1
|
Kumar R, Kumari P, Kumar R. Central Nervous System Response Against Ionizing Radiation Exposure: Cellular, Biochemical, and Molecular Perspectives. Mol Neurobiol 2025; 62:7268-7295. [PMID: 39875779 DOI: 10.1007/s12035-025-04712-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Gamma radiation is known to induce several detrimental effects on the nervous system. The hippocampus region, specifically the dentate gyrus (DG) and subventricular zone (SVZ), have been identified as a radiation-sensitive neurogenic niche. Radiation alters the endogenous redox status of neural stem cells (NSCs) and other proliferative cells, especially in the hippocampus region, leading to oxidative stress, neuroinflammation, and cell death. Planned (i.e., radiotherapy of brain tumor patients) or unplanned radiation exposure (i.e., accidental radiation exposure) can induce nonspecific damage to neuronal tissues, resulting in chronic or acute radiation syndrome. Although anatomical alterations in the neuronal tissues have been reported at higher doses of gamma radiation, biochemical and molecular perturbations may be evident even at much lower radiation doses. They may manifest in the form of neuronal deficits and cognitive impairment. In the present review, several molecular events and signaling pathways, such as oxidative stress, neuroinflammation, apoptosis, cognition, neuroplasticity, and neurotoxicity induced in neuronal cells upon ionizing radiation exposure, are reviewed. Furthermore, brain-specific radioprotectors and mitigators that protect normal neuronal cells and tissues against ionizing radiation during radiotherapy of cancer patients or nuclear emergencies are also discussed.
Collapse
Affiliation(s)
- Ravi Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Pratibha Kumari
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
2
|
Wang P, Liu J, Zhang M, Yang J, Lian P, Cheng X, Qin J. Radiation Exposure Induced Blood-Brain Barrier Injury via Mitochondria-Mediated Sterile Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02356. [PMID: 40433769 DOI: 10.1002/advs.202502356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Radiation-induced brain injury (RIBI) is caused by exposure to high doses of ionizing radiation and characterized by severe cognitive dysfunction and brain necrosis. However, the pathogenesis of RIBI is not fully understood, and no effective intervention is available. This work describes a blood-brain barrier (BBB) microphysiological system (MPS), that allowed to explore the responses of BBB and distinct brain cells to radiation exposure. Following acute exposure to radiation of X-ray or γ-ray, characteristic RIBI-associated pathological responses are observed, including BBB compromise, DNA breaks, inhibited cell proliferation, cell hypertrophy, and proinflammatory cytokine release. Among the distinctive types of cells, brain endothelial cells show the highest radiosensitivity as compared to other cells in the MPS. Intriguingly, X-ray and γ-ray radiation consistently induce prominent sterile inflammation responses, especially type I interferon response, in the BBB MPS. These responses are mediated by radiation-induced mitochondrial DNA release and subsequent activation of cGAS-STING signaling pathway. Furthermore, it is found abrocitinib (JAK1 inhibitor) and idebenone (mitochondrial protectant) can attenuate radiation-induced inflammation and ameliorate injuries in the BBB MPS. These findings reveal the involvement of mitochondria-mediated sterile inflammation in RIBI pathogenesis, identifying mitochondria as a potential target for new radioprotective measures.
