Skip to main content

Interplay between endocannabinoids and dopamine in the basal ganglia: implications for pain in Parkinson’s disease

Abstract

Pain is a complex phenomenon, and basal ganglia circuitry integrates many aspects of pain including motor, emotional, autonomic, and cognitive responses. Perturbations in dopamine (DA) signaling are implicated in the pathogenesis of chronic pain due to its involvement in both pain perception and relief. Several lines of evidence support the role of endocannabinoids (eCBs) in the regulation of many electrical and chemical aspects of DAergic neuron function including excitability, synaptic transmission, integration, and plasticity. However, eCBs play an even more intricate and intimate relationship with DA, as indicated by the adaptive changes in the eCB system following DA depletion. Although the precise mechanisms underlying DA control on pain are not fully understood, given the high correlation of eCB and DAergic system, it is conceivable that eCBs may be part of these mechanisms.

In this brief survey, we describe the reciprocal regulation of eCB-DA neurotransmission with a particular emphasis on the actions of eCBs on ionic and synaptic signaling in DAergic neurons mediated by CB receptors or independent on them. Furthermore, we analyze the eCB-DA imbalance which characterizes pain condition and report the implications of reduced DA levels for pain in Parkinson’s disease. Lastly, we discuss the potential of the eCB-DA system in the development of future therapeutic strategies for the treatment of pain.

Introduction

The identification of the cannabinoids receptors (CB-Rs) in the early 1990s [1, 2] suggested that they serve not merely as binding sites for psychotropic substances extracted from Cannabis sativa plant flowers (cannabinoids) but rather function as mediators of endogenous signaling molecules, physiologically operating as messengers in the mammalian central nervous system (CNS). This discovery prompted the search for endogenous agonists, or endocannabinoids (eCBs), which mimic the psychomotor effects of cannabinoids [3].

Since then, the eCB system has been demonstrated to play important physiological roles in the control of emotional responses, cognition, feeding, pain, motivated behaviors, and motor control. However, the precise mechanisms employed by eCBs to mediate their effects remain partially understood, due to the multiplicity and complexity of their actions. In fact, both CB1 and CB2 are G protein-coupled receptors (GPCRs), which influence the biochemical and electrical state of a cell through complex mechanisms involving kinases, phosphatases, transcription factors, and ion channels. Moreover, the interaction with other systems of neurotransmitters and the modulation of their activity can induce changes in synaptic transmission, promoting long-term changes in synapse efficacy and modifying the structural organization of neuronal circuitries.

CB-Rs are widely distributed throughout the CNS. Of note, their robust expression within the basal ganglia circuitry has stimulated investigation on the possible interplay between eCBs and dopamine (DA) system, not only in the context of movement disorders but also in pain research. Several preclinical and clinical studies have demonstrated that eCBs are crucial modulators of DAergic transmission [4], and that DA agonists may have an antinociceptive effect [5]. Conversely, DA depletion leads to enhanced nociceptive responses [6,7,8,9,10,11] and induces adaptive changes in the eCB system [12,13,14,15]. Accordingly, Parkinson’s disease (PD) patients report comorbid pain conditions and higher pain rating [16, 17].

Despite such evidence, the specific mechanisms through which intracellular effectors recruited by eCBs directly or indirectly interact with DA-dependent signaling effectors to affect pain perception have been only partially identified. However, a review of all the potential cellular and molecular mechanisms is beyond the scope of this paper, and we refer the reader to a number of excellent comprehensive reviews (see [18,19,20,21,22]). Our brief survey will rather focus on the available evidence on the interactions between eCBs and DA. In PD, DA depletion provides an ideal clue to review and discuss the compensatory changes of the eCB system, as a potential mechanism contributing to pain in PD. Advancing our knowledge on these basic mechanisms might reveal potential molecular targets for drug development in PD-related pain.

Endocannabinoid-dopamine crosstalk

eCB and CB-Rs in the basal ganglia

The presence of eCB-Rs within the basal ganglia circuitry has been demonstrated by several neuroanatomical studies. The expression of the CB1-R has been found in the glutamatergic corticostriatal afferences in both dorsal and ventral striatum [23, 24] and the projections of striatal GABAergic neurons innervating the globus pallidus (GPi and GPe) and the substantia nigra pars reticulata (SNpr) [25, 26]. Some studies have also confirmed the distribution of CB1-R in the glutamatergic terminals innervating the subthalamic nucleus (STN) and the GPi/SNpr [27] (Fig. 1).

Fig. 1
figure 1

Distribution of CB-Rs within the basal ganglia circuitry and eCB/CB-dependent modulation of DA release via direct and indirect mechanisms. eCBs can control both excitatory and inhibitory neurotransmitter release and indirectly modulate axonal and somatodendritic DA release. A direct action of eCBs on DA axons and cell bodies occurs following binding to CB2-R or K+ channels which results in inhibition or increase in DA release

Also, CB2-R was found in the striatum, GP, DAergic neurons of the ventral tegmental area (VTA) and SN, and in the basal thalamus [28,29,30,31,32] (Fig. 1).

In addition to the classical CB-Rs, other putative eCB receptors exist. Among these, the transient receptor potential vanilloid type-1 (TRPV1) channel can be activated by several exogenous and endogenous stimuli. TRPV-1 has been identified in both the axonal terminals and the soma of nigrostriatal DAergic neurons [33,34,35] (Fig. 1).

CB-Rs and the putative receptors are activated by eicosanoids such as N-arachidonoyl-ethanolamide (AEA) or anandamide, and 2-arachidonoyl-glicerol (2-AG), and by cannabinoids and their analogues.

As GPCRs, both CB1-R and CB2-R activate heterotrimeric G proteins and use similar transduction systems. CB1-R is coupled to Gαi and Gαo proteins which inhibit protein kinase A (PKA). PKA mediates most of the effects of CB1-R by phosphorylating and regulating the function of a wide array of cellular substrates such as voltage-gated K+, Na+, and Ca2+ channels (Fig. 2), ionotropic glutamate, and GABA receptors and transcription factors. Noteworthy, the modulation of ion channels following CB1-R activation can either depend on its coupling with Gαolf protein or be independent of cAMP/PKA signaling. A good example is the gating of G protein-coupled inwardly rectifying K+ channels (GIRKs) at presynaptic level in the nucleus accumbens (NAc) [36]. Also, CB1-R can modulate intracellular Ca2+ levels and regulate ligand- and voltage-gated ion channels through the liberation of the Gβγ subunit upon receptor activation.

Fig. 2
figure 2

Schematic representation of eCB- and CB-mediated changes in neuronal excitability via action at ion channel level. The release of eCBs causes activation or inhibition of several classes of ion channels. In some cases, the modulation of the ion channels by eCBs (or CBs) is consequent to the activation of CB-Rs, whereas in others, the action of eCBs on these channels is direct or occurs independently of known CB-Rs

CB1-R is mainly expressed in presynaptic terminals in homo- and heterodimeric structures; however, it can also be present at postsynaptic level where it can form heterodimers with other GPCRs including DA receptors [37,38,39]. Depending on the DA-R bound, CB1-R can contribute in different ways to the modulation of both excitatory and inhibitory synaptic functions as well as to the decrease of neurotransmitter release in the basal ganglia circuitry (for rev., see [40]).

Similar to CB1-R, CB2-R is coupled to Gαi and Gαo proteins and its stimulation results in PKA inhibition. Although its role in the CNS is still controversial, some studies have shown its involvement in modulation of ion channels and DA levels [41,42,43,44].

CB1-Rs and CB2-Rs can also activate other intracellular signaling cascades, including the mitogen-activated protein kinase (MAPK) cascade, the phosphatidylinositol 3-kinase (PI3K) cascade, and nitric oxide production [45].

