Wiring and firing neuronal networks: endocannabinoids take center stage

https://doi.org/10.1016/j.conb.2008.08.007Get rights and content

Endocannabinoids (eCBs) function as retrograde messengers at both excitatory and inhibitory synapses, and control various forms of synaptic plasticity in the adult brain. The molecular machinery required for specific eCB functions during synaptic plasticity is well established. However, eCB signaling plays surprisingly fundamental roles in controlling the acquisition of neuronal identity during CNS development. Recent work suggests that selective recruitment of regulatory signaling networks to CB1 cannabinoid receptors dictates neuronal state-change decisions. In addition, the spatial localization and temporal precision of eCB actions emerges as a novel organizer in developing neuronal networks. Current challenges include fitting novel molecular candidates into regulatory eCB signaling pathways, and defining the temporal dynamics of context-dependent signaling mechanisms underpinning particular neuronal specification events.

Introduction

Synaptic communication in complex neuronal networks relies on the coincident activity of integrated feedback mechanisms allowing optimal temporal refinement of activity-dependent synaptic connectivity [1]. Generation and maintenance of the structural and functional coherence of neuronal circuits is the basic cellular principle of higher brain functions. Thus, multiple mechanisms have evolved during brain development to allow the temporal refinement of neuronal excitability. Although redundancy at the level of coexistent signaling networks with partially overlapping functions ensures the continuous adaptation of presynaptic and postsynaptic components between connected neurons, retrograde synaptic signaling emerges as a uniquely powerful means to tune the temporal and spatial efficacy of synaptic information transfer.

During the past decade, several retrograde messengers have been identified (Figure 1) that exhibit robust differences in their speed of affecting synaptic integration, temporal flexibility and efficiency, and spatial precision [2, 3•, 4•, 5, 6, 7]. Endocannabinoid (eCB) signaling represents a key retrograde signaling pathway [8] for tuning both homosynaptic and heterosynaptic plasticity [9] in the postnatal brain. The general molecular paradigm is that eCBs are synthesized postsynaptically in an activity-dependent manner and engage presynaptic CB1 cannabinoid receptors (CB1Rs) on both excitatory and inhibitory afferents thus decreasing neurotransmitter release. Four basic forms of eCB-mediated synaptic plasticity have been described (Figure 1) [8, 9, 10]: Firstly, in depolarization-induced suppression of inhibition (DSI), the depolarization of a postsynaptic neuron stimulates eCB production that activates presynaptic CB1Rs at GABAergic interneuron terminals, leading to a decrease in inhibitory neurotransmission. eCBs produced in the same fashion acting on CB1R on excitatory neurons evoke depolarization-induced suppression of excitation (DSE). Secondly, in metabotropic suppression of inhibition (MSI), the activation of postsynaptic Gq/11-linked receptors (typically M1 or M3 muscarinic acetylcholine receptors or group I metabotropic glutamate receptors [mGluRs]) leads to production of eCBs activating presynaptic CB1Rs thus decreasing inhibitory neurotransmission. As before, when eCBs produced in this fashion activate CB1Rs on excitatory terminals, the process is termed metabotropic suppression of excitation (MSE). Thirdly, eCB-mediated long-term depression (LTD) is evoked during the sustained stimulation of group I mGluRs, as might happen during the prolonged low-frequency stimulation of excitatory pathways. LTD can affect either the stimulated pathway (homosynaptic LTD) or a neighboring pathway (heterosynaptic LTD), if the terminals of the neighboring pathway express CB1Rs. Fourthly, in slow-self inhibition (SSI), eCBs are produced following the repetitive depolarization of a neuron and activate CB1Rs on the same neuron, opening inwardly rectifying potassium channels and causing sustained hyperpolarization of the neuron [11]. SSI is remarkable since in this form of eCB-mediated plasticity eCBs are produced and act in the same cell, as opposed to being retrograde messengers in the other forms. It should be noted that since eCBs can inhibit both excitatory and inhibitory neurotransmission, their net effect at the circuit level, also influenced by the coincident presence of other factors, can be either inhibitory or stimulatory.

Molecular determinants of eCB-mediated retrograde signaling at central synapses appear to be developmentally organized such that they can feedback to control the earliest events of presynaptic neurotransmitter release during the transition from synaptogenesis to synaptic communication in developing neuronal circuits [12••, 13]. This leads to the question whether molecular underpinnings of eCB signaling loops acting so efficiently in the postnatal brain subserve particular physiological functions during brain development. The answer appears to be yes, but we are far from understanding the molecular logic and temporal dynamics of eCB signaling networks in the embryonic brain, and how their specific neurodevelopmental functions relate to and define their retrograde control of neurotransmitter release at mature synapses. Important open questions include: where and when eCBs are produced in the developing brain; the molecular identity of eCBs and whether they represent ‘active’ signals; whether respective receptors and intracellular signal transduction cascades differ from those in the postnatal brain; how eCB signaling integrates with other regulatory systems; and how the relative power of this newly emerging signaling entity contributes to the defining of neurodevelopmental processes. In this review, we focus on the recent discoveries establishing eCB-driven cellular identification events in the developing cerebrum, and define a unifying concept of how eCB signaling provides positional signals for excitatory and inhibitory afferents along the dendritic tree of cortical neurons, thus shaping the complexity of cortical connectivity.

Section snippets

Molecular logic of endocannabinoid signaling sculpted by developmental principles

2-Arachidonoylglycerol (2-AG) and anandamide (AEA), members of the eCB family of neuroactive lipids, are primarily synthesized by sn-1-diacylglycerol lipase α/β (DAGLα/β) [14••] and α/β-hydrolase 4/glycerophosphodiesterase 1 (ABDH4/GDE-1) [15] and bind to cannabinoid receptors in the brain (Figure 1) and at the periphery. 2-AG and AEA promiscuously activate CB1, CB2 (CB1/2R), and other cannabinoid receptors including GPR55 [16]. However, the identity of eCBs and bioactive lipids stimulating

Expression dictates function: context-dependent signaling at CB1 cannabinoid receptors

The existence of eCB ligands and CB1Rs in the developing rodent and human brain triggered an initial wave of interest when CB1Rs were unequivocally identified as the targets of Δ9-tetrahydrocannabinol (THC) from cannabis [30]. It took almost another decade for the cellular specificity, functions, and interacting partners of eCB signaling networks affecting CNS patterning to emerge. In fact, both 2-AG and AEA are present in the developing CNS from the very early stages of differentiation, though

Endocannabinoids define synapse positioning

The establishment of long-range excitatory axons by pyramidal cells precedes the neurochemical specification and synapse patterning of GABAergic interneurons during corticogenesis. Prominent DAGLα/β localization to pyramidal cell axons is required for axonal elongation through cell-autonomous signaling [33••, 37] (Figure 3). De novo synthesized 2-AG can exert either autocrine regulation via CB1Rs distributed along the longitudinal axis of the growing axon, or paracrine signaling among

Therapeutic implications

The neurodevelopmental impact of THC exposure during pregnancy has recently gained considerable attention given the long-lasting effects of prenatal cannabis use on emotional control, social behaviors, and cognition in affected offspring [46]. THC may be deleterious, acting as an agonist or an antagonist. Since THC is an intermediate potency, low efficacy CB1R agonist, THC can potentially antagonize the actions of the more efficacious 2-AG in vivo. Thus, in addition to activating CB1Rs, THC

Conclusions

The eCB system is emerging as a key regulatory signaling network fundamental to the wiring of the brain during development with an array of functions ranging from lineage segregation of stem cells to refinement of synaptic functions in complex neuronal networks. We have recently experienced a quantum leap in understanding the molecular hierarchy and signaling principles selectively governed by eCB signals in the embryonic brain. Nevertheless, the key unresolved question remains to define the

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

This work was supported by the Swedish Medical Research Council (TH), Scottish Universities Life Science Alliance (TH), Alzheimer’s Association (KM, TH), European Molecular Biology Organization Young Investigator Programme (TH), European Commission (HEALTH-F2-2007-201159; TH), National Institutes of Health Grants DA023214 (TH), DA11322 (KM), DA15916 (KM), DA21969 (KM), and BBSRC (PD).

References (50)

  • A. Ahmad-Annuar et al.

    Signaling across the synapse: a role for Wnt and Dishevelled in presynaptic assembly and neurotransmitter release

    J Cell Biol

    (2006)
  • J.P. Magby et al.

    Single-cell characterization of retrograde signaling by brain-derived neurotrophic factor

    J Neurosci

    (2006)
  • M. Hartmann et al.

    Synaptic secretion of BDNF after high-frequency stimulation of glutamatergic synapses

    EMBO J

    (2001)
  • T. Harkany et al.

    Endocannabinoid-independent retrograde signaling at inhibitory synapses in layer 2/3 of neocortex: involvement of vesicular glutamate transporter 3

    J Neurosci

    (2004)
  • R.I. Wilson et al.

    Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses

    Nature

    (2001)
  • V. Chevaleyre et al.

    Endocannabinoid-mediated synaptic plasticity in the CNS

    Annu Rev Neurosci

    (2006)
  • A. Straiker et al.

    Metabotropic suppression of excitation in murine autaptic hippocampal neurons

    J Physiol

    (2007)
  • A. Bacci et al.

    Long-lasting self-inhibition of neocortical interneurons mediated by endocannabinoids

    Nature

    (2004)
  • P. Berghuis et al.

    Hardwiring the brain: endocannabinoids shape neuronal connectivity

    Science

    (2007)
  • C. Bernard et al.

    Altering cannabinoid signaling during development disrupts neuronal activity

    Proc Natl Acad Sci U S A

    (2005)
  • T. Bisogno et al.

    Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain

    J Cell Biol

    (2003)
  • G.M. Simon et al.

    Anandamide biosynthesis catalyzed by the phosphodiesterase GDE1 and detection of glycerophospho-N-acyl ethanolamine precursors in mouse brain

    J Biol Chem

    (2008)
  • D. Piomelli

    The molecular logic of endocannabinoid signalling

    Nat Rev Neurosci

    (2003)
  • J.E. Lauckner et al.

    GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current

    Proc Natl Acad Sci U S A

    (2008)
  • E. Ryberg et al.

    The orphan receptor GPR55 is a novel cannabinoid receptor

    Br J Pharmacol

    (2007)
  • Cited by (94)

    • Endocannabinoid signaling at excitatory and inhibitory synapses

      2023, Neurobiology and Physiology of the Endocannabinoid System
    • GPR18 drives FAAH inhibition-induced neuroprotection against HIV-1 Tat-induced neurodegeneration

      2021, Experimental Neurology
      Citation Excerpt :

      Levels of the endogenous cannabinoid ligands, anandamide (N-arachidonoylethanolamine, AEA) and 2-arachidonoylglycerol (2-AG), are heightened in individuals diagnosed with neurodegenerative disorders and negatively correlate with severity of disease progression (Chen et al., 2011; More and Choi, 2015; Pryce et al., 2003). These ligands exert their effects primarily via cannabinoid receptors type 1 and 2 (CB1R and CB2R), but also through other G protein-coupled receptors (GPCRs) such as GPR55 and GRP18 (Harkany et al., 2008; Henstridge, 2012; McHugh, 2012; Ross et al., 2012). Activated microglia express CB2Rs, which regulate microglial immune function (Ashton and Glass, 2007; Ehrhart et al., 2005; Malek et al., 2015), and stimulation of this receptor blunts neurotoxic cytokine release mediated by HIV-1 gp120 protein (Hu et al., 2013).

    • Review of the Endocannabinoid System

      2021, Biological Psychiatry: Cognitive Neuroscience and Neuroimaging
    • Mini-review: The therapeutic role of cannabinoids in neuroHIV

      2021, Neuroscience Letters
      Citation Excerpt :

      The two main eCB ligands are N-arachidonoylethanolamine (AEA, also known as anandamide) and 2-arachidonoylglycerol (2-AG). These ligands as well as exogenous cannabinoids, such as delta-9-tetrahydrocannabinol (Δ9-THC) found in cannabis, act predominantly via cannabinoid type 1 and/or cannabinoid type 2 receptors (CB1R and CB2R, respectively), but can also activate the transient receptor potential vanilloid (TRPV) ion channels [102,117], peroxisome proliferator-activated receptors (PPARs) [174,224], and/or other G-protein-coupled receptors, including GPR55 and GPR18 [42,50,93,202]. The CB1R is the most abundant G-protein-coupled receptor in the CNS, mainly expressed on neurons [150] and is responsible for the psychoactive effects of Δ9-THC, which is a primary compound of cannabis [65,220].

    View all citing articles on Scopus
    View full text