Collapse
Affiliation(s)
- Peng Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jiayue Liu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Min Zhang
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Juan Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, P. R. China
| | - Peihan Lian
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Xiu Cheng
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jianhua Qin
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100000, China
| |
Collapse
|
3
|
Xu J, Zhang L, Liu T, Liu Q, Zhu J. Comparison of the effects of IMRT and IMPT on MRI features of normal brain tissue: a retrospective study. BMC Med Imaging 2025; 25:176. [PMID: 40399870 PMCID: PMC12096751 DOI: 10.1186/s12880-025-01724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
OBJECTIVE This study aims to analyze magnetic resonance imaging (MRI) feature changes across different dose regions to investigate the differences in imaging feature alterations induced by Intensity-Modulated Radiation Therapy (IMRT) and Intensity-Modulated Proton Therapy (IMPT) in normal brain tissue. And evaluate the potential application value of radiomic features in dose optimization and biological dose assessment. METHODS A retrospective analysis included 113 patients undergoing brain-target radiotherapy: 44 receiving IMPT (average total dose: 5713.63 ± 774.32 cGy) and 69 receiving IMRT (average total dose: 5489.86 ± 627.05 cGy). There was no significant difference in prescribed doses between groups (p < 0.05). MRI data were collected pre-treatment and one month post-treatment, including T1, enhanced T1, T2, T2-FLAIR, and ADC sequences. Images underwent preprocessing and rigid registration on the 3D Slicer platform. Dose regions (90%, 70%, 50%, 30% of the total dose) were segmented and classified into high (70-90%), medium (50-70%), and low (30-50%) dose regions as volumes of interest (VOI). Radiomic features were extracted from pre- and post-treatment MRI datasets, and Delta features were calculated as the difference between pre- and post-treatment data. Non-parametric tests, t-tests, and Mann-Whitney U tests were used to identify significant feature changes, with effect size analysis to compare the magnitude of changes. Multiple testing correction was applied to reduce false positives. Finally, box plots and the Mapping of Dose Distribution and Differences in Radiomics Feature Variation were utilized to visually illustrate the extent of brain tissue changes in each dose region following radiotherapy. RESULTS Non-parametric tests indicated that more radiomic features exhibited significant changes in the IMRT group (p < 0.001). Effect size analysis also showed that the majority of features demonstrated greater and more pronounced changes within dose regions in the IMRT group (p < 0.05). Further correlation analysis revealed that most features (33/34) had weak correlations with dose-volume parameters (r < 0.3), suggesting that treatment modality itself may be the primary driver of these alterations rather than dose distribution alone. This finding highlights the need to refine traditional dose-based radiotherapy evaluation by incorporating biological effect considerations. Additionally, Moreover, the feature Delta_wavelet-HHL.65 exhibited the most significant change, with an effect size of -0.8 in the IMRT group. It demonstrated a moderate-to-high correlation with dose-volume parameters in the medium-to-high dose regions, with a maximum correlation coefficient of 0.65 (p < 0.001). This suggests that Delta_wavelet-HHL.65 may serve as a potential biomarker for reflecting local dose effects, contributing to the optimization of radiotherapy dose distribution. CONCLUSIONS This study revealed that IMRT and IMPT induce distinct radiomic feature changes under comparable dose conditions, and these differences are not solely determined by dose distribution. This suggests that radiotherapy evaluation should extend beyond physical dose metrics to incorporate biological effect dimensions. Furthermore, Delta_wavelet-HHL.65 demonstrated the most pronounced change in the IMRT group and exhibited correlations with dose-volume parameters in the low-to-medium dose regions, highlighting its potential for radiotherapy optimization. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jian Xu
- Department of Radiation Physics Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital), Jinan, 250117, P. R. China
| | - Lili Zhang
- Liaocheng People's Hospital of Shandong Province, Liaocheng, 252000, P. R. China
| | - Tingting Liu
- Sun Yat-sen University Cancer Center Gansu Hospital, Lanzhou, 730050, P. R. China
| | - Qingzeng Liu
- Department of Radiation Physics Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital), Jinan, 250117, P. R. China
| | - Jian Zhu
- Department of Radiation Physics Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, P. R. China.
