Excitotoxicity, peri-infarct depolarization, oxidative stress, apoptosis, and inflammation contribute to the development of cerebral injury after ischemia.
1- Dirnagl U.
- Iadecola C.
- Moskowitz M.A.
Pathobiology of ischaemic stroke: an integrated view.
To develop effective therapeutic strategies for postischemic brain injury, it is crucial to understand the highly diverse temporal profiles (eg, onset and duration) of these individual factors and to interrupt their pathophysiological cascades. The
N-methyl-
d-aspartate (NMDA) receptor blocker MK-801 markedly reduces ischemic brain injury; however, MK-801 has a brief therapeutic time window: the neuroprotective effect of MK-801 is obtained only when therapy is given within at most 1 hour after the onset of focal ischemia in rats and gerbils.
2- Margaill I.
- Parmentier S.
- Callebert J.
- Allix M.
- Boulu R.G.
- Plotkine M.
Short therapeutic window for MK-801 in transient focal cerebral ischemia in normotensive rats.
, 3- Hatfield R.H.
- Gill R.
- Brazell C.
The dose-response relationship and therapeutic window for dizocilpine (MK-801) in a rat focal ischaemia model.
Furthermore, MK-801 only postpones postischemic neuronal death; it does not improve either neurological recovery or endpoint cell survival at weeks after treatment.
4- Valtysson J.
- Hillered L.
- Andiné P.
- Hagberg H.
- Persson L.
Neuropathological endpoints in experimental stroke pharmacotherapy: the importance of both early and late evaluation.
, 5- Von Lubitz D.K.
- McKenzie R.J.
- Lin R.C.
- Devlin T.M.
- Skolnick P.
MK-801 is neuroprotective but does not improve survival in severe forebrain ischemia.
Similarly, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor antagonists also did not significantly protect neuronal loss at 28 days after middle cerebral artery occlusion (MCAO).
6- Colbourne F.
- Li H.
- Buchan A.M.
- Clemens J.A.
Continuing postischemic neuronal death in CA1: influence of ischemia duration and cytoprotective doses of NBQX and SNX-111 in rats.
This short therapeutic window and lack of long-term effect of NMDA or AMPA receptor antagonists suggests that these receptors play only a transient role in the early ischemic cascade. Thus, some pathophysiological processes that are not affected by one of these treatments are thought to contribute to the delayed cerebral ischemic damage. In this context, much attention has been paid to inflammation, which starts after excitotoxicity and lasts for up to several days and even weeks, as an attractive therapeutic target to prevent the evolution of massive tissue damage in ischemia.
7- Price C.J.
- Warburton E.A.
- Menon D.K.
Human cellular inflammation in the pathology of acute cerebral ischaemia.
The A3 adenosine receptor, one of the four adenosine receptor subtypes (A1, A2A, A2B, and A3), is widely distributed in human and rat brain.
8- Dixon A.K.
- Gubitz A.K.
- Sirinathsinghji D.J.
- Richardson P.J.
- Freeman T.C.
Tissue distribution of adenosine receptor mRNAs in the rat.
, 9- Atkinson M.R.
- Townsend-Nicholson A.
- Nicholl J.K.
- Sutherland G.R.
- Schofield P.R.
Cloning, characterisation and chromosomal assignment of the human adenosine A3 receptor (ADORA3) gene.
Expression of A3AR is reported to increase in activated inflammatory cells and modulate their activities
10- Bar-Yehuda S.
- Silverman M.H.
- Kerns W.D.
- Ochaion A.
- Cohen S.
- Fishman P.
The anti-inflammatory effect of A3 adenosine receptor agonists: a novel targeted therapy for rheumatoid arthritis.
, 11- Ochaion A.
- Bar-Yehuda S.
- Cohen S.
- Barer F.
- Patoka R.
- Amital H.
- Reitblat T.
- Reitblat A.
- Ophir J.
- Konfino I.
- Chowers Y.
- Ben-Horin S.
- Fishman P.
The anti-inflammatory target A(3) adenosine receptor is over-expressed in rheumatoid arthritis, psoriasis and Crohn's disease.
Moreover, the A3AR agonists are known to have anti-inflammatory effects in various animal models.
12- Haskó G.
- Németh Z.H.
- Vizi E.S.
- Salzman A.L.
- Szabó C.
An agonist of adenosine A3 receptors decreases interleukin-12 and interferon-gamma production and prevents lethality in endotoxemic mice.
, 13- Wagner R.
- Ngamsri K.C.
- Stark S.
- Vollmer I.
- Reutershan J.
Adenosine receptor A3 is a critical mediator in LPS-induced pulmonary inflammation.
, 14- Ge Z.D.
- van der Hoeven D.
- Maas J.E.
- Wan T.C.
- Auchampach J.A.
A(3) adenosine receptor activation during reperfusion reduces infarct size through actions on bone marrow-derived cells.
Recently, several studies using A3AR knockout mice support the possibility of A3AR as a new target to relieve cerebral ischemic injury.
15- Chen G.J.
- Harvey B.K.
- Shen H.
- Chou J.
- Victor A.
- Wang Y.
Activation of adenosine A3 receptors reduces ischemic brain injury in rodents.
, 16- Fedorova I.M.
- Jacobson M.A.
- Basile A.
- Jacobson K.A.
Behavioral characterization of mice lacking the A3 adenosine receptor: sensitivity to hypoxic neurodegeneration.
Furthermore, although their effects are variable, depending on experimental conditions (eg, dosage, time window, administration route, location or severity of ischemic injury, and experimental models), administration of A3AR agonists before or immediately after ischemic insults was shown to significantly protect brain tissues in rodent ischemic models.