TRPV1 channels are characterized by weak voltage sensitivity and nonselective permeability to monovalent and divalent cations including Mg2+, Ca2+, and Na+. Their activation has been shown to modulate synaptic transmission and neurotransmitter release [46,47,48].

Dual regulation of DA/eCB neurotransmission

DA neurons are fine-tuned by eCBs, which can regulate DA neurotransmission at midbrain cell bodies, at DA axon terminal endings or by acting on DA neuron effector sites in the striatum (Fig. 1).

The modulation of DA release by eCBs in midbrain neurons occurs via different mechanisms, according to the target receptor activated.

DA neurons in midbrain do not express CB1-Rs [49]; however, many other neuronal populations connected with DAergic cells present CB1-R and are modulated by eCBs. Thus, eCBs indirectly control DA cells through modulation of local circuitry, facilitating or suppressing DAergic neuron activity [40, 50, 51], primarily through 2-AG (Fig. 1).

In contrast to CB1-Rs which are not expressed by midbrain DAergic neurons, CB2-Rs are present on cell bodies [29], and their activation by exogenous agonists inhibits DAergic neuronal firing [52, 53] (Fig. 1); however, whether endogenous molecules driving CB2-R signaling act in a similar way to reduce DA release has not been established yet.

In the SN, an increase in the excitatory neurotransmission through a tonic modulation of the glutamatergic synaptic inputs onto DAergic neurons by the eCB AEA has been measured. Such enhancement occurs after activation of TRPV1, and the effect of AEA on DAergic transmission results in an increase in DA levels [46, 47] (Fig. 1). Of note, the modulation of DA release by AEA occurs also in response to peripheral noxious stimulation in freely moving animals [47]. While TRPV1 channels have been found also on DAergic cells, it is unclear whether their activation has a direct action at postsynaptic level.

At striatal level, DA neurotransmission is modulated locally at presynaptic axonal release sites by several afferent inputs (i.e., cholinergic and glutamatergic inputs). In such context, also eCBs participate in the local modulation of axonal DA release. Specifically, it has been observed that CB1-R activation inhibits the evoked release of DA [54] (Fig. 1). The exact cellular localization of CB1-Rs responsible for such inhibition is not clear yet; however, polysynaptic and indirect mechanisms have been implicated in the decrease of DA levels.

Interestingly, also CB2-R activation on DA terminals seems responsible for causing inhibition of striatal DA release [55]. This phenomenon has been observed following exogenous activation; whether endogenous agonists do the same influencing DA tone and DA-dependent behavior is not established yet (Fig. 1).

Finally, the localization of TRPV1 channels at striatal level [34] implicates a possible involvement of these receptors in the modulation of the DA tone. These channels have already been demonstrated to be involved in the control of GABA synapses and in the regulation of excitatory transmission [48, 56], thus, it is plausible that their activation indirectly may impact DAergic transmission.

The regulation of DA neurotransmission by eCBs at striatal sites occurs also at the level of the regulatory mechanisms that control the concentration of DA at synaptic cleft. Specifically, AEA increases extracellular DA levels [57] via inhibition of DA transporter (DAT) activity [58] (Fig. 1). The inhibition of DA uptake by eCBs and other cannabinoids has been observed in native tissue but also in heterologous expression systems [59,60,61]. Collectively, these data clearly show that eCBs control DA neurotransmission at multiple sites of action within the basal ganglia.

The mutual interaction between eCBs and DA is not only dependent on the ability of eCBs to modulate DA release but also vice versa. Indeed, DAergic neurons can themselves produce eCBs from their somata and dendrites [62], thereby modulating the retrograde signaling. The release of eCBs from DAergic neurons is consequent to an increase in the firing of DAergic cells which causes the mobilization of 2-AG (Fig. 1). Moreover, it has been observed that DA, following binding to D2-R, can promote the postsynaptic release of eCBs from striatal spiny projection neurons (SPNs) which, in turn, influence transmitter exocytosis through the activation of presynaptic GPCRs [63].

In general, the stimulation of D2-Rs by DA causes increases of striatal eCB levels due to an increased synthesis and an inhibited degradation [64, 65]. Interestingly, D2-R and CB1-R are co-expressed, and CB1-R activation by AEA can enhance the effects of D2-R activation [66, 67], thereby amplifying e-CB production.

eCB action independent of CB-Rs: focus on ion channels

Endogenous cannabinoids, as well as exogenous and synthetic congeners, have been shown to target other molecular substrates in addition to the classical CB-Rs. In particular, these molecules can modify the gating of several classes of ion channels and affect neuronal excitability and neurotransmitter release.

In fact, both eCBs and cannabidiol (CBD), a phytocannabinoid extracted from cannabis sativa, are able to interact with voltage-gated sodium channels (Nav). Specifically, 2-AG is able to induce channel block and inhibit Na+ currents or induce changes in biophysical properties of activation/inactivation of Nav channels, thereby affecting their function [68] (Fig. 2). Similarly, CBD has inhibitory effects on Na+ conductance without specificity to any of Nav subfamily [69,70,71,72,73] and can directly interact with Nav channels to inhibit their activity [74, 75] (Fig. 2). Interestingly, CBD binds the channel in more than a single site, and the binding sites are not positioned to physically occlude the pore. Moreover, the inhibition is allosteric and determines a stabilization of the inactivated state.

In addition to Nav channels, cannabinoids have been reported to interact also with K+ channels decreasing the peak currents [76]. In this regard, it has been shown that the inhibition of native A-type K+ channels by 2-AG in DAergic neurons causes an increase in their excitability, inducing a transition from tonic firing to burst discharge and thus affecting DA release [77] (Fig. 2).

With these results in mind, and considering the close relationship between cannabinoids and DAergic system, it can be speculated that CBD and eCBs could exert their analgesic effects, at least in part, through a direct action on ion channels whose modulation causes changes in neuronal excitability and in DA release.

Endocannabinoid-DA interaction: pathological imbalance and implications for pain

eCB-DA imbalance in PD patients

Clinical studies support the evidence for a reciprocal interplay between DA and eCBs. In individuals with PD, the levels of eCBs measured in the cerebrospinal fluid (CSF) were significantly higher as compared to control subjects. This finding was observed either in drug-naive, recently diagnosed subjects, or in patients with a longer disease duration, undergoing drug washout [15]. Of interest, eCBs levels did not differ in these two subgroups of patients, indicating that elevations of CSF eCBs are independent of disease duration. However, in patients undergoing DAergic treatment, a normal eCB content was measured, not different from controls, suggesting that an adaptive, compensatory mechanism occurs during DA denervation. Evidence for a similar, adaptive change emerges from binding studies performed in post-mortem samples from PD patients. Measurement of CB1-R binding and its coupling to G proteins revealed higher stimulation by CB1 receptor agonists of [35S] GTPcS binding both in the caudate and putamen, along with an enhanced CB1 receptor binding [78]. Overall, the reported changes in eCB levels with or without DA replacement suggest that the DA/eCB interplay is highly dynamic.

The general view of this dual interaction is further confirmed by additional clinical hints and supported by preclinical findings (see below). Indeed, while DA supplementation is known to improve dramatically the motor performance observed in hypokinetic disorders, such as PD, the therapeutic use of CBs has been approved for hyperkinetic disorders, such as Tourette’s syndrome and other tic disorders [79, 80], further supporting the notion of a dual interplay between DA and eCB system.

eCB-DA imbalance in models of PD

The close interaction between DA and eCBs is clearly evident not only in PD patients but also in experimental PD models. In fact, in multiple toxin-induced rodent and nonhuman primate PD models, as well as in genetic rodent PD models, eCB system is reorganized with changes measurable at level of concentration, clearance, and activity.

In particular, in 6-OHDA-denervated rats and in reserpine-treated rats, enhanced levels of eCBs have been measured in the basal ganglia circuitry [12, 81]. However, the observed enhancement is a consequence of a decreased cleavage and not dependent on an increased synthesis [12]. In line with rodent data, both AEA and 2-AG levels were increased in the caudate of non-human primates treated with MPTP [82].

In contrast to toxin-induced PD models, eCB levels were unchanged in a genetic mouse model of PD carrying the recessive mutation in the PTEN-induced putative kinase 1 (PINK1) gene responsible for early-onset PD [83]. However, in spite of unaltered eCB levels, the synaptic responses to CB1-R activation were impaired, and a significant decrease of binding capacity of CB1R agonists was found in PINK1 knockout mice, again supporting the notion of a mutual interplay between DA and eCBs.

Although some differences exist among the various PD experimental models regarding the level of eCBs altered following DA depletion, the changes observed in the eCB system were in all the models reverted after DAergic therapy with levodopa or a D2-R agonist [13, 15, 82, 83] suggesting that the upregulation of the eCB system consequent to DA depletion is an adaptive, dynamic homeostatic mechanism aimed at compensating the effects of DA loss in the basal ganglia.

Such interplay is further corroborated by a number of preclinical findings. CB-R agonists potentiated reserpine-induced hypokinesia [84] and DA receptor antagonist-induced catalepsy [85]. Conversely, they were able to reduce quinpirole-induced hyperlocomotion [86] and amphetamine-induced hyperactivity (for rev., see [87]).

Effects of soluble products of inflammation on eCB-DA signaling in PD

Neuroinflammation is a well-established feature of PD, and a growing body of evidence suggests that both peripheral and central immune dysregulation can upregulate cytokines such as IL-1β, IL-6, and TNF-α [88], which participate in the development and persistence of pain states during PD. The increased levels of these cytokines in PD patients’ serum and CSF can affect different neural networks and influence synaptic transmission with significant changes in eCB-DA signaling as well as behavioral implications. Intriguingly, a modulatory effect on both excitatory and inhibitory neurotransmissions at striatal level has been reported for IL-1β with involvement of eCB and DAergic systems. As a matter of fact, the exposure to this cytokine causes enhancement of spontaneous excitatory currents via the activation of presynaptic TRPV1 [89] and a reduced sensitivity of CB1-R [90]. In addition, IL-1β reduces GABAergic inhibition inducing SPN hyperactivation [91]. Analogously, TRPV1 and CB1-R seem to be implicated in such inhibitory modulation [92, 93]. The striatal excitatory/inhibitory imbalance induced by IL-1β is coupled to an alteration of DAergic transmission as suggested by the finding that evoked axonal DA release is reduced when IL-1β is upregulated and restored following administration of IL-1 receptor antagonist (IL-1ra) [94]. Moreover, IL-1ra application also rescues striatal CB1-R sensitivity and DA-dependent behavior [95]. Although not exhaustive, these observations clearly confirm the strong connection between eCB and DAergic system and their sensitivity to inflammatory mediators suggesting a contribution for these molecules in causing the eCB-DA imbalance. In addition to IL-1β, other pro-inflammatory cytokines have been proven to affect DA neurotransmission [96]; however, whether this impairment also exerts influence on eCB system and has implications for pain process is still unknown and deserves to be investigated.

eCB-DA imbalance during pain

The role of DA in pain can be better appreciated in conditions of DA loss such as in PD. Indeed, compelling evidence indicates that up to 80% of individuals with PD manifest pain during the disease course [17]. The degeneration of nigrostriatal DA pathway in PD patients causes hyperalgesia [97], and the administration of levodopa induces an increase in the pain threshold [98, 99]. Intriguingly, toxin-induced rodent models of PD (i.e., MPTP and 6-OHDA) have provided a valuable contribution in understanding the role of DA in pain process. Similar to humans, DA-depleted animals show an increased nociception and decreased nociceptive threshold [10, 100,101,102,103,104] suggesting that the loss of DA in the basal ganglia is involved in the reduction of pain threshold [105]. Of note, the elevation of striatal DA following electrical stimulation of the SN or the direct striatal administration of apomorphine, a D1/D2-R agonist, induces pain inhibition [106]. Similarly, systemic administration of DA agonists determines hypoalgesia by modulating D2 receptors in dorsolateral striatum [107]. In spite of such evidence, both in humans and in models of PD, replacement with DA therapy does not promote recovery from pain, thereby suggesting that other neurotransmitters may play a role in the pain signaling process.

Although the great therapeutic potential of eCB system in the modulation of pain has been widely recognized [108, 109], the involvement of DA/eCB system within basal ganglia circuitry in chronic pain has been less extensively explored [110]. eCBs are elevated at various sites in nociceptive pathways in chronic pain [19] highlighting their role as endogenous analgesics. Based on the existence of a concurrent decrease in DA concentrations observed during pain, the increase in eCB levels might compensate DA decrease. The imbalance in eCB-DA system during pain condition is somehow reminiscent of the alteration observed during PD. However, eCB effects on nociception and pain following DA depletion have been scarcely investigated. Also, whether DAergic drugs are able to restore resting levels of eCBs as in animal models of PD [13] needs to be clarified.

Recently, Nascimento and colleagues [111] analyzed the effect of CBD on nociception in experimental PD, highlighting the ability of this drug to exert antinociception via activation of CB1-R. Moreover, the authors suggest that the analgesic effect induced by CBD is also dependent on an increase in AEA which likely potentiates the activation of CB1. An elevation in AEA levels has been found in 6-OHDA-denervated animals and is dependent on a decreased cleavage [12]; thus, it is plausible that this drug may cause an increase in DA levels acting at the same sites used by eCBs to directly modulate somatodendritic or axonal DA release in the basal ganglia.

Conclusions

Although the role of DA in pain perception in PD has not been fully elucidated, it is now evident that an altered DA neurotransmission correlates with impaired eCB levels. Hence, the eCB system would represent, in principle, a promising target for the treatment of pain in PD. Additional work is needed to clarify the functional meaning of these interactions, in order to gain a better understanding of the mechanisms through which eCBs affect DA neurotransmission, which may translate into novel opportunities for improving pain treatment.

Availability of data and materials

No datasets were generated or analysed during the current study.

References

  1. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI (1990) Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346(6284):561–564. https://doi.org/10.1038/346561a0

    Article  CAS  PubMed  Google Scholar 

  2. Munro S, Thomas KL, Abu-Shaar M (1993) Molecular characterization of a peripheral receptor for cannabinoids. Nature 365(6441):61–65. https://doi.org/10.1038/365061a0

    Article  CAS  PubMed  Google Scholar 

  3. Di Marzo V, Fontana A, Cadas H, Schinelli S, Cimino G, Schwartz JC, Piomelli D (1994) Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 372(6507):686–691. https://doi.org/10.1038/372686a0

    Article  PubMed  Google Scholar 

  4. Melis M, Pistis M (2007) Endocannabinoid signaling in midbrain dopamine neurons: more than physiology? Curr Neuropharmacol 5(4):268–277. https://doi.org/10.2174/157015907782793612

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dennis SG, Melzack R (1983) Effects of cholinergic and dopaminergic agents on pain and morphine analgesia measured by three pain tests. Exp Neurol 81(1):167–176. https://doi.org/10.1016/0014-4886(83)90166-8

    Article  CAS  PubMed  Google Scholar 

  6. Saadé NE, Atweh SF, Bahuth NB, Jabbur SJ (1997) Augmentation of nociceptive reflexes and chronic deafferentation pain by chemical lesions of either dopaminergic terminals or midbrain dopaminergic neurons. Brain Res 751(1):1–12. https://doi.org/10.1016/s0006-8993(96)01164-x

    Article  PubMed  Google Scholar 

  7. Dieb W, Ouachikh O, Durif F, Hafidi A (2014) Lesion of the dopaminergic nigrostriatal pathway induces trigeminal dynamic mechanical allodynia. Brain Behav 4(3):368–380. https://doi.org/10.1002/brb3.214

    Article  PubMed  PubMed Central  Google Scholar 

  8. Ogata M, Noda K, Akita H, Ishibashi H (2015) Characterization of nociceptive response to chemical, mechanical, and thermal stimuli in adolescent rats with neonatal dopamine depletion. Neuroscience 289:43–55. https://doi.org/10.1016/j.neuroscience.2015.01.002

    Article  CAS  PubMed  Google Scholar 

  9. Taylor AMW, Beccker S, Schweinhardt P, Cahill C (2016) Mesolimbic dopamine signaling in acute and chronic pain: implications for motivation, analgesia, and addiction. Pain 157(6):1194–1198. https://doi.org/10.1097/j.pain.0000000000000494

    Article  PubMed  PubMed Central  Google Scholar 

  10. Charles KA, Naudet F, Bouali-Banazzouz R, Landry M, De Deurwaerdère P, Fossat P, Banazzouz A (2018) Alteration of nociceptive integration in the spinal cord of a rat model of Parkinson’s disease. Mov Disord 33(6):1010–1015. https://doi.org/10.1002/mds.27377

    Article  CAS  PubMed  Google Scholar 

  11. Ziółkowska B (2021) Corticostriatal glutamatergic transmission in chronic pain. Brain Sci 11(10):1311. https://doi.org/10.3390/brainsci11101311

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gubellini P, Picconi B, Bari M, Battista N, Calabresi P, Centonze D, Bernardi G, Finazzi-Agrò A, Maccarone M (2002) Experimental parkinsonism alters endocannabinoid degradation: implications for striatal glutamatergic transmission. J Neurosci 22(16):6900–6907. https://doi.org/10.1523/JNEUROSCI.22-16-06900.2002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Maccarrone M, Gubellini P, Bari M, Picconi B, Battista N, Centonze D, Bernardi G, Finazzi-Agrò A, Calabresi P (2003) Levodopa treatment reverses endocannabinod system abnormalities in experimental parkinsonism. J Neurochem 85(4):1018–1025. https://doi.org/10.1046/j.1459.2003.01759.x

    Article  CAS  PubMed  Google Scholar 

  14. Pisani A, Fezza F, Galati S, Battista N, Napolitano S, Finazzi-Agrò A, Bernardi G, Brusa L, Pirantozzi M, Stanzione P, Maccarrone M (2005) High endogenous cannabinoid levels in the cerebrospinal fluid of untreated Parkinson’s disease patients. Ann Neurol 57(5):777–779. https://doi.org/10.1002/ana.20462

    Article  PubMed  Google Scholar 

  15. Pisani V, Madeo G, Tassone A, Sciamanna G, Maccarrone M, Stanzione P, Pisani A (2011) Homeostatic changes of the endocannabinoid system in Parkinson’s disease. Mov Disord 26(2):216–222. https://doi.org/10.1002/mds.23457

    Article  PubMed  Google Scholar 

  16. Nebe A, Ebersbach G (2009) Pain intensity on and off levodopa in patients with Parkinson’s disease. Mov Disord 24(8):1233–1237. https://doi.org/10.1002/mds.22546

    Article  PubMed  Google Scholar 

  17. Ghosh P, Imbriani P, Caputi N, Natoli S, Schirinzi T, Di Lazzaro G, Covington L, Sparks AD, Salnikova Y, Rukaniva K, Chaudhuri KR, Pisani A (2020) A dual centre study of pain in Parkinson’s disease and its relationship with other non-motor symptoms. J Parkinsons Dis 10(4):1817–1825. https://doi.org/10.3233/JPD-202088

    Article  PubMed  Google Scholar 

  18. Maldonado R, Baños JE, Cabañero D (2016) The endocannabinoid system and neuropathic pain. Pain 157(Suppl 1):S23–S32. https://doi.org/10.1097/j.pain.0000000000000428

    Article  CAS  PubMed  Google Scholar 

  19. Woodhams SG, Chapman V, Finn DP, Hohmann AG, Neugebauer V (2017) The cannabinoid system and pain. Neuropharmacology 124:105–120. https://doi.org/10.1016/j.neuropharm.2017.06.015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Donvito G, Nass SR, Wilkerson JL, Curry ZA, Schurman LD, Kinsey SG, Lichtman AH (2018) The endogenous cannabinoid system: a budding source of targets for treating inflammatory and neuropathic pain. Neuropsychopharmacology 43(1):52–79. https://doi.org/10.1038/npp.2017.204

    Article  CAS  PubMed  Google Scholar 

  21. Cristino L, Bisogno T, Di Marzo V (2020) Cannabinoids and the expanded endocannabinoids system in neurological disorders. Nat Rev Neurol 16(1):9–29. https://doi.org/10.1038/s41582-019-0284

    Article  PubMed  Google Scholar 

  22. Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, Rice ASC (2021) Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Pain 162(Suppl 1):S5–S25. https://doi.org/10.1097/j.pain.0000000000002268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gerdeman G, Lovinger DM (2001) CB1 cannabinoid receptor inhibits synaptic release of glutamate in rat dorsolateral striatum. J Neurophysiol 85(1):468–471. https://doi.org/10.1152/jn.2001.85.1.468

    Article  CAS  PubMed  Google Scholar 

  24. Köfalvi A, Rodrigues RJ, Ledent C, Mackie K, Sylvester Vizi E, Cunha RA, Sperlágh B (2005) Involvement of cannabinoid receptors in the regulation of neurotransmitter release in the rodent striatum: a combined immunochemical and pharmacological analysis. J Neurosci 25(11):2874–2884. https://doi.org/10.1523/JNEUROSCI.4232-04.2005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Herkenham M, Lynn AB, de Costa BR, Richfield EK (1991) Neuronal localization of cannabinoid receptors in the basal ganglia of the rat. Brain Res 547(2):267–274. https://doi.org/10.1016/0006-8993(91)90970-7

    Article  CAS  PubMed  Google Scholar 

  26. Tsou K, Brown S, Sañudo-Peña MC, Mackie K, Walker JM (1998) Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience 83(2):393–411. https://doi.org/10.1016/s0306-4522(97)00436-3

    Article  CAS  PubMed  Google Scholar 

  27. Sañudo-Peña MC, Walker JM (1997) Role of the subthalamic nucleus in cannabinoid actions in the substantia nigra of the rat. J Neurophysiol 77(3):1635–1638. https://doi.org/10.1152/jn.1997.77.3.1635

    Article  PubMed  Google Scholar 

  28. Sierra S, Luquin N, Rico AJ, Gómez-Bautista V, Roda E, Dopeso-Reyes IG, Vázquez A, Martínez-Pinilla E, Labandeira-García JL, Franco R, Lanciego JL (2015) Detection of cannabinoid receptors CB1 and CB2 within basal ganglia output neurons in macaques: changes following experimental parkinsonism. Brain Struct Funct 220(5):2721–2738. https://doi.org/10.1007/s00429-014-0823-8

    Article  CAS  PubMed  Google Scholar 

  29. Zhang HY, Bi GH, Li X, Li J, Qu H, Zhang SJ, Li CY, Onaivi ES, Gardner EL, Xi ZX, Liu QR (2015) Species differences in cannabinoid receptor 2 and receptor responses to cocaine self-administration in mice and rats. Neuropsychopharmacology 40(4):1037–1051. https://doi.org/10.1038/npp.2014.297

    Article  CAS  PubMed  Google Scholar 

  30. Sánchez-Zavaleta R, Cortés H, Avalos-Fuentes JA, García U, Segovia Vila J, Erlij D, Florán B (2018) Presynaptic cannabinoid CB2 receptors modulate [3 H]-glutamate release at subthalamo-nigral terminals of the rat. Synapse 72(11):e22061. https://doi.org/10.1002/syn.22061

    Article  CAS  PubMed  Google Scholar 

  31. Canseco-Alba A, Schanz N, Sanabria B, Zhao J, Lin Z, Liu QR, Onaivi ES (2019) Behavioral effects of psychostimulants in mutant mice with cell-type specific deletion of CB2 cannabinoid receptors in dopamine neurons. Behav Brain Res 360:286–297. https://doi.org/10.1016/j.bbr.2018.11.043

    Article  CAS  PubMed  Google Scholar 

  32. Jordan CJ, Xi ZX (2019) Progress in brain cannabinoid CB2 receptor research: from genes to behavior. Neurosci Biobehav Rev 98:208–220. https://doi.org/10.1016/j.neubiorev.2018.12.026

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mezey E, Tóth ZE, Cortright DN, Arzubi MK, Krause JE, Elde R, Guo A, Blumberg PM, Szallasi A (2000) Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci U S A 97(7):3655–3660. https://doi.org/10.1073/pnas.97.7.3655

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cristino L, de Petrocellis L, Pryce G, Baker D, Guglielmotti V, Di Marzo V (2006) Immunohistochemical localization of cannabinoid type 1 and vanilloid transient receptor potential vanilloid type 1 receptors in the mouse brain. Neuroscience 139(4):1405–1415. https://doi.org/10.1016/j.neuroscience.2006.02.074

    Article  CAS  PubMed  Google Scholar 

  35. Marinelli S, Di Marzo V, Florenzano F, Fezza F, Viscomi MT, van der Stelt M, Bernardi G, Molinari M, Maccarrone M, Mercuri NB (2007) N-arachidonoyl-dopamine tunes synaptic transmission onto dopaminergic neurons by activating both cannabinoid and vanilloid receptors. Neuropsychopharmacology 32(2):298–308. https://doi.org/10.1038/sj.npp.1301118

    Article  CAS  PubMed  Google Scholar 

  36. Robbe D, Alonso G, Duchamp F, Bockaert J, Manzoni OJ (2001) Localization and mechanisms of action of cannabinoid receptors at the glutamatergic synapses of the mouse nucleus accumbens. J Neurosci 21(1):109–116. https://doi.org/10.1523/JNEUROSCI.21-01-00109.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Przybyla JA, Watts VJ (2010) Ligand-induced regulation and localization of cannabinoid CB1 and dopamine D2L receptor heterodimers. J Pharmacol Exp Ther 332(3):710–719. https://doi.org/10.1124/jpet.109.162701

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Khan SS, Lee FJS (2014) Delineation of domains within the cannabinoid CB1 and dopamine D2 receptors that mediate the formation of the heterodimer complex. J Mol Neurosci 53(1):10–21. https://doi.org/10.1007/s12031-013-0181-7

    Article  CAS  PubMed  Google Scholar 

  39. Bagher AM, Laprairie RB, Kelly ME, Denovan-Wright EM (2016) Antagonism of dopamine receptor 2 long affects cannabinoid receptor 1 signaling in a cell culture model of striatal medium spiny projection neuros. Mol Pharmacol 89(6):652–666. https://doi.org/10.1124/mol.116.103465

    Article  CAS  PubMed  Google Scholar 

  40. Garcia C, Palomo-Garo C, Gómez-Gálvez Y, Fernández-Ruiz J (2016) Cannabinoid-dopamine interactions in physiology and physiopathology of the basal ganglia. Br J Pharmacol 173(13):2069–2079

    Article  CAS  PubMed  Google Scholar 

  41. Demuth DG, Molleman A (2006) Cannabinoid signaling. Life Sci 78(6):549–563. https://doi.org/10.1016/j.lfs.2005.05.055

    Article  CAS  PubMed  Google Scholar 

  42. Xi ZX, Peng XQ, Li X, Song R, Zhang HY, Liu QR, Yang HJ, Bi GH, Li J, Gardner EL (2011) Brain cannabinoid CB2 receptors modulate cocaine’s actions in mice. Nat Neurosci 14(9):1160–1166. https://doi.org/10.1038/nn.2874

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ma Z, Gao F, Larsen B, Gao M, Luo Z, Chen D, Ma X, Qiu S, Zhou Y, Xie J, Xi ZX, Wu J (2019) Mechanisms of cannabinoid CB2 receptor-mediated reduction of dopamine neuronal excitability in mouse ventral tegmental area. EBioMedicine 42:225–237. https://doi.org/10.1016/j.ebiom.2019.03.040

    Article  PubMed  PubMed Central  Google Scholar 

  44. Galaj E, Bi GH, Yang HJ, Xi ZX (2020) Cannabidiol attenuates the rewarding effects of cocaine in rats by CB2, 5-HT1A and TRPV1 receptor mechanisms. Neuropharmacology 167:107740. https://doi.org/10.1016/j.neuropharm.2019.107740

    Article  CAS  PubMed  Google Scholar 

  45. Tadijan A, Vlašić I, Vlainić J, Dikić D, Oršlić N, Jazvinšćak Jembrek M (2022) Intracellular molecular targets and signaling pathways involved in antioxidative and neuroprotective effects of cannabinoids in neurodegenerative conditions. Antioxidant (Basel) 11(10):2049. https://doi.org/10.3390/antiox11102049

    Article  CAS  Google Scholar 

  46. Marinelli S, Di Marzo V, Berretta N, Matias I, Maccarrone M, Bernardi G, Mercuri NB (2003) Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J Neurosci 23(8):3136–3144. https://doi.org/10.1523/JNEUROSCI.23-08-03136.2003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Marinelli S, Pascucci T, Bernardi G, Puglisi-Allegra S, Mercuri NB (2005) Activation of TRPV1 in the VTA excites dopaminergic neurons and increases chemical- and noxious-induced dopamine release in the nucleus accumbens. Neuropsychopharmacology 30(5):864–870. https://doi.org/10.1038/sj.npp.1300615

    Article  CAS  PubMed  Google Scholar 

  48. Maccarrone M, Rossi S, Bari M, De Chiara V, Fezza F, Musella A, Gasperi V, Prosperetti C, Bernardi G, Finazzi-Agrò A, Cravatt BF, Centonze D (2008) Anandamide inhibits metabolism and physiological actions of 2-arachidonoylglycerol in the striatum. Nat Neurosci 11(2):152–159. https://doi.org/10.1038/nn2042

    Article  CAS  PubMed  Google Scholar 

  49. Julian MD, Martin AB, Cuellar B, Rodriguez De Fonseca F, Navarro M, Moratalla R, Garcia-Segura LM (2003) Neuroanatomical relationship between type 1 cannabinoid receptors and dopaminergic systems in the rat basal ganglia. Neuroscience 119(1):309–318. https://doi.org/10.1016/s0306-4522(03)00070-8

    Article  CAS  PubMed  Google Scholar 

  50. Melis M, Perra S, Muntoni AL, Pillolla G, Lutz B, Marsicano G, Di Marzo V, Gessa GL, Pistis M (2004) Prefrontal cortex stimulation induces 2-arachidonoyl-glycerol-mediated suppression of excitation in dopamine neurons. J Neurosci 24(47):10707–10715. https://doi.org/10.1523/JNEUROSCI.3502-04.2004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Szabo B, Siemes S, Wallmichrath I (2002) Inhibition of GABAergic neurotransmission in the ventral tegmental area by cannabinoids. Eur J Neurosci 15(12):2057–2061. https://doi.org/10.1046/j.1460-9568.2002.02041.x

    Article  PubMed  Google Scholar 

  52. Zhang HY, Gao M, Liu QR, Bi GH, Li X, Yang HJ, Gardner EL, Wu J, Xi ZX (2014) Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc Natl Acad Sci U S A 111(46):E5007–E5015. https://doi.org/10.1073/pnas.1413210111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang HY, Gao M, Shen H, Bi GH, Yang HJ, Liu QR, Wu J, Gardner EL, Bonci A, Xi ZX (2017) Expression of functional cannabinoid CB2 receptor in VTA dopamine neurons in rats. Addict Biol 22(3):752–765. https://doi.org/10.1111/adb.12367

    Article  CAS  PubMed  Google Scholar 

  54. Sidló Z, Reggio PH, Rice ME (2008) Inhibition of striatal dopamine release by CB1 receptor activation requires nonsynaptic communication involving GABA, H2O2, and KATP channels. Neurochem Int 52(1–2):80–88. https://doi.org/10.1016/j.neuint.2007.07.014

    Article  CAS  PubMed  Google Scholar 

  55. Foster DJ, Wilson JM, Remke DH, Mahmood MS, Uddin MJ, Wess J, Patel S, Marnett LJ, Niswender CM, Jones CK, Xiang Z, Lindsley CW, Rook JM, Conn PJ (2016) Antipsychotic-like effects of M4 positive allosteric modulators are mediated by CB2 receptor-dependent inhibition of dopamine release. Neuron 91(6):1244–1252. https://doi.org/10.1016/j.neuron.2016.08.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Musella A, De Chiara V, Rossi S, Prosperetti C, Bernardi G, Maccarrone M, Centonze D (2009) TRPV1 channels facilitate glutamate transmission in the striatum. Mol Cell Neurosci 40(1):89–97. https://doi.org/10.1016/j.mcn.2008.09.001

    Article  CAS  PubMed  Google Scholar 

  57. Solinas M, Justinova Z, Goldberg SR, Tanda G (2006) Anandamide administration alone and after inhibition of fatty acid amide hydrolase (FAAH) increases dopamine levels in the nucleus accumbens shell in rats. J Neurochem 98(2):408–419. https://doi.org/10.1111/j.1471-4159.2006.03880.x

    Article  CAS  PubMed  Google Scholar 

  58. Oz M, Jaligam V, Galadari S, Petroianu G, Shuba YM, Shippenberg TS (2010) The endogenous cannabinoid, anandamide, inhibits dopamine transporter function by a receptor-independent mechanism. J Neurochem 112(6):1454–1464. https://doi.org/10.1111/j.1471-4159.2009.06557.x

    Article  CAS  PubMed  Google Scholar 

  59. Chen N, Appell M, Berfield JL, Reith ME (2003) Inhibition by arachidonic acid and other fatty acids of dopamine uptake at the human dopamine transporter. Eur J Pharmacol 478(2–3):89–95. https://doi.org/10.1016/j.ejphar.2003.08.045

    Article  CAS  PubMed  Google Scholar 

  60. Steffens M, Feuerstein TJ (2004) Receptor-independent depression of DA and 5-HT uptake by cannabinoids in rat neocortex–involvement of Na(+)/K(+)-ATPase. Neurochem Int 44(7):529–538. https://doi.org/10.1016/j.neuint.2003.08.009

    Article  CAS  PubMed  Google Scholar 

  61. Price DA, Owens WA, Gould GG, Frazer A, Roberts JL, Daws LC, Giuffrida A (2007) CB1-independent inhibition of dopamine transporter activity by cannabinoids in mouse dorsal striatum. J Neurochem 101(2):389–396. https://doi.org/10.1111/j.1471-4159.2006.04383.x

    Article  CAS  PubMed  Google Scholar 

  62. Seutin V (2005) Dopaminergic neurones: much more than dopamine? Br J Pharmacol 146(2):167–169. https://doi.org/10.1038/sj.bjp.0706328

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Wang W, Dever D, Lowe J, Storey GP, Bhansali A, Eck EK, Nitulescu I, Weimer J, Bamford NS (2012) Regulation of prefrontal excitatory neurotransmission by dopamine in the nucleus accumbens core. J Physiol 590(16):3743–3769. https://doi.org/10.1113/jphysiol.2012.235200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Giuffrida A, Parsons LH, Kerr TM, Rodríguez de Fonseca F, Navarro M, Piomelli D (1999) Dopamine activation of endogenous cannabinoid signaling in dorsal striatum. Nat Neurosci 2(4):358–363. https://doi.org/10.1038/7268

    Article  CAS  PubMed  Google Scholar 

  65. Beltramo M, de Fonseca FR, Navarro M, Calignano A, Gorriti MA, Grammatikopoulos G, Sadile AG, Giuffrida A, Piomelli D (2000) Reversal of dopamine D(2) receptor responses by an anandamide transport inhibitor. J Neurosci 20(9):3401–3407. https://doi.org/10.1523/JNEUROSCI.20-09-03401.2000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Alonso R, Voutsinos B, Fournier M, Labie C, Steinberg R, Souilhac J, Le Fur G, Soubrié P (1999) Blockade of cannabinoid receptors by SR141716 selectively increases Fos expression in rat mesocorticolimbic areas via reduced dopamine D2 function. Neuroscience 91(2):607–620. https://doi.org/10.1016/s0306-4522(98)00675-7

    Article  CAS  PubMed  Google Scholar 

  67. Meschler JP, Conley TJ, Howlett AC (2000) Cannabinoid and dopamine interaction in rodent brain: effects on locomotor activity. Pharmacol Biochem Behav 67(3):567–573. https://doi.org/10.1016/s0091-3057(00)00390-7

    Article  CAS  PubMed  Google Scholar 

  68. Okada Y, Imendra KG, Miyazaki T, Hotokezaka H, Fujiyama R, Zeredo JL, Miyamoto T, Toda K (2005) Biophysical properties of voltage-gated Na+ channels in frog parathyroid cells and their modulation by cannabinoids. J Exp Biol 208(Pt 24):4747–4756. https://doi.org/10.1242/jeb.01967

    Article  CAS  PubMed  Google Scholar 

  69. Hill AJ, Jones NA, Smith I, Hill CL, Williams CM, Stephens GJ, Whalley BJ (2014) Voltage-gated sodium (NaV) channel blockade by plant cannabinoids does not confer anticonvulsant effects per se. Neurosci Lett 566:269–274. https://doi.org/10.1016/j.neulet.2014.03.013

    Article  CAS  PubMed  Google Scholar 

  70. Patel RR, Barbosa C, Brustovetsky T, Brustovetsky N, Cummins T (2016) Aberrant epilepsy-associated mutant Nav1.6 sodium channel activity can be targeted with cannabidiol. Brain 139(Pt 8):2164–2181. https://doi.org/10.1093/brain/aww129

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ghovanloo MR, Shuart NG, Mezeyova J, Dean RA, Ruben PC, Goodchild SJ (2018) Inhibitory effects of cannabidiol on voltage-dependent sodium currents. J Biol Chem 293(43):16546–16558. https://doi.org/10.1074/jbc.RA118.004929

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Mason ER, Cummins TR (2020) Differential inhibition of human Nav1.2 resurgent and persistent sodium currents by cannabidiol and GS967. Int J Mol Sci 21(7):2454. https://doi.org/10.3390/ijms21072454

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Milligan CJ, Anderson LL, Bowen MT, Banister SD, McGregor IS, Arnold JC, Petrou S (2022) A nutraceutical product, extracted from Cannabis sativa, modulates voltage-gated sodium channel function. J Cannabis Res 4(1):30. https://doi.org/10.1186/s42238-022-00136-x

    Article  PubMed  PubMed Central  Google Scholar 

  74. Sait LG, Sula A, Ghovanloo MR, Hollingworth D, Ruben PC, Wallace BA (2020) Cannabidiol interactions with voltage-gated sodium channels. Elife 9:e58593. https://doi.org/10.7554/eLife.58593

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Huang J, Fan X, Jin X, Jo S, Zhang HB, Fujita A, Bean BP, Yan N (2023) Cannabidiol inhibits Nav channels through two distinct binding sites. Nat Commun 14(1):3613. https://doi.org/10.1038/s41467-023-39307-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Watkins AR (2019) Cannabinoid interactions with ion channels and receptors. Channels (Austin) 13(1):162–167. https://doi.org/10.1080/19336950.2019.1615824

    Article  PubMed  Google Scholar 

  77. Gantz SC, Bean BP (2017) Cell-autonomous excitation of midbrain dopamine neurons by endocannabinoid-dependent lipid signaling. Neuron 93(6):1375–1387.e2. https://doi.org/10.1016/j.neuron.2017.02.025

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lastres-Becker I, Cebeira M, de Ceballos ML, Zeng BY, Jenner P, Ramos JA, Fernández-Ruiz JJ (2001) Increased cannabinoid CB1 receptor binding and activation of GTP-binding proteins in the basal ganglia of patients with Parkinson’s syndrome and of MPTP-treated marmosets. Eur J Neurosci 14(11):1827–1832. https://doi.org/10.1046/j.0953-816x.2001.01812.x

    Article  CAS  PubMed  Google Scholar 

  79. Koppel BS (2015) Cannabis in the treatment of dystonia, dyskinesias, and tics. Neurotherapeutics 12(4):788–792. https://doi.org/10.1007/s13311-015-0376-4

    Article  PubMed  PubMed Central  Google Scholar 

  80. Roessner V, Eichele H, Stern JS, Skov L, Rizzo R, Debes NM, Nagy P, Cavanna AE, Termine C, Ganos C, Münchau A, Szejko N, Cath D, Müller-Vahl KR, Verdellen C, Hartmann A, Rothenberger A, Hoekstra PJ, Plessen KJ (2022) European clinical guidelines for Tourette syndrome and other tic disorders-version 2.0. Part III: pharmacological treatment. Eur Child Adolesc Psychiatry 31(3):425–411. https://doi.org/10.1007/s00787-021-01899-z

    Article  PubMed  Google Scholar 

  81. Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM (2000) Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson’s disease. FASEB J 14(10):1432–1438. https://doi.org/10.1096/fj.14.10.1432

    Article  PubMed  Google Scholar 

  82. van der Stelt M, Fox SH, Hill M, Crossman AR, Petrosino S, Di Marzo V, Brotchie JM (2005) A role for endocannabinoids in the generation of parkinsonism and levodopa-induced dyskinesia in MPTP-lesioned non-human primate models of Parkinson’s disease. FASEB J 19(9):1140–1142. https://doi.org/10.1096/fj.04-3010fje

    Article  CAS  PubMed  Google Scholar 

  83. Madeo G, Schirinzi T, Maltese M, Martella G, Rapino C, Fezza F, Mastrangelo N, Bonsi P, Maccarrone M, Pisani A (2016) Dopamine-dependent CB1 receptor dysfunction at corticostriatal synapses in homozygous PINK1 knockout mice. Neuropharmacology 101:460–470. https://doi.org/10.1016/j.neuropharm.2015.10.021

    Article  CAS  PubMed  Google Scholar 

  84. Moss DE, McMaster SB, Rogers J (1981) Tetrahydrocannabinol potentiates reserpine-inuced hypokinesia. Pharmacol Biochem Behav 15(5):779–783. https://doi.org/10.1016/0091-3057(81)90022-8

    Article  CAS  PubMed  Google Scholar 

  85. Anderson JJ, Kask AM, Chase TN (1996) Effects of cannabinoi receptor stimulation and blockade on catalepsy produced by dopamine receptor antagonists. Eur J Pharmacol 295(2–3):163–168. https://doi.org/10.1016/0014-2999(95)00661-3

    Article  CAS  PubMed  Google Scholar 

  86. Marcellino D, Carriba P, Filip M, Borgkvist A, Frankowska M, Bellido I, Tanganelli S, Müller CE, Fisone G, Lluis C, Agnati LF, Franco R, Fuxe K (2008) Antagonistic cannabinoid CB1/dopamine D2 receptor interactions in striatal CB1/D2 heteromers. A combined neurochemical and behavioral analysis. Neuropharmacology 54(5):815–823. https://doi.org/10.1016/j.neuropharm.2007.12.011

    Article  CAS  PubMed  Google Scholar 

  87. Fernández-Ruiz J (2009) The endocannabinoid system as a target for the treatment of motor dysfunction. Br J Pharmacol 156(7):1029–1040. https://doi.org/10.1111/j.1476-5381.2008.00088.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Karpenko MN, Vasilishina AA, Gromova EA, Muruzheva ZM, Bernadotte A (2018) Interleukin-1beta, interleukin-1 receptor antagonist, interleukin-6, interleukin-10, and tumor necrosis factor-alpha levels in CSF and serum in relation to the clinical variety of Parkinson’s disease. Cell Immunol 334:99. https://doi.org/10.1016/j.cellimm.2018.08.007

    Article  CAS  PubMed  Google Scholar 

  89. Rossi S, Furlan R, De Chiara V, Motta C, Studer V, Mori F, Musella A, Bergami A, Muzio L, Bernardi G, Battistini L, Martino G, Centonze D (2012) Interleukin-1β causes synaptic hyperexcitability in multiple sclerosis. Ann Neurol 71(1):76–83. https://doi.org/10.1002/ana.22512

    Article  CAS  PubMed  Google Scholar 

  90. De Chiara V, Motta C, Rossi S, Studer V, Barbieri F, Lauro D, Bernardi G, Centonze D (2013) Interleukin-1β alters the sensitivity of cannabinoid CB1 receptors controlling glutamate transmission in the striatum. Neuroscience 250:232–239. https://doi.org/10.1016/j.neuroscience.2013.06.069

    Article  CAS  PubMed  Google Scholar 

  91. Rossi S, Studer V, Motta C, De Chiara V, Barbieri F, Bernardi G, Centonze D (2012) Inflammation inhibits GABA transmission in multiple sclerosis. Mult Scler 18(11):1633–1635. https://doi.org/10.1177/1352458512440207

    Article  CAS  PubMed  Google Scholar 

  92. Musumeci G, Grasselli G, Rossi S, De Chiara V, Musella A, Motta C, Studer V, Bernardi G, Haji N, Sepman H, Fresegna D, Maccarrone M, Mandolesi G, Centonze D (2011) Transient receptor potential vanilloid 1 channels modulate the synaptic effects of TNF-α and of IL-1β in experimental autoimmune encephalomyelitis. Neurobiol Dis 43(3):669–677. https://doi.org/10.1016/j.nbd.2011.05.018

    Article  CAS  PubMed  Google Scholar 

  93. Rossi S, Sacchetti L, Napolitano F, De Chiara V, Motta C, Studer V, Musella A, Barbieri F, Bari M, Bernardi G, Maccarrone M, Usiello A, Centonze D (2012) Interleukin-1b causes anxiety by interacting with the endocannabinoid system. J Neurosci 32(40):13896–13905. https://doi.org/10.1523/JNEUROSCI.1515-12.2012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Gentile A, Fresegna D, Federici M, Musella A, Rizzo FR, Sepman H, Bullitta S, De Vito F, Haji N, Rossi S, Mercuri NB, Usiello A, Mandolesi G, Centonze D (2015) Dopaminergic dysfunction is associated with IL-1β-dependent mood alterations in experimental autoimmune encephalomyelitis. Neurobiol Dis 74:347–358. https://doi.org/10.1016/j.nbd.2014.11.022

    Article  CAS  PubMed  Google Scholar 

  95. Gentile A, Fresegna D, Musella A, Sepman H, Bullitta S, De Vito F, Fantozzi R, Usiello A, Maccarrone M, Mercuri NB, Lutz B, Mandolesi G, Centonze D (2016) Interaction between interleukin1b and type-1 cannabinoid receptor is involved in anxiety-like behavior in experimental autoimmune encephalomyelitis. J Neuroinflammation 13(1):231. https://doi.org/10.1186/s12974-016-0682-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mancini M, Natoli S, Gardoni F, Di Luca M, Pisani A (2023) Dopamine transmission imbalance in neuroinflammation: perspectives on long-term COVID-19. Int J Mol Sci 24(6):5618. https://doi.org/10.3390/ijms24065618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Fil A, Cano-de-la-Cuerda R, Muñoz-Hellín E, Vela L, Ramiro-González M, Fernández-de-Las-Peñas C (2013) Pain in Parkinson disease: a review of the literature. Parkinsonism Relat Disord 19(3):285–294. https://doi.org/10.1016/j.parkreldis.2012.11.009. discussion 285

    Article  PubMed  Google Scholar 

  98. Brefel-Courbon C, Payoux P, Thalamas C, Ory F, Quelven I, Chollet F, Montastruc JL, Rascol O (2005) Effect of levodopa on pain threshold in Parkinson’s disease: a clinical and positron emission tomography study. Mov Disord 20(12):1557–1563. https://doi.org/10.1002/mds.20629

    Article  PubMed  Google Scholar 

  99. Gerdelat-Mas A, Simonetta-Moreau M, Thalamas C, Ory-Magne F, Slaoui T, Rascol O, Brefel-Courbon C (2007) Levodopa raises objective pain threshold in Parkinson’s disease: a RIII reflex study. J Neurol Neurosurg Psychiatry 78(10):1140–1142. https://doi.org/10.1136/jnnp.2007.120212

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Zengin-Toktas Y, Ferrier J, Durif F, Llorca PM, Authier N (2013) Bilateral lesions of the nigrostriatal pathways are associated with chronic mechanical pain hypersensitivity in rats. Neurosci Res 76(4):261–264. https://doi.org/10.1016/j.neures.2013.05.003

    Article  PubMed  Google Scholar 

  101. Gee LE, Chen N, Ramirez-Zamora A, Shin DS, Pilitsis JG (2015) The effects of subthalamic deep brain stimulation on mechanical and thermal thresholds in 6OHDA-lesioned rats. Eur J Neurosci 42(4):2061–2069. https://doi.org/10.1111/ejn.12992

    Article  PubMed  PubMed Central  Google Scholar 

  102. Kaszuba BC, Walling I, Gee LE, Shin DS, Pilitsis JG (2017) Effects of subthalamic deep brain stimulation with duloxetine on mechanical and thermal thresholds in 6OHDA lesioned rats. Brain Res 1655:233–241. https://doi.org/10.1016/j.brainres.2016.10.025

    Article  CAS  PubMed  Google Scholar 

  103. Gómez-Paz A, Drucker-Colín R, Milán-Aldaco D, Palomero-Rivero M, Ambriz-Tututi M (2018) Intrastriatal chromospheres’ transplant reduces nociception in hemiparkinsonian rats. Neuroscience 387:123–134. https://doi.org/10.1016/j.neuroscience.2017.08.052

    Article  CAS  PubMed  Google Scholar 

  104. Nascimento GC, Bariotto-Dos-Santos K, Leite-Panissi CRA, Del-Bel EA, Bortolanza M (2018) Nociceptive response to L-DOPA-induced dyskinesia in hemiparkinsonian rats. Neurotox Res 34(4):799–807. https://doi.org/10.1007/s12640-018-9896-0

    Article  CAS  PubMed  Google Scholar 

  105. Wasner G, Deuschl G (2012) Pains in Parkinson disease–many syndromes under one umbrella. Nat Rev Neurol 8(5):284–294. https://doi.org/10.1038/nrneurol.2012.54

    Article  CAS  PubMed  Google Scholar 

  106. Lin MT, Wu JJ, Chandra A, Tsay BL (1981) Activation of striatal dopamine receptors induces pain inhibition in rats. J Neural Transm 51(3–4):213–222. https://doi.org/10.1007/BF01248953

    Article  CAS  PubMed  Google Scholar 

  107. Magnusson JE, Fisher K (2000) The involvement of dopamine in nociception: the role of D(1) and D(2) receptors in the dorsolateral striatum. Brain Res 855(2):260–266. https://doi.org/10.1016/s0006-8993(99)02396-3

    Article  CAS  PubMed  Google Scholar 

  108. Guindon J, Hohmann A (2009) The endocannabinoid system and pain. CNS Neurol Disord Drug Targets 8(6):403–421. https://doi.org/10.2174/187152709789824660

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. McDonagh MS, Morasco BJ, Wagner J, Ahmed AY, Fu R, Kansagara D, Chou R (2022) Cannabis-based products for chronic pain: a systematic review. Ann Intern Med 175(8):1143–1153. https://doi.org/10.7326/M21-4520

    Article  PubMed  Google Scholar 

  110. Mlost J, Wąsik A, Starowicz K (2019) Role of endocannabinoid system in dopamine signalling within the reward circuits affected by chronic pain. Pharmacol Res 143:40–47. https://doi.org/10.1016/j.phrs.2019.02.029

    Article  CAS  PubMed  Google Scholar 

  111. Crivelaro do Nascimento G, Ferrari DP, Guimaraes FS, Del Bel EA, Bortolanza M, Ferreira-Junior NC (2020) Cannabidiol increases the nociceptive threshold in a preclinical model of Parkinson’s disease. Neuropharmacology 163:107808. https://doi.org/10.1016/j.neuropharm.2019.107808

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

Work supported by #NEXTGENERATIONEU (NGEU) and funded by the Ministry of University and Research (MUR), National Recovery and Resilience Plan (NRRP), project MNESYS (PE0000006) — a multiscale integrated approach to the study of the nervous system in health and disease (DN. 1553 11.10.2022).

Author information

Authors and Affiliations

Authors

Contributions

M.M. wrote and edited the manuscript and prepared figures. A.C. drafted the manuscript. D.D.M. revised the article. A.P. conceived the work, supervised the work, wrote and edited the manuscript. M.M., A.C., D.D.M., A.P. reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Antonio Pisani.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mancini, M., Calculli, A., Di Martino, D. et al. Interplay between endocannabinoids and dopamine in the basal ganglia: implications for pain in Parkinson’s disease. J Anesth Analg Crit Care 4, 33 (2024). https://doi.org/10.1186/s44158-024-00169-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s44158-024-00169-z

Keywords