Next Article in Journal
Sodium Thiosulfate Improves Hypertension in Rats with Adenine-Induced Chronic Kidney Disease
Next Article in Special Issue
Iron Dysregulation in Mitochondrial Dysfunction and Alzheimer’s Disease
Previous Article in Journal
Alcohol and Head and Neck Cancer: Updates on the Role of Oxidative Stress, Genetic, Epigenetics, Oral Microbiota, Antioxidants, and Alkylating Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy

by
Ryszard Pluta
1,*,
Jacek Kiś
2,
Sławomir Januszewski
1,
Mirosław Jabłoński
3 and
Stanisław J. Czuczwar
4
1
Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland
2
Department of Urology, 1st Military Clinical Hospital with the Outpatient Clinic, Al. Racławickie 23, 20-049 Lublin, Poland
3
Department of Rehabilitation and Orthopedics, Medical University of Lublin, Jaczewskiego 8 Str., 20-090 Lublin, Poland
4
Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b Str., 20-090 Lublin, Poland
*
Author to whom correspondence should be addressed.
Antioxidants 2022, 11(1), 146; https://doi.org/10.3390/antiox11010146
Submission received: 18 December 2021 / Revised: 5 January 2022 / Accepted: 9 January 2022 / Published: 11 January 2022
(This article belongs to the Special Issue Energy Disorders in Neurodegenerative Diseases)

Abstract

:
Recent years have seen remarkable progress in research into free radicals oxidative stress, particularly in the context of post-ischemic recirculation brain injury. Oxidative stress in post-ischemic tissues violates the integrity of the genome, causing DNA damage, death of neuronal, glial and vascular cells, and impaired neurological outcome after brain ischemia. Indeed, it is now known that DNA damage and repair play a key role in post-stroke white and gray matter remodeling, and restoring the integrity of the blood-brain barrier. This review will present one of the newly characterized mechanisms that emerged with genomic and proteomic development that led to brain ischemia to a new level of post-ischemic neuropathological mechanisms, such as the presence of amyloid plaques and the development of neurofibrillary tangles, which further exacerbate oxidative stress. Finally, we hypothesize that modified amyloid and the tau protein, along with the oxidative stress generated, are new key elements in the vicious circle important in the development of post-ischemic neurodegeneration in a type of Alzheimer’s disease proteinopathy.

1. Introduction

The human brain accounts for only 2% of the total body weight, uses about 20% of the oxygen supplied to the whole body [1,2] and produces more free radicals than other organs of the body [2,3]. The brain’s enormous energy consumption makes it more susceptible to oxidative stress than any other organ in the human body [1,3]. The main structural and functional part of the brain, namely neurons, are particularly susceptible to oxidative injury due to the high level of metabolism compared to other cells of the nervous system [4]. The human brain is the most complex organ that controls all of the body’s responses. It contains billions of neurons that continuously control the proper functioning of the body. Neurons oversee and transmit signals that enable us to speak, think, move and achieve everything we do [5]. The brain’s neurons are closely interconnected to form a neural network. Therefore, the slightest disconnection between neurons can lead to disruption of various activities controlled by the brain, causing serious damage to the brain [5]. Disrupting the transmission of information between neurons in the brain has devastating effects, affecting human brain functions such as memory, movement, intelligence, speech and many other activities [5,6].
Human ischemic stroke is an age-related neurodegenerative disorder that is the leading cause of long-term disability and mortality, placing a huge burden on patients and their carers, the community and the healthcare system [7]. Currently available therapies for ischemic stroke are intravenous injection of tissue plasminogen activator and thrombectomy. However, both treatments are limited by a narrow therapeutic window and should be performed within 4.5 and 12 h, respectively, of the onset of an ischemic stroke. These limitations mean that the vast majority of post-ischemic patients are inadequately treated and therefore develop severe neurological deficits, with frequent full-blown dementia [8,9,10]. So, restoring blood flow following cerebral ischemia is an important challenge for stroke researchers and clinicians. Therefore, the effective restoration of blood circulation to the area of the ischemia is the main goal after the onset of the acute phase of an ischemic stroke. It should be emphasized that there may also be complications during the restoration of blood flow, among which one of the most serious further and complicated risks is the brain reperfusion itself. Restoration of blood flow is an inevitable problem once circulation is restored by therapy at the site of a stroke and carries with it important additional neuropathological changes. Despite the successful restoration of blood flow, neurological progress is still not observed in a significant number of patients due to the presence of neuropathological phenomena that prevent the return to normal activity [11,12]. After reversible cerebral ischemia with recirculation changes, now referred to as proteomic, genomic and biochemical cascades, they damage the ischemic parenchyma of the brain, counteracting the beneficial effects of restoring cerebral circulation [13,14]. During post-ischemic reperfusion, neuropathological pathways include excitotoxicity, intracellular Ca2+ accumulation, neuroinflammation, free radical and amyloid generation, modification of tau protein, necrosis, apoptosis, lipolysis, and elastin degradation to polypeptides [15,16,17,18,19]. Recently, elastin-derived polypeptides have been reported to induce overproduction of β-amyloid peptides in a model of Alzheimer’s disease [20,21,22]. Among these various neuropathological phenomena, free radical changes in brain tissue play an important role in the process of ischemic brain damage during recovery of blood flow at the primary ischemic focus.
The oxidation of nucleic acids, proteins and lipids in neurons is crucial for the survival or death of neuronal cells during recirculation at the area of cerebral ischemia. Neurons contain large amounts of polyunsaturated fatty acids that interact with reactive oxygen species, triggering a self-propelling cascade and initiating, for example, increased lipid peroxidation [23]. It should be emphasized that neuronal cells are low in glutathione, an essential antioxidant for scavenging free radicals [24]. Consequently, neuronal cells are extremely susceptible to oxidative stress. Accordingly, post-ischemic stroke patients are more predisposed to cognitive impairment and Alzheimer’s disease type dementia, which significantly affects the patient’s quality of life. All of these symptoms are related to secondary brain damage after stroke [25].
Oxidative stress indicators are not limited to ischemia of the injured brain [26], but are also present in serum or cells of the immune system and this is a universal phenomenon [27]. Therefore, in systemic blood circulation, after ischemia, elevated components of oxidative stress and mediators of chronic inflammation were found in patients, and in addition to the impaired immune response, polynuclear and mononuclear leukocytes were activated [26,28]. Leukocytes release, in addition to the overproduction of oxidation products, including reactive oxygen species and pro-inflammatory cytokines, including interleukin-6 or interleukin-1β [28]. As a result, the overproduction of reactive oxygen species through increased activity of a number of oxidative enzymes (e.g., myeloperoxidase, xanthine oxidase, etc.) or through dysregulation of the mitochondrial respiratory chain along with the elimination of antioxidants (e.g., catalase, superoxide dismutase, glutathione, glutathione peroxidase, etc.) finally causes oxidation of molecules (e.g., DNA, lipids and proteins), which ultimately damage cells and tissues. Moreover, since oxidation and inflammation are linked phenomena [26], the accumulation of oxidative factors trigger the release of pro-inflammatory mediators, and then strengthen and continue the vicious cycle of oxidative damage, inducing progressive neurodegeneration of the brain tissue [28].
Recently, an intensively researched post-ischemic neuropathology has emerged in connection with genomic and proteomic development, which has transferred ischemia-reperfusion brain injury to a new level of neuropathological mechanisms, due to the presence of amyloid plaques and the development of neurofibrillary tangles, which further exacerbate oxidative stress in the brain in addition to the classic processes described so far after ischemia. The major amyloid plaque molecule are β-amyloid peptide 1-42 or 1-40, which is derived from proteolytic metabolism of the amyloid protein precursor. Additionally hyperphosphorylated tau protein is the major component of neurofibrillary tangles. Therefore, oxidative stress is a common and key factor in post-ischemic brain injury during recirculation. It consists of an increased generation of reactive oxygen species and reactive nitrogen species that damage various components of the cell such as DNA, lipids and proteins [29]. DNA oxidative injury is one of the deadliest end effects of increased oxidative stress post-ischemia [30,31]. Oxidative damage to DNA occurs within minutes in an ischemic brain [30,31], but oxidative breakdown can continue for up to six months after a stroke [27]. Problems with repairing damaged DNA result in the activation of various mechanisms that trigger the death of neuronal cells through apoptosis that threatens functional regeneration after stroke, with a high probability of developing full-blown dementia [31].
Finally, in this review we will discuss the hypothesis that modified amyloid with tau protein hyperphosphorylation and oxidative stress are new key elements in the vicious cycle important in post-ischemic neurodegeneration with Alzheimer’s disease-type proteinopathy. In addition, we will present a new look at the oxidative stress generated by amyloid and tau protein, and at the same time show the effect of oxidative stress on the behavior of amyloid and tau protein in the context of post-ischemic brain neurodegeneration with the development of Alzheimer’s disease-type proteinopathy. The data show a progressive large redox imbalance due to the overproduction and accumulation of oxidative factors in the cells and brain tissue, which exceeds the clearance capacity by the antioxidant system. We will also focus on the role of two main factors, such as amyloid and tau protein, which trigger oxidative stress in post-ischemic reperfusion injury in the vicious cycle.

2. Free Radicals in Recirculation

Free radicals fall into two main groups: reactive oxygen species and reactive nitrogen species [2]. Reactive oxygen species and reactive nitrogen species play a key role in many pathological processes, including ischemic-reperfusion injury. Therefore, the toxicity of free radicals in ischemic-reperfusion injury is currently being intensively studied. Based on the above data, the concept of “oxidative stress” was introduced by Helmut Sies [32] as an imbalance between the production of oxidants and antioxidants that can cause damage to various organs and organisms. Since then, the field of redox biology has developed the concept of the important role of oxidative stress in the development of various pathologies, including cerebral ischemia [33,34]. In the following sections, we will discuss some of the mechanisms involved in the toxicity of free radicals to the brain during ischemia-reperfusion injury.

2.1. Reactive Oxygen Species in Recirculation

Oxidative stress is triggered by the overproduction of reactive oxygen species. The main types of reactive oxygen species include superoxide anion, hydroxyl radicals and hydrogen peroxide. Under normal circumstances, superoxide dismutase, glutathione peroxidase, catalase, and other antioxidants protect the brain from oxidative stress. In the following, we will discuss some of the mechanisms involved in the toxicity of reactive oxygen species to brain tissue during post-ischemic reperfusion [2]. During post-ischemic reperfusion, the production of free radicals increases significantly, leading to the breakdown of the antioxidant processes [2]. According to previous research, overproduction of reactive oxygen species involves four pathways: NADPH oxidase, mitochondrial respiratory chain dysfunction, xanthine reaction mediated by xanthine oxidase and arachidonic acid catalyzed by cyclooxygenase 2 [2,35]. In the early stages of cerebral ischemia, reactive oxygen species are mainly produced by the mitochondria [2].
Following ischemia, the brain lacks oxygen and glucose, which negatively affects ATP production. In the case of ATP deficiency, calcium levels in neurons increase, causing massive production of reactive oxygen species through the depolarization of the mitochondria [2,36,37,38]. Along with the penetration of macrophages and other immune cells into the brain during the development of neuroinflammation, the activation of NADPH oxidase in these cells contributes to the production of reactive oxygen species [39]. With recirculation of the blood, an abundance of oxygen appears which causes oxidative damage. It is well known that oxidative stress can activate pro-apoptotic mechanisms such as the cytochrome c pathway, autophagy, inducing DNA damage, affecting protein structure and function, especially lipid peroxidation during reperfusion [40,41,42].

