Skip to main content
Erschienen in: Experimental & Translational Stroke Medicine 1/2015

Open Access 01.12.2015 | Research

Docosahexaenoic acid improves behavior and attenuates blood–brain barrier injury induced by focal cerebral ischemia in rats

verfasst von: Sung-Ha Hong, Larissa Khoutorova, Nicolas G Bazan, Ludmila Belayev

Erschienen in: Experimental & Translational Stroke Medicine | Ausgabe 1/2015

Abstract

Background

Ischemic brain injury disrupts the blood–brain barrier (BBB) and then triggers a cascade of events, leading to edema formation, secondary brain injury and poor neurological outcomes. Recently, we have shown that docosahexaenoic acid (DHA) improves functional and histological outcomes following experimental stroke. However, little is known about the effect of DHA on BBB dysfunction after cerebral ischemia-reperfusion injury. The present study was designed to determine whether DHA protects against BBB disruption after focal cerebral ischemia in rats.

Methods

Physiologically-controlled SD rats received 2 h middle cerebral artery occlusion (MCAo). DHA (5 mg/kg) or vehicle (saline) was administered I.V. at 3 h after onset of MCAo. Fluorometric quantitation of Evans Blue dye (EB) was performed in eight brain regions at 6 h, 24 h or 72 h after MCAo. Fluorescein isothiocynate (FITC) - dextran leakage and histopathology was evaluated on day 3 after stroke.

Results

Physiological variables were stable and showed no significant differences between groups. DHA improved neurological deficits at 24 h, 48 h and 72 h and decreased EB extravasation in the ischemic hemisphere at 6 h (by 30%), 24 h (by 48%) and 72 h (by 38%). In addition, EB extravasation was decreased by DHA in the cortex and total hemisphere as well. FITC-dextran leakage was reduced by DHA treatment on day 3 by 68% compared to the saline group. DHA treatment attenuated cortical (by 50%) and total infarct volume (by 38%) compared to vehicle-treated rats on day 3 after stroke.

Conclusions

DHA therapy diminishes BBB damage accompanied with the acceleration of behavioral recovery and attenuation of the infarct volume. It is reasonable to propose that DHA has the potential for treating focal ischemic stroke in the clinical setting.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Author contributions to the study and manuscript preparation include the following. Conception and design: LB, NGB, SH. Acquisition of data: SH, LK. Analysis and interpretation: all authors. Drafting the article: SH, LB. Critically revising the article: all authors. Reviewed submitted version of manuscript: all authors. Approved the final version of the manuscript on behalf of all authors: LB. Statistical analysis: SH, LB. Administrative/technical/material support: LB. Study supervision: SH, NGB, LB.

Introduction

Cerebral ischemia can cause blood–brain barrier (BBB) disruption and increased cerebral vascular permeability, leading to the formation of brain edema [1,2]. Clinically, BBB disruption occurs in more than one third of stroke patients and is associated with poor outcomes and lower survival rates following stroke [3,4]. In animal models of cerebral ischemia–reperfusion, BBB disruption is believed to be biphasic, which implies an early first opening (at 3–6 h) followed by a refractory period when the BBB is closed, and a delayed second opening (at 48–72 h) [5,6]. Since cerebral ischemia and BBB damage are closely correlated, so approaches for protecting BBB integrity and reducing BBB permeability could help elucidate the pathophysiological mechanism in brain ischemia, leading to novel therapies as well as providing a mechanistic roadmap to evaluate efficacy of treatment [7].
Recently, we have shown that docosahexaenoic acid (DHA; 22:6, n-3), a member of the essential omega-3 fatty acid family, improves functional and histological outcomes in experimental stroke [8,9]. DHA is highly concentrated in the brain and is involved in cognition and other brain functions [10,11]. DHA is the precursor of the docosanoids, and neuroprotectin D1 (NPD1; 10R,17S-dihydroxy-docosa-4Z,7Z,11E,15E,19Z hexaenoic acid), being the first identified member of this group of mediators, inhibits oxidative stress-induced proinflammatory gene expression and promotes cell survival [12,13]. DHA is involved in excitable membrane function, neuronal signaling and has been implicated in neuroprotection [8,9]. DHA administration improves histological and neurological outcome following focal cerebral ischemia in rats when treatment is initiated as late as 5 hours after ischemia onset [9].
Many methods for assessing the disruption of the blood–brain barrier have been adopted in experimental models [14]. Small (EB, 68 kDa) and large (FITC-dextran, 2000 kDa) tracers were used here to evaluate BBB permeability. Although distribution of these two markers are similar (primarily, if not exclusively, extracellular), intracellular and extracellular uptake of EB was reported [15]. Serum albumin-bound EB crosses the easily broken BBB, and its regional fluorometric quantification provides the precise assessment of the degree of BBB disruption in ischemic injury [16]. Subsequently, the presence or absence of highly localized extracellular FITC-dextran leakage has been used to identify brain regions undergoing vascular disruption and to label the severely damaged BBB [15,17]. The present study was designed to determine whether DHA protects against BBB disruption after focal cerebral ischemia in rats.

Materials and methods

Animal preparation

The present study was conducted in accordance with the NIH guidelines for the care and use of animals in research and under protocols approved by the Institutional Animal Care and Use Committee of the Louisiana State University Health Sciences Center, New Orleans. Male Sprague–Dawley (SD) rats (34 months-old); Charles River Laboratory, Wilmington, MA) were fasted overnight with free access to water prior to the surgical procedure. Anesthesia was induced by the inhalation of 3% isoflurane in 70% NO and 30% O2 mixed gases, and then maintained with 1% isoflurane in the same mixed gases during the procedure. Orally-intubated animals were mechanically ventilated after the immobilization by injection of pancronium bromide (0.5 mg/kg, I.V.). The catheters were implanted into the right femoral artery and vein for the blood sampling and infusion of drug, EB and FITC-dextran. Serial analyses of arterial blood gases and plasma glucose were conducted before and during surgical procedure. Rectal (CMA/150 Temperature Controller, CMA/Microdialysis AB, Stockholm, Sweden) and cranial (temporalis muscle; Omega Engineering, Stamford, CT) temperatures were closely monitored before, during and after MCAo. Rectal temperature and body weight were monitored daily and before sacrifice during the survival period.

