Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Pharmacology & Therapeutics 244 (2023) 108394

Contents lists available at ScienceDirect

Pharmacology & Therapeutics

journal homepage: www.elsevier.com/locate/pharmthera

Inhibiting degradation of 2-arachidonoylglycerol as a therapeutic


strategy for neurodegenerative diseases
Chu Chen ⁎
Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX
78229, USA

a r t i c l e i n f o a b s t r a c t

Article history: Endocannabinoids are endogenous lipid signaling mediators that participate in a variety of physiological and
Received 9 February 2023 pathological processes. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid and is a full ago-
Received in revised form 17 March 2023 nist of G-protein-coupled cannabinoid receptors (CB1R and CB2R), which are targets of Δ9-tetrahydrocannabinol
Accepted 21 March 2023
(Δ9-THC), the main psychoactive ingredient in cannabis. While 2-AG has been well recognized as a retrograde
Available online 24 March 2023
messenger modulating synaptic transmission and plasticity at both inhibitory GABAergic and excitatory gluta-
Associate editor: S.J. Enna matergic synapses in the brain, growing evidence suggests that 2-AG also functions as an endogenous terminator
of neuroinflammation in response to harmful insults, thus maintaining brain homeostasis. Monoacylglycerol
lipase (MAGL) is the key enzyme that degrades 2-AG in the brain. The immediate metabolite of 2-AG is arachi-
Keywords:: donic acid (AA), a precursor of prostaglandins (PGs) and leukotrienes. Several lines of evidence indicate that
Endocannabinoid pharmacological or genetic inactivation of MAGL, which boosts 2-AG levels and reduces its hydrolytic metabo-
2-Arachidonoylglycerol lites, resolves neuroinflammation, mitigates neuropathology, and improves synaptic and cognitive functions in
Monoacylglycerol lipase animal models of neurodegenerative diseases, including Alzheimer’s disease (AD), multiple sclerosis (MS),
Neurodegenerative disease
Parkinson’s disease (PD), and traumatic brain injury (TBI)-induced neurodegenerative disease. Thus, it has
Alzheimer’s disease
been proposed that MAGL is a potential therapeutic target for treatment of neurodegenerative diseases. As the
Traumatic brain injury
main enzyme hydrolyzing 2-AG, several MAGL inhibitors have been identified and developed. However, our
understanding of the mechanisms by which inactivation of MAGL produces neuroprotective effects in neurode-
generative diseases remains limited. A recent finding that inhibition of 2-AG metabolism in astrocytes, but not in
neurons, protects the brain from TBI-induced neuropathology might shed some light on this unsolved issue. This
review provides an overview of MAGL as a potential therapeutic target for neurodegenerative diseases and
discusses possible mechanisms underlying the neuroprotective effects of restraining degradation of 2-AG in
the brain.
© 2023 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY license (http://
creativecommons.org/licenses/by/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Synthesis and metabolism of 2-AG . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. 2-AG signaling in resolving neuroinflammation and protecting neurons . . . . . . . . . . . . . . . . . . . . 6
4. MAGL in neurodegenerative diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
5. Signaling pathways mediating neuroprotection following inactivation of MAGL . . . . . . . . . . . . . . . . . 10

Abbreviations: ABHD6/12, α/β-hydrolase domain containing 6/12; AC, adenylyl cyclase; AD, Alzheimer’s disease; AEA, anandamide; AA, arachidonic acid; 2-AG, 2-
arachidonoylglycerol; BBB, blood-brain barrier; CB1R, cannabinoid receptor type 1; CB2R, cannabinoid receptor type 2; CBD, cannabidiol; CHI, closed head injury; COX-2, cyclooxygen-
ase-2; DAGLα/β, diacylglycerol lipase α/β; EET, epoxyeicosatrienoic acids; ERK1/2, extracellular signal-regulated kinases 1 and 2; FAAH, fatty acid amide hydrolase; FABPs, fatty-acid
binding proteins; GABA, gamma-aminobutyric acid; HETE, hydroxyeicosatetraene; HPETE, hydroperoxyeicosatetraenoic acid; IL-1β,6, interleukin-1β,6; LTP, long-term potentiation;
MAGL, monoacylglycerol lipase; MS, multiple sclerosis; NAPE, N-arachidonoyl phosphatidyl ethanolamine; NAPE-PLD, NAPE phospholipase D; NF-κB, nuclear factor kappa-light-
chain-enhancer of activated B cells; PD, Parkinson’s disease; PG, prostaglandin; PG-G, prostaglandin glycerol; PPARγ, peroxisome proliferator-activated receptor γ; TBI, traumatic brain
injury; Δ9-THC, Δ9-tetrahydrocannabinol; TNFα, tumor necrosis factor α.
⁎ Corresponding author at: Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229,
USA.
E-mail address: chenc7@uthscsa.edu (C. Chen).

https://doi.org/10.1016/j.pharmthera.2023.108394
0163-7258/© 2023 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

6. MAGL inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7. Perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12

1. Introduction & Newquist, 2013; Hoffman et al., 2007; Messinis, Kyprianidou,


Malefaki, & Papathanasopoulos, 2006; Pope Jr. & Yurgelun-Todd, 1996;
Marijuana has been used by humans over a few thousand years for Puighermanal et al., 2009; Solowij, et al., 2002; Taffe, 2012; Volkow,
both recreational and medical purposes. The active ingredients in Baler, Compton, & Weiss, 2014), medical use of Δ9-THC (Dronabinol or
marijuana remained unknown until the mid-1960 when Dr. Raphael Marinol) is limited to some conditions for treatment of nausea and
Mechoulam, the “Father of Cannabinoid Research”, isolated Δ9- vomiting caused by cancer chemotherapy or weight loss and for poor
tetrahydrocannabinol (Δ9-THC) and identified it to be the main psycho- appetite in patients with acquired immunodeficiency syndrome
active constituent in cannabis (Mechoulam, 1970; Mechoulam & Parker, (AIDS). Since then, over 400 chemicals in marijuana have been identi-
2013). The Mechoulam team also identified another prevalent fied, of which more than 100 are chemically and biosynthetically related
phytocannabinoid in cannabis, cannabidiol (CBD) (Mechoulam & cannabinoids (Ligresti, De Petrocellis, & Di Marzo, 2016). Synthetic can-
Shvo, 1963). A most recent review article provides an overview on nabinoids, including CP-55940, Hu-210, and Win55,212-2, emerged
similarities and differences between Δ9-THC and CBD in terms of their from the identification of Δ9-THC (Fig. 1). In the mid-1980s, the ground-
molecular structures, mechanisms of biological effects on brain func- breaking work by Dr. Allyn Howlett’s lab using radiolabeled [3H]-
tions, and side effects (Stella, 2023). CBD has been thought CP55940 provided solid and conclusive evidence that cannabinoids
non-psychotropic and fewer side effects. However, recent studies sug- inhibit adenylyl cyclase (AC) through GTP-binding proteins (Devane,
gested that CBD should be considered psychotropic as well (Stella, Dysarz 3rd, Johnson, Melvin, & Howlett, 1988; Howlett, 1987; Howlett
2023). In rodent studies, it has been observed that CBD impairs memory et al., 2004; Howlett & Fleming, 1984; Howlett, Qualy, & Khachatrian,
reconsolidation, affects motor behavior, and impairs attentional 1986). These findings led to identification and cloning of two types of
set-shifting and spatial working memory (Bayer et al., 2022; Espejo- cannabinoid receptors, type 1 (CB1R) and type 2 (CB2R) in the early
Porras, Fernández-Ruiz, Pertwee, Mechoulam, & García, 2013; Stella, 1990s in both animals and humans (Herkenham et al., 1990;
2023; Szkudlarek et al., 2019). It has long been known that Δ9-THC Herkenham et al., 1991; Howlett et al., 1990; Matsuda, Lolait,
induces neuropsychological and cognitive side effects, including Brownstein, Young, & Bonner, 1990; Munro, Thomas, & Abu-Shaar,
anxiety-like and impaired locomotor behavior, synaptic and memory 1993; Van Sickle et al., 2005). Both CB1R and CB2R are seven-
impairments (Carlini, 2004; Chen et al., 2013; Hoffman, Oz, Yang, transmembrane domain receptors, and they display similar pharmaco-
Lichtman, & Lupica, 2007; Stella, 2023). Both Δ9-THC and CBD are FDA logical and biochemical properties, coupling to GTP binding proteins
approved cannabinoid medicines. However, due to the undesirable to inhibit activity of AC (Fig. 2) (Felder et al., 1995; Howlett & Abood,
side effects (Amin & Ali, 2019; Chen et al., 2013; Fitzgerald, Bronstein, 2017). While CB1R is primarily expressed in the brain, CB2R is

Fig. 1. Δ9-tetrahydrocannabinol (Δ9-THC) and synthetic cannabinoids.

2
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

predominantly expressed in the peripheral immune system and in It has been almost six decades since the Mechoulam’s group isolated
microglial cells, the primary innate immune cells of the brain (Howlett and identified Δ9-THC as the primary psychoactive constituent of can-
et al., 2002; Howlett et al., 2004; Mackie, 2005, 2008; Pertwee, 2006). nabis (Howlett et al., 2004; Mechoulam, 1970; Mechoulam, Braun, &
Discovery and identification of CB1R and CB2R, and especially the Gaoni, 1967; Mechoulam & Gaoni, 1967; Mechoulam, Hanuš, Pertwee,
discovery of high levels of CB1R expression in the brain, provided & Howlett, 2014). Their pioneering work opened a new era of cannabi-
clues to the existence of endogenous lipid ligands that bind to and acti- noid research and greatly enhanced our understanding of the ingredi-
vate CB1R or CB2R, prompting the search for endogenous cannabinoid ents of marijuana and actions of cannabinoids. Their research
ligands. The first endocannabinoid, N-arachidonylethanolamine (AEA), promoted and facilitated discovery of the endogenous cannabinoid sys-
also known as anandamide, was isolated and identified in 1992 tem. Through extensive and rigorous research by scientists over the past
(Devane et al., 1992; Felder et al., 1993). Soon thereafter, in 1995, a sec- 30 years, we have now recognized and confirmed the existence of the
ond endocannabinoid, 2-arachidonoylglycerol (2-AG), was indepen- endogenous cannabinoid system, which elicits biological effects when
dently identified by two groups of investigators, Drs. Mechoulam and it is activated (Lu & Mackie, 2021). The endocannabinoid system con-
Sugiura (Mechoulam et al., 1995; Sugiura et al., 1995; Sugiura, sists of cannabinoid receptors (CB1R and CB2R), endocannabinoids
Kishimoto, Oka, & Gokoh, 2006). These discoveries were soon con- (e.g., AEA and 2-AG), transporters, and the enzymes that synthesize
firmed by other groups (Gonsiorek et al., 2000; Hillard, 2000; Stella, and degrade endocannabinoids (Fig. 4). In addition, there are a few ad-
Schweitzer, & Piomelli, 1997) and several putative endogenous canna- ditional putative cannabinoid receptors, one of which is GPR55 (Kano
binoids have been identified (Fig. 3). AEA and 2-AG have been investi- et al., 2009; Ligresti et al., 2016; Lu & Mackie, 2021). Recent evidence
gated most thoroughly and they differ in several aspects in terms of shows that fatty acid-binding protein 5 (FABP5) is an important trans-
binding activity for CB1R or CB2R. For instance, 2-AG is the most abun- porter for endocannabinoids (Haj-Dahmane et al., 2018; Kaczocha,
dant endocannabinoid and a full agonist for CB1R and CB2R, whereas Glaser, & Deutsch, 2009; Kaczocha & Haj-Dahmane, 2022). It is clear
AEA is a partial agonist for CB1R and CB2R (Howlett & Abood, 2017; now that the endocannabinoid system participates in a variety of phys-
Howlett et al., 2002; Howlett et al., 2004; Kano, Ohno-Shosaku, iological and pathological processes, which have been discussed in
Hashimotodani, Uchigashima, & Watanabe, 2009; Piomelli, 2003; several previous reviews in details (Baggelaar, Maccarrone, & van der
Sugiura et al., 2006). AEA is also involved in pain sensation and chronic Stelt, 2018; Castillo, Younts, Chávez, & Hashimotodani, 2012; Cristino,
inflammation through activation of a transient receptor potential cation Bisogno, & Di Marzo, 2020; Di Marzo, Stella, & Zimmer, 2015; Egmond,
channel subfamily V member 1 (TRPV1), also known as a vanilloid Straub, & der Stelt, 2021; Freund, Katona, & Piomelli, 2003; Howlett
receptor (Aghazadeh Tabrizi et al., 2017; Muller, Morales, & Reggio, et al., 2004; Kaczocha & Haj-Dahmane, 2022; Kano et al., 2009; Katona
2018; Starowicz, Nigam, & Di Marzo, 2007). & Freund, 2012; Ligresti et al., 2016; Lu & Mackie, 2021; Mechoulam
et al., 2014; Mechoulam & Parker, 2013; Pertwee, 2006, 2015). This re-
view primarily focuses on the latest developments in 2-AG signaling
and its metabolism by inactivation of monoacylglycerol lipase
(MAGL), also known as monoglyceride lipase, the primary enzyme
that hydrolyzes 2-AG, in animal models of neurodegenerative diseases
(Blankman, Simon, & Cravatt, 2007; Dinh et al., 2002; Dinh, Freund, &
Piomelli, 2002; Long, Nomura, & Cravatt, 2009; Viader et al., 2015).

2. Synthesis and metabolism of 2-AG

Since 2-AG was identified as an endogenous cannabinoid, 2-AG


functioning as a retrograde messenger in modulation of synaptic trans-
mission and plasticity at both GABAergic and glutamatergic synapses
has been extensively studied. It has been recognized that 2-AG is re-
leased from postsynaptic neurons and activates presynaptically
expressed CB1R, resulting in a reduction of neurotransmitter release
through inhibition of voltage-gated Ca2+ channel activity and a con-
comitant increase in K+ channel conductance in presynaptic nerve ter-
minals (Alger, 2002; Castillo et al., 2012; Chevaleyre & Castillo, 2003,
2004; Freund & Hajos, 2003; Gao et al., 2010; Gerdeman, Ronesi, &
Lovinger, 2002; Hashimotodani, Ohno-Shosaku, & Kano, 2007; Heifets
& Castillo, 2009; Iversen, 2003; Kano et al., 2009; Katona & Freund,
2012; Kreitzer & Regehr, 2002; Lovinger, 2008; Pitler & Alger, 1994;
Stella et al., 1997; Tanimura et al., 2010; Vigh & von Gersdorff, 2007;
Wilson, Kunos, & Nicoll, 2001; Wilson & Nicoll, 2001, 2002). Accumulat-
ing evidence indicates that 2-AG possesses anti-inflammatory and
neuroprotective effects in response to proinflammatory, neurotoxic,
and mechanical insults and maintains brain homeostasis (Chen, 2015,
2023; Du, Chen, Zhang, & Chen, 2011; Panikashvili, Mechoulam, Beni,
Alexandrovich, & Shohami, 2005; Panikashvili et al., 2006; Panikashvili
et al., 2001; Song, Zhang, & Chen, 2015; Sugiura et al., 2006; Zhang &
Chen, 2008; Zhang, Hu, Teng, Tang, & Chen, 2014), suggesting that
2-AG is an important endogenous lipid mediator involved in multiple
Fig. 2. GTP-binding protein-coupled cannabinoid receptors (CB1R and CB2R). Cannabinoid functions in the brain.
(Δ9-THC) or endocannabinoid (2-AG) binds to CB1R or CB2R to activate the receptor- 2-AG is the most abundant endocannabinoid and has been recog-
coupled G protein, resulting in dissociation of the Gαi subunit from Gβγ subunits. Disso-
ciated Gαi subunit inhibits activity of adenylyl cyclase (AC) that catalyzes ATP to cAMP,
nized as a true natural ligand for both CB1R and CB2R (Sugiura et al.,
leading to reduction of protein kinase A (PKA) activity. Dissociated Gβγ subunits interact 2006). 2-AG is generated from membrane phospholipids (e.g., phos-
with other effectors to regulate biological function through different signaling pathways. phatidylinositol, phosphatidylinositol 4,5-bisphosphate, triacylglycerol,

3
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Fig. 3. Substances that have been identified as endogenous cannabinoids. Anandamide (AEA) and 2-arachidonoylglycerol (2-AG) are the most studied endocannabinoids (Adapted from
Kano et al., 2009 with permission from American Physiological Society).