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital), Jinan, 250117, P. R. China.
| |
Collapse
|
4
|
Kiang JG, Cannon G, Zhai M, Olson MG, Woods AK, Cleveland KS, Ellery H, Xu F, Xiao M. A Combined Therapy of Pegylated G-CSF with Ciprofloxacin Mitigates Damage Induced by Lethal Ionizing Radiation to the Bone Marrow, Spleen, and Ileum by Increasing AKT Activation but Decreasing IL-18, C3, and miR-34a. Radiat Res 2025; 203:341-356. [PMID: 40181563 DOI: 10.1667/rade-24-00266.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Ciprofloxacin (CIP) was found to enhance pegylated G-CSF therapy (PEG, Neulasta®)-induced survival from 30% to 85% after ionizing radiation exposure. This combined therapy significantly mitigated radiation-induced brain hemorrhage through its capability to improve platelet recovery. This study tested whether this combined treatment also mitigated gastrointestinal damage from radiation. B6D2F1 female mice were exposed to 60Co γ radiation. CIP was fed daily to mice for up to 14 days. PEG was injected on day 1, and then weekly up to day 14. For the early time point study, blood, femurs, spleen, and ileum were collected on days 2, 4, 9, and 15 postirradiation. Bone marrow cells were counted; spleen weights and splenocyte counts were measured; and ileum histopathology was examined and analyzed. AKT, ERK, JNK, p38, claudin 2, NF-kB, Bax, Bcl-2, and gasdermin D were measured in ileum lysates using Western blotting while miR-34a was measured by reverse transcription followed by real-time-PCR, and citrulline was measured by colorimetric assay. In serum, interleukin-18 (IL-18) was measured by Luminex assay and complement protein 3 (C3) was detected by ELISA. The bacterial DNA load in livers was measured by real-time PCR. Radiation depleted bone marrow cells in femurs beginning day 2 through day 15 postirradiation, which was mitigated by PEG or CIP+PEG on day 9 through day 15 and by CIP on day 15, respectively. Radiation exposure led to decreased spleen weight on day 2 through day 15, while PEG or CIP+PEG significantly mitigated the reduction on day 9 through day 15. Radiation exposure reduced splenocyte counts on day 2 through day 15 postirradiation, but that was mitigated by PEG or CIP+PEG on day 15. Ileum histology showed that radiation decreased villus height on day 2 through day 15; CIP mitigated the reduction on day 15, whereas PEG+CIP mitigated it on day 2 through 15. Villus widths were increased on day 2 through day 15, while PEG+CIP effectively decreased them on day 4 through day 15. Crypt depth was reduced by radiation on day 2, but returned to the baseline on day 4 through 15. CIP or CIP+PEG transiently increased the depth only on day 4. Crypt counts were reduced by radiation on days 2 and 4, but returned to the baseline on days 9 and 15, regardless of individual drugs or combinations. Citrulline data confirmed the villus height recovery. Radiation significantly increased pro-inflammatory cytokine IL-18 on days 4 and 9, which was mitigated by PEG alone or PEG+CIP, but not by CIP alone. Radiation increased C3 on day 9 in ileum and serum. The serum C3 was positively associated with the serum IL-18 levels and negatively correlated with the crypt depth. Radiation-induced decreases in claudin 2 (a tight junction marker) in ileum and increases in bacterial DNA in livers were mitigated by PEG+CIP. Radiation did not reduce NF-kB and its activation but reduced Bcl-2 expression, which was not significantly recovered by any individual drug or combination. However, the PEG and CIP combination significantly decreased NF- kB and BAX. In contrast, radiation increased miR-34a and cleaved gasdermin D, which CIP+PEG effectively mitigated. This was confirmed by immunohistochemistry. The results taken together suggest that PEG+CIP combined treatment was effective in mitigating the radiation-induced bone marrow, spleen, and ileum injury. The mitigative effect of this combined treatment was mediated by increases in G-CSF levels that suppress miR-34a, thereby probably leading to decreased gasdermin D-mediated pyroptosis.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Georgetta Cannon
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Min Zhai
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Matthew G Olson
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Akeylah K Woods
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Katherine S Cleveland
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Hengying Ellery
- Department of Large Animal Research, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Feng Xu
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Mang Xiao
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| |
Collapse
|
5
|
Wang Q, Guo C, Wang T, Shuai P, Wu W, Huang S, Li Y, Zhao P, Zeng C, Yi L. Drug protection against radiation-induced neurological injury: mechanisms and developments. Arch Toxicol 2025; 99:851-863. [PMID: 39724149 DOI: 10.1007/s00204-024-03933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
In daily life, individuals are frequently exposed to various forms of radiation, which, when adhering to safety standards, typically result in relatively minor health effects. However, accidental exposure to radiation levels that exceed these safety standards can lead to significant health consequences. This study focuses on the analysis of radiation-induced damage to the nervous system and the mechanisms of pharmacological protection. The findings indicate that radiation can adversely affect neural structures, memory, and neurobehaviour. A range of pharmacological agents, including traditional Chinese medicine, Western medicine, and other therapeutic drugs, can be employed to safeguard the nervous system from radiation damage. The primary protective mechanisms of these agents encompass antioxidant effects, attenuation of apoptosis, and reduction of neurogenesis. A comprehensive review of these topics will offer new insights for the development and investigation of drugs aimed at mitigating radiation-induced damage to the nervous system.
