Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2003 Jun 30;139(8):1539–1547. doi: 10.1038/sj.bjp.0705385

LF 16-0687 Ms, a bradykinin B2 receptor antagonist, reduces ischemic brain injury in a murine model of transient focal cerebral ischemia

Li Ding-Zhou 1, Isabelle Margaill 1, Bruno Palmier 1, Didier Pruneau 2, Michel Plotkine 1, Catherine Marchand-Verrecchia 1,*
PMCID: PMC1573979  PMID: 12922942

Abstract

  1. Bradykinin promotes neuronal damage and brain edema through the activation of the B2 receptor. The neuroprotective effect of LF 16-0687 Ms, a B2 receptor antagonist, has been described when given prior to induction of transient focal cerebral ischemia in rat, but there are no data regarding the consequence of a treatment when given after injury. Therefore, in a murine model of transient middle cerebral artery occlusion (MCAO), we evaluated the effect of LF 16-0687 Ms given prior to and/or after the onset of ischemia on neurological deficit, infarct volume and inflammatory responses including cerebral edema, blood–brain barrier (BBB) disruption and neutrophil accumulation.

  2. LF 16-0687 Ms (1, 2 and 4 mg kg−1) administered 0.5 h before and, 1.25 and 6 h after MCAO, decreased the infarct volume by a maximum of 33% and significantly improved the neurological recovery.

  3. When given at 0.25 and 6.25 h after MCAO, LF 16-0687 Ms (1.5, 3 and 6 mg kg−1) decreased the infarct volume by a maximum of 25% and improved the neurological score.

  4. Post-treatment with LF 16-0687 Ms (1.5 mg kg−1) significantly decreased brain edema (−28%), BBB disruption (−60%) and neutrophil accumulation (−65%) induced by ischemia. Physiological parameters were not modified by LF 16-0687 Ms.

  5. These data emphasize the role of bradykinin B2 receptor in the development of infarct lesion, neurological deficit and inflammatory responses resulting from transient focal cerebral ischemia. Therefore, B2 receptor antagonist might represent a new therapeutic approach in the pharmacological treatment of stroke.

Keywords: Bradykinin B2 receptor antagonist, focal cerebral ischemia, mice, neuroprotection, brain edema, inflammation

Introduction

Bradykinin is generated in plasma and tissue from kininogen by the action of kallikrein (Bhoola et al., 1992). All the components of the kallikrein–kinin system have been identified in brain tissue from various species including human (Kizuki et al., 1994; Walker et al., 1995; Raidoo et al., 1996). The actions of bradykinin are mediated through the G-protein coupled B2 (constitutive) and B1 (inducible) receptor subtypes; however the B2 receptor accounts for the majority of acute physiological effects of bradykinin (Marceau, 1995; Regoli et al., 1998). The B2 receptor has been located on cerebral endothelial cells (Wahl et al., 1996), neurons (Raidoo & Bhoola, 1997; Chen et al., 2000), astrocytes and microglia (Hosli & Hosli, 1993). In neuronal and neuroglial tissues, activation of the B2 receptor induces the production and release of proinflammatory mediators such as reactive oxygen species (Rosenblum, 1987; Ellis, 1990; Sobey et al., 1997), nitric oxide (Katusic et al., 1993; Gorlach & Wahl, 1996), prostanoids (Gecse et al., 1989), excitatory amino-acid neurotransmitters (Parpura et al., 1994; Jeftinija et al., 1996) and cytokines (Cunha et al., 1992). The release of such mediators triggers inflammatory reactions resulting in cerebral arteriolar dilatation, loss of cerebrovascular autoregulation, endothelial cell lesion and increased cerebral vascular permeability (Whalley & Wahl, 1983; Unterberg et al., 1984; Wahl et al., 1996). These events lead to edema formation, blood–brain barrier (BBB) disruption and ultimately neuronal injury and cell death (Unterberg et al., 1986; Francel, 1992; Walker et al., 1995). Bradykinin is considered as an important mediator of inflammatory responses associated to cerebral injury (Unterberg & Baethmann, 1984; Francel, 1992). An increase in plasma bradykinin levels was observed during global cerebral ischemia in rat (Kamiya et al., 1993) and activation of the kallikrein–kinin pathway was reported in patients with cerebral infarct (Wagner et al., 2002). Although a number of experimental studies showed that the nonpeptide B2 receptor antagonist, LF 16-0687 Ms (Pruneau et al., 1999b), reduced cerebral edema and improved neurological function in rodent models of traumatic brain injury including closed head trauma (Pruneau et al., 1999a; Verrecchia et al., 2000; Rachinsky et al., 2001; Kaplanski et al., 2002), controlled cortical impact injury (Stover et al., 2000) and cold lesion trauma (Schulz et al., 2000; Gorlach et al., 2001; Plesnila et al., 2001), only few studies have investigated the contribution of the B2 receptor in cerebral ischemia. Relton et al. (1997) demonstrated that CP-0597, a peptide B2 receptor antagonist, significantly reduced brain swelling, infarct size and improved behavioral outcomes and body weight loss in a rat model of transient focal cerebral ischemia. Using the same model, Zausinger et al. (2002) reported that LF 16-0687 Ms was neuroprotective, further supporting a detrimental role of B2 receptor in the development of ischemic brain damage. However, in this study, LF 16-0687 Ms was given before the induction of ischemia, and to date, there are no data regarding the effect of LF 16-0687 Ms administered after the onset of ischemia. In addition, there is no experimental study examining the consequence of a treatment with a B2 receptor antagonist in ischemic mice brain.

Therefore, the main purpose of the present study was to evaluate the potential therapeutic value of LF 16-0687 Ms administered after the onset of ischemia on neurological deficit, histological damage and inflammatory responses including cerebral edema, BBB disruption and neutrophil accumulation following transient middle cerebral artery occlusion (MCAO) in mice.

Methods

Drugs

LF 16-0687 Ms (1-[[3-[(2,4-dimethylquinolin-8-yl)oxymethyl]-2,4-dichlorophenyl]sulfonyl]-N-[3-[[4-(aminoiminomethyl)-phenyl]carbonyl-amino]propyl]-2(S)-pyrrolidinecarboxyamide dimesylate salt) was synthesized as a mesylate salt at Laboratoires Fournier (Daix, France). This compound was dissolved in saline and injected subcutaneously (10 ml kg−1) in all the experiments. Doses of LF 16-0687 Ms used in the present study refer to the base form.

Transient focal cerebral ischemia

All experiments were performed on male Swiss mice weighing 22–25 g (Charles River, France) housed under a controlled temperature, 12 h light/dark cycle and with access to food and water ad libitum. Animal care was in compliance with French regulations on protection of animals used for experimental and other scientific purposes (D2001-486), as well as with the EEC regulations (Official Journal of European Community L358 12/18/1986).

Mice were anesthetized with chloral hydrate (Sigma, France) at the dose of 400 mg kg−1 (i.p. 10 ml kg−1). Body temperature was maintained at 37±0.5°C by means of a heating blanket and a heating lamp throughout the entire experimental procedure. Focal cerebral ischemia was induced by left MCAO with the use of an intraluminal filament technique, as previously described (Ding-Zhou et al., 2002). Briefly, the left common and external carotid arteries were isolated and ligated with a 4-0 silk suture through a midline neck incision. An arteriotomy was fashioned in the common carotid artery just proximal to the carotid bifurcation. A 6-0 nylon monofilament (Ethicon), blunted at the tip with an open flame, was introduced through this incision into the internal carotid artery and advanced approximately 13 mm distal to the carotid bifurcation for occlusion of the origin of the middle cerebral artery. The thread was carefully withdrawn 15 min after MCAO. After recovery from anesthesia, animals were returned to their cages and placed in a heating incubator at 29°C with free access to food and water during the entire postoperative period (24 or 48 h). In sham-operated mice, the carotid arteries were prepared surgically, but the filament was not inserted (Kamii et al., 1994).