or lysophosphatidylinositol) through multiple pathways (Murataeva, hydrolysis of 2-AG displays tissue-specificity. For example, systemic ad-
Straiker, & Mackie, 2014). However, the primary pathway of biosynthe- ministration of a potent and irreversible MAGL inhibitor JZL184 results
sis 2-AG is via conversion of membrane phospholipids to diacylglycerols in a dramatic elevation of 2-AG levels in the brain, but has less effect
(DAG) by phospholipase C-β (PLCβ) and then hydrolysis of DAG to 2-AG in peripheral tissues (Long, Nomura, & Cravatt, 2009), suggesting that
by diacylglycerol lipase α and diacylglycerol lipase β (DAGLα/β) 2-AG in the brain is predominantly metabolized by MAGL. Indeed, it
(Fig. 5). Diacylglycerol lipases were cloned in 2003 (Bisogno et al., has been estimated that 85% of 2-AG in the brain is hydrolyzed by
2003), advancing our understanding of the key pathway for biosynthe- MAGL, whereas about 12% of 2-AG is hydrolyzed by ABHD6 and
sis of 2-AG. There is evidence that DAGLα is the major biosynthetic en- ABHD12 (Alhouayek, Masquelier, & Muccioli, 2014; Blankman et al.,
zyme for 2-AG in neurons and astrocytes in the brain, while DAGLβ is 2007; Dinh, Carpenter, et al., 2002; Dinh, Freund, & Piomelli, 2002;
responsible for synthesis of 2-AG in microglia (Gao et al., 2010; Fiskerstrand et al., 2010; Labar, Wouters, & Lambert, 2010; Long,
Murataeva et al., 2014; Shin et al., 2018; Tanimura et al., 2010; Viader Nomura, & Cravatt, 2009; Marrs et al., 2010; Muccioli et al., 2007;
et al., 2016; Xue et al., 2019; Yoshida et al., 2006). The presence of Murataeva et al., 2014; Navia-Paldanius, Savinainen, & Laitinen, 2012;
DAGLα in the postsynaptic site and loss of retrograde endocannabinoid Nomura et al., 2011; Savinainen, Saario, & Laitinen, 2012; Scalvini,
signaling when DAGLα is pharmacologically or genetically inactivated Piomelli, & Mor, 2016; Schlosburg et al., 2010). ABHD6 has been identi-
further support the notion that 2-AG functions as a retrograde signaling fied in postsynaptic dendrites (Marrs et al., 2010), suggesting that
messenger modulating synaptic transmission and plasticity (Castillo ABHD6 may play an important role in maintaining the efficacy of
et al., 2012; Diana & Marty, 2004; Gao et al., 2010; Hashimotodani 2-AG-mediated retrograde signaling in synaptic transmission
et al., 2007; Kano et al., 2009; Murataeva et al., 2014; Ogasawara et al., (Baggelaar et al., 2018; Marrs et al., 2010). The latest studies demon-
2016; Ohno-Shosaku & Kano, 2014; Tanimura et al., 2010; Yoshida strated that MAGL is the primary enzyme hydrolyzing 2-AG in neurons
et al., 2006). and astrocytes, whereas ABHD12 is the major enzyme degrading 2-AG
Like the synthesis of 2-AG, there are several pathways involving en- in microglial cells (X. Liu et al., 2016; Viader et al., 2015; Viader et al.,
zymes that degrade 2-AG (Fig. 5), including MAGL, α/β hydrolase 2016). These studies provide important information suggesting involve-
domain-containing protein 6 and 12 (ABHD6 and ABHD12), and ment of diverse pathways in biosynthesis and metabolism of 2-AG in
cyclooxygenase-2 (COX-2). MAGL was first identified as an enzyme hy- the brain. Moreover, 2-AG can be oxidatively catalyzed by COX-2 to
drolyzing 2-AG by the Piomelli’s lab where they found that MAGL par- form new types of prostaglandins, specifically prostaglandin glycerols
ticipates in 2-AG inactivation (Dinh, Carpenter, et al., 2002; Dinh, (PG-Gs), when expression and activity of COX-2 are elevated during in-
Freund, & Piomelli, 2002). While MAGL is expressed in most cells, its flammation (Kingsley, Rouzer, Morgan, Patel, & Marnett, 2019; Morgan

4
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Fig. 4. The endocannabinoid system. There are five components of the endocannabinoid system consisting of endocannabinoids, cannabinoid receptors, enzymes synthesizing and
degrading endocannabinoids, and endocannabinoid transporters. AEA is a partial agonist for CB1R and CB2R but is a full agonist for TRPV1. 2-AG is a full agonist for CB1R and CB2R.
AA: arachidonic acid; ABHD6/12: α/β-hydrolase domain containing 6 and 12; AEA: Anandamide; 2-AG: 2-arachidonoylglycerol; COX-2: cyclooxygenase-2; DAG: diacylglycerol;
DAGLα/β: diacylglycerol lipase α/β; FAAH: fatty acid amide hydrolase; FABP5: fatty acid-binding protein 5; GPR55: G protein-coupled receptor 55; MAGL: monoacylglycerol lipase;
NAPE: N-arachidonyl-phosphatidyl ethanolamine; NAPE-PLD: N-acetylphosphatidylethanolamine-hydrolysing phospholipase D; PG-EA: prostaglandin ethanolamine; PG-G: prostaglan-
din ethanolamine glycerol; TRPV1: transient receptor potential vanilloid 1

et al., 2018; Rouzer & Marnett, 2008). There are two isoforms of Masquelier, Cani, Lambert, & Muccioli, 2013; Alhouayek & Muccioli,
cyclooxygenases, COX-1 and COX-2, which convert arachidonic acid 2014; Hu, Bradshaw, Chen, Tan, & Walker, 2008; Kingsley et al., 2019).
(AA) to prostaglandins (PGs), including PGE2, PGD2, PGF2α, PGI2, TXA2 Therefore, COX-2 plays unique roles not only in catalyzing AA to
(Vane, Bakhle, & Botting, 1998; Yang & Chen, 2008). COX-1 is constitu- form PGs, but also in oxidatively metabolizing 2-AG to PG-Gs
tively expressed in all tissues and functions as a housekeeper. However, (Alhouayek & Muccioli, 2014; Baggelaar et al., 2018; Guindon &
COX-2 is inducible and its expression is immediately upregulated in re- Hohmann, 2008; Sang & Chen, 2006). In addition, carboxylesterases
sponse to infection or insults (Chen & Bazan, 2005; Vane et al., 1998; have been reported to hydrolyze 2-AG in peripheral tissues, includ-
Yang & Chen, 2008). Basal expression of COX-2 is only detected in the ing lung, spleen, and macrophages (Szafran, Borazjani, Lee, Ross, &
brain, kidney, and testis under resting conditions. In the early 2000s, it Kaplan, 2015; Szafran et al., 2022; Xie et al., 2010). However, there
was found that endocannabinoids AEA and 2-AG can be oxidatively me- are not reports about 2-AG hydrolysis by carboxylesterases in the
tabolized by COX-2, but not by COX-1. Pioneering work by the Marnett brain.
group identified 2-AG as a substrate for COX-2 (Duggan et al., 2011; Arachidonic acid is the immediate metabolite of 2-AG hydrolyzed by
Hermanson et al., 2013; Hermanson, Gamble-George, Marnett, & Patel, MAGL, ABHD6 or ABHD12 and is a precursor of prostaglandins (PGs)
2014; Kingsley et al., 2019; Kozak, Prusakiewicz, & Marnett, 2004; and leukotrienes (Fig. 5). AA is an important lipid mediator that partic-
Kozak, Prusakiewicz, Rowlinson, Schneider, & Marnett, 2001; Kozak, ipates in numerous biological events through conversion to PGs,
Rowlinson, & Marnett, 2000; Morgan et al., 2018; Rouzer & Marnett, hydroperoxyeicosatetraenoic acids (HPETEs), hydroxyeicosatetraenes
2008). Oxygenation of 2-AG by COX-2 produces PG-Gs, which are (HETEs), leukotrienes, lipoxins, and epoxyeicosatrienoic acids (EETs)
distinct from classical PGs derived from AA. The biological function of catalyzed by COX-1/2, lipoxygenases (LOX), and cytochrome P450 oxi-
PG-Gs remained unclear, and it is uncertain whether there are specific dases (CYP), respectively (Fig. 5). Certain metabolites of AA (e.g., PGE2
receptors for these 2-AG metabolites catalyzed by COX-2 (Alhouayek and LTB4) are proinflammatory mediators (Henderson Jr., 1994;
& Muccioli, 2014; Kingsley et al., 2019; Murataeva et al., 2014). Ricciotti & FitzGerald, 2011; Salmon & Higgs, 1987; Xu & Chen, 2015;
However, earlier studies showed that PGE2-G enhances both excitatory Yao & Narumiya, 2019). It has been long thought that AA is primarily de-
and inhibitory synaptic transmission and induces neurotoxicity, rived from membrane phospholipids through phospholipase A2 (PLA2)/
whereas 2-AG suppresses synaptic transmission, resolves neuroinflam- PLC pathways. However, it has been shown that 2-AG hydrolysis-
mation, and protects neurons from harmful insults, suggesting that the delivered AA contributes a significant proportion to the AA pool
effects of COX-2 metabolites of 2-AG are opposite to that of 2-AG (Chen, (Nomura et al., 2011), suggesting that control of 2-AG metabolism by
Zhang, & Chen, 2011; Lindgren et al., 2013; Sang & Chen, 2006; Sang, manipulation of the various enzymes that degrade 2-AG will have a sig-
Zhang, & Chen, 2006, 2007; Zhang & Chen, 2008). Currently available nificant impact on AA-mediated signaling cascades in physiology and
information indicates that different COX-2 metabolites of 2-AG may diseases (Alhouayek et al., 2014; Baggelaar et al., 2018; Grabner,
display distinct effects on inflammatory responses (Alhouayek, Zimmermann, Schicho, & Taschler, 2017).

5
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Fig. 5. Biosynthesis and metabolism of 2-arachidonoylglycerol (2-AG). 2-AG is generated from membrane phospholipids (e.g., phosphatidylinositol and phosphatidylinositol 4,5-
bisphosphate) through phospholipase C-β (PLCβ) to diacylglycerols (DAG). DAG is then converted to 2-AG by two isoforms of diacylglycerol lipase α and diacylglycerol lipase β
(DAGLα/β). 2-AG is primarily degraded by monoacylglycerol lipase (MAGL) as well as by α/β-hydrolase domain containing 6 and 12 (ABHD6/12) to arachidonic acid (AA). 2-AG is
also oxidatively converted by cyclooxygenase-2 (COX-2) to prostaglandin glycerols (PG-Gs). AA is further metabolized by COX-1/2 to prostaglandins (PGs) and by lipoxygenase (LOX)
to hydroperoxyeicosatetraenoic acids (HPETEs), hydroxyeicosatetraenes (HETEs), and leukotrienes (LT4s). AA is also catalyzed by cytochrome P450 oxidases (CYP) to HETEs or
epoxyeicosatrienoic acids (EETs).

3. 2-AG signaling in resolving neuroinflammation and protecting endogenous terminator of inflammation in response to harmful insults
neurons that elicit inflammatory responses (Chen, 2023). This assumption has
been supported by several studies. For example, 2-AG produces a
The earliest evidence that 2-AG acts to resolve neuroinflammation dose-dependent suppression of proinflammatory factor lipopolysaccha-
and protect neurons comes from the studies of Mechoulam and ride (LPS)- or excitotoxic kainic acid-induced COX-2 expression and
Shohami in which animals received a traumatic brain injury (TBI), spe- prevents IL-1β- or glutamate-induced neurodegeneration. These effects
cifically a closed head injury (Panikashvili et al., 2001). The authors ob- are mediated through CB1R-dependnet inhibition of mitogen-activated
served that TBI induces release of 2-AG in the brain and that direct protein kinase (MAPK)/NF-κB signaling (Zhang & Chen, 2008). Appar-
administration of synthetic 2-AG significantly reduces TBI-induced ently, 2-AG inhibition of COX-2 is primarily restricted to glial cells. How-
edema and cell death. The protective effects appear to be mediated by ever, synthetic cannabinoids CP-55940 and Win55,212-2 failed to
G protein-coupled CB1R as the beneficial effects of 2-AG are attenuated prevent LPS-induced elevation of COX-2 in vitro (Zhang & Chen,
by SR141716 (rimonabant), a potent and selective antagonist of CB1R, 2008). In addition, LPS-induced COX-2 is reduced by enhancement of
or by genetic deletion of CB1R (Panikashvili et al., 2001; Panikashvili endogenous 2-AG by URB602, a MAGL inhibitor but not by URB597,
et al., 2005). They provided further evidence that 2-AG-produced neu- an inhibitor of fatty acid amide hydrolase (FAAH), which hydrolyzes
roprotection in TBI is a consequence of CB1R-mediated inhibition of nu- AEA. These results suggest that 2-AG functions as an endogenous inhib-
clear factor kappa B (NF-κB), which results in inhibition of transcription itor of COX-2, which protects neurons from harmful insults, as elevated
and expression of cytokines, including tumor necrosis factor α (TNFα), COX-2 is a key player and also an important indicator in many
interleukin-1β (IL-1β), and IL-6 (Mechoulam, Spatz, & Shohami, 2002; neuroinflammation-associated neurodegenerative diseases, including
Panikashvili et al., 2001; Panikashvili et al., 2005; Panikashvili et al., Alzheimer’s disease (AD), multiple sclerosis (MS), Parkinson’s disease
2006; Shohami, Cohen-Yeshurun, Magid, Algali, & Mechoulam, 2011). (PD), and TBI-induced neurodegenerative disease (Ferrer et al., 2019;
Neuroinflammation is one of the early neurochemical responses after Hoozemans & O'Banion, 2005; Hurley, Olschowka, & O'Banion, 2002;
TBI (Jassam, Izzy, Whalen, McGavern, & El Khoury, 2017). The neuropro- Minghetti, 2004; Teismann et al., 2003; Turini & DuBois, 2002; Vane
tective effects of 2-AG are largely associated with suppression of neuro- et al., 1998). Consistent with the studies described above, other studies
inflammatory responses following TBI, suggesting that 2-AG is an also found that direct application of synthetic 2-AG or blockade of

6
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

endogenous 2-AG degradation by MAGL inhibitors URB602 or JZL184 Finally, monoacylglycerols are hydrolyzed by MAGL to fatty acid
attenuate β-amyloid (Aβ)-induced neuronal apoptosis and degenera- and glycerol. MAGL was cloned in 1997 (Karlsson et al., 2001;
tion via CB1R-mediated suppression of extracellular signal-regulated ki- Karlsson, Contreras, Hellman, Tornqvist, & Holm, 1997). It has a mo-
nases 1 and 2 (ERK1/2), NF-κB, and COX-2 in cultured hippocampal lecular weight of 33 kDa and is expressed in cytoplasm and plasma
neurons (X. Chen et al., 2011). Other studies also found similar neuro- membrane of many tissues and cells and in lipid droplets (Gulyas
protective effects of 2-AG in caudate nucleus (CN) neurons against LPS et al., 2004; Zechner et al., 2009). Importantly, MAGL is not only an
stimulus by CB1R-dependent suppression of MAPK-NF-κB and COX-2 intracellular α/β serine hydrolase that hydrolyzes monoacylglycerol
(Y. Lu, Peng, Dong, & Yang, 2014). The Chen group provided further ev- but is also the main enzyme that degrades 2-AG in the brain
idence that peroxisome proliferator-activated receptor gamma γ (Blankman et al., 2007; Dinh, Carpenter, et al., 2002; Dinh, Freund,
(PPARγ) is a downstream signaling molecule of CB1R in mediating 2- & Piomelli, 2002; Long & Cravatt, 2011; Long, Nomura, & Cravatt,
AG-induced suppression of LPS- or cytokine-induced increases in syn- 2009; Nomura et al., 2011; Scalvini et al., 2016; Schlosburg et al.,
aptic release of glutamate, expression of COX-2, and phosphorylation 2010). MAGL is highly expressed in the brain, particularly, in presyn-
of NF-κB (Du et al., 2011). They observed that exogenous or endogenous aptic nerve terminals, suggesting that MAGL plays an important role
2-AG-induced suppression of these changes in response to LPS or IL-1β in terminating 2-AG signaling to precisely modulate or tune synaptic
stimulus is blocked by GW9662, a PPARγ antagonist, whereas 15- activity (Dinh, Freund, & Piomelli, 2002; Ludányi et al., 2011). Use of
deoxy-Δ-12,14-prostaglandin J2 (15d-PGJ2), an endogenous PPARγ pharmacological and genetic approaches to manipulate MAGL activ-
agonist, or rosiglitazone, a synthetic PPARγ agonist mimics the effects ity have provided significant insights into 2-AG metabolism in phys-
of 2-AG-induced suppression of synaptic release of glutamate, expres- iology and disease.
sion of COX-2, and phosphorylation of NF-κB (Du et al., 2011). Using a While ‘on demand’ synthesis and release of 2-AG are important for
PPARγ luciferase assay and immunoblot analysis, it was found that 2- brain homeostasis, 2-AG is rapidly degraded by several enzymes, in-
AG is capable of activating PPARγ and increasing expression of PPARγ, cluding MAGL, ABHD6/12, or COX-2 upon its formation. Higher levels
suggesting that 2-AG is an endogenous PPARγ agonist (Du et al., 2011; of 2-AG can be maintained in the brain by increasing 2-AG synthesis
Hu et al., 2022; Zhang et al., 2014). It was also observed that direct ad- by increasing DAGLα/β activity or by reducing degradation. However,
ministration of 2-AG delays disease onset, reduces axonal pathology, increasing 2-AG levels by enhancing its synthesis may accelerate me-
and reduces mortality in experimental autoimmune encephalomyelitis tabolism of 2-AG and release of AA, resulting in elevation of metabolites
(EAE), a widely used model of MS. This effect is likely associated with of AA. This approach may not achieve ideal beneficial effects as desired
activation of cannabinoid receptors (Lourbopoulos et al., 2011). How- since certain types of AA metabolites such as prostaglandins (i.e., PGE2)
ever, other studies showed that inhibition of LPS-stimulated expression and leukotrienes (LTB4) are proinflammatory mediators and neurotoxic
and release of TNFα in microglial cells by 2-AG are independent of CB1R (Hein & O'Banion, 2009; Henderson Jr., 1994; Ricciotti & FitzGerald,
or CB2R (Facchinetti, Del Giudice, Furegato, Passarotto, & Leon, 2003). In 2011; Salmon & Higgs, 1987; Xu & Chen, 2015; Yao & Narumiya,
addition, there are studies showing that 2-AG-induced suppression of 2019). Because 2-AG in the brain is predominantly metabolized by
IL-2 in T-cells is independent of CB1R or CB2R but dependent on MAGL (Blankman et al., 2007; Long, Nomura, & Cravatt, 2009; Nomura
PPARγ (Rockwell, Snider, Thompson, Vanden Heuvel, & Kaminski, et al., 2011), inactivation of MAGL would produce “a dual hit,” which
2006). These findings suggest that resolving inflammation by 2-AG would augment anti-inflammatory and neuroprotective 2-AG signaling,
may involve CB1R/2R-dependent and independent mechanisms and and concurrently reduce proinflammatory and neurotoxic eicosanoids
that PPARγ is a downstream signaling mediator of CB1R/2R in 2-AG- (Chen, 2023; Hu et al., 2022). In addition, prolonged inactivation of
produced anti-inflammatory and neuroprotective effects (Chen, 2023; MAGL with pharmacological inhibitors or genetic knockout of MAGL
Chen et al., 2012; Du et al., 2011; Hu et al., 2022; Xu & Chen, 2015; has been shown to enhance LTP and spatial learning and memory
Zhang et al., 2014). (Lysenko et al., 2014; Pan et al., 2011; Zhang et al., 2015). Studies
Several previous studies revealed that 2-AG, but not AEA, is re- using pharmacological or genetic inactivation of MAGL to inhibit 2-AG
leased in the brain following TBI, infusion of Aβ, or by high frequency metabolism in experimental animal models have provided evidence
stimulation that induces long-term potentiation (LTP) in brain slices suggesting that MAGL is a promising therapeutic target for neurodegen-
(Panikashvili et al., 2001; Stella et al., 1997; van der Stelt et al., erative diseases (Chen, 2016, 2022; Chen et al., 2012; Gil-Ordonez,
2006). Correspondingly, exogenous administration of 2-AG or aug- Martin-Fontecha, Ortega-Gutierrez, & Lopez-Rodriguez, 2018; Grabner
mentation of 2-AG levels by inactivation of MAGL to prevent 2-AG et al., 2017; Mulvihill & Nomura, 2013; Wenzel & Klegeris, 2018; Xu &
breakdown attenuates TBI-induced neuroinflammation and neuropa- Chen, 2015; Zanfirescu, Ungurianu, Mihai, Radulescu, & Nitulescu,
thology (Hu et al., 2022; Katz et al., 2015; Panikashvili et al., 2001; Piro 2021; Zhu, Gao, & Chen, 2021).
et al., 2018; Zhang, Teng, Song, Hu, & Chen, 2015), suggesting that re-
lease of 2-AG is an endogenous defense mechanism to increase resil- 4.1. Alzheimer’s disease
ience of the brain in response to injury, infection, or other stimuli,
thereby maintaining brain homeostasis. Thus, it is likely that 2-AG Alzheimer’s disease (AD) is the most common cause of dementia
maintains brain homeostatic conditions primarily through modulat- in the elderly. It is a neurodegenerative disease characterized by
ing synaptic transmission and plasticity, resolving neuroinflamma- accumulation and deposition of extracellular Aβ plaques and intra-
tion, and protecting neurons from harmful insults (Chen, 2015, cellular hyperphosphorylated tau proteins that form neurofibrillary
2023; Mulvihill & Nomura, 2013; Xu & Chen, 2015). tangles, sustained neuroinflammation, synaptic malfunction, pro-
gressive deterioration of cognitive function, and loss of memory in
4. MAGL in neurodegenerative diseases association with widespread neuronal death. No effective therapies
are currently available for preventing or treating AD or delaying pro-
MAGL was originally purified from adipose tissues and recog- gression of the disease. This lack is largely attributed to our limited
nized as the rate-limiting enzyme involved in the last step of hydro- understanding of the mechanisms underlying development of AD.
lyzing triacylglycerol to fatty acid and glycerol in white adipose While the etiology of AD is multifactorial and complex and is linked
tissue (Fredrikson, Tornqvist, & Belfrage, 1986; Tornqvist & to multiple mechanisms and signaling pathways, it is generally ac-
Belfrage, 1976; Zechner, Kienesberger, Haemmerle, Zimmermann, cepted that chronic inflammation as a root cause significantly con-
& Lass, 2009). Triacylglycerol is first cleaved by adipose triglyceride tributes to pathogenesis of AD (Akiyama et al., 2000; Heneka et al.,
lipase (ATGL) to yield diacylglycerols (DAGs), which are then con- 2015; Kwon & Koh, 2020). Since 2-AG displays profound anti-
verted by hormone-sensitive lipase (HSL) into monoacylglycerols. inflammatory and neuroprotective properties and its hydrolytic