Collapse
Affiliation(s)
- Qingyu Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Caimao Guo
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tiantian Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Peimeng Shuai
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenyu Wu
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shuqi Huang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuanyuan Li
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pei Zhao
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengkai Zeng
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Lan Yi
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
Li X, Hua S, Zhong D, Zhou M, Ding Z. Metal-organic framework-edaravone nanoparticles for radiotherapy-induced brain injury treatment. Biomaterials 2025; 314:122868. [PMID: 39413653 DOI: 10.1016/j.biomaterials.2024.122868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/10/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
Cranial radiotherapy may cause damage to normal brain tissues and induce cognitive dysfunction, so developing an effective strategy to prevent radiotherapy-induced brain injury is essential. Metal-organic frameworks (MOFs) can be used as vectors for the delivery of neuroprotective drugs due to their high drug loading capacity and low toxicity. In this study, we synthesized MIL-53(Cr) nanoparticles, which were used to deliver edaravone, and modified the surface of the nanoparticles with polyethylene glycol and Angiopep-2 (EDA@MIL-53(Cr)-P/A) to improve their oral bioavailability and ability to cross the blood-brain barrier (BBB). We confirmed that MIL-53(Cr)-P/A nanoparticles could achieve the sustained release of edaravone and enhance its ability to cross the BBB. The results of in vitro experiments showed that EDA@MIL-53(Cr)-P/A could exert radioprotective effects on HT22 and BV2 cells. We also demonstrated that EDA@MIL-53(Cr)-P/A could alleviate brain injury and cognitive dysfunction in mice receiving whole-brain irradiation. Mechanistically, EDA@MIL-53(Cr)-P/A alleviated irradiation-induced brain damage by inhibiting oxidative stress, DNA damage, apoptosis and inflammatory reactions. This study provides a new strategy for the protection against radiotherapy-induced brain injury.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Zhejiang University, Haining, 314400, China
| | - Shiyuan Hua
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Danni Zhong
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Zhejiang University, Haining, 314400, China; State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, 310009, China.
