, Abood ME. Mutation of a highly conserved aspartate residue in the second transmembrane domain of the cannabinoid receptors, CB1 and CB2, disrupts G-protein coupling. J Pharmacol Exp Ther. 1998;285(2):651–658. [PubMed]
107.
Barnett-Norris J, Hurst DP, Lynch DL. et al. Conformational memories and the endocannabinoid binding site at the cannabinoid CB1 receptor. J Med Chem. 2002;45(17):3649–3659. [PubMed]
Copyright © 2000-2013, Landes Bioscience.
potp font 1