7
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

metabolites are proinflammatory and neurotoxic (X. Chen et al., patients with AD at advanced stages. However, 2-AG signaling and the
2011; Du et al., 2011; Henderson Jr., 1994; Mulvihill & Nomura, levels of DAGLα, which synthesizes 2-AG, are increased in the vicinity
2013; Panikashvili et al., 2006; Panikashvili et al., 2001; Ricciotti & of Aβ plaques in the brains of AD patients (Mulder et al., 2011). This
FitzGerald, 2011; Salmon & Higgs, 1987; Yao & Narumiya, 2019; might be a homeostatic defense mechanism as infusion of Aβ into the
Zhang & Chen, 2008; Zhang et al., 2014), investigators have asked brain or brain trauma has been observed to trigger release of 2-AG,
whether inhibition of 2-AG degradation by inactivation of MAGL which might protect the brain from injury and insults (Panikashvili
would prevent or modify AD neuropathology in experimental animal et al., 2001; van der Stelt et al., 2006). Elevated expression or activity
models of AD. Systemic administration of JZL184, a potent and irre- of MAGL in the hippocampus of patients with AD and of animal models
versible MAGL inhibitor (Long et al., 2009; Long, Nomura, & of AD has also been reported and elevation of MAGL proceeds in an age-
Cravatt, 2009), resulted in of 6 to 7-fold increases in 2-AG levels in dependent manner (Farooqui, Liss, & Horrocks, 1988; Syal et al., 2020).
the brain, while AA and PGE 2 were reduced by roughly 75% and Given the role of MAGL in degradation of 2-AG, the observation of in-
50%, respectively in 5xFAD mice (Chen et al., 2012), a widely used creased expression of MAGL in aging and AD suggests that 2-AG degra-
animal model of AD (Oakley et al., 2006). Of significance, production dation in the brain is also increased in aging and AD as MAGL is the main
and accumulation of total Aβ and Aβ42 peptides and expression of β- enzyme hydrolyzing 2-AG (Blankman et al., 2007; Dinh, Carpenter,
site amyloid precursor protein cleaving enzyme 1 (BACE1), a key en- et al., 2002; Dinh, Freund, & Piomelli, 2002; Long, Nomura, & Cravatt,
zyme responsible for synthesis of Aβ, were significantly reduced in 2009; Nomura et al., 2011). This remains speculative, however, due to
5xFAD mice (Chen et al., 2012). Chronic inhibition of MAGL also de- the difficulty in obtaining brain tissues or cerebrospinal fluid from pa-
creases reactivity of astrocytes and microglia and reduces the num- tients with AD for analysis. The levels of plasma 2-AG in patients with
ber of degenerating neurons. Importantly, AD mice treated with AD have been reported previously, but the results are inconsistent.
JZL184 display improved long-term synaptic plasticity in terms of One study showed increased plasma levels of 2-AG, and one did not
long-term potentiation (LTP), spatial learning and memory. These (Altamura et al., 2015; Koppel et al., 2009). Studies that assessed brain
synaptic and cognitive protective effects of MAGL inhibition are 2-AG in AD animals also arrived at conflicting results. Brain 2-AG levels
likely associated with the rescue of the reduced expression of gluta- were not altered in 5xFAD TG mice (Vázquez et al., 2015), but in the
mate receptor subunits and density of dendritic spines (Chen et al., AβPPswe/PS1ΔE9 mouse model of AD, there are no differences in 2-
2012). At the same time, another group observed similar anti- AG levels in the hippocampus and frontal cortex between WT and APP
inflammation and neuroprotective effects following genetic deletion TG mice. However, 2-AG was significantly reduced in the striatum of
of mgll (the gene encoding MAGL) in PS1/APP+ mice. Genetic inacti- 8-month-old mice (Maroof, Ravipati, Pardon, Barrett, & Kendall,
vation of mgll reduces total Aβ, Aβ40 and Aβ42, cytokine production, 2014). Another study provided evidence that there is an age-related de-
including IL-1β, IL-6, and TNFα, and expression of astrocyte and mi- cline in 2-AG but not AEA in the hippocampus of CD1 and C57BL/6
croglia markers (Piro et al., 2012). These beneficial effects are similar (Piyanova et al., 2015). Thus, the status of 2-AG in the brains of patients
to those of pharmacological inactivation of MAGL (Chen et al., 2012). with AD and animal models of AD remain uncertain.
Several other studies also provided evidence that pharmacological
inactivation of MAGL reduces neuroinflammation and Aβ and im- 4.2. Parkinson’s disease
proves learning and memory in APP TG mice (Pihlaja et al., 2015;
Zhang et al., 2014; Zhang & Chen, 2018). Since accumulation and de- Parkinson’s disease (PD) is another devastating neurodegenerative
position of extracellular Aβ plaques are one of the neuropathological disease resulting from loss of neurons that produce dopamine in the
hallmarks in AD, the early studies were primarily focused on resolu- substantia nigra. Neuroinflammation and immune dysfunctions are
tion of neuroinflammation and reduction of amyloidosis by inactiva- well recognized as prominent features in PD (Tansey et al., 2022).
tion of MAGL in animal models of Aβ pathology (Chen et al., 2012; While only a few studies involving MAGL in PD have been reported,
Pihlaja et al., 2015; Piro et al., 2012; Zhang & Chen, 2018; Zhang these studies provide clear evidence that limiting 2-AG degradation by
et al., 2014). However, intracellular accumulation of neurofibrillary inactivation of MAGL is neuroprotective in animal models of PD. Using
tangles consisting primarily of hyperphosphorylated tau (p-tau) the 1-methyl-4-phenyl-tetrahydropyridine (MPTP) mouse model of
proteins is another important neuropathological hallmark of AD. PD, it was found that pharmacological or genetic inactivation of MAGL
Using the P301S/PS19 mice, a widely used tau mouse model of AD reduces MPTP-induced dopaminergic neurodegeneration and elevates
(Yoshiyama et al., 2007), a recent study provided evidence that phar- dopamine levels in the striatum and substantia nigra (Nomura et al.,
macological inactivation of MAGL also mitigates neuroinflammation, 2011). Chronic treatment with MAGL inhibitor JZL184 prevents MPTP-
including reduced expression of cytokines, reduced reactivity of as- induced motor impairment and preserves the nigrostriatal pathway
trocytes and microglia, and reduced phosphorylated NF-κB. These ef- (Fernández-Suárez et al., 2014). Increasing levels of 2-AG by direct ad-
fects might lead to decreases in tauopathies, including decreases in ministration of exogenous 2-AG or by pharmacological inactivation of
phosphorylated GSK3β, p-tau Thr181, and p-tau Ser202/Thr205 MAGL to restrain endogenous 2-AG degradation provide neuroprotec-
(AT8) (Hashem, Hu, Zhang, Gao, & Chen, 2021). Importantly, inacti- tion against MPTP-induced cell death (Mounsey et al., 2015). In addition,
vation of MAGL reduces cell apoptosis and prevents losses in expres- pharmacological inhibition of MAGL, but not of FAAH, attenuates MPTP-
sion of synaptic proteins, including PSD-95 and glutamate receptor induced effects on striatal dopamine levels, which is accompanied by an
subunits, and prevents deterioration in cognitive function in PS19 increase in 2-AG levels (Pasquarelli et al., 2017). The neuroprotective ef-
tau TG mice (Hashem et al., 2021). Alleviation of both Aβ and tau pa- fects of MAGL inactivation are likely associated with neuroprotective and
thologies in APP and tau mouse models of AD by inactivation of anti-inflammatory factors released from astrocytes and microglial cells,
MAGL suggests that MAGL is a promising therapeutic target for AD and CB1R and CB2R in astrocytes and microglial cells may be involved
(Chen, 2022; Chen et al., 2012; Gil-Ordonez et al., 2018; Grabner in mediating these effects (Bernal-Chico et al., 2023; Fernández-Suárez
et al., 2017; Hashem et al., 2021; Mulvihill & Nomura, 2013; Zhang et al., 2014). The results from these studies suggest that modulation of
et al., 2014). 2-AG levels by manipulation of MAGL may provide an approach for pre-
Other studies also provide evidence to support MAGL as a therapeu- vention and treatment of this movement disorder.
tic target for AD. Immunoblot analysis shows that expression of MAGL
in the post-mortem hippocampus from patients with AD is significantly 4.3. Traumatic brain injury
elevated and the expression level exhibits a positive correlation with
the Braak stage, which represents AD progression (Mulder et al., TBI is a temporary or permanent disruption of brain function caused
2011), suggesting that 2-AG degradation is escalated in the brain of by external forces. TBI has been recognized as an important risk factor

8
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

for development of AD and dementia later in life (Dams-O'Connor, These neuroprotective effects produced by MAGL inhibitors likely result
Guetta, Hahn-Ketter, & Fedor, 2016; Fleminger, Oliver, Lovestone, from enhanced 2-AG signaling and concurrently decreased eicosanoid
Rabe-Hesketh, & Giora, 2003; Graves et al., 1990; Johnson, Stewart, & levels, suggesting that control of 2-AG degradation by MAGL is an im-
Smith, 2010; Mortimer, French, Hutton, & Schuman, 1985; Zhu et al., portant mechanism in maintaining brain homeostasis and in terminat-
2021). Single severe or repeated TBI may induce chronic traumatic en- ing neuroinflammation that occurs in TBI.
cephalopathy (CTE), a long-term progressive AD-like neurodegenera- Pharmacological inactivation of MAGL resolves neuroinflammation,
tive disease, which includes hyperphosphorylated tau, aggregation reduces neuropathology, and improves synaptic and cognitive functions
and translocation of transactive response DNA binding protein 43 in TBI. However, it remained unclear whether genetic inactivation of
(TDP-43), persistent neuroinflammation, neurodegeneration, and syn- MAGL would produce neuroprotective effects similar to those of phar-
aptic and cognitive declines (Al-Dahhak, Khoury, Qazi, & Grossberg, macological inhibition. A recent study by Hu and colleagues has filled
2018; Blennow, Hardy, & Zetterberg, 2012; DeKosky, Blennow, this gap (Hu et al., 2022). In their study, an mgllflox/flox knockout (KO)
Ikonomovic, & Gandy, 2013; Gao, Hu, Zhang, Hashem, & Chen, 2022; mouse model was used to generate total (tKO), neuronal (nKO), and as-
Li, Liang, & Fu, 2021; McKee et al., 2013; Turner, Lucke-Wold, Robson, trocytic (aKO) MAGL KO mice. In wild-type (WT) mice, repeated mild
Lee, & Bailes, 2016; Wu et al., 2021). However, no effective therapies CHI resulted in chronic neuroinflammation, excessive production of
are currently available for prevention or treatment of TBI-induced neu- TDP-43 and phosphorylated tau, neurodegeneration, deterioration in
rodegenerative disease. The acute brain damage after TBI results not expression of synaptic proteins, and impairments in LTP and cognitive
only from the primary injury, which is the result of an external mechan- function. These TBI-induced changes were diminished in tKO and aKO
ical force, but also from the secondary injury associated with a complex mice. However, inactivation of MAGL in neurons did not produce
cascade of molecular, cellular and immune responses (Zhu et al., 2021). these neuroprotective effects (Hu et al., 2022). These findings suggest
Neuroinflammation plays a critical role in causing secondary brain in- that previously observed neuroprotective effects produced by pharma-
jury following TBI (Simon et al., 2017). The extent of the neuroinflam- cological inactivation of MAGL in TBI result largely from restraining 2-
matory response is closely correlated with the outcome following TBI AG degradation in astrocytes, rather than in neurons (Chen, 2023;
(Woodcock & Morganti-Kossmann, 2013). The implication of this is Fucich et al., 2020; Hu et al., 2022; Katz et al., 2015; Mayeux et al.,
that while the primary injury from TBI may not be preventable, appro- 2017; Selvaraj et al., 2021; Zhang, Teng, et al., 2015). Moreover, the au-
priate and timely intervention to resolve neuroinflammation following thors observed that expression of MAGL in astrocytes was significantly
the primary injury would be key to preventing further brain damage, increased in hippocampal astrocytes following TBI, suggesting that 2-
neuropathological changes, and synaptic and cognitive declines (Xu & AG degradation was likely increased, promoting neuroinflammation
Chen, 2015; Zhang, Teng, et al., 2015; Zhu et al., 2021). Because of the and neuropathology (Hu et al., 2022).
profound anti-inflammatory and neuroprotective property of 2-AG, a
boost of endogenous 2-AG or administration of exogenous 2-AG 4.4. Multiple sclerosis
would likely ameliorate TBI-induced neuroinflammation. Indeed, early
studies showed that application of synthetic 2-AG produces CB1R- Multiple sclerosis (MS) is an incurable, chronic inflammatory disor-
dependent maintenance of the integrity of the blood-brain barrier der of the central nervous system. It is traditionally considered to be an
(BBB), reduction of proinflammatory cytokines, and suppression of autoimmune demyelinating disease. A 2020 study estimated that more
NF-κB activation, providing neuroprotection against CHI (Panikashvili than 2.8 million people are living with MS worldwide (Walton et al.,
et al., 2001; Panikashvili et al., 2005; Panikashvili et al., 2006). These 2020). In consideration of the anti-inflammatory properties of 2-AG,
studies provided evidence that 2-AG signaling plays an important role several studies have been conducted to assess the effect of inactivation
in protecting the brain from trauma by resolving TBI-induced neuroin- of MAGL on MS (Brindisi et al., 2016; Chiurchiù, van der Stelt, Centonze,
flammation (Mechoulam et al., 2002; Shohami et al., 2011). & Maccarrone, 2018; Punt, van der Vliet, & van der Stelt, 2022; van
As 2-AG is subject to rapid degradation, a good strategy to enhance Egmond, Straub, & van der Stelt, 2021). It was found that administration
2-AG signaling is to inhibit its degradation. It has been shown that a sin- of JZL184 under a therapeutic regimen decreased clinical severity,
gle injection of JZL184 following TBI in rats improves neurological and prevented demyelination, and reduced inflammation in chronic exper-
behavioral functions and protects the integrity of BBB assessed 24 imental autoimmune encephalomyelitis (Bernal-Chico et al., 2015). In
hours after lateral fluid percussion (Katz et al., 2015). In a mouse addition, MAGL inactivation also robustly preserved myelin integrity
model of repeated mild CHI, it was found that multiple injections of and suppressed microglial activation in the cuprizone-induced model
JZL184 following the impact promoted neurologic recovery, reduced ex- of T-cell-independent demyelination (Bernal-Chico et al., 2015). Inacti-
pression of proinflammatory cytokines, and reduced the reactivity of vation of MAGL with different inhibitors also reduces neuroinflamma-
astroglial cells (Zhang, Teng, et al., 2015). Importantly, pharmacological tion, decreases chondroitin sulfate proteoglycan deposition around
inactivation of MAGL significantly suppressed TBI-induced increases in demyelinated lesions in the spinal cord of Theiler's murine encephalo-
expression of BACE1, neurodegeneration, aberrant production of TDP- myelitis virus-infected mice, and ameliorates the clinical progression
43 protein, and phosphorylated tau. Inhibition of 2-AG degradation in a MS mouse model (Feliú et al., 2017; Hernández-Torres et al.,
also prevents TBI-induced decreases in expression of glutamate recep- 2014). The protective effects of MAGL inactivation are apparently asso-
tor subunits and reduces impairments in basal synaptic transmission, ciated with increased 2-AG tone, which promotes remyelination to
long-term synaptic plasticity, and spatial learning and memory in ani- ameliorate motor dysfunction in a model of progressive multiple sclero-
mals exposed to repeated mild CHI (Zhang, Teng, et al., 2015). These sis. These studies provide evidence that modulation of 2-AG signaling by
studies provide evidence that pharmacological inactivation of MAGL inactivating MAGL attenuates symptoms and promotes remyelination
produces neuroprotection against TBI-induced neuroinflammation, dis- in experimental models of MS, providing evidence supporting MAGL
ruption of BBB integrity, neuropathology, and synaptic and cognitive as a target for treatment of MS.
impairments (Katz et al., 2015; Xu & Chen, 2015; Zhang, Teng, et al.,
2015; Zhu et al., 2021). Several recent studies provide further evidence 4.5. Other brain disorders
that pharmacological modulation of 2-AG metabolism by different
MAGL inhibitors ameliorates TBI-induced neuronal and synaptic alter- Inactivation of MAGL is beneficial for other neurodegenerative dis-
ations, production of inflammatory cytokines, reactivity of astrocytes eases (e.g., amyotrophic lateral sclerosis), and other neurological or neu-
and microglia, glutamate dyshomeostasis, and impairments in spatial ropsychiatric disorders, including seizure/epilepsy and mental
learning and memory (Fucich et al., 2020; Mayeux, Katz, Edwards, disorders (Bedse, Hill, & Patel, 2020; Colangeli et al., 2023; Naidoo
Middleton, & Molina, 2017; Selvaraj, Tanaka, Wen, & Zhang, 2021). et al., 2012; Pan et al., 2011; Pasquarelli et al., 2017; Ratano et al.,