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
7
|
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. [PMID: 39803653 PMCID: PMC11711531 DOI: 10.62347/beau4974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
Radioactive brain injury, a severe complication ensuing from radiotherapy for head and neck malignancies, frequently manifests as cognitive impairment and substantially diminishes patients' quality of life. Despite its profound impact, the pathogenesis of this condition remains inadequately elucidated, and efficacious treatments are notably absent in clinical practice. Consequently, contemporary interventions predominantly focus on symptom alleviation rather than achieving a radical cure or reversing the injury process. This article provides a comprehensive review of the various pathogenic mechanisms and therapeutic strategies associated with radioactive brain injury, offering insights that may guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yaru Wang
- Department of Oncology, Anhui Medical UniversityHefei 230000, Anhui, China
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical UniversityHefei 230000, Anhui, China
| | - Xiaoqing Bao
- Department of Oncology, Anhui Medical UniversityHefei 230000, Anhui, China
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical UniversityHefei 230000, Anhui, China
| | - Yu Zhang
- Department of Oncology, Anhui Medical UniversityHefei 230000, Anhui, China
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical UniversityHefei 230000, Anhui, China
| | - Qibing Wu
- Department of Oncology, Anhui Medical UniversityHefei 230000, Anhui, China
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical UniversityHefei 230000, Anhui, China
| |
Collapse
|
8
|
Kirti, Sharma AK, Yashavarddhan MH, Kumar R, Shaw P, Kalonia A, Shukla SK. Exosomes: A new perspective for radiation combined injury as biomarker and therapeutics. Tissue Cell 2024; 91:102563. [PMID: 39270512 DOI: 10.1016/j.tice.2024.102563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
Radiation Combined Injuries (RCI) pose formidable public health risks, particularly in the context of nuclear incidents, necessitating specialized treatments and development of biomarkers. RCI encompasses instances where ionizing radiation exposure coincides with burns, wounds, or trauma. However, the limited understanding of cellular responses hinders progress in developing effective therapies. This article underscores the pivotal role of exosomes, nano-sized particles (30-120 nm) actively secreted by cells, in addressing the intricate challenges posed by RCI. Exosomes serve as vehicles for the transportation of bioactive molecules, including proteins, lipids, and miRNA, thereby facilitating processes critical to radiotherapy, burn injury, and wound healing. Exosomes hold significant promise for the transformation of RCI management by reducing inflammation, promoting wound healing, managing sepsis, altering immunological responses, and modulating signal transduction pathways. Moreover, exosomes are also being explored as biomarker for various diseases and stress conditions including radiation exposure and associated injuries. This comprehensive review highlights the burgeoning potential of exosomes in advancing the management of RCI, thereby enhancing public health preparedness and response.
Collapse
Affiliation(s)
- Kirti
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India
| | - Ajay Kumar Sharma
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India.
| | - M H Yashavarddhan
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India
| | - Rishav Kumar
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India
| | - Priyanka Shaw
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India
| | - Aman Kalonia
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India
| | - Sandeep Kumar Shukla
- Radiation Combined Injuries Research Department, Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Timarpur, Delhi 110054, India.
| |
Collapse
|
9
|
Wang X, Wang Y, Li Y, Lu H, Mo D, Liu Z, Gao L, Zhao Y, Zhao L, Huang Y, Fan Y, Wang D. The initial implementation of the transverse bone transport technique in the post-radiation region of the mandible. A pre-clinical in vivo study. BMC Oral Health 2024; 24:1434. [PMID: 39587575 PMCID: PMC11587573 DOI: 10.1186/s12903-024-05175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND To link the treatment of radiation injury with angiogenesis, and to design and seek a new therapeutic technique for the prevention and treatment of radiation injury. METHODS The transverse bone transport device for rabbit mandible was designed and manufactured. Eighteen New Zealand white rabbits were randomly divided into a radiotherapy group and a normal group. The radiotherapy group received 18 Gy of radiation, and the device was implanted two weeks later. After a 7-day incubation period, transverse transportation was performed at a speed of 0.5 circles (0.4 mm) per day, with an 8-day cycle and a total traction distance of 3.2 mm. CBCT, Micro CT, and histological staining were employed to assess the dynamics of movement, osteogenesis, and angiogenesis. RESULTS The transverse bone transport model of rabbit mandible was successfully established. CBCT revealed that the transport height in the normal and radiotherapy groups were 3.24 ± 0.17 mm and 3.22 ± 0.19 mm respectively. Micro CT analysis showed an increase in BV/TV and Tb.N over time, while Tb.Sp decreased; differences in BV/TV existed at 2 weeks but disappeared thereafter; differences in Tb.N and Tb.Sp persisted at 2 and 4 weeks. Histological staining using HE, Masson, and IHC demonstrated good bone maturity accompanied by rich neovascularization, and this was also confirmed by ImageJ software analysis. CONCLUSIONS The transverse bone transport was employed for the first time in the radiation-induced mandibular damage, thereby establishing a basis for further investigation into its clinical efficacy, application value, and underlying mechanisms. This breakthrough offers novel prospects for clinical interventions.