2.2. Brain Damage Due to Reactive Oxygen Species during Recirculation

Oxidative stress can induce cell death by modifying the structure and function of proteins, DNA damage and lipid peroxidation. DNA injury includes active and passive DNA damage. Oxidative stress causes particularly passive DNA injury. Active DNA injury (mediated by DNA endonucleases, which mainly contain caspase-activated deoxynuclease, endonuclease G, and an apoptosis-inducing factor) causes double-stranded DNA fragmentation (Figure 1). Passive DNA injury is caused by the direct reaction of DNA with reactive oxygen species or the indirect reaction of DNA with the products of the reaction of reactive oxygen species with proteins or lipids, leading to modification of nucleotide bases or the development of single or double strand breaks (Figure 1) [2,31].
The hydroxyl radical, a form of reactive oxygen species produced by the Fenton reaction, can cause lipid peroxidation. As a result of the reaction of hydroxyl radicals with unsaturated fatty acids, an alkyl radical is formed, which when reacted with molecular oxygen, can form a peroxyl radical. Then the peroxyl radical receives hydrogen from another fatty acid and produces lipid hydroperoxide and a second alkyl radical, which causes another lipid peroxidation [2]. Lipid peroxidation damages membranes, leading to organelle dysfunction and changes in ion transport [2,43]. Lipid peroxidation products play an important role in damage from oxidative stress. These products are 4-hydroxynonenal, malondialdehyde and acrolein [2]. They can cause protein dysfunction, leading to more dangerous damage from oxidative stress [2]. Protein 53 is a key molecule in the process of generating reactive oxygen species during the death of neurons in the brain following ischemia [2,44,45]. Reactive oxygen species can cause a protein 53 reaction with cyclophilin D, which opens the mitochondrial transition pores, causing mitochondrial swelling and cells necrosis (Figure 2) [2,46]. Reactive oxygen species increase the permeability of the mitochondrial membrane and affect the released cytochrome c, which forms an inhibitory complex with proteins of the Bcl-2 family, such as Bid and Bax, and protein 53. Cytochrome c can generate active caspases by forming a complex with procaspase-9, an apoptotic protease activating factor-1 and ATP, resulting in apoptosis of pyramidal neurons in the CA1 region of the hippocampus following reversible global cerebral ischemia in rats (Figure 2) [2,47]. Protein 53 apoptosis modulator is a key pro-apoptotic protein that belongs to the Bcl-2 protein family [2,48]. Some evidence suggests that inhibition of oxidative stress by abundant superoxide dismutase 1 may suppress upregulated apoptosis modulator by protein 53, indicating an association between protein 53 upregulated apoptosis modulator and oxidative stress in death of pyramidal neurons in the CA1 region of the hippocampus after reversible global ischemic brain injury [2,45]. Another most important cell death regulating mechanism is related to the mitogen-activated protein kinase, which is associated with the activity of reactive oxygen species. Mitogen-activated protein kinase has been shown to induce the death of neuronal cells in the cortex and hippocampus in transient cerebral ischemia in mice [2,49]. The mechanism of the mitogen-activated protein kinase consists of three main components: extracellular signal-regulated kinase 1/2, c-Jun NH2-terminal kinase, and protein 38 mitogen-activated protein kinase. C-Jun NH2-terminal kinase and protein 38 mitogen-activated protein kinase play an important role in apoptosis. The mechanisms of the c-Jun NH2 terminal kinase and the protein 38 mitogen-activated protein kinase can be activated by the apoptosis signal regulating kinase 1, which is activated by reactive oxygen species, inducing post-ischemic apoptosis [2]. Additionally, as post-ischemic neuroinflammation develops, immune cells containing NADPH oxidase produce significant amounts of reactive oxygen species, worsening damage from oxidative stress [2]. Also, reactive oxygen species can activate inflammatory cells. Reactive oxygen species activate neutrophils, microglia and macrophages by the nuclear factor kappa B pathway. Leukocytes possess myeloperoxidase that can trigger the production of hydrochloric acid, a strong oxidant that is essential for the survival and plasticity of neuronal cells post-ischemia [2,50].

2.3. Reactive Nitrogen Species in Recirculation

There are two specific types of reactive nitrogen species, oxidative peroxynitrite and NO, which dominate the progression of post-ischemic brain injury [2]. NO is produced by the reaction of oxygen with L-arginine, which is controlled by three types of nitric oxide synthase, including neuronal nitric oxide synthase, endothelial nitric oxide synthase and induced nitric oxide synthase. Neuronal nitric oxide synthase and endothelial nitric oxide synthase are calcium dependent, while induced nitric oxide synthase is calcium independent. Overall, the NO generated from induced nitric oxide synthase and neuronal nitric oxide synthase is harmful to cells. It was shown that mice lacking endothelial nitric oxide synthase had a greater infarct volume than control mice in post-ischemic brain injury [2,51]. Endothelial nitric oxide synthase produces huge amounts of NO at the onset of ischemia in endothelial cells, causing the blood vessels to widen [2]. In contrast, NO, produced mainly from activated induced nitric oxide synthase and neuronal nitric oxide synthase, is detrimental to focal ischemic brain injury [2]. In addition, it has been shown that overactive endothelial nitric oxide synthase may be harmful, and its limited inhibition may act preventively in post-ischemic conditions by inhibiting the development of peroxynitrite [2,52]. Additionally, NO reacts with O2− and generates a strong oxidant and peroxynitrite, which is associated with a stronger oxidation than both O2− and NO together [2,53].

2.4. Brain Damage Due to Reactive Nitrogen Species during Recirculation

Too much NO can lead to loss of control of the blood-brain barrier, neuroinflammation, and death of neurons [2]. Both matrix metalloproteinase and tight junction proteins are influenced by NO, which opens the blood-brain barrier after ischemia [54,55,56]. NO stimulates matrix metalloproteinase-2 in the first phase, and in the second phase, it activates matrix metalloproteinase-9, which affects the opening of the blood-brain barrier [2]. Activated matrix metalloproteinases disintegrate the extracellular matrix and tight junction proteins of the walls of blood vessels [57]. Excess NO causes neuronal death by protein modification, mitochondrial dysfunction and the formation of peroxynitrite [2]. During brain ischemia, NO strongly inhibits cytochrome c oxidase in the respiratory chain of the mitochondria [2]. In contrast, the reaction of NO with proteins leads to the nitrosylation of proteins or the formation of nitrosothiol [2,58]. NO also increases the activity of cyclooxygenase-2, which can mediate glutamate toxicity by generating additional oxygen reactive species and participate in the neuroinflammatory response by pro-inflammatory production of prostaglandin E2 [59]. Serious damage to the brain after ischemia by oxidative peroxynitrite is the death of neurons and changes in the permeability of the blood-brain barrier. The reaction of the oxidant peroxynitrite with tyrosine produces 3-nitrotyrosine, which causes damage to the proteins of the cytoskeleton and inhibition of enzymatic activity, resulting in the death of neurons [2,60]. In addition, the oxidant peroxynitrite reacts with DNA elements, such as the sugar phosphate backbone and guanine nucleotides, causing DNA damage and ultimately activating poly (adenosine diphosphate-ribose) polymerase (PARP) processes [2]. Increased PARP-1 activation depletes NAD+, leading to cell death, including neuronal cells [2,61]. In addition, oxidative peroxynitrite causes mitochondrial dysfunction by affecting the I-V complexes of the mitochondrial respiratory chain [2,62,63]. Peroxidation of membrane lipids by oxidative peroxynitrite also causes cells death [2,64]. There are also observations that oxidative peroxynitrite can activate matrix metalloproteinase-1, -2 and -9, leading to dysfunction of the tight junction proteins of blood vessels [65,66], finally triggering the permeability of the blood-brain barrier after ischemia-reperfusion injury to the brain [2].

3. Amyloid Accumulation in Post-Ischemic Brain

The key processes related to amyloid accumulation and its effects on ischemic brain injury are virtually unclear. The following is the amyloidogenic processing of the post-ischemic amyloid protein precursor that is associated with the production and deposition of amyloid in the brain following ischemia [67,68,69]. In addition, it has been shown that patients with a history of ischemic stroke have an increase in elastin-derived polypeptides in the cerebrospinal fluid that contribute to the accumulation of amyloid in the extracellular space [19,20,21,22]. The accumulation of amyloid in the brain after ischemia [67,70] and its presence in the blood [71,72] indicate an important role of amyloid in the progression of ischemic brain injury with Alzheimer’s disease proteinopathy.
The expression of the amyloid protein precursor (APP) gene in the CA1 area of the hippocampus decreased within 2 days, in contrast, it increased 7–30 days after ischemia (Table 1) [68]. Expression of the β-secretase gene (BACE1) increased during 2–7 days and decreased 30 days following ischemia (Table 1) [68]. Presenilin 1 and 2 (PSEN1 and 2) genes increased in 2–7 days and decreased in 30 days after ischemia (Table 1) [68].
In the CA3 region of the hippocampus, the expression of the APP gene fluctuated around the control values within 2 and 30 days after ischemia (Table 1) [69]. But 7 days post-ischemia, expression of the above gene increased [69]. The expression of the α-secretase (ADAM10) gene fell below control during 2, 7, 30 days post-ischemia (Table 1) [69]. Expression of the BACE1 gene in the above region decreased in 2–7 days and increased 30 days after ischemia (Table 1) [69]. The PSEN1 gene was elevated within 2–7 days and was fluctuating around control 30 days following ischemia (Table 1). In this area, post-ischemic PSEN2 gene expression fluctuated around control within 2 days, while it decreased on day 7, but increased above control on day 30 (Table 1) [69].
The expression of the APP gene in the temporal cortex decreased within 2 days of survival and increased above the control 7–30 days after ischemia (Table 2) [73]. The highest increase in BACE1 gene expression was recorded within 2 days, but 7–30 days after ischemia, it oscillated around the control (Table 2) [73]. PSEN1 gene expression fluctuated about control within 2, 7 and 30 days post-ischemia (Table 2) [74]. The highest increase in the expression of the PSEN2 gene was observed within 2 days, while in 7–30 days after ischemia, the expression of this gene oscillated around the control (Table 2) [74].
After transient experimental cerebral ischemia with a survival of up to 2 years, amyloid accumulation in the intra- and extracellular space was observed [67,75,76,77,78,79,80,81,82,83,84,85,86]. Accumulation of amyloid in the extracellular space has been observed as diffuse or senile amyloid plaques [67,76,82,85,87,88,89,90,91,92,93]. Amyloid plaques have been found in the hippocampus, cerebral cortex, thalamus, around the lateral ventricles, and in the corpus callosum. The accumulation of amyloid inside neuronal and neuroglial cells indicates the importance of amyloid in the progressive neurodegeneration of the brain after ischemia with the development of Alzheimer’s disease proteinopathy [67,80,87,88,94,95,96,97,98]. These data indicate that after experimental ischemia, amyloid production is responsible for additional neurodegenerative processes that may worsen post-ischemic neurological outcomes due to the constant wasting of neurons [7,76,82,85,86,99,100]. Amyloid is a toxic molecule that activates intracellular mechanisms in ischemic astrocytes, neurons and microglia, which further induce damage and loss of neuronal and glial cells following ischemia. Following ischemia in the human brain, both diffuse and senile amyloid plaques have been observed in the brain cortex and hippocampus [70,101,102,103]. In addition, clinical trials showed that blood amyloid was increased in patients with post-ischemic brain damage [71,72]. The rise in blood amyloid negatively correlated with the neurological outcome after cerebral ischemia [72].

4. Amyloid versus Oxidative Stress in Recirculation

Studies have shown that methionine 35 from β-amyloid peptide is important for oxidative stress and its toxicity induced by amyloid [104,105,106]. Methionine undergoes two-electron oxidation to form methionine sulfoxide [106,107]. Oxidation of methionine to sulfoxide plays a significant role in reducing the antioxidant defense of cells [108]. In addition, the presence of methionine sulfoxide reductase suggests that sulfoxide reductase may influence the antioxidant effect [106,109,110,111]. Additionally, methionine residing in side amyloid in the lipid bilayer can undergo single-electron oxidation to form a sulfuranyl free radical. Since amyloid is generated from an amyloid protein precursor, it is suggested that the amyloid once produced may insert as small oligomer into the lipid bilayer adopting the α-helical conformation [106]. This suggests that the secondary structure of amyloid is a peptide neurotoxicity factor. It has been suggested that molecular oxygen or Cu2+ may be the key to the oxidation of methionine to the sulfuranyl radical. In the absence of oxygen, amyloid cannot lead to the formation of free radicals [106]. In vitro studies have shown that β-amyloid peptide can promote the reduction of Cu2+ to Cu+ by binding to Cu2+ and forming hydrogen peroxide. Then, the Cu+ can react with hydrogen peroxide to form highly reactive hydroxyl free radicals [106,112]. The importance of copper in amyloid-induced toxicity is suggested by a study where methionine 35 was replaced with valine, which showed an increase in toxicity, suggesting that this substitution may change the amyloid conformation from α-helix to a mixture of α-helical and β-sheet conformations, thus increasing Cu2+ binding followed by associated toxicity [106]. In addition, the replacement of histidine 6, 13 and 14 in the β-amyloid peptide 1–42 by tyrosine, which binds to Cu2+ with a lower affinity than histidine, showed that this did not affect oxidative stress and neurotoxicity, further emphasizing the importance of methionine in inducing oxidative stress and its toxicity by amyloid [106].
After generating the sulfuranyl free radical, it can detach allylic H atoms from the acyl chains of unsaturated fatty acids in the lipid bilayer to initiate the lipid peroxidation process and, consequently, affect the lipid bilayer [106]. The oxidation products further diffuse across the membrane, affecting other cellular organelles, greatly enhancing the action of the primary amyloid free radical, leading to neuronal cells death and post-ischemic brain neurodegeneration.
Studies with transgenic Caenorhabditis elegans expressing human β-amyloid peptide 1-42 presented increased oxidation which correlated with worm paralysis [106,113]. However, some studies have found that the 33-35 area of the β-amyloid peptide 25-35 is critical to the aggregation and neurotoxic properties of the β-amyloid peptide [106]. Study in transgenic mice presented that methionine residing in the β-amyloid peptide affects neurotoxicity, oxidative stress, and the formation of amyloid plaques [114].