Experimental protocols

Study I: effect of pre- and postischemic treatment with LF 16-0687 Ms

Treatment with LF 16-0687 Ms or its vehicle (saline) was started 0.5 h before ischemia, and repeated 1.25 and 6 h after the onset of ischemia. Mice were randomly assigned to five groups: group 1, non-operated mice; group 2, ischemic mice treated with saline; groups 3–5, ischemic animals treated with LF 16-0687 Ms at the dose of 1, 2 and 4 mg kg−1. The mice thus received a cumulative dose of 3, 6 and 12 mg kg−1 respectively. This dosing schedule has been selected according to the studies of Zausinger et al. (2002). These authors have shown that LF 16-0687 Ms at low dose, given 0.5 h before ischemia, at onset of reperfusion and 6 h after reperfusion, reduced ischemic brain damage in rats subjected to 1.5 h MCAO. At 48 h after ischemia, neurological deficit and brain infarction were determined.

Study II: effect of postischemic treatment with LF 16-0687 Ms

Treatment with LF 16-0687 Ms or its vehicle (saline) was given 0.25 and 6.25 h after the onset of ischemia. Mice submitted to MCAO were randomly assigned to five groups: group 1, non-operated mice; group 2, ischemic mice treated with saline; groups 3–5, ischemic mice treated with LF 16-0687 Ms at the dose of 1.5, 3 and 6 mg kg−1. Thus, here again, the mice received a cumulative dose of 3, 6 and 12 mg kg−1 respectively. Neurological deficit and infarct volume were determined 48 h after ischemia.

Furthermore, the effect of LF 16-0687 Ms given postischemia was investigated on cerebral edema, BBB disruption and neutrophil accumulation. In each experiment, four groups were studied: a group of non-operated mice and a group of sham-operated mice served as controls, and two ischemic groups were treated with either saline or LF 16-0687 Ms at the dose of 1.5 mg kg−1 given 15 min and 6.25 h after MCAO (total dose of LF 16-0687 Ms 3 mg kg−1). Cerebral edema and the BBB disruption were examined 24 h after ischemia, and neutrophil accumulation was evaluated 48 h after MCAO. These time points were chosen on the basis of our preliminary studies concerning the time course of these ischemic outcomes in our model (Ding-Zhou et al., 2002; unpublished observations).

Evaluation of neurological deficit

Sensorimotor neurological deficit was assessed by a grip test (Hall, 1985). Each mouse was picked up by the tail and placed on a taut string 60 cm long suspended 40 cm above a table. Grip score was measured as the length of time (in seconds) that the mouse remained on the string in some manner (using one or more paws, tail, tail plus paws), for a maximum of 30 s. Each experiment was conducted randomly and blindly. Neurological examinations were assessed only once in non-operated, vehicle- and LF 16-0687 Ms-treated ischemic mice, so the neurogical evaluation was not performed before the treatment. This choice is made on the basis of studies of Hall (1985) who has reported that if mice were placed repeatedly on the string, some of them could be made to perform better. This phenomena is probably related to the effect of training and memory. So it is useful to allow each mouse an equal opportunity when performing this neurological test.

Measurement of infarct volume

After neurological score evaluation, mice were killed with an overdose of sodium pentobarbitone (200 mg kg−1, i.p. , Sanofi, France), their brains were removed and sectioned into six 1 mm-thick coronal slices using a tissue Chopper (McIlwain, The Mickle laboratory Engineering, England). Coronal brain slices were immediately immersed into 2% 2,3,5,-triphenyltetrazolium chloride (Sigma, France) for 20 min at room temperature in the dark followed by fixation in a 4% paraformaldehyde solution (Prolabo, France) overnight prior to analysis (Bederson et al., 1986). The infarction area, outlined in white, was measured on the posterior surface of each section using a computer image analysis system (Imstar, Paris, France) and corrected for brain edema according to Golanov & Reis (1995). Infarct volume, expressed in mm3, was calculated by a linear integration of the corrected lesion areas.

Evaluation of cerebral edema

Cerebral edema was determined by measuring the brain water content (BWC) according to the wet–dry method (Hatashita et al., 1988). The mice were killed and brains were immediately removed and placed on a frozen plate. Tissue samples were dissected out from infarct areas in ischemic mice and from corresponding areas in sham-operated and non-operated animals. Samples were immediately weighed to obtain wet weight. Then, samples were dried in a desiccating oven at 110°C for 24 h and weighed again to obtain the dry weight. BWC was calculated as follows: BWC (%)=(wet weight−dry weight) × 100/wet weight.

Evaluation of BBB permeability

The integrity of the BBB was investigated using Evans blue extravasation (Chan et al., 1991; Ikeda et al., 1994). Evans blue at 1% in saline (100 μl) was injected into the tail vein. After 60 min, the chest wall was opened under sodium pentobartitone anesthesia (55 mg kg−1, i.p.). Animals were perfused transcardially with saline at 100 mmHg pressure until blue color was absent from the effluent. Brains were then removed, tissue samples were dissected out as described above, weighed, placed in 400 μl of pure formamide (Sigma, France) and incubated for 72 h in the dark at 50°C. The optical density of the formamide solution was measured at 620 nm. Data are expressed as μg Evans blue g−1 tissue.

Evaluation of neutrophil accumulation

Myeloperoxidase (MPO) activity was used as an indicator of neutrophil accumulation in brain parenchyma (Barone et al., 1991). Mice were anesthetized with sodium pentobarbitone (55 mg kg−1, i.p.) and transcardially perfused with 200 ml of saline at 100 mmHg. Brains were removed, samples were dissected out as previously described and immediately frozen at −40°C until the assay. Frozen tissue samples were weighted and homogenized with an ultra thurax (Ika Labortehnik, Germany) in 20 w v−1 of 5 mmol l−1 potassium phosphate buffer (4°C, pH 6) followed by centrifugation for 30 min at 25,000 × g at 4°C. Supernatants were discarded, and the pellets were washed again as described above. MPO was extracted by suspension in 0.5% hexadecyltrymethylammonium bromide in 10 w v−1 50 mmol l−1 potassium phosphate buffer (pH 6, 25°C). The samples were immediately frozen in methyl-2 butane at −40°C, and three freeze–thaw cycles were performed with 10 s sonication (Vibra Cell, Fischer Bioblock Scientific, France) between each cycle at 25°C. Samples were then incubated at 4°C for 20 min and centrifuged at 12,500 × g for 15 min at 4°C. An aliquot of 7 μl of the supernatant was then allowed to react at 25°C with 200 μl of mixed solution containing 0.167 mg ml−1 o-dianisidine dihydrochloride (Sigma, France) and 0.0005% hydrogen peroxide (Sigma, France) in 50 mmol l−1 potassium phosphate buffer (pH 6).

The kinetic change in absorbance at 460 nm during 2 min was assayed using a colorimetric microplate reader (Dynex, MRX France). One unit of MPO activity was defined as the degradation of 1 μmol of hydrogen peroxide per minute at 25°C and was expressed in units per gram weight of wet tissue (U MPO g−1).

