Next Article in Journal
Sleep Quality Moderates the Impact of Place-Based Social Adversity on Physical Health in Women with Breast Cancer Transitioning from Active Treatment to Survivorship
Previous Article in Journal
BCMA CAR-T: From Multiple Myeloma to Light-Chain Amyloidosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Depression and Anxiety After Radiation-Induced Brain Injury: A Review of Current Research Progress

Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Curr. Oncol. 2025, 32(8), 419; https://doi.org/10.3390/curroncol32080419 (registering DOI)
Submission received: 2 July 2025 / Revised: 22 July 2025 / Accepted: 24 July 2025 / Published: 26 July 2025
(This article belongs to the Section Psychosocial Oncology)

Simple Summary

Radiation therapy is a cornerstone treatment for brain tumors but frequently causes radiation-induced brain injury, damaging healthy brain tissue. A significant consequence of radiation-induced brain injury is the development of affective disorders, particularly major depression and anxiety, severely impacting patients’ quality of life. The underlying pathophysiology involves complex mechanisms like neuroinflammation, oxidative stress, blood–brain barrier disruption, and white matter damage. Current management relies on antidepressants, corticosteroids, and neuroprotective agents, while promising new therapies target neuroinflammation and neural repair pathways. This review examines the pathogenesis of radiation-induced brain injury-related affective disorders and evaluates both established and emerging treatment strategies, aiming to inform the development of more effective interventions to improve patient outcomes.

Abstract

Radiation therapy serves as a fundamental treatment for primary and metastatic brain tumors, whether used alone or combined with surgery and chemotherapy. Despite its oncological efficacy, this treatment paradigm frequently induces radiation-induced brain injury (RBI), a progressive neuropathological condition characterized by structural and functional damage to healthy cerebral parenchyma. Patients with RBI frequently develop affective disorders, particularly major depressive disorder and generalized anxiety disorder, which profoundly impair psychosocial functioning and quality of life. The pathophysiology involves complex mechanisms such as neuroinflammation, oxidative stress, blood–brain barrier disruption, and white matter damage. Current management strategies include antidepressants, corticosteroids, and neuroprotective agents, while emerging therapies targeting neuroinflammation and neural repair show promise. This review comprehensively examines the pathogenesis of RBI-related affective disorders and evaluates both conventional and novel treatment approaches. By synthesizing current evidence, we aim to provide insights for developing more effective interventions to improve patient outcomes and quality of life.

1. Introduction

Brain tumors are one of the most leading causes of cancer-related death globally, especially in children [1,2]. Metastatic brain tumors, typically from lung cancer, breast cancer and melanoma, jeopardize about 10 to 30 percent of adult cancer patients and 6 to 10 percent of childhood cancer patients [3,4]. At present, radiation therapy is the primary treatment for brain tumors. However, although the survival time of tumor patients has continuously extended with the rapid development of radiotherapy technology, the probability of radiation-induced brain injury (RBI) has also aroused the attention of doctors in the meantime [5,6,7,8,9]. Affective disorders such as depression and anxiety, generally appear several years after brain radiotherapy [10]. Specifically, depression is mainly manifested as low mood, insomnia, and fatigue [11]; anxiety disorders are primarily featured with stress and autonomic hyperactivity [12]. As the above symptoms all seriously affect the quality of patients’ life, RBI undoubtedly causes tremendous economic pressure not only on society but also on the medical and health environment.
The pathological mechanism of affective disorders after radiotherapy is complicated. Currently, the main hypotheses include chronic neuroinflammation, damage to emotion-related brain areas such as decreased hippocampal neurogenesis, lack of neurotrophic factors, synaptic ultrastructure impairment, and functional defects, etc.
The purpose of this review is to systematically examine the clinical manifestations, underlying mechanisms, and therapeutic approaches for RBI-associated affective disorders. This synthesis of current evidence will facilitate further investigation into RBI and its neuropsychiatric sequelae, with the ultimate goals of expanding treatment paradigms, optimizing clinical interventions, and improving patients’ quality of life.

2. The Association Between RBI and Symptoms of Depression and Anxiety

2.1. Clinical Trials

The current clinical trials are still in their early stages. As early as 1997s, a meta-analytical review conducted by Van’t Spijker et al. demonstrated that the incidence of depression in cancer patients was often higher than that in the general population [13]. Moreover, after Whole-brain radiation therapy, attention decline is common [14]; and this symptom is especially apparent in patients with attention problems [15]. The incidence of depression is 54%, and the mortality rate of cancer patients suffering from depression is twice that of other cancer patients [16]. A study that, respectively, examined the Baker Depression and Anxiety Survey of 13 brain tumor patients before and after radiation therapy showed that depression and anxiety were remarkably higher in those who were administrated with radiation therapy than those who were not [17]. A study conducted by Wang et al. in 2022 revealed a significant increase in the rates of depression and anxiety following radiation therapy for nasopharyngeal cancer. Specifically, the prevalence of anxiety among nasopharyngeal cancer patients increased from 34% prior to radiation therapy to 64% afterward, while the prevalence of depression rose from 25% to 56% [18]. Compared with adults, children receiving craniocerebral radiotherapy are more susceptible to severe depression and anxiety [18]. Another paired case-control study, which included 46 patients with NPS as the case group and 46 patients without NPS as the control group, found that the depression and anxiety levels in the case group were drastically higher than those in the control group [10]. Although clinical trials do not directly prove that RBI causes affective disorders, they suggest that RBI is a potential hazard for depression and anxiety in cancer patients and may be an underlying cause. Moreover, the prevalence of anxiety and depression was the highest in patients with head and neck tumors after radiotherapy [19]. Atrophy of certain brain area such as amygdala was proved to connect with poor memory, mood and emotional health. This pathological change may also happen during RBI. As expected, a recent longitudinal clinical trial showed bilateral amygdala and nucleus atrophy occurred in a time and dose-dependent manner after brain radiotherapy. Therefore, treatment plans that preserve the amygdala may preserve neurocognitive and neuropsychiatric functions in this population [20,21] (Table 1).

2.2. Animal Tests

During cancer diagnosis and treatment, patients will inevitably be bothered with negative emotions, which is likely to trigger psychiatric comorbidities [11]. For this reason, experimental models used to study depressive symptoms after cancer treatment have been carefully chosen. However, as early as 1994, Miyachi, Y., in the process of studying the effects of low-dose X-ray, found that mice showed significant behavioral inhibition after 5 to 15 cGy total body X-ray irradiation [22], but the higher dose (25–35 cGy) did not play the same role. This may be explained by the radiation dose-dependent genetic changes; brain tissue in mice after total-body irradiation transcriptome spectrum analysis showed that low-dose exposure (10 cGy) induced genes are not affected by high-dose radiation (2 Gy), and the gene induced by low dose also has its unique signal pathways and functions [23]. In 2014, Son et al. reported that mice exposed to a single 10 Gy irradiation showed significant depression-like behaviors at 30- and 90-day post-exposure [24]. However, in 2019, Ueno, H et al. irradiated the brains of mature mice with 20 Gy ionizing radiation to study the behavioral disorders of mice in the subacute stage. In the forced swimming experiment one week later, the irradiated mice had substantially less immobility time than the control group, and the mice showed depression-like behaviors compared with the control group [25]. All of these above animal experimentations provide evidence for RBI-related depression.
The results of animal experiments on depression caused by RBI are relatively simple, mostly showing an increase in depressive behavior. However, results from animal studies related to anxiety have been controversial, despite the significant stress generated by potential radiation toxicity in the brain [26]. In 2020, Dey, D. et. Al. reported that five weeks after radiotherapy combined with TMZ chemotherapy, the mice exhibited anxiety-like behaviors, and anxiety-like along with depression-like behaviors were observed at week 15. Notably, neither irradiation nor TMZ treatment alone induced significant anxiety-like behaviors [27]. In 2016, Brown, R. J. et al. administered fraction-dose or single high-dose whole-brain irradiation to young rats, and no significant difference was observed in open field tests during the subacute phase, but the change in exercise habits and anxiety-like behavior increased [28]. At the same time, Ueno, H. et al. irradiated the brains of mature mice with 20 Gy, and the tail suspension test showed no abnormality after 3–6 days, which indicated that severe anxiety behavior was unlikely to be caused in the subacute phase, but it may also be related to the dose of irradiation, as low-dose irradiation seems to be more likely to cause anxiety disorder compared with high dose [25]. In 2012, Trivedi, R. et al. reported that mice irradiated with radiation doses of 3, 5, and 8Gy evaluated the emotion-related behaviors. The results found that on day 5 post-irradiation, both the 3 Gy and 8 Gy doses exhibited antianxiety effects (as evidenced by prolonged center time), whereas the 5 Gy dose showed no significant effect. By day 10, anxiety-like behavior demonstrated a trend toward dose-dependent modulation; however, no statistically significant difference was observed between the 5 Gy and 8 Gy groups [29]. In 2017, Zhou K et al. found that the duration of staying in the open central area in the irradiated group (12 Gy) was 71.4% higher than that in the non-irradiated group after 28 days of birth [30]. The reasons accounting for above controversial results are complicated, including different experimental conditions (including radiation dose, type, time, etc.), variances in animal species, and lack of consistent anxiety-related behavior tests (Table 2).

3. Possible Mechanism of RBI Leading to Depression and Anxiety

Theoretically, brain necrosis will inevitably cause affective disorders when RBI affects cognition-related brain regions. Although the mechanism of RBI and its underlying molecular basis remain unclear, researchers have reached a consensus that RBI pathogenesis is complex, continuous, and dynamic. This process involves multiple aspects, including radiation-induced blood–brain barrier disruption, injury to various neural cell types, microvascular damage, neuroinflammation, impaired neurogenesis, and other contributing factors [35]. These complex mechanisms ultimately lead to diverse pathological manifestations, ranging from focal radiation necrosis to diffuse leukoencephalopathy with cerebral atrophy [36].

3.1. Hippocampal Dysfunction

The hippocampus is an important part of the cerebral limbic system, which plays a predominant role in cognitive and emotional regulation [37]. The dorsal part of the hippocampus mainly regulates learning and memory functions, while the ventral part is primarily responsible for emotional behavior and stress response [38,39]. It has also been confirmed that hippocampal dysfunction is associated with the development of neurological and psychiatric disorders [11]. Numerous data have demonstrated that craniocerebral radiation can induce hippocampal-dependent behavior disorders, including depression-like behavior [40,41].

3.1.1. Reduced Hippocampal Neurogenesis

Adult neurogenesis persists throughout life, albeit with a gradual age-related decline. In the mature mammalian brain, neural stem cells are primarily located in the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus [42]. Therefore, these two areas will differentiate into new neurons when stimulated. Radiotherapy can inhibit hippocampal neurogenesis, hence leading to numerous changes in the brain including a shift from long-term potentiation (LTP) to long-term depression and a permanent alternation in synaptic plasticity. And this pathological process is more pronounced in adolescents than in adults [43,44]. Some previous studies have suggested that changes in adult hippocampal neurons may be fundamentally involved in the pathophysiology of depressive symptoms [45,46,47]. Sustained neurogenesis is defined as the continuous development of new brain neurons, which is indispensable for maintaining the normal behavioral functions. When neurogenesis is inhibited through pharmacological means, individuals may experience symptoms commonly found in depressed patients [48,49]. This finding was further supported by a study, where Santarelli et al. discovered that antidepressants induced neurogenesis disruption prevented the expected behavioral responses to these medications [50]. NRBF2 is essential part of the autophagy-associated phosphatidylinositol 3-kinase complex, which contains PIK3C3/VPS34 [51]. Increasing the levels of NRBF2 in the adult neural stem cells of the DG region of the brain was able to reverse the negative effects on neurogenesis and alleviate symptoms of depression in the mice [52]. Overall, these findings highlight the importance of sustained neurogenesis for maintaining behavioral homeostasis and provide insights into the molecular mechanisms underlying the link between neurogenesis and depression. Although reduced neurogenesis is thought to be closely related to mood disorders, its exact mechanism of action remains controversial. Several animal studies have demonstrated a direct relationship between neurogenetic disorders in the hippocampus and depression under specific conditions of ionizing radiation. Earlier in 2011, Snyder et al., using either transgenic or radiation methods to inhibit adult neurogenesis specifically, found that mice with neurogenic defects showed increased immobility during forced swimming tests and decreased preference for sucrose compared to controls [53]. These findings confirm that neurons in the dentate gyrus are critical to the negative regulation of the HPA axis in the hippocampus and support the direct role of neurogenesis in post-radiation depression in adults. Son et al. (2014) reported that mice exhibiting depressive-like behaviors during the 90-day period following 10 Gy irradiation showed a progressive reduction in hippocampal doublecortin (DCX, an immunohistochemical marker of neurogenesis)-positive cells [24]. Kang, J. et al. also observed similar depressive behavior in the mouse model 30 days after 2 Gy skull irradiation, which was accompanied by a downregulation of the BrdU/Neun and DCX-positive populations, as well as an upregulation of MAP-2 expression in the hippocampus [32]. After analyzing the brain transcriptional files of radiation-induced depressed mice, they found that radiation-induced depressed mice increased expression of thyroxine transporter protein (TTR). This suggests that depression, one side effect of cranial radiotherapy, could be mediated by the inhibition of a pathophysiological procedure called retinol-regulated hippocampal neurogenesis [32]. However, in another study, hippocampal neurogenesis was reduced after a single dose of 5 Gy irradiation, but there was no significant change in the immobility time of the mice during the TST test [31]. The difference between them can be explained by differences in the type or dose of radiation. Therefore, further research is needed to determine whether hippocampal neurogenesis contributes to the improvement of depression-like behavior. At the same time, various factors, such as age, radiation source, dose, sex, and genes, as well as the duration of the behavioral studies, may influence the observation of behavioral changes. It is generally believed that women are more likely to be bothered with mood disorders. For instance, Roughton, K. et al. irradiated the whole brain of male and female mice with a dose of 8 Gy on the 14th day after birth. After ionizing radiation, the proliferation of neural stem cells in the subgranular region of the dentate gyrus of the hippocampus was sharply suppressed, especially in females, who were more inclined to behavior anxiously in the open field experiment. Therefore, there may be a long-term sex difference in craniocerebral irradiation [33]. In addition to gender, the role of genes has also become an increasingly hot research topic. In 2012, Brackett, J. Et al. reported that homozygous antioxidant enzyme related gene defects were more likely to produce delayed mood effect [54]. Another study demonstrated that female ApoE-deficient mice exhibited elevated anxiety-like behaviors [34]. These studies shed light on the potential genetic influence on radiation-induced affective disorders.

3.1.2. Nerve Growth Factor

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophic factor family, is abundant in the hippocampus and indispensable to the survival, proliferation, and differentiation of neurons [55,56]. During central nervous system development, neurotrophic factors are highly sensitive to radiation, so the regulation of BDNF and its receptor expression may be altered by very low doses of radiation (0.02 Gy) [57]. Moreover, the downregulation of BDNF mRNA or protein expression in hippocampus dentate gyrus of anxious or depressed patients is thought to be associated with psychiatric disorders [58,59,60]. This evidence indicates that BDNF play a potential role in hippocampal-dependent emotion regulation. Antidepressants exert therapeutic effects by inducing the expression of neurotrophic factors while targeting apoptotic proteins [61,62,63]. Accordingly, radiation may influence the emotion by altering BDNF levels as well [64]. As expected, a study showed that at 30 and 90 days post-10 Gy irradiation, the downregulation of hippocampal BDNF and GDNF mRNA levels coincided with the emergence of depressive behaviors in rats [24]. The levels of BDNF mRNA and protein in the hippocampus have been proved to be affected by various factors. However, there is still a lack of detailed study on how radiotherapy directly or indirectly affects BDNF mediated mood disorders. It is already well known that intestinal flora could mediate cognitive dysfunction according to the brain–gut axis. Interestingly, this mechanism may also apply to the radiation-induced affective disorders. For example, studies have shown that radiation therapy can affect the composition of intestinal flora, and probiotics can control the expression level of BDNF to alleviate anxiety behavior [38,65]. Therefore, one possible mechanism is that radiotherapy affects emotional regulation by altering the expression of BDNF in the hippocampus through the brain–gut axis.