9
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

2018; Shonesy et al., 2014; Silveira, Wegener, & Joca, 2021; Terrone from these studies that the signaling pathways that mediate MAGL
et al., 2018; Vickstrom et al., 2021; von Ruden, Bogdanovic, Wotjak, & inactivation-induced neuroprotective effects are complex, and the sig-
Potschka, 2015; Zhang, Wang, et al., 2015; Zhong et al., 2014), which naling pathways involved in the protective events likely depend on
are not discussed here. the disease context (e.g., AD versus TBI).
Peroxisome proliferator-activated receptor-γ (PPARγ), a member of
5. Signaling pathways mediating neuroprotection following nuclear receptors, displays anti-inflammatory and neuroprotective ef-
inactivation of MAGL fects through interaction with NF-κB, resulting in reductions in tran-
scription and expression of genes involved in inflammation and
As 2-AG displays profound anti-inflammatory and neuroprotective neurodegeneration (Bensinger & Tontonoz, 2008; Bright, Kanakasabai,
properties, it is reasonable to ask whether augmentation of 2-AG signal- Chearwae, & Chakraborty, 2008; Heneka et al., 2005; Khan et al., 2019;
ing by inactivating MAGL would resolve inflammatory responses, Villapol, 2018). Earlier studies showed that PPARγ is a target of canna-
thereby preventing or treating some brain disorders. Not surprisingly, binoids via CB1R- or CB2R-dependent and -independent pathways
MAGL has been proposed as a therapeutic target for treating neurode- (O'Sullivan, 2007, 2016). It has been shown that 2-AG suppression of
generative diseases (Baggelaar et al., 2018; Bajaj, Jain, Vyas, Bawa, & IL-1 in Jurkat T cells is not mediated via CB1R or CB2R but via activation
Vohora, 2021; Chen, 2022; Chen et al., 2012; Gil-Ordonez et al., 2018; of PPARγ (Rockwell et al., 2006). Du et al., reported that PPARγ is a tar-
Hashem et al., 2021; M. Hu et al., 2022; Mulvihill & Nomura, 2013; get of 2-AG in resolving neuroinflammation and neuroprotection
Nomura et al., 2011; Piro et al., 2012; Ren, Wang, Zhang, & Chen, (Du et al., 2011). Administration of 2-AG or MAGL inhibitors reduces
2020; Xu & Chen, 2015; Zhang & Chen, 2018; Zhang et al., 2014; LPS- or cytokine-induced increases in expression of COX-2, phosphory-
Zhang, Teng, et al., 2015; Zhu et al., 2021). However, the molecular lated NF-κB, and excitatory synaptic transmission in cultured hippo-
mechanisms underlying the neuroprotective effects of MAGL inactiva- campal neurons, and these effects are blocked by a CB1R antagonist or
tion in neurodegenerative diseases remain to be defined. Inflammation PPARγ antagonist but mimicked by 15d-PGJ2, an endogenous PPARγ
has been recognized as a common mechanism of disease (Chen, 2010), agonist (Du et al., 2011). The authors also observed that treatment of
and neuroinflammation is a known root cause of neurodegenerative the cultures with LPS or cytokines resulted in a decrease in expression
diseases (Heneka et al., 2015; McGeer, Rogers, & McGeer, 2016; of PPARγ, but the decrease was prevented by 2-AG or inactivation of
Ransohoff, 2016). Therefore, strategies to resolve neuroinflammation MAGL. These results provide evidence that the anti-inflammatory ef-
represent promising avenues toward preventing development of neu- fects of 2-AG or of MAGL inactivation, at least in vitro, are mediated
rodegenerative diseases. Because 2-AG displays anti-inflammatory and via CB1R and PPARγ signaling. Although anti-inflammatory and neuro-
neuroprotective properties (Chen, 2015, 2023; Du et al., 2011; protective effects of pharmacological or genetic inactivation of MAGL in
Panikashvili et al., 2005; Panikashvili et al., 2006; Panikashvili et al., animal models of AD are not mediated via CB1R or CB2R (Chen et al.,
2001; Song et al., 2015; Sugiura et al., 2006; Zhang & Chen, 2008; 2012; Piro et al., 2012; Zhang & Chen, 2018), the MAGL inactivation-
Zhang et al., 2014), but its metabolites (e.g., PGE2 and LTB4) are induced neuroprotective effects are eliminated by antagonism of
proinflammatory and neurotoxic and contribute to neurodegenerative PPARγ in 5xFAD APP mice (Zhang et al., 2014). This study also provides
diseases (Hein & O'Banion, 2009; Henderson Jr., 1994; Ricciotti & evidence that 2-AG is an endogenous PPARγ agonist, as 2-AG increases
FitzGerald, 2011; Salmon & Higgs, 1987; Xu & Chen, 2015; Yao & PPARγ luciferase activity, a finding which has been further supported by
Narumiya, 2019), inactivation of MAGL may provide the means to pro- a study showing that the 2-AG-mediated increase in activity of PPARγ
duce anti-inflammatory and neuroprotective effects through multiple luciferase is blocked by a CB1R or PPARγ antagonist (Hu et al., 2022).
signaling pathways in neurodegenerative diseases based on the proper- Importantly, the authors discovered that inactivation of MAGL in astro-
ties of its substrate 2-AG and the proinflammatory and neurotoxic prop- cytes prevents TBI-induced neuropathology, as well as synaptic and
erties of 2-AG hydrolytic metabolites. cognitive impairments and that the neuroprotective effects are dimin-
Early studies showed that 2-AG functions as an endogenous neuro- ished by silencing of PPARγ in astrocytes (Hu et al., 2022). They also ob-
protective mediator against brain trauma by a CB1R-dependent mecha- served that TBI-induced neuropathology and deficits in learning and
nism of resolution of inflammatory responses and by maintenance of memory in WT animals can be averted by overexpression of human
BBB integrity. It was shown that neuroprotective effects mediated by PPARγ in astrocytes. These studies provide evidence that PPARγ likely
2-AG were attenuated by a selective CB1R antagonist or by genetic de- acts downstream of CB1R in mediating 2-AG- or MAGL inactivation-
letion of CB1R (Panikashvili et al., 2001; Panikashvili et al., 2005; produced anti-inflammatory and neuroprotective effects in neurode-
Panikashvili et al., 2006). The CB1R-mediated anti-inflammatory and generative diseases (Chen, 2023; Du et al., 2011; Hu et al., 2022;
neuroprotective effects in TBI have been confirmed by a recent study Zhang et al., 2014). However, the precise mechanisms of how CB1R
in which pharmacological inactivation of MAGL in CB1R knockout interacts with PPARγ remain unclear, which warrant further
mice failed to produce neuroprotection in TBI (Hu et al., 2022). These investigation.
studies provide evidence that 2-AG- or MAGL inactivation-induced neu- Single cell transcriptomic analysis has shown that genetic inactiva-
roprotection in TBI is mediated via CB1R. However, alleviation of neuro- tion of MAGL alters expression of immune- and inflammation-related
inflammation in an LPS-induced animal model of inflammation and genes in microglia and astrocytes and enhances immune/inflammatory
neuronal loss in an MPTP-induced animal model of PD by pharmacolog- vigilance in these glial cells (Zhu, Zhang, Hashem, Gao, & Chen, 2023).
ical or genetic inactivation of MAGL appeared to not involve 2-AG sig- This has been confirmed by showing that TBI-upregulated expression
naling to activate CB1R or CB2R; instead, these effects were mediated of genes associated with inflammation is significantly reduced in mice
through prostaglandin signaling as a result of decreases in AA and PGs lacking MAGL in astrocytes, while TBI-downregulated expression of
(Nomura et al., 2011). Consistent with these observations, anti- genes associated with anti-inflammation or maintaining brain homeo-
inflammatory and neuroprotective effects by pharmacological or ge- stasis is not significantly changed in astrocytic mgll KO mice (Hu et al.,
netic inactivation of MAGL in animal models of AD are also not mediated 2022). Of note, TBI-upregulated expression of genes associated with in-
via CB1R nor via CB2R (Chen et al., 2012; Piro et al., 2012; Zhang & Chen, flammation in astrocytic MAGL knockout mice is robustly reduced in
2018). There are reports that restorative astroglia and microglia activa- microglia, and TBI-increased microglial reactivity is also reduced, sug-
tion and the release of neuroprotective and anti-inflammatory mole- gesting that 2-AG generated in astrocytes is an important signaling me-
cules may contribute to MAGL inactivation-produced neuroprotection diator that interacts with microglia to terminate microglia-mediated
in PD animals (Fernández-Suárez et al., 2014). In an animal model of inflammatory responses. These observations provide evidence that
MS, MAGL inactivation-induced protection of oligodendrocytes was as- restraining 2-AG metabolism in astrocytes enhances resilience to brain
sociated with activation of CB1R (Bernal-Chico et al., 2015). It appears trauma by suppressing TBI-induced upregulation of genes associated

10
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

with inflammation and reducing TBI-induced downregulation of genes (Blankman et al., 2007; Burston et al., 2008; Hohmann et al., 2005;
with anti-inflammatory properties or genes involved in maintenance King et al., 2007; Saario et al., 2005). However, JZL184, which was
of brain homeostasis. In line with these observations, a previous study invented by the Benjamin Cravatt’s group (Long, Li, et al., 2009; Long,
demonstrated that AA and PGs derived from 2-AG metabolism primar- Nomura, & Cravatt, 2009; Schlosburg et al., 2010; Schlosburg et al.,
ily result from 2-AG synthesized in astrocytes but not in neurons 2014), is the most widely used MAGL inhibitor in experimental animals
(Nomura et al., 2011), which might be one of the mechanisms underly- because of its high selectivity and potency (Long, Li, et al., 2009; Long,
ing astrocytic MAGL inactivation-produced anti-inflammatory and neu- Nomura, & Cravatt, 2009; Schlosburg et al., 2010; Schlosburg et al.,
roprotective effects in neurodegenerative diseases (Fig. 6). This suggests 2014). Although many MAGL inhibitors have been identified, currently
that MAGL in astrocytes controls 2-AG signaling in neurodegenerative only ABX-1431 (Lu AG06466), which is an irreversible inhibitor for
diseases and inactivation of astrocytic MAGL resolves neuroinflamma- MAGL, is in clinical trials (Cisar et al., 2018; Jiang & van der Stelt,
tion and protects neurons by enhancement of 2-AG signaling and lower- 2018; Müller-Vahl et al., 2021; Punt et al., 2022). It is imperative to iden-
ing of the levels of 2-AG hydrolytic metabolites eicosanoids in astrocytes tify and develop novel MAGL inhibitors, especially reversible inhibitors,
(Fig. 6). that can be used for neurodegenerative diseases. MAGL inhibitors are
more thoroughly discussed in several reviews (Alhouayek et al., 2014;
6. MAGL inhibitors Bajaj et al., 2021; Bononi et al., 2021; Deng & Li, 2020; Gil-Ordonez
et al., 2018; Scalvini et al., 2016; van Egmond et al., 2021).
Giving that, in the brain, MAGL is the main enzyme that hydrolyzes
2-AG, and 2-AG exhibits profound anti-inflammatory and neuroprotec- 7. Perspective
tive properties, while its hydrolytic metabolites are proinflammatory
and neurotoxic, MAGL has emerged as an attractive drug target for Since 2-AG displays profound anti-inflammatory and neuroprotec-
brain disorders (Alhouayek et al., 2014; Baggelaar et al., 2018; Bajaj tive properties and 2-AG hydrolytic metabolites are proinflammatory
et al., 2021; Gil-Ordonez et al., 2018; Grabner et al., 2017; Mulvihill & and neurotoxic, restraint of 2-AG degradation to enhance 2-AG signal-
Nomura, 2013; van Egmond et al., 2021; Xu & Chen, 2015; Zhu et al., ing and to reduce its metabolites appear to provide a mechanism to ter-
2021). This has led to identification and development of several com- minate inflammatory responses, thereby preventing or treating brain
pounds that inhibit activity of MAGL. MAGL inhibitors are classified as disorders. Thus, MAGL has been proposed as a therapeutic target for
reversible or irreversible based on their mechanisms of action (Gil- neurodegenerative diseases (Alhouayek et al., 2014; Baggelaar et al.,
Ordonez et al., 2018; Scalvini et al., 2016). URB602 and N-arachidonyl 2018; Bajaj et al., 2021; Brindisi et al., 2016; Chen, 2022; Chen et al.,
maleimide (NAM) were the earliest identified MAGL inhibitors 2012; Cisar et al., 2018; Gil-Ordonez et al., 2018; Grabner et al., 2017;

Fig. 6. Signaling pathways that mediate astrocytic MAGL inactivation-produced neuroprotection in neurodegenerative diseases. Pharmacological or genetic inactivation of MAGL in astro-
cytes boosts 2-AG levels and reduces its immediate metabolite AA, leading to reduction of the levels of prostaglandins (PGs) and leukotrienes (LT4s). Enhanced 2-AG signaling activates
PPARγ directly or indirectly through binding to CB1R as well as increases expression of PPARγ. PPARγ interacts with NF-κB to inhibit its transcription and expression of genes involved in
inflammatory responses in astrocytes. Reduced release of inflammatory factors from astrocytes alleviates NF-κB-mediated neuropathology in neurons, including phosphorylated tau (p-
tau), synthesis of Aβ, and production of TDP-43. Reduced release of inflammatory factors from astrocytes also decreases NF-κB-mediated inflammatory cascades in microglia to suppress
production and release of cytokines, chemokines, complements, and prostaglandins (PGs) from microglia. It is still not clear whether increased 2-AG levels in astrocytes could activate
PPARγ directly or indirectly through binding to CB2R in microglial cells, leading to reduction of NF-κB-mediated transcription and expression of genes involved in inflammatory responses
in microglia. Reduced neuroinflammation, astrogliosis, and microgliosis by inactivation of MAGL in astrocytes prevent or ameliorate neuropathology, synaptic dysfunction, neurodegen-
eration, impairments in learning and memory through maintaining the brain in homeostasis (Chen, 2023).

11
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Hashem et al., 2021; Hu et al., 2022; Mulvihill & Nomura, 2013; Akiyama, H., Barger, S., Barnum, S., Bradt, B., Bauer, J., Cole, G. M., ... Wyss-Coray, T. (2000).
Inflammation and Alzheimer’s disease. Neurobiology of Aging 21, 383–421.
Pasquarelli, Engelskirchen, et al., 2017; Piomelli & Mabou Tagne, 2022; Al-Dahhak, R., Khoury, R., Qazi, E., & Grossberg, G. T. (2018). Traumatic brain injury,
Wenzel & Klegeris, 2018; Zhang & Chen, 2018; Zhang et al., 2014; chronic traumatic encephalopathy, and alzheimer disease. Clinics in Geriatric
Zhang, et al., 2015; Zhu et al., 2022). However, recent studies have Medicine 34, 617–635.
shown that global inactivation of MAGL induces some adverse effects, Alger, B. E. (2002). Retrograde signaling in the regulation of synaptic transmission: focus
on endocannabinoids. Progress in Neurobiology 68, 247–286.
including an increased incidence of lung adenocarcinoma, worsened Alhouayek, M., Masquelier, J., Cani, P. D., Lambert, D. M., & Muccioli, G. G. (2013). Implica-
heart function after acute myocardial infarction, impaired fine motor tion of the anti-inflammatory bioactive lipid prostaglandin D2-glycerol ester in the
coordination, and bone loss (R. Liu et al., 2018; Marino et al., 2020; control of macrophage activation and inflammation by ABHD6. Proceedings of the
National Academy of Sciences of the United States of America 110, 17558–17563.
Marino et al., 2019; Martinez-Torres et al., 2019; Schloss et al., 2019).
Alhouayek, M., Masquelier, J., & Muccioli, G. G. (2014). Controlling 2-arachidonoylglycerol
In addition, early studies also demonstrated that chronic inactivation metabolism as an anti-inflammatory strategy. Drug Discovery Today 19, 295–304.
of MAGL causes functional antagonism of the endocannabinoid system, Alhouayek, M., & Muccioli, G. G. (2014). COX-2-derived endocannabinoid metabolites as
primarily resulting from functional tolerance of CB1R (Chanda et al., novel inflammatory mediators. Trends in Pharmacological Sciences 35, 284–292.
Altamura, C., Ventriglia, M., Martini, M. G., Montesano, D., Errante, Y., Piscitelli, F., ... Di
2010; Imperatore et al., 2015; Schlosburg et al., 2010), which may Marzo, V. (2015). Elevation of plasma 2-arachidonoylglycerol levels in Alzheimer’s
reduce the effectiveness of MAGL inhibitors. A recent study by Hu disease patients as a potential protective mechanism against neurodegenerative de-
et al. revealed that inactivation of MAGL in neurons does not prevent cline. Journal of Alzheimers Disease 46, 497–506.
TBI-induced neuropathology or deficits in synaptic plasticity, learning Amin, M. R., & Ali, D. W. (2019). Pharmacology of medical cannabis. Advances in
Experimental Medicine and Biology 1162, 151–165.
and memory (Hu et al., 2022). In contrast, inactivation of MAGL in astro- Baggelaar, M. P., Maccarrone, M., & van der Stelt, M. (2018). 2-Arachidonoylglycerol: A
cytes produces anti-inflammatory and neuroprotective effects in TBI signaling lipid with manifold actions in the brain. Progress in Lipid Research 71, 1–17.
(Hu et al., 2022). Mass spectrometry analysis in global and cell type- Bajaj, S., Jain, S., Vyas, P., Bawa, S., & Vohora, D. (2021). The role of endocannabinoid path-
way in the neuropathology of Alzheimer's disease: can the inhibitors of MAGL and
specific mgll KO mice has demonstrated that most 2-AG in the brain is
FAAH prove to be potential therapeutic targets against the cognitive impairment as-
generated in neurons, and only small amounts of 2-AG are synthesized sociated with Alzheimer's disease? Brain Research Bulletin 174, 305–322.
in astrocytes (Hu et al., 2022; Viader et al., 2015). However, 2-AG syn- Bayer, H., Stern, C. A. J., Troyner, F., Gazarini, L., Guimarães, F. S., & Bertoglio, L. J. (2022).
thesized in astrocytes produces profound anti-inflammatory and neuro- Medial prefrontal cortex mechanisms of cannabidiol-induced aversive memory
reconsolidation impairments. Neuropharmacology 205, Article 108913.
protective effects despite the small amounts of 2-AG generated in Bedse, G., Hill, M. N., & Patel, S. (2020). 2-Arachidonoylglycerol modulation of anxiety and
astrocytes (Hu et al., 2022). A previous study also reported that inactiva- stress adaptation: from grass roots to novel therapeutics. Biological Psychiatry 88,
tion of MAGL in astrocytes attenuates LPS-induced production of proin- 520–530.
flammatory PGE2 and cytokines without causing cannabinoid receptor Bensinger, S. J., & Tontonoz, P. (2008). Integration of metabolism and inflammation by
lipid-activated nuclear receptors. Nature 454, 470–477.
desensitization (Grabner et al., 2016). In addition, a recent study Bernal-Chico, A., Canedo, M., Manterola, A., Victoria Sánchez-Gómez, M., Pérez-Samartín,
showed that overexpression of miR-30b, which targets molecules im- A., Rodríguez-Puertas, R., Matute, C., & Mato, S. (2015). Blockade of monoacylglycerol
portant for synapses (Song, Hu, Zhang, Teng, & Chen, 2019), alters syn- lipase inhibits oligodendrocyte excitotoxicity and prevents demyelination in vivo.
Glia 63, 163–176.
aptic transmission and impairs LTP, and these changes are reversed by
Bernal-Chico, A., Tepavcevic, V., Manterola, A., Utrilla, C., Matute, C., & Mato, S. (2023).
inactivation of MAGL in astrocytes, but not in neurons (Zhu et al., Endocannabinoid signaling in brain diseases: Emerging relevance of glial cells. Glia
2023). The findings from these studies suggest that neuroprotective 71, 103–126.
effects of global inactivation of MAGL by pharmacological inhibitors or Bisogno, T., Howell, F., Williams, G., Minassi, A., Cascio, M. G., Ligresti, A., ... Doherty, P.
(2003). Cloning of the first sn1-DAG lipases points to the spatial and temporal regu-
genetic deletion of mgll observed previously result primarily from lation of endocannabinoid signaling in the brain. The Journal of Cell Biology 163,
restraining degradation of 2-AG in astrocytes rather than in neurons 463–468.
(Hu et al., 2022). Thus, global inactivation of MAGL might not be an op- Blankman, J. L., Simon, G. M., & Cravatt, B. F. (2007). A comprehensive profile of brain en-
zymes that hydrolyze the endocannabinoid 2-arachidonoylglycerol. Chemistry &
timal approach to achieving ideal therapeutic goals for neurodegenera-
Biology 14, 1347–1356.
tive diseases. Perhaps targeting astroglial cells would be an ideal Blennow, K., Hardy, J., & Zetterberg, H. (2012). The neuropathology and neurobiology of
therapeutic strategy for neurological disorders (Lee, Wheeler, & traumatic brain injury. Neuron 76, 886–899.
Quintana, 2022; Santello, Toni, & Volterra, 2019). However, current Bononi, G., Poli, G., Rizzolio, F., Tuccinardi, T., Macchia, M., Minutolo, F., & Granchi, C.
(2021). An updated patent review of monoacylglycerol lipase (MAGL) inhibitors
pharmacotherapies do not have capacity to target a molecule in a spe- (2018-present). Expert Opinion on Therapeutic Patents 31, 153–168.
cific type of cells. Thus, development of nanoparticle-, exosome-, or Bright, J. J., Kanakasabai, S., Chearwae, W., & Chakraborty, S. (2008). PPAR regulation of in-
viral vector-mediated therapies might be approaches to target MAGL flammatory signaling in CNS diseases. PPAR Research 2008, Article 658520.
in specific type of cells. While inhibiting 2-AG degradation, which aug- Brindisi, M., Maramai, S., Gemma, S., Brogi, S., Grillo, A., Di Cesare Mannelli, L., ... Butini, S.
(2016). Development and pharmacological characterization of selective blockers of
ments 2-AG signaling and reduces hydrolytic metabolites of 2-AG, is be- 2-arachidonoyl glycerol degradation with efficacy in rodent models of multiple scle-
lieved to provide a therapeutic strategy for treating neurodegenerative rosis and pain. Journal of Medicinal Chemistry 59, 2612–2632.
diseases, deciphering the cell type-specific roles of 2-AG metabolism Burston, J. J., Sim-Selley, L. J., Harloe, J. P., Mahadevan, A., Razdan, R. K., Selley, D. E., &
Wiley, J. L. (2008). N-arachidonyl maleimide potentiates the pharmacological and
will enable us to develop more comprehensive therapeutic strategies
biochemical effects of the endocannabinoid 2-arachidonylglycerol through inhibition
for treating brain disorders (Chen, 2023). of monoacylglycerol lipase. Journal of Pharmacology and Experimental Therapeutics
327, 546–553.
Carlini, E. A. (2004). The good and the bad effects of (-) trans-delta-9-tetrahydrocannab-
inol (Delta 9-THC) on humans. Toxicon 44, 461–467.
Declaration of Competing Interest
Castillo, P. E., Younts, T. J., Chávez, A. E., & Hashimotodani, Y. (2012). Endocannabinoid sig-
naling and synaptic function. Neuron 76, 70–81.
The authors declare that there are no conflicts of interest. Chanda, P. K., Gao, Y., Mark, L., Btesh, J., Strassle, B. W., Lu, P., ... Samad, T. A. (2010).
Monoacylglycerol lipase activity is a critical modulator of the tone and integrity of
the endocannabinoid system. Molecular Pharmacology 78, 996–1003.
Acknowledgement
Chen, C. (2010). COX-2's new role in inflammation. Nature Chemical Biology 6, 401–402.
Chen, C. (2015). Homeostatic regulation of brain functions by endocannabinoid signaling.
This work was supported by National Institutes of Health grants Neural Regeneration Research 10, 691–692.
RF1NS076815 from NINDS and NIA, R01MH113535 from NIMH, and Chen, C. (2016). Endocannabinoid metabolism in neurodegenerative diseases.
Neuroimmunology & Neuroinflammation 3, 268–270.
R01AG058621 from NIA.
Chen, C. (2022). Endocannabinoid metabolism and Alzheimer's disease. Neural
Regeneration Research 17, 1987–1988.
References Chen, C. (2023). Endocannabinoid control of neuroinflammation in traumatic brain injury
by monoacylglycerol lipase in astrocytes. Neural Regeneration Research 18,
Aghazadeh Tabrizi, M., Baraldi, P. G., Baraldi, S., Gessi, S., Merighi, S., & Borea, P. A. (2017). 1023–1024.
Medicinal chemistry, pharmacology, and clinical implications of trpv1 receptor an- Chen, C., & Bazan, N. G. (2005). Lipid signaling: sleep, synaptic plasticity, and neuroprotec-
tagonists. Medicinal Research Reviews 37, 936–983. tion. Prostaglandins & Other Lipid Mediators 77, 65–76.

12
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Chen, R., Zhang, J., Fan, N., Teng, Z. Q., Wu, Y., Yang, H., ... Chen, C. (2013). Delta9-THC- and alters glial cell phenotype in the chronic MPTP mouse model. Neurobiology of
caused synaptic and memory impairments are mediated through COX-2 signaling. Aging 35, 2603–2616.
Cell 155, 1154–1165. Ferrer, M. D., Busquets-Cortés, C., Capó, X., Tejada, S., Tur, J. A., Pons, A., & Sureda, A.
Chen, R., Zhang, J., Wu, Y., Wang, D., Feng, G., Tang, Y. P., ... Chen, C. (2012). (2019). Cyclooxygenase-2 inhibitors as a therapeutic target in inflammatory diseases.
Monoacylglycerol lipase is a therapeutic target for Alzheimer’s disease. Cell Reports Current Medicinal Chemistry 26, 3225–3241.
2, 1329–1339. Fiskerstrand, T., H’Mida-Ben Brahim, D., Johansson, S., M’Zahem, A., Haukanes, B. I.,
Chen, X., Zhang, J., & Chen, C. (2011). Endocannabinoid 2-arachidonoylglycerol protects Drouot, N., ... Knappskog, P. M. (2010). Mutations in ABHD12 cause the neurodegen-
neurons against beta-amyloid insults. Neuroscience 178, 159–168. erative disease PHARC: An inborn error of endocannabinoid metabolism. American
Chevaleyre, V., & Castillo, P. E. (2003). Heterosynaptic LTD of hippocampal GABAergic Journal of Human Genetics 87, 410–417.
synapses: a novel role of endocannabinoids in regulating excitability. Neuron 38, Fitzgerald, K. T., Bronstein, A. C., & Newquist, K. L. (2013). Marijuana poisoning. Topics in
461–472. Companion Animal Medicine 28, 8–12.
Chevaleyre, V., & Castillo, P. E. (2004). Endocannabinoid-mediated metaplasticity in the Fleminger, S., Oliver, D. L., Lovestone, S., Rabe-Hesketh, S., & Giora, A. (2003). Head injury
hippocampus. Neuron 43, 871–881. as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication.
Chiurchiù, V., van der Stelt, M., Centonze, D., & Maccarrone, M. (2018). The endocannabi- Journal of Neurology, Neurosurgery, and Psychiatry 74, 857–862.
noid system and its therapeutic exploitation in multiple sclerosis: Clues for other Fredrikson, G., Tornqvist, H., & Belfrage, P. (1986). Hormone-sensitive lipase and
neuroinflammatory diseases. Progress in Neurobiology 160, 82–100. monoacylglycerol lipase are both required for complete degradation of adipocyte tri-
Cisar, J. S., Weber, O. D., Clapper, J. R., Blankman, J. L., Henry, C. L., Simon, G. M., ... Grice, C. acylglycerol. Biochimica et Biophysica Acta 876, 288–293.
A. (2018). Identification of ABX-1431, a selective inhibitor of monoacylglycerol lipase Freund, T. F., & Hajos, N. (2003). Excitement reduces inhibition via endocannabinoids.
and clinical candidate for treatment of Neurological disorders. Journal of Medicinal Neuron 38, 362–365.
Chemistry 61, 9062–9084. Freund, T. F., Katona, I., & Piomelli, D. (2003). Role of endogenous cannabinoids in synap-
Colangeli, R., Morena, M., Werner, A., Thompson, R. J., van der Stelt, M., Pittman, Q. J., ... tic signaling. Physiological Reviews 83, 1017–1066.
Teskey, G. C. (2023). 2-AG-mediated control of GABAergic signaling is impaired in a Fucich, E. A., Stielper, Z. F., Cancienne, H. L., Edwards, S., Gilpin, N. W., Molina, P. E., &
model of epilepsy. Journal of Neuroscience 43, 571–583. Middleton, J. W. (2020). Endocannabinoid degradation inhibitors ameliorate neuro-
Cristino, L., Bisogno, T., & Di Marzo, V. (2020). Cannabinoids and the expanded endocan- nal and synaptic alterations following traumatic brain injury. Journal of
nabinoid system in neurological disorders. Nature Reviews. Neurology 16, 9–29. Neurophysiology 123, 707–717.
Dams-O’Connor, K., Guetta, G., Hahn-Ketter, A. E., & Fedor, A. (2016). Traumatic brain in- Gao, F., Hu, M., Zhang, J., Hashem, J., & Chen, C. (2022). TDP-43 drives synaptic and cogni-
jury as a risk factor for Alzheimer’s disease: current knowledge and future directions. tive deterioration following traumatic brain injury. Acta Neuropathologica 144,
Neurodegenerative Disease Management 6, 417–429. 187–210.
DeKosky, S. T., Blennow, K., Ikonomovic, M. D., & Gandy, S. (2013). Acute and chronic Gao, Y., Vasilyev, D. V., Goncalves, M. B., Howell, F. V., Hobbs, C., Reisenberg, M., ... Doherty,
traumatic encephalopathies: pathogenesis and biomarkers. Nature Reviews. P. (2010). Loss of retrograde endocannabinoid signaling and reduced adult
Neurology 9, 192–200. neurogenesis in diacylglycerol lipase knock-out mice. Journal of Neuroscience 30,
2017–2024.
Deng, H., & Li, W. (2020). Monoacylglycerol lipase inhibitors: modulators for lipid metab-
olism in cancer malignancy, neurological and metabolic disorders. Acta Gerdeman, G. L., Ronesi, J., & Lovinger, D. M. (2002). Postsynaptic endocannabinoid re-
Pharmaceutica Sinica B 10, 582–602. lease is critical to long-term depression in the striatum. Nature Neuroscience 5,
446–451.
Devane, W. A., Dysarz, F. A., 3rd, Johnson, M. R., Melvin, L. S., & Howlett, A. C. (1988). De-
Gil-Ordonez, A., Martin-Fontecha, M., Ortega-Gutierrez, S., & Lopez-Rodriguez, M. L.
termination and characterization of a cannabinoid receptor in rat brain. Molecular
(2018). Monoacylglycerol lipase (MAGL) as a promising therapeutic target.
Pharmacology 34, 605–613.
Biochemical Pharmacology 157, 18–32.
Devane, W. A., Hanus, L., Breuer, A., Pertwee, R. G., Stevenson, L. A., Griffin, G., ...
Gonsiorek, W., Lunn, C., Fan, X., Narula, S., Lundell, D., & Hipkin, R. W. (2000). Endocanna-
Mechoulam, R. (1992). Isolation and structure of a brain constituent that binds to
binoid 2-arachidonyl glycerol is a full agonist through human type 2 cannabinoid re-
the cannabinoid receptor. Science 258, 1946–1949.
ceptor: antagonism by anandamide. Molecular Pharmacology 57, 1045–1050.
Di Marzo, V., Stella, N., & Zimmer, A. (2015). Endocannabinoid signalling and the deteri-
Grabner, G. F., Eichmann, T. O., Wagner, B., Gao, Y., Farzi, A., Taschler, U., ... Zimmermann,
orating brain. Nature Reviews. Neuroscience 16, 30–42.
R. (2016). Deletion of Monoglyceride Lipase in Astrocytes Attenuates
Diana, M. A., & Marty, A. (2004). Endocannabinoid-mediated short-term synaptic plastic-
Lipopolysaccharide-induced Neuroinflammation. The Journal of Biological Chemistry
ity: depolarization-induced suppression of inhibition (DSI) and depolarization-
291, 913–923.
induced suppression of excitation (DSE). British Journal of Pharmacology 142, 9–19.
Grabner, G. F., Zimmermann, R., Schicho, R., & Taschler, U. (2017). Monoglyceride lipase as
Dinh, T. P., Carpenter, D., Leslie, F. M., Freund, T. F., Katona, I., Sensi, S. L., ... Piomelli, D.
a drug target: At the crossroads of arachidonic acid metabolism and endocannabinoid
(2002). Brain monoglyceride lipase participating in endocannabinoid inactivation.
signaling. Pharmacology & Therapeutics 175, 35–46.
Proceedings of the National Academy of Sciences of the United States of America 99,
Graves, A. B., White, E., Koepsell, T. D., Reifler, B. V., van Belle, G., Larson, E. B., & Raskind,
10819–10824.
M. (1990). The association between head trauma and Alzheimer’s disease. American
Dinh, T. P., Freund, T. F., & Piomelli, D. (2002). A role for monoglyceride lipase in 2- Journal of Epidemiology 131, 491–501.
arachidonoylglycerol inactivation. Chemistry and Physics of Lipids 121, 149–158.
Guindon, J., & Hohmann, A. G. (2008). A physiological role for endocannabinoid-derived
Du, H., Chen, X., Zhang, J., & Chen, C. (2011). Inhibition of COX-2 expression by endocan- products of cyclooxygenase-2-mediated oxidative metabolism. British Journal of
nabinoid 2-arachidonoylglycerol is mediated via PPAR-gamma. British Journal of Pharmacology 153, 1341–1343.
Pharmacology 163, 1533–1549. Gulyas, A. I., Cravatt, B. F., Bracey, M. H., Dinh, T. P., Piomelli, D., Boscia, F., & Freund, T. F.
Duggan, K. C., Hermanson, D. J., Musee, J., Prusakiewicz, J. J., Scheib, J. L., Carter, B. D., ... (2004). Segregation of two endocannabinoid-hydrolyzing enzymes into pre- and
Marnett, L. J. (2011). (R)-Profens are substrate-selective inhibitors of endocannabi- postsynaptic compartments in the rat hippocampus, cerebellum and amygdala.
noid oxygenation by COX-2. Nature Chemical Biology 7, 803–809. European Journal of Neuroscience 20, 441–458.
van Egmond, N., Straub, V. M., & van der Stelt, M. (2021). Targeting endocannabinoid sig- Haj-Dahmane, S., Shen, R. Y., Elmes, M. W., Studholme, K., Kanjiya, M. P., Bogdan, D., ...
naling: FAAH and MAG lipase inhibitors. Annual Review of Pharmacology and Kaczocha, M. (2018). Fatty-acid-binding protein 5 controls retrograde endocannabi-
Toxicology 61, 441–463. noid signaling at central glutamate synapses. Proceedings of the National Academy of
Egmond, N. V., Straub, V. M., & der Stelt, M. V. (2021). Targeting endocannabinoid signal- Sciences of the United States of America 115, 3482–3487.
ing: FAAH and MAG lipase inhibitors. Annual Review of Pharmacology and Toxicology Hashem, J., Hu, M., Zhang, J., Gao, F., & Chen, C. (2021). Inhibition of 2-
61, 441–463. arachidonoylglycerol metabolism alleviates neuropathology and improves cognitive
Espejo-Porras, F., Fernández-Ruiz, J., Pertwee, R. G., Mechoulam, R., & García, C. (2013). function in a tau mouse model of Alzheimer's disease. Molecular Neurobiology 58,
Motor effects of the non-psychotropic phytocannabinoid cannabidiol that are medi- 4122–4133.
ated by 5-HT1A receptors. Neuropharmacology 75, 155–163. Hashimotodani, Y., Ohno-Shosaku, T., & Kano, M. (2007). Presynaptic monoacylglycerol
Facchinetti, F., Del Giudice, E., Furegato, S., Passarotto, M., & Leon, A. (2003). Cannabinoids lipase activity determines basal endocannabinoid tone and terminates retrograde
ablate release of TNFalpha in rat microglial cells stimulated with lypopolysaccharide. endocannabinoid signaling in the hippocampus. Journal of Neuroscience 27,
Glia 41, 161–168. 1211–1219.
Farooqui, A. A., Liss, L., & Horrocks, L. A. (1988). Stimulation of lipolytic enzymes in Heifets, B. D., & Castillo, P. E. (2009). Endocannabinoid signaling and long-term synaptic
Alzheimer’s disease. Annals of Neurology 23, 306–308. plasticity. Annual Review of Physiology 71, 283–306.
Felder, C. C., Briley, E. M., Axelrod, J., Simpson, J. T., Mackie, K., & Devane, W. A. (1993). Hein, A. M., & O’Banion, M. K. (2009). Neuroinflammation and memory: the role of pros-
Anandamide, an endogenous cannabimimetic eicosanoid, binds to the cloned taglandins. Molecular Neurobiology 40, 15–32.
human cannabinoid receptor and stimulates receptor-mediated signal transduction. Henderson, W. R., Jr. (1994). The role of leukotrienes in inflammation. Annals of Internal
Proceedings of the National Academy of Sciences of the United States of America 90, Medicine 121, 684–697.
7656–7660. Heneka, M. T., Carson, M. J., El Khoury, J., Landreth, G. E., Brosseron, F., Feinstein, D. L., ...
Felder, C. C., Joyce, K. E., Briley, E. M., Mansouri, J., Mackie, K., Blond, O., ... Mitchell, R. L. Kummer, M. P. (2015). Neuroinflammation in Alzheimer’s disease. Lancet Neurology
(1995). Comparison of the pharmacology and signal transduction of the human can- 14, 388–405.
nabinoid CB1 and CB2 receptors. Molecular Pharmacology 48, 443–450. Heneka, M. T., Sastre, M., Dumitrescu-Ozimek, L., Hanke, A., Dewachter, I., Kuiperi, C., ...
Feliú, A., Bonilla Del Río, I., Carrillo-Salinas, F. J., Hernández-Torres, G., Mestre, L., Puente, Landreth, G. E. (2005). Acute treatment with the PPARgamma agonist pioglitazone
N., ... Guaza, C. (2017). 2-arachidonoylglycerol reduces proteoglycans and enhances and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I trans-
remyelination in a progressive model of demyelination. Journal of Neuroscience 37, genic mice. Brain 128, 1442–1453.
8385–8398. Herkenham, M., Lynn, A. B., Johnson, M. R., Melvin, L. S., de Costa, B. R., & Rice, K. C. (1991).
Fernández-Suárez, D., Celorrio, M., Riezu-Boj, J. I., Ugarte, A., Pacheco, R., González, H., ... Characterization and localization of cannabinoid receptors in rat brain: a quantitative
Aymerich, M. S. (2014). Monoacylglycerol lipase inhibitor JZL184 is neuroprotective in vitro autoradiographic study. Journal of Neuroscience 11, 563–583.

