Endocanabinoides en Manejo Del Dolor

Embed Size (px)

Citation preview

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    1/17

    R E V I E W

    A R T I C L E

    The role of endocannabinoids in pain

    modulation

    Panagiotis Zogopoulos, Ioanna Vasileiou, Efstratios Patsouris, Stamatios

    E. Theocharis*

    First Department of Pathology, Medical School, University of Athens, 75 Mikras Asias Street, Goudi, 11527 Athens,Greece

    Keywords

    2-arachidonoylglycerol,

    analgesia,

    anandamide,

    N-arachidonoylethanol-

    amine,

    antinociception,

    CB receptors,

    endocannabinoids

    Received 28 November 2011;

    revised 3 September 2012;

    accepted 21 September 2012

    *Correspondence and reprints:

    [email protected]

    A B S T R A C T

    The endocannabinoid system (ES) is comprised of cannabinoid (CB) receptors, their

    endogenous ligands (endocannabinoids), and proteins responsible for their metabo-

    lism. Endocannabinoids serve as retrograde signaling messengers in GABAergic and

    glutamatergic synapses, as well as modulators of postsynaptic transmission, that

    interact with other neurotransmitters. Physiological stimuli and pathological condi-

    tions lead to differential increases in brain endocannabinoids that regulate distinctbiological functions. Furthermore, endocannabinoids modulate neuronal, glial, and

    endothelial cell function and exert neuromodulatory, anti-excitotoxic, anti-inflam-

    matory, and vasodilatory effects. Analgesia is one of the principal therapeutic tar-

    gets of cannabinoids. Cannabinoid analgesia is based on the suppression of spinal

    and thalamic nociceptive neurons, but peripheral sites of action have also been

    identified. The chronic pain that occasionally follows peripheral nerve injury differs

    fundamentally from inflammatory pain and is an area of considerable unmet thera-

    peutic need. Over the last years, considerable progress has been made in under-

    standing the role of the ES in the modulation of pain. Endocannabinoids have been

    shown to behave as analgesics in models of both acute nociception and clinical pain

    such as inflammation and painful neuropathy. The framework for such analgesic

    effects exists in the CB receptors, which are found in areas of the nervous systemimportant for pain processing and in immune cells that regulate the neuro-immune

    interactions that mediate the inflammatory hyperalgesia. The purpose of this review

    is to present the available research and clinical data, up to date, regarding the ES

    and its role in pain modulation, as well as its possible therapeutic perspectives.

    I N T R O D U C T I O N

    Cannabinoids, first discovered in the 1940s, are a class

    of chemical compounds that include the phytocannabi-

    noids (oxygen-containing C21 aromatic hydrocarboncompounds found in the cannabis plant) and chemical

    compounds that mimic the actions of phytocannabi-

    noids or have a similar chemical structure [1,2]. Syn-

    thetic cannabinoids encompass a variety of distinct

    chemical classes: the classical cannabinoids structurally

    related to D9-tetrahydrocannabinol (D9-THC), the non-

    classical cannabinoids including the aminoalkylindoles,

    1,5-diarylpyrazoles, quinolines, and arylsulfonamides,

    as well as eicosanoids related to the endocannabinoids.

    D9-THC (the primary psychoactive component of the

    cannabis plant), cannabidiol (CBD), and cannabinol

    (CBN) are the most prevalent natural cannabinoids

    and have been mostly studied. Cannabinoids can beadministered by smoking, vaporizing, oral ingestion,

    transdermal patch, intravenous injection, sublingual

    absorption, or rectal suppository. Most cannabinoids

    are metabolized in the liver, especially by cytochrome

    P450 (CYP) mixed-function oxidases, mainly CYP 2C9

    [1,2].

    Over the last decades, several studies have reported the

    existence of an endogenous lipid signaling system with

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    64 Fundamental & Clinical Pharmacology 27 (2013) 6480

    doi: 10.1111/fcp.12008

    Funda

    mental&ClinicalPharmacology

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    2/17

    cannabimimetic actions, referred to as endocannabinoid

    system (ES). Recent pharmacological advances have

    enabled the study of the physiological roles played by the

    ES, including its role in analgesia. The purpose of this

    review is to present current knowledge about the role of

    the ES in pain modulation and consequently the future

    prospects of developing potential analgesic agents.

    T H E E N D O C A N N A B I N O I D S Y S T E M

    The ES is involved in a variety of physiological pro-

    cesses including nociception (pain sensation), appetite,

    lipid metabolism, gastrointestinal motility, cardiovascu-

    lar modulation, motor activity, mood, and memory

    [35]. It is comprised of cannabinoid receptor type-1

    (CB1) and type-2 (CB2), which are seven-transmem-

    brane, G-protein coupled receptors negatively coupled

    to adenylyl cyclase and positively coupled to mitogen-activated protein kinase (MAPK)[6,7]. It also includes

    their endogenous lipid-based ligands, the endocannabi-

    noids, of which anandamide (N-arachidonoylethanol-

    amine, AEA) and 2-arachidonoylglycerol (2-AG) are

    mostly studied [8,9], and the proteins that are respon-

    sible for their biosynthesis, transport, and degradation

    [10].

    Cannabinoid receptors

    CB1 receptors are most abundantly expressed in the

    mammalian brain and also in peripheral tissues

    [11,12]. They are highly expressed in regions of thebrain, such as the cortex, limbic system, hippocampus,

    cerebellum, brainstem, and several nuclei in the basal

    ganglia that are associated with emotion, cognition,

    memory, motor and executive function [13]. More

    specifically, they are expressed in brain areas involved

    in nociceptive transmission and processing, including

    the periaqueductal gray (PAG), anterior cingulate cor-

    tex (ACC), and thalamus in addition to the dorsal horn

    of the spinal cord and dorsal root ganglion (DRG)[14

    16]. CB1 receptors are found primarily at the terminals

    and also at the axons, at cell bodies, and at dendrites

    of central and peripheral neurons, where they typicallymediate the inhibition of amino acid and monoamine

    neurotransmitter release, as occurs with the inhibitory

    neurotransmitter gamma-aminobutyric acid (GABA)

    [17,18].

    CB2 receptors in the brain are expressed primarily in

    the perivascular microglial cells [19,20] and astrocytes

    [21,22], where they modulate the immune responses

    [2325]. They are also expressed in cerebromicrovas-

    cular endothelial cells [26] and in central (brainstem)

    and peripheral neurons [2729]. Furthermore, CB2

    receptors are found on the cells of the immune system

    throughout the whole body [i.e., B lymphocytes, mac-

    rophages, natural killer (NK) cells] [30,31], and they

    are also expressed in the myocardium, the human cor-

    onary endothelial cells, the smooth muscle cells, and

    the liver [11,12,32].

    Ligands

    Endocannabinoids are endogenous metabolites of

    eicosanoid fatty acids. They are lipid signaling

    mediators of the same CB receptors that mediate the

    effects of D9-THC [33,34]. They are derivatives of ara-

    chidonic acid conjugated with either ethanolamine or

    glycerol. Apart from anandamide (AEA) and 2-arachi-

    donoylglycerol (2-AG), which are the best described

    endocannabinoids, N-arachidonoyldopamine (NADA),

    2-arachidonoylglyceryl ether (2-AGE, noladin ether), and

    O-arachidonoylethanolamine (OAE, virodhamine) are also

    included [8,3537] (Figure 1).

    AEA, the first endocannabinoid to be identified [8],

    appears to be a partial agonist for CB1 receptor [38]

    with modest affinity [Ki = 61 nM (rat) and 240 nM

    (human)] and a relatively weak CB2 receptor ligand

    (Ki = 4401930 nM for rodent and human CB2 recep-

    tors) with low overall efficacy. More recent data sug-

    gest that it might also interact directly with other

    molecular targets, including non-CB1, non-CB2 G-pro-

    tein coupled receptors, gap junctions, and various ionchannels [3942].

    2-AG, the second identified CB receptor ligand

    [43,44], is the most abundant endocannabinoid in the

    central nervous system (CNS) and a full agonist for

    both CB1 and CB2 receptors [34,38,45] with lower

    affinity (Ki = 472 and 1400 nM, respectively) and

    greater efficacy relatively to AEA [46,47].

    NADA was discovered in 2000 and preferentially

    binds to the CB1 receptor [48]. The distribution pattern

    of endogenous NADA in various brain areas differs

    from that of AEA, with the highest levels found in the

    striatum and hippocampus [35], while it, also, exists inthe DRG at low levels [49]. 2-AGE, isolated in 2001

    from porcine brain, binds primarily to the CB1 receptor

    (Ki = 21.2 nM) and only weakly to the CB2 receptor

    [50].

    OAE, discovered in 2002, is a compound similar to

    AEA in being formed from arachidonic acid and

    ethanolamine, but OAE contains an ester linkage rather

    than AEAs amide linkage. Although it is a full agonist

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 65

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    3/17

    for CB2 receptor and a partial agonist for CB1 receptor,

    it behaves as a CB1 antagonist in vivo. In rats, OAE was

    found to be present at comparable or slightly lower con-

    centrations than AEA in the brain, but 2- to 9-fold

    higher concentrations peripherally [36].

    Biosynthesis

    Unlike traditional neurotransmitters such as acetylcho-

    line and dopamine, endogenous cannabinoids are notstored in vesicles after synthesis, but are synthesized on

    demand from phospholipid precursors residing in the

    cell membrane in response to a rise in intracellular cal-

    cium levels [4,37,51]. However, some evidence sug-

    gests that a pool of synthesized endocannabinoids

    (namely, 2-AG) may exist without the requirement of

    on-demand synthesis [52].

    Endocannabinoid levels are elevated in brain paren-

    chyma as part of internal repair responses to traumatic

    brain and spinal cord injuries [53,54]. Enzymatic syn-

    thesis of both AEA and 2-AG draws upon pools of mem-

    brane phospholipids such as phosphatidylethanolamine(PE), phosphatidylcholine (PC), and phosphatidylinositol

    4,5-bisphosphate [55,56]. It is worth mentioning that

    hormones of the gonadal axis, such as estradiol, regu-

    late the expression of the enzymes involved in the syn-

    thesis and metabolism of endocannabinoids in different

    peripheral tissues [57].

    AEA and its precursor, N-arachidonoylphosphatidyl-

    ethanolamine (NAPE), are normally expressed at low

    concentrations in the rat brain, but increase in a cal-

    cium-dependent manner postmortem [58] and after

    severe neuronal injury [38,59]. A two-step biosynthesis

    pathway of AEA has been suggested, involving the

    sequential action of a calcium-dependent transacylase

    (Ca-TA, N-acyltransferase) and a NAPE-selective phos-

    pholipase D (NAPE-PLD). N-acyltransferase transfers a

    fatty acyl chain from a membrane phospholipid mole-

    cule onto the primary amine of membrane, phosphati-dyl-ethanolamine, to generate NAPE, and NAPE-PLD

    hydrolyzes NAPE to N-acylethanolamines (NAEs) such

    as AEA [6062] (Figure 2). AEA can also be formed by

    the stimulation of dopamine D2 receptors in a G-pro-

    tein-coupled process [63].

    2-AG is synthesized from diacylglycerol (DAG) by

    diacylglycerol lipase (DAGL). DAGL is found at

    increased levels after neuronal injury [28,64] and

    catalyzes the hydrolysis of DAG, releasing a free fatty

    acid and monoacylglycerol, which is then converted to

    2-AG [13,65] (Figure 3). AEA levels increase is fol-

    lowed by 2-AG upregulation [54]. The accumulation of2-AG at the site of injury has been described to present

    a peak 4 h after injury and sustained up to at least

    24 h postinjury [66].

    Transport and metabolism

    Endocannabinoids serve as retrograde signaling

    messengers in GABAergic and glutamatergic synapses,

    as well as modulators of postsynaptic transmission,

    Figure 1 Biosynthesis, transport anddegradation of endocannabinoids.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    66 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    4/17

    interacting with other neurotransmitters, including

    norepinephrine and dopamine [67]. 2-AG and AEA are

    removed from extracellular space and transported into

    cells through a diffusion-facilitated transporter system

    or uptaken via membrane-associated carrier and simple

    diffusion [68]. Thus, endocannabinoid signaling func-

    tions are efficiently terminated by cellular uptake and

    rapid, enzyme-catalyzed hydrolytic inactivation. Fatty

    acid amide hydrolase (FAAH) and monoacylglycerol

    lipase (MAGL) are the primary catabolic enzymes of

    AEA and 2-AG, respectively [6972]. A partly cytosolic

    variant of FAAH, termed FAAH-like anandamide trans-

    porter, has been shown to bind AEA with low micro-

    molar affinity and facilitate its translocation into cells

    [73].

    FAAH is highly expressed by neurons in the mamma-

    lian brain as an integral membrane protein and is upreg-

    ulated after neuronal injury [28,64]. It is localized in

    endoplasmic reticulum of hippocampus, neocortex, and

    cerebellum [55,74] and catalyzes the hydrolysis of sev-

    eral endogenous, biologically active lipids, including

    AEA, oleoyl ethanolamide (OEA), and palmitoyl ethan-

    olamide (PEA)[75]. AEA has a short half-life, as it is rap-

    idly hydrolyzed by FAAH, and its resting concentrations

    in the CNS are very low. FAAH degrades AEA into

    arachidonic acid and ethanolamine, after its release from

    neurons [72,76]. Enhanced cannabinoid signaling can

    be achieved by preventing AEA hydrolysis/inactivation

    by FAAH. A number of FAAH inhibitors exist that can

    increase the level of AEA in the brain of experimental

    animals [55].

    On the other hand, FAAH has been also demon-

    strated to catalyze AEA synthesis from arachidonic acid

    and ethanolamine, with a reported Km for ethanol-

    amine of at least 36 mM [77]. Several studies have

    shown that recombinant FAAH protein acts as an AEA

    synthetase if the concentration of ethanolamine is very

    high (100 mM) and is capable of catalyzing the reverse

    of the hydrolase reaction [78,79].

    2-AG is hydrolyzed into arachidonic acid and

    glycerol by either FAAH or preferably by MAGL

    [51,75,80]. 2-AG has been shown to be a substrate for

    FAAH both in vitro [69,81] and in vivo [82].

    Recent evidence reveals that endogenous cannabi-

    noids are also substrates for cyclo-oxygenase (COX) and

    can be selectively oxygenated by a COX-2 pathway to

    form new classes of prostaglandins (prostaglandin glyc-

    erol esters and prostaglandin ethanolamides) [8385].

    Therefore, this is another pathway in degrading

    endocannabinoids in addition to their well-known

    Figure 2 The AEA biosynthesis

    pathway.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 67

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    5/17

    hydrolysis pathways. Metabolites of AEA and 2-AG,

    derived from COX-2, have biological activity, including

    the activation of protein kinase C (PKC) and also have

    effects on the contractility of smooth muscle prepara-

    tions [86,87]. Prostanoids derived from both AEA and

    2-AG are significantly more stable metabolically than

    free-acid prostaglandins, suggesting that COX-2 action

    on endocannabinoids may provide oxygenated lipids

    with sufficiently long half-lives to act as systemic

    mediators or prodrugs [88,89].

    Signaling pathways and molecular targets

    The neuromodulatory and anti-inflammatory effects of

    cannabinoids are mediated by induction of apoptosis,

    inhibition of cell proliferation, suppression of cytokine

    production, and induction of T-regulatory cells. One

    major mechanism of immunosuppression by cannabi-

    noids is the induction of cell death or apoptosis in

    immune cell populations, thus playing a protective role

    in autoimmune conditions [37,90].

    In vitro and in vivo studies have shown that cannabi-

    noids can act on glia and neurons to inhibit the release

    of pro-inflammatory cytokines [tumor necrosis factor-a

    (TNF-a), interleukin (IL)-6, and IL-1b] and enhance

    the release of anti-inflammatory factors such as the

    cytokines IL-4 and IL-10 [21,9193]. AEA, via the

    activation of CB1 receptors, enhances the synthesis of

    IL-6, which has both pro- and anti-inflammatory prop-

    erties, and reduces the synthesis of the proinflammato-

    ry cytokine TNF-a in Theilers virusinfected astrocytes

    [94].

    CB receptors initiate different signaling pathways

    including adenylyl cyclase and protein kinase A (PKA)

    inhibition and regulation of ionic channels. CB1 agon-

    ists reduce calcium influx by blocking the activity of

    voltage-dependent N-, P/Q-, and L-type calcium (Ca2+)

    channels [95,96]. This leads to reduced activity of neu-

    ronal nitric oxide synthase (nNOS) but also to the

    reduction of other potentially damaging reactive oxy-

    gen species [9799]. CB1 activation can also initiate

    the opening of inwardly rectifying K+ channels and the

    inhibition of adenylyl cyclase activity, resulting in a

    decrease in cytosolic cAMP [34,100,101]. In addition,

    regulation of neuronal gene expression by CB1 recep-

    tors depends on the recruitment of complex networks

    of intracellular protein kinases, such as the phosphati-

    Figure 3 The 2-AG biosynthesis

    pathway.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    68 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    6/17

    dylinositol 3-kinase/Akt, the extracellular signal-regu-

    lated kinase (ERK), and the focal adhesion kinase,

    which become activated when hippocampal brain tis-

    sue is treated with cannabinoid agonists according to

    the findings of experimental studies [102,103]. CB1

    receptors also modulate the generation of sphingolipid-

    derived signaling mediators and cell death pathways

    (e.g., caspase activation and the endoplasmic reticulum

    stress response) [104].

    AEA can inhibit a number of different ion channels

    [105], and it appears that there is a direct extracellular

    binding site for AEA on these channels. AEA has been

    demonstrated to activate the transient receptor poten-

    tial vanilloid 1 (TRPV1) both in vitro and in vivo

    [45,106,107] and to upregulate genes involved in pro-

    inflammatory microglial-related responses [108,109].

    TRPV1 receptors are nonselective ion channels whose

    location in sensory neurons allows them to gate

    responses to painful stimuli such as high temperature

    and low pH. Activation of TRPV1 leads to an increased

    influx of Ca2+ [110], glutamate release [111], and sub-

    stantial contribution to neuronal excitotoxicity that

    leads to apoptosis [108,112].

    CB2 receptors mediate anti-inflammatory actions of

    cannabinoids on astrocytes and microglia. In particu-

    lar, they decrease the activity of antigen-presenting

    cells and downregulate cytokine (IFN-c, TNF-a, and

    IL-6) production during inflammatory responses in in

    vivo and in vitro studies [113,114]. The anti-inflamma-

    tory effects of cannabinoids on glial cells involve theinhibition of nuclear factor jB (NF-jB)-induced tran-

    scription of proinflammatory chemokines and cytokines

    [115,116]. Moreover, CB2 receptors might control

    immune cell proliferation by coupling to ERK activa-

    tion (independent of cAMP) via regulation of mkp-1

    gene expression by histone H3 phosphorylation [117].

    AEA induces rapid phosphorylation of histone H3 on

    the mkp-1 gene and also induces mkp-1 expression in

    microglial cells of inflammatory brain lesions, which

    suppresses nitric oxide (NO) release and inflammatory

    damage in living brain tissue [118].

    Endocannabinoids mainly induce an inhibitory effecton both GABAergic and glutamatergic neurotransmis-

    sion and neurotransmitter release, although the results

    are somewhat variable [119121]. In some cases,

    cannabinoids diminish the effects of GABA, while in

    others they can augment the effects of GABA. The

    effect of activating a receptor depends on where it is

    expressed on the neuron: if CB receptors are

    presynaptic and inhibit the release of GABA, cannabi-

    noids would diminish GABA effects; the net effect

    would be stimulation. However, if CB receptors are

    postsynaptic and on the same cell as GABA receptors,

    they would probably mimic the effects of GABA; in

    that case, the net effect would be inhibition [122]. En-

    docannabinoids induce these effects via the phenome-

    non of depolarization-induced suppression of inhibition

    (DSI). DSI refers to endocannabinoid-induced suppres-

    sion of GABAergic synaptic transmission. In DSI,

    strong depolarization of a postsynaptic neuron induces

    a release of signal that acts on the presynaptic CB1

    receptor and transiently inhibits the release of GABA.

    Such retrograde signaling by endocannabinoid-medi-

    ated DSI occurs in the hippocampus but has also been

    shown outside the hippocampus at interneuron-princi-

    pal cell synapses [123]. Thereafter, a similar phenome-

    non has been demonstrated for glutamatergic synaptic

    transmission and has been designated depolarization-

    induced suppression of excitation (DSE) [124,125].

    Cannabinoids attenuate glutamate-induced injury by

    inhibiting glutamate release via presynaptic CB1 recep-

    tors coupled to G-proteins and N-type voltage-gated

    calcium channels [97,126]. 2-AG, but not AEA, is pos-

    sibly a signaling molecule in mediating CB1-dependent

    DSI or DSE [34,127]. Moreover, enzymes that synthe-

    size 2-AG are present in postsynaptic dendritic spines,

    providing direct evidence that 2-AG is synthesized in

    postsynaptic sites and acts on presynaptic CB1 recep-

    tors [128,129]. Thus, endocannabinoids (especially

    2-AG) are proposed to serve as retrograde messengersin modulating both GABAergic and glutamatergic

    synaptic transmission [120,121,130,131].

    E N D O C A N N A B I N O I D S A N D P A I N

    M O D U L A T I O N

    Analgesia is one of the principal therapeutic targets of

    cannabinoids. The chronic pain that occasionally fol-

    lows peripheral nerve injury differs fundamentally from

    inflammatory pain and is an area of considerable,

    unmet therapeutic need [64,132135]. Several investi-

    gations have gathered important experimental andclinical data about the analgesic properties of cannabi-

    noids and their endogenous counterparts.

    Experimental data

    Sites of analgesic action

    Cannabinoid administration has been found to suppress

    behavioral and neurophysiological responses to all

    types of nociceptive stimuli tested. It suppresses both

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 69

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    7/17

    wide dynamic range neurons (mediating response to

    touch and pain) and nociceptive-specific neurons that

    mediate response to pain only, but does not affect low-

    threshold mechanoreceptive neurons that mediate

    response to touch only [136]. Cannabinoids have been

    found in many studies to exert their analgesic effects

    by an action in the brain via descending modulation,

    by a direct spinal action, and/or by an action on the

    peripheral nerve [136140]. As highly lipophilic

    compounds, cannabinoids can readily penetrate the

    bloodbrain barrier and access the brain [23,24]. This

    is followed by the induction of antinociceptive effects

    through actions in the PAG and the rostral ventrolat-

    eral medulla (RVM), whose circuits inhibit spinal noci-

    ceptive neurotransmission [136140]. The ACC is part

    of the medial pain pathway and has been shown to be

    involved in the affective component of pain processing

    [141]. A recent research has shown that CB receptor

    mediated G-protein activity in the rostral anterior cin-

    gulate cortex (rACC) of mice is decreased after 10 days

    of chronic constriction injury (CCI), perhaps in an

    attempt to minimize the feeling of pain [142]. Experi-

    mental studies have demonstrated that microinjection

    of cannabinoids into sites such as the dorsolateral

    PAG, dorsal raphe nucleus, RVM, amygdala, lateral

    posterior and submedius regions of the thalamus, supe-

    rior colliculus, and noradrenergic A5 region produces

    antinociception [137140,143145]. Furthermore, sys-

    temically inactive doses of cannabinoids have been

    shown to attenuate carrageenan-evoked allodynia andhyperalgesia, when administered peripherally, and sup-

    press carrageenan-evoked Fos protein (a marker of

    neuronal activity) expression in the lumbar dorsal horn

    of the spinal cord in rats [146]. At the level of the

    spinal cord, CB2 receptors activation has analgesic

    effects in neuropathic rats [147,162]. Thus, both

    peripheral and spinal cord injections of cannabinoids

    have been shown to be antinociceptive.

    Pain models

    Animal studies have firmly established cannabinoid-

    induced analgesia in a wide array of pain models

    [143,144] (Table I). In models of acute or physiologi-

    cal pain, cannabinoids are highly effective against

    thermal [148], mechanical [149], and chemical pain

    [149,150], and typically, cannabinoids were compara-

    ble with opiates (both in potency and efficacy) in pro-

    ducing antinociception [148]. On the other hand, in

    models of tonic or chronic pain, both inflammatory

    [151] and neuropathic [152] cannabinoids have

    shown even greater potency and efficacy. D9-THC,

    which has approximately equal affinity for the CB1

    and CB2 receptors, appears to ease moderate pain andto be neuroprotective [2]. It is likely that high doses of

    D9-THC would be effective in pain management, but,

    unfortunately, these doses also produce undesirable

    CNS effects.

    Neuropathic pain is defined as pain arising as a

    direct consequence of a lesion or disease affecting the

    somatosensory system. It can be caused by several dis-

    orders, like nerve injury, diabetes, viral infection, and

    chemotherapic agents [134,152]. Both CB and TRPV1

    receptors are upregulated in the spinal cord and DRG

    of neuropathic rats [64,152]. Molecules that are inhibi-

    tors of endocannabinoid cellular re-uptake and are alsoagonists for TRPV1 receptors, such as AM404 and

    arvanil, are very effective against both thermic hyperal-

    gesia and mechanical allodynia in the CCI model of

    neuropathic pain [153,154]. The antinociceptive

    responses to D9-THC and other cannabinoids are

    Table I Antinociceptive effects of cannabinoids on various pain models

    Agent Animal subject Pain test Pain model Results References

    AEA Rats Thermal allodynia Inflammatory Reduced hyperalgesia Karbarz et al. (2009) [75]

    2-AG Mice CHI Acute/Inflammatory Inhibition of pro-inflammatory

    cytokines

    Panikashvili et al.

    (2006) [196]

    AEA/2-AG Rats CCI

    mechanical/thermal

    allodynia

    Neuropathic Anti-nociception Petrosino et al.,

    (2007) [157]

    URB597 (FAAH

    inhibitor)

    Mice CCI mechanical/cold

    allodynia

    Neuropathic Attenuation of allodynia Kinsey et al. (2009) [159]

    OL135 (FAAH

    inhibitor) + AEA/2-AG

    Mice MTI mec ha nic al al lodynia Acute Att enuat ion of a ll odynia Pal mer et a l. (2008) [195]

    D9-THC Rats Thermal allodynia Acute Antinociception Martin et al. (1999)

    [139,140]

    AEA, anandamide, N-arachidonoylethanolamine; CCI, chronic constriction injury, MTI, mild thermal injury; CHI, closed head injury.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    70 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    8/17

    absent or markedly attenuated in CB1 knockout

    mice [155]. Moreover, peripheral deletion of CB1 on

    nociceptors (with CB1 preserved in the CNS) can

    block the analgesic effects of locally and systemically

    administered cannabinoids [156]. In the spinal cord,

    the elevation of AEA levels appears to be an early

    observed already after 3 days and strong event

    accompanying CCI of the sciatic nerve, followed also

    by a significant elevation in 2-AG levels 7 days after

    the surgery [157]. This differential effect on the two

    major endocannabinoids might have a functional sig-

    nificance as 2-AG is able to activate both CB1 and CB2

    receptors, whereas AEA can only activate the former

    receptor type, but can instead gate TRPV1 channels.

    Therefore, it can be assumed that AEA levels are ele-

    vated at both 3 and 7 days from CCI as an adaptive

    response aimed at targeting first the CB1 receptor,

    which is already present in the spinal cord even prior

    to the development of pain following nerve constric-

    tion. The TRPV1 receptor is activated later, when the

    expression of this protein is strongly elevated and par-

    ticipates in thermal hyperalgesia [158]. Likewise, 2-AG

    levels might be elevated only 7 days after CCI to acti-

    vate CB2 receptors, which are also upregulated only

    following the full development of neuropathic pain (i.e.,

    starting 4 days from surgery). CB2 receptor stimulation

    has been found to decrease allodynia at the level of

    peripheral nociceptors, spinal nerves, and afferents, or

    supraspinally [64,157,159]. The levels of the two

    major endocannabinoids, AEA and 2-AG, increase fol-lowing CCI of the sciatic nerve in both the spinal cord

    and in some supraspinal areas involved in the descend-

    ing control of nociception and of some of its emotional

    components. Therefore, the ES might become chroni-

    cally activated as an adaptive response to neuropathic

    pain aiming at counteracting pain transmission [157].

    It can be, thus, explained why inhibitors of endocanna-

    binoid inactivation can exert analgesic effects in this

    experimental model of pain.

    There have been several reports on the analgesic

    efficacy of pharmacological inhibition of FAAH using

    different chemical classes of inhibitors such as a-keto-heterocycle compounds, carbamates, and analogues of

    N-arachidonoyl serotonin [75]. FAAH inhibition

    enhances the analgesic properties of AEA, as they would

    accumulate to higher levels in the absence of hydrolysis.

    This was confirmed in the FAAH knockout mice pro-

    duced by Cravatt et al. [76] that had enhanced levels of

    AEA and exhibited a hypoalgesic phenotype in several

    pain models. The anti-allodynic effects of the FAAH

    inhibitors, in several clinical studies, were fully reversed

    by pretreatment with either CB1 (i.e., SR141716A) or

    CB2 receptor antagonists, but were unaffected by the

    TRPV1 receptor antagonist, capsazepine [159].

    CB receptor agonists are active in animal models of

    acute pain when they are either administered peripher-

    ally or injected directly into the brain or spinal cord.

    Locally administered AEA has been shown to attenuate

    carrageenan-induced thermal hyperalgesia and cutane-

    ous edema [160]. Endogenous fatty acid derivatives

    such as oleamide, palmitoylethanolamide, 2-lineoylglyc-

    erol, 2-palmitoylglycerol, and a family of arachidonoyl

    amino acids may also interact with endocannabinoids in

    the modulation of pain sensitivity [136]. NADA elicits a

    host of cannabimimetic effects, including analgesia after

    systemic administration. It is noteworthy that NADA,

    through the activation of TRPV1, causes hyperalgesia

    when administered peripherally [35,161]. Given that

    NADA is capable of eliciting analgesia upon systemic

    administration and hyperalgesia upon intradermal injec-

    tion, it is possible that endogenous NADA may acti-

    vate either CB1 or TRPV1 depending on location

    and circumstance. Apart from NADA, 2-AGE has been

    reported, in experimental models, to induce sedation,

    hypothermia, intestinal immobility, and mild antinoci-

    ception in mice [2]. Animal experimental models using

    tail flick, hot plate, or radiant heat paw withdrawal tests

    have shown that cannabinoids can interact synergisti-

    cally with opioid receptor agonists in the production of

    antinociception. This synergism seems to be receptormediated as it can be blocked by both cannabinoid and

    opioid receptor antagonists [162,163]. The suppression

    of motor responses to noxious stimuli induced by CB1

    receptor agonists in animal pain models does not seem

    to stem from the known ability of these agents to impair

    motor function or to induce hypothermia. Thus, the

    analgesic effects of cannabinoids have been found to be

    due to the suppression of spinal and thalamic nocicep-

    tive neurons and independent of any actions on

    either the motor system or sensory neurons that trans-

    mit messages related to non-nociceptive stimulation

    [136].

    Synthetic cannabinoids

    The antinociceptive potency of D9-THC is no less than

    that of morphine, an agent already known to induce

    receptor-mediated analgesia [143, 162164]. A number

    of cannabinoids show even greater potency than D9-

    THC, for example, in the mouse tail flick test, after intra-

    venous administration [137,144]. A wide variety of

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 71

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    9/17

    synthetic cannabinoids have been produced that interact

    with CB receptors. Experimental studies have shown

    that activation of the CB1 receptor by synthetic agon-

    ists, and pharmacological elevation of endocannabinoid

    levels, suppress hyperalgesia and allodynia in animal

    models of neuropathic pain [165,166]. Systemic admin-

    istration of the CB receptor agonist Win55,212-2 dose-

    dependently reversed the thermal hyperalgesia and the

    mechanical and cold allodynia by a CB1, but not CB2

    receptor-mediated effect [167]. Win55,212-2 has also

    been reported to be effective after intrathecal and

    peripheral administration in doses not systemically

    active, suggesting that a potential peripheral site of

    action may be exploited to divorce the psychotropic

    effects of cannabinoids from their analgesic effects

    [167]. Bay 38-7271, another synthetic cannabinoid

    agonist, exerts analgesic and neuroprotective effects after

    traumatic brain injury in rats [168]. The selective

    CB1 receptor antagonist, SR141716A, can prevent the

    antinociceptive effects of CB receptor agonists at an

    appropriately high potency. It is important to bear in

    mind that, although SR141716A is CB1-selective, it is

    not CB1 specific and will, at sufficiently high doses,

    block CB2 as well as CB1 receptors [169].

    Clinical data

    It is generally accepted that opioid analgesics are less

    effective when used for the treatment of neuropathic

    pain in comparison with inflammatory pain. One

    explanation for this is that following peripheral nerveinjury, there is a depletion of opioid receptor expression

    in the spinal dorsal horn [170,171]. However, the

    destruction of primary afferent input to the dorsal

    horn, by dorsal rhizotomy [16] or neonatal capsaicin

    therapy [171], is not associated with such a depletion

    of CB1 receptor-like immunoreactivity or binding, thus

    giving cannabinoids a potential therapeutic advantage

    over opioids in neuropathic pain. Moreover, cannabi-

    noids may also be particularly efficacious in patient

    populations where the emetic effects of opioids are

    poorly tolerated, for example, in patients with cancer

    and patients with HIV infection [37].

    Side effects

    The development of cannabinoid agonists as analgesics

    has been hampered due to psychotropic and debilitating

    side effects. The most common adverse events associ-

    ated with the use of cannabis are headache, dry eyes,

    burning sensation in areas of neuropathic pain, dizzi-

    ness, numbness, cough, and effects on memory and on

    motor control that occur as a result of indiscriminate

    activation of CB receptors at sites other than those

    involved in the transmission of nociceptive stimuli

    [172,173]. Efforts are currently under way to develop

    inhalational forms ofD9-THC that may possibly be more

    effective than an oral formulation in managing cancer

    pain, either alone or in combination with other analge-

    sics [130,174]. It is noteworthy that in vivo cannabi-

    noid administration has been reported to be neurotoxic

    [175]. Moreover, there is a significant abuse potential,

    which has hindered their development as therapeutic

    agents [176]. Nevertheless, the synthetic cannabinoid

    abn-CBD represents a promising candidate for treatment

    of neuronal injury in vivo because it does not bind to

    CB1 and CB2 receptors and may thus produce less

    undesired side effects [177]. Therefore, a possible way

    to have its benefits, while you avoid its side effects, is to

    manipulate the endogenous cannabinoid system.

    Endocannabinoid analgesia

    As endocannabinoids exert their actions through the

    same targets (CB1 and CB2 receptors) with D9-THC

    and other exogenous cannabinoids, there are indica-

    tions that they share common analgesic properties.

    Multiple lines of evidence indicate that endocannabi-

    noids serve naturally to suppress pain. Physiological

    stimuli and pathological conditions lead to differential

    increases in brain endocannabinoids that regulate dis-

    tinct biological functions. Physiological stimuli lead to

    rapid and transient (seconds to minutes) increases inendocannabinoids that activate neuronal CB1 recep-

    tors, modulate ion channels, and inhibit neurotrans-

    mission [125], whereas pathological conditions lead

    to much slower and sustained (hours to days)

    increases in the endocannabinoid tone that changes

    gene expression, implementing molecular mechanisms

    that prevent the production and diffusion of harmful

    mediators [178180].

    While a proportion of the peripheral analgesic effects

    of endocannabinoids can be attributed to a neuronal

    mechanism acting through CB1 receptors expressed by

    primary afferent neurones, the anti-inflammatoryactions of endocannabinoids are mediated through CB2

    receptors that have also been found to participate in

    pain modulation [181]. Activation of the CB1 receptor

    inhibits the transmitter release from nociceptive pri-

    mary afferent fibers both at the periphery and the CNS.

    The GABA release within the PAG and RVM and the

    glutamate release within the spinal cord are inhibited

    [160]. Presynaptic inhibition is a particularly powerful

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    72 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    10/17

    mechanism of neural modulation, as it can have the

    final determinant influence on the output signal of a

    neuron and its subsequent communication to other

    neurons.

    Clinical trials

    In human clinical trials, administration of CB receptor

    agonists has been shown to be effective in treating neu-

    ropathic pain conditions and, in some instances, rival

    the analgesic efficacy of morphine [182,183]. A clinical

    trial in healthy volunteers revealed that low doses of

    smoked cannabis (D9-THC) can reduce the pain

    induced by intradermal capsaicin, while at higher doses

    the pain is increased [184]. Another study in patients

    with advanced cancer showed that the combined

    administration of D9-THC and CBD resulted in a statis-

    tically significant reduction in pain [185]. Smoked can-

    nabis has also been shown to effectively relieve chronicneuropathic pain from HIV-associated sensory neuropa-

    thy, and its use is generally well tolerated [186]. Cann-

    abinoids that have been approved in USA or Europe

    include dronabinol (MarinolTM, approved in USA, UK,

    and Canada), nabilone (CesametTM, approved in USA,

    UK, Canada, and Mexico), and GW-100 (SativexTM, a

    combination ofD9-THC and CBD, approved in UK, Can-

    ada, Spain, Czech Republic, Germany, and Denmark).

    However, all of these drugs contain D9-THC or an ana-

    logue and are nonselective with respect to CB1 and

    CB2 receptors. In several clinical studies, patients with

    multiple sclerosis have reported the benefits of D

    9

    -THCand the CB receptor agonist nabilone, in treating spas-

    ticity, pain, tremor, and ataxia [187,188].

    C O N C L U S I O N

    Over the last years, considerable progress has been

    made in understanding the role of endocannabinoids in

    pain modulation. The ES represents a local messenger

    between the nervous and immune system and is

    obviously involved in the control of immune activation

    and neuroprotection. Manipulation of endocannabi-

    noids and/or use of exogenous cannabinoids in vivocan constitute a potent treatment modality against

    inflammatory disorders. Cannabinoids have been tested

    in several experimental models of autoimmune disor-

    ders such as multiple sclerosis, rheumatoid arthritis,

    colitis, and hepatitis and have been shown to protect

    the host from the pathogenesis through induction of

    multiple anti-inflammatory pathways and consequently

    they also contribute to antinociception.

    Endocannabinoid signaling may be enhanced indi-

    rectly to therapeutic levels through FAAH inhibition,

    thus prolonging the duration of action of endogenously

    released AEA. Therefore, FAAH serves as an attrac-

    tive pharmacotherapeutic target and selective FAAH

    inhibitors as promising analgesic candidates for various

    neurological and neurodegenerative/neuroinflammato-

    ry disorders, including seizures of diverse etiology, multi-

    ple sclerosis, Alzheimers, Huntingtons, and Parkinsons

    diseases [9,189192]. Pharmacological inhibition of FAAH

    is antinociceptive in models of acute and inflammatory

    pain. Furthermore, inhibition of FAAH and MAGL

    reduces neuropathic pain through distinct receptor

    mechanisms of action and presents viable targets for

    the development of analgesic therapeutics [159]. The

    site- and event-specific character of the pharmacological

    inhibition of endocannabinoid-deactivating enzymes

    such as FAAH and MAGL may offer increased selectiv-

    ity with less risk of the undesirable side effects that have

    been observed with CB receptor agonists capable of acti-

    vating all accessible receptors indiscriminately [46, 47].

    The broad distribution of CB1 receptors in the brain

    underpins both their therapeutic effects, such as analge-

    sia, as well as their side effects. Several studies have dem-

    onstrated analgesic effects of CB2 receptor agonists in

    models of acute and chronic pain. Peripheral antinoci-

    ception without CNS effects, mediated by the ES, is con-

    sistent with the peripheral distribution of CB2 receptors.

    More specific drugs acting selectively on peripheral CB2

    receptors, and enzyme inhibitors preventing the break-down of endocannabinoids, offer a potential to separate

    the analgesic effects from the undesirable side effects of

    the drug and point to a mainstream role of cannabinoid

    medicines in the management of pain.

    Over the last decades, numerous studies have revealed

    several secrets of the ES [193,194]. Although, further

    information is still needed before the ES is completely com-

    prehended, pharmacological modulation of the ES seems,

    nowadays, a viable target that will pave the way for the

    therapeutic intervention at a wide spectrum of diseases.

    A B B R E V I A T I O N S

    2-AG 2-arachidonoylglycerol

    2-AGE 2-arachidonoylglyceryl ether, noladin ether

    ACC anterior cingulate cortex

    AEA anandamide, N-arachidonoylethanolamine

    APC antigen-presenting cells

    CB1/CB2 receptors cannabinoid 1/cannabinoid 2

    receptors

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 73

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    11/17

    CBD cannabidiol

    CBN cannabinol

    CCI chronic constriction injury

    CNS central nervous system

    COX cyclo-oxygenase

    CYP cytochrome P450

    DAG diacylglycerol

    DAGL diacylglycerol lipase

    DRG dorsal root ganglion

    DSE depolarization-induced suppression of excitation

    DSI depolarization-induced suppression of inhibition

    ERK extracellular signal-regulated kinase

    ES endocannabinoid system

    FAAH fatty acid amide hydrolase

    FAK focal adhesion kinase

    GABA gamma-aminobutyric acid

    IL interleukin

    MAGL monoacylglycerol lipase

    MAPK mitogen-activated protein kinase

    NADA N-arachidonoyldopamine

    NAE N-acylethanolamine

    NAPE N-arachidonoylphosphatidyl-ethanolamine

    NAPE-PLD NAPE-selective phospholipase D

    NF-jB nuclear factor jB

    NK natural killer (cells)

    nNOS neuronal nitric oxide synthase

    NO nitric oxide

    OAE O-arachidonoylethanolamine, virodhamine

    OEA oleoyl ethanolamide

    PAG periaqueductal grayPC phosphatidylcholine

    PEA palmitoyl ethanolamide

    PE phosphatidylethanolamine

    PK protein kinase

    ROS reactive oxygen species

    RVM rostral ventrolateral medulla

    TNF-a tumor necrosis factor-a

    TRPV1 transient receptor potential vanilloid 1

    D9-THC D9-tetrahydrocannabinol

    R E F E R E N C E S

    1 Pertwee R.G. The therapeutic potential of drugs that

    target cannabinoid receptors or modulate the tissue levels or

    actions of endocannabinoids. AAPS J. (2005) 7 625654.

    2 Grotenhermen F. Cannabinoids. Curr. Drug Targets CNS

    Neurol. Disord. (2005) 4 507530.

    3 Lichtman A.H. SR141716A enhances spatial memory as

    assessed in a radial-arm maze task in rats. Eur. J. Pharmacol.

    (2000) 404 175179.

    4 Rodriguez de Fonseca F., Del Arco I., Bermudez-Silva F.J.,

    Bilbao A., Cippitelli A., Navarro M. The endocannabinoid

    system: physiology and pharmacology. Alcohol Alcohol.

    (2005) 40 214.

    5 Izzo A.A., Sharkey K.A. Cannabinoids and the gut: new

    developments and emerging concepts. Pharmacol. Ther.

    (2010) 126 21

    38.6 Matsuda L.A., Lolait S.J., Brownstein M.J., Young A.C., Bonner

    T.I. Structure of a cannabinoid receptor and functional

    expression of the cloned cDNA. Nature (1990) 346 561564.

    7 Guzman M., Sanchez C., Galve-Roperh I. Control of the cell

    survival/death decision by cannabinoids. J. Mol. Med. (2001)

    78 613625.

    8 Devane W.A., Hanus L., Breuer A. et al. Isolation and

    structure of a brain constituent that binds to the

    cannabinoid receptor. Science (1992) 258 19461949.

    9 Bisogno T., Martire A., Petrosino S., Popoli P., Di Marzo V.

    Symptom-related changes of endocannabinoid and

    palmitoylethanolamide levels in brain areas of R6/2 mice, a

    transgenic model of Huntingtons disease. Neurochem. Int.

    (2008) 52 307

    313.

    10 Bari M., Battista N., Fezza F., Gasperi V., Maccarrone M. New

    insights into endocannabinoid degradation and its therapeutic

    potential. Mini. Rev. Med. Chem. (2006) 6 257268.

    11 Rajesh M., Mukhopadhyay P., Batkai S. et al. Cannabidiol

    attenuates high glucose-induced endothelial cell

    inflammatory response and barrier disruption. Am. J.

    Physiol. Heart Circ. Physiol. (2007) 293 610619.

    12 Rajesh M., Mukhopadhyay P., Hasko G., Huffman J.W.,

    Mackie K., Pacher P. CB2 cannabinoid receptor agonists

    attenuate TNF-a-induced human vascular smooth muscle

    cell proliferation and migration. Br. J. Pharmacol. (2008)

    153 347357.

    13 Piomelli D. The molecular logic of endocannabinoid

    signalling. Nat. Rev. Neurosci. (2003) 4 873884.

    14 Herkenham M. Characterization and localization of

    cannabinoid receptors in brain: an in vitro technique using

    slide-mounted tissue sections. NIDA Res. Monogr. (1991)

    112 129145.

    15 Herkenham M., Lynn A.B., Johnson M.R., Melvin L.S., de

    Costa B.R., Rice K.C. Characterization and localization of

    cannabinoid receptors in rat brain: a quantitative in vitro

    autoradiographic study. J. Neurosci. (1991) 11 563583.

    16 Farquhar-Smith W.P., Egertova M., Bradbury E.J., McMahon

    S.B., Rice A.S., Elphick M.R. Cannabinoid CB(1) receptor

    expression in rat spinal cord. Mol. Cell. Neurosci. (2000) 15

    510521.

    17 Iversen L. Cannabis and the brain. Brain (2003) 12612521270.

    18 Matyas F., Yanovsky Y., Mackie K., Kelsch W., Misgeld U.,

    Freund T.F. Subcellular localization of type 1 cannabinoid

    receptors in the rat basal ganglia. Neuroscience (2006) 137

    337361.

    19 Carrier E.J., Kearn C.S., Barkmeier A.J. et al. Cultured rat

    microglial cells synthesize the endocannabinoid

    2-arachidonylglycerol, which increases proliferation via a

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    74 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    12/17

    CB2 receptor-dependent mechanism. Mol. Pharmacol.

    (2004) 65 9991007.

    20 Gong J.P., Onaivi E.S., Ishiguro H. et al. Cannabinoid CB2

    receptors: immunohistochemical localization in rat brain.

    Brain Res. (2006) 1071 1023.

    21 Sheng W.S., Hu S., Min X., Cabral G.A., Lokensgard J.R.,

    Peterson P.K. Synthetic cannabinoid WIN55, 212

    2 inhibitsgeneration of inflammatory mediators by IL-1beta-stimulated

    human astrocytes. Glia (2005) 49 211219.

    22 Onaivi E.S., Ishiguro H., Gong J.P. et al. Discovery of the

    presence and functional expression of cannabinoid CB2

    receptors in brain. Ann. N. Y. Acad. Sci. (2006) 1074

    514536.

    23 Cabral G.A., Raborn E.S., Griffin L., Dennis J., Marciano-

    Cabral F. CB2 receptors in the brain: role in central immune

    function. Br. J. Pharmacol. (2008) 153 240251.

    24 Cabral G.A., Griffin-Thomas L. Cannabinoids as therapeutic

    agents for ablating neuroinflammatory disease. Endocr.

    Metab. Immune Disord. Drug Targets (2008) 8 159172.

    25 Sagredo O., Gonzalez S., Aroyo I. et al. Cannabinoid CB2

    receptor agonists protect the striatum against malonate

    toxicity: relevance for Huntingtons disease. Glia (2009) 57

    11541167.

    26 Golech S.A., McCarron R.M., Chen Y. et al. Human brain

    endothelium: coexpression and function of vanilloid and

    endocannabinoid receptors. Brain Res. Mol. Brain Res.

    (2004) 132 8792.

    27 Van Sickle M.D., Duncan M., Kingsley P.J. et al.

    Identification and functional characterization of

    brainstem cannabinoid CB2 receptors. Science (2005)

    310 329332.

    28 Wotherspoon G., Fox A., McIntyre P., Colley S., Bevan S.,

    Winter J. Peripheral nerve injury induces cannabinoid

    receptor 2 protein expression in rat sensory neurons.

    Neuroscience (2005) 135 235245.

    29 Ashton J.C., Friberg D., Darlington C.L., Smith P.F.

    Expression of the cannabinoid CB2 receptor in the rat

    cerebellum: an immunohistochemical study. Neurosci. Lett.

    (2006) 396 113116.

    30 Galiegue S., Mary S., Marchand J. et al. Expression of central

    and peripheral cannabinoid receptors in human immune

    tissues and leukocyte subpopulations. Eur. J. Biochem.

    (1995) 232 5461.

    31 Schatz A.R., Lee M., Condie R.B., Pulaski J.T., Kaminski N.E.

    Cannabinoid receptors CB1 and CB2: a characterization of

    expression and adenylate cyclase modulation within the

    immune system. Toxicol. Appl. Pharmacol. (1997) 142 278

    287.

    32 Rajesh M., Pan H., Mukhopadhyay P. et al. Cannabinoid-2

    receptor agonist HU-308 protects against hepatic ischemia/

    reperfusion injury by attenuating oxidative stress,

    inflammatory response, and apoptosis. J. Leukoc. Biol.

    (2007) 82 13821389.

    33 McAllister S.D., Glass M. CB(1) and CB(2) receptor-mediated

    signalling: a focus on endocannabinoids. Prostaglandins

    Leukot. Essent. Fatty Acids (2002) 66 161171.

    34 Mackie K. Cannabinoid receptors as therapeutic targets.

    Annu. Rev. Pharmacol. Toxicol. (2006) 46 101122.

    35 Huang S.M., Bisogno T., Trevisani M. et al. An endogenous

    capsaicin-like substance with high potency at recombinant

    and native vanilloid VR1 receptors. Proc. Natl Acad. Sci.

    USA (2002) 99 84008405.

    36 Porter A.C., Sauer J.M., Knierman M.D. et al.Characterization of a novel endocannabinoid, virodhamine,

    with antagonist activity at the CB1 receptor. J. Pharmacol.

    Exp. Ther. (2002) 301 10201024.

    37 Nagarkatti P., Pandey R., Rieder S.A., Hegde V.L.,

    Nagarkatti M. Cannabinoids as novel anti-inflammatory

    drugs. Future Med Chem (2009) 1 13331349.

    38 Sugiura T., Kondo S., Kishimoto S. et al. Evidence that

    2-arachidonoylglycerol but not N-palmitoylethanolamine or

    anandamide is the physiological ligand for the cannabinoid

    CB2 receptorcomparison of the agonistic activities of

    various cannabinoid receptor ligands in HL-60 cells. J. Biol.

    Chem. (2000) 275 605612.

    39 Venance L., Piomelli D., Glowinski J., Giaume C.

    Inhibition by anandamide of gap junctions and

    intercellular calcium signalling in striatal astrocytes.

    Nature (1995) 376 590594.

    40 Sagan S., Venance L., Torrens Y., Cordier J., Glowinski J.,

    Giaume C. Anandamide and WIN 55212-2 inhibit cyclic

    AMP formation through G-protein-coupled receptors distinct

    from CB1 cannabinoid receptors in cultured astrocytes. Eur.

    J. Neurosci. (1999) 11 691699.

    41 Di Marzo V., Bisogno T., De Petrocellis L. Endocannabinoids:

    new targets for drug development. Curr. Pharm. Des. (2000)

    6 13611380.

    42 Szoke E., Balla Z., Csernoch L., Czeh G., Szolcsanyi J. Interacting

    effects of capsaicin and anandamide on intracellular calcium in

    sensory neurones. NeuroReport (2000) 11 19491952.

    43 Mechoulam R., Ben-Shabat S., Hanus L. et al. Identification

    of an endogenous 2-monoglyceride, present in canine gut,

    that binds to cannabinoid receptors. Biochem. Pharmacol.

    (1995) 50 8390.

    44 Sugiura T., Kondo S., Sukagawa A. et al.

    2-Arachidonoylglycerol: a possible endogenous cannabinoid

    receptor ligand in brain. Biochem. Biophys. Res. Commun.

    (1995) 215 8997.

    45 Di Marzo V., Petrosino S. Endocannabinoids and the

    regulation of their levels in health and disease. Curr. Opin.

    Lipidol. (2007) 18 129140.

    46 Vemuri V.K., Janero D.R., Makriyannis A.

    Pharmacotherapeutic targeting of the endocannabinoid

    signaling system: drugs for obesity and the metabolicsyndrome. Physiol. Behav. (2008) 93 671686.

    47 Janero D.R., Vadivel S.K., Makriyannis A.

    Pharmacotherapeutic modulation of the endocannabinoid

    signalling system in psychiatric disorders: drug-discovery

    strategies. Int. Rev. Psychiatry (2009) 21 122133.

    48 Bisogno T., Melck D., Bobrov M. et al. N-acyl-dopamines:

    novel synthetic CB1 cannabinoid-receptor ligands and

    inhibitors of anandamide inactivation with

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 75

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    13/17

    cannabimimetic activity in vitro and in vivo. Biochem. J.

    (2000) 1 817824.

    49 Walker M.J., Krey F.J., Chu C.J., Huang S.M.

    Endocannabinoids and related fatty acid derivatives in pain

    modulation. Chem. Phys. Lipids (2002) 121 159172.

    50 Hanus L. et al. 2-Arachidonoyl glycerol ether, endogenous

    agonist of the cannabinoid CB1 receptor. Proc. Natl Acad.Sci. U S A (2001) 98 36623665.

    51 Di Marzo V., Bisogno T., De Petrocellis L. et al. Biosynthesis

    and inactivation of the endocannabinoid 2-

    arachidonoylglycerol in circulating and tumoral

    macrophages. Eur. J. Biochem. (1999) 264 258267.

    52 Longhua Z., Meina W., Bisogno Tiziana B., Di Marzo V.,

    Alger B.E. Endocannabinoids Generated by Ca2 + or by

    Metabotropic Glutamate Receptors Appear to Arise from

    Different Pools of Diacylglycerol Lipase. PLoS One (2011) 6

    16305.

    53 van Der Stelt M., Veldhuis W.B., van Haaften G.W. et al.

    Exogenous anandamide protects rat brain against

    acute neuronal injury in vivo. J. Neurosci. (2001) 21

    8765

    8771.

    54 Garcia-Ovejero D., Arevalo-Martin A., Petrosino S. et al. The

    endocannabinoid system is modulated in response to spinal

    cord injury in rats. Neurobiol. Dis. (2009) 33 5771.

    55 Ahn K., McKinney M.K., Cravatt B.F. Enzymatic pathways

    that regulate endocannabinoid signaling in the nervous

    system. Chem. Rev. (2008) 108 16871707.

    56 Lovinger D.M. Presynaptic modulation by endocannabinoids.

    Handb. Exp. Pharmacol. (2008) 184 435477.

    57 Lopez R., Mateos Vicente B., Romero-Zerbo S.Y. et al.

    Estradiol decreases cortical reactive astrogliosis after brain

    injury by a mechanism involving cannabinoid receptors.

    Cereb. Cortex (2011) 21 20462055.

    58 Schmid P.C., Krebsbach R.J., Perry S.R., Dettmer T.M.,

    Maasson J.L., Schmid H.H. Occurrence and postmortem

    generation of anandamide and other long-chain N-

    acylethanolamines in mammalian brain. FEBS Lett. (1995)

    375 143147.

    59 Hansen H.H., Schmid P.C., Bittigau P. et al. Anandamide,

    but not 2-arachidonoylglycerol, accumulates during in vivo

    neurodegeneration. J. Neurochem. (2001) 78 14151427.

    60 Natarajan V., Schmid P.C., Reddy P.V., Zuzarte-Augustin

    M.L., Schmid H.H. Biosynthesis of Nacylethanolamine

    phospholipids by dog brain preparations. J. Neurochem.

    (1983) 41 13031312.

    61 Natarajan V., Schmid P.C., Reddy P.V., Schmid H.H.

    Catabolism of N-acylethanolamine phospholipids by dog

    brain preparations. J. Neurochem. (1984) 41 1303

    1312.62 Cadas H., di Tomaso E., Piomelli D. Occurrence and

    biosynthesis of endogenous cannabinoid precursor,

    N-arachidonoyl phosphatidylethanolamine, in rat brain.

    J. Neurosci. (1997) 17 12261242.

    63 Giuffrida A., Parsone L.H., Kerr T.M., Rodriguez de Fonseca

    F., Navarro M., Pomelli D. Dopamine activation of

    endogenous cannabinoid signaling in dorsal striatum. Nat.

    Neurosci. (1999) 2 358363.

    64 Zhang J., Hoffert C., Vu H.K., Groblewski T., Ahmad S.,

    ODonnell D. Induction of CB2 receptor expression in the rat

    spinal cord of neuropathic but not inflammatory chronic

    pain models. Eur. J. Neurosci. (2003) 17 27502754.

    65 Sugiura T., Kishimoto S., Oka S., Gokoh M., Wake K.

    Metabolism and physiological significance of anadamide and

    2-arachidonoylglycerol, endogenous cannabinoid receptorligands, in: Fonteh A.N., Wykle R.L. (Eds), Arachidonate

    remodeling and inflammation, Birkhduser Verlag, Basel,

    Switzerland, 2004, pp. 211237.

    66 Mechoulam R., Shohami E. Endocannabinoids and traumatic

    brain injury. Mol. Neurobiol. (2007) 36 6874.

    67 Miller A.S., Walker J.M. Effects of a cannabinoid on spontaneous

    and evoked neuronal activity in the substantia nigra pars

    reticulata. Eur. J. Pharmacol. (1995) 279 179185.

    68 Croxford J.L., Yamamura T. Cannabinoids and the immune

    system: potential for the treatment of inflammatory diseases.

    J. Neuroimmunol. (2005) 166 318.

    69 Cravatt B.F., Giang D.K., Mayfield S.P., Boger D.L., Lerner

    R.A., Gilula N.B. Molecular characterization of an enzyme

    that degrades neuromodulatory fatty acid amides. Nature

    (1996) 384 8387.

    70 Fegley D., Kathuria S., Mercier R. et al. Anandamide transport

    is independent of fatty-acid amide hydrolase activity and is

    blocked by the hydrolysis-resistant inhibitor AM1172. Proc.

    Natl Acad. Sci. USA (2004) 101 87568761.

    71 Blankman J.L., Simon G.M., Cravatt B.F. A comprehensive

    profile of brain enzymes that hydrolyze the endocannabinoid

    2-arachidonoylglycerol. Chem. Biol. (2007) 14 13471356.

    72 Di Marzo V. Endocannabinoids: synthesis and degradation.

    Rev. Physiol. Biochem. Pharmacol. (2008) 160 124.

    73 Fu J., Bottegoni G., Sasso O. et al. A catalytically silent

    FAAH-1 variant drives anandamide transport in neurons.

    Nat. Neurosci. (2011) 15 6469.

    74 Egertova M., Giang D.K., Cravatt B.F., Elphick M.R. A new

    perspective on cannabinoid signalling: complementary

    localization of fatty acid amide hydrolase and the CB1

    receptor in rat brain. Proc. Biol. Sci. (1998) 265

    20812085.

    75 Karbarz M.J., Luo L., Chang L. et al. Biochemical and

    Biological Properties of 4-(3-phenyl-[1,2,4] thiadiazol-5-yl)-

    piperazine-1-carboxylic acid phenylamide, a Mechanism-

    Based Inhibitor of Fatty Acid Amide Hydrolase. Anesth.

    Analg. (2009) 108 316329.

    76 Cravatt B.F., Demarest K., Patricelli M.P. et al.

    Supersensitivity to anandamide and enhanced endogenous

    cannabinoid signaling in mice lacking fatty acid amide

    hydrolase. Proc. Natl Acad. Sci. USA (2001) 98 9371

    9376.77 Katayama K., Ueda N., Katoh I., Yamamoto S. Equilibrium

    in the hydrolysis and synthesis of cannabimimetic

    anandamide demonstrated by a purified enzyme. Biochim.

    Biophys. Acta (1999) 1440 205214.

    78 Kurahashi Y., Ueda N., Suzuki H., Suzuki M., Yamamoto S.

    Reversible hydrolysis and synthesis of anandamide

    demonstrated by recombinant rat fatty-acid amide hydrolase.

    Biochem. Biophys. Res. Commun. (1997) 237 512515.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    76 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    14/17

    79 Arreaza G., Devane W., Omeir R.L. et al. The cloned rat

    hydrolytic enzyme responsible for the breakdown of

    anandamide also catalyzes its formation via the

    condensation of arachidonic acid and ethanolamine.

    Neurosci. Lett. (1997) 234 5962.

    80 Walter L., Dinh T., Stella N. ATP induces a rapid and

    pronounced increase in 2-arachidonoylglycerol productionby astrocytes, a response limited by monoacylglycerol lipase.

    J. Neurosci. (2004) 24 80688074.

    81 Goparaju S.K., Udea N., Yamaguchi H., Yamamoto S.

    Anandamide aminohydrolase reacting with

    2-arachidonoylglycerol, another cannabinoid receptor ligand.

    FEBS Lett. (1998) 422 6973.

    82 Maione S., De Petrocellis L., de Novellis V. et al. Analgesic

    actions of N-arachidonoyl-serotonin, a fatty acid amide

    hydrolase inhibitor with antagonistic activity at vanilloid

    TRPV1 receptors. Br. J. Pharmacol. (2007) 150

    766781.

    83 Yu M., Ives D., Ramesha C.S. Synthesis of prostaglandin E2

    ethanolamide from anandamide by cyclooxygenase-2. J. Biol.

    Chem. (1997) 272 21181

    21186.

    84 Sang N., Chen C. Lipid signaling and synaptic plasticity.

    Neuroscientist (2006) 2 425434.

    85 Sang N., Zhang J., Chen C. COX-2 oxidative metabolite of

    endocannabinoid 2-AG enhances excitatory glutamatergic

    synaptic transmission and induces neurotoxicity. J.

    Neurochem. (2007) 102 19661977.

    86 Ross R.A., Craib S.J., Stevenson L.A. et al. Pharmacological

    characterization of the anandamide cyclooxygenase

    metabolite:prostaglandin E2 ethanolamide. J. Pharmacol.

    Exp. Ther. (2002) 301 900907.

    87 Nirodi C.S., Crew B.C., Kozak K.R., Rorrow J.D., Marnett L.J.

    The glyceryl ester of prostaglandin E2 mobilize calcium and

    activates signal transduction in RAW 264.7 cells. Proc. Natl

    Acad. Sci. USA (2004) 101 18401845.

    88 Kozak K.R., Prusakiewicz J.L., Marnett L.J. Oxidative

    metabolism of endocannabinoids by COX-2. Curr.

    Pharmaceut. Design (2004) 10 659667.

    89 Patrignani P., Tacconelli S., Sciulli M.G., Capone M.L. New

    insights into COX-2 biology and inhibition. Brain Res. Rev.

    (2005) 48 352359.

    90 Hengartner M.O. The biochemistry of apoptosis. Nature

    (2000) 407 770776.

    91 Shohami E., Beit-Yannai E., Horowitz M., Kohen R. Oxidative

    stress in closed-head injury: brain antioxidant capacity as an

    indicator of functional outcome. J. Cereb. Blood Flow Metab.

    (1997) 17 10071019.

    92 Shohami E., Gallily R., Mechoulam R., Bass R., Ben Hur T.Cytokine production in the brain following closed head

    injury: dexanabinol (HU-211) is a novel TNF-alpha inhibitor

    and an effective neuroprotectant. J. Neuroimmunol. (1997)

    72 169177.

    93 Cabral G.A., Harmon K.N., Carlisle S.J. Cannabinoid-

    mediated inhibition of inducible nitric oxide production by

    rat microglial cells: evidence for CB1 receptor participation.

    Adv. Exp. Med. Biol. (2001) 493 207214.

    94 Molina-Holgado F., Molina-Holgado E., Guaza C. The

    endogenous cannabinoid anandamide potentiates IL-6

    production by astrocytes infected with Theilers murine

    encephalomyelitis virus by a receptor- mediated pathway.

    FEBS Lett. (1998) 433 139142.

    95 Choi S., Lovinger D.M. Metabotropic glutamate receptor

    modulation of voltage-gated Ca2+

    channels involvesmultiple receptor subtypes in cortical neurons. J. Neurosci.

    (1996) 16 3645.

    96 Twitchell W., Brown S., Mackie K. Cannabinoids inhibit

    N- and P/Q-type calcium channels in cultured rat

    hippocampal neurons. J. Neurophysiol. (1997) 78 4350.

    97 Shen M., Piser T.M., Seybold V.S., Thayer S.A. Cannabinoid

    receptor agonists inhibit glutamatergic synaptic

    transmission in rat hippocampal cultures. J. Neurosci.

    (1996) 16 43224334.

    98 van der Stelt M., Veldhuis W.B., Maccarrone M. et al. Acute

    neuronal injury, excitotoxicity, and the endocannabinoid

    system. Mol. Neurobiol. (2002) 26 317346.

    99 Mehta S.L., Manhas N., Raghubir R. Molecular targets in

    cerebral ischemia for developing novel therapeutics. Brain

    Res. Rev. (2007) 54 3466.

    100 Howlett A.C., Fleming R.M. Cannabinoid inhibition of

    adenylate cyclase. Pharmacology of the response in

    neuroblastoma cell membranes. Mol. Pharmacol. (1984) 26

    532538.

    101 Chevaleyre V., Takahashi K.A., Castillo P.E.

    Endocannabinoid-mediated synaptic plasticity in the CNS.

    Annu. Rev. Neurosci. (2006) 29 3776.

    102 Derkinderen P., Toutant M., Burgaya F. et al. Regulation of

    a neuronal form of focal adhesion kinase by anandamide.

    Science (1996) 273 17191722.

    103 Derkinderen P., Valjent E., Toutant M. et al. Regulation of

    extracellular signal-regulated kinase by cannabinoids in

    hippocampus. J. Neurosci. (2003) 23 23712382.

    104 Guzman M. Cannabinoids: potential anticancer agents. Nat.

    Rev. Cancer (2003) 3 745755.

    105 Oz M. Receptor-independent actions of cannabinoids on cell

    membranes: focus on endocannabinoids. Pharmacol. Ther.

    (2006) 111 114144.

    106 Zygmunt P.M., Peterson J., Andersson D.A. et al. Vanilloid

    receptors on sensory nerves mediate the vasodilator action

    of anandamide. Nature (1999) 400 452457.

    107 De Petrocellis L., Di Marzo V. Lipids as regulators of the

    activity of transient receptor potential type V1 (TRPV1)

    channels. Life Sci. (2005) 77 16511666.

    108 Maccarrone M., Lorenzon T., Bari M., Melino G., Finazzi-

    Agro A. Anandamide induces apoptosis in human cellsvia vanilloid receptors - evidence for a protective role of

    cannabinoid receptors. J. Biol. Chem. (2000) 275

    3193831945.

    109 Cernak I., Vink R., Natale J. et al. The dark side of

    endocannabinoids: a neurotoxic role for anandamide.

    J. Cereb. Blood Flow Metab. (2004) 24 564578.

    110 Szallasi A., Blumberg P.M. Vanilloid (Capsaicin) receptors

    and mechanisms. Pharmacol. Rev. (1999) 51 159212.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 77

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    15/17

    111 Marinelli S., Vaughan C.W., Christie M.J., Connor M.

    Capsaicin activation of glutamatergic synaptic transmission

    in the rat locus coeruleus in vitro. J. Physiol. (2002) 543

    531540.

    112 Yue H.Y., Fujita T., Kawasaki Y., Kumamoto E.

    AM404 enhances the spontaneous release of

    L-glutamate in a manner sensitive to capsazepine inadult rat substantia gelatinosa neurones. Brain Res.

    (2004) 1018 283287.

    113 Berdyshev E.V. Cannabinoid receptors and the regulation of

    immune response. Chem. Phys. Lipids (2000) 108 169190.

    114 Lombard C., Nagarkatti M., Nagarkatti P. CB2 cannabinoid

    receptor agonist, JWH-015, triggers apoptosis in immune

    cells: potential role for CB2-selective ligands as immuno-

    suppressive agents. Clin. Immunol. (2007) 122 259270.

    115 Jacobsson S.O., Rongard E., Stridh M., Tiger G., Fowler C.J.

    Serum-dependent effects of tamoxifen and cannabinoids

    upon C6 glioma cell viability. Biochem. Pharmacol. (2000)

    60 18071813.

    116 Panikashvili D., Mechoulam R., Beni S.M., Alexandrovich

    A., Shohami E.. CB1 cannabinoid receptors are involved in

    neuroprotection via NF-kappa B inhibition. J. Cereb. Blood

    Flow Metab. (2005) 25 477484.

    117 Sarker K.P., Obara S., Nakata M., Kitajima I., Maruyama I.

    Anandamide induces apoptosis of PC-12 cells: involvement

    of superoxide and caspase-3. FEBS Lett. (2000) 472 3944.

    118 Eljaschewitsch E., Witting A., Mawrin C. et al. The

    endocannabinoid anandamide protects neurons during CNS

    inflammation by induction of MKP-1 in microglial cells.

    Neuron (2006) 49 6779.

    119 Pitler T.A., Alger B.E. Depolarization-induced suppression of

    GABAergic inhibition in rat hippocampal pyramidal cells: G

    protein involvement in a presynaptic mechanism. Neuron

    (1994) 13 14471455.

    120 Wilson R.I., Nicoll R.A. Endogenous cannabinoids mediate

    retrograde signalling at hippocampal synapses. Nature

    (2001) 410 588592.

    121 Wilson R.I., Kunos G., Nicoll R.A. Presynaptic specificity of

    endocannabinoid signaling in the hippocampus. Neuron

    (2001) 31 453462.

    122 Alsasua del Valle A. Implication of cannabinoids in neurological

    diseases. Cell. Mol. Neurobiol. (2006) 26 579591.

    123 Trettel J., Levine E.S. Endocannabinoids mediate rapid

    retrograde signaling at interneuron right-arrow pyramidal

    neuron synapses of the neocortex. J. Neurophysiol. (2003)

    89 23342338.

    124 Kreitzer A.C., Regehr W.G. Retrograde inhibition of

    presynaptic calcium influx by endogenous cannabinoids atexcitatory synapses onto Purkinje cells. Neuron (2001) 29

    717727.

    125 Freund T.F., Katona I., Piomelli D. Role of endogenous

    cannabinoids in synaptic signaling. Physiol. Rev. (2003) 83

    10171066.

    126 Mackie K., Hille B. Cannabinoids inhibit N-type calcium

    channels in neuroblastoma-glioma cells. Proc. Natl Acad.

    Sci. USA (1992) 89 38253829.

    127 Kim J., Alger B.E. Inhibition of cyclooxygenase-2 potentiates

    retrograde endocannabinoid effects in hippocampus. Nat.

    Neurosci. (2004) 7 697698.

    128 Katona I., Urban G.M., Wallace M. et al. Molecular

    composition of the endocannabinoid system at glutamatergic

    synapses. J. Neurosci. (2006) 26 56285637.

    129 Yoshida T., Fukaya M., Uchigashima M. et al. Localizationof diacylglycerol lipase-alpha around postsynaptic spine

    suggests close proximity between production site of an

    endocannabinoid, 2-arachidonoyl-glycerol, and

    presynaptic cannabinoid CB1 receptor. J. Neurosci. (2006)

    26 47404751.

    130 Wilson R.I., Nicoll R.A. Endocannabinoid signaling in the

    brain. Science (2002) 296 678682.

    131 Alger B.E. Retrograde signaling in the regulation of synaptic

    transmission: focus on endocannabinoids. Prog. Neurobiol.

    (2002) 68 247286.

    132 Tsou K., Lowitz K.A., Hohmann A.G. et al. Suppression of

    noxious stimulus evoked expression of fos-like

    immunoreactivity in rat spinal cord by a selective

    cannabinoid agonist. Neuroscience (1995) 70 791

    798.

    133 Herzberg U., Eliav E., Bennett G.J., Kopin I.J. The analgesic

    effects of R(+)-WIN55,2122 mesylate, a high affinity

    cannabinoid agonist, in a rat model of neuropathic pain.

    Neurosci. Lett. (1997) 221 157160.

    134 Wang L.X., Wang Z.J. Animal and cellular models of

    chronic pain. Adv. Drug Deliv. Rev. (2003) 55

    949965.

    135 Jaggi A.S., Jain V., Singh N. Animal models of neuropathic

    pain. Fundam. Clin. Pharmacol. (2011) 25 128.

    136 Walker J.M., Huang S.M. Cannabinoid analgesia.

    Pharmacol. Ther. (2002) 95 127135.

    137 Martin W.J., Patrick S.L., Coffin P.O., Tsou K., Walker J.M.

    An examination of the central sites of action of

    cannabinoid-induced antinociception in the rat. Life Sci.

    (1995) 56 21032109.

    138 Martin L.J., Al-Abdulla N.A., Brambrink A.M., Kirsch

    J.R., Sieber F.E., Portera-Caill iau C. Neurodegene ration in

    excitotoxicity, global cerebral ischemia, and target

    deprivation: a perspective on the contributions of

    apoptosis and necrosis. Brain Res. Bull. (1998) 46

    281309.

    139 Martin W.J., Loo C.M., Basbaum A.I. Spinal cannabinoids

    are anti-allodynic in rats with persistent inflammation. Pain

    (1999) 82 199205.

    140 Martin W.J., Coffin P.O., Attias E., Balinsky M., Tsou K.,

    Walker J.M. Anatomical basis for cannabinoid-induced

    antinociception as revealed by intracerebral microinjections.Brain Res. (1999) 822 237242.

    141 Treede R.D., Kenshalo D.R., Gracely R.H., Jones A.K. The

    cortical representation of pain. Pain (1999) 79 105111.

    142 Hoot M.R., Sim-Selley L.J., Poklis J.L., Rehab A. Abdullah,

    Krista L. Scoggins, Dana E. Selley, William L. Dewey.

    Chronic constriction injury reduces cannabinoid receptor 1

    activity in the rostral anterior cingulate cortex of mice.

    Brain Res. (2010) 1339 1825.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de Therapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    78 P. Zogopoulos et al.

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    16/17

    143 Martin B.R., Lichtman A.H. Cannabinoid transmission and

    pain perception. Neurobiol. Dis. (1998) 5 447461.

    144 Martin W.J., Tsou K., Walker J.M. Cannabinoid receptor-

    mediated inhibition of the rat tail-flick reflex after

    microinjection into the rostral ventromedial medulla.

    Neurosci. Lett. (1998) 242 3336.

    145 Martin B.R., Thomas B.F., Razdan R.K. Structuralrequirements for cannabinoid receptor probes, in: Pertwee

    R.G. (Ed.), Cannabinoid receptors, Academic Press, London,

    1995b, pp. 3585.

    146 Nackley A.G., Suplita R.L., Hohmann A.G. A peripheral

    cannabinoid mechanism suppresses spinal fos protein

    expression and pain behavior in a rat model of

    inflammation. Neuroscience (2003) 117 659670.

    147 Sagar D.R., Kelly S., Millns P.J., OShaughnessey C.T.,

    Kendall D.A., Chapman V. Inhibitory effects of CB1 and CB2

    receptor agonists on responses of DRG neurons and dorsal

    horn neurons in neuropathic rats. Eur. J. Neurosci. (2005)

    22 371379.

    148 Khanna N., Malhotra R.S., Mehta A.K., Garg G.R., Halder

    S., Sharma K.K. Interaction of morphine and potassium

    channel openers on experimental models of pain in mice.

    Fundam. Clin. Pharmacol. (2011) 25 479484.

    149 Bloom A.S., Dewey W.L., Harris L.S., Brosius K.K. 9-nor-

    9bhydroxyhexahydrocannabinol a cannabinoid with potent

    antinociceptive activity: comparisons with morphine.

    J. Pharmacol. Exp. Ther. (1997) 200 263270.

    150 Sofia R.D., Nalepa S.D., Harakal J.J., Vassar H.B. Anti-edema

    and analgesic properties of D9-tetrahydrocannabinol THC.

    J. Pharmacol. Exp. Ther. (1973) 186 646655.

    151 Formukong E.A., Evans A.T., Evans F.J. Analgesic and

    antiinflammatory activity of constituents of Cannabis sativa

    L. Inflammation (1988) 12 361371.

    152 Walczak J.S., Pichette V., Leblond F., Desbiens K., Beaulieu

    P. Characterization of chronic constriction of the saphenous

    nerve, a model of neuropathic pain in mice showing rapid

    molecular and electrophysiological changes. J. Neurosci. Res.

    (2006) 83 13101322.

    153 Costa B., Colleoni M., Trovato A.E., Conti S., Pomelli F.,

    Franke C. Anandamide uptake and hydrolysis as targets for

    the treatment of neuropathic pain: effect of AM404 and

    URB597 in rats with chronic constriction injury of the

    sciatic nerve. XV Symposium on the Cannabinoids,

    Clearwater Beach, FL, 24-29 June 2005, p. 33.

    154 Costa B., Siniscalco D., Trovato A.E. et al. AM404, an

    inhibitor of anandamide uptake, prevents pain behaviour

    and modulates cytokine and apoptotic pathways in a rat

    model of neuropathic pain. Br. J. Pharmacol. (2006) 14810221032.

    155 Ledent C., Valverde O., Cossu G. et al. Unresponsiveness to

    cannabinoids and reduced addictive effects of opiates in CB1

    receptor knockout mice. Science (1999) 283

    401404.

    156 Agarwal N., Pacher P., Tegeder I. et al. Cannabinoids mediate

    analgesia largely via peripheral type 1 cannabinoid receptors

    in nociceptors. Nat. Neurosci. (2007) 10 870879.

    157 Petrosino S., Palazzo E., de Novellis V. et al. Changes in

    spinal and supraspinal endocannabinoid levels in

    neuropathic rats. Neuropharmacology (2007) 52 415422.

    158 Kanai Y., Nakazato E., Fujiuchi A., Hara T., Imai A.

    Involvement of an increased spinal TRPV1 sensitization

    through its up-regulation in mechanical allodynia of CCI

    rats. Neuropharmacology (2005) 49 977

    984.159 Kinsey S.G., Long J.Z., ONeal S.T. et al. Blockade of

    endocannabinoid-degrading enzymes attenuates neuropathic

    pain. J. Pharmacol. Exp. Ther. (2009) 330 902910.

    160 Richardson J.D., Kilo S., Hargreaves K.M. Cannabinoids

    reduce hyperalgesia and inflammation via interaction with

    peripheral CB1 receptors. Pain (1998) 75 111119.

    161 Bisogno T., Ligresti A., Di Marzo V. The endocannabinoid

    signalling system: biochemical aspects. Pharmacol. Biochem.

    Behav. (2005) 81 224238.

    162 Welch S.P., Stevens D.L. Antinociceptive activity of

    intrathecally administered cannabinoids alone, and in

    combination with morphine, in mice. J. Pharmacol. Exp.

    Ther. (1992) 262 1018.

    163 Cichewicz D.L., Martin Z.L., Smith F.L., Welch S.P.

    Enhancement of m opioid antinociception by oral D9-

    tetrahydrocannabinol: dose-response analysis and receptor

    identification. J. Pharmacol. Exp. Ther. (1999) 289 859867.

    164 Gonzalez-Rodr guez S., Llames S., Hidalgo A., Baamonde A.,

    Menendez L. Potentiation of acute morphine-induced

    analgesia measured by a thermal test in bone cancer-bearing

    mice. Fundam. Clin. Pharmacol. (2012) 26 363372.

    165 Goya P., Jagerovic N., Hernandez-Folgado L., Martin M.I.

    Cannabinoids and neuropathic pain. Mini Rev. Med. Chem.

    (2003) 3 765772.

    166 Cravatt B.F., Lichtman A.H. The endogenous cannabinoid

    system and its role in nociceptive behavior. J. Neurobiol.

    (2004) 61 149160.

    167 Bridges D., Rice A.S.C., Egertova M., Elphick M.R., Winter J.

    The cannabinoid CB l receptor is localised predominantly on

    N52 within the rat dorsal root ganglion. Soc. Neurosci.

    Abstr. (2001) 27 716.3.

    168 Mauler F., Hinz V., Augstein K.H., Fassbender M., Horvath

    E. Neuroprotective and brain edema-reducing efficacy of the

    novel cannabinoid receptor agonist BAY 38-7271. Brain

    Res. (2003) 989 99111.

    169 Pertwee R.G. Pharmacology of cannabinoid receptor ligands.

    Curr. Med. Chem. (1999) 6 635664.

    170 Besse D., Lombard M.C., Perrot S., Besson J.M. Regulation of

    opioid binding sites in the superficial dorsal horn of the rat

    spinal cord following loose ligation of the sciatic nerve:

    comparison with sciatic nerve section and lumbar dorsalrhizotomy. Neuroscience (1992) 50 921933.

    171 Hohmann A.G., Tsou K., Walker J.M. Cannabinoid

    modulation of wide dynamic range neurons in the lumbar

    dorsal horn of the rat by spinally administered WIN55,

    212-2. Neurosci. Lett. (1998) 257 119122.

    172 Carlini E.A. The good and the bad effects of (-)trans-delta-

    9-tetrahydrocannabinol (Delta 9 THC) on humans. Toxicon

    (2004) 44 461467.

    2012 The Authors Fundamental and Clinical Pharmacology 2012 Societe Francaise de Pharmacologie et de The rapeutique

    Fundamental & Clinical Pharmacology 27 (2013) 6480

    Endocannabinoids and pain 79

  • 7/29/2019 Endocanabinoides en Manejo Del Dolor

    17/17

    173 Ware M., Wang T., Shapiro S., Robinson A. et al. Smoked

    cannabis for chronic neuropathic pain: a randomized

    controlled trial. CMAJ (2010) 182 694701.

    174 Lichtman A.H., Peart J., Poklis J.L. et al. Pharmacological

    evaluation of aerosolized cannabinoids in mice. Eur. J.

    Pharmacol. (2000) 399 141149.

    175 Landfield P.W., Cadwallader L.B., Vinsant S. Quantitativechanges in hippocampal structure following long-term

    exposure to delta 9-tetrahydrocannabinol: possible mediation

    by glucocorticoid systems. Brain Res. (1988) 443 4762.

    176 Gardner E.L. Endocannabinoids ignaling system andb rain

    reward: emphasis on dopamine. Pharmacol. Biochem.

    Behav. (2005) 81 263284.

    177 Kreutz S., Koch M., Bottger C., Ghadban C., Korf H.W.,

    Dehghani F. 2-Arachidonoylglycerol elicits neuroprotective

    effects on excitotoxically lesioned dentate gyrus granule cells

    via abnormal- cannabidiol sensitive receptors on microglial

    cells. Glia (2009) 57 286294.

    178 Nagayama T., Sinor A.D., Simon R.P. et al. Cannabinoids and

    neuroprotection in global and focal cerebral ischemia and in

    neuronal cultures. J. Neurosci. (1999) 19 2987

    2995.

    179 Marsicano G., Goodenough S., Monory K. et al. CB1

    cannabinoid receptors and on-demand defense against

    excitotoxicity. Science (2003) 302 8488.

    180 Stella N. Cannabinoid signaling in glial cells. Glia (2004) 48

    267277.

    181 Rice A.S.C., Farquhar-Smith W.P., Nagy I.

    Endocannabinoids and pain: spinal and peripheral analgesia

    in inflammation and neuropathy. Prostaglandins Leukot.

    Essent. FattyAcids (2002) 66 243256.

    182 Blake D.R., Robson P., Ho M., Jubb R.W., McCabe C.S.

    Preliminary assessment of the efficacy, tolerability and safety

    of a cannabis-based medicine (Sativex) in the treatment of

    pain caused by rheumatoid arthritis. Rheumatology (Oxford)

    (2006) 45 5052.

    183 Ashton J.C., Milligan E.D. Cannabinoids for the treatment of

    neuropathic pain: clinical evidence. Curr. Opin. Investig.

    Drugs (2008) 9 6575.

    184 Wallace M., Schulteis G., Atkinson J.H. et al. Dose-

    dependent effects of smoked cannabis on capsaicin-induced

    pain and hyperalgesia in healthy volunteers. Anesthesiology

    (2007) 107 785796.

    185 Johnson J.R., Burnell-Nugent M., Lossignol D., Ganae-Motan

    E.D., Potts R., Fallon M.T. Multicenter, double-blind,

    randomized, placebo-controlled, parallel-group study of the

    efficacy, safety and tolerability of THC:CBD extract and THC

    extract in patients with intractable cancer-related pain. J.

    Pain. Symptom. Manag. (2010) 39 167179.

    186 Abrams D.I., Jay C.A., Shade S.B. et al. Cannabis in painful

    HIV-associated sensory neuropathy. Neurology (2007) 68

    515521.

    187 Pertwee R.G., Ross R.A. Cannabinoid receptors and theirligands. Prostaglandins Leukot. Essent. Fatty Acids (2002)

    66 101121.

    188 Pollmann W., Feneberg W. Current management of pain

    associated with multiple sclerosis. CNS Drugs (2008) 22

    291324.

    189 Benito C., Nunez E., Tolon R.M. et al. Cannabinoid CB2

    receptors and fatty acid amide hydrolase are selectively

    overexpressed in neuritic plaque-associated glia in Alzheimers

    disease brains. J. Neurosci. (2003) 23 1113611141.

    190 Maccarrone M., Gubellini P., Bari M. et al. Levodopa

    treatment reverses endocannabinoid system abnormalities

    in exper