3.1.3. Alterations of Synaptic Plasticity

The interconnection of neural networks is indispensable to the normal central nervous system operation. The establishment and maintenance of such a network require precise regulation of neuronal dendrites and dendritic spine growth. The connections between them are dependent on the synaptic genesis. Apart from the morphology of the synapses that is composed of synapses structural plasticity and the number of synapses, the chemical plasticity is also closely related to synaptic density. Decrease in dendritic complexity, loss of synaptic connections, and impaired synaptic genesis are observed in major depressive disorder (MDD) patients [66,67,68]. The synaptic pathology charactered with synaptic plasticity reduction may play an important role in the onset and progression of affective disorders [67,69,70,71,72]. Many researches have confirmed that radiation can deteriorate synaptic plasticity [73,74]. For example, in 2022, Wu, M. Y. Et. Al. observed a remarkable reduction in peak discharge and excitatory synaptic inputs in hippocampal CA1 pyramidal neurons, while inhibitory inputs were greatly enhanced in the mice after receiving 10 min of cranial irradiation at a dose rate of 3 Gy/min. Corresponding to electrophysiological outcomes, they found suppressed expression of one synaptic plasticity marker called VGLUT1, a vesicular excitatory neurotransmitter transporter, along with the increased expression of VGAT, a vesicular inhibitory neurotransmitter transporter. In addition, in the irradiated mice, long-term enhancement of the hippocampus was diminished and GluR1 expression was suppressed, which suggested that radiation compromises not only the intrinsic excitability but also synaptic plasticity of hippocampal CA1 pyramidal neurons [75]. In 2013, Parihar, V. K. Et al. found that cranial radiation (1 and 10 Gy) drastically reduced the complexity of dendritic structures in mice 10 and 30 days after exposure. Crucial neuron morphology index including dendritic branches, length, and area degenerated in a dose-dependent way (>50%). Under the same dose and time, the structure of hippocampal dentate gyrus neuron dendritic spines was also seriously deteriorated, namely a 20–35% reduction in the number accompanied with a 40–70% decrease in density [76]. In contrast to the previously found long-term loss of dendritic spines, Duman, JG et al. in 2018 demonstrated that the acute response to radiotherapy was an increase in spinous processes and excitatory synapses before a spinal synaptic density reduction. Therefore, reversing the synaptic remodeling may become a promising therapeutic strategy for preventing the acute radiation-induced toxicity, and the memantine is one of the representative drugs [77]. Several previous researches have demonstrated that cranial IR alters LTP, N-methyl-D-aspartate receptor subunits, and glutamine transportability, which all play essential roles in maintaining synaptic plasticity [43,78,79]. Similarly, the abnormal glutamatergic system featured with different postsynaptic NMDA (N-methyl-D-aspartic acid) and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxalo-propionic acid) receptor subunits expression in the hippocampus and prefrontal cortex (PFC) has also been illustrated in a rat model of depression [80]. Pro-inflammatory cytokines released after radiation, such as TNF-α and IL-1, can have a direct function on neurons via binding to the extracellular domain of the cytokine receptor and then activating downstream signaling pathways, thereby altering excitability, synaptic strength, and synaptic scaling [81,82,83]. RBI may affect the transduction of neurotrophic factors and related signals by interfering with synaptic plasticity, thus contributing to post-IR mood disorders.

3.2. Chronic Inflammation

Although the specific mechanism of RBI remains unclear, it is generally believed that chronic inflammation and oxidative stress play an essential role in the occurrence and progression of RBI. During the inflammation, no matter acute or chronic phase, the mRNA expression of pro-inflammatory mediators, such as tumor necrosis factor-α (TNF-α), Interleukin (IL-1β, IL-4, IL-6, and IL-8), Monocyte chemoattractant protein 1 (MCP-1), Inducible nitric oxide synthase (iNOS) and Intercellular adhesion molecule 1 (ICAM-1) was substantially upregulated in the mice brain [83,84,85]. Increased levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), CCl2, C-reactive protein (CRP), and TNF-α in the blood and cerebrospinal fluid were also detected in the patients haunting by depression. The “depression cytokine hypothesis” implies that these inflammatory cytokines play a crucial part in developing depression [86,87,88,89,90,91]. According to the above interesting hypothesis, alternations in cytokine-induced reward pathways and dopamine neurotransmission may be the underlying mechanism of mood disorders after RBI.
Brain irradiation triggers an inflammatory storm including the over-activation of microglia and excessive secretion of inflammatory cytokines by macrophages [88,92]. The total population of microglia decreased after IR, while the activated microglia upregulated. Similar results were also found in the postnatal rats 7 days after a single 8 Gy brain irradiation and in the elderly rats receiving 10 Gy whole-brain radiation [93,94]. And there is evidence that depression could be deemed as a microglia-mediated chronic neuroinflammation disease [95,96,97]. Setiawan, E. et al. found that in MDD patients, transporter density, as measured by distributed volume (TSPO VT), increased in activated microglia, which is a distinctive feature of neuroinflammation [96]. Therefore, one of the possible mechanisms is the activation of microglia induced by radiation-induced chronic inflammation. The decreased levels of neurotrophic factors in inactive microglia combined with the activated microglia stimulated by pro-inflammatory mediators lead to hippocampal dysfunction. Activation of microglia also modulates noradrenergic and serotonergic neurotransmission and can induce depressive- and anxiety-like behaviors through the NLRP3 inflammasome/IL-1β signaling pathway [97,98,99]. Additionally, behavioral tests supplemented by magnetic resonance imaging and immunohistochemistry yielded consistent results showing radiation-induced post-necrotic mood disorders and microglial activation in the right primary somatosensory cortex of Fischer rats [100]. The decline in the number of microglia in adult mouse brains 30 and 90 days after a single 10 Gy irradiation was consistent with the occurrence of depression-like behavior [24]. In a word, activated microglia may be one of the crucial causes of mood disorder after radiotherapy.
Ionizing radiation produces reactive oxygen species (ROS) that break DNA double strands, followed by the spread of inflammatory responses involving cytokines and chemokines [36]. Inflammation and oxidative stress are concerned with the injury of glutamate neurotransmitters. Astrocytes release chemical transmitters (including glutamate) in a calcium-dependent manner. Cytokines can suppress the expression of glutamate transporters in astrocytes and increase the release of glutamate [101]. Notably, the glutamate released by astrocytes preferentially binds to the extracellular domain of NMDA receptors, leading to reduced production of trophic factors (including BDNF), ultimately compromising neuronal integrity [102,103]. Associated with depression is increased glutamate content in the frontal cortex of patients with mood disorders, along with evidence of increased microglia activation and QUIN expression [98,104,105,106]. Dysregulation of inflammatory cytokine production and signaling in the brain can contribute to cognitive and emotional deficits by activating inflammation-related enzymes such as indoleamine 2,3-dioxygenase (IDO), which are expressed by activated astrocytes, microglia, and infiltrating macrophages [107,108,109]. IDO is a tryptophan degradation enzyme, and inflammatory cytokines can enhance its activity. In chronic inflammation, the over-activated IDO accelerate the consumption of tryptophan, one raw material for serotonin synthesis. Activation of IDO facilitates the tryptophan metabolic reprogramming that converts the tryptophan into the kynurenine pathway instead of generating serotonin, producing neurotoxic metabolites such as 3-hydroxykynurenine, quinolinic acid [108,110,111,112]. Besides their stimulant toxicity, these anxiety- and depression-related metabolites also activate monoamine oxidase (MAO), the enzymatic target of antidepressants that degrades serotonin (5-HT), dopamine (DA), and norepinephrine (NE) [90,113]. In the pathogenesis and treatment of depression, IDO have the potential to become a new pharmacological research target. However, whether it is a crucial factor or just a participating factor in the complex mechanism of depression remains to be further studied. This association has not been observed in radiation-induced animal models. However, clinical studies have shown significantly increased IDO activity (calculated by the Kyn/Trp ratio) in non-small cell lung cancer patients after receiving a biologically equivalent dose (BED) > 70 Gy, compared to pre-radiotherapy levels [114]. Similarly, cytokines may also disturb the synthesis of dopamine DA by reducing the bioavailability of tetrahydrobiopterin (BH4). BH4, a key enzyme cofactor involved in the production of dopamine DA, is highly sensitive to REDOX and easily oxidized to dihydrobiopterin (BH2). What is more, this process is irreversible [115,116]. Cytokines also have an impact on dopamine packaging, release, and reuptake. Dopamine relies on vesicular monoamine transporter 2 (VMAT2) to encapsulate cytoplasmic dopamine for release. There is evidence that inflammatory cytokines (IL-1β and TNF-α) can impair VMAT2 function [117]. It is worth noting that mitochondrion, an important oxidative stress victim, has also become a significant target for inflammation-related damage. The upregulation of radiation-induced pro-inflammatory factors (including IL-6, TNF-α, and ROS) impairs mitochondrial function and induces interferon-α (IFN-α) production, which exhibits both neuropsychological and neurotoxic effects [118,119,120,121]. These factors together influence hippocampal neurogenesis and lead to behavioral dysfunction. Studies have demonstrated that decreasing levels of TNF-α, IL-6, iNOS, and IL-1β can mitigate neuropsychiatric symptom severity. Furthermore, inhibition of the STAT3/NF-κB pathway activity—a key inflammatory regulatory pathway—also ameliorates these symptoms [122]. More detailed researches are needed to understand the mechanisms of radiation-induced neuroinflammation and its exact role in affective disorders.

3.3. Other Possible Mechanisms

In addition, there are some other mechanisms, including vascular damage, nerve demyelination, and endocrine changes, caused by RBI [123,124]. Brain irradiation exerts profound effects on the cerebrovasculature, potentially leading to delayed radiation-induced necrosis [125]. Ten to fifty-two weeks after irradiation, the dose of 5 Gy/ fraction (8 fractions in total) can reduce the endothelial cells population, vascular density, and vascular length [126]. Blood–brain barrier permeability and vascular endothelial growth factor (VEGF) expression increased within 24 h after exposure to 6 Gy radiation [127]. Inflammatory factors induced by radiotherapy, such as iNOS, also increase BBB permeability [128]. The high permeability of the blood–brain barrier enables the peripheral inflammatory cytokines enter the cerebrospinal fluid and these inflammatory factors in turn impair the integrity of the blood–brain barrier. This positive feedback loop results in a cascade neuroinflammatory response, finally leading to emotional disorders.
In a model of chronic depression induced by stress, loss of myelin sheath may play an indispensable role shown by diffusion tensor imaging [129]. Also, several studies have demonstrated that depression and anxiety are common symptoms of demyelinating diseases such as epilepsy and multiple sclerosis [130,131,132]. One of the main pathological manifestations of RBI is the demyelination of oligodendrocytes, so it is speculated that radiotherapy can cause affective disorders through demyelination as well [36]. Conpanzo et al. found that ruptured and demyelinated splenial fiber bundles in the right hemisphere of Fischer rats 54 days post-irradiation, along with selective behavioral changes (including depressive- and anxiety-like behaviors), could be attributed to these ultrastructural lesions [100]. In summary, the underlying mechanism of emotional change can be ascribed to the neuron connectivity alternations between the limbic area and the PFC [129].
Radiotherapy can distinctly reduce the secretion of melatonin in the pineal gland of patients, aggravating severe sleep difficulties and prevalence of depression [133]. Therefore, the occurrence of depression and anxiety may be explained by the post-radiotherapy neuroendocrine changes. Studies have reported that radiation affects the functions of the growth hormone/insulin-like growth factor-I axis, restricting physical and mental development [134]. Moreover, radiation can markedly reduce the rate of glucose metabolism in the temporal and occipital lobes. Therefore, brain metabolic suppression is proposed to become one of the possible ways to induce metabolic depression [135]. For example, depression and anxiety are representative clinical manifestations of post-cranial irradiation somnolence syndrome [136,137]. The pathophysiology of somnolence syndrome may be associated with the transient demyelination of white matter or the nonspecific effects of chronic diseases and poor emotional perception.

4. Potential Diagnostic, Assessment, and Preventive Strategies

4.1. Early Neuropsychological Assessment

Traditional psychiatric assessment tools, such as the hospital anxiety and depression scale (HADS), are effective in screening for anxiety and depressive symptoms caused by radiation-induced brain injury. However, they exhibit insufficient sensitivity in detecting subclinical cognitive dysfunctions resulting from such damage. Given that the onset of radiation-induced anxiety and depression is closely associated with hippocampal dysfunction, evaluating hippocampal injury plays a crucial role in the early diagnosis of these conditions.
The Wechsler memory scale-revised (WMS-R) is a widely used neuropsychological tool for assessing an individual’s memory function, including short-term memory, long-term memory, and working memory [138]. It measures various memory dimensions through multiple core subscales, such as attention, general memory, visual memory, verbal memory, and delayed memory [139]. Its standardized scoring system uses the memory quotient (MQ) to quantify results, with an MQ score of 80 or above indicating normal functioning. Scores between 70 and 79 suggest a borderline condition, while scores of 69 or below indicate impairment. Moreover, lower scores correspond to more severe deficits. The WMS-R has significant clinical value in evaluating hippocampal damage [140]. Numerous studies have found that patients with hippocampal damage often have very low WMS-R scores, particularly in delayed memory (less than 50) [141,142,143]. The California verbal learning test (CVLT) is a widely used tool in clinical and neuropsychological assessments, primarily aimed at evaluating an individual’s verbal learning and memory abilities [144]. This test involves a series of tasks, including immediate recall, interference list, short-delay free recall, and cued recall, to assess memory performance and learning strategies at various stages [144]. The CVLT holds substantial practical value in clinical settings [145]. It aids clinicians in identifying and evaluating various neuropsychological disorders, such as Alzheimer’s disease (AD), traumatic brain injury, and others [146,147].
Although no studies have yet specifically applied the WMS-R and the CVLT for the early identification of RBI and its associated anxiety and depression, these neuropsychological assessment tools hold significant potential for future applications. Given their role in assessing hippocampal dysfunction, they may be valuable in the early detection of RBI and related psychological symptoms.

4.2. Advanced Imaging Techniques

With the rapid advancement of neuroimaging technologies, many advanced imaging techniques have provided new breakthroughs in understanding the mechanisms of radiation-induced brain injury. These techniques offer unique insights into the structural and functional abnormalities of the hippocampus, providing important evidence for clinical diagnosis, evaluation, and treatment planning.
Resting-state functional magnetic resonance imaging (rs-fMRI) detects spontaneous neural activity in the brain, enabling precise assessment of the functional connectivity between the hippocampus and the default mode network [148]. Task-based fMRI, on the other hand, directly observes hippocampal functional activation patterns by designing specific memory encoding or emotional processing tasks [149]. In patients with AD, rs-fMRI can detect early abnormalities in hippocampal activity, while task-based fMRI demonstrates characteristic hippocampal activation reduction, often preceding clinical symptoms [150,151]. Additionally, studies have shown that after whole-brain radiotherapy (WBRT), patients exhibit widespread hyperconnectivity in rs-fMRI, along with significant increases in global modularity and local network variability, which are strongly correlated with cognitive decline [152].
Arterial Spin Labeling (ASL) Perfusion Imaging provides a new dimension for assessing the hippocampus. This technique does not require exogenous contrast agents and allows for the repeated measurement of cerebral blood flow (CBF) in the hippocampal region [153]. In patients with radiation-induced brain injury, ASL-CBF values are typically lower, indicating reduced cerebral blood perfusion [154]. Magnetic Resonance Spectroscopy (MRS) quantifies the concentration of metabolites such as N-acetylaspartate (NAA) and myo-inositol, which can sensitively reflect the degree of neuronal damage [155]. MRS exhibits higher detection sensitivity for radiation-induced brain injury, with a characteristic increase in the NAA/Choline (Cho) ratio in affected patients [156,157]. Fluorodeoxyglucose Positron Emission Tomography (FDG-PET) detects changes in glucose metabolism and can identify early hippocampal functional abnormalities [158]. Studies have shown that a reduction in central nervous system glucose metabolism following brain radiotherapy is closely associated with cognitive decline [159].
Advanced imaging technologies hold significant value in the clinical assessment of radiation-induced brain injury. By integrating these advanced techniques, a comprehensive evaluation of the neurobiological changes in brain injury can be achieved from multiple dimensions, thereby significantly enhancing the sensitivity and specificity of early diagnosis [152,154,156,157,159]. The complementary application of these techniques not only helps to distinguish radiation-induced brain injury from neurodegenerative diseases such as AD but also provides objective evidence for treatment monitoring and prognostic assessment [150,151,152]. In the clinical context of radiation-induced brain injury-related anxiety and depression, these advanced imaging techniques can be applied to patients with depressive and anxiety symptoms following brain radiotherapy. They reveal the specific neurobiological alterations in such patients, enabling effective differentiation between secondary and primary mood disorders and predicting the risk of symptom progression, thus offering objective support for personalized treatment strategies [160].

4.3. Hippocampus Avoidance Radiotherapy Strategy

Hippocampal Avoidance Whole-Brain Radiotherapy (HA-WBRT) is an innovative radiation therapy technique designed to reduce the risk of cognitive dysfunction induced by conventional WBRT by precisely avoiding the hippocampal region. [161]. This technique primarily relies on advanced radiation therapy methods, such as Intensity-Modulated Radiotherapy (IMRT). IMRT dynamically adjusts the intensity distribution of the radiation beams to ensure adequate dose delivery to the tumor target while minimizing exposure to critical neural structures, such as the hippocampus [162,163]. In clinical practice, the hippocampal anatomy is accurately located using MRI-CT fusion imaging, and a 5–10 mm safety margin is set around it as the avoidance region. Treatment planning was performed to deliver 30 Gy in 10 fractions to the whole brain while maintaining hippocampal dose below protocol-specified limits (D100% < 9 Gy, Dmax < 16 Gy) in accordance with RTOG 0933 guidelines [164]. Studies have shown that this approach can reduce the average radiation dose to the hippocampus by up to 87%, significantly outperforming traditional radiotherapy techniques [163]. Furthermore, other technologies such as Volumetric Modulated Arc Therapy (VMAT) can also be utilized within HA-WBRT to enhance treatment precision and efficiency [165].
In terms of neurocognitive protection, HA-WBRT has demonstrated clear short-term and long-term cognitive benefits [161]. Studies have shown that no significant cognitive improvement is observed within two months post-radiotherapy, but protective effects gradually emerge starting from three months [161,166]. At three months, significant differences were noted in memory/verbal learning (p < 0.0001) and overall cognitive function (p = 0.001) [166,167,168]. At six months, improvements in memory/verbal learning continued (p < 0.1) [161,167,169], and at 9–24 months, overall cognitive function maintained a significant advantage (p < 0.05) [168,170]. It is noteworthy that the improvement in memory function shows subtype specificity, with long-term memory benefiting more significantly than short-term memory [171]. Additionally, different cognitive dimensions exhibit varied responses to HA-WBRT. This technique is particularly effective in enhancing overall cognitive function, memory, and verbal learning, but improvements in executive function, visual learning, attention, and information processing speed are relatively limited [172].
In terms of survival outcomes, HA-WBRT demonstrates comparable efficacy to conventional WBRT. Long-term follow-up data from several key clinical trials confirm that there are no significant statistical differences between the two treatment modalities in key survival indicators, such as overall survival (OS) and progression-free survival (PFS) [161,167,169,173]. These findings strongly suggest that, despite the significant reduction in radiation dose to the hippocampal region, HA-WBRT does not compromise tumor control. The clinical evidence provides strong support for the dual goals of HA-WBRT, achieving both neurocognitive protection and survival benefits.
Although existing studies primarily focus on cognitive outcomes, given the hippocampus’s close involvement in emotional regulation, HA-WBRT may theoretically improve mood symptoms by preserving hippocampal structural integrity [174]. However, direct evidence for improvements in depression and anxiety is currently lacking, and prospective studies combining emotional scales with hippocampal volume/function MRI parameters are needed for systematic evaluation. Overall, HA-WBRT represents a significant advancement in neuroprotective radiotherapy, and its potential value in emotional regulation suggests that it could serve as a promising strategy for preventing radiation-induced brain damage leading to depression and anxiety. Further validation of its comprehensive clinical benefits through multi-center randomized controlled trials is warranted.

5. Potential Treatments

While multiple therapeutic interventions exist for RBI, current evidence regarding their efficacy in managing associated depression and anxiety remains limited. This review systematically evaluates pharmacological agents with demonstrated effectiveness in treating RBI-related mood disorders.

5.1. Inhibitors of Glycogen Synthase

Glycogen synthase kinase 3 (GSK-3) is a serine/threonine kinase composed of two subunits, namely GSK-3α and GSK-3β [175,176]. It mainly phosphorylates Tau protein to increase Tau protein content in neuronal fibers and causes neuronal defects together with β-amyloid plaques, which are associated with neurodegenerative diseases such as Alzheimer and Parkinson [177,178]. As a result, GSK-3 has emerged as a promising therapeutic target for neurodegenerative diseases. Recent studies have demonstrated the efficacy of synthetic GSK-3 inhibitors in mitigating AD, Parkinson’s disease (PD), mood disorders, and other neurological conditions [179,180,181]. Lithium is the first identified GSK3 inhibitor, which can inhibit stem cell or progenitor cell death, attenuate neuroinflammation, improve synaptic plasticity, stimulate stem cell and progenitor cell proliferation and neurogenesis, and thus play a powerful neuroprotective role after cranial irradiation [182,183,184,185,186,187]. Even better, lithium not only does not protect cancer cells against radiotherapy but may serve as a radiation sensitizer in some cases [188,189]. In a radiation-induced rodent model, lithium treatment attenuated radiation-induced progenitor cell death in the hippocampal subgranular zone, enhanced neurogenesis and astrocytogenesis in juvenile rats, and ameliorated radiation-associated anxiety-like behaviors [30]. Thus, glycogen synthase inhibitors are undoubtedly effective agents for treating post-radiotherapy mood disorders, but further investigations are still needed to elucidate the efficacy of drugs other than lithium.

5.2. Fluoxetine

Fluoxetine, a selective serotonin reuptake inhibitor (SSRI) antidepressant, has been widely used to treat the adult major depressive disorder, obsessive-compulsive disorder, bulimia neuropathy in clinical. Also, fluoxetine may play a protective role in cognitive disorders such as vascular dementia through increasing neurogenesis in the dentate gyrus of the hippocampus, enhancing synaptic plasticity, and upregulating the expression of BDNFs in the hippocampus [190,191,192,193]. A study reported that 5HT1A serotonin receptor levels declined by 50 percent in both early and late hippocampi, and a reduction of 37 percent in serotonin levels was observed at 15 weeks after radiotherapy. Chronic fluoxetine treatment was adequate to reverse depression-like behavior induced by the combined chemoradiotherapy [27]. One study found that mice with 6 weeks temozolomide treatment after single cranial irradiation of 9Gy spent less time in open arms during the elevated plus maze tests, whereas fluoxetine reversed this anxiety-like behavior [194]. Although studies have confirmed the efficacy of fluoxetine in alleviating post-radiation anxiety, the potential impact of temozolomide in combined interventions should not be overlooked. Further research is needed to clarify the relationship between post-radiation anxiety and fluoxetine response and to elucidate its underlying mechanisms. Additionally, certain side effects warrant careful consideration, including sexual dysfunction, manic switching, and even persistent pulmonary hypertension [195,196].

5.3. Allantoin and Neferine

Retinol and its major metabolite retinoic acid (RA) play a crucial role in hippocampal neurogenesis, anxiety-like behavior, and hippocampal-dependent memory in adulthood [197,198,199,200]. Moreover, dysregulated retinol signaling pathway is associated with neurological diseases including cerebral ischemia and AD, which are featured with age-related memory decline, long-term hippocampal enhancement, and neurogenesis [201,202,203,204]. Overexpression of TTR inhibits retinol-mediated neurogenesis. Allantoin and neferine, the active constituents of lotus, enhance retinol-mediated neuronal maturation via repressing TTR expression and promoting PAK1 phosphorylation. Studies have shown that these toxic-free natural substances can relieve depressive symptoms after craniocerebral radiotherapy. With the treatment of allantoin and neferine, the amount of BrdU+/Neun+ in irradiated mice dramatically reduced, which enhanced hippocampal neurogenesis and relieved neurodegeneration, thus alleviating the depressive symptoms caused by radiation [32]. Allantoin and neferine may serve as potential therapeutic agents for depression, potentially exerting antidepressant effects by mitigating hippocampal neurodegeneration caused by cranial irradiation.

5.4. Other Potential Treatment

Minocycline is a clinically available antibiotic that is charactered with neuroprotective ability in various neurodegenerative diseases through its direct antioxidant activity [205,206]. Since ionizing radiation triggers caspase-3-dependent apoptosis primarily through ROS generation, ROS scavengers such as minocycline can inhibit this apoptotic pathway in neural stem cells. Studies have shown that minocycline can significantly alleviate radiation-induced cognitive impairment and defend newborn neurons against radiation-induced apoptosis, thereby reducing the loss of new neurons [207]. In recent years, it has also been reported that minocycline can also treat emotional disorders. And inhibitory caspase-1 can regulate intestinal microorganisms to affect the inflammatory signal regulation pathway and then indirectly block the activation of IDO, thereby changing brain function and affecting depression and anxiety-like behaviors [208,209]. Therefore, it can be speculated that minocycline may be an effective drug for treating the post-radiotherapy affective disorder. P7C3 compounds are one type of neuroprotective agents that enhance hippocampal neurogenesis and have a more predominant neurogenic effect than antidepressants on the market. Its neuroprotective effects have been shown to protect people from cognitive deficits and depression-like behavior by promoting neurons to survive independent of early disease-specific pathology [210]. The study found that P7C3 compounds exhibited antidepressant effects in mice exposed to chronic social frustration, stress or caloric restriction, but ablation of hippocampal nerves by cranial irradiation could eliminate this antidepressant effect [211]. However, further clinical and animal experiments are needed to explore and prove whether it can exert an anti-depressive effect after radiotherapy. In addition to conventional medical treatments, mindfulness-based interventions (MBIs) have demonstrated significant efficacy in managing anxiety and depression symptoms among adult cancer patients. Notably, the 2023 ASCO Clinical Practice Guideline for managing anxiety and depression in adults with cancer strongly recommends various complementary interventions—including MBIs, yoga, relaxation techniques, music therapy, reflexology, and aromatherapy (via inhalation)—for alleviating anxiety symptoms during active cancer treatment. Furthermore, these non-pharmacological approaches are widely recognized for their sustained benefits in anxiety management during post-treatment survivorship care [19]. In addition to the above-mentioned interventions, the ASCO Guideline also suggests the use of MBIs, yoga, acupuncture, tai chi, and/or Qigong, along with reflexology, for treating anxiety symptoms after cancer treatment. Furthermore, for depressive symptoms during cancer treatment, the ASCO Guideline recommends the integration of MBIs, yoga, music therapy, relaxation techniques, and reflexology. These interventions have been shown to be effective in relieving symptoms of depression and promoting emotional well-being during the challenging period of cancer treatment. After cancer treatment, the ASCO Guideline emphasizes the importance of continuing MBIs, yoga, tai chi, and/or Qigong as potential treatments for depressive symptoms. In a word, these mind–body practices not only provide emotional support but also contribute to overall recovery and improved quality of life for cancer survivors in a subtle way [212].

6. Conclusions

Brain radiotherapy induced affective disorders such as depression and anxiety are predominant symptoms of the RBI. While many possible mechanisms including hippocampal dysfunction and chronic neuro-inflammation have been proposed, the research on its exact mechanism is still insufficient, and we need to rule out the spirit of comorbidities. Other confounding factors require further study to determine whether there is a causal relationship between them. Although it is hard to figure out which mechanisms directly contribute to affective disorders following radiotherapy, this review may provide insights and identify the underlying molecular mechanisms. Future research directions may include developing new protective agents to prevent or mitigate adverse reactions in patients with RBI. This review helps provide new targets for preventing emotional disorders caused by RBI to improve patients’ quality of life and extend life expectancy.

Author Contributions

G.H., Q.L., and W.Z. contributed to the concept and design; F.Y. and R.L. were responsible for manuscript writing; X.P., N.L., and M.F. performed critical revision and editing of the scientific content. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the National Natural Sciences Foundation of China (grant number 82173311 and 82303830), Hubei Provincial Natural Science Foundation of China (grant number 2023AFB727), China Postdoctoral Science Foundation (2024M761051).

Institutional Review Board Statement

All the data in this review are derived from published literature and do not involve new ethical approvals. The original ethical statements of the relevant literature can be found in their corresponding papers.

Informed Consent Statement

The case data cited in this review are all from published literature, and the written informed consent forms of the patients have been obtained for the original studies. This article does not contain any new information that can identify the patient’s identity.

Data Availability Statement

All the data cited in this review are from published literature. The relevant original data can be obtained through corresponding papers or public databases. No new experimental data were generated in this study.

Acknowledgments

The authors would like to express sincere gratitude to Guangyuan Hu for his critical guidance on this review.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have influenced the work reported in this paper.

Abbreviations

The following abbreviations are used in this manuscript:
RBIRadiation-induced brain injury
LTPLong-term potentiation
TTRThyroxine transporter protein
BDNFBrain-derived neurotrophic factor
MDDMajor depressive disorder
PFCPrefrontal cortex
TNF-αTumor necrosis factor-α
MCP-1Monocyte chemoattractant protein 1
iNOSInducible nitric oxide synthase
ICAM-1Intercellular adhesion molecule 1
IL-6Interleukin-6
IL-1βInterleukin-1β
CRPC-reactive protein
ROSReactive oxygen species
IDOIndoleamine 2,3-dioxygenase
MAOMonoamine oxidase
DADopamine
NENorepinephrine
BEDBiologically equivalent dose
VMAT2Vesicular monoamine transporter 2
IFN-αInterferon-α
VEGFVascular endothelial growth factor
HADSHospital anxiety and depression scale
WMS-RWechsler memory scale-revised
MQMemory quotient
CVLTCalifornia verbal learning test
ADAlzheimer’s disease
rs-fMRIResting-state functional magnetic resonance imaging
WBRTWhole-brain radiotherapy
ASLArterial Spin Labeling
CBFCerebral blood flow
MRSMagnetic Resonance Spectroscopy
NAAN-acetylaspartate
ChoCholine
FDG-PETFluorodeoxyglucose Positron Emission Tomography
HA-WBRTHippocampal Avoidance Whole-Brain Radiotherapy
IMRTIntensity-Modulated Radiotherapy
VMATVolumetric Modulated Arc Therapy
OSOverall survival
PFSProgression-free survival
GSK-3Glycogen synthase kinase 3
PDParkinson’s disease
SSRISelective serotonin reuptake inhibitor
RARetinoic acid
MBIsMindfulness-based interventions

References

  1. Bishop, A.J.; McDonald, M.W.; Chang, A.L.; Esiashvili, N. Infant Brain Tumors: Incidence, Survival, and the Role of Radiation Based on Surveillance, Epidemiology, and End Results (Seer) Data. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 341–347. [Google Scholar] [CrossRef]
  2. Yu, J.; Shi, W.E.; Zhao, R.; Gao, X.; Li, H. Epidemiology of Brain Tumors in Children Aged Two and Under: A 10-Year Single-Institute Study. Oncol. Lett. 2015, 9, 1651–1656. [Google Scholar] [CrossRef]
  3. Johnson, J.D.; Young, B. Demographics of Brain Metastasis. Neurosurg. Clin. N. Am. 1996, 7, 337–344. [Google Scholar] [CrossRef]
  4. Tanzhu, G.; Chen, L.; Ning, J.; Xue, W.; Wang, C.; Xiao, G.; Yang, J.; Zhou, R. Metastatic Brain Tumors: From Development to Cutting-Edge Treatment. MedComm 2025, 6, e70020. [Google Scholar] [CrossRef]
  5. Ewend, M.G.; Elbabaa, S.; Carey, L.A. Current Treatment Paradigms for the Management of Patients with Brain Metastases. Neurosurgery 2005, 57, S66–S77; discussion S71–S74. [Google Scholar] [CrossRef]
  6. Peacock, K.H.; Lesser, G.J. Current Therapeutic Approaches in Patients with Brain Metastases. Curr. Treat. Options Oncol. 2006, 7, 479–489. [Google Scholar] [CrossRef]
  7. He, Y.Q.; Wang, T.M.; Yang, D.W.; Xue, W.Q.; Deng, C.M.; Li, D.H.; Zhang, W.L.; Liao, Y.; Xiao, R.W.; Luo, L.T.; et al. A Comprehensive Predictive Model for Radiation-Induced Brain Injury in Risk Stratification and Personalized Radiotherapy of Nasopharyngeal Carcinoma. Radiother. Oncol. 2024, 190, 109974. [Google Scholar] [CrossRef]
  8. Jiménez-Labaig, P.; Aymerich, C.; Rullan, A.; Cacicedo, J.; Braña, I.; Nutting, C.; Newbold, K.; Harrington, K.J.; Catalan, A. Prevalence of Depressive and Anxiety Symptoms in Patients with Head and Neck Cancer Undergoing Radiotherapy: A Systematic Review and Meta-Analysis of Longitudinal Studies. Radiother. Oncol. 2025, 202, 110649. [Google Scholar] [CrossRef]
  9. Jovanovic Ristivojevic, J.; Jovanovic Korda, N.; Vujanac, V.; Nikitovic, M.; Arsenijevic, T. Non-Endemic Non-Keratinizing Nasopharyngeal Carcinoma: Long-Term Toxicity Following Chemoradiation. Oncol. Lett. 2025, 29, 283. [Google Scholar] [CrossRef]
  10. Tang, Y.; Luo, D.; Rong, X.; Shi, X.; Peng, Y. Psychological Disorders, Cognitive Dysfunction and Quality of Life in Nasopharyngeal Carcinoma Patients with Radiation-Induced Brain Injury. PLoS ONE 2012, 7, e36529. [Google Scholar] [CrossRef]
  11. Pereira Dias, G.; Hollywood, R.; Bevilaqua, M.C.; da Luz, A.C.; Hindges, R.; Nardi, A.E.; Thuret, S. Consequences of Cancer Treatments on Adult Hippocampal Neurogenesis: Implications for Cognitive Function and Depressive Symptoms. Neuro-Oncology 2014, 16, 476–492. [Google Scholar] [CrossRef]
  12. Roy-Byrne, P. Treatment-Refractory Anxiety; Definition, Risk Factors, and Treatment Challenges. Dialogues Clin. Neurosci. 2015, 17, 191–206. [Google Scholar] [CrossRef]
  13. van’t Spijker, A.; Trijsburg, R.W.; Duivenvoorden, H.J. Psychological Sequelae of Cancer Diagnosis: A Meta-Analytical Review of 58 Studies after 1980. Sci. Med. 1997, 59, 280–293. [Google Scholar] [CrossRef]
  14. Wong, E.; Rowbottom, L.; Tsao, M.; Zhang, L.; McDonald, R.; Danjoux, C.; Barnes, E.; Chan, S.; Chow, E. Correlating Symptoms and Their Changes with Survival in Patients with Brain Metastases. Ann. Palliat. Med. 2016, 5, 253–266. [Google Scholar] [CrossRef]
  15. Oh, Y.; Seo, H.; Sung, K.W.; Joung, Y.S. The Effects of Attention Problems on Psychosocial Functioning in Childhood Brain Tumor Survivors: A 2-Year Postcraniospinal Irradiation Follow-Up. J. Pediatr. Hematol. Oncol. 2017, 39, e46–e53. [Google Scholar] [CrossRef]
  16. Shekelle, R.B.; Raynor, W.J., Jr.; Ostfeld, A.M.; Garron, D.C.; Bieliauskas, L.A.; Liu, S.C.; Maliza, C.; Paul, O. Psychological Depression and 17-Year Risk of Death from Cancer. Psychosom. Med. 1981, 43, 117–125. [Google Scholar] [CrossRef]
  17. Yuksek, E.; Eroz, S.; Yassa, A.; Akturk, D.; Zakirov, F.; Akcam, F.E.; Emul, M. The Influences of Whole Brain Radiotherapy on Social Cognition and Association with Hippocampal and Frontal Dosimetry. Psychiatr. Q. 2015, 86, 533–543. [Google Scholar] [CrossRef]
  18. Wang, C.; Chen, J.; Su, L.; Hua, Y.; Ye, J.; Song, X.; Lv, W.; Zhang, M.; Huang, F.; Tian, J.; et al. The Psychological Status in Patients with Nasopharyngeal Carcinoma During Radiotherapy. Eur. Arch. Oto-Rhino-Laryngol. 2022, 279, 1035–1042. [Google Scholar] [CrossRef]
  19. Sharma, N.; Purkayastha, A. Prevalence of Anxiety and Depression in Cancer Patients During Radiotherapy: A Rural Indian Perspective. J. Cancer Res. Ther. 2021, 17, 218–224. [Google Scholar] [CrossRef]
  20. Unnikrishnan, S.; Karunamuni, R.; Salans, M.A.; Gudipati, S.; Qian, A.S.; Yu, J.; Connor, M.; Huynh-Le, M.-P.; Tibbs, M.D.; Hermann, G.; et al. Dose-Dependent Atrophy in Bilateral Amygdalae and Nuclei after Brain Radiation Therapy and Its Association with Mood and Memory Outcomes on a Longitudinal Clinical Trial. Int. J. Radiat. Oncol. Biol. Phys. 2023, 117, 834–845. [Google Scholar] [CrossRef]
  21. You, H.; He, L.; Ouyang, Z.; Yang, Y.; Xie, S.; Zhou, J.; Zhang, Y.; Shi, J. Case Report: Intracranial Lesions in a Patient with Anxiety and Depression: Tumor Recurrence or Radiation Encephalopathy? Front. Oncol. 2024, 14, 1422765. [Google Scholar] [CrossRef]
  22. Miyachi, Y.; Yamada, T. Low-Dose X-Ray-Induced Depression of Sexual Behavior in Mice. Behav. Brain Res. 1994, 65, 113–115. [Google Scholar] [CrossRef]
  23. Lowe, X.R.; Bhattacharya, S.; Marchetti, F.; Wyrobek, A.J. Early Brain Response to Low-Dose Radiation Exposure Involves Molecular Networks and Pathways Associated with Cognitive Functions, Advanced Aging and Alzheimer’s Disease. Radiat. Res. 2009, 171, 53–65. [Google Scholar] [CrossRef]
  24. Son, Y.; Yang, M.; Kim, J.S.; Kim, J.; Kim, S.H.; Kim, J.C.; Shin, T.; Wang, H.; Jo, S.K.; Jung, U.; et al. Hippocampal Dysfunction During the Chronic Phase Following a Single Exposure to Cranial Irradiation. Exp. Neurol. 2014, 254, 134–144. [Google Scholar] [CrossRef]
  25. Ueno, H.; Suemitsu, S.; Murakami, S.; Kitamura, N.; Wani, K.; Matsumoto, Y.; Okamoto, M.; Ishihara, T. Region-Specific Reduction of Parvalbumin Neurons and Behavioral Changes in Adult Mice Following Single Exposure to Cranial Irradiation. Int. J. Radiat. Biol. 2019, 95, 611–625. [Google Scholar] [CrossRef]
  26. Smart, D. Radiation Radiation Toxicity in the Central Nervous System: Mechanisms and Strategies for Injury Reduction. Semin. Radiat. Oncol. 2017, 27, 332–339. [Google Scholar] [CrossRef]
  27. Dey, D.; Parihar, V.K.; Szabo, G.G.; Klein, P.M.; Tran, J.; Moayyad, J.; Ahmed, F.; Nguyen, Q.-A.; Murry, A.; Merriott, D.; et al. Neurological Impairments in Mice Subjected to Irradiation and Chemotherapy. Radiat. Res. 2020, 193, 407–424. [Google Scholar] [CrossRef]
  28. Brown, R.J.; Jun, B.J.; Cushman, J.D.; Nguyen, C.; Beighley, A.H.; Blanchard, J.; Iwamoto, K.; Schaue, D.; Harris, N.G.; Jentsch, J.D.; et al. Changes in Imaging and Cognition in Juvenile Rats after Whole-Brain Irradiation. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 470–478. [Google Scholar] [CrossRef]
  29. Trivedi, R.; Khan, A.R.; Rana, P.; Haridas, S.; Hemanth Kumar, B.S.; Manda, K.; Rathore, R.K.; Tripathi, R.P.; Khushu, S. Radiation-Induced Early Changes in the Brain and Behavior: Serial Diffusion Tensor Imaging and Behavioral Evaluation after Graded Doses of Radiation. J. Neurosci. Res. 2012, 90, 2009–2019. [Google Scholar] [CrossRef]
  30. Zhou, K.; Xie, C.; Wickström, M.; Dolga, A.M.; Zhang, Y.; Li, T.; Xu, Y.; Culmsee, C.; Kogner, P.; Zhu, C.; et al. Lithium Protects Hippocampal Progenitors, Cognitive Performance and Hypothalamus-Pituitary Function after Irradiation to the Juvenile Rat Brain. Oncotarget 2017, 8, 34111–34127. [Google Scholar] [CrossRef]
  31. Wong-Goodrich, S.J.; Pfau, M.L.; Flores, C.T.; Fraser, J.A.; Williams, C.L.; Jones, L.W. Voluntary Running Prevents Progressive Memory Decline and Increases Adult Hippocampal Neurogenesis and Growth Factor Expression after Whole-Brain Irradiation. Cancer Res. 2010, 70, 9329–9338. [Google Scholar] [CrossRef]
  32. Kang, J.; Kim, W.; Seo, H.; Kim, E.; Son, B.; Lee, S.; Park, G.; Jo, S.; Moon, C.; Youn, H.; et al. Radiation-Induced Overexpression of Transthyretin Inhibits Retinol-Mediated Hippocampal Neurogenesis. Sci. Rep. 2018, 8, 8394. [Google Scholar] [CrossRef]
  33. Roughton, K.; Kalm, M.; Blomgren, K. Sex-Dependent Differences in Behavior and Hippocampal Neurogenesis after Irradiation to the Young Mouse Brain. Eur. J. Neurosci. 2012, 36, 2763–2772. [Google Scholar] [CrossRef]
  34. Acevedo, S.E.; McGinnis, G.; Raber, J. Effects of 137cs Gamma Irradiation on Cognitive Performance and Measures of Anxiety in Apoe-/- and Wild-Type Female Mice. Radiat. Res. 2008, 170, 422–428. [Google Scholar] [CrossRef]
  35. Wang, Y.; Bao, X.; Zhang, Y.; Wu, Q. The Current Research Status of the Mechanisms and Treatment of Radioactive Brain Injury. Am. J. Cancer Res. 2024, 14, 5598–5613. [Google Scholar] [CrossRef]
  36. Ali, F.S.; Arevalo, O.; Zorofchian, S.; Patrizz, A.; Riascos, R.; Tandon, N.; Blanco, A.; Ballester, L.Y.; Esquenazi, Y. Cerebral Cerebral Radiation Necrosis: Incidence, Pathogenesis, Diagnostic Challenges, and Future Opportunities. Curr. Oncol. Rep. 2019, 21, 66. [Google Scholar] [CrossRef]
  37. Li, Y.D.; Luo, Y.J.; Chen, Z.K.; Quintanilla, L.; Cherasse, Y.; Zhang, L.; Lazarus, M.; Huang, Z.L.; Song, J. Hypothalamic Modulation of Adult Hippocampal Neurogenesis in Mice Confers Activity-Dependent Regulation of Memory and Anxiety-Like Behavior. Nat. Neurosci. 2022, 25, 630–645. [Google Scholar] [CrossRef]
  38. Ogrodnik, M.; Zhu, Y.; Langhi, L.G.P.; Tchkonia, T.; Kruger, P.; Fielder, E.; Victorelli, S.; Ruswhandi, R.A.; Giorgadze, N.; Pirtskhalava, T.; et al. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019, 29, 1233. [Google Scholar] [CrossRef]
  39. Li, K.; Koukoutselos, K.; Sakaguchi, M.; Ciocchi, S. Distinct Ventral Hippocampal Inhibitory Microcircuits Regulating Anxiety and Fear Behaviors. Nat. Commun. 2024, 15, 8228. [Google Scholar] [CrossRef]
  40. Rao, A.A.; Ye, H.; Decker, P.A.; Howe, C.L.; Wetmore, C. Therapeutic Doses of Cranial Irradiation Induce Hippocampus-Dependent Cognitive Deficits in Young Mice. J. Neuro-Oncol. 2011, 105, 191–198. [Google Scholar] [CrossRef]
  41. Winocur, G.; Wojtowicz, J.M.; Sekeres, M.; Snyder, J.S.; Wang, S. Inhibition of Neurogenesis Interferes with Hippocampus-Dependent Memory Function. Hippocampus 2006, 16, 296–304. [Google Scholar] [CrossRef]
  42. Lopez-Virgen, V.; Gonzalez-Morales, O.; Gonzalez-Perez, O. The Ventricular-Subventricular, Subgranular and Subcallosal Zones: Three Niches of Neural Stem Cells in the Postnatal Brain. Exp. Brain Res. 2023, 241, 1463–1470. [Google Scholar] [CrossRef]
  43. Zanni, G.; Zhou, K.; Riebe, I.; Xie, C.; Zhu, C.; Hanse, E.; Blomgren, K. Irradiation of the Juvenile Brain Provokes a Shift from Long-Term Potentiation to Long-Term Depression. Dev. Neurosci. 2015, 37, 263–272. [Google Scholar] [CrossRef]
  44. Leskinen, S.; Alsalek, S.; Wernicke, A.G. Effects of Radiotherapy on the Hippocampus and Hippocampal Neurogenesis: A systematic Review of Preclinical Studies. Strahlenther. Onkol. 2025, 201, 383–397. [Google Scholar] [CrossRef]
  45. Mateus-Pinheiro, A.; Pinto, L.; Sousa, N. Epigenetic (De)Regulation of Adult Hippocampal Neurogenesis: Implications for Depression. Clin. Epigenet. 2011, 3, 5. [Google Scholar] [CrossRef]
  46. Cheng, K.; Chen, C.; Zhou, Q.; Chen, X.; Xie, P. Deficit of Neuronal Eaat2 Impairs Hippocampus Ca3 Neuron’s Activity and May Induce Depressive Like Behaviors. J. Adv. Res. 2025. [Google Scholar] [CrossRef]
  47. Zhang, K.; Wang, F.; Zhai, M.; He, M.; Hu, Y.; Feng, L.; Li, Y.; Yang, J.; Wu, C. Hyperactive Neuronal Autophagy Depletes Bdnf and Impairs Adult Hippocampal Neurogenesis in a Corticosterone-Induced Mouse Model of Depression. Theranostics 2023, 13, 1059–1075. [Google Scholar] [CrossRef]
  48. Mateus-Pinheiro, A.; Pinto, L.; Bessa, J.M.; Morais, M.; Alves, N.D.; Monteiro, S.; Patrício, P.; Almeida, O.F.X.; Sousa, N. Sustained Remission from Depressive-Like Behavior Depends on Hippocampal Neurogenesis. Transl. Psychiatry 2013, 3, e210. [Google Scholar] [CrossRef]
  49. Liu, S.; Lei, T.; Wang, L.; Chen, F.; Hu, X.; Song, G.; Tang, X.; Wu, G.; Chen, H.; Sun, X.; et al. Taurine Reverses Arsenic-Induced Inhibition of Hippocampal Neurogenesis and Depression-Like Behavior in Mice. J. Biochem. Mol. Toxicol. 2024, 38, e70037. [Google Scholar] [CrossRef]
  50. Santarelli, L.; Saxe, M.; Gross, C.; Surget, A.; Battaglia, F.; Dulawa, S.; Weisstaub, N.; Lee, J.; Duman, R.; Arancio, O.; et al. Requirement of Hippocampal Neurogenesis for the Behavioral Effects of Antidepressants. Science 2003, 301, 805–809. [Google Scholar] [CrossRef]
  51. Young, L.N.; Cho, K.; Lawrence, R.; Zoncu, R.; Hurley, J.H. Dynamics and Architecture of the Nrbf2-Containing Phosphatidylinositol 3-Kinase Complex I of Autophagy. Proc. Natl. Acad. Sci. USA 2016, 113, 8224–8229. [Google Scholar] [CrossRef]
  52. Zhang, S.-Q.; Deng, Q.; Zhu, Q.; Hu, Z.-L.; Long, L.-H.; Wu, P.-F.; He, J.-G.; Chen, H.-S.; Yue, Z.; Lu, J.-H.; et al. Cell Type-Specific Nrbf2 Orchestrates Autophagic Flux and Adult Hippocampal Neurogenesis in Chronic Stress-Induced Depression. Cell Discov. 2023, 9, 90. [Google Scholar] [CrossRef]
  53. Snyder, J.S.; Soumier, A.; Brewer, M.; Pickel, J.; Cameron, H.A. Adult Hippocampal Neurogenesis Buffers Stress Responses and Depressive Behaviour. Nature 2011, 476, 458–461. [Google Scholar] [CrossRef]
  54. Brackett, J.; Krull, K.R.; Scheurer, M.E.; Liu, W.; Srivastava, D.K.; Stovall, M.; Merchant, T.E.; Packer, R.J.; Robison, L.L.; Okcu, M.F. Antioxidant Enzyme Polymorphisms and Neuropsychological Outcomes in Medulloblastoma Survivors: A Report from the Childhood Cancer Survivor Study. Neuro-Oncology 2012, 14, 1018–1025. [Google Scholar] [CrossRef]
  55. Airaksinen, M.S.; Saarma, M. The Gdnf Family: Signalling, Biological Functions and Therapeutic Value. Nat. Rev. Neurosci. 2002, 3, 383–394. [Google Scholar] [CrossRef]
  56. Treble-Barna, A.; Heinsberg, L.W.; Stec, Z.; Breazeale, S.; Davis, T.S.; Kesbhat, A.A.; Chattopadhyay, A.; VonVille, H.M.; Ketchum, A.M.; Yeates, K.O.; et al. Brain-Derived Neurotrophic Factor (BDNF) Epigenomic Modifications and Brain-Related Phenotypes in Humans: A Systematic Review. Neurosci. Biobehav. Rev. 2023, 147, 105078. [Google Scholar] [CrossRef]
  57. Dimberg, Y.; Vazquez, M.; Soderstrom, S.; Ebendal, T. Effects of X-Irradiation on Nerve Growth Factor in the Developing Mouse Brain. Toxicol. Lett. 1997, 90, 35–43. [Google Scholar] [CrossRef]
  58. Colla, M.; Kronenberg, G.; Deuschle, M.; Meichel, K.; Hagen, T.; Bohrer, M.; Heuser, I. Hippocampal Volume Reduction and Hpa-System Activity in Major Depression. J. Psychiatr. Res. 2007, 41, 553–560. [Google Scholar] [CrossRef]
  59. Ibi, M.; Liu, J.; Arakawa, N.; Kitaoka, S.; Kawaji, A.; Matsuda, K.I.; Iwata, K.; Matsumoto, M.; Katsuyama, M.; Zhu, K.; et al. Depressive-Like Behaviors Are Regulated by Nox1/Nadph Oxidase by Redox Modification of Nmda Receptor 1. J. Neurosci. 2017, 37, 4200–4212. [Google Scholar] [CrossRef]
  60. Xue, Y.; Liang, H.; Yang, R.; Deng, K.; Tang, M.; Zhang, M. The Role of Pro- and Mature Neurotrophins in the Depression. Behav. Brain Res. 2021, 404, 113162. [Google Scholar] [CrossRef]
  61. Harmer, C.J.; Duman, R.S.; Cowen, P.J. How Do Antidepressants Work? New Perspectives for Refining Future Treatment Approaches. Lancet Psychiatry 2017, 4, 409–418. [Google Scholar] [CrossRef]
  62. McKernan, D.P.; Dinan, T.G.; Cryan, J.F. “Killing the Blues”: A Role for Cellular Suicide (Apoptosis) in Depression and the Antidepressant Response? Prog. Neurobiol. 2009, 88, 246–263. [Google Scholar] [CrossRef]
  63. Gold, P.W.; Wong, M.L. Comment On: Antidepressants Act by Directly Binding to Trkb Neurotrophin Receptors. Mol. Psychiatry 2024, 29, 3926–3927. [Google Scholar] [CrossRef]
  64. Liu, X.; Ding, Y.; Jiang, C.; Ma, X.; Xin, Y.; Li, Y.; Zhang, S.; Shao, B. Astragaloside Iv Ameliorates Radiation-Induced Nerve Cell Damage by Activating the Bdnf/Trkb Signaling Pathway. Phytother. Res. 2023, 37, 4102–4116. [Google Scholar] [CrossRef]
  65. da Fonseca, S.T.O.; Alves, C.C.; Dias, C.T.; Mendes-da-Silva, C. Probiotics and Undernourishment Impact on Brain 5-Hydroxytryptamine System and Neurotrophin Bdnf in Rats: Risk of Depression and Anxiety? Nutrition 2025, 132, 112680. [Google Scholar] [CrossRef]
  66. Bremner, J.D.; Krystal, J.H.; Southwick, S.M.; Charney, D.S. Functional Neuroanatomical Correlates of the Effects of Stress on Memory. J. Trauma. Stress 1995, 8, 527–553. [Google Scholar] [CrossRef]
  67. Holmes, S.E.; Scheinost, D.; Finnema, S.J.; Naganawa, M.; Davis, M.T.; DellaGioia, N.; Nabulsi, N.; Matuskey, D.; Angarita, G.A.; Pietrzak, R.H.; et al. Lower Synaptic Density Is Associated with Depression Severity and Network Alterations. Nat. Commun. 2019, 10, 1529. [Google Scholar] [CrossRef]
  68. Kang, H.J.; Voleti, B.; Hajszan, T.; Rajkowska, G.; Stockmeier, C.A.; Licznerski, P.; Lepack, A.; Majik, M.S.; Jeong, L.S.; Banasr, M.; et al. Decreased Expression of Synapse-Related Genes and Loss of Synapses in Major Depressive Disorder. Nat. Med. 2012, 18, 1413–1417. [Google Scholar] [CrossRef]
  69. Duman, R.S.; Aghajanian, G.K.; Sanacora, G.; Krystal, J.H. Synaptic Plasticity and Depression: New Insights from Stress and Rapid-Acting Antidepressants. Nat. Med. 2016, 22, 238–249. [Google Scholar] [CrossRef]
  70. Vose, L.R.; Stanton, P.K. Synaptic Plasticity, Metaplasticity and Depression. Curr. Neuropharmacol. 2017, 15, 71–86. [Google Scholar] [CrossRef]
  71. Yoshino, Y.; Roy, B.; Kumar, N.; Shahid Mukhtar, M.; Dwivedi, Y. Molecular Pathology Associated with Altered Synaptic Transcriptome in the Dorsolateral Prefrontal Cortex of Depressed Subjects. Transl. Psychiatry 2021, 11, 73. [Google Scholar] [CrossRef]
  72. Sasibhushana, R.B.; Shankaranarayana Rao, B.S.; Srikumar, B.N. Anxiety-, and Depression-Like Behavior Following Short-Term Finasteride Administration Is Associated with Impaired Synaptic Plasticity and Cognitive Behavior in Male Rats. J. Psychiatr. Res. 2024, 174, 304–318. [Google Scholar] [CrossRef]
  73. Zou, Y.; Corniola, R.; Leu, D.; Khan, A.; Sahbaie, P.; Chakraborti, A.; Clark, D.J.; Fike, J.R.; Huang, T.T. Extracellular Superoxide Dismutase Is Important for Hippocampal Neurogenesis and Preservation of Cognitive Functions after Irradiation. Proc. Natl. Acad. Sci. USA 2012, 109, 21522–21527. [Google Scholar] [CrossRef]
  74. Sun, L.; Chen, M.; Wang, H.; Dong, J.; Zhao, L.; Peng, R. Camkiiδ Promotes Synaptic Plasticity under Terahertz Wave Radiation by Activation of the Nf-Κb Pathway. J. Phys. Chem. Lett. 2022, 13, 5925–5931. [Google Scholar] [CrossRef]
  75. Wu, M.-Y.; Zou, W.-J.; Yu, P.; Yang, Y.; Li, S.-J.; Liu, Q.; Xie, J.; Chen, S.-Q.; Lin, W.-J.; Tang, Y. Cranial Irradiation Impairs Intrinsic Excitability and Synaptic Plasticity of Hippocampal Ca1 Pyramidal Neurons with Implications for Cognitive Function. Neural Regen. Res. 2022, 17, 2253–2259. [Google Scholar] [CrossRef]
  76. Parihar, V.K.; Limoli, C.L. Cranial Irradiation Compromises Neuronal Architecture in the Hippocampus. Proc. Natl. Acad. Sci. USA 2013, 110, 12822–12827. [Google Scholar] [CrossRef]
  77. Duman, J.G.; Dinh, J.; Zhou, W.; Cham, H.; Mavratsas, V.; Paveškovic, M.; Mulherkar, S.; McGovern, S.; Tolias, K.; Grosshans, D. Memantine Prevents Acute Radiation-Induced Toxicities at Hippocampal Excitatory Synapses. Neuro-Oncology 2018, 20, 655–665. [Google Scholar] [CrossRef]
  78. Shi, L.; Adams, M.M.; Long, A.; Carter, C.C.; Bennett, C.; Sonntag, W.E.; Nicolle, M.M.; Robbins, M.; D’Agostino, R.; Brunso-Bechtold, J.K. Spatial Learning and Memory Deficits after Whole-Brain Irradiation Are Associated with Changes in Nmda Receptor Subunits in the Hippocampus. Radiat. Res. 2006, 166, 892–899. [Google Scholar] [CrossRef]
  79. Rohde, B.H.; Rea, M.A.; Simon, J.R.; McBride, W.J. Effects of X-Irradiation Induced Loss of Cerebellar Granule Cells on the Synaptosomal Levels and the High Affinity Uptake of Amino Acids. J. Neurochem. 1979, 32, 1431–1435. [Google Scholar] [CrossRef]
  80. Treccani, G.; Gaarn du Jardin, K.; Wegener, G.; Müller, H.K. Differential Expression of Postsynaptic Nmda and Ampa Receptor Subunits in the Hippocampus and Prefrontal Cortex of the Flinders Sensitive Line Rat Model of Depression. Synapse 2016, 70, 471–474. [Google Scholar] [CrossRef]
  81. Lewitus, G.M.; Konefal, S.C.; Greenhalgh, A.D.; Pribiag, H.; Augereau, K.; Stellwagen, D. Microglial Tnf-A Suppresses Cocaine-Induced Plasticity and Behavioral Sensitization. Neuron 2016, 90, 483–491. [Google Scholar] [CrossRef]
  82. Vezzani, A.; Maroso, M.; Balosso, S.; Sanchez, M.A.; Bartfai, T. Il-1 Receptor/Toll-Like Receptor Signaling in Infection, Inflammation, Stress and Neurodegeneration Couples Hyperexcitability and Seizures. Brain Behav. Immun. 2011, 25, 1281–1289. [Google Scholar] [CrossRef]
  83. Sultana, N.; Sun, C.; Katsube, T.; Wang, B. Biomarkers of Brain Damage Induced by Radiotherapy. Dose-Response 2020, 18, 1559325820938279. [Google Scholar] [CrossRef]
  84. Hong, J.H.; Chiang, C.S.; Campbell, I.L.; Sun, J.R.; Withers, H.R.; McBride, W.H. Induction of Acute Phase Gene Expression by Brain Irradiation. Int. J. Radiat. Oncol. Biol. Phys. 1995, 33, 619–626. [Google Scholar] [CrossRef]
  85. Kyrkanides, S.; Moore, A.H.; Olschowka, J.A.; Daeschner, J.C.; Williams, J.P.; Hansen, J.T.; Kerry O’Banion, M. Cyclooxygenase-2 Modulates Brain Inflammation-Related Gene Expression in Central Nervous System Radiation Injury. Brain Res. Mol. Brain Res. 2002, 104, 159–169. [Google Scholar] [CrossRef]
  86. Dulcich, M.S.; Hartman, R.E. Pomegranate Supplementation Improves Affective and Motor Behavior in Mice after Radiation Exposure. Evid. Based Complement. Altern. Med. 2013, 2013, 940830. [Google Scholar] [CrossRef]
  87. Oh, S.B.; Park, H.R.; Jang, Y.J.; Choi, S.Y.; Son, T.G.; Lee, J. Baicalein Attenuates Impaired Hippocampal Neurogenesis and the Neurocognitive Deficits Induced by Gamma-Ray Radiation. Br. J. Pharmacol. 2013, 168, 421–431. [Google Scholar] [CrossRef]
  88. Chan, K.; Cathomas, F.; Russo, S. Central and Peripheral Inflammation Link Metabolic Syndrome and Major Depressive Disorder. Physiology 2019, 34, 123–133. [Google Scholar] [CrossRef]
  89. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and Its Discontents: The Role of Cytokines in the Pathophysiology of Major Depression. Biol. Psychiatry 2009, 65, 732–741. [Google Scholar] [CrossRef]
  90. Schiepers, O.J.; Wichers, M.C.; Maes, M. Cytokines and Major Depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2005, 29, 201–217. [Google Scholar] [CrossRef]
  91. Liu, F.; Yang, Y.; Fan, X.W.; Zhang, N.; Wang, S.; Shi, Y.J.; Hu, W.J.; Wang, C.X. Impacts of Inflammatory Cytokines on Depression: A Cohort Study. BMC Psychiatry 2024, 24, 195. [Google Scholar] [CrossRef]
  92. Pazzaglia, S.; Briganti, G.; Mancuso, M.; Saran, A. Neurocognitive Decline Following Radiotherapy: Mechanisms and Therapeutic Implications. Cancers 2020, 12, 146. [Google Scholar] [CrossRef]
  93. Kalm, M.; Lannering, B.; Björk-Eriksson, T.; Blomgren, K. Irradiation-Induced Loss of Microglia in the Young Brain. J. Neuroimmunol. 2009, 206, 70–75. [Google Scholar] [CrossRef]
  94. Hua, K.; Schindler, M.K.; McQuail, J.A.; Forbes, M.E.; Riddle, D.R. Regionally Distinct Responses of Microglia and Glial Progenitor Cells to Whole Brain Irradiation in Adult and Aging Rats. PLoS ONE 2012, 7, e52728. [Google Scholar] [CrossRef]
  95. Iwata, M.; Ota, K.T.; Duman, R.S. The Inflammasome: Pathways Linking Psychological Stress, Depression, and Systemic Illnesses. Brain Behav. Immun. 2013, 31, 105–114. [Google Scholar] [CrossRef]
  96. Setiawan, E.; Wilson, A.A.; Mizrahi, R.; Rusjan, P.M.; Miler, L.; Rajkowska, G.; Suridjan, I.; Kennedy, J.L.; Rekkas, P.V.; Houle, S.; et al. Role of Translocator Protein Density, a Marker of Neuroinflammation, in the Brain During Major Depressive Episodes. JAMA Psychiatry 2015, 72, 268–275. [Google Scholar] [CrossRef]
  97. Han, Q.; Li, W.; Chen, P.; Wang, L.; Bao, X.; Huang, R.; Liu, G.; Chen, X. Microglial Nlrp3 Inflammasome-Mediated Neuroinflammation and Therapeutic Strategies in Depression. Neural Regen. Res. 2024, 19, 1890–1898. [Google Scholar] [CrossRef]
  98. Steiner, J.; Bielau, H.; Brisch, R.; Danos, P.; Ullrich, O.; Mawrin, C.; Bernstein, H.G.; Bogerts, B. Immunological Aspects in the Neurobiology of Suicide: Elevated Microglial Density in Schizophrenia and Depression Is Associated with Suicide. J. Psychiatr. Res. 2008, 42, 151–157. [Google Scholar] [CrossRef]
  99. Wang, Y.; Han, Q.; Gong, W.; Pan, D.; Wang, L.; Hu, W.; Yang, M.; Li, B.; Yu, J.; Liu, Q. Microglial Activation Mediates Chronic Mild Stress-Induced Depressive- and Anxiety-Like Behavior in Adult Rats. J. Neuroinflamm. 2018, 15, 21. [Google Scholar] [CrossRef]
  100. Constanzo, J.; Midavaine, É.; Fouquet, J.; Lepage, M.; Descoteaux, M.; Kirby, K.; Tremblay, L.; Masson-Côté, L.; Geha, S.; Longpré, J.M.; et al. Brain Irradiation Leads to Persistent Neuroinflammation and Long-Term Neurocognitive Dysfunction in a Region-Specific Manner. Prog. Neuro-Psychopharmacol Biol. Psychiatry 2020, 102, 109954. [Google Scholar] [CrossRef]
  101. Ida, T.; Hara, M.; Nakamura, Y.; Kozaki, S.; Tsunoda, S.; Ihara, H. Cytokine-Induced Enhancement of Calcium-Dependent Glutamate Release from Astrocytes Mediated by Nitric Oxide. Neurosci. Lett. 2008, 432, 232–236. [Google Scholar] [CrossRef]
  102. Haydon, P.G.; Carmignoto, G. Astrocyte Control of Synaptic Transmission and Neurovascular Coupling. Physiol. Rev. 2006, 86, 1009–1031. [Google Scholar] [CrossRef]
  103. Tyurikova, O.; Shih, P.Y.; Dembitskaya, Y.; Savtchenko, L.P.; McHugh, T.J.; Rusakov, D.A.; Semyanov, A. K(+) Efflux through Postsynaptic Nmda Receptors Suppresses Local Astrocytic Glutamate Uptake. Glia 2022, 70, 961–974. [Google Scholar] [CrossRef]
  104. Matute, C.; Domercq, M.; Sánchez-Gómez, M.V. Glutamate-Mediated Glial Injury: Mechanisms and Clinical Importance. Glia 2006, 53, 212–224. [Google Scholar] [CrossRef]
  105. Hashimoto, K.; Sawa, A.; Iyo, M. Increased Levels of Glutamate in Brains from Patients with Mood Disorders. Biol. Psychiatry 2007, 62, 1310–1316. [Google Scholar] [CrossRef]
  106. Ritter, C.; Buchmann, A.; Müller, S.T.; Volleberg, M.; Haynes, M.; Ghisleni, C.; Noeske, R.; Tuura, R.; Hasler, G. Evaluation of Prefrontal Γ-Aminobutyric Acid and Glutamate Levels in Individuals with Major Depressive Disorder Using Proton Magnetic Resonance Spectroscopy. JAMA Psychiatry 2022, 79, 1209–1216. [Google Scholar] [CrossRef]
  107. Andre, C.; Dinel, A.L.; Ferreira, G.; Laye, S.; Castanon, N. Diet-Induced Obesity Progressively Alters Cognition, Anxiety-Like Behavior and Lipopolysaccharide-Induced Depressive-Like Behavior: Focus on Brain Indoleamine 2,3-Dioxygenase Activation. Brain Behav. Immun. 2014, 41, 10–21. [Google Scholar] [CrossRef]
  108. Guillemin, G.J.; Brew, B.J.; Noonan, C.E.; Takikawa, O.; Cullen, K.M. Indoleamine 2,3 Dioxygenase and Quinolinic Acid Immunoreactivity in Alzheimer’s Disease Hippocampus. Neuropathol. Appl. Neurobiol. 2005, 31, 395–404. [Google Scholar] [CrossRef]
  109. Chen, J.; Li, J.; Qiao, H.; Hu, R.; Li, C. Disruption of Ido Signaling Pathway Alleviates Chronic Unpredictable Mild Stress-Induced Depression-Like Behaviors and Tumor Progression in Mice with Breast Cancer. Cytokine 2023, 162, 156115. [Google Scholar] [CrossRef]
  110. Badawy, A.A. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int. J. Tryptophan. Res. 2017, 10, 1178646917691938. [Google Scholar] [CrossRef]
  111. Maes, M.; Leonard, B.E.; Myint, A.M.; Kubera, M.; Verkerk, R. The New ‘5-Ht’ Hypothesis of Depression: Cell-Mediated Immune Activation Induces Indoleamine 2,3-Dioxygenase, Which Leads to Lower Plasma Tryptophan and an Increased Synthesis of Detrimental Tryptophan Catabolites (Trycats), Both of Which Contribute to the Onset of Depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 702–721. [Google Scholar] [CrossRef]
  112. Gagliardi, F.; De Domenico, P.; Snider, S.; Roncelli, F.; Comai, S.; Mortini, P. Immunomodulatory Mechanisms Driving Tumor Escape in Glioblastoma: The Central Role of Ido and Tryptophan Metabolism in Local and Systemic Immunotolerance. Crit. Rev. Oncol./Hematol. 2025, 209, 104657. [Google Scholar] [CrossRef]
  113. Guglielmi, P.; Carradori, S.; D’Agostino, I.; Campestre, C.; Petzer, J.P. An Updated Patent Review on Monoamine Oxidase (Mao) Inhibitors. Expert Opin. Ther. Pat. 2022, 32, 849–883. [Google Scholar] [CrossRef]
  114. Zhu, Y.; Jiang, C.; Liu, Y.; Li, Y.; Wu, H.; Feng, J.; Xu, Y. Association between Ido Activity and Prognosis in Patients with Non-Small Cell Lung Cancer after Radiotherapy. Ann. Transl. Med. 2020, 8, 1169. [Google Scholar] [CrossRef]
  115. Felger, J.C.; Miller, A.H. Cytokine Effects on the Basal Ganglia and Dopamine Function: The Subcortical Source of Inflammatory Malaise. Front. Neuroendocr. 2012, 33, 315–327. [Google Scholar] [CrossRef]
  116. Wilson, S.K.; Thomas, J. Bh4 as a Therapeutic Target for Adhd: Relevance to Neurotransmitters and Stress-Driven Symptoms. J. Atten. Disord. 2024, 28, 161–167. [Google Scholar] [CrossRef]
  117. Kazumori, H.; Ishihara, S.; Rumi, M.A.; Ortega-Cava, C.F.; Kadowaki, Y.; Kinoshita, Y. Transforming Growth Factor-Alpha Directly Augments Histidine Decarboxylase and Vesicular Monoamine Transporter 2 Production in Rat Enterochromaffin-Like Cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 286, G508–G514. [Google Scholar] [CrossRef]
  118. Voloboueva, L.A.; Giffard, R.G. Inflammation, Mitochondria, and the Inhibition of Adult Neurogenesis. J. Neurosci. Res. 2011, 89, 1989–1996. [Google Scholar] [CrossRef]
  119. Valentine, A.D.; Meyers, C.A.; Kling, M.A.; Richelson, E.; Hauser, P. Mood and Cognitive Side Effects of Interferon-Alpha Therapy. Semin. Oncol. 1998, 25, 39–47. [Google Scholar]
  120. Velusamy, P.; Buckley, D.J.; Greaney, J.L.; Case, A.J.; Fadel, P.J.; Trott, D.W. Il-6 Induces Mitochondrial Ros Production and Blunts No Bioavailability in Human Aortic Endothelial Cells. Am. J. Physiology. Regul. Integr. Comp. Physiol. 2025, 328, R509–R514. [Google Scholar] [CrossRef]
  121. Rackov, G.; Tavakoli Zaniani, P.; Colomo Del Pino, S.; Shokri, R.; Monserrat, J.; Alvarez-Mon, M.; Martinez, A.C.; Balomenos, D. Mitochondrial Reactive Oxygen Is Critical for Il-12/Il-18-Induced Ifn-Γ Production by Cd4(+) T Cells and Is Regulated by Fas/Fasl Signaling. Cell Death Dis. 2022, 13, 531. [Google Scholar] [CrossRef]
  122. Zhou, Z.; Zhou, Y.; Huang, Z.; Wang, M.; Jiang, J.; Yan, M.; Xiang, W.; Li, S.; Yu, Y.; Chen, L.; et al. Notopterol Improves Cognitive Dysfunction and Depression-Like Behavior Via Inhibiting Stat3/Nf-ĸb Pathway Mediated Inflammation in Glioma-Bearing Mice. Int. Immunopharmacol. 2023, 118, 110041. [Google Scholar] [CrossRef]
  123. Acharya, M.M.; Christie, L.A.; Lan, M.L.; Giedzinski, E.; Fike, J.R.; Rosi, S.; Limoli, C.L. Human Neural Stem Cell Transplantation Ameliorates Radiation-Induced Cognitive Dysfunction. Cancer Res. 2011, 71, 4834–4845. [Google Scholar] [CrossRef]
  124. Huang, X.; Li, M.; Zhou, D.; Deng, Z.; Guo, J.; Huang, H. Endothelial Progenitor Cell Transplantation Restores Vascular Injury in Mice after Whole-Brain Irradiation. Brain Res. 2020, 1746, 147005. [Google Scholar] [CrossRef]
  125. Miller, K.B.; Mi, K.L.; Nelson, G.A.; Norman, R.B.; Patel, Z.S.; Huff, J.L. Ionizing Radiation, Cerebrovascular Disease, and Consequent Dementia: A Review and Proposed Framework Relevant to Space Radiation Exposure. Front. Physiol. 2022, 13, 1008640. [Google Scholar] [CrossRef]
  126. Brown, W.R.; Blair, R.M.; Moody, D.M.; Thore, C.R.; Ahmed, S.; Robbins, M.E.; Wheeler, K.T. Capillary Loss Precedes the Cognitive Impairment Induced by Fractionated Whole-Brain Irradiation: A Potential Rat Model of Vascular Dementia. J. Neurol. Sci. 2007, 257, 67–71. [Google Scholar] [CrossRef]
  127. Jin, X.; Liang, B.; Chen, Z.; Liu, X.; Zhang, Z. The Dynamic Changes of Capillary Permeability and Upregulation of Vegf in Rats Following Radiation-Induced Brain Injury. Microcirculation 2014, 21, 171–177. [Google Scholar] [CrossRef]
  128. Zhong, Q.; Yu, H.; Huang, C.; Zhong, J.; Wang, H.; Xu, J.; Cheng, Y. Fcpr16, a Novel Phosphodiesterase 4 Inhibitor, Produces an Antidepressant-Like Effect in Mice Exposed to Chronic Unpredictable Mild Stress. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 90, 62–75. [Google Scholar] [CrossRef]
  129. Hemanth Kumar, B.; Mishra, S.; Trivedi, R.; Singh, S.; Rana, P.; Khushu, S. Demyelinating Evidences in Cms Rat Model of Depression: A Dti Study at 7 T. Neuroscience 2014, 275, 12–21. [Google Scholar] [CrossRef]
  130. Ma, T.; Li, B.; Le, Y.; Xu, Y.; Wang, F.; Tian, Y.; Cai, Q.; Liu, Z.; Xiao, L.; Li, H. Demyelination Contributes to Depression Comorbidity in a Rat Model of Chronic Epilepsy Via Dysregulation of Olig2/Lingo-1 and Disturbance of Calcium Homeostasis. Exp. Neurol. 2019, 321, 113034. [Google Scholar] [CrossRef]
  131. Simpson, S.; Tan, H.; Otahal, P.; Taylor, B.; Ponsonby, A.; Lucas, R.; Blizzard, L.; Valery, P.; Lechner-Scott, J.; Shaw, C.; et al. Anxiety, Depression and Fatigue at 5-Year Review Following Cns Demyelination. Acta Neurol. Scand. 2016, 134, 403–413. [Google Scholar] [CrossRef]
  132. Margoni, M.; Preziosa, P.; Rocca, M.A.; Filippi, M. Depressive Symptoms, Anxiety and Cognitive Impairment: Emerging Evidence in Multiple Sclerosis. Transl. Psychiatry 2023, 13, 264. [Google Scholar] [CrossRef]
  133. Rosenkranz, E.; Thissen, A.; Siegel, S.; Piroth, M.; Clusmann, H.; Gebauer, J.; Brabant, G.; Kreitschmann-Andermahr, I. Melatonin Secretion Following Brain Midline Irradiation Is Diminished, but Not Correlated with Subjective Sleep Disturbances. Clin. Endocrinol. 2018, 89, 870–877. [Google Scholar] [CrossRef]
  134. Forbes, M.E.; Paitsel, M.; Bourland, J.D.; Riddle, D.R. Systemic Effects of Fractionated, Whole-Brain Irradiation in Young Adult and Aging Rats. Radiat. Res. 2013, 180, 326–333. [Google Scholar] [CrossRef]
  135. Mineura, K.; Sasajima, T.; Kowada, M.; Saitoh, H.; Shishido, F. Case Report: Radiation-Induced Vasculopathy Implicated by Depressed Blood Flow and Metabolism in a Pineal Glioma. Br. J. Radiol. 1993, 66, 727–733. [Google Scholar] [CrossRef]
  136. Proctor, S.J.; Kernaham, J.; Taylor, P. Depression as Component of Post-Cranial Irradiation Somnolence Syndrome. Lancet 1981, 1, 1215–1216. [Google Scholar] [CrossRef]
  137. Faithfull, S.; Brada, M. Somnolence Syndrome in Adults Following Cranial Irradiation for Primary Brain Tumours. Clin. Oncol. 1998, 10, 250–254. [Google Scholar] [CrossRef]
  138. Atkinson, L. Three Standard Errors of Measurement and the Wechsler Memory Scale—Revised. Psychol. Assess. J. Consult. Clin. Psychol. 1991, 3, 136. [Google Scholar] [CrossRef]
  139. Elwood, R.W. The Wechsler Memory Scale-Revised: Psychometric Characteristics and Clinical Application. Neuropsychol. Rev. 1991, 2, 179–201. [Google Scholar] [CrossRef]
  140. Rempel-Clower, N.L.; Zola, S.M.; Squire, L.R.; Amaral, D.G. Three Cases of Enduring Memory Impairment after Bilateral Damage Limited to the Hippocampal Formation. J. Neurosci. 1996, 16, 5233–5255. [Google Scholar] [CrossRef]
  141. Hamann, S.B.; Squire, L.R. Intact Perceptual Memory in the Absence of Conscious Memory. Behav. Neurosci. 1997, 111, 850–854. [Google Scholar] [CrossRef]
  142. Cave, C.B.; Squire, L.R. Intact Verbal and Nonverbal Short-Term Memory Following Damage to the Human Hippocampus. Hippocampus 1992, 2, 151–163. [Google Scholar] [CrossRef]
  143. Hamann, S.B.; Cahill, L.; McGaugh, J.L.; Squire, L.R. Intact Enhancement of Declarative Memory for Emotional Material in Amnesia. Learn. Mem. 1997, 4, 301–309. [Google Scholar] [CrossRef]
  144. Delis, D.; Kramer, J.; Kaplan, E.; Ober, B.A. California Verbal Learning Test Research Edition Manual; The Psychological Corporation: San Antonio, TX, USA, 1987. [Google Scholar]
  145. Elwood, R.W. The California Verbal Learning Test: Psychometric Characteristics and Clinical Application. Neuropsychol. Rev. 1995, 5, 173–201. [Google Scholar] [CrossRef]
  146. DeJong, J.; Donders, J. Cluster Subtypes on the California Verbal Learning Test-Second Edition (Cvlt-II) in a Traumatic Brain Injury Sample. J. Clin. Exp. Neuropsychol. 2010, 32, 953–960. [Google Scholar] [CrossRef]
  147. Beck, I.R.; Gagneux-Zurbriggen, A.; Berres, M.; Taylor, K.I.; Monsch, A.U. Comparison of Verbal Episodic Memory Measures: Consortium to Establish a Registry for Alzheimer’s Disease—Neuropsychological Assessment Battery (Cerad-Nab) Versus California Verbal Learning Test (CVLT). Arch. Clin. Neuropsychol. 2012, 27, 510–519. [Google Scholar] [CrossRef]
  148. van den Heuvel, M.P.; Hulshoff Pol, H.E. Exploring the Brain Network: A Review on Resting-State Fmri Functional Connectivity. Eur. Neuro-Psychopharmacol. 2010, 20, 519–534. [Google Scholar] [CrossRef]
  149. Wehrheim, M.H.; Faskowitz, J.; Schubert, A.L.; Fiebach, C.J. Reliability of Variability and Complexity Measures for Task and Task-Free Bold Fmri. Hum. Brain Mapp. 2024, 45, e26778. [Google Scholar] [CrossRef]
  150. Greicius, M.D.; Srivastava, G.; Reiss, A.L.; Menon, V. Default-Mode Network Activity Distinguishes Alzheimer’s Disease from Healthy Aging: Evidence from Functional Mri. Proc. Natl. Acad. Sci. USA 2004, 101, 4637–4642. [Google Scholar] [CrossRef]
  151. Celone, K.A.; Calhoun, V.D.; Dickerson, B.C.; Atri, A.; Chua, E.F.; Miller, S.L.; DePeau, K.; Rentz, D.M.; Selkoe, D.J.; Blacker, D.; et al. Alterations in Memory Networks in Mild Cognitive Impairment and Alzheimer’s Disease: An Independent Component Analysis. J. Neurosci. 2006, 26, 10222–10231. [Google Scholar] [CrossRef]
  152. Morrison, M.A.; Walter, S.; Mueller, S.; Felton, E.; Jakary, A.; Stoller, S.; Molinaro, A.M.; Braunstein, S.E.; Hess, C.P.; Lupo, J.M. Functional Network Alterations in Young Brain Tumor Patients with Radiotherapy-Induced Memory Impairments and Vascular Injury. Front. Neurol. 2022, 13, 921984. [Google Scholar] [CrossRef]
  153. Ferré, J.C.; Bannier, E.; Raoult, H.; Mineur, G.; Carsin-Nicol, B.; Gauvrit, J.Y. Arterial Spin Labeling (Asl) Perfusion: Techniques and Clinical Use. Diagn. Interv. Imaging 2013, 94, 1211–1223. [Google Scholar] [CrossRef]
  154. Zhang, J.; Wu, Y.; Wang, Y.; Zhang, X.; Lei, Y.; Zhu, G.; Mao, C.; Zhang, L.; Ma, L. Diffusion-Weighted Imaging and Arterial Spin Labeling Radiomics Features May Improve Differentiation between Radiation-Induced Brain Injury and Glioma Recurrence. Eur. Radiol. 2023, 33, 3332–3342. [Google Scholar] [CrossRef]
  155. Niederer, P.F. Basic Elements of Nuclear Magnetic Resonance for Use in Medical Diagnostics: Magnetic Resonance Imaging (Mri) and Magnetic Resonance Spectroscopy (Mrs). Technol. Health Care 2011, 19, 373–389. [Google Scholar] [CrossRef]
  156. Schlemmer, H.P.; Bachert, P.; Herfarth, K.K.; Zuna, I.; Debus, J.; van Kaick, G. Proton Mr Spectroscopic Evaluation of Suspicious Brain Lesions after Stereotactic Radiotherapy. AJNR Am. J. Neuroradiol. 2001, 22, 1316–1324. [Google Scholar]
  157. Kinoshita, K.; Tada, E.; Matsumoto, K.; Asari, S.; Ohmoto, T.; Itoh, T. Proton Mr Spectroscopy of Delayed Cerebral Radiation in Monkeys and Humans after Brachytherapy. AJNR Am. J. Neuroradiol. 1997, 18, 1753–1761. [Google Scholar]
  158. Ishibashi, K.; Onishi, A.; Wagatsuma, K.; Fujiwara, Y.; Ishii, K. Longitudinal 18f-Fdg Images in Patients with Alzheimer Disease over More Than 9 Years from a Preclinical Stage. Clin. Nucl. Med. 2020, 45, e185–e189. [Google Scholar] [CrossRef]
  159. Hahn, C.A.; Zhou, S.M.; Raynor, R.; Tisch, A.; Light, K.; Shafman, T.; Wong, T.; Kirkpatrick, J.; Turkington, T.; Hollis, D.; et al. Dose-Dependent Effects of Radiation Therapy on Cerebral Blood Flow, Metabolism, and Neurocognitive Dysfunction. Int. J. Radiat. Oncol. Biol. Phys. 2009, 73, 1082–1087. [Google Scholar] [CrossRef]
  160. Savitz, J.B.; Rauch, S.L.; Drevets, W.C. Clinical Application of Brain Imaging for the Diagnosis of Mood Disorders: The Current State of Play. Mol. Psychiatry 2013, 18, 528–539. [Google Scholar] [CrossRef]
  161. 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]
  162. Taylor, A.; Powell, M.E. Intensity-Modulated Radiotherapy—What Is It? Cancer Imaging 2004, 4, 68–73. [Google Scholar] [CrossRef]
  163. Gondi, V.; Tolakanahalli, R.; Mehta, M.P.; Tewatia, D.; Rowley, H.; Kuo, J.S.; Khuntia, D.; Tomé, W.A. Hippocampal-Sparing Whole-Brain Radiotherapy: A “How-to” Technique Using Helical Tomotherapy and Linear Accelerator-Based Intensity-Modulated Radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 1244–1252. [Google Scholar] [CrossRef]
  164. 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]
  165. Zhang, H.W.; Hu, B.; Pang, H.W. Dosimetric Comparison of Helical Tomotherapy and Volumetric Modulated Arc Therapy in Hippocampal Avoidance Whole-Brain Radiotherapy. J. Appl. Clin. Med. Phys. 2024, 25, e14218. [Google Scholar] [CrossRef]
  166. Westover, K.D.; Mendel, J.T.; Dan, T.; Kumar, K.; Gao, A.; Pulipparacharuv, S.; Iyengar, P.; Nedzi, L.; Hannan, R.; Anderson, J.; et al. Phase II Trial of Hippocampal-Sparing Whole Brain Irradiation with Simultaneous Integrated Boost for Metastatic Cancer. Neuro-Oncology 2020, 22, 1831–1839. [Google Scholar] [CrossRef]
  167. Rodríguez de Dios, N.; Couñago, F.; Murcia-Mejía, M.; Rico-Oses, M.; Calvo-Crespo, P.; Samper, P.; Vallejo, C.; Luna, J.; Trueba, I.; Sotoca, A.; et al. Randomized Phase Iii Trial of Prophylactic Cranial Irradiation with or without Hippocampal Avoidance for Small-Cell Lung Cancer (Premer): A Gicor-Goecp-Seor Study. J. Clin. Oncol. 2021, 39, 3118–3127. [Google Scholar] [CrossRef]
  168. Shang, W.; Yao, H.; Sun, Y.; Mu, A.; Zhu, L.; Li, X. Preventive Effect of Hippocampal Sparing on Cognitive Dysfunction of Patients Undergoing Whole-Brain Radiotherapy and Imaging Assessment of Hippocampal Volume Changes. BioMed Res. Int. 2022, 2022, 4267673. [Google Scholar] [CrossRef]
  169. Yang, W.C.; Chen, Y.F.; Yang, C.C.; Wu, P.F.; Chan, H.M.; Chen, J.L.; Chen, G.Y.; Cheng, J.C.; Kuo, S.H.; Hsu, F.M. Hippocampal Avoidance Whole-Brain Radiotherapy without Memantine in Preserving Neurocognitive Function for Brain Metastases: A Phase Ii Blinded Randomized Trial. Neuro-Oncology 2021, 23, 478–486. [Google Scholar] [CrossRef]
  170. Wang, B.; Fu, S.; Huang, Y.; Liu, L.; Liang, Y.; An, W.; Fan, Y.; Zhao, Y. The Effect of Hippocampal Avoidance Whole Brain Radiotherapy on the Preservation of Long-Term Neurocognitive Function in Non-Small Cell Lung Cancer Patients with Brain Metastasis. Technol. Cancer Res. Treat. 2021, 20, 15330338211034269. [Google Scholar] [CrossRef]
  171. Lin, S.Y.; Yang, C.C.; Wu, Y.M.; Tseng, C.K.; Wei, K.C.; Chu, Y.C.; Hsieh, H.Y.; Wu, T.H.; Pai, P.C.; Hsu, P.W.; et al. Evaluating the Impact of Hippocampal Sparing During Whole Brain Radiotherapy on Neurocognitive Functions: A Preliminary Report of a Prospective Phase II Study. Biomed. J. 2015, 38, 439–449. [Google Scholar] [CrossRef]
  172. Leskinen, S.; Shah, H.A.; Yaffe, B.; Schneider, S.J.; Ben-Shalom, N.; Boockvar, J.A.; D’Amico, R.S.; Wernicke, A.G. Hippocampal Avoidance in Whole Brain Radiotherapy and Prophylactic Cranial Irradiation: A Systematic Review and Meta-Analysis. J. Neuro-Oncol. 2023, 163, 515–527. [Google Scholar] [CrossRef]
  173. Belderbos, J.S.A.; De Ruysscher, D.K.M.; De Jaeger, K.; Koppe, F.; Lambrecht, M.L.F.; Lievens, Y.N.; Dieleman, E.M.T.; Jaspers, J.P.M.; Van Meerbeeck, J.P.; Ubbels, F.; et al. Phase 3 Randomized Trial of Prophylactic Cranial Irradiation with or without Hippocampus Avoidance in Sclc (Nct01780675). J. Thorac. Oncol. 2021, 16, 840–849. [Google Scholar] [CrossRef]
  174. Cao, B.; Passos, I.C.; Mwangi, B.; Amaral-Silva, H.; Tannous, J.; Wu, M.J.; Zunta-Soares, G.B.; Soares, J.C. Hippocampal Subfield Volumes in Mood Disorders. Mol. Psychiatry 2017, 22, 1352–1358. [Google Scholar] [CrossRef]
  175. Kaidanovich-Beilin, O.; Woodgett, J.R. Gsk-3: Functional Insights from Cell Biology and Animal Models. Front. Mol. Neurosci. 2011, 4, 40. [Google Scholar] [CrossRef]
  176. Marosi, M.; Arman, P.; Aceto, G.; D’Ascenzo, M.; Laezza, F. Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int. J. Mol. Sci. 2022, 23, 4413. [Google Scholar] [CrossRef]
  177. Sequeira, R.C.; Godad, A. Understanding Glycogen Synthase Kinase-3: A Novel Avenue for Alzheimer’s Disease. Mol. Neurobiol. 2024, 61, 4203–4221. [Google Scholar] [CrossRef]
  178. Wang, C.; Cui, Y.; Xu, T.; Zhou, Y.; Yang, R.; Wang, T. New Insights into Glycogen Synthase Kinase-3: A Common Target for Neurodegenerative Diseases. Biochem. Pharmacol. 2023, 218, 115923. [Google Scholar] [CrossRef]
  179. Saha, S.; Pal, D.; Nimse, S. Recent Advances in the Discovery of Gsk-3 Inhibitors from Synthetic Origin in the Treatment of Neurological Disorders. Curr. Drug Targets 2021, 22, 1437–1462. [Google Scholar] [CrossRef]
  180. Karati, D.; Meur, S.; Roy, S.; Mukherjee, S.; Debnath, B.; Jha, S.K.; Sarkar, B.K.; Naskar, S.; Ghosh, P. Glycogen Synthase Kinase 3 (Gsk3) Inhibition: A Potential Therapeutic Strategy for Alzheimer’s Disease. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 398, 2319–2342. [Google Scholar] [CrossRef]
  181. Arciniegas Ruiz, S.M.; Eldar-Finkelman, H. Glycogen Synthase Kinase-3 Inhibitors: Preclinical and Clinical Focus on Cns-a Decade Onward. Front. Mol. Neurosci. 2021, 14, 792364. [Google Scholar] [CrossRef]
  182. Stambolic, V.; Ruel, L.; Woodgett, J.R. Lithium Inhibits Glycogen Synthase Kinase-3 Activity and Mimics Wingless Signalling in Intact Cells. Curr. Biol. 1996, 6, 1664–1668. [Google Scholar] [CrossRef]
  183. Li, Q.; Li, H.; Roughton, K.; Wang, X.; Kroemer, G.; Blomgren, K.; Zhu, C. Lithium Reduces Apoptosis and Autophagy after Neonatal Hypoxia-Ischemia. Cell Death Dis. 2010, 1, e56. [Google Scholar] [CrossRef]
  184. Contestabile, A.; Greco, B.; Ghezzi, D.; Tucci, V.; Benfenati, F.; Gasparini, L. Lithium Rescues Synaptic Plasticity and Memory in Down Syndrome Mice. J. Clin. Investig. 2013, 123, 348–361. [Google Scholar] [CrossRef]
  185. Zanni, G.; Di Martino, E.; Omelyanenko, A.; Andäng, M.; Delle, U.; Elmroth, K.; Blomgren, K. Lithium Increases Proliferation of Hippocampal Neural Stem/Progenitor Cells and Rescues Irradiation-Induced Cell Cycle Arrest In Vitro. Oncotarget 2015, 6, 37083–37097. [Google Scholar] [CrossRef]
  186. Huo, K.; Sun, Y.; Li, H.; Du, X.; Wang, X.; Karlsson, N.; Zhu, C.; Blomgren, K. Lithium Reduced Neural Progenitor Apoptosis in the Hippocampus and Ameliorated Functional Deficits after Irradiation to the Immature Mouse Brain. Mol. Cell. Neurosci. 2012, 51, 32–42. [Google Scholar] [CrossRef]
  187. Rybakowski, J.K. Antiviral, Immunomodulatory, and Neuroprotective Effect of Lithium. J. Integr. Neurosci. 2022, 21, 68. [Google Scholar] [CrossRef]
  188. Yazlovitskaya, E.M.; Edwards, E.; Thotala, D.; Fu, A.; Osusky, K.L.; Whetsell, W.O., Jr.; Boone, B.; Shinohara, E.T.; Hallahan, D.E. Lithium Treatment Prevents Neurocognitive Deficit Resulting from Cranial Irradiation. Cancer Res. 2006, 66, 11179–11186. [Google Scholar] [CrossRef]
  189. Zhukova, N.; Ramaswamy, V.; Remke, M.; Martin, D.; Castelo-Branco, P.; Zhang, C.; Fraser, M.; Tse, K.; Poon, R.; Shih, D.; et al. Wnt Activation by Lithium Abrogates Tp53 Mutation Associated Radiation Resistance in Medulloblastoma. Acta Neuropathol. Commun. 2014, 2, 174. [Google Scholar] [CrossRef]
  190. Malberg, J.E.; Eisch, A.J.; Nestler, E.J.; Duman, R.S. Chronic Antidepressant Treatment Increases Neurogenesis in Adult Rat Hippocampus. J. Neurosci. 2000, 20, 9104–9110. [Google Scholar] [CrossRef]
  191. Wang, J.W.; David, D.J.; Monckton, J.E.; Battaglia, F.; Hen, R. Chronic Fluoxetine Stimulates Maturation and Synaptic Plasticity of Adult-Born Hippocampal Granule Cells. J. Neurosci. 2008, 28, 1374–1384. [Google Scholar] [CrossRef]
  192. Liu, X.; Zhang, J.; Sun, D.; Fan, Y.; Zhou, H.; Fu, B. Effects of Fluoxetine on Brain-Derived Neurotrophic Factor Serum Concentration and Cognition in Patients with Vascular Dementia. Clin. Interv. Aging 2014, 9, 411–418. [Google Scholar] [CrossRef]
  193. Shi, H.J.; Wu, D.L.; Chen, R.; Li, N.; Zhu, L.J. Requirement of Hippocampal Dg Nnos-Capon Dissociation for the Anxiolytic and Antidepressant Effects of Fluoxetine. Theranostics 2022, 12, 3656–3675. [Google Scholar] [CrossRef]
  194. Gan, H.; Zhang, Q.; Zhu, B.; Wu, S.; Chai, D. Fluoxetine Reverses Brain Radiation and Temozolomide-Induced Anxiety and Spatial Learning and Memory Defect in Mice. J. Neurophysiol. 2019, 121, 298–305. [Google Scholar] [CrossRef]
  195. Gordon, M.; Melvin, G. Selective Serotonin Re-Uptake Inhibitors—A Review of the Side Effects in Adolescents. Aust. Fam. Physician 2013, 42, 620–623. [Google Scholar]
  196. Ng, Q.X.; Venkatanarayanan, N.; Ho, C.Y.X.; Sim, W.S.; Lim, D.Y.; Yeo, W.S. Selective Serotonin Reuptake Inhibitors and Persistent Pulmonary Hypertension of the Newborn: An Update Meta-Analysis. J. Womens Health 2019, 28, 331–338. [Google Scholar] [CrossRef]
  197. Kang, J.B.; Koh, P.O. Retinoic Acid Has Neuroprotective Effects by Modulating Thioredoxin in Ischemic Brain Damage and Glutamate-Exposed Neurons. Neuroscience 2023, 521, 166–181. [Google Scholar] [CrossRef]
  198. Sato, Y.; Meller, R.; Yang, T.; Taki, W.; Simon, R.P. Stereo-Selective Neuroprotection against Stroke with Vitamin a Derivatives. Brain Res. 2008, 1241, 188–192. [Google Scholar] [CrossRef]
  199. Liang, N.; Li, H.; Zhang, K.; Wang, Y.; Xiang, L.; Xiao, L.; Luo, G. Association of Dietary Retinol Intake and Serum Neurofilament Light Chain Levels: Results from Nhanes 2013-2014. Nutrients 2024, 16, 1763. [Google Scholar] [CrossRef]
  200. Khatib, T.; Chisholm, D.R.; Whiting, A.; Platt, B.; McCaffery, P. Decay in Retinoic Acid Signaling in Varied Models of Alzheimer’s Disease and In-Vitro Test of Novel Retinoic Acid Receptor Ligands (Rar-Ms) to Regulate Protective Genes. J. Alzheimer’s Dis. 2020, 73, 935–954. [Google Scholar] [CrossRef]
  201. Bonhomme, D.; Minni, A.M.; Alfos, S.; Roux, P.; Richard, E.; Higueret, P.; Moisan, M.P.; Pallet, V.; Touyarot, K. Vitamin a Status Regulates Glucocorticoid Availability in Wistar Rats: Consequences on Cognitive Functions and Hippocampal Neurogenesis? Front. Behav. Neurosci. 2014, 8, 20. [Google Scholar] [CrossRef]
  202. Jiang, W.; Yu, Q.; Gong, M.; Chen, L.; Wen, E.Y.; Bi, Y.; Zhang, Y.; Shi, Y.; Qu, P.; Liu, Y.X.; et al. Vitamin a Deficiency Impairs Postnatal Cognitive Function Via Inhibition of Neuronal Calcium Excitability in Hippocampus. J. Neurochem. 2012, 121, 932–943. [Google Scholar] [CrossRef]
  203. Saponaro, F.; Kim, J.H.; Chiellini, G. Transthyretin Stabilization: An Emerging Strategy for the Treatment of Alzheimer’s Disease? Int. J. Mol. Sci. 2020, 21, 8672. [Google Scholar] [CrossRef]
  204. Surya, K.; Manickam, N.; Jayachandran, K.S.; Kandasamy, M.; Anusuyadevi, M. Resveratrol Mediated Regulation of Hippocampal Neuroregenerative Plasticity Via Sirt1 Pathway in Synergy with Wnt Signaling: Neurotherapeutic Implications to Mitigate Memory Loss in Alzheimer’s Disease. J. Alzheimer’s Dis. 2023, 94, S125–S140. [Google Scholar] [CrossRef]
  205. Kraus, R.L.; Pasieczny, R.; Lariosa-Willingham, K.; Turner, M.S.; Jiang, A.; Trauger, J.W. Antioxidant Properties of Minocycline: Neuroprotection in an Oxidative Stress Assay and Direct Radical-Scavenging Activity. J. Neurochem. 2005, 94, 819–827. [Google Scholar] [CrossRef]
  206. Parvardeh, S.; Sheikholeslami, M.A.; Ghafghazi, S.; Pouriran, R.; Mortazavi, S.E. Minocycline Improves Memory by Enhancing Hippocampal Synaptic Plasticity and Restoring Antioxidant Enzyme Activity in a Rat Model of Cerebral Ischemia-Reperfusion. Basic Clin. Neurosci. 2022, 13, 225–235. [Google Scholar] [CrossRef]
  207. Zhang, L.; Li, K.; Sun, R.; Zhang, Y.; Ji, J.; Huang, P.; Yang, H.; Tian, Y. Minocycline Ameliorates Cognitive Impairment Induced by Whole-Brain Irradiation: An Animal Study. Radiat. Oncol. 2014, 9, 281. [Google Scholar] [CrossRef]
  208. Wong, M.; Inserra, A.; Lewis, M.; Mastronardi, C.; Leong, L.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S.; et al. Inflammasome Signaling Affects Anxiety- and Depressive-Like Behavior and Gut Microbiome Composition. Mol. Psychiatry 2016, 21, 797–805. [Google Scholar] [CrossRef]
  209. O’Connor, J.C.; Lawson, M.A.; André, C.; Moreau, M.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide-Induced Depressive-Like Behavior Is Mediated by Indoleamine 2,3-Dioxygenase Activation in Mice. Mol. Psychiatry 2009, 14, 511–522. [Google Scholar] [CrossRef]
  210. Voorhees, J.; Remy, M.; Cintrón-Pérez, C.; El Rassi, E.; Khan, M.; Dutca, L.; Yin, T.; McDaniel, L.; Williams, N.; Brat, D.; et al. (-)-P7c3-S243 Protects a Rat Model of Alzheimer’s Disease from Neuropsychiatric Deficits and Neurodegeneration without Altering Amyloid Deposition or Reactive Glia. Biol. Psychiatry 2018, 84, 488–498. [Google Scholar] [CrossRef]
  211. Walker, A.K.; Rivera, P.D.; Wang, Q.; Chuang, J.C.; Tran, S.; Osborne-Lawrence, S.; Estill, S.J.; Starwalt, R.; Huntington, P.; Morlock, L.; et al. The P7c3 Class of Neuroprotective Compounds Exerts Antidepressant Efficacy in Mice by Increasing Hippocampal Neurogenesis. Mol. Psychiatry 2015, 20, 500–508. [Google Scholar] [CrossRef]
  212. Carlson, L.E.; Ismaila, N.; Addington, E.L.; Asher, G.N.; Atreya, C.; Balneaves, L.G.; Bradt, J.; Fuller-Shavel, N.; Goodman, J.; Hoffman, C.J.; et al. Integrative Oncology Care of Symptoms of Anxiety and Depression in Adults with Cancer: Society for Integrative Oncology-Asco Guideline. J. Clin. Oncol. 2023, 41, 4562–4591. [Google Scholar] [CrossRef]
Table 1. Clinical trials.
Table 1. Clinical trials.
Study DesignPopulation CharacteristicsAssessment TimepointsPsychological ScalesKey FindingsFollow-Up DurationReference
Case-control13 WBRT patients vs. 13 healthy controls1-month post WBRTBDI, BAIThe WBRT group showed significantly higher scores on the BDI (16.40 ± 12.16 vs. 4.00 ± 2.38) and BAI (14.47 ± 11.96 vs. 4.54 ± 3.30) compared to the control group (p < 0.05)Weekly during treatment[17]
Retrospective cohort232 newly diagnosed NPC patientsPre-RT, week 4, RT completionHADSAnxiety incidence: 34.0% → 55.1% → 64.0% (p < 0.001);
Depression incidence: 25.0% → 43.9% → 56.0% (p < 0.001)
N/A[18]
Case-control46 RBI vs. 46 non-RBI NPC patientsCase group: post-RT: 6.0 ± 3.5
Control group: post-RT: 5.7 ± 3.1
SAS, SDSThe RBI group had higher incidence rates of depression (84.8%) and anxiety (87.0%). The SDS (63.48 ± 8.11 vs. 58.67 ± 7.52, p = 0.008) and SAS (67.36 ± 10.41 vs. 60.34 ± 9.76, p = 0.005) scores were significantly higher in the RBI groupN/A[10]
Cross-sectional100 cancer patients undergoing RTPre-RT, mid-RT, RT completionHADSMaximum prevalence of anxiety and depression was seen in patients having head and neck malignanciesN/A[19]
RBI: Radiation-induced brain injury; SDS: Self-rating Depression Scale; SAS: Self-rating Anxiety Scale; BDI: depression inventory; BAI: Beck anxiety inventory; HADS: Hospital Anxiety and Depression Scale; WBRT: Whole-brain radiotherapy; RT: Radiotherapy.
Table 2. Animal tests.
Table 2. Animal tests.
SpeciesAgeTime After WBIRadiation DoseRadiation TypeType of ExperimentResultsReference
Male Fischer rats28 days4 months27 Gy in 9 daily fractions of 3 Gy, or 34 Gy, where the last 3 Gy fraction being replaced with a 10 Gy boostX-rayEPMThe experimental group spending less time in the open arms
relative to controls (27Gy: p = 0.002, 34Gy: p = 0.02).
[28]
Male strain A mice6–10 weeks0, 5, 10 days3 Gy/single
5 Gy/single
8 Gy/single
γ-rayOFTOn day 5 post-irradiation (PI), an anxiolytic effect was observed at the 3 Gy and 8 Gy dose levels, as evidenced by a significant increase in time spent in the center compared to controls. In contrast, the 5 Gy group showed no statistical difference from the control group.
By day 10 PI, anxiety-like behavior exhibited an approximately dose-dependent response. However, no significant difference was detected between the 5 Gy and 8 Gy dose groups.
[29]
Male Wistar rat pups11 days28, 60 days6 Gy/singleX-rayOFTThe time spent in the open central zone was 71.4% greater in the irradiated rats compared to the non-irradiated controls (p < 0.05).[30]
Male C57BL/6 mice11 weeksEPM, OFT: 3 days
FST, TST: 6 days
20 Gy/singleX-rayEPM
OFT
TST
FST
EPM: There were no significant differences between irradiated and control mice in the total distance traveled (p = 0.8345), number of total entries into the arms (p = 0.4982), or the time spent in the open arms (p = 0.7938).
OFT: Irradiated mice had significantly lower total distance and time spent in the central area compared with control mice (p = 0.0128; p = 0.0056). There was a significantly lower number of entries to the central area by irradiated mice than by control mice (p = 0.0346).
FST: Irradiated mice spent significantly less time immobile in each 1 min period during the 10 min test period than did control mice on day 1 (p = 0.0106). However, on day 2, there were no significant differences in the percentage of immobility between irradiated and control mice (p = 0.9824).
TST: There were no significant differences between irradiated and control mice (p = 0.3913; p = 0.4321).
[25]
Male C57BL/6 mice8 weeks1, 3
months
10 Gy/singleγ-rayTSTAt 30 days (p < 0.01) and 90 days (p < 0.001) post-irradiation, mice treated with 10 Gy showed significantly longer immobility times than sham-irradiated controls.[24]
Female C57BL/6J mice4 weeks2 weeks,
2.5 months
5 Gy/singleX-rayTSTWBI did not significantly alter total immobility time.[31]
Male C57BL/6 mice6 weeks30 days2 Gy/singleX-rayTST
FST
TST: Mice exposed to radiation displayed a significant increase in immobility duration in the TST at 30 days post-irradiation compared to non-irradiated controls (p < 0.0001). Additionally, irradiation markedly reduced active twisting and curling movements while prolonging passive swaying time during mobile phases.
FST: Irradiated mice exhibited significantly prolonged immobility at 30 days post-exposure (p < 0.0001) relative to sham-treated controls.
[32]
Male and female
C57BL/6J mice
14 days4 months8 Gy/singleX-rayOFTA significant reduction in center zone time was observed in irradiated female mice (p = 0.025), but not in males (ns).[33]
Female C57BL/6J mice6 months3 months10 Gy/singleγ-rayOFTThere were no significant differences between irradiated and control mice.[34]
OFT: Open Field Test; TST: Tail Suspension Test; FST: Forced Swim Test; EPM: Elevated Plus Maze Test; ns: not significant (p > 0.05).
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

Yang, F.; Liu, R.; Peng, X.; Luo, N.; Fu, M.; Zhu, W.; Li, Q.; Hu, G. Depression and Anxiety After Radiation-Induced Brain Injury: A Review of Current Research Progress. Curr. Oncol. 2025, 32, 419. https://doi.org/10.3390/curroncol32080419

AMA Style

Yang F, Liu R, Peng X, Luo N, Fu M, Zhu W, Li Q, Hu G. Depression and Anxiety After Radiation-Induced Brain Injury: A Review of Current Research Progress. Current Oncology. 2025; 32(8):419. https://doi.org/10.3390/curroncol32080419

Chicago/Turabian Style

Yang, Feng, Rundong Liu, Xiaohong Peng, Na Luo, Min Fu, Wenjun Zhu, Qianxia Li, and Guangyuan Hu. 2025. "Depression and Anxiety After Radiation-Induced Brain Injury: A Review of Current Research Progress" Current Oncology 32, no. 8: 419. https://doi.org/10.3390/curroncol32080419

APA Style

Yang, F., Liu, R., Peng, X., Luo, N., Fu, M., Zhu, W., Li, Q., & Hu, G. (2025). Depression and Anxiety After Radiation-Induced Brain Injury: A Review of Current Research Progress. Current Oncology, 32(8), 419. https://doi.org/10.3390/curroncol32080419

Article Metrics

Back to TopTop