Open Access

Experimental models and plant‑based therapy for experimental cerebral ischemia (Review)

  • Authors:
    • Nada M. Ezzelarab
    • Naela Saleh
    • Eman A. Khalil
    • Ahmed Abdellatif
  • View Affiliations

  • Published online on: August 11, 2020     https://doi.org/10.3892/ijfn.2020.5
  • Article Number: 5
  • Copyright: © Ezzelarab et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cerebral ischemia is a leading cause of mortality worldwide. Available treatments are mainly thrombolytic agents for restoring blood flow to the brain. However, this approach has a very narrow treatment window. Despite extensive research, there is still a need for further investigations to identify and develop novel treatment approaches. The present review aimed to summarize and discuss evidence from the literature regarding the best models with which to study cerebral ischemia and the available herbal sources that may provide potential treatment strategies for cerebral ischemia. The present review was based on research published between 1990 and 2020. Herbal remedies provide a promising research area that warrants further attention from researchers in the field. Different models have been used to investigate the pathophysiology of cerebral ischemia/reperfusion, and to examine various treatment approaches. The plant kingdom is rich in various phytochemicals with neuroprotective functions. From the literature search performed herein, it can be concluded that middle cerebral and bilateral common carotid artery occlusion models are the most convenient, cost‑effective and easily reproducible models. A number of plants, particularly those from Southeast Asia, have used for cerebral ischemia research; however, many more need to be investigated, particularly plants from Africa.

1. Introduction

Cerebral ischemia occurs when the blood flow to the brain is restricted, and it claims the lives of millions worldwide (1,2). In total, 16% of humans will have a stroke during their lifetime, with >15 million cases noted annually (1,3). Stroke is a complex disease with a narrow time window for therapeutic intervention to restore the blood supply and prevent permanent brain tissue damage (2). As a result, currently available strategies are considered inadequate (2). Therefore, there is a need for further research in order to understand the pathophysiology of the disease and to identify techniques that can reduce its severe complications (2).

Clinically relevant models are essential for cerebral ischemia research. These models should be clinically relevant and reproducible to aid in the understanding of the pathophysiology of ischemic stroke, as well as to function as a platform for the development of novel therapeutic approaches for stroke treatment.

For a number of years, plants and natural remedies have been the primary tool for folk medicine. Medicinal plants provide a cost-effective source of drugs with significant therapeutic benefits and few side-effects in comparison to commercial synthetic drugs. Herbal remedies may provide a source of novel compounds that may present novel therapeutic tools for cerebral ischemia and stroke. However, with the variants of models of cerebral ischemia, the literature lacks the link between the efficacy of the plant and the model used, which may represent a possible strategy with which to understand the mechanisms of these natural remedies.

The present review aimed to summarize and discuss the literature for data related to animal models utilized in the research of cerebral ischemia, along with the herbal-based treatment approaches used in cerebral ischemia, in order to draw a full picture of the model used for treatment. The present review also aimed to illustrate the possible association between the natural ingredient in question that proved effective and the models of cerebral ischemia.

2. Data collection methods

Search strategy

Following the guidelines of Preferred Reporting Items For Systematic Reviews And Meta-Analyses (PRISMA) (4), the present review was conducted. The present review aimed to illustrate the data from peer-reviewed original articles on cerebral ischemia phytotherapy or from studies using in vivo models of cerebral ischemia. A search was conducted to obtain targeted articles through PubMed (https://pubmed.ncbi.nlm.nih.gov/) using the following keywords: ‘Cerebral ischemia and herbal medicine’. The search span was between 1990 and 2020. In addition, the results were restricted to studies in the English language.

Selection criteria

The inclusion criteria were as follows: i) Only original studies between 1990 and 2020 were included in the present review; ii) any original article that assessed or used herbal extracts as a treatment approach for animal models of cerebral ischemia. On the other hand, the exclusion criteria were expanded to the following: i) Articles that reported the combination of plants as a formula/recipe; ii) research that was not published in the English language and were between 1990 and 2020; iii) case reports, review articles, or any secondary publications.

Data extraction

Search results were imported into Endnote X8 (Thompson Reuter) for the deletion of duplicates. The references were then screened by 3 reviewers, independently, using the eligibility criteria. The included articles were then reviewed, and data extraction was performed by 3 independent reviewers. Any disagreement in the extraction steps was raised to the supervisor to reach a consensus.

Risk of bias for individual studies

The included articles were investigated through the ‘The Cochrane Collaboration's tool for assessing the risk of bias’ (5). Any disagreements were discussed between authors to reach a consensus.

3. Search results

Search results and study characteristics

The search revealed 830 records; following title/abstract screening, 370 articles were selected. Following the screening of the full text of the articles, 52 studies were included in the present review.

Using the Cochrane risk of bias tool, we were uncertain of the bias regarding domains 4, 5 and 6(5). However, most of the included studies showed a low risk of bias in the other domains.

Pathophysiology of cerebral ischemia

Ischemic stroke accounts for approximately 90% of all stroke cases in humans, followed by intracerebral hemorrhage (9%) and subarachnoid hemorrhage (3%). Ischemic stroke occurs due to the blockage of the middle cerebral artery (MCA). Cerebral tissue hypoxia and ischemia follow within minutes, leading to neuronal cell death and permanent damage to the brain (6). Thrombolytics and the rapid restoration of the blood supply remain the only treatment options with which to prevent further neuronal damage and decrease disability (6).

Ischemic damage to both white and grey matter causes permanent damage to brain tissue (7-9). Chronic cerebral hypoperfusion causes microglia/astrocyte activation, matrix metalloproteinase stimulation, blood-brain barrier disruption and endothelial abnormalities (10-12). Chronic cerebral hypoperfusion generates neuroinflammation, oxidative stress and apoptosis of the oligodendroglia (10-12). Aging, diabetes, atherosclerosis and hypertension are the most common risk factors that lead to chronic cerebral hypoperfusion (10-12).

In vitro models of cerebral ischemia

Testing different treatment approaches on human cells in vitro provides highly valuable, cost-effective and high-throughput systems for studies on stroke (13,14). Although in vivo models are preferred in cerebral ischemia, genetic differences and structural variations, as well as molecular differences exist, which renders clinical translation problematic (13); therefore in vitro models are still critical for the understanding of the molecular mechanisms of the disease. With the introduction of new technologies, there is an excellent opportunity for the development of in vitro systems to model stroke and improve drug discovery (15). For a full review of the in vitro models of cerebral ischemia, please see the study by Holloway and Gavins, 2016(15).

Animal models in cerebral ischemia
Brain structure and function: Humans vs. animals

Although there are apparent differences between the human brain and the brains of other species, animal studies are critical in translational research. The debate continues as to whether these differences render the use of animal models in stroke studies irrelevant to the clinical application (16). These differences are evident in infarct localization (16). Another significant difference is the amount of white matter in the brain. The white matter accounts for 60% of brain tissue in humans, compared to 35% in dogs, 20% in rabbits, 15% in rats and only 10% in mice (17). This white matter difference poses a problem, as the ischemic damage of the white matter is a key player in the pathophysiology of stroke in humans (18).

Small vs. large animal models in cerebral ischemia. A number of animal species are used to investigate the mechanisms underlying cerebral ischemia (19-22). Different methods are used to generate cerebral ischemia, such as bilateral common carotid artery (CCA) occlusion (BCCAO) in rats (19), bilateral CCA stenosis (20), or asymmetric CCA surgery in mice (21), and three-vessel occlusions (3VO) in primates (22). The pros and cons of these models are illustrated in Fig. 1.

The majority of the stroke preclinical studies are conducted using small animals, particularly rodents. These studies have assisted researchers in understanding the molecular and biochemical processes within the ischemic tissue (23), as well as in understanding the different aspects of the injury mechanisms (24). However, despite the ease of handling rodents and the cognitive impairment produced as a consequence of chronic cerebral hypoperfusion, no motor abnormalities or white matter infarcts have been generated (10,25,26).

The guidelines of the Stroke Therapy Academic Industry Roundtable (STAIR), dictate that clinical studies cannot be performed on humans before testing the new proposed pharmaceutical drugs on higher animal species (27-29). Nonetheless, the use large animal models in research is associated with various issues. One of these is the need for invasive surgery to generate and monitor ischemia and this causes a high mortality rate (24). Furthermore, large animal models are costly to maintain and are labor-intensive (24). Moreover, animal rights organizations have raised several concerns regarding the use of large animals (16).

One of the apparent advantages of using large animals, such as dogs, cats, pigs, sheep and primates (30) is that imaging is easier than with the use of small animals (31). It is also more suitable to monitor the physiology, e.g., blood pressure, and blood gases in large animals compared to small ones (24).

Another advantage is that the brain of large animals is similar to the human brain in terms of functionality and structure, i.e. gyrencephalic, while rodents have lissencephalic brains (24,32). In addition, the ratio of the neocortex to the basal ganglia and the volume of white matter indicate that large animals are closer to the human neuroanatomy (17,33,34). Furthermore, large animals are considerably similar to humans in several behavioral aspects, as well as sensorimotor integration (35). Primates exhibit similarities to humans regarding the cerebral structure and high white matter percentage (29,36). Furthermore, the baboon 3-vessel occlusion (3VO) model has exhibited a tau pathology and amyloid-β deposition similar to humans (37).

On the other hand, small animals, particularly rodents, are less costly to maintain compared to large animals (17,38). Moreover, the rat physiology and cerebrovascular anatomy are similar to those of humans (38,39), and mice have homogeneous genes, and as a result, genetic mutations are easily achievable to generate transgenic mice, commonly used to investigate the molecular pathophysiology of stroke (40,41). The small brain size in rats and mice may be seen as an advantage, as various fixation procedures can be performed for neurochemical and biochemical investigations (21).

Methods used to induce cerebral ischemia in animal models. Ischemic stroke in humans occurs, in most cases, due to middle cerebral artery (MCA) occlusion (MCAO) (42). A number of models are designed to restrict cerebral blood flow, permanently or transiently, either directly by occlusion of the MCA or indirectly by common carotid artery ligation (43). A craniectomy is required for the direct occlusion of one or more cerebral vessels, through ligation, clipping, with hooks, or electrocoagulation (44). This model resembles human cases in terms of transient and permanent ischemia (44). Cerebral ischemia (CI) in this approach affects most of the cortices (45) and protects the thalamic, hypothalamic, hippocampal and midbrain regions from damage, since it produces small infarcts, unlike other MCAO models (45). It also creates a high percentage of infarct size and neurological deficits. The visual confirmation of successful MCAO is achieved during the surgery, with subsequent reperfusion of the ischemic areas (46).

On the other hand, this technique can lead to the injury of the underlying cortex or rupture of vessels by drilling or electrocoagulation (46). Additionally, the intracranial pressure and blood-brain barrier (BBB) function are greatly affected, and, as a result, it requires superior surgical skills (46). All in all, this method is highly invasive, with several complications. Therefore, other models have been introduced to avoid such complications.

MCAO models. This technique is similar to 70% of cerebral ischemic strokes that occur in humans (33,47). Koizumi et al (48) first established the intraluminal suture technique in 1986 in rats, and it was modified in the 1990s for use in mice (49). The intraluminal suture model offers two options: Transient ischemia with reperfusion or a permanent occlusion based on the time in which the suture is left in place (50). A craniotomy is not required in this model (50). A significant disadvantage is that it stimulates a larger infarction beyond the MCA zone to spread to the hippocampus and thalamus (51).

This model is reproducible in terms of primary ischemic injury and, consequently, cell death, blood-brain barrier (BBB) damage and glial activation (33,52). It is also appropriate for neuroprotection studies, due to the considerable existence of ischemic penumbra at the early stages post-occlusion (33). The infarct size in this model can be affected by the rat or mouse strain and the coating material for sutures. Strokes in spontaneously hypertensive rats (SHRs) are comparatively large and consistent in size. By contrast, in Sprague-Dawley (SD) rats, the infarcts are small and vary in size (53). An inadequate suture type may result in insufficient MCAO, leading to vessel rupture and subsequent subarachnoid hemorrhage (SAH). Silicone or poly-l-lysine coating suture is more adherent to neighboring vascular endothelium, compared to the uncoated suture, which leads to larger infarcts and minimizes inter-animal variability (54,55).

Unlike humans, following 60 min of MCAO, hypothalamic damage occurs (56). Following MCAO, hyperthermia occurs in rats and mice for at least 1 day due to hypothalamic ischemia (56,57). Approximately 12 h is needed in the transient MCAO mouse model for the damage to be repaired and for recovery to take place, which is a long therapeutic time, unlike stroke in humans (58). These two different mechanisms in the two MCAO models lead to varying results, which may be the reasons for the failure of neuroprotective agents in clinical experiments (58).

BCCAO. In several human cases of stroke, cerebral ischemia is a complication of thromboembolic conditions, such as carotid artery stenosis (59), atrial fibrillation (60) and heart failure (61). The BCCAO model is a relatively rapid and easy rat model that can be used to produce temporary or chronic cerebral ischemia (25). Rats usually exhibit white matter damage accompanied by cognitive impairments resembling those associated with stroke in humans. However, the affection of the visual pathways following the rat BCCAO compromises behavioral assessments. Therefore, C57/Bl mice have been used instead of rats, since the mouse model does not cause visual impairment and is, therefore, suitable for behavioral studies. This model is relatively easy to use, and training researchers on this mouse model decreases the mortality rate to <2%.

The bilateral common carotid artery stenosis (BCAS) model is a modification that causes carotid stenosis; the severity of cerebral hypoperfusion can be easily controlled by changing the diameter of micro-coils inserted in the carotid artery. This model is used worldwide and can be regarded as one of the most promising models of chronic cerebral hypoperfusion (10,20,62,63). This model mimics the white matter lesions induced by chronic cerebral hypoperfusion in humans (64).

Thromboembolic models. Thromboembolism is another cause of stroke. The thromboembolic clot model depends on the application route, the number and area of the clots (38). The thromboembolic clot can be spontaneous from autologous blood, which leads to vessel occlusion, causing infarcts (38). An advantage of this model is the spontaneous lysis of the clots, followed by reperfusion, which is similar to the case in humans (65).

An alternative method is the direct injection of thrombin into the MCA or internal carotid artery (ICA) to cause vascular occlusion (66). In this model, polymerized fibrin with a low number of cells and platelets are used to produce clots, while most of the human clots contain an accumulation of both platelets and fibrin, a deposition of neutrophils and monocytes and a high aggregation percentage of erythrocyte (67). In addition, the intravascular introduction of clots causes, in most cases, multifocal infarcts with noticeable variance in size and the localization of the lesion (68).

Another method with which to induce embolic stroke is by using microsphere/macrospheres to block blood flow. The major discrepancy with this model is that the fabricated spheres lead to permanent ischemia as they do not dissolve (65). An advantage associated with this model is the fact that the occlusion in rats may be postponed, while the animal is under monitor by PET or a magnetic resonance imaging (MRI) device (69).

Endothelin-1 model (ET-1). The reversible occlusion of the MCA can be achieved using a potent vasoconstrictor, such as endothelin-1 (ET-1) (33,38,70,71). A rapid blood flow reduction to the brain can be noted after the ET-1 injection, followed by reperfusion several hours later (72). Occlusion can be achieved through various mechanisms, directly by topical application onto the exposed MCA, or by intracerebral injection (73). A primary advantage of the injection method is the low invasiveness and the low mortality rate. Although ET1 is effective in rats, it is not as effective in mice (74). Another problem with this model is the variability of the lesion size (75).

Photo thrombosis model. This model requires an intravenous injection in rats or an intraperitoneal injection in mice (76,77) of a photosensitive dye, such as Rose Bengal or erythrosin B, followed by exposing the skull laser (78). Cerebral ischemia can be induced in specific brain regions depending on the purpose of the study. Once the dye is activated, reactive oxygen species (ROS) are generated, leading to endothelial damage, platelet activation and aggregation in pial and intraparenchymal vessels, forming thrombi (17). An advantage of this model is the rapid production of ischemic cell death (79). A further advantage is that it decreases the invasion and mortality rates. A significant disadvantage is that the ischemic injury is associated with early intracellular and extracellular edema formation (80), which differ from those observed in human stroke, as intracellular edema is the main indication of acute cerebral ischemia in humans (81).

In conclusion, as demonstrated through various sudies, there are various animal models, and associated techniques to produce cerebral ischemia/reperfusion, and each model has its advantages and weaknesses. The selection of the model should be based on the objectives and goal of the research study, bearing in mind that none of these models is identical to the human stroke pathophysiology.

4. Assessment of functional deficits in cerebral ischemia models

Since stroke is associated with problems in the sensory and motor pathways, researchers have focused on studying the behavioral and cognitive aspects post-stroke (82). Various functional tests (Table I) are available for use in animals, including the Rotarod, the grip and string test, the wire hanging test, the adhesive removal test, the open field maze and the water maze test (83-95).

Table I

Assessment of functional neurological deficits.

Table I

Assessment of functional neurological deficits.

TestUseMethodOutcome
Rotarod testThe rotarod test assesses the rodent 's motor coordination and balance, and in particular, detects abnormalities in the cerebellar function (84).It requires putting the rodents on a cylinder, which rotates at different rates, and the time to fall is determined (85).A significant decrease in the time the animal remains on the rod following stroke induction (86). Animal pre-training to balance on the rotarod before surgery is recommended.
Grip strength testThe grip strength test assesses the muscle force of rodents and detects any impairment in the limb strength (87).The rodent grasps a bar or a grid attached to a force transducer to determine the strength of rodents while pulling it away (87).Grip strength decreases significantly after stroke induction (88).
Wire hang testAs rodents tend to grasp any wire to avoid falling, the wire hang test is a cost-effective and easy method to evaluate the rodents' muscle performance.The time that a rodent can hang, and this depends on several factors, including the weight of the rodent, its sex, muscle size, physical properties, and age (89).In cerebral ischemia, muscle impairment is expected, and the ability to grasp the wire decreases (90). The main problem is that the recorded results are not always consistent (89).
The adhesive removal testA sensitive technique to evaluate the rodent's sensorimotor deficiency (91).The time to recognize the presence of adhesive tape placed on the rodent's forepaw and to remove it is recorded (91).The prolonged time indicated a loss of sensory function.
Open field mazeThe open field maze is one of the most widely used methods to investigate the rodents' behavior. It is a rapid and straight forwardmethod to evaluate the activity, both qualitatively and quantitatively (92,93).The open field measures movements in an enclosure of varied shapes, either circular, square, or rectangular, with a surrounding fence to prevent rodents from escaping (93).Studies have shown an increase in the crossed squares, rearing and grooming activities after ischemic stroke (148).
Water mazeThe water maze method tests rodents' spatial understanding and learning (94).The water maze test requires the placement of the rodent in a water pool, where there is a hidden platform under the water surface (94).Memory and learning abilities are determined based on the time the rodent takes to reach the platform.
Modified neurological severity score (mNSS) pointsThe mNSS test (95) evaluates the motor, sensory, reflex and the beam balance on a scale from 0 to 18.A score of 0 is normal and one of 18, is a maximal deficit.
Clark general and focal scalesThe Clark scales divided into the general neurological scale and focal neurological scale (96).The general test addresses the hair, ears, eyes, posture, spontaneous activity as well as the presence of epileptic behavior in the animals. The scores in the 6 areas are added to provide a total score ranging from 0 to 28. The focal test addresses the body and front limb symmetry, gait, climbing and circling behavior, and whisker response. The scores are added to provide a focal score ranging from 0 to 28.The clinical score is highly correlated with the infarct volume.

[i] Numbers in parentheses indicate relevant references.

5. Herbal remedies as neuroprotective agents in cerebral ischemia

The MCAO model has been extensively studied with numerous natural herbal extracts. Cerebral ischemic injury results in the production of ROS, which cause lipid, DNA and protein oxidation, therefore causing cell damage and death (96). A number of these herbs possess antioxidant, anti-inflammatory and neuroprotective activity, such as Artemisia absinthium L. (97), Lavandula angustifolia (98), Scutellaria baicalensis (99) and several others. Their active ingredients exert significant neuroprotective effects and improvement in behavioral function when used before or after ischemic injury (98-100).

Chinese plants provide a rich source of herbal extracts for medicinal purposes. Some of these have been widely investigated in cerebral ischemia. Plants, such as Erigeron breviscapus (101,102) contain breviscapine, which is considered to target autophagy mechanisms, leading to a reduction in infarct size and functional improvements in rats. Scutellarin is another component of Erigeron breviscapus, which exerts a decrease in infarct size and functional improvement when injected into rats (102). Other Chinese plants include Fructus Chebulae (103) and Fructus Schisandrae (Chinese magnolia vine fruit) (104), which protect against metalloproteinase degradation. In rats, these fruits lead to a reduction in the expression of TNF-a and IL-1b, as well as reduction in the degradation of the metalloproteinases, MMP-2 and MMP-9, in ischemic hemispheres, which leads to a reduction in infarct size (104). Panax ginseng (105-107) reduces infarcts size in rats and mice and improves function through the downregulation of calpain I and caspase 3(107), and the induction of Nrf 2 downstream targets (106). Panax ginseng has also been used in global and focal ischemic models (108); it causes a decrease in lipid peroxidation and mitochondrial swelling (108).

Salvia miltiorrhiza is a potent antioxidant that is used in MCAO (100) and in spinal cord ischemia/reperfusion injury in rats (109). It exerts a decrease in edema and infarct volume, as well as functional improvements in both models. Antioxidants, such as Camellia sinensis (110,111) and Araucaria bidwillii (112) cause functional improvements when used in global and focal ischemic models by inhibiting lipid peroxidation, and decreasing apoptosis and neuronal cell death.

Other herbal extracts and their active ingredients used in rodent models of MCAO are summarized in Table II (97-107,113-129). In Table III, the commonly used herbal extracts and their active ingredients used in other rodent models of cerebral ischemia are also listed (98,108-112,130-147). The possible mechanisms of action of each are highlighted.

Table II

Herbal extracts and their active ingredients used in rodent models of middle cerebral artery occlusion (MCAO).

Table II

Herbal extracts and their active ingredients used in rodent models of middle cerebral artery occlusion (MCAO).

Plant usedDoses/routeActive ingredients/action Outcomes/results(Refs.)
Artemisia absinthium L.(100 mg/kg and 200 mg/kg, orally)AntioxidantRat, ↓ brain oxidative stress and damage, and behavioral deficits(97)
Eleutherococcus senticosus barkOrally 3, 30 and 300 mg/kg twice at times of 0 and 90 min after reperfusionAnti-inflammatory properties through the inhibition of COX-2 expression, microglia, and astrocyte expression.Rat, vessel occlusion (4-VO)↓; hippocampal CA1 neuronal death at 300 mg/kg; ↓ COX-2, GFAP, and OX-42 in the hippocampal CA1 region(115)
Embelia ribes Burm.(100 and 200 mg/kg body weight; p.o.) for 30 daysAntioxidantRat, ↑ the grip strength activity, and GSH, GPx, GR and GST levels in hippocampus and frontal cortex; ↓ LDH levels in serum and TBARS levels in the hippocampus and frontal cortex(113)
Erigeron breviscapus (Hand-Mazz)i.p., 0.33 mg/kgBreviscapine; targeting autophagy mechanismsRat, ↓ infarct volume, brain water content and neurofunctional deficiency(101)
Intraperitoneal injections of scutellarin (20 and 60 mg/kg)Scutellarin; enhancing cellular antioxidant defense capacityRat, ↓ neurological deficits and brain infarct volume; ↑ endogenous antioxidant activity(102)
EriodictyolOral eriodictyol (1, 2 and 4 mg/kg) 30 min before pMCAO, 2 h after, and once daily for 5 days.Inhibition of neuroinflammationRat, ↓ neuronal death, infarct area, neurological and memory deficits; ↓ MPO activity, TNF-α, iNOS, and GFAP expression(114)
Fructus Chebulae300 and 500 mg/kg orallyInhibition of oxidative damagesRat, ↓ cerebral infarct volume(103)
Fructus Schisandrae(10, 30 mg/kg, IP) 30 min before the onset of ischemia, and 2 h after reperfusionSchisandrin B; inhibits Inflammation, and protects against metalloproteinase degradation.Rat, ↓ infarct volumes; ↓ protein expression of TNF-a and IL-1b and degradation of MMP-2 and MMP-9 in ischemic hemispheres(104)
Gastrodia elata (GE) BlumeIP 4-HBA (20 mg/kg) 1 h after MCAO4-Hydroxybenzyl alcohol (4-HBA); antioxidant, anti-inflammatory, anti-excitotoxic, and anti-apoptotic effectsRat, ↓ infarct volumes, motor impairments and neurological deficits; ↓ Zn2+-induced cell death, ROS generation, and PARP-1 induction(116)
Ginkgo biloba45 mg/kg injectedEGb761; activating the Akt/CREB/BDNF pathwayRat, ↑ behavior scores; ↑ phosphorylation of AKT, CREB and BDNF in the brain(105)
Lavandula angustifoliaOrally once/day for 3 days before ischemia and once 2 h after ischemiaLavender oil AntioxidantMice, ↓ neurological deficits, infarct size, MDA levels, carbonyl, ROS ↑ antioxidant capacity(99)
Nigella sativa seeds400 mg/kg, per orally for 7 daysAntioxidant, free radical scavenging, and anti-inflammatory propertiesRat, ↓TBARS levels ↑ glutathione, SOD and catalase levels(117)
Panax ginsengIP, 10 and 20 mg kg; orally, 100 mg/kg for 7 days before MCAO; oral, (0.5, 1, 5 or 10 mg/kg), every 3 days20(R)-ginsenoside Rg(3); downregulation of calpain I and caspase 3; through Nrf2 pathways; ginsenoside Rb1Rat, ↓ cerebral infarct volumes; ↓ calpain I and caspase-3 mRNA; mice, ↓ acute sensorimotor deficits; ↑ induction of Nrf2-downstream targets; mice, ↓ oxidative stress(105-107)
Polygonum cuspidatum Sieb (knotgrass)2.5, 5, 10 mg/kg tail vein injection 15 min after occlusion Emodin-8-O-beta-D-glucoside; antioxidative effectsRat, ↓ neurological deficits and the cerebral infarct area; ↑antioxidative; ↓ MDA level(118)
Astragali radixIntragastric calycosin (7.5, 15, 30 mg/kg)Calycosin; antioxidantRat, ↓ neurological deficit and infarct volume; ↓ malondialdehyde (MDA), and reactive oxygen species (ROS); ↑the activity of superoxide dismutase (SOD), catalase and glutathione peroxidase (GSH-Px); ↓ expression of 4-hydroxy-2-nonenal (4-HNE)(119)
Pueraria radix(25 and 50 mg/kg; intraperitoneally) 10 min before MCAOPuerarin; inhibition of both HIF-1α and TNF-αRat, ↓infarct size; ↓ (HIF-1), (iNOS) and active caspase-3 protein expression; ↓ mRNA of TNF-α in ischemic regions(120)
Scrophularia radix2.4 g/kg-1Regulating MAPK pathwaysMice, ↓ infarct volume, brain water content, (NO), (MDA), neurological deficits and LDH; ↑ antioxidant capacity(121)
Salvia miltiorrhiza (Red sage) and cacaoOrally high dose (270 mg/kg) and low (27 mg/kg)Antioxidants: Reducing the production of free radicalsRat, ↓ ischemic cell death within the peri-infarct area; ↑ performance in routine motor and neurological tasks(122)
Salvia miltiorrhiza (Red sage)i.p. (15, 30 and 60 mg/kg); 30 and 60 mg/kg injected every 24 h for 5 days; i.p. 16 mg/kgMagnesium lithospermate B; upregulation of p-Akt; vasodilation; ↓ platelet aggregation; ↓inflammation; sodium danshensu [3-(3,4-dihydroxyphenyl) lactic acid; inhibition of apoptosis by activating the PI3K/Akt pathway; tanshinonesRat, ↓ neurological deficits; ↓ brain water content, glutamate levels, and cerebral infarct zones; rat, ↑ survival rate; ↓ infarct volume; ↓ neuronal death; ↓ number of apoptotic cells; ↑ ratio of Bcl-2/Bax; mice, 30% reduction in infarct size; improved neurological deficit(100,123,124)
Scutellaria baicalensis (Chinese skullcap)i.p. dose of 100 mg/kgBaicalin; inhibits the expression of PAR-1↑ Rat neurological function; ↓ cerebral infarct volume(99)
Sophora flavescens Ait.i.p. oxymatrine 120 mg/kg after MCAOOxymatrine; downregulation of 12/15-LOX, phospho-p38 MAPK and cPLA2Rat; ↓ brain water content, and infarct volume; ↓ overexpression of 12/15-LOX, phospho-p38; MAPK and cPLA2(125)
0.5 mg kg, i.p.Flavonoids kurarinone (45.5%) and sophoraflavone G (14.7%); inhibition of caspase-3 activation and reduction of DNA fragmentationRat; ↓ caspase-3 enzyme activity, and DNA fragmentation; ↓ cell apoptosis(126)
Tripterygium wilfor0dii Hook.Pre-treatment with TP (0.2 mg/kg) and DAHP (0.5 g/kg) by i.p. injection 0.2 mg/kg, IP 24 h before MCAODAHP and triptolide Activation of the PI3K/Akt/mTOR pathway and inactivation of the ERK1/2 pathway Inhibition of NF-κB activationRat, ↓ ischemic lesion volume, and neuronal cell death; ↓ astrocyte numbers, ↓ levels of Bax and caspase 3, ↑ NF-κB; ↑Bcl-2 expression;↑expression of PI3K, Akt, and mTOR; ↓ ERK1 and ERK2 phosphorylation (both studies) ↓ iNOS, COX-2, GFAP and NF-κB expression(127,128)
Zizyphus jujuba and Silymarin100, 250 and 500 mg/kg, p.o., or Silymarin (250 mg/kg, p.o.) for 3 days before MCAOAmelioration of oxidative stressRat, ↓ neurological deficits, motor impairment, and cerebral infarction volume; ↓ oxidative stress(129)

[i] ↓, decrease; ↑, increase.

Table III

Herbal extracts and their active ingredients used in other rodent models of cerebral ischemia.

Table III

Herbal extracts and their active ingredients used in other rodent models of cerebral ischemia.

Experimental modelPlant usedDose/routeActive ingredients/actions Outcomes/results(Refs.)
Bilateral common carotid artery occlusion (BCAO) in ratsAraucaria bidwilliiSeven days pre-treatment with a bi-flavone fraction (BFR), 100 and 200 mg/kgBiflavones; antioxidant↑ Superoxide dismutase (SOD), catalase (CAT), glutathione (GSH); ↓ lipid peroxidation (LPO) in various brain regions; ↓ neurological deficit and sensory-motor function(112)
Camellia sinensisGreen tea extract (0.5%) orally administeredFlavanol methylxanthines, theobromine, and theophylline; increased levels of hydrogen peroxide and inhibition of lipid peroxidation products in the ipsilateral hemisphere by the ischemia/reperfusion↑ Hydrogen peroxide, and also inhibited the increased production of lipid peroxidation products; ↓apoptosis, ↓neuronal cell death(110,111)
Magnolia officinalis10 and 30 mg/kg intravenous injectionMagnolol; downregulation of p38/MAPK, CHOP, and nitrotyrosine↓ Infarct volume; ↓ inflammatory cytokines; ↓ production of nitrotyrosine, 4-hydroxy2-nonenal (4-HNE), inducible NO synthase (iNOS), various phosphorylated p38 mitogen-activated protein kinases, and different C/EBP homologs; ↓reactive oxygen species; ↑ expression of p-Akt and (NF-κB)(130)
Global and focal cerebral ischemia/ reperfusion in rats and micePanax ginsengIn vivo Re (5, 10 or 20 mg kg 21, oral for 7 days, once a day) before occlusionGinsenoside↓ MDA level AND mitochondrial swelling(108)
Momordica charantia50, 100, 200 mg/kg at 30 min before cerebral ischemia, or 100 or 200 mg/kg, 30 min after cerebral ischemiaM. charantia polysaccharide (MCP); inhibiting oxidative stress↓ NO, O2, ONOO and lipid peroxidation; ↓ activation of JNK3/c-Jun/Fas-L and JNK3/cytochrome c/caspase-3; signaling cascades in ischemic brains(131)
Ocimum basilicum100 and 200 mg/kg 3,7-dimethyl-1,6-octadien-3-ol (linalool; 3.94 mg/g), 1-methoxy-4-(2-propenyl) benzene (estragole; 2.03 mg/g), methyl cinnamate (1.28 mg/g), 4-allyl-2-methoxyphenol (eugenol; 0.896 mg/g), and 1, 8-cineole (0.288 mg/g); anticonvulsant, anti-inflammatory, and neurodegenerative; restoration of endogenous antioxidants; elevated brain glutathione content↓ Infarct size and lipid peroxidation(132-134)
Ocimum sanctumOrally in doses of 200 mg/kg/dayMethanolic extract of OS leaves; anti-inflammatory, antioxidant, immunomodulatory and anti-stress properties↑ SOD activity; prevented the rise in methane dicarboxylic aldehyde (MDA) levels(135)
O. europaea (olive oil)Virgin olive oil consumption at 0.75 ml/kg/dayMonounsaturated fatty acids and polyphenols; oral low-dose (2.5 mg/kg of body weight) of hydroxytyrosol and a high-dose (10 mg/kg of body weight)↓ Infarct volume, brain edema; ↑brain cerebroside levels(136-138)
Spinal cord ischemia/reperfusion (I/R) injury in rats.Salvia miltiorrhiza (red sage)1, 10, or 50 mg/kgSalvianolic acid B; ERK activation↓ Spinal cord edema and infarct volume; ↑ motor function of the hind limbs; ↓ generation of oxidative products; ↑ antioxidant defense activities(109)
Transient hippocampal ischemia in ratsMelissa officinalis (lemon balm)100 µg/ml orallyMelissa oil; inhibition of HIF-1α and oxidative stress, followed by the inhibition of apoptosis↓ Caspase-3 activity and malondialdehyde level; ↑ antioxidant capacity in the hippocampus; ↓ HIF-1α gene expression(139)
Transient focal cerebral ischemia in miceLavandula officinalis200 mg of lavender officinalisLinalool-octanone, camphor, Caryophyllene, terpinen-4-ol, and flavonoids; decrease neurological deficit scores, infarct size, the levels of MDA, carbonyl and ROS, and attenuate neuronal damage, upregulated SOD, CAT, GSH-Px activities, and GSH/GSSG ratio↓ Neurological deficit scores, infarct size, MDA, carbonyl and ROS, ↓ neuronal damage, antioxidant effects(98,140)
Transient global ischemia in gerbilsBaicalin and JasminoidinBaicalin and jasminoidin, or nimodipine) were intravenously treatedBaicalin and jasminoidin; reduce neuronal damage in Gerbils hippocampus; lower MDA content, higher SOD, GSH, and GSH-PX activities↓ Infarction area; ↓ lipid peroxidation; ↓ caspase-3(141-143)
Gastrodia elataFree radical scavenging and antioxidant activity; increased expression of antioxidant genesProtection of hippocampal neurons; ↓ infarct size in cortex and striatum(144,145)
Ginkgo biloba L.Ginkgo biloba extract (37,5-150 mg/kg) orallyGinkgo-flavone glycosides and terpenoids; antioxidant and free radical scavenging effects; decrease neurons against oxidative stress; decrease neuronal injuryPre-treatment with G. biloba extract improved blood flow and reduced edema in the hippocampal region in a dose-dependent manner(146,147)

[i] ↓, decrease; ↑, increase.

6. Conclusions

Different in vitro and animal models are available for the study of cerebral ischemia/reperfusion injury. The pathophysiology of cerebral ischemia/reperfusion is complex, and animal models are superior, particularly when testing various treatment approaches. It is suggested that the MCAO and BCCAO models the most convenient, cost-effective and easily reproducible models. Herbal extracts and phytochemicals provide a wide variety of neuroprotective agents that may be of value to research in cerebral ischemia. Further investigations are required to identify the active ingredients of such plants, and further testing is warranted. The literature provides a wealth of knowledge regarding herbal medicine in cerebral ischemia research, mostly using plants from south-East Asia. Plants from Africa and other regions warrant further investigation as they provide attractive targets for the development of novel therapeutic drugs.

Acknowledgements

Not applicable.

Funding

The present study was partially funded by a research support grant (SSE-BIOL-A.A-FY20) from the American University in Cairo.

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

NME, NS, EK, AA were involved in the conception and design of the study, and in the writing and revision of the manuscript. NE and NS were involved in the production of the figure and tables.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Després JP, Fullerton HJ, Howard VJ, et al: Heart disease and stroke statistics-2015 update: A report from the American Heart Association. Circulation. 131:e29–e322. 2015.PubMed/NCBI View Article : Google Scholar

2 

Dong B, Yang Y, Zhang Z, Xie K, Su L and Yu Y: Hemopexin alleviates cognitive dysfunction after focal cerebral ischemia-reperfusion injury in rats. BMC Anesthesiol. 19(13)2019.PubMed/NCBI View Article : Google Scholar

3 

Di Carlo A: Human and economic burden of stroke. Age Ageing. 38:4–5. 2009.PubMed/NCBI View Article : Google Scholar

4 

Moher D, Liberati A, Tetzlaff J and Altman DG: Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Ann Intern Med. 151:264–269. 2009.PubMed/NCBI View Article : Google Scholar

5 

Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, Savovic J, Schulz KF, Weeks L, Sterne JA, et al: The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ. 343(d5928)2011.PubMed/NCBI View Article : Google Scholar

6 

Woodruff TM, Thundyil J, Tang SC, Sobey CG, Taylor SM and Arumugam TV: Pathophysiology, treatment, and animal and cellular models of human ischemic stroke. Mol Neurodegener. 6(11)2011.PubMed/NCBI View Article : Google Scholar

7 

O'Brien JT and Thomas A: Vascular dementia. Lancet. 386:1698–1706. 2015.PubMed/NCBI View Article : Google Scholar

8 

Kalaria RN: Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol. 131:659–685. 2016.PubMed/NCBI View Article : Google Scholar

9 

Hainsworth AH and Markus HS: Do in vivo experimental models reflect human cerebral small vessel disease? A systematic review. J Cereb Blood Flow Metab. 28:1877–1891. 2008.PubMed/NCBI View Article : Google Scholar

10 

Bink DI, Ritz K, Aronica E, Van Der Weerd L and Daemen MJ: Mouse models to study the effect of cardiovascular risk factors on brain structure and cognition. J Cereb Blood Flow Metab. 33:1666–1684. 2013.PubMed/NCBI View Article : Google Scholar

11 

Gorelick PB, Counts SE and Nyenhuis D: Vascular cognitive impairment and dementia. Biochim Biophys Acta. 1862:860–868. 2016.PubMed/NCBI View Article : Google Scholar

12 

Venkat P, Chopp M and Chen J: Models and mechanisms of vascular dementia. Exp Neurol. 272:97–108. 2015.PubMed/NCBI View Article : Google Scholar

13 

Hanke T: Lessons from TGN1412. Lancet. 368:1569–1570; author reply 1570. 2006.PubMed/NCBI View Article : Google Scholar

14 

Römer PS, Berr S, Avota E, Na SY, Battaglia M, ten Berge I, Einsele H and Hünig T: Preculture of PBMCs at high cell density increases sensitivity of T-cell responses, revealing cytokine release by CD28 superagonist TGN1412. Blood. 118:6772–6782. 2011.PubMed/NCBI View Article : Google Scholar

15 

Holloway PM and Gavins FN: Modeling ischemic stroke in vitro: status quo and future perspectives. Stroke. 47:561–569. 2016.PubMed/NCBI View Article : Google Scholar

16 

Cook DJ and Tymianski M: Nonhuman primate models of stroke for translational neuroprotection research. Neurotherapeutics. 9:371–379. 2012.PubMed/NCBI View Article : Google Scholar

17 

Krafft PR, Bailey EL, Lekic T, Rolland WB, Altay O, Tang J, Wardlaw JM, Zhang JH and Sudlow CL: Etiology of stroke and choice of models. Int J Stroke. 7:398–406. 2012.PubMed/NCBI View Article : Google Scholar

18 

Ahmad AS, Satriotomo I, Fazal J, Nadeau SE and Doré S: Considerations for the optimization of induced white matter injury preclinical models. Front Neurol. 6(172)2015.PubMed/NCBI View Article : Google Scholar

19 

Edrissi H, Schock SC, Cadonic R, Hakim AM and Thompson CS: Cilostazol reduces blood brain barrier dysfunction, white matter lesion formation and motor deficits following chronic cerebral hypoperfusion. Brain Res. 1646:494–503. 2016.PubMed/NCBI View Article : Google Scholar

20 

Shibata M, Ohtani R, Ihara M and Tomimoto H: White matter lesions and glial activation in a novel mouse model of chronic cerebral hypoperfusion. Stroke. 35:2598–2603. 2004.PubMed/NCBI View Article : Google Scholar

21 

Hattori Y, Enmi J, Kitamura A, Yamamoto Y, Saito S, Takahashi Y, Iguchi S, Tsuji M, Yamahara K, Nagatsuka K, et al: A novel mouse model of subcortical infarcts with dementia. J Neurosci. 35:3915–3928. 2015.PubMed/NCBI View Article : Google Scholar

22 

Chen A, Akinyemi RO, Hase Y, Firbank MJ, Ndung'u MN, Foster V, Craggs LJ, Washida K, Okamoto Y, Thomas AJ, et al: Frontal white matter hyperintensities, clasmatodendrosis and gliovascular abnormalities in ageing and post-stroke dementia. Brain. 139:242–258. 2015.PubMed/NCBI View Article : Google Scholar

23 

McCabe C, Arroja MM, Reid E and Macrae IM: Animal models of ischaemic stroke and characterisation of the ischaemic penumbra. Neuropharmacology. 134:169–177. 2018.PubMed/NCBI View Article : Google Scholar

24 

Traystman RJ: Animal models of focal and global cerebral ischemia. ILAR J. 44:85–95. 2003.PubMed/NCBI View Article : Google Scholar

25 

Farkas E, Luiten PG and Bari F: Permanent, bilateral common carotid artery occlusion in the rat: A model for chronic cerebral hypoperfusion-related neurodegenerative diseases. Brain Res Rev. 54:162–180. 2007.PubMed/NCBI View Article : Google Scholar

26 

Nishio K, Ihara M, Yamasaki N, Kalaria RN, Maki T, Fujita Y, Ito H, Oishi N, Fukuyama H, Miyakawa T, et al: A mouse model characterizing features of vascular dementia with hippocampal atrophy. Stroke. 41:1278–1284. 2010.PubMed/NCBI View Article : Google Scholar

27 

Stem Cell Therapies as an Emerging Paradigm in Stroke Participants: Stem cell therapies as an emerging paradigm in stroke (STEPS): Bridging basic and clinical science for cellular and neurogenic factor therapy in treating stroke. Stroke 40: 510-515, 2009.

28 

Savitz SI, Chopp M, Deans R, Carmichael S, Phinney D and Wechsler L: STEPS Participants: Stem cell therapy as an emerging paradigm for stroke (STEPS) II. Stroke. 42:825–829. 2011.PubMed/NCBI View Article : Google Scholar

29 

Stroke Therapy Academic Industry Roundtable (STAIR): Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke 30: 2752-2758, 1999.

30 

Bacigaluppi M, Comi G and Hermann DM: Animal models of ischemic stroke. Part two: Modeling cerebral ischemia. Open Neurol J. 4:34–38. 2010.PubMed/NCBI View Article : Google Scholar

31 

Marshall J, Ridley R, Baker H, Hall L, Carpenter T and Wood N: Serial MRI, functional recovery, and long-term infarct maturation in a non-human primate model of stroke. Brain Res Bull. 61:577–585. 2003.PubMed/NCBI View Article : Google Scholar

32 

Cook DJ, Teves L and Tymianski M: Treatment of stroke with a PSD-95 inhibitor in the gyrencephalic primate brain. Nature. 483:213–217. 2012.PubMed/NCBI View Article : Google Scholar

33 

Howells DW, Porritt MJ, Rewell SS, O'collins V, Sena ES, Van Der Worp HB, Traystman RJ and Macleod MR: Different strokes for different folks: The rich diversity of animal models of focal cerebral ischemia. J Cereb Blood Flow Metab. 30:1412–1431. 2010.PubMed/NCBI View Article : Google Scholar

34 

Macrae I: Preclinical stroke research-advantages and disadvantages of the most common rodent models of focal ischaemia. Br J Pharmacol. 164:1062–1078. 2011.PubMed/NCBI View Article : Google Scholar

35 

Canazza A, Minati L, Boffano C, Parati E and Binks S: Experimental models of brain ischemia: A review of techniques, magnetic resonance imaging, and investigational cell-based therapies. Front Neurol. 5(19)2014.PubMed/NCBI View Article : Google Scholar

36 

Madigan JB, Wilcock DM and Hainsworth AH: Vascular contributions to cognitive impairment and dementia: Topical review of animal models. Stroke. 47:1953–1959. 2016.PubMed/NCBI View Article : Google Scholar

37 

Ndung'u M, Härtig W, Wegner F, Mwenda J, Low R, Akinyemi R and Kalaria RN: Cerebral amyloid β(42) deposits and microvascular pathology in ageing baboons. Neuropathol Appl Neurobiol. 38:487–499. 2012.PubMed/NCBI View Article : Google Scholar

38 

Durukan A and Tatlisumak T: Acute ischemic stroke: Overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav. 87:179–197. 2007.PubMed/NCBI View Article : Google Scholar

39 

Liu F and McCullough LD: Middle cerebral artery occlusion model in rodents: Methods and potential pitfalls. J Biomed Biotechnol. 2011(464701)2011.PubMed/NCBI View Article : Google Scholar

40 

Kraft P, Göb E, Schuhmann MK, Göbel K, Deppermann C, Thielmann I, Herrmann AM, Lorenz K, Brede M, Stoll G, et al: FTY720 ameliorates acute ischemic stroke in mice by reducing thrombo-inflammation but not by direct neuroprotection. Stroke. 44:3202–3210. 2013.PubMed/NCBI View Article : Google Scholar

41 

Göb E, Reymann S, Langhauser F, Schuhmann MK, Kraft P, Thielmann I, Göbel K, Brede M, Homola G, Solymosi L, et al: Blocking of plasma kallikrein ameliorates stroke by reducing thromboinflammation. Ann Neurol. 77:784–803. 2015.PubMed/NCBI View Article : Google Scholar

42 

Dirnagl U and Macleod MR: Stroke research at a road block: The streets from adversity should be paved with meta-analysis and good laboratory practice. Br J Pharmacol. 157:1154–1156. 2009.PubMed/NCBI View Article : Google Scholar

43 

Yanamoto H, Nagata I, Niitsu Y, Xue JH, Zhang Z and Kikuchi H: Evaluation of MCAO stroke models in normotensive rats: Standardized neocortical infarction by the 3VO technique. Exp Neurol. 182:261–274. 2003.PubMed/NCBI View Article : Google Scholar

44 

Dirnagl U: Rodent models of stroke: Springer, 2010.

45 

Buchan AM, Xue D and Slivka A: A new model of temporary focal neocortical ischemia in the rat. Stroke. 23:273–279. 1992.PubMed/NCBI View Article : Google Scholar

46 

Sugimori H, Yao H, Ooboshi H, Ibayashi S and Iida M: Krypton laser-induced photothrombotic distal middle cerebral artery occlusion without craniectomy in mice. Brain Res Brain Res Protoc. 13:189–196. 2004.PubMed/NCBI View Article : Google Scholar

47 

Bogousslavsky J, Van Melle G and Regli F: The Lausanne Stroke Registry: Analysis of 1,000 consecutive patients with first stroke. Stroke. 19:1083–1092. 1988.PubMed/NCBI View Article : Google Scholar

48 

Koizumi J, Yoshida Y, Nakazawa T and Ooneda G: Experimental studies of ischemic brain edema. 1. A new experimental model of cerebral embolism in rats in which recirculation can be introduced in the ischemic area. Jpn J Stroke. 8:1–8. 1986.

49 

Smith HK, Russell JM, Granger DN and Gavins FN: Critical differences between two classical surgical approaches for middle cerebral artery occlusion-induced stroke in mice. J Neurosci Methods. 249:99–105. 2015.PubMed/NCBI View Article : Google Scholar

50 

Chiang T, Messing RO and Chou WH: Mouse model of middle cerebral artery occlusion. J Vis Exp. (e2761)2011.PubMed/NCBI View Article : Google Scholar

51 

Garcia JH, Liu KF and Ho KL: Neuronal necrosis after middle cerebral artery occlusion in Wistar rats progresses at different time intervals in the caudoputamen and the cortex. Stroke. 26:636–643, Discussion 643. 1995.PubMed/NCBI View Article : Google Scholar

52 

Kuraoka M, Furuta T, Matsuwaki T, Omatsu T, Ishii Y, Kyuwa S and Yoshikawa Y: Direct experimental occlusion of the distal middle cerebral artery induces high reproducibility of brain ischemia in mice. Exp Anim. 58:19–29. 2009.PubMed/NCBI View Article : Google Scholar

53 

Duverger D and MacKenzie ET: The quantification of cerebral infarction following focal ischemia in the rat: Influence of strain, arterial pressure, blood glucose concentration, and age. J Cereb Blood Flow Metab. 8:449–461. 1988.PubMed/NCBI View Article : Google Scholar

54 

Belayev L, Alonso OF, Busto R, Zhao W and Ginsberg MD: Middle cerebral artery occlusion in the rat by intraluminal suture. Neurological and pathological evaluation of an improved model. Stroke. 27:1616–1623. 1996.PubMed/NCBI View Article : Google Scholar

55 

Schmid-Elsaesser R, Zausinger S, Hungerhuber E, Baethmann A and Reulen HJ: A critical reevaluation of the intraluminal thread model of focal cerebral ischemia. Stroke. 29:2162–2170. 1998.PubMed/NCBI View Article : Google Scholar

56 

Li F, Omae T and Fisher M: Spontaneous hyperthermia and its mechanism in the intraluminal suture middle cerebral artery occlusion model of rats. Stroke. 30:2464–2470, Discussion 2470-2471. 1999.PubMed/NCBI View Article : Google Scholar

57 

Barber PA, Hoyte L, Colbourne F and Buchan AM: Temperature-regulated model of focal ischemia in the mouse: A study with histopathological and behavioral outcomes. Stroke. 35:1720–1725. 2004.PubMed/NCBI View Article : Google Scholar

58 

Hossmann KA: The two pathophysiologies of focal brain ischemia: Implications for translational stroke research. J Cereb Blood Flow Metab. 32:1310–1316. 2012.PubMed/NCBI View Article : Google Scholar

59 

Demarin V, Zavoreo I and Kes VB: Carotid artery disease and cognitive impairment. J Neurol Sci. 322:107–111. 2012.PubMed/NCBI View Article : Google Scholar

60 

de Bruijn RF, Heeringa J, Wolters FJ, Franco OH, Stricker BH, Hofman A, Koudstaal PJ and Ikram MA: Association between atrial fibrillation and dementia in the general population. JAMA Neurol. 72:1288–1294. 2015.PubMed/NCBI View Article : Google Scholar

61 

Adelborg K, Szépligeti S, Sundbøll J, Horváth-Puhó E, Henderson VW, Ording A, Pedersen L and Sørensen HT: Risk of stroke in patients with heart failure: A population-based 30-year cohort study. Stroke. 48:1161–1168. 2017.PubMed/NCBI View Article : Google Scholar

62 

Shibata M, Yamasaki N, Miyakawa T, Kalaria RN, Fujita Y, Ohtani R, Ihara M, Takahashi R and Tomimoto H: Selective impairment of working memory in a mouse model of chronic cerebral hypoperfusion. Stroke. 38:2826–2832. 2007.PubMed/NCBI View Article : Google Scholar

63 

Ihara M, Taguchi A, Maki T, Washida K and Tomimoto H: A mouse model of chronic cerebral hypoperfusion characterizing features of vascular cognitive impairment. Methods Mol Biol. 1135:95–102. 2014.PubMed/NCBI View Article : Google Scholar

64 

Washida K, Hattori Y and Ihara M: Animal models of chronic cerebral hypoperfusion: From mouse to primate. Int J Mol Sci. 20(6176)2019.PubMed/NCBI View Article : Google Scholar

65 

Sommer CJ: Ischemic stroke: Experimental models and reality. Acta Neuropathol. 133:245–261. 2017.PubMed/NCBI View Article : Google Scholar

66 

Orset C, Macrez R, Young AR, Panthou D, Angles-Cano E, Maubert E, Agin V and Vivien D: Mouse model of in situ thromboembolic stroke and reperfusion. Stroke. 38:2771–2778. 2007.PubMed/NCBI View Article : Google Scholar

67 

Smith WS, Sung G, Starkman S, Saver JL, Kidwell CS, Gobin YP, Lutsep HL, Nesbit GM, Grobelny T, Rymer MM, et al: Safety and efficacy of mechanical embolectomy in acute ischemic stroke: Results of the MERCI trial. Stroke. 36:1432–1438. 2005.PubMed/NCBI View Article : Google Scholar

68 

Niessen F, Hilger T, Hoehn M and Hossmann KA: Differences in clot preparation determine outcome of recombinant tissue plasminogen activator treatment in experimental thromboembolic stroke. Stroke. 34:2019–2024. 2003.PubMed/NCBI View Article : Google Scholar

69 

Walberer M and Rueger MA: The macrosphere model-an embolic stroke model for studying the pathophysiology of focal cerebral ischemia in a translational approach. Ann Transl Med. 3(123)2015.PubMed/NCBI View Article : Google Scholar

70 

Macrae IM, Robinson MJ, Graham DI, Reid JL and McCulloch J: Endothelin-1-induced reductions in cerebral blood flow: Dose dependency, time course, and neuropathological consequences. J Cereb Blood Flow Metab. 13:276–284. 1993.PubMed/NCBI View Article : Google Scholar

71 

Bogaert L, Scheller D, Moonen J, Sarre S, Smolders I, Ebinger G and Michotte Y: Neurochemical changes and laser Doppler flowmetry in the endothelin-1 rat model for focal cerebral ischemia. Brain Res. 887:266–275. 2000.PubMed/NCBI View Article : Google Scholar

72 

Biernaskie J, Corbett D, Peeling J, Wells J and Lei H: A serial MR study of cerebral blood flow changes and lesion development following endothelin-1-induced ischemia in rats. Magn Reson Med. 46:827–830. 2001.PubMed/NCBI View Article : Google Scholar

73 

Hughes PM, Anthony DC, Ruddin M, Botham MS, Rankine EL, Sablone M, Baumann D, Mir AK and Perry VH: Focal lesions in the rat central nervous system induced by endothelin-1. J Neuropathol Exp Neurol. 62:1276–1286. 2003.PubMed/NCBI View Article : Google Scholar

74 

Horie N, Maag AL, Hamilton SA, Shichinohe H, Bliss TM and Steinberg GK: Mouse model of focal cerebral ischemia using endothelin-1. J Neurosci Methods. 173:286–290. 2008.PubMed/NCBI View Article : Google Scholar

75 

Ansari S, Azari H, Caldwell KJ, Regenhardt RW, Hedna VS, Waters MF, Hoh BL and Mecca AP: Endothelin-1 induced middle cerebral artery occlusion model for ischemic stroke with laser Doppler flowmetry guidance in rat. J Vis Exp. (50014)2013.PubMed/NCBI View Article : Google Scholar

76 

Kim GW, Sugawara T and Chan PH: Involvement of oxidative stress and caspase-3 in cortical infarction after photothrombotic ischemia in mice. J Cereb Blood Flow Metab. 20:1690–1701. 2000.PubMed/NCBI View Article : Google Scholar

77 

Kleinschnitz C, Braeuninger S, Pham M, Austinat M, Nölte I, Renné T, Nieswandt B, Bendszus M and Stoll G: Blocking of platelets or intrinsic coagulation pathway-driven thrombosis does not prevent cerebral infarctions induced by photothrombosis. Stroke. 39:1262–1268. 2008.PubMed/NCBI View Article : Google Scholar

78 

Watson BD, Dietrich WD, Busto R, Wachtel MS and Ginsberg MD: Induction of reproducible brain infarction by photochemically initiated thrombosis. Ann Neurol. 17:497–504. 1985.PubMed/NCBI View Article : Google Scholar

79 

Dietrich WD, Ginsberg MD, Busto R and Watson BD: Photochemically induced cortical infarction in the rat. 1. Time course of hemodynamic consequences. J Cereb Blood Flow Metab. 6:184–194. 1986.PubMed/NCBI View Article : Google Scholar

80 

Lee VM, Burdett NG, Carpenter A, Hall LD, Pambakian PS, Patel S, Wood NI and James MF: Evolution of photochemically induced focal cerebral ischemia in the rat. Magnetic resonance imaging and histology. Stroke. 27:2110–2119. 1996.PubMed/NCBI View Article : Google Scholar

81 

Provenzale JM, Jahan R, Naidich TP and Fox AJ: Assessment of the patient with hyperacute stroke: Imaging and therapy. Radiology. 229:347–359. 2003.PubMed/NCBI View Article : Google Scholar

82 

DeVries AC, Nelson RJ, Traystman RJ and Hurn PD: Cognitive and behavioral assessment in experimental stroke research: Will it prove useful? Neurosci Biobehav Rev. 25:325–342. 2001.PubMed/NCBI View Article : Google Scholar

83 

Shiotsuki H, Yoshimi K, Shimo Y, Funayama M, Takamatsu Y, Ikeda K, Takahashi R, Kitazawa S and Hattori N: A rotarod test for evaluation of motor skill learning. J Neurosci Methods. 189:180–185. 2010.PubMed/NCBI View Article : Google Scholar

84 

Balkaya M, Kröber JM, Rex A and Endres M: Assessing post-stroke behavior in mouse models of focal ischemia. J Cereb Blood Flow Metab. 33:330–338. 2013.PubMed/NCBI View Article : Google Scholar

85 

Lee JK, Park MS, Kim YS, Moon KS, Joo SP, Kim TS and Kim SH: Photochemically induced cerebral ischemia in a mouse model. Surg Neurol. 67:620–625. 2007.PubMed/NCBI View Article : Google Scholar

86 

De Luca A, Tinsley J, Aartsma-Rus A, van Putten M, Nagaraju K, de La Porte S, Dubach-Powell J and Carlson G: Use of grip strength meter to assess the limb strength of mdx mice. SOP DMD_M.2. 2008.

87 

Ishrat T, Sayeed I, Atif F and Stein DG: Effects of progesterone administration on infarct volume and functional deficits following permanent focal cerebral ischemia in rats. Brain Res. 1257:94–101. 2009.PubMed/NCBI View Article : Google Scholar

88 

Hoffman E and Winder SJ: A modified wire hanging apparatus for small animal muscle function testing. PLoS Curr 8. (ecurrents.md.1e2bec4e78697b7b0ff80ea25a1d38be)2016.PubMed/NCBI View Article : Google Scholar

89 

Gerlai R, Thibodeaux H, Palmer JT, van Lookeren Campagne M and Van Bruggen N: Transient focal cerebral ischemia induces sensorimotor deficits in mice. Behav Brain Res. 108:63–71. 2000.PubMed/NCBI View Article : Google Scholar

90 

Bouet V, Boulouard M, Toutain J, Divoux D, Bernaudin M, Schumann-Bard P and Freret T: The adhesive removal test: A sensitive method to assess sensorimotor deficits in mice. Nat Protoc. 4:1560–1564. 2009.PubMed/NCBI View Article : Google Scholar

91 

Seibenhener ML and Wooten MC: Use of the Open Field Maze to measure locomotor and anxiety-like behavior in mice. J Vis Exp. (e52434)2015.PubMed/NCBI View Article : Google Scholar

92 

Gould TD, Dao DT and Kovacsics CE: The open field test. Mood and anxiety related phenotypes in mice. Springer, pp1-20, 2009.

93 

Vorhees CV and Williams MT: Morris water maze: Procedures for assessing spatial and related forms of learning and memory. Nat Protoc. 1:848–858. 2006.PubMed/NCBI View Article : Google Scholar

94 

Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, Sanchez-Ramos J and Chopp M: Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 32:2682–2688. 2001.PubMed/NCBI View Article : Google Scholar

95 

Clark WM, Lessov NS, Dixon MP and Eckenstein F: Monofilament intraluminal middle cerebral artery occlusion in the mouse. Neurol Res. 19:641–648. 1997.PubMed/NCBI View Article : Google Scholar

96 

Niizuma K, Endo H and Chan PH: Oxidative stress and mitochondrial dysfunction as determinants of ischemic neuronal death and survival. J Neurochem. 109:133–138. 2009.PubMed/NCBI View Article : Google Scholar

97 

Bora KS and Sharma A: Neuroprotective effect of Artemisia absinthium L. on focal ischemia and reperfusion-induced cerebral injury. J Ethnopharmacol. 129:403–409. 2010.PubMed/NCBI View Article : Google Scholar

98 

Wang D, Yuan X, Liu T, Liu L, Hu Y, Wang Z and Zheng Q: Neuroprotective activity of lavender oil on transient focal cerebral ischemia in mice. Molecules. 17:9803–9817. 2012.PubMed/NCBI View Article : Google Scholar

99 

Dai J, Qiu YM, Ma ZW, Yan GF, Zhou J, Li SQ, Wu H, Jin YC and Zhang XH: Neuroprotective effect of baicalin on focal cerebral ischemia in rats. Neural Regen Res. 13:2129–2133. 2018.PubMed/NCBI View Article : Google Scholar

100 

Cao ZQ, Quan W, Hou SX, Guo C, Ma SB, Zhang W and Li X: The natural therapeutic magnesium lithospermate B potently provides neuroprotective effects on cerebral ischemia/reperfusion injury in rats. J Ethnopharmacol. 162:191–198. 2015.PubMed/NCBI View Article : Google Scholar

101 

Pengyue Z, Tao G, Hongyun H, Liqiang Y and Yihao D: Breviscapine confers a neuroprotective efficacy against transient focal cerebral ischemia by attenuating neuronal and astrocytic autophagy in the penumbra. Biomed Pharmacother. 90:69–76. 2017.PubMed/NCBI View Article : Google Scholar

102 

Guo H, Hu LM, Wang SX, Wang YL, Shi F, Li H, Liu Y, Kang LY and Gao XM: Neuroprotective effects of scutellarin against hypoxic-ischemic-induced cerebral injury via augmentation of antioxidant defense capacity. Chin J Physiol. 54:399–405. 2011.PubMed/NCBI View Article : Google Scholar

103 

Gaire BP and Kim HJ: Neuroprotective effects of Fructus Chebulae extracts on experimental models of cerebral ischemia. J Tradit Chin Med. 34:69–75. 2014.PubMed/NCBI View Article : Google Scholar

104 

Lee TH, Jung CH and Lee DH: Neuroprotective effects of Schisandrin B against transient focal cerebral ischemia in Sprague-Dawley rats. Food Chem Toxicol. 50:4239–4245. 2012.PubMed/NCBI View Article : Google Scholar

105 

He B, Chen P, Yang J, Yun Y, Zhang X, Yang R and Shen Z: Neuroprotective effect of 20(R)-ginsenoside Rg(3) against transient focal cerebral ischemia in rats. Neurosci Lett. 526:106–111. 2012.PubMed/NCBI View Article : Google Scholar

106 

Liu L, Vollmer MK, Fernandez VM, Dweik Y, Kim H and Doré SJ: Korean red ginseng pretreatment protects against long-term sensorimotor deficits after ischemic stroke likely through Nrf2. Front Cell Neurosci. 12(74)2018.PubMed/NCBI View Article : Google Scholar

107 

Dong X, Zheng L, Lu S and Yang YJ: Neuroprotective effects of pretreatment of ginsenoside Rb1 on severe cerebral ischemia-induced injuries in aged mice: Involvement of anti-oxidant signaling. Geriatr Gerontol Int. 17:338–345. 2017.PubMed/NCBI View Article : Google Scholar

108 

Chen LM, Zhou XM, Cao YL and Hu WX: Neuroprotection of ginsenoside Re in cerebral ischemia-reperfusion injury in rats. J Asian Nat Prod Res. 10:439–445. 2008.PubMed/NCBI View Article : Google Scholar

109 

Duan W, Wang L, Lv J, Gao K, Lu Y, Qin S, Ma X, Li J and Ge X: Metabolomics study on the effects of salvianolic acid B and borneol for treating cerebral ischemia in rats by ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry. Rejuvenation Res. 22:313–324. 2019.PubMed/NCBI View Article : Google Scholar

110 

Hong JT, Ryu SR, Kim HJ, Lee JK, Lee SH, Kim DB, Yun YP, Ryu JH, Lee BM and Kim PY: Neuroprotective effect of green tea extract in experimental ischemia-reperfusion brain injury. Brain Res Bull. 53:743–749. 2000.PubMed/NCBI View Article : Google Scholar

111 

Graham HN: Green tea composition, consumption, and polyphenol chemistry. Prev Med. 21:334–350. 1992.PubMed/NCBI View Article : Google Scholar

112 

Mukherjee PK, Ahamed KN, Kumar V, Mukherjee K and Houghton PJ: Protective effect of biflavones from Araucaria bidwillii Hook in rat cerebral ischemia/reperfusion induced oxidative stress. Behav Brain Res. 178:221–228. 2007.PubMed/NCBI View Article : Google Scholar

113 

Nazam Ansari M, Bhandari U, Islam F and Tripathi CD: Evaluation of antioxidant and neuroprotective effect of ethanolic extract of Embelia ribes Burm in focal cerebral ischemia/reperfusion-induced oxidative stress in rats. Fundam Clin Pharmacol. 22:305–314. 2008.PubMed/NCBI View Article : Google Scholar

114 

Ferreira Ede O, Fernandes MY, Lima NM, Neves KR, Carmo MR, Lima FA, Fonteles AA, Menezes AP and Andrade GM: Neuroinflammatory response to experimental stroke is inhibited by eriodictyol. Behav Brain Res. 312:321–332. 2016.PubMed/NCBI View Article : Google Scholar

115 

Lee D, Park J, Yoon J, Kim MY, Choi HY and Kim HJ: Neuroprotective effects of Eleutherococcus senticosus bark on transient global cerebral ischemia in rats. J Ethnopharmacol. 139:6–11. 2012.PubMed/NCBI View Article : Google Scholar

116 

Luo L, Kim SW, Lee HK, Kim ID, Lee H and Lee JK: Anti-Zn2+-toxicity of 4-hydroxybenzyl alcohol in astrocytes and neurons contribute to a robust neuroprotective effects in the postischemic brain. Cell Mol Neurobiol. 38:615–626. 2018.PubMed/NCBI View Article : Google Scholar

117 

Akhtar M, Maikiyo AM, Najmi AK, Khanam R, Mujeeb M and Aqil M: Neuroprotective effects of chloroform and petroleum ether extracts of Nigella sativa seeds in stroke model of rat. J Pharm Bioallied Sci. 5(119)2013.PubMed/NCBI View Article : Google Scholar

118 

Wang C, Zhang D, Ma H and Liu JJ: Neuroprotective effects of emodin-8-O-beta-d-glucoside in vivo and in vitro. Eur J Pharmacol. 577:58–63. 2007.PubMed/NCBI View Article : Google Scholar

119 

Guo C, Tong L, Xi M, Yang H, Dong H and Wen AJ: Neuroprotective effect of calycosin on cerebral ischemia and reperfusion injury in rats. Cell Physiol Biochem. 144:768–774. 2012.PubMed/NCBI View Article : Google Scholar

120 

Chang Y, Hsieh CY, Peng ZA, Yen TL, Hsiao G, Chou DS, Chen CM and Sheu JR: Neuroprotective mechanisms of puerarin in middle cerebral artery occlusion-induced brain infarction in rats. J Biomed Sci. 16(9)2009.PubMed/NCBI View Article : Google Scholar

121 

Meng X, Xie W, Xu Q, Liang T, Xu X, Sun G and Sun X: Neuroprotective effects of radix scrophulariae on cerebral ischemia and reperfusion injury via MAPK pathways. Molecules. 23(2401)2018.PubMed/NCBI View Article : Google Scholar

122 

Kaneko Y, Eve DJ, Yu S, Shojo H, Bae EC, Park DH, Roschek B Jr, Alberte RS, Sanberg PR, Sanberg CD, et al: Acute treatment with herbal extracts provides neuroprotective benefits in in vitro and in vivo stroke models, characterized by reduced ischemic cell death and maintenance of motor and neurological functions. Cell Med. 1:137–142. 2010.PubMed/NCBI View Article : Google Scholar

123 

Guo C, Yin Y, Duan J, Zhu Y, Yan J, Wei G, Guan Y, Wu X, Wang Y, Xi M and Wen A: Neuroprotective effect and underlying mechanism of sodium danshensu [3-(3,4-dihydroxyphenyl) lactic acid from Radix and Rhizoma Salviae miltiorrhizae=Danshen] against cerebral ischemia and reperfusion injury in rats. Phytomedicine. 22:283–289. 2015.PubMed/NCBI View Article : Google Scholar

124 

Lam BY, Lo AC, Sun X, Luo HW, Chung SK and Sucher NJ: Neuroprotective effects of tanshinones in transient focal cerebral ischemia in mice. Phytomedicine. 10:286–291. 2003.PubMed/NCBI View Article : Google Scholar

125 

Cui L, Zhang X, Yang R, Wang L, Liu L, Li M and Du W: Neuroprotection and underlying mechanisms of oxymatrine in cerebral ischemia of rats. Neurol Res. 33:319–324. 2011.PubMed/NCBI View Article : Google Scholar

126 

Park S, Nam K, Lee H, Cho EY, Koo U and Mar W: Neuroprotective effects of an alkaloid-free ethyl acetate extract from the root of Sophora flavescens Ait. against focal cerebral ischemia in rats. Phytomedicine. 16:1042–1051. 2009.PubMed/NCBI View Article : Google Scholar

127 

Li W, Yang Y, Hu Z, Ling S and Fang M: Neuroprotective effects of DAHP and Triptolide in focal cerebral ischemia via apoptosis inhibition and PI3K/Akt/mTOR pathway activation. Front Neuroanat. 9(48)2015.PubMed/NCBI View Article : Google Scholar

128 

Lee HF, Lee TS and Kou YR: Anti-inflammatory and neuroprotective effects of triptolide on traumatic brain injury in rats. Respir Physiol Neurobiol. 182:1–8. 2012.PubMed/NCBI View Article : Google Scholar

129 

Gupta S and Gupta YK: Combination of Zizyphus jujuba and silymarin showed better neuroprotective effect as compared to single agent in MCAo-induced focal cerebral ischemia in rats. J Ethnopharmacol. 197:118–127. 2017.PubMed/NCBI View Article : Google Scholar

130 

Chen JH, Kuo HC, Lee KF and Tsai TH: Magnolol protects neurons against ischemia injury via the downregulation of p38/MAPK, CHOP and nitrotyrosine. Toxicol Appl Pharmacol. 279:294–302. 2014.PubMed/NCBI View Article : Google Scholar

131 

Gong J, Sun F, Li Y, Zhou X, Duan Z, Duan F, Zhao L, Chen H, Qi S and Shen J: Momordica charantia polysaccharides could protect against cerebral ischemia/reperfusion injury through inhibiting oxidative stress mediated c-Jun N-terminal kinase 3 signaling pathway. Neuropharmacology. 91:123–134. 2015.PubMed/NCBI View Article : Google Scholar

132 

Bora KS, Arora S and Shri R: Role of Ocimum basilicum L. in prevention of ischemia and reperfusion-induced cerebral damage, and motor dysfunctions in mice brain. J Ethnopharmacol. 137:1360–1365. 2011.PubMed/NCBI View Article : Google Scholar

133 

Dringen R: Metabolism and functions of glutathione in brain. Prog Neurobiol. 62:649–671. 2000.PubMed/NCBI View Article : Google Scholar

134 

Siddiqui BS, Aslam H, Ali ST, Begu S and Khatoon N: Two new triterpenoids and a steroidal glycoside from the aerial parts of Ocimum basilicum. Chem Pharm Bull (Tokyo). 55:516–519. 2007.PubMed/NCBI View Article : Google Scholar

135 

Yanpallewar S, Rai S, Kumar M and Acharya SB: Evaluation of antioxidant and neuroprotective effect of Ocimum sanctum on transient cerebral ischemia and long-term cerebral hypoperfusion. Pharmacol Biochem Behav. 79:155–164. 2004.PubMed/NCBI View Article : Google Scholar

136 

Fki I, Sahnoun Z and Sayadi S: Hypocholesterolemic effects of phenolic extracts and purified hydroxytyrosol recovered from olive mill wastewater in rats fed a cholesterol-rich diet. J Agric Food Chem. 55:624–631. 2007.PubMed/NCBI View Article : Google Scholar

137 

Mohagheghi F, Bigdeli MR, Rasoulian B, Zeinanloo AA and Khoshbaten A: Dietary virgin olive oil reduces blood brain barrier permeability, brain edema, and brain injury in rats subjected to ischemia-reperfusion. ScientificWorldJournal. 10:180–191. 2010.PubMed/NCBI View Article : Google Scholar

138 

Rabiei Z, Bigdeli MR and Rasoulian B: Neuroprotection of dietary virgin olive oil on brain lipidomics during stroke. Curr Neurovasc Res. 10:231–237. 2013.PubMed/NCBI View Article : Google Scholar

139 

Bayat M, Azami Tameh A, Hossein Ghahremani M, Akbari M, Mehr SE, Khanavi M and Hassanzadeh G: Neuroprotective properties of Melissa officinalis after hypoxic-ischemic injury both in vitro and in vivo. Daru. 20(42)2012.PubMed/NCBI View Article : Google Scholar

140 

Rabiei Z and Rafieian-Kopaei M: Neuroprotective effect of pretreatment with Lavandula officinalis ethanolic extract on blood-brain barrier permeability in a rat stroke model. Asian Pac J Trop Med. 7S1:S421–S426. 2014.PubMed/NCBI View Article : Google Scholar

141 

Cao Y, Maoa X, Sun C, Zheng P, Gao J, Wang X, Min D, Sun H, Xie N and Cai J: Baicalin attenuates global cerebral ischemia/reperfusion injury in gerbils via anti-oxidative and anti-apoptotic pathways. Brain Res Bull. 85:396–402. 2011.PubMed/NCBI View Article : Google Scholar

142 

Han BH, D'Costa A, Back SA, Parsadanian M, Patel S, Shah AR, Gidday JM, Srinivasan A, Deshmukh M and Holtzman DM: BDNF blocks caspase-3 activation in neonatal hypoxia-ischemia. Neurobiol Dis. 7:38–53. 2000.PubMed/NCBI View Article : Google Scholar

143 

Zhang ZJ, Li P, Wang Z, Li PT, Zhang WS, Sun ZH, Zhang XJ and Wang YY: A comparative study on the individual and combined effects of baicalin and jasminoidin on focal cerebral ischemia-reperfusion injury. Brain Res. 1123:188–195. 2006.PubMed/NCBI View Article : Google Scholar

144 

Kim HJ, Lee SR and Moon KD: Ether fraction of methanol extracts of Gastrodia elata, medicinal herb protects against neuronal cell damage after transient global ischemia in gerbils. Phytother Res. 17:909–912. 2003.PubMed/NCBI View Article : Google Scholar

145 

Yu SJ, Kim JR, Lee CK, Han JE, Lee JH, Kim HS, Hong JH and Kang SG: Gastrodia elata blume and an active component, p-hydroxybenzyl alcohol reduce focal ischemic brain injury through antioxidant related gene expressions. Biol Pharm Bull. 28:1016–1020. 2005.PubMed/NCBI View Article : Google Scholar

146 

Joyeux M, Lobstein A, Anton R and Mortier F: Comparative antilipoperoxidant, antinecrotic and scavenging properties of terpenes and biflavones from Ginkgo and some flavonoids. Planta Med. 61:126–129. 1995.PubMed/NCBI View Article : Google Scholar

147 

Calapai G, Crupi A, Firenzuoli F, Marciano MC, Squadrito F, Inferrera G, Parisi A, Rizzo A, Crisafulli C, Fiore A and Caputi AP: Neuroprotective effects of Ginkgo biloba extract in brain ischemia are mediated by inhibition of nitric oxide synthesis. Life Sci. 67:2673–2683. 2000.PubMed/NCBI View Article : Google Scholar

148 

Yan XB, Wang SS, Hou HL, Ji R and Zhou JN: Lithium improves the behavioral disorder in rats subjected to transient global cerebral ischemia. Behav Brain Res. 177:282–289. 2007.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-October 2020
Volume 1 Issue 1

Print ISSN: 2634-7989
Online ISSN:2634-7237

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ezzelarab NM, Saleh N, Khalil EA and Abdellatif A: Experimental models and plant‑based therapy for experimental cerebral ischemia (Review). Int J Funct Nutr 1: 5, 2020
APA
Ezzelarab, N.M., Saleh, N., Khalil, E.A., & Abdellatif, A. (2020). Experimental models and plant‑based therapy for experimental cerebral ischemia (Review). International Journal of Functional Nutrition, 1, 5. https://doi.org/10.3892/ijfn.2020.5
MLA
Ezzelarab, N. M., Saleh, N., Khalil, E. A., Abdellatif, A."Experimental models and plant‑based therapy for experimental cerebral ischemia (Review)". International Journal of Functional Nutrition 1.1 (2020): 5.
Chicago
Ezzelarab, N. M., Saleh, N., Khalil, E. A., Abdellatif, A."Experimental models and plant‑based therapy for experimental cerebral ischemia (Review)". International Journal of Functional Nutrition 1, no. 1 (2020): 5. https://doi.org/10.3892/ijfn.2020.5