Effect of LF 16-0687 Ms on physiological variables

The carotid artery was cannulated to study the effect of LF 16-0687 Ms on mean arterial blood pressure (MABP), arterial blood gases and pH. The arterial catheter was connected to a pressure transducer (EMKA Technologies). Once MABP was stabilized, animals were treated with either LF 16-0687 Ms at the dose of 1.5, 3 and 6 mg kg−1 or its vehicle. MABP was recorded during 2 h. Arterial blood samples were collected 15, 30, 60, 90 and 120 min after treatment to analyze pH, partial pressure in arterial oxygen and carbon dioxide (PaO2 and PaCO2), using a blood gas and pH analyzer (ABL 330, Radiometer, France). Body temperature was maintained at 37±0.5°C by means of a heating blanket throughout the experiment.

Data expression and statistical analysis

Data are expressed as means±s.e.m. (standard error of mean) of n observations, where n represents the number of animals or samples. Comparisons between multiple groups were evaluated by one-way analysis of variance (ANOVA) followed by protected least-significant difference (PLSD) Fisher's test. The effect of surgical procedure was first evaluated by comparing sham-operated group versus the non-operated group using one-way ANOVA followed by Dunnett's test. Comparisons between sham-operated and ischemic groups were then evaluated by one-way ANOVA followed by PLSD Fisher's test. When measures were repeated (physiological variables), data were analyzed by a two-factorial ANOVA for repeated measures. A P-value less than 0.05 was considered to be statistically significant.

Results

Study I: effect of pre- and postischemic treatment with LF 16-0687 Ms

Effect on neurological deficit

At 48 h after MCAO, the grip score reflected a severe impairment of sensory motor function in vehicle-treated ischemic mice as compared with non-operated mice (9.8±4.0 versus 29.7±0.3 s, P<0.001) (Figure 1a). LF 16−0687 Ms improved the grip score in a dose-related manner with a significant effect at 2 and 4 mg kg−1 (22.3±3.4 and 25.2±3.1 s, P<0.01 versus vehicle-treated ischemic mice).

Figure 1.

Figure 1

Effect of pre- and postischemic treatment with LF 16-0687 Ms on (a) grip test and (b) infarct volume 48 h after MCAO. Vehicle and LF 16-0687 Ms at 1, 2 and 4 mg kg−1 were given (s.c.) 0.5 h before, 1.25 and 6 h after MCAO. Experimental groups consisted of 9–11 mice. Values are means±s.e.m. †††P<0.001 versus non-operated mice, *P<0.05 and **P<0.01 versus vehicle-treated mice.

Effect on infarct volume

At 48 h after transient MCAO, an infarct developed in the cerebral cortex and the striatum (80±4 mm3) (Figure 1b). Administration of LF 16-0687 Ms at 1, 2 and 4 mg kg−1 significantly reduced the total infarct volume by 33% (54±10 mm3, P<0.01), 23% (62±7 mm3, P<0.05) and 33% (54±7 mm3, P<0.01) respectively.

Study II: effect of postischemic treatment with LF 16-0687 Ms

Effect on neurological deficit

At 48 h after transient MCAO, LF 16-0687 Ms given 0.25. and 6.25 h postischemia at doses of 1.5, 3 and 6 mg kg−1 improved the grip score (23.1±2.4 s, P<0.001; 21.9±2.1 s, P<0.001 and 20.1±2.9 s, P<0.01 versus 8.8±3.3 s in vehicle-treated ischemic mice, respectively) (Figure 2a).

Figure 2.

Figure 2

Effect of postischemic treatment with LF 16-0687 Ms on (a) grip test and (b) infarct volume 48 h after MCAO. Vehicle and LF 16-0687 Ms at 1.5, 3 and 6 mg kg−1 were given (s.c.) 0.25 and 6.25 h after MCAO. Experimental groups consisted of 10–15 mice. Values are means±s.e.m. †††P<0.001 versus non-operated mice, *P<0.05, **P<0.01 and ***P<0.001 versus vehicle-treated mice.

Effect on infarct volume

At 48 h after transient MCAO, administration of LF 16-0687 Ms at 1.5, 3 and 6 mg kg−1 significantly reduced the total infarct volume by 25% (65±5 mm3, P<0.01), 25% (65±5 mm3, P<0.01) and 17% (71±6 mm3, P<0.05), respectively, compared with vehicle-treated ischemic mice (86±3 mm3) (Figure 2b). This histological protection was more prominent in the cortical infarct area (25–35%) than in the striatal lesion (10–14%).

Effect on brain water content

At 24 h after transient MCAO, the BWC in the ischemic area was significantly increased in vehicle-treated mice compared with that of sham-operated mice (85.8±0.5 versus 80.1±0.1%, P<0.001) (Figure 3). LF 16-0687 Ms at a dose of 1.5 mg kg−1, given 0.25 and 6.25 h after MCAO reduced the BWC increase (84.2±0.7%, P<0.05 versus vehicle-treated mice). The BWC in sham-operated mice was not significantly different from that of non-operated mice.

Figure 3.

Figure 3

Effect of LF 16-0687 Ms on the brain water content (BWC) measured 24 h after MCAO. LF 16-0687 Ms (1.5 mg kg−1) was given (s.c.) 0.25 and 6.25 h after MCAO. Experimental groups consisted of 10 mice. Values are means±s.e.m. †††P<0.001 versus sham-operated mice, *P<0.05 versus vehicle-treated mice.

Effect on Evans blue extravasation

Evans blue content in brain tissue was similar in sham-operated and non-operated mice (0.6±0.3μg g−1 tissue versus 0.1±0.0 μg g−1 tissue) (Figure 4). At 24 h after MCAO, Evans blue content in the area of infarction was markedly increased in the vehicle-treated group (14.6±3.1 μg g−1 tissue, P<0.001 versus sham-operated mice). In mice treated with 1.5 mg kg−1 of LF 16-0687 Ms, 0.25 and 6.25 h after MCAO, Evans blue content was reduced by 60% (5.8±3.9 μg g−1 tissue, P<0.05 versus vehicle-treated group).

Figure 4.

Figure 4

Effect of LF 16-0687 Ms on Evans blue extravasation measured 24 h after MCAO. LF 16-0687 Ms (1.5 mg kg−1) was given (s.c.) 0.25 and 6.25 h after MCAO. Experimental groups consisted of 9–11 mice. Values are means±s.e.m. †††P<0.001 versus sham-operated mice, *P<0.05 versus vehicle-treated mice.

Effect on brain myeloperoxidase activity

At 48 h after ischemia, vehicle-treated mice exhibited a massive increase in MPO activity compared with sham-operated mice (0.189±0.004 versus 0.010±0.003 U g−1, P<0.001) (Figure 5). When LF 16-0687 Ms was given at a dose of 1.5 mg kg−1, 0.25 and 6.25 h after MCAO, the increased MPO activity was reduced by 65% (0.066±0.025 U g−1, P<0.001 versus vehicle-treated mice). There was no significant difference between MPO activity in sham-operated and non-operated mice.

Figure 5.

Figure 5

Effect of LF 16-0687 Ms on myeloperoxidase activity (MPO) measured 48 h after MCAO. LF 16-0687 Ms (1.5 mg kg−1) was given (s.c.) 0.25 and 6.25 h after MCAO. Experimental groups consisted of 14–15 mice. Values are means±s.e.m. †††P<0.001 versus sham-operated mice, ***P<0.001 versus vehicle-treated mice.

Effect on physiological variables

Administration of LF 16-0687 Ms at 1.5, 3 and 6 mg kg−1 did not result in any effect on MABP, PaO2, PaCO2 and pH (Table 1). Physiological variables were within the normal physiological ranges throughout the experiment without significant difference between vehicle-treated and LF 16-0687 Ms-treated animals.