Transient Middle Cerebral Artery occlusion (MCAo)

The right MCA was occluded for 2 h by intraluminal filament, as we described previously [18]. Briefly, the right common carotid artery (CCA) and external carotid artery (ECA) were exposed through midline neck incision, and then completely isolated from the surrounding nerves. The occipital branches of the ECA and pterygopalatine artery were ligated. A 4-cm of 3–0 nylon filament, coated with poly-L-lysine was advanced to the origin of MCA through the proximal ECA via internal carotid artery. The filament was inserted 20 to 22 mm from the bifurcation of the CCA, according to the animal’s body weight. The neck incision was then closed and rats were returned to their cages. After 2 h of MCAo, the rats were re-anesthetized with the same anesthetic combination and the intraluminal filament was gently removed. The animals were allowed to survive for different times according the experimental protocol with free access to water and food.

Behavioral tests

Behavioral tests were conducted before, during MCAo (at 60 min), and then at 6, 24, 48 and 72 h after MCAo by an investigator who was blinded to the experimental groups. The battery consisted of two tests, (1) postural reflex to examine the upper body posture when the rat was suspended by tail, and (2) forelimb placing test to assess the forelimb placing responses to visual, tactile and proprioceptive stimuli [18]. Neurologic function was graded on a scale of 0 to 12 (normal =0, maximal deficits = 12), as we described previously [18]. The severity of stroke injury was assessed by behavioral examination of each rat at 60 min after onset of MCAo. Rats that did not demonstrate high-grade contralateral deficit (score, 10–11) were excluded from further study. Two animals were excluded for this reason.

Treatment groups

Animals were randomly assigned to DHA (5 mg/kg, Cayman, Ann Arbor, MI) or vehicle (0.9% saline) treatment groups. All treatments were administered intravenously at 3 h after the onset of MCAo at a constant rate over 3 min using an infusion pump. The DHA dose–response study in rats with transient focal cerebral ischemia showed that a 5 mg/kg dosage was highly neuroprotective [8]; thus this dose was applied in this study.

Study protocols

Two protocols were used. In series 1, BBB leakage was studied by EB dye. The neurological status was evaluated at 6, 24 and 72 h followed by EB measurement at 6, 24 or 72 h (n = 8-12 rats per group). In series 2, BBB leakage was assessed by FITC-dextran. Behavioral score was measured on days 1, 2 and 3. On Day 3, FITC-dextran leakage was analyzed followed by histopathology (n = 5-7 rats per groups). EB, FITC and histopathological analyses were conducted by an investigator who was blinded to the experimental groups.

Evaluation of BBB leakage by Evans Blue (EB) dye

The integrity of the BBB was investigated using EB extravasation [19]. EB (2%, 4 ml/kg, in saline, Sigma-Aldrich, MO) was injected intravenously either at 5 h, 23 h or 71 h after MCAo. After 1 h of EB circulation, rats were anesthetized with isoflurane and were transcardially perfused with saline until colorless fluid was observed from the right atrium. After decapitation, the removed brains were divided into two coronal blocks that included the bregma levels + 0.3 and – 1.8 mm. Coronal blocks were divided into right and left hemisphere, and were cut into eight regions for local measurement of EB dye (see Figure 1). EB extravasation into the brain was measured as we previously described [19]. Samples were weighed and placed in 50% of tricholoroacetic acid. Following homogenization and centrifugation at 10000 RPM for 20 min, extracted EB dye was diluted with ethanol (1:3) and its fluorescence was determined (excitation at 620 nm and emission at 680 nm) by a Spectra Max M5e (Molecular Devices, CA). External standards were prepared in a range of 100 – 500 ng/ml in the same solvent. The tissue content of EB was calculated based on the linear standard curve and expressed as μg of EB/g of tissue.

Evaluation of BBB leakage by fluorescein isothiocyanate dextran (FITC-dextran)

FITC-dextran (2000 kDa, 1 ml, 50 mg/ml, in saline, Sigma-Aldrich, St. Louis, MO) was intravenously injected at 72 h after MCAo [20,21]. After 1 min of circulation, brains were removed, fixed in 4% paraformaldehyde for 24 h, cryoprotected in 30% sucrose for 48 h and then coronally sectioned (20 μm-thickness) using the cryostat. Mosaic fluorescent images of the sections were obtained at six bregma levels (+2.7, + 1.2, − 0.3, − 1.3, − 1.8 and - 3.8 mm) with fluorescence motorized microscope (BX61VS, Olympus, Japan) at a 10 X magnification objective lens. Regional FITC-dextran fluorescent intensity in the ipsi- and contra-lateral cortex, subcortex and hemisphere were measured by Image J (National Institute of Health, Bethesda, MD). Optical density was measured by applying the following formula: (Right fluorescent intensity – Left fluorescent intensity)/Left fluorescent intensity × 100.

Histopathology

Histopathology was performed after the evaluation of BBB leakage by FITC-dextran at 72 h as we previously described [18]. Adjacent coronal sections (20 μm-thickness) from FITC-dextran study were stained with thionine (Nissl). Stained sections at nine standardized bregma levels were digitized, and then cortical, subcortical infarct areas were outlined and measured as well as the both hemispheres using MCID core imaging software (InterFocus Imaging Ltd., Cambridge, England). Infarct volume was quantified as an integrated product of cross-sectional infarct areas and intersectional distance and corrected for brain swelling. Computer generated mosaic images of stained sections were obtained by motorized microscope (BX61VS, Olympus, Japan) at a 10 X magnification objective lens.

Statistical analysis

Repeated-measures analysis of variance (ANOVA), followed by Bonferroni tests, was used for multiple-group comparisons. Two-tailed Student’s T-test was used for two-group comparisons. A value of p < 0.05 was regarded as statistically significant. Values are presented as means ± SEM. Statistical analyses were conducted by Prism 5 (Graph Pad Software, Inc., La Jolla, CA).

Results

There were no significant differences in the rectal and cranial (temporalis muscle) temperatures, body weight, arterial blood gases, plasma glucose and hematocrit among the groups (Table 1). Four animals died during the experiment: 3 rats in the saline group (on days 1, 2 and 3) and 1 rat in the DHA group (on day 1).
Table 1
Physiological variables
 
Evans-Blue Study
FITC-dexran Study
 
Saline - 6 h
DHA - 6 h
Saline - 24 h
DHA - 24 h
Saline - 72 h
DHA - 72 h
Saline - 72 h
DHA - 72 h
 
(n=9)
(n=9)
(n=9)
(n=8)
(n=12)
(n=12)
(n=7)
(n=6)
Before MCAo (15min)
        
Cranial Temperature (°C)
37.2 ± 0.1
37.4 ± 0.1
37.1 ± 0.1
37.1 ± 0.2
37.4 ± 0.1
37.2 ± 0.1
37.2 ± 0.2
37.2 ± 0.2
Rectal Temperature (°C)
37.2 ± 0.1
37.3 ± 0.1
37.2 ± 0.1
37.3 ± 0.2
37.3 ± 0.1
37.2 ± 0.1
37.0 ± 0.1
37.0 ± 0.2
pH
7.49 ± 0.0
7.53 ± 0.0
7.50 ± 0.1
7.47 ± 0.0
7.47 ± 0.0
7.53 ± 0.0
7.4 ± 0.1
7.5 ± 0.0
pO2 (mmHg)
113.3 ± 4.1
120.7 ± 4.6
113.8 ± 6.8
114.9 ± 5.0
119.8 ± 3.7
120.4 ± 4.2
126.3 ± 8.5
113.7 ± 0.7
pCO2 (mmHg)
39.3 ± 0.4
38.7 ± 0.7
40.1 ± 0.6
38.4 ± 0.8
39.0 ± 0.5
38.1 ± 0.6
39.1 ± 0.9
38.3 ± 5.1
Plasma Glucose (mg/dL)
167.1 ± 12.7
171.2 ± 8.8
166.3 ± 9.9
194.9 ± 12.7
171.1 ± 13.0
161.7 ± 8.0
196.9 ± 6.1
204.7 ± 13.2
Hematocrit (%)
45.7 ± 12.7
45.8 ± 0.9
44.4 ± 0.8
45.8 ± 0.5
57.2 ± 11.7
45.8 ± 0.6
45.6 ± 0.6
45.8 ± 1.0
During MCAo (15 min)
        
Cranial Temprature (°C)
37.6 ± 0.1
37.6 ± 0.1
37.4 ± 0.1
37.3 ± 0.1
37.7 ± 0.1
37.2 ± 0.1
37.6 ± 0.2
37.6 ± 0.1
Rectal Temperature (°C)
37.6 ± 0.1
37.4 ± 0.1
37.5 ± 0.1
37.5 ± 0.1
37.7 ± 0.1
37.5 ± 0.1
37.4 ± 0.1
37.3 ± 0.1
pH
7.51 ± 0.1
7.51 ± 0.0
7.46 ± 0.0
7.48 ± 0.0
7.46 ± 0.0
7.52 ± 0.0
7.5 ± 0.1
7.6 ± 0.1
pO2 (mmHg)
109.7 ± 4.4
114.0 ± 4.3
106.0 ± 4.8
106.9 ± 2.7
109.3 ± 3.4
114.6 ± 4.0
108.1 ± 4.9
105.0 ± 4.3
pCO2 (mmHg)
39.8 ± 0.3
39.8 ± 0.8
39.6 ± 0.6
39.0 ± 0.4
38.8 ± 0.4
38.5 ± 0.5
39.6 ± 0.7
39.2 ± 0.8
Plasma Glucose (mg/dL)
167.0 ± 10.6
164.0 ± 4.4
164.0 ± 4.6
193.3 ± 13.0
173.2 ± 15.8
155.0 ± 5.1
185.7 ± 6.9
197.3 ± 12.5
Hematocrit (%)
45.2 ± 0.6
46.3 ± 0.1
44.3 ± 0.8
45.6 ± 0.3
57.2 ± 11.7
46.0 ± 0.6
44.7 ± 0.8
46.7 ± 1.0
After treatment (15 min)
        
Cranial Temprature (°C)
37.4 ± 0.1
37.1 ± 0.1
37.2 ± 0.2
37.1 ± 0.2
37.2 ± 0.2
37.0 ± 0.2
36.7 ± 0.2
36.9 ± 0.2
Rectal Temperature (°C)
37.7 ± 0.1
37.4 ± 0.2
37.8 ± 0.2
37.9 ± 0.2
37.7 ± 0.2
37.5 ± 0.1
36.9 ± 0.2
37.4 ± 0.2
After treatment (24 h)
        
Rectal Temperature (°C)
  
38.2 ± 0.3
37.9 ± 0.3
37.4 ± 0.9
38.0 ± 0.2
37.8 ± 0.3
38.1 ± 0.4
Body Weight (g)
  
264.2 ± 5
268.3 ± 6.3
302.1 ± 6.9
275.5 ± 21.9
299.4 ± 17.0
287.5 ± 19.5
After treatment (48 h)
        
Rectal Temperature (°C)
    
37.3 ± 0.1
36.9 ± 0.3
37.6 ± 0.2
37.5 ± 0.2
Body Weight (g)
    
287.4 ± 8.3
266.8 ± 21.8
288.3 ± 15.4
279.7 ± 23.4
After treatment (72 h)
        
Rectal Temperature (°C)
    
37.0 ± 0.2
36.6 ± 0.4
36.2 ± 0.8
36.9 ± 0.5
Body Weight (g)
    
274.3 ± 10.5
258.8 ± 21.6
281.3 ± 15.8
277.5 ± 26.6

Evans Blue study

Total neurologic scores were improved in the 24 and 72 h DHA groups compared to the corresponding vehicle-treated groups (Figure 2). There were no differences in the behavioral score between DHA and saline 6 h groups (Figure 2). Tissue leakage of EB was clearly related to cerebral ischemia and presented in Figure 3. Tissue leakage of EB was clearly related to cerebral ischemia, as presented in Figure 3. The brains of the saline-treated rats showed intense blue staining at 6 and 72 h, which were observed predominantly in the ventromedial striatum and the cortex ipsilateral to the MCAo (Figure 3). On the contrary, in the DHA-treated rats, cortical and subcortical regions showed only faint blue staining (Figure 3). DHA treatment significantly reduced EB dye extravasation (Figure 4) into the cortex (at 72 h) and subcortex (at 6, 24 and 72 h). In addition, EB content of the right hemisphere and total EB from the whole brain (Figure 4) were also significantly decreased in all DHA-treated groups compared with the corresponding vehicle group at 6, 24 and 72 h.

FITC-dextran study

DHA-treated rats displayed a significantly improved total neurologic score at 24, 48 and 72 h after MCAo compared to the vehicle group (Figure 5). Representative fluorescent images of brains at the six bregma levels from the DHA or vehicle rats at 72 h is presented in Figure 6. Intensive FITC–dextran leakage was observed in the whole ischemic hemisphere, cortex and subcortex in the saline-treated rats. By contrast, FITC-dextran leakage was dramatically reduced by DHA treatment from the microvessels of the ischemic area in both the cortex and subcortex (Figure 6). The alteration of FITC-dextran leakage was measured by the relative FITC-dextran fluorescent intensity to the comparable regions of non-ischemic hemisphere. DHA treatment significantly decreased the relative fluorescence intensity at multiple bregma levels in the cortex and the entire hemisphere compared to the vehicle group (Figure 7A). In addition, total relative fluorescent intensity in the cortex and hemisphere (Figure 7B) was reduced by DHA treatment compared to the vehicle group. Treatment with DHA demonstrated a consistent smaller lesion involving cortical and subcortical regions in the right hemisphere at 72 h after MCAo compared to vehicle group (Figure 8A). DHA treated group significantly reduced the cortical and total infarct areas at multiple bregma levels compared to vehicle treated group (Figure 8B). Moreover, total and cortical infarct volumes in the DHA-treated rats were remarkably decreased compared to saline-treated rats (Figure 8C).

Discussion

In the present study, we have established a protective effect of DHA treatment against BBB disruption after experimental focal cerebral ischemia. Post-ischemic administration of DHA robustly protected against ischemia-induced BBB disruption, as evidenced by reduced EB extravasation and FITC-dextran leakage, improved the neurological deficits and attenuated infarct volume. Moreover, there were no adverse behavioral side effects observed with DHA administration. Our results suggest that this approach might be adaptable as a therapeutic avenue.
Cerebral ischemia results in rapid accumulation of free fatty acids, including arachidonic acid (ArAc; 20:4) and DHA [10,11]. These fatty acids are released from membrane phospholipids. Both fatty acids are derived from dietary essential fatty acids; however, only DHA, from the omega-3 polyunsaturated fatty acyl chain, is concentrated in phospholipids of various cells of the brain and retina [22]. Synaptic membranes and photoreceptors share the highest content of DHA of all cell membranes. DHA, one of the main structural lipids in the mammalian brain, plays crucial roles in the development and function of brain neurons [10,12]. It is involved in memory formation, promotes neurogenesis both in vitro and in vivo; and has been implicated in neuroprotection [8,9,23-25]. In vivo, the active DHA supply to the brain (provided by the liver through the blood stream) is necessary for cell development and function. It also may play a critical role in conditions where, due to enhanced oxidative stress, the polyunsaturated fatty acyl chains of membrane phospholipids are decreased as a consequence of lipid peroxidation, as occurs in aging, retinal degenerations, and neurodegenerations such as Alzheimer disease [26,27]. In ischemia, there is also loss of brain DHA due to breakdown of enhanced phospholipase A2-activated DHA-containing phospholipids [23].
Beneficial effects of DHA in preventing and ameliorating stroke damage have been attributed to generation of the stereospecific derivative, neuroprotectin D1 (NPD1), which is a key survival signaling event leading to neuroprotection [22,28,29]. Recently, we have identified NPD1 following cerebral ischemia-reperfusion in the mouse [28]. NPD1 was found to serve an endogenous neuroprotective role by inhibiting apoptotic DNA damage, upregulating anti-apoptotic and downregulating pro-apoptotic proteins, and also binding toxic peroxides [26,28,29]. We demonstrated that NPD1 synthesis takes place in the ipsilateral side of ischemic brain and peaks at 8 h of reperfusion, and then decreases and is still detectable 25 h after reperfusion [24,28]. Although DHA in the brain can produce NPD1, additional DHA from acute administration is beneficial because lipidomic analysis shows that it potentiates NPD1 synthesis in the penumbra [9,30].
A variety of biologic effects of DHA have been demonstrated with fish or fish oil supplements in humans [31]. They have been found to reduce cholesterol, lower blood pressure, block clot-promoting platelet activation, prevent heart arrhythmias, prevent vascular inflammation and improve vascular function, and protect the heart muscle following a heart attack [32,33]. Epidemiologic studies provide evidence for a beneficial effect of omega-3 fatty acids on manifestations of coronary heart disease and ischemic stroke, particularly with respect to sudden cardiac death in patients with established disease [34]. Clinically important anti-inflammatory effects in humans are further suggested by trials demonstrating benefits of omega-3 fatty acids in rheumatoid arthritis, psoriasis, asthma, inflammatory bowel disorders, osteoporosis, sepsis and cancer [31].
Vascular damage during cerebral ischemia occurs early and it progresses in a biphasic manner [5], contributing to the development of brain edema, hemorrhagic transformation, and worsened clinical outcome in stroke patients [35]. The BBB has a central role in stroke pathogenesis and it is a therapeutic target. Many drugs have been screened to protect BBB permeability integrity and reduce ischemic brain damage by targeting different mechanisms [36]. Unfortunately, so far there are no effective therapeutic interventions for BBB disruption [3]. DHA presence in vascular endothelial cells in the brain suggests that this fatty acid is incorporated into these cells from the systemic circulation [37-39]. DHA in the cell membrane endows fluidity and proper functioning [40]. Furthermore, DHA enriched phospholipids in cellular membranes influences the function of BBB and signaling properties of neurons through fostering a dynamic environment to membrane-associated receptors or enzymes of the BBB compartment [41]. Little is known about the effect of DHA on BBB dysfunction after cerebral ischemia-reperfusion injury. The beneficial effects of pre-treatment with DHA have been reported on BBB disruption, brain edema and inflammatory cell infiltration [42,43]. In contrast, one study showed that post-treatment with DHA (500 nmol/kg, i.p.at 60 min after 90 min of MCAo) exacerbated cerebral ischemic injury and increased BBB permeability in the ischemic cortex at 24 h after reperfusion [44]; note that the DHA dosage applied was 100 times less than in our study. Our study revealed that DHA administration decreased EB extravasation in the ischemic hemisphere at 6 h (by 30%), 24 h (by 48%) and 72 h (by 38%) compared to vehicle-treated rats. In addition, EB extravasation was decreased by DHA treatment in the cortex at 72 h (by 36%) and subcortex at 6 h (by 32%), at 24 h (by 45%) and at 72 h (by 40%) as well as the reduction into the entire brain at 6 h (by 30%), at 24 h (by 49%) and at 72 h (by 31%) compared to the corresponding vehicle groups. We also found that treatment with DHA also suppressed FITC-dextran leakage into the cortex (by 67%) and entire hemisphere (by 71%) compared to the saline-treated animals at 72 h after onset of stroke. In addition, DHA attenuated cortical (by 50%) and total infarct volumes (by 38%) compared to the saline-treated rats at 72 h. These results strongly demonstrate that DHA protects the severely-damaged BBB, accompanied by a relief of abnormal BBB permeability in the delayed ischemic reperfusion damage.
The exact mechanism by which DHA protects the BBB requires further investigation. The beneficial effect of DHA has been shown in a well-controlled animal model of MCAo. In the present study, and in recently published observations [9,13], we have employed a poly-l-lysine coated filament and have found that this technique leads to a 100% incidence of infarction following 2 h of MCAo, predictable size and location of the infarct, and a high degree of inter-animal reproducibility of infarct size (coefficient of variation 8%) [18]. We demonstrated that DHA did not have direct effects on body and cranial temperatures or arterial blood gases because these variables were carefully controlled and did not differ among groups.

Conclusions

Our results show that DHA is markedly neuroprotective in an experimental stroke model in rats, diminishing BBB damage accompanied with an improvement of behavioral functions and attenuation of the infarct volume. We therefore suggest that this agent offers great promise for developing therapies for cerebral ischemia in patients with acute ischemic stroke.

Acknowledgments

This study was supported by R01 NS046741 (NGB) and R01 NS065786 (LB) from the National Institute of Neurological Disorders and Stroke. We thank Daniela Anzola, Darlene Guillot and Ryan Labadens for their expert technical assistance.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Author contributions to the study and manuscript preparation include the following. Conception and design: LB, NGB, SH. Acquisition of data: SH, LK. Analysis and interpretation: all authors. Drafting the article: SH, LB. Critically revising the article: all authors. Reviewed submitted version of manuscript: all authors. Approved the final version of the manuscript on behalf of all authors: LB. Statistical analysis: SH, LB. Administrative/technical/material support: LB. Study supervision: SH, NGB, LB.
Literatur
1.
Zurück zum Zitat Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16:1–13.PubMedCrossRef Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16:1–13.PubMedCrossRef
4.
Zurück zum Zitat Warach S, Latour LL. Evidence of reperfusion injury, exacerbated by thrombolytic therapy, in human focal brain ischemia using a novel imaging marker of early blood–brain barrier disruption. Stroke. 2004;35(11 suppl 1):2659–61.PubMedCrossRef Warach S, Latour LL. Evidence of reperfusion injury, exacerbated by thrombolytic therapy, in human focal brain ischemia using a novel imaging marker of early blood–brain barrier disruption. Stroke. 2004;35(11 suppl 1):2659–61.PubMedCrossRef
5.
Zurück zum Zitat Belayev L, Busto R, Zhao W, Ginsberg MD. Quantitative evaluation of blood–brain barrier permeability following middle cerebral artery occlusion in rats. Brain Res. 1996;739:88–96.PubMedCrossRef Belayev L, Busto R, Zhao W, Ginsberg MD. Quantitative evaluation of blood–brain barrier permeability following middle cerebral artery occlusion in rats. Brain Res. 1996;739:88–96.PubMedCrossRef
6.
Zurück zum Zitat Huang ZG, Xue D, Preston E, Karbalai H, Buchan AM. Biphasic opening of the blood–brain barrier following transient focal ischemia: Effects of hypothermia. Can J Neurol Sci. 1999;26:298–304.PubMedCrossRef Huang ZG, Xue D, Preston E, Karbalai H, Buchan AM. Biphasic opening of the blood–brain barrier following transient focal ischemia: Effects of hypothermia. Can J Neurol Sci. 1999;26:298–304.PubMedCrossRef
7.
Zurück zum Zitat Lo EH, Moskowitz MA, Jacobs TP. Exciting, radical, suicidal: how brain cells die after stroke. Stroke. 2005;36:189–92.PubMedCrossRef Lo EH, Moskowitz MA, Jacobs TP. Exciting, radical, suicidal: how brain cells die after stroke. Stroke. 2005;36:189–92.PubMedCrossRef
8.
Zurück zum Zitat Belayev L, Khoutorova L, Atkins KD, Bazan NG. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Stroke. 2009;40:3121–6.PubMedCentralPubMedCrossRef Belayev L, Khoutorova L, Atkins KD, Bazan NG. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Stroke. 2009;40:3121–6.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Belayev L, Khoutorova L, Atkins KD, Eady TN, Hong S, Lu Y, et al. Docosahexaenoic acid therapy of experimental ischemic stroke. Transl Stroke Res. 2011;2:33–41.PubMedCentralPubMedCrossRef Belayev L, Khoutorova L, Atkins KD, Eady TN, Hong S, Lu Y, et al. Docosahexaenoic acid therapy of experimental ischemic stroke. Transl Stroke Res. 2011;2:33–41.PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat Bazan NG. Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care. 2007;10:136–41.PubMedCrossRef Bazan NG. Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care. 2007;10:136–41.PubMedCrossRef
11.
Zurück zum Zitat Bazan NG. The onset of brain injury and neurodegeneration triggers the synthesis of docosanoid neuroprotective signaling. Cell Mol Neurobiol. 2006;26:901–13.PubMedCrossRef Bazan NG. The onset of brain injury and neurodegeneration triggers the synthesis of docosanoid neuroprotective signaling. Cell Mol Neurobiol. 2006;26:901–13.PubMedCrossRef
12.
Zurück zum Zitat Bazan NG, Musto AE, Knott EJ. Endogenous signaling by omega-3 docosahexaenoic acid-derived mediators sustains homeostatic synaptic and circuitry integrity. Mol Neurobiol. 2011;44:216–22.PubMedCentralPubMedCrossRef Bazan NG, Musto AE, Knott EJ. Endogenous signaling by omega-3 docosahexaenoic acid-derived mediators sustains homeostatic synaptic and circuitry integrity. Mol Neurobiol. 2011;44:216–22.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Eady TN, Belayev L, Khoutorova L, Atkins KD, Zhang C, Bazan NG. Docosahexaenoic acid signaling modulates cell survival in experimental ischemic stroke penumbra and initiates long-term repair in young and aged rats. PLoS One. 2012;7(10):e46151.PubMedCentralPubMedCrossRef Eady TN, Belayev L, Khoutorova L, Atkins KD, Zhang C, Bazan NG. Docosahexaenoic acid signaling modulates cell survival in experimental ischemic stroke penumbra and initiates long-term repair in young and aged rats. PLoS One. 2012;7(10):e46151.PubMedCentralPubMedCrossRef
14.
Zurück zum Zitat Sandoval KE, Witt KA. Blood–brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis. 2008;32:200–19.PubMedCrossRef Sandoval KE, Witt KA. Blood–brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis. 2008;32:200–19.PubMedCrossRef
15.
Zurück zum Zitat Nagaraja TN, Keenan KA, Fenstermacher JD, Knight RA. Acute leakage patterns of fluorescent plasma flow markers after transient focal cerebral ischemia suggest large openings in blood–brain barrier. Microcirculation. 2008;15:1–14.PubMedCrossRef Nagaraja TN, Keenan KA, Fenstermacher JD, Knight RA. Acute leakage patterns of fluorescent plasma flow markers after transient focal cerebral ischemia suggest large openings in blood–brain barrier. Microcirculation. 2008;15:1–14.PubMedCrossRef
16.
Zurück zum Zitat Uyama O, Okamura N, Yanase M, Narita M, Kawabata K, Sugita M. Quantitative evaluation of vascular permeability in the gerbil brain after transient ischemia using evans blue fluorescence. J Cereb Blood Flow Metab. 1988;8:282–4.PubMedCrossRef Uyama O, Okamura N, Yanase M, Narita M, Kawabata K, Sugita M. Quantitative evaluation of vascular permeability in the gerbil brain after transient ischemia using evans blue fluorescence. J Cereb Blood Flow Metab. 1988;8:282–4.PubMedCrossRef
17.
Zurück zum Zitat Chen B, Friedman B, Cheng Q, Tsai P, Schim E, Kleinfeld D, et al. Severe blood–brain barrier disruption and surrounding tissue injury. Stroke. 2009;40:e666–74.PubMedCentralPubMedCrossRef Chen B, Friedman B, Cheng Q, Tsai P, Schim E, Kleinfeld D, et al. Severe blood–brain barrier disruption and surrounding tissue injury. Stroke. 2009;40:e666–74.PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Belayev L, Alonso OF, Busto R, Zhao W, Ginsberg MD. Middle cerebral artery occlusion in the rat by intraluminal suture. neurological and pathological evaluation of an improved model. Stroke. 1996;27:1616–22. discussion 1623.PubMedCrossRef Belayev L, Alonso OF, Busto R, Zhao W, Ginsberg MD. Middle cerebral artery occlusion in the rat by intraluminal suture. neurological and pathological evaluation of an improved model. Stroke. 1996;27:1616–22. discussion 1623.PubMedCrossRef
19.
Zurück zum Zitat Belayev L, Busto R, Ikeda M, Rubin LL, Kajiwara A, Morgan L, et al. Protection against blood–brain barrier disruption in focal cerebral ischemia by the type IV phosphodiesterase inhibitor BBB022: a quantitative study. Brain Res. 1998;787:277–85.PubMedCrossRef Belayev L, Busto R, Ikeda M, Rubin LL, Kajiwara A, Morgan L, et al. Protection against blood–brain barrier disruption in focal cerebral ischemia by the type IV phosphodiesterase inhibitor BBB022: a quantitative study. Brain Res. 1998;787:277–85.PubMedCrossRef
20.
Zurück zum Zitat Hoffmann A, Bredno J, Wendland M, Derugin N, Ohara P, Wintermark M. High and low molecular weight fluorescein isothiocyanate (FITC)-dextrans to assess blood–brain barrier disruption: technical considerations. Transl Stroke Res. 2011;2:106–11.PubMedCentralPubMedCrossRef Hoffmann A, Bredno J, Wendland M, Derugin N, Ohara P, Wintermark M. High and low molecular weight fluorescein isothiocyanate (FITC)-dextrans to assess blood–brain barrier disruption: technical considerations. Transl Stroke Res. 2011;2:106–11.PubMedCentralPubMedCrossRef
21.
Zurück zum Zitat Jin X, Liu J, Yang Y, Liu KJ, Yang Y, Liu W. Spatiotemporal evolution of blood brain barrier damage and tissue infarction within the first 3 h after ischemia onset. Neurobiol Dis. 2012;48:309–16.PubMedCrossRef Jin X, Liu J, Yang Y, Liu KJ, Yang Y, Liu W. Spatiotemporal evolution of blood brain barrier damage and tissue infarction within the first 3 h after ischemia onset. Neurobiol Dis. 2012;48:309–16.PubMedCrossRef
22.
Zurück zum Zitat Bazan NG. Cell survival matters: docosahexaenoic acid signaling, neuroprotection and photoreceptors. Trends Neurosci. 2006;29:263–71.PubMedCrossRef Bazan NG. Cell survival matters: docosahexaenoic acid signaling, neuroprotection and photoreceptors. Trends Neurosci. 2006;29:263–71.PubMedCrossRef
23.
Zurück zum Zitat Bazan NG. Neuroprotectin D1 (NPD1): a DHA-derived mediator that protects brain and retina against cell injury-induced oxidative stress. Brain Pathol. 2005;15:159–66.PubMedCrossRef Bazan NG. Neuroprotectin D1 (NPD1): a DHA-derived mediator that protects brain and retina against cell injury-induced oxidative stress. Brain Pathol. 2005;15:159–66.PubMedCrossRef
24.
Zurück zum Zitat Belayev L, Marcheselli VL, Khoutorova L, de Turco EB R, Busto R, Ginsberg MD, et al. Docosahexaenoic acid complexed to albumin elicits high-grade ischemic neuroprotection. Stroke. 2005;36:118–23.PubMedCrossRef Belayev L, Marcheselli VL, Khoutorova L, de Turco EB R, Busto R, Ginsberg MD, et al. Docosahexaenoic acid complexed to albumin elicits high-grade ischemic neuroprotection. Stroke. 2005;36:118–23.PubMedCrossRef
25.
Zurück zum Zitat Hong SH, Belayev L, Khoutorova L, Obenaus A, Bazan NG. Docosahexaenoic acid confers enduring neuroprotection in experimental stroke. J Neurol Sci. 2014;338:135–41.PubMedCrossRef Hong SH, Belayev L, Khoutorova L, Obenaus A, Bazan NG. Docosahexaenoic acid confers enduring neuroprotection in experimental stroke. J Neurol Sci. 2014;338:135–41.PubMedCrossRef
26.
Zurück zum Zitat Bazan NG, Molina MF, Gordon WC. Docosahexaenoic acid signalolipidomics in nutrition: significance in aging, neuroinflammation, macular degeneration, Alzheimer's, and other neurodegenerative diseases. Annu Rev Nutr. 2011;31:321–51.PubMedCentralPubMedCrossRef Bazan NG, Molina MF, Gordon WC. Docosahexaenoic acid signalolipidomics in nutrition: significance in aging, neuroinflammation, macular degeneration, Alzheimer's, and other neurodegenerative diseases. Annu Rev Nutr. 2011;31:321–51.PubMedCentralPubMedCrossRef
27.
Zurück zum Zitat Scott BL, Bazan NG. Membrane docosahexaenoate is supplied to the developing brain and retina by the liver. Proc Natl Acad Sci U S A. 1989;86:2903–7.PubMedCentralPubMedCrossRef Scott BL, Bazan NG. Membrane docosahexaenoate is supplied to the developing brain and retina by the liver. Proc Natl Acad Sci U S A. 1989;86:2903–7.PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Marcheselli VL, Hong S, Lukiw WJ, Tian XH, Gronert K, Musto A, et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J Biol Chem. 2003;278:43807–17.PubMedCrossRef Marcheselli VL, Hong S, Lukiw WJ, Tian XH, Gronert K, Musto A, et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J Biol Chem. 2003;278:43807–17.PubMedCrossRef
30.
Zurück zum Zitat de Turco EB R, Belayev L, Liu Y, Busto R, Parkins N, Bazan NG, et al. Systemic fatty acid responses to transient focal cerebral ischemia: Influence of neuroprotectant therapy with human albumin. J Neurochem. 2002;83:515–24.CrossRef de Turco EB R, Belayev L, Liu Y, Busto R, Parkins N, Bazan NG, et al. Systemic fatty acid responses to transient focal cerebral ischemia: Influence of neuroprotectant therapy with human albumin. J Neurochem. 2002;83:515–24.CrossRef
31.
Zurück zum Zitat Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp Biol Med (Maywood). 2008;233:674–88.CrossRef Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp Biol Med (Maywood). 2008;233:674–88.CrossRef
32.
Zurück zum Zitat Das UN. Essential fatty acids and their metabolites could function as endogenous HMG-CoA reductase and ACE enzyme inhibitors, anti-arrhythmic, anti-hypertensive, anti-atherosclerotic, anti-inflammatory, cytoprotective, and cardioprotective molecules. Lipids Health Dis. 2008;7:37.PubMedCentralPubMedCrossRef Das UN. Essential fatty acids and their metabolites could function as endogenous HMG-CoA reductase and ACE enzyme inhibitors, anti-arrhythmic, anti-hypertensive, anti-atherosclerotic, anti-inflammatory, cytoprotective, and cardioprotective molecules. Lipids Health Dis. 2008;7:37.PubMedCentralPubMedCrossRef
33.
Zurück zum Zitat Mori TA, Beilin LJ. Omega-3 fatty acids and inflammation. Curr Atheroscler Rep. 2004;6:461–7.PubMedCrossRef Mori TA, Beilin LJ. Omega-3 fatty acids and inflammation. Curr Atheroscler Rep. 2004;6:461–7.PubMedCrossRef
34.
Zurück zum Zitat Van Bilsen M, Planavila A. Fatty acids and cardiac disease: fuel carrying a message. Acta Physiol (Oxf). 2014;211:476–90.CrossRef Van Bilsen M, Planavila A. Fatty acids and cardiac disease: fuel carrying a message. Acta Physiol (Oxf). 2014;211:476–90.CrossRef
35.
Zurück zum Zitat Fagan SC, Hess DC, Hohnadel EJ, Pollock DM, Ergul A. Targets for vascular protection after acute ischemic stroke. Stroke. 2004;35:2220–5.PubMedCrossRef Fagan SC, Hess DC, Hohnadel EJ, Pollock DM, Ergul A. Targets for vascular protection after acute ischemic stroke. Stroke. 2004;35:2220–5.PubMedCrossRef
36.
Zurück zum Zitat Borlongan CV, Rodrigues Jr AA, Oliveira MC. Breaking the barrier in stroke: What should we know? A mini-review. Curr Pharm Des. 2012;18:3615–23.PubMedCentralPubMedCrossRef Borlongan CV, Rodrigues Jr AA, Oliveira MC. Breaking the barrier in stroke: What should we know? A mini-review. Curr Pharm Des. 2012;18:3615–23.PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Innis SM. Dietary (n-3) fatty acids and brain development. J Nutr. 2007;137:855–9.PubMed Innis SM. Dietary (n-3) fatty acids and brain development. J Nutr. 2007;137:855–9.PubMed
38.
Zurück zum Zitat Rapoport SI, Rao JS, Igarashi M. Brain metabolism of nutritionally essential polyunsaturated fatty acids depends on both the diet and the liver. Prostaglandins Leukot Essent Fatty Acids. 2007;77:251–61.PubMedCentralPubMedCrossRef Rapoport SI, Rao JS, Igarashi M. Brain metabolism of nutritionally essential polyunsaturated fatty acids depends on both the diet and the liver. Prostaglandins Leukot Essent Fatty Acids. 2007;77:251–61.PubMedCentralPubMedCrossRef
39.
41.
Zurück zum Zitat Farkas E, de Wilde MC, Kiliaan AJ, Luiten PG. Systemic effects of dietary n-3 PUFA supplementation accompany changes of CNS parameters in cerebral hypoperfusion. Ann N Y Acad Sci. 2002;977:77–86.PubMedCrossRef Farkas E, de Wilde MC, Kiliaan AJ, Luiten PG. Systemic effects of dietary n-3 PUFA supplementation accompany changes of CNS parameters in cerebral hypoperfusion. Ann N Y Acad Sci. 2002;977:77–86.PubMedCrossRef
42.
Zurück zum Zitat Cao D, Li M, Xue R, Zheng W, Liu Z, Wang X. Chronic administration of ethyl docosahexaenoate decreases mortality and cerebral edema in ischemic gerbils. Life Sci. 2005;78:74–81.PubMedCrossRef Cao D, Li M, Xue R, Zheng W, Liu Z, Wang X. Chronic administration of ethyl docosahexaenoate decreases mortality and cerebral edema in ischemic gerbils. Life Sci. 2005;78:74–81.PubMedCrossRef
43.
Zurück zum Zitat Pan HC, Kao TK, Ou YC, Yang DY, Yen YJ, Wang CC, et al. Protective effect of docosahexaenoic acid against brain injury in ischemic rats. J Nutr Biochem. 2009;20:715–25.PubMedCrossRef Pan HC, Kao TK, Ou YC, Yang DY, Yen YJ, Wang CC, et al. Protective effect of docosahexaenoic acid against brain injury in ischemic rats. J Nutr Biochem. 2009;20:715–25.PubMedCrossRef
44.
Zurück zum Zitat Yang DY, Pan HC, Yen YJ, Wang CC, Chuang YH, Chen SY, et al. Detrimental effects of post-treatment with fatty acids on brain injury in ischemic rats. Neurotoxicology. 2007;28:1220–9.PubMedCrossRef Yang DY, Pan HC, Yen YJ, Wang CC, Chuang YH, Chen SY, et al. Detrimental effects of post-treatment with fatty acids on brain injury in ischemic rats. Neurotoxicology. 2007;28:1220–9.PubMedCrossRef
Metadaten
Titel
Docosahexaenoic acid improves behavior and attenuates blood–brain barrier injury induced by focal cerebral ischemia in rats
verfasst von
Sung-Ha Hong
Larissa Khoutorova
Nicolas G Bazan
Ludmila Belayev
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Experimental & Translational Stroke Medicine / Ausgabe 1/2015
Elektronische ISSN: 2040-7378
DOI
https://doi.org/10.1186/s13231-014-0012-0

Neu in den Fachgebieten Neurologie und Psychiatrie

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Vierten reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.