Collapse
Affiliation(s)
- Xian Wang
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Yuetong Wang
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Yuetao Li
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Haoyu Lu
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Dongqin Mo
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Zhiqing Liu
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Linjing Gao
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Yanfei Zhao
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Lixiang Zhao
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Yude Huang
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Yiyang Fan
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China
| | - Daiyou Wang
- College & Hospital of Stomatology, Guangxi Medical University, No.10 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, P.R. China.
| |
Collapse
|
10
|
Li X, Ding Z. Cognitive dysfunction induced by cranial radiotherapy: mechanisms and therapeutic methods. Brain Res Bull 2024; 218:111106. [PMID: 39447765 DOI: 10.1016/j.brainresbull.2024.111106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cranial radiotherapy can damage normal brain tissues, inducing cognitive dysfunction in patients. Radiotherapy-induced cognitive dysfunction is associated with hippocampal injury, white matter damage and microvascular injury. In this study, the mechanisms of cognitive dysfunction induced by cranial radiotherapy and combined chemoradiotherapy are reviewed, and the advances in therapeutic methods for radiotherapy-induced brain injury are summarized. The mechanisms of radiotherapy-induced brain injury include a decline of neurogenesis, impairment of neurons and glial cells, vascular injury, oxidative stress and DNA damage, cell death, and inflammatory response. Disruption of the bloodbrain barrier (BBB) increases the exposure of the brain to chemotherapeutic agents, thus exacerbating radiotherapy-induced brain damage. The current methods used to prevent radiotherapy-induced brain injury mainly include precision radiotherapy, stem cell transplantation, and treatment with neuroprotective drugs. The combined application of precision radiotherapy and neuroprotective drugs, including antioxidants, anti-inflammatory agents and other drugs, might exert better neuroprotective effects. To resolve the issues of neuroprotective drugs, such as difficulty in crossing the BBB, nanoenzymes and drug delivery nano-systems could be applied in the future.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Wu Y, Wang H, Liang Z, Cui J, Liu S, Chen Y, Pi D, Ouyang M. Elucidating the mechanism of traditional Chinese medicine formula (Yifei-sanjie Pill) in alleviating the chemobrain based on network pharmacology and experimental verification. J Tradit Complement Med 2024. [DOI: 10.1016/j.jtcme.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
12
|
Chamseddine I, Shah K, Lee H, Ehret F, Schuemann J, Bertolet A, Shih HA, Paganetti H. Decoding Patient Heterogeneity Influencing Radiation-Induced Brain Necrosis. Clin Cancer Res 2024; 30:4424-4433. [PMID: 39106090 PMCID: PMC11444871 DOI: 10.1158/1078-0432.ccr-24-1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/27/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE In radiotherapy (RT) for brain tumors, patient heterogeneity masks treatment effects, complicating the prediction and mitigation of radiation-induced brain necrosis. Therefore, understanding this heterogeneity is essential for improving outcome assessments and reducing toxicity. EXPERIMENTAL DESIGN We developed a clinically practical pipeline to clarify the relationship between dosimetric features and outcomes by identifying key variables. We processed data from a cohort of 130 patients treated with proton therapy for brain and head and neck tumors, utilizing an expert-augmented Bayesian network to understand variable interdependencies and assess structural dependencies. Critical evaluation involved a three-level grading system for each network connection and a Markov blanket analysis to identify variables directly impacting necrosis risk. Statistical assessments included log-likelihood ratio, integrated discrimination index, net reclassification index, and receiver operating characteristic (ROC). RESULTS The analysis highlighted tumor location and proximity to critical structures such as white matter and ventricles as major determinants of necrosis risk. The majority of network connections were clinically supported, with quantitative measures confirming the significance of these variables in patient stratification (log-likelihood ratio = 12.17; P = 0.016; integrated discