Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3680 2023-08-03 13:59:00 |
2 format correct Meta information modification 3680 2023-08-04 09:06:06 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Pluta, R.; Miziak, B.; Czuczwar, S.J. Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment. Encyclopedia. Available online: https://encyclopedia.pub/entry/47616 (accessed on 01 July 2024).
Pluta R, Miziak B, Czuczwar SJ. Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment. Encyclopedia. Available at: https://encyclopedia.pub/entry/47616. Accessed July 01, 2024.
Pluta, Ryszard, Barbara Miziak, Stanisław J. Czuczwar. "Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment" Encyclopedia, https://encyclopedia.pub/entry/47616 (accessed July 01, 2024).
Pluta, R., Miziak, B., & Czuczwar, S.J. (2023, August 03). Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment. In Encyclopedia. https://encyclopedia.pub/entry/47616
Pluta, Ryszard, et al. "Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment." Encyclopedia. Web. 03 August, 2023.
Honey and Its Ingredients in Post-Ischemic Neurodegeneration Treatment
Edit

Neurodegeneration of the brain after ischemia is a major cause of severe, long-term disability, dementia, and mortality, which is a global problem. These phenomena are attributed to excitotoxicity, changes in the blood–brain barrier, neuroinflammation, oxidative stress, vasoconstriction, cerebral amyloid angiopathy, amyloid plaques, neurofibrillary tangles, and ultimately neuronal death. In addition, genetic factors such as post-ischemic changes in genetic programming in the expression of amyloid protein precursor, β-secretase, presenilin-1 and -2, and tau protein play an important role in the irreversible progression of post-ischemic neurodegeneration. Numerous studies have shown that the high contents of flavonoids and phenolic acids in honey have antioxidant, anti-inflammatory, anti-apoptotic, anti-amyloid, anti-tau protein, anticholinesterase, serotonergic, and AMPAK activities, influencing signal transmission and neuroprotective effects.

brain ischemia Alzheimer’s disease proteinopathy honey excitotoxicity neuroinflammation apoptosis amyloid tau protein neurodegeneration

1. Introduction

Ischemic stroke is a disease characterized by high mortality, morbidity, recurrence, and low cure rate [1]. Worldwide, ischemic stroke is the leading cause of severe long-term disability and the second leading cause of death (5.5–6.0 million) after ischemic heart disease [2][3][4][5][6][7][8]. Ischemic stroke, caused by occlusion of an artery leading to the brain, is the most common form of stroke, accounting for roughly 85–90% of all strokes [3][5][8][9][10][11]. The incidence of stroke increases with age in both men and women [7], but in some countries, especially India and China, the incidence of stroke in people under 40 has recently increased, representing a serious problem for public health [12]. In the coming years, the global trend of extending life expectancy will cause a parallel increase in the incidence of stroke.
Approximately 70% of ischemic strokes and 87% of stroke-connected deaths and disability-adjusted life years occur in low- and middle-income countries [13]. In these countries, the number of ischemic stroke cases has more than doubled in the last four decades [13]. In contrast, over the last forty years, the incidence of ischemic stroke decreased by 42% in developed countries [13]. Ischemic stroke occurs on average 15 years earlier and causes more deaths in people in developing countries compared to people in developed countries [14]. As many as 84% cases of ischemic strokes in developing countries die within three years of stroke, compared with 16% in developed countries [13]. Current epidemiological statistics evaluate that approximately 14–17 million people suffer from ischemic stroke annually, of whom approximately half die within a year [2][3][4][5][6][7][8][15]. It is now believed that one in six people in the world will suffer a stroke in their lifetime [15]. It is also known that the number of post-ischemic patients all over the world has reached approximately 33 million [2][6]. Interestingly, in the 21st century, the number of cases of ischemic stroke in young adults has increased to about 2 million per year [16]. Due to aging and obesity among adults in the European community, it is estimated that by 2025, the incidence of stroke in this group will increase to 1.5 million [16].
There has been a global decline in age-adjusted mortality and stroke rates for the last 25 years, but the total number of stroke cases has increased as life expectancy has increased [10]. Ischemic stroke is the most common cause of brain ischemia in humans in developed countries, and the number of cases is 10 times higher than that of hemorrhagic stroke, while in developing countries this difference is much smaller [10]. Although ischemic stroke death rates are dropping, it is believed that up to 50% of stroke-connected deaths can be attributed to improper preventive or medicinal management of modifiable risk factors [10]. The risk of recurrence of ischemic stroke during the first month of recirculation is high; 1 in 25 patients have been shown to have a repeated stroke in this time frame [10]. According to the latest forecasts, the number of ischemic stroke cases will increase to 77 million in 2030 [2][6]. If the tendency of growing ischemic stroke indicators in the world continues, by 2030 there will be about 12 million deaths worldwide, 70 million patients will suffer a stroke, and more than 200 million disability-adjusted life years will be recorded annually [6].
Despite progress in diagnosis and symptomatic treatment of ischemic stroke, it is calculated that the number of strokes will more than double by 2050, and long-term disability following stroke will increase in equal measure due to demographic shift and the growing amount of stroke survivors [17][18]. It is estimated that 30–40% of ischemic strokes cases are cryptogenic, i.e., they have no known cause [9]. Symptoms usually vary depending on the extent of the stroke and the area of the brain affected, and include sensory and motor dysfunctions that are often permanent. Approximately 30–50% of stroke survivors do not return to functional independence [9].
Brain ischemia has been shown to trigger a sequence of phenomena called the “ischemic cascade” that can last from minutes to days [9][19]. These phenomena include energy failure, excitotoxicity, oxidative stress, disruption of the blood–brain barrier, neuroinflammation, and ultimately cell death [8][9][20]. Ischemic lesions cause cortical and subcortical infarcts, white matter damage, cerebral amyloid angiopathy, and microbleeds [21][22][23]. This additionally causes hypoperfusion, ischemia of adjacent structures, chronic neuroinflammation, accumulation of amyloid plaques and neurofibrillary tangles, gliosis, neuronal death, and finally brain atrophy [24][25][26][27][28][29][30]. Post-ischemic neurodegeneration commonly involves injury to the following brain regions: the cerebral cortex, temporal lobe, hippocampus, amygdala, entorhinal cortex, and parahippocampus. After ischemia, these regions are involved in memory and cognitive deficits, and their progressive neurodegeneration also triggers behavioral changes. Since most regions affected by pathology are related to both cognition and behavior, this makes behavioral changes strongly correlate with cognitive dysfunction. Ischemia-related cognitive impairment ranges from mild to severe, occurring in about 35–70% of survivors one-year post-stroke, with higher rates seen soon post-stroke [31][32][33][34][35][36]. Approximately 20% of patients with mild cognitive impairment post-stroke make a full recovery, with the highest rate of recovery seen soon after insult [36][37]. However, cognitive improvement without returning to pre-stroke levels is more common than fully recovery [36][38][39]. Interestingly, the risk of developing dementia in the future increases after ischemic brain damage, even in patients with transient cognitive impairment [36][40]. Brain ischemia has been shown to speed up the beginning of dementia by 10 years [41]. Approximately 8–13% of patients experience dementia soon after a first ischemic stroke and more than 40% after a second ischemic stroke [6][35][41]. In addition, the estimated progress of dementia in patients surviving 25 years post-stroke is approximately 48% [24][41].
It should be emphasized that patients experiencing an ischemic stroke have significant implications for caregivers, society, and the economy, especially in developing countries where the projected increase in the incidence of stroke is the highest. Not surprisingly, the socioeconomic influence of strokes is huge and increasing over time, with an annual cost in the EU of EUR 38 billion in 2012, EUR 45 billion in 2015 and EUR 60 billion in 2017 [42].
