Skip to main content

Main menu

  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE

User menu

  • Log out
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Neuroscience
  • Log out
  • Log in
  • My Cart
Journal of Neuroscience

Advanced Search

Submit a Manuscript
  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE
PreviousNext
Cellular/Molecular

ATP Induces a Rapid and Pronounced Increase in 2-Arachidonoylglycerol Production by Astrocytes, a Response Limited by Monoacylglycerol Lipase

Lisa Walter, Thien Dinh and Nephi Stella
Journal of Neuroscience 15 September 2004, 24 (37) 8068-8074; https://doi.org/10.1523/JNEUROSCI.2419-04.2004
Lisa Walter
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thien Dinh
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nephi Stella
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

The cytoplasm of neural cells contain millimolar amounts of ATP, which flood the extracellular space after injury, activating purinergic receptors expressed by glial cells and increasing gliotransmitter production. These gliotransmitters, which are thought to orchestrate neuroinflammation, remain widely uncharacterized. Recently, we showed that microglial cells produce 2-arachidonoylglycerol (2-AG), an endocannabinoid known to prevent the propagation of harmful neuroinflammation, and that ATP increases this production by threefold at 2.5 min (Witting et al., 2004). Here we show that ATP increases 2-AG production from mouse astrocytes in culture, a response that is more rapid (i.e., significant within 10 sec) and pronounced (i.e., 60-fold increase at 2.5 min) than any stimulus-induced increase in endocannabinoid production reported thus far. Increased 2-AG production from astrocytes requires millimolar amounts of ATP, activation of purinergic P2X7 receptors, sustained rise in intracellular calcium, and diacylglycerol lipase activity. Furthermore, we show that astrocytes express monoacylglycerol lipase (MGL), the main hydrolyzing enzyme of 2-AG, the pharmacological inhibition of which potentiates the ATP-induced 2-AG production (up to 113-fold of basal 2-AG production at 2.5 min). Our results show that ATP greatly increases, and MGL limits, 2-AG production from astrocytes. We propose that 2-AG may function as a gliotransmitter, with MGL inhibitors potentiating this production and possibly restraining the propagation of harmful neuroinflammation.

  • 2-AG
  • ATP
  • lipase
  • cannabinoid
  • astrocyte
  • glia

Introduction

In the CNS, two endocannabinoids have been identified and well characterized: arachidonoylethanolamide (AEA; also known as anandamide) and 2-AG, which activate cannabinoid receptors with distinct pharmacology (Devane et al., 1992; Sugiura et al., 1995; Stella et al., 1997). Recent studies suggest that short-term versus long-term activation of cannabinoid receptors by endocannabinoids induces unique cellular responses. For example, endocannabinoid activation of neuronal CB1 receptors for seconds modulates presynaptic ion channels and inhibits neurotransmitter release (Wilson and Nicoll, 2002), whereas sustained increases in endocannabinoid production and associated activation of cannabinoid receptors for minutes to hours constitutes an “on-demand” defense mechanism limiting cell damage (Marsicano et al., 2003; van der Stelt et al., 2003; Walter and Stella, 2004). In favor of the neuroprotective role of endocannabinoids are studies showing that brain injury leads to a significant increase in endocannabinoid production (Sugiura et al., 2000, 2001; Panikashvili et al., 2001; Baker et al., 2001; Franklin et al., 2003), improving cell survival after neuronal toxicity induced by kainate, ouabain, closed-head injury, ischemia, and experimental autoimmune encephalomyelitis (Sinor et al., 2000; Baker et al., 2001; Panikashvili et al., 2001; van der Stelt et al., 2001; Parmentier-Batteur et al., 2002; Marsicano et al., 2003). The molecular mechanisms downstream of this protective effect include presynaptic inhibition of glutamate release, decreased release of cytotoxins from resident microglial cells and invading immune cells, increased growth factor expression, and lowering of blood pressure and edema (Waksman et al., 1999; Gallily et al., 2000; Cabral et al., 2001; Kreitzer and Regehr, 2001; Parmentier-Batteur et al., 2002; Khaspekov et al., 2004). Whereas the modes of action of endocannabinoids are starting to be understood, their cellular source and the molecular mechanisms controlling their production and inactivation remain unclear.

Endocannabinoid levels are finely tuned by both their production and inactivation. AEA is produced by phospholipase D and hydrolyzed by fatty acid amide hydrolase (FAAH) (Cravatt et al., 2001; Okamoto et al., 2004), and 2-AG is produced by diacylglycerol lipase (DGL) and hydrolyzed by monoacylglycerol lipase (MGL) (Dinh et al., 2002; Bisogno et al., 2003). Many laboratories have reported receptor-mediated increases in endocannabinoid production. To our knowledge the largest and fastest receptor-mediated increase in endocannabinoid production in any cell type has been reported in macrophages. Specifically, platelet-activating factor applied for 45 sec increases 2-AG production by 12-fold in mouse macrophages (Berdyshev et al., 2001). Several reports on the ability of neural cells to produce endocannabinoids are also available. Stimulation of primary rat neurons with glutamate plus carbachol for 2.5 min increases AEA production by 12-fold and 2-AG production by fivefold (Stella and Piomelli, 2001). The same stimulus applied to primary mouse neurons increases AEA production by twofold and 2-AG production by eightfold (Walter et al., 2003). Recently, we showed that ATP increases 2-AG production in primary mouse microglial cells, a response that is not significant at 80 sec (N. Stella, unpublished observation), yet reaches a significant threefold increase at 2.5 min (Witting et al., 2004). Although it is known that astrocytes also produce AEA and 2-AG (Walter et al., 2002; Walter and Stella, 2003), whether ATP affects this production is unknown.

Physiological stimuli lead to synaptic release of ATP in the nanomolar to micromolar range (Edwards and Gibb, 1993), whereas neuropathological conditions associated with cell damage lead to cytoplasmic ATP release in the millimolar range (Dubyak and El-Moatassim, 1993). One pivotal function ascribed to astrocytes is to maintain CNS homeostasis and orchestrate neuroinflammation through their ability to release gliotransmitters (Chen and Swanson, 2003). For example, astrocytes express metabotropic P2Y1,2,4,6&12 and ionotropic P2X1-7 purinergic receptors (James and Butt, 2002; Fumagalli et al., 2003), suggesting that these cells can sense and respond to ATP, and initiate the production of gliotransmitters (John et al., 2001). Because sustained production of endocannabinoids is known to reduce neuroinflammation propagation, and astrocytes produce AEA and 2-AG in a calcium-dependent manner (Walter et al., 2002; Walter and Stella, 2003), we sought to determine the effect of ATP on endocannabinoid production by astrocytes.

Materials and Methods

Materials. ATP, ADP, AMP, adenosine, adenosine 5′-triphosphate-2′,3′-dialdehyde (oxidized ATP), palmityl trifluoromethyl ketone (PTFMK), phenylmethanesulfonyl fluoride (PMSF), methyl arachidonyl fluorophosphonate (MAFP), EGTA, and all other reagents unless specified otherwise were from Sigma (St. Louis, MO). Pertussis toxin was from Calbiochem (La Jolla, CA). RHC80267 was from Biomol (Plymouth Meeting, PA). Arachidonyl trifluoromethyl ketone (ATFMK) was from Tocris Cookson (Ballwin, MO). Radioactive anandamide and 2-AG were from American Radiolabeled Chemicals (St. Louis, MO).

Cell culture. Mouse astrocytes and neurons in primary cultures were prepared from C57BL/6 mice as described (Walter et al., 2002), according to the guidelines of the Institutional Animal Care and Use Committee of the University of Washington (Seattle, WA). Briefly, 6- to 8-week-old astrocytes prepared from postnatal day 1 mice were plated in DMEM supplemented with 10% fetal bovine serum (HyClone, Logan, UT) and penicillin-streptomycin (100 U/ml, 100 μg/ml) for an additional 1 week and allowed to grow to confluency on 100 and 35 mm culture dishes (Corning, Corning, NY) and 13 mm glass coverslips (Fisher Scientific, Houston, TX) in 24-well plates (Corning). One day before use, astrocytes (94% pure as determined by immunofluorescent glial fibrillary acidic protein labeling) (Walter et al., 2002) were rinsed with PBS with high glucose (33 mm), and media was replaced with serum-free MEM supplemented with l-glutamine (2 mm), HEPES (10 mm), NaHCO3 (5 mm), and penicillin-streptomycin (100 U/ml and 100 μg/ml).

Neurons were prepared from day 16 mice embryos using Neurobasal (Invitrogen, San Diego, CA) supplemented with 2% B27 (Invitrogen), l-glutamate (0.024 mm), l-glutamine (0.5 mm), and penicillin-streptomycin (10 U/ml and 10 μg/ml) on 100 mm dishes 1 week before use (Stella et al., 1995).

Lipid extraction and purification. Endocannabinoids and related lipids from cells plated in 100 mm dishes were extracted and purified as previously described (Walter et al., 2002; Walter and Stella, 2003). Briefly, cells maintained in 9 ml of culture media (37°C) were stimulated by adding 1 ml of media containing drugs prepared at 10×. Stimulations were stopped by adding 10 ml of ice-cold methanol and placing dishes on ice. Total lipids were extracted with 20 ml of chloroform containing six internal standards [200 pmol [2H4]-AEA, [2H4]-homo-γ-linolenoyl ethanolamide (HEA), [2H4]-docosatetraenoyl ethanolamide (DEA), [2H4]oleoyl ethanolamide (OEA), [2H4]-palmitoyl ethanolamide (PEA), and [2H8]-2-AG]. Endocannabinoids and related lipids present in organic phases were further purified by open-bed silica gel chromatography followed by HPLC.

Endocannabinoid quantification. Endocannabinoid amounts were quantified by chemical ionization gas chromatography-mass spectrometry (CI GC-MS) as described (Walter et al., 2002; Walter and Stella, 2003), with two important changes.

First, the internal standard used to quantify 2-AG was [2H8]-2-AG instead of [2H4]-AEA, as previously described (Walter and Stella, 2003). When injected into the GC-MS, [2H8]-2-AG yielded a mass spectrum with the base peak at m/z = 441, corresponding to the protonated molecule with the neutral loss of one TMS alcohol ([M + H -90]+) (Fig. 1a). A second major ion was m/z = 457, resulting from the loss of one TMS group ([M + H -74]+) (Fig. 1a). We selected the base peak for 2-AG quantification by isotope dilution.

  Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Chemical ionization gas chromatography-mass spectrometry analysis of endocannabinoids and related lipids. a, Chemical ionization mass spectrum of 200 pmol [2H8]-2-AG yields a base peak at m/z = 441. b, Two hundred picomoles of: PEA, OEA, 2-AG, AEA, HEA, and DEA were coinjected and monitored by selected ion monitoring, yielding clear separation between peaks.

Second, we found that calibration curves generated by directly injecting standards into the GC-MS were different from calibration curves generated with standards added to cell culture media and processed through the HPLC. In particular, the ratio of unlabeled standards to deuterium-labeled standards was higher when measured in media. To be closer to our experimental conditions, we generated calibration curves by adding 0, 2, 20, and 200 pmol of unlabeled standards and 200 pmol deuterium-labeled standards to 10 ml serum-free MEM and extracted and purified them in the same manner as cells. This allows a more accurate quantification of endocannabinoids and related lipids by correcting possible artifacts caused by sample processing. Using this procedure, we monitored six distinct peaks indicating adequately gas chromatography separation (Fig. 1b). Furthermore, this procedure generated linear calibration curves (r2 = 0.99-1.00) and reliable detection limits (2 pmol) for each lipid (data not shown).

Values are mean ± SE of independent quantifications (amount lipid per milligram of protein), each performed on one 100 mm dish of cells. Statistical analyses including ANOVA and Student's t test were performed by Prism (Graph Pad Software, San Diego, CA). Protein in 35 mm dishes of primary astrocytes from the same cell culture preparation was quantified using the DC protein assay (Bio-Rad, Hercules, CA).

Calcium imaging. Calcium imaging was performed as described (Moller et al., 1997). Briefly, astrocytes grown on 13 mm coverslips placed in 24-well plates were incubated in HEPES-bicarbonate buffer containing fura-2-AM (5 μm; Molecular Probes, Eugene, OR) for 45 min at room temperature. Cells were placed in a perfusion chamber on an inverted microscope (Diaphot 200; Nikon, New York, NY) equipped with a 40×, 1.3 numerical aperture oil immersion objective. Fura-2 was excited using a Lambda DG-4 filter system (Sutter Instruments, Novato, CA) at 340 and 380 nm, and fluorescence emission was collected at 510 ± 20 nm via a bandpass filter. Fluorescence acquisition and image analysis were performed using a digital imaging system (R3; Inovision, Cleveland, OH). Ratios were collected at time intervals of 2 sec. To reproduce the experimental conditions used to quantify endocannabinoid production, drugs were added to the chamber under nonperfused conditions.

Reverse-transcription-PCR. Reverse transcription (RT) was performed using superscript first-strand synthesis (Invitrogen). The following mouse MGL primers were used: MGL forward, 5′-CAA GGC CCT CAT CTT TGT GT-3′; and MGL reverse, 5′-ACT TGG AAG TCC GAC ACC AC-3′. Amplification was performed by 35 cycles of: 94°C for 30 sec, 61°C for 30 sec, and 72°C for 2 min. Amplification cycles were followed by 72°C for 10 min. RT-PCR products were separated on 1.2% agarose gels, sequenced, and validated by comparison with the National Center for Biotechnology Information database. Absence of RT-PCR product in the “no RT” reaction confirmed the absence of genomic DNA in the samples.

Western blot. Western blot was performed as described (Walter et al., 2003). Briefly, astrocytes and neurons grown in 100 mm dishes were lysed, and membrane protein fractions were collected after centrifugation at 16,000 × g for 15 min. Samples were electrophoresed (SDS-PAGE), transferred to nitrocellulose, and immunoblotted using an antibody directed against rodent MGL with or without the presence of the immunizing peptide.

MGL and FAAH activity. Cells grown in 100 mm dishes were rinsed with HEPES-bicarbonate buffer, lysed, and homogenized in 50 mm Tris, pH 7.5. For each condition, homogenate (1 ml, 35 μg protein) was incubated with 75 pm [3H(N)]-2-AG for MGL activity or [3H(N)]-AEA for FAAH activity (American Radiolabeled Chemicals) for 5 min at 37°C. Reactions were stopped by adding methanol (1 ml), and lipids were extracted with chloroform containing arachidonic acid and 1-AG for MGL activity or AEA for FAAH activity (each at 20 μm; Cayman Chemicals, Ann Arbor, MI). Samples were separated on silica gel plates (Analtech) using chloroform:methanol:ammonium hydroxide (80:20:1) as solvent system. Lipids were visualized with phosphomolybdic acid (10% in ethanol), bands were scraped off of plates, and radioactivity was determined by scintillation counting. Background radioactivity levels obtained by performing the reaction in the absence of cell homogenate were systematically subtracted.

Results

ATP increases 2-AG but not other endocannabinoids in astrocytes

Quantification of endocannabinoid production by CI GC-MS showed that primary cultures of mouse astrocytes under basal conditions produced 2-AG at 6.88 ± 0.61 pmol/mg, AEA at 3.1 ± 1.2 pmol/mg, HEA at 0.4 ± 0.2 pmol/mg, DEA at 0.5 ± 0.2 pmol/mg, PEA at 13.4 ± 3.6 pmol/mg, and OEA at 0.7 ± 0.1 pmol/mg (n = 35), amounts similar to those previously reported (Walter et al., 2002; Walter and Stella, 2003). When astrocytes were treated with increasing concentrations of ATP for 30 sec, 2-AG levels increased in a dose-dependent manner (Fig. 2a), whereas levels of other endocannabinoids and related lipids measured were unchanged (data not shown).

  Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

ATP selectively, dose-dependently, and time-dependently increases 2-AG production by mouse astrocytes. Mouse astrocytes in primary culture were treated with culture media (basal levels, dotted line) or ATP at increasing concentrations for 30 sec (EC50 = 1.4 mm) (a) or with ATP at 100 μm (closed circles) or 3 mm (open squares) for indicated times (b). Endocannabinoids and related lipids were extracted, purified, and quantified by CI GC-MS. Levels of PEA, OEA, AEA, HEA, and DEA were not significantly affected by ATP (data not shown). Basal levels of all endocannabinoids measured did not change significantly at any time points examined (data not shown). *ANOVA followed by Dunnett's post-test; p < 0.05; n = 4-53.

We determined the time course of this response, using ATP at two concentrations: 100 μm, which reflects physiological levels, and 3 mm, which corresponds to the levels reached during injury. At 100 μm, ATP increased 2-AG production by ninefold at 30 sec (Fig. 2b), which was the only time point with a significant response. At 3 mm, ATP increased 2-AG production 25-fold at 10 sec, a response that reached 60-fold at 2.5 min and decreased thereafter (Fig. 2b). To our knowledge, this is the fastest and largest neurotransmitter-induced increase in 2-AG reported for any cell type.

EctoATPases hydrolyze ATP into ADP, AMP, and adenosine, which also elicit cellular responses (Joseph et al., 2003). To rule out the possibility that the effects of ATP on 2-AG were caused by one of its metabolites, endocannabinoid levels were measured in astrocytes treated with ADP, AMP, and adenosine, each at 100 μm for 30 sec. No significant change in any lipid levels was observed (data not shown), suggesting that ATP itself increased 2-AG in these cells. Because 2-AG production was maximally increased by 3 mm of ATP incubated for 2.5 min, we characterized this response further.

ATP increases 2-AG via purinergic P2X7 receptors and requires extracellular calcium and diacylglycerol lipase activity

Two lines of evidence suggest the involvement of P2X7 receptors in the ATP-induced increase in 2-AG production. First, millimolar concentrations of ATP are required to fully induce this response, and P2X7 is the only purinergic receptor subtype requiring millimolar concentration of ATP for activation, EC50 = 780 μm (Chessell et al., 1998). Second, pretreating astrocytes with oxidized ATP, a P2X7 receptor antagonist (Murgia et al., 1993; Wiley et al., 1994), abolished the 3 mm ATP-induced 2-AG production (Fig. 3a). Note that pertussis toxin treatment, which inhibits Gi/o proteins, did not alter the effect of 3 mm ATP-induced 2-AG production (Fig. 3a), suggesting that P2Y receptors coupled to Gi/o proteins are not involved.

  Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

ATP increases 2-AG via P2X7 receptors and requires extracellular calcium and DGL activity. a, Primary mouse astrocytes were pretreated with culture media (control), P2X7 antagonist oxidized ATP (oATP, 300 μm) for 2 hr, pertussis toxin (PTX, 0.3 μg/ml) for 4 hr, EGTA (5 mm) for 10 min, or DGL inhibitor RHC80267 (30 μm) for 10 min before ATP (3 mm) treatment for 2.5 min. Endocannabinoids and related lipids were extracted, purified, and quantified by CI GC-MS. 2-AG production is expressed as percentage of 3 mm ATP-induced 2-AG response. *ANOVA followed by Dunnett's post-test; p < 0.05; n = 5-13. b, Primary mouse astrocytes were loaded with fura-2-AM dye to measure intracellular calcium levels. Fluorescence was imaged when cells were treated with ATP (3 mm) in the presence (control) or absence of calcium in the buffer (+EGTA), or with ATP (100 μm). Traces are average responses of all the cells in the field of view (∼30 cells), representative of n = 3 separate cultures.

Previous reports indicate that 2-AG production requires extracellular calcium entering cells through ionotropic receptors, which leads to a sustained increase in intracellular calcium ([Ca2+]i) increasing DGL activity (Stella and Piomelli, 2001; Bisogno et al., 2003; Witting et al., 2004). Indeed, preincubation of astrocytes with EGTA, which chelates extracellular calcium and eliminates the [Ca2+]i plateau (Fig. 3b), significantly reduced ATP-induced 2-AG production (Fig. 3a). ATP at 100 μm also yielded a rapid rise in [Ca2+]i, but this response was followed by a much lower [Ca2+]i plateau (Fig. 3b), in agreement with the lower increase in 2-AG levels compared with ATP at 3 mm (Fig. 2b).

DGL, an enzyme that has recently been cloned, is known to generate 2-AG by hydrolyzing diacylglycerol (Bisogno et al., 2003). ATP-induced 2-AG production required DGL activity, because the DGL inhibitor RHC80267 significantly reduced this response (Fig. 3a).

Astrocytes express functional MGL

A rat MGL was recently cloned and shown to be the primary metabolizing enzyme of 2-AG in neurons (Dinh et al., 2002). Three lines of evidence suggest that astrocytes also express MGL. First, using total RNA extracted from cultured astrocytes, we amplified by RT-PCR an amplicon of appropriate size and sequence, suggesting that these cells express mRNA encoding for MGL (Fig. 4a). Second, using an antibody directed against MGL and Western blot analysis (Dinh et al., 2002), we found that cultured astrocytes express a protein that migrates at ∼30 kDa, corresponding to the expected size of MGL as shown in neurons (Fig. 4b), suggesting that mouse astrocytes in culture express MGL protein. Third, cell homogenates of cultured astrocytes hydrolyzed 2-AG radioactively labeled on its arachidonic acid moiety into arachidonic acid in a time-dependent manner (Fig. 4d). Addition of increasing concentrations of radioactive 2-AG (7.5, 25, 75, and 250 pm) led to increased radioactivity in the arachidonic acid fraction (data not shown). This enzymatic activity was inhibited by ATFMK and MAFP, two MGL inhibitors (Dinh et al., 2002), whereas PTFMK and PMSF, two FAAH and PLA2 inhibitors, had no effect (Fig. 4e). In agreement with the noninvolvement of FAAH in this hydrolysis of 2-AG, astrocyte homogenates demonstrated no FAAH activity; i.e., homogenates did not convert AEA into arachidonic acid (Fig. 4c). Finally, MGL activity in homogenates of astrocytes was not significantly affected by ATP, calcium, or EGTA (Fig. 4e), suggesting that this enzyme is not directly regulated by ATP and calcium levels.

  Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Mouse astrocytes express a functional MGL. a, RT-PCR was performed with primers that recognize mouse MGL mRNA. We used reverse-transcribed total RNA from primary mouse astrocytes. RT-PCR products were the appropriate size (162 bp) and sequence. b, Western blot revealed that primary mouse neurons and astrocytes express a protein recognized by a MGL antibody at the appropriate size (∼30 kDa), representative of n = 3 separate cultures. Labeling at this size was eliminated when antibody incubation was performed in the presence of immunizing peptide (data not shown). Astrocyte cell homogenates were incubated with [3H(N)]-AEA (c) or [3H(N)]-2-AG (d) for increasing time points at 37°C. Lipids were chloroform-extracted and analyzed by thin-layer chromatography. Bands with the same retention time as arachidonic acid and AEA (c) or 2-AG (d) were scraped off, and radioactivity was therein determined (n = 3-6). e, MGL activity, as measured by labeled arachidonic acid production, was significantly inhibited by ATFMK (10 μm) and MAFP (10 μm), whereas PTFMK (100 μm) and PMSF (100 μm) had no effect. ATP (3 mm), EGTA (5 mm), and extra calcium (CaCl2, 3 mm) did not affect MGL activity. *ANOVA followed by Dunnett's post-test; p < 0.05; n = 6-16.

Pharmacological control of 2-AG degradation

To determine if MGL controls 2-AG production, cultured astrocytes were pretreated with a MGL inhibitor, treated with ATP (100 μm or 3 mm), and endocannabinoid production was determined. In pilot experiments, we found that MGL activity was maximally inhibited by 10 μm of either ATFMK or MAFP (data not shown), thus we used these concentrations to examine alterations in endocannabinoid production. Basal 2-AG levels were enhanced by fivefold by ATFMK and by fourfold by MAFP (Fig. 5). Although neither inhibitor significantly affected the maximum increase in 2-AG production induced by physiological (100 μm, 30 sec) and injury-level ATP (3 mm, 2.5 min) (data not shown), both inhibitors augmented the sustained response determined at 10 min, a time point when 2-AG production is normally reduced (Fig. 1b). Specifically, ATFMK and MAFP approximately tripled the ATP-induced (100 μm) 2-AG levels (from sevenfold to 24-fold and 19-fold, respectively), and approximately doubled the ATP-induced (3 mm) 2-AG levels (from 46-fold to 74-fold and 113-fold, respectively) (Fig. 5). Note that 100 μm PTFMK did not affect the ATP (3 mm)-induced 2-AG levels determined at 10 min (data not shown). Together, these results suggest the decline in ATP-induced 2-AG production observed at 10 min is caused by MGL activity, and its pharmacological inhibition significantly enhances and prolongs 2-AG production.

  Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Pharmacological inhibition of MGL increases ATP-induced 2-AG production. Primary mouse astrocytes were pretreated with ATFMK (10 μm) or MAFP (10 μm) for 10 min followed by treatment with culture media, 100 μm ATP, or 3 mm ATP (each for 10 min). Endocannabinoids and related lipids were extracted, purified, and quantified by CI GC-MS. 2-AG production is expressed as a percentage of basal 2-AG levels. *Student's t test; p < 0.05; n = 6-14.

Discussion

In this study, we show that ATP increases 2-AG production in mouse astrocytes in primary culture. This response is fast and robust when ATP is applied at injury-level concentrations, occurs via P2X7 receptors, and requires both extracellular calcium and DGL activity. Furthermore, we show that astrocytes express a functional MGL enzyme, the pharmacological inhibition of which further increases and prolongs 2-AG production. To our knowledge, this is the first study reporting that a MGL inhibitor can potentiate receptor-dependent 2-AG production in cells.

Amounts of 2-AG in mouse brain are reported to be between 0.5 and 45 pmol/mg (Baker et al., 2001; Panikashvili et al., 2001; Marsicano et al., 2002; Watanabe et al., 2003; Hanus et al., 2003), and in rat brain between 0.2 and 19 pmol/mg (Sugiura et al., 1995, 2000, 2001; Stella et al., 1997; Bisogno et al., 1999a,b; Kathuria et al., 2003; Ferrer et al., 2003; Di Marzo et al., 2000; Hansen et al., 2001). Which neural cell type accounts for this production of endocannabinoids? Primary cultures of rat neurons produce 16.5 ± 3.3 pmol of 2-AG per milligram of protein (Stella and Piomelli, 2001), whereas primary cultures of mouse neurons only produce 0.4 ± 0.1 pmol of 2-AG per milligram of protein (Walter et al., 2003). Primary cultures of mouse microglial cells produce 7.0 ± 0.8 pmol of 2-AG per milligram (Witting et al., 2004). Here we show that astrocytes produce 6.88 ± 0.61 pmol of 2-AG per milligram of protein [a previously reported measurement was ∼2 pmol/mg (Walter and Stella, 2003)]. Thus, because astrocytes are the most abundant cell type in the CNS, our results indicate that a substantial portion of 2-AG production in the brain may come from these cells.

Our data reinforce the notion that a sustained rise in [Ca2+]i fueled by extracellular calcium is required to increase 2-AG production by cells. Indeed, activation of the P2X7 receptor results in a prolonged opening of its pore, allowing large quantities of extracellular calcium to enter cells (North, 2002). We show that the sustained rise in [Ca2+]i induced by 3 mm of ATP, as well as the increased 2-AG production, are both reduced when EGTA chelates extracellular calcium. Do increases in 2-AG production systematically correlate with sustained rises in [Ca2+]i? This seems to be the case. In neurons, activation of NMDA receptors enhances [Ca2+]i and increases 2-AG production (Stella and Piomelli, 2001). In macrophage-like cells, both platelet-activating factor and lipopolysaccharide induce sustained increases in [Ca2+]i (Khoo et al., 2001; Hoffman et al., 2003) and increase 2-AG production (Berdyshev et al., 2001; Matias et al., 2002). In microglial cells, activation of P2X7 receptors also controls 2-AG production, although this response is much slower and less pronounced (Witting et al., 2004).

Several laboratories have shown that astrocytes inactivate en- docannabinoids. However, whether this occurs through FAAH or MGL remained unclear until recently. Neurons account for the majority of FAAH expressed in brain, as assessed by immunohistochemistry; whereas limited expression of FAAH has been reported in astrocytes, suggesting that these cells play a minor role in AEA inactivation. Indeed, compared with liver homogenates, which express high FAAH activity (Cravatt et al., 1996), homogenates from astrocytes in culture hydrolyze little, if any, AEA (Shivachar et al., 1996) (Fig. 4c). Although FAAH has been suggested to participate in 2-AG breakdown in vitro (Goparaju et al., 1998, 1999), this scenario is unlikely to occur in vivo because FAAH knock-out mice have elevated levels of AEA in the CNS (Cravatt et al., 2001), whereas their 2-AG levels remain unchanged (B. Cravatt, personal communication). Furthermore, although PLA2 enzymes have been involved in 2-AG production (Bisogno et al., 1999a,b), and both ATFMK and MAFP are know to inhibit these enzymes (Huang et al., 1996; Lio et al., 1996), we show that PTFMK, a PLA2 enzyme inhibitor, neither affects 2-AG hydrolysis nor augments the ATP-induced 2-AG production, thus ruling out the involvement of these lipases in this response. Thus, our results suggest that astrocytes express MGL, which hydrolyzes 2-AG and finely tunes its production.

The MGL activity in cultured astrocytes differs from that of cultured microglial cells in one important manner: calcium does not affect MGL activity in astrocytes (Fig. 4e), although it inhibits this activity in microglial cells (Witting et al., 2004). Whether this differential sensitivity toward calcium is caused by astrocytes and microglial cells expressing different variants of MGL or different regulating enzymes altogether might account for the differences in kinetics of increased 2-AG production, a topic currently investigated in our laboratory.

In situ, astrocytes express P2X7 receptors (James and Butt, 2002). Thus, injured neural cells releasing millimolar amounts of ATP might increase 2-AG production from astrocytes as a protective mechanism. Sustained activation of cannabinoid receptors on neurons will inhibit the release of the excitotoxic neurotransmitter glutamate (Shen and Thayer, 1998; Sullivan, 1999). It is also possible that this pronounced production of 2-AG constitutes a means to recruit microglial cells and invading macrophages, because these cells migrate toward 2-AG (Kishimoto et al., 2003; Walter et al., 2003). Because cannabinoids inhibit the release of cytotoxins from microglial cells and invading macrophages and increase their proliferation rate (Waksman et al., 1999; Gallily et al., 2000; Cabral et al., 2001; Carrier et al., 2004), as well as induce growth factor production and reduce edema (Jarai et al., 1999; Marsicano et al., 2003), 2-AG production by astrocytes may aid in slowing or blocking the propagation of neuroinflammation and allied cell damage.

The functions of endocannabinoids in brain are starting to be understood. Here, we suggest that the activity-dependent production of 2-AG by the most prevalent cell type in the CNS may participate in the orchestration of neuroinflammation, possibly by preventing its propagation and harmful effects on neural cells. Pharmacological modulation of the targets involved in endocannabinoid production and degradation may be beneficial for augmenting the neuroprotective properties of the endocannabinoid signaling system.

Footnotes

  • This work was supported by National Institutes of Health Grant DA14486 (N.S.) and United States Public Health Service National Research Service Award GrantT32 GM07270 from the National Institute of General Medical Sciences (L.W.). We thank Dr. Thomas Möller (NS044337) for access to calcium imaging and Dr. Daniele Piomelli (DA12447) for providing MGL antibody. We also thank Dr. Anke Witting for significant scientific input.

  • Correspondence should be addressed to Nephi Stella, Department of Pharmacology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195-7280. E-mail: nstella{at}u.washington.edu.

  • Copyright © 2004 Society for Neuroscience 0270-6474/04/248068-07$15.00/0

References

  1. ↵
    Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Makriyannis A, Khanolkar A, Layward L, Fezza F, Bisogno T, Di Marzo V (2001) Endocannabinoids control spasticity in a multiple sclerosis model. FASEB J 15: 300-302.
    OpenUrlFREE Full Text
  2. ↵
    Berdyshev EV, Schmid PC, Krebsbach RJ, Schmid HH (2001) Activation of PAF receptors results in enhanced synthesis of 2-arachidonoylglycerol (2-AG) in immune cells. FASEB J 15: 2171-2178.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    Bisogno T, Melck D, De Petrocellis L, Di Marzo V (1999a) Phosphatidic acid as the biosynthetic precursor of the endocannabinoid 2-arachidonoylglycerol in intact mouse neuroblastoma cells with ionomycin. J Neurochem 72: 2213-2219.
    OpenUrl
  4. ↵
    Bisogno T, Berrendero F, Ambrosino G, Cebeira M, Ramos JA, Fernandez-Ruiz JJ, Di Marzo V (1999b) Brain regional distribution of endocannabinoids: implications for their biosynthesis and biological function. Biochem Biophys Res Commun 256: 377-380.
    OpenUrlCrossRefPubMed
  5. ↵
    Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A, Matias I, Schiano-Moriello A, Paul P, Williams EJ, Gangadharan U, Hobbs C, Di Marzo V, Doherty P (2003) Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J Cell Biol 163: 463-468.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Cabral GA, Harmon KN, Carlisle SJ (2001) Cannabinoid-mediated inhibition of inducible nitric oxide production by rat microglial cells: evidence for CB1 receptor participation. Adv Exp Med Biol 493: 207-214.
    OpenUrlPubMed
  7. ↵
    Carrier EJ, Kearn CS, Barkmeier AJ, Breese NM, Yang W, Nithipatikom K, Pfister SL, Campbell WB, Hillard CJ (2004) Cultured rat microglial cells synthesize the endocannabinoid 2-arachidonylglycerol, which increases proliferation viaaCB2 receptor-dependent mechanism. Mol Pharm 65: 999-1007.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Chen Y, Swanson RA (2003) Astrocytes and brain injury. J Cereb Blood Flow Metab 23: 137-149.
    OpenUrlCrossRefPubMed
  9. ↵
    Chessell IP, Michel AD, Humphrey PP (1998) Effects of antagonists at the human recombinant P2X7 receptor. Br J Pharmacol 124: 1314-1320.
    OpenUrlCrossRefPubMed
  10. ↵
    Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB (1996) Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 384: 83-87.
    OpenUrlCrossRefPubMed
  11. ↵
    Cravatt BF, Demarest K, Patricelli MP, Bracey MH, Giang DK, Martin BR, Lichtman AH (2001) Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc Natl Acad Sci USA 98: 9371-9376.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R (1992) Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258: 1946-1949.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM (2000) Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson's disease. FASEB J 14: 1432-1438.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, Kathuria S, Piomelli D (2002) Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci USA 99: 10819-10824.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Dubyak GR, El-Moatassim C (1993) Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. Am J Physiol 265: C577-606.
  16. ↵
    Edwards FA, Gibb AJ (1993) ATP -a fast neurotransmitter. FEBS Lett 325: 86-89.
    OpenUrlCrossRefPubMed
  17. ↵
    Ferrer B, Asbrock N, Kathuria S, Piomelli D, Giuffrida A (2003) Effects of levodopa on endocannabinoid levels in rat basal ganglia: implications for the treatment of levodopa-induced dyskinesias. Eur J Neurosci 18: 1607-1614.
    OpenUrlCrossRefPubMed
  18. ↵
    Franklin A, Parmentier-Batteur S, Walter L, Greenberg DA, Stella N (2003) Palmitoylethanolamide increases after focal cerebral ischemia and potentiates microglial cell motility. J Neurosci 23: 7767-7775.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Fumagalli M, Brambilla R, D'Ambrosi N, Volonte C, Matteoli M, Verderio C, Abbracchio MP (2003) Nucleotide-mediated calcium signaling in rat cortical astrocytes: role of P2X and P2Y receptors. Glia 43: 218-230.
    OpenUrlCrossRefPubMed
  20. ↵
    Gallily R, Breuer A, Mechoulam R (2000) 2-Arachidonylglycerol, an endogenous cannabinoid, inhibits tumor necrosis factor-alpha production in murine macrophages, and in mice. Eur J Pharmacol 406: 5-7.
  21. ↵
    Goparaju SK, Ueda N, Yamaguchi H, Yamamoto S (1998) Anandamide amidohydrolase reacting with 2-arachidonoylglycerol, another cannabinoid receptor ligand. FEBS Lett 422: 69-73.
    OpenUrlCrossRefPubMed
  22. ↵
    Goparaju SK, Ueda N, Taniguchi K, Yamamoto S (1999) Enzymes of porcine brain hydrolyzing 2-arachidonoylglycerol, an endogenous ligand of cannabinoid receptors. Biochem Pharmacol 57: 417-423.
    OpenUrlCrossRefPubMed
  23. ↵
    Hansen HH, Schmid PC, Bittigau P, Lastres-Becker I, Berrendero F, Manzanares J, Ikonomidou C, Schmid HH, Fernandez-Ruiz JJ, Hansen HS (2001) Anandamide, but not 2-arachidonoylglycerol, accumulates during in vivo neurodegeneration. J Neurochem 78: 1415-1427.
    OpenUrlCrossRefPubMed
  24. ↵
    Hanus L, Avraham Y, Ben-Shushan D, Zolotarev O, Berry EM, Mechoulam R (2003) Short-term fasting and prolonged semistarvation have opposite effects on 2-AG levels in mouse brain. Brain Res 983: 144-151.
    OpenUrlCrossRefPubMed
  25. ↵
    Hoffman A, Kann O, Ohlemeyer C, Hanisch UK, Kettenmann H (2003) Elevation of basal intracellular calcium as a central element in the activation of brain macrophages (microglia): suppression of receptor-evoked calcium signaling and control of release function. J Neurosci 23: 4410-4419.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Huang Z, Payette P, Abdullah K, Cromlish WA, Kennedy BP (1996) Functional identification of the active-site nucleophile of the human 85-kDa cytosolic phospholipase A2. Biochemistry 35: 3712-3721.
    OpenUrlCrossRefPubMed
  27. ↵
    James G, Butt AM (2002) P2Y and P2X purinoceptor mediated Ca(2+) signalling in glial cell pathology in the central nervous system. Eur J Pharmacol 447: 247-260.
    OpenUrlCrossRefPubMed
  28. ↵
    Jarai Z, Wagner JA, Varga K, Lake KD, Compton DR, Martin BR, Zimmer AM, Bonner TI, Buckley NE, Mezey E, Razdan RK, Zimmer A, Kunos G (1999) Cannabinoid-induced mesenteric vasodilation through an endothelial site distinct from CB1 or CB2 receptors. Proc Natl Acad Sci USA 96: 14136-14141.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    John GR, Simpson JE, Woodroofe MN, Lee SC, Brosnan CF (2001) Extracellular nucleotides differentially regulate interleukin-1beta signaling in primary human astrocytes: implications for inflammatory gene expression. J Neurosci 21: 4134-4142.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Joseph SM, Buchakjian MR, Dubyak GR (2003) Colocalization of ATP release sites and ecto-ATPase activity at the extracellular surface of human astrocytes. J Biol Chem 278: 23331-23342.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, Mor M, Tarzia G, La Rana G, Calignano A, Giustino A, Tattoli M, Palmery M, Cuomo V, Piomelli D (2003) Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med 9: 76-81.
    OpenUrlCrossRefPubMed
  32. ↵
    Khaspekov LG, Brenz Verca MS, Frumkina LE, Hermann H, Marsicano G, Lutz B (2004) Involvement of brain-derived neurotrophic factor in cannabinoid receptor-dependent protection against excitotoxicity. Eur J Neurosci 19: 1691-1698.
    OpenUrlCrossRefPubMed
  33. ↵
    Khoo C, Helm J, Choi HB, Kim SU, McLarnon JG (2001) Inhibition of store-operated Ca(2+) influx by acidic extracellular pH in cultured human microglia. Glia 36: 22-30.
    OpenUrlCrossRefPubMed
  34. ↵
    Kishimoto S, Gokoh M, Oka S, Muramatsu M, Kajiwara T, Waku K, Sugiura T (2003) 2-arachidonoylglycerol induces the migration of HL-60 cells differentiated into macrophage-like cells and human peripheral blood monocytes through the cannabinoid CB2 receptor-dependent mechanism. J Biol Chem 278: 24469-24475.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Kreitzer AC, Regehr WG (2001) Retrograde inhibition of presynaptic calcium influx by endogenous cannabinoids at excitatory synapses onto Purkinje cells. Neuron 29: 717-727.
    OpenUrlCrossRefPubMed
  36. ↵
    Lio YC, Reynolds LJ, Balsinde J, Dennis EA (1996) Irreversible inhibition of Ca(2+)-independent phospholipase A2 by methyl arachidonyl fluorophosphonate. Biochim Biophys Acta 1302: 55-60.
    OpenUrlPubMed
  37. ↵
    Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, Hermann H, Tang J, Hofmann C, Zieglgansberger W, Di Marzo V, Lutz B (2002) The endogenous cannabinoid system controls extinction of aversive memories. Nature 418: 530-534.
    OpenUrlCrossRefPubMed
  38. ↵
    Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, Azad SC, Cascio MG, Gutierrez SO, van der Stelt M, Lopez-Rodriguez ML, Casanova E, Schutz G, Zieglgansberger W, Di Marzo V, Behl C, Lutz B (2003) CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science 302: 84-88.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Matias I, Pochard P, Orlando P, Salzet M, Pestel J, Di Marzo V (2002) Presence and regulation of the endocannabinoid system in human dendritic cells. Eur J Biochem 269: 3771-3778.
    OpenUrlPubMed
  40. ↵
    Moller T, Nolte C, Burger R, Verkhratsky A, Kettenmann H (1997) Mechanisms of C5a and C3a complement fragment-induced [Ca2+]i signaling in mouse microglia. J Neurosci 17: 615-624.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Murgia M, Hanau S, Pizzo P, Rippa M, Di Virgilio F (1993) Oxidized ATP. An irreversible inhibitor of the macrophage purinergic P2Z receptor. J Biol Chem 268: 8199-8203.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    North RA (2002) Molecular physiology of P2X receptors. Physiol Rev 82: 1013-1067.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N (2004) Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem 279: 5298-5305.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Panikashvili D, Simeonidou C, Ben-Shabat S, Hanus L, Breuer A, Mechoulam R, Shohami E (2001) An endogenous cannabinoid (2-AG) is neuroprotective after brain injury. Nature 413: 527-531.
    OpenUrlCrossRefPubMed
  45. ↵
    Parmentier-Batteur S, Jin K, Mao XO, Xie L, Greenberg DA (2002) Increased severity of stroke in CB1 cannabinoid receptor knock-out mice. J Neurosci 22: 9771-9775.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Shen M, Thayer SA (1998) Cannabinoid receptor agonists protect cultured rat hippocampal neurons from excitotoxicity. Mol Pharmacol 54: 459-462.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Shivachar AC, Martin BR, Ellis EF (1996) Anandamide- and delta9-tetrahydrocannabinol-evoked arachidonic acid mobilization and blockade by SR141716A [N-(Piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboximide hydrochloride]. Biochem Pharmacol 51: 669-676.
    OpenUrlCrossRefPubMed
  48. ↵
    Sinor AD, Irvin SM, Greenberg DA (2000) Endocannabinoids protect cerebral cortical neurons from in vitro ischemia in rats. Neurosci Lett 278: 157-160.
    OpenUrlCrossRefPubMed
  49. ↵
    Stella N, Piomelli D (2001) Receptor-dependent formation of endogenous cannabinoids in cortical neurons. Eur J Pharmacol 425: 189-196.
    OpenUrlCrossRefPubMed
  50. ↵
    Stella N, Pellerin L, Magistretti PJ (1995) Modulation of the glutamate-evoked release of arachidonic acid from mouse cortical neurons: involvement of a pH-sensitive membrane phospholipase A2. J Neurosci 15: 3307-3317.
    OpenUrlAbstract
  51. ↵
    Stella N, Schweitzer P, Piomelli D (1997) A second endogenous cannabinoid that modulates long-term potentiation. Nature 388: 773-778.
    OpenUrlCrossRefPubMed
  52. ↵
    Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, Yamashita A, Waku K (1995) 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun 215: 89-97.
    OpenUrlCrossRefPubMed
  53. ↵
    Sugiura T, Yoshinaga N, Kondo S, Waku K, Ishima Y (2000) Generation of 2-arachidonoylglycerol, an endogenous cannabinoid receptor ligand, in picrotoxinin-administered rat brain. Biochem Biophys Res Commun 271: 654-658.
    OpenUrlCrossRefPubMed
  54. ↵
    Sugiura T, Yoshinaga N, Waku K (2001) Rapid generation of 2-arachidonoylglycerol, an endogenous cannabinoid receptor ligand, in rat brain after decapitation. Neurosci Lett 297: 175-178.
    OpenUrlCrossRefPubMed
  55. ↵
    Sullivan JM (1999) Mechanisms of cannabinoid-receptor-mediated inhibition of synaptic transmission in cultured hippocampal pyramidal neurons. J Neurophysiol 82: 1286-1294.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    van der Stelt M, Veldhuis WB, van Haaften GW, Fezza F, Bisogno T, Bar PR, Veldink GA, Vliegenthart JF, Di Marzo V, Nicolay K (2001) Exogenous anandamide protects rat brain against acute neuronal injury in vivo. J Neurosci 21: 8765-8771.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    van der Stelt M, Hansen HH, Veldhuis WB, Bar PR, Nicolay K, Veldink GA, Vliegenthart JFG, Hansen HS (2003) Biosynthesis of endocannabinoids and their modes of action in neurodegenerative diseases. Neurotox Res 5: 183-200.
    OpenUrlPubMed
  58. ↵
    Waksman Y, Olson JM, Carlisle SJ, Cabral GA (1999) The central cannabinoid receptor (CB1) mediates inhibition of nitric oxide production by rat microglial cells. J Pharmacol Exp Ther 288: 1357-1366.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Walter L, Stella N (2003) Endothelin-1 increases 2-arachidonoyl glycerol (2-AG) production in astrocytes. Glia 44: 85-90.
    OpenUrlCrossRefPubMed
  60. ↵
    Walter L, Stella N (2004) Cannabinoids and neuroinflammation. Br J Pharmacol 141: 775-785.
    OpenUrlCrossRefPubMed
  61. ↵
    Walter L, Franklin A, Witting A, Moller T, Stella N (2002) Astrocytes in culture produce anandamide and other acylethanolamides. J Biol Chem 26: 26.
    OpenUrl
  62. ↵
    Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, Mackie K, Stella N (2003) Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci 23: 1398-1405.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Watanabe S, Doshi M, Hamazaki T (2003) n-3 Polyunsaturated fatty acid (PUFA) deficiency elevates and n-3 PUFA enrichment reduces brain 2-arachidonoylglycerol level in mice. Prostaglandins Leukot Essent Fatty Acids 69: 51-59.
    OpenUrlCrossRefPubMed
  64. ↵
    Wiley JS, Chen JR, Snook MB, Jamieson GP (1994) The P2Z-purinoceptor of human lymphocytes: actions of nucleotide agonists and irreversible inhibition by oxidized ATP. Br J Pharmacol 112: 946-950.
    OpenUrlPubMed
  65. ↵
    Wilson RI, Nicoll RA (2002) Endocannabinoid signaling in the brain. Science 296: 678-682.
    OpenUrlAbstract/FREE Full Text
  66. ↵
    Witting A, Walter L, Wacker J, Moller T, Stella N (2004) P2X7 receptors control 2-arachidonoylglycerol production by microglial cells. Proc Natl Acad Sci USA 101: 3214-3219.
    OpenUrlAbstract/FREE Full Text
Back to top

In this issue

The Journal of Neuroscience: 24 (37)
Journal of Neuroscience
Vol. 24, Issue 37
15 Sep 2004
  • Table of Contents
  • About the Cover
  • Index by author
Email

Thank you for sharing this Journal of Neuroscience article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
ATP Induces a Rapid and Pronounced Increase in 2-Arachidonoylglycerol Production by Astrocytes, a Response Limited by Monoacylglycerol Lipase
(Your Name) has forwarded a page to you from Journal of Neuroscience
(Your Name) thought you would be interested in this article in Journal of Neuroscience.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
View Full Page PDF
Citation Tools
ATP Induces a Rapid and Pronounced Increase in 2-Arachidonoylglycerol Production by Astrocytes, a Response Limited by Monoacylglycerol Lipase
Lisa Walter, Thien Dinh, Nephi Stella
Journal of Neuroscience 15 September 2004, 24 (37) 8068-8074; DOI: 10.1523/JNEUROSCI.2419-04.2004

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Respond to this article
Request Permissions
Share
ATP Induces a Rapid and Pronounced Increase in 2-Arachidonoylglycerol Production by Astrocytes, a Response Limited by Monoacylglycerol Lipase
Lisa Walter, Thien Dinh, Nephi Stella
Journal of Neuroscience 15 September 2004, 24 (37) 8068-8074; DOI: 10.1523/JNEUROSCI.2419-04.2004
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Responses to this article

Respond to this article

Jump to comment:

No eLetters have been published for this article.

Related Articles

Cited By...

More in this TOC Section

  • Sex differences in histamine regulation of striatal dopamine
  • A Critical Role of Neuroligin 2 C-Terminus in OCD and Social Behavior
  • Time-Dependent Actions of Corticosterone on Infralimbic Cortex Pyramidal Neurons of Adult Male Rats
Show more Cellular/Molecular
  • Home
  • Alerts
  • Follow SFN on BlueSky
  • Visit Society for Neuroscience on Facebook
  • Follow Society for Neuroscience on Twitter
  • Follow Society for Neuroscience on LinkedIn
  • Visit Society for Neuroscience on Youtube
  • Follow our RSS feeds

Content

  • Early Release
  • Current Issue
  • Issue Archive
  • Collections

Information

  • For Authors
  • For Advertisers
  • For the Media
  • For Subscribers

About

  • About the Journal
  • Editorial Board
  • Privacy Notice
  • Contact
  • Accessibility
(JNeurosci logo)
(SfN logo)

Copyright © 2025 by the Society for Neuroscience.
JNeurosci Online ISSN: 1529-2401

The ideas and opinions expressed in JNeurosci do not necessarily reflect those of SfN or the JNeurosci Editorial Board. Publication of an advertisement or other product mention in JNeurosci should not be construed as an endorsement of the manufacturer’s claims. SfN does not assume any responsibility for any injury and/or damage to persons or property arising from or related to any use of any material contained in JNeurosci.