4,383
Views
77
CrossRef citations to date
0
Altmetric
Review Article

Cannabinoids and the skeleton: From marijuana to reversal of bone loss

, &
Pages 560-567 | Published online: 21 Dec 2009

Abstract

The active component of marijuana, Δ9-tetrahydrocannabinol, activates the CB1 and CB2 cannabinoid receptors, thus mimicking the action of endogenous cannabinoids. CB1 is predominantly neuronal and mediates the cannabinoid psychotropic effects. CB2 is predominantly expressed in peripheral tissues, mainly in pathological conditions. So far the main endocannabinoids, anandamide and 2-arachidonoylglycerol, have been found in bone at ‘brain’ levels. The CB1 receptor is present mainly in skeletal sympathetic nerve terminals, thus regulating the adrenergic tonic restrain of bone formation. CB2 is expressed in osteoblasts and osteoclasts, stimulates bone formation, and inhibits bone resorption. Because low bone mass is the only spontaneous phenotype so far reported in CB2 mutant mice, it appears that the main physiologic involvement of CB2 is associated with maintaining bone remodeling at balance, thus protecting the skeleton against age-related bone loss. Indeed, in humans, polymorphisms in CNR2, the gene encoding CB2, are strongly associated with postmenopausal osteoporosis. Preclinical studies have shown that a synthetic CB2-specific agonist rescues ovariectomy-induced bone loss. Taken together, the reports on cannabinoid receptors in mice and humans pave the way for the development of 1) diagnostic measures to identify osteoporosis-susceptible polymorphisms in CNR2, and 2) cannabinoid drugs to combat osteoporosis.

Introduction

The marijuana plant Cannabis Sativa has been cultivated for thousands of years for medical and recreational use in the form of marijuana or hashish. Its psychoactive effects have made it the most common drug of abuse. However, it affects not only the brain but virtually every organ system in the body. We now know that the active component of marijuana, Δ9-tetrahydrocannabinol (THC), acts on two distinct receptors that are distributed throughout the body, only one of which mediates the psychotropic effects. These receptors respond to endogenous ligands, termed endocannabinoids, with THC just mimicking the activity of these physiological activators. The endocannabinoids are produced and degraded by specific enzymes. Together, the receptors, ligands, and enzymes comprise the endocannabinoid system. The on-going discovery of this system during the last two decades and its relevance for many organ systems has fueled extensive research and tremendous interest from pharmaceutical companies for potential therapeutic applications. Progress in this field has exploded in the last decade. There is a huge literature, growing by the day, investigating the endocannabinoid system in neural and non-neural tissues. Numerous excellent reviews have appeared in the last several years that treat the history, biochemistry, pharmacology, and therapeutic potential of this system Citation[1–8].

In ancient times, Cannabis was used therapeutically to relieve pain, reduce inflammation, and as a sedative. It was also used extensively to treat migraine headaches and ulcers. It is now well established that THC produces numerous beneficial effects, including analgesia, appetite stimulation, nausea reduction, and reduction of intraocular pressure. THC also affects fertility, short-term memory, tumor growth, and motor co-ordination Citation[9], Citation[10]. The therapeutic use of THC has been hampered by psychotropic effects that have prevented general acceptance by the Federal Drug Administration (FDA). Marinol™, a synthetic THC in sesame oil, is the only FDA-approved cannabinoid agonist for use in the US. It is prescribed as an appetite stimulant in AIDS, gastric by-pass, and chemotherapy patients and also as an antiemetic for chemotherapy. Sativex™ was approved by Health Canada in 2005 to relieve pain and spasticity in multiple sclerosis. It is a sublingual spray made by blending two of the main active ingredients of cannabis, THC and cannabidiol (CBD), and used for the relief of neuropathic pain. It has been recently (December 2008) approved by the FDA for stage III clinical trials in the US. An antagonist of the type I cannabinoid receptor (CB1), marketed as Acomplia™ by Sanofi Aventis, was approved for use as an antiobesity drug in the European Union in 2006. It also blocks the weight-gain associated with nicotine withdrawal, reduces visceral fat content, and lowers LDL levels. However, its marketing in Europe as well as stage III clinical trials of Acomplia™ in the US have been recently halted due to a 2-fold increase in psychiatric (depression, anxiety, irritability) and gastrointestinal (nausea) side-effects Citation[11].

Key messages

  • Several key components of the endocannabinoid system have been identified in bone.

  • The main physiologic involvement of CB2 (type 2 cannabinoid receptor) is associated with maintaining bone remodeling at balance.

  • CB2 agonists are possible candidates for a combined antiresorptive and anabolic therapy for osteoporosis.

Recently, there has been a rapidly growing interest in the role of cannabinoids in the regulation of skeletal remodeling and bone mass, addressed in basic, translational, and clinical research. A recent citation search revealed as many as 80 publications addressing the skeletal cannabinoid system since the first publications in 2005 Citation[12], Citation[13]. This review focuses on the skeletal cannabinoid system as a therapeutic target for patients with osteoporosis and other skeletal deficits.