Table 1.

Physiological variables for saline and LF 16-0687 Ms-treated mice

  MABP (mmHg) PaO2 (mmHg) PaCO2 (mmHg) pH
Vehicle
 Before 89±8 122±6 37.5±2.2 7.38±0.03
 15 min after 89±9 118±6 40.3±2.8 7.38±0.02
 30 min after 87±9 122±3 42.5±1.6 7.34±0.02
 60 min after 90±9 121±3 37.1±2.0 7.33±0.03
 90 min after 90±7 121±5 41.6±2.0 7.35±0.03
 120 min after 90±5 116±6 39.4±2.2 7.39±0.02
LF 16-0687 Ms (1.5 mg kg−1)
 Before 87±5 122±6 37.5±2.2 7.38±0.03
 15 min after 90±5 113±2 40.5±2.1 7.39±0.03
 30 min after 93±3 118±5 37.8±1.2 7.38±0.03
 60 min after 90±5 114±6 38.6±2.6 7.33±0.03
 90 min after 86±7 125±3 40.1±2.2 7.34±0.03
 120 min after 87±8 120±4 38.7±1.8 7.38±0.01
LF 16-0687 Ms (3 mg kg−1)
 Before 87±7 122±6 37.5±2.2 7.38±0.03
 15 min after 91±7 117±3 36.7±1.0 7.36±0.03
 30 min after 90±5 120±6 40.5±1.8 7.34±0.03
 60 min after 89±5 119±7 39.1±2.0 7.34±0.03
 90 min after 84±7 113±7 41.9±1.9 7.34±0.03
 120 min after 89±7 111±4 40.0±2.5 7.38±0.03
LF 16-0687 Ms (6 mg kg−1)
 Before 90±6 122±6 37.5±2.2 7.38±0.03
 15 min after 91±6 112±6 42.3±2.6 7.36±0.02
 30 min after 94±6 120±7 42.0±3.1 7.34±0.01
 60 min after 92±7 119±7 41.2±2.3 7.33±0.03
 90 min after 88±7 120±7 42.3±3.0 7.34±0.03
 120 min after 90±7 114±5 40.3±2.4 7.40±0.03

Values of MABP, PaCO2, PaO2 and pH were measured before and after treatment of LF 16-0687 Ms (1.5, 3 and 6 mg kg−1) or its vehicle. There were no significant differences between groups. Values are means±s.e.m. (n=6 per group).

Discussion

In the present study, we evaluated the neuroprotective effect of LF 16-0687 Ms, a selective bradykinin B2 receptor antagonist, in mice submitted to transient focal cerebral ischemia. We demonstrated that LF 16-0687 Ms reduced the infarct volume and the neurological deficit even when the treatment was initiated after the onset of MCAO. Furthermore, we showed that LF 16-0687 Ms decreased cerebral edema, BBB disruption and neutrophil accumulation following ischemia. These results suggest that bradykinin, through the activation of B2 receptor, plays a detrimental role in the development of ischemic damage and support the development of LF 16-0687 Ms as a powerful therapeutic approach of stroke.

LF 16-0687 Ms is a selective nonpeptide antagonist of the bradykinin B2 receptor (Pruneau et al., 1999a) which is about 1000-fold more potent than CP-0127, another B2 receptor antagonist (Cheronis et al., 1992; Pruneau et al., 2000) and which is devoid of partial agonist activity (Pruneau et al., 1999b) in contrast with Hoe 140, another B2 receptor antagonist (Hock et al., 1991; Wirth et al., 1991; Feletou et al., 1994). Pharmacokinetic studies in mice indicated that LF 16-0687 Ms given s.c. had a rapid onset of action with a maximal plasma value achieved at 1 h. In agreement with previous observations (Pruneau et al., 1999a; Stover et al., 2000; Plesnila et al., 2001), we showed that LF 16-0687 Ms had no adverse effects on MABP, blood gases and pH.

The mechanism by which bradykinin contibutes to the development of ischemic brain injury remains unclear. Previous studies that investigated the differential role of B1 and B2 receptors after focal cerebral ischemia (Relton et al., 1997) and cold cortical injury (Gorlach et al., 2001) demonstrated that blockade of the B2 receptor was protective against neuronal deficit and survival in agreement with the present study, while the B1 receptor was apparently not involved. In the brain of mammals, B2 receptor is present on endothelial cells, glial cells and neurons (Walker et al., 1995; Raidoo et al., 1996; Chen et al., 2000). Activation of this receptor results in stimulation of the membrane phosphatidyl–inositol cascade, activation of protein kinase C leading to generation of inositol-1,4,5,-triphosphate and diacylglycerol (Raidoo & Bhoola, 1998). Activation of these intracellular second-messenger systems ultimately leads to the production of prostaglandins (Gecse et al., 1989), nitric oxide (Katusic et al., 1993; Gorlach & Wahl, 1996) and free radicals (Rosenblum, 1987; Ellis, 1990; Sobey et al., 1997), which activate Ca2+-dependent K+ channels resulting in an increase of neuronal intracellular calcium levels (Parpura et al., 1994; Sobey et al., 1997). These endogenous substances are well-known mediators of cell damage. Bradykinin also triggers the production of other inflammatory mediators such as cytokines (Cunha et al., 1992; Schwaninger et al., 1999), P-selectin (Rozsa et al., 1998) and platelet-activating factor (McIntyre et al., 1985; Satoh et al., 1995). In addition, bradykinin has been shown to induce glutamate and aspartate release from cultured astrocytes (Parpura et al., 1994; Jeftinija et al., 1996) and neurons (Rydh-Rinder et al., 2001), these two excitatory amino acids playing a detrimental role in ischemic situations. All of these actions are believed to contribute to pathophysiological disturbance following cerebral ischemia including the development of cerebral edema, an increase of BBB permeability, neutrophil accumulation and ultimately cell destruction (Francel, 1992). These deleterious actions of bradykinin together with the identification of bradykinin receptor throughout the brain (Hosli & Hosli, 1993; Wahl et al., 1996; Raidoo et al., 1996; Raidoo & Bhoola, 1997; Chen et al., 2000) as well as the activation of the kallikrein–kinin system following cerebral injury (Maier-Hauff et al., 1984; Kamiya et al., 1993; Makevnina et al., 1994), provide a strong rationale for the anti-ischemic action of LF 16-0687 Ms. LF 16-0687 Ms was shown to attenuate brain damage and edema formation in rodent models of traumatic brain injury including closed head injury (Pruneau et al., 1999a; Verrecchia et al., 2000; Kaplanski et al., 2002), controlled cortical impact injury (Stover et al., 2000) and cryogenic brain lesion (Schulz et al., 2000; Plesnila et al., 2001). In a rat model of transient focal cerebral ischemia, Zausinger et al. (2002) have reported that 4 mg kg−1 of LF 16-0687 Ms given 0.5 h before ischemia, at onset of reperfusion and 6 h after reperfusion (i.e. 12 mg kg−1 day−1) significantly attenuated brain damage and cerebral edema, while higher doses failed to affect infarct volume. This significant dose-dependent effect of LF 16-0687 Ms, together with other consistent findings in traumatic brain injury (Verrecchia et al., 2000; Plesnila et al., 2001), led us to the dosing regime used in the present experiments. The neuroprotective effects of LF 16-0687 Ms were first confirmed by using a similar pre- and post-treatment strategy. Then, we examined the effect of a post-treatment alone. One of the principal findings of this study is that LF 16-0687 Ms at low doses, given even after the onset of MCAO, can still reduce the infarct volume and improve neurological function with a similar magnitude as to the treatment begun before MCAO. In accordance with our finding, it was shown in a rat model of brain trauma that initiation of LF 16-0687 Ms treatment either 1 h or prior to after the insult was similarly efficient in improving neurological function recovery and in reducing cerebral edema (Rachinsky et al., 2001). Verrecchia et al. (2000) reported that protection could even be accomplished when the onset of treatment was delayed up to 2 h after closed head injury in mice.

