Elsevier

European Journal of Pharmacology

Volume 722, 5 January 2014, Pages 134-146
European Journal of Pharmacology

Review
Regulation of nausea and vomiting by cannabinoids and the endocannabinoid system

https://doi.org/10.1016/j.ejphar.2013.09.068Get rights and content

Abstract

Nausea and vomiting (emesis) are important elements in defensive or protective responses that animals use to avoid ingestion or digestion of potentially harmful substances. However, these neurally-mediated responses are at times manifested as symptoms of disease and they are frequently observed as side-effects of a variety of medications, notably those used to treat cancer. Cannabis has long been known to limit or prevent nausea and vomiting from a variety of causes. This has led to extensive investigations that have revealed an important role for cannabinoids and their receptors in the regulation of nausea and emesis. With the discovery of the endocannabinoid system, novel ways to regulate both nausea and vomiting have been discovered that involve the production of endogenous cannabinoids acting centrally. Here we review recent progress in understanding the regulation of nausea and vomiting by cannabinoids and the endocannabinoid system, and we discuss the potential to utilize the endocannabinoid system in the treatment of these frequently debilitating conditions.

Introduction

Reflex mechanisms that serve to protect a host from injury and disability represent important and frequently well-conserved adaptations to a hostile external environment. Rarely do these adaptations, such as blinking or sneezing, become “hijacked” by physiological or pathophysiological processes in the body, not involving the organ they evolved to protect. Unfortunately, that is not the case for nausea and vomiting. Nausea is an aversive experience that often precedes emesis (vomiting), but is distinct from it (Borison and Wang, 1953, Carpenter, 1990, Horn, 2008, Andrews and Horn, 2006, Stern et al., 2011). Retching and vomiting lead to the forceful expulsion of gastric and/or upper intestinal contents, the primary function of which is to remove ingested materials or food that may be contaminated or potentially harmful. Nausea associated with emesis serves as an unconditioned stimulus for learning and memory; food that becomes associated with nausea and vomiting will be avoided in future encounters (Borison and Wang, 1953, Carpenter, 1990, Horn, 2008, Andrews and Horn, 2006, Stern et al., 2011).

In the natural environment, as a protective reflex, nausea and vomiting are very important adaptations found in most vertebrate species (Borison et al., 1981). However, possibly because of its importance, the sensitivity of this reflex is very low, making it easily activated. In various disease states, e.g. diabetes and labyrinthitis (Koch, 1999, Schmäl, 2013), the inappropriate activation of this reflex leads to severe and debilitating symptoms. Many central nervous system conditions, including elevated intracranial pressure, migraine headache and concussion also cause nausea and vomiting (Edvinsson et al., 2012, Mott et al., 2012, Stern et al., 2011). Nausea and vomiting are frequent, unwanted, side-effects of a range of medications used to treat a variety of conditions, notably cancer chemotherapeutic agents (Hesketh, 2005, Rojas and Slusher, 2012). Pregnancy-induced nausea and vomiting are reportedly adaptive mechanisms, but hyperemesis gravidarum can severely compromise both the health of the mother and the developing fetus (Patil et al., 2012, Sanu and Lamont, 2011, Sherman and Flaxman, 2002). Finally, motion sickness, which results from a sensory conflict between visual and vestibular stimuli, can be of immense discomfort, and severely limit certain activities (Schmäl, 2013, Yates et al., 1998). Nausea and vomiting are significant in our society and understanding them represents both an important goal and a major challenge; the former because of the substantial health implications, but the latter because it is hard to judge if an experimental animal is nauseated and commonly used laboratory animals are some of the few species that do not vomit! Nevertheless, significant progress has been made in our understanding of the processes of nausea and vomiting, which has led to new and improved pharmacological treatments for these disorders in the last 20–30 years, as described in many of the accompanying articles in this volume and previous reviews (Rojas and Slusher, 2012, Sanger and Andrews, 2006, Schmäl, 2013).

One of the oldest pharmacological remedies for nausea and vomiting is the plant cannabis (Kalant, 2001). In clinical trials, cannabis-based medicines have been found to be effective anti-emetics and even surpass some modern treatments in their potential to alleviate nausea (Cotter, 2009, Tramèr et al., 2001). However, it was not until the early 1990s that the mechanism of action of cannabis was established following the cloning of the “cannabinoid” (CB) receptors (Howlett et al., 2002, Pertwee et al., 2010). The significance of this discovery was enhanced when it was realized that these receptors were part of an endogenous cannabinoid (endocannabinoid) system in the brain and elsewhere in the body (Di Marzo and De Petrocellis, 2012, Izzo and Sharkey, 2010, Mechoulam and Parker, 2013, Piomelli, 2003). The endocannabinoid system serves to modulate the expression of nausea and vomiting when activated by central or peripheral emetic stimuli (Darmani and Chebolu, 2013, Parker et al., 2011).

In this article we will outline the endocannabinoid system and then describe what is known about this system in relation to the neural circuits of nausea and vomiting. We will describe recent findings on the anti-emetic effects of cannabinoids and show how manipulation of elements of the endocannabinoid system can modify the expression of emesis. We will discuss at some length the evidence that cannabinoids and the endocannabinoid system can regulate nausea, because this is an area that has been not been considered so fully in the past. We will then briefly describe the paradoxical effect of chronic exposure to high doses of cannabis that in some people causes a cyclic vomiting syndrome. Finally, we will conclude with some future directions for this research by identifying gaps in our knowledge of the regulation of nausea and vomiting by cannabinoids and the endocannabinoid system.

Section snippets

The endocannabinoid system

The isolation of Δ9-tetrahydrocannabinol (Δ9-THC) as the major psychoactive ingredient in cannabis was an important milestone in neuropharmacology (Howlett et al., 2002, Pertwee et al., 2010). This discovery provided the impetus for extensive investigations that led to an understanding of many of the central and peripheral sites of action of cannabis and ultimately to the cloning of the two G-protein coupled cannabinoid receptors; CB1 and CB2. CB1 receptors are distributed throughout the

The endocannabinoid system at sites in the brain and gastrointestinal tract involved in nausea and vomiting

The key components of the brain-gut circuitry mediating emesis have been well described (Andrews and Horn, 2006, Hornby, 2001). As outlined above, emesis can be initiated peripherally or centrally. However, most commonly, emesis is evoked from the gastrointestinal tract by ingestion of toxins, including bacteria or bacterial products, or food that is not tolerated. It may also be caused by drugs such as the cancer chemotherapeutic agent cisplatin and radiation. In most of these examples, the

Anti-emetic effects of cannabinoids and endocannabinoids

Cannabis is a well-known anti-emetic whose actions have been extensively reviewed (Cotter, 2009, Darmani and Chebolu, 2013, Izzo and Sharkey, 2010, Parker et al., 2011, Tramèr et al., 2001). Following the isolation of Δ9-THC, the mechanism and site of action of cannabinoids were established. In humans and animal models, plant-derived cannabinoids, synthetic cannabinoids and endocannabinoids inhibit emesis evoked peripherally or centrally with drugs or natural stimuli. Cannabinoids block both

Cannabinoids and endocannabinoids in the control of nausea in humans

There is clearly a need of treatments for acute, delayed and anticipatory nausea in chemotherapy treatment (e.g., Poli-Bigelli et al., 2003). One of the first recognized medicinal benefits of cannabis was for the treatment of nausea (Iversen, 2008). The most investigated compound has been Δ9-THC (see Cotter, 2009, Tramèr et al., 2001 for reviews); however, other nonpsychoactive compounds in the cannabis plant have recently been reported to also have benefits in preclinical models of nausea and

Cannabinoid and endocannabinoid regulation of nausea in animal models

Animal models of vomiting have been valuable in elucidating the neural mechanisms of the emetic reflex (Hornby, 2001); however, the central mechanisms regulating nausea are still not well understood (Andrews and Horn, 2006). Considerably greater progress has been made toward the control of vomiting than the control of nausea. One reason is that nausea is much more difficult to quantify than is vomiting, and therefore, preclinical model development has been challenging. Although vomiting is a

Contextually-elicited conditioned gaping reactions: a model of anticipatory nausea

Rats not only display conditioned gaping reactions when re-exposed to a flavor previously paired with a nausea-inducing drug, but they also display conditioned gaping reactions when re-exposed to a context previously paired with a nausea-inducing drug (Chan et al., 2009, Limebeer et al., 2008, Rock et al., 2008;). As well, the house musk shrew also displays conditioned retching when re-exposed to a context previously paired with toxin-induced vomiting (Parker and Kemp, 2001, Parker et al., 2006

Cannabis and hyperemesis: the paradoxical effect of chronic exposure to cannabis

Heavy chronic cannabis use in some people, frequently young ones, leads to a constellation of symptoms that include abdominal pain, recurrent nausea and intractable cyclic vomiting (Galli et al., 2011, Nicolson et al., 2012, Simonetto et al., 2012). This syndrome was first reported about 10 years ago (Allen et al., 2004). These symptoms are, of course, exactly the opposite of what has been outlined above and hence represent a paradoxical effect of cannabis. Relief from these symptoms can be

Future directions in using the endocannabinoid system in the treatment of nausea and vomiting

As can be appreciated from the discussion in the previous sections, we believe that the endocannabinoid system has the potential to be used for the treatment of nausea and likely as an adjunct therapy for the treatment of emesis, particularly delayed emesis, where current therapies are limited in their degree of efficacy. There are, however, many gaps in our knowledge, most of which were highlighted above. One of the biggest limitations is the very widespread nature of the CB1 receptor and the

Acknowledgements

Original work in the authors' laboratories is supported by the Canadian Institutes of Health Research (KAS), the Natural Sciences and Engineering Research Council of Canada (LAP) and NIH grants-NIDA 12605 and CA115331 (ND). KAS is the recipient of a Killam Annual Professorship and holds the Crohn's & Colitis Foundation of Canada Chair in Inflammatory Bowel Disease Research at the University of Calgary. LAP is the recipient of a Tier 1 Canada Research Chair in behavioural neuroscience at

References (199)

  • H. Catenoix et al.

    The role of the anterior insular cortex in ictal vomiting: a stereotactic electroencephalography study

    Epilepsy Behav.

    (2008)
  • M.Y. Chan et al.

    Lipopolysaccharide (LPS) blocks the acquisition of LiCl-induced gaping in a rodent model of anticipatory nausea

    Neurosci. Lett.

    (2009)
  • K.M. Chu et al.

    Olvanil: a non-pungent TRPV1 activator has anti-emetic properties in the ferret

    Neuropharmacology

    (2010)
  • D. Cunningham et al.

    A randomized trial of oral nabilone and prochlorperazine compared to intravenous metoclopramide and dexamethasone in the treatment of nausea and vomiting induced by chemotherapy regimens containing cisplatin or cisplatin analogues

    Eur. J. Cancer Clin. Oncol.

    (1988)
  • N.A. Darmani

    Delta(9)-tetrahydrocannabinol and synthetic cannabinoids prevent emesis produced by the cannabinoid CB(1) receptor antagonist/inverse agonist SR 141716A

    Neuropsychopharmacology

    (2001)
  • N.A. Darmani

    Delta-9-tetrahydrocannabinol differentially suppresses cisplatin-induced emesis and indices of motor function via cannabinoid CB(1) receptors in the least shrew

    Pharmacol. Biochem. Behav

    (2001)
  • N.A. Darmani

    The cannabinoid CB1 receptor antagonist SR 141716A reverses the antiemetic and motor depressant actions of WIN 55, 212-2

    Eur. J. Pharmacol

    (2001)
  • N.A. Darmani et al.

    Delta-9-tetrahydrocannabinol differentially suppresses emesis versus enhanced locomotor activity produced by chemically diverse dopamine D2/D3 receptor agonists in the least shrew (Cryptotis parva)

    Pharmacol. Biochem. Behav

    (2005)
  • N.A. Darmani et al.

    Behaviorally active doses of the CB1 receptor antagonist SR 141716A increase brain serotonin and dopamine levels and turnover

    Pharmacol. Biochem. Behav.

    (2003)
  • N.A. Darmani et al.

    Cisplatin increases brain 2-arachidonoylglycerol (2-AG) and concomitantly reduces intestinal 2-AG and anandamide levels in the least shrew

    Neuropharmacology

    (2005)
  • N.A. Darmani et al.

    Antiemetic and motor-depressive actions of CP55,940: cannabinoid CB1 receptor characterization, distribution, and G-protein activation

    Eur. J. Pharmacol.

    (2003)
  • J. De Vry et al.

    Behavioral mechanisms underlying inhibition of food-maintained responding by the cannabinoid receptor antagonist/inverse agonist SR141716A

    Eur. J. Pharmacol.

    (2004)
  • L. Edvinsson et al.

    Basic mechanisms of migraine and its acute treatment

    Pharmacol. Ther.

    (2012)
  • M. Fiol et al.

    Ictus emeticus and the insular cortex

    Epilepsy Res.

    (1988)
  • B. Forget et al.

    Granular insular cortex inactivation as a novel therapeutic strategy for nicotine addiction

    Biol. Psychiatry

    (2010)
  • J.P. Gong et al.

    Cannabinoid CB2 receptors: immunohistochemical localization in rat brain

    Brain Res.

    (2006)
  • H.C. Grill et al.

    The taste reactivity test. I: Mimetic responses to gustatory stimuli in neurologically normal rats

    Brain Res.

    (1978)
  • P. Gyombolai et al.

    Regulation of endocannabinoid release by G proteins: a paracrine mechanism of G protein-coupled receptor action

    Mol. Cell Endocrinol.

    (2012)
  • K. Hillsley et al.

    Serotonin and cholecystokinin activate different populations of rat mesenteric vagal afferents

    Neurosci. Lett.

    (1998)
  • C.C. Horn

    Why is the neurobiology of nausea and vomiting so important?

    Appetite

    (2008)
  • C.C. Horn et al.

    Differential effects on gastrointestinal and hepatic vagal afferent fibers in the rat by the anti-cancer agent cisplatin

    Auton. Neurosci

    (2004)
  • A.A. Izzo et al.

    Cannabinoids and the gut: new developments and emerging concepts

    Pharmacol. Ther.

    (2010)
  • J. Jelsing et al.

    The effect of leptin receptor deficiency and fasting on cannabinoid receptor 1 mRNA expression in the rat hypothalamus, brainstem and nodose ganglion

    Neurosci. Lett

    (2009)
  • M. Kaczocha et al.

    Fatty acid-binding proteins transport N-acylethanolamines to nuclear receptors and are targets of endocannabinoid transport inhibitors

    J. Biol. Chem.

    (2012)
  • M.R. Landauer et al.

    Attenuation of cyclophosphamide-induced taste aversions in mice by prochlorperazeine, delta 9-tetraydrocannabinol, nbilone and levonantradol, Pharmacol. Biochem. Behav.

    (1985)
  • S. Ahmedzai et al.

    Anti-emetic efficacy and toxicity of nabilone, a synthetic cannabinoid, in lung cancer chemotherapy

    Br. J. Can

    (1983)
  • J.H. Allen et al.

    Cannabinoid hyperemesis: cyclical hyperemesis in association with chronic cannabis abuse

    Gut

    (2004)
  • J.H. Baek et al.

    Cannabinoid CB2 receptor expression in the rat brainstem cochlear and vestibular nuclei

    Acta Otolaryngol.

    (2008)
  • I. Billig et al.

    Plasma hormone levels and central c-Fos expression in ferrets after systemic administration of cholecystokinin

    Am. J. Physiol. Regul. Integr. Comp. Physiol

    (2001)
  • J.L. Blankman et al.

    Chemical probes of endocannabinoid metabolism

    Pharmacol. Rev.

    (2013)
  • F.M. Boissonade et al.

    Fos expression in ferret dorsal vagal complex after peripheral emetic stimuli

    Am. J. Physiol.

    (1994)
  • D. Bolognini et al.

    Cannabidiolic acid prevents vomiting in Suncus murinus and nausea-induced behaviour in rats by enhancing 5-HT1A receptor activation

    Br. J. Pharmacol.

    (2013)
  • H.L. Borison et al.

    Phylogenic and neurologic aspects of the vomiting process

    J. Clin. Pharmacol.

    (1981)
  • H.L. Borison et al.

    Physiology and pharmacology of vomiting

    Pharmacol. Rev.

    (1953)
  • A. Brusco et al.

    Ultrastructural localization of neuronal brain CB2 cannabinoid receptors

    Ann. N.Y. Acad. Sci

    (2008)
  • G. Burdyga et al.

    Expression of cannabinoid CB1 receptors by vagal afferent neurons is inhibited by cholecystokinin

    J. Neurosci.

    (2004)
  • G. Burdyga et al.

    Expression of cannabinoid CB1 receptors by vagal afferent neurons: kinetics and role in influencing neurochemical phenotype

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2010)
  • M.P. Carey et al.

    Delta-9-tetrahydrocannabinol in cancer chemotherapy: research problems and issues

    Ann. Intern. Med.

    (1983)
  • D.O. Carpenter

    Neural mechanisms of emesis

    Can. J. Physiol. Pharmacol.

    (1990)
  • D.F. Cechetto et al.

    Evidence for a viscerotopic sensory representation in the cortex and thalamus in the rat

    J. Comp. Neurol.

    (1987)
  • Cited by (156)

    View all citing articles on Scopus
    View full text