Skeletal remodeling

Bone structure displays sequential stages throughout life, comprising 1) a rapid skeletal growth phase accompanied by accrual of peak bone mass; 2) a steady state phase whereby bone mass remains constant; and 3) age-related bone loss Citation[14]. These changes are the consequence of a continuous process of resorption and formation of the mineralized matrix, referred to as bone remodeling. The remodeling process occurs concomitantly in multiple foci that encompass approximately 5% of trabecular, endosteal, and osteonal surfaces Citation[15]. The remodeling cycle in individual foci consists of a relatively rapid (i.e. a few weeks in humans) resorption of pre-existing mineralized matrix by a specific hematopoietic, monocyte-derived cell type, the osteoclast Citation[16]. It is then followed by a slower (i.e. a few months) stage of bone formation by another bone-specific, fibroblast-like cell type, the osteoblast Citation[17]. Different foci present different phases of the remodeling cycle. Healthy adults retain an overall balance between bone resorption and formation. The significance of balanced bone remodeling is demonstrated by osteoporosis, the most common metabolic bone disease in developed societies, which results from a net increase in bone resorption and bone loss, weakening of the skeleton, and increased fracture risk.

Abbreviations

The co-ordinated occurrence of multiple remodeling sites is suggestive of a complex hierarchical regulation consisting of local, autocrine/paracrine, and systemic endocrine regulatory systems Citation[18]. Indeed, numerous studies have demonstrated paracrine control of osteoclast formation and activity by factors such as receptor activator of NFκB ligand (RANKL), osteoprotegerin (OPG), macrophage colony-stimulating factor (M-CSF) and interleukin 6 (IL-6), which are derived from neighboring stromal cells, including osteoblasts and their precursors Citation[19–24]. Locally, osteoblasts are regulated mainly by bone morphogenetic proteins (BMPs) and Wnts Citation[25], Citation[26]. Systemically, it is well established that depletion of gonadal hormones in females and males favors bone loss Citation[27–29]. In addition, parathyroid hormone (PTH) Citation[30], Citation[31], calcitonin Citation[32], insulin-like growth factor I (IGF-I) Citation[33], osteogenic growth peptide (OGP) Citation[34], and duodenum-derived serotonin Citation[35] are involved in the control of bone formation. Bone remodeling is also subject to a central control by hypothalamic leptin and neuropeptide Y signaling as well as downstream sympathetic signaling through osteoblastic β2-adrenergic receptors (β2AR) Citation[36–38]. Furthermore, imbalances in bone remodeling, previously attributed to excessive thyroid activity and estrogen depletion, may result from the interaction between thyroid and follicular stimulating hormones and their receptors, which are expressed in bone cells Citation[39], Citation[40].

Cannabinoid receptors and ligands

The actions of cannabinoids are mediated mainly through the activation of G protein-coupled cannabinoid CB1 and CB2 receptors Citation[41]. CB1 and CB2 share 44% overall identity (68% identity for the transmembrane domains). CB1 is perhaps the most abundantly expressed G protein-coupled receptor in the central nervous system. It is also present in peripheral neurons and the gonads and to some extent in several other peripheral tissues. CB2 is expressed in the skeleton, immune system, cirrhotic liver, arteriosclerotic plaques, inflamed gastrointestinal mucosa, and in glial and inflammatory cells in pathological brain conditions Citation[42], Citation[43]. That CB1 and CB2 are not functionally identical is demonstrated by the presence of cannabinoid agonists and antagonists with distinct binding specificities to either receptor Citation[44], Citation[45]. Both receptors signal via the Gi/o subclass of G proteins, inhibiting stimulated adenylyl cyclase activity. Further downstream, the CBs induce the activation of p42/44 mitogen-activated protein kinase (MAPK), p38 MAPK, c-Jun N-terminal kinase, AP-1, the neural form of focal adhesion kinase, protein kinase B, and K+ and Ca2 + transients Citation[46]. It has been proposed that GPR55 and transient receptor potential vanilloid type 1 receptor (TRPV1) are also involved in endocannabinoid triggering of these events Citation[47–49].

The main CB1 and CB2 endogenous ligands are N-arachidonoylethanolamine (AEA or anandamide) and 2-arachidonoylglycerol (2-AG) Citation[50], Citation[51]. Anandamide is present in a variety of tissues, such as the brain, kidney, liver, spleen, testis, uterus, and blood, in picomol/g concentrations, with the highest levels reported in the central nervous system. The low anandamide concentrations have been attributed to low substrate (arachidonic acid esterified at the 1-position) levels for this pathway and/or the short anandamide half-life in vivo (t1/2<5 min) Citation[52], Citation[53]. Anandamide is biosynthesized through N-acyl phosphatidyl ethanolamine phospholipase D (NAPE-PLD)-dependent and -independent pathways Citation[54]. The main anandamide-degrading enzyme is fatty acid amide hydrolase (FAAH), a membrane-associated serine hydrolase enriched in the brain and liver Citation[55]. In general, the tissue distribution of 2-AG is similar to that of anandamide. However, its concentration in is 300–1,000 higher (ng/g range). 2-AG production has been demonstrated in the central nervous system, platelets, and macrophages, especially in response to stimulation by inflammatory agents such as lipopolysaccharide Citation[56], Citation[57]. It is generated from arachidonic acid-enriched membrane phospholipids, such as inositol phospholipids, through the combined actions of phospholipase C and diacylglycerol lipases (DAGLα and DAGLβ) Citation[58], Citation[59]. It has been proposed that, like other monoacylglycerols, 2-AG is metabolized by a monoacylglycerol lipase Citation[60].

A couple of striking observations lead us to assess the occurrence and role of a skeletal endocannabinoid system. One is that bone formation and bone mass, as well as the central production of at least one major endocannabinoid, 2-AG, are subject to negative control by leptin Citation[61]. The second observation is that traumatic brain injury enhances both bone formation Citation[62], Citation[63] and central 2-AG production Citation[64].

The skeletal endocannabinoid system

Several key components of the endocannabinoid system have been identified in bone. The main endocannabinoids, anandamide and 2-AG, are present in this tissue in pmol/g and nmol/g concentrations, respectively, levels similar to those found in the brain. Because the blood endocannabinoid levels are several orders of magnitude lower than those found in bone, it is very likely that anandamide and 2-AG are synthesized locally in the skeleton Citation[65]. Indeed, both ligands are produced by osteoblasts and osteoclasts in culture. In addition, diacylglycerol lipases (DAGLs) α and β, enzymes critically involved in 2-AG biosynthesis, are expressed in osteoblasts, osteocytes, and bone-lining cells Citation[66]. The respective anandamide biosynthetic and degrading enzymes, NAPE-PLD and FAAH Citation[55], are also expressed in bone cells (Citation[67] and our unpublished results). Although both 2-AG and anandamide are perceived as non-selective agonists of CB1 and CB2, findings in bone and bone cell cultures suggest differential effects of these ligands. While 2-AG activates CB1 in the sympathetic nerve terminals following a single or chronic administration to mice, thus stimulating bone formation (Citation[66] and our unpublished results), it has no effect on osteoblasts and may even act as an inverse agonist in these cells Citation[66], Citation[68]. Like the CB2-selective agonist, anandamide stimulates in vitro osteoblast proliferation (our unpublished results). In addition, the number of osteoclasts in culture is increased by a direct challenge with anandamide Citation[13] or through the action of the FAAH inhibitor URB597 that leads to increased anandamide levels endogenously Citation[67]. It remains to be seen whether these actions of anandamide are mediated by CB1, CB2, GPR55, and/or TRPV1.

Effects on bone cell differentiation and activity

Activation of CB2 has different effects in early osteoblast progenitors and in more mature osteoblastic cells. In the early precursors, represented by bone-marrow-derived, partially differentiated osteoblastic cells that show limited CB2 expression, the specific CB2 agonist HU-308 Citation[45], but not the specific CB1 agonist noladin ether Citation[69], triggers a Gi protein-mediated mitogenic effect and consequent expansion of the preosteoblastic pool. Ex vivo osteoblastic colony (CFU-Ob) formation by bone-marrow stromal cb2-/- cells is markedly diminished, whereas CFU-Ob formation by wild-type cells is stimulated by HU-308 Citation[70], Citation[71]. In mature osteoblastic cells, represented by the MC3T3 E1 cell line, the same ligand stimulates osteoblast-differentiated functions such as alkaline phosphatase activity and matrix mineralization Citation[12], Citation[70]. Hence, CB2 signaling is involved in several regulatory, pro-osteogenic processes along the osteoblast lineage.

In mouse bone-marrow-derived osteoclastogenic cultures and in the RAW 264.7 cell line, CB2 activation inhibits osteoclast formation by restraining mitogenesis at the monocytic stage, prior to incubation with RANKL. It also suppresses osteoclast formation by repressing RANKL expression in osteoblasts and osteoblast progenitors Citation[70]. Likewise, it has been recently shown that the cannabinoid receptor agonist ajulemic acid also suppresses osteoclastogenesis Citation[72]. By contrast, another study reported the stimulation of osteoclast formation and bone resorption by cannabinoid receptor agonists and their inhibition by antagonists Citation[13], Citation[73]. These allegedly paradoxical effects may be species- and/or agonist-dependent, as in human osteoclasts and other cells anandamide has been shown to activate also TRPV1 Citation[49]. TRPV1 activation in the human osteoclasts and osteoclast precursors enhances osteoclast formation and activity Citation[67] and may modify the effect of selective CB2 agonists. In addition to CB2, low levels of CB1 mRNA have been also reported in bone cell cultures Citation[13], Citation[67], Citation[70]. However, before drawing any conclusions related to the functional significance of CB1 in bone cells, its expression in these cells must be further evaluated at the protein level and in vivo using conditional gene ablation.

Skeletal phenotypes of cannabinoid receptor-deficient mice

Cannabinoid receptor mutant mice have been used to assess the physiologic role of CB1 and CB2 in the control of bone mass. In the case of CB1, the skeletal phenotype depends on the mouse strain and/or the construct used for gene mutation. In one CB1-deficient line, back-crossed to CD1 mice (CD1CB1-/-), the N-terminal 233 codons of the CNR1 gene (which encodes CB1) were ablated Citation[74]. The effect of this mutation shows a clear gender disparity. Females have normal trabecular bone with a slight cortical expansion, whereas male CD1CB1-/- mice exhibit high bone mass Citation[75]. Sexually mature CD1CB1-/- mice of either gender display normal bone formation and resorption parameters, suggesting that the male phenotype is acquired early in life, during the developmental phase when peak bone mass is determined. A similar male phenotype was reported when mice carrying the same mutated CNR1 gene were further back-crossed to Biozzi ABH mice Citation[13], Citation[65], Citation[76]. In the second line, back-crossed to C57BL6J mice (C57CB1-/-), almost the entire protein-encoding sequence was removed Citation[77]. Both male and female C57CB1-/- have a low bone mass phenotype accompanied by increased osteoclast counts and decreased bone formation rate Citation[75]. Our recent findings suggest that CB1 controls osteoblast function by negatively regulating norepinephrine (NE) release from sympathetic nerve terminals in the immediate vicinity of these cells. NE suppresses bone formation by binding to osteoblastic β2AR Citation[38]; this suppression is alleviated by activation of sympathetic CB1 Citation[66].

Animals with a CNR2-mutated gene (which encodes CB2) have a gender-independent skeletal phenotype. During their first 2–3 months of life, CNR2-/- mice accrue a normal peak trabecular bone mass but later display a markedly enhanced age-related bone loss; their trabecular bone volume density at 1 year of age is approximately half compared to wild-type controls Citation[70]. Reminiscent of human postmenopausal osteoporosis Citation[78], the CNR2-/- mice have a high bone turn-over with increases in both bone resorption and formation which are at a net negative balance Citation[70]. Importantly, low bone mass is the only spontaneous phenotype so far reported in these mice. Hence, because healthy CB2 mutant mice are otherwise normal, it appears that the main physiologic involvement of CB2 is associated with maintaining bone remodeling at balance.

The endocannabinoid system as a target for antiosteoporotic therapy and osteoporosis risk assessment

Unlike CB1, CB2 is not associated with the cannabinoid psychotropic effects. Therefore, CB2-specific ligands could offer an opportunity to prevent and/or rescue bone loss while avoiding the psychological adverse effects of cannabinoids. Indeed, the specific, non-psychoactive CB2 agonist HU-308 attenuates bone loss induced by estrogen depletion in ovariectomized (OVX) animals using either ‘preventive’ or ‘rescue’ protocols Citation[65], Citation[70]. In the preventive approach, HU-308 administration commenced immediately after OVX. To assess reversal of bone loss, the drug was given beginning 6 weeks post-OVX to allow for bone loss to occur. In either protocol, the cannabinoid treatment consisted of daily intraperitoneal injections for 4–6 weeks. The attenuation of bone mass reflected both inhibition of bone resorption and stimulation of bone formation. Hence, CB2 agonists are possible candidates for a combined antiresorptive and anabolic therapy for osteoporosis.

Interestingly, marijuana smoke inhalation was recently reported to inhibit endosseous implant anchorage in rats, negatively affecting both the bone-implant contact and peri-implant bone Citation[79]. This is not necessarily in contradiction to the bone anabolic activity of well defined cannabinoid receptor agonists, as marijuana contains a mixture of biologically active phytocannabinoid whose skeletal effects have not been tested yet. In addition, the peri-implant healing process may differ considerably from remodeling of the non-traumatized skeleton and thus respond differently to cannabinoids. Another potentially confounding issue is the non-selectivity/non-specificity of many cannabinoid ligands, either endogenous, plant-derived, or synthetic. The skeletal relevance of this issue has been recently demonstrated in a study showing that the ‘so-called’ CB2-selective inverse agonist AM360 at a daily dose of 0.1 mg/kg prevented OVX-induced bone loss in wild-type but not in CNR2-/- mice, therefore indicating CB2 selectivity at this low dose. However, the same preparation was equally effective in preventing bone loss in wild-type and CNR2-/- mice at higher doses Citation[73]. Hence, in the skeleton, and probably elsewhere, cannabinoid ligands may have CB1- and/or CB2-independent effects, depending on concentrations or doses used.