5. Tau Protein Modification in Post-Ischemic Recirculation

Recent studies have documented an association between the expression of the tau protein (MAPT) gene in the CA1 and CA3 regions of the hippocampus after ischemia with a 30-day survival (Table 1) [69]. In the CA1 area, the expression of the MAPT gene increased significantly during 2 days following the ischemic brain injury (Table 1). In contrast, expression of the above gene was reduced 7–30 days post-ischemia (Table 1). After ischemia in the CA3 area, an oscillation of the expression of the MAPT gene around the control was observed 2 days after ischemia (Table 1) [69]. In contrast, the expression of the above gene increased 7–30 days after ischemia (Table 1) [69].
Studies of ischemic brains in animals and humans have documented the accumulation of tau protein in neurons and glial cells in the hippocampus, cortex, and thalamus [83,115,116,117,118,119,120,121]. An increase in soluble tau protein in brain tissue using microdialysis in an ischemic brain is also shown [122]. In addition, studies have indicated that the modified tau protein inhibits the movement of neurofilaments, organelles, vesicles with an amyloid protein precursor, and increases oxidative stress in neurons [123].
An increase in blood tau protein level was observed after global cerebral ischemia in humans with a maximum increase on days 2 and 4 of survival [124]. This observation correlated with two types of neuronal death in the brain: necrosis and apoptosis, respectively [125]. The above information suggests that blood tau protein levels may be a prognostic factor for neurological outcome post-ischemia [124,125]. Increased levels of tau protein have also been found in the blood of patients after ischemic stroke, which is probably a factor in the progression of damage in neuronal cells [126]. In addition, an increase in tau protein has been observed in humans in the cerebrospinal fluid following an ischemic stroke [126]. Moreover, post-ischemia, blood-derived tau protein [126] crosses the ischemic blood-brain barrier in two directions and can enhance its own pathology in the brain [127]. Finally, ischemic blood-brain barrier insufficiency may exacerbate tau protein neuropathology in the brain and also suggests that post-ischemic brain injury may be part of the cause responsible for the increase in plasma tau protein concentration [126,127].
After transient local ischemic damage to the brain, the tau protein underwent excessive hyperphosphorylation and accumulated at the border of the ischemic focus [128]. Following forebrain ischemia in gerbils, tau protein phosphorylation has been shown to be controlled by mitogen-activated protein kinase, cyclin-dependent kinase 5, and glycogen synthase kinase 3 [129]. New data show that after ischemia, the tau protein undergoes hyperphosphorylation in neuronal cells and accompanies their apoptosis [120,121,128,130,131].
The information presented argues that the tau protein undergoes continuous hyperphosphorylation after a transient episode of cerebral ischemia. This triggers post-ischemia the generation of paired helical filaments [132], neurofibrillary tangle-like [128,130,131] and neurofibrillary tangles [133,134]. This points to the involvement of various modifications of the tau protein in the death of neurons in the brain following ischemia. The above evidence also explains a different principle regarding the time of death of neuronal pyramidal cells in the CA1 and CA3 areas of the hippocampus after ischemia in a manner dependent on tau protein hyperphosphorylation and modification of its structure.

6. Tau Protein versus Oxidative Stress in Recirculation

Several studies have documented that oxidative stress triggers tau protein phosphorylation in neuronal cells in vitro [135,136,137]. Additionally, carbonyl-4HNE facilitates hyperphosphorylation of the tau protein and aggregation in cultures [137,138,139,140]. In addition, oxidative stress also generates the oxidation of fatty acids which have been found to promote tau protein polymerization in cultures [137,141]. In mice deficient in mitochondrial superoxide dismutase 2, tau protein hyperphosphorylation, oxidative stress, and mitochondrial dysfunction were observed in parallel [137,142]. In addition, mitochondrial oxidative stress has been shown to induce hyperphosphorylation of the tau protein [137,142]. It was shown that zebrafish deficient in folic acid induced oxidative stress related to hyperphosphorylation of tau protein and its aggregation and accumulation of amyloid plaques [137,143,144]. Damage to mitochondria, development of oxidative stress, increased tau protein phosphorylation and nuclear translocation of glycogen synthase kinase 3 beta were observed in rabbits after intracisternal injection of β-amyloid peptide 1-42. It was shown that in vitro treatment of rat cortical neurons with cuprizone, a copper chelator, with Fe2+ and H2O2 oxidizing molecules, significantly increased the activity of glycogen synthase kinase 3 beta and tau protein hyperphosphorylation [137,145]. However, simultaneous addition of lithium chloride to these cultures lowers the activity of glycogen synthase kinase 3 beta and tau protein phosphorylation, recognizing glycogen synthase kinase 3 beta as an enzyme involved in tau protein phosphorylation after oxidative stress in this model [137,145]. This was supported by the promotion of the hyperphosphorylation of the tau protein observed in rat cortical neurons in vitro by glycogen synthase kinase 3 beta and oxidative stress [137,145]. Taken together, this evidence suggests that glycogen synthase kinase 3 beta plays an important role in the pathology of tau protein and that effective modulation of its action may prevent apoptosis and tau protein phosphorylation induced by oxidative stress [137,146]. However, in addition to the glycogen synthase kinase 3 beta, oxidative stress affects other signaling pathways and kinases that mediate tau protein hyperphosphorylation. In fact, 4-hydroxynonenal has also been found to directly activate stress-activated kinases, such as c-Jun NH2-terminal kinase and protein 38, in neurons in vitro [147]. Another possible element in the interaction between oxidative stress and tau protein phosphorylation is peptidyl prolyl cis-trans isomerase 1. Therefore, it has been found that the accumulation of hyperphosphorylated tau protein promotes the production of reactive oxygen species and oxidative stress (Figure 1 and Figure 2) [137]. Additionally, this evidence strongly suggests that oxidative stress, in turn, directly induces hyperphosphorylation of the tau protein [137]. In this context, tau protein hyperphosphorylation and oxidative stress emerge as two components of a vital “vicious circle” leading to a progressive increase in radical oxygen species and modified tau protein, and ultimately to neuronal death [137]. The data presented above clearly prove that the tau protein in patients and animals after cerebral ischemia causes oxidative stress in neuronal cells [137]. On the other hand, the line of evidence also suggests that the generation of reactive oxygen species may directly induce hyperphosphorylation and aggregation of the tau protein.

7. DNA Damage in Brain Cells during Recirculation

Ischemic brain damage leads to an increase in free radicals oxidative stress, which causes damage to DNA, gray and white matter and the blood-brain barrier. Oxidative damage to DNA occurs immediately after cerebral ischemia [30]. Oxidative damage to DNA in the brain following ischemia has been studied frequently in neurons. Unlike neurons, microglia, astrocyte, oligodendrocyte, and endothelial cells, which are also prone to ischemic damage, glial DNA damage is less studied than neuronal DNA damage.

7.1. Neurons

Research suggests that post-ischemic brain injury produces base modifications, apurinic/apyrimidinic sites, double- and single-strand breaks of DNA in neuronal cells, all of which activate pro-death signaling pathways and accelerate neuronal death (Figure 1) [148]. Study found that the amount of urine base modification was negatively correlated with neurological outcome after brain ischemia and significantly decreased after rehabilitation [148]. This information suggests that an increased amount of base modification in urine could be used as a biomarker for neurological outcome post-ischemia, while a decreased amount of base modification after rehabilitation is associated with a better prognosis. This evidence supports the idea that damage to DNA structure, for example the development of base modification, may serve as a prognostic marker after ischemia and that DNA repair may be a potential therapeutic target post-ischemia.

7.2. Microglia

Microglia cells are responding rapidly to cerebral ischemia, including production reactive oxygen species [2,148]. After ischemic damage, microglia shows oxidative DNA damage, including the generation of base modifications [31]. Microglia cells in mice can increase oxidative stress by the voltage-gated proton channel, thus contributing to DNA damage [148]. It should be noted that microglia plays a dual role in relation to cerebral ischemia due to different phenotypes, namely with a pro-inflammatory phenotype it threatens the life of neurons and with a repair phenotype it supports the survival of neurons [29,31,148]. Thus, modulation of the microglia phenotype may have a favorable effect on post-ischemia recovery.

7.3. Astrocytes

Astrocytes are also sensitive to DNA oxidative damage. Cerebral ischemia is known to induce astrocyte activity, and this phenomenon can cause DNA damage in astrocytes [148]. Activated astrocytes in the ischemic focus show oxidative damage to DNA, such as single- and double-strand breaks and base modifications (Figure 1) [148]. The activity of astrocytes influences the formation and transformation of the glial scar, the structure and function of neuronal cells and the blood-brain barrier after ischemia. Given the pivotal role of astrocytes in repairing blood-brain barrier, they are expected to facilitate the rebuilding of the blood-brain barrier and improve neurological outcomes following cerebral ischemia.

7.4. Oligodendrocytes

Since oligodendrocytes determine myelination, damage to the DNA in these cells can cause axonal demyelination following ischemia [148,149]. DNA damage and repair in oligodendrocyte cells has been demonstrated in studies after ischemic brain injury [149,150].

7.5. Endothelial Cells

DNA oxidative damage is also documented in the vascular endothelium. Experimental studies prove that endothelial cells in small vessels of the brain show signs of DNA single-strand breaks within 3 days after focal transient cerebral ischemia [148]. DNA damage in the vascular endothelium causes cognitive impairment, the development of cerebral edema and infiltration of inflammatory cells, which may directly affect the neurological outcome after cerebral ischemia [148,151].

8. Side Effects of Post-Ischemic Recirculation

Recirculation triggers serious complications, such as failure of the blood-brain barrier, development of cerebral edema and hemorrhagic transformation of the ischemic focus and the generation of free radicals. Patients who develop life-threatening complications have poorer neurological outcomes that can often be fatal.

8.1. Blood-Brain Barrier Insufficiency

Matrix metalloproteinases, a key factor in damage to the blood-brain barrier (Figure 3), can cause oxidative damage to DNA, resulting in neuronal death [148]. Brain ischemia may induce abnormal behavior of gelatinase A and B, which results in increased permeability of the blood-brain barrier (Figure 3) and enlargement of the ischemic focus [54,148]. Gelatinases, which cause damage to the blood-brain barrier, also act as nucleases that enter the nucleus of the neuron and break down nuclear DNA repair proteins in post-ischemic neuronal cells [148]. This evidence supports the existence of internal DNA damage that induces the death of neuronal cells following ischemia [148].

8.2. Brain Edema Development

Edema development induced by free radical oxygen stress includes cytotoxic and vasogenic edema (Figure 3). Cytotoxic edema is associated with abnormal ion transport across membranes. Ion transport proteins, which are oxidized by reactive oxygen species, are involved in ion pumps, ion channels, ion exchangers and ion cotransporters. Reactive oxygen species oxidize sulfhydryl groups located on ion transport proteins, peroxidate membrane phospholipids, reduce the amount of ATP and inhibit oxidative phosphorylation, which causes abnormal ion transport, causing cytotoxic edema (Figure 3) [152,153]. In addition, reactive oxygen species are involved in inhibiting glutamate uptake by Na+/glutamate transport [2]. The enormous release of glutamate during reperfusion injury [15] destroys K+, Na+ and Ca2+ homeostasis, causing membrane dysfunction and development of cytotoxic edema (Figure 3) [154]. Vasogenic edema is caused by a failure of the blood-brain barrier, and the likely mechanisms leading to damage to the blood-brain barrier are described above. (Figure 3).

8.3. Hemorrhagic Transformation of the Ischemic Focus

Hemorrhagic transformation is directly related to uncontrolled permeability of the blood-brain barrier for plasma and blood cells to the brain (Figure 3). The blood-brain barrier consists of endothelial cells, basement membrane, tight junctions, astrocytes and pericytes [2,155]. These cells have enzymes such as nitric oxide synthase and NADPH oxidase [2,52,156,157,158]. Following ischemia-reperfusion injury and treatment with thrombectomy or thrombolysis, H2O2 generated by NADPH oxidase induces changes in the blood-brain barrier by altering tight junction proteins and thereby increasing monocyte migration [2,159,160,161]. Free radicals, especially NO and peroxynitrite, activate the mechanisms of matrix metalloproteinase’s, which lead to the degradation of laminin and collagen in the basement membrane, which results in uncontrolled permeability of the blood-brain barrier after cerebral ischemia [157,161,162,163,164,165,166]. Matrix metalloproteinases have been shown to be involved in the development of hemorrhage associated with plasminogen activator [2,167]. Thus, it is possible that NO is involved in the hemorrhagic conversion of the ischemic brain following plasminogen activator therapy mediated by matrix metalloproteinases. In addition, plasminogen activator may increase matrix metalloproteinases, especially matrix metalloproteinase-9, via the lipoprotein receptor mechanism, but the ultimate effect of NO and plasminogen activator on hemorrhagic conversion following embolic ischemia of brain is still unknown [54,167,168,169,170]. It has been shown that peroxynitrite can inactivate plasminogen activator, influencing the course of action of thrombolysis [2,171,172,173].