15- Chen G.J.
- Harvey B.K.
- Shen H.
- Chou J.
- Victor A.
- Wang Y.
Activation of adenosine A3 receptors reduces ischemic brain injury in rodents.
, 17- Von Lubitz D.K.
- Lin R.C.
- Popik P.
- Carter M.F.
- Jacobson K.A.
Adenosine A3 receptor stimulation and cerebral ischemia.
, 18- Von Lubitz D.K.
- Simpson K.L.
- Lin R.C.
Right thing at a wrong time? Adenosine A3 receptors and cerebroprotection in stroke.
The underlying mechanism, however, remains largely unknown.
In the present study, therefore, we investigated the anti-ischemic mechanism of the selective A3AR agonist LJ529 [2-chloro-
N6-(3-iodobenzyl)-5′-
N-methylcarbamoyl-4′-thioadenosine], a 4′-thio analog of the well-known Cl-IB-MECA. Cl-IB-MECA displays inhibition constant
Ki values of 820, 470, and 0.33 nmol/L for rA1AR, rA2AAR, and rA3AR, respectively.
19- Kim H.O.
- Ji X.D.
- Siddiqi S.M.
- Olah M.E.
- Stiles G.L.
- Jacobson K.A.
2-Substitution of N6-benzyladenosine-5′-uronamides enhances selectivity for A3 adenosine receptors.
For human adenosine receptors, the
Ki values of Cl-IB-MECA are 220, 5400, and 1.4 nmol/L for hA1AR, hA2AAR, and hA3AR, respectively.
20- Tchilibon S.
- Joshi B.V.
- Kim S.K.
- Duong H.T.
- Gao Z.G.
- Jacobson K.A.
(N)-methanocarba 2,N6-disubstituted adenine nucleosides as highly potent and selective A3 adenosine receptor agonists.
We previously showed that LJ529 possesses higher potency and better selectivity to human A3AR than Cl-IB-MECA does: the
Ki values of LJ529 are 0.38 nmol/L at hA3AR, but only 193 and 223 nmol/L at hA1AR and hA2AAR, respectively.
21- Jeong L.S.
- Jin D.Z.
- Kim H.O.
- Shin D.H.
- Moon H.R.
- Gunaga P.
- Chun M.W.
- Kim Y.C.
- Melman N.
- Gao Z.G.
- Jacobson K.A.
N6-substituted D-4′-thioadenosine-5′-methyluronamides: potent and selective agonists at the human A3 adenosine receptor.
In the present study, we found that intraperitoneal injection of LJ529 at 2 and 7 hours after ischemia (ie, 0.5 and 5.5 hours after starting reperfusion) markedly reduced cerebral ischemic injury, and that this anti-ischemic effect might be related to reduced migration of inflammatory cells into the ischemic lesion via suppression of polarized expression of Rho GTPases.
Materials and Methods
Animals
Sprague-Dawley male rats (260 to 270 g; Charles River Laboratories International, Seoul, Korea) were acclimated to their environment for 5 days before use. All experimental procedures using animals were in accordance with the NIH Guide (7th Edition) for the Care and Use of Laboratory Animals and were approved by the Committee of Korea University College of Medicine.
Focal Cerebral Ischemia Model
Rats were anesthetized with 3.0% isoflurane in N
2O and O
2 (70:30 v/v) mixture via facemask and were maintained in 2% isoflurane. Focal cerebral ischemia was achieved by right-sided endovascular MCAO, as described previously.
22- Belayev L.
- Alonso O.F.
- Busto R.
- Zhao W.
- Ginsberg M.D.
Middle cerebral artery occlusion in the rat by intraluminal suture Neurological and pathological evaluation of an improved model.
Briefly, a 3-0 heat-blunted monofilament nylon suture (Ethicon Johnson & Johnson, Brussels, Belgium) were inserted into the lumen of the right external carotid artery stump and advanced 17.5 mm into the internal carotid artery to occlude the ostium of the MCA. The suture was removed after 1.5 hours to allow animals to recover. Sham-operated controls were subjected to the same surgical procedures, except for MCAO. Throughout experiments, the body temperature was monitored with a rectal thermometer and maintained at 37°C ± 0.3°C with heating pads. Physiological values were measured 15 minutes before MCAO and at 15 minutes after reperfusion. Mean arterial blood pressure was monitored for 5 minutes using a DigiMed blood pressure analyzer (Micro-Med, Louisville, KY). Blood pH, PaO
2, PaCO
2, and glucose were monitored using an automatic pH/blood gas analyzer (Ciba Corning Diagnostics, Medfield, MA). LJ529 was intraperitoneally administrated to rats twice (1 or 2 mg/kg for each) at 2 and 7 hours after starting MCAO (ie, 0.5 and 5.5 hours after reperfusion). If necessary, either the A3AR antagonist MRS1523 (
Ki = 113 nmol/L for rA3AR
23- Li A.H.
- Moro S.
- Melman N.
- Ji X.D.
- Jacobson K.A.
Structure-activity relationships and molecular modeling of 3,5-diacyl-2,4-dialkylpyridine derivatives as selective A3 adenosine receptor antagonists.
; 1 mg/kg, i.p.) or the A2AAR antagonist SCH58261 (
Ki = 1.3 nmol/L for rA2AAR
24- Zocchi C.
- Ongini E.
- Conti A.
- Monopoli A.
- Negretti A.
- Baraldi P.G.
- Dionisotti S.
The non-xanthine heterocyclic compound SCH 58261 is a new potent and selective A2a adenosine receptor antagonist.