In view of the above data, there is an urgent need to develop new therapies capable of preventing or reducing brain ischemia related to Alzheimer’s disease proteinopathy damage. Currently, there are no causal treatments available that could prevent the disease or effectively treat its sequelae. It is understood that ischemic prophylaxis should be started as early as possible to reverse the natural progression of the disease. Currently, of course, reperfusion remains the only and immediate therapeutic option in ischemic stroke. At the present, despite limited effectiveness, thrombolysis with recombinant tissue plasminogen activator (rtPA) and thrombectomy are used in the therapy of acute stroke [5]. Less than 10% of ischemic stroke cases receive rtPA treatment [43]. However, the clinical profit is less than three percent due to the restricted time window for optimal medical therapy and the potential for extensive bleeding following drug administration [44][45]. In addition, reperfusion strategies, despite their limited effectiveness, are applicable only to a small percentage of patients due to the short time window of therapy, contraindications, and costs connected with maintaining the infrastructure for performing procedures [5]. In addition, the possibility of further therapeutic intervention after reperfusion procedures in the form of a neuroprotective effect is now proposed. Neuroprotection is not an alternate to thrombolytic treatment and thrombectomy, but aims to brake alternations in brain parenchyma and preclude the spread of damages to adjacent areas or structures. The idea of neuroprotection is promising in preclinical studies but has not been translated into clinical success [46][47]. A fresh notable example is nerinetide, which interferes with postsynaptic density protein 95, an excitatory neuronal protein. In experimental studies, it has shown promise in the therapy of cerebral ischemia, but has not shown treatment advantages in human stroke [47]. One of the reasons why neuroprotective strategies fail is that they mainly target neuronal cells. It should be added that the brain is composed of different cells, i.e., astrocytes, microglia, oligodendrocytes, endothelial cells, and pericytes, all of which influence the function and survival of neuronal cells by releasing different substances that act either positively or negatively. These cells are under-studied as therapeutic targets in post-ischemic stroke, but understanding of their post-ischemic behavior, both harmful and beneficial, continues to grow and is being intensively studied.

2. Therapeutic Potential of Honey and Its Ingredients in Post-Ischemic Neurodegeneration

Honey administered orally before and after brain ischemia reduced damaged pyramidal neurons in the hippocampus, and significantly improved spatial learning and memory performance [48][49].
Quercetin and its glycosides, including isoquercetin, have a beneficial effect on pathological changes in various models of ischemic brain injury [50][51][52][53][54][55][56][57][58]. The beneficial effect of quercetin results from its anti-inflammatory, anti-apoptotic, and antioxidant effects, and from inhibiting metalloproteinase activity, which prevents blood–brain barrier failure after cerebral ischemia [50][51][52][53][54][55][56][57][58]. It is suggested that the factor Nrf2 may be involved in the antioxidant and anti-apoptotic effects [50][51][52][53][54][55][56][57][58].
Myricetin has been studied for multiple medical effects, including anti-apoptotic, anti-inflammatory, and antioxidant properties [59][60]. Studies have shown that myricetin works against brain damage after local ischemia [59][60][61]. Among the proposed molecular mechanisms of myricetin action, inhibition of p38 MAPK and enhancement of AKT and Nrf2 factors have been indicated [60][62].
A protective effect has been demonstrated with kaempferol in transient local cerebral ischemia in rats, e.g., in reducing amyloid protein precursor [63][64][65]. Studies indicate that treatment with kaempferol after ischemia prevents the development of neuroinflammation by reducing the activation of NF-kB/RelA and STAT3 [63][64][65].
Naringenin exhibits neuroprotective properties in a model of cerebral ischemia, reducing apoptosis, inflammation, oxidative stress, and neurological deficits by modulating claudin-5, MMP9, and Nrf2 [66][67][68]. In addition, naringin, a naringenin-7-O-glycoside, prevented brain microvascular thrombosis in spontaneously hypertensive rats [69].
Luteolin has been shown to have a neuroprotective effect, i.e., anti-apoptotic and blood–brain barrier stabilization, on ischemic brain damage by increasing claudin-5 and inhibiting MMP9, reducing oxidative stress, and enhancing autophagy by activating the Nrf2 pathway and reducing inflammatory changes [70][71][72][73].
Administration of caffeic acid before or after ischemia had a protective effect on the brain by improving the neurological outcome in various models of cerebral ischemia [74][75][76][77][78]. The neuroprotection provided by this substance was probably mediated by the inhibition of 5-lipoxygenase and both antioxidant and anti-inflammatory effects [69][74][75][76][77][78].
The beneficial effect of ferulic acid was confirmed in animal models of global and local brain ischemia [79][80][81][82][83][84]. There are many indications that the neuroprotective effect of ferulic acid is related to the anti-inflammatory and neurotrophic effects related to a reduction in the activity of intercellular adhesion molecule-1, an increased level of erythropoietin in the brain, and granulocyte colony-stimulating factor [69][79][80][81][82][83][84]. In addition, it has been shown that ferulic acid extends the therapeutic window after focal cerebral ischemia, which is currently very useful in the clinic [81].
On the other hand, p-coumaric acid has shown neuroprotective effects in models of local and global cerebral ischemia by inhibiting apoptosis and the production of reactive oxygen species [85][86].
However, chlorogenic acid given before or after ischemia diminished infarct size, blood–brain barrier damage, and behavioral deficits in focal cerebral ischemia by affecting the activation of MMP, increasing the levels of erythropoietin, HIF-1α, and nerve growth factor in the brain [87][88][89][90][91][92][93]. In addition, the substance supported neuroprotection in rats by affecting the Nrf2 path in a model of brain ischemia induced by ligation of both common carotid arteries [89]. It is worth noting that the other substance contained in honey, chlorogenic acid in combination with rtPA, effectively reduced behavioral deficits in focal cerebral ischemia in rabbits and extended the time window of rtPA treatment [69][94].
Another honey ingredient, ellagic acid, had a protective effect after experimental ischemic brain injury by affecting the regulation of Bcl-2/Bax activity [95].
Gallic acid protects against experimental, transient focal, and global ischemic brain injury by decreasing oxidative stress with elevated antioxidant levels and reducing markers responsible for inflammatory answer [96][97][98][99][100]. The neuroprotective effect of gallic acid is attributed to its ability to enter the brain through the blood–brain barrier, directly decreasing the concentration of reactive oxygen and nitrogen species and chelating transition metal ions [100]. Gallic acid has been documented to interrupt the vicious cycle of oxidative stress during brain injury due to ischemia [96][97][98][99][100].
In addition to the primary anti-inflammatory, antioxidant, and anti-apoptotic potential shown above, most honey ingredients, such as luteolin, myricetin, naringenin, quercetin, kaempferol, caffeic acid, ellagic acid, ferulic acid, gallic acid, and p-coumaric acid, also have a therapeutic effect on the progression of neurodegeneration associated with amyloid pathology in Alzheimer’s disease [101] and ischemia-related brain neurodegeneration of Alzheimer’s disease proteinopathy [64][65][66]. Additionally, naringenin, quercetin, naringin, ellagic acid, and caffeic acid also decrease tau protein phosphorylation in the brain in models of Alzheimer’s disease [101]. Honey components reduce the expression of genes involved in amyloidogenic neurodegeneration, such as amyloid protein precursor, β-secretase, and presenilin 1 [101][102][103][104]. In addition, myricetin and ellagic acid prevent the production of amyloid by increasing the expression and activity of α-secretase, which leads to a decrease in the cleavage of the amyloid protein precursor to soluble amyloid, and thus prevents the synaptic deposition of the latter [102][103]. These data suggest that the phenolic ingredients of honey participate in the regulation of the expression of genes involved in the reduction of oxidative stress and the development of amyloid fibrils [101]. Also, flavonoids and phenolic acids elevate the expression of the Nrf2 transcription factor, which is in charge for the induction of antioxidant genes, thus improving protection against oxidative damage [101]. Additionally, by reducing the induction of inflammatory factors, honey ingredients also weaken the immune response of microglia and astrocytes in the hippocampus, entorhinal cortex, and amygdala. Further, honey ingredients reduce tau protein hyperphosphorylation, which prevents the development of neurofibrillary tangles [105] and reduces the production and accumulation of amyloid in the form of plaques [106][107]. They also appear to exert neuroprotective effects by preventing neuronal damage and apoptosis and regulating the cholinergic system, similar to curcumin analogs in scopolamine-induced amnesia, where they increase acetylcholine and choline acetyltransferase and decrease butyrylcholinesterase [104][108][109]. Chlorogenic acid, ellagic acid, caffeic acid, gallic acid, myricetin, naringenin, quercetin, and kaempferol lower the level of acetylcholinesterase [101] like curcumin analogs in an experimental model of amnesia [108][109]. Additionally, caffeic acid and gallic acid also reduce level of butyrylcholinesterase [101]. It has also been shown that the action of chlorogenic acid leads to a reduction in memory and cognitive deficits in humans [110]. Since honey contains many flavonoids and phenolic acids, it can be expected that its consumption and proper preparation as a medicinal substance will have great potential in the prevention and/or treatment of post-ischemic brain pathology with the Alzheimer’s disease phenotype. The therapeutic potential of honey and its ingredients in preclinical models of focal and global brain ischemia (regardless of whether the administration was started before, during, or after ischemia), its effective concentration, dose, and duration of treatment, and the main effects are presented in Table 1.
