Back to Journals » Cancer Management and Research » Volume 17

The Pathogenesis and Prevention Strategies of Radiation-Induced Brain Injury

Authors Wang Y, Wu J, Wang Y, Song W, Ren H, Han X, Dong X, Guo Z

Received 26 March 2025

Accepted for publication 1 July 2025

Published 17 July 2025 Volume 2025:17 Pages 1433—1440

DOI https://doi.org/10.2147/CMAR.S525791

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Bilikere Dwarakanath



Yong Wang,1,* Jun Wu,2,* Ya Wang,1 Weiyuan Song,3 Hongjian Ren,4 Xu Han,5 Xiaoqi Dong,6 Zhiqiang Guo1

1Oncology Department, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China; 2Nuclear Medicine Department, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China; 3Nephrology Department, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China; 4Tumor Radiotherapy, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China; 5Anesthesiology Department, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China; 6Pharmacy Department, Shanxi Fenyang Hospital, Lüliang City, Shanxi Province, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Zhiqiang Guo, Email [email protected]

Abstract: Radiation-induced brain injury (RBI) encompasses the severe symptoms resulting from radiation-induced damage to the normal tissue surrounding tumors in patients undergoing radiotherapy for head and neck malignancies. The primary symptoms include skin erythema, pain, and may extend to headache, syncope, nausea, vomiting, memory impairment, alterations in mental status, visual disturbances, drowsiness, and other neurological abnormalities localized to the area of treatment. These side effects both limit the effectiveness of radiation therapy and reduce the patient’s quality of life. During radiotherapy, while killing tumor cells, the radiation will damage the cerebral microvascular endothelial cells, cause cerebrovascular inflammatory response, destroy the blood-brain barrier, aggravate the cerebral oxidative stress response, and induce apoptosis of nerve cells. This review summarizes the mechanisms underlying the occurrence and progression of radiation-induced brain injury and discusses promising strategies for prevention and treatment that may be applicable to clinical patients suffering from this condition.

Keywords: radiation-induced brain injury, radiation protection agents, oxidative stress, apoptosis, cognitive function

Background

Radiation-induced brain injury (RBI) is the most serious complication of neurological impairment in patients with head and neck tumors who receive radiation therapy.1 According to a 2021 Global Cancer Survey, cancer has become the leading cause of death worldwide in 2020. Among them, the overall incidence of head and neck tumors reached 9.5% (3.0% for thyroid cancer, 2.0% for lip and oral cancer, 1.6% for brain and nervous system cancer, 1.0% for laryngeal cancer, 0.7% for nasopharyngeal, oropharyngeal, and laryngopharyngeal cancers, and 0.3% for salivary gland cancers).2 In China, the disease with the highest mortality rate is cancer, of which head and neck tumors rank sixth.3 At present, with the optimization of clinical radiotherapy strategies, the side effects of radiotherapy have also been greatly reduced. However, numerous studies have shown that there is a persistent inflammatory response in the brain after radiation therapy, which is a major contributor to RBI.4 Therefore, this article will review the mechanism of inflammation and its prevention and treatment strategies in the occurrence and development of RBI.

RBI Overview

Radiation-induced brain injury refers to a series of pathological changes related to healthy brain tissue damage in patients with head and neck tumors after radiotherapy, including a series of biochemical and cellular disorders such as capillary damage, blood-brain barrier disruption, abnormal autoimmune response, imbalance between oxidation and antioxidant balance, and inhibition of nerve cell development, as well as loss of endogenous neurogenesis, demyelination, and ablation of endogenous oligodendrocyte progenitor cells.5 Combined with the state of chronic neuroinflammation, Xi and memory dysfunction, local neurological deficits, and progressive dementia can eventually occur.6

Mechanism of Radiation-Induced Brain Injury

Ionizing Radiation Leads to Vascular Damage and Destruction of the Blood-Brain Barrier