13
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Herkenham, M., Lynn, A. B., Little, M. D., Johnson, M. R., Melvin, L. S., de Costa, B. R., & Rice, brain barrier integrity, and neuroinflammation following mild traumatic brain injury.
K. C. (1990). Cannabinoid receptor localization in brain. Proceedings of the National Journal of Neurotrauma 32, 297–306.
Academy of Sciences of the United States of America 87, 1932–1936. Khan, M. A., Alam, Q., Haque, A., Ashafaq, M., Khan, M. J., Ashraf, G. M., & Ahmad, M.
Hermanson, D. J., Gamble-George, J. C., Marnett, L. J., & Patel, S. (2014). Substrate-selective (2019). Current Progress on peroxisome proliferator-activated receptor gamma ago-
COX-2 inhibition as a novel strategy for therapeutic endocannabinoid augmentation. nist as an emerging therapeutic approach for the treatment of Alzheimer’s disease:
Trends in Pharmacological Sciences 35, 358–367. an update. Current Neuropharmacology 17, 232–246.
Hermanson, D. J., Hartley, N. D., Gamble-George, J., Brown, N., Shonesy, B. C., Kingsley, P. J., King, A. R., Duranti, A., Tontini, A., Rivara, S., Rosengarth, A., Clapper, J. R., ... Piomelli, D.
... Patel, S. (2013). Substrate-selective COX-2 inhibition decreases anxiety via endo- (2007). URB602 inhibits monoacylglycerol lipase and selectively blocks 2-
cannabinoid activation. Nature Neuroscience 16, 1291–1298. arachidonoylglycerol degradation in intact brain slices. Chemistry & Biology 14,
Hernández-Torres, G., Cipriano, M., Hedén, E., Björklund, E., Canales, Á., Zian, D., ... López- 1357–1365.
Rodríguez, M. L. (2014). A reversible and selective inhibitor of monoacylglycerol li- Kingsley, P. J., Rouzer, C. A., Morgan, A. J., Patel, S., & Marnett, L. J. (2019). Aspects of pros-
pase ameliorates multiple sclerosis. Angewandte Chemie (International Ed. in English) taglandin glycerol ester biology. Advances in Experimental Medicine and Biology 1161,
53, 13765–13770. 77–88.
Hillard, C. J. (2000). Biochemistry and pharmacology of the endocannabinoids Koppel, J., Bradshaw, H., Goldberg, T. E., Khalili, H., Marambaud, P., Walker, M. J., ... Davies,
arachidonylethanolamide and 2-arachidonylglycerol. Prostaglandins & Other Lipid P. (2009). Endocannabinoids in Alzheimer’s disease and their impact on normative
Mediators 61, 3–18. cognitive performance: a case-control and cohort study. Lipids in Health and Disease
Hoffman, A. F., Oz, M., Yang, R., Lichtman, A. H., & Lupica, C. R. (2007). Opposing actions of 8, 2.
chronic Delta9-tetrahydrocannabinol and cannabinoid antagonists on hippocampal Kozak, K. R., Prusakiewicz, J. J., & Marnett, L. J. (2004). Oxidative metabolism of
long-term potentiation. Learning & Memory 14, 63–74. endocannabinoids by COX-2. Current Pharmaceutical Design 10, 659–667.
Hohmann, A. G., Suplita, R. L., Bolton, N. M., Neely, M. H., Fegley, D., Mangieri, R., ... Kozak, K. R., Prusakiewicz, J. J., Rowlinson, S. W., Schneider, C., & Marnett, L. J. (2001).
Piomelli, D. (2005). An endocannabinoid mechanism for stress-induced analgesia. Amino acid determinants in cyclooxygenase-2 oxygenation of the endocannabinoid
Nature 435, 1108–1112. 2-arachidonylglycerol. The Journal of Biological Chemistry 276, 30072–30077.
Hoozemans, J. J., & O’Banion, M. K. (2005). The role of COX-1 and COX-2 in Alzheimer’s Kozak, K. R., Rowlinson, S. W., & Marnett, L. J. (2000). Oxygenation of the endocannabi-
disease pathology and the therapeutic potentials of non-steroidal anti- noid, 2-arachidonylglycerol, to glyceryl prostaglandins by cyclooxygenase-2. The
inflammatory drugs. Current Drug Targets. CNS and Neurological Disorders 4, 307–315. Journal of Biological Chemistry 275, 33744–33749.
Howlett, A. C. (1987). Cannabinoid inhibition of adenylate cyclase: relative activity of con- Kreitzer, A. C., & Regehr, W. G. (2002). Retrograde signaling by endocannabinoids. Current
stituents and metabolites of marihuana. Neuropharmacology 26, 507–512. Opinion in Neurobiology 12, 324–330.
Howlett, A. C., & Abood, M. E. (2017). CB(1) and CB(2) receptor pharmacology. Advances Kwon, H. S., & Koh, S. H. (2020). Neuroinflammation in neurodegenerative disorders: the
in Pharmacology 80, 169–206. roles of microglia and astrocytes. Translational Neurodegeneration 9, 42.
Howlett, A. C., Barth, F., Bonner, T. I., Cabral, G., Casellas, P., Devane, W. A., ... Pertwee, R. G. Labar, G., Wouters, J., & Lambert, D. M. (2010). A review on the monoacylglycerol lipase:
(2002). International Union of pharmacology. XXVII. Classification of cannabinoid re- at the interface between fat and endocannabinoid signalling. Current Medicinal
ceptors. Pharmacological Reviews 54, 161–202. Chemistry 17, 2588–2607.
Howlett, A. C., Bidaut-Russell, M., Devane, W. A., Melvin, L. S., Johnson, M. R., & Lee, H. G., Wheeler, M. A., & Quintana, F. J. (2022). Function and therapeutic value of as-
Herkenham, M. (1990). The cannabinoid receptor: biochemical, anatomical and be- trocytes in neurological diseases. Nature Reviews. Drug Discovery 21, 339–358.
havioral characterization. Trends in Neurosciences 13, 420–423. Li, L., Liang, J., & Fu, H. (2021). An update on the association between traumatic brain in-
jury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction.
Howlett, A. C., Breivogel, C. S., Childers, S. R., Deadwyler, S. A., Hampson, R. E., & Porrino, L.
Neuroscience and Biobehavioral Reviews 120, 372–386.
J. (2004). Cannabinoid physiology and pharmacology: 30 years of progress.
Neuropharmacology 47(Suppl. 1), 345–358. Ligresti, A., De Petrocellis, L., & Di Marzo, V. (2016). From phytocannabinoids to cannabi-
noid receptors and endocannabinoids: pleiotropic physiological and pathological
Howlett, A. C., & Fleming, R. M. (1984). Cannabinoid inhibition of adenylate cyclase. Phar-
roles through complex pharmacology. Physiological Reviews 96, 1593–1659.
macology of the response in neuroblastoma cell membranes. Molecular Pharmacology
Lindgren, C. A., Newman, Z. L., Morford, J. J., Ryan, S. B., Battani, K. A., & Su, Z. (2013). Cy-
26, 532–538.
clooxygenase-2, prostaglandin E2 glycerol ester and nitric oxide are involved in
Howlett, A. C., Qualy, J. M., & Khachatrian, L. L. (1986). Involvement of Gi in the inhibition
muscarine-induced presynaptic enhancement at the vertebrate neuromuscular junc-
of adenylate cyclase by cannabimimetic drugs. Molecular Pharmacology 29, 307–313.
tion. The Journal of Physiology 591, 4749–4764.
Hu, M., Zhu, D., Zhang, J., Gao, F., Hashem, J., Kingsley, P., ... Chen, C. (2022). Enhancing
Liu, R., Wang, X., Curtiss, C., Landas, S., Rong, R., Sheikh, M. S., & Huang, Y. (2018). Mono-
endocannabinoid signalling in astrocytes promotes recovery from traumatic brain in-
glyceride lipase gene knockout in mice leads to increased incidence of lung adenocar-
jury. Brain 145, 179–193.
cinoma. Cell Death & Disease 9, 36.
Hu, S. S., Bradshaw, H. B., Chen, J. S., Tan, B., & Walker, J. M. (2008). Prostaglandin E2 glyc-
Liu, X., Chen, Y., Vickstrom, C. R., Li, Y., Viader, A., Cravatt, B. F., & Liu, Q. S. (2016). Coordi-
erol ester, an endogenous COX-2 metabolite of 2-arachidonoylglycerol, induces hy-
nated regulation of endocannabinoid-mediated retrograde synaptic suppression in
peralgesia and modulates NFkappaB activity. British Journal of Pharmacology 153,
the cerebellum by neuronal and astrocytic monoacylglycerol lipase. Scientific
1538–1549.
Reports 6, 35829.
Hurley, S. D., Olschowka, J. A., & O’Banion, M. K. (2002). Cyclooxygenase inhibition as a
Long, J. Z., & Cravatt, B. F. (2011). The metabolic serine hydrolases and their functions in
strategy to ameliorate brain injury. Journal of Neurotrauma 19, 1–15.
mammalian physiology and disease. Chemical Reviews 111, 6022–6063.
Imperatore, R., Morello, G., Luongo, L., Taschler, U., Romano, R., De Gregorio, D., Belardo, C., Long, J. Z., Li, W., Booker, L., Burston, J. J., Kinsey, S. G., Schlosburg, J. E., ... Cravatt, B. F.
Maione, S., Di Marzo, V., & Cristino, L. (2015). Genetic deletion of monoacylglycerol (2009). Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabi-
lipase leads to impaired cannabinoid receptor CB(1)R signaling and anxiety-like be- noid behavioral effects. Nature Chemical Biology 5, 37–44.
havior. Journal of Neurochemistry 135, 799–813.
Long, J. Z., Nomura, D. K., & Cravatt, B. F. (2009). Characterization of monoacylglycerol li-
Iversen, L. (2003). Cannabis and the brain. Brain 126, 1252–1270. pase inhibition reveals differences in central and peripheral endocannabinoid metab-
Jassam, Y. N., Izzy, S., Whalen, M., McGavern, D. B., & El Khoury, J. (2017). olism. Chemistry & Biology 16, 744–753.
Neuroimmunology of traumatic brain injury: time for a paradigm shift. Neuron 95, Lourbopoulos, A., Grigoriadis, N., Lagoudaki, R., Touloumi, O., Polyzoidou, E., Mavromatis,
1246–1265. I., Tascos, N., Breuer, A., Ovadia, H., Karussis, D., Shohami, E., Mechoulam, R., &
Jiang, M., & van der Stelt, M. (2018). Activity-based protein profiling delivers selective Simeonidou, C. (2011). Administration of 2-arachidonoylglycerol ameliorates both
drug candidate ABX-1431, a monoacylglycerol lipase inhibitor, to control lipid metab- acute and chronic experimental autoimmune encephalomyelitis. Brain Research
olism in neurological disorders. Journal of Medicinal Chemistry 61, 9059–9061. 1390, 126–141.
Johnson, V. E., Stewart, W., & Smith, D. H. (2010). Traumatic brain injury and amyloid- Lovinger, D. M. (2008). Presynaptic modulation by endocannabinoids. Handbook of
beta pathology: a link to Alzheimer’s disease? Nature Reviews. Neuroscience 11, Experimental Pharmacology, 435–477.
361–370. Lu, H. C., & Mackie, K. (2021). Review of the Endocannabinoid System. Biological
Kaczocha, M., Glaser, S. T., & Deutsch, D. G. (2009). Identification of intracellular carriers Psychiatry: Cognitive Neuroscience and Neuroimaging 6, 607–615.
for the endocannabinoid anandamide. Proceedings of the National Academy of Lu, Y., Peng, F., Dong, M., & Yang, H. (2014). Endocannabinoid 2-arachidonylglycerol pro-
Sciences of the United States of America 106, 6375–6380. tects primary cultured neurons against LPS-induced impairments in rat caudate nu-
Kaczocha, M., & Haj-Dahmane, S. (2022). Mechanisms of endocannabinoid transport in cleus. Journal of Molecular Neuroscience 54, 49–58.
the brain. British Journal of Pharmacology 179, 4300–4310. Ludányi, A., Hu, S. S., Yamazaki, M., Tanimura, A., Piomelli, D., Watanabe, M., ... Katona, I.
Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M., & Watanabe, M. (2009). (2011). Complementary synaptic distribution of enzymes responsible for synthesis
Endocannabinoid-mediated control of synaptic transmission. Physiological Reviews and inactivation of the endocannabinoid 2-arachidonoylglycerol in the human hippo-
89, 309–380. campus. Neuroscience 174, 50–63.
Karlsson, M., Contreras, J. A., Hellman, U., Tornqvist, H., & Holm, C. (1997). cDNA cloning, Lysenko, L. V., Kim, J., Henry, C., Tyrtyshnaia, A., Kohnz, R. A., Madamba, F., ...
tissue distribution, and identification of the catalytic triad of monoglyceride lipase. Kleschevnikov, A. M. (2014). Monoacylglycerol lipase inhibitor JZL184 improves be-
Evolutionary relationship to esterases, lysophospholipases, and haloperoxidases. havior and neural properties in Ts65Dn mice, a model of down syndrome. PLoS One
The Journal of Biological Chemistry 272, 27218–27223. 9, Article e114521.
Karlsson, M., Reue, K., Xia, Y. R., Lusis, A. J., Langin, D., Tornqvist, H., & Holm, C. (2001). Mackie, K. (2005). Distribution of cannabinoid receptors in the central and peripheral
Exon-intron organization and chromosomal localization of the mouse monoglyceride nervous system. Handbook of Experimental Pharmacology, 299–325.
lipase gene. Gene 272, 11–18. Mackie, K. (2008). Cannabinoid receptors: where they are and what they do. Journal of
Katona, I., & Freund, T. F. (2012). Multiple functions of endocannabinoid signaling in the Neuroendocrinology 20(Suppl. 1), 10–14.
brain. Annual Review of Neuroscience 35, 529–558. Marino, S., Carrasco, G., Li, B., Shah, K. M., Lath, D. L., Sophocleous, A., ... Idris, A. I. (2020).
Katz, P. S., Sulzer, J. K., Impastato, R. A., Teng, S. X., Rogers, E. K., & Molina, P. E. (2015). JZL184, a monoacylglycerol lipase inhibitor, induces bone loss in a multiple myeloma
Endocannabinoid degradation inhibition improves neurobehavioral function, blood- model of immunocompetent mice. Calcified Tissue International 107, 72–85.

14
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Marino, S., de Ridder, D., Bishop, R. T., Renema, N., Ponzetti, M., Sophocleous, A., ... Idris, A. Oakley, H., Cole, S. L., Logan, S., Maus, E., Shao, P., Craft, J., ... Vassar, R. (2006).
I. (2019). Paradoxical effects of JZL184, an inhibitor of monoacylglycerol lipase, on Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in trans-
bone remodelling in healthy and cancer-bearing mice. EBioMedicine 44, 452–466. genic mice with five familial Alzheimer’s disease mutations: potential factors in am-
Maroof, N., Ravipati, S., Pardon, M. C., Barrett, D. A., & Kendall, D. A. (2014). Reductions in yloid plaque formation. Journal of Neuroscience 26, 10129–10140.
endocannabinoid levels and enhanced coupling of cannabinoid receptors in the stri- Ogasawara, D., Deng, H., Viader, A., Baggelaar, M. P., Breman, A., den Dulk, H., ... van der
atum are accompanied by cognitive impairments in the AbetaPPswe/PS1DeltaE9 Stelt, M. (2016). Rapid and profound rewiring of brain lipid signaling networks by
mouse model of Alzheimer’s disease. Journal of Alzheimers Disease 42, 227–245. acute diacylglycerol lipase inhibition. Proceedings of the National Academy of
Marrs, W. R., Blankman, J. L., Horne, E. A., Thomazeau, A., Lin, Y. H., Coy, J., ... Stella, N. Sciences of the United States of America 113, 26–33.
(2010). The serine hydrolase ABHD6 controls the accumulation and efficacy of 2- Ohno-Shosaku, T., & Kano, M. (2014). Endocannabinoid-mediated retrograde modulation
AG at cannabinoid receptors. Nature Neuroscience 13, 951–957. of synaptic transmission. Current Opinion in Neurobiology 29, 1–8.
Martinez-Torres, S., Cutando, L., Pastor, A., Kato, A., Sakimura, K., de la Torre, R., Valjent, E., O’Sullivan, S. E. (2007). Cannabinoids go nuclear: evidence for activation of peroxisome
Maldonado, R., Kano, M., & Ozaita, A. (2019). Monoacylglycerol lipase blockade im- proliferator-activated receptors. British Journal of Pharmacology 152, 576–582.
pairs fine motor coordination and triggers cerebellar neuroinflammation through O’Sullivan, S. E. (2016). An update on PPAR activation by cannabinoids. British Journal of
cyclooxygenase-2. Brain, Behavior, and Immunity 81, 399–409. Pharmacology 173, 1899–1910.
Matsuda, L. A., Lolait, S. J., Brownstein, M. J., Young, A. C., & Bonner, T. I. (1990). Structure Pan, B., Wang, W., Zhong, P., Blankman, J. L., Cravatt, B. F., & Liu, Q. S. (2011). Alterations of
of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346, endocannabinoid signaling, synaptic plasticity, learning, and memory in
561–564. monoacylglycerol lipase knock-out mice. Journal of Neuroscience 31, 13420–13430.
Mayeux, J., Katz, P., Edwards, S., Middleton, J. W., & Molina, P. E. (2017). Inhibition of Panikashvili, D., Mechoulam, R., Beni, S. M., Alexandrovich, A., & Shohami, E. (2005). CB1
endocannabinoid degradation improves outcomes from mild traumatic brain injury: cannabinoid receptors are involved in neuroprotection via NF-kappa B inhibition.
a mechanistic role for synaptic hyperexcitability. Journal of Neurotrauma 34, 436–443. Journal of Cerebral Blood Flow and Metabolism 25, 477–484.
McGeer, P. L., Rogers, J., & McGeer, E. G. (2016). Inflammation, antiinflammatory agents, Panikashvili, D., Shein, N. A., Mechoulam, R., Trembovler, V., Kohen, R., Alexandrovich, A.,
and Alzheimer’s disease: the last 22 years. Journal of Alzheimers Disease 54, 853–857. & Shohami, E. (2006). The endocannabinoid 2-AG protects the blood-brain barrier
McKee, A. C., Stern, R. A., Nowinski, C. J., Stein, T. D., Alvarez, V. E., Daneshvar, D. H., ... after closed head injury and inhibits mRNA expression of proinflammatory cytokines.
Cantu, R. C. (2013). The spectrum of disease in chronic traumatic encephalopathy. Neurobiology of Disease 22, 257–264.
Brain 136, 43–64. Panikashvili, D., Simeonidou, C., Ben-Shabat, S., Hanus, L., Breuer, A., Mechoulam, R., &
Mechoulam, R. (1970). Marihuana chemistry. Science 168, 1159–1166. Shohami, E. (2001). An endogenous cannabinoid (2-AG) is neuroprotective after
Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N. E., Schatz, A. R., ... brain injury. Nature 413, 527–531.
Compton, D. R., et al. (1995). Identification of an endogenous 2-monoglyceride, pres- Pasquarelli, N., Engelskirchen, M., Hanselmann, J., Endres, S., Porazik, C., Bayer, H., Buck, E.,
ent in canine gut, that binds to cannabinoid receptors. Biochemical Pharmacology 50, Karsak, M., Weydt, P., Ferger, B., & Witting, A. (2017). Evaluation of monoacylglycerol
83–90. lipase as a therapeutic target in a transgenic mouse model of ALS. Neuropharmacology
Mechoulam, R., Braun, P., & Gaoni, Y. (1967). A stereospecific synthesis of (-)-delta 1- and 124, 157–169.
(-)-delta 1(6)-tetrahydrocannabinols. Journal of the American Chemical Society 89, Pasquarelli, N., Porazik, C., Bayer, H., Buck, E., Schildknecht, S., Weydt, P., Witting, A., &
4552–4554. Ferger, B. (2017). Contrasting effects of selective MAGL and FAAH inhibition on dopa-
Mechoulam, R., & Gaoni, Y. (1967). The absolute configuration of delta-1-tetrahydrocan- mine depletion and GDNF expression in a chronic MPTP mouse model of Parkinson's
nabinol, the major active constituent of hashish. Tetrahedron Letters 12, 1109–1111. disease. Neurochemistry International 110, 14–24.
Mechoulam, R., Hanuš, L. O., Pertwee, R., & Howlett, A. C. (2014). Early phytocannabinoid Pertwee, R. G. (2006). Cannabinoid pharmacology: the first 66 years. British Journal of
chemistry to endocannabinoids and beyond. Nature Reviews. Neuroscience 15, Pharmacology 147(Suppl. 1), S163–S171.
757–764. Pertwee, R. G. (2015). Endocannabinoids and Their Pharmacological Actions. Handbook of
Mechoulam, R., & Parker, L. A. (2013). The endocannabinoid system and the brain. Annual Experimental Pharmacology 231, 1–37.
Review of Psychology 64, 21–47. Pihlaja, R., Takkinen, J., Eskola, O., Vasara, J., Lopez-Picon, F. R., Haaparanta-Solin, M., &
Mechoulam, R., & Shvo, Y. (1963). Hashish. I. The structure of cannabidiol. Tetrahedron 19, Rinne, J. O. (2015). Monoacylglycerol lipase inhibitor JZL184 reduces neuroinflamma-
2073–2078. tory response in APdE9 mice and in adult mouse glial cells. Journal of
Mechoulam, R., Spatz, M., & Shohami, E. (2002). Endocannabinoids and neuroprotection. Neuroinflammation 12, 81.
Science's STKE 2002, re5. Piomelli, D. (2003). The molecular logic of endocannabinoid signalling. Nature Reviews.
Messinis, L., Kyprianidou, A., Malefaki, S., & Papathanasopoulos, P. (2006). Neuropsycho- Neuroscience 4, 873–884.
logical deficits in long-term frequent cannabis users. Neurology 66, 737–739. Piomelli, D., & Mabou Tagne, A. (2022). Endocannabinoid-Based Therapies. Annual Review
Minghetti, L. (2004). Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain of Pharmacology and Toxicology 62, 483–507.
diseases. Journal of Neuropathology and Experimental Neurology 63, 901–910. Piro, J. R., Benjamin, D. I., Duerr, J. M., Pi, Y., Gonzales, C., Wood, K. M., ... Samad, T. A.
Morgan, A. J., Kingsley, P. J., Mitchener, M. M., Altemus, M., Patrick, T. A., Gaulden, A. D., ... (2012). A dysregulated endocannabinoid-eicosanoid network supports pathogenesis
Patel, S. (2018). Detection of cyclooxygenase-2-derived oxygenation products of the in a mouse model of Alzheimer’s disease. Cell Reports 1, 617–623.
endogenous cannabinoid 2-arachidonoylglycerol in mouse brain. ACS Chemical Piro, J. R., Suidan, G. L., Quan, J., Pi, Y., O’Neill, S. M., Ilardi, M., ... Samad, T. A. (2018). Inhi-
Neuroscience 9, 1552–1559. bition of 2-AG hydrolysis differentially regulates blood brain barrier permeability
Mortimer, J. A., French, L. R., Hutton, J. T., & Schuman, L. M. (1985). Head injury as a risk after injury. Journal of Neuroinflammation 15, 142.
factor for Alzheimer’s disease. Neurology 35, 264–267. Pitler, T. A., & Alger, B. E. (1994). Depolarization-induced suppression of GABAergic inhi-
Mounsey, R. B., Mustafa, S., Robinson, L., Ross, R. A., Riedel, G., Pertwee, R. G., & Teismann, bition in rat hippocampal pyramidal cells: G protein involvement in a presynaptic
P. (2015). Increasing levels of the endocannabinoid 2-AG is neuroprotective in the 1- mechanism. Neuron 13, 1447–1455.
methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Piyanova, A., Lomazzo, E., Bindila, L., Lerner, R., Albayram, O., Ruhl, T., Lutz, B., Zimmer, A.,
Experimental Neurology 273, 36–44. & Bilkei-Gorzo, A. (2015). Age-related changes in the endocannabinoid system in the
Muccioli, G. G., Xu, C., Odah, E., Cudaback, E., Cisneros, J. A., Lambert, D. M., ... Stella, N. mouse hippocampus. Mechanisms of Ageing and Development 150, 55–64.
(2007). Identification of a novel endocannabinoid-hydrolyzing enzyme expressed Pope, H. G., Jr., & Yurgelun-Todd, D. (1996). The residual cognitive effects of heavy mari-
by microglial cells. Journal of Neuroscience 27, 2883–2889. juana use in college students. JAMA 275, 521–527.
Mulder, J., Zilberter, M., Pasquare, S. J., Alpar, A., Schulte, G., Ferreira, S. G., ... Harkany, T. Puighermanal, E., Marsicano, G., Busquets-Garcia, A., Lutz, B., Maldonado, R., & Ozaita, A.
(2011). Molecular reorganization of endocannabinoid signalling in Alzheimer’s dis- (2009). Cannabinoid modulation of hippocampal long-term memory is mediated
ease. Brain 134, 1041–1060. by mTOR signaling. Nature Neuroscience 12, 1152–1158.
Muller, C., Morales, P., & Reggio, P. H. (2018). Cannabinoid ligands targeting TRP channels. Punt, J. M., van der Vliet, D., & van der Stelt, M. (2022). Chemical Probes to Control and
Frontiers in Molecular Neuroscience 11, 487. Visualize Lipid Metabolism in the Brain. Accounts of Chemical Research 55, 3205–3217.
Müller-Vahl, K. R., Fremer, C., Beals, C., Ivkovic, J., Loft, H., & Schindler, C. (2021). Ransohoff, R. M. (2016). How neuroinflammation contributes to neurodegeneration.
Monoacylglycerol lipase inhibition in tourette syndrome: a 12-week, randomized, Science 353, 777–783.
controlled study. Movement Disorders 36, 2413–2418. Ratano, P., Petrella, C., Forti, F., Passeri, P. P., Morena, M., Palmery, M., ... Campolongo, P.
Mulvihill, M. M., & Nomura, D. K. (2013). Therapeutic potential of monoacylglycerol lipase (2018). Pharmacological inhibition of 2-arachidonoilglycerol hydrolysis enhances
inhibitors. Life Sciences 92, 492–497. memory consolidation in rats through CB2 receptor activation and mTOR signaling
Munro, S., Thomas, K. L., & Abu-Shaar, M. (1993). Molecular characterization of a periph- modulation. Neuropharmacology 138, 210–218.
eral receptor for cannabinoids. Nature 365, 61–65. Ren, S. Y., Wang, Z. Z., Zhang, Y., & Chen, N. H. (2020). Potential application of endocanna-
Murataeva, N., Straiker, A., & Mackie, K. (2014). Parsing the players: 2- binoid system agents in neuropsychiatric and neurodegenerative diseases-focusing
arachidonoylglycerol synthesis and degradation in the CNS. British Journal of on FAAH/MAGL inhibitors. Acta Pharmacologica Sinica 41, 1263–1271.
Pharmacology 171, 1379–1391. Ricciotti, E., & FitzGerald, G. A. (2011). Prostaglandins and inflammation. Arteriosclerosis,
Naidoo, V., Karanian, D. A., Vadivel, S. K., Locklear, J. R., Wood, J. T., Nasr, M., ... Bahr, B. A. Thrombosis, and Vascular Biology 31, 986–1000.
(2012). Equipotent inhibition of fatty acid amide hydrolase and monoacylglycerol li- Rockwell, C. E., Snider, N. T., Thompson, J. T., Vanden Heuvel, J. P., & Kaminski, N. E. (2006).
pase - dual targets of the endocannabinoid system to protect against seizure pathol- Interleukin-2 suppression by 2-arachidonyl glycerol is mediated through peroxisome
ogy. Neurotherapeutics 9, 801–813. proliferator-activated receptor gamma independently of cannabinoid receptors 1 and
Navia-Paldanius, D., Savinainen, J. R., & Laitinen, J. T. (2012). Biochemical and pharmaco- 2. Molecular Pharmacology 70, 101–111.
logical characterization of human α/β-hydrolase domain containing 6 (ABHD6) and Rouzer, C. A., & Marnett, L. J. (2008). Non-redundant functions of cyclooxygenases: oxy-
12 (ABHD12). Journal of Lipid Research 53, 2413–2424. genation of endocannabinoids. The Journal of Biological Chemistry 283, 8065–8069.
Nomura, D. K., Morrison, B. E., Blankman, J. L., Long, J. Z., Kinsey, S. G., Marcondes, M. C., ... von Ruden, E. L., Bogdanovic, R. M., Wotjak, C. T., & Potschka, H. (2015). Inhibition of
Cravatt, B. F. (2011). Endocannabinoid hydrolysis generates brain prostaglandins that monoacylglycerol lipase mediates a cannabinoid 1-receptor dependent delay of kin-
promote neuroinflammation. Science 334, 809–813. dling progression in mice. Neurobiology of Disease 77, 238–245.

15
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

Saario, S. M., Salo, O. M., Nevalainen, T., Poso, A., Laitinen, J. T., Järvinen, T., & Niemi, R. Szafran, B. N., Borazjani, A., Scheaffer, H. L., Crow, J. A., McBride, A. M., Adekanye, O., ...
(2005). Characterization of the sulfhydryl-sensitive site in the enzyme responsible Ross, M. K. (2022). Carboxylesterase 1d inactivation augments lung inflammation
for hydrolysis of 2-arachidonoyl-glycerol in rat cerebellar membranes. Chemistry & in mice. ACS Pharmacology & Translational Science 5, 919–931.
Biology 12, 649–656. Szkudlarek, H. J., Desai, S. J., Renard, J., Pereira, B., Norris, C., Jobson, C. E. L., ... Laviolette, S.
Salmon, J. A., & Higgs, G. A. (1987). Prostaglandins and leukotrienes as inflammatory me- R. (2019). Δ-9-Tetrahydrocannabinol and Cannabidiol produce dissociable effects on
diators. British Medical Bulletin 43, 285–296. prefrontal cortical executive function and regulation of affective behaviors.
Sang, N., & Chen, C. (2006). Lipid signaling and synaptic plasticity. Neuroscientist 12, Neuropsychopharmacology 44, 817–825.
425–434. Taffe, M. A. (2012). Delta(9)Tetrahydrocannabinol impairs visuo-spatial associative learn-
Sang, N., Zhang, J., & Chen, C. (2006). PGE2 glycerol ester, a COX-2 oxidative metabolite of ing and spatial working memory in rhesus macaques. Journal of Psychopharmacology
2-arachidonoyl glycerol, modulates inhibitory synaptic transmission in mouse hippo- 26, 1299–1306.
campal neurons. The Journal of Physiology 572, 735–745. Tanimura, A., Yamazaki, M., Hashimotodani, Y., Uchigashima, M., Kawata, S., Abe, M., Kita,
Sang, N., Zhang, J., & Chen, C. (2007). COX-2 oxidative metabolite of endocannabinoid 2- Y., Hashimoto, K., Shimizu, T., Watanabe, M., Sakimura, K., & Kano, M. (2010). The
AG enhances excitatory glutamatergic synaptic transmission and induces neurotoxic- endocannabinoid 2-arachidonoylglycerol produced by diacylglycerol lipase alpha
ity. Journal of Neurochemistry 102, 1966–1977. mediates retrograde suppression of synaptic transmission. Neuron 65, 320–327.
Santello, M., Toni, N., & Volterra, A. (2019). Astrocyte function from information process- Tansey, M. G., Wallings, R. L., Houser, M. C., Herrick, M. K., Keating, C. E., & Joers, V. (2022).
ing to cognition and cognitive impairment. Nature Neuroscience 22, 154–166. Inflammation and immune dysfunction in Parkinson disease. Nature Reviews.
Savinainen, J. R., Saario, S. M., & Laitinen, J. T. (2012). The serine hydrolases MAGL, ABHD6 Immunology 22, 657–673.
and ABHD12 as guardians of 2-arachidonoylglycerol signalling through cannabinoid Teismann, P., Vila, M., Choi, D. K., Tieu, K., Wu, D. C., Jackson-Lewis, V., & Przedborski, S.
receptors. Acta Physiologica (Oxford, England) 204, 267–276. (2003). COX-2 and neurodegeneration in Parkinson’s disease. Annals of the New
Scalvini, L., Piomelli, D., & Mor, M. (2016). Monoglyceride lipase: Structure and inhibitors. York Academy of Sciences 991, 272–277.
Chemistry and Physics of Lipids 197, 13–24. Terrone, G., Pauletti, A., Salamone, A., Rizzi, M., Villa, B. R., Porcu, L., ... Vezzani, A. (2018).
Schlosburg, J. E., Blankman, J. L., Long, J. Z., Nomura, D. K., Pan, B., Kinsey, S. G., ... Cravatt, B. Inhibition of monoacylglycerol lipase terminates diazepam-resistant status epilepti-
F. (2010). Chronic monoacylglycerol lipase blockade causes functional antagonism of cus in mice and its effects are potentiated by a ketogenic diet. Epilepsia 59, 79–91.
the endocannabinoid system. Nature Neuroscience 13, 1113–1119. Tornqvist, H., & Belfrage, P. (1976). Purification and some properties of a
Schlosburg, J. E., Kinsey, S. G., Ignatowska-Jankowska, B., Ramesh, D., Abdullah, R. A., Tao, monoacylglycerol-hydrolyzing enzyme of rat adipose tissue. The Journal of Biological
Q., ... Lichtman, A. H. (2014). Prolonged monoacylglycerol lipase blockade causes Chemistry 251, 813–819.
equivalent cannabinoid receptor type 1 receptor-mediated adaptations in fatty acid Turini, M. E., & DuBois, R. N. (2002). Cyclooxygenase-2: a therapeutic target. Annual
amide hydrolase wild-type and knockout mice. Journal of Pharmacology and Review of Medicine 53, 35–57.
Experimental Therapeutics 350, 196–204. Turner, R. C., Lucke-Wold, B. P., Robson, M. J., Lee, J. M., & Bailes, J. E. (2016). Alzheimer’s
Schloss, M. J., Horckmans, M., Guillamat-Prats, R., Hering, D., Lauer, E., Lenglet, S., ... disease and chronic traumatic encephalopathy: Distinct but possibly overlapping dis-
Steffens, S. (2019). 2-Arachidonoylglycerol mobilizes myeloid cells and worsens ease entities. Brain Injury 30, 1279–1292.
heart function after acute myocardial infarction. Cardiovascular Research 115, Van Sickle, M. D., Duncan, M., Kingsley, P. J., Mouihate, A., Urbani, P., Mackie, K., ... Sharkey,
602–613. K. A. (2005). Identification and functional characterization of brainstem cannabinoid
Selvaraj, P., Tanaka, M., Wen, J., & Zhang, Y. (2021). The novel monoacylglycerol lipase in- CB2 receptors. Science 310, 329–332.
hibitor MJN110 suppresses neuroinflammation, normalizes synaptic composition Vane, J. R., Bakhle, Y. S., & Botting, R. M. (1998). Cyclooxygenases 1 and 2. Annual Review of
and improves behavioral performance in the repetitive traumatic brain injury Pharmacology and Toxicology 38, 97–120.
mouse model. Cells 10. Vázquez, C., Tolón, R. M., Grande, M. T., Caraza, M., Moreno, M., Koester, E. C., ... Romero, J.
Shin, M., Snyder, H. W., Donvito, G., Schurman, L. D., Fox, T. E., Lichtman, A. H., ... Hsu, K. L. (2015). Endocannabinoid regulation of amyloid-induced neuroinflammation.
(2018). Liposomal delivery of diacylglycerol lipase-beta inhibitors to macrophages Neurobiology of Aging 36, 3008–3019.
dramatically enhances selectivity and efficacy in vivo. Molecular Pharmaceutics 15, Viader, A., Blankman, J. L., Zhong, P., Liu, X., Schlosburg, J. E., Joslyn, C. M., ... Cravatt, B. F.
721–728. (2015). Metabolic interplay between astrocytes and neurons regulates endocannabi-
Shohami, E., Cohen-Yeshurun, A., Magid, L., Algali, M., & Mechoulam, R. (2011). noid action. Cell Reports 12, 798–808.
Endocannabinoids and traumatic brain injury. British Journal of Pharmacology 163, Viader, A., Ogasawara, D., Joslyn, C. M., Sanchez-Alavez, M., Mori, S., Nguyen, W., ... Cravatt,
1402–1410. B. F. (2016). A chemical proteomic atlas of brain serine hydrolases identifies cell type-
Shonesy, B. C., Bluett, R. J., Ramikie, T. S., Báldi, R., Hermanson, D. J., Kingsley, P. J., ... Patel, specific pathways regulating neuroinflammation. Elife 5, Article e12345.
S. (2014). Genetic disruption of 2-arachidonoylglycerol synthesis reveals a key role Vickstrom, C. R., Liu, X., Liu, S., Hu, M. M., Mu, L., Hu, Y., ... Liu, Q. S. (2021). Role of endo-
for endocannabinoid signaling in anxiety modulation. Cell Reports 9, 1644–1653. cannabinoid signaling in a septohabenular pathway in the regulation of anxiety- and
Silveira, K. M., Wegener, G., & Joca, S. R. L. (2021). Targeting 2-arachidonoylglycerol sig- depressive-like behavior. Molecular Psychiatry 26, 3178–3191.
nalling in the neurobiology and treatment of depression. Basic & Clinical Vigh, J., & von Gersdorff, H. (2007). Endocannabinoids mediate synaptic plasticity at
Pharmacology & Toxicology 129, 3–14. mixed synapses. Neuron 56, 945–946.
Simon, D. W., McGeachy, M. J., Bayır, H., Clark, R. S., Loane, D. J., & Kochanek, P. M. (2017). Villapol, S. (2018). Roles of peroxisome proliferator-activated receptor gamma on brain
The far-reaching scope of neuroinflammation after traumatic brain injury. Nature and peripheral inflammation. Cellular and Molecular Neurobiology 38, 121–132.
Reviews. Neurology 13, 171–191. Volkow, N. D., Baler, R. D., Compton, W. M., & Weiss, S. R. (2014). Adverse health effects of
Solowij, N., Stephens, R. S., Roffman, R. A., Babor, T., Kadden, R., Miller, M., ... Marijuana marijuana use. The New England Journal of Medicine 370, 2219–2227.
Treatment Project Research, G (2002). Cognitive functioning of long-term heavy can- Walton, C., King, R., Rechtman, L., Kaye, W., Leray, E., Marrie, R. A., ... Baneke, P. (2020).
nabis users seeking treatment. JAMA 287, 1123–1131. Rising prevalence of multiple sclerosis worldwide: Insights from the Atlas of MS.
Song, Y., Hu, M., Zhang, J., Teng, Z. Q., & Chen, C. (2019). A novel mechanism of synaptic third editionMultiple Sclerosis 26, 1816–1821.
and cognitive impairments mediated via microRNA-30b in Alzheimer’s disease. Wenzel, T. J., & Klegeris, A. (2018). Novel multi-target directed ligand-based strategies for
EBioMedicine 39, 409–421. reducing neuroinflammation in Alzheimer’s disease. Life Sciences 207, 314–322.
Song, Y., Zhang, J., & Chen, C. (2015). Fine-tuning of synaptic upscaling at excitatory syn- Wilson, R. I., Kunos, G., & Nicoll, R. A. (2001). Presynaptic specificity of endocannabinoid
apses by endocannabinoid signaling is mediated via the CB1 receptor. Scientific signaling in the hippocampus. Neuron 31, 453–462.
Reports 5, 16257. Wilson, R. I., & Nicoll, R. A. (2001). Endogenous cannabinoids mediate retrograde signal-
Starowicz, K., Nigam, S., & Di Marzo, V. (2007). Biochemistry and pharmacology of ling at hippocampal synapses. Nature 410, 588–592.
endovanilloids. Pharmacology & Therapeutics 114, 13–33. Wilson, R. I., & Nicoll, R. A. (2002). Endocannabinoid signaling in the brain. Science 296,
Stella, N. (2023). THC and CBD: Similarities and differences between siblings. Neuron 111, 678–682.
302–327. Woodcock, T., & Morganti-Kossmann, M. C. (2013). The role of markers of inflammation
Stella, N., Schweitzer, P., & Piomelli, D. (1997). A second endogenous cannabinoid that in traumatic brain injury. Frontiers in Neurology 4, 18.
modulates long-term potentiation. Nature 388, 773–778. Wu, Y., Wu, H., Zeng, J., Pluimer, B., Dong, S., Xie, X., ... Zhao, Z. (2021). Mild traumatic
van der Stelt, M., Mazzola, C., Esposito, G., Matias, I., Petrosino, S., De Filippis, D., Micale, V., brain injury induces microvascular injury and accelerates Alzheimer-like pathogene-
Steardo, L., Drago, F., Iuvone, T., & Di Marzo, V. (2006). Endocannabinoids and beta- sis in mice. Acta Neuropathologica Communications 9, 74.
amyloid-induced neurotoxicity in vivo: effect of pharmacological elevation of endo- Xie, S., Borazjani, A., Hatfield, M. J., Edwards, C. C., Potter, P. M., & Ross, M. K. (2010). Inac-
cannabinoid levels. Cellular and Molecular Life Sciences 63, 1410–1424. tivation of lipid glyceryl ester metabolism in human THP1 monocytes/macrophages
Sugiura, T., Kishimoto, S., Oka, S., & Gokoh, M. (2006). Biochemistry, pharmacology and by activated organophosphorus insecticides: role of carboxylesterases 1 and 2.
physiology of 2-arachidonoylglycerol, an endogenous cannabinoid receptor ligand. Chemical Research in Toxicology 23, 1890–1904.
Progress in Lipid Research 45, 405–446. Xu, J. Y., & Chen, C. (2015). Endocannabinoids in Synaptic Plasticity and Neuroprotection.
Sugiura, T., Kondo, S., Sukagawa, A., Nakane, S., Shinoda, A., Itoh, K., Yamashita, A., & Neuroscientist 21, 152–168.
Waku, K. (1995). 2-Arachidonoylglycerol: a possible endogenous cannabinoid recep- Xue, F., Xue, S. S., Liu, L., Sang, H. F., Ma, Q. R., Tan, Q. R., ... Peng, Z. W. (2019). Early inter-
tor ligand in brain. Biochemical and Biophysical Research Communications 215, 89–97. vention with electroacupuncture prevents PTSD-like behaviors in rats through en-
Syal, C., Kosaraju, J., Hamilton, L., Aumont, A., Chu, A., Sarma, S. N., ... Wang, J. (2020). Dys- hancing hippocampal endocannabinoid signaling. Progress in Neuro-
regulated expression of monoacylglycerol lipase is a marker for anti-diabetic drug Psychopharmacology & Biological Psychiatry 93, 171–181.
metformin-targeted therapy to correct impaired neurogenesis and spatial memory Yang, H., & Chen, C. (2008). Cyclooxygenase-2 in synaptic signaling. Current
in Alzheimer’s disease. Theranostics 10, 6337–6360. Pharmaceutical Design 14, 1443–1451.
Szafran, B., Borazjani, A., Lee, J. H., Ross, M. K., & Kaplan, B. L. (2015). Lipopolysaccharide Yao, C., & Narumiya, S. (2019). Prostaglandin-cytokine crosstalk in chronic inflammation.
suppresses carboxylesterase 2g activity and 2-arachidonoylglycerol hydrolysis: A British Journal of Pharmacology 176, 337–354.
possible mechanism to regulate inflammation. Prostaglandins & Other Lipid Yoshida, T., Fukaya, M., Uchigashima, M., Miura, E., Kamiya, H., Kano, M., & Watanabe, M.
Mediators 121, 199–206. (2006). Localization of diacylglycerol lipase-alpha around postsynaptic spine

16
C. Chen Pharmacology & Therapeutics 244 (2023) 108394

suggests close proximity between production site of an endocannabinoid, 2- Zhang, J., Teng, Z., Song, Y., Hu, M., & Chen, C. (2015). Inhibition of monoacylglycerol li-
arachidonoyl-glycerol, and presynaptic cannabinoid CB1 receptor. Journal of pase prevents chronic traumatic encephalopathy-like neuropathology in a mouse
Neuroscience 26, 4740–4751. model of repetitive mild closed head injury. Journal of Cerebral Blood Flow and
Yoshiyama, Y., Higuchi, M., Zhang, B., Huang, S. M., Iwata, N., Saido, T. C., ... Lee, V. M. Metabolism 35, 443–453.
(2007). Synapse loss and microglial activation precede tangles in a P301S tauopathy Zhang, Z., Wang, W., Zhong, P., Liu, S. J., Long, J. Z., Zhao, L., ... Liu, Q. S. (2015). Blockade of
mouse model. Neuron 53, 337–351. 2-arachidonoylglycerol hydrolysis produces antidepressant-like effects and enhances
Zanfirescu, A., Ungurianu, A., Mihai, D. P., Radulescu, D., & Nitulescu, G. M. (2021). adult hippocampal neurogenesis and synaptic plasticity. Hippocampus 25, 16–26.
Targeting monoacylglycerol lipase in pursuit of therapies for neurological and neuro- Zhong, P., Wang, W., Pan, B., Liu, X., Zhang, Z., Long, J. Z., ... Liu, Q. S. (2014).
degenerative diseases. Molecules 26. Monoacylglycerol lipase inhibition blocks chronic stress-induced depressive-like be-
Zechner, R., Kienesberger, P. C., Haemmerle, G., Zimmermann, R., & Lass, A. (2009). Adi- haviors via activation of mTOR signaling. Neuropsychopharmacology 39, 1763–1776.
pose triglyceride lipase and the lipolytic catabolism of cellular fat stores. Journal of Zhu, D., Gao, F., & Chen, C. (2021). Endocannabinoid metabolism and traumatic brain in-
Lipid Research 50, 3–21. jury. Cells 10.
Zhu, D., Zhang, J., Gao, F., Hu, M., Hashem, J., & Chen, C. (2022). Augmentation of 2-
Zhang, J., & Chen, C. (2008). Endocannabinoid 2-arachidonoylglycerol protects neurons by
arachidonoylglycerol signaling in astrocytes maintains synaptic functionality by reg-
limiting COX-2 elevation. The Journal of Biological Chemistry 283, 22601–22611.
ulation of miRNA-30b. Experimental Neurology 361, Article 114292.
Zhang, J., & Chen, C. (2018). Alleviation of neuropathology by inhibition of Zhu, D., Zhang, J., Gao, F., Hu, M., Hashem, J., & Chen, C. (2023). Augmentation of 2-
monoacylglycerol lipase in APP transgenic mice lacking CB2 receptors. Molecular arachidonoylglycerol signaling in astrocytes maintains synaptic functionality by reg-
Neurobiology 55, 4802–4810. ulation of miRNA-30b. Experimental Neurology 361, Article 114292.
Zhang, J., Hu, M., Teng, Z., Tang, Y. P., & Chen, C. (2014). Synaptic and cognitive improve- Zhu, D., Zhang, J., Hashem, J., Gao, F., & Chen, C. (2023). Inhibition of 2-
ments by inhibition of 2-AG metabolism are through upregulation of microRNA-188- arachidonoylglycerol degradation enhances glial immunity by single-cell transcripto-
3p in a mouse model of Alzheimer’s disease. Journal of Neuroscience 34, 14919–14933. mic analysis. Journal of Neuroinflammation 20, 17.

17

You might also like