; 70 μg/kg, i.p.) was simultaneously administrated with LJ529. The dose of each antagonist used in the present study was determined based on the
in vivo effect observed in previous studies
25- Ge Z.D.
- Peart J.N.
- Kreckler L.M.
- Wan T.C.
- Jacobson M.A.
- Gross G.J.
- Auchampach J.A.
Cl-IB-MECA [2-chloro-N6-(3-iodobenzyl)adenosine-5′-N-methylcarboxamide] reduces ischemia/reperfusion injury in mice by activating the A3 adenosine receptor.
, 26- Melani A.
- Pantoni L.
- Bordoni F.
- Gianfriddo M.
- Bianchi L.
- Vannucchi M.G.
- Bertorelli R.
- Monopoli A.
- Pedata F.
The selective A2A receptor antagonist SCH 58261 reduces striatal transmitter outflow, turning behavior and ischemic brain damage induced by permanent focal ischemia in the rat.
, 27- Popoli P.
- Reggio R.
- Pezzola A.
Effects of SCH 58261, an adenosine A(2A) receptor antagonist, on quinpirole-induced turning in 6-hydroxydopamine-lesioned rats Lack of tolerance after chronic caffeine intake.
and in our preliminary experiments. At doses used in the present study, antagonists alone did not show any effect on infarction size. All
in vivo experiments and the subsequent data analysis were performed in a double-blind and randomized manner.
Microinjection of LPS into Corpus Callosum
After rats were anesthetized with chloral hydrate (300 mg/kg), LPS (Escherichia coli serotype 055:B5; Sigma-Aldrich, St. Louis, MO; 5 μg/5 μL) or vehicle (saline) was microinjected into the corpus callosum at a rate of 0.5 μL/min using a microinjection pump (PDH 2000; Harvard Apparatus, Holliston, MA), with the following coordinates: 0.1 mm posterior from bregma, 1.8 mm lateral from the sagittal suture, and 3.2 mm below the dura mater.
Measurement of Infarct Volume
Rats were anesthetized with chloral hydrate and decapitated at 1 day after MCAO. Coronal sections of brain (2 mm) were stained with 2% triphenyltetrazolium chloride (Sigma-Aldrich) at 37°C for 30 minutes, fixed with 4% paraformaldehyde (pH 7.4) in 0.1 mol/L phosphate buffer for 1 day, and subsequently cryoprotected in phosphate buffer containing 30% sucrose at 4°C for 2 days. The cross-sectional area of infarction between the bregma levels of +4 mm (anterior) and −6 mm (posterior) were determined with Optimas version 5.1 image analysis software (Bioscan, Edmonds, WA). Brain infarct size was measured manually by outlining the margins of infarct areas. The total infarction volume was integrated from six chosen sections, expressed as a percentage of the total brain volume, and compensated for brain edema, as described previously.
28Quantification of infarct size on focal cerebral ischemia model of rats using a simple and economical method.
, 29- Swanson R.A.
- Morton M.T.
- Tsao-Wu G.
- Savalos R.A.
- Davidson C.
- Sharp F.R.
A semiautomated method for measuring brain infarct volume.
Cerebral edema was determined in a double-blind manner by the percent increase of the ipsilateral/contralateral hemisphere area: % edema volume = [(ipsilateral volume − contralateral volume)/contralateral volume] × 100. Thereafter, the tissues were frozen, cut into 10- or 30-μm coronal sections on a Leica 3050 cryostat (Leica, Nussloch, Germany) and stored at −20°C.
Immunohistochemistry
The brain sections were quenched with 0.3% hydroperoxide, blocked with 10% normal horse serum, and then stained with mouse anti-ED1 antibody overnight at room temperature (diluted 1:200; Serotec, Oxford, UK). After further staining with biotinylated anti-mouse IgG (Vector Laboratories, Burlingame, CA) and peroxidase-conjugated streptavidin (diluted 1:200; Vector Laboratories), the antigens were visualized with 5 minutes of incubation at 37°C in 0.1 mol/L phosphate buffer containing 0.02% 3,3-diaminobenzidine and 0.0045% hydrogen peroxide (ABC method). The number of ED1-positive cells was manually counted in 10 grids (0.1 mm2/grid) using a DMC2 digital microscope camera (Polaroid, Minnetonka, MN), and averaged from three to five adjacent sections.
Microglial Cell Culture
Pure microglial cells were prepared from primary mixed glial cell culture. Cerebral cortices from neonatal Sprague-Dawley rats (1 to 2 days old) were triturated to single cells. They were then plated into poly-d-lysine (1 μg/mL; Sigma-Aldrich) coated 75-cm2 T-flasks and maintained in modified Eagle's medium (MEM) containing 10% fetal bovine serum. At 7or 8 days after plating, microglia were detached from the flasks by mild shaking (37°C, 2 minutes at 200/min) and plated onto the experimental plates. After 6 hours, microglia were replaced with serum-free MEM and were used for experiments after an overnight.
Enzyme-Linked Immunosorbent Assay
The concentrations of IL-1β, TNF-α, and MCP-1 were measured by enzyme-linked immunosorbent assay using monoclonal antibodies according to the manufacturer's procedures (KOMA Biotech, Seoul, Korea).