Table 1. Activity of honey and selected its flavonoids and phenolic acids in various models of brain ischemia.
Substance Model Treatment Effects References
    Honey    
Malaysian Tualang honey p2VO Pre: 1.2 g/kg for 10 days with Post: 10 weeks ↓ Hippocampal CA1 region damage ↑ Spatial learning, memory performance [48][50]
    Flavonoids    
Quercetin tMCAO Post: 20 mg/kg/d for 3 days ↓ Oxidative stress,
necrosis, apoptosis, brain edema, brain injury, neurological deficits
[54]
Quercetin pMCAO Post: 30 mg/kg single dose ↓ Brain injury [51]
Quercetin Photothrombotic model Post: 25 µmol/kg every 12 h for 3 days ↓ BBB injury, brain edema, neurological deficits
↑ Functional outcomes
[53]
Quercetin 2VO Pre: 50 mg/kg 30 min before and immediately
post-ischemia, then daily for 2 days
↓ BBB injury, delayed neuronal damage in CA1, CA2, brain injury [52]
Quercetin tMCAO Pre: 10 mg/kg 30 min before ↓ Neurological deficits, behavioral changes
↑ Parvalbumin expression
[58]
Quercetin tMCAO Pre: 10 mg/kg 1 h before ↓ Brain edema, damage in brain cortex, neurological deficits
↑ Thioredoxin, interaction of apoptosis signal-regulating kinase 1 and thioredoxin
[56]
Quercetin tMCAO Pre: 10 mg/kg 30 min before ↓ Infarct volume, neurological deficit
↑ Protein phosphatase 2A
[55]
Quercetin tMCAO Post: 10, 30, 50 mg/kg at the onset of reperfusion ↓ BBB injury, ROS, infarct volume, neurological deficit [59]
Quercetin pMCAO Pre: 10 mg/kg 1 h before ↓ Intracellular calcium overload, glutamate excitotoxicity, caspase-3. [57]
Myricetin tMCAO Pre: 20 mg/kg 2 h before and daily for 2 days after ischemia
Pre: 25 mg/kg daily for 7 days
↓ Oxidative stress,
apoptosis, neuronal loss, inflammation,
infarct volume, ROS,
neurological deficits
↑ Antioxidant enzymes, mitochondrial function, Nrf2 nuclear translocation, HO-1 expression
[60]
Myricetin pMCAO Pre: 1 mg/kg, 5 mg/kg, 25 mg/kg for 7 days ↓ IL-1β, IL-6, TNF-α, MDA, p38 MAPK,
NF-κB/p65, apoptosis, infarct area, neurological deficit
↑ GSH/GSSG ratio, SOD, phosphorylated AKT
[63]
Myricetin tMCAO Pre: 25 mg/kg for 7 days ↓ Excitotoxicity, oxidative stress, inflammation, apoptosis [62]
Kaempferol tMCAO Pre: 10,15 μmol/l 30 min before and
immediately after ischemia
Post: 7.5, 10 mg/kg single dose
Post: 25, 50, 100 mg/kg daily for 7 days
↓ Metalloproteinase, anti-laminin staining, nitrosative-oxidative stress, caspase-9, apoptosis, poly-(ADP-ribose) polymerase, amyloid protein precursor, glial fibrillary acidic protein, phosphorylated STAT3, NF-κB p65, nuclear content of NF-κB p65, tumor necrosis factor α, interleukin 1β, intercellular adhesion molecule 1, matrix metallopeptidase 9, inducible nitric oxide synthase, myeloperoxidase, neuroinflammation, BBB injury, microglia activity, brain injury, neurological deficits [64][65][71]
Naringenin pMACO Pre: 100 mg/kg daily for 4 days ↓ Neuroinflammation, edema, NOD2, RIP2, NF-κB, MMP-9, BBB injury, infarct volume, neurological deficits
↑ Claudin-5
[68]
Naringenin tMCAO Pre: 50 mg/kg daily for 21 days
Post: 80 µM single dose
↓ Apoptosis, oxidative stress, edema, NF-κB, myeloperoxidase, nitric oxide, cytokines, neuroinflammation, glial activation, injury volume, neurological
deficits
↑ Cortical neurons proliferation
[67][70]
Luteolin tMCAO Post: 20, 40, 80 mg/kg 0 and 12 h after ischemia ↓ Injury volume, edema, IL-1β, TNF-α, iNOS, COX-2, NF-κB, inflammation,
neurological deficits
↑ Nrf2, PPARγ.
[71]
Luteolin tMCAO Post: 5, 10, 25 mg/kg single dose ↓ Oxidative stress,
apoptosis, mRNA and protein of MMP9, infarct volume, neurological deficits
↑ PI3K/Akt
[72]
Luteolin pMCAO Post: 10, 25 mg/kg single dose post-ischemia ↓ MDA, Bax, oxidative stress, apoptosis, edema, infarct volume, neurological deficits
↑ SOD1, CAT, Bcl-2, claudin-5
[73]
Luteolin pMCAO Post: 5, 10 mg/kg 0 h and daily for 3 days survival ↓ Brain edema, TLR4, TLR5, p-p38, NF-κB infarct size, neurological deficit
↑Phospho-ERK
[74]
    Phenolic acids    
Caffeic acid tMCAO Pre: 10, 50 mg/kg 30 min before, 0, 1, 2 h, and every 12 h for 4 days after ischemia
Pre: 0.1, 1, 10 µg/kg 15 min before, single dose
↓ Neuroinflammation, leukotrienes, neuron loss, 5-lipoxygenase, astrocyte proliferation, infarct volume, brain atrophy, neurological dysfunction
↑ NO
[75][76]
Caffeic acid pMCAO Post: 10 µmol/kg daily for 7 days ↓ MDA, CAT, XO, oxidative stress, lipid peroxidation, infarct size, neurological deficits
↑ GSH, NO
[77]
Caffeic acid Global ischemia Post: 10, 30, 50 mg/kg single dose ↓ Hippocampus injury, NF-κBp65, MDA, 5-LO, oxidative stress, memory deficits
↑ SOD
[79]
Caffeic acid tMCAO Post: 3, 10, 30 mg/kg 0, 2 h after ischemia ↓ MMP-2, MMP-9, edema, damage in penumbra, infarct volume, sensory-motor deficits, behavioral deficits [78]
Ferulic
acid
Global ischemia Post: 28, 56, 112 mg/kg daily for 5 days ↓ Oxidative stress, mRNA caspase 3, mRNA Bax, hippocampus apoptosis, memory impairment
↑ mRNA Bcl-2, SOD
[85]
Ferulic
acid
tMCAO Post: 50, 100, 200 mg/kg daily for 7 days ↓ Hippocampus injury, neurological deficits
↑ In hippocampus, EPO and granulocyte colony-stimulating factor
[83]
Ferulic
acid
tMCAO Post: 100 mg/kg 0 h post-ischemia
Post: 100 mg/kg 2 h post-ischemia
Post: 100 mg/kg 24 h
Pre: 100 mg/kg 24 h before ischemia
Pre: 100 mg/kg 2 h before ischemia
Pretreatment 2 h before ischemia and posttreatment 2 h after ischemia ↓ Bax, astrocytosis, infarction volume [84]
Ferulic
acid
tMCAO Pre: 80, 100 mg/kg
Post: 100 mg/kg 30 min after ischemia
↓ Superoxide radicals, ICAM-1, NF-κB, infarct size, neurological deficits [80]
Ferulic
acid
tMCAO Post; 100 mg/kg 0 h after ischemia ↓ ICAM-1 mRNA, Mac-1 mRNA, Mac-1, 4-HNE, 8-OHdG positive cells, TUNEL positive cells, caspase 3, microglia activity, apoptosis, macrophages, oxidative stress, inflammation [81]
Ferulic
acid
tMCAO Post: 100 mg/kg 0 h, or 30 min or 2 h after ischemia ↓ PSD-95, nNOS, iNOS, nitrotyrosine, caspase-3, apoptosis, Bax, cytochrome c, MAP kinase
↑ Gamma-aminobutyric acid type B receptor, therapeutic window
[82]
P-coumaric
acid
pMCAO Post: 100 mg/kg single dose ↓ Oxidative damage, MDA, apoptosis, caspase-3, caspase-9, edema, infarct volume, neurological deficits
↑ SOD, NRF-1
[86]
P-coumaric
acid
Global ischemia Pre: 100 mg/kg for 2 weeks before ischemia ↓ MDA, oxidative stress, hippocampal neuronal death, infarct volume, brain damage
↑ Catalase, superoxide dismutase
[87]
Chlorogenic
acid
tMCAO Post: 3, 10, 30 mg/kg 0, 2 h after ischemia
Pre: 15, 30, 60 mg/kg for 1 week
↓ BBB, oxidative stress, MMP-2, MMP-9, edema, infarct volume, sensory-motor deficits, behavioral deficits [88][89]
Chlorogenic
acid
tMCAO Post: 30 mg/kg 2 h after ischemia ↓ Cytochrome c, caspase-3, cleaved caspase-3, neurological deficits
↑ Phospho-PDK1, phospho-Akt, phospho-Bad
[92]
Chlorogenic
acid
tMCAO Pre: 15, 30, 60 mg/kg once a day for 1 week ↓ Mortality, infarction area, injury of hippocampus, cortex lesions, neurological deficit
↑ EPO, HIF-1α, NGF
[89]
Chlorogenic
acid
Repeated global ischemia Post: 20, 100, 500 mg/kg single dose ↓ Oxidative stress,
apoptosis, MMPs, infarct volume, memory deficits
↑ SOD, GSH
[90]
Chlorogenic
acid
Embolic strokes with rtPA Post: 50 mg/kg 5 min, 1, 3 h after ischemia ↓ Behavioral deficits
↑ Therapeutic window
[95]
Chlorogenic
acid
tMCAO Post: 30 mg/kg 2 h after ischemia ↓ TUNEL-positive cells, caspase-3 and -7, oxidative stress, edema, infarct size, neurological damage [91]
Chlorogenic
acid
tMCAO Post: 30 mg/kg 2 h post-ischemia ↓ Reactive oxygen species, oxidative stress, NF-κB, IL-1β, TNF-α, microglia, astrocyte activation, inflammation, cortex pathology [94]
Chlorogenic
acid
tMCAO Post: 30 mg/kg/d 3 days after ischemia ↓ Cerebral cortex apoptosis, infarct volume
↑ Angiogenesis, VEGFA, PI3K/Akt signaling
[92]
Gallic acid tMCAO Pre: 50 mg/kg daily for 7 days
Pre: 50 mg/kg single dose
↓ Oxidative stress, apoptosis, neuroinflammation, mitochondrial
dysfunction, injury size, neurological deficits
[97][99]
Gallic acid Global ischemia Pre: 100 mg/kg/d for 10 days ↓ BBB injury, MDA, oxidative stress, hippocampus EEG changes, anxiety,
behavioral deficits
[100]
Gallic acid Global ischemia Post: 25, 50 mg/kg/d for 1 week ↓ Oxidative stress, depressive symptoms [98]
Ellagic
acid
Photothrombotic model Pre: 10, 30 mg/kg 24 h before and 0 h post-ischemia ↓ Apoptotic cells, infarct size, neurological deficits [96]
P2VO—permanent occlusion of both common carotid arteries, tMCAO—transient middle cerebral artery occlusion, pMCAO—permanent middle cerebral artery occlusion, BBB—blood–brain barrier, ROS—reactive oxygen species, Nrf2—nuclear factor erythroid 2-related factor 2, HO-1—heme oxygenase 1, STAT3—signal transducer and activator of transcription 3, NF-κB p65—nuclear-kappa B factor p65, NOD2—nucleotide-binding oligomerization domain containing 2, RIP2—receptor-interacting-serine/threonine-protein kinase 2, MMP-9—matrix metallopeptidase-9, IL-1β—interleukin-1 beta, IL-6—interleukin-6, MDA—malondialdehyde, p38 MAPK—p38 mitogen activated protein kinase, GSH/GSSG—glutathione/glutathione disulfide, SOD—superoxide dismutase, AKT—protein kinase B, TNF-α—tumor necrosis factor-α, MAPKs—mitogen-activated protein kinases, ERK—extracellular signal-regulated kinase, iNOS—inducible nitric oxide synthase, COX-2—cyclooxygenase-2, PPARγ—peroxisome proliferator-activated receptor gamma, PI3K—phosphatidylinositol 3-kinase, CAT—catalase, TLR4—toll-like receptor 4, p-p38—phospho-p38, XO—xanthine oxidase, NO—nitric oxide, 5-LO—5-lipoxygenase, EPO—erythropoietin, ICAM-1—intercellular adhesion molecule 1, NRF1—nuclear respiratory factor 1, VEGFA—vascular endothelial growth factor A, EEG—electroencephalography. ↓—decrease, ↑—increase.

References

  1. Mandzia, J.; Cipriano, L.E.; Kapral, M.K.; Fang, J.; Hachinski, V.; Sposato, L.A. Intravenous thrombolysis after first-ever ischemic stroke and reduced incident dementia rate. Stroke 2022, 53, 1170–1177.
  2. Bejot, Y.; Daubail, B.; Giroud, M. Epidemiology of stroke and transient ischemic attacks: Current knowledge and perspectives. Rev. Neurol. 2016, 172, 59–68.
  3. Xu, S.; Lu, J.; Shao, A.; Zhang, J.H.; Zhang, J. Glial Cells: Role of the immune response in ischemic stroke. Front. Immunol. 2020, 11, 294.
  4. Feigin, V.L.; Stark, B.A.; Johnson, C.O.; Roth, G.A.; Bisignano, C.; Abady, G.G.; Abbasifard, M.; Abbasi-Kangevari, M.; Abd-Allah, F.; Abedi, V.; et al. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820.
  5. Patabendige, A.; Singh, A.; Jenkins, S.; Sen, J.; Chen, R. Astrocyte activation in neurovascular damage and repair following ischaemic stroke. Int. J. Mol. Sci. 2021, 22, 4280.
  6. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Neuroinflammation in post-ischemic neurodegeneration of the brain: Friend, foe, or both? Int. J. Mol. Sci. 2021, 22, 4405.
  7. Virani, S.S.; Alonso, A.; Aparicio, H.J.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Cheng, S.; Delling, F.N.; et al. Heart disease and stroke statistics—2021 update: A report from the American heart association. Circulation 2021, 143, e254–e743.
  8. Dang, H.; Mao, W.; Wang, S.; Sha, J.; Lu, M.; Cong, L.; Meng, X.; Li, H. Systemic inflammation response index as a prognostic predictor in patients with acute ischemic stroke: A propensity score matching analysis. Front. Neurol. 2023, 13, 1049241.
  9. Hernández, I.H.; Villa-González, M.; Martín, G.; Soto, M.; Pérez-Álvarez, M.J. Glial Cells as therapeutic approaches in brain ischemia-reperfusion injury. Cells 2021, 10, 1639.
  10. Kamarova, M.; Baig, S.; Patel, H.; Monks, K.; Wasay, M.; Ali, A.; Redgrave, J.; Majid, A.; Bell, S.M. Antiplatelet use in ischemic stroke. Ann. Pharmacother. 2022, 56, 1159–1173.
  11. Wang, Y.; Leak, R.K.; Cao, G. Microglia-mediated neuroinflammation and neuroplasticity after stroke. Front. Cell Neurosci. 2022, 16, 980722.
  12. Venketasubramanian, N.; Yoon, B.W.; Pandian, J.; Navarro, J.C. Stroke epidemiology in south, east, and south-east Asia: A review. J. Stroke 2017, 19, 286–294.
  13. Johnson, W.; Onuma, O.; Owolabi, M.; Sachdev, S. Stroke: A global response is needed. Bull. World Health Organ. 2016, 94, 634.
  14. Owolabi, M.O.; Akarolo-Anthony, S.; Akinyemi, R.; Arnett, D.; Gebregziabher, M.; Jenkins, C.; Tiwari, H.; Arulogun, O.; Akpalu, A.; Sarfo, F.S.; et al. Members of the H3 Africa Consortium. Members of the H3 Africa Consortium. The burden of stroke in Africa: A glance at the present and a glimpse into the future. Cardiovasc. J. Afr. 2015, 26 (Suppl. 1), S27–S38.
  15. Farina, M.; Vieira, L.E.; Buttari, B.; Profumo, E.; Saso, L. The Nrf2 pathway in ischemic stroke: A review. Molecules 2021, 26, 5001.
  16. Bulygin, K.V.; Beeraka, N.M.; Saitgareeva, A.R.; Nikolenko, V.N.; Gareev, I.; Beylerli, O.; Akhmadeeva, L.R.; Mikhaleva, L.M.; Torres Solis, L.F.; Solís Herrera, A.; et al. Can miRNAs be considered as diagnostic and therapeutic molecules in ischemic stroke pathogenesis?—Current Status. Int. J. Mol. Sci. 2020, 21, 6728.
  17. Howard, G.; Goff, D.C. Population shifts and the future of stroke: Forecasts of the future burden of stroke. Ann. N. Y. Acad. Sci. 2012, 1268, 14–20.
  18. Simats, A.; Liesz, A. Systemic inflammation after stroke: Implications for post-stroke comorbidities. EMBO Mol. Med. 2022, 14, e16269.
  19. Xing, C.; Arai, K.; Lo, E.H.; Hommel, M. Pathophysiologic cascades in ischemic stroke. Int. J. Stroke 2012, 7, 378–385.
  20. Kim, J.H.; Kim, S.Y.; Kim, B.; Lee, S.R.; Cha, S.H.; Lee, D.S.; Lee, H.J. Prospects of therapeutic target and directions for ischemic stroke. Pharmaceuticals 2021, 14, 321.
  21. Goulay, R.; Mena Romo, L.; Hol, E.M.; Dijkhuizen, R.M. From stroke to dementia: A Comprehensive review exposing tight interactions between stroke and amyloid-β formation. Transl. Stroke Res. 2020, 11, 601–614.
  22. Rost, N.S.; Brodtmann, A.; Pase, M.P.; van Veluw, S.J.; Biffi, A.; Duering, M.; Hinman, J.D.; Dichgans, M. Post-stroke cognitive impairment and dementia. Circ. Res. 2022, 130, 1252–1271.
  23. Chen, Y.; Ye, M. Risk factors and their correlation with severity of cerebral microbleed in acute large artery atherosclerotic cerebral infarction patients. Clin. Neurol. Neurosurg. 2022, 221, 107380.
  24. Snowdon, D.A.; Greiner, L.H.; Mortimer, J.A.; Riley, K.P.; Greiner, P.A.; Markesbery, W.R. Brain infarction and the clinical expression of Alzheimer disease: The Nun Study. JAMA 1997, 277, 813–817.
  25. Van Groen, T.; Puurunen, K.; Mäki, H.M.; Sivenius, J.; Jolkkonen, J. Transformation of diffuse beta-amyloid precursor protein and beta-amyloid deposits to plaques in the thalamus after transient occlusion of the middle cerebral artery in rats. Stroke 2005, 36, 1551–1556.
  26. Qi, J.; Wu, H.; Yang, Y.; Wand, D.; Chen, Y.; Gu, Y.; Liu, T. Cerebral ischemia and Alzheimer’s disease: The expression of amyloid-β and apolipoprotein E in human hippocampus. J. Alzheimers Dis. 2007, 12, 335–341.
  27. Pluta, R.; Ułamek, M.; Jabłoński, M. Alzheimer’s mechanisms in ischemic brain degeneration. Anat. Rec. 2009, 292, 1863–1881.
  28. Sekeljic, V.; Bataveljic, D.; Stamenkovic, S.; Ułamek, M.; Jabłoński, M.; Radenovic, L.; Pluta, R.; Andjus, P.R. Cellular markers of neuroinflammation and neurogenesis after ischemic brain injury in the long-term survival rat model. Brain Struct. Funct. 2012, 217, 411–420.
  29. Hatsuta, H.; Takao, M.; Nogami, A.; Uchino, A.; Sumikura, H.; Takata, T.; Morimoto, S.; Kanemaru, K.; Adachi, T.; Arai, T.; et al. Tau and TDP-43 accumulation of the basal nucleus of Meynert in individuals with cerebral lobar infarcts or hemorrhage. Acta Neuropathol. Commun. 2019, 7, 49.
  30. Radenovic, L.; Nenadic, M.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J.; Andjus, P.R.; Pluta, R. Heterogeneity in brain distribution of activated microglia and astrocytes in a rat ischemic model of Alzheimer’s disease after 2 years of survival. Aging 2020, 12, 12251–12267.
  31. Ihle-Hansen, H.; Thommessen, B.; Wyller, T.B.; Engedal, K.; Oksengard, A.R.; Stenset, V.; Loken, K.; Aaberg, M.; Fure, B. Incidence and subtypes of MCI and dementia 1 year after first-ever stroke in patients without pre-existing cognitive impairment. Dement. Geriatr. Cogn. Disord. 2011, 32, 401–407.
  32. Douiri, A.; Rudd, A.G.; Wolfe, C.D. Prevalence of poststroke cognitive impairment: South London Stroke Register 1995–2010. Stroke 2013, 44, 138–145.
  33. Jacquin, A.; Binquet, C.; Rouaud, O.; Graule-Petot, A.; Daubail, B.; Osseby, G.V.; Bonithon-Kopp, C.; Giroud, M.; Bejot, Y. Post-stroke cognitive impairment: High prevalence and determining factors in a cohort of mild stroke. J. Alzheimers Dis. 2014, 40, 1029–1038.
  34. Lo, J.W.; Crawford, J.D.; Desmond, D.W.; Godefroy, O.; Jokinen, H.; Mahinrad, S.; Bae, H.J.; Lim, J.S.; Kohler, S.; Douven, E.; et al. Stroke and Cognition (STROKOG) Collaboration. Profile of and risk factors for poststroke cognitive impairment in diverse ethnoregional groups. Neurology 2019, 93, e2257–e2271.
  35. Hashim, S.; Ahmad, S.; Al Hatamleh, M.A.I.; Mustafa, M.Z.; Mohamed, M.; Mohamud, R.; Kadir, R.; Kub, T.N.T. Trigona honey as a potential supplementary therapy to halt the progression of post-stroke vascular cognitive impairment. Int. Med. J. 2021, 28, 335–338.
  36. El Husseini, N.; Katzan, I.L.; Rost, N.S.; Blake, M.L.; Byun, E.; Pendlebury, S.T.; Aparicio, H.J.; Marquine, M.J.; Gottesman, R.F.; Smith, E.E.; et al. cognitive impairment after ischemic and hemorrhagic stroke: A scientific statement from the american heart association/american stroke association. Stroke 2023, 54, e272–e291.
  37. Rasquin, S.M.; Lodder, J.; Verhey, F.R. Predictors of reversible mild cognitive impairment after stroke: A 2-year follow-up study. J. Neurol. Sci. 2005, 229–230, 21–25.
  38. Liu, G.; Xie, W.; He, A.D.; Da, X.W.; Liang, M.L.; Yao, G.Q.; Xiang, J.Z.; Gao, C.J.; Ming, Z.Y. Antiplatelet activity of chrysin via inhibiting platelet αIIbβ3-mediated signaling pathway. Mol. Nutr. Food Res. 2016, 60, 1984–1993.
  39. Dichgans, M.; Leys, D. Vascular cognitive impairment. Circ. Res. 2017, 120, 573–591.
  40. Pendlebury, S.T.; Wadling, S.; Silver, L.E.; Mehta, Z.; Rothwell, P.M. Transient cognitive impairment in TIA and minor stroke. Stroke 2011, 42, 3116–3121.
  41. Elman-Shina, K.; Efrati, S. Ischemia as a common trigger for Alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 1012779.
  42. Neurology, T.L. A unified European action plan on stroke. Lancet Neurol. 2020, 19, 963.
  43. Tao, T.; Liu, M.; Chen, M.; Luo, Y.; Wang, C.; Xu, T.; Jiang, Y.; Guo, Y.; Zha, J.H. Natural medicine in neuroprotection for ischemic stroke: Challenges and prospective. Pharmacol. Ther. 2020, 216, 107695.
  44. Kremers, F.; Venema, E.; Duvekot, M.; Yo, L.; Bokkers, R.; Lycklama, À.; Nijeholt, G.; van Es, A.; van der Lugt, A.; Majoie, C.; et al. Outcome prediction models for endovascular treatment of ischemic stroke: Systematic review and external validation. Stroke 2022, 53, 825–836.
  45. Xiong, X.Y.; Liu, L.; Yang, Q.W. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog. Neurobiol. 2016, 142, 23–44.
  46. Herson, P.S.; Traystman, R.J. Animal models of stroke: Translational potential at present and in 2050. Future Neurol. 2014, 9, 541–551.
  47. Hill, M.D.; Goyal, M.; Menon, B.K.; Nogueira, R.G.; McTaggart, R.A.; Demchuk, A.M.; Poppe, A.Y.; Buck, B.H.; Field, T.S.; Dowlatshahi, D.; et al. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPE-NA1): A multicentre, double-blind, randomised controlled trial. Lancet 2020, 395, 878–887.
  48. Saxena, A.K.; Phyu, H.P.; Al-Ani, I.M.; Talib, N.A. Potential protective effect of honey against chronic cerebral hypoperfusion-induced neurodegeneration in rats. J. Anat. Soc. India 2014, 63, 151–155.
  49. Saxena, A.K.; Phyu, H.; Al-Ani, I.; Oothuman, P. Improved spatial learning and memory performance following tualang honey treatment during cerebral hypoperfusion-induced neurodegeneration. J. Transl. Sci. 2016, 2, 264–271.
  50. Rivera, F.; Urbanavicius, J.; Gervaz, E.; Morquio, A.; Dajas, F. Some aspects of the in vivo neuroprotective capacity of flavonoids: Bioavailability and structure-activity relationship. Neurotox. Res. 2004, 6, 543–553.
  51. Cho, J.Y.; Kim, I.S.; Jang, Y.H.; Kim, A.R.; Lee, S.R. Protective effect of quercetin, a natural flavonoid against neuronal damage after transient global cerebral ischemia. Neurosci. Lett. 2006, 404, 330–335.
  52. Lee, J.K.; Kwak, H.J.; Piao, M.S.; Jang, J.W.; Kim, S.H.; Kim, H.S. Quercetin reduces the elevated matrix metalloproteinases-9 level and improves functional outcome after cerebral focal ischemia in rats. Acta Neurochir. 2011, 153, 1321–1329.
  53. Dai, Y.; Zhang, H.; Zhang, J.; Yan, M. Isoquercetin attenuates oxidative stress and neuronal apoptosis after ischemia/reperfusion injury via Nrf2-mediated inhibition of the NOX4/ROS/NF-κB pathway. Chem. Biol. Interact. 2018, 284, 32–40.
  54. Park, D.J.; Kang, J.B.; Shah, M.A.; Koh, P.O. Quercetin alleviates the injury-induced decrease of protein phosphatase 2A subunit B in cerebral ischemic animal model and glutamate-exposed HT22 cells. J. Vet. Med. Sci. 2019, 81, 1047–1054.
  55. Park, D.J.; Kang, J.B.; Shah, F.A.; Jin, Y.B.; Koh, P.O. Quercetin attenuates decrease of thioredoxin expression following focal cerebral ischemia and glutamate-induced neuronal cell Damage. Neuroscience 2020, 428, 38–49.
  56. Park, D.J.; Jeonm, S.J.; Kang, J.B.; Koh, P.O. Quercetin Reduces Ischemic Brain Injury by Preventing Ischemia-induced decreases in the neuronal calcium sensor protein hippocalcin. Neuroscience 2020, 430, 47–62.
  57. Park, D.J.; Kang, J.B.; Shah, F.A.; Koh, P.O. Quercetin attenuates the reduction of parvalbumin in middle cerebral artery occlusion animal model. Lab. Anim. Res. 2021, 37, 9.
  58. Yang, R.; Shen, Y.-J.; Chen, M.; Zhao, J.-Y.; Chen, S.-H.; Zhang, W.; Song, J.-K.; Li, L.; Du, G.-H. Quercetin attenuates ischemia reperfusion injury by protecting the blood-brain barrier through Sirt1 in MCAO rats. J. Asian Nat. Prod. Res. 2021, 24, 278–289.
  59. Wu, S.; Yue, Y.; Peng, A.; Zhang, L.; Xiang, J.; Cao, X.; Ding, H.; Yin, S. Myricetin ameliorates brain injury and neurological deficits via Nrf2 activation after experimental stroke in middle-aged rats. Food Funct. 2016, 7, 2624–2634.
  60. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Myricetin as a Promising Molecule for the Treatment of Post-Ischemic Brain Neurodegeneration. Nutrients 2021, 13, 342.
  61. Zhang, Y.; Fu, K.; Wang, C.; Ma, C.; Gong, L.; Zhou, H.; Xue, X.; Peng, C.; Li, Y. Protective effects of dietary quercetin on cerebral ischemic injury: Pharmacology, pharmacokinetics and bioavailability-enhancing nanoformulations. Food Funct. 2023, 14, 4470–4489.
  62. Sun, L.; Xu, P.; Fu, T.; Huang, X.; Song, J.; Chen, M.; Tian, X.; Yin, H.; Han, J. Myricetin against ischemic cerebral injury in rat middle cerebral artery occlusion model. Mol. Med. Rep. 2018, 17, 3274–3280.
  63. López-Sánchez, C.; Martín-Romero, F.J.; Sun, F.; Luis, L.; Samhan-Arias, A.K.; García-Martínez, V.; Gutiérrez-Merino, C. Blood micromolar concentrations of kaempferol afford protection against ischemia/reperfusion-induced damage in rat brain. Brain Res. 2007, 1182, 123–137.
  64. Yu, L.; Chen, C.; Wang, L.F.; Kuang, X.; Liu, K.; Zhang, H.; Du, J.R. Neuroprotective effect of kaempferol glycosides against brain injury and neuroinflammation by inhibiting the activation of NF-κB and STAT3 in transient focal stroke. PLoS ONE 2013, 8, e55839.
  65. Li, W.H.; Cheng, X.; Yang, Y.L.; Liu, M.; Zhang, S.S.; Wang, Y.H.; Du, G.H. Kaempferol attenuates neuroinflammation and blood brain barrier dysfunction to improve neurological deficits in cerebral ischemia/reperfusion rats. Brain Res. 2019, 1722, 146361.
  66. Raza, S.S.; Khan, M.M.; Ahmad, A.; Ashafaq, M.; Islam, F.; Wagner, A.P.; Safhi, M.M.; Islam, F. Neuroprotective effect of naringenin is mediated through suppression of NF-κB signaling pathway in experimental stroke. Neuroscience 2013, 230, 157–171.
  67. Bai, X.; Zhang, X.; Chen, L.; Zhang, J.; Zhang, L.; Zhao, X.; Zhao, T.; Zhao, Y. Protective effect of naringenin in experimental ischemic stroke: Down-regulated NOD2. RIP2. NF-κB. MMP-9 and up-regulated claudin-5 expression. Neurochem. Res. 2014, 39, 1405–1415.
  68. Wang, K.; Chen, Z.; Huang, J.; Huang, L.; Luo, N.; Liang, X.; Liang, M.; Xie, W. Naringenin prevents ischaemic stroke damage via anti-apoptotic and anti-oxidant effects. Clin. Exp. Pharmacol. Physiol. 2017, 44, 862–871.
  69. Parrella, E.; Gussago, C.; Porrini, V.; Benarese, M.; Pizzi, M. From preclinical stroke models to humans: Polyphenols in the prevention and treatment of stroke. Nutrients 2020, 29, 85.
  70. Li, Q.; Tian, Z.; Wang, M.; Kou, J.; Wang, C.; Rong, X.; Li, J.; Xie, X.; Pang, X. Luteoloside attenuates neuroinflammation in focal cerebral ischemia in rats via regulation of the PPAR/Nrf2/NF-κB signaling pathway. Int. Immunopharmacol. 2019, 66, 309–316.
  71. Luo, S.; Li, H.; Mo, Z.; Lei, J.; Zhu, L.; Huang, Y.; Fu, R.; Li, C.; Huang, Y.; Liu, K.; et al. Connectivity map identifies luteolin as a treatment option of ischemic stroke by inhibiting MMP9 and activation of the PI3K/Akt signaling pathway. Exp. Mol. Med. 2019, 51, 1–11.
  72. Qiao, H.; Dong, L.; Zhang, X.; Zhu, C.; Wang, L.; Liu, Z.; Chen, L.; Xing, Y.; Wang, C.; Li, Y. Protective effect of luteolin in experimental ischemic stroke: Upregulated SOD1. CAT. Bcl-2 and claudin-5. down-regulated MDA and Bax expression. Neurochem. Res. 2012, 37, 2014–2024.
  73. Qiao, H.; Zhang, X.; Zhu, C.; Dong, L.; Wang, L.; Xing, Y.; Wang, C.; Ji, Y.; Cao, X. Luteolin downregulates TLR4. TLR5. NF-kappaB and p-p38MAPK expression. upregulates the p-ERK expression. and protects rat brains against focal ischemia. Brain Res. 2012, 1448, 71–81.
  74. Tsai, S.K.; Lin, M.J.; Liao, P.H.; Yang, C.Y.; Lin, S.M.; Liu, S.M.; Lin, R.H.; Chih, C.L.; Huang, S.S. Caffeic acid phenethyl ester ameliorates cerebral infarction in rats subjected to focal cerebral ischemia. Life Sci. 2006, 78, 2758–2762.
  75. Zhou, Y.; Fang, S.H.; Ye, Y.L.; Chu, L.S.; Zhang, W.P.; Wang, M.L.; Wei, E.Q. Caffeic acid ameliorates early and delayed brain injuries after focal cerebral ischemia in rats. Acta Pharmacol. Sin. 2006, 27, 1103–1110.
  76. Altug, M.E.; Serarslan, Y.; Bal, R.; Kontaş, T.; Ekici, F.; Melek, I.M.; Aslan, H.; Duman, T. Caffeic acid phenethyl ester protects rabbit brains against permanent focal ischemia by antioxidant action: A biochemical and planimetric study. Brain Res. 2008, 1201, 135–142.
  77. Lee, K.; Lee, B.J.; Bu, Y. Protective Effects of Dihydrocaffeic Acid, a Coffee Component Metabolite, on a Focal Cerebral Ischemia Rat Model. Molecules 2015, 20, 11930–11940.
  78. Liang, G.; Shi, B.; Luo, W.; Yang, J. The protective effect of caffeic acid on global cerebral ischemia-reperfusion injury in rats. Behav. Brain Funct. 2015, 11, 18.
  79. Cheng, C.Y.; Ho, T.Y.; Lee, E.J.; Su, S.Y.; Tang, N.Y.; Hsieh, C.L. Ferulic acid reduces cerebral infarct through its antioxidative and anti-inflammatory effects following transient focal cerebral ischemia in rats. Am. J. Chin. Med. 2008, 36, 1105–1119.
  80. Cheng, C.Y.; Su, S.Y.; Tang, N.Y.; Ho, T.Y.; Chiang, S.Y.; Hsieh, C.L. Ferulic acid provides neuroprotection against oxidative stress-related apoptosis after cerebral ischemia/reperfusion injury by inhibiting ICAM-1 mRNA expression in rats. Brain Res. 2008, 1209, 136–150.
  81. Cheng, C.Y.; Su, S.Y.; Tang, N.Y.; Ho, T.Y.; Lo, W.Y.; Hsieh, C.L. Ferulic acid inhibits nitric oxide-induced apoptosis by enhancing GABA(B1) receptor expression in transient focal cerebral ischemia in rats. Acta Pharmacol. Sin. 2010, 31, 889–899.
  82. Zhang, L.; Wang, H.; Wang, T.; Jiang, N.; Yu, P.; Chong, Y.; Fu, F. Ferulic acid ameliorates nerve injury induced by cerebral ischemia in rats. Exp. Ther. Med. 2015, 9, 972–976.
  83. Cheng, C.Y.; Tang, N.Y.; Kao, S.T.; Hsieh, C.L. Ferulic Acid Administered at Various Time Points Protects against Cerebral Infarction by Activating p38 MAPK/p90RSK/CREB/Bcl-2 Anti-Apoptotic Signaling in the Subacute Phase of Cerebral Ischemia-Reperfusion Injury in Rats. PLoS ONE 2016, 11, e0155748.
  84. Ren, Z.; Zhang, R.; Li, Y.; Li, Y.; Yang, Z.; Yang, H. Ferulic acid exerts neuroprotective effects against cerebral ischemia/reperfusioninduced injury via antioxidant and anti-apoptotic mechanisms in vitro and in vivo. Int. J. Mol. Med. 2017, 40, 1444–1456.
  85. Guven, M.; Aras, A.B.; Akman, T.; Sen, H.M.; Ozkan, A.; Salis, O.; Sehitoglu, I.; Kalkan, Y.; Silan, C.; Deniz, M.; et al. Neuroprotective effect of p-coumaric acid in rat model of embolic cerebral ischemia. Iran. J. Basic Med. Sci. 2015, 18, 356–363.
  86. Sakamula, R.; Thong-Asa, W. Neuroprotective effect of p-coumaric acid in mice with cerebral ischemia reperfusion injuries. Metab. Brain Dis. 2018, 33, 765–773.
  87. Lee, K.; Lee, J.S.; Jang, H.J.; Kim, S.M.; Chang, M.S.; Park, S.H.; Kim, K.S.; Bae, J.; Park, J.W.; Lee, B.; et al. Chlorogenic acid ameliorates brain damage and edema by inhibiting matrix metalloproteinase-2 and 9 in a rat model of focal cerebral ischemia. Eur. J. Pharmacol. 2012, 689, 89–95.
  88. Miao, M.; Cao, L.; Li, R.; Fang, X.; Miao, Y. Protective effect of chlorogenic acid on the focal cerebral ischemia reperfusion rat models. Saudi Pharm. J. 2017, 25, 556–563.
  89. Liu, D.; Wang, H.; Zhang, Y.; Zhang, Z. Protective Effects of Chlorogenic Acid on Cerebral Ischemia/Reperfusion Injury Rats by Regulating Oxidative Stress-Related Nrf2 Pathway. Drug Des. Dev. Ther. 2020, 14, 51–60.
  90. Shah, M.A.; Kang, J.B.; Park, D.J.; Kim, M.O.; Koh, P.O. Chlorogenic acid alleviates neurobehavioral disorders and brain damage in focal ischemia animal models. Neurosci. Lett. 2021, 760, 136085.
  91. Fan, Y.; Li, Y.; Yang, Y.; Lin, K.; Lin, Q.; Luo, S.; Zhou, X.; Lin, Q.; Zhang, F. Chlorogenic acid promotes angiogenesis and attenuates apoptosis following cerebral ischaemia-reperfusion injury by regulating the PI3K-Akt signalling. Pharm. Biol. 2022, 60, 1646–1655.
  92. Shah, M.A.; Kang, J.B.; Kim, M.O.; Koh, P.O. Chlorogenic acid alleviates the reduction of Akt and Bad phosphorylation and of phospho-Bad and 14-3-3 binding in an animal model of stroke. J. Vet. Sci. 2022, 23, e84.
  93. Shah, M.A.; Kang, J.B.; Park, D.J.; Kim, M.O.; Koh, P.O. Chlorogenic acid alleviates cerebral ischemia-induced neuroinflammation via attenuating nuclear factor kappa B activation. Neurosci. Lett. 2022, 773, 136495.
  94. Lapchak, P.A. The phenylpropanoid micronutrient chlorogenic acid improves clinical rating scores in rabbits following multiple infarct ischemic strokes: Synergism with tissue plasminogen activator. Exp. Neurol. 2007, 205, 407–413.
  95. Liu, Q.S.; Deng, R.; Li, S.; Li, X.; Li, K.; Kebaituli, G.; Li, X.; Liu, R. Ellagic acid protects against neuron damage in ischemic stroke through regulating the ratio of Bcl-2/Bax expression. Appl. Physiol. Nutr. Metab. 2017, 42, 855–860.
  96. Sun, J.; Li, Y.Z.; Ding, Y.H.; Wang, J.; Geng, J.; Yang, H.; Ren, J.; Tang, J.Y.; Gao, J. Neuroprotective effects of gallic acid against hypoxia/reoxygenation-induced mitochondrial dysfunctions in vitro and cerebral ischemia/reperfusion injury in vivo. Brain Res. 2014, 1589, 126–139.
  97. Nabavi, S.F.; Habtemariam, S.; Di Lorenzo, A.; Sureda, A.; Khanjani, S.; Nabavi, S.M.; Daglia, M. Post-Stroke depression modulation and in vivo antioxidant activity of gallic acid and its synthetic derivatives in a murine model system. Nutrients 2016, 8, 248.
  98. Zhao, Y.; Li, D.; Zhu, Z.; Sun, Y. Improved Neuroprotective effects of gallic acid-loaded chitosan nanoparticles against ischemic stroke. Rejuvenation Res. 2019, 23, 284–292.
  99. Mirshekari Jahangiri, H.; Sarkaki, A.; Farbood, Y.; Dianat, M.; Goudarzi, G. Gallic acid affects blood-brain barrier permeability, behaviors, hippocampus local EEG, and brain oxidative stress in ischemic rats exposed to dusty particulate matter. Environ. Sci. Pollut. Res. 2020, 27, 5281–5292.
  100. Abdelsalam, S.A.; Renu, K.; Zahra, H.A.; Abdallah, B.M.; Ali, E.M.; Veeraraghavan, V.P.; Sivalingam, K.; Ronsard, L.; Ammar, R.B.; Vidya, D.S.; et al. Polyphenols Mediate Neuroprotection in Cerebral Ischemic Stroke—An Update. Nutrients 2023, 15, 1107.
  101. Shaikh, A.; Ahmad, F.; Teoh, S.L.; Kumar, J.; Yahaya, M.F. Honey and Alzheimer’s Disease-current understanding and future prospects. Antioxidants 2023, 12, 427.
  102. Rezai-Zadeh, K.; Shytle, R.D.; Bai, Y.; Tian, J.; Hou, H.; Mori, T.; Zeng, J.; Obregon, D.; Town, T.; Tan, J. Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer’s disease β-amyloid production. J. Cell. Mol. Med. 2009, 13, 574–588.
  103. Mori, T.; Koyama, N.; Guillot-Sestier, M.-V.; Tan, J.; Town, T. Ferulic Acid Is a Nutraceutical β-Secretase Modulator That Improves Behavioral Impairment and Alzheimer-like Pathology in Transgenic Mice. PLoS ONE 2013, 8, e55774.
  104. Ogunsuyi, O.B.; Oboh, G.; Oluokun, O.O.; Ademiluyi, A.O.; Ogunruku, O.O. Gallic Acid Protects against Neurochemical Alterations in Transgenic Drosophila Model of Alzheimer’s Disease. Adv. Tradit. Med. 2020, 20, 89–98.
  105. Zheng, Q.; Kebede, M.T.; Kemeh, M.M.; Islam, S.; Lee, B.; Bleck, S.D.; Wurfl, L.A.; Lazo, N.D. Inhibition of the Self-Assembly of Aβand of Tau by Polyphenols: Mechanistic Studies. Molecules 2019, 24, 2316.
  106. Harakeh, S.; Qari, M.H.; Ramadan, W.S.; Al Jaouni, S.K.; Almuhayawi, M.S.; Al Amri, T.; Ashraf, G.M.; Bharali, D.J.; Mousa, S.A. A Novel Nanoformulation of Ellagic Acid Is Promising in Restoring Oxidative Homeostasis in Rat Brains with Alzheimer’s Disease. Curr. Drug Metab. 2021, 22, 299–307.
  107. Rashno, M.; Gholipour, P.; Salehi, I.; Komaki, A.; Rashidi, K.; Khoshnam, S.E.; Ghaderi, S. P-Coumaric acid mitigates passive avoidance memory and hippocampal synaptic plasticity impairments in aluminum chloride-induced Alzheimer’s Disease rat model. J. Funct. Foods 2022, 94, 105117.
  108. Hussain, H.; Ahmad, S.; Shah, S.W.A.; Ghias, M.; Ullah, A.; Rahman, S.U.; Kamal, Z.; Khan, F.A.; Khan, N.M.; Muhammad, J.; et al. Neuroprotective Potential of Synthetic Mono-Carbonyl Curcumin Analogs Assessed by Molecular Docking Studies. Molecules 2021, 26, 7168.
  109. Hussain, H.; Ahmad, S.; Shah, S.W.A.; Ullah, A.; Ali, N.; Almehmadi, M.; Ahmad, M.; Khalil, A.A.K.; Jamal, S.B.; Ahmad, H.; et al. Attenuation of Scopolamine-Induced Amnesia via Cholinergic Modulation in Mice by Synthetic Curcumin Analogs. Molecules 2022, 27, 2468.
  110. Ochiai, R.; Saitou, K.; Suzukamo, C.; Osaki, N.; Asada, T. Effect of Chlorogenic Acids on Cognitive Function in Mild Cognitive Impairment: A Randomized Controlled Crossover Trial. J. Alzheimers Dis. 2019, 72, 1209–1216.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 432
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 04 Aug 2023
1000/1000
Video Production Service