Previous Article in Journal
Resting-State Functional Connectivity Predicts Attention Problems in Children: Evidence from the ABCD Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Neuroprotection against Radiation-Induced Brain Injury: A Review of Key Compounds

by
Lucas González-Johnson
1,2,3,*,
Ariel Fariña
4,5,
Gonzalo Farías
1,2,
Gustavo Zomosa
2,
Víctor Pinilla-González
1,6 and
Catalina Rojas-Solé
1,6
1
Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile
2
University of Chile Clinical Hospital, Santiago 8380453, Chile
3
Biomedical Neuroscience Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile
4
Fundación Arturo López Pérez, Santiago 7500921, Chile
5
Faculty of Medicine, Universidad de los Andes, Santiago 12455, Chile
6
Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8330111, Chile
*
Author to whom correspondence should be addressed.
NeuroSci 2024, 5(4), 462-484; https://doi.org/10.3390/neurosci5040034
Submission received: 1 September 2024 / Revised: 8 October 2024 / Accepted: 9 October 2024 / Published: 12 October 2024

Abstract

:
Brain radiation is a crucial tool in neuro-oncology for enhancing local tumor control, but it can lead to mild-to-profound and progressive impairments in cognitive function. Radiation-induced brain injury is a significant adverse effect of radiotherapy for cranioencephalic tumors, primarily caused by indirect cellular damage through the formation of free radicals. This results in late neurotoxicity manifesting as cognitive impairment due to free radical production. The aim of this review is to highlight the role of different substances, such as drugs used in the clinical setting and antioxidants such as ascorbate, in reducing the neurotoxicity associated with radiation-induced brain injury. Currently, there is mainly preclinical and clinical evidence supporting the benefit of these interventions, representing a cost-effective and straightforward neuroprotective strategy.

1. Introduction

Radiotherapy (RT) is commonly used to treat various central nervous system (CNS) tumors, including glioblastomas, germinomas, vestibular schwannomas, and brain metastases (BMs) [1]. It is estimated that 20–50% of patients with malignant neoplasms will develop brain metastases during the course of their disease [2]. In this context, radiosurgery and Whole Brain Radiation Therapy (WBRT) play a crucial role, becoming key therapeutic tools for managing both primary and secondary brain tumors [3]. These techniques are particularly useful for treating tumors located in encephalic regions that are difficult to access surgically and as an adjuvant therapy [4,5]. The standard WBRT regimen for BM consists of 30 Gy administered in 10 fractions (10 × 3 Gy) over 2 weeks in most centers globally [6], while radiosurgery can be delivered in a single session or in 3 to 5 fractions [5].
Both brain tumors and their treatments can result in neurocognitive impairment [7]. Traditional neuro-oncological perspectives tend to oversimplify the brain as a series of functional units organized in parallel circuits, often overlooking the significance of CNS volume in radiation dose tolerance. While small volumes of brain tissue can tolerate high radiation doses with minimal functional impact, WBRT, which affects larger volumes, is more likely to cause neurocognitive deficits [3,8]. This is primarily due to damage to normal brain parenchyma, particularly in the hippocampus, where neural stem cells reside [9]. Several randomized controlled trials have demonstrated cognitive impairment in patients following WBRT [10,11,12].
One strategy to reduce the risk of cognitive decline is hippocampal-sparing WBRT, a technique that combines intensity-modulated radiation therapy (IMRT) with hippocampal avoidance. This approach has been shown to lower the risk of cognitive impairment from 30% to 7% [13]. Another method is radiosurgery, which targets metastases with high doses of radiation while sparing normal brain tissue, maintaining effective tumor control without compromising overall survival [14]. However, these advanced techniques are available only in centers equipped with modern radiotherapy technology, leaving many patients without access to hippocampal-sparing WBRT or radiosurgery.
In this context, radiation-induced brain injury (RIBI) remains a significant adverse effect of radiotherapy for cranial tumors, with limited options available for its prevention [15]. Over the past decade, various pharmacological agents have been investigated to mitigate the cognitive toxicity associated with radiotherapy, but their results have been largely suboptimal, partly due to an incomplete understanding of the underlying mechanisms [16,17]. Recent evidence highlights oxidative stress as a key mediator of RIBI, as both in vitro and in vivo models have demonstrated that radiotherapy generates free radicals (FRs), causing indirect cellular damage through sublethal injury and promoting neuronal apoptosis [16,18]. This has spurred interest in antioxidant agents as potential therapies to alleviate RIBI. In light of this, the present manuscript reviews the role of oxidative stress in radiation-induced brain injury and explores various pharmacological agents with antioxidant properties that have been studied as radioprotectants, with a particular emphasis on ascorbic acid (AA), one of the most well-researched compounds in this field.

2. Principles of Radiotherapy in Normal and Cancerous Cells

The response of both tumor and normal cells to multiple doses of radiation is governed by five key factors, commonly referred to as the 5 Rs of radiobiology: (1) DNA repair, (2) redistribution in the cell cycle, (3) reoxygenation, (4) repopulation, and (5) radiosensitivity [19]. These factors are essential in optimizing radiotherapy (RT) treatments.
A critical concept in radiotherapy is the α/β ratio, a metric that describes the sensitivity of tumors and normal tissues to fractionation [1,20]. Tissue response to radiation and fractionated dosing is modeled using various frameworks, with the Linear Quadratic (LQ) model being the most widely applied. This model predicts the biological response of tissues by calculating the surviving fraction of cells after a given dose. The LQ equation incorporates two key parameters: a linear dose coefficient (α), which is more relevant at low doses, and a quadratic dose coefficient (β), which becomes significant at higher doses, typically in the 1–8 Gy range. Clinically, this model distinguishes between early-responding tissues, which have a high α/β ratio, and late-responding tissues, which have a low α/β ratio. Malignant tumors generally exhibit high α/β ratios, while slow-growing benign tumors tend to have lower ratios [21].
For late-responding normal tissues like the brain and spinal cord, the α/β ratio is typically 2–3 Gy, reflecting the low regenerative capacity of the central nervous system (CNS). In contrast, early-responding tissues such as the skin or gastrointestinal tract, along with most squamous cell carcinomas, exhibit an α/β ratio of around 10 Gy [22].
In this context, this section will present the main effects of radiation on the cells of our organism, with emphasis on the mechanisms of damage to the genetic material and its repair, as well as the main mechanisms of cell death involved.

2.1. Radiation-Induced DNA Damage

Radiation causes DNA damage primarily in the form of single-strand breaks (SSBs) and double-strand breaks (DSBs). These lesions are recognized by sensor proteins that activate downstream signaling pathways to initiate the DNA damage response (DDR) [23,24]. The p53 protein plays a central role in DDR, with its concentration and phosphorylation status determining whether the cell will survive or undergo apoptosis [25]. Additionally, cyclin-dependent kinase inhibitors and checkpoint kinases are involved in cell cycle arrest, allowing for DNA repair and the maintenance of genomic stability [25,26,27,28] (Figure 1).

2.2. Double-Strand Break Repair Mechanisms (DSB)

Double-strand breaks (DSBs) can be repaired through two primary mechanisms: non-homologous end-joining (NHEJ) and homologous recombination (HR) [27]. NHEJ, although error-prone, is active throughout the entire cell cycle, particularly in the G1 phase. Its inaccuracy stems from the processing of DNA ends before ligation, which can result in short insertions or deletions, potentially leading to the loss of genetic information or misrepair by ligating ends from different DSBs, resulting in translocations and rearrangements that may cause aneuploidy [27].
In contrast, HR is an error-free process but requires an intact sister chromatid as a repair template, making it only functional during the S and G2 phases of the cell cycle. When cellular damage exceeds repair capacity, cell death (CD) ensues [27].
The choice between NHEJ and HR for DNA repair is regulated by the competition between DNA-end protection and resection, primarily governed by BRCA1 and the X-ray repair cross-complementing proteins. Normal cells possess redundancy in DNA repair pathways, enhancing their repair capacity, whereas cancer cells often exhibit impaired DNA repair mechanisms, leading to a higher occurrence of DNA breaks. If DNA damage is successfully repaired, the cell proceeds with its cycle; otherwise, cell death is triggered [27].

2.3. Cell Death Mechanisms

Radiation-induced cell death (CD) as a result of impaired DNA repair depends on several factors, including cell type, TP53 status, oxygen supply, DNA repair capacity, the stage of the cell cycle during irradiation, microenvironment characteristics, radiation dose, and quality [27,29]. In most solid tumors, mitotic CD is the predominant form of cell death, primarily through mitotic catastrophe, with apoptosis playing a secondary role. In contrast, normal tissues typically undergo senescence (Figure 1).

2.3.1. Apoptosis

Apoptosis is a tightly regulated form of CD characterized by pyknosis, cell shrinkage, and internucleosomal DNA fragmentation [30]. There are three main pathways leading to apoptosis: (1) intrinsic/mitochondrial, (2) extrinsic, and (3) membrane stress/ceramide pathway [27]. Apoptosis has a limited role in the response of most solid tumors to treatment, due to the loss of pro-apoptotic mechanisms during oncogenesis [31,32]. Radiation predominantly activates the intrinsic apoptotic pathway (IAP).
The IAP is initiated by DNA damage, such as single-strand breaks (SSBs) and double-strand breaks (DSBs). When DNA repair fails, prolonged activation of p53 increases the likelihood of apoptosis over cell cycle arrest by disrupting the balance between pro- and anti-apoptotic factors. This imbalance leads to the release of cytochrome c from mitochondria, activating caspase 9 and forming the apoptosome [31]. The extrinsic apoptotic pathway is initiated by external signals, such as tumor necrosis factor (TNF) ligands binding to death receptors on the plasma membrane, which activates caspase 8 [33,34]. Radiation can upregulate death receptors, making cells more susceptible to extrinsic apoptosis. The ceramide pathway, independent of DNA damage and p53, is activated by oxidative stress and membrane damage, leading to ceramide production, which acts as a second messenger in apoptosis signaling [35].
All three pathways ultimately converge on the activation of effector caspases (caspases 3 and 7), initiating the demolition phase of apoptosis, which results in controlled degradation of cellular components [35].

2.3.2. Mitotic Catastrophe (MC)

Mitotic catastrophe is the primary form of p53-independent CD induced by ionizing radiation, particularly in apoptosis-resistant cells [31]. It refers to cell death occurring during, or as a result of, aberrant mitosis [36]. This process is triggered by the premature induction of mitosis before the completion of the S and G2 phases, leading to cell cycle arrest and subsequent regulated death, either during the first mitotic division (mitotic death) or in subsequent divisions via delayed apoptosis or necrosis [31,37]. This delayed response explains the delayed cell death often observed in solid tumors after RT, occurring 2–6 days post-irradiation [38]. Aberrant mitoses and failed cytokinesis result in atypical chromosome segregation and division, producing tetraploid giant cells, aneuploidy, micronuclei formation, and centrosome hyper-amplification, particularly in cells lacking functional p53 [39,40,41]. These cells become incapable of further replication [37,42,43,44].

2.3.3. Senescence

Senescence refers to permanent cell cycle arrest, characterized microscopically by enlarged, flattened cells with increased granularity [45,46]. Radiation-induced senescence is triggered by DNA damage and activation of functional p53 and pRb. Although senescent cells are clonogenically “dead,” they remain metabolically active and viable for extended periods (dormant), secreting factors that can alter the tumor microenvironment (TME) and potentially promote tumor growth and progression [47,48]. In some tumors, senescence may be a mechanism to escape radiation-induced cytotoxicity, and senescent cells can potentially “reawaken” after months or years due to external stimuli in the TME [31]. Other forms of radiation-induced CD include necrosis and autophagy.

3. Oxidative Stress and Antioxidant Defenses in the CNS: Implications for Radiation-Induced Brain Injury

Redox reactions generate pro-oxidant reactive species that, at appropriate concentrations, are essential for various cellular and organismal functions, including defense against microorganisms, intracellular communication, and transcription factor activation. However, when these species exceed optimal levels, they can cause cellular damage, which antioxidant mechanisms strive to counteract [49,50]. The balance between oxidants and antioxidants is critical for maintaining cellular function, but various conditions can disrupt this redox homeostasis, resulting in oxidative stress, where pro-oxidants overwhelm the body’s antioxidant defenses [50].
At the cellular level, reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the principal pro-oxidants, and their effects are modulated by both enzymatic and non-enzymatic antioxidant systems. Key antioxidant enzymes include superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), and thioredoxin (TRx), among others [51]. Oxidative stress induces cellular damage through lipid peroxidation, protein oxidation, and DNA strand breaks, with tissues such as the central nervous system (CNS) and muscle, which have low cell turnover, being particularly vulnerable [52,53,54].
The CNS is especially susceptible to oxidative damage, for several reasons. Firstly, it consumes approximately 20% of the body’s oxygen, which leads to excessive ROS production under pathological conditions [55,56]. Moreover, ROS are involved in critical CNS processes, such as neuronal plasticity, axonal regeneration, and neurotransmission [57]. Despite these essential roles, the CNS has limited antioxidant defenses, with relatively low levels of glutathione and catalase compared to other tissues [58,59,60]. Additionally, the abundance of redox-active transition metals and the generation of hydrogen peroxide through monoaminergic metabolism exacerbate oxidative imbalance [61,62]. The high concentration of polyunsaturated fatty acids (PUFAs) in CNS lipid membranes further increases susceptibility to lipid peroxidation and ferroptosis [63,64].
Oxidative stress also plays a critical role in the pathogenesis of radiation-induced brain injury (RIBI). Ionizing radiation generates substantial amounts of ROS and free radicals through the radiolysis of water, leading to extensive cellular damage, particularly to DNA [65] (Figure 2). While it was previously believed that DNA strand breaks were caused directly by high-energy photons, recent evidence suggests that most of the damage is due to oxidative stress [66,67]. In addition to DNA damage, oxidative stress activates redox-sensitive kinases such as Src, PI3K-Akt, and MAPK, including pathways like Erk, JNK, and p38 [68,69,70]. These kinases regulate transcription factors via phosphorylation and have been implicated in cognitive impairment associated with RIBI, especially in the hippocampus’s CA1 region [71,72].
The mitogen-activated protein kinase (MAPK) pathways consist of kinase cascades that regulate key processes such as cell proliferation, differentiation, survival, and apoptosis [73]. Persistent activation of JNK or p38 pathways has been linked to neuronal apoptosis [74]. Ionizing radiation activates all three MAPK pathways, though the intensity of activation depends on cell type [75], with oxidative stress particularly stimulating p38 MAPK [76,77]. Radiation-induced ROS can also activate the ERK cascade, enhancing c-Jun transcriptional activity and upregulating pro-inflammatory genes such as cyclooxygenase-2, interleukin-1β, and tumor necrosis factor-α [78]. Direct exposure of cells to exogenous hydrogen peroxide (H2O2), mimicking oxidative stress, has similarly been shown to activate MAPK pathways [79,80].
Consequently, the CNS is highly vulnerable to ionizing radiation, highlighting the importance of developing protective therapies [81], with antioxidants emerging as a promising strategy to mitigate such damage [66].

4. Hallmarks of Brain Injury Induced by Radiation

Radiation-induced brain injury can manifest with several hallmark features, depending on the stage and severity of the exposure. These effects are typically classified into early (acute and subacute) and late stages; however, these stages can overlap over time.
These effects reflect the cumulative impact of oxidative stress, inflammation, vascular injury, and neural damage triggered by radiation. The severity of these symptoms often depends on the radiation dose, the area of the brain exposed, and the patient’s individual sensitivity to radiation.

4.1. Inflammation

This is an immediate response where inflammatory cytokines are released, leading to swelling and disruption of the blood–brain barrier. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the CNS and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing radiation-induced brain injury [82].
Following irradiation, activated microglia release inflammatory factors, exacerbating neuroinflammation and facilitating damage progression. Controlling microglial activation and suppressing the secretion of these factors is therefore crucial for preventing such injuries. While microglial activation is central to neuroinflammation, the precise mechanisms by which radiation triggers this response remain unclear, involving multiple signaling pathways [83,84]. Investigating the interactions among microglia, neurons, astrocytes, and peripheral immune cells may offer strategies to mitigate microglial activation, reduce inflammatory agent release, and limit peripheral immune cell infiltration into the brain.

4.2. Brain Edema

After brain trauma, ischemia, inflammation, or the presence of a tumor, water enters astrocytes through AQP4 channels. This leads to brain tissue swelling, cytotoxic edema, and elevated intracranial pressure, all of which can significantly increase morbidity and mortality. Brain edema in radiation-induced brain injury refers to the abnormal accumulation of fluid in the brain tissues as a result of radiation exposure [85]. This swelling occurs because radiation disrupts the blood–brain barrier. Some studies revealed that the water channel protein aquaporin-4 (AQP4) is expressed in perivascular astrocytes end-feet, and regulates water movement across the barrier membrane [86]. In hypoxia- induced cell swelling and damage, AQP4 surface expression increases through a calmodulin-dependent mechanism. The inhibition with trifluoperazine, a typical antipsychotic, reduced AQP4 localization. Therefore, this drug treatment eliminated CNS edema, and promoted faster functional recovery, suggesting that AQP4 inhibition could be a useful therapeutic approach for treating brain edema [87,88]. Indeed, AQP4 trafficking in primary human astrocytes and its vesicular translocation mechanisms are important to the edema treatment [85], and further studies are necessary to determine the effects of this drug on radiation-induced brain injury and in related CNS edema therapies.
In this sense, even the treatment with resveratrol showed that this antioxidant compound ameliorates oxidative stress and inhibits AQP4 in a rat cerebral ischemia-reperfusion injury, being a therapeutic target [89].

4.3. Astrogliosis

Astrocytes provide both structural and functional support to neurons and they contribute to angiogenesis, neurogenesis, synaptogenesis, dendrogenesis, and axogenesis, helping to create an environment conducive to recovery. After a brain injury, they suffer a switch in their phenotype into reactive astrocytes and become activated, leading to astrogliosis. This process plays a significant role, acting as both a protective and potentially harmful response in brain injury [90,91].
Astrogliosis involves the proliferation and hypertrophy of astrocytes, which initially help to protect neural tissue by forming a glial scar and preventing further spread of damage, and which have a protective role in recovery from inflammatory and ischemic disease, as well as their role in degenerative disorders [92].
However, chronic or excessive astrogliosis can contribute to long-term detrimental effects. It can exacerbate inflammation, disrupt the blood–brain barrier, and interfere with neuronal function, plasticity and regeneration. Additionally, excessive accumulation of reactive astrocytes can create a non-permissive environment for neural repair, leading to persistent neurological deficits, cognitive impairments, and exacerbation of radiation-induced tissue damage [93,94].
Some studies on strokes showed that the acute inhibition of AQP4 promoted neurological recovery by diminishing brain edema at the early stage and attenuating peri-infarct astrogliosis and AQP4 depolarization at the subacute stage [95]. Indeed, preconditioned extracellular vesicles derived from hypoxic microglia alleviate post-stroke AQP4 depolarization, restore disrupted cerebrospinal fluid flow, and reduce astrogliosis and neuroinflammation [96].
Therefore, due to their dual role in both supporting and hindering recovery, astrocytes have emerged as promising therapeutic targets for pharmacological interventions aimed at improving functional outcomes and neurological recovery.
Additionally, there are other side effects, such as neurological symptoms, including trigeminal nerve deficit, hydrocephalus, ataxia, and dizziness, which may appear in the acute phase [97]. Vascular damage can also occur, as radiation induces fibrosis and damages blood vessels, leading to chronic ischemia and an increased risk of more injury [98,99]. Another serious consequence is radiation necrosis [100], which involves tissue death in localized areas of the brain, often resulting in permanent neurological deficits during the late stage.

5. Use of Other Pharmacological Approaches against RIBI in Clinical Setting

In recent years, numerous studies have explored various pharmacological strategies to mitigate neurocognitive toxicity resulting from whole-brain radiotherapy (WBRT). Potential treatments to reduce these hallmarks include memantine, alpha tocopherol, indomethacin, renin–angiotensin system blockers, ACE inhibitors, PPAR-α, melatonin, and metformin [101,102,103]. However, there is a lack of human studies with these compounds and neuroprotective outcomes. An exception is memantine, which can reduce ROS and pyroptosis via NLRP3/NLRC4/Caspase-1 in RIBI [104]. Moreover, a recent study showed that pretreatment with metformin reduces inflammation and decreases DNA damage in the in vitro and downstream pathways involved in RIBI [102].

5.1. Memantine

Memantine is a noncompetitive, low-affinity, open-channel antagonist of the N-methyl-D-aspartate receptor (NMDAR). Glutamate serves as the primary excitatory neurotransmitter in cortical and hippocampal neurons, with the NMDAR playing a critical role in learning and memory. WBRT is known to induce profound capillary rarefaction, reduce vascular density, and impair vasculogenesis, all of which contribute to radiation-induced cognitive decline [16].
Memantine has been shown to block ischemia-induced NMDA excitation and has proven effective in treating vascular dementia [105]. As such, it is posited that memantine may provide neuroprotection against radiation-induced cognitive impairment, making it a promising candidate for prophylactic use during radiation therapy [106]. Preclinical models have demonstrated its neuroprotective effects [107,108,109], and in two placebo-controlled Phase III trials, memantine was validated as an effective treatment for small-vessel disease [110,111].
According to Brown et al. (2013), a placebo-controlled Phase III trial involving patients with brain metastases receiving WBRT showed that memantine significantly extended the time to cognitive decline, with neurocognitive function preservation improved by up to 31% in the memantine group [112]. However, it is concerning that nearly 70% of brain-metastases patients treated with WBRT still experienced neurocognitive deterioration within six months, and this figure rose to between 50% and 90% within three to six months following fractionated WBRT [112,113].
In a Phase II multi-institutional clinical trial (RTOG 0933), Gondi et al. (2014) demonstrated that conformal hippocampal avoidance (HA) using intensity-modulated radiation therapy during WBRT is associated with better memory preservation compared to historical control series [13].
Further supporting this, Brown et al. (2020) found in the Phase III clinical trial NRG CC001 that the combination of memantine with HA-WBRT should be considered standard care for patients with a good performance status who are set to undergo WBRT (excluding metastases in the hippocampal region), in order to preserve cognitive function without compromising overall survival or intracranial progression-free survival [114].
Given the persistent rate of neurocognitive decline, additional neuroprotective strategies continue to be investigated to enhance the benefits of memantine and HA protocols [3,112]. A summary of other pharmacological approaches as preclinical interventions for RIBI is presented in Table 1.

5.2. Vitamin C or Ascorbic Acid

Vitamin C, also known as ascorbic acid (AA), is one of the most potent antioxidants. It is crucial for the development and maintenance of connective tissues and plays a key role in bone formation, wound healing, and the health of gums. Metabolically, it is involved in several vital processes, including the activation of vitamin B, folic acid, the conversion of cholesterol to bile acids, and the transformation of amino acids, such as tryptophan and serotonin. Its antioxidant properties protect the body from free radical damage, and it has been proposed as a therapeutic agent for various diseases [115].
In particular, AA is an essential micronutrient for the CNS, as will be discussed in the following sections.

5.2.1. Dynamics of Ascorbate in the CNS

Ascorbic acid is highly concentrated in the CNS, particularly in the gray matter, including the hippocampus [116,117]. It is a potent water-soluble antioxidant [107] and an essential micronutrient for the CNS [118,119]. AA enters the CNS via SVCT2 transporters at the choroid plexus, which are stereospecific for the L-isomer [120,121]. From the cerebrospinal fluid (CSF), AA diffuses into the brain’s extracellular fluid, with concentrations ranging from 200 to 400 μM [122,123,124]. In fresh brain tissue, AA concentrations reach 1 to 2.6 mM, approximately one-fifth of glutamate levels [116].
When functioning as an antioxidant, AA is oxidized to dehydroascorbic acid (DHA) [125]. DHA crosses the blood–brain barrier via GLUT1 transporters [126]. Cells subjected to oxidative stress, such as epithelial cells, microglia, and stromal cells, produce superoxide via NADPH oxidase, which oxidizes AA to DHA, facilitating its transport [115]. DHA is then taken up by astrocytes through GLUT1 and reduced back to AA, preventing the loss of oxidized AA [115] (Figure 3).
This recycling of AA is facilitated by enzymes such as semidehydroascorbate reductase, which converts the ascorbyl radical back to ascorbate, and dehydroascorbate reductase, which performs the same conversion on DHA [127,128]. These mechanisms are highly active in brain regions rich in ascorbate, ensuring its recirculation [117] (Figure 3).
Astrocytes, with their higher glutathione concentrations, are primarily responsible for reducing DHA to AA. AA is then released into the interstitial space, providing extracellular antioxidant protection, a process stimulated by glutamate release [126,129]. Some AA is taken up by neurons via SVCT2 transporters, delivering intracellular protection [130].
Within neurons, AA can inhibit glucose consumption and stimulate lactate transport. It also accumulates in mitochondria, where it protects mitochondrial membranes and DNA from free radicals [122,131]. The high concentrations of AA in brain tissue and CSF underscore its vital role in maintaining CNS homeostasis. AA has been shown to modulate both glutamate- and dopamine-mediated neurotransmission [104] and promote myelin formation by Schwann cells, while scavenging free radicals [130,132].

5.2.2. Antioxidant Effects of Ascorbic Acid

Ascorbic acid (AA) is likely the most important water-soluble antioxidant in the brain’s extracellular fluid. It plays a key role in reducing excitotoxicity [133] and is essential for regenerating reduced α-tocopherol in cell membranes [134], thereby protecting lipids, proteins, and DNA from oxidative damage and maintaining their normal structure and biological function (Figure 4).
AA halts oxidative stress by donating a hydrogen atom to reactive oxygen species (ROS), leading to the formation of a stable ascorbyl radical—a direct antioxidant mechanism. This action provides protection against oxygen-derived molecular species [135]. Oxidative damage to biomolecules produces measurable by-products, such as 8-oxodeoxyguanosine from DNA [136], F2-isoprostanes from lipids [137] and carbonyl derivatives from proteins [138]. These markers offer valuable methods to assess AA’s antioxidant effects. Additionally, the ascorbyl radical can be monitored using electron paramagnetic resonance spectroscopy to evaluate oxidative stress, as has been carried out in patients with sepsis [139,140].
Due to its potent antioxidant properties, AA is considered an important neuroprotective agent [141]. However, while AA counteracts oxidative stress, it can also form reactive oxidants, particularly in the presence of transition metals [142]. Furthermore, AA exerts indirect antioxidant effects by inhibiting ROS-producing enzymes and reducing inflammatory responses via NF-κB pathway inhibition [66] (Figure 3).

5.3. The Role of Ascorbic Acid in Preventing Radiation-Induced Brain Injury (RIBI)

The protective role of AA in preventing RIBI has garnered significant attention in recent studies. AA exhibits potent antioxidative properties that can mitigate the cellular damage caused by ionizing radiation. Research indicates that the lipophilic vitamin C derivative, 6-o-palmitoylascorbate, shows superior efficacy compared to regular ascorbate in reducing X-ray-induced DNA damage, lipid peroxidation, and protein carbonylation in human lymphocytes [143]. This derivative not only enhances cell viability but also prevents the depletion of crucial antioxidants like glutathione, thus offering substantial protection against oxidative stress induced by radiation.
In addition to its protective effects, AA has been shown to significantly reduce DNA double-strand breaks (DSBs) associated with radiation exposure. A randomized, double-blind, placebo-controlled trial found that pre-treatment with AA led to an impressive 87% reduction in DSBs in patients exposed to high radiation doses during cardiac examinations [144]. Moreover, oral administration of AA prior to abdominal contrast-enhanced CT scans resulted in a 61% reduction in the mean increase of γ-H2AX foci, further underscoring its capacity to protect against genetic damage from radiation [145].
The cumulative evidence suggests that AA, especially when used in conjunction with other antioxidants like N-acetylcysteine, may play a crucial role in enhancing cellular defenses against RIBI. Its ability to scavenge free radicals and stabilize genomic integrity highlights its potential as a therapeutic agent in clinical settings involving radiation exposure [146]. Thus, incorporating vitamin C into pre-radiation protocols could be a valuable strategy for reducing the risk of RIBI and improving patient outcomes.

5.4. The Dual Effect of Ascorbic Acid in Cancer

Ascorbic acid (AA) has been shown to enhance tumor radiosensitization while simultaneously reducing radiation-induced toxicity in normal tissues, particularly in non-CNS models of pancreatic cancer [147]. However, the overall role of antioxidants in cancer remains a topic of considerable debate. On one hand, antioxidants serve a protective function by neutralizing free radicals, thereby reducing oxidative stress and potentially lowering the risk of cancer development by preventing DNA damage. This protective mechanism is crucial, as oxidative stress is a well-established factor in the initiation of cancer [148,149].
Conversely, once cancer has developed, the effects of antioxidants can become paradoxical [150,151]. High doses of AA have been reported to exhibit various antitumor effects, including the proteolysis of hypoxia-inducible factor alpha (HIFα), epigenetic regulation, and a pro-oxidant effect. This oxidative-promoting action at elevated concentrations can be detrimental to cancer cells, as noted by Cockfield and Schafer [152], who discuss the context-specific vulnerabilities of cancer cells to antioxidant defenses. Furthermore, by reducing oxidative stress, antioxidants may inadvertently protect cancer cells from damage caused by reactive oxygen species, which could otherwise be therapeutically employed to induce cancer cell death. This protective mechanism may facilitate tumor progression and confer resistance to certain cancer treatments. Thus, while antioxidants are beneficial in the prevention of cancer, their role in established cancers is complex and may vary based on the context and specific cancer type, necessitating further investigation beyond just brain cancers.
Interestingly, Levine et al. highlight that AA can deliver hydrogen peroxide (H2O2) to tumor cells and participate in Fenton reactions involving redox-active intracellular iron, leading to oxidative damage [153]. In glioblastoma, a Phase 2 clinical trial involving pharmacologic ascorbate combined with chemoradiation demonstrated a significant increase in median overall survival, rising from 14.6 months in historical controls to 19.6 months in the trial cohort [154]. This evidence underscores the nuanced role of vitamin C in cancer therapy and the need for tailored approaches in its application.
Table 1. Studies accounting for pharmacological strategies against RIBI.
Table 1. Studies accounting for pharmacological strategies against RIBI.
Details of the StudyModelGroupsIrradiation ProcedureDrug TestedCognitive TestingOther EvaluationsResultsRef.
MitoQKMDivided into four groups, 10 mice each:
1. G1: ip PSS 0.9% for 3 days
2. G2: ip MitoQ
3. G3: WBI
4. G4: ip MitoQ + WBI
WBI of mice was performed using a high-LET 56-Fe ions beams at the energy of 160 MeV/μ. Each mouse received 2 Gy doses at a dose rate of 0.5 Gy/min, and the mice were placed in the plateau region. MitoQ groups received MitoQ (5 mg/kg/day) for 3 days (-)-Determination of oxidative stress parameters
(PCO, MDA, SOD, CAT)
-Mitochondrial respiration measurements (O2 consumption and RCR)
-Measurement of mitochondria-generated ROS
-mtDNA damage assay
-mitochondrial dynamics protein (Mfn2, Drp1, bcl-2, bax, cyto c)
-Gene expression analysis (BA; Casp3; SOD2; Opa1)
MitoQ reduced radiation-induced oxidative stress with decreased lipid peroxidation and reduced protein and DNA oxidation. MitoQ protected mitochondrial respiration after RT. MitoQ increased Mfn2 and OPA1 and decreased Drp1.
MitoQ also suppressed mitochondrial DNA damage, cyto c release, and caspase-3 activity in RT-treated mice compared to the control group [152].
[155]
QuercetinWARDivided into 4 groups (n = 8/each):
1. control group
2. G QUER: quercetin
3. G RAD was given only irradiation
4. G RAD + QUER: quercetin + irradiation
RAD groups were
subjected to cranium irradiation with a single dose of 20 Gy of photons using a 6 MV LINAC at a dose rate of ~1 Gy/min, with the source–axis distance technique, with 1.0 cm of bolus material on the surface.
QUER groups received
Quercetin 50 mg/kg body weight (BW) daily in distilled water and 0.25 mL PS for 15 days.
(-)-Total antioxidant status and MDA
-Brain histopathological evaluation
Tissue samples and biochemical levels of tissue-injury markers in the four groups were compared. In all measured parameters of oxidative stress, administration of quercetin significantly demonstrated favorable effects. Both plasma and tissue levels of MDA and total antioxidant status significantly changed in favor of antioxidant activity. Histopathological evaluation of the tissues also demonstrated a significant decrease in cellular degeneration and infiltration parameters after quercetin administration. Quercetin demonstrated significant neuroprotection after radiation-induced brain injury.[156]
Date syrup WARDivided into 4 groups, 15 rats each.
1. G1 (Control); received 1 mL 0.9% saline solution orally
for 4 weeks and served as control; 2. G2 (Irradiated); was exposed to radiation at a dose level of 6 Gy and
sacrificed after 48 h.
3. G3 (Date syrup);
4. G4 (Irradiated + Date syrup)
Whole-body gamma-irradiation. Animals were irradiated at an acute
Single-dose level of 6 Gy delivered at a dose rate of 0.713 rad/s.
Date syrup group received daily date syrup by stomach intubation at a dose of 4 mL/kg body weight for 4 weeks. (-)-Serum biochemical analysis.
-Assessment of oxidant/antioxidant biomarkers (lipid peroxidation, DNA damage, GSH, CAT activity
-Assessment of MMP-9
-q RT-PCR evaluation for TNF-α gene expression
-Liver histopathological examination
Pretreatment of rats with Date syrup ameliorated the tissue damage induced by radiation as evidenced
by the improvement in liver function, antioxidant status and reduction in DNA damage. Moreover, liver
TNF-α expression and serum MMP-9 activity were reduced.
[157]
NSI-189LERDivided into 3 groups (n = 15–16/each):
1. controls receiving oral gavage (vehicle only) and sham irradiation
2. cohorts receiving oral gavage (vehicle only) and 27 Gy head-only fractionated exposure
3. cohorts receiving oral gavage (NSI-189, 30 mg/kg) and 27 Gy head-only fractionated exposure
For CI, animals were positioned under a collimated (1 cm2 diameter) beam for head-only irradiation delivered at a dose rate of 1 Gy/min. Fractionation of 27 Gy was delivered over 3 separate doses of 8.67 Gy, which were administered 48 h apart.NSI 189
The drug was administered by daily oral gavage at a concentration adjusted to the weight of the animals. The daily dosing was set at 2 mL/kg, setting the target daily dose of 30 mg/kg. Thus, the daily volume of the drug typically varied between 0.6 and 1.0 mL/rat.
cognitive testing 1 week after termination of oral gavage (5 weeks post-RT). Cognitive testing was performed over the course of three weeks and included four different spontaneous exploration tasks (novel place recognition, novel object recognition, object in place and temporal order) followed by contextual and cued fear conditioning-Assessment of neurogenesis
-Determination of hippocampal volume
-Assessment of activated microglia
NSI-189 treatment resulted in significantly improved performance in four of these tasks: novel-place recognition, novel-object recognition, object in place and temporal order. In addition, there was a trend for improved performance in the contextual phase of the fear-conditioning task. Importantly, enhanced cognition in the NSI-189-treated cohort was found to persist one month after the cessation of drug treatment. These neurocognitive benefits of NSI-189 coincided with a significant increase in neurogenesis and a significant decrease in the numbers of activated microglia compared to the irradiated cohort that was given the vehicle alone.[158]
FingolimodCMDivided into 4 groups:
1. G1: methylcellulose vehicle alone
2. G2: vehicle + radiation
3. G3: FTY720
4. G4: FTY720 + radiation
For irradiation, a Gammacell 40 irradiator with a dose rate of 95. cGy/minute was used. A single dose of 7 Gy was administered to each animal.FTY720 groups received three ip of 0.5 mg/kg FTY720 in the week prior to irradiation.
They then received 3 ip/week of vehicle or 0.5 mg/kg FTY720 for 6 weeks.
Fear conditioning and MWM were then employed to test learning and memory. -IF and IHC of brain tissue (antibodies: anti S1PR1, nestin, GFAP, doublecortin, NeuN, Tubulin III/Tuj1)
-qRT-PCR of BDNF vs. B2
The learning deficits were fully restored by FTY720. In irradiated brains, FTY720 maintained the cytoarchitecture of the dentate gyrus granular cell layer and partially restored the pool of NPC. In mice harboring BTSC xenografts, FTY720 delayed tumor growth and improved survival.[159]
mNGFSDRDivided into 3 groups:
G1: control (n = 15)
G2: mNGF + CI (n = 20)
G3: PSS + CI (n = 20)
CI at a single dose of 12 Gy by X-ray.?MWM experimentEB leakage of the brain, and expressions of neuN, vWF, ZO-1 in hippocampus by immunofluorescence, and expressions of neuN, vWF, ZO-1, VEGF and GFAP in hippocampus by WB mNGF decreases the damage by RT, improving the latency time of escape in the Morris water maze, and decreases the EB leakage.
In the IF, mNGF increases the expression of neunN, vWF abd ZO-1. In WB, mNGF increases the expression of neuN, vWF and ZO-1.
[160]
Kukoamine (KuA)WARDivided into 5 groups (n = 5–8/group):
1. G1: sham irradiation
2. G2: CI
3. G3: CI + KuA low dose
4. G4: CI + KuA middle dose
5. G5: CI + KuA high dose
CI was performed
with 6-MeV electron beams delivered by a LINAC. Irradiated rats received a single dose of 30 Gy X-rays at a
dose rate of 250 cGy/min.
KuA was administered
at a dose of 5, (G3) 10 (G4) and 20 mg/kg (G5) body weight.
(-)-MDA, GSH level and SOD, CAT activity assays
-Nissl Staining and TUNEL staining
-WB, using antibodies anti: BDNF, Casp3, CytC, Bax, Bcl2, GAPDH, BA
Whole brain irradiation led to the neuronal abnormality and
it was alleviated by KuA. KuA decreased MDA level, increased GSH level, SOD and CAT activities, as well
as alleviated neuronal apoptosis by regulating the expression of cleaved caspase-3, cytochrome C, Bax and Bcl2. Additionally, KuA increased the expression of BDNF.
[15]
AcanthopanaxKMDivided into 3 large groups (n = 32 each)
G1: behavioral test
G2: pathological sections
G3: metabolomics analysis.
Each large group was divided into 4 small groups for the experiments (n = 8 per group)
g1: normal control
g2: model set (CI)
g3: treatment group AS + CI
g4: treatment group V + CI
Irradiated by 60 Co-γ ray irradiation with the mean LET of 62.2 KeV/μm at a dose of 4 Gy and a dose rate of 0.1 Gy/min.AS was administered at a dose of 235.7 mg/kg/day.
V was administered at a dose of 13.75 mg/kg/day.
MWM and sucrose preference test-Production of pathological sections for brain tissues (PFC)
-Metabolomics analysis based on 1H NMR
AS significantly improved the decline of low LET-induced learning ability and spatial memory capacity, increased the sensitivity of the nervous system and, to a certain degree, prevented brain tissue lesions caused by radiation. In our study, we also observed that AS had a better effect on brain tissue development and brain–glutamate-cycle balance compared with a chemical drug (Venlafaxine).[161]
NOTES: WAR: Wistar albino rats; LER: Long–Evans rats; KM: Kunming mice; MDA: malondialdehyde; WBI: whole body irradiation; LINAC: linear accelerator; ip: intraperitoneal injection; PSS: physiological saline solution; G: group; PCO: protein carbonyl; SOD: superoxide dismutase; CAT: catalase; RCR: respiratory control ratio; CM: C57/Bl/6J mice; BTSC: brain tumor stem cell; IF: Immunofluorescence; IHC: immunohistochemistry; SDR: Sprague Dawley rat; CI: cranial irradiation; EB: Evans blue; WB: Western blot; mNGF: mouse nerve growth factor; BA: beta-actin; MWM: Morris water maze; V: venlafaxine; AS: acanthopanax senticosus; PFC: prefrontal cortex.

6. Discussion and Conclusions

In conclusion, emerging evidence highlights the protective role of AA and other compounds such as memantine in mitigating RIBI. AA’s potent antioxidative properties can significantly reduce cellular damage caused by ionizing radiation. Notably, the lipophilic derivative 6-o-palmitoylascorbate has shown greater efficacy than standard ascorbate in minimizing X-ray-induced DNA damage, lipid peroxidation, and protein carbonylation in human lymphocytes. This derivative enhances cell viability and preserves essential antioxidants like glutathione, providing substantial protection against oxidative stress from radiation exposure.
Moreover, clinical studies demonstrate AA’s ability to substantially decrease DNA double-strand breaks associated with radiation. For instance, a randomized trial revealed an impressive 87% reduction in double-strand breaks in patients pre-treated with AA before high-dose radiation procedures. Similar results were observed with oral AA administration prior to CT scans, resulting in a 61% decrease in the mean increase of γ-H2AX foci.
While the role of MAPK-mediated signaling in RIBI is under investigation, the specific impact of antioxidants like AA and other drugs remains largely unexplored. Concerns about potential detrimental effects in anticancer therapies exist; however, recent research indicates that AA does not compromise the efficacy of radiotherapy. There is an urgent need for comprehensive studies to elucidate the relationship between AA and standard pharmacological treatments in clinical settings aimed at preventing RIBI.
Incorporating AA or memantine into pre-radiation protocols could prove invaluable for reducing RIBI risk and enhancing patient outcomes. Future clinical trials should focus on evidence-based assessments, including neurocognitive tests, to evaluate the effectiveness of antioxidant interventions and develop more effective strategies for managing RIBI.

Author Contributions

L.G.-J.: hypothesis design; L.G.-J. and C.R.-S.: scientific study rationale; L.G.-J., C.R.-S. and V.P.-G.: pharmacological bases; A.F., G.F. and G.Z.: clinical aspects; C.R.-S. and L.G.-J.: image design; L.G.-J., V.P.-G., C.R.-S., G.F., G.Z. and A.F.: scientific discussion. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No datasets were generated or analyzed during the current study.

Acknowledgments

This research project was presented in the Medical Science Summer School Oncology for Medical Students 2018; Groningen, The Netherlands by L.G.-J.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

AAascorbic acid
AQP4aquaporin-4
BMsbrain metastases
CATcatalase
CDcell death
CNScentral nervous system
CSFcerebrospinal fluid
DDRDNA damage response
DHAdehydroascorbic acid
DSBdouble-strand break
ERKextracellular-signal-regulated kinase
FRsfree radicals
GPxglutathione peroxidase
GSHreduced glutathione
HRhomologous recombination
IAPintrinsic apoptotic pathway
JNKc-Jun N-terminal kinase
LQlinear quadratic
MAPKmitogen-activated protein kinase
NHJEnon-homologous end-joining
NMDARN-methyl-D-aspartate receptor
RIBIradiation-induced brain injury
ROSreactive oxygen species
RTradiotherapy
SODsuperoxide dismutase
SSBsingle-strand break
TMEtumor microenvironment
WBRTwhole brain radiation therapy

References

  1. Rahman, R.; Sulman, E.; Haas-Kogan, D.; Cagney, D.N. Update on Radiation Therapy for Central Nervous System Tumors. Hematol. Oncol. Clin. N. Am. 2022, 36, 77–93. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, X.; Zhang, W.; Cao, W.-D.; Cheng, G.; Liu, B.; Cheng, J. A Review of Current Management of Brain Metastases. Ann. Surg. Oncol. 2012, 19, 1043–1050. [Google Scholar] [CrossRef] [PubMed]
  3. Wefel, J.S.; Parsons, M.W.; Gondi, V.; Brown, P.D. Neurocognitive Aspects of Brain Metastasis. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2018; pp. 155–165. [Google Scholar]
  4. Khuntia, D.; Brown, P.; Li, J.; Mehta, M.P. Whole-Brain Radiotherapy in the Management of Brain Metastasis. J. Clin. Oncol. 2006, 24, 1295–1304. [Google Scholar] [CrossRef]
  5. Gondi, V.; Bauman, G.; Bradfield, L.; Burri, S.H.; Cabrera, A.R.; Cunningham, D.A.; Eaton, B.R.; Hattangadi-Gluth, J.A.; Kim, M.M.; Kotecha, R.; et al. Radiation Therapy for Brain Metastases: An ASTRO Clinical Practice Guideline. Pract. Radiat. Oncol. 2022, 12, 265–282. [Google Scholar] [CrossRef]
  6. Rades, D.; Bohlen, G.; Lohynska, R.; Veninga, T.; Stalpers, L.J.A.; Schild, S.E.; Dunst, J. Whole-Brain Radiotherapy with 20 Gy in 5 Fractions for Brain Metastases in Patients with Cancer of Unknown Primary (CUP). Strahlenther. Onkol. 2007, 183, 631–636. [Google Scholar] [CrossRef] [PubMed]
  7. Li, J.; Bentzen, S.M.; Renschler, M.; Mehta, M.P. Regression after Whole-Brain Radiation Therapy for Brain Metastases Correlates with Survival and Improved Neurocognitive Function. J. Clin. Oncol. 2007, 25, 1260–1266. [Google Scholar] [CrossRef]
  8. Achrol, A.S.; Rennert, R.C.; Anders, C.; Soffietti, R.; Ahluwalia, M.S.; Nayak, L.; Peters, S.; Arvold, N.D.; Harsh, G.R.; Steeg, P.S.; et al. Brain Metastases. Nat. Rev. Dis. Primer 2019, 5, 5. [Google Scholar] [CrossRef]
  9. Gondi, V.; Tomé, W.A.; Mehta, M.P. Why Avoid the Hippocampus? A Comprehensive Review. Radiother. Oncol. 2010, 97, 370–376. [Google Scholar] [CrossRef]
  10. Brown, P.D.; Jaeckle, K.; Ballman, K.V.; Farace, E.; Cerhan, J.H.; Anderson, S.K.; Carrero, X.W.; Barker, F.G.; Deming, R.; Burri, S.H.; et al. Effect of Radiosurgery Alone vs Radiosurgery With Whole Brain Radiation Therapy on Cognitive Function in Patients With 1 to 3 Brain Metastases: A Randomized Clinical Trial. JAMA 2016, 316, 401–409. [Google Scholar] [CrossRef]
  11. Brown, P.D.; Ballman, K.V.; Cerhan, J.H.; Anderson, S.K.; Carrero, X.W.; Whitton, A.C.; Greenspoon, J.; Parney, I.F.; Laack, N.N.I.; Ashman, J.B.; et al. Postoperative Stereotactic Radiosurgery Compared with Whole Brain Radiotherapy for Resected Metastatic Brain Disease (NCCTG N107C/CEC·3): A Multicentre, Randomised, Controlled, Phase 3 Trial. Lancet Oncol. 2017, 18, 1049–1060. [Google Scholar] [CrossRef]
  12. Chang, E.L.; Wefel, J.S.; Hess, K.R.; Allen, P.K.; Lang, F.F.; Kornguth, D.G.; Arbuckle, R.B.; Swint, J.M.; Shiu, A.S.; Maor, M.H.; et al. Neurocognition in Patients with Brain Metastases Treated with Radiosurgery or Radiosurgery plus Whole-Brain Irradiation: A Randomised Controlled Trial. Lancet Oncol. 2009, 10, 1037–1044. [Google Scholar] [CrossRef] [PubMed]
  13. Gondi, V.; Pugh, S.L.; Tome, W.A.; Caine, C.; Corn, B.; Kanner, A.; Rowley, H.; Kundapur, V.; DeNittis, A.; Greenspoon, J.N.; et al. Preservation of Memory with Conformal Avoidance of the Hippocampal Neural Stem-Cell Compartment during Whole-Brain Radiotherapy for Brain Metastases (RTOG 0933): A Phase II Multi-Institutional Trial. J. Clin. Oncol. 2014, 32, 3810–3816. [Google Scholar] [CrossRef] [PubMed]
  14. Tsao, M.N.; Xu, W.; Wong, R.K.; Lloyd, N.; Laperriere, N.; Sahgal, A.; Rakovitch, E.; Chow, E. Whole Brain Radiotherapy for the Treatment of Newly Diagnosed Multiple Brain Metastases. Cochrane Database Syst. Rev. 2018, 1, CD003869. [Google Scholar] [CrossRef]
  15. Zhang, Y.; Cheng, Z.; Wang, C.; Ma, H.; Meng, W.; Zhao, Q. Neuroprotective Effects of Kukoamine a against Radiation-Induced Rat Brain Injury through Inhibition of Oxidative Stress and Neuronal Apoptosis. Neurochem. Res. 2016, 41, 2549–2558. [Google Scholar] [CrossRef]
  16. Warrington, J.P.; Ashpole, N.; Csiszar, A.; Lee, Y.W.; Ungvari, Z.; Sonntag, W.E. Whole Brain Radiation-Induced Vascular Cognitive Impairment: Mechanisms and Implications. J. Vasc. Res. 2013, 50, 445–457. [Google Scholar] [CrossRef]
  17. Yang, L.; Yang, J.; Li, G.; Li, Y.; Wu, R.; Cheng, J.; Tang, Y. Pathophysiological Responses in Rat and Mouse Models of Radiation-Induced Brain Injury. Mol. Neurobiol. 2017, 54, 1022–1032. [Google Scholar] [CrossRef] [PubMed]
  18. Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The Tumour Microenvironment after Radiotherapy: Mechanisms of Resistance and Recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
  19. Steel, G.G.; McMillan, T.J.; Peacock, J.H. The 5Rs of Radiobiology. Int. J. Radiat. Biol. 1989, 56, 1045–1048. [Google Scholar] [CrossRef]
  20. Fowler, J.F. Review: Total Doses in Fractionated Radiotherapy--Implications of New Radiobiological Data. Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 1984, 46, 103–120. [Google Scholar] [CrossRef]
  21. Santacroce, A.; Kamp, M.A.; Budach, W.; Hänggi, D. Radiobiology of Radiosurgery for the Central Nervous System. BioMed Res. Int. 2013, 2013, 362761. [Google Scholar] [CrossRef]
  22. Sminia, P.; Guipaud, O.; Viktorsson, K.; Ahire, V.; Baatout, S.; Boterberg, T.; Cizkova, J.; Dostál, M.; Fernandez-Palomo, C.; Filipova, A.; et al. Clinical Radiobiology for Radiation Oncology. In Radiobiology Textbook; Baatout, S., Ed.; Springer International Publishing: Cham, Switzerland, 2023; pp. 237–309. ISBN 978-3-031-18810-7. [Google Scholar]
  23. Shiloh, Y. ATM and Related Protein Kinases: Safeguarding Genome Integrity. Nat. Rev. Cancer 2003, 3, 155–168. [Google Scholar] [CrossRef] [PubMed]
  24. Abraham, R.T. Cell Cycle Checkpoint Signaling through the ATM and ATR Kinases. Genes Dev. 2001, 15, 2177–2196. [Google Scholar] [CrossRef] [PubMed]
  25. Maier, P.; Hartmann, L.; Wenz, F.; Herskind, C. Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int. J. Mol. Sci. 2016, 17, 102. [Google Scholar] [CrossRef] [PubMed]
  26. Lukas, J.; Lukas, C.; Bartek, J. Mammalian Cell Cycle Checkpoints: Signalling Pathways and Their Organization in Space and Time. DNA Repair 2004, 3, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  27. Sia, J.; Szmyd, R.; Hau, E.; Gee, H.E. Molecular Mechanisms of Radiation-Induced Cancer Cell Death: A Primer. Front. Cell Dev. Biol. 2020, 8, 41. [Google Scholar] [CrossRef]
  28. Helton, E.S.; Chen, X. P53 Modulation of the DNA Damage Response. J. Cell. Biochem. 2007, 100, 883–896. [Google Scholar] [CrossRef]
  29. Abend, M. Reasons to Reconsider the Significance of Apoptosis for Cancer Therapy. Int. J. Radiat. Biol. 2003, 79, 927–941. [Google Scholar] [CrossRef]
  30. Shinomiya, N. New Concepts in Radiation-Induced Apoptosis: “premitotic Apoptosis” and “Postmitotic Apoptosis. J. Cell. Mol. Med. 2001, 5, 240–253. [Google Scholar] [CrossRef]
  31. Eriksson, D.; Stigbrand, T. Radiation-Induced Cell Death Mechanisms. Tumour Biol. 2010, 31, 363–372. [Google Scholar] [CrossRef]
  32. Igney, F.H.; Krammer, P.H. Death and Anti-Death: Tumour Resistance to Apoptosis. Nat. Rev. Cancer 2002, 2, 277–288. [Google Scholar] [CrossRef]
  33. Scaffidi, C.; Fulda, S.; Srinivasan, A.; Friesen, C.; Li, F.; Tomaselli, K.J.; Debatin, K.M.; Krammer, P.H.; Peter, M.E. Two CD95 (APO-1/Fas) Signaling Pathways. EMBO J. 1998, 17, 1675–1687. [Google Scholar] [CrossRef] [PubMed]
  34. Kastan, M. On the TRAIL from P53 to Apoptosis? Nat. Genet. 1997, 17, 130–131. [Google Scholar] [CrossRef] [PubMed]
  35. Jin, Z.; El-Deiry, W.S. Overview of Cell Death Signaling Pathways. Cancer Biol. Ther. 2005, 4, 147–171. [Google Scholar] [CrossRef] [PubMed]
  36. Galluzzi, L.; Maiuri, M.C.; Vitale, I.; Zischka, H.; Castedo, M.; Zitvogel, L.; Kroemer, G. Cell Death Modalities: Classification and Pathophysiological Implications. Cell Death Differ. 2007, 14, 1237–1243. [Google Scholar] [CrossRef]
  37. Roninson, I.B.; Broude, E.V.; Chang, B.D. If Not Apoptosis, Then What? Treatment-Induced Senescence and Mitotic Catastrophe in Tumor Cells. Drug Resist. Updat. Rev. Comment. Antimicrob. Anticancer Chemother. 2001, 4, 303–313. [Google Scholar] [CrossRef]
  38. Ruth, A.C.; Roninson, I.B. Effects of the Multidrug Transporter P-Glycoprotein on Cellular Responses to Ionizing Radiation. Cancer Res. 2000, 60, 2576–2578. [Google Scholar]
  39. Weaver, B.A.A.; Cleveland, D.W. Decoding the Links between Mitosis, Cancer, and Chemotherapy: The Mitotic Checkpoint, Adaptation, and Cell Death. Cancer Cell 2005, 8, 7–12. [Google Scholar] [CrossRef]
  40. Yamada, H.Y.; Gorbsky, G.J. Spindle Checkpoint Function and Cellular Sensitivity to Antimitotic Drugs. Mol. Cancer Ther. 2006, 5, 2963–2969. [Google Scholar] [CrossRef]
  41. Ianzini, F.; Bertoldo, A.; Kosmacek, E.A.; Phillips, S.L.; Mackey, M.A. Lack of P53 Function Promotes Radiation-Induced Mitotic Catastrophe in Mouse Embryonic Fibroblast Cells. Cancer Cell Int. 2006, 6, 11. [Google Scholar] [CrossRef]
  42. Eriksson, D.; Löfroth, P.-O.; Johansson, L.; Riklund, K.A.; Stigbrand, T. Cell Cycle Disturbances and Mitotic Catastrophes in HeLa Hep2 Cells Following 2.5 to 10 Gy of Ionizing Radiation. Clin. Cancer Res. 2007, 13, 5501s–5508s. [Google Scholar] [CrossRef]
  43. Somosy, Z. Radiation Response of Cell Organelles. Micron 2000, 31, 165–181. [Google Scholar] [CrossRef] [PubMed]
  44. Castedo, M.; Kroemer, G. Mitotic catastrophe: A special case of apoptosis. J. Soc. Biol. 2004, 198, 97–103. [Google Scholar] [CrossRef] [PubMed]
  45. Hayflick, L.; Moorhead, P.S. The Serial Cultivation of Human Diploid Cell Strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  46. Stein, G.H.; Dulić, V. Origins of G1 Arrest in Senescent Human Fibroblasts. BioEssays News Rev. Mol. Cell. Dev. Biol. 1995, 17, 537–543. [Google Scholar] [CrossRef]
  47. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
  48. Novakova, Z.; Hubackova, S.; Kosar, M.; Janderova-Rossmeislova, L.; Dobrovolna, J.; Vasicova, P.; Vancurova, M.; Horejsi, Z.; Hozak, P.; Bartek, J.; et al. Cytokine Expression and Signaling in Drug-Induced Cellular Senescence. Oncogene 2010, 29, 273–284. [Google Scholar] [CrossRef] [PubMed]
  49. Fuchs-Tarlovsky, V. Role of Antioxidants in Cancer Therapy. Nutr. Burbank Los Angel. Cty. Calif 2013, 29, 15–21. [Google Scholar] [CrossRef] [PubMed]
  50. Shirazi, A.; Mihandoost, E.; Ghobadi, G.; Mohseni, M.; Ghazi-khansari, M. Evaluation of Radio-Protective Effect of Melatonin on Whole Body Irradiation Induced Liver Tissue Damage. Cell J. Yakhteh 2013, 14, 292–297. [Google Scholar]
  51. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C.S. Oxidative Stress and Metabolic Disorders: Pathogenesis and Therapeutic Strategies. Life Sci. 2016, 148, 183–193. [Google Scholar] [CrossRef]
  52. Pinilla-González, V.; Montecinos-Barrientos, B.; Martin-Kommer, C.; Chichiarelli, S.; Saso, L.; Rodrigo, R. Exploring Antioxidant Strategies in the Pathogenesis of ALS. Open Life Sci. 2024, 19, 20220842. [Google Scholar] [CrossRef]
  53. Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging Hallmarks and the Role of Oxidative Stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef] [PubMed]
  54. Ionescu-Tucker, A.; Cotman, C.W. Emerging Roles of Oxidative Stress in Brain Aging and Alzheimer’s Disease. Neurobiol. Aging 2021, 107, 86–95. [Google Scholar] [CrossRef] [PubMed]
  55. Kirkman, H.N.; Gaetani, G.F. Mammalian Catalase: A Venerable Enzyme with New Mysteries. Trends Biochem. Sci. 2007, 32, 44–50. [Google Scholar] [CrossRef] [PubMed]
  56. Baxter, P.S.; Hardingham, G.E. Adaptive Regulation of the Brain’s Antioxidant Defences by Neurons and Astrocytes. Free Radic. Biol. Med. 2016, 100, 147–152. [Google Scholar] [CrossRef]
  57. Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 Reasons Why the Brain Is Susceptible to Oxidative Stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef]
  58. Magistretti, P.J.; Allaman, I. A Cellular Perspective on Brain Energy Metabolism and Functional Imaging. Neuron 2015, 86, 883–901. [Google Scholar] [CrossRef]
  59. Goyal, M.S.; Hawrylycz, M.; Miller, J.A.; Snyder, A.Z.; Raichle, M.E. Aerobic Glycolysis in the Human Brain Is Associated with Development and Neotenous Gene Expression. Cell Metab. 2014, 19, 49–57. [Google Scholar] [CrossRef]
  60. Ren, X.; Zou, L.; Zhang, X.; Branco, V.; Wang, J.; Carvalho, C.; Holmgren, A.; Lu, J. Redox Signaling Mediated by Thioredoxin and Glutathione Systems in the Central Nervous System. Antioxid. Redox Signal. 2017, 27, 989–1010. [Google Scholar] [CrossRef]
  61. Que, E.L.; Domaille, D.W.; Chang, C.J. Metals in Neurobiology: Probing Their Chemistry and Biology with Molecular Imaging. Chem. Rev. 2008, 108, 1517–1549. [Google Scholar] [CrossRef]
  62. Edmondson, D.E. Hydrogen Peroxide Produced by Mitochondrial Monoamine Oxidase Catalysis: Biological Implications. Curr. Pharm. Des. 2014, 20, 155–160. [Google Scholar] [CrossRef]
  63. Bazinet, R.P.; Layé, S. Polyunsaturated Fatty Acids and Their Metabolites in Brain Function and Disease. Nat. Rev. Neurosci. 2014, 15, 771–785. [Google Scholar] [CrossRef] [PubMed]
  64. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  65. Burns, T.C.; Awad, A.J.; Li, M.D.; Grant, G.A. Radiation-Induced Brain Injury: Low-Hanging Fruit for Neuroregeneration. Neurosurg. Focus 2016, 40, E3. [Google Scholar] [CrossRef] [PubMed]
  66. Sotomayor, C.G.; González, C.; Soto, M.; Moreno-Bertero, N.; Opazo, C.; Ramos, B.; Espinoza, G.; Sanhueza, Á.; Cárdenas, G.; Yévenes, S.; et al. Ionizing Radiation-Induced Oxidative Stress in Computed Tomography-Effect of Vitamin C on Prevention of DNA Damage: PREVIR-C Randomized Controlled Trial Study Protocol. J. Clin. Med. 2024, 13, 3866. [Google Scholar] [CrossRef]
  67. Azzam, E.I.; Jay-Gerin, J.-P.; Pain, D. Ionizing Radiation-Induced Metabolic Oxidative Stress and Prolonged Cell Injury. Cancer Lett. 2012, 327, 48–60. [Google Scholar] [CrossRef]
  68. Benhar, M.; Engelberg, D.; Levitzki, A. ROS, Stress-Activated Kinases and Stress Signaling in Cancer. EMBO Rep. 2002, 3, 420–425. [Google Scholar] [CrossRef] [PubMed]
  69. Son, Y.; Cheong, Y.-K.; Kim, N.-H.; Chung, H.-T.; Kang, D.G.; Pae, H.-O. Mitogen-Activated Protein Kinases and Reactive Oxygen Species: How Can ROS Activate MAPK Pathways? J. Signal Transduct. 2011, 2011, 792639. [Google Scholar] [CrossRef]
  70. Selim, K.A.; Abdelrasoul, H.; Aboelmagd, M.; Tawila, A.M. The Role of the MAPK Signaling, Topoisomerase and Dietary Bioactives in Controlling Cancer Incidence. Diseases 2017, 5, 13. [Google Scholar] [CrossRef]
  71. Dai, H.-L.; Hu, W.-Y.; Jiang, L.-H.; Li, L.; Gaung, X.-F.; Xiao, Z.-C. P38 MAPK Inhibition Improves Synaptic Plasticity and Memory in Angiotensin II-Dependent Hypertensive Mice. Sci. Rep. 2016, 6, 27600. [Google Scholar] [CrossRef]
  72. He, L.; Deng, Y.; Gao, J.; Zeng, L.; Gong, Q. Icariside II Ameliorates Ibotenic Acid-Induced Cognitive Impairment and Apoptotic Response via Modulation of MAPK Pathway in Rats. Phytomed. Int. J. Phytother. Phytopharm. 2018, 41, 74–81. [Google Scholar] [CrossRef]
  73. Zhang, W.; Liu, H.T. MAPK Signal Pathways in the Regulation of Cell Proliferation in Mammalian Cells. Cell Res. 2002, 12, 9–18. [Google Scholar] [CrossRef] [PubMed]
  74. Gaitanaki, C.; Papatriantafyllou, M.; Stathopoulou, K.; Beis, I. Effects of Various Oxidants and Antioxidants on the P38-MAPK Signalling Pathway in the Perfused Amphibian Heart. Mol. Cell. Biochem. 2006, 291, 107–117. [Google Scholar] [CrossRef]
  75. Chiarini, A.; Dal Pra, I.; Marconi, M.; Chakravarthy, B.; Whitfield, J.F.; Armato, U. Calcium-Sensing Receptor (CaSR) in Human Brain’s Pathophysiology: Roles in Late-Onset Alzheimer’s Disease (LOAD). Curr. Pharm. Biotechnol. 2009, 10, 317–326. [Google Scholar] [CrossRef]
  76. Dent, P.; Yacoub, A.; Fisher, P.B.; Hagan, M.P.; Grant, S. MAPK Pathways in Radiation Responses. Oncogene 2003, 22, 5885–5896. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, E.K.; Choi, E.-J. Pathological Roles of MAPK Signaling Pathways in Human Diseases. Biochim. Biophys. Acta 2010, 1802, 396–405. [Google Scholar] [CrossRef]
  78. Deng, Z.; Sui, G.; Rosa, P.M.; Zhao, W. Radiation-Induced c-Jun Activation Depends on MEK1-ERK1/2 Signaling Pathway in Microglial Cells. PLoS ONE 2012, 7, e36739. [Google Scholar] [CrossRef] [PubMed]
  79. Ruffels, J.; Griffin, M.; Dickenson, J.M. Activation of ERK1/2, JNK and PKB by Hydrogen Peroxide in Human SH-SY5Y Neuroblastoma Cells: Role of ERK1/2 in H2O2-Induced Cell Death. Eur. J. Pharmacol. 2004, 483, 163–173. [Google Scholar] [CrossRef]
  80. Dabrowski, A.; Boguslowicz, C.; Dabrowska, M.; Tribillo, I.; Gabryelewicz, A. Reactive Oxygen Species Activate Mitogen-Activated Protein Kinases in Pancreatic Acinar Cells. Pancreas 2000, 21, 376–384. [Google Scholar] [CrossRef]
  81. Zomosa, G.; Lühr, C.; Bova, F.; González-Johnson, L.; Rojas-Solé, C.; Troncoso, L.; Miranda, G.; Lorenzoni, J.; Zomosa, G.; Lühr, C.; et al. Radiosurgery for Intracranial Meningiomas. In Meningioma—The Essentials From Bench to Bedside; IntechOpen: London, UK, 2024; ISBN 978-0-85466-145-9. [Google Scholar]
  82. Wang, Y.; Tian, J.; Liu, D.; Li, T.; Mao, Y.; Zhu, C. Microglia in Radiation-Induced Brain Injury: Cellular and Molecular Mechanisms and Therapeutic Potential. CNS Neurosci. Ther. 2024, 30, e14794. [Google Scholar] [CrossRef]
  83. Boyd, A.; Byrne, S.; Middleton, R.J.; Banati, R.B.; Liu, G.-J. Control of Neuroinflammation through Radiation-Induced Microglial Changes. Cells 2021, 10, 2381. [Google Scholar] [CrossRef]
  84. Schmal, Z.; Rübe, C.E. Region-Specific Effects of Fractionated Low-Dose Versus Single-Dose Radiation on Hippocampal Neurogenesis and Neuroinflammation. Cancers 2022, 14, 5477. [Google Scholar] [CrossRef] [PubMed]
  85. Markou, A.; Kitchen, P.; Aldabbagh, A.; Repici, M.; Salman, M.M.; Bill, R.M.; Balklava, Z. Mechanisms of Aquaporin-4 Vesicular Trafficking in Mammalian Cells. J. Neurochem. 2024, 168, 100–114. [Google Scholar] [CrossRef] [PubMed]
  86. Mader, S.; Brimberg, L. Aquaporin-4 Water Channel in the Brain and Its Implication for Health and Disease. Cells 2019, 8, 90. [Google Scholar] [CrossRef]
  87. Kitchen, P.; Salman, M.M.; Halsey, A.M.; Clarke-Bland, C.; MacDonald, J.A.; Ishida, H.; Vogel, H.J.; Almutiri, S.; Logan, A.; Kreida, S.; et al. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell 2020, 181, 784–799.e19. [Google Scholar] [CrossRef]
  88. Sylvain, N.J.; Salman, M.M.; Pushie, M.J.; Hou, H.; Meher, V.; Herlo, R.; Peeling, L.; Kelly, M.E. The Effects of Trifluoperazine on Brain Edema, Aquaporin-4 Expression and Metabolic Markers during the Acute Phase of Stroke Using Photothrombotic Mouse Model. Biochim. Biophys. Acta Biomembr. 2021, 1863, 183573. [Google Scholar] [CrossRef] [PubMed]
  89. Li, W.; Tan, C.; Liu, Y.; Liu, X.; Wang, X.; Gui, Y.; Qin, L.; Deng, F.; Yu, Z.; Hu, C.; et al. Resveratrol Ameliorates Oxidative Stress and Inhibits Aquaporin 4 Expression Following Rat Cerebral Ischemia-Reperfusion Injury. Mol. Med. Rep. 2015, 12, 7756–7762. [Google Scholar] [CrossRef]
  90. Alhadidi, Q.M.; Bahader, G.A.; Arvola, O.; Kitchen, P.; Shah, Z.A.; Salman, M.M. Astrocytes in Functional Recovery Following Central Nervous System Injuries. J. Physiol. 2024, 602, 3069–3096. [Google Scholar] [CrossRef]
  91. Burda, J.E.; Bernstein, A.M.; Sofroniew, M.V. Astrocyte Roles in Traumatic Brain Injury. Exp. Neurol. 2016, 275, 305–315. [Google Scholar] [CrossRef]
  92. Linnerbauer, M.; Rothhammer, V. Protective Functions of Reactive Astrocytes Following Central Nervous System Insult. Front. Immunol. 2020, 11, 573256. [Google Scholar] [CrossRef]
  93. Zhou, Y.; Shao, A.; Yao, Y.; Tu, S.; Deng, Y.; Zhang, J. Dual Roles of Astrocytes in Plasticity and Reconstruction after Traumatic Brain Injury. Cell Commun. Signal. CCS 2020, 18, 62. [Google Scholar] [CrossRef]
  94. Pekny, M.; Pekna, M. Astrocyte Reactivity and Reactive Astrogliosis: Costs and Benefits. Physiol. Rev. 2014, 94, 1077–1098. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, C.; Lin, L.; Yin, L.; Hao, X.; Tian, J.; Zhang, X.; Ren, Y.; Li, C.; Yang, Y. Acutely Inhibiting AQP4 With TGN-020 Improves Functional Outcome by Attenuating Edema and Peri-Infarct Astrogliosis After Cerebral Ischemia. Front. Immunol. 2022, 13, 870029. [Google Scholar] [CrossRef] [PubMed]
  96. Xin, W.; Pan, Y.; Wei, W.; Tatenhorst, L.; Graf, I.; Popa-Wagner, A.; Gerner, S.T.; Huber, S.; Kilic, E.; Hermann, D.M.; et al. Preconditioned Extracellular Vesicles from Hypoxic Microglia Reduce Poststroke AQP4 Depolarization, Disturbed Cerebrospinal Fluid Flow, Astrogliosis, and Neuroinflammation. Theranostics 2023, 13, 4197–4216. [Google Scholar] [CrossRef] [PubMed]
  97. Bin Alamer, O.; Palmisciano, P.; Mallela, A.N.; Labib, M.A.; Gardner, P.A.; Couldwell, W.T.; Lunsford, L.D.; Abou-Al-Shaar, H. Stereotactic Radiosurgery in the Management of Petroclival Meningiomas: A Systematic Review and Meta-Analysis of Treatment Outcomes of Primary and Adjuvant Radiosurgery. J. Neurooncol. 2022, 157, 207–219. [Google Scholar] [CrossRef]
  98. Kozin, S.V. Vascular Damage in Tumors: A Key Player in Stereotactic Radiation Therapy? Trends Cancer 2022, 8, 806–819. [Google Scholar] [CrossRef] [PubMed]
  99. Yamada, M.K. A Link between Vascular Damage and Cognitive Deficits after Whole-Brain Radiation Therapy for Cancer: A Clue to Other Types of Dementia? Drug Discov. Ther. 2016, 10, 79–81. [Google Scholar] [CrossRef]
  100. Lee, D.; Riestenberg, R.A.; Haskell-Mendoza, A.; Bloch, O. Brain Metastasis Recurrence Versus Radiation Necrosis: Evaluation and Treatment. Neurosurg. Clin. N. Am. 2020, 31, 575–587. [Google Scholar] [CrossRef]
  101. Pazzaglia, S.; Briganti, G.; Mancuso, M.; Saran, A. Neurocognitive Decline Following Radiotherapy: Mechanisms and Therapeutic Implications. Cancers 2020, 12, 146. [Google Scholar] [CrossRef]
  102. Xiang, J.; Lu, Y.; Quan, C.; Gao, Y.; Zhou, G. Metformin Protects Radiation-Induced Early Brain Injury by Reducing Inflammation and DNA Damage. Brain Sci. 2023, 13, 645. [Google Scholar] [CrossRef]
  103. Hladik, D.; Tapio, S. Effects of Ionizing Radiation on the Mammalian Brain. Mutat. Res. Mutat. Res. 2016, 770, 219–230. [Google Scholar] [CrossRef]
  104. Xu, K.; Sun, G.; Wang, Y.; Luo, H.; Wang, Y.; Liu, M.; Liu, H.; Lu, X.; Qin, X. Mitigating Radiation-Induced Brain Injury via NLRP3/NLRC4/Caspase-1 Pyroptosis Pathway: Efficacy of Memantine and Hydrogen-Rich Water. Biomed. Pharmacother. 2024, 177, 116978. [Google Scholar] [CrossRef] [PubMed]
  105. Yang, Z.; Bai, S.; Gu, B.; Peng, S.; Liao, W.; Liu, J. Radiation-Induced Brain Injury After Radiotherapy for Brain Tumor. In Molecular Considerations and Evolving Surgical Management Issues in the Treatment of Patients with a Brain Tumor; IntechOpen: London, UK, 2015; ISBN 978-953-51-2031-5. [Google Scholar]
  106. Dye, N.B.; Gondi, V.; Mehta, M.P. Strategies for Preservation of Memory Function in Patients with Brain Metastases. Chin. Clin. Oncol. 2015, 4, 24. [Google Scholar] [CrossRef] [PubMed]
  107. Chen, H.S.; Pellegrini, J.W.; Aggarwal, S.K.; Lei, S.Z.; Warach, S.; Jensen, F.E.; Lipton, S.A. Open-Channel Block of N-Methyl-D-Aspartate (NMDA) Responses by Memantine: Therapeutic Advantage against NMDA Receptor-Mediated Neurotoxicity. J. Neurosci. 1992, 12, 4427–4436. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, H.S.; Lipton, S.A. Mechanism of Memantine Block of NMDA-Activated Channels in Rat Retinal Ganglion Cells: Uncompetitive Antagonism. J. Physiol. 1997, 499, 27–46. [Google Scholar] [CrossRef] [PubMed]
  109. Pellegrini, J.W.; Lipton, S.A. Delayed Administration of Memantine Prevents N-Methyl-D-Aspartate Receptor-Mediated Neurotoxicity. Ann. Neurol. 1993, 33, 403–407. [Google Scholar] [CrossRef]
  110. Orgogozo, J.-M.; Rigaud, A.-S.; Stöffler, A.; Möbius, H.-J.; Forette, F. Efficacy and Safety of Memantine in Patients with Mild to Moderate Vascular Dementia: A Randomized, Placebo-Controlled Trial (MMM 300). Stroke 2002, 33, 1834–1839. [Google Scholar] [CrossRef]
  111. Wilcock, G.; Möbius, H.J.; Stöffler, A. MMM 500 group A Double-Blind, Placebo-Controlled Multicentre Study of Memantine in Mild to Moderate Vascular Dementia (MMM500). Int. Clin. Psychopharmacol. 2002, 17, 297–305. [Google Scholar] [CrossRef]
  112. Brown, P.D.; Pugh, S.; Laack, N.N.; Wefel, J.S.; Khuntia, D.; Meyers, C.; Choucair, A.; Fox, S.; Suh, J.H.; Roberge, D.; et al. Memantine for the Prevention of Cognitive Dysfunction in Patients Receiving Whole-Brain Radiotherapy: A Randomized, Double-Blind, Placebo-Controlled Trial. Neuro-Oncology 2013, 15, 1429–1437. [Google Scholar] [CrossRef]
  113. Greene-Schloesser, D.; Robbins, M.E.; Peiffer, A.M.; Shaw, E.G.; Wheeler, K.T.; Chan, M.D. Radiation-Induced Brain Injury: A Review. Front. Oncol. 2012, 2, 73. [Google Scholar] [CrossRef]
  114. Brown, P.D.; Gondi, V.; Pugh, S.; Tome, W.A.; Wefel, J.S.; Armstrong, T.S.; Bovi, J.A.; Robinson, C.; Konski, A.; Khuntia, D.; et al. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients With Brain Metastases: Phase III Trial NRG Oncology CC001. J. Clin. Oncol. 2020, 38, 1019–1029. [Google Scholar] [CrossRef]
  115. Chambial, S.; Dwivedi, S.; Shukla, K.K.; John, P.J.; Sharma, P. Vitamin C in Disease Prevention and Cure: An Overview. Indian J. Clin. Biochem. IJCB 2013, 28, 314–328. [Google Scholar] [CrossRef] [PubMed]
  116. Castro, M.A.; Beltrán, F.A.; Brauchi, S.; Concha, I.I. A Metabolic Switch in Brain: Glucose and Lactate Metabolism Modulation by Ascorbic Acid. J. Neurochem. 2009, 110, 423–440. [Google Scholar] [CrossRef] [PubMed]
  117. Grünewald, R.A. Ascorbic Acid in the Brain. Brain Res. Brain Res. Rev. 1993, 18, 123–133. [Google Scholar] [CrossRef] [PubMed]
  118. Nishikimi, M.; Yagi, K. Biochemistry and Molecular Biology of Ascorbic Acid Biosynthesis. In Subcellular Biochemistry: Ascorbic Acid: Biochemistry and Biomedical Cell Biology; Harris, J.R., Ed.; Springer: Boston, MA, USA, 1996; pp. 17–39. ISBN 978-1-4613-0325-1. [Google Scholar]
  119. Hammarström, L. Autoradiographic Studies on the Distribution of C14-Labelled Ascorbic Acid and Dehydroascorbic Acid. Acta Physiol. Scand. 1966, 70, 1–83. [Google Scholar] [CrossRef]
  120. May, J.M. Vitamin C Transport and Its Role in the Central Nervous System. In Water Soluble Vitamins: Clinical Research and Future Application; Subcellular Biochemistry; Springer: Dordrecht, The Netherlands, 2012; Volume 56, pp. 85–103. [Google Scholar] [CrossRef]
  121. Rice, M.E. Ascorbate Regulation and Its Neuroprotective Role in the Brain. Trends Neurosci. 2000, 23, 209–216. [Google Scholar] [CrossRef]
  122. Bürzle, M.; Suzuki, Y.; Ackermann, D.; Miyazaki, H.; Maeda, N.; Clémençon, B.; Burrier, R.; Hediger, M.A. The Sodium-Dependent Ascorbic Acid Transporter Family SLC23. Mol. Aspects Med. 2013, 34, 436–454. [Google Scholar] [CrossRef] [PubMed]
  123. Stamford, J.A.; Kruk, Z.L.; Millar, J. Regional Differences in Extracellular Ascorbic Acid Levels in the Rat Brain Determined by High Speed Cyclic Voltammetry. Brain Res. 1984, 299, 289–295. [Google Scholar] [CrossRef]
  124. Schenk, J.O.; Miller, E.; Gaddis, R.; Adams, R.N. Homeostatic Control of Ascorbate Concentration in CNS Extracellular Fluid. Brain Res. 1982, 253, 353–356. [Google Scholar] [CrossRef]
  125. Miele, M.; Fillenz, M. In Vivo Determination of Extracellular Brain Ascorbate. J. Neurosci. Methods 1996, 70, 15–19. [Google Scholar] [CrossRef]
  126. Huang, J.; Agus, D.B.; Winfree, C.J.; Kiss, S.; Mack, W.J.; McTaggart, R.A.; Choudhri, T.F.; Kim, L.J.; Mocco, J.; Pinsky, D.J.; et al. Dehydroascorbic Acid, a Blood-Brain Barrier Transportable Form of Vitamin C, Mediates Potent Cerebroprotection in Experimental Stroke. Proc. Natl. Acad. Sci. USA 2001, 98, 11720–11724. [Google Scholar] [CrossRef]
  127. Astuya, A.; Caprile, T.; Castro, M.; Salazar, K.; García, M.D.L.A.; Reinicke, K.; Rodríguez, F.; Vera, J.C.; Millán, C.; Ulloa, V.; et al. Vitamin C Uptake and Recycling among Normal and Tumor Cells from the Central Nervous System. J. Neurosci. Res. 2005, 79, 146–156. [Google Scholar] [CrossRef] [PubMed]
  128. Diliberto, E.J.; Dean, G.; Carter, C.; Allen, P.L. Tissue, Subcellular, and Submitochondrial Distributions of Semidehydroascorbate Reductase: Possible Role of Semidehydroascorbate Reductase in Cofactor Regeneration. J. Neurochem. 1982, 39, 563–568. [Google Scholar] [CrossRef] [PubMed]
  129. Rice, M.E.; Russo-Menna, I. Differential Compartmentalization of Brain Ascorbate and Glutathione between Neurons and Glia. Neuroscience 1998, 82, 1213–1223. [Google Scholar] [CrossRef] [PubMed]
  130. Castro, M.A.; Pozo, M.; Cortés, C.; García, M.D.L.A.; Concha, I.I.; Nualart, F. Intracellular Ascorbic Acid Inhibits Transport of Glucose by Neurons, but Not by Astrocytes. J. Neurochem. 2007, 102, 773–782. [Google Scholar] [CrossRef] [PubMed]
  131. Castro, M.; Caprile, T.; Astuya, A.; Millán, C.; Reinicke, K.; Vera, J.C.; Vásquez, O.; Aguayo, L.G.; Nualart, F. High-Affinity Sodium-Vitamin C Co-Transporters (SVCT) Expression in Embryonic Mouse Neurons. J. Neurochem. 2001, 78, 815–823. [Google Scholar] [CrossRef]
  132. Padayatty, S.J.; Katz, A.; Wang, Y.; Eck, P.; Kwon, O.; Lee, J.-H.; Chen, S.; Corpe, C.; Dutta, A.; Dutta, S.K.; et al. Vitamin C as an Antioxidant: Evaluation of Its Role in Disease Prevention. J. Am. Coll. Nutr. 2003, 22, 18–35. [Google Scholar] [CrossRef]
  133. Majewska, M.D.; Bell, J.A. Ascorbic Acid Protects Neurons from Injury Induced by Glutamate and NMDA. Neuroreport 1990, 1, 194–196. [Google Scholar] [CrossRef]
  134. Niki, E. Action of Ascorbic Acid as a Scavenger of Active and Stable Oxygen Radicals. Am. J. Clin. Nutr. 1991, 54, 1119S–1124S. [Google Scholar] [CrossRef]
  135. Rebec, G.V.; Pierce, R.C. A Vitamin as Neuromodulator: Ascorbate Release into the Extracellular Fluid of the Brain Regulates Dopaminergic and Glutamatergic Transmission. Prog. Neurobiol. 1994, 43, 537–565. [Google Scholar] [CrossRef]
  136. Dizdaroglu, M.; Jaruga, P.; Birincioglu, M.; Rodriguez, H. Free Radical-Induced Damage to DNA: Mechanisms and Measurement. Free Radic. Biol. Med. 2002, 32, 1102–1115. [Google Scholar] [CrossRef]
  137. Milne, G.L.; Musiek, E.S.; Morrow, J.D. F2-Isoprostanes as Markers of Oxidative Stress in Vivo: An Overview. Biomark. Biochem. Indic. Expo. Response Susceptibility Chem. 2005, 10 (Suppl. 1), S10–S23. [Google Scholar] [CrossRef]
  138. Reznick, A.Z.; Packer, L. Oxidative Damage to Proteins: Spectrophotometric Method for Carbonyl Assay. Methods Enzymol. 1994, 233, 357–363. [Google Scholar] [CrossRef] [PubMed]
  139. Galley, H.F.; Davies, M.J.; Webster, N.R. Ascorbyl Radical Formation in Patients with Sepsis: Effect of Ascorbate Loading. Free Radic. Biol. Med. 1996, 20, 139–143. [Google Scholar] [CrossRef] [PubMed]
  140. Rodrigo, R.; Guichard, C.; Charles, R. Clinical Pharmacology and Therapeutic Use of Antioxidant Vitamins. Fundam. Clin. Pharmacol. 2007, 21, 111–127. [Google Scholar] [CrossRef]
  141. Padh, H. Cellular Functions of Ascorbic Acid. Biochem. Cell Biol. Biochim. Biol. Cell. 1990, 68, 1166–1173. [Google Scholar] [CrossRef]
  142. Tomé, A.D.R.; Ferreira, P.M.P.; Freitas, R.M. de Inhibitory Action of Antioxidants (Ascorbic Acid or Alpha-Tocopherol) on Seizures and Brain Damage Induced by Pilocarpine in Rats. Arq. Neuropsiquiatr. 2010, 68, 355–361. [Google Scholar] [CrossRef] [PubMed]
  143. Xiao, L.; Tsutsui, T.; Miwa, N. The Lipophilic Vitamin C Derivative, 6-o-Palmitoylascorbate, Protects Human Lymphocytes, Preferentially over Ascorbate, against X-Ray-Induced DNA Damage, Lipid Peroxidation, and Protein Carbonylation. Mol. Cell. Biochem. 2014, 394, 247–259. [Google Scholar] [CrossRef]
  144. Stehli, J.; Fuchs, T.A.; Ghadri, J.R.; Gaemperli, O.; Fiechter, M.; Kaufmann, P.A. Antioxidants Prevent DNA Double-Strand Breaks from X-Ray-Based Cardiac Examinations: A Randomized, Double-Blinded, Placebo-Controlled Trial. J. Am. Coll. Cardiol. 2014, 64, 117–118. [Google Scholar] [CrossRef]
  145. Tao, S.M.; Zhou, F.; Joseph Schoepf, U.; Fischer, A.M.; Giovagnoli, D.; Lin, Z.X.; Zhou, C.S.; Lu, G.M.; Zhang, L.J. The Effect of Prophylactic Oral Vitamin C Use on DNA Double-Strand Breaks after Abdominal Contrast-Enhanced CT: A Preliminary Study. Eur. J. Radiol. 2019, 117, 69–74. [Google Scholar] [CrossRef]
  146. Rostami, A.; Moosavi, S.A.; Dianat Moghadam, H.; Bolookat, E.R. Micronuclei Assessment of The Radioprotective Effects of Melatonin and Vitamin C in Human Lymphocytes. Cell J. 2016, 18, 46–51. [Google Scholar] [CrossRef]
  147. Du, J.; Cieslak, J.A.; Welsh, J.L.; Sibenaller, Z.A.; Allen, B.G.; Wagner, B.A.; Kalen, A.L.; Doskey, C.M.; Strother, R.K.; Button, A.M.; et al. Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer. Cancer Res. 2015, 75, 3314–3326. [Google Scholar] [CrossRef] [PubMed]
  148. Didier, A.J.; Stiene, J.; Fang, L.; Watkins, D.; Dworkin, L.D.; Creeden, J.F. Antioxidant and Anti-Tumor Effects of Dietary Vitamins A, C, and E. Antioxidants 2023, 12, 632. [Google Scholar] [CrossRef] [PubMed]
  149. Kaffash Farkhad, N.; Asadi-Samani, M.; Asadi-Samani, F.; Asadi-Samani, H. The Role of Natural Antioxidants in Reducing Oxidative Stress in Cancer. In Plant Antioxidants and Health; Ekiert, H.M., Ramawat, K.G., Arora, J., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–16. ISBN 978-3-030-45299-5. [Google Scholar]
  150. Asadi-Samani, M.; Farkhad, N.K.; Mahmoudian-Sani, M.R.; Shirzad, H.; Asadi-Samani, M.; Farkhad, N.K.; Mahmoudian-Sani, M.R.; Shirzad, H. Antioxidants as a Double-Edged Sword in the Treatment of Cancer. In Antioxidants; IntechOpen: London, UK, 2019; ISBN 978-1-78923-920-1. [Google Scholar]
  151. Zahra, K.F.; Lefter, R.; Ali, A.; Abdellah, E.-C.; Trus, C.; Ciobica, A.; Timofte, D. The Involvement of the Oxidative Stress Status in Cancer Pathology: A Double View on the Role of the Antioxidants. Oxid. Med. Cell. Longev. 2021, 2021, 9965916. [Google Scholar] [CrossRef]
  152. Cockfield, J.A.; Schafer, Z.T. Antioxidant Defenses: A Context-Specific Vulnerability of Cancer Cells. Cancers 2019, 11, 1208. [Google Scholar] [CrossRef]
  153. Zaher, A.; Petronek, M.S.; Allen, B.G.; Mapuskar, K.A. Balanced Duality: H2O2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues. Int. J. Mol. Sci. 2024, 25, 8885. [Google Scholar] [CrossRef] [PubMed]
  154. Petronek, M.S.; Monga, V.; Bodeker, K.L.; Kwofie, M.; Lee, C.-Y.; Mapuskar, K.A.; Stolwijk, J.M.; Zaher, A.; Wagner, B.A.; Smith, M.C.; et al. Magnetic Resonance Imaging of Iron Metabolism with T2* Mapping Predicts an Enhanced Clinical Response to Pharmacologic Ascorbate in Patients with GBM. Clin. Cancer Res. 2024, 30, 283–293. [Google Scholar] [CrossRef]
  155. Gan, L.; Wang, Z.; Si, J.; Zhou, R.; Sun, C.; Liu, Y.; Ye, Y.; Zhang, Y.; Liu, Z.; Zhang, H. Protective Effect of Mitochondrial-Targeted Antioxidant MitoQ against Iron Ion 56Fe Radiation Induced Brain Injury in Mice. Toxicol. Appl. Pharmacol. 2018, 341, 1–7. [Google Scholar] [CrossRef]
  156. Kale, A.; Piskin, Ö.; Bas, Y.; Aydin, B.G.; Can, M.; Elmas, Ö.; Büyükuysal, Ç. Neuroprotective Effects of Quercetin on Radiation-Induced Brain Injury in Rats. J. Radiat. Res. 2018, 59, 404–410. [Google Scholar] [CrossRef] [PubMed]
  157. Abou-Zeid, S.M.; El-Bialy, B.E.; El-Borai, N.B.; AbuBakr, H.O.; Elhadary, A.M.A. Radioprotective Effect of Date Syrup on Radiation- Induced Damage in Rats. Sci. Rep. 2018, 8, 7423. [Google Scholar] [CrossRef]
  158. Allen, B.D.; Acharya, M.M.; Lu, C.; Giedzinski, E.; Chmielewski, N.N.; Quach, D.; Hefferan, M.; Johe, K.K.; Limoli, C.L. Remediation of Radiation-Induced Cognitive Dysfunction through Oral Administration of the Neuroprotective Compound NSI-189. Radiat. Res. 2018, 189, 345–353. [Google Scholar] [CrossRef]
  159. Stessin, A.M.; Banu, M.A.; Clausi, M.G.; Berry, N.; Boockvar, J.A.; Ryu, S. FTY720/Fingolimod, an Oral S1PR Modulator, Mitigates Radiation Induced Cognitive Deficits. Neurosci. Lett. 2017, 658, 1–5. [Google Scholar] [CrossRef] [PubMed]
  160. He, G.Y.; Huang, H.W.; Deng, Z.Z.; Guo, J.J. Effect of mouse nerve growth factor on cognitive impairment in whole brain irradiation rats. Zhonghua Yi Xue Za Zhi 2016, 96, 1530–1534. [Google Scholar] [CrossRef] [PubMed]
  161. Zhou, A.Y.; Song, B.W.; Fu, C.Y.; Baranenko, D.D.; Wang, E.J.; Li, F.Y.; Lu, G.W. Acanthopanax Senticosus Reduces Brain Injury in Mice Exposed to Low Linear Energy Transfer Radiation. Biomed. Pharmacother. Biomedecine Pharmacother. 2018, 99, 781–790. [Google Scholar] [CrossRef]
Figure 1. Cellular response to radiation-induced DNA damage. Ionizing radiation induces DNA damage in cells in the form of single- or double-strand breaks via ROS formation, thus blocking their ability to divide and proliferate further. DNA damage is sensed by cells, and results in various cellular responses, depending on the level of DNA damage (repairable or irreparable) and cell type (normal or cancer cell). Failure to activate normal p53-dependent DNA damage response may cause mitotic catastrophe or the generation of aneuploid cells which contribute to the progression of cancer. Signaling pathways that promote DNA repair and inhibition of cell death can protect cancer cells from irradiation-induced cytotoxicity, promoting survival and subsequent radiation resistance of cancer cells.
Figure 1. Cellular response to radiation-induced DNA damage. Ionizing radiation induces DNA damage in cells in the form of single- or double-strand breaks via ROS formation, thus blocking their ability to divide and proliferate further. DNA damage is sensed by cells, and results in various cellular responses, depending on the level of DNA damage (repairable or irreparable) and cell type (normal or cancer cell). Failure to activate normal p53-dependent DNA damage response may cause mitotic catastrophe or the generation of aneuploid cells which contribute to the progression of cancer. Signaling pathways that promote DNA repair and inhibition of cell death can protect cancer cells from irradiation-induced cytotoxicity, promoting survival and subsequent radiation resistance of cancer cells.
Neurosci 05 00034 g001
Figure 2. Principles of radiotherapy in normal and cancerous cells. This figure illustrates the distinct responses of normal and tumor cells to radiation. It emphasizes the pathways through which radiation induces DNA damage, ultimately leading to various forms of cell death, including apoptosis, mitotic catastrophe, and senescence. Abbreviations: CNS, Central Nervous System; ROS, Reactive Oxygen Species.
Figure 2. Principles of radiotherapy in normal and cancerous cells. This figure illustrates the distinct responses of normal and tumor cells to radiation. It emphasizes the pathways through which radiation induces DNA damage, ultimately leading to various forms of cell death, including apoptosis, mitotic catastrophe, and senescence. Abbreviations: CNS, Central Nervous System; ROS, Reactive Oxygen Species.
Neurosci 05 00034 g002
Figure 3. Ascorbate dynamics CNS: uptake and regulation. This figure outlines the distribution and dynamics of ascorbic acid (AA) as it moves from the blood supply into the central nervous system (CNS) to function as an antioxidant. Ascorbate enters the CNS via glucose transporters (GLUTs) as dehydroascorbic acid (DHA) and is reduced to AA in astrocytes. AA is then taken up into neurons from the extracellular fluid (ECF) through sodium-dependent vitamin C transporter 2 (SVCT2), acting as an intracellular antioxidant and oxidizing back to DHA. The recycling of DHA from neurons to the ECF is facilitated by astrocytes. The extracellular concentration of AA is homeostatically regulated and influenced by glutamate release, enhancing transport from astrocytes to the ECF. However, AA recycling may be compromised in pathophysiological conditions. Abbreviations: DHA: Dehydroascorbic acid; GLUT: Glucose transporter; AA: Ascorbic acid; SVCT2: Sodium-ascorbate co-transporters; ROS: reactive oxygen species.
Figure 3. Ascorbate dynamics CNS: uptake and regulation. This figure outlines the distribution and dynamics of ascorbic acid (AA) as it moves from the blood supply into the central nervous system (CNS) to function as an antioxidant. Ascorbate enters the CNS via glucose transporters (GLUTs) as dehydroascorbic acid (DHA) and is reduced to AA in astrocytes. AA is then taken up into neurons from the extracellular fluid (ECF) through sodium-dependent vitamin C transporter 2 (SVCT2), acting as an intracellular antioxidant and oxidizing back to DHA. The recycling of DHA from neurons to the ECF is facilitated by astrocytes. The extracellular concentration of AA is homeostatically regulated and influenced by glutamate release, enhancing transport from astrocytes to the ECF. However, AA recycling may be compromised in pathophysiological conditions. Abbreviations: DHA: Dehydroascorbic acid; GLUT: Glucose transporter; AA: Ascorbic acid; SVCT2: Sodium-ascorbate co-transporters; ROS: reactive oxygen species.
Neurosci 05 00034 g003
Figure 4. Mechanism of AA uptake and its effects in the cell. Studies with cultured cells have shown that AA can affect gene expression mediated by its redox effects. Abbreviations: DHA, dehydroascorbic acid; Fe3+, ferric iron; Fe2+, ferrous iron; GPX, glutathione peroxidase; GSH, reduced glutathione; GSSG, oxidized glutathione; GST, glutathione transferase; Vit C, vitamin C; Vit E, vitamin E.
Figure 4. Mechanism of AA uptake and its effects in the cell. Studies with cultured cells have shown that AA can affect gene expression mediated by its redox effects. Abbreviations: DHA, dehydroascorbic acid; Fe3+, ferric iron; Fe2+, ferrous iron; GPX, glutathione peroxidase; GSH, reduced glutathione; GSSG, oxidized glutathione; GST, glutathione transferase; Vit C, vitamin C; Vit E, vitamin E.
Neurosci 05 00034 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

González-Johnson, L.; Fariña, A.; Farías, G.; Zomosa, G.; Pinilla-González, V.; Rojas-Solé, C. Exploring Neuroprotection against Radiation-Induced Brain Injury: A Review of Key Compounds. NeuroSci 2024, 5, 462-484. https://doi.org/10.3390/neurosci5040034

AMA Style

González-Johnson L, Fariña A, Farías G, Zomosa G, Pinilla-González V, Rojas-Solé C. Exploring Neuroprotection against Radiation-Induced Brain Injury: A Review of Key Compounds. NeuroSci. 2024; 5(4):462-484. https://doi.org/10.3390/neurosci5040034

Chicago/Turabian Style

González-Johnson, Lucas, Ariel Fariña, Gonzalo Farías, Gustavo Zomosa, Víctor Pinilla-González, and Catalina Rojas-Solé. 2024. "Exploring Neuroprotection against Radiation-Induced Brain Injury: A Review of Key Compounds" NeuroSci 5, no. 4: 462-484. https://doi.org/10.3390/neurosci5040034

Article Metrics

Back to TopTop