9. Conclusions

Free radicals are physiological byproducts of cellular metabolism and are therefore inevitable. On the other hand, the imbalance between pro-oxidative and antioxidant cellular molecules leads to a vicious cycle where free radical oxidative stress causes oxidation of DNA, lipids and proteins in neurons and ultimately their death after ischemia. Oxidation can be considered a neuropathological marker in cerebral ischemia. It contributes to disease progression by increasing amyloid accumulation, tau protein hyperphosphorylation, and loss of neurons and synapses, leading to post-ischemic brain neurodegeneration in the form of Alzheimer’s disease proteinopathy (Figure 4). Additionally, several lines of evidence indicate that amyloid itself causes oxidative stress (Figure 4). Amyloid-triggered oxidative stress occurs in a bilayer in which the β-amyloid peptide is inserted as oligomer and serves as a source of reactive oxygen species [106].
It is worth noting that oxidative stress can modify the tau protein by phosphorylation which mediates the neurodegeneration e.g., in Drosophila melanogaster [174]. Phosphorylation of the tau protein blocks the movement of organelles, neurofilaments and vesicles of the amyloid protein precursor in neurons and increases oxidative stress post-ischemia (Figure 4) [123]. Oxidative stress is an important factor contributing to the accumulation and hyperphosphorylation of tau protein with the development of neurofibrillary tangles, indicating that it plays a key role in the pathogenesis of post-ischemic brain neurodegeneration in the form of Alzheimer’s disease proteinopathy (Figure 4). Oxidative stress has been shown to stimulate tau protein hyperphosphorylation and amyloid production, but the accumulation of amyloid and hyperphosphorylated tau protein also induces oxidative stress. Moreover, the close relationship between amyloid, hyperphosphorylated tau protein and oxidative stress suggests that this phenomenon is a key element in the vicious cycle that plays a significant role in the pathological process of modifying tau protein and amyloid, resulting in post-ischemic neurodegeneration of the brain in the form of Alzheimer’s disease proteinopathy (Figure 4).
In addition to the accumulation of free radicals, deficiency of superoxide dismutase, glutathione peroxidase and catalase has also been found after cerebral ischemia. Free radicals, especially reactive oxygen species and reactive nitrogen species, have intense oxidation or nitrification abilities in the brain. During post-ischemic brain recirculation with re-flow of blood, the massive production of reactive oxygen species and reactive nitrogen species leads to the death of neurons through DNA damage, protein dysfunction and lipid peroxidation. Oxidative/nitrosative stress in brain ischemia-reperfusion injury also plays a key role in inducing hemorrhagic transformation of the ischemic focus in conjunction with changes in the blood-brain barrier and development of cerebral edema (Figure 3). Research indicates that mitochondrial dysfunction [25,175], metal dyshomeostasis [41,176,177,178], tau protein hyperphosphorylation [123,174], neuroinflammation and amyloid accumulation [106] are the primary processes that induce oxidative stress. Inactivation and deficiency of antioxidant enzymes reduces the removal of free radicals from the brain during recirculation. This indicates that oxidative stress is an important neuropathological hallmark of the post-ischemic brain [175,179].
Broadly speaking, in order to survive cerebral ischemia, neuronal and glial cells developed integrated responses, so-called longevity safety processes, consisting of several genes called vitagenes, including but not limited to members of the heat shock protein system to detect and control various forms of pathological processes [180]. All of these considerations strongly support the idea that the smooth functioning of the maintenance and repair processes is crucial for the survival of brain cells under conditions of oxidative and nitrosative injury, and that tolerance to brain stress can be achieved through modulation of a multigene system such as the hormesis-vitagene network [180]. The efficient functioning of repair and maintenance processes is crucial for both survival and quality of life after ischemic brain injury. This is accomplished by a complex network of so-called longevity processes, which are made up of several genes of vitagene network, including the heat shock system, a highly conserved mechanism responsible for the repair and preservation of cellular macromolecules such as proteins, RNAs and DNA. During inflammation after cerebral ischemia, there is a gradual decline in the strength of the heat shock response, which may prevent protein damage from repairing, leading to the death of neuronal cells. In this situation, the concept of dose-response hormetic neuroprotection should be applied with the key role of heat shock proteins as elements of the vitagene network in neuroprotection and redox proteomics as redox modulators of stress-responsive vitagenes [181,182]. The use of hormetic machinery makes it possible to apply specific cell, tissue and/or pathological pathways therapies in a timely manner in post-ischemic brain. Natural antioxidants [3] are able to activate vitagenes such as heat shock protein 70, heme oxygenase, thioredoxin reductase and sirtuins, which are part of the integrated cell stress tolerance system [181]. Thus, activation of the vitagene system, with elevation of thioredoxin reductase, heme oxygenase 1, glutathione and sirtuins, results in a reduction of pro-oxidative conditions [181]. Dietary antioxidants have been shown to have neuroprotective effects by activating hormetic pathways, including vitagene. Therefore, regulation of endogenous cellular defense processes through the heat shock system, natural and pharmacological antioxidant substances may be an interesting therapeutic approach in ischemic neurodegeneration of the brain. Thus, by sustaining or restoring vitagenes activity, it is possible to delay the development of post-ischemic injury and thereby increase life expectancy [180]. Therefore, preventing oxidative damage to DNA in brain cells and restoring gray and white matter and normal blood-brain barrier activity is critical to the successful treatment of post-ischemic brain.

