Next Article in Journal
Cre-Recombinase Induces Apoptosis and Cell Death in Enterocyte Organoids
Next Article in Special Issue
Association between Dietary Total Antioxidant Capacity of Antioxidant Vitamins and the Risk of Stroke among US Adults
Previous Article in Journal
Preventive Effect of Cocoa Flavonoids via Suppression of Oxidative Stress-Induced Apoptosis in Auditory Senescent Cells
Previous Article in Special Issue
Puerarin Attenuates Oxidative Stress and Ferroptosis via AMPK/PGC1α/Nrf2 Pathway after Subarachnoid Hemorrhage in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease

Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2022, 11(8), 1447; https://doi.org/10.3390/antiox11081447
Submission received: 12 July 2022 / Revised: 22 July 2022 / Accepted: 25 July 2022 / Published: 26 July 2022

Abstract

:
Despite the reality that a death from cerebrovascular accident occurs every 3.5 min in the United States, there are few therapeutic options which are typically limited to a narrow window of opportunity in time for damage mitigation and recovery. Novel therapies have targeted pathological processes secondary to the initial insult, such as oxidative damage and peripheral inflammation. One of the greatest challenges to therapy is the frequently permanent damage within the CNS, attributed to a lack of sufficient neurogenesis. Thus, recent use of cell-based therapies for stroke have shown promising results. Unfortunately, stroke-induced inflammatory and oxidative damage limit the therapeutic potential of these stem cells. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been implicated in endogenous antioxidant and anti-inflammatory activity, thus presenting an attractive target for novel therapeutics to enhance stem cell therapy and promote neurogenesis. This review assesses the current literature on the concomitant use of stem cell therapy and Nrf2 targeting via pharmaceutical and natural agents, highlighting the need to elucidate both upstream and downstream pathways in optimizing Nrf2 treatments in the setting of cerebrovascular disease.

1. Introduction

There are two major types of cerebrovascular accidents, hemorrhagic (HS) and ischemic (IS) stroke. IS refers to an obstruction of blood flow to a region in the brain, ultimately leading to significant CNS damage. HS involves blood vessel damage and rupture in the cerebral vasculature, also resulting in substantial CNS damage. Approximately 87% of strokes are ischemic, with the remainder being hemorrhagic [1]. Nearly a quarter of patients with IS die, and half of patients with HS [2]. One-sixth of people around the world will have a stroke in their lifetime, and the yearly incidence of stroke is nearly 14 million. Furthermore, it is the fifth leading cause of death in the United Sates and the leading cause of long-term disability [3,4,5]. Despite these grave statistics, treatments for both types of strokes are severely limited. For IS, there are narrow time windows where thrombolytic therapies, such as tissue plasminogen activator (tPA) or mechanical thrombectomy (MT) can be employed. If these treatments are attempted outside of a 4.5 h time window, hemorrhagic transformation can occur and further detriment a patient’s prognosis [6,7,8,9,10]. HS has no proven clinical therapies [11]. With poor prognoses and little to no treatment available, it is vital that greater research focus is attributed to stroke therapy development.
While substantial brain damage occurs because of IS or HS conditions alone, it can be argued that an even greater contributor to stroke pathophysiology is secondary cell death mechanisms [3,12]. After both types of strokes, major cell death mechanisms include excitotoxicity, oxidative stress, free radical accumulation, mitochondrial dysfunction, inflammation, and impaired neurogenesis, angiogenesis, and vasculogenesis [13,14,15]. With such a wide range of pathophysiological mediators, ample treatment targets exist to be studied as potential stroke therapeutics. Nuclear factor erythroid 2-related factor 2 (NFE2L2/Nrf2) and its associated pathway is one such attractive target due to its role in sequestration of oxidative stress that will remain as the focus of this review.
Nrf2 binds to promoters of antioxidant genes and thus serves a crucial role in the attenuation of reactive oxygen/nitrogen species. Seeing that the brain is vulnerable to oxidative stress [16], activation of Nrf2 is an attractive target in the setting of ischemia such as cerebrovascular accidents. An E3 ubiquitin ligase complex consisting of several proteins including Kelch-like ECH-Associated Protein 1 (KEAP1), Cullin 3 (CUL3), and RING-box protein 1 (RBX1) serves to inhibit Nrf2 in the normal physiological setting. At baseline, the complex binds to Nrf2 for ubiquitination and subsequent proteasomal degradation of Nrf2 [17]. In the event of oxidative stress, KEAP1′s conformation adjusts via modification of cysteine thiols [18], allowing Nrf2 to accumulate. Nrf2 then travels to the cell nucleus, binds with small musculoaponeurotic fibrosarcoma (sMaf) proteins, and drives transcription of >250 cytoprotective genes. Many genes driven by Nrf2 are involved with glutathione synthesis or action. De novo synthesis of glutathione begins with the action of the enzyme γ-glutamylcysteine synthetase, which uses ATP hydrolysis to combine glutamate with cysteine to produce γ-glutamylcysteine. The enzyme glutathione synthetase then utilizes ATP to add glycine to the dipeptide to form glutathione [19,20]. Glutathione conjugates to xenobiotics to eliminate them; glutathione S-transferases (GSTs) are enzymes that assist in this manner [21]. Glutathione must be in its reduced form to act properly, which the enzyme glutathione reductase ensures [22]. Other examples of Nrf2 promoted genes include superoxide dismutase (SOD), catalase, heme oxygenase 1 (HO-1), and NAD(P)H quinone dehydrogenase 1 (NQO1).
Nrf2 plays a vital role in managing excessive oxidative stress following stroke, with a vital role in heme and iron metabolism, antioxidant proliferation, glutathione regeneration, thioredoxin, and protein recycling [23,24]. The major producers of Nrf2 in the brain include microglia and macrophages [25,26]. In fact, microglia and astrocytes produce an extreme amount of Nrf2 within 24 h following an induced middle cerebral artery occlusion (MCAO) [27]. In IS, hydrogen peroxide is elevated after ischemia and reperfusion, serving as a major stimulus for Nrf2 activation [5,28,29,30,31]. IS models in Nrf2-/- rats have greater damage after IS due to the loss of the protective Nrf2 properties [28,32,33]. While targeting Nrf2 has clear benefits for IS treatment, similar toxic ROS accumulation in HS suggests that Nrf2 may also be beneficial for HS treatment. HS Nrf2-/- models have poor prognoses, with larger hematomas, more functional defects, greater cell death, ROS, and cell damage [34,35]. The primary contributors heme, hemoglobin, and iron have been shown to induce Nrf2 signaling, further elucidating this signaling mechanism’s role in lessening oxidative damage following HS [36,37]. Within its producer cells, Nrf2 downstream proteins clear red blood cell debris and reduce oxidative damage [38]. One Nrf2-related protein mentioned above, HO-1, is an antioxidant which produces protective substances, carbon monoxide and biliverdin, and increases microglial capabilities to phagocytose HS damaged tissue [36,39]. HO-1, the stress-induced isoform of heme oxygenase, is significantly increased within 3–5 days following HS in rodents [40,41]. Other than HO-1 simply degrading proinflammatory heme into anti-inflammatory carbon monoxide and eventual bilirubin, it also decreases NF-κB signaling [42], which subsequently decreases proinflammatory cytokines including TNF-α and IL-6. Targeting any of the aforementioned ameliorators of oxidative stress has therapeutic potential, however, we argue that targeting the most upstream protein, Nrf2, will show the greatest therapeutic benefit.
A schematic representation of the Nrf2 pathway, downstream effectors, and molecular crosstalk with the NF-κB pathway can be seen in Figure 1.
Despite the described therapeutic effects of Nrf2 upregulation, it must be noted that such benefits of Nrf2 are dependent on disease state. Recent reviews have pointed to uncontrolled Nrf2 expression as the reason for chemoresistance through inhibition of oxidative stress in ovarian cancer [43] and gastric cancer [44]. Thus, the therapeutic benefits of Nrf2 are limited.
This review will probe scientific evidence for the reduction in oxidative stress following stroke by targeting the Nrf2 signaling pathway.

2. Neuroinflammation and Oxidative Stress in Stroke

Oxidative stress is a common feature of HS and IS; however, the processes vary due to different mechanisms of infarction. In IS, a cascade of events prompted by nutrient and oxygen deprivation promote cell death. Without ample oxygen, mitochondrial respiration is shut down, and ATP levels are depleted. Thus, ATPase pumps vital to cell equilibrium, including the Na+/K+ pump, are inoperative. Further along this cascade, Na+/Ca2+ pumps are disrupted, and Ca2+ accumulation occurs within neurons, promoting lipases, proteases, and other cell death pathways [45]. Mitochondria undergo programmed death processes and release cytochrome C, leading to apoptosis [46]. Further expression of the Fas ligand also instigates cell death pathways. This excessive cell death increases levels of reactive oxygen species (ROS) and exacerbates mitochondrial dysfunction and ATP shortages [47,48]. Furthermore, alterations in mitochondrial metabolic processes also increase ROS. Despite an already exorbitant number of pathological processes, cell death also causes excitotoxicity due to excessive glutamate release. Glutamate furthers the ongoing processes by stimulating even more Ca2+ influx, perpetuating a cycle of inflammation and cell death [49]. Apoptotic neurons, necrotic neurons, and their released products serve as damage-associated molecular patterns activating a cascade of inflammatory immune cells within the infarction area. Local microglia, astrocytes, and neutrophils release ROS, perpetuating oxidative stress within the brain after IS [47,50,51]. Later invasion of peripheral immune cells further worsens oxidative stress. Ultimately, the brain is faced with an overwhelmingly toxic proliferation of ROS after IS.
While cell death and release of ROS is also seen in HS, oxidative stress following HS has additional pathologic ROS contributions, primarily related to the large influx of hemoglobin and its metabolites [12]. Once a blood vessel ruptures, red blood cells flood the cerebral parenchyma and release hemoglobin after loss of membrane integrity [52]. Hemoglobin, composed of heme, is also broken down by macrophages and microglia. Primary constituents of heme include iron, biliverdin, and carbon monoxide [36,52]. Iron and heme ultimately contribute to the oxidative damage seen after HS, causing damage to the local neurovasculature and blood–brain barrier [39,53,54,55]. Iron promotes the Fenton reaction, a notorious contributor of free radicals [36,56,57]. Furthermore, iron can promote ferroptosis, an iron-dependent cell death, further enhancing oxidative damage via ROS, lipid peroxidation, mitochondrial changes, and inflammatory pathways [55,58,59]. Both IS and HS demonstrate extreme ROS damage, perpetuating a cycle of cell death and an injurious local environment. Reducing oxidative stress should be a central therapeutic goal for stroke, possibly revolutionizing stroke treatments.

3. Peripheral Inflammation and Nrf2

3.1. Mechanistic Interrogation of Peripheral Inflammation

In addition to the drastic oxidative damage observed after stroke, inflammation undoubtedly plays a major role in the clinical and functional outcomes of patients with ischemic or hemorrhagic stroke. Growing scientific evidence indicates that stroke should be treated as a multi-organ disease, rather than the traditional compartmentalization of the CNS and other affected organ systems (Figure 2) [60]. Local inflammatory processes have been clearly documented to cause damage to brain tissue [61], and a more recent body of evidence points to peripheral inflammation as also playing an important role in the pathogenesis of stroke. For example, plasma concentrations of IL-6 were closely correlated with infarct volume and stroke severity [62]. Other inflammatory markers such as C-Reactive Protein are also elevated upon the onset of stroke [63].
Organs that influence peripheral inflammation include but are not limited to the spleen [64], cervical lymph nodes [65], and bone marrow stem cells [66]. Of note, the spleen has been shown to decrease in size following stroke in rats subjected to MCAO [64,67,68]. This is largely due to the release of lymphocytes, monocytes, and neutrophils from the spleen due to upregulated catecholamines, as seen in stroke models such as MCAO [69,70,71]. These splenocytes play a significant role in inflammation, and accordingly, rats with splenectomy prior to the MCAO display an 82.3% decrease in infarct volume compared to those with an intact spleen [64]. Clinical studies also show an inverse correlation between spleen size and blood lymphocyte counts, while there is a positive correlation with neutrophil counts, further supporting evidence of the spleen’s role in peripheral inflammation [72]. Bone marrow has a relatively less understood role in peripheral inflammation following ischemic stroke, but recent studies in mice subjected to MCAO show activation of hematopoiesis via bone marrow β3 adrenoreceptors [66]. Similar to splenectomy, the removal of cervical lymph nodes decreases the extent of brain damage in the days after ischemic stroke. Additionally, the possible activation of VEGFR3 tyrosine kinase receptors on cervical lymph nodes by VEGF-C secretion from the brain stands as a potential cause for the initiation of peripheral immune response [73], tying together the multi-organ web that carries out the damaging changes we see from peripheral inflammation.
Although much of the damage occurs due to the inflammation itself, the immunodepression that follows poses a sizable challenge that cannot be ignored. Immunodepression is the body’s response to the acute inflammation that occurs post-stroke, but it can precipitate a wide range of infections, the most common of which are pneumonia and urinary tract infections [74]. This primarily occurs through the activation of the β-arrestin2-NF-κB [75] or cAMP-PKA-NF-κB pathways [76], exciting the adrenal medulla and increasing plasma catecholamine levels which ultimately decreases the amount of plasma lymphocytes [77]. Other pathways, such as the cholinergic anti-inflammatory pathway [78] and the hypothalamus–pituitary–adrenal axis [79], also exacerbate the risk of infection.
Nrf2′s role in post-stroke peripheral inflammation is still a topic of exploration. Recent research has shown Nrf2′s role to lie primarily in its complex transcriptional regulation of inflammatory pathways such as the NF-κB system [80] and subsequently, cytokines such as IL-6 and IL-1β [81,82]. Nrf2 has also been shown to regulate other cytokines such as COX-2 and iNOS in mouse models [83], although this has not yet been shown in MCAO mice. More research needs to be conducted to fully understand Nrf2′s role in post-stroke peripheral inflammation, particularly whether it impacts any of the organ systems described above, but the general wealth of knowledge on Nrf2′s role in other pathologies gives researchers a substantial incentive to investigate Nrf2 in stroke.

3.2. Stem Cells and Peripheral Inflammation

While stem cell therapy for stroke is an area of heavy focus, there is comparatively less research conducted on the role of stem cells in the context of peripheral inflammation. A perceived problem was that stem cells administered peripherally could not cross the blood–brain barrier, which limits their applications. However, recent research has shown that there is potential for stem cell therapy to modulate the peripheral immune response. Infusion of human umbilical cord blood cells was shown to restore spleen size and function in rats with MCAO [84,85], coincident with significant reduction in ischemic brain damage and improved behavioral performance [86]. Subsequently, multipotent adult progenitor cells have been shown to suppress groups of genes that upregulate inflammatory response from within the spleen [87]. This resulted in decreased levels of inflammatory cytokines such as IL-1β and TNF-α. These studies all show that targeting the spleen can significantly benefit stroke outcomes. More recent research has shown that bone marrow mesenchymal stem cells implanted into rat brains with MCAO preferentially migrate to the spleen [88]. Unfortunately, less is known about the applications of stem cells in the other organs involved in peripheral immunity during a stroke. As more is revealed about the intricacies of the brain’s post-stroke communication with organs in the body, researchers will be able to develop more targeted therapies to address peripheral immune response more precisely and effectively. Considering the substantial literature on Nrf2, alongside emerging literature using cell-based therapies for stroke, combining these two therapeutic modalities may be incredibly beneficial.

4. Current Nrf2-Directed Treatment Modalities

4.1. Stem Cell Therapies and Nrf2

Several initiatives have been implemented to examine the therapeutic role of exogenous stem cells for stroke treatments [13,89]. It is hypothesized that these cell-based therapies work via a bystander effect [90]. As opposed to the transplanted cells replacing the dead or dying ischemic cells, bystander effects refer to the grafted stem cells releasing protective factors against neuroinflammation and oxidative stress [91,92,93]. This ultimately promotes neurogenesis, angiogenesis, oligodendrogenesis, vasculogenesis, and synaptogenesis by releasing anti-inflammatory and repair signals [94]. Clinically and pre-clinically, stem cells such as fetal-derived neural stem cells, embryonic stem cells, bone marrow stem cells, umbilical cord stem cells, adipose stem cells, and induced pluripotent stem cells have been examined for stroke treatment and demonstrate promising results [74,95,96,97,98,99]. Unfortunately, the use of stem cells after stroke poses some difficulty due to the exorbitant inflammatory and oxidative stressors present within the infarcted area. Oxidative stress can induce autophagy, or cell-induced recycling of intracellular components. While beneficial and homeostatic in some instances, this process of ROS-induced autophagy can be of detriment to stem cell viability [100]. To optimize stem cell treatments and amplify cell survival for the most beneficial results, oxidative stress should be reduced prior to or concurrently with stem cell administration.
Endogenously, adult human neurogenesis is incredibly limited to only certain locations of the brain such as the subgranular zone of the dentate gyrus and subventricular zone of the lateral ventricles [101], but neural stem cells (NSCs) do reside there and are able to differentiate into neurons and certain glia, including oligodendrocytes and astrocytes [102,103]. NSC survival and differentiation is promoted by Nrf2 secondary to its role in the reduction in intracellular ROS [104,105]. In rats, it has been noted that variations in expression levels of Nrf2 are tied with the survival of NSCs [106], and in mice, a deficiency in Nrf2 expression or function resulting in the reduction in NSC proliferation could be improved with administration of Nrf2 [107]. However, following an ischemic cerebrovascular accident, neurogenesis by NSCs is insufficient for complete recovery due to relatively low yields of new cells [108,109]. Thus, a possible therapy is to administer exogenous NSCs obtained from neuroectoderm in fetuses or from the neurogenic sites in adults, after they have been cultured and exposed to basic fibroblast growth factor and epidermal growth factor [110,111]. Unfortunately, the host brain often rejects transplant NSCs [112], although it has been observed that delivery of human NSC-derived extracellular vesicles improves transplantation outcomes by stimulating nuclear localization of Nrf2, which subsequently decreases oxidative stress and stimulates axon elongation [113,114]. The following section investigates drug therapy that may be used concomitantly with NSC therapy to improve outcomes in patients suffering from cerebrovascular disease.

4.2. Targeting Nrf2 to Enhance Stem Cell Therapy

While stem cell therapy is undoubtedly a promising stroke treatment [115], its overall efficacy may benefit from adjunctive therapeutics that ameliorate inflammation, such as Nrf2 treatments (Figure 3). The following reviews a number of potential agents for co-administration with NSCs as a potential treatment modality.
Dimethyl fumarate (DMF), derived from fumaric acid, is a known treatment for psoriasis and multiple sclerosis due to its immunomodulatory effects [116]. Considerable research into its precise mechanism and interactions is ongoing, however, it is known to bind KEAP1, resulting in upregulation of antioxidant genes, including HO-1 [117], γ-glutamylcysteine synthetase, glutathione synthetase, and glutathione reductase [118], following nuclear translocation of Nrf2 [119]. A study in an MCAO model on Sprague-Dawley rats demonstrated significantly reduced infarct volumes in the group treated with DMF compared to control counterparts [120]. As for its use in stem cell therapy, DMF delivered to mouse and rat models following a treatment with hydrogen peroxide to trigger oxidative stress resulted in increased NSC survival. Additionally, DMF enhanced the survival of motor neurons and reduced both the production of ROS and the rates of apoptosis following hydrogen peroxide treatment [103].
Berberine (BBR), a natural alkaloid isolated from medicinal herbs, is a known treatment for diarrhea that has recently become a multitarget drug for neurological disorders owed in part to antioxidant and anti-inflammatory properties [121]. The interaction of BBR with the Nrf2 pathway was demonstrated through siRNA inactivation of Nrf2 signaling, which diminished antioxidant effects following BBR’s administration [122,123]. In MCAO-induced stroke mice, BBR activated peroxisome proliferator-activated receptor-δ (PPARδ), which in turn upregulated Nrf2, along with other known antioxidants, to scavenge ROS in NSCs, thereby promoting their proliferation and improving recovery [124].
Carbon monoxide (CO) has recently been shown to have a significant role as an antioxidant, stimulating the bilirubin/biliverdin redox cycling system and the pentose-phosphate pathway to produce NADPH, a reducing equivalent [125]. CO plays a protective role against iron overload, making it an attractive treatment for administration with transplanted NSCs following hemorrhagic stroke. This has been previously demonstrated in mouse models through modulation of the crosstalk between Nrf2 and NF-κB, in which CO inhibited NF-κB while inducing ROS scavenging through Nrf2 activation [126].
Sulforaphane (SFN) is an isothiocyanate found in a number of cruciferous vegetables with known antioxidant, anti-inflammatory, and anti-tumor activity [127]. Specifically, the antioxidant properties of SFN are derived from well-documented activation of the Nrf2 pathway [128,129,130] including downstream activation of GSTs, NQO-1, and HO-1 [28]. MCAO-induced Sprague-Dawley rats treated with SFN demonstrated heightened Nrf2 levels within the cerebral microvasculature after 24 h, resulting in reduction in blood–brain barrier (BBB) disruption and lesion progression [131]. Later experimentation into Sprague-Dawley rats revealed that SFN treatment at concentrations less than 10 µM stimulated NSC differentiation and proliferation [132]. Thus, SFN shows significant promise in stroke treatment with and without concurrent stem cell treatment.
Doxycycline (DOX) is a tetracycline derivative known to be well-tolerated and demonstrate broad-spectrum efficacy as an antibiotic that also exhibits antioxidant and anti-apoptotic activity [133,134]. Antioxidant activity has been determined using electron paramagnetic resonance to confirm scavenging of superoxide by DOX and has been related to modulation of the Nrf2 pathway [134], while its anti-apoptotic role in neuroprotection has been attributed to inhibition of microglial activation [135]. Additionally, DOX has been tied to the upregulation of tight junctions with subsequent inhibition of the matrix metalloproteinases (MMP) MMP-2 and MMP-9 and protein kinase C delta for protection of the BBB [136]. This neuroprotective role explains the historic success of DOX in reducing damage from ischemic stroke modeled in rabbits and MCAO-induced rats [137,138]. As an aspect of stem cell treatment, DOX has been documented to upregulate Nrf2 mRNA and protein levels in NSCs for an overall increase in cell survival in preconditioned fetal rat brains [139,140,141].
Minocycline, a semisynthetic tetracycline, has also been associated with significant neuroprotection through a number of properties including antioxidation, anti-apoptosis, anti-inflammation, and vascular protection [142,143]. A systematic review and meta-analysis by Malhotra et al. of randomized control trials of minocycline application in stroke patients indicated success in reducing damage attributed to acute ischemic stroke and a need for further research into a potential role in acute intracerebral hemorrhage [143]. In a number of experimental studies on rat models, NSCs pretreated with minocycline resulted in a noted upregulation of Nrf2 and downstream genes such as NQO1 and HO-1, which correlated with increased NSC viability and proliferation [112,144]. Further, minocycline resulted in the NSCs releasing neuroprotective factors such as brain-derived neurotrophic factor, nerve growth factor, glial cell-derived neurotrophic factor, and vascular endothelial growth factor [112]. Outcomes of neuroinflammation were mitigated by minocycline, inhibiting microglial activation and counteracting proinflammatory cytokines that normally inhibit neurogenesis, restoring the neurogenic and oligodendrogliogenic potential of NSCs [145,146,147]. Minocycline has also been implicated in the success of bone marrow-derived mesenchymal stem cell transplantation in animal stroke models in which the combination of therapies results in reduced infarct size and enhanced recovery [148,149,150].
Tert-butylhydroquinone (tBHQ), a synthetic phenol, is a common food additive known to have low toxicity and antioxidant properties. Pulse-chase assay has demonstrated a stabilizing effect of tBHQ on Nrf2 in NSCs, making it an attractive molecule for research into neuroprotection from oxidative stress [151]. tBHQ has been studied extensively in cerebral ischemia, sufficiently activating Nrf2 in rat models to reduce cortical damage and sensorimotor deficits following ischemia-reperfusion [33]. Further studies into cerebral ischemia in mice reveal that activation of Nrf2 by tBHQ also enhances angiogenesis and astrocyte activation [152]. For intracerebral hemorrhages, tBHQ treatment reduced oxidative brain damage, microglial activation, and release of proinflammatory cytokines to improve outcomes in CD1 mice [153]. Models of subarachnoid hemorrhage treated with tBHQ highlighted the upregulation of KEAP1, Nrf2, HO-1 and NQO1to reduce early brain injury and cognitive dysfunction [154]. tBHQ has recently been implicated in the generation of multipotent stem cells from normal brain pericytes placed under oxidative stress for a key role in regeneration of cerebral vasculature, suggesting the potential for future research into tBHQ and in vivo generation of stem cells [155].
Resveratrol, a natural stilbene, is produced by a number of fruits and vegetables with known antioxidant and antitumor activity [156]. In response to vascular injury, resveratrol has been indicated to mobilize endothelial cells and increase endothelial progenitor cell proliferation [157,158]. A recent meta-analysis in rodents has demonstrated an overall neuroprotective effect of resveratrol in ischemic stroke models [159]. On a molecular level, this effect is achieved through upregulation of Nrf2 and downstream antioxidant genes, including HO-1 [160], that enhance neurogenesis and increase viability of NSCs by reducing apoptosis [140,161].
Ginseng, along with its bioactive ingredients ginsenosides, is a staple of herbal medicine predominantly utilized in Asia. Rodent and human models have highlighted a number of important pharmaceutical properties that include antioxidant, anti-inflammatory, and immunomodulatory effects [162]. These effects are achieved through inhibition of the proinflammatory NF-κB pathway and subsequent upregulation of Nrf2 [163]. Panax notoginseng was also shown to reduce the expression of Nogo-A and Nogo Receptor, molecules that typically inhibit axonal regeneration after ischemic injury [162]. As it pertains to stem cell therapy, ginseng has been implicated in regeneration during inflammatory diseases such as stroke, inducing neurogenesis and angiogenesis that has been shown to preserve cognitive function [164]. For instance, pretreatment of MCAO-induced Wistar rats with Ginseng total saponins resulted in improved neurological scores after a two-week recovery period, highlighting Ginseng’s inductive effect on NSCs to promote regeneration [165].
Theaflavin (TFA), a polyphenolic compound, is a pigment contained in black tea known to exert antioxidant [166] and anti-inflammatory effects [167], among a number of other health benefits [168]. TFA’s mechanism of action revolves around the nuclear translocation of Nrf2, activating the Nrf2/ARE pathway to upregulate HO-1 [169]. Li et al. showed that NSCs treated with TFA can result in a decreased infarct volume and improved cognitive function through Nrf2′s protective effects against oxidative stress [170]. Additionally promising for stem cell viability is a reported dose-response relationship between TFA and B-cell lymphoma 2 (Bcl-2) overexpression to downregulate the mitochondrial apoptotic pathway in ischemic stroke models [171].
Curcumin is a natural polyphenol that is isolated from turmeric. This compound has low toxicity and owes both its antioxidant and anti-apoptotic properties to activation of the Nrf2 pathway by modulating the demethylation of CpG islands to promote gene transcription [172], which also leads to enhanced activation of HO-1 and NQO1 [173,174]. Curcumin has been shown to exert a neuroprotective effect in MCAO-induced rat models [175], and post-stroke injections reduced damage to hippocampal CA1 neurons [176]. In addition to oxidative stress, the far-reaching effects of curcumin in ischemia-reperfusion injury are involved in BBB disruption, platelet adhesion and aggregation, and immune functions [177]. Endogenously, curcumin regulates NSC proliferation, differentiation, and migration, with a number of potential applications to stroke and other neurological disorders [178]. In stem cell transplantation, embryonic stem cell exosomes loaded with curcumin resulted in significant cerebrovascular regeneration in mouse models [179].
With both treatment modalities alone showing promise in treating stroke, combining pharmaceuticals or natural products with stem cell therapy can efficiently target the Nrf2 pathway to promote broad effects on cell viability and proliferation.

4.3. Downstream Targets of the Nrf2 Pathway

In addition to the potential stroke therapies described above, there exist a number of compounds known to target key mediators related to the oxidative and inflammatory response that exist downstream of the Nrf2 pathway. The following will summarize a number of such treatment options while emphasizing the advantages of targeting Nrf2 upstream for broader therapeutic outcomes.
HO-1 is an intriguing target considering it has the greatest amount of antioxidant response elements on its promoter, when compared to other key downstream targets such as NQO1, GST, and γ-glutamylcysteine synthetase [180]. As discussed earlier, HO-1 metabolizes heme into various components including: ferrous iron (which promotes ferritin expression and thus iron sequestration, preventing iron-mediated cell injury) [181,182], biliverdin-IXa which is converted to bilirubin-IXa via biliverdin reductase (both components have antioxidant and anti-inflammatory properties) [183], and carbon monoxide (with anti-inflammatory, vasorelaxant, and anti-apoptotic properties) [184]. Animal studies have demonstrated protection against experimental stroke with various compounds activating HO-1 including some drugs previously mentioned: sulforaphane, Gingko biloba (EGb 761), curcumin, resveratrol, triterpenoids, and dimethyl fumarate [117,160,173,185,186,187,188,189]. Melatonin specifically increases expression of HO-1 downstream of Nrf2, resulting in improvement of motor skills and reduction in infarction size in experimental stroke models [190]. With regard to stem cells, melatonin has been shown to enhance neurogenesis in peri-infarct regions [191]. Other inducers of downstream HO-1 expression include oleanolic acid, hemopexin, and propofol, all of which have brought about improved outcomes in stroke models [192,193,194].
NQO1 is an important downstream enzyme involved in detoxifying reactive species. In rats who suffered stroke, intraperitoneal injection of 300 mg/kg curcumin was shown to induce expression of NQO1 [174], as well as reduce oxidative stress and improve binding of Nrf2 to ARE. Animal studies also showed increased concentrations of mRNA and protein product of NQO1 (as well as Nrf2, KEAP1, and HO-1) after treatment of tBHQ [154].
GSTs are enzymes that utilize the supply of reduced glutathione to detoxify xenobiotics. As previously noted, sulforaphane activates the Nrf2 pathway, inducing GSTs, as well as HO-1 and NQO-1 [28]. γ-glutamylcysteine synthetase is another enzyme important in the glutathione pathway, and its expression (along with glutathione reductase and glutathione synthetase) was seen to be induced in astrocytes and microglia in the setting of dimethyl fumarate administration [118].
NF-κB, a mediator demonstrating previously discussed antagonistic crosstalk with the Nrf2 pathway, is a key regulator of neuroinflammation that leads to the common complications of edema, hemorrhage, and necrosis following stroke [195,196,197]. For this reason, a significant body of research exists into post-stroke inhibition of the NF-κB pathway. Experimental models of cerebral ischemia have indicated that statins, such as simvastatin [198] and atorvastatin [199], substantially reduce NF-κB expression in brain tissue through transcriptional inhibition [200]. In exogenous stem cell therapy, simvastatin has been shown to aid bone-marrow-derived mesenchymal stem cell migration, while both statins have demonstrated activity in proliferation, viability, and differentiation of endogenous NSCs [201,202]. Naloxone has been shown to promote NF-κB inhibition through increased expression of the inhibitory protein, IκBα, and a reduction in NF-κB p65 nuclear translocation, resulting in decreased neuronal apoptosis and a dose-dependent decrease in infarction volume in animal models of cerebral ischemia [203]. An additional inhibitor of NF-κB nuclear translocation is artesunate, a drug prescribed for cerebral malaria that has recently found use in a mouse model of ischemic stroke, and ameliorated neuroinflammation by suppressing neutrophil infiltration and microglial activation [204]. In the context of stem cells, artesunate has been utilized to enhance NSC proliferation in the subventricular zone and peri-infarct cortex [205,206]. Aspirin, a common prophylactic in stroke prevention, may also have a role in treatment, having been shown to downregulate NF-κB-mediated endoplasmic reticulum stress in cerebrovascular endothelial cells following cerebral infarction in a mouse model [207]. Aspirin treatment alongside human umbilical cord matrix-derived stem cells improved learning and memory via the Morris water maze test [208]. Isosteviol sodium (STVNA), obtained from stevioside (a natural sweetener), exerts neuroprotective effects by interfering with the NF-κB signaling pathway [209]. In models of ischemia, STVNA has been implicated in inhibition of astrogliosis [210], modulation of microglia/macrophage polarization [211], and preservation of volume control in endothelial cells [212]. Flavonoids constitute a class of natural products with a significant role in the inhibition of microglial polarization and management of oxidative stress, with a number of compounds acting upon the NF-κB signaling pathway [213,214,215,216,217,218,219,220]. A number of the flavonoids have been implicated in the activity of stem cells, including hesperetin in NSC proliferation [221], icariin in neurogenesis and angiogenesis through release of BDNF and VEGF [222], and quercetin in enhancing stem cell viability and proliferation while reducing apoptosis [223].
While targeting of NF-κB and other downstream molecules mentioned above will certainly hold a central role in the future of stroke treatment, there exists limitations in this approach that are not yet fully appreciated. For instance, corticosteroids are commonly prescribed for controlling inflammation and have long been known as potent inhibitors of NF-κB through induction of its inhibitory protein, IκBα [224,225]. Despite this, a systematic review first published in 1997 and updated in 2011 consistently found the corticosteroids dexamethasone and betamethasone to have no effect on death or neurological/functional outcomes in acute ischemic stroke [226]. Treatment targeting Nrf2 upstream, as opposed to specific downstream targets, offers durability to treatment by enhancing a number of antioxidant, anti-inflammatory, and neuroprotective pathways should specific downstream approaches fail to achieve desired outcomes.
A comprehensive list of compounds and associated targets upstream or downstream within the Nrf2 pathway, along with the recognized effects in stroke treatment and stem cell therapy, is outlined in Table 1. Studies were completed in vivo (typically middle cerebral artery occlusion/reperfusion technique) or in vitro (typically oxygen-glucose deprivation/reoxygenation technique) in rodent models.

5. Conclusions and Future Directions

This review consolidates a plethora of information regarding oxidative damage and inflammation, both centrally and peripherally, following IS and HS. Furthermore, an elaborate discussion on the power of stem cells encourages ongoing studies to determine optimal safety and efficient uses of cell-based treatments for stroke. Considering both focuses, we argue that enhancing the antioxidant properties of the Nrf2 pathway concurrently with stem cell administration is a novel and imperative future research focus. Currently, the very few treatment options for IS and HS offer little reprieve from permanent functional impairments. Thus, greater initiatives are necessary to amplify treatment options for stroke. Furthermore, this review has revealed a number of potential directions for future research, including the oxidative processes that occur following cerebrovascular accident to enhance targeted therapies for improved stem cell viability and the role of Nrf2 in peripheral inflammation. Ultimately, we propose optimizing antioxidant and cell-based therapies by targeting the most upstream regulator of the antioxidant Nrf2 pathway, Nrf2 itself, to reduce secondary cell death, peripheral inflammation, and improve stem cell survival and effect.

Author Contributions

Conceptualization, J.G., G.L., M.M. and A.A.; investigation, J.G., G.L., M.M. and A.A.; supervision, C.V.B.; visualization, J.G., G.L., M.M. and A.A.; writing—original draft preparation, J.G., G.L., M.M. and A.A.; writing—review and editing, J.G., G.L., M.M., A.A. and C.V.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We acknowledge the work of the artists Brgfx, pikisuperstar, macrovector, and storyset, whose vector art was downloaded from freepik.com (accessed on 11 July 2022) and used within the figure design.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics—2022 Update: A Report from the American Heart Association. Circulation 2022, 145, e153–e639. [Google Scholar] [CrossRef] [PubMed]
  2. Andersen, K.K.; Olsen, T.S.; Dehlendorff, C.; Kammersgaard, L.P. Hemorrhagic and ischemic strokes compared: Stroke severity, mortality, and risk factors. Stroke 2009, 40, 2068–2072. [Google Scholar] [CrossRef] [Green Version]
  3. Marcet, P.; Santos, N.; Borlongan, C.V. When friend turns foe: Central and peripheral neuroinflammation in central nervous system injury. Neuroimmunol. Neuroinflamm. 2017, 4, 82–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mozaffarian, D.; Benjamin, E.J.; Go, A.S.; Arnett, D.K.; Blaha, M.J.; Cushman, M.; de Ferranti, S.; Despres, J.P.; Fullerton, H.J.; Howard, V.J.; et al. Heart disease and stroke statistics—2015 update: A report from the American Heart Association. Circulation 2015, 131, e29–e322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Acosta, S.A.; Tajiri, N.; de la Pena, I.; Bastawrous, M.; Sanberg, P.R.; Kaneko, Y.; Borlongan, C.V. Alpha-synuclein as a pathological link between chronic traumatic brain injury and Parkinson’s disease. J. Cell. Physiol. 2015, 230, 1024–1032. [Google Scholar] [CrossRef]
  6. Gumbinger, C.; Reuter, B.; Stock, C.; Sauer, T.; Wietholter, H.; Bruder, I.; Rode, S.; Kern, R.; Ringleb, P.; Hennerici, M.G.; et al. Time to treatment with recombinant tissue plasminogen activator and outcome of stroke in clinical practice: Retrospective analysis of hospital quality assurance data with comparison with results from randomised clinical trials. BMJ 2014, 348, g3429. [Google Scholar] [CrossRef] [Green Version]
  7. Kaesmacher, J.; Kaesmacher, M.; Maegerlein, C.; Zimmer, C.; Gersing, A.S.; Wunderlich, S.; Friedrich, B.; Boeckh-Behrens, T.; Kleine, J.F. Hemorrhagic Transformations after Thrombectomy: Risk Factors and Clinical Relevance. Cerebrovasc. Dis. 2017, 43, 294–304. [Google Scholar] [CrossRef]
  8. Nguyen, H.; Lee, J.Y.; Sanberg, P.R.; Napoli, E.; Borlongan, C.V. Eye Opener in Stroke. Stroke 2019, 50, 2197–2206. [Google Scholar] [CrossRef]
  9. Lees, K.R.; Bluhmki, E.; von Kummer, R.; Brott, T.G.; Toni, D.; Grotta, J.C.; Albers, G.W.; Kaste, M.; Marler, J.R.; Hamilton, S.A.; et al. Time to treatment with intravenous alteplase and outcome in stroke: An updated pooled analysis of ECASS, ATLANTIS, NINDS, and EPITHET trials. Lancet 2010, 375, 1695–1703. [Google Scholar] [CrossRef]
  10. Mao, L.; Li, P.; Zhu, W.; Cai, W.; Liu, Z.; Wang, Y.; Luo, W.; Stetler, R.A.; Leak, R.K.; Yu, W.; et al. Regulatory T cells ameliorate tissue plasminogen activator-induced brain haemorrhage after stroke. Brain 2017, 140, 1914–1931. [Google Scholar] [CrossRef]
  11. Cordonnier, C.; Demchuk, A.; Ziai, W.; Anderson, C.S. Intracerebral haemorrhage: Current approaches to acute management. Lancet 2018, 392, 1257–1268. [Google Scholar] [CrossRef]
  12. Imai, T.; Matsubara, H.; Hara, H. Potential therapeutic effects of Nrf2 activators on intracranial hemorrhage. J. Cereb. Blood Flow Metab. 2021, 41, 1483–1500. [Google Scholar] [CrossRef] [PubMed]
  13. Anthony, S.; Cabantan, D.; Monsour, M.; Borlongan, C.V. Neuroinflammation, Stem Cells, and Stroke. Stroke 2022, 53, 1460–1472. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, S.; Li, F.; Leak, R.K.; Chen, J.; Hu, X. Regulation of Neuroinflammation through Programed Death-1/Programed Death Ligand Signaling in Neurological Disorders. Front. Cell. Neurosci. 2014, 8, 271. [Google Scholar] [CrossRef] [Green Version]
  15. Stonesifer, C.; Corey, S.; Ghanekar, S.; Diamandis, Z.; Acosta, S.A.; Borlongan, C.V. Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog. Neurobiol. 2017, 158, 94–131. [Google Scholar] [CrossRef]
  16. Saeed, S.A.; Shad, K.F.; Saleem, T.; Javed, F.; Khan, M.U. Some new prospects in the understanding of the molecular basis of the pathogenesis of stroke. Exp. Brain Res. 2007, 182, 1. [Google Scholar] [CrossRef]
  17. Kobayashi, A.; Kang, M.I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell. Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef] [Green Version]
  18. Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011, 16, 123–140. [Google Scholar] [CrossRef]
  19. Huang, C.S.; Anderson, M.E.; Meister, A. Amino acid sequence and function of the light subunit of rat kidney gamma-glutamylcysteine synthetase. J. Biol. Chem. 1993, 268, 20578–20583. [Google Scholar] [CrossRef]
  20. Meister, A. Glutathione, metabolism and function via the gamma-glutamyl cycle. Life Sci. 1974, 15, 177–190. [Google Scholar] [CrossRef]
  21. Boyland, E.; Chasseaud, L.F. The role of glutathione and glutathione S-transferases in mercapturic acid biosynthesis. Adv. Enzymol. Relat. Areas Mol. Biol. 1969, 32, 173–219. [Google Scholar] [CrossRef] [PubMed]
  22. Forman, H.J.; Zhang, H.; Rinna, A. Glutathione: Overview of its protective roles, measurement, and biosynthesis. Mol. Aspects Med. 2009, 30, 1–12. [Google Scholar] [CrossRef] [Green Version]
  23. Mazur, A.; Fangman, M.; Ashouri, R.; Arcenas, A.; Dore, S. Nrf2 as a therapeutic target in ischemic stroke. Expert Opin. Ther. Targets 2021, 25, 163–166. [Google Scholar] [CrossRef] [PubMed]
  24. Leonardo, C.C.; Dore, S. Dietary flavonoids are neuroprotective through Nrf2-coordinated induction of endogenous cytoprotective proteins. Nutr. Neurosci. 2011, 14, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kim, E.; Cho, S. Microglia and Monocyte-Derived Macrophages in Stroke. Neurotherapeutics 2016, 13, 702–718. [Google Scholar] [CrossRef] [PubMed]
  26. Christopher, E.; Loan, J.J.M.; Samarasekera, N.; McDade, K.; Rose, J.; Barrington, J.; Hughes, J.; Smith, C.; Al-Shahi Salman, R. Nrf2 activation in the human brain after stroke due to supratentorial intracerebral haemorrhage: A case-control study. BMJ Neurol. Open 2022, 4, e000238. [Google Scholar] [CrossRef] [PubMed]
  27. Takagi, T.; Kitashoji, A.; Iwawaki, T.; Tsuruma, K.; Shimazawa, M.; Yoshimura, S.; Iwama, T.; Hara, H. Temporal activation of Nrf2 in the penumbra and Nrf2 activator-mediated neuroprotection in ischemia-reperfusion injury. Free Radic. Biol. Med. 2014, 72, 124–133. [Google Scholar] [CrossRef]
  28. Farina, M.; Vieira, L.E.; Buttari, B.; Profumo, E.; Saso, L. The Nrf2 Pathway in Ischemic Stroke: A Review. Molecules 2021, 26, 5001. [Google Scholar] [CrossRef]
  29. Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [Green Version]
  30. Morooka, H.; Hirotsune, N.; Wani, T.; Ohmoto, T. Histochemical demonstration of free radicals (H2O2) in ischemic brain edema and protective effects of human recombinant superoxide dismutase on ischemic neuronal damage. Acta Neurochir. Suppl. 1994, 60, 307–309. [Google Scholar] [CrossRef]
  31. Purdom-Dickinson, S.E.; Sheveleva, E.V.; Sun, H.; Chen, Q.M. Translational control of nrf2 protein in activation of antioxidant response by oxidants. Mol. Pharmacol. 2007, 72, 1074–1081. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, L.; Vollmer, M.K.; Fernandez, V.M.; Dweik, Y.; Kim, H.; Dore, S. Korean Red Ginseng Pretreatment Protects Against Long-Term Sensorimotor Deficits After Ischemic Stroke Likely through Nrf2. Front. Cell. Neurosci. 2018, 12, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Shih, A.Y.; Li, P.; Murphy, T.H. A small-molecule-inducible Nrf2-mediated antioxidant response provides effective prophylaxis against cerebral ischemia in vivo. J. Neurosci. 2005, 25, 10321–10335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wang, J.; Fields, J.; Zhao, C.; Langer, J.; Thimmulappa, R.K.; Kensler, T.W.; Yamamoto, M.; Biswal, S.; Dore, S. Role of Nrf2 in protection against intracerebral hemorrhage injury in mice. Free Radic. Biol. Med. 2007, 43, 408–414. [Google Scholar] [CrossRef] [Green Version]
  35. Zhao, X.; Sun, G.; Zhang, J.; Strong, R.; Dash, P.K.; Kan, Y.W.; Grotta, J.C.; Aronowski, J. Transcription factor Nrf2 protects the brain from damage produced by intracerebral hemorrhage. Stroke 2007, 38, 3280–3286. [Google Scholar] [CrossRef]
  36. Wagner, K.R.; Sharp, F.R.; Ardizzone, T.D.; Lu, A.; Clark, J.F. Heme and iron metabolism: Role in cerebral hemorrhage. J. Cereb. Blood Flow Metab. 2003, 23, 629–652. [Google Scholar] [CrossRef] [Green Version]
  37. Duan, X.; Wen, Z.; Shen, H.; Shen, M.; Chen, G. Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. Oxid. Med. Cell. Longev. 2016, 2016, 1203285. [Google Scholar] [CrossRef] [Green Version]
  38. Zhao, X.; Sun, G.; Ting, S.M.; Song, S.; Zhang, J.; Edwards, N.J.; Aronowski, J. Cleaning up after ICH: The role of Nrf2 in modulating microglia function and hematoma clearance. J. Neurochem. 2015, 133, 144–152. [Google Scholar] [CrossRef]
  39. Belcher, J.D.; Beckman, J.D.; Balla, G.; Balla, J.; Vercellotti, G. Heme degradation and vascular injury. Antioxid. Redox Signal. 2010, 12, 233–248. [Google Scholar] [CrossRef] [Green Version]
  40. Chen, M.; Regan, R.F. Time course of increased heme oxygenase activity and expression after experimental intracerebral hemorrhage: Correlation with oxidative injury. J. Neurochem. 2007, 103, 2015–2021. [Google Scholar] [CrossRef]
  41. Shang, H.; Yang, D.; Zhang, W.; Li, T.; Ren, X.; Wang, X.; Zhao, W. Time course of Keap1-Nrf2 pathway expression after experimental intracerebral haemorrhage: Correlation with brain oedema and neurological deficit. Free Radic. Res. 2013, 47, 368–375. [Google Scholar] [CrossRef] [PubMed]
  42. Chi, X.; Yao, W.; Xia, H.; Jin, Y.; Li, X.; Cai, J.; Hei, Z. Elevation of HO-1 Expression Mitigates Intestinal Ischemia-Reperfusion Injury and Restores Tight Junction Function in a Rat Liver Transplantation Model. Oxid. Med. Cell. Longev. 2015, 2015, 986075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tossetta, G.; Fantone, S.; Montanari, E.; Marzioni, D.; Goteri, G. Role of NRF2 in Ovarian Cancer. Antioxidants 2022, 11, 663. [Google Scholar] [CrossRef] [PubMed]
  44. Farkhondeh, T.; Pourbagher-Shahri, A.M.; Azimi-Nezhad, M.; Forouzanfar, F.; Brockmueller, A.; Ashrafizadeh, M.; Talebi, M.; Shakibaei, M.; Samarghandian, S. Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies. Molecules 2021, 26, 3157. [Google Scholar] [CrossRef]
  45. Iadecola, C.; Anrather, J. The immunology of stroke: From mechanisms to translation. Nat. Med. 2011, 17, 796–808. [Google Scholar] [CrossRef]
  46. Sekerdag, E.; Solaroglu, I.; Gursoy-Ozdemir, Y. Cell Death Mechanisms in Stroke and Novel Molecular and Cellular Treatment Options. Curr. Neuropharmacol. 2018, 16, 1396–1415. [Google Scholar] [CrossRef]
  47. Anrather, J.; Iadecola, C. Inflammation and Stroke: An Overview. Neurotherapeutics 2016, 13, 661–670. [Google Scholar] [CrossRef]
  48. Moskowitz, M.A.; Lo, E.H.; Iadecola, C. The science of stroke: Mechanisms in search of treatments. Neuron 2010, 67, 181–198. [Google Scholar] [CrossRef] [Green Version]
  49. Choi, D.W. Excitotoxicity: Still Hammering the Ischemic Brain in 2020. Front. Neurosci. 2020, 14, 579953. [Google Scholar] [CrossRef]
  50. Hernandez-Ontiveros, D.G.; Tajiri, N.; Acosta, S.; Giunta, B.; Tan, J.; Borlongan, C.V. Microglia activation as a biomarker for traumatic brain injury. Front. Neurol. 2013, 4, 30. [Google Scholar] [CrossRef] [Green Version]
  51. Nathan, C. Neutrophils and immunity: Challenges and opportunities. Nat. Rev. Immunol. 2006, 6, 173–182. [Google Scholar] [CrossRef] [PubMed]
  52. Ziai, W.C. Hematology and inflammatory signaling of intracerebral hemorrhage. Stroke 2013, 44, S74–S78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Imai, T.; Iwata, S.; Hirayama, T.; Nagasawa, H.; Nakamura, S.; Shimazawa, M.; Hara, H. Intracellular Fe2+ accumulation in endothelial cells and pericytes induces blood-brain barrier dysfunction in secondary brain injury after brain hemorrhage. Sci. Rep. 2019, 9, 6228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Meguro, T.; Chen, B.; Lancon, J.; Zhang, J.H. Oxyhemoglobin induces caspase-mediated cell death in cerebral endothelial cells. J. Neurochem. 2001, 77, 1128–1135. [Google Scholar] [CrossRef] [Green Version]
  55. Zille, M.; Karuppagounder, S.S.; Chen, Y.; Gough, P.J.; Bertin, J.; Finger, J.; Milner, T.A.; Jonas, E.A.; Ratan, R.R. Neuronal Death After Hemorrhagic Stroke In Vitro and In Vivo Shares Features of Ferroptosis and Necroptosis. Stroke 2017, 48, 1033–1043. [Google Scholar] [CrossRef] [Green Version]
  56. Wu, J.; Hua, Y.; Keep, R.F.; Nakamura, T.; Hoff, J.T.; Xi, G. Iron and iron-handling proteins in the brain after intracerebral hemorrhage. Stroke 2003, 34, 2964–2969. [Google Scholar] [CrossRef] [Green Version]
  57. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  58. Fang, Y.; Gao, S.; Wang, X.; Cao, Y.; Lu, J.; Chen, S.; Lenahan, C.; Zhang, J.H.; Shao, A.; Zhang, J. Programmed Cell Deaths and Potential Crosstalk With Blood-Brain Barrier Dysfunction After Hemorrhagic Stroke. Front. Cell. Neurosci. 2020, 14, 68. [Google Scholar] [CrossRef]
  59. Li, Q.; Weiland, A.; Chen, X.; Lan, X.; Han, X.; Durham, F.; Liu, X.; Wan, J.; Ziai, W.C.; Hanley, D.F.; et al. Ultrastructural Characteristics of Neuronal Death and White Matter Injury in Mouse Brain Tissues After Intracerebral Hemorrhage: Coexistence of Ferroptosis, Autophagy, and Necrosis. Front. Neurol. 2018, 9, 581. [Google Scholar] [CrossRef] [Green Version]
  60. Ma, S.; Zhao, H.; Ji, X.; Luo, Y. Peripheral to central: Organ interactions in stroke pathophysiology. Exp. Neurol. 2015, 272, 41–49. [Google Scholar] [CrossRef]
  61. Allan, S.M.; Rothwell, N.J. Inflammation in central nervous system injury. Philos. Trans. R. Soc. London Ser. B Biol. Sci. 2003, 358, 1669–1677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Smith, C.J.; Emsley, H.C.; Gavin, C.M.; Georgiou, R.F.; Vail, A.; Barberan, E.M.; del Zoppo, G.J.; Hallenbeck, J.M.; Rothwell, N.J.; Hopkins, S.J.; et al. Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome. BMC Neurol. 2004, 4, 2. [Google Scholar] [CrossRef] [PubMed]
  63. Emsley, H.C.; Smith, C.J.; Gavin, C.M.; Georgiou, R.F.; Vail, A.; Barberan, E.M.; Hallenbeck, J.M.; del Zoppo, G.J.; Rothwell, N.J.; Tyrrell, P.J.; et al. An early and sustained peripheral inflammatory response in acute ischaemic stroke: Relationships with infection and atherosclerosis. J. Neuroimmunol. 2003, 139, 93–101. [Google Scholar] [CrossRef]
  64. Ajmo, C.T., Jr.; Vernon, D.O.; Collier, L.; Hall, A.A.; Garbuzova-Davis, S.; Willing, A.; Pennypacker, K.R. The spleen contributes to stroke-induced neurodegeneration. J. Neurosci. Res. 2008, 86, 2227–2234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Planas, A.M.; Gómez-Choco, M.; Urra, X.; Gorina, R.; Caballero, M.; Chamorro, Á. Brain-derived antigens in lymphoid tissue of patients with acute stroke. J. Immunol. 2012, 188, 2156–2163. [Google Scholar] [CrossRef]
  66. Courties, G.; Herisson, F.; Sager, H.B.; Heidt, T.; Ye, Y.; Wei, Y.; Sun, Y.; Severe, N.; Dutta, P.; Scharff, J.; et al. Ischemic stroke activates hematopoietic bone marrow stem cells. Circ. Res. 2015, 116, 407–417. [Google Scholar] [CrossRef] [Green Version]
  67. Gendron, A.; Teitelbaum, J.; Cossette, C.; Nuara, S.; Dumont, M.; Geadah, D.; du Souich, P.; Kouassi, E. Temporal effects of left versus right middle cerebral artery occlusion on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain Res. 2002, 955, 85–97. [Google Scholar] [CrossRef]
  68. Offner, H.; Subramanian, S.; Parker, S.M.; Wang, C.; Afentoulis, M.E.; Lewis, A.; Vandenbark, A.A.; Hurn, P.D. Splenic atrophy in experimental stroke is accompanied by increased regulatory T cells and circulating macrophages. J. Immunol. 2006, 176, 6523–6531. [Google Scholar] [CrossRef] [Green Version]
  69. Seifert, H.A.; Hall, A.A.; Chapman, C.B.; Collier, L.A.; Willing, A.E.; Pennypacker, K.R. A transient decrease in spleen size following stroke corresponds to splenocyte release into systemic circulation. J. Neuroimmune Pharmacol. 2012, 7, 1017–1024. [Google Scholar] [CrossRef] [Green Version]
  70. Dotson, A.L.; Chen, Y.; Zhu, W.; Libal, N.; Alkayed, N.J.; Offner, H. Partial MHC Constructs Treat Thromboembolic Ischemic Stroke Characterized by Early Immune Expansion. Transl. Stroke Res. 2016, 7, 70–78. [Google Scholar] [CrossRef] [Green Version]
  71. Ajmo, C.T., Jr.; Collier, L.A.; Leonardo, C.C.; Hall, A.A.; Green, S.M.; Womble, T.A.; Cuevas, J.; Willing, A.E.; Pennypacker, K.R. Blockade of adrenoreceptors inhibits the splenic response to stroke. Exp. Neurol. 2009, 218, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chiu, N.L.; Kaiser, B.; Nguyen, Y.V.; Welbourne, S.; Lall, C.; Cramer, S.C. The Volume of the Spleen and Its Correlates after Acute Stroke. J. Stroke Cerebrovasc. Dis. 2016, 25, 2958–2961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Esposito, E.; Ahn, B.J.; Shi, J.; Nakamura, Y.; Park, J.H.; Mandeville, E.T.; Yu, Z.; Chan, S.J.; Desai, R.; Hayakawa, A.; et al. Brain-to-cervical lymph node signaling after stroke. Nat. Commun. 2019, 10, 5306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Miller, C.M.; Behrouz, R. Impact of Infection on Stroke Morbidity and Outcomes. Curr. Neurol. Neurosci. Rep. 2016, 16, 83. [Google Scholar] [CrossRef] [PubMed]
  75. Deng, Q.W.; Yang, H.; Yan, F.L.; Wang, H.; Xing, F.L.; Zuo, L.; Zhang, H.Q. Blocking Sympathetic Nervous System Reverses Partially Stroke-Induced Immunosuppression but does not Aggravate Functional Outcome After Experimental Stroke in Rats. Neurochem. Res. 2016, 41, 1877–1886. [Google Scholar] [CrossRef]
  76. Zuo, L.; Shi, L.; Yan, F. The reciprocal interaction of sympathetic nervous system and cAMP-PKA-NF-kB pathway in immune suppression after experimental stroke. Neurosci. Lett. 2016, 627, 205–210. [Google Scholar] [CrossRef]
  77. Liu, D.D.; Chu, S.F.; Chen, C.; Yang, P.F.; Chen, N.H.; He, X. Research progress in stroke-induced immunodepression syndrome (SIDS) and stroke-associated pneumonia (SAP). Neurochem. Int. 2018, 114, 42–54. [Google Scholar] [CrossRef]
  78. Martelli, D.; McKinley, M.J.; McAllen, R.M. The cholinergic anti-inflammatory pathway: A critical review. Auton. Neurosci. 2014, 182, 65–69. [Google Scholar] [CrossRef]
  79. Mracsko, E.; Liesz, A.; Karcher, S.; Zorn, M.; Bari, F.; Veltkamp, R. Differential effects of sympathetic nervous system and hypothalamic-pituitary-adrenal axis on systemic immune cells after severe experimental stroke. Brain Behav. Immun. 2014, 41, 200–209. [Google Scholar] [CrossRef]
  80. Wardyn, J.D.; Ponsford, A.H.; Sanderson, C.M. Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochem. Soc. Trans. 2015, 43, 621–626. [Google Scholar] [CrossRef] [Green Version]
  81. Kobayashi, E.H.; Suzuki, T.; Funayama, R.; Nagashima, T.; Hayashi, M.; Sekine, H.; Tanaka, N.; Moriguchi, T.; Motohashi, H.; Nakayama, K.; et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat. Commun. 2016, 7, 11624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Clausen, B.H.; Lundberg, L.; Yli-Karjanmaa, M.; Martin, N.A.; Svensson, M.; Alfsen, M.Z.; Flæng, S.B.; Lyngsø, K.; Boza-Serrano, A.; Nielsen, H.H.; et al. Fumarate decreases edema volume and improves functional outcome after experimental stroke. Exp. Neurol. 2017, 295, 144–154. [Google Scholar] [CrossRef]
  83. Khor, T.O.; Huang, M.T.; Kwon, K.H.; Chan, J.Y.; Reddy, B.S.; Kong, A.N. Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium-induced colitis. Cancer Res. 2006, 66, 11580–11584. [Google Scholar] [CrossRef] [Green Version]
  84. Vendrame, M.; Gemma, C.; Pennypacker, K.R.; Bickford, P.C.; Davis Sanberg, C.; Sanberg, P.R.; Willing, A.E. Cord blood rescues stroke-induced changes in splenocyte phenotype and function. Exp. Neurol. 2006, 199, 191–200. [Google Scholar] [CrossRef] [PubMed]
  85. Seifert, H.A.; Offner, H. The splenic response to stroke: From rodents to stroke subjects. J. Neuroinflamm. 2018, 15, 195. [Google Scholar] [CrossRef] [Green Version]
  86. Vendrame, M.; Cassady, J.; Newcomb, J.; Butler, T.; Pennypacker, K.R.; Zigova, T.; Sanberg, C.D.; Sanberg, P.R.; Willing, A.E. Infusion of human umbilical cord blood cells in a rat model of stroke dose-dependently rescues behavioral deficits and reduces infarct volume. Stroke 2004, 35, 2390–2395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yang, B.; Hamilton, J.A.; Valenzuela, K.S.; Bogaerts, A.; Xi, X.; Aronowski, J.; Mays, R.W.; Savitz, S.I. Multipotent Adult Progenitor Cells Enhance Recovery After Stroke by Modulating the Immune Response from the Spleen. Stem Cells 2017, 35, 1290–1302. [Google Scholar] [CrossRef] [Green Version]
  88. Xu, K.; Lee, J.Y.; Kaneko, Y.; Tuazon, J.P.; Vale, F.; van Loveren, H.; Borlongan, C.V. Human stem cells transplanted into the rat stroke brain migrate to the spleen via lymphatic and inflammation pathways. Haematologica 2019, 104, 1062–1073. [Google Scholar] [CrossRef]
  89. Kawabori, M.; Shichinohe, H.; Kuroda, S.; Houkin, K. Clinical Trials of Stem Cell Therapy for Cerebral Ischemic Stroke. Int. J. Mol. Sci. 2020, 21, 7380. [Google Scholar] [CrossRef]
  90. Suzuki, T.; Sato, Y.; Kushida, Y.; Tsuji, M.; Wakao, S.; Ueda, K.; Imai, K.; Iitani, Y.; Shimizu, S.; Hida, H.; et al. Intravenously delivered multilineage-differentiating stress enduring cells dampen excessive glutamate metabolism and microglial activation in experimental perinatal hypoxic ischemic encephalopathy. J. Cereb. Blood Flow Metab. 2021, 41, 1707–1720. [Google Scholar] [CrossRef]
  91. Borlongan, C.V.; Hadman, M.; Sanberg, C.D.; Sanberg, P.R. Central nervous system entry of peripherally injected umbilical cord blood cells is not required for neuroprotection in stroke. Stroke 2004, 35, 2385–2389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Doeppner, T.R.; El Aanbouri, M.; Dietz, G.P.; Weise, J.; Schwarting, S.; Bähr, M. Transplantation of TAT-Bcl-xL-transduced neural precursor cells: Long-term neuroprotection after stroke. Neurobiol. Dis. 2010, 40, 265–276. [Google Scholar] [CrossRef] [PubMed]
  93. Islam, R.; Drecun, S.; Varga, B.V.; Vonderwalde, I.; Siu, R.; Nagy, A.; Morshead, C.M. Transplantation of Human Cortically-Specified Neuroepithelial Progenitor Cells Leads to Improved Functional Outcomes in a Mouse Model of Stroke. Front. Cell. Neurosci. 2021, 15, 4290. [Google Scholar] [CrossRef]
  94. Acosta, S.A.; Tajiri, N.; Hoover, J.; Kaneko, Y.; Borlongan, C.V. Intravenous Bone Marrow Stem Cell Grafts Preferentially Migrate to Spleen and Abrogate Chronic Inflammation in Stroke. Stroke 2015, 46, 2616–2627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ghuman, H.; Perry, N.; Grice, L.; Gerwig, M.; Moorhead, J., Jr.; Nitzsche, F.; Poplawsky, A.J.; Ambrosio, F.; Modo, M. Physical therapy exerts sub-additive and suppressive effects on intracerebral neural stem cell implantation in a rat model of stroke. J. Cereb. Blood Flow Metab. 2022, 42, 826–843. [Google Scholar] [CrossRef] [PubMed]
  96. Lyu, Z.; Park, J.; Kim, K.M.; Jin, H.J.; Wu, H.; Rajadas, J.; Kim, D.H.; Steinberg, G.K.; Lee, W. A neurovascular-unit-on-a-chip for the evaluation of the restorative potential of stem cell therapies for ischaemic stroke. Nat. Biomed. Eng. 2021, 5, 847–863. [Google Scholar] [CrossRef]
  97. Park, Y.J.; Borlongan, C.V. Recent advances in cell therapy for stroke. J. Cereb. Blood Flow Metab. 2021, 41, 2797–2799. [Google Scholar] [CrossRef]
  98. Borlongan, M.C.; Farooq, J.; Sadanandan, N.; Wang, Z.J.; Cozene, B.; Lee, J.Y.; Steinberg, G.K. Stem Cells for Aging-Related Disorders. Stem Cell Rev. Rep. 2021, 17, 2054–2058. [Google Scholar] [CrossRef]
  99. Berlet, R.; Anthony, S.; Brooks, B.; Wang, Z.J.; Sadanandan, N.; Shear, A.; Cozene, B.; Gonzales-Portillo, B.; Parsons, B.; Salazar, F.E.; et al. Combination of Stem Cells and Rehabilitation Therapies for Ischemic Stroke. Biomolecules 2021, 11, 1316. [Google Scholar] [CrossRef]
  100. Prakash, R.; Fauzia, E.; Siddiqui, A.J.; Yadav, S.K.; Kumari, N.; Shams, M.T.; Naeem, A.; Praharaj, P.P.; Khan, M.A.; Bhutia, S.K.; et al. Oxidative Stress-induced Autophagy Compromises Stem Cell Viability. Stem Cells 2022, 40, 468–478. [Google Scholar] [CrossRef]
  101. Niklison-Chirou, M.V.; Agostini, M.; Amelio, I.; Melino, G. Regulation of Adult Neurogenesis in Mammalian Brain. Int. J. Mol. Sci. 2020, 21, 4869. [Google Scholar] [CrossRef] [PubMed]
  102. Martino, G.; Pluchino, S. The therapeutic potential of neural stem cells. Nat. Rev. Neurosci. 2006, 7, 395–406. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, Q.; Chuikov, S.; Taitano, S.; Wu, Q.; Rastogi, A.; Tuck, S.J.; Corey, J.M.; Lundy, S.K.; Mao-Draayer, Y. Dimethyl Fumarate Protects Neural Stem/Progenitor Cells and Neurons from Oxidative Damage through Nrf2-ERK1/2 MAPK Pathway. Int. J. Mol. Sci. 2015, 16, 13885–13907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Noble, M.; Smith, J.; Power, J.; Mayer-Pröschel, M. Redox state as a central modulator of precursor cell function. Ann. N. Y. Acad. Sci. 2003, 991, 251–271. [Google Scholar] [CrossRef] [PubMed]
  105. Rafalski, V.A.; Brunet, A. Energy metabolism in adult neural stem cell fate. Prog. Neurobiol. 2011, 93, 182–203. [Google Scholar] [CrossRef] [PubMed]
  106. Corenblum, M.J.; Ray, S.; Remley, Q.W.; Long, M.; Harder, B.; Zhang, D.D.; Barnes, C.A.; Madhavan, L. Reduced Nrf2 expression mediates the decline in neural stem cell function during a critical middle-age period. Aging Cell 2016, 15, 725–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Robledinos-Antón, N.; Rojo, A.I.; Ferreiro, E.; Núñez, Á.; Krause, K.H.; Jaquet, V.; Cuadrado, A. Transcription factor NRF2 controls the fate of neural stem cells in the subgranular zone of the hippocampus. Redox Biol. 2017, 13, 393–401. [Google Scholar] [CrossRef]
  108. Kernie, S.G.; Parent, J.M. Forebrain neurogenesis after focal Ischemic and traumatic brain injury. Neurobiol. Dis. 2010, 37, 267–274. [Google Scholar] [CrossRef] [Green Version]
  109. Kokaia, Z.; Lindvall, O. Neurogenesis after ischaemic brain insults. Curr. Opin. Neurobiol. 2003, 13, 127–132. [Google Scholar] [CrossRef]
  110. Boese, A.C.; Le, Q.E.; Pham, D.; Hamblin, M.H.; Lee, J.P. Neural stem cell therapy for subacute and chronic ischemic stroke. Stem Cell Res. Ther. 2018, 9, 154. [Google Scholar] [CrossRef]
  111. Lee, J.P.; McKercher, S.; Muller, F.J.; Snyder, E.Y. Neural stem cell transplantation in mouse brain. Curr. Protoc. Neurosci. 2008, 42, 3.10.1–3.10.23. [Google Scholar] [CrossRef] [PubMed]
  112. Sakata, H.; Niizuma, K.; Yoshioka, H.; Kim, G.S.; Jung, J.E.; Katsu, M.; Narasimhan, P.; Maier, C.M.; Nishiyama, Y.; Chan, P.H. Minocycline-preconditioned neural stem cells enhance neuroprotection after ischemic stroke in rats. J. Neurosci. 2012, 32, 3462–3473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kahroba, H.; Davatgaran-Taghipour, Y. Exosomal Nrf2: From anti-oxidant and anti-inflammation response to wound healing and tissue regeneration in aged-related diseases. Biochimie 2020, 171–172, 103–109. [Google Scholar] [CrossRef]
  114. Liu, Q.; Tan, Y.; Qu, T.; Zhang, J.; Duan, X.; Xu, H.; Mu, Y.; Ma, H.; Wang, F. Therapeutic mechanism of human neural stem cell-derived extracellular vesicles against hypoxia-reperfusion injury in vitro. Life Sci. 2020, 254, 117772. [Google Scholar] [CrossRef]
  115. Singh, M.; Pandey, P.K.; Bhasin, A.; Padma, M.V.; Mohanty, S. Application of Stem Cells in Stroke: A Multifactorial Approach. Front. Neurosci. 2020, 14, 473. [Google Scholar] [CrossRef]
  116. Bomprezzi, R. Dimethyl fumarate in the treatment of relapsing–remitting multiple sclerosis: An overview. Ther. Adv. Neurol. Disord. 2015, 8, 20–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Yao, Y.; Miao, W.; Liu, Z.; Han, W.; Shi, K.; Shen, Y.; Li, H.; Liu, Q.; Fu, Y.; Huang, D.; et al. Dimethyl Fumarate and Monomethyl Fumarate Promote Post-Ischemic Recovery in Mice. Transl. Stroke Res. 2016, 7, 535–547. [Google Scholar] [CrossRef] [Green Version]
  118. Lin, S.X.; Lisi, L.; Dello Russo, C.; Polak, P.E.; Sharp, A.; Weinberg, G.; Kalinin, S.; Feinstein, D.L. The anti-inflammatory effects of dimethyl fumarate in astrocytes involve glutathione and haem oxygenase-1. ASN Neuro 2011, 3, AN20100033. [Google Scholar] [CrossRef]
  119. Chen, H.; Assmann, J.C.; Krenz, A.; Rahman, M.; Grimm, M.; Karsten, C.M.; Köhl, J.; Offermanns, S.; Wettschureck, N.; Schwaninger, M. Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective effect in EAE. J. Clin. Investig. 2014, 124, 2188–2192. [Google Scholar] [CrossRef]
  120. Safari, A.; Badeli-Sarkala, H.; Namavar, M.R.; Kargar-Abarghouei, E.; Anssari, N.; Izadi, S.; Borhani-Haghighi, A. Neuroprotective effect of dimethyl fumarate in stroke: The role of nuclear factor erythroid 2-related factor 2. Iran. J. Neurol. 2019, 18, 108–113. [Google Scholar] [CrossRef]
  121. Shou, J.W.; Shaw, P.C. Therapeutic Efficacies of Berberine against Neurological Disorders: An Update of Pharmacological Effects and Mechanisms. Cells 2022, 11, 796. [Google Scholar] [CrossRef] [PubMed]
  122. Choi, Y.H. Berberine Hydrochloride Protects C2C12 Myoblast Cells Against Oxidative Stress-Induced Damage via Induction of Nrf-2-Mediated HO-1 Expression. Drug Dev. Res. 2016, 77, 310–318. [Google Scholar] [CrossRef] [PubMed]
  123. Ashrafizadeh, M.; Fekri, H.S.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Therapeutic and biological activities of berberine: The involvement of Nrf2 signaling pathway. J. Cell. Biochem. 2020, 121, 1575–1585. [Google Scholar] [CrossRef] [PubMed]
  124. Shou, J.W.; Li, X.X.; Tang, Y.S.; Lim-Ho Kong, B.; Wu, H.Y.; Xiao, M.J.; Cheung, C.K.; Shaw, P.C. Novel mechanistic insight on the neuroprotective effect of berberine: The role of PPARδ for antioxidant action. Free Radic. Biol. Med. 2022, 181, 62–71. [Google Scholar] [CrossRef]
  125. Stucki, D.; Steinhausen, J.; Westhoff, P.; Krahl, H.; Brilhaus, D.; Massenberg, A.; Weber, A.P.M.; Reichert, A.S.; Brenneisen, P.; Stahl, W. Endogenous Carbon Monoxide Signaling Modulates Mitochondrial Function and Intracellular Glucose Utilization: Impact of the Heme Oxygenase Substrate Hemin. Antioxidants 2020, 9, 652. [Google Scholar] [CrossRef]
  126. Zhengxing, X.; Aiying, H.; Zongqiang, Z.; Zengli, M. Carbon Monoxide Protects Neural Stem Cells Against Iron Overload by Modulating the Crosstalk Between Nrf2 and NF-κB Signaling. Neurochem. Res. 2022, 47, 1383–1394. [Google Scholar] [CrossRef]
  127. Sun, Y.; Yang, T.; Mao, L.; Zhang, F. Sulforaphane Protects against Brain Diseases: Roles of Cytoprotective Enzymes. Austin J. Cerebrovasc. Dis. Stroke 2017, 4, 1054. [Google Scholar] [CrossRef] [Green Version]
  128. Sun, Y.; Yang, T.; Leak, R.K.; Chen, J.; Zhang, F. Preventive and Protective Roles of Dietary Nrf2 Activators Against Central Nervous System Diseases. CNS Neurol. Disord. Drug Targets 2017, 16, 326–338. [Google Scholar] [CrossRef] [Green Version]
  129. Ping, Z.; Liu, W.; Kang, Z.; Cai, J.; Wang, Q.; Cheng, N.; Wang, S.; Wang, S.; Zhang, J.H.; Sun, X. Sulforaphane protects brains against hypoxic-ischemic injury through induction of Nrf2-dependent phase 2 enzyme. Brain Res. 2010, 1343, 178–185. [Google Scholar] [CrossRef]
  130. Bai, Y.; Wang, X.; Zhao, S.; Ma, C.; Cui, J.; Zheng, Y. Sulforaphane Protects against Cardiovascular Disease via Nrf2 Activation. Oxid. Med. Cell. Longev. 2015, 2015, 407580. [Google Scholar] [CrossRef] [Green Version]
  131. Alfieri, A.; Srivastava, S.; Siow, R.C.M.; Cash, D.; Modo, M.; Duchen, M.R.; Fraser, P.A.; Williams, S.C.R.; Mann, G.E. Sulforaphane preconditioning of the Nrf2/HO-1 defense pathway protects the cerebral vasculature against blood–brain barrier disruption and neurological deficits in stroke. Free Radic. Biol. Med. 2013, 65, 1012–1022. [Google Scholar] [CrossRef] [PubMed]
  132. Han, Z.; Xu, Q.; Li, C.; Zhao, H. Effects of sulforaphane on neural stem cell proliferation and differentiation. Genesis 2017, 55, e23022. [Google Scholar] [CrossRef] [PubMed]
  133. Griffin, M.O.; Fricovsky, E.; Ceballos, G.; Villarreal, F. Tetracyclines: A pleitropic family of compounds with promising therapeutic properties. Review of the literature. Am. J. Physiol. Cell Physiol. 2010, 299, C539–C548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Clemens, D.L.; Duryee, M.J.; Sarmiento, C.; Chiou, A.; McGowan, J.D.; Hunter, C.D.; Schlichte, S.L.; Tian, J.; Klassen, L.W.; O’Dell, J.R.; et al. Novel Antioxidant Properties of Doxycycline. Int. J. Mol. Sci. 2018, 19, 4078. [Google Scholar] [CrossRef] [Green Version]
  135. Yrjänheikki, J.; Keinänen, R.; Pellikka, M.; Hökfelt, T.; Koistinaho, J. Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc. Natl. Acad. Sci. USA 1998, 95, 15769–15774. [Google Scholar] [CrossRef] [Green Version]
  136. Wang, Z.; Xue, Y.; Jiao, H.; Liu, Y.; Wang, P. Doxycycline-mediated protective effect against focal cerebral ischemia-reperfusion injury through the modulation of tight junctions and PKCδ signaling in rats. J. Mol. Neurosci. 2012, 47, 89–100. [Google Scholar] [CrossRef]
  137. Clark, W.M.; Calcagno, F.A.; Gabler, W.L.; Smith, J.R.; Coull, B.M. Reduction of central nervous system reperfusion injury in rabbits using doxycycline treatment. Stroke 1994, 25, 1411–1415; discussion 1416. [Google Scholar] [CrossRef] [Green Version]
  138. Clark, W.M.; Lessov, N.; Lauten, J.D.; Hazel, K. Doxycycline treatment reduces ischemic brain damage in transient middle cerebral artery occlusion in the rat. J. Mol. Neurosci. 1997, 9, 103–108. [Google Scholar] [CrossRef]
  139. Malik, Y.S.; Sheikh, M.A.; Zhu, X. Doxycycline can stimulate cytoprotection in neural stem cells with oxygen-glucose deprivation-reoxygenation injury: A potential approach to enhance effectiveness of cell transplantation therapy. Biochem. Biophys. Res. Commun. 2013, 432, 355–358. [Google Scholar] [CrossRef]
  140. Kahroba, H.; Ramezani, B.; Maadi, H.; Sadeghi, M.R.; Jaberie, H.; Ramezani, F. The role of Nrf2 in neural stem/progenitors cells: From maintaining stemness and self-renewal to promoting differentiation capability and facilitating therapeutic application in neurodegenerative disease. Ageing Res. Rev. 2021, 65, 101211. [Google Scholar] [CrossRef]
  141. Othman, F.A.; Tan, S.C. Preconditioning Strategies to Enhance Neural Stem Cell-Based Therapy for Ischemic Stroke. Brain Sci. 2020, 10, 893. [Google Scholar] [CrossRef] [PubMed]
  142. Yrjänheikki, J.; Tikka, T.; Keinänen, R.; Goldsteins, G.; Chan, P.H.; Koistinaho, J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc. Natl. Acad. Sci. USA 1999, 96, 13496–13500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Malhotra, K.; Chang, J.J.; Khunger, A.; Blacker, D.; Switzer, J.A.; Goyal, N.; Hernandez, A.V.; Pasupuleti, V.; Alexandrov, A.V.; Tsivgoulis, G. Minocycline for acute stroke treatment: A systematic review and meta-analysis of randomized clinical trials. J. Neurol. 2018, 265, 1871–1879. [Google Scholar] [CrossRef] [PubMed]
  144. Rueger, M.A.; Muesken, S.; Walberer, M.; Jantzen, S.U.; Schnakenburg, K.; Backes, H.; Graf, R.; Neumaier, B.; Hoehn, M.; Fink, G.R.; et al. Effects of minocycline on endogenous neural stem cells after experimental stroke. Neuroscience 2012, 215, 174–183. [Google Scholar] [CrossRef] [PubMed]
  145. Yong, V.W.; Wells, J.; Giuliani, F.; Casha, S.; Power, C.; Metz, L.M. The promise of minocycline in neurology. Lancet Neurol. 2004, 3, 744–751. [Google Scholar] [CrossRef]
  146. Vay, S.U.; Blaschke, S.; Klein, R.; Fink, G.R.; Schroeter, M.; Rueger, M.A. Minocycline mitigates the gliogenic effects of proinflammatory cytokines on neural stem cells. J. Neurosci. Res. 2016, 94, 149–160. [Google Scholar] [CrossRef]
  147. Liu, Z.; Fan, Y.; Won, S.J.; Neumann, M.; Hu, D.; Zhou, L.; Weinstein, P.R.; Liu, J. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke 2007, 38, 146–152. [Google Scholar] [CrossRef] [Green Version]
  148. Tsai, M.J.; Liou, D.Y.; Chu, Y.C.; Chen, Y.; Huang, M.C.; Huang, W.C.; Cheng, H.; Tsai, S.K.; Huang, S.S. Minocycline exhibits synergism with conditioned medium of bone marrow mesenchymal stem cells against ischemic stroke. J. Tissue Eng. Regen. Med. 2021, 15, 279–292. [Google Scholar] [CrossRef]
  149. Franco, E.C.; Cardoso, M.M.; Gouvêia, A.; Pereira, A.; Gomes-Leal, W. Modulation of microglial activation enhances neuroprotection and functional recovery derived from bone marrow mononuclear cell transplantation after cortical ischemia. Neurosci. Res. 2012, 73, 122–132. [Google Scholar] [CrossRef]
  150. Cho, D.Y.; Jeun, S.S. Combination therapy of human bone marrow-derived mesenchymal stem cells and minocycline improves neuronal function in a rat middle cerebral artery occlusion model. Stem Cell Res. Ther. 2018, 9, 309. [Google Scholar] [CrossRef]
  151. Li, J.; Johnson, D.; Calkins, M.; Wright, L.; Svendsen, C.; Johnson, J. Stabilization of Nrf2 by tBHQ confers protection against oxidative stress-induced cell death in human neural stem cells. Toxicol. Sci. 2005, 83, 313–328. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, Y.; Zhang, X.; Yang, Y.; Zhang, L.; Cui, L.; Zhang, C.; Chen, R.; Xie, Y.; He, J.; He, W. Tert-butylhydroquinone enhanced angiogenesis and astrocyte activation by activating nuclear factor-E2-related factor 2/heme oxygenase-1 after focal cerebral ischemia in mice. Microvasc. Res. 2019, 126, 103891. [Google Scholar] [CrossRef] [PubMed]
  153. Sukumari-Ramesh, S.; Alleyne, C.H., Jr. Post-Injury Administration of Tert-butylhydroquinone Attenuates Acute Neurological Injury After Intracerebral Hemorrhage in Mice. J. Mol. Neurosci. 2016, 58, 525–531. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, Z.; Ji, C.; Wu, L.; Qiu, J.; Li, Q.; Shao, Z.; Chen, G. Tert-butylhydroquinone alleviates early brain injury and cognitive dysfunction after experimental subarachnoid hemorrhage: Role of Keap1/Nrf2/ARE pathway. PLoS ONE 2014, 9, e97685. [Google Scholar] [CrossRef] [Green Version]
  155. Sakuma, R.; Kobayashi, M.; Kobashi, R.; Onishi, M.; Maeda, M.; Kataoka, Y.; Imaoka, S. Brain pericytes acquire stemness via the Nrf2-dependent antioxidant system. Stem Cells 2022, sxac024. [Google Scholar] [CrossRef]
  156. Kalantari, H.; Das, D.K. Physiological effects of resveratrol. Biofactors 2010, 36, 401–406. [Google Scholar] [CrossRef]
  157. Gu, J.; Wang, C.Q.; Fan, H.H.; Ding, H.Y.; Xie, X.L.; Xu, Y.M.; Wang, B.Y.; Huang, D.J. Effects of resveratrol on endothelial progenitor cells and their contributions to reendothelialization in intima-injured rats. J. Cardiovasc. Pharmacol. 2006, 47, 711–721. [Google Scholar] [CrossRef]
  158. Xia, L.; Wang, X.X.; Hu, X.S.; Guo, X.G.; Shang, Y.P.; Chen, H.J.; Zeng, C.L.; Zhang, F.R.; Chen, J.Z. Resveratrol reduces endothelial progenitor cells senescence through augmentation of telomerase activity by Akt-dependent mechanisms. Br. J. Pharmacol. 2008, 155, 387–394. [Google Scholar] [CrossRef] [Green Version]
  159. Liu, J.; He, J.; Huang, Y.; Hu, Z. Resveratrol has an Overall Neuroprotective Role in Ischemic Stroke: A Meta-Analysis in Rodents. Front. Pharmacol. 2021, 12, 795409. [Google Scholar] [CrossRef]
  160. Bastianetto, S.; Ménard, C.; Quirion, R. Neuroprotective action of resveratrol. Biochim. Biophys. Acta 2015, 1852, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
  161. Shen, C.; Cheng, W.; Yu, P.; Wang, L.; Zhou, L.; Zeng, L.; Yang, Q. Resveratrol pretreatment attenuates injury and promotes proliferation of neural stem cells following oxygen-glucose deprivation/reoxygenation by upregulating the expression of Nrf2, HO-1 and NQO1 in vitro. Mol. Med. Rep. 2016, 14, 3646–3654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Ong, W.Y.; Farooqui, T.; Koh, H.L.; Farooqui, A.A.; Ling, E.A. Protective effects of ginseng on neurological disorders. Front. Aging Neurosci. 2015, 7, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Yang, S.; Li, F.; Lu, S.; Ren, L.; Bian, S.; Liu, M.; Zhao, D.; Wang, S.; Wang, J. Ginseng root extract attenuates inflammation by inhibiting the MAPK/NF-κB signaling pathway and activating autophagy and p62-Nrf2-Keap1 signaling in vitro and in vivo. J. Ethnopharmacol. 2022, 283, 114739. [Google Scholar] [CrossRef] [PubMed]
  164. Kim, M.; Mok, H.; Yeo, W.S.; Ahn, J.H.; Choi, Y.K. Role of ginseng in the neurovascular unit of neuroinflammatory diseases focused on the blood-brain barrier. J. Ginseng Res. 2021, 45, 599–609. [Google Scholar] [CrossRef] [PubMed]
  165. Zheng, G.Q.; Cheng, W.; Wang, Y.; Wang, X.M.; Zhao, S.Z.; Zhou, Y.; Liu, S.J.; Wang, X.T. Ginseng total saponins enhance neurogenesis after focal cerebral ischemia. J. Ethnopharmacol. 2011, 133, 724–728. [Google Scholar] [CrossRef]
  166. Peluso, I.; Serafini, M. Antioxidants from black and green tea: From dietary modulation of oxidative stress to pharmacological mechanisms. Br. J. Pharmacol. 2017, 174, 1195–1208. [Google Scholar] [CrossRef] [Green Version]
  167. Wu, Y.; Jin, F.; Wang, Y.; Li, F.; Wang, L.; Wang, Q.; Ren, Z.; Wang, Y. In vitro and in vivo anti-inflammatory effects of theaflavin-3,3′-digallate on lipopolysaccharide-induced inflammation. Eur. J. Pharmacol. 2017, 794, 52–60. [Google Scholar] [CrossRef]
  168. Takemoto, M.; Takemoto, H. Synthesis of Theaflavins and Their Functions. Molecules 2018, 23, 918. [Google Scholar] [CrossRef] [Green Version]
  169. Li, Y.; Shi, J.; Sun, X.; Li, Y.; Duan, Y.; Yao, H. Theaflavic acid from black tea protects PC12 cells against ROS-mediated mitochondrial apoptosis induced by OGD/R via activating Nrf2/ARE signaling pathway. J. Nat. Med. 2020, 74, 238–246. [Google Scholar] [CrossRef]
  170. Li, R.; Li, X.; Wu, H.; Yang, Z.; Fei, L.; Zhu, J. Theaflavin attenuates cerebral ischemia/reperfusion injury by abolishing miRNA-128-3p-mediated Nrf2 inhibition and reducing oxidative stress. Mol. Med. Rep. 2019, 20, 4893–4904. [Google Scholar] [CrossRef] [Green Version]
  171. Mu, D.; Qin, H.; Jiao, M.; Hua, S.; Sun, T. Modeling the neuro-protection of theaflavic acid from black tea and its synergy with nimodipine via mitochondria apoptotic pathway. J. Zhejiang Univ. Sci. B 2021, 22, 123–135. [Google Scholar] [CrossRef] [PubMed]
  172. Ashrafizadeh, M.; Ahmadi, Z.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Curcumin Activates the Nrf2 Pathway and Induces Cellular Protection Against Oxidative Injury. Curr. Mol. Med. 2020, 20, 116–133. [Google Scholar] [CrossRef] [PubMed]
  173. Yang, C.; Zhang, X.; Fan, H.; Liu, Y. Curcumin upregulates transcription factor Nrf2, HO-1 expression and protects rat brains against focal ischemia. Brain Res. 2009, 1282, 133–141. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, J.; Li, Q.; Wang, X.; Yu, S.; Li, L.; Wu, X.; Chen, Y.; Zhao, J.; Zhao, Y. Neuroprotection by curcumin in ischemic brain injury involves the Akt/Nrf2 pathway. PLoS ONE 2013, 8, e59843. [Google Scholar] [CrossRef] [Green Version]
  175. Wang, Q.; Sun, A.Y.; Simonyi, A.; Jensen, M.D.; Shelat, P.B.; Rottinghaus, G.E.; MacDonald, R.S.; Miller, D.K.; Lubahn, D.E.; Weisman, G.A.; et al. Neuroprotective mechanisms of curcumin against cerebral ischemia-induced neuronal apoptosis and behavioral deficits. J. Neurosci. Res. 2005, 82, 138–148. [Google Scholar] [CrossRef]
  176. Chen, J.; Tang, X.Q.; Zhi, J.L.; Cui, Y.; Yu, H.M.; Tang, E.H.; Sun, S.N.; Feng, J.Q.; Chen, P.X. Curcumin protects PC12 cells against 1-methyl-4-phenylpyridinium ion-induced apoptosis by bcl-2-mitochondria-ROS-iNOS pathway. Apoptosis 2006, 11, 943–953. [Google Scholar] [CrossRef]
  177. Bavarsad, K.; Barreto, G.E.; Hadjzadeh, M.A.; Sahebkar, A. Protective Effects of Curcumin Against Ischemia-Reperfusion Injury in the Nervous System. Mol. Neurobiol. 2019, 56, 1391–1404. [Google Scholar] [CrossRef]
  178. Ma, W.; Xu, D.; Zhao, L.; Yuan, M.; Cui, Y.L.; Li, Y. Therapeutic role of curcumin in adult neurogenesis for management of psychiatric and neurological disorders: A scientometric study to an in-depth review. Crit. Rev. Food Sci. Nutr. 2022, 1–13. [Google Scholar] [CrossRef]
  179. Kalani, A.; Chaturvedi, P.; Kamat, P.K.; Maldonado, C.; Bauer, P.; Joshua, I.G.; Tyagi, S.C.; Tyagi, N. Curcumin-loaded embryonic stem cell exosomes restored neurovascular unit following ischemia-reperfusion injury. Int. J. Biochem. Cell Biol. 2016, 79, 360–369. [Google Scholar] [CrossRef] [Green Version]
  180. Chumboatong, W.; Thummayot, S.; Govitrapong, P.; Tocharus, C.; Jittiwat, J.; Tocharus, J. Neuroprotection of agomelatine against cerebral ischemia/reperfusion injury through an antiapoptotic pathway in rat. Neurochem. Int. 2017, 102, 114–122. [Google Scholar] [CrossRef]
  181. Balla, G.; Jacob, H.S.; Balla, J.; Rosenberg, M.; Nath, K.; Apple, F.; Eaton, J.W.; Vercellotti, G.M. Ferritin: A cytoprotective antioxidant strategem of endothelium. J. Biol. Chem. 1992, 267, 18148–18153. [Google Scholar] [CrossRef]
  182. Balla, J.; Vercellotti, G.M.; Jeney, V.; Yachie, A.; Varga, Z.; Jacob, H.S.; Eaton, J.W.; Balla, G. Heme, heme oxygenase, and ferritin: How the vascular endothelium survives (and dies) in an iron-rich environment. Antioxid. Redox Signal. 2007, 9, 2119–2137. [Google Scholar] [CrossRef] [PubMed]
  183. Fredenburgh, L.E.; Merz, A.A.; Cheng, S. Haeme oxygenase signalling pathway: Implications for cardiovascular disease. Eur. Heart J. 2015, 36, 1512–1518. [Google Scholar] [CrossRef] [Green Version]
  184. Motterlini, R.; Otterbein, L.E. The therapeutic potential of carbon monoxide. Nat. Rev. Drug Discov. 2010, 9, 728–743. [Google Scholar] [CrossRef] [PubMed]
  185. Ding, Y.; Chen, M.; Wang, M.; Li, Y.; Wen, A. Posttreatment with 11-Keto-β-Boswellic Acid Ameliorates Cerebral Ischemia-Reperfusion Injury: Nrf2/HO-1 Pathway as a Potential Mechanism. Mol. Neurobiol. 2015, 52, 1430–1439. [Google Scholar] [CrossRef]
  186. Shah, Z.A.; Li, R.C.; Ahmad, A.S.; Kensler, T.W.; Yamamoto, M.; Biswal, S.; Doré, S. The flavanol (-)-epicatechin prevents stroke damage through the Nrf2/HO1 pathway. J. Cereb. Blood Flow Metab. 2010, 30, 1951–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Shah, Z.A.; Nada, S.E.; Doré, S. Heme oxygenase 1, beneficial role in permanent ischemic stroke and in Gingko biloba (EGb 761) neuroprotection. Neuroscience 2011, 180, 248–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Zhang, F.; Wang, S.; Zhang, M.; Weng, Z.; Li, P.; Gan, Y.; Zhang, L.; Cao, G.; Gao, Y.; Leak, R.K.; et al. Pharmacological induction of heme oxygenase-1 by a triterpenoid protects neurons against ischemic injury. Stroke 2012, 43, 1390–1397. [Google Scholar] [CrossRef]
  189. Zhao, J.; Kobori, N.; Aronowski, J.; Dash, P.K. Sulforaphane reduces infarct volume following focal cerebral ischemia in rodents. Neurosci. Lett. 2006, 393, 108–112. [Google Scholar] [CrossRef]
  190. Parada, E.; Buendia, I.; León, R.; Negredo, P.; Romero, A.; Cuadrado, A.; López, M.G.; Egea, J. Neuroprotective effect of melatonin against ischemia is partially mediated by alpha-7 nicotinic receptor modulation and HO-1 overexpression. J. Pineal Res. 2014, 56, 204–212. [Google Scholar] [CrossRef]
  191. Chern, C.M.; Liao, J.F.; Wang, Y.H.; Shen, Y.C. Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice. Free Radic. Biol. Med. 2012, 52, 1634–1647. [Google Scholar] [CrossRef]
  192. Lin, K.; Zhang, Z.; Zhang, Z.; Zhu, P.; Jiang, X.; Wang, Y.; Deng, Q.; Lam Yung, K.K.; Zhang, S. Oleanolic Acid Alleviates Cerebral Ischemia/Reperfusion Injury via Regulation of the GSK-3β/HO-1 Signaling Pathway. Pharmaceuticals 2021, 15, 1. [Google Scholar] [CrossRef] [PubMed]
  193. Yang, Y.; Dong, B.; Lu, J.; Wang, G.; Yu, Y. Hemopexin reduces blood-brain barrier injury and protects synaptic plasticity in cerebral ischemic rats by promoting EPCs through the HO-1 pathway. Brain Res. 2018, 1699, 177–185. [Google Scholar] [CrossRef] [PubMed]
  194. Liang, C.; Cang, J.; Wang, H.; Xue, Z. Propofol attenuates cerebral ischemia/reperfusion injury partially using heme oxygenase-1. J. Neurosurg. Anesthesiol. 2013, 25, 311–316. [Google Scholar] [CrossRef] [PubMed]
  195. Howell, J.A.; Bidwell, G.L.B., III. Targeting the NF-κB pathway for therapy of ischemic stroke. Ther. Deliv. 2020, 11, 113–123. [Google Scholar] [CrossRef] [PubMed]
  196. Harari, O.A.; Liao, J.K. NF-κB and innate immunity in ischemic stroke. Ann. N. Y. Acad. Sci. 2010, 1207, 32–40. [Google Scholar] [CrossRef]
  197. Jover-Mengual, T.; Hwang, J.-Y.; Byun, H.-R.; Court-Vazquez, B.L.; Centeno, J.M.; Burguete, M.C.; Zukin, R.S. The Role of NF-κB Triggered Inflammation in Cerebral Ischemia. Front. Cell. Neurosci. 2021, 15, 3610. [Google Scholar] [CrossRef]
  198. Sironi, L.; Banfi, C.; Brioschi, M.; Gelosa, P.; Guerrini, U.; Nobili, E.; Gianella, A.; Paoletti, R.; Tremoli, E.; Cimino, M. Activation of NF-kB and ERK1/2 after permanent focal ischemia is abolished by simvastatin treatment. Neurobiol. Dis. 2006, 22, 445–451. [Google Scholar] [CrossRef]
  199. Wang, H.Y.; Hou, Y.; Sun, J.; Xu, Q.L.; Zhang, D.Q.; Han, Y.C. Effect of atorvastatin on expression of TLR4 and NF-κB in stroke rats and its protective effect on brain. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 10799–10805. [Google Scholar] [CrossRef]
  200. Hölschermann, H.; Schuster, D.; Parviz, B.; Haberbosch, W.; Tillmanns, H.; Muth, H. Statins prevent NF-kappaB transactivation independently of the IKK-pathway in human endothelial cells. Atherosclerosis 2006, 185, 240–245. [Google Scholar] [CrossRef]
  201. Jahromi, G.P.; Shabanzadeh, A.P.; Hashtjini, M.M.; Sadr, S.S.; Vani, J.R.; Sarshoori, J.R.; Charish, J. Bone marrow-derived mesenchymal stem cell and simvastatin treatment leads to improved functional recovery and modified c-Fos expression levels in the brain following ischemic stroke. Iran. J. Basic Med. Sci. 2018, 21, 1004–1012. [Google Scholar] [CrossRef]
  202. Choi, N.Y.; Kim, J.Y.; Hwang, M.; Lee, E.H.; Choi, H.; Lee, K.Y.; Lee, Y.J.; Koh, S.H. Atorvastatin Rejuvenates Neural Stem Cells Injured by Oxygen-Glucose Deprivation and Induces Neuronal Differentiation through Activating the PI3K/Akt and ERK Pathways. Mol. Neurobiol. 2019, 56, 2964–2977. [Google Scholar] [CrossRef]
  203. Wang, X.; Sun, Z.J.; Wu, J.L.; Quan, W.Q.; Xiao, W.D.; Chew, H.; Jiang, C.M.; Li, D. Naloxone attenuates ischemic brain injury in rats through suppressing the NIK/IKKα/NF-κB and neuronal apoptotic pathways. Acta Pharmacol. Sin. 2019, 40, 170–179. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, Y.; Dang, W.; Zhang, S.; Wang, L.; Zhang, X. Artesunate attenuates inflammatory injury and inhibits the NF-κB pathway in a mouse model of cerebral ischemia. J. Int. Med. Res. 2021, 49, 03000605211053549. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, K.; Yang, Y.; Ge, H.; Wang, J.; Chen, X.; Lei, X.; Zhong, J.; Zhang, C.; Xian, J.; Lu, Y.; et al. Artesunate promotes the proliferation of neural stem/progenitor cells and alleviates Ischemia-reperfusion Injury through PI3K/Akt/FOXO-3a/p27(kip1) signaling pathway. Aging 2020, 12, 8029–8048. [Google Scholar] [CrossRef]
  206. Zhang, K.; Yang, Y.; Ge, H.; Wang, J.; Lei, X.; Chen, X.; Wan, F.; Feng, H.; Tan, L. Neurogenesis and Proliferation of Neural Stem/Progenitor Cells Conferred by Artesunate via FOXO3a/p27Kip1 Axis in Mouse Stroke Model. Mol. Neurobiol. 2022. [Google Scholar] [CrossRef]
  207. Wang, X.; Shen, B.; Sun, D.; Cui, X. Aspirin ameliorates cerebral infarction through regulation of TLR4/NF-κB-mediated endoplasmic reticulum stress in mouse model. Mol. Med. Rep. 2018, 17, 479–487. [Google Scholar] [CrossRef]
  208. Sheibani, V.; Esmaeilpour, K.; Eslaminejad, T.; Nematollahi-Mahani, S.N. Coadministration of the Human Umbilical Cord Matrix-Derived Mesenchymal Cells and Aspirin Alters Postischemic Brain Injury in Rats. J. Stroke Cerebrovasc. Dis. 2015, 24, 2005–2016. [Google Scholar] [CrossRef]
  209. Zhang, H.; Sun, X.; Xie, Y.; Zan, J.; Tan, W. Isosteviol Sodium Protects Against Permanent Cerebral Ischemia Injury in Mice via Inhibition of NF-κB-Mediated Inflammatory and Apoptotic Responses. J. Stroke Cerebrovasc. Dis. 2017, 26, 2603–2614. [Google Scholar] [CrossRef]
  210. Zhang, H.; Sun, X.; Xie, Y.; Tian, F.; Hu, H.; Tan, W. Isosteviol Sodium Inhibits Astrogliosis after Cerebral Ischemia/Reperfusion Injury in Rats. Biol. Pharm. Bull. 2018, 41, 575–584. [Google Scholar] [CrossRef] [Green Version]
  211. Zhang, H.; Lu, M.; Zhang, X.; Kuai, Y.; Mei, Y.; Tan, Q.; Zhong, K.; Sun, X.; Tan, W. Isosteviol Sodium Protects against Ischemic Stroke by Modulating Microglia/Macrophage Polarization via Disruption of GAS5/miR-146a-5p sponge. Sci. Rep. 2019, 9, 12221. [Google Scholar] [CrossRef] [PubMed]
  212. Rösing, N.; Salvador, E.; Güntzel, P.; Kempe, C.; Burek, M.; Holzgrabe, U.; Soukhoroukov, V.; Wunder, C.; Förster, C. Neuroprotective Effects of Isosteviol Sodium in Murine Brain Capillary Cerebellar Endothelial Cells (cerebEND) After Hypoxia. Front. Cell. Neurosci. 2020, 14, 3950. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, J.; Jiang, H.; Wu, F.; Chi, X.; Pang, Y.; Jin, H.; Sun, Y.; Zhang, S. Neuroprotective Effects of Hesperetin in Regulating Microglia Polarization after Ischemic Stroke by Inhibiting TLR4/NF-κB Pathway. J. Healthc. Eng. 2021, 2021, 9938874. [Google Scholar] [CrossRef] [PubMed]
  214. Ran, Y.; Qie, S.; Gao, F.; Ding, Z.; Yang, S.; Tian, G.; Liu, Z.; Xi, J. Baicalein ameliorates ischemic brain damage through suppressing proinflammatory microglia polarization via inhibiting the TLR4/NF-κB and STAT1 pathway. Brain Res. 2021, 1770, 147626. [Google Scholar] [CrossRef]
  215. Xue, X.; Qu, X.J.; Yang, Y.; Sheng, X.H.; Cheng, F.; Jiang, E.N.; Wang, J.H.; Bu, W.; Liu, Z.P. Baicalin attenuates focal cerebral ischemic reperfusion injury through inhibition of nuclear factor κB p65 activation. Biochem. Biophys. Res. Commun. 2010, 403, 398–404. [Google Scholar] [CrossRef]
  216. Dai, M.; Chen, B.; Wang, X.; Gao, C.; Yu, H. Icariin enhance mild hypothermia-induced neuroprotection via inhibiting the activation of NF-κB in experimental ischemic stroke. Metab. Brain Dis. 2021, 36, 1779–1790. [Google Scholar] [CrossRef]
  217. Deng, Y.; Xiong, D.; Yin, C.; Liu, B.; Shi, J.; Gong, Q. Icariside II protects against cerebral ischemia-reperfusion injury in rats via nuclear factor-κB inhibition and peroxisome proliferator-activated receptor up-regulation. Neurochem. Int. 2016, 96, 56–61. [Google Scholar] [CrossRef]
  218. Liu, C.; Liu, S.; Xiong, L.; Zhang, L.; Li, X.; Cao, X.; Xue, J.; Li, L.; Huang, C.; Huang, Z. Genistein-3′-sodium sulfonate Attenuates Neuroinflammation in Stroke Rats by Down-Regulating Microglial M1 Polarization through α7nAChR-NF-κB Signaling Pathway. Int. J. Biol. Sci. 2021, 17, 1088–1100. [Google Scholar] [CrossRef]
  219. Nabavi, S.F.; Daglia, M.; Tundis, R.; Loizzo, M.R.; Sobarzo-Sanchez, E.; Orhan, I.E.; Nabavi, S.M. Genistein: A Boon for Mitigating Ischemic Stroke. Curr. Top. Med. Chem. 2015, 15, 1714–1721. [Google Scholar] [CrossRef]
  220. Le, K.; Song, Z.; Deng, J.; Peng, X.; Zhang, J.; Wang, L.; Zhou, L.; Bi, H.; Liao, Z.; Feng, Z. Quercetin alleviates neonatal hypoxic-ischemic brain injury by inhibiting microglia-derived oxidative stress and TLR4-mediated inflammation. Inflamm. Res. 2020, 69, 1201–1213. [Google Scholar] [CrossRef]
  221. Paramita, P.; Sethu, S.N.; Subhapradha, N.; Ragavan, V.; Ilangovan, R.; Balakrishnan, A.; Srinivasan, N.; Murugesan, R.; Moorthi, A. Neuro-protective effects of nano-formulated hesperetin in a traumatic brain injury model of Danio rerio. Drug Chem. Toxicol. 2022, 45, 507–514. [Google Scholar] [CrossRef] [PubMed]
  222. Liu, D.; Ye, Y.; Xu, L.; Yuan, W.; Zhang, Q. Icariin and mesenchymal stem cells synergistically promote angiogenesis and neurogenesis after cerebral ischemia via PI3K and ERK1/2 pathways. Biomed. Pharmacother. 2018, 108, 663–669. [Google Scholar] [CrossRef] [PubMed]
  223. Zhang, L.L.; Zhang, H.T.; Cai, Y.Q.; Han, Y.J.; Yao, F.; Yuan, Z.H.; Wu, B.Y. Anti-inflammatory Effect of Mesenchymal Stromal Cell Transplantation and Quercetin Treatment in a Rat Model of Experimental Cerebral Ischemia. Cell. Mol. Neurobiol. 2016, 36, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
  224. Auphan, N.; DiDonato, J.A.; Rosette, C.; Helmberg, A.; Karin, M. Immunosuppression by glucocorticoids: Inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 1995, 270, 286–290. [Google Scholar] [CrossRef] [PubMed]
  225. Nelson, G.; Wilde, G.J.C.; Spiller, D.G.; Kennedy, S.M.; Ray, D.W.; Sullivan, E.; Unitt, J.F.; White, M.R.H. NF-κB signalling is inhibited by glucocorticoid receptor and STAT6 via distinct mechanisms. J. Cell Sci. 2003, 116, 2495–2503. [Google Scholar] [CrossRef] [Green Version]
  226. Sandercock, P.A.; Soane, T. Corticosteroids for acute ischaemic stroke. Cochrane Database Syst. Rev. 2011, 2011, CD000064. [Google Scholar] [CrossRef]
  227. Azadi, R.; Mousavi, S.E.; Kazemi, N.M.; Yousefi-Manesh, H.; Rezayat, S.M.; Jaafari, M.R. Anti-inflammatory efficacy of Berberine Nanomicelle for improvement of cerebral ischemia: Formulation, characterization and evaluation in bilateral common carotid artery occlusion rat model. BMC Pharmacol. Toxicol. 2021, 22, 54. [Google Scholar] [CrossRef]
  228. Zeynalov, E.; Doré, S. Low doses of carbon monoxide protect against experimental focal brain ischemia. Neurotox. Res. 2009, 15, 133–137. [Google Scholar] [CrossRef] [Green Version]
  229. Ye, R.; Kong, X.; Yang, Q.; Zhang, Y.; Han, J.; Li, P.; Xiong, L.; Zhao, G. Ginsenoside rd in experimental stroke: Superior neuroprotective efficacy with a wide therapeutic window. Neurotherapeutics 2011, 8, 515–525. [Google Scholar] [CrossRef] [Green Version]
  230. Nada, S.E.; Tulsulkar, J.; Shah, Z.A. Heme oxygenase 1-mediated neurogenesis is enhanced by Ginkgo biloba (EGb 761®) after permanent ischemic stroke in mice. Mol. Neurobiol. 2014, 49, 945–956. [Google Scholar] [CrossRef]
  231. Li, R.; Si, M.; Jia, H.Y.; Ma, Z.; Li, X.W.; Li, X.Y.; Dai, X.R.; Gong, P.; Luo, S.Y. Anfibatide alleviates inflammation and apoptosis via inhibiting NF-kappaB/NLRP3 axis in ischemic stroke. Eur. J. Pharmacol. 2022, 926, 175032. [Google Scholar] [CrossRef]
  232. Cai, J.; Liang, J.; Zhang, Y.; Shen, L.; Lin, H.; Hu, T.; Zhan, S.; Xie, M.; Liang, S.; Xian, M.; et al. Cyclo-(Phe-Tyr) as a novel cyclic dipeptide compound alleviates ischemic/reperfusion brain injury via JUNB/JNK/NF-κB and SOX5/PI3K/AKT pathways. Pharmacol. Res. 2022, 180, 106230. [Google Scholar] [CrossRef] [PubMed]
  233. Chen, B.; Cao, P.; Guo, X.; Yin, M.; Li, X.; Jiang, L.; Shao, J.; Chen, X.; Jiang, C.; Tao, L.; et al. Maraviroc, an inhibitor of chemokine receptor type 5, alleviates neuroinflammatory response after cerebral Ischemia/reperfusion injury via regulating MAPK/NF-κB signaling. Int. Immunopharmacol. 2022, 108, 108755. [Google Scholar] [CrossRef] [PubMed]
  234. Sun, X.; Liu, B. Donepezil ameliorates oxygen-glucose deprivation/reoxygenation-induced brain microvascular endothelial cell dysfunction via the SIRT1/FOXO3a/NF-κB pathways. Bioengineered 2022, 13, 7760–7770. [Google Scholar] [CrossRef] [PubMed]
  235. Wang, J.; Fu, X.; Zhang, D.; Yu, L.; Li, N.; Lu, Z.; Gao, Y.; Wang, M.; Liu, X.; Zhou, C.; et al. ChAT-positive neurons participate in subventricular zone neurogenesis after middle cerebral artery occlusion in mice. Behav. Brain Res. 2017, 316, 145–151. [Google Scholar] [CrossRef] [Green Version]
  236. Man, J.; Cui, K.; Fu, X.; Zhang, D.; Lu, Z.; Gao, Y.; Yu, L.; Li, N.; Wang, J. Donepezil promotes neurogenesis via Src signaling pathway in a rat model of chronic cerebral hypoperfusion. Brain Res. 2020, 1736, 146782. [Google Scholar] [CrossRef]
  237. Liu, W.; Shao, C.; Zang, C.; Sun, J.; Xu, M.; Wang, Y. Protective effects of dexmedetomidine on cerebral ischemia/reperfusion injury via the microRNA-214/ROCK1/NF-κB axis. BMC Anesthesiol. 2021, 21, 203. [Google Scholar] [CrossRef]
  238. Xian, M.; Cai, J.; Zheng, K.; Liu, Q.; Liu, Y.; Lin, H.; Liang, S.; Wang, S. Aloe-emodin prevents nerve injury and neuroinflammation caused by ischemic stroke via the PI3K/AKT/mTOR and NF-κB pathway. Food Funct. 2021, 12, 8056–8067. [Google Scholar] [CrossRef]
  239. Zhang, D.; Feng, Y.; Pan, H.; Xuan, Z.; Yan, S.; Mao, Y.; Xiao, X.; Huang, X.; Zhang, H.; Zhou, F.; et al. 9-Methylfascaplysin exerts anti-ischemic stroke neuroprotective effects via the inhibition of neuroinflammation and oxidative stress in rats. Int. Immunopharmacol. 2021, 97, 107656. [Google Scholar] [CrossRef]
  240. Wang, Q.; Zhao, H.; Gao, Y.; Lu, J.; Xie, D.; Yu, W.; He, F.; Liu, W.; Hisatome, I.; Yamamoto, T.; et al. Uric acid inhibits HMGB1-TLR4-NF-κB signaling to alleviate oxygen-glucose deprivation/reoxygenation injury of microglia. Biochem. Biophys. Res. Commun. 2021, 540, 22–28. [Google Scholar] [CrossRef]
  241. Kao, M.H.; Wu, J.S.; Cheung, W.M.; Chen, J.J.; Sun, G.Y.; Ong, W.Y.; Herr, D.R.; Lin, T.N. Clinacanthus nutans Mitigates Neuronal Death and Reduces Ischemic Brain Injury: Role of NF-κB-driven IL-1β Transcription. Neuromol. Med. 2021, 23, 199–210. [Google Scholar] [CrossRef] [PubMed]
  242. Liu, J.; Xu, J.; Mi, Y.; Yang, Y.; Li, Q.; Zhou, D.; Wei, K.; Chen, G.; Li, N.; Hou, Y. Pterostilbene alleviates cerebral ischemia and reperfusion injury in rats by modulating microglial activation. Food Funct. 2020, 11, 5432–5445. [Google Scholar] [CrossRef] [PubMed]
  243. Ling, C.; Liang, J.; Zhang, C.; Li, R.; Mou, Q.; Qin, J.; Li, X.; Wang, J. Synergistic Effects of Salvianolic Acid B and Puerarin on Cerebral Ischemia Reperfusion Injury. Molecules 2018, 23, 564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Zhuang, P.; Zhang, Y.; Cui, G.; Bian, Y.; Zhang, M.; Zhang, J.; Liu, Y.; Yang, X.; Isaiah, A.O.; Lin, Y.; et al. Direct stimulation of adult neural stem/progenitor cells in vitro and neurogenesis in vivo by salvianolic acid B. PLoS ONE 2012, 7, e35636. [Google Scholar] [CrossRef] [Green Version]
  245. Kim, D.C.; Quang, T.H.; Oh, H.; Kim, Y.C. Steppogenin Isolated from Cudrania tricuspidata Shows Antineuroinflammatory Effects via NF-κB and MAPK Pathways in LPS-Stimulated BV2 and Primary Rat Microglial Cells. Molecules 2017, 22, 2130. [Google Scholar] [CrossRef] [Green Version]
  246. Bai, S.; Hu, Z.; Yang, Y.; Yin, Y.; Li, W.; Wu, L.; Fang, M. Anti-Inflammatory and Neuroprotective Effects of Triptolide via the NF-κB Signaling Pathway in a Rat MCAO Model. Anat. Rec. 2016, 299, 256–266. [Google Scholar] [CrossRef] [Green Version]
  247. El-Sahar, A.E.; Safar, M.M.; Zaki, H.F.; Attia, A.S.; Ain-Shoka, A.A. Sitagliptin attenuates transient cerebral ischemia/reperfusion injury in diabetic rats: Implication of the oxidative-inflammatory-apoptotic pathway. Life Sci. 2015, 126, 81–86. [Google Scholar] [CrossRef]
  248. Tian, M.; Yang, M.; Li, Z.; Wang, Y.; Chen, W.; Yang, L.; Li, Y.; Yuan, H. Fluoxetine suppresses inflammatory reaction in microglia under OGD/R challenge via modulation of NF-κB signaling. Biosci. Rep. 2019, 39, BSR20181584. [Google Scholar] [CrossRef]
  249. Wei, N.; Li, C.; Zhu, Y.; Zheng, P.; Hu, R.; Chen, J. Fluoxetine regulates the neuronal differentiation of neural stem cells transplanted into rat brains after stroke by increasing the 5HT level. Neurosci. Lett. 2022, 772, 136447. [Google Scholar] [CrossRef]
Figure 1. The Nrf2 pathway and downstream effectors. Note the inhibition of the KEAP1 complex by ROS to promote the release of Nrf2 for subsequent nuclear translocation and transcription of antioxidant proteins. Downstream, HO-1 inhibits NF-κB signaling to downregulate the inflammatory response.
Figure 1. The Nrf2 pathway and downstream effectors. Note the inhibition of the KEAP1 complex by ROS to promote the release of Nrf2 for subsequent nuclear translocation and transcription of antioxidant proteins. Downstream, HO-1 inhibits NF-κB signaling to downregulate the inflammatory response.
Antioxidants 11 01447 g001
Figure 2. Peripheral contributions to oxidative and inflammatory responses after stroke. This figure illustrates the bidirectional inflammatory and oxidative signaling between the damaged central nervous system tissue and peripheral organs, such as the spleen, lymph nodes, and bone marrow. The Nrf2 pathway can mitigate this exacerbated response via its antioxidative protein products.
Figure 2. Peripheral contributions to oxidative and inflammatory responses after stroke. This figure illustrates the bidirectional inflammatory and oxidative signaling between the damaged central nervous system tissue and peripheral organs, such as the spleen, lymph nodes, and bone marrow. The Nrf2 pathway can mitigate this exacerbated response via its antioxidative protein products.
Antioxidants 11 01447 g002
Figure 3. Crosstalk between Nrf2 and stem cells for targeting stroke-induced inflammation. This figure exemplifies local (neutrophils, monocytes, lymphocytes, and microglia) and peripheral (spleen, lymph nodes, and bone marrow) inflammatory and oxidative stressors due to ischemic and hemorrhagic stroke. The use of Nrf2 pathway amplification concurrent with stem cell therapy can enhance the efficacy of stem cell treatments to reduce secondary cell death following stroke.
Figure 3. Crosstalk between Nrf2 and stem cells for targeting stroke-induced inflammation. This figure exemplifies local (neutrophils, monocytes, lymphocytes, and microglia) and peripheral (spleen, lymph nodes, and bone marrow) inflammatory and oxidative stressors due to ischemic and hemorrhagic stroke. The use of Nrf2 pathway amplification concurrent with stem cell therapy can enhance the efficacy of stem cell treatments to reduce secondary cell death following stroke.
Antioxidants 11 01447 g003
Table 1. Nrf2-based compounds and downstream molecules that interact with stem cell therapy for stroke.
Table 1. Nrf2-based compounds and downstream molecules that interact with stem cell therapy for stroke.
CompoundTarget(s)Effect on Stroke TreatmentCitationEffect on Stem Cells in Setting of StrokeCitation
Dimethyl FumarateNrf2 Pathway via KEAP1
  • Reduced infarct volume
  • Inhibited leukocyte infiltration
  • Supported subcellular localization of tight junctions
  • Decreased neurological deficits
  • Reduced edema volume
[120]
  • Enhanced NSC survival
  • Increased motor neuron survival
  • Reduced ROS production
  • Decreased apoptosis
[103]
BerberineNrf2 Pathway via PPARδ
  • Reduced inflammation by upregulation of inhibitory cytokines
  • Reduced infarct volume
  • Reduced edema volume
[227]
  • Promoted NSC proliferation
  • Promoted ROS scavenging
[121]
Carbon MonoxideNrf2 and NF-κB Pathways
  • Reduced infarct volume
  • Reduced cerebral edema
  • Improved neurological function
[228]
  • Modulated NSC tolerance to iron overload
  • Increased NSC proliferation
[126]
SulforaphaneNrf2 Pathway Upstream
  • Reduced BBB disruption
  • Reduced lesion progression
  • Decreased neurological deficits
[131]
  • Promoted NSC proliferation
  • Promoted NSC differentiation
[132]
DoxycyclineNrf2 Pathway Upstream
  • Protected BBB via inhibition of MMP-2, MMP-9, and PKCδ
  • Blocked leukocyte adhesion
[136,137,138]
  • Inhibited microglial activation
  • Promoted superoxide scavenging
  • Reduced NSC apoptosis
[134,135]
MinocyclineNrf2 Pathway Upstream
  • Preserved BBB via MMP-9 inhibition
  • Improved neurological/functional outcomes
[143]
  • Increased NSC viability
  • Increased NSC proliferation
  • Promoted NSC release of neuroprotective factors
  • Restored neurogenesis
[112,147]
Tert-butylhydroquinoneNrf2 Pathway Upstream
  • Reduced cortical damage
  • Reduced sensorimotor deficits
[33]
  • Inhibited microglial activation
  • Decreased release of proinflammatory cytokines
  • Increased angiogenesis
[153,155]
ResveratrolNrf2 Pathway Upstream
  • Reduced infarct volume
  • Improved neurobehavioral scores
[159]
  • Increased proliferation and mobilization of endothelial cell progenitors
  • Enhanced NSC survival and proliferation
  • Reduced NSC apoptosis
[157,158,161]
GinsengNrf2 and NF-κB Pathways
  • Reduced infarct volume
  • Reduced edema volume
  • Improved neurological outcomes
[229]
  • Promoted proliferation of endothelial precursor cells and NSCs
  • Enhanced neurogenesis, angiogenesis, and synaptic plasticity
  • Induced NSC differentiation
[164]
TheaflavinNrf2 Pathway Upstream (via nuclear translocation)
  • Reduced infarct volume and neuronal injury
  • Improved memory impairment and learning ability
[170]
  • Increased Bcl-2 overexpression
  • Inhibited mitochondrial apoptotic pathway
[171]
CurcuminNrf2 Pathway Upstream (via gene transcription)
  • Decreased neuronal cell death
  • Decreased lipid peroxidation
  • Protected hippocampal CA1 neurons
  • Prevented BBB disruption
[172,175,176]
  • Increased NSC proliferation, differentiation, and migration
  • Enhanced viability of embryonic stem cell exosomes
[178,179]
Gingko bilobaHO-1 Downstream
  • Improved infarction volume
[187]
  • Improved infarction volume and motor skills
  • Enhanced proliferation of NSCs
[230]
2-cyano-3,12 dioxooleana-1,9 dien-28-oyl imidazolineHO-1 Downstream
  • Upregulated HO-1
  • Increased neuronal survival
  • Improved neurological dysfunction and infarct volume
[188]--
MelatoninHO-1 Downstream
  • Increased expression of HO-1
  • Improved infarct size and motor skills
[190]
  • Enhanced endogenous neurogenesis and cell proliferation in peri-infarct regions
[191]
Oleanolic acidHO-1 Downstream
  • Attenuated cytotoxicity and overproduction of intracellular ROS via suppression of GSK-3β activation and enhancement of HO-1 expression
  • Improved area of cerebral infarction and neurological scores
[192]--
HemopexinHO-1 Downstream
  • Induced expression of HO-1
  • Promoted migration and differentiation of endothelial progenitor cells
  • Facilitated angiogenesis
[193]--
PropofolHO-1 Downstream
  • Improved neurological deficits and infarct volume
  • Attenuated neuron apoptosis
  • Increased HO-1 protein expression in ischemic penumbra
[194]--
SimvastatinNF-κB Pathway
  • Abolished NF-κB activation
[198]
  • Increased bone-marrow-derived mesenchymal stem cell relocation, endogenous neurogenesis, arteriogenesis, astrocyte activation
  • Decreased neuronal damage
[201]
AtorvastatinNF-κB Pathway
  • Decreased expression of TLR4 and NF-κB
  • Improved neurological deficit scores
[199]
  • Restored survival, proliferation, migration, and differentiation of NSCs
[202]
NaloxoneNF-κB Pathway
  • Decreased brain edema, infarction volume, and morphological injury
  • Improved motor behavioral function
  • Inhibited nuclear translocation of NF-κB p65
  • Decreased concentrations of nuclear NF-κB p65 in the ischemic penumbra
  • Increased IκBα
  • Attenuated phosphorylated NIK and IKKα levels in the ischemic penumbra
  • Increased Bcl-2 and decreased Bax
  • Stabilized mitochondrial transmembrane potential
  • Inhibited cytochrome C release and activation of caspase 3 and caspase 9
[203]--
ArtesunateNF-κB Pathway
  • Improved neurological deficit scores and infarct volumes
  • Reduced neutrophil infiltration and microglia activation
  • Downregulated TNF-α and IL-1β expression
  • Inhibited nuclear translocation of NF-κB
[204]
  • Promoted proliferation of NSCs in ipsilateral subventricular zone and peri-infarct cortex
[205,206]
AspirinNF-κB Pathway
  • Suppressed TLR4 and NF-κB expression in cerebrovascular endothelial cells
  • Improved infarct area
[207]
  • Improved learning and memory with human umbilical cord matrix-derived stem cells
[208]
Isosteviol sodiumNF-κB Pathway
  • Improved infarct volume and neurological scores
  • Increased number of restored neurons and decreased astrocytes
  • Downregulated mRNA expression of inhibitor of nuclear factor kappa-B kinase-α, inhibitor of nuclear factor kappa-B kinase-β, NF-κB, inhibitor of NF-κB-α, tumor necrosis factor-α, interleukin-1 beta, Bcl-2-associated X protein, and caspase 3
  • Upregulated mRNA of Bcl-2
[209]--
HesperetinNF-κB Pathway
  • Improved neurological deficit
  • Regulated polarization of microglia
[213]
  • Induced proliferation of NSCs
[221]
BaicaleinNF-κB Pathway
  • Improved infarct volume and sensorimotor function
  • Decreased proinflammatory markers, release of proinflammatory cytokines, and nitric oxide
  • Increased anti-inflammatory markers CD206 and Arg-1
  • Reduced TLR4, phosphorylation of IKBα and p65, and nuclear translocation of NF-κB p65
  • Inhibited phosphorylation of signal transducer and activator of transcription 1 (STAT1)
[214]--
IcariinNF-κB Pathway
  • Reduced cerebral infarct volume, neurological deficit, cerebral cell death of rats
  • Downregulated expression of TNF-α, IL-6, C-caspase 3, and Bax
  • Upregulated expression of Bcl-2
  • Downregulated activation of PPARs/Nrf2/NF-κB and JAK2/STAT3 pathways
[216]
  • Increased expression of BDNF and VEGF in the hippocampus and frontal cortex
  • Promoted angiogenesis and neurogenesis
  • Improved brain infarction volumes, motor and somatosensory deficits, and neurobehavioral outcomes
[222]
Genistein-3′-sodium sulfonateNF-κB Pathway
  • Improved brain infarct volume and neurological function
  • Reduced microglia M1 polarization and IL-1β levels
  • Inhibited activation of NF-κB signaling in the ischemic penumbra
[218]--
QuercetinNF-κB Pathway
  • Improved cerebral infarct volumes
  • Improved cognitive and motor deficits
[220]
  • Improved neurological functional recovery
  • Reduced proinflammatory cytokines IL- 1β and IL-6
  • Increased anti-inflammatory cytokines IL-4, IL-10, and TGF-β1
  • Inhibited cell apoptosis
  • Improved survival rate of human umbilical mesenchymal stromal cells
[223]
AnfibatideNF-κB Pathway
  • Improved neurological deficit, neurobehavioral impairment, and infarct volume
  • Increased cell viability
  • Decreased LDH release
  • Inhibited expression of p-IκBα, p-p65, NLRP3, ASC, cleaved caspase 1, Bax, and cleaved caspase3
  • Promoted expression of Bcl-2
  • Decreased TUNEL-positive cell number and concentration of IL-β and IL-18
[231]--
Cyclo-(Phe-Tyr)NF-κB Pathway
  • Decreased size of cerebral infarct
  • Improved neurological scores
  • Blocked inflammatory and oxidative factor release
[232]--
Maraviroc NF-κB Pathway
  • Improved neurological deficits and infarct volumes
  • Decreased levels of apoptosis and inflammation
  • Increased viability of primary microglia
  • Decreased secretion of and expression of IL-1β, IL-6, and TNF-α in microglia
  • Inhibited activity of NF-κB pathway and JNK pathway
[233]--
DonepezilNF-κB Pathway
  • Increased cell viability of human brain microvascular endothelial cells
  • Promoted cell migration and angiogenesis
  • Decreased cell permeability
  • Increased expression of tight junction proteins
  • Regulated expression of SIRT1/FOXO3a/NF-κB
[234]
  • Enhanced post-stroke neurogenic effects that naturally occur in the subventricular zone such as:
    Upregulated metabotropic acetylcholine receptors, phosphorylated protein kinase C, and p-38
    Increased number of BrdU/doublecortin-positive cells, protein count of phosphorylated-neural cell adhesion molecules, and mammalian achaete scute homolog-1
  • Induced proliferation of NSCs and neuroblasts in subventricular zone
[235,236]
DexmedetomidineNF-κB Pathway
  • Reduced infarction area
  • Increased miR-214 expression
[237]--
Aloe-emodinNF-κB Pathway
  • Improved infarct size and behavioral score
  • Decreased expression of TNF-α, MDA, LDH, caspase 3, and NF-κB
  • Increased expression of SOD, Bcl-2/Bax, PI3K, AKT, and mTOR
[238]--
9-MethylfascaplysinNF-κB Pathway
  • Improved motor impairments and infarct size
  • Reduced activation of microglia/macrophage in ischemic penumbra
  • Reduced expression of proinflammatory factors
  • Inhibited oxidative stress and activation of NF-κB and NLRP3 inflammasome
[239]--
Uric acidNF-κB Pathway
  • Attenuated severity of cerebral infarction and activation of microglia in cerebral cortex
  • Reduced release of proinflammatory cytokines TNF-α, IL1β, and IL6
  • Improved cell viability
  • Decreased LDH release
[240]--
Clinacanthus nutansNF-κB Pathway
  • Inhibited IL-1β transcription
  • Attenuated IκBα degradation
  • Decreased production of IL-6 and TNFα
[241]--
PterostilbeneNF-κB Pathway
  • Improved neurological scores, edema, and infarct volume
  • Increased number of mature neurons
  • Decreased microglia activation
  • Reduced iNOS and IL-1β mRNA expression
  • Promoted IκBα expression
  • Inhibited expression of inflammatory cytokines
  • Suppressed NADPH activity
  • Decreased ROS production
[242]--
Salvianolic Acid B and PuerarinNF-κB Pathway
  • Reduced ROS levels
  • Inhibited apoptosis
  • Improved mitochondrial membrane potential
  • Improved neurological deficit scores and infarct area
  • Inhibited expression of proinflammatory cytokines (TNF-α, IL-1β, IL-6)
[243]Salvianolic Acid B:
  • Induced proliferation of NSCs
  • Improved cognitive impairment
[244]
SteppogeninNF-κB Pathway
  • Inhibited nuclear translocation of NF-κB
  • Suppressed JNK and p38 MAPK signaling
[245]--
TriptolideNF-κB Pathway
  • Attenuated brain infarction volume, water content, neurological deficits, and neuronal cell death rate
  • Downregulated iNOS, COX-2, and GFAP
  • Increased expression of Bcl-2
  • Suppressed Bax and caspase 3
[246]--
SitagliptinNF-κB Pathway
  • Suppressed IL-6 and TNF-α
  • Increased anti-inflammatory IL-10
  • Reduced neutrophil infiltration, lipid peroxides, and nitric oxide associated with replenished reduced glutathione
  • Decreased glutamate
  • Decreased cytochrome C and caspase 3
[247]--
FluoxetineNF-κB Pathway
  • Decreased TNF-α, IL-1β, IL-6, and NF-κB subunits p65 and p50
  • Increased IκBα
[248]
  • Increased NSC differentiation
  • Upregulated neurogenin1 expression
  • Downregulated ERK2 phosphorylation
[249]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gordon, J.; Lockard, G.; Monsour, M.; Alayli, A.; Borlongan, C.V. The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease. Antioxidants 2022, 11, 1447. https://doi.org/10.3390/antiox11081447

AMA Style

Gordon J, Lockard G, Monsour M, Alayli A, Borlongan CV. The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease. Antioxidants. 2022; 11(8):1447. https://doi.org/10.3390/antiox11081447

Chicago/Turabian Style

Gordon, Jonah, Gavin Lockard, Molly Monsour, Adam Alayli, and Cesario V. Borlongan. 2022. "The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease" Antioxidants 11, no. 8: 1447. https://doi.org/10.3390/antiox11081447

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