The time course of brain edema formation and BBB damage has been previously studied in our murine model of transient MCAO. We showed that the BWC significantly increased 4 h after MCAO, an effect that remained significant for up to 48 h (unpublished observation), and that Evans blue extravasation occurred 6 h after MCAO with a maximum at 24 h (Ding-Zhou et al., 2002). The reduction of cerebral edema and of BBB disruption after a postischemic treatment with LF 16-0687 Ms suggests that BBB damage, leading predominantly to vasogenic brain edema, is a target of LF 16-0687 Ms. Previous data showed that the progression of edema closely correlated with the rise of plasma and tissue bradykinin levels after global ischemia in rat (Kamiya et al., 1993). Accordingly, a significant enhancement of brain kininogen content, which was accompanied by protein leakage and increase of cerebral water content, was detected in rat after fluid percussion injury (Ellis et al., 1989). These results are consistent with other reports, showing that bradykinin synthesis inhibitors and B2 receptor antagonists decreased cerebral edema in both global and transient focal cerebral ischemia in rat (Kamiya et al., 1993; Relton et al., 1997; Zausinger et al., 2002). Cytotoxic edema is also known to participate in brain swelling and neuronal death. However, the present study did not allow to assess if the reduction of brain edema by LF 16-0687 Ms is also due to reduction of the cytotoxic edema.

The inflammatory reaction is a major contibutor to progression of ischemic infarction (Sornas et al., 1972; Kochanek & Hallenbeck, 1992; Matsuo et al., 1995; Liao et al., 2001) and some reports support the view that bradykinin is a chemotactic agent for neutrophils (Bhoola et al., 1992; Bhoola, 1996; Carl et al., 1996). As we previously observed a maximum neutrophil accumulation 48 h after MCAO (unpublished data), this led us to investigate the effect of the blockade of B2 receptor on neutrophil accumulation. We found that the MPO activity increase induced by MCAO was significantly inhibited by the post-treatment with LF 16-0687 Ms, suggesting that the inhibition of neutrophil accumulation participated in the neuroprotective effect of LF 16-0687 Ms. Interestingly, B2 receptor has been localized on the external surface of neutrophils (Haasemann et al., 1994). Reduction of MPO activity in a model of traumatic shock in intestinal tissue (Christopher et al., 1994) and attenuation of neutrophil rolling and sticking following global cerebral ischemia (Lehmberg et al., 1998, 2000) have also been reported with CP-0127, another B2 receptor antagonist. Similarly, the neutrophil accumulation induced by either bradykinin or the venom of Phoneutria nigriventer spider in rat pleural exudate (Costa et al., 2002) and the increased leukocyte adhesion induced by high concentration of bradykinin in rat mesentery (Shigematsu et al., 1999) were markedly attenuated by Hoe-140, another B2 receptor antagonist. In addition to its local production in ischemic inflamed tissue and transudation from plasma due to BBB leakage, bradykinin can also reach inflammatory sites through infiltrating neutrophils which represent another effective way of delivering high concentrations of bradykinin at the site of inflammation (Bockmann & Paegelow, 2000; Bhoola et al., 2001). In this respect, there are data suggesting that neutrophils contain the complete system for the generation of bradykinin (Gustafson et al., 1989; Bockmann & Paegelow, 2000).

In conclusion, our results show that a post-treatment with LF 16-0687 Ms significantly reduced infarct volume, neurological deficit and inflammatory responses including cerebral edema, BBB disruption and neutrophil accumulation. The present findings provide further support for the therapeutic development of LF 16-0687 Ms in stroke and emphasize the key role of bradykinin B2 receptor as a mediator of secondary brain damage.

Abbreviations

BBB

blood–brain barrier

BWC

brain water content

LF 16-0687 MS

1-[[3-[(2,4-dimethylquinolin-8-yl)oxymethyl]−2,4-dichlorophenyl] sulfonyl]-N-[3-[[4-(aminoimino-methyl)phenyl]carbonyl-amino]propyl]-2(S)-pyrrolidine carboxyamide dimesylate salt

MABP

mean arterial blood pressure

MCAO

middle cerebral artery occlusion

MPO

myeloperoxidase

PaO2

arterial pressure in O2

PaCO2

arterial pressure in CO2

References

  1. BARONE F.C., HILLEGASS L.M., PRICE W.J., WHITE R.F., LEE E.V., FEUERSTEIN G.Z., SARAU H.M., CLARK R.K., GRISWOLD D.E. Polymorphonuclear leukocyte infiltration into cerebral focal ischemic tissue: myeloperoxidase activity assay and histologic verification. J. Neurosci. Res. 1991;29:336–345. doi: 10.1002/jnr.490290309. [DOI] [PubMed] [Google Scholar]
  2. BEDERSON J.B., PITTS L.H., GERMANO S.M., NISHIMURA M.C., DAVIS R.L., BARTKOWSKI H.M. Evaluation of 2,3,5-triphenyltetrazolium chloride as a stain for detection and quantification of experimental cerebral infarction in rats. Stroke. 1986;17:1304–1308. doi: 10.1161/01.str.17.6.1304. [DOI] [PubMed] [Google Scholar]
  3. BHOOLA K., RAMSAROOP R., PLENDL J., CASSIM B., DLAMINI Z., NAICKER S. Kallikrein and kinin receptor expression in inflammation and cancer. Biol. Chem. 2001;382:77–89. doi: 10.1515/BC.2001.013. [DOI] [PubMed] [Google Scholar]
  4. BHOOLA K.D. Translocation of the neutrophil kinin moiety and changes in the regulation of kinin receptors in inflammation. Immunopharmacology. 1996;33:247–256. doi: 10.1016/0162-3109(96)00067-7. [DOI] [PubMed] [Google Scholar]
  5. BHOOLA K.D., FIGUEROA C.D., WORTHY K. Bioregulation of kinins: kallikreins, kininogens, and kininases. Pharmacol. Rev. 1992;44:1–80. [PubMed] [Google Scholar]
  6. BOCKMANN S., PAEGELOW I. Kinins and kinin receptors: importance for the activation of leukocytes. J. Leukoc. Biol. 2000;68:587–592. [PubMed] [Google Scholar]
  7. CARL V.S., MOORE E.E., MOORE F.A., WHALLEY E.T. Involvement of bradykinin B1 and B2 receptors in human PMN elastase release and increase in endothelial cell monolayer permeability. Immunopharmacology. 1996;33:325–329. doi: 10.1016/0162-3109(96)00055-0. [DOI] [PubMed] [Google Scholar]
  8. CHAN P.H., YANG G.Y., CHEN S.F., CARLSON E., EPSTEIN C.J. Cold-induced brain edema and infarction are reduced in transgenic mice overexpressing CuZn-superoxide dismutase. Ann. Neural. 1991;29:482–486. doi: 10.1002/ana.410290506. [DOI] [PubMed] [Google Scholar]
  9. CHEN E.Y., EMERICH D.F., BARTUS R.T., KORDOWER J.H. B2 bradykinin receptor immunoreactivity in rat brain. J. Comp. Neurol. 2000;427:1–18. [PubMed] [Google Scholar]
  10. CHERONIS J.C., WHALLEY E.T., NGUYEN K.T., EUBANKS S.R., ALLEN L.G., DUGGAN M.J., LOY S.D., BONHAM K.A., BLODGETT J.K. A new class of bradykinin antagonists: synthesis and in vitro activity of bissuccinimidoalkane peptide dimers. J. Med. Chem. 1992;35:1563–1572. doi: 10.1021/jm00087a010. [DOI] [PubMed] [Google Scholar]
  11. CHRISTOPHER T.A., MA X.L., GAUTHIER T.W., LEFER A.M. Beneficial actions of CP-0127, a novel bradykinin receptor antagonist, in murine traumatic shock. Am. J. Physiol. 1994;266:H867–H873. doi: 10.1152/ajpheart.1994.266.3.H867. [DOI] [PubMed] [Google Scholar]
  12. COSTA S.K., MORENO R.A., ESQUISATTO L.C., JULIANO L., BRAIN S.D., DE NUCCI G., ANTUNES E. Role of kinins and sensory neurons in the rat pleural leukocyte migration induced by Phoneutria nigriventer spider venom. Neuroscience. 2002;318:158–162. doi: 10.1016/s0304-3940(01)02498-3. [DOI] [PubMed] [Google Scholar]
  13. CUNHA F.Q., POOLE S., LORENZETTI B.B., FERREIRA S.H. The pivotal role of tumour necrosis factor alpha in the development of inflammatory hyperalgesia. Br. J. Pharmacol. 1992;107:660–664. doi: 10.1111/j.1476-5381.1992.tb14503.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. DING-ZHOU L., MARCHAND-VERRECCHIA C., CROCI N., PLOTKINE M., MARGAILL I. L-NAME reduces infarction, neurological deficit and blood–brain barrier disruption following cerebral ischemia in mice. Eur. J. Pharmacol. 2002;457:137–146. doi: 10.1016/s0014-2999(02)02686-9. [DOI] [PubMed] [Google Scholar]
  15. ELLIS E.F.Initiation of eicosanoid and free radical formation following brain injury: the role of the kallikrein–kinin system New Trends in Lipid Mediators Research 1990Basel, Switzerland: Karger; 129–145.ed. Braquet, P. & Robinson, L.P [Google Scholar]
  16. ELLIS E.F., CHAO J., HEIZER M.L. Brain kininogen following experimental brain injury: evidence for a secondary event. J. Neurosurg. 1989;71:437–442. doi: 10.3171/jns.1989.71.3.0437. [DOI] [PubMed] [Google Scholar]
  17. FELETOU M., GERMAIN M., THURIEAU C., FAUCHERE J.L., CANET E. Agonistic and antagonistic properties of the bradykinin B2 receptor antagonist, Hoe 140, in isolated blood vessels from different species. Br. J. Pharmacol. 1994;112:683–689. doi: 10.1111/j.1476-5381.1994.tb13130.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. FRANCEL P.C. Bradykinin and neuronal injury. J. Neurotrauma. 1992;9:S27–S45. [PubMed] [Google Scholar]
  19. GECSE A., MEZEI Z., TELEGDY G. The effect of bradykinin and its fragments on the arachidonate cascade of brain microvessels. Adv. Exp. Med. Biol. 1989;247:249–254. doi: 10.1007/978-1-4615-9543-4_36. [DOI] [PubMed] [Google Scholar]
  20. GOLANOV E.V., REIS D.J. Contribution of cerebral edema to the neuronal salvage elicited by stimulation of cerebellar fastigial nucleus after occlusion of the middle cerebral artery in rat. J. Cereb. Blood Flow Metab. 1995;15:172–174. doi: 10.1038/jcbfm.1995.19. [DOI] [PubMed] [Google Scholar]
  21. GORLACH C., HORTOBAGYI T., HORTOBAGYI S., BENYO Z., RELTON J., WHALLEY E.T., WAHL M. Bradykinin B2, but not B1, receptor antagonism has a neuroprotective effect after brain injury. J. Neurotrauma. 2001;18:833–838. doi: 10.1089/089771501316919193. [DOI] [PubMed] [Google Scholar]
  22. GORLACH C., WAHL M. Bradykinin dilates rat middle cerebral artery and its large branches via endothelial B2 receptors and release of nitric oxide. Peptides. 1996;17:1373–1378. doi: 10.1016/s0196-9781(96)00223-9. [DOI] [PubMed] [Google Scholar]
  23. GUSTAFSON E.J., SCHMAIER A.H., WACHTFOGEL Y.T., KAUFMAN N., KUCICH U., COLMAN R.W. Human neutrophils contain and bind high molecular weight kininogen. J. Clin. Invest. 1989;84:28–35. doi: 10.1172/JCI114151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. HAASEMANN M., FIGUEROA C.D., HENDERSON L., GRIGORIEV S., ABD ALLA S., GONZALEZ C.B., DUNIA I., HOEBEKE J., JARNAGIN K., CARTAUD J. Distribution of bradykinin B2 receptors in target cells of kinin action. Visualization of the receptor protein in A431 cells, neutrophils and kidney sections. Braz. J. Med. Biol. Res. 1994;27:1739–1756. [PubMed] [Google Scholar]
  25. HALL E.D. High-dose glucocorticoid treatment improves neurological recovery in head-injured mice. J. Neurosurg. 1985;62:882–887. doi: 10.3171/jns.1985.62.6.0882. [DOI] [PubMed] [Google Scholar]
  26. HATASHITA S., HOFF J.T., SALAMAT S.M. Ischemic brain edema and the osmotic gradient between blood and brain. J. Cereb. Blood Flow Metab. 1988;8:552–559. doi: 10.1038/jcbfm.1988.96. [DOI] [PubMed] [Google Scholar]
  27. HOCK F.J., WIRTH K., ALBUS U., LINZ W., GERHARDS H.J., WIEMER G., HENKE S., BREIPOHL G., KONIG W., KNOLLE J. Hoe 140 a new potent and long acting bradykinin-antagonist: in vitro studies. Br. J. Pharmacol. 1991;102:769–773. doi: 10.1111/j.1476-5381.1991.tb12248.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. HOSLI E., HOSLI L. Autoradiographic localization of binding sites for neuropeptide Y and bradykinin on astrocytes. Neuroreport. 1993;4:159–162. doi: 10.1097/00001756-199302000-00011. [DOI] [PubMed] [Google Scholar]
  29. IKEDA Y., WANG M., NAKAZAWA S. Simple quantitative evaluation of blood–brain barrier disruption in vasogenic brain edema. Acta Neurochir. Suppl. (Wien) 1994;60:119–120. doi: 10.1007/978-3-7091-9334-1_31. [DOI] [PubMed] [Google Scholar]
  30. JEFTINIJA S.D., JEFTINIJA K.V., STEFANOVIC G., LIU F. Neuroligand-evoked calcium-dependent release of excitatory amino acids from cultured astrocytes. J. Neurochem. 1996;66:676–684. doi: 10.1046/j.1471-4159.1996.66020676.x. [DOI] [PubMed] [Google Scholar]
  31. KAMII H., KINOUCHI H., SHARP F.R., EPSTEIN C.J., SAGAR S.M., CHAN P.H. Expression of c-fos mRNA after a mild focal cerebral ischemia in SOD-1 transgenic mice. Brain Res. 1994;662:240–244. doi: 10.1016/0006-8993(94)90818-4. [DOI] [PubMed] [Google Scholar]
  32. KAMIYA T., KATAYAMA Y., KASHIWAGI F., TERASHI A. The role of bradykinin in mediating ischemic brain edema in rats. Stroke. 1993;24:571–576. doi: 10.1161/01.str.24.4.571. [DOI] [PubMed] [Google Scholar]
  33. KAPLANSKI J., PRUNEAU D., ASA I., ARTRU A.A., AZEZ A., IVASHKOVA Y., RUDICH Z., SHAPIRA Y. LF 16-0687 Ms, a bradykinin B2 receptor antagonist, reduces brain edema and improves long-term neurological function recovery after closed head trauma in rats. J. Neurotrauma. 2002;19:953–964. doi: 10.1089/089771502320317104. [DOI] [PubMed] [Google Scholar]
  34. KATUSIC Z.S., MILDE J.H., COSENTINO F., MITROVIC B.S. Subarachnoid hemorrhage and endothelial L-arginine pathway in small brain stem arteries in dogs. Stroke. 1993;24:392–399. doi: 10.1161/01.str.24.3.392. [DOI] [PubMed] [Google Scholar]
  35. KIZUKI K., SUZUKI T., KUDO M., NOGUCHI T. Immunohistochemical demonstration of tissue kallikrein in the neurons of rat brain. Brain Res. 1994;634:305–309. doi: 10.1016/0006-8993(94)91934-8. [DOI] [PubMed] [Google Scholar]
  36. KOCHANEK P.M., HALLENBECK J.M. Polymorphonuclear leukocytes and monocytes/macrophages in the pathogenesis of cerebral ischemia and stroke. Stroke. 1992;23:1367–1379. doi: 10.1161/01.str.23.9.1367. [DOI] [PubMed] [Google Scholar]
  37. LEHMBERG J., BECK J., BAETHMANN A., UHL E. Influence of the bradykinin B2-receptor-antagonist CP-0597 on the cerebral microcirculation and outcome on global cerebral ischemia. Cerebrovasc. Dis. 1998;8 Suppl:76. doi: 10.1007/978-3-7091-6346-7_8. [DOI] [PubMed] [Google Scholar]
  38. LEHMBERG J., BECK J., BAETHMANN A., UHL E. Influence of the bradykinin B1/B2-receptor-antagonist B 9430 on the cerebral microcirculation and outcome of gerbils from global cerebral ischemia. Acta Neurochir. Suppl. 2000;76:39–41. doi: 10.1007/978-3-7091-6346-7_8. [DOI] [PubMed] [Google Scholar]
  39. LIAO S.L., CHEN W.Y., RAUNG S.L., KUO J.S., CHEN C.J. Association of immune responses and ischemic brain infarction in rat. Neuroreport. 2001;12:1943–1947. doi: 10.1097/00001756-200107030-00034. [DOI] [PubMed] [Google Scholar]
  40. MAIER-HAUFF K., BAETHMANN A.J., LANGE M., SCHURER L., UNTERBERG A. The kallikrein–kinin system as mediator in vasogenic brain edema. Part 2: studies on kinin formation in focal and perifocal brain tissue. J. Neurosurg. 1984;61:97–106. doi: 10.3171/jns.1984.61.1.0097. [DOI] [PubMed] [Google Scholar]
  41. MAKEVNINA L.G., LOMOVA I.P., ZUBKOVYUN, SEMENYUTIN V.B. Kininogen consumption in cerebral circulation of humans during brain ischemia and postischemic reperfusion. Braz. J. Med. Biol. Res. 1994;27:1955–1963. [PubMed] [Google Scholar]
  42. MARCEAU F. Kinin B1 receptors: a review. Immunopharmacology. 1995;30:1–26. doi: 10.1016/0162-3109(95)00011-h. [DOI] [PubMed] [Google Scholar]
  43. MATSUO Y., KIHARA T., IKEDA M., NINOMIYA M., ONODERA H., KOGURE K. Role of neutrophils in radical production during ischemia and reperfusion of the rat brain: effect of neutrophil depletion on extracellular ascorbyl radical formation. J. Cereb. Blood Flow Metab. 1995;15:941–947. doi: 10.1038/jcbfm.1995.119. [DOI] [PubMed] [Google Scholar]
  44. MCINTYRE T.M., ZIMMERMAN G.A., SATOH K., PRESCOTT S.M. Cultured endothelial cells synthesize both platelet-activating factor and prostacyclin in response to histamine, bradykinin, and adenosine triphosphate. J. Clin. Invest. 1985;76:271–280. doi: 10.1172/JCI111957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. PARPURA V., BASARSKY T.A., LIU F., JEFTINIJA K., JEFTINIJA S., HAYDON P.G. Glutamate-mediated astrocyte-neuron signalling. Nature. 1994;369:744–747. doi: 10.1038/369744a0. [DOI] [PubMed] [Google Scholar]
  46. PLESNILA N., SCHULZ J., STOFFEL M., ERISKAT J., PRUNEAU D., BAETHMANN A. Role of bradykinin B2 receptors in the formation of vasogenic brain edema in rats. J. Neurotrauma. 2001;18:1049–1058. doi: 10.1089/08977150152693746. [DOI] [PubMed] [Google Scholar]
  47. PRUNEAU D., CHORNY I., BENKOVITZ V., ARTRU A., ROITBLAT L., SHAPIRA Y. Effect of LF 16-0687 MS, a new nonpeptide bradykinin B2 receptor antagonist, in a rat model of closed head trauma. J. Neurotrauma. 1999a;16:1057–1065. doi: 10.1089/neu.1999.16.1057. [DOI] [PubMed] [Google Scholar]
  48. PRUNEAU D., PAQUET J.L., LUCCARINI J.M., DEFRENE E., FOUCHET C., FRANCK R.M., LOILLIER B., ROBERT C., BELICHARD P., DUCLOS H., CREMERS B., DODEY P. Pharmacological profile of LF 16-0687, a new potent non-peptide bradykinin B2 receptor antagonist. Immunopharmacology. 1999b;43:187–194. doi: 10.1016/s0162-3109(99)00128-9. [DOI] [PubMed] [Google Scholar]
  49. PRUNEAU D., PAQUET J.L., LUCCARINI J.M., DEFRENE E., FOUCHET C., FRANCK R.M., LOILLIER B., ROBERT C. LF 16-0687, a new potent non-peptide bradykinin B2 receptor antagonist. Restor. Neural. Neurosci. 2000;16:177. doi: 10.1016/s0162-3109(99)00128-9. [DOI] [PubMed] [Google Scholar]
  50. RACHINSKY M., PRUNEAU D., ARTRU A.A., KAPULER V., ALONCHIN A., SMOLANEZKI Y., SHAPIRA Y. The importance of kinin antagonist treatment timing in closed head trauma. J. Trauma. 2001;51:944–948. doi: 10.1097/00005373-200111000-00020. [DOI] [PubMed] [Google Scholar]
  51. RAIDOO D.M., BHOOLA K.D. Kinin receptors on human neurones. J. Neuroimmunol. 1997;77:39–44. doi: 10.1016/s0165-5728(97)00048-9. [DOI] [PubMed] [Google Scholar]
  52. RAIDOO D.M., BHOOLA K.D. Pathophysiology of the kallikrein–kinin system in mammalian nervous tissue. Pharmacol. Ther. 1998;79:105–127. doi: 10.1016/s0163-7258(98)00011-4. [DOI] [PubMed] [Google Scholar]
  53. RAIDOO D.M., RAMSAROOP R., NAIDOO S., BHOOLA K.D. Regional distribution of tissue kallikrein in the human brain. Immunopharmacology. 1996;32:39–47. doi: 10.1016/0162-3109(96)00007-0. [DOI] [PubMed] [Google Scholar]
  54. REGOLI D., NSA ALLOGHO S., RIZZI A., GOBEIL F.J. Bradykinin receptors and their antagonists. Eur. J. Pharmacol. 1998;348:1–10. doi: 10.1016/s0014-2999(98)00165-4. [DOI] [PubMed] [Google Scholar]
  55. RELTON J.K., BECKEY V.E., HANSON W.L., WHALLEY E.T. CP-0597, a selective bradykinin B2 receptor antagonist, inhibits brain injury in a rat model of reversible middle cerebral artery occlusion. Stroke. 1997;28:1430–1436. doi: 10.1161/01.str.28.7.1430. [DOI] [PubMed] [Google Scholar]
  56. ROSENBLUM W.I. Hydroxyl radical mediates the endothelium-dependent relaxation produced by bradykinin in mouse cerebral arterioles. Circ. Res. 1987;61:601–603. doi: 10.1161/01.res.61.4.601. [DOI] [PubMed] [Google Scholar]
  57. ROZSA Z., RUSSELL J., SALTER J., GRANGER D.N. Bradykinin mediates P-selectin expression in rat gastrointestinal microcirculation. Gastroenterology. 1998;114:A411. [Google Scholar]
  58. RYDH-RINDER M., KEREKES N., SVENSSON M., HOKFELT T. Glutamate release from adult primary sensory neurons in culture is modulated by growth factors. Regul. Pept. 2001;102:69–79. doi: 10.1016/s0167-0115(01)00297-x. [DOI] [PubMed] [Google Scholar]
  59. SATOH K., YOSHIDA H., IMAIZUMI T.A., KOYAMA M., TAKAMATSU S. Production of platelet-activating factor by porcine brain microvascular endothelial cells in culture. Thromb. Haemost. 1995;74:1335–1339. [PubMed] [Google Scholar]
  60. SCHULZ J., PLESNILA N., ERISKAT J., STOFFEL M., PRUNEAU D., BAETHMANN A. LF16-0687 a novel non-peptide bradykinin B2 receptor antagonist reduces vasogenic brain edema from a focal lesion in rats. Acta Neurochir. Suppl. 2000;76:137–139. doi: 10.1007/978-3-7091-6346-7_28. [DOI] [PubMed] [Google Scholar]
  61. SCHWANINGER M., SALLMANN S., PETERSEN N., SCHNEIDER A., PRINZ S., LIBERMANN T.A., SPRANGER M. Bradykinin induces interleukin-6 expression in astrocytes through activation of nuclear factor-kappaB. J. Neurochem. 1999;73:1461–1466. doi: 10.1046/j.1471-4159.1999.0731461.x. [DOI] [PubMed] [Google Scholar]
  62. SHIGEMATSU S., ISHIDA S., GUTE D.C., KORTHUIS R.J. Concentration-dependent effects of bradykinin on leukocyte recruitment and venular hemodynamics in rat mesentery. Am. J. Physiol. 1999;277:H152–H160. doi: 10.1152/ajpheart.1999.277.1.H152. [DOI] [PubMed] [Google Scholar]
  63. SOBEY C.G., HEISTAD D.D., FARACI F.M. Mechanisms of bradykinin-induced cerebral vasodilatation in rats. Evidence that reactive oxygen species activate K+ channels. Stroke. 1997;28:2290–2295. doi: 10.1161/01.str.28.11.2290. [DOI] [PubMed] [Google Scholar]
  64. SORNAS R., OSTLUND H., MULLER R. Cerebrospinal fluid cytology after stroke. Arch. Neurol. 1972;26:489–501. doi: 10.1001/archneur.1972.00490120029002. [DOI] [PubMed] [Google Scholar]
  65. STOVER J.F., DOHSE N.K., UNTERBERG A.W. Bradykinin 2 receptor antagonist LF 16-0687Ms reduces posttraumatic brain edema. Acta Neurochir. Suppl. 2000;76:171–175. doi: 10.1007/978-3-7091-6346-7_34. [DOI] [PubMed] [Google Scholar]
  66. UNTERBERG A., BAETHMANN A.J. The kallikrein–kinin system as mediator in vasogenic brain edema. Part 1: cerebral exposure to bradykinin and plasma. J. Neurosurg. 1984;61:87–96. doi: 10.3171/jns.1984.61.1.0087. [DOI] [PubMed] [Google Scholar]
  67. UNTERBERG A., DAUTERMANN C., BAETHMANN A.J., MULLERESTER W. The kallikrein–kinin system as mediator in vasogenic brain edema. Part 3: inhibition of the kallikrein–kinin system in traumatic brain swelling. J. Neurosurg. 1986;64:269–276. doi: 10.3171/jns.1986.64.2.0269. [DOI] [PubMed] [Google Scholar]
  68. UNTERBERG A., WAHL M., BAETHMANN A. Effects of bradykinin on permeability and diameter of pial vessels in vivo. J. Cereb. Blood Flow Metab. 1984;4:574–585. doi: 10.1038/jcbfm.1984.82. [DOI] [PubMed] [Google Scholar]
  69. VERRECCHIA C., MESENGE C., PLOTKINE M. Neuroprotective effect of LF 16-0687 Ms, a bradykinin B2 receptor antagonist, on a model of traumatic brain injury in mice. Restor. Neurol. Neurosci. 2000;16:177–178. [Google Scholar]
  70. WAGNER S., KALB P., LUKOSAVA M., HILGENFELDT U., SCHWANINGER M. Activation of the tissue kallikrein–kinin system in stroke. J. Neurol. Sci. 2002;202:75–76. doi: 10.1016/s0022-510x(02)00208-3. [DOI] [PubMed] [Google Scholar]
  71. WAHL M., WHALLEY E.T., UNTERBERG A., SCHILLING L., PARSONS A.A., BAETHMANN A., YOUNG A.R. Vasomotor and permeability effects of bradykinin in the cerebral microcirculation. Immunopharmacology. 1996;33:257–263. doi: 10.1016/0162-3109(96)00068-9. [DOI] [PubMed] [Google Scholar]
  72. WALKER K., PERKINS M., DRAY A. Kinins and kinin receptors in the nervous system. Neurochem. Int. 1995;26:1–26. doi: 10.1016/0197-0186(94)00114-a. [DOI] [PubMed] [Google Scholar]
  73. WHALLEY E.T., WAHL M. Analysis of bradykinin receptor mediating relaxation of cat cerebral arteries in vivo and in vitro. Naunyn Schmiedebergs Arch. Pharmacol. 1983;323:66–71. doi: 10.1007/BF00498830. [DOI] [PubMed] [Google Scholar]
  74. WIRTH K., BREIPOHL G., STECHL J., KNOLLE J., HENKE S., SCHOLKENS B. DesArg9-D-Arg[Hyp3,Thi5,D-Tic7, Oic8]bradykinin (desArg10-[Hoe140]) is a potent bradykinin B1 receptor antagonist. Eur. J. Pharmacol. 1991;205:217–218. doi: 10.1016/0014-2999(91)90824-a. [DOI] [PubMed] [Google Scholar]
  75. ZAUSINGER S., LUMENTA D., PRUNEAU D., SCHMID-ELSAESSER R., PLESNILA N., BAETHMANN A. Effects of LF 16-0687 Ms, a bradykinin B(2) receptor antagonist, on brain edema formation and tissue damage in a rat model of temporary focal cerebral ischemia. Brain Res. 2002;950:268–278. doi: 10.1016/s0006-8993(02)03053-6. [DOI] [PubMed] [Google Scholar]

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

RESOURCES