Chemotaxis
Microphotographs of migrating microglial cells in the live cell chamber (Live Cell Instrument, Seoul, Korea) were monitored with inverted confocal microscopy system (Leica DMIRE2, TLC_SP2) and differential interference contrast transmission image programs (Leica, Ver. 2.5). Fifteen minutes after LJ529 was applied by micromanipulator (Narishige, Japan), the MCP-1 gradient (100 ng/mL; BD Pharmingen, San Jose, CA) was generated using a Harvard Apparatus microinjection device (0.1 μL/min; PHD 2000 programmable). MRS1523 (1 μmol/L) or SCH58261 (100 nmol/L) was applied to the cells for 20 minutes before treatment with MCP-1. For quantitative analysis, MCP-1 was placed in the bottom chamber of a chemotaxis chamber (Neuro Probe, Cabin John, MD) and microglia (5 × 10
4 cells/mL in serum-free MEM) in the upper chamber were allowed to migrate to the bottom part for 2 hours, through membrane pore (8 mm
2 filter area; Neuro Probe). Migrated cells on the bottom-side filter were stained for nuclei with Harris's hematoxylin and then counted. The movement or morphological change of microglia was recorded in video for 20 minutes starting from the application of MCP-1 (see
Supplemental Videos S1–S4 at
http://ajp.amjpathol.org).
Immunocytochemistry
After stimulation with gradient MCP-1 for 5 minutes, microglia were fixed and stained with primary antibodies against Rac, Cdc42, and Rho (diluted 1:500, 1:250 and 1:250, respectively; Millipore-Upstate Biotechnology, Lake Placid, NY) and Alexa Fluor 488-phalloidin (diluted 1:2000; Molecular Probes, Carlsbad, CA) for filamentous actin.
Activation of Rho GTPase
Active forms of GTPase (GTP-bound forms) were measured by pull-down kits for Rho GTPases (Millipore-Upstate). Briefly, microglia were stimulated with MCP-1 (100 ng/mL), then lysed with kit-supplied buffer, and centrifuged at 14,000 × g for 5 minutes. Equal amounts of the supernatant fractions obtained were incubated with either PAK-1 PBD-agarose (which binds Rac-GTP or Cdc42-GTP) or rhotekin RBD-agarose (which binds Rho-GTP) for 1 hour at 4°C, followed by washing three times. Proteins bound to the beads were eluted in Laemmli sample buffer and subjected to Western blot analysis using mouse monoclonal antibodies specific to Rac, Cdc42, and Rho, respectively.
Cortical Neuronal Culture
Cortical neurons (5 × 105 cells/mL) were prepared from fetal rats (17 to 18 embryonic days old) and plated on poly-d-lysine (100 μg/mL)/laminin (4 μg/mL)-precoated plates using 10% fetal bovine serum-DMEM with 2 mmol/L glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin. At 3 days after culture, 5 μmol/L cytosine arabinoside was applied to block non-neuronal cell division. The medium was then replaced twice a week. Experiments were performed at 14 to 16 days in vitro.
Oxygen-Glucose Deprivation (OGD) and Reoxygenation and Excitotoxicity
For in vitro hypoxic-ischemic insult, cultured neurons were placed in an anaerobic chamber (partial pressure of oxygen <2 mmHg) at 37°C for 1 hour. OGD was discontinued by replacing with oxygenated DMEM containing 25 mmol/L glucose and returning the cells to normoxic conditions. Control cells, not exposed to OGD, were maintained in DMEM containing glucose (25 mmol/L), aerated with an aerobic gas mix (95% air, 5% CO2). LJ529 was applied immediately after reoxygenation. To evoke excitotoxicity, neurons were treated with 100 μmol/L NMDA for 10 minutes in the presence or absence of LJ529.
Assessment of Cell Injury or Death
Cell injury or death was assessed by the amount of lactate dehydrogenase released into the bathing medium using a diagnostic kit (Sigma-Aldrich) and expressed as a percentage of total lactate dehydrogenase, which was measured in sister cultures frozen and thawed after the experiments.
30- Ju C.
- Oh Y.J.
- Han B.H.
- Kim H.S.
- Kim H.C.
- Kim W.K.
Intracellular pH-dependent peroxynitrite-evoked synergistic death of glucose-deprived astrocytes.
Measurement of Plasma Membrane Potential
Plasma membrane potential was monitored by using bisoxonol fluorescent dye [(bis-[1,3-diethyl-thio-barbiturate]-trimethineoxonol) (DiBAC
4(3)] (excitation at 540 nm; emission at 565 nm); the increase in intensity indicates the membrane depolarization.
31Dye indicators of membrane potential.
Neurons were preincubated with 5 μmol/L DiBAC
4(3) for 10 minutes before LJ529 treatment. Immediately after cells were exposed to 100 μmol/L NMDA, time-lapse fluorescence intensity (excitation at 540 nm; emission at 565 nm) was measured with a fluorescence plate reader (SpectraMAX GeminiEM; Molecular Devices, Sunnyvale, CA) and compensated with autofluorescence [ie, fluorescence in non-loaded cells with DiBAC
4(3)].
Measurement of Antioxidant Activities
For direct scavenging activity of LJ529, peroxynitrite donor 3-morpholinosydnonimine (SIN-1, 200 μmol/L) or H
2O
2 (1 mmol/L) was added to Earle's balanced salt solution buffer containing 10 μmol/L dihydrorhodamine 123 in the absence and presence of LJ529. After 10 minutes at room temperature, rhodamine 123 fluorescence (excitation at 490 nm; emission at 530 nm) was measured with a fluorescence microplate reader (SpectraMAX GeminiEM; Molecular Devices). To measure oxidant levels, 30 μmol/L dichlorodihydro-fluorescein diacetate (H
2DCF-DA) was applied for 10 minutes in Earle's balanced salt solution buffer containing 0.1% bovine serum albumin and 2.5 mmol/L probenecid after LPS treatment for 18 hours. After washing out H
2DCF-DA supernatant, fluorescence was measured (excitation at 488 nm; emission at 525 nm). Catalase and superoxide dismutase were added 30 minutes before measurement to remove hydrogen peroxide or superoxide, respectively. For the measurement of nitric oxide (NO) production, nitrite levels in the supernatants were determined by the Griess test, as described previously.
32- Schmidt H.H.
- Warner T.D.
- Nakane M.
- Förstermann U.
- Murad F.
Regulation and subcellular location of nitrogen oxide synthases in RAW264.7 macrophages.
Statistical Analysis
Data were analyzed for statistical significance by one-way analysis of variance followed by Scheffé's test for multiple comparisons. Data are reported as means ± SEM (for in vivo infarction and edema volume) or SD.
Discussion
The recent recognition that cerebral stroke and other ischemic injury are manifestations of chronic progressive inflammation has had great influence on the development of therapeutic and preventative strategies.
7- Price C.J.
- Warburton E.A.
- Menon D.K.
Human cellular inflammation in the pathology of acute cerebral ischaemia.
, 47- Amantea D.
- Nappi G.
- Bernardi G.
- Bagetta G.
- Corasaniti M.T.
Post-ischemic brain damage: pathophysiology and role of inflammatory mediators.
In most previous studies, A3AR agonists were administered before or immediately after stroke.
15- Chen G.J.
- Harvey B.K.
- Shen H.
- Chou J.
- Victor A.
- Wang Y.
Activation of adenosine A3 receptors reduces ischemic brain injury in rodents.
, 17- Von Lubitz D.K.
- Lin R.C.
- Popik P.
- Carter M.F.
- Jacobson K.A.
Adenosine A3 receptor stimulation and cerebral ischemia.
, 18- Von Lubitz D.K.
- Simpson K.L.
- Lin R.C.
Right thing at a wrong time? Adenosine A3 receptors and cerebroprotection in stroke.
The present study is the first to demonstrate anti-ischemic protection achieved by postischemic treatment (ie, at 2 and 7 hours after the onset of ischemia) of the A3AR agonist LJ529. The A3AR antagonist abolished this anti-ischemic effect of LJ529 (
Figure 1). Excitotoxicity and oxidative stress are major factors causing neuronal cell death in the early stage of cerebral ischemic insults
1- Dirnagl U.
- Iadecola C.
- Moskowitz M.A.
Pathobiology of ischaemic stroke: an integrated view.
, 48The complexity of neurobiological processes in acute ischemic stroke.
However, the anti-ischemic effect of LJ529 appears not to be related with blockade of excitotoxicity and oxidative stress (
Figure 5,
Figure 6). Rather, LJ529 reduces cerebral ischemic injury through activation of the A3AR, potentially via suppression of postischemic inflammatory responses such as infiltration/migration of inflammatory cells.
Neuroinflammatory mechanisms are activated after an ischemic insult by excitotoxicity and oxidative stress and play a pivotal role in prolonged brain damage in ischemia.
1- Dirnagl U.
- Iadecola C.
- Moskowitz M.A.
Pathobiology of ischaemic stroke: an integrated view.
, 7- Price C.J.
- Warburton E.A.
- Menon D.K.
Human cellular inflammation in the pathology of acute cerebral ischaemia.
, 45- Jordan J.
- Segura T.
- Brea D.
- Galindo M.F.
- Castillo J.
Inflammation as therapeutic objective in stroke.
A rapid activation of resident microglial cells and infiltration of peripheral inflammatory monocytes has been shown to modulate the extent of ischemic brain damage.
1- Dirnagl U.
- Iadecola C.
- Moskowitz M.A.
Pathobiology of ischaemic stroke: an integrated view.
, 45- Jordan J.
- Segura T.
- Brea D.
- Galindo M.F.
- Castillo J.
Inflammation as therapeutic objective in stroke.
We also found that LPS-activated microglia and monocytes accelerated and aggravated cerebral ischemic injury,
34- Lee J.C.
- Cho G.S.
- Kim H.J.
- Lim J.H.
- Oh Y.K.
- Nam W.
- Chung J.H.
- Kim W.K.
Accelerated cerebral ischemic injury by activated macrophages/microglia after lipopolysaccharide microinjection into rat corpus callosum.
and that depletion of peripheral leukocytes by γ-irradiation before ischemia reduced the cerebral infarction caused by MCAO/R (unpublished data). In the present study, LJ529 was found to inhibit the infiltration/migration of microglia/monocytes induced by MCAO/R or injection of LPS into the corpus callosum (
Figure 1). It is quite reasonable, therefore, to confirmed that LJ529 reduces postischemic cerebral injury via suppression of infiltration/migration of inflammatory cells and their immune responses.
Many earlier studies have demonstrated that activation of A3AR reduces the production/release of proinflammatory cytokines in activated inflammatory cells,
12- Haskó G.
- Németh Z.H.
- Vizi E.S.
- Salzman A.L.
- Szabó C.
An agonist of adenosine A3 receptors decreases interleukin-12 and interferon-gamma production and prevents lethality in endotoxemic mice.
, 36- Lee J.Y.
- Jhun B.S.
- Oh Y.T.
- Lee J.H.
- Choe W.
- Baik H.H.
- Ha J.
- Yoon K.S.
- Kim S.S.
- Kang I.
Activation of adenosine A3 receptor suppresses lipopolysaccharide-induced TNF-alpha production through inhibition of PI 3-kinase/Akt and NF-kappaB activation in murine BV2 microglial cells.
, 38- Bowlin T.L.
- Borcherding D.R.
- Edwards 3rd, C.K.
- McWhinney C.D.
Adenosine A3 receptor agonists inhibit murine macrophage tumor necrosis factor-alpha production in vitro and in vivo.
, 49- Martin L.
- Pingle S.C.
- Hallam D.M.
- Rybak L.P.
- Ramkumar V.
Activation of the adenosine A3 receptor in RAW 264.7 cells inhibits lipopolysaccharide-stimulated tumor necrosis factor-alpha release by reducing calcium-dependent activation of nuclear factor-kappaB and extracellular signal-regulated kinase 1/2.
, 50- Szabó C.
- Scott G.S.
- Virág L.
- Egnaczyk G.
- Salzman A.L.
- Shanley T.P.
- Haskó G.
Suppression of macrophage inflammatory protein (MIP)-1alpha production and collagen-induced arthritis by adenosine receptor agonists.
which were widely accepted as a key detrimental factor in cerebral ischemic injury.
47- Amantea D.
- Nappi G.
- Bernardi G.
- Bagetta G.
- Corasaniti M.T.
Post-ischemic brain damage: pathophysiology and role of inflammatory mediators.
The A3AR agonist Cl-IB-MECA inhibits LPS-induced TNF-α production
in vivo, decreasing its effect in A3AR knockout mice.
51- Salvatore C.A.
- Tilley S.L.
- Latour A.M.
- Fletcher D.S.
- Koller B.H.
- Jacobson M.A.
Disruption of the A(3) adenosine receptor gene in mice and its effect on stimulated inflammatory cells.
Another A3AR agonist, IB-MECA, has been shown to suppress the production of IL-12 and IFN-γ, and to reduce lethality in endotoxemic mice.
12- Haskó G.
- Németh Z.H.
- Vizi E.S.
- Salzman A.L.
- Szabó C.
An agonist of adenosine A3 receptors decreases interleukin-12 and interferon-gamma production and prevents lethality in endotoxemic mice.
IB-MECA also suppresses LPS-induced production of either TNF-α
49- Martin L.
- Pingle S.C.
- Hallam D.M.
- Rybak L.P.
- Ramkumar V.
Activation of the adenosine A3 receptor in RAW 264.7 cells inhibits lipopolysaccharide-stimulated tumor necrosis factor-alpha release by reducing calcium-dependent activation of nuclear factor-kappaB and extracellular signal-regulated kinase 1/2.
or the chemokine macrophage inflammatory protein (MIP)-1α
12- Haskó G.
- Németh Z.H.
- Vizi E.S.
- Salzman A.L.
- Szabó C.
An agonist of adenosine A3 receptors decreases interleukin-12 and interferon-gamma production and prevents lethality in endotoxemic mice.
in macrophageal RAW 264.7 cells. Similarly, activation of adenosine A3 receptor suppresses LPS-induced TNF-α production in murine BV2 microglial cells.
36- Lee J.Y.
- Jhun B.S.
- Oh Y.T.
- Lee J.H.
- Choe W.
- Baik H.H.
- Ha J.
- Yoon K.S.
- Kim S.S.
- Kang I.
Activation of adenosine A3 receptor suppresses lipopolysaccharide-induced TNF-alpha production through inhibition of PI 3-kinase/Akt and NF-kappaB activation in murine BV2 microglial cells.
Consistent with previous studies, we have demonstrated that LJ529 significantly reduced the release of IL-1β, TNF-α, and MCP-1 in LPS-treated microglial cells (
Figure 2). Notably, however, the IC
50 values of LJ529 for the inhibition of cytokine/chemokine release were ∼10 μmol/L. Compared with the nanomolar level of
Ki values for A3AR,
19- Kim H.O.
- Ji X.D.
- Siddiqi S.M.
- Olah M.E.
- Stiles G.L.
- Jacobson K.A.
2-Substitution of N6-benzyladenosine-5′-uronamides enhances selectivity for A3 adenosine receptors.
, 20- Tchilibon S.
- Joshi B.V.
- Kim S.K.
- Duong H.T.
- Gao Z.G.
- Jacobson K.A.
(N)-methanocarba 2,N6-disubstituted adenine nucleosides as highly potent and selective A3 adenosine receptor agonists.
, 21- Jeong L.S.
- Jin D.Z.
- Kim H.O.
- Shin D.H.
- Moon H.R.
- Gunaga P.
- Chun M.W.
- Kim Y.C.
- Melman N.
- Gao Z.G.
- Jacobson K.A.
N6-substituted D-4′-thioadenosine-5′-methyluronamides: potent and selective agonists at the human A3 adenosine receptor.
these relatively high IC
50 values of LJ529 and also of Cl-IB-MECA in the present study suggest that A3AR may not play a major role in LPS-induced cytokine release in microglia. Because the potency and efficacy of A3AR agonists depend on the quantities of receptors and second messenger profiles present in a certain cell type,
52Adenosine A3 receptors: novel ligands and paradoxical effects.
it is also possible that the IC
50 values of A3AR agonists are cell type-dependent (eg, at micromolar levels particularly in microglia), as shown in the present study and by others.
36- Lee J.Y.
- Jhun B.S.
- Oh Y.T.
- Lee J.H.
- Choe W.
- Baik H.H.
- Ha J.
- Yoon K.S.
- Kim S.S.
- Kang I.
Activation of adenosine A3 receptor suppresses lipopolysaccharide-induced TNF-alpha production through inhibition of PI 3-kinase/Akt and NF-kappaB activation in murine BV2 microglial cells.
Also, the effect of adenosine receptor agonists on cytokine release has been shown to be subject to the specific Toll-like receptor used for immune stimulation in a cell-type-dependent manner,
53- Ramakers B.P.
- Riksen N.P.
- Rongen G.A.
- van der Hoeven J.G.
- Smits P.
- Pickkers P.
The effect of adenosine receptor agonists on cytokine release by human mononuclear cells depends on the specific Toll-like receptor subtype used for stimulation.
and Toll-like receptor activation by LPS induces an up-regulation of A2AAR and a down-regulation of A3AR in primary rhesus monkey microglia.
54- van der Putten C.
- Zuiderwijk-Sick E.A.
- van Straalen L.
- de Geus E.D.
- Boven L.A.
- Kondova I.
- IJzerman A.P.
- Bajramovic J.J.
Differential expression of adenosine A3 receptors controls adenosine A2A receptor-mediated inhibition of TLR responses in microglia.
This high level of LJ529 concentration in blood might not be obtained
in vivo by dual injection of LJ529 (2 mg/kg each), as in the present study. A pharmacokinetic analysis showed that the peak plasma concentration of LJ529 was below micromolar level (unpublished data). Moreover, although the effect of LJ529 on cytokine/chemokine release was shown to be diminished by both A3AR and A2AAR antagonists (
Figure 3), only the A3AR antagonist (and not the A2AAR antagonist) blocked the anti-ischemic effect of LJ529
in vivo (
Figure 1). Thus, attenuation of cerebral infarction by LJ529 may not be fully attributed to the decreased production of inflammatory cytokines.
LJ529 directly inhibited MCP-1-induced microglial chemotaxis through A3AR (
Figure 3). This result is consistent with previous studies in human lung eosinophils
55- Walker B.A.
- Jacobson M.A.
- Knight D.A.
- Salvatore C.A.
- Weir T.
- Zhou D.
- Bai T.R.
Adenosine A3 receptor expression and function in eosinophils.
and mouse bone marrow neutrophils,
14- Ge Z.D.
- van der Hoeven D.
- Maas J.E.
- Wan T.C.
- Auchampach J.A.
A(3) adenosine receptor activation during reperfusion reduces infarct size through actions on bone marrow-derived cells.
, 46- van der Hoeven D.
- Wan T.C.
- Auchampach J.A.
Activation of the A(3) adenosine receptor suppresses superoxide production and chemotaxis of mouse bone marrow neutrophils.
showing that selective activation of the A3AR regulates the chemotaxis of inflammatory cells. Because L529 exerts the anti-chemotactic effect at nanomolar concentrations, we further examined the effect of an A3AR agonist on cellular chemotactic machinery, especially the Rho family of small GTPases, as a putative mechanism underlying the anti-ischemic effect of LJ529. The Rho GTPases are expressed in inflammatory cells, and their inhibitors are known to prevent the migration of inflammatory monocytes or leukocytes into brain and to attenuate the inflammatory process in multiple sclerosis animal models.
42- Hendriks J.J.
- Alblas J.
- van der Pol S.M.
- van Tol E.A.
- Dijkstra C.D.
- de Vries H.E.
Flavonoids influence monocytic GTPase activity and are protective in experimental allergic encephalitis.
, 56- Walters C.E.
- Pryce G.
- Hankey D.J.
- Sebti S.M.
- Hamilton A.D.
- Baker D.
- Greenwood J.
- Adamson P.
Inhibition of Rho GTPases with protein prenyltransferase inhibitors prevents leukocyte recruitment to the central nervous system and attenuates clinical signs of disease in an animal model of multiple sclerosis.
For directed cell migration, cells have to be polarized through the expression cytoskelectal change and the activation and polarized expression of Rho GTPases is known to play a key role in regulating the polarized actin cytoskeleton that organizes the membrane protrusions and focal adhesions.
39Rho GTPases and the actin cytoskeleton.
, 40Cellular signaling in macrophage migration and chemotaxis.
, 41- Sánchez-Madrid F.
- del Pozo M.A.
Leukocyte polarization in cell migration and immune interactions.
, 42- Hendriks J.J.
- Alblas J.
- van der Pol S.M.
- van Tol E.A.
- Dijkstra C.D.
- de Vries H.E.
Flavonoids influence monocytic GTPase activity and are protective in experimental allergic encephalitis.
, 43- Honing H.
- van den Berg T.K.
- van der Pol S.M.
- Dijkstra C.D.
- van der Kammen R.A.
- Collard J.G.
- de Vries H.E.
RhoA activation promotes transendothelial migration of monocytes via ROCK.
Forward protrusion to move (lamellipodia or filopodia) is associated with actin polymerization, which is coupled to an actin-myosin II-mediated contraction at the posterior of the cell (uropod).
57- Allen W.E.
- Jones G.E.
- Pollard J.W.
- Ridley A.J.
Rho, Rac and Cdc42 regulate actin organization and cell adhesion in macrophages.
, 58- Worthylake R.A.
- Lemoine S.
- Watson J.M.
- Burridge K.
RhoA is required for monocyte tail retraction during transendothelial migration.
, 59- Xu J.
- Wang F.
- Van Keymeulen A.
- Herzmark P.
- Straight A.
- Kelly K.
- Takuwa Y.
- Sugimoto N.
- Mitchison T.
- Bourne H.R.
Divergent signals and cytoskeletal assemblies regulate self-organizing polarity in neutrophils.
Although the A3AR agonist Cl-IB-MECA has been reported to induce Rho-mediated stress fibers in human astroglioma cells,
60- Abbracchio M.P.
- Rainaldi G.
- Giammarioli A.M.
- Ceruti S.
- Brambilla R.
- Cattabeni F.
- Barbieri D.
- Franceschi C.
- Jacobson K.A.
- Malorni W.
The A3 adenosine receptor mediates cell spreading, reorganization of actin cytoskeleton, and distribution of Bcl-XL: studies in human astroglioma cells.
the relationship between A3AR agonist and Rho GTPases in immune cells remains obscure. Moreover, because microglia do not possess stress fibers,
61- Abd-El-Basset E.M.
- Prashanth J.
- Ananth Lakshmi K.V.
Up-regulation of cytoskeletal proteins in activated microglia.
Rho GTPases cannot act on stress fibers in these cells. Instead, we demonstrated for the first time in the present study that LJ529 inhibits the activity and polarized expression of Rac, Cdc42, and Rho in microglia (
Figure 4), possibly inhibiting actin polarity and cytoskeletal change and thus leading to decreased MCP-1-induced chemotaxis and migration. Further studies using a constitutively active form or an RNA interference of Rho GTPases in combination with LJ529 may confirm our assumptions in microglia.
In cultured cortical neurons, we found that postischemic treatment with LJ529 did not prevent OGD/reoxygenation-evoked neurotoxicity. The lack of neuroprotection by postischemic treatment with LJ529 may not be surprising, because inflammatory responses are not involved in OGD/reoxygenation-evoked neurotoxicity. Some researchers have, however, reported anti-ischemic effect of A3AR agonists in cultured neurons. For example, chronic preischemic treatment of an A3AR agonist protected cortical neurons against OGD/reoxygenation-induced damage.
15- Chen G.J.
- Harvey B.K.
- Shen H.
- Chou J.
- Victor A.
- Wang Y.
Activation of adenosine A3 receptors reduces ischemic brain injury in rodents.
Also, prolonged treatment of A3AR agonist before/during ischemia, but not with short exposure, reduced anoxic depolarization-evoked neuronal death and disrupted excitatory neurotransmission by OGD in rat hippocampal slices
62- Pugliese A.M.
- Coppi E.
- Volpini R.
- Cristalli G.
- Corradetti R.
- Jeong L.S.
- Jacobson K.A.
- Pedata F.
Role of adenosine A3 receptors on CA1 hippocampal neurotransmission during oxygen-glucose deprivation episodes of different duration.
or against forebrain ischemia in gerbils.
17- Von Lubitz D.K.
- Lin R.C.
- Popik P.
- Carter M.F.
- Jacobson K.A.
Adenosine A3 receptor stimulation and cerebral ischemia.
Because long-term treatment with an A3AR agonist may affect various cellular functions, the exact reason for disparities in results obtained by us and by other researchers needs to be further clarified. Although this is speculative, the treatment time-dependent anti-ischemic effect of A3AR-mediated effects may be due to temporal difference of endogenous adenosine outflow under ischemia,
63- Hagberg H.
- Andersson P.
- Lacarewicz J.
- Jacobson I.
- Butcher S.
- Sandberg M.
Extracellular adenosine, inosine, hypoxanthine, and xanthine in relation to tissue nucleotides and purines in rat striatum during transient ischemia.
, 64- Latini S.
- Bordoni F.
- Corradetti R.
- Pepeu G.
- Pedata F.
Temporal correlation between adenosine outflow and synaptic potential inhibition in rat hippocampal slices during ischemia-like conditions.
as well as changes in receptor desensitization, internalization, and/or down-regulation of adenosine receptors.
65- Klaasse E.C.
- Ijzerman A.P.
- de Grip W.J.
- Beukers M.W.
Internalization and desensitization of adenosine receptors.
In summary, although LJ529 does not modulate the excitotoxicity and oxidative stress that are well-known neurotoxic factors in the early phase of ischemia, it markedly reduces cerebral infarct through activation of the A3AR, possibly via suppression of microglia/monocyte accumulation into the ischemic lesions and their inflammatory responses. Considering that the short therapeutic time window for effective treatment of cerebral ischemic stroke may be due to delayed but sustained inflammatory responses, A3AR agonists (including LJ529) could represent an attractive therapeutic drug for the treatment of postischemic injury. Moreover, because multiple pathways leading to neuronal death are activated in cerebral ischemia, a combination of drugs rather than single-drug treatment may be required for efficient neuroprotection. In support of this notion, the three-drug cocktail of minocycline (an anti-microbial agent with anti-inflammatory properties), riluzole (a glutamate antagonist), and nimodipine (a calcium channel blocker) has been reported to exert significant neuroprotection in a focal ischemic mouse model.
66Differential neuroprotective effects of a minocycline-based drug cocktail in transient and permanent focal cerebral ischemia.
Thus, an optimal therapy combining the anti-inflammatory drug LJ529 with certain drugs with antioxidant and/or anti-excitotoxic activities may also be advantageous.
Article info
Publication history
Published online: August 19, 2011
Accepted:
July 1,
2011
Footnotes
Supported by grants from Brain Research Center of the 21st Century Frontier Research Program (2010K000808 to W.-K.K.) the Bio & Medical Technology Development Program (No. 2011-0019440) and from Mid-career Researcher Program (2010-0026203 to L.S.J.) through the National Research Foundation funded by the Ministry of Education, Science and Technology, Republic of Korea.
I.-Y.C, and J.-C.L. contributed equally to the present work.
Supplemental material for this article can be found at http://ajp.amjpathol.org or at doi: 10.1016/j.ajpath.2011.07.006.
Copyright
© 2011 American Society for Investigative Pathology. Published by Elsevier Inc.