Hemodynamic effects of cannabinoids: coronary and cerebral vasodilation mediated by cannabinoid CB1 receptors

https://doi.org/10.1016/S0014-2999(01)01112-8Get rights and content

Abstract

Activation of peripheral cannabinoid CB1 receptors elicits hypotension. Using the radioactive microsphere technique, we examined the effects of cannabinoids on systemic hemodynamics in anesthetized rats. The potent cannabinoid CB1 receptor agonist HU-210 ({−}-11-OH-Δ9 tetrahydrocannabinol dimethylheptyl, 10 μg/kg i.v.) reduced mean blood pressure by 57±5 mm Hg by decreasing cardiac index from 37±1 to 23±2 ml/min/100 g (P<0.05) without significantly affecting systemic vascular resistance index. HU-210 elicited a similar decrease in blood pressure following ganglionic blockade and vasopressin infusion. The endogenous cannabinoid anandamide (arachidonyl ethanolamide, 4 mg/kg i.v.) decreased blood pressure by 40±7 mm Hg by reducing systemic vascular resistance index from 3.3±0.1 to 2.3±0.1 mm Hg min/ml/100 g (P<0.05), leaving cardiac index and stroke volume index unchanged. HU-210, anandamide, and its metabolically stable analog, R-methanandamide, lowered vascular resistance primarily in the coronaries and the brain. These vasodilator effects remained unchanged when autoregulation was prevented by maintaining blood pressure through volume replacement, but were prevented by pretreatment with the cannabinoid CB1 receptor antagonist SR141716A (N-{piperidin-1-yl}-5-{4-chlorophenyl}-1-{2,4-dichlorophenyl}-4-methyl-1H-pyrazole-3-carboxamide HCl; 3 mg/kg i.v.). Only anandamide and R-methanandamide were vasodilators in the mesentery. We conclude that cannabinoids elicit profound coronary and cerebral vasodilation in vivo by direct activation of vascular cannabinoid CB1 receptors, rather than via autoregulation, a decrease in sympathetic tone or, in the case of anandamide, the action of a non-cannabinoid metabolite. Differences between the hemodynamic profile of various cannabinoids may reflect quantitative differences in cannabinoid CB1 receptor expression in different tissues and/or the involvement of as-yet-unidentified receptors.

Introduction

The psychoactive properties of cannabinoids, constituents of the marijuana plant, have long been recognized, and similar effects can be elicited by the endogenous ligand arachidonyl ethanolamide (anandamide) (Hillard, 2000). Recent research has revealed that cannabinoids elicit not only neurobehavioral but also cardiovascular effects (Kunos et al., 2000). The biological effects of cannabinoids are mediated by specific receptors, of which two subtypes have been identified, cannabinoid CB1 receptors present in the brain but also in some peripheral tissues (Matsuda et al., 1990) and cannabinoid CB2 receptors expressed by immune cells (Munro et al., 1993). Through the use of a selective cannabinoid CB1 receptor antagonist, SR141716A (N-{piperidin-1-yl}-5-{4-chlorophenyl}-1-{2,4-dichlorophenyl}-4-methyl-1H-pyrazole-3-carboxamide HCl; Rinaldi-Carmona et al., 1994), the hypotensive and bradycardic effects of cannabinoids in rodents could be attributed to activation of peripheral cannabinoid CB1 receptors Varga et al., 1995, Járai et al., 1999. This conclusion has been confirmed by the absence of such effects in cannabinoid CB1 receptor-knockout mice Járai et al., 1999, Ledent et al., 1999.

Recent findings implicate endogenous cannabinoids in cardiovascular regulation. Circulating macrophages and platelets activated by hemorrhage or by bacterial endotoxin synthesize anandamide and another endogenous cannabinoid, 2-arachidonoyl glycerol (2-AG), and these endocannabinoids contribute to the hypotension of hemorrhagic and septic shock via the activation of peripheral cannabinoid CB1 receptors Wagner et al., 1997, Varga et al., 1998. Vascular endothelial cells also contain anandamide (Deutsch et al., 1997) and 2-AG Mechoulam et al., 1998, Sugiura et al., 1998, and functional cannabinoid CB1 receptors are present on cerebrovascular smooth muscle cells (Gebremedhin et al., 1999). It has been proposed that the endothelium-derived hyperpolarizing factor (EDHF) released by various vasodilators may be an endocannabinoid (Randall et al., 1996), although this has been contested Plane et al., 1997, Pratt et al., 1998.

There is evidence that various cannabinoids produce strong vasodilation in vitro Chataigneau et al., 1998, Deutsch et al., 1997, Ellis et al., 1995, Wagner et al., 1999, but the underlying mechanisms are unclear (Pratt et al., 1998). However, there is a lack of information on the in vivo effects of cannabinoids on blood flow to various organs, and on the hemodynamic changes underlying their hypotensive action. In a study of the cardiovascular effects of a series of cannabinoids in anesthetized rats, the synthetic cannabinoid HU-210 ({−}-11-OH-Δ9-tetrahydrocannabinol dimethylheptyl) was found to be the most potent analog in producing hypotension and bradycardia with ED50's (doses producing half-maximal effects) of 2 and 90 μg/kg, respectively (Lake et al., 1997a). Moreover, at a maximally effective i.v. dose, HU-210 lowered blood pressure by 75–80 mm Hg for up to 2 h, whereas a maximal hypotensive dose of anandamide lowered blood pressure transiently (∼15 min) by no more than 45 mm Hg (Lake et al., 1997a). The way this different degree of blood pressure reduction is achieved, e.g. reduction of cardiac output or of systemic vascular resistance, is unknown. Our aim was to analyze and compare the regional and systemic hemodynamic changes in response to anandamide, its metabolically stable analog R-methanandamide (Abadji et al., 1994), and HU-210, and the sensitivity of these changes to inhibition by SR141716A in urethane-anesthetized rats. The results indicate that cannabinoids differently decrease cardiac output and total peripheral resistance, and that all three cannabinoids are strong vasodilators in the heart and the brain via the activation of vascular cannabinoid CB1 receptors. Furthermore, anandamide and R-methanandamide cause mesenteric vasodilation that does not appear to involve activation of cannabinoid CB1 receptors.

Section snippets

Drugs, chemicals

The source of the cannabinoids used has been provided elsewhere Lake et al., 1997a, Járai et al., 1999. Hexamethonium, arginine vasopressin, and urethane were from Sigma (St. Louis, MO). 46Scandium and 57Cobalt-labeled microspheres (diameter 15.5±0.1 μm) were from NEN Dupont (Boston, MA).

Animals, surgery

A total of 80 male Sprague–Dawley rats (300–380 g) were used, according to procedures approved by the Institutional Animal Care and Use Committee. The rats were anesthetized with urethane, 0.7 g/kg i.v.+0.3

Basal cardiovascular parameters

Fig. 1 contains the basal cardiovascular parameters in urethane-anesthetized controls and rats treated with 3 mg/kg SR141716A i.v. 30 min prior to the measurements. Of note, basal mean blood pressure and heart rate were not different from values we reported in conscious, chronically cannulated rats (Lake et al., 1997b). The well-documented hypotension following intraperitoneal administration of urethane can be avoided by the treatment protocol described by Maggi and Meli (1986) and employed

Discussion

The profound and long lasting hypotension elicited via activation of peripheral cannabinoid CB1 receptors is unsurpassed by hypotension triggered through any other mechanism (Lake et al., 1997a). Known ligands for these receptors include not only plant-derived and synthetic cannabinoids, but also endogenous substances such as anandamide (Devane et al., 1992). Both anandamide and the synthetic agonist HU-210 elicit hypotension, and although anandamide is less efficacious and its effect is much

Acknowledgements

This work was supported by grants R01-HL59257 and R01-HL49938 from the National Institutes of Health (to G.K.), a fellowship (to J.A.W.) from the Deutsche Forschungsgemeinschaft, a fellowship (to Z.J.) from Sanofi Recherche (Montpellier, France), and a Martin Rodbell Visiting Scientist award sponsored by Philip Morris (to S.B.). We thank Dr. Jerry Hirsch for help with the gamma spectrometry.

References (43)

  • Y. Chen et al.

    Human brain capillary endothelium: 2-arachidonyl-glycerol interacts with endothelin-1

    Circ. Res.

    (2000)
  • A.A. Coutts et al.

    Inhibition by cannabinoid receptor agonists of acetylcholine release from the guinea-pig myenteric plexus

    Br. J. Pharmacol.

    (1997)
  • D.G. Deutsch et al.

    Production and physiological actions of anandamide in the vasculature of the rat kidney

    J. Clin. Invest.

    (1997)
  • W.A. Devane et al.

    Isolation and structure of a brain constituent that binds to the cannabinoid receptor

    Science

    (1992)
  • E.F. Ellis et al.

    Anandamide and Δ9-THC-dilation of cerebral arterioles is blocked by indomethacin

    Am. J. Physiol.

    (1995)
  • S. Galiegue et al.

    Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations

    Eur. J. Biochem.

    (1995)
  • N. Garcia et al.

    Systemic and portal hemodynamic effects of anandamide

    Am. J. Physiol.

    (2001)
  • D. Gebremedhin et al.

    Cannabinoid CB1 receptor of cat cerebral arterial muscle functions to inhibit L-type Ca2+ channel current

    Am. J. Physiol.

    (1999)
  • A. Gulati et al.

    Cardiovascular effects of centrally administered endothelin-1 and its relationship to changes in cerebral blood flow

    Life Sci.

    (1996)
  • M.A. Huestis et al.

    Blockade of effects of smoked marijuana by the CB1-selective cannabinoid receptor antagonist SR141716A

    Arch. Gen. Psychiatry

    (2001)
  • E.J.N. Ishac et al.

    Inhibition of exocytotic noradrenaline release by presynaptic cannabinoid CB1 receptors on peripheral sympathetic nerves

    Br. J. Pharmacol.

    (1996)
  • Cited by (164)

    • Cardiac death with THC found in blood: A study of 33 cases

      2018, Toxicologie Analytique et Clinique
    View all citing articles on Scopus
    1

    Current address: Department of Medicine, University of Würzburg, Würzburg, Germany.

    2

    Current address: St. Emery Municipal Hospital, Budapest, Hungary.

    3

    Current address: National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA.

    View full text