Polymorphisms in the human CNR1 and CNR2 loci were studied to assess the cannabinoid receptors as targets for the risk assessment and treatment of osteoporosis. The CNR1 locus is located on chromosome 5q15 and encompasses a single coding exon that is preceded by several non-coding 5′ exons, indicating a complex transcriptional regulation of this gene by different promoters Citation[80], Citation[81]. The CNR2 locus is located on chromosome 1p36. This genomic region and its mouse ortholog on chromosome 4 have been linked to bone mineral density (BMD) and osteoporosis in several independent association analyses Citation[82–84]. However, these analyses did not consider CNR2 as a potential candidate gene. Like CNR1, the CNR2 gene also consists of a single coding exon, which is preceded by a non-coding upstream exon.

Thus far, two genetic association studies have been reported dealing with the relationship between polymorphisms in CNR genes and osteoporosis. The first study was carried out in a French Caucasian sample comparing postmenopausal osteoporotic women with a low bone mineral density (BMD) and age-matched healthy controls Citation[85]. Analysis of four single nucleotide polymorphisms (SNPs) spanning nearly 20 kb around the CB1 coding exon revealed no significant association with the osteoporosis phenotype, suggesting that the CNR1 locus does not have a major role in this sample.

In the CNR2 gene a total of 26 SNPs were analyzed, spanning approximately 300 kb around the CNR2 locus. Several of these SNPs showed a significant association with the disease phenotype, suggesting that CNR2 polymorphisms are important genetic risk factors for osteoporosis. The most significant P-values for allele and genotype associations were observed with SNPs located within the CB2 coding region (P = 0.0014 and P = 0.00073, respectively). Furthermore, when BMD at the lumbar spine was analyzed as a quantitative trait, highly significant differences were found in BMD between individuals carrying different SNPs in the CB2 coding region. Indeed, sequencing the CB2 coding exon in all patients and controls identified two missense variants, Gln63Arg and His316Tyr, with the Arg63 variant being more common in the osteoporotic patients than in the healthy controls Citation[85]. Taken together, these findings suggest that a common variant of the CB2 receptor contributes to the etiology of osteoporosis in humans.

The second is a prospective study, which analyzed several candidate quantitative trait loci in BMD, including CNR2, in a cohort of 1,110 women and 1,128 Japanese men, 40–79 years of age Citation[86]. For the CNR2 locus, a single SNP (rs2501431, A→G) was assessed, which had shown the strongest association in the previously published French sample. BMD, measured by peripheral quantitative computed tomography or dual-energy X-ray absorptiometry, was consistently lower in women with the AA genotype compared to the AG and GG genotypes. Together, these studies strongly suggest that CNR2 is the susceptibility gene for low BMD and osteoporosis on chromosome 1p36.

Conclusions

Recent studies in mice and humans suggest an important role for the endocannabinoid system in the regulation of skeletal remodeling and the consequent implications on bone mass and biomechanical function. Although the CB1 cannabinoid receptor has been identified in sympathetic terminals innervating the skeleton, its role in controlling bone turn-over remains to be elucidated. The CB2 cannabinoid receptor is expressed in bone cells. Its bone anabolic action, including some of the mechanisms involved, has been reported in some detail, and is also inferred from human genetic studies. These studies portray polymorphisms in CNR2, the gene encoding CB2, as important genetic risk factors for osteoporosis. Taken together, the reports on cannabinoid receptors in mice and humans pave the way for the development of 1) diagnostic measures to identify osteoporosis-susceptible polymorphisms in CNR2, and 2) cannabinoid drugs to combat osteoporosis.

Acknowledgements

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Piomelli D. The molecular logic of endocannabinoid signalling. Nat Rev Neurosci. 2003; 4: 873–84
  • Lambert DM, Fowler CJ. The endocannabinoid system: drug targets, lead compounds, and potential therapeutic applications. J Med Chem. 2005; 48: 5059–87
  • Hohmann AG, Suplita RL 2nd. Endocannabinoid mechanisms of pain modulation. AAPS J. 2006; 8: E693–708
  • Pertwee RG. Cannabinoid pharmacology: the first 66 years. Br J Pharmacol. 2006; 147(Suppl 1)S163–71
  • Kogan NM, Mechoulam R. Cannabinoids in health and disease. Dialogues Clin Neurosci. 2007; 9: 413–30
  • Marx J. Drug development. Drugs inspired by a drug. Science. 2006; 311: 322–5
  • Yazulla S. Endocannabinoids in the retina: From marijuana to neuroprotection. Prog Retin Eye Res. 2008; 27: 501–26
  • Iversen L. The science of marijuana. Oxford University Press, New York 2000
  • Mechoulam R. Discovery of endocannabinoids and some random thoughts on their possible roles in neuroprotection and aggression. Prostaglandins Leukot Essent Fatty Acids. 2002; 66: 93–9
  • Iversen L. Cannabis and the brain. Brain. 2003; 126: 1252–70
  • Akbas F, Gasteyger C, Sjodin A, Astrup A, Larsen TM. A critical review of the cannabinoid receptor as a drug target for obesity management. Obes Rev. 2009; 10: 58–67
  • Bab I. The skeleton: stone bones and stoned heads?. Cannabinoids as therapeutics. Milestones in drug therapy series, R Mechoulam. Birkhäuser Verlag, Basel 2005; 201–6
  • Idris AI, van 't Hof RJ, Greig IR, Ridge SA, Baker D, Ross RA, et al. Regulation of bone mass, bone loss and osteoclast activity by cannabinoid receptors. Nat Med. 2005; 11: 774–9
  • Bab I, Hajbi-Yonissi C, Gabet Y, Müller R. Micro-tomographic atlas of the mouse skeleton. Springer, New York 2007; 195–9
  • Parfitt AM. The coupling of bone formation to bone resorption: a critical analysis of the concept and of its relevance to the pathogenesis of osteoporosis. Metab Bone Dis Relat Res. 1982; 4: 1–6
  • Roodman GD. Cell biology of the osteoclast. Exp Hematol. 1999; 27: 1229–41
  • Bab I, Ashton BA, Gazit D, Marx G, Williamson MC, Owen ME. Kinetics and differentiation of marrow stromal cells in diffusion chambers in vivo. J Cell Sci. 1986; 84: 139–51
  • Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000; 21: 115–37
  • Poli V, Balena R, Fattori E, Markatos A, Yamamoto M, Tanaka H, et al. Interleukin-6 deficient mice are protected from bone loss caused by estrogen depletion. EMBO J. 1994; 13: 1189–96
  • Suda T, Kobayashi K, Jimi E, Udagawa N, Takahashi N. The molecular basis of osteoclast differentiation and activation. Novartis Found Symp. 2001; 232: 235–47
  • Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Lüthy R, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997; 89: 309–19
  • Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998; 93: 165–76
  • Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, et al. osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev. 1998; 12: 1260–8
  • Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999; 397: 315–23
  • Yoshida Y, Tanaka S, Umemori H, Minowa O, Usui M, Ikematsu N, et al. Negative regulation of BMP/Smad signaling by Tob in osteoblasts. Cell. 2000; 103: 1085–97
  • Bodine PV, Komm BS. Wnt signaling and osteoblastogenesis. Rev Endocr Metab Disord. 2006; 7: 33–9
  • Most W, van der Wee-Pals L, Ederveen A, Papapoulos S, Lowik C. Ovariectomy and orchidectomy induce a transient increase in the osteoclastogenic potential of bone marrow cells in the mouse. Bone. 1997; 20: 27–30
  • Gabet Y, Kohavi D, Muller R, Chorev M, Bab I. Intermittently administered parathyroid hormone 1–34 reverses bone loss and structural impairment in orchiectomized adult rats. Osteoporos Int. 2005; 16: 1436–43
  • Alexander JM, Bab I, Fish S, Müller R, Uchiyama T, Gronowicz G, et al. Human parathyroid hormone 1–34 reverses bone loss in ovariectomized mice. J Bone Miner Res. 2001; 16: 1665–73
  • Potts JT, Juppner H. Parathyroid hormone and parathyroid hormone-related peptide in calcium homeostasis, bone metabolism, and bone development: the proteins, their genes, and receptors. Academic Press, San Diego 1998
  • Gunther T, Chen ZF, Kim J, Priemel M, Rueger JM, Amling M, et al. Genetic ablation of parathyroid glands reveals another source of parathyroid hormone. Nature. 2000; 406: 199–203
  • Nicholson GC, Moseley JM, Sexton PM, Mendelsohn FA, Martin TJ. Abundant calcitonin receptors in isolated rat osteoclasts. Biochemical and autoradiographic characterization. J Clin Invest. 1986; 78: 355–60
  • Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL, Wu Y, Liu JL, et al. Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest. 2002; 110: 771–81
  • Bab I, Chorev M. Osteogenic growth peptide: from concept to drug design. Biopolymers. 2002; 66: 33–48
  • Yadav VK, Ryu JH, Suda N, Tanaka KF, Gingrich JA, Schütz G, et al. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell. 2008; 135: 825–37
  • Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell. 2000; 100: 197–207
  • Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, et al. Hypothalamic Y2 receptors regulate bone formation. J Clin Invest. 2002; 109: 915–21
  • Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, et al. Leptin regulates bone formation via the sympathetic nervous system. Cell. 2002; 111: 305–17
  • Abe E, Marians RC, Yu W, Wu XB, Ando T, Li Y, et al. TSH is a negative regulator of skeletal remodeling. Cell. 2003; 115: 151–62
  • Sun L, Peng Y, Sharrow AC, Iqbal J, Zhang Z, Papachristou DJ, et al. FSH directly regulates bone mass. Cell. 2006; 125: 247–60
  • Howlett AC. The cannabinoid receptors. Prostaglandins Other Lipid Mediat. 2002; 68–69: 619–31
  • Julien B, Grenard P, Teixeira-Clerc F, Van Nhieu JT, Li L, Karsak M, et al. Antifibrogenic role of the cannabinoid receptor CB2 in the liver. Gastroenterology. 2005; 128: 742–55
  • Steffens S, Veillard NR, Arnaud C, Pelli G, Burger F, Staub C, et al. Low dose oral cannabinoid therapy reduces progression of atherosclerosis in mice. Nature. 2005; 434: 782–6
  • Shire D, Calandra B, Bouaboula M, Barth F, Rinaldi-Carmona M, Casellas P, et al. Cannabinoid receptor interactions with the antagonists SR 141716A and SR 144528. Life Sci. 1999; 65: 627–35
  • Hanus L, Breuer A, Tchilibon S, Shiloah S, Goldenberg D, Horowitz M, et al. HU-308: a specific agonist for CB(2), a peripheral cannabinoid receptor. Proc Natl Acad Sci U S A. 1999; 96: 14228–33
  • Howlett, AC. Cannabinoid receptor signaling. Handb Exp Pharmacol 2005; 53–79
  • Lauckner JE, Jensen JB, Chen HY, Lu HC, Hille B, Mackie K. GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc Natl Acad Sci U S A. 2008; 105: 2699–704
  • Pertwee RG. GPR55: a new member of the cannabinoid receptor clan?. Br J Pharmacol. 2007; 152: 984–6
  • De Petrocellis L, Bisogno T, Maccarrone M, Davis JB, Finazzi-Agro A, Di Marzo V. The activity of anandamide at vanilloid VR1 receptors requires facilitated transport across the cell membrane and is limited by intracellular metabolism. J Biol Chem. 2001; 276: 12856–63
  • Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992; 258: 1946–9
  • Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995; 50: 83–90
  • Hansen HS, Moesgaard B, Hansen HH, Petersen G. N-Acylethanolamines and precursor phospholipids—relation to cell injury. Chem Phys Lipids. 2000; 108: 135–50
  • Willoughby KA, Moore SF, Martin BR, Ellis EF. The biodisposition and metabolism of anandamide in mice. J Pharmacol Exp Ther. 1997; 282: 243–7
  • Simon GM, Cravatt BF. Endocannabinoid biosynthesis proceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/beta-hydrolase 4 in this pathway. J Biol Chem. 2006; 281: 26465–72
  • Cravatt BF, Demarest K, Patricelli MP, Bracey MH, Giang DK, Martin BR, et al. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc Natl Acad Sci U S A. 2001; 98: 9371–6
  • Varga K, Wagner JA, Bridgen DT, Kunos G. Platelet- and macrophage-derived endogenous cannabinoids are involved in endotoxin-induced hypotension. FASEB J. 1998; 12: 1035–44
  • Di Marzo V, Bisogno T, De Petrocellis L, Melck D, Orlando P, Wagner JA, et al. Biosynthesis and inactivation of the endocannabinoid 2-arachidonoylglycerol in circulating and tumoral macrophages. Eur J Biochem. 1999; 264: 258–67
  • Stella N, Schweitzer P, Piomelli D. A second endogenous cannabinoid that modulates long-term potentiation. Nature. 1997; 388: 773–8
  • Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A, et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J Cell Biol. 2003; 163: 463–8
  • Konrad RJ, Major CD, Wolf BA. Diacylglycerol hydrolysis to arachidonic acid is necessary for insulin secretion from isolated pancreatic islets: sequential actions of diacylglycerol and monoacylglycerol lipases. Biochemistry. 1994; 33: 13284–94
  • Di Marzo V, Goparaju SK, Wang L, Liu J, Bátkai S, Járai Z, et al. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature. 2001; 410: 822–5
  • Orzel JA, Rudd TG. Heterotopic bone formation: clinical, laboratory, and imaging correlation. J Nucl Med. 1985; 26: 125–32
  • Wildburger R, Zarkovic N, Tonkovic G, Skoric T, Frech S, Hartleb M, et al. Post-traumatic hormonal disturbances: prolactin as a link between head injury and enhanced osteogenesis. J Endocrinol Invest. 1998; 21: 78–86
  • Panikashvili D, Simeonidou C, Ben-Shabat S, Hanus L, Breuer A, Mechoulam R, et al. An endogenous cannabinoid (2-AG) is neuroprotective after brain injury. Nature. 2001; 413: 527–31
  • Bab I, Ofek O, Tam J, Rehnelt J, Zimmer A. Endocannabinoids and the regulation of bone metabolism. J Neuroendocrinol 2008; 20(Suppl 1)69–74
  • Tam J, Trembovler V, Di Marzo V, Petrosino S, Leo G, Alexandrovich A, et al. The cannabinoid CB1 receptor regulates bone formation by modulating adrenergic signaling. FASEB J. 2008; 22: 285–94
  • Rossi F, Siniscalco D, Luongo L, De Petrocellis L, Bellini G, Petrosino S, et al. The endovanilloid/endocannabinoid system in human osteoclasts: possible involvement in bone formation and resorption. Bone. 2009; 44: 476–84
  • Bab I, Zimmer A. Cannabinoid receptors and the regulation of bone mass. Br J Pharmacol. 2008; 153: 182–8
  • Hanus L, Abu-Lafi S, Fride E, Breuer A, Vogel Z, Shalev DE, et al. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A. 2001; 98: 3662–5
  • Ofek O, Karsak M, Leclerc N, Fogel M, Frenkel B, Wright K, et al. Peripheral cannabinoid receptor, CB2, regulates bone mass. Proc Natl Acad Sci U S A. 2006; 103: 696–701
  • Scutt A, Williamson EM. Cannabinoids stimulate fibroblastic colony formation by bone marrow cells indirectly via CB2 receptors. Calcif Tissue Int. 2007; 80: 50–9
  • George KL, Saltman LH, Stein GS, Lian JB, Zurier RB. Ajulemic acid, a nonpsychoactive cannabinoid acid, suppresses osteoclastogenesis in mononuclear precursor cells and induces apoptosis in mature osteoclast-like cells. J Cell Physiol. 2008; 214: 714–20
  • Idris AI, Sophocleous A, Landao-Bassonga E, van 't Hof RJ, Ralston SH. Regulation of bone mass, osteoclast function, and ovariectomy-induced bone loss by the type 2 cannabinoid receptor. Endocrinology. 2008; 149: 5619–26
  • Ledent C, Valverde O, Cossu G, Petitet F, Aubert JF, Beslot F, et al. Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science. 1999; 283: 401–4
  • Tam J, Ofek O, Fride E, Ledent C, Gabet Y, Müller R, et al. Involvement of neuronal cannabinoid receptor CB1 in regulation of bone mass and bone remodeling. Mol Pharmacol. 2006; 70: 786–92
  • Amor S, Smith PA, Hart B, Baker D. Biozzi mice: of mice and human neurological diseases. J Neuroimmunol. 2005; 165: 1–10
  • Zimmer A, Zimmer AM, Hohmann AG, Herkenham M, Bonner TI. Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proc Natl Acad Sci U S A. 1999; 96: 5780–5
  • Brown JP, Delmas PD, Malaval L, Edouard C, Chapuy MC, Meunier PJ. Serum bone Gla-protein: a specific marker for bone formation in postmenopausal osteoporosis. Lancet. 1984; 1: 1091–3
  • Nogueira-Filho Gda R, Cadide T, Rosa BT, Neiva TG, Tunes R, Peruzzo D, et al. Cannabis sativa smoke inhalation decreases bone filling around titanium implants: a histomorphometric study in rats. Implant Dent. 2008; 17: 461–70
  • McCaw E, Hu H, Gomez GT, Hebb AL, Kelly ME, Denovan-Wright EM. Structure, expression and regulation of the cannabinoid receptor gene (CB1) in Huntington's disease transgenic mice. Eur J Biochem. 2004; 271: 4909–20
  • Zhang P, Ishiguro H, Ohtsuki T, Hess J, Carillo F, Walther D, et al. Human cannabinoid receptor 1: 5′ exons, candidate regulatory regions, polymorphisms, haplotypes and association with polysubstance abuse. Mol Psychiatry. 2004; 9: 916–31
  • Devoto M, Shimoya K, Caminis J, Ott J, Tenenhouse A, Whyt MP, et al. First-stage autosomal genome screen in extended pedigrees suggests genes predisposing to low bone mineral density on chromosomes 1p, 2p and 4q. Eur J Hum Genet. 1998; 6: 151–7
  • Devoto M, Specchia C, Li HH, Caminis J, Tenenhouse A, Rodriguez H, et al. Variance component linkage analysis indicates a QTL for femoral neck bone mineral density on chromosome 1p36. Hum Mol Genet. 2001; 10: 2447–52
  • Devoto M, Spotila LD, Stabley DL, Wharton GN, Rydbeck H, Korkko J, et al. Univariate and bivariate variance component linkage analysis of a whole-genome scan for loci contributing to bone mineral density. Eur J Hum Genet. 2005; 13: 781–8
  • Karsak M, Cohen-Solal M, Freudenberg J, Ostertag A, Morieux C, Kornak U, et al. Cannabinoid receptor type 2 gene is associated with human osteoporosis. Hum Mol Genet. 2005; 14: 3389–96
  • Yamada Y, Ando F, Shimokata H. Association of candidate gene polymorphisms with bone mineral density in community-dwelling Japanese women and men. Int J Mol Med. 2007; 19: 791–801

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.