The blood-brain barrier is a highly organized, multicellular structure that maintains immunochemical homeostasis in brain tissue by regulating molecular transendothelial transport between the parenchyma and systemic circulation, while also limiting the translocation of peripheral immune cells. It has been reported that, compared to brain parenchyma and vascular cells, low-dose radiation can induce damage and apoptosis in glial cells and vascular endothelial progenitor cells located in the ventricular zone.5 In addition, a growing number of clinical and preclinical studies have shown that the effects of radiation on parenchymal cells and brain physiology are exacerbated by associated cerebral microvascular endothelial injury, resulting in cerebrovascular inflammation, disruption of the blood-brain barrier, and imbalance of metabolic homeostasis.7–11 The vascular hypothesis is supported by a large body of data describing radiation-induced changes in vascular structure, including vasodilation, vascular lesions and endothelial progenitor cell depletion, senescence, and the appearance of abnormal endothelial cells, as well as activation of endothelial apoptosis and vascular inflammation.12,13 Quantitative studies of the brains of irradiated animals have also shown that endothelial cell nuclei and vascular length decrease with time and dose increase.14 The response of vascular tissue to radiation exhibits a two-phase pattern: the first is that the acute phase can occur within 24 hours of radiation, and the underlying mechanism is related to radiation-induced endothelial apoptosis. The second is the advanced stage, which takes several months to develop and is characterized by capillary collapse, basement membrane thickening, and loss of endothelial cell clonal activity.15 Dimitrievich et al16,17 have demonstrated that capillary sensitivity to a single X-ray dose of 200 to 2000 rad is significantly higher than that of larger vessels. Their study also showed that radiation injury was characterized by capillary destruction, extravasation of blood components, and pro-inflammatory changes. Evidence has been reported that the high vulnerability of the capillary layer to radiation is specifically due to the high radiation sensitivity of its endothelial cells, which are the major structural components of the capillary wall.18 In radiation-induced brain diseases, the microvascular system is the primary tissue-blood interface in the central nervous system that maintains and controls nerve tissue homeostasis.19 Extensive damage to parenchymal and/or stromal cells and progressive loss of their progenitor cells can lead to worsening of neuroinflammation and disruption of the blood-brain barrier (BBB), ultimately leading to white matter lesions.20–24 The endothelial cells of the microvascular bed include specific cellular phenotypes and represent part of the blood-brain barrier, which tightly controls brain immunochemical homeostasis.19 Thus, damage to the normal capillary endothelium by radiation can lead to disruption of the brain-blood barrier, which can lead to serious health consequences.

Ionizing Radiation Leads to Oxidative Stress

Early studies have shown that damage to the central nervous system is often associated with an inflammatory response. The mechanism of the inflammatory response in the brain differs from that of peripheral tissues, which may be related to stronger immunosuppression in this organ.25 As effector cells of the innate immune system of the brain and the most important antigen-presenting cells in the cerebral cortex, microglia play a key role in the initiation, development, and recovery of neuroinflammation.26

Studies have found that when the body receives radiotherapy, ionizing radiation deposits energy into tissue cells, and at the same time damages mitochondria, it will decompose water to produce reactive oxygen species (ROS), and the reactive oxygen species produced induce a series of strong oxidative stress responses, The causes of neuroinflammation are complex and diverse. Measuring inflammatory response indicators can serve as a method to evaluate the degree of oxidative stress. Researchers detected the levels of inflammatory responses in vitro or in vivo after irradiation with different doses of radiation, and the results showed that: tumor necrosis factor- α (TNF-α), increased expression of proinflammatory factors such as interleukin-1β (IL-1β), cyclic oxygenase 2 (COX-2), activation of transcription factors (AP-1), nuclear factors NFκB, CREB, and mRNA levels of several chemokines (MCP1/CCL2, Gro/KC/CXCL1) are upregulated.27–30 This, in turn, leads to further damage to cellular nucleic acids, proteins, and lipids, as well as extracellular matrix components, and local tissue damage promotes microglial activation.25,31 Initial acute radiation-induced microglial activation and neuroinflammatory responses, followed by persistent microglia secreting inflammatory cytokines and production of reactive oxygen species, have led to a cascade of chronic neuroinflammatory responses associated with hippocampal nerve development and cognitive impairment.21,30,32 Studies have demonstrated that the transcriptional profile of irradiated microglia is very similar to that obtained by microglia during aging, which is the greatest risk factor for cognitive decline and neurodegenerative diseases. In recent years, more and more studies have shown that hippocampal neurogenesis is related to cognitive function. Although the mechanism is not fully understood, a link has been reported between sustained microglial activation and impaired hippocampal neurogenesis.19,33

Ionizing Radiation Causes Nerve Cell Death

In recent years, a large number of studies have shown that radiation-induced brain injury is not the result of damage to a single cell or structure, but is caused by the interaction of multiple types of cells, among which astrocytes, endothelial cells, microglia, neurons and oligodendrocytes are the main ones. More recently, preclinical studies have focused on the hippocampus, one of two sites in the brain for adult neurogenesis that plays an important role in learning and memory9. The hippocampus is made up of three regions, the dentate gyrus (DG), CA3 and CA1, which are involved in both rodent and human cognition. In addition, DG is one of two sites of adult neurogenesis in the mammalian brain. Neuronal stem cells (NSCs) in DGs are both self-renewing and giving rise to new neurons, astrocytes, and oligodendrocytes.34,35 Endothelial cells and astrocytes have been shown to promote or modulate neurogenesis, creating a specific microenvironment conducive to this process. Additionally, a dose-dependent decrease in the number of neural stem cells (NSCs) has been observed, along with a reduction in the proliferation of surviving NSCs and a decrease in their differentiation into neurons.31,36 Radiation has been demonstrated to induce apoptosis in central nervous system (CNS) cells, with this phenomenon being particularly pronounced during the neonatal and early postnatal periods. Additionally, it has been observed in the brains of young adult rats. Neurons and glial cells in the lateral ventricles (LV), olfactory bulb (OB), neocortex, piriform and entorhinal cortex, dentate gyrus (DG), striatum, thalamus, amygdala, brainstem, and subcompartmental zone (SVZ) of the cerebellar white matter were all subject to apoptosis after a single dose of 2 Gy.37 A single high-dose (2–10 Gy) of rat whole-brain irradiation increases dramatically in hippocampal DG cell apoptosis within 3 to 6 hours and plateaus within 6 to 12 hours after radiation therapy.38–40 The number of apoptotic cells remains unchanged for one to nine months after radiation therapy.40–42

Low doses of ionizing radiation can also adversely affect metabolic pathways that are not directly involved in regulating apoptosis, such as acute or chronic irradiation affecting the ERK1/ERK2 (extracellular signal-regulated kinase) signaling pathway, as well as changes in Trp53 phosphorylation and p21 protein levels, thereby affecting neuronal survival after irradiation.43,44 At the same time, ionizing radiation can interfere with the expression of cell cycle regulatory proteins, leading to subsequent apoptosis, which is one of the main mechanisms by which ionizing radiation causes cell death within a few hours of irradiation.45 In addition, it has been found that radiation irradiation can lead to down-regulation of the anti-apoptotic gene Bax and up-regulation of the pro-apoptotic gene Bcl-2 in the mitochondrial pathway, promotes the release of cytochrome C (Cytc), activate Caspase-mediated apoptosis, and induce the occurrence of neuronal apoptosis.44,46

Prevention and Treatment of RBI

A large amount of research has shown that the reduction in hippocampal neurogenesis is related to spatial memory and learning.47,48 Researchers have conducted whole brain or local irradiation of rodent heads and subsequently found significant impairment in cognitive function in animal behavior tests such as Morris water maze (MWM), passive avoidance, Barnes maze, novel object recognition.49–53 Currently, in order to prevent the occurrence of Radiation-induced brain injury (RBI), many methods for preventing and treating radiation damage induced by ionizing radiation have been developed. The main methods used in clinics include physical protection and medical protection. The main drugs for prevention/treatment are as follows (Table 1):

Table 1 Summary of Therapeutic Strategies and Mechanisms for Radiation-Induced Brain Injury

  1. Anti-inflammatory treatments encompass a range of medications, including nonsteroidal anti-inflammatory drugs (NSAIDs), steroidal drugs, COX inhibitors, and PPAR (peroxisome proliferator-activated receptor) agonists, all of which suppress inflammatory responses. Additionally, hydroxychloroquine, morphine, tetrahydrofuran, and methoxyquinoline are utilized, alongside dextrocorticoids and neuroprotective agents such as carbamazepine, which inhibits damaging oxygen free radicals. Furthermore, various vitamins and antioxidants, including vitamin E, vitamin C, and selenium, also play a role in anti-inflammatory treatment.33,51,52 Clinically prescribed medications, including PPAR α and γ agonists, as well as RAS receptor blockers, prevent radiation-induced neuroinflammation and cognitive impairment. In addition, amifostine and hydrogen-rich water can attenuate damage caused by ionizing radiation by reducing inflammation and inhibiting oxidative stress through meso-oxygen and hydroxyl radicals.53–57 Some scholars have utilized hydrogen-rich water to treat rats with radiation-induced brain injury. The results indicate that a specific dosage of hydrogen-rich water intervention can significantly reduce the inflammatory response in rat brain tissue and assist in the restoration of cognitive function. This effect may be mediated through signaling pathways related to factors such as PI3K and Akt.58,59
  2. Neural stem cell transplantation: NSCs transplantation is considered to be an effective treatment strategy for various neurological diseases, characterized by a central nervous system (CNS) repair mechanism that restores tissue damage and salvages lost function. Cellular sources of NSCs include fetal and adult CNS-derived NSCs, neural progenitor cells, and a wide range of non-neural stem cells such as mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs). Since the function of normal NSCs may be adversely affected by radiation, there is a strong rationale for improving cognition after cranial irradiation based on the strategy of transplanting neural stem cells.60,61 Many preclinical studies have focused on xenografting human pluripotent NSCs (embryonic stem cells, ESC) into rodent hosts, and irradiation of rats two days after intrahippocampal transplantation of human NSCs has improved hippocampal-dependent cognitive impairment and preserved neurogenesis.35,62,63 A recent study examined the transplantation of human embryonic stem cell-derived oligodendrocytes into the brains of mice after a regimen of four weeks of fractionated irradiation at clinically relevant doses. The behavioral test results indicated that the cognitive function of C57BL/6J adult female mice was fully restored ten weeks post-oligodendrocyte brain transplantation. However, the recovery of motor function was found to necessitate the simultaneous transplantation of the cerebellum.64 Simultaneously, the research team optimized a protocol for deriving and selectively enriching late-stage oligodendrocyte progenitor cells from human embryonic stem cells (ESCs). They demonstrated that these cells have the capability to remyelinate the brain and improve behavioral deficits. The clinical impact of these studies can be substantial as the need to address quality of life in cancer survivors grows more pressing.65
  3. Targeted drug therapy: Radiotargeted therapy is an emerging technology that consists of specific small molecule antigens, such as nuclides or isoforms, that can bind to the cell surface, thereby reducing the cytotoxicity of Radiation-induced brain injury. Bevacizumab is a humanized monoclonal antibody that blocks vascular endothelial growth factor (VEGF) and competitively inhibits the binding of VEGF to endothelial cell surface receptors, reducing endothelial cell proliferation and neovascularization, and reducing vascular permeability by binding to VEGF.66 In a randomized, single-blind, prospective clinical study comparing bevacizumab with conventional corticosteroid therapy for Radiation-induced brain injury (RBI), 62.1% of patients in the bevacizumab group experienced significant improvement in symptoms of RBI, compared with 42.6% in the conventional hormone therapy group. Furthermore, bevacizumab was demonstrated to reduce symptoms of Radiation-induced brain injury and enhance overall outcomes.67 In addition to therapeutic approaches, emerging studies have suggested the potential utility of plasma-based biomarkers in detecting early radiation-induced brain injury, offering a non-invasive tool for patient monitoring and risk stratification.68

Summary and Outlook

At present, a large number of scientific studies have been conducted on the mechanism of injury and the prevention and treatment of RBI, and there are some medical measures to prevent the occurrence of Radiation-induced brain injury, such as the use of goggles, Optimize radiotherapy regimens and the avoidance of excessive use of radioactive technology.69 At the same time, many drugs can also reduce RBI in clinical practice, and these drugs can also improve oxygen metabolism, promote nerve cell regeneration, reduce inflammatory response and vascular inflammation, prevent brain cell damage, and promote further recovery of brain function and performance. In addition to drug therapy, there are a number of other treatments for Radiation-induced brain injury, including radiotargeted therapy, spinal deformity modification, and psychological support. Finally, psychoactive care can help patients better cope with Radiation-induced brain injury, rebuild self-confidence, reduce anxiety and depression, and urge patients to implement effective rehabilitation therapies. There is a growing interest in the prevention of radiation injury, especially cognitive impairment caused by radiotherapy for head and neck tumors.

In addition, in recent years, a large number of animal experimental studies have found that many drugs have radiation protection effects, which are expected to become new radiation protection agents, following amifostine, with the in-depth study of the role and mechanism of hydrogen-rich water, it is clearly found that hydrogen-rich water can significantly improve cognitive and learning and memory dysfunction after RBI, to reduce the occurrence of Radiation-induced brain injury, its mechanism of action may be closely related to inhibiting nerve cell apoptosis, promoting vascular structural integrity and nerve cell regeneration, and improving and reshaping synaptic plasticity. The translation of these promising preclinical discoveries into clinical practice offers hope for improving the quality of life of patients with brain tumors after radiotherapy. However, the overall mechanism of the RBI is complex and sophisticated, and so far, there are still many detailed regulatory mechanisms and integration mechanisms that we are not fully aware of. Therefore, a more in-depth and comprehensive discussion is still needed for RBI to provide more experimental basis and theoretical basis for the clinical treatment of Radiation-induced brain injury.

Data Sharing Statement

No new data were created.

Acknowledgments

We sincerely acknowledge the efforts of every author. Their valuable suggestions and the time and energy they invested in the initial draft writing and subsequent revisions of this paper have been of great significance.

Funding

This research received no external funding.

Disclosure

The authors declare no conflicts of interest. This paper has been uploaded to **preprint server ** as a preprint: https://papers.ssrn.com/sol3/papers.cfm?abstract_id=5064576.

References

1. Zhang Z, Jiang J, He Y, et al. Pregabalin mitigates microglial activation and neuronal injury by inhibiting HMGB1 signaling pathway in radiation-induced brain injury. J Neuroinflamm. 2022;19(1). doi:10.1186/s12974-022-02596-7

2. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca A Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.21660

3. Kantapan J, Intachai N, Khamto N, et al. Pentagalloyl glucose and cisplatin combination treatment exhibits a synergistic anticancer effect in 2D and 3D models of head and neck carcinoma. Pharmaceuticals. 2022;15(7):830. doi:10.3390/ph15070830

4. Bosch LVD, Schaaf AVD, Laan HPVD, et al. Comprehensive toxicity risk profiling in radiation therapy for head and neck cancer: a new concept for individually optimised treatment. Radiother Oncol. 2021;157:147–154. doi:10.1016/j.radonc.2021.01.024

5. Gorbunov NV, Kiang JG. Brain damage and patterns of neurovascular disorder after ionizing irradiation. complications in radiotherapy and radiation combined injury. Radiation Res. 2021;196(1):1–16. doi:10.1667/RADE-20-00147.1

6. Brown PD, Asher AL, Ballman KV, et al. NCCTG N0574 (Alliance): a Phase III randomized trial of whole brain radiation therapy (WBRT) in addition to radiosurgery (SRS) in patients with 1 to 3 brain metastases. J Clin Oncol. 2015;33(18_suppl):LBA4–LBA4. doi:10.1200/jco.2015.33.18_suppl.lba4

7. Naveed M, Zhou Q-G, Han F. Cerebrovascular inflammation: a critical trigger for neurovascular injury? Neurochem Int. 2019;126:165–177. doi:10.1016/j.neuint.2019.03.011

8. Development of Blood Vessel-Related Radiation Damage in the Fimbria of the Central Nervous System – PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/2298633/. Accessed February 19, 2025.

9. Greene-Schloesser DM, Moore E, Robbins ME. Molecular pathways: radiation-induced cognitive impairment. Clin Cancer Res. 2013;19(9):2294–2300. doi:10.1158/1078-0432.CCR-11-2903

10. Yin E, Nelson DO, Coleman MA, Peterson LE, Wyrobek AJ. Gene expression changes in mouse brain after exposure to low-dose ionizing radiation. Int J Radiation Biol. 2003;79(10):759–775. doi:10.1080/09553000310001610961

11. Hopewell’ JW, Campling D, Calvo W, Reinhold HS, Wilkinson’ JH, Yeung’ TK. Vascular irradiation damage: its cellular basis and likely consequences. British J Cancer. 1986;53:181–191.

12. O’Connor MM, Mayberg MR. Effects of radiation on cerebral vasculature: a review. Neurosurgery. 2000;46(1):138–151. doi:10.1093/neurosurgery/46.1.138

13. Yang Y, Li H, Xu Y, et al. Notch signaling mediates radiation-induced smooth muscle cell hypermuscularization and cerebral vasculopathy. Stroke. 2022;53(12):3751–3762. doi:10.1161/STROKEAHA.122.040697

14. Burns TC, Awad AJ, Li MD, Grant GA. Radiation-induced brain injury: low-hanging fruit for neuroregeneration. Neurosurg Focus. 2016;40(5):E3. doi:10.3171/2016.2.FOCUS161

15. Ungvari Z, Podlutsky A, Sosnowska D, et al. Ionizing radiation promotes the acquisition of a senescence-associated secretory phenotype and impairs angiogenic capacity in cerebromicrovascular endothelial cells: role of increased DNA damage and decreased DNA repair capacity in microvascular radiosensitivity. J Gerontol. 2013;68(12):1443–1457. doi:10.1093/gerona/glt057

16. Ljubimova BNV, Levitman MK, Plotnikova ED, Eidus LK. Endothelial cell population dynamics in rat brain after local irradiation. British J Radiol. 1991;64(766):934–940. doi:10.1259/0007-1285-64-766-934

17. Mendonca MS, Chin-Sinex H, Dhaemers R, Mead LE, Yoder MC, Ingram DA. Differential mechanisms of X-ray-induced cell death in human endothelial progenitor cells isolated from cord blood and adults. Radiation Res. 2011;176(2):208–216. doi:10.1667/RR2427.1

18. Peper M, Steinvorth S, Schraube P, et al. Neurobehavioral Toxicity Of Total Body Irradiation: A Follow-Up In Long-Term Survivors. Int J Radiation Oncol Biol Physics. 2000;46(2):303–311. doi:10.1016/S0360-3016(99)00442-3

19. Monje ML, Mizumatsu S, Fike JR, Palmer TD. Irradiation Induces Neural Precursor-Cell Dysfunction. Nature Med. 2002;8(9):955–962. doi:10.1038/nm749

20. Song H, Stevens CF, Gage FH. Astroglia induce neurogenesis from adult neural stem cells; 2002.

21. Casciati A, Dobos K, Antonelli F, et al. Age-related effects of X-ray irradiation on mouse hippocampus. Oncotarget. 2016;7:28040.

22. Makale MT, McDonald CR, Hattangadi-Gluth JA, Kesari S. Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours. Nat Rev Neurol. 2016;13(1):52–64. doi:10.1038/nrneurol.2016.185

23. Maazen RWMVD, Kleiboer BJ, Verhagen I, Kogel AJVD. Irradiation in vitro discriminates between different O-2A progenitor cell subpopulations in the perinatal central nervous system of rats. Radiation Res. 1991;128. doi:10.2307/3578068

24. Coderre JA, Morris GM, Micca PL, et al. Late effects of radiation on the central nervous system: role of vascular endothelial damage and glial stem cell survival. Radiation Res. 2006;166(3):495–503. doi:10.1667/RR3597.1

25. Gebicke-Haerter PJ. Microglia in neurodegeneration. Mol Aspects. 2001;54:47–58.

26. Yuan X, Yu Y, Chao Y, Yan H, Yang L, Fang A. Effects of RT-03 on biological behavior of irradiated microglia. 2021

27. Kyrkanides S, Olschowka JA, Williams JP, Hansen JT, O’banion MK. TNFa and IL-1b mediate intercellular adhesion molecule-1 induction via microglia-astrocyte interaction in CNS radiation injury. J Neuroimmunol. 1999;95(1–2):95–106. doi:10.1016/s0165-5728(98)00270-7

28. Ramanan S, Kooshki M, Zhao W, Hsu FC, Robbins ME. PPARα ligands inhibit radiation-induced microglial inflammatory responses by negatively regulating NF-κB and AP-1 pathways. Free Radic Biol Med. 2008;45(12):1695–1704. doi:10.1016/j.freeradbiomed.2008.09.002

29. Kalm M, Fukuda A, Fukuda H, et al. Transient inflammation in neurogenic regions after irradiation of the developing brain. Radiation Res. 2009;171(1):66–76. doi:10.1667/RR1269.1

30. Lee WH, Sonntag WE, Mitschelen M, Yan H, Lee YW. Irradiation induces regionally specific alterations in pro-inflammatory environments in rat brain. Int J Radiation Biol. 2010;86(2):132–144. doi:10.3109/09553000903419346

31. Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR. Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res. 2003;63:4021–4027.

32. Hong J-H, Chiang C-S, Campbell IL, Sun J-R, Withers HR, McBride WH. Induction of acute phase gene expression by brain irradiation. Int J Radiation Oncol Biol Physics. 1995;33(3):619–626. doi:10.1016/0360-3016(95)00279-8

33. Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302(5651):1760–1765. doi:10.1126/science.1088417

34. Adult Neurogenesis, Glia, and the Extracellular Matrix –PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/31051133/. Accessed February 19, 2025.

35. Ihunwo AO, Perego J, Martino G, Vicenzi E, Panina-Bordignon P. Neurogenesis and viral infection. Front Immunol. 2022;13:826091.

36. Bellinzona M, Gobbel GT, Shinohara C, Fike JR. Apoptosis is induced in the subependyma of young adult rats by ionizing irradiation. Neurosci Lett. 1996;208(3):163–166. doi:10.1016/0304-3940(96)12572-6

37. Ferrer I, Macaya A, Blanco TR, et al. Evidence of internucleosomal DNA fragmentation and identification of dying cells in x-ray-induced cell death in the developing brain. Int J Develop Neurosci. 1995;13(1):21–28. doi:10.1016/0736-5748(94)00064-a

38. Sasaki R, Matsumoto A, Itoh K, et al. Target cells of apoptosis in the adult murine dentate gyrus and o4 immunoreactivity after ionizing radiation. Neurosci Lett. 2000;279(1):57–60. doi:10.1016/S0304-3940(99)00910-6

39. Shinohara C, Gobbel GT, Lamborn KR, Tada E, Fike JR. Apoptosis in the subependyma of young adult rats after single and fractionated doses of X-rays. Cancer Res. 1997;57:2694–702.

40. Wei M, Feng S, Zhang L, et al. Active fraction combination from liuwei dihuang decoction improves adult hippocampal neurogenesis and neurogenic microenvironment in cranially irradiated mice. Front Pharmacol. 2021;12. doi:10.3389/fphar.2021.717719

41. Rola R, Raber J, Rizk A, et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol. 2004;188(2):316–330. doi:10.1016/j.expneurol.2004.05.005

42. Fan Y, Liu Z, Weinstein PR, Fike JR, Liu J. Environmental enrichment enhances neurogenesis and improves functional outcome after cranial irradiation. Eur J Neurosci. 2007;25(1):38–46. doi:10.1111/j.1460-9568.2006.05269.x

43. Silasi G, Diaz-Heijtz R, Besplug J, et al. Selective brain responses to acute and chronic low-dose x-ray irradiation in males and females. Biochem Biophys Res Commun. 2004;325(4):1223–1235. doi:10.1016/j.bbrc.2004.10.166

44. Limoli CL, Giedzinski E, Rola R, Otsuka S, Palmer TD, Fike JR. Radiation response of neural precursor cells: linking cellular sensitivity to cell cycle checkpoints, apoptosis and oxidative stress. Radiation Res. 2004;161(1):17–27. doi:10.1667/RR3112

45. Kee N, Sivalingam S, Boonstra R, Wojtowicz JM. The utility of Ki-67 and BrdU as proliferative markers of adult neurogenesis. J Neurosci Meth. 2002;115(1):97–105. doi:10.1016/S0165-0270(02)00007-9

46. Winocur G, Wojtowicz JM, Sekeres M, Snyder JS, Wang S. Inhibition of neurogenesis interferes with hippocampus-dependent memory function. Hippocampus. 2006;16(3):296–304. doi:10.1002/hipo.20163

47. Madsen TM, Kristjansen PEG, Bolwig TG, Wörtwein G. Arrested neuronal proliferation and impaired hippocampal function following fractionated brain irradiation in the adult rat. Neuroscience. 2003;119(3):635–642. doi:10.1016/S0306-4522(03)00199-4

48. Akiyama K, Tanaka R, Sato M, Takeda N. Cognitive dysfunction and histological findings in adult rats one year after whole brain irradiation. Neurologia Medico-Chirurgica. 2001;41(12):590–598. doi:10.2176/nmc.41.590

49. Schnegg CI, Greene-Schloesser D, Kooshki M, Payne VS, Hsu FC, Robbins ME. The PPARδ agonist GW0742 inhibits neuroinflammation, but does not restore neurogenesis or prevent early delayed hippocampal-dependent cognitive impairment after whole-brain irradiation. Free Radic Biol Med. 2013;61:1–9. doi:10.1016/j.freeradbiomed.2013.03.002

50. Lee TC, Greene-Schloesser D, Payne V, et al. Chronic administration of the angiotensin-converting enzyme inhibitor, ramipril, prevents fractionated whole-brain irradiation-induced perirhinal cortex-dependent cognitive impairment. Radiation Res. 2012;178(1):45–56. doi:10.1667/RR2731.1

51. Greene-Schloesser D, Payne V, Peiffer AM, et al. The peroxisomal proliferator-activated receptor (PPAR) α agonist, fenofibrate, prevents fractionated whole-brain irradiation-induced cognitive impairment. Radiation Res. 2014;181(1):33–44. doi:10.1667/RR13202.1

52. Microglia in Radiation-Induced Brain Injury: cellular and Molecular Mechanisms and Therapeutic Potential –PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/38867379/. Accessed February 19, 2025.

53. Reichman HR, Farrell CL, Maestro RFD. Effects of steroids and nonsteroid anti-inflammatory agents on vascular permeability in a rat glioma model. J Neurosurg. 1986;65. doi:10.3171/jns.1986.65.2.0233

54. Portnow J, Suleman S, Grossman SA, Eller S, Carson K. A Cyclooxygenase-2 (COX-2) inhibitor compared with dexamethasone in a survival study of rats with intracerebral 9L gliosarcomas. Neuro-Oncology. 2002;4. doi:10.1215/15228517-4-1-22

55. Feng Z, Zhang Y, Yang C, et al. Bioinspired and inflammation-modulatory glycopeptide hydrogels for radiation-induced chronic skin injury repair. Adv Healthcare Mater. 2023:12. doi:10.1002/adhm.202201671

56. Boutros SW, Zimmerman B, Nagy SC, Lee JS, Perez R, Raber J. Amifostine (WR-2721) mitigates cognitive injury induced by heavy ion radiation in male mice and alters behavior and brain connectivity. Front Physiol. 2021;12. doi:10.3389/fphys.2021.770502

57. Zhang J, Xue X, Han X, et al. Hydrogen-rich water ameliorates total body irradiation-induced hematopoietic stem cell injury by reducing hydroxyl radical. Oxid Med Cell Longev. 2017;2017(1). doi:10.1155/2017/8241678

58. Rochette L, Zeller M, Cottin Y, Vergely C. Antitumor activity of protons and molecular hydrogen: underlying mechanisms. Cancers. 2021;14(1):13. doi:10.3390/cancers14010013

59. Wang Y, Chen X, Liu M, et al. Effect of hydrogen-rich water on brain injury in rats with acute radiation based on PI3K/AKT/ Caspase-9 signaling pathway. J Environ Occup Med. 2023;40. doi:10.11836/JEOM22406

60. Zhai QY, Ren YQ, Ni QS, Song ZH, Ge KL, Guo YL. Transplantation of human umbilical cord mesenchymal stem cells-derived neural stem cells pretreated with neuregulin1β ameliorate cerebral ischemic reperfusion injury in rats. Biomolecules. 2022;12(3):428. doi:10.3390/biom12030428

61. Kremer LPM, Cerrizuela S, Dehler S, et al. High throughput screening of novel AAV capsids identifies variants for transduction of adult NSCs within the subventricular zone. Mol Ther Meth Clin Develop. 2021;23:33–50. doi:10.1016/j.omtm.2021.07.001

62. Brown C, McKee C, Halassy S, Kojan S, Feinstein DL, Chaudhry GR. Neural stem cells derived from primitive mesenchymal stem cells reversed disease symptoms and promoted neurogenesis in an experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. Stem Cell Res Ther. 2021;12(1):12. doi:10.1186/s13287-021-02563-8

63. Acharya MM, Christie LA, Lan ML, et al. Human neural stem cell transplantation ameliorates radiation-induced cognitive dysfunction. Cancer Res. 2011;71. doi:10.1158/0008-5472.CAN-11-0027

64. Piao J, Major T, Auyeung G, et al. Human embryonic stem cell-derived oligodendrocyte progenitors remyelinate the brain and rescue behavioral deficits following radiation. Cell Stem Cell. 2015;16(2):198–210. doi:10.1016/j.stem.2015.01.004

65. Acharya MM, Christie LA, Lan ML, et al. Rescue of radiation-induced cognitive impairment through cranial transplantation of human embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2009, 106, doi:10.1073/pnas.0909293106.

66. Xu Y, Rong X, Hu W, et al. Bevacizumab monotherapy reduces radiation-induced brain necrosis in nasopharyngeal carcinoma patients: a randomized controlled trial. Int J Radiat Oncol Biol Phys. 2018;101(5):1087–1095. doi:10.1016/j.ijrobp.2018.04.068

67. Intra-Cisterna-Magna Bevacizumab Injection (ICM-BI) Reproduces Pathological Alterations of Cerebral Small Vessel Diseases. Available from: https://pubmed.ncbi.nlm.nih.gov/39450471/. Accessed February 19, 2025.

68. Makranz C, Lubotzky A, Zemmour H, et al. Short report: plasma based biomarkers detect radiation induced brain injury in cancer patients treated for brain metastasis: a pilot study. PLoS One. 2023;18(11):e0285646. doi:10.1371/journal.pone.0285646IF:2.9Q1

69. Wu J, Lu AD, Zhang LP, Zuo YX, Jia YP. Study of clinical outcome and prognosis in pediatric core binding factor-acute myeloid leukemia. Zhonghua Xue Ye Xue Za Zhi. 2019;40(1):52–57. [PMID: 30704229; PMCID: PMC7351698]. doi:10.3760/cma.j.issn.0253-2727.2019.01.010

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.