Author Contributions

Conceptualization, R.P., J.K., M.J. and S.J.C.; Methodology, R.P.; Software, S.J.; Validation, R.P., J.K., M.J. and S.J.C.; Formal Analysis, R.P., J.K., M.J. and S.J.C.; Investigation, S.J.; Resources, R.P., J.K., M.J. and S.J.C.; Data Curation, R.P.; Writing—Original Draft Preparation, R.P., J.K., M.J. and S.J.C.; Writing—Review & Editing, R.P., J.K., M.J. and S.J.C.; Visualization, S.J.; Supervision, R.P.; Project Administration, R.P.; Funding Acquisition, R.P., M.J. and S.J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the financial support from the Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland (T3-RP) and the Medical University of Lublin, Lublin, Poland (DS 475/21-SJC).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chen, Z.; Zhong, C. Oxidative stress in Alzheimer’s disease. Neurosci. Bull. 2014, 30, 271–281. [Google Scholar] [CrossRef]
  2. Sun, M.S.; Jin, H.; Sun, X.; Huang, S.; Zhang, F.-L.; Guo, Z.-N.; Yang, Y. Free radical damage in ischemia-reperfusion injury: An obstacle in acute ischemic stroke after revascularization therapy. Oxid. Med. Cell. Longev. 2018, 2018, 3804979. [Google Scholar] [CrossRef]
  3. Rahman, M.H.; Akter, R.; Bhattacharya, T.; Abdel-Daim, M.M.; Alkahtani, S.; Arafah, M.W.; Al-Johani, N.S.; Alhoshani, N.M.; Alkeraishan, N.; Alhenaky, A.; et al. Resveratrol and neuroprotection: Impact and its therapeutic potential in Alzheimer’s disease. Front. Pharmacol. 2020, 11, 619024. [Google Scholar] [CrossRef]
  4. Tholey, G.; Ledig, M. Neuronal and astrocytic plasticity: Metabolic aspects. Ann. Med. Interne 1990, 141 (Suppl. S1), 13–18. [Google Scholar]
  5. Behl, T.; Makkar, R.; Sehgal, A.; Singh, S.; Sharma, N.; Zengin, G.; Bungau, S.; Andronie-Cioara, F.L.; Munteanu, M.A.; Brisc, M.C.; et al. Current trends in neurodegeneration: Cross talks between oxidative stress, cell death, and inflammation. Int. J. Mol. Sci. 2021, 22, 7432. [Google Scholar] [CrossRef] [PubMed]
  6. Gitler, A.D.; Dhillon, P.; Shorter, J. Neurodegenerative disease: Models, mechanisms, and a new hope. Dis. Model. Mech. 2017, 10, 499–502. [Google Scholar] [CrossRef] [Green Version]
  7. Pluta, R.; Jolkkonen, J.; Cuzzocrea, S.; Pedata, F.; Cechetto, D.; PopaWagner, A. Cognitive impairment with vascular impairment and degeneration. Curr. Neurovasc. Res. 2011, 8, 342–350. [Google Scholar] [CrossRef] [PubMed]
  8. Kiryk, A.; Pluta, R.; Figiel, I.; Mikosz, M.; Ułamek, M.; Niewiadomska, G.; Jabłoński, M.; Kaczmarek, L. Transient brain ischemia due to cardiac arrest causes irreversible long-lasting cognitive injury. Behav. Brain Res. 2011, 219, 1–7. [Google Scholar] [CrossRef]
  9. 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. Profile of and risk factors for post-stroke cognitive impairment in diverse ethno-regional groups. Neurology 2019, 93, e2257–e2271. [Google Scholar] [CrossRef] [Green Version]
  10. Yuan, Y.; Shan, X.; Men, W.; Zhai, H.; Qiao, X.; Geng, L.; Li, C. The effect of crocin on memory, hippocampal acetylcholine level, and apoptosis in a rat model of cerebral ischemia. Biomed. Pharmacother. 2020, 130, 110543. [Google Scholar] [CrossRef] [PubMed]
  11. Molina, C.A.; Saver, J.L. Extending reperfusion therapy for acute ischemic stroke: Emerging pharmacological, mechanical, and imaging strategies. Stroke 2005, 36, 2311–2320. [Google Scholar] [CrossRef] [PubMed]
  12. Dalkara, T.; Arsava, E.M. Can restoring incomplete microcirculatory reperfusion improve stroke outcome after thrombolysis? JCBFM 2012, 32, 2091–2099. [Google Scholar] [CrossRef] [PubMed]
  13. Aronowski, J.; Strong, R.; Grotta, J.C. Reperfusion injury: Demonstration of brain damage produced by reperfusion after transient focal ischemia in rats. JCBFM 1997, 17, 1048–1056. [Google Scholar] [CrossRef]
  14. Pan, J.; Konstas, A.A.; Bateman, B.; Ortolano, G.A.; Pile-Spellman, J. Reperfusion injury following cerebral ischemia: Pathophysiology, MR imaging, and potential therapies. Neuroradiology 2007, 49, 93–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Pluta, R.; Salinska, E.; Puka, M.; Stafiej, A.; Łazarewicz, J.W. Early changes in extracellular amino acids and calcium concentrations in rabbit hippocampus following complete 15-min cerebral ischemia. Resuscitation 1988, 16, 193–210. [Google Scholar] [CrossRef]
  16. Wang, X.; Lo, E.H. Triggers and mediators of hemorrhagic transformation in cerebral ischemia. Mol. Neurobiol. 2003, 28, 229–244. [Google Scholar] [CrossRef]
  17. Sekeljic, V.; Bataveljic, D.; Stamenkovic, S.; Ułamek, M.; Jabłonski, 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. [Google Scholar] [CrossRef]
  18. 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. [Google Scholar] [CrossRef]
  19. Nicoloff, G.; Tzvetanov, P.; Christova, P.; Baydanoff, S. Detection of elastin derived peptides in cerebrospinal fluid of patients with first ever ischaemic stroke. Neuropeptides 2008, 42, 277–282. [Google Scholar] [CrossRef]
  20. Ma, C.; Su, J.; Sun, Y.; Feng, Y.; Shen, N.; Li, B.; Liang, Y.; Yang, X.; Wu, H.; Zhang, H.; et al. Significant upregulation of Alzheimer’s β-amyloid levels in a living system induced by extracellular elastin polypeptides. Angew. Chem. Int. Ed. Engl. 2019, 58, 18703–18709. [Google Scholar] [CrossRef] [Green Version]
  21. Ma, J.; Ma, C.; Li, J.; Sun, Y.; Ye, F.; Liu, K.; Zhang, H. Extracellular matrix proteins involved in Alzheimer’s disease. Chemistry 2020, 26, 12101–12110. [Google Scholar] [CrossRef]
  22. Szychowski, K.A.; Skóra, B.; Wójtowicz, A.K. Elastin-derived peptides in the central nervous system: Friend or foe. Cell Mol. Neurobiol. 2021; online ahead of print. [Google Scholar] [CrossRef]
  23. Nunomura, A.; Castellani, R.J.; Zhu, X.; Moreira, P.I.; Perry, G.; Smith, M.A. Involvement of oxidative stress in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2006, 65, 631–641. [Google Scholar] [CrossRef] [Green Version]
  24. Pocernich, C.; Butterfield, D. Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim. Biophys. Acta 2012, 1822, 625–630. [Google Scholar] [CrossRef] [Green Version]
  25. Federico, A.; Cardaioli, E.; Da Pozzo, P.; Formichi, P.; Gallus, G.N.; Radi, E. Mitochondria, oxidative stress and neurodegeneration. J. Neurol. Sci. 2012, 322, 254–262. [Google Scholar] [CrossRef]
  26. De la Fuente, M.; Miquel, J. An update of the Oxidation-Inflammation theory of aging: The involvement of the immune system in oxi-inflamm-aging. Curr. Pharm. Des. 2009, 15, 3003–3026. [Google Scholar] [CrossRef] [PubMed]
  27. Pascotini, E.T.; Flores, A.E.; Kegler, A.; Gabbi, P.; Bochi, G.V.; Algarve, T.D.; Prado, A.L.; Duarte, M.M.; da Cruz, I.B.; Moresco, R.N.; et al. Apoptotic markers and DNA damage are related to late phase of stroke: Involvement of dyslipidemia and inflammation. Physiol. Behav. 2015, 151, 369–378. [Google Scholar] [CrossRef] [PubMed]
  28. Mihai, S.; Codrici, E.; Popescu, I.D.; Enciu, A.M.; Albulescu, L.; Necula, L.G.; Mambet, C.; Anton, G.; Tanase, C. Inflammation related mechanisms in chronic kidney disease prediction, progression, and outcome. J. Immunol. Res. 2018, 2018, 2180373. [Google Scholar] [CrossRef] [PubMed]
  29. Zhao, H.; Han, Z.; Ji, X.; Luo, Y. Epigenetic regulation of oxidative stress in ischemic stroke. Aging Dis. 2016, 7, 295–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Cui, J.; Holmes, E.H.; Greene, T.G.; Liu, P.K. Oxidative DNA damage precedes DNA fragmentation after experimental stroke in rat brain. FASEB J. 2000, 14, 955–967. [Google Scholar] [CrossRef] [PubMed]
  31. Li, P.; Hu, X.; Gan, Y.; Gao, Y.; Liang, W.; Chen, J. Mechanistic insight into DNA damage and repair in ischemic stroke: Exploiting the base excision repair pathway as a model of neuroprotection. Antioxid. Redox Signal. 2011, 14, 1905–1918. [Google Scholar] [CrossRef]
  32. Sies, H. Oxidative stress: Introductory remarks. In Oxidative Stress; Sies, H., Ed.; Academic Press: London, UK, 1985; pp. 1–8. [Google Scholar]
  33. Flohé, L. Looking back at the early stages of redox biology. Antioxidants 2020, 9, 1254. [Google Scholar] [CrossRef] [PubMed]
  34. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 30, 1–21. [Google Scholar] [CrossRef] [PubMed]
  35. Margaill, I.; MPlotkine, M.; Lerouet, D. Antioxidant strategies in the treatment of stroke. Free Radic. Biol. Med. 2005, 39, 429–443. [Google Scholar] [CrossRef]
  36. Drose, S.; Brandt, U. Molecular mechanisms of superoxide production by the mitochondrial respiratory chain. Adv. Exp. Med. Biol. 2012, 748, 145–169. [Google Scholar]
  37. Kalogeris, T.; Bao, Y.; Korthuis, R.J. Mitochondrial reactive oxygen species: A double edged sword in ischemia/reperfusion vs preconditioning. Redox Biol. 2014, 2, 702–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Shenoda, B. The role of Na+/Ca2+ exchanger subtypes in neuronal ischemic injury. Transl. Stroke Res. 2015, 6, 181–190. [Google Scholar] [CrossRef] [PubMed]
  39. Walder, C.E.; Green, S.P.; Darbonne, W.C.; Mathias, J.; Rae, J.; Dinauer, M.C.; Curnutte, J.T.; Thomas, G.R. Ischemic stroke injury is reduced in mice lacking a functional NADPH oxidase. Stroke 1997, 28, 2252–2258. [Google Scholar] [CrossRef] [PubMed]
  40. Lee, S.J.; Cho, K.S.; Koh, J.Y. Oxidative injury triggers autophagy in astrocytes: The role of endogenous zinc. Glia 2009, 57, 1351–1361. [Google Scholar] [CrossRef] [PubMed]
  41. Lee, S.J.; Koh, J.Y. Roles of zinc and metallothionein-3 in oxidative stress-induced lysosomal dysfunction, cell death, and autophagy in neurons and astrocytes. Mol. Brain 2010, 3, 30. [Google Scholar] [CrossRef] [Green Version]
  42. Granzotto, A.; Sensi, S.L. Intracellular zinc is a critical intermediate in the excitotoxic cascade. Neurobiol. Dis. 2015, 81, 25–37. [Google Scholar] [CrossRef]
  43. Nigam, S.; Schewe, T. Phospholipase A(2)s and lipid peroxidation. Biochim. Biophys. Acta 2000, 1488, 167–181. [Google Scholar] [CrossRef]
  44. Saito, A.; Hayashi, T.; Okuno, S.; Nishi, T.; Chan, P.H. Modulation of p53 degradation via MDM2-mediated ubiquitylation and the ubiquitin–proteasome system during reperfusion after stroke: Role of oxidative stress. JCBFM 2005, 25, 267–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Niizuma, K.; Endo, H.; Nito, C.; Myer, D.J.; Chan, P.H. Potential role of PUMA in delayed death of hippocampal CA1 neurons after transient global cerebral ischemia. Stroke 2009, 40, 618–625. [Google Scholar] [CrossRef] [Green Version]
  46. Vaseva, A.V.; Marchenko, N.D.; Ji, K.; Tsirka, S.E.; Holzmann, S.; Moll, U.M. p53 opens the mitochondrial permeability transition pore to trigger necrosis. Cell 2012, 149, 1536–1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Endo, H.; Kamada, H.; Nito, C.; Nishi, T.; Chan, P.H. Mitochondrial translocation of p53 mediates release of cytochrome c and hippocampal CA1 neuronal death after transient global cerebral ischemia in rats. J. Neurosci. 2006, 26, 7974–7983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Shan, Z.; Liu, Q.; Li, Y.; Wu, J.; Sun, D.; Gao, Z. PUMA decreases the growth of prostate cancer PC-3 cells independent of p53. Oncol. Lett. 2017, 13, 1885–1890. [Google Scholar] [CrossRef] [Green Version]
  49. Takagi, Y.; Nozaki, K.; Sugino, T.; Hattori, I.; Hashimoto, N. Phosphorylation of c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase after transient forebrain ischemia in mice. Neurosc. Lett. 2000, 294, 117–120. [Google Scholar] [CrossRef]
  50. Mattson, M.P. NF-κB in the survival and plasticity of neurons. Neurochem. Res. 2005, 30, 883–893. [Google Scholar] [CrossRef]
  51. Huang, Z.; Huang, P.L.; Ma, J.; Meng, W.; Ayata, C.; Fishman, M.C.; Moskowitz, M.A. Enlarged infarcts in endothelial nitric oxide synthase knockout mice are attenuated by nitro-L-arginine. JCBFM 1996, 16, 981–987. [Google Scholar] [CrossRef] [PubMed]
  52. Gursoy-Ozdemir, Y.; Can, A.; Dalkara, T. Reperfusion induced oxidative/nitrative injury to neurovascular unit after focal cerebral ischemia. Stroke 2004, 35, 1449–1453. [Google Scholar] [CrossRef] [Green Version]
  53. Pacher, P.; Beckman, J.S.; Liaud, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Aoki, T.; Sumii, T.; Mori, T.; Wang, X.; Lo, E.H. Blood-brain barrier disruption and matrix metalloproteinase-9 expression during reperfusion injury: Mechanical versus embolic focal ischemia in spontaneously hypertensive rats. Stroke 2002, 33, 2711–2717. [Google Scholar] [CrossRef] [Green Version]
  55. Pfefferkorn, T.; Rosenberg, G.A. Closure of the blood-brain barrier by matrix metalloproteinase inhibition reduces rtPA-mediated mortality in cerebral ischemia with delayed reperfusion. Stroke 2003, 34, 2025–2030. [Google Scholar] [CrossRef] [Green Version]
  56. Yang, Y.; Estrada, E.Y.; Thompson, J.F.; Liu, W.; Rosenberg, G.A. Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. JCBFM 2007, 27, 697–709. [Google Scholar] [CrossRef]
  57. Rosenberg, G.A.; Yang, Y. Vasogenic edema due to tight junction disruption by matrix metalloproteinases in cerebral ischemia. Neurosurg. Focus 2007, 22, E4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Matsushita, K.; Morrell, C.N.; Cambien, B.; Yang, S.X.; Yamakuchi, M.; Bao, C.; Hara, M.R.; Quick, R.A.; Cao, W.; O’Rourke, B.; et al. Nitric oxidere gulates exocytosis by S-nitrosylation of N-ethylmaleimi desensitive factor. Cell 2003, 115, 139–150. [Google Scholar] [CrossRef] [Green Version]
  59. Iadecola, C.; Gorelick, P.B. The Janus face of cyclooxygenase-2 in ischemic stroke: Shifting toward downstream targets. Stroke 2005, 36, 182–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kuhn, D.M.; Sakowski, S.A.; Sadidi, M.; Geddes, T.J. Nitrotyrosine as a marker for peroxynitrite-induced neurotoxicity:the beginning or the end of the end of dopamine neurons? J. Neurochem. 2004, 89, 529–536. [Google Scholar] [CrossRef]
  61. Besson, V.C. Drug targets for traumatic brain injury from poly(ADP-ribose)polymerase pathway modulation. Br. J. Pharmacol. 2009, 157, 695–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Riobó, N.A.; Clementi, E.; Melani, M.; Boveris, A.; Cadenas, E.; Moncada, S.; Poderoso, J.J. Nitric oxide inhibits mitochondrial NADH:ubiquinone reductase activity through peroxynitrite formation. Biochem. J. 2001, 359, 139–145. [Google Scholar] [CrossRef]
  63. Aulak, K.S.; Koeck, T.; Crabb, J.W.; Stuehr, D.J. Dynamics of protein nitration in cells and mitochondria. Am. J. Physiol. Heart Circ. Physiol. 2004, 286, H30–H38. [Google Scholar] [CrossRef] [Green Version]
  64. Guy, R.A.; Maguire, G.F.; Crandall, I.; Connelly, P.W.; Kain, K.C. Characterization of peroxynitrite-oxidized low density lipoprotein binding to human CD36. Atherosclerosis 2001, 155, 19–28. [Google Scholar] [CrossRef]
  65. Okamoto, T.; Akaike, T.; Sawa, T.; Miyamoto, Y.; van der Vliet, A.; Maeda, H. Activation of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation via disulfide S-oxide formation. J. Biol. Chem. 2001, 276, 29596–29602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Viappiani, S.; Nicolescu, A.C.; Holt, A.; Sawicki, G.; Crawford, B.D.; León, H.; van Mulligen, T.; Schulz, R. Activation and modulation of 72 kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochem. Pharmacol. 2009, 77, 826–834. [Google Scholar] [CrossRef] [PubMed]
  67. Pluta, R.; Kida, E.; Lossinsky, A.S.; Golabek, A.A.; Mossakowski, M.J.; Wisniewski, H.M. Complete cerebral ischemia with short-term survival in rats induced by cardiac arrest: I. Extracellular accumulation of Alzheimer’s β-amyloid protein precursor in the brain. Brain Res. 1994, 649, 323–328. [Google Scholar] [CrossRef]
  68. Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Januszewski, S.; Jabłoński, M.; Gil-Kulik, P.; Brzozowska, J.; Petniak, A.; Furmaga-Jabłońska, W.; Bogucki, J.; et al. Dysregulation of amyloid precursor protein, β-secretase, presenilin 1 and 2 genes in the rat selectively vulnerable CA1 subfield of hippocampus following transient global brain ischemia. J. Alzheimers Dis. 2015, 47, 1047–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Pluta, R.; Ułamek-Kozioł, M.; Kocki, J.; Bogucki, J.; Januszewski, S.; Bogucka-Kocka, A.; Czuczwar, S.J. Expression of the tau protein and amyloid protein precursor processing genes in the CA3 area of the hippocampus in the ischemic model of Alzheimer’s disease in the rat. Mol. Neurobiol. 2020, 57, 1281–1290. [Google Scholar] [CrossRef] [Green Version]
  70. 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. [Google Scholar] [CrossRef]
  71. Lee, P.H.; Bang, O.Y.; Hwang, E.M.; Lee, J.S.; Joo, U.S.; Mook-Jung, I.; Huh, K. Circulating beta amyloid protein is elevated in patients with acute ischemic stroke. J. Neural Transm. 2005, 112, 1371–1379. [Google Scholar] [CrossRef]
  72. Zetterberg, H.; Mörtberg, E.; Song, L.; Chang, L.; Provuncher, G.K.; Patel, P.P.; Ferrell, E.; Fournier, D.R.; Kan, C.W.; Campbell, T.G.; et al. Hypoxia due to cardiac arrest induces a time-dependent increase in serum amyloid β levels in humans. PLoS ONE 2011, 6, e28263. [Google Scholar] [CrossRef] [Green Version]
  73. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Petniak, A.; Gil-Kulik, P.; Januszewski, S.; Bogucki, J.; Jabłoński, M.; Brzozowska, J.; Furmaga-Jabłońska, W.; et al. Discrepancy in expression of β-secretase and amyloid-β protein precursor in Alzheimer-related genes in the rat medial temporal lobe cortex following transient global brain ischemia. J. Alzheimers Dis. 2016, 51, 1023–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Gil-Kulik, P.; Januszewski, S.; Jabłoński, M.; Petniak, A.; Brzozowska, J.; Bogucki, J.; et al. Alzheimer associated presenilin 2 gene is dysregulated in rat medial temporal lobe cortex after complete brain ischemia due to cardiac arrest. Pharmacol. Rep. 2016, 68, 155–161. [Google Scholar] [CrossRef] [PubMed]
  75. Pluta, R.; Barcikowska, M.; Dębicki, G.; Ryba, M.; Januszewski, S. Changes in amyloid precursor protein and apolipoprotein E immunoreactivity following ischemic brain injury in rat with long-term survival: Influence of idebenone treatment. Neurosci. Lett. 1997, 232, 95–98. [Google Scholar] [CrossRef]
  76. Pluta, R.; Barcikowska, M.; Mossakowski, M.J.; Zelman, I. Cerebral accumulation of beta-amyloid following ischemic brain injury with long-term survival. Acta Neurochir. 1998, 71, 206–208. [Google Scholar]
  77. Hall, E.D.; Oostveen, J.A.; Dunn, E.; Carter, D.B. Increased amyloid protein precursor and apolipoprotein E immunoreactivity in the selectively vulnerable hippocampus following transient forebrain ischemia in gerbils. Exp. Neurol. 1995, 135, 17–27. [Google Scholar] [CrossRef]
  78. Tomimoto, H.; Akiguchi, I.; Wakita, H.; Nakamura, S.; Kimura, J. Ultrastructural localization of amyloid protein precursor in the normal and postischemic gerbil brain. Brain Res. 1995, 672, 187–195. [Google Scholar] [CrossRef]
  79. Ishimaru, H.; Ishikawa, K.; Haga, S.; Shoji, M.; Ohe, Y.; Haga, C.; Sasaki, A.; Takashashi, A.; Maruyama, Y. Accumulation of apolipoprotein E and β-amyloid-like protein in a trace of the hippocampal CA1 pyramidal cell layer after ischaemic delayed neuronal death. Neuro Rep. 1996, 7, 3063–3067. [Google Scholar] [CrossRef] [PubMed]
  80. Yokota, M.; Saido, T.C.; Tani, E.; Yamaura, I.; Minami, N. Cytotoxic fragment of amyloid precursor protein accumulates in hippocampus after global forebrain ischemia. JCBFM 1996, 16, 1219–1223. [Google Scholar] [CrossRef] [Green Version]
  81. Lin, B.; Schmidt-Kastner, R.; Busto, R.; Ginsberg, M.D. Progressive parenchymal deposition of β-amyloid precursor protein in rat brain following global cerebral ischemia. Acta Neuropathol. 1999, 97, 359–368. [Google Scholar] [CrossRef]
  82. Pluta, R. The role of apolipoprotein E in the deposition of β-amyloid peptide during ischemia–reperfusion brain injury. A model of early Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2000, 903, 324–334. [Google Scholar] [CrossRef]
  83. Sinigaglia-Coimbra, R.; Cavalheiro, E.A.; Coimbra, C.G. Postischemic hypertermia induces Alzheimer-like pathology in the rat brain. Acta Neuropathol. 2002, 103, 444–452. [Google Scholar] [CrossRef]
  84. Fujioka, M.; Taoka, T.; Matsuo, Y.; Mishima, K.; Ogoshi, K.; Kondo, Y.; Isuda, M.; Fujiwara, M.; Asano, T.; Sakaki, T.; et al. Magnetic resonance imaging shows delayed ischemic striatal neurodegeneration. Ann. Neurol. 2003, 54, 732–747. [Google Scholar] [CrossRef]
  85. Pluta, R.; Ułamek, M.; Jabłoński, M. Alzheimer’s mechanisms in ischemic brain degeneration. Anat. Rec. 2009, 292, 1863–1881. [Google Scholar] [CrossRef]
  86. Jabłoński, M.; Maciejewski, R.; Januszewski, S.; Ułamek, M.; Pluta, R. One year follow up in ischemic brain injury and the role of Alzheimer factors. Physiol. Res. 2011, 60 (Suppl. S1), 113–119. [Google Scholar] [CrossRef] [PubMed]
  87. Pluta, R. Glial expression of the β-amyloid peptide in cardiac arrest. J. Neurol Sci. 2002, 203–204, 277–280. [Google Scholar] [CrossRef]
  88. Pluta, R. Astroglial expression of the beta-amyloid in ischemia-reperfusion brain injury. Ann. N. Y. Acad. Sci. 2002, 977, 102–108. [Google Scholar] [CrossRef]
  89. Pluta, R.; Januszewski, S.; Jabłoński, M.; Ułamek, M. Factors in creepy delayed neuronal death in hippocampus following brain ischemia-reperfusion injury with long-term survival. Acta Neurochir. 2010, 106, 37–41. [Google Scholar]
  90. Pluta, R. Blood–brain barrier dysfunction and amyloid precursor protein accumulation in microvascular compartment following ischemia–reperfusion brain injury with 1-year survival. Acta Neurochir. 2003, 86, 117–122. [Google Scholar]
  91. Pluta, R. Pathological opening of the blood–brain barrier to horseradish peroxidase and amyloid precursor protein following ischemia–reperfusion brain injury. Chemotherapy 2005, 51, 223–226. [Google Scholar] [CrossRef] [PubMed]
  92. Pluta, R.; Ułamek, M.; Januszewski, S. Micro-blood–brain barrier openings and cytotoxic fragments of amyloid precursor protein accumulation in white matter after ischemic brain injury in long-lived rats. Acta Neurochir. 2006, 96, 267–271. [Google Scholar]
  93. Van Groen, T.; Puurunen, K.; Maki, 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. [Google Scholar] [CrossRef] [Green Version]
  94. Banati, R.B.; Gehrmann, J.; Wießner, C.; Hossmann, K.A.; Kreutzberg, G.W. Glial expression of the β-amyloid precursor protein (APP) in global ischemia. JCBFM 1995, 15, 647–654. [Google Scholar] [CrossRef]
  95. Palacios, G.; Mengod, G.; Tortosa, A.; Ferrer, I.; Palacios, J.M. Increased β-amyloid precursor protein expression in astrocytes in the gerbil hippocampus following ischaemia: Association with proliferation of astrocytes. Eur. J. Neurosci. 1995, 7, 501–510. [Google Scholar] [CrossRef] [PubMed]
  96. Nihashi, T.; Inao, S.; Kajita, Y.; Kawai, T.; Sugimoto, T.; Niwa, M.; Kabeya, R.; Hata, N.; Hayashi, S.; Yoshida, J. Expression and distribution of beta amyloid precursor protein and beta amyloid peptide in reactive astrocytes after transient middle cerebral artery occlusion. Acta Neurochir. 2001, 143, 287–295. [Google Scholar] [CrossRef] [PubMed]
  97. Badan, I.; Platt, D.; Kessler, C.; Popa-Wagner, A. Temporal dynamics of degenerative and regenerative events associated with cerebral ischemia in aged rats. Gerontology 2003, 49, 356–365. [Google Scholar] [CrossRef] [PubMed]
  98. Badan, I.; Dinca, I.; Buchhold, B.; Suofu, Y.; Walker, L.; Gratz, M.; Platt, D.; Kessler, C.H.; Popa-Wagner, A. Accelerated accumulation of N- and C-terminal beta APP fragments and delayed recovery of microtubule-associated protein 1B expression following stroke in aged rats. Eur. J. Neurosci. 2004, 19, 2270–2280. [Google Scholar] [CrossRef]
  99. Pluta, R.; Kocki, J.; Maciejewski, R.; Ułamek-Kozioł, M.; Jabłoński, M.; Bogucka-Kocka, A.; Czuczwar, S.J. Ischemia signaling to Alzheimer-related genes. Folia Neuropathol. 2012, 50, 322–329. [Google Scholar] [CrossRef] [Green Version]
  100. Pluta, R.; Jabłoński, M.; Czuczwar, S.J. Postischemic dementia with Alzheimer phenotype: Selectively vulnerable versus resistant areas of the brain and neurodegeneration versus β-amyloid peptide. Folia Neuropathol. 2012, 50, 101–109. [Google Scholar]
  101. Jendroska, K.; Poewe, W.; Daniel, S.E.; Pluess, J.; Iwerssen-Schmidt, H.; Paulsen, J.; Barthel, S.; Schelosky, L.; Cervos-Navarro, J.; DeArmond, S.J. Ischemic stress induces deposition of amyloid beta immunoreactivity in human brain. Acta Neuropathol. 1995, 90, 461–466. [Google Scholar] [CrossRef]
  102. Wisniewski, H.M.; Maslinska, D. Beta-protein immunoreactivity in the human brain after cardiac arrest. Folia Neuropathol. 1996, 34, 65–71. [Google Scholar]
  103. Jendroska, K.; Hoffmann, O.M.; Patt, S. Amyloid β peptide and precursor protein (APP) in mild and severe brain ischemia. Ann. N. Y. Acad. Sci. 1997, 826, 401–405. [Google Scholar] [CrossRef]
  104. Yatin, S.M.; Varadarajan, S.; Butterfield, D.A. Vitamin E prevents Alzheimer’s amyloid beta-peptide (1-42)-induced neuronal protein oxidation and reactive oxygen species production. J. Alzheimers Dis. 2000, 2, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Butterfield, D.A.; Boyd-Kimball, D. The critical role of methionine 35 in Alzheimer’s amyloid beta-peptide (1-42)-induced oxidative stress and neurotoxicity. Biochim. Biophys. Acta 2005, 1703, 149–156. [Google Scholar] [CrossRef] [PubMed]
  106. Butterfield, D.A.; Swomley, A.M.; Sultana, R. Amyloid β-peptide (1-42)-induced oxidative stress in Alzheimer disease: Importance in disease pathogenesis and progression. Antioxid. Redox Signal. 2013, 19, 823–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Schoneich, C.; Pogocki, D.; Hug, G.L.; Bobrowski, K. Free radical reactions of methionine in peptides: Mechanisms relevant to beta-amyloid oxidation and Alzheimer’s disease. J. Am. Chem. Soc. 2003, 125, 13700–13713. [Google Scholar] [CrossRef]
  108. Stadtman, E.R. Cyclic oxidation and reduction of methionine residues of proteins in antioxidant defense and cellular regulation. Arch Biochem. Biophys. 2004, 423, 2–5. [Google Scholar] [CrossRef]
  109. Varadarajan, S.; Yatin, S.; Aksenova, M.; Butterfield, D.A. Review: Alzheimer’s amyloid beta-peptide-associated free radical oxidative stress and neurotoxicity. J. Struct Biol. 2000, 130, 184–208. [Google Scholar] [CrossRef] [PubMed]
  110. Luo, S.; Levine, R.L. Methionine in proteins defends against oxidative stress. FASEB J. 2009, 23, 464–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Misiti, F.; Clementi, M.E.; Giardina, B. Oxidation of methionine 35 reduces toxicity of the amyloid beta-peptide(1-42) in neuroblastoma cells (IMR-32) via enzyme methionine sulfoxide reductase a expression and function. Neurochem. Int. 2010, 56, 597–602. [Google Scholar] [CrossRef]
  112. Jomova, K.; Vondrakova, D.; Lawson, M.; Valko, M. Metals, oxidative stress and neurodegenerative disorders. Mol. Cell Biochem. 2010, 345, 91–104. [Google Scholar] [CrossRef]
  113. Wan, L.; Nie, G.; Zhang, J.; Luo, Y.; Zhang, P.; Zhang, Z.; Zhao, B. Beta-amyloid peptide increases levels of iron content and oxidative stress in human cell and Caenorhabditis elegans models of Alzheimer disease. Free Radic. Biol. Med. 2010, 50, 122–129. [Google Scholar] [CrossRef]
  114. Butterfield, D.A.; Galvan, V.; Lange, M.B.; Tang, H.; Sowell, R.A.; Spilman, P.; Fombonne, J.; Gorostiza, O.; Zhang, J.; Sultana, R.; et al. In vivo oxidative stress in brain of Alzheimer disease transgenic mice: Requirement for methionine 35 in amyloid beta-peptide of APP. Free Radic. Biol. Med. 2010, 48, 136–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Dewar, D.; Graham, D.I.; Teasdale, G.M.; McCulloch, J. Cerebral ischemia induces alterations in tau and ubiquitin proteins. Dementia 1994, 5, 168–173. [Google Scholar] [CrossRef] [PubMed]
  116. Geddes, J.W.; Schwab, C.; Craddock, S.; Wilson, J.L.; Pettigrew, L.C. Alterations in tau immunostaining in the rat hippocampus following transient cerebral ischemia. JCBFM 1994, 14, 554–564. [Google Scholar]
  117. Dewar, D.; Dawson, D. Tau protein is altered by focal cerebral ischaemia in the rat: An immunohistochemical and immunoblotting study. Brain Res. 1995, 684, 70–78. [Google Scholar] [CrossRef]
  118. Irving, E.A.; Yatsushiro, K.; McCulloch, J.; Dewar, D. Rapid alteration of tau in oligodendrocytes after focal ischemic injury in the rat: Involvement of free radicals. JCBFM 1997, 17, 612–622. [Google Scholar] [CrossRef] [PubMed]
  119. Uchihara, T.; Nakamura, A.; Arai, T.; Ikeda, K.; Tsuchiya, K. Microglial tau undergoes phosphorylation-independent modification after ischemia. Glia 2004, 45, 180–187. [Google Scholar] [CrossRef]
  120. Majd, S.; Power, J.H.; Koblar, S.A.; Grantham, H.J. Early glycogen synthase kinase-3 and protein phosphatase 2A independent tau dephosphorylation during global brain ischaemia and reperfusion following cardiac arrest and the role of the adenosine monophosphate kinase pathway. Eur. J. Neurosci. 2016, 44, 1987–1997. [Google Scholar] [CrossRef] [Green Version]
  121. Fujii, H.; Takahashi, T.; Mukai, T.; Tanaka, S.; Hosomi, N.; Maruyama, H.; Sakai, N.; Matsumoto, M. Modifications of tau protein after cerebral ischemia and reperfusion in rats are similar to those occurring in Alzheimer’s disease—Hyperphosphorylation and cleavage of 4- and 3-repeat tau. JCBFM 2017, 37, 2441–2457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Schiefecker, A.J.; Putzer, G.; Braun, P.; Martini, J.; Strapazzon, G.; Antunes, A.P.; Mulino, M.; Pinggera, D.; Glodny, B.; Brugger, H.; et al. Total tau protein as investigated by cerebral microdialysis increases in hypothermic cardiac arrest: A pig study. Ther. Hypothermia Temp. Manag. 2021, 11, 28–34. [Google Scholar] [CrossRef]
  123. Stamer, K.; Vogel, R.; Thies, E.; Mandelkow, E.; Mandelkow, E.M. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J. Cell Biol. 2002, 156, 1051–1063. [Google Scholar] [CrossRef]
  124. Mörtberg, E.; Zetterberg, H.; Nordmark, J.; Blennow, K.; Catry, C.; Decraemer, H.; Vanmechelen, E.; Rubertsson, S. Plasma tau protein in comatose patients after cardiac arrest treated with therapeutic hypothermia. Acta Anaesthesiol. Scand. 2011, 55, 1132–1138. [Google Scholar] [CrossRef]
  125. Randall, J.; Mörtberg, E.; Provuncher, G.K.; Fournier, D.R.; Duffy, D.C.; Rubertsson, S.; Blennow, K.; Zetterberg, H.; Wilson, D.H. Tau proteins in serum predict neurological outcome after hypoxic brain injury from cardiac arrest: Results of a pilot study. Resuscitation 2013, 84, 351–356. [Google Scholar] [CrossRef]
  126. De Vos, A.; Bjerke, M.; Brouns, R.; De Roeck, N.; Jacobs, D.; Van den Abbeele, L.; Guldolf, K.; Zetterberg, H.; Blennow, K.; Engelborghs, S.; et al. Neurogranin and tau in cerebrospinal fluid and plasma of patients with acute ischemic stroke. BMC Neurol. 2017, 17, 170. [Google Scholar] [CrossRef] [Green Version]
  127. Banks, W.A.; Kovac, A.; Majerova, P.; Bullock, K.M.; Shi, M.; Zhang, J. Tau proteins cross the blood-brain barrier. J. Alzheimers Dis. 2017, 597, 411–419. [Google Scholar] [CrossRef] [PubMed]
  128. Wen, Y.; Yang, S.; Liu, R.; Simpkins, J.W. Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res. 2004, 1022, 30–38. [Google Scholar] [CrossRef] [PubMed]
  129. Morioka, M.; Kawao, T.; Yano, S.; Kai, Y.; Tsuiki, H.; Yoshinaga, Y.; Mtsumoto, J.; Maeda, T.; Hamada, J.; Yamamoto, H.; et al. Hyperphosphorylation at serine 199/202 of tau factor in the gerbil hippocampus after transient forebrain ischemia. Biochem. Biophys. Res. Commun. 2006, 347, 273–278. [Google Scholar] [CrossRef]
  130. Wen, Y.; Yang, S.; Liu, R.; Brun-Zinkernagel, A.M.; Koulen, P.; Simpkins, J.W. Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and Alzheimer’s disease-like tauopathy in female rats. J. Biol. Chem. 2004, 279, 22684–22692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Wen, Y.; Yang, S.H.; Liu, R.; Perez, E.J.; Brun-Ziukemagel, A.M.; Koulen, P.; Simpkins, J.W. Cdk5 is involved in NFT-like tauopathy induced by transient cerebral ischemia in female rats. Biochim. Biophys. Acta 2007, 1772, 473–483. [Google Scholar] [CrossRef]
  132. Khan, S.; Yuldasheva, N.Y.; Batten, T.F.C.; Pickles, A.R.; Kellett, K.A.B.; Saha, S. Tau pathology and neurochemical changes associated with memory dysfunction in an optimized murine model of global cerebral ischaemia–A potential model for vascular dementia? Neurochem. Int. 2018, 118, 134–144. [Google Scholar] [CrossRef] [Green Version]
  133. Kato, T.; Hirano, A.; Katagiri, T.; Sasaki, H.; Yamada, S. Neurofibrillary tangle formation in the nucleus basalis of Meynert ipsilateral to a massive cerebral infarct. Ann. Neurol. 1988, 23, 620–623. [Google Scholar] [CrossRef]
  134. 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. [Google Scholar] [CrossRef] [PubMed]
  135. Zhu, X.; Lee, H.G.; Casadesus GAvila, J.; Drew, K.; Perry, G.; Smith, M.A. Oxidative imbalance in Alzheimer’s disease. Mol. Neurobiol. 2005, 31, 205–217. [Google Scholar] [CrossRef]
  136. Su, B.; Wang, X.; Lee, H.-G.; Tabaton, M.; Perry, G.; Smith, M.A.; Zhu, X. Chronic oxidative stress causes increased tau phosphorylation in M17 neuroblastoma cells. Neurosc. Lett. 2010, 468, 267–271. [Google Scholar] [CrossRef] [PubMed]
  137. Alavi Naini, S.M.; Soussi-Yanicostas, N. Tau hyperphosphorylation and oxidative stress, a critical vicious circle in neurodegenerative tauopathies? Oxid. Med. Cell. Longev. 2015, 2015, 151979. [Google Scholar] [CrossRef] [Green Version]
  138. Perez, M.; Cuadros, R.; Smith, M.A.; Perry, G.; Avila, J. Phosphorylated, but not native, tau protein assembles following reaction with the lipid peroxidation product, 4-hydroxy-2-nonenal. FEBS Lett. 2000, 486, 270–274. [Google Scholar] [CrossRef] [Green Version]
  139. Gomez-Ramos, A.; JDıaz-Nido, J.; Smith, M.A.; Perry, G.; Avila, J. Effect of the lipid peroxidation product acrolein on tau phosphorylation in neural cells. J. Neurosci. Res. 2003, 71, 863–870. [Google Scholar] [CrossRef] [PubMed]
  140. Liu, Q.; Smith, M.A.; Avila, J.; DeBernardis, J.; Kansal, M.; Takeda, A.; Zhu, X.; Nunomura, A.; Honda, K.; Moreira, P.I.; et al. Alzheimer-specific epitopes of tau represent lipid peroxidation-induced conformations. Free Radic. Biol. Med. 2005, 38, 746–754. [Google Scholar] [CrossRef]
  141. Gamblin, T.C.; King, M.E.; Kuret, J.; Berry, R.W.; Binder, L.I. Oxidative regulation of fatty acid-induced tau polymerization. Biochemistry 2000, 39, 14203–14210. [Google Scholar] [CrossRef]
  142. Melov, S.; Adlard, P.A.; Morten, K.; Johnson, F.; Golden, T.R.; Hinerfeld, D.; Schilling, B.; Mavros, C.; Masters, C.L.; Volitakis, I.; et al. Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS ONE 2007, 2, e536. [Google Scholar] [CrossRef]
  143. Stover, P.J. One-carbon metabolism-genome interactions in folate-associated pathologies. J. Nutr. 2009, 139, 2402–2405. [Google Scholar] [CrossRef] [Green Version]
  144. Kao, T.T.; Chu, C.Y.; Lee, G.H.; Hsiao, T.H.; Cheng, N.W.; Chang, N.S.; Chen, B.H.; Fu, T.F. Folate deficiency-induced oxidative stress contributes to neuropathy in young and aged zebrafish-implication in neural tube defects and Alzheimer’s diseases. Neurobiol. Dis. 2014, 71, 234–244. [Google Scholar] [CrossRef] [PubMed]
  145. Lovell, M.A.; Xiong, S.; Xie, C.; Davies, P.; Markesbery, W.R. Induction of hyperphosphorylated tau in primary rat cortical neuron cultures mediated by oxidative stress and glycogen synthase kinase-3. J. Alzheimers Dis. 2004, 6, 659–671; discussion 673–681. [Google Scholar] [CrossRef] [PubMed]
  146. Lee, K.Y.; Koh, S.H.; Noh, M.Y.; Park, K.W.; YJ Lee, Y.J.; Kim, S.H. Glycogen synthase kinase-3β activity plays very important roles in determining the fate of oxidative stress inflicted neuronal cells. Brain Res. 2007, 1129, 89–99. [Google Scholar] [CrossRef]
  147. Tamagno, E.; Parola, M.; Bardini, P.; Piccini, A.; Borghi, R.; Guglielmotto, M.; Santoro, G.; Davit, A.; Danni, O.; Smith, M.A.; et al. β-site APP cleaving enzyme up-regulation induced by 4-hydroxynonenal is mediated by stress-activated protein kinases pathways. J. Neurochem. 2005, 92, 628–636. [Google Scholar] [CrossRef] [PubMed]
  148. Li, P.; Stetler, A.; Leak, R.K.; Shi, Y.; Li, Y.; Yu, W.; Bennett, M.V.L.; Chen, J. Oxidative stress and DNA damage after cerebral ischemia: Potential therapeutic targets to preserve the genome and improve stroke recovery. Neuropharmacology 2018, 1349 Pt B, 208–217. [Google Scholar] [CrossRef]
  149. Madsen, P.M.; Pinto, M.; Patel, S.; McCarthy, S.; Gao, H.; Taherian, M.; Karmally, S.; Pereira, C.V.; Dvoriantchikova, G.; Ivanov, D.; et al. Mitochondrial DNA double-strand breaks in oligodendrocytes cause demyelination, axonal injury, and CNS inflammation. J. Neurosci. 2017, 37, 10185–10199. [Google Scholar] [CrossRef] [Green Version]
  150. Stetler, R.A.; Gao, Y.; Leak, R.K.; Weng, Z.; Shi, Y.; Zhang, L.; Pu, H.; Zhang, F.; Hu, X.; Hassan, S.; et al. APE1/Ref-1 facilitates recovery of gray and white matter and neurological function after mild stroke injury. Proc. Natl. Acad. Sci. USA 2016, 113, E3558–E3567. [Google Scholar] [CrossRef] [Green Version]
  151. Jadavji, N.M.; Farr, T.D.; Lips, J.; Khalil, A.A.; Boehm-Sturm, P.; Foddis, M.; Harms, C.; Fuchtemeier, M.; Dirnagl, U. Elevated levels of plasma homocysteine, deficiencies in dietary folic acid and uracil-DNA glycosylase impair learning in a mouse model of vascular cognitive impairment. Behav. Brain Res. 2015, 283, 215–226. [Google Scholar] [CrossRef] [Green Version]
  152. Rohn, T.T.; Hinds, T.R.; Vincenzi, F.F. Inhibition of Ca2+-pump ATPase and the Na+/K+-pump ATPase by iron-generated free radicals: Protection by 6,7-dimethyl-2,4-di-1-pyrrolidinyl-7h-pyrrolo [2,3-d]pyrimidine sulfate (U-89843D), a potent, novel, antioxidant/free radical scavenger. Biochem. Pharmacol. 1996, 51, 471–476. [Google Scholar] [CrossRef]
  153. Kourie, J.I. Interaction of reactive oxygen species with ion transport mechanisms. Am. J. Physiol. 1998, 275, C1–C24. [Google Scholar] [CrossRef]
  154. Sombati, S.; Coulter, D.A.; DeLorenzo, R.J. Neurotoxic activation of glutamate receptors induces an extended neuronal depolarization in cultured hippocampal neurons. Brain Res. 1991, 566, 316–319. [Google Scholar] [CrossRef]
  155. Sandoval, K.E.; Witt, K.A. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol. Dis. 2008, 32, 200–219. [Google Scholar] [CrossRef]
  156. Guix, F.X.; Uribesalgo, I.; Coma, M.; Munoz, F.J. The physiology and pathophysiology of nitric oxide in the brain. Prog. Neurobiol. 2005, 76, 126–152. [Google Scholar] [CrossRef] [PubMed]
  157. Miller, A.A.; Drummond, G.R.; De Silva, T.M.; Mast, A.E.; Hickey, H.; Williams, J.P.; Broughton, B.R.; Sobey, C.G. NADPH oxidase activity is higher in cerebral versus systemic arteries of four animal species: Role of Nox2. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H220–H225. [Google Scholar] [CrossRef] [PubMed]
  158. Mathiisen, T.M.; Lehre, K.P.; Danbolt, N.C.; Ottersen, O.P. The perivascular astroglial sheath provides a complete covering of the brain microvessels: An electron microscopic 3D reconstruction. Glia 2010, 58, 1094–1103. [Google Scholar] [CrossRef] [PubMed]
  159. Van der Goes, A.; Wouters, D.; Van Der Pol, S.M.; Huizinga, R.; Ronken, E.; Adamson, P.; Greenwood, J.; Dijkstra, C.D.; De Vries, H.E. Reactive oxygen species enhance the migration of monocytes across the blood-brain barrier in vitro. FASEB J. 2001, 15, 1852–1854. [Google Scholar] [CrossRef]
  160. Lee, H.S.; Namkoong, K.; Kim, D.H.; Kim, K.J.; Cheong, Y.H.; Kim, S.S.; Lee, W.B.; Kim, K.Y. Hydrogen peroxide-induced alterations of tight junction proteins in bovine brain microvascular endothelial cells. Microvasc. Res. 2004, 68, 231–238. [Google Scholar] [CrossRef]
  161. Kahles, T.; Luedike, P.; Endres, M.; Galla, H.J.; Steinmetz, H.; Busse, R.; Neumann-Haefelin, T.; Brandes, R.P. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 2007, 38, 3000–3006. [Google Scholar] [CrossRef] [Green Version]
  162. Adam-Vizi, V. Production of reactive oxygen species in brain mitochondria: Contribution by electron transport chain and non–electron transport chain sources. Antioxid. Redox Signal. 2005, 7, 1140–1149. [Google Scholar] [CrossRef]
  163. Chrissobolis, S.; Faraci, F.M. The role of oxidative stress and NADPH oxidase in cerebrovascular disease. Trends Mol. Med. 2008, 14, 495–502. [Google Scholar] [CrossRef] [Green Version]
  164. Lochhead, J.J.; McCaffrey, G.; Quigley, C.E.; Finch, J.; DeMarco, K.M.; Nametz, N.; Davis, T.P. Oxidative stress increases blood–brain barrier permeability and induces alterations in occludin during hypoxia–reoxygenation. JCBFM 2010, 30, 1625–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Fraser, P.A. The role of free radical generation in increasing cerebrovascular permeability. Free Radic. Biol. Med. 2011, 51, 967–977. [Google Scholar] [CrossRef]
  166. Lehner, C.; Gehwolf, R.; Tempfer, H.; Krizbai, I.; Hennig, B.; Bauer, H.C.; Bauer, H. Oxidative stress and blood–brain barrier dysfunction under particular consideration of matrix metalloproteinases. Antioxid. Redox Signal. 2011, 15, 1305–1323. [Google Scholar] [CrossRef]
  167. Sumii, T.; Lo, E.H. Involvement of matrix metalloproteinase in thrombolysis-associated hemorrhagic transformation after embolic focal ischemia in rats. Stroke 2002, 33, 831–836. [Google Scholar] [CrossRef] [Green Version]
  168. Wang, X.; Lee, S.R.; Arai, K.; Lee, S.R.; Tsuji, K.; Rebeck, G.W.; Lo, E.H. Lipoprotein receptor–mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat. Med. 2003, 9, 1313–1317. [Google Scholar] [CrossRef] [PubMed]
  169. Zhao, B.Q.; Tejima, E.; Lo, E.H. Neurovascular proteases in brain injury, hemorrhage and remodeling after stroke. Stroke 2007, 38, 748–752. [Google Scholar] [CrossRef] [Green Version]
  170. Ramos-Fernandez, M.; Bellolio, M.F.; Stead, L.G. Matrix metalloproteinase-9 as a marker for acute ischemic stroke: A systematic review. Stroke Cerebrovasc. Dis. 2011, 20, 47–54. [Google Scholar] [CrossRef] [PubMed]
  171. Gugliucci, A. Human plasminogen is highly susceptible to peroxynitrite inactivation. Clin. Chem. Lab. Med. 2003, 41, 1064–1068. [Google Scholar] [CrossRef]
  172. Nielsen, V.G.; Crow, J.P.; Zhou, F.; Parks, D.A. Peroxynitrite inactivates tissue plasminogen activator. Anesth. Analg. 2004, 98, 1312–1317. [Google Scholar] [CrossRef]
  173. Taffi, R.; Nanetti, L.; Mazzanti, L.; Bartolini, M.; Vignini, A.; Raffaelli, F.; Pasqualetti, P.; Vernieri, F.; Provinciali, L.; Silvestrini, M. Plasma levels of nitric oxide and stroke outcome. J. Neurol. 2008, 255, 94–98. [Google Scholar] [CrossRef]
  174. Dias-Santagata, D.; Fulga, T.A.; Duttaroy, A.; Feany, M.B. Oxidative stress mediates tau-induced neurodegeneration in Drosophila. J. Clin. Investig. 2007, 117, 236–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Katz, A.; Brosnahan, S.B.; Papadopoulos, J.; Parnia, S.; Lam, J.Q. Pharmacologic neuroprotection in ischemic brain injury after cardiac arrest. Ann. N. Y. Acad. Sci. 2021; online ahead of print. [Google Scholar] [CrossRef]
  176. Zhang, M.; Li, W.; Wang, Y.; Wang, T.; Ma, M.; Tian, C. Association between the change of serum copper and ischemic stroke: A systematic review and meta-analysis. J. Mol. Neurosci. 2020, 70, 475–480. [Google Scholar] [CrossRef] [PubMed]
  177. Buendia, I.; Tenti, G.; Michalska, P.; Méndez-López, I.; Luengo, E.; Satriani, M.; Padín-Nogueira, F.; López, M.G.; Ramos, M.T.; García, A.G.; et al. ITH14001, a CGP37157-nimodipine hybrid designed to regulate calcium homeostasis and oxidative stress, exerts neuroprotection in cerebral ischemia. ACS Chem. Neurosci. 2017, 8, 67–81. [Google Scholar] [CrossRef]
  178. De Gregorio-Rocasolano, N.; Martí-Sistac, O.; Gasull, T. Deciphering the iron side of stroke: Neurodegeneration at the crossroads between iron dyshomeostasis, excitotoxicity, and ferroptosis. Front. Neurosci. 2019, 13, 85. [Google Scholar] [CrossRef] [Green Version]
  179. Yoshitomi, T.; Nagasaki, Y. Self-assembling antioxidants for ischemia-reperfusion injuries. Antioxid. Redox Signal. 2021; online ahead of print. [Google Scholar] [CrossRef]
  180. Calabrese, V.; Boyd-Kimball, D.; Scapagnini, G.; Butterfield, D.A. Nitric oxide and cellular stress response in brain aging and neurodegenerative disorders: The role of vitagenes. In Vivo 2004, 18, 245–267. [Google Scholar]
  181. Calabrese, V.; Stella, A.M.; Butterfield, D.A.; Scapagnini, G. Redox regulation in neurodegeneration and longevity: Role of the heme oxygenase and HSP70 systems in brain stress tolerance. Antioxid. Redox Signal. 2004, 6, 895–913. [Google Scholar] [PubMed] [Green Version]
  182. Calabrese, E.J.; Agathokleous, E. Hormesis: Transforming disciplines that rely on the dose response. IUBMB Life 2022, 74, 8–23. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cross talks between amyloid, tau protein and oxidative stress during DNA injury and fragmentation and brain cells death in post-ischemic brain.
Figure 1. Cross talks between amyloid, tau protein and oxidative stress during DNA injury and fragmentation and brain cells death in post-ischemic brain.
Antioxidants 11 00146 g001
Figure 2. The interplay among oxidative stress, amyloid and tau protein in development necrosis, apoptosis and neurodegeneration. ↑—increase.
Figure 2. The interplay among oxidative stress, amyloid and tau protein in development necrosis, apoptosis and neurodegeneration. ↑—increase.
Antioxidants 11 00146 g002
Figure 3. Complications in brain after post-ischemic recirculation. BBB-blood-brain barrier.
Figure 3. Complications in brain after post-ischemic recirculation. BBB-blood-brain barrier.
Antioxidants 11 00146 g003
Figure 4. The interplay among amyloid, tau protein and oxidative stress during post-ischemic brain neurodegeneration of Alzheimer’s disease proteinopathy. ↓—decrease, ↑—increase, APP-amyloid protein precursor, ROS-reactive oxygen species, RNS-reactive nitrogen species.
Figure 4. The interplay among amyloid, tau protein and oxidative stress during post-ischemic brain neurodegeneration of Alzheimer’s disease proteinopathy. ↓—decrease, ↑—increase, APP-amyloid protein precursor, ROS-reactive oxygen species, RNS-reactive nitrogen species.
Antioxidants 11 00146 g004
Table 1. Post-ischemic expression of genes associated with Alzheimer’s disease in the CA1 and CA3 regions of the hippocampus.
Table 1. Post-ischemic expression of genes associated with Alzheimer’s disease in the CA1 and CA3 regions of the hippocampus.
GenesAPPADAM10BACE1PSEN1PSEN2MAPT
Survival
CA1 area
2 daysn.a.↑↑↑↑↑↑
7 daysn.a.
30 daysn.a.
CA3 area
2 days
7 days
30 days
Expression: ↑ increase; ↑↑ increase; ↓ decrease, ↔ oscillation around control values; n.a. not available. Genes: APP-amyloid protein precursor, BACE1-β-secretase, PSEN1-presnilin 1, PSEN2-presenilin 2, MAPT-tau protein.
Table 2. Post-ischemic expression of genes associated with Alzheimer’s disease in the medial temporal cortex.
Table 2. Post-ischemic expression of genes associated with Alzheimer’s disease in the medial temporal cortex.
GenesAPPBACE1PSEN1PSEN2
Survival
2 days↑↑↑↑
7 days
30 days
Expression: ↑ increase; ↑↑ increase; ↓ decrease, ↔ oscillation around control values. Genes: APP-amyloid protein precursor, BACE1-β-secretase, PSEN1-presnilin 1, PSEN2-presenilin 2.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pluta, R.; Kiś, J.; Januszewski, S.; Jabłoński, M.; Czuczwar, S.J. Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy. Antioxidants 2022, 11, 146. https://doi.org/10.3390/antiox11010146

AMA Style

Pluta R, Kiś J, Januszewski S, Jabłoński M, Czuczwar SJ. Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy. Antioxidants. 2022; 11(1):146. https://doi.org/10.3390/antiox11010146

Chicago/Turabian Style

Pluta, Ryszard, Jacek Kiś, Sławomir Januszewski, Mirosław Jabłoński, and Stanisław J. Czuczwar. 2022. "Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy" Antioxidants 11, no. 1: 146. https://doi.org/10.3390/antiox11010146

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop