Skip to main content
Canna~Fangled Abstracts

A new cannabinoid CB2 receptor agonist HU-910 attenuates oxidative stress, inflammation and cell death associated with hepatic ischaemia/reperfusion injury

By September 8, 2013No Comments
pub med big
Logo of brjpharm

Br J Pharmacol. 2012 April; 165(8): 2462–2478.
PMCID: PMC3423243

A new cannabinoid CB2 receptor agonist HU-910 attenuates oxidative stress, inflammation and cell death associated with hepatic ischaemia/reperfusion injury

This article has been cited by other articles in PMC.

Abstract

BACKGROUND AND PURPOSE

Cannabinoid CB2 receptor activation has been reported to attenuate myocardial, cerebral and hepatic ischaemia-reperfusion (I/R) injury.

EXPERIMENTAL APPROACH

We have investigated the effects of a novel CB2 receptor agonist ((1S,4R)-2-(2,6-dimethoxy-4-(2-methyloctan-2-yl)phenyl)-7,7-dimethylbicyclo[2.2.1]hept-2-en-1-yl)methanol (HU-910) on liver injury induced by 1 h of ischaemia followed by 2, 6 or 24 h of reperfusion, using a well-established mouse model of segmental hepatic I/R.

KEY RESULTS

Displacement of [3H]CP55940 by HU-910 from specific binding sites in CHO cell membranes transfected with human CB2 or CB1 receptors (hCB1/2) yielded Ki values of 6 nM and 1.4 µM respectively. HU-910 inhibited forskolin-stimulated cyclic AMP production by hCB2 CHO cells (EC50= 162 nM) and yielded EC50 of 26.4 nM in [35S]GTPγS binding assays using hCB2 expressing CHO membranes. HU-910 given before ischaemia significantly attenuated levels of I/R-induced hepatic pro-inflammatory chemokines (CCL3 and CXCL2), TNF-α, inter-cellular adhesion molecule-1, neutrophil infiltration, oxidative stress and cell death. Some of the beneficial effect of HU-910 also persisted when given at the beginning of the reperfusion or 1 h after the ischaemic episode. Furthermore, HU-910 attenuated the bacterial endotoxin-triggered TNF-α production in isolated Kupffer cells and expression of adhesion molecules in primary human liver sinusoidal endothelial cells stimulated with TNF-α. Pretreatment with a CB2 receptor antagonist attenuated the protective effects of HU-910, while pretreatment with a CB1 antagonist tended to enhance them.

CONCLUSION AND IMPLICATIONS

HU-910 is a potent CB2 receptor agonist which may exert protective effects in various diseases associated with inflammation and tissue injury.

LINKED ARTICLES

This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visithttp://dx.doi.org/10.1111/bph.2011.163.issue-7

Keywords: cannabinoids, oxidative stress, inflammation, ischaemia-reperfusion

Introduction

Ischaemia followed by reperfusion (I/R) is a major mechanism leading to end-organ damage which complicates the course of circulatory shock, organ transplantation, myocardial infarction, stroke and may accompany surgical interventions involving vascular occlusion. The common pathology of these conditions is that the transient disruption of the normal blood supply to target organs followed by reperfusion induces an acute generation of reactive oxygen and nitrogen species subsequent to reoxygenation upon vascular reopening (Ferdinandy and Schulz, 2003Pacher et al., 2007). These changes initiate a chain of deleterious cellular responses leading to inflammation, cell death and eventually culminating in target organ dysfunction or failure (Liaudet et al., 2003Pacher and Hasko, 2008).

Activation of cannabinoid CB2 receptors (nomenclature follows Alexander et al., 2011) has been reported to attenuate injury in preclinical models of myocardial (Montecucco et al., 2009), cerebral (Zhang et al., 2009a,b) and hepatic I/R injury (Batkai et al., 2007Rajeshet al., 2007) in addition to numerous other inflammatory disorders (Pacher et al., 2006Di Marzo, 2008Pacher and Mechoulam, 2011). Furthermore, CB2 receptors protect liver against development of fibrosis (Julien et al., 2005Lotersztajn et al., 2008) and may also play an important role in liver regeneration (Teixeira-Clerc et al., 2010) and protection against alcohol-induced liver injury.

We have already demonstrated that the CB2 receptor agonists HU-308 and JWH-133 afforded protection against hepatic I/R injury only in doses of 10 and 20 mg·kg−1 (given i.p.) (Batkai et al., 2007Rajesh et al., 2007). In this study, we have investigated the effects of a novel CB2 receptor agonist ((1S,4R)-2-(2,6-dimethoxy-4-(2-methyloctan-2-yl)phenyl)-7,7-dimethylbicyclo[2.2.1]hept-2-en-1-yl)methanol (HU-910; Figure 1) with greater in vivo efficacy (effective from 1 mg·kg−1 i.p.) compared with HU-308 and JWH-133, on liver injury induced by 1 h of ischaemia followed by 2, 6 or 24 h of reperfusion, using a well-established mouse model of segmental hepatic I/R (Rajesh et al., 2007Moonet al., 2008Abe et al., 2009Mukhopadhyay et al., 2011b). We have also explored the effects of HU-910 on the production of TNF-α by murine Kupffer cells (key resident macrophage-derived inflammatory cells of the liver) triggered by bacterial endotoxin/lipopolysaccharide (LPS), and on the expression of adhesion molecules in primary human liver sinusoidal endothelial cells (HLSEC) stimulated with TNF-α.

Figure 1

Chemical structure and representative graphs of binding assays of HU-910. (A) Chemical structure of HU-910. (B) Competition binding assays were performed with either CHO-CB1 or CHO-CB2 cellular membranes by displacement of 2.5 nM [3H]CP 55,940. (C) Representative 

Our findings strengthen the potential of HU-910 for the prevention or treatment of I/R injury and other inflammatory disorders.

Methods

Cell line generation and maintenance

The cDNA clones for human CB1 and CB2 receptors tagged with three haemagglutinin (HA) sequences were obtained from the Missouri S&T cDNA Resource Center (http://www.cdna.org) in cloning vector pcDNA3.1 + (pcDNA 3 × HA hCB1/2). The vector containing the human CB2 receptor was transfected directly into CHO-KI cells obtained from ATCC. The HA-tagged human CB1 receptor sequence was subcloned into the pef4-V5-HisA vector with Kpn1 (Roche) and Pme1 (New England Biolabs) restriction enzymes and subsequently transfected into CHO-K1 cells using Lipofectamine 2000 according to the manufacturer’s instructions (Invitrogen). Cells were clonally isolated by limited dilution and screened by immunocytochemistry for expression of the HA tag. Clones expressing the HA tag were also screened by reverse transcription PCR to confirm expression of human CB1 and human CB2 receptor mRNA transcripts.

Cells were maintained in Dulbecco’s modified Eagle medium: nutrient mixture F-12 (DMEM/F12) media supplemented with 10% fetal bovine serum (FBS), 100 units·mL−1penicillin and 100 µg·mL−1 streptomycin and 2 mM L-glutamine. Transfected cell lines were maintained with additional 250 µg·mL−1 zeocin for CHO-CB1 transfected cells and 500 µg·mL−1 G-418 for CHO-CB2 transfected cells (all reagents obtained from Invitrogen).

Membrane preparation

Cells were grown to 90–100% confluence and harvested in ice-cold phosphate buffered saline with 5 mM EDTA. Cells were centrifuged at 200×g for 10 min and frozen at −80°C until required. Cell pellets were thawed with cold 0.32 M sucrose and homogenized with a glass homogenizer. The homogenate was centrifuged at 1000×g for 10 min at 4°C and the supernatant centrifuged in a Sorvall ultracentrifuge for 30 min at 100 000×g. The pellet was then washed in ice-cold Tris wash buffer and re- centrifuged twice more. The final pellet was resuspended in 50 mM Tris pH 7.5, 0.5 mM EDTA. Protein concentration was determined using the Dc protein assay kit (Bio-Rad, Hercules, CA, USA).

Competition binding assay

The Kd of CP 55,940 in the isolated CB1 and CB2 receptor expressing membranes was previously determined to be 2.3 nM and 1.5 nM respectively. Competition binding assays at 2.5 nM [3H]-CP 55,940 (PerkinElmer) were carried out to determine the Ki values for tested compounds. Membranes (5–10 µg) were incubated with radioligand and a range of concentrations of test compounds in binding buffer (50 mM Tris pH 7.4, 5 mM MgCl2, 1 mM EDTA) with 0.5% (w/v) bovine serum albumin (BSA) (ICP Bio, New Zealand), at 30°C for 60 min. Stock solutions of putative cannabinoid ligands were prepared in DMSO to a concentration of 10 mM. Six different final concentrations of compounds were used ranging from 0.1 nM to 50 µM. Non-specific binding was determined in the presence of 1 µM non-radioactive CP 55,940 (Tocris Cookson). Assays were terminated by addition of 3 mL ice-cold binding buffer and filtration through GF/C filters (Whatman) pre-soaked in cold binding buffer, followed by two washes in the same buffer.

Radioactivity was determined by incubation of filters with Irgasafe scintillation fluid (PerkinElmer) and scintillation counting in a Wallac Trilux using Microbeta Trilux software. Data was analysed using the Prism 4.02 program (GraphPad Software, San Deigo, CA, USA).

cAMP assay

CHO-CB1 and CHO-CB2 cells were seeded at a density of 104 cells per well in poly-L-lysine treated 96-well culture plates (BD Biosciences). The following day wells were incubated with 40 µL DMEM/F12 containing 0.5% (w/v) BSA and 0.5 mM 3-isobutyl-1-methylxanthine (Sigma-Aldrich) for 30 min prior to 15 min stimulation with 50 µM forskolin (Tocris Cookson) and varying concentrations of indicated compounds at 37°C, 5% CO2. Assays were stopped by removal of media and addition of 100% ice-cold ethanol. Plates were then frozen for a minimum of 2 h before complete evaporation of ethanol. The well contents were then reconstituted in 50 µL cAMP assay buffer (20 mM HEPES pH 7.5 and 5 mM EDTA). Half of the reconstituted sample was transferred to round bottom 96-well plates (Greiner Bio-One GmbH) with 50 µL 0.01% w/v cAMP dependent protein kinase A [PKA (Sigma-Aldrich) in 1 mM sodium citrate pH 6.5 with 2 mM dithiothreitol] and 25 µL [3H]-cAMP (at 22 nM in cAMP assay buffer) (GE Healthcare, Life Sciences). Samples were then allowed to equilibrate for 3–18 h. Following this, a charcoal slurry [5% (w/v) activated charcoal and 0.2% (w/v) BSA in cAMP assay buffer] was added to the samples and the plates centrifuged at 3000×g, 4°C for 5 min. A sample of the supernatant was then transferred to 96-well flexible microplates (PerkinElmer) and 200 µL Irgasafe scintillation fluid (PerkinElmer) added. Plates were sealed, vigourously agitated and scintillation counting performed by a Wallac Trilux using Microbeta Trilux software.

[35S]GTPγS binding assay

Human CB2 receptor expressing CHO-K1 membranes (5 µg per incubation mixture) were diluted in 50 mM Tris-HCl (pH 7.5) and 0.5 mM EDTA and added to the indicated compounds in a pre-mixed incubation cocktail. Final incubation concentrations were 55 mM Tris-HCl (pH 7.4), 1 mM EDTA, 100 mM NaCl, 5 mM MgCl2, 0.5% BSA, 50 µM GDP, 0.2 nM [35S]GTPγS (PerkinElmer) with varied concentrations of compounds (0.1 nM–10 µM) and 5 µg membrane. Incubations were continued for 60 min at 30°C in a shaking water bath. Assays were terminated by addition of 2 mL ice-cold wash buffer (50 mM Tris-HCl, pH 7.5 and 5 mM MgCl2) and filtration through pre-soaked GF/C filters (Whatman), followed by two further washes. Radioactivity was determined as described for competition binding assays.

Hepatic ischaemia-reperfusion

All animal care and experimental procedures complied with the National Institutes of Health (NIH) guidelines for the care and use of laboratory animals and were approved by the Institutional Animal Care and Use Committees of the National Institute on Drug Abuse (NIAAA). Male C57BL/6J mice (25–30 g; Jackson Laboratories, Bar Harbor, ME, USA) were anesthetized with pentobarbital sodium (65 mg·kg−1 i.p.). We used the model of segmental (70%) hepatic ischaemia, as described by Mukhopadhyay et al., (2011b). Briefly, the liver was exposed by midline laparotomy and the hepatic artery and the portal vein were clamped using an atraumatic micro-serrefine. This method of partial ischaemia prevents mesenteric venous congestion by allowing portal decompression throughout the right and caudate lobes of the liver. The duration of hepatic ischaemia was 60 min, after which the vascular clips were removed and liver was reperfused for 2, 6 or 24 h, as indicated. Sham surgeries were identical except that hepatic blood vessels were not clamped with a micro-serrefine. The liver was kept moist at 37°C with gauze soaked in 0.9% saline. Body temperature was monitored with a rectal temperature probe and was maintained at 37°C by a heating blanket. Treatments with HU-910 0.3, 1, 3 and 10 mg·kg−1or vehicle (i.p.), started 2 h before I/R or were given after ischaemia at the moment of reperfusion, as indicated in the text. CB1 and CB2 receptor antagonists were given 2 h before ischaemia or HU-910 treatment. At the experimental end points, blood was collected and liver samples were removed and snap-frozen in liquid nitrogen for determining biochemical parameters or fixed in 4% buffered formalin for histopathological evaluation (Moon et al., 2008).

Serum aspartate amino-transferase (AST) and alanine amino-transferase (ALT) levels

The activities of AST and ALT, indicators of liver cellular damage (necrosis), were measured in serum samples using a clinical chemistry analyser system (VetTest 8008, IDEXX laboratories, Westbrook, ME, USA) (Moon et al., 2008).

Histological examination of liver sections

Liver samples were fixed in 4% buffered formalin. After embedding and cutting 5 µm slices, all sections were stained with haematoxylin/eosin. Neutrophils were stained for myeloperoxidase (MPO) using anti-MPO antibody, according to the manufacturer’s protocol (Biocare Medical, Concord, CA, USA), and samples were counter-stained with nuclear fast red as described (Mukhopadhyay et al., 2011b). Histological evaluation was performed without knowledge of the treatments.

Real-time PCR analyses of mRNA

Total RNA was isolated from liver homogenate using TRIzol reagents (Invitrogen, Carlsbad, CA, USA) according to manufacturer’s instructions. The isolated RNA was treated with RNase-free DNase (Ambion, Austin, TX, USA) to remove traces of genomic DNA contamination. Samples (1µg) of total RNA were reverse-transcribed to cDNA using the SuperScript II (Invitrogen, Carlsbad, CA, USA). The target gene expression was quantified with Power SYBER Green PCR Master Mix using an ABI HT7900 real-time PCR instrument (Applied Biosystems, Foster City, CA, USA). Each amplified sample in all wells was analysed for homogeneity using dissociation curve analysis. After denaturation at 95°C for 2 min, 40 cycles were performed at 95°C for 10 s and at 60°C for 30 s. Relative quantification was calculated using the comparative CT method (2-ΔΔCt method: ΔΔCt =ΔCt sample −ΔCt reference). Lower ΔCT values and lower ΔΔCT reflect a relatively higher amount of gene transcript.

Primers used were as follows:

CCL3, 5′-TGCCCTTGCTGTTCTTCTCTG-3′ and 5′-CAACGATGAATTGGCGTGG-3′; CXCL2, 5′-AGTGAACTGCGCTGTCAATGC-3′ and 5′-AGGCAAACTTTTTGACCGCC-3′; TNF-α, 5′-AAGCCTGTAGCCCACGTCGTA-3′ and 5′-AGGTACAACCCATCGGCTGG-3′; ICAM-1, 5′-AACTTTTCAGCTCCGGTCCTG-3′ and 5′-TCAGTGTGAATTGGACCTGCG-3′; CB1, 5′-ATGAAGTCGATCCTAGATGGCCTTGCAGA-3′ and 5′TCACAGAGCCTCGGCAGACGTG-3′; CB2, 5′-GACCTTCACAGCCTCTGTGGGTA-3 and 5′-GATTTTCCCATCAGCCTCTGTCT-3; and actin, 5′-TGCACCACCAACTGCTTAG-3′ and 5′-GGATGCAGGGATGATGTTC-3′.

Hepatic 4-hydroxynonenal (HNE) content

Lipid peroxides are unstable indicators of oxidative stress in cells that decompose to form more complex and reactive compounds such as HNE, which has been shown to be capable of binding to proteins and forming stable HNE adducts. HNE in the hepatic tissues was determined using a kit (Cell Biolabs, CA, USA). In brief, BSA or hepatic tissue extracts (10 µg·mL−1) are adsorbed onto a 96-well plate for 12 h at 4°C. HNE adducts present in the sample or standard were probed with anti-HNE antibody, followed by an horseradish peroxidase (HRP)-conjugated secondary antibody. The content of HNE-protein adducts in an experimental sample was determined by comparing with a standard curve (Mukhopadhyay et al., 2010).

Detection of hepatic carbonyl adducts

Carbonyl content in liver tissues was determined by OxiSelect Protein Carbonyl elisa Kit (Cell Biolabs, CA, USA) (Mukhopadhyay et al., 2011a). In brief, BSA standards or protein samples (10 µg·mL−1) were adsorbed onto a 96-well plate for 2 h at 37°C. The protein carbonyls present in the sample or standard were derivatized to DNP hydrazone and probed with an anti-DNP antibody, followed by an HRP-conjugated secondary antibody. The protein carbonyl content in unknown sample was determined by comparing with a standard curve, prepared from reduced and oxidized BSA standards.

Detection of apoptosis by caspase 3/7 activity assays

Caspase 3/7 activity in hepatic tissue lysate was measured using the Apo-One Homogenous Caspase-3/7 assay kit (Promega Corp., Madison, WI, USA). An aliquot of caspase reagent was added to each well and mixed on a plate shaker for 1 h at room temperature shielded from light, and the fluorescence was measured (Rajesh et al., 20092010).

Hepatic DNA fragmentation elisa

The quantitative determinations of cytoplasmic histone-associated-DNA-fragmentation (mono and oligonucleosomes) due to cell death in liver homogenizates were measured using elisa kit (Roche Diagnostics GmbH) (Mukhopadhyay et al., 2009Rajesh et al., 2010).

Isolation and stimulation of hepatic Kupffer cells

Livers were perfused in deeply anaesthetized mice and the livers were excised for isolation of Kupffer cells. Hepatic cellular contents were released from the stroma by digestion with the perfusion medium (hepatocyte wash media) containing collagenase type 1 (Sigma, St. Louis, MO, USA) and 2% Penicillin and Streptomycin (Invitrogen, Carlsbad, CA, USA). Kupffer cells were isolated using Optiprep gradient. Kupffer cell fraction was aspirated and washed twice with RPMI 1640 medium. Homogenous Kupffer cells population was obtained by using negative selection (anti-CD146) LS column according to the manufacturer’s instruction (Miltenyi Biotec, Auburn, CA, USA) as described earlier (Mukhopadhyay et al., 2011b). After additional washes, Kupffer cells were plated on to 96-well or six-well plates in RPMI 1640 media, containing 10% FBS and penicillin-streptomycin, and incubated overnight in a CO2 incubator at 37°C and 5% CO2. Cells were maintained for 2 h with RPMI 1640 medium containing FBS (2%). HU-910 or HU-308 or the CB2 receptor antagonists (SR144528 or AM-630) at indicated concentrations were added 1 h prior to LPS treatment. Cells were treated for 6 h with LPS (Escherichia coliO127:BB catalogue # L3129, Sigma Chemicals, St Louis, MO, USA). After the end of treatments, culture supernatants were removed and snap frozen in liquid nitrogen and assayed for the TNF-α concentrations with the use of elisa kit (Mouse TNF-α; catalogue # SMTA00; R&D Systems, Minneapolis, MN, USA) as described by Mukhopadhyay et al., (2011b).

Cell surface inter-cellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expression assay

Cell surface expression of ICAM-1 and VCAM-1 in the HLSEC was measured by in situelisaas described (Rajesh et al., 2007). In brief, HLSEC cells were grown in 96-well plates. After treatments as described in Figure 14in situelisa was performed with anti-mouse ICAM-1 or VCAM-1 monoclonal antibodies (1:1500 dilution; R&D Systems) and by measuring the absorbance colorimetrically at 450 nm using the HRP-3,3′,5,5′-tetramethylbenzidine developing system (Sigma, St. Louis, MO, USA). Each treatment was performed in triplicate, and the experiments were repeated three times.

Figure 14

HU-910 attenuates the TNFα-induced adhesion molecules expression in human liver sinusoidal endothelial cells (HLSEC). Treatment of HLSEC cells with 50 ng·mL−1 TNF-α for 6 h, markedly enhances the expression of adhesion 

Analysis of data

For binding data the Ki was determined from IC50 values derived from competition binding data fitted with one site competition non-linear regression analysis by Prism 4.02 using the Kd values shown in Table 1. pIC50 values were determined from cAMP assays by fitting a sigmoidal concentration response curve. Results shown were generated by averaging at least three independently determined pIC50 values. Data shown is mean IC50(95% confidence interval). Emax values were calculated as a percentage of the maximal response detected in parallel cAMP assays with HU-210 or HU-308 for CB1 and CB2receptor expressing cells respectively. Data are displayed as the mean ± SEM.

Table 1

HU-910 activates CB2 receptors in vitro

For all experiments presented in Figures 214, values have been expressed as means and variability as SEM. Statistical significance among groups was determined by one-way anovafollowed by Newman–Keuls post hoc analysis using GraphPad Prism 5 software (San Diego, CA, USA). Probability values of P < 0.05 were considered significant.

Figure 2

HU-910 attenuates hepatic ischaemia/reperfusion (I/R) injury. (A,B) Serum transaminase alanine amino-transferase (ALT) (A) and aspartate amino-transferase (AST) (B) levels in sham operated mice treated with vehicle (Veh) or HU-910 (n = 4–5) or 

Materials

The chemical structure of HU-910 is shown in Figure 1. The synthesis of HU-910 has been presented at a meeting of the International Cannabinoid Research Society (Magid et al., 2010a) and in a patent (Magid et al., 2010b). HU-910 has a unique bicyclic structure, in the non-aromatic portion of the molecule. Its synthesis follows a number of steps starting with a Suzuki cross-coupling reaction using (+)-camphor-10-sulphonyl chloride (Sigma-Aldrich) and 2,6-dimethoxy-4-(2-methyloctan-2-yl)benzene (Dominianni et al., 1977).

The CB1 and CB2 receptor antagonists/inverse agonists SR141716A and SR144528 were obtained from the National Institute on Drug Abuse (NIDA) Drug Supply Program. For in vivo administration, all drugs were dissolved in vehicle solution (one drop of Tween-80 in 3 mL 2.5% dimethyl sulphoxide in saline) as previously described (Batkai et al., 2007). Vehicle solution was used in control experiments.

Results

As shown in Table 1 and in Figure 1B–D, HU-910 behaved in all assays as a potent agonist at human CB2 receptors (comparable to the reference CB2 receptor agonist HU-308).

HU-910 attenuates markers of hepatic I/R injury (ALT, AST)

For assessments of hepatocellular damage of the post-ischaemic liver, the serum transaminase activities (AST and ALT), markers of necrosis, were measured. After 1 h of ischaemia and a subsequent 2 or 6 h of reperfusion (I/R 2 h and I/R 6 h respectively), a dramatic increase in liver enzyme activities were observed in vehicle-treated C57Bl6/J mice as compared with sham-operated controls, which almost returned to baseline at 24 h of reperfusion (I/R 24 h) (Figure 2). HU-910 was able to attenuate these increases in the markers of hepatic I/R injury at all measured time points, while HU-910 alone had no effects on ALT and AST levels in the sham animals compared with the vehicle-treated group (Figure 2).

Because the peak elevation of ALT and AST is known to occur around 6 h of reperfusion (Abe et al., 2009Mukhopadhyay et al., 2011b), as also demonstrated in Figure 2, this time point was chosen to evaluate the maximal hepatocellular injury (necrosis) in the subsequent experiments.

Pretreatment with HU-910 (0.3, 1, 3 or 10 mg·kg−1 i.p.) 2 h before the induction of the ischaemia dose-dependently attenuated the serum transaminase elevations (ALT and AST) at 6 h of reperfusion compared with vehicle (Figure 3). Furthermore, pretreatment with the CB2 receptor antagonist SR144528 (3 mg·kg−1 i.p.) significantly attenuated the effect of 3 or 10 mg·kg−1 HU-910 at the time of the peak serum ALT and AST elevations, implying a role for CB2 receptors in the beneficial effects of HU-910 (Figure 3). Because the protective effect of 3 and 10 mg·kg−1 of HU-910 were similar (Figure 3), and the protection afforded by the lower dose could almost completely be prevented by pretreatment with SR144528 (3 mg·kg−1 i.p.), 3 mg·kg−1 HU-910 was used in the subsequent experiments. Given alone, SR144528 (3 mg·kg−1) did not affect the I/R-induced increase in liver enzymes, while pretreatment with the CB1 receptor antagonist SR141716A (3 mg·kg−1 and 10 mg·kg−1) alone attenuated the markers of hepatic I/R injury and potentiated the effect of HU-910, when given in combination (Figure 3). At higher doses (above 10 mg·kg−1), SR144528 alone did attenuate the hepatic I/R injury (data not shown).

Figure 3

HU-910 dose-dependently attenuates hepatic ischaemia/reperfusion (I/R) injury: role of CB1/2receptors. (A,B) Serum transaminase alanine amino-transferase (ALT) (A) and aspartate amino-transferase (AST) (B) levels in sham operated mice treated with vehicle 

HU-910 treatment given immediately after the induction of the ischaemia (Figure 4A) or 1 h following the reperfusion (Figure 4B) still attenuated, although to a lesser extent than in the pretreatment experiments, the hepatic injury measured at 6 h of reperfusion.

Figure 4

HU-910 treatment at reperfusion or 1 h later attenuates hepatic ischaemia/reperfusion (I/R) injury. Serum transaminase alanine amino-transferase (ALT) and aspartate amino-transferase (AST) levels in mice exposed to 1 h of hepatic ischaemia followed by 

HU-910 improves I/R-induced histological damage

Haematoxylin and eosin staining of representative liver sections after 24 h of reperfusion showed (Figure 5) that I/R induced marked coagulation necrosis (lighter areas, with marked inflammatory cell infiltration), which was dramatically reduced and became more focal in mice treated with HU-910 (3 mg·kg−1 i.p.). Pretreatment with the CB2 receptor antagonist/inverse agonist SR144528 (3 mg·kg−1 i.p.) almost completely prevented the protective effect of HU-910, as seen on liver histology. Vehicle or HU-910 treatment in the sham animals had no effect on liver histopathology. A similar histological profile was seen throughout the group (n = 3–5 mice).

Figure 5

HU-910 attenuates histological damage at 24 h following ischaemia. Haematoxylin and eosin staining of representative liver sections of sham mice treated with vehicle (Sham), and mice exposed to 1 h of ischaemia followed by 24 h of reperfusion treated 

HU-910 attenuates the marked neutrophil infiltration induced by I/R

Neutrophils are important mediators of the delayed tissue injury following I/R. Most of the hepatic neutrophil infiltration is known to occur between 6 and 24 h of reperfusion (Abe et al., 2009Mukhopadhyay et al., 2011b). An indicator of neutrophil infiltration is the tissue MPO activity. In sham-treated mice, MPO staining was barely detectable (Figure 6). In contrast, there was a marked increase in infiltrating MPO-positive immune cells (brown staining) after 24 h of reperfusion in vehicle treated animals, which was significantly attenuated by HU-910 3 mg·kg−1 i.p. (Figure 6). Pretreatment with the CB2 receptor antagonist SR144528 (3 mg·kg−1 i.p.) almost completely prevented the effect of HU-910 seen on hepatic neutrophil infiltration. Vehicle or HU-910 treatment in the sham animals had no effect on liver MPO staining. A similar histological profile was seen throughout the group (n = 3–5).

Figure 6

HU-910 attenuates the ischaemia/reperfusion (I/R)-induced increased neutrophil infiltration. (A) Myeloperoxidase (MPO) staining (brown) of representative liver sections of sham mice treated with vehicle (Sham), and mice exposed to 1 h of ischaemia followed 

HU-910 attenuates the hepatic pro-inflammatory chemokine, cytokine and adhesion molecule expression induced by I/R

I/R greatly increased the expression of mRNA of pro-inflammatory chemokines CCL3, CXCL2 and CCL2 (Figure 7A–C), the pro-inflammatory cytokine TNF-α (Figure 8A) and the adhesion molecule ICAM-1 (Figure 8B) in liver tissue as documented by real-time PCR, which was attenuated by HU-910 (3 mg·kg−1 i.p.) given before the ischaemia (Figures 7and ​and8).8). Hepatic TNF-α, CCL3, CXCL2 and CCL2 and ICAM-1 mRNA (part of the acute inflammatory response orchestrated by activated Kupffer and endothelial cells) were maximal at 2 h of reperfusion (Figures 7 and ​and8)8) and decreased by 24 h of reperfusion almost to control levels. HU-910 given before ischaemia significantly decreased the peak values of CCL3, CXCL2 and CCL2 (Figure 7A–C), TNF-α (Figure 8A) and ICAM-1 (Figure 8B) mRNA in liver. These effects could be largely attenuated by pretreatment with the CB2 receptor antagonist SR144528 (3 mg·kg−1 i.p.) (Figures 7 and ​and88).

Figure 7

HU-910 attenuates the ischaemia/reperfusion (I/R)-induced acute pro-inflammatory chemokine response in the liver. Real-time PCR shows significant increase of hepatic pro-inflammatory chemokine CCL3 (A), CXCL2 (B), CCL2 (C) mRNA levels at 2 h of reperfusion 
Figure 8

HU-910 attenuates the ischemia/reperfusion (I/R)-induced acute pro-inflammatory cytokine and adhesion molecule response in the liver. Real-time PCR shows significant increase of pro-inflammatory cytokine TNF-α (A), and adhesion molecule ICAM-1 

HU-910 decreases the increased oxidative stress and hepatocyte cell death induced by I/R

The rate of oxidative posttranslational modification of proteins and lipid peroxidation was negligible in sham livers, as indicated by the low level of carbonyl adducts and HNE. We found a time-dependent increase in HNE peaking at 24 h of reperfusion, which was significantly attenuated by HU-910 (3 mg·kg−1 i.p.) (Figure 9A). Hepatic content of carbonyl adducts also peaked at 24 h of reperfusion, which was significantly attenuated by HU-910 (3 mg·kg−1 i.p.) (Figure 9B).

Figure 9

HU-910 attenuates the ischaemia/reperfusion (I/R)-induced increased oxidative stress and apoptotic cell death. (A) HNE adducts (a marker for lipid peroxidation/oxidative stress); (B) Oxidative modification of proteins measured by carbonyl adducts; (C,D) 

Apoptotic cell death in the liver following I/R was evaluated by monitoring caspase 3/7 activity and DNA fragmentation (Figure 9C,D). All markers of cell death in the liver were markedly increased at 24 h of reperfusion following 60 min of ischaemia. HU-910 (3 mg·kg−1 i.p.) significantly attenuated the increase of both markers (Figure 9C,D).

All effects of HU-910 on oxidative and cell death markers were largely prevented by the CB2 receptor antagonist SR144528 (3 mg·kg−1 i.p.) (Figure 9A–D).

HU-910 treatment (administered after ischaemia) attenuates histological damage, oxidative stress, cell death and acute pro-inflammatory response following hepatic ischaemia-reperfusion injury

I/R induced marked coagulation necrosis (lighter areas, with marked inflammatory cell infiltration), which was dramatically reduced and became more focal in mice treated with HU-910 (3 mg·kg−1 given immediately after the ischaemic period) (Figure 10A). HU-910 also attenuated the observed increase in infiltrating MPO-positive immune cells (brown staining) after 24 h of reperfusion in vehicle-treated animals (Figure 10B), and also the markers of oxidative stress (protein carbonyl, HNE) and apoptotic cell death (caspase 3/7 activity and DNA fragmentation) (Figure 11A–D).

Figure 10

HU-910 treatment (administered after ischaemia) attenuates histological damage at 24 h following ischaemia. (A) Haematoxylin and eosin staining of representative liver sections of mice exposed to 1 h of ischaemia followed by 24 h of reperfusion treated 
Figure 11

HU-910 treatment (administered after ischaemia) attenuates oxidative stress and cell death at 24 h following ischaemia. Ischaemia/reperfusion (I/R) induced significant increases at 24 h of reperfusion in hepatic oxidative stress markers [protein carbonyl 

Furthermore, HU-910 (3 mg·kg−1; administered immediately after the ischaemic period) was also able to attenuate the increased expression of mRNA of CCL3, CXCL2 and CCL2 (Figure 12A–C), of the adhesion molecule ICAM-1 (Figure 12D) and of TNF-α (Figure 12E), after 6 h of reperfusion.

Figure 12

HU-910 (administered after ischaemia) attenuates the ischaemia/reperfusion (I/R)-induced pro-inflammatory response in the liver at 6 h following ischaemia. Real-time PCR shows significant increase of mRNA for chemokines CCL3 (A), CXCL2 (B), CCL2 (C), 

Only the higher dose of HU-910 (10 mg·kg−1) was able to improve the histopathological injury and attenuate the neutrophil infiltration when it was given 1 h after the ischaemic period (data not shown).

HU-910 attenuates the TNF-α production in mouse Kupffer cells induced by LPS

Because the acute inflammatory response in the liver is orchestrated mainly by activated Kupffer and endothelial cells, which express CB2 receptors (Rajesh et al., 2007Hall et al., 2010Pacher and Mechoulam, 2011), we also tested the concentration-dependent effect of HU-910 on inflammatory responses of these cell types. Kupffer cells were isolated from mice as described in the Methods section, and then treated with bacterial LPS (100 ng·mL−1) in the presence or absence of HU-910 (10 nM–10 µM), and the CB2 receptor antagonist SR144528 (1–6 µM). As shown in Figure 13, LPS treatment drastically enhanced TNF-α production in Kupffer cells, which was attenuated in a concentration-dependent manner by HU-910. The effect of 3 µM HU-910 was largely prevented by pretreatment with the CB2 receptor antagonist SR144528 at 1 µM. Interestingly, higher concentrations of SR144528 by themselves attenuated the TNF-α production induced by LPS, indicating a non-specific effect at these concentrations. Addition of vehicle, HU-910 (10 µM) or SR144528 (1 µM) had no effect on the negligible baseline TNF-α levels in unstimulated Kupffer cells.

Figure 13

HU-910 attenuates lipopolysaccharide (LPS)-induced TNF-α secretion in Kupffer cells. HU-910 (10 nM-10 µM) attenuates LPS-induced TNF-α secretion of Kupffer cells in a concentration-dependent manner. The CB2 receptor antagonist 

HU-910 attenuates the adhesion molecules expression in HLSEC induced by TNF-α

Treatment of HLSEC cells with TNF-α (50 ng·mL−1) for 6 h markedly enhanced the production of the adhesion molecules, ICAM-1 and VCAM-1 (Figure 14). This was inhibited when cells were treated with HU-910 (10 nM–3 µM). The effect of HU-910 (3 µM) was largely attenuated by pretreatment with the CB2 receptor antagonist SR144528 at 1 µM. Vehicle, HU-910 (3 µM) or SR144528 (1 µM) had no effect on baseline TNF-α levels in unstimulated HLSEC.

Discussion

In the current study, using various in vitro assays we have demonstrated that HU-910 was a potent agonist of CB2 receptors. In these in vitro assays, the pharmacological properties of HU-910 on CB2 receptors were similar to those of HU-308 (Hanus et al., 1999) and JWH-133 (Huffman, 2005Pertwee, 2005). However, HU-910 had greater in vivo efficacy compared with the two earlier CB2 receptor agonists, by at least one order of magnitude, in terms of attenuating acute and delayed inflammatory response and interrelated oxidative stress and cell death in a murine model of hepatic I/R (Batkai et al., 2007Rajesh et al., 2007), which is at least in part mediated by activation of CB2 receptors. We also showed that HU-910 attenuated LPS-triggered TNF-α production in isolated Kupffer cells, and the expression of adhesion molecules in primary HLSEC stimulated with TNF-α.

I/R injury in the liver is a common complication of prolonged surgical procedures, liver transplantation and circulatory shock. The initial damage is inflicted by a sequence of events, when the liver is transiently deprived of its blood supply followed by reoxygenation. These include acute generation of reactive oxygen and nitrogen species from the activation of various cellular and subcellular sources, such as xanthine oxido-reductases and mitochondria (Engerson et al., 1987Pacher et al., 2006)] during the early reperfusion leading to increased lipid peroxidation, oxidative modification of key proteins involved in cell survival or death, energy metabolism or energy supply (Moon et al., 2008), and oxidative DNA damage (Gero and Szabo, 2006Jaeschke, 2003). The first line of defence against tissue injury is provided by the endothelial cells which respond to oxidative stress by activation, resulting in release of various inflammatory mediators and increased expression of adhesion molecules. In concert with the endothelial cell activation, the resident macrophages of the liver, the Kupffer cells, are also rapidly activated by reperfusion, generating large amounts of pro-inflammatory chemokines and cytokines which prime and facilitate the recruitment of neutrophils and other inflammatory cells into the liver vasculature upon reperfusion. The inflammatory cells attached to the activated endothelium release more reactive oxidants and pro-inflammatory mediators resulting in endothelial dysfunction and disruption of the endothelial barrier. Through the damaged endothelium, inflammatory cells can transmigrate to the parenchyma and attach to and damage hepatocytes by releasing proteolytic enzymes and oxidants, ultimately leading to cell death (both apoptotic and necrotic) and organ failure (Jaeschke, 2006Pacher and Hasko, 2008).

Consistent with earlier reports (Moon et al., 2008Abe et al., 2009Mukhopadhyay et al., 2011b), we found that the acute inflammatory response in our hepatic I/R model peaked between 2 and 6 h of reperfusion, with marked increases (up to over 30-fold) in mRNA for TNF-α, CCL3, CXCL2 and CCL2 and ICAM-1 in liver, which declined thereafter by 24 h of reperfusion (Moon et al., 2008Abe et al., 2009Mukhopadhyay et al., 2011b). The peak hepatocyte necrosis occurred at 6 h of reperfusion (indicated by peak elevations of serum ALT and AST levels), which gradually returned close to normal levels by 24 h of reperfusion, indicating that the predominant type of cell death at the earlier time points of reperfusion is necrotic. As the chronic inflammatory reaction develops (from 12 h of reperfusion), the histological picture of post-ischaemic hepatic morphology at 24 h of reperfusion is characterized by marked coagulation necrosis (lighter areas) with massive inflammatory MPO-positive neutrophil cell infiltration (Figures 5​,66 and ​and10),10), which parallels with increases in oxidative stress (indicated by HNE and protein carbonyl levels) and apoptotic cell death (caspase 3/7 activity and DNA fragmentation).

There is considerable interest in the development of novel selective CB2 receptor agonists, which lack the psychoactive properties of CB1 receptor agonists, for the treatment of various inflammatory and other disorders (Pacher and Mechoulam, 2011). As mentioned earlier, numerous recent studies using potent CB2 receptor agonists and/or knockout mice have provided compelling evidence that CB2 receptor activation is protective against myocardial (Montecucco et al., 2009), cerebral (Zhang et al., 20072009a,bMurikinati et al., 2010) and hepatic (Batkai et al., 2007Rajesh et al., 2007) I/R injuries by decreasing the endothelial cell activation/inflammatory response [e.g. expression of adhesion molecules, secretion of chemokines (Batkai et al., 2007Rajesh et al., 2007)], and by attenuating the leukocyte chemotaxis, rolling, adhesion to endothelium, activation and transendothelial migration, and interrelated oxidative/nitrosative damage (Pacher and Hasko, 2008Zhang et al., 2009a).

In agreement with the above mentioned reports, we found that the peak elevations of serum liver transaminases (ALT/AST) occurred at 6 h following reperfusion (Figure 2), and were dose-dependently (1–10 mg·kg−1) attenuated by HU-910 given before the induction of I/R. 1 mg·kg−1 of HU-910 was sufficient to achieve similar or greater protection (as shown by decreases in I/R-induced elevated serum ALT/AST) during hepatic I/R injury, compared with the effects reported with HU-308 and JWH-133 at doses of 10 and 20 mg·kg−1 (Batkai et al., 2007Rajesh et al., 2007) respectively. The selected optimal dose (3 mg·kg−1) for mechanistic studies significantly decreased tissue oxidative stress (HNE and carbonyl adducts), ameliorated acute and chronic hepatic inflammatory responses (CCL3, CXCL2 and CCL2, TNF-α, ICAM-1/CD54 mRNA levels and tissue neutrophil infiltration), and both necrotic (ALT/AST levels and coagulation necrosis) and apoptotic (caspase 3/7 activity, DNA fragmentation) cell death at various relevant (2, 6 and/or 24 h following ischaemia) time points. Furthermore, HU-910 concentration-dependently attenuated theLPS-triggered TNF-α production in isolated Kupffer cells, and expression of adhesion molecules in primary HLSEC stimulated with TNF-α. These beneficial effects of HU-910 (3 mg·kg-1) were largely attenuated by the CB2receptor antagonist/inverse agonist SR144528 (3 mg·kg−1), which had no significant effect on I/R injury or the inflammatory response by itself; indicating that these effects were, at least in part, mediated by CB2 receptor activation. The fact that the effect of the highest dose of HU-910 used (10 mg·kg−1) was only partially attenuated by SR144528 could indicate some protective effects of the compound unrelated to CB2 receptor activation. However, the absence of the complete reversal can also be explained by the existence of non-specific anti-inflammatory effects of SR144528. This is also supported by aggravated hepatic I/R injury in CB2 knockout mice compared with their wild-type littermates, which cannot be mimicked by pretreatment of mice exposed to hepatic I/R with SR144528 at 3 mg·kg−1 i.p. However, this dose of SR144528 was able to attenuate the protective effect of CB2 agonist JWH-133 given at 20 mg·kg−1 i.p. (Batkai et al., 2007). Pretreatment with the CB1 receptor antagonist SR141716 (rimonabant) did not prevent the protective effect of HU-910 on liver necrosis; in fact, it reduced the serum ALT/AST levels by itself, an effect more pronounced when HU-910 was given in combination with SR141716. The protective effect of the CB1 receptor antagonist/inverse agonist on hepatic I/R injury described in our current report is in agreement with the protection observed in a model of rat liver I/R complicated with endotoxaemia (Caraceni et al., 2009) and in other models of I/R injury (Pacher and Hasko, 2008Zhang et al., 2009b), as well as with the proposition of combining CB1 antagonists with CB2 receptor agonists for the treatment of reperfusion injury (Pacher and Hasko, 2008Zhang et al., 2009b). Importantly, some of the protective effects of HU-910 also persisted when it was administered either at the beginning of the reperfusion or 1 h after the ischaemic episode (see Figures 4 and ​and101012 and Results).

Collectively, our results suggest that HU-910 is a novel CB2 receptor agonist which may exert protective effects in various diseases associated with inflammation and tissue injury. Its greater efficacy in vivo, despite similar pharmacological properties in vitro on CB2receptors to HU-308 or JWH-133, could be explained, at least in part, by its better penetration to injured tissues, or perhaps some additional beneficial properties unrelated to CB2 receptor activation, which together with its therapeutic window should be explored in the future studies. The beneficial effects of HU-910 against hepatic I/R injury are particularly exciting, because CB2 receptors are emerging as important targets in various liver (Lotersztajn et al., 2008Pacher and Gao, 2008) and other (Pacher and Mechoulam, 2011) diseases and their complications, and may also be crucial in liver regeneration (Teixeira-Clerc et al., 2010).

Acknowledgments

This study was supported by the Intramural Research Program of NIH/NIAAA (to P. P.) and by NIDA grant #9789 (to R. M.). Dr Béla Horváth is a recipient of a Hungarian Research Council Scientific Research Fund Fellowship (OTKA-NKTH-EU MB08-80238). The authors are indebted to Drs George Kunos and Bin Gao for providing key resources and support.

Glossary

4-HNE
4-hydroxy-2-nonenal (marker of lipid peroxidation)
CB2 or CB1 receptor
cannabinoid 1 or 2 receptor
HU-910
((1S,4R)-2-(2,6-dimethoxy-4-(2-methyloctan-2-yl)phenyl)-7,7-dimethylbicyclo[2.2.1]hept-2-en-1-yl)methanol
I/R
ischaemia/reperfusion
ICAM-1
inter-cellular adhesion molecule 1, CD54;CCL2, monocyte chemotactic protein-1
CCL3
macrophage inflammatory protein-1α
CXCL2
macrophage inflammatory protein-2α
VCAM-1
vascular cell adhesion molecule 1

Conflict of interest

Dr R. Mechoulam and L. Magid have submitted a patent on HU-910, other authors have no conflicts to disclose.

References

  • Abe Y, Hines IN, Zibari G, Pavlick K, Gray L, Kitagawa Y, et al. Mouse model of liver ischemia and reperfusion injury: method for studying reactive oxygen and nitrogen metabolites in vivo. Free Radic Biol Med. 2009;46:1–7. [PMC free article][PubMed]
  • Alexander SPH, Mathie A, Peters JA. Guide to Receptors and Channels (GRAC), 5th Edition. Br J Pharmacol. 2011;164(Suppl. 1):S1–S324. [PMC free article][PubMed]
  • Batkai S, Osei-Hyiaman D, Pan H, El-Assal O, Rajesh M, Mukhopadhyay P, et al. Cannabinoid-2 receptor mediates protection against hepatic ischemia/reperfusion injury. FASEB J. 2007;21:1788–1800. [PMC free article] [PubMed]
  • Caraceni P, Pertosa AM, Giannone F, Domenicali M, Grattagliano I, Principe A, et al. Antagonism of the cannabinoid CB-1 receptor protects rat liver against ischaemia-reperfusion injury complicated by endotoxaemia. Gut. 2009;58:1135–1143.[PubMed]
  • Di Marzo V. Targeting the endocannabinoid system: to enhance or reduce? Nat Rev Drug Discov. 2008;7:438–455. [PubMed]
  • Dominianni SJ, Ryan CW, DeArmitt CW. Synthesis of 5-(tert-alkyl)resorcinols. J Org Chem. 1977;42:344–346. [PubMed]
  • Engerson TD, McKelvey TG, Rhyne DB, Boggio EB, Snyder SJ, Jones HP. Conversion of xanthine dehydrogenase to oxidase in ischemic rat tissues. J Clin Invest.1987;79:1564–1570. [PMC free article] [PubMed]
  • Ferdinandy P, Schulz R. Nitric oxide, superoxide, and peroxynitrite in myocardial ischaemia-reperfusion injury and preconditioning. Br J Pharmacol.2003;138:532–543. [PMC free article] [PubMed]
  • Gero D, Szabo C. Role of the peroxynitrite-poly (ADP-ribose) polymerase pathway in the pathogenesis of liver injury. Curr Pharm Des. 2006;12:2903–2910. [PubMed]
  • Hall D, Poussin C, Velagapudi VR, Empsen C, Joffraud M, Beckmann J, et al. Peroxisomal and microsomal lipid pathways associated with resistance to hepatic steatosis and reduced pro-inflammatory state. J Biol Chem. 2010;285:31011–31023. [PMC free article] [PubMed]
  • Hanus L, Breuer A, Tchilibon S, Shiloah S, Goldenberg D, Horowitz M, et al. HU-308: a specific agonist for CB2, a peripheral cannabinoid receptor. Proc Natl Acad Sci U S A. 1999;96:14228–14233. [PMC free article] [PubMed]
  • Huffman JW. CB2 receptor ligands. Mini Rev Med Chem. 2005;5:641–649.[PubMed]
  • Jaeschke H. Role of reactive oxygen species in hepatic ischemia-reperfusion injury and preconditioning. J Invest Surg. 2003;16:127–140. [PubMed]
  • Jaeschke H. Mechanisms of Liver Injury. II. Mechanisms of neutrophil-induced liver cell injury during hepatic ischemia-reperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol. 2006;290:G1083–G1088.[PubMed]
  • Julien B, Grenard P, Teixeira-Clerc F, Van Nhieu JT, Li L, Karsak M, et al. Antifibrogenic role of the cannabinoid receptor CB2 in the liver. Gastroenterology.2005;128:742–755. [PubMed]
  • Liaudet L, Szabo G, Szabo C. Oxidative stress and regional ischemia-reperfusion injury: the peroxynitrite-poly(ADP-ribose) polymerase connection. Coron Artery Dis. 2003;14:115–122. [PubMed]
  • Lotersztajn S, Teixeira-Clerc F, Julien B, Deveaux V, Ichigotani Y, Manin S, et al. CB2receptors as new therapeutic targets for liver diseases. Br J Pharmacol.2008;153:286–289. [PMC free article] [PubMed]
  • Magid L, Goodfellow CE, Srebnik M, Glass M, Alexandrovich A, Hanuš L, et al. Design, synthesis and neuroprotrective potential of novel CB2 receptor selective agonists. 20th Annual Symposium on the Cannabinoids; International Cannabinoid Research Society, Research Triangle Park, NC, USA. 2010a. Abstracts, p 42.
  • Magid L, Mechoulam R, Shohami E, Bab I. Novel arylated camphenes, processes for their preparation and uses thereof. 2010b. Israeli Patent PCT/IL2010/000970.
  • Montecucco F, Lenglet S, Braunersreuther V, Burger F, Pelli G, Bertolotto M, et al. CB2 cannabinoid receptor activation is cardioprotective in a mouse model of ischemia/reperfusion. J Mol Cell Cardiol. 2009;46:612–620. [PubMed]
  • Moon KH, Hood BL, Mukhopadhyay P, Rajesh M, Abdelmegeed MA, Kwon YI, et al. Oxidative inactivation of key mitochondrial proteins leads to dysfunction and injury in hepatic ischemia reperfusion. Gastroenterology. 2008;135:1344–1357.[PMC free article] [PubMed]
  • Mukhopadhyay P, Rajesh M, Batkai S, Kashiwaya Y, Hasko G, Liaudet L, et al. Role of superoxide, nitric oxide, and peroxynitrite in doxorubicin-induced cell death in vivo and in vitro. Am J Physiol Heart Circ Physiol. 2009;296:H1466–H1483.[PMC free article] [PubMed]
  • Mukhopadhyay P, Rajesh M, Batkai S, Patel V, Kashiwaya Y, Liaudet L, et al. CB1cannabinoid receptors promote oxidative stress and cell death in murine models of doxorubicin-induced cardiomyopathy and in human cardiomyocytes. Cardiovasc Res. 2010;85:773–784. [PMC free article] [PubMed]
  • Mukhopadhyay P, Horvath B, Rajesh M, Matsumoto S, Saito K, Batkai S, et al. Fatty acid amide hydrolase is a key regulator of endocannabinoid-induced myocardial tissue injury. Free Radic Biol Med. 2011a;50:179–195. [PMC free article][PubMed]
  • Mukhopadhyay P, Rajesh M, Horvath B, Batkai S, Park O, Tanchian G, et al. Cannabidiol protects against hepatic ischemia/reperfusion injury by attenuating inflammatory signaling and response, oxidative/nitrative stress, and cell death.Free Radic Biol Med. 2011b;50:1368–1381. [PMC free article] [PubMed]
  • Murikinati S, Jüttler E, Keinert T, Ridder DA, Muhammad S, Waibler Z, et al. Activation of cannabinoid 2 receptors protects against cerebral ischemia by inhibiting neutrophil recruitment. FASEB J. 2010;24:788–798. [PubMed]
  • Pacher P, Gao B. Endocannabinoids and liver disease. III. Endocannabinoid effects on immune cells: implications for inflammatory liver diseases. Am J Physiol Gastrointest Liver Physiol. 2008;294:G850–G854. [PMC free article] [PubMed]
  • Pacher P, Hasko G. Endocannabinoids and cannabinoid receptors in ischaemia-reperfusion injury and preconditioning. Br J Pharmacol. 2008;153:252–262.[PMC free article] [PubMed]
  • Pacher P, Mechoulam R. Is lipid signaling through cannabinoid 2 receptors part of a protective system? Prog Lipid Res. 2011;50:193–211. [PMC free article] [PubMed]
  • Pacher P, Batkai S, Kunos G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev. 2006;58:389–462. [PMC free article][PubMed]
  • Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007;87:315–424. [PMC free article] [PubMed]
  • Pertwee RG. Pharmacological actions of cannabinoids. Handb Exp Pharmacol.2005;168:1–51. [PubMed]
  • Rajesh M, Pan H, Mukhopadhyay P, Batkai S, Osei-Hyiaman D, Hasko G, et al. Cannabinoid-2 receptor agonist HU-308 protects against hepatic ischemia/reperfusion injury by attenuating oxidative stress, inflammatory response, and apoptosis. J Leukoc Biol. 2007;82:1382–1389. [PMC free article][PubMed]
  • Rajesh M, Mukhopadhyay P, Batkai S, Mukhopadhyay B, Patel V, Hasko G, et al. Xanthine oxidase inhibitor allopurinol attenuates the development of diabetic cardiomyopathy. J Cell Mol Med. 2009;13:2330–2341. [PMC free article][PubMed]
  • Rajesh M, Mukhopadhyay P, Bátkai S, Patel V, Saito K, Matsumoto S, et al. Cannabidiol attenuates cardiac dysfunction, oxidative stress, fibrosis, and inflammatory and cell death signaling pathways in diabetic cardiomyopathy. J Am Coll Cardiol. 2010;56:2115–2125. [PMC free article] [PubMed]
  • Teixeira-Clerc F, Belot MP, Manin S, Deveaux V, Cadoudal T, Chobert MN, et al. Beneficial paracrine effects of cannabinoid receptor 2 on liver injury and regeneration. Hepatology. 2010;52:1046–1059. [PMC free article] [PubMed]
  • Zhang M, Martin BR, Adler MW, Razdan RK, Jallo JI, Tuma RF. Cannabinoid CB2receptor activation decreases cerebral infarction in a mouse focal ischemia/reperfusion model. J Cereb Blood Flow Metab. 2007;27:1387–1396.[PMC free article] [PubMed]
  • Zhang M, Adler MW, Abood ME, Ganea D, Jallo J, Tuma RF. CB2 receptor activation attenuates microcirculatory dysfunction during cerebral ischemic/reperfusion injury. Microvasc Res. 2009a;78:86–94. [PMC free article] [PubMed]
  • Zhang M, Martin BR, Adler MW, Razdan RJ, Kong W, Ganea D, et al. Modulation of cannabinoid receptor activation as a neuroprotective strategy for EAE and stroke.J Neuroimmune Pharmacol. 2009b;4:249–259. [PMC free article] [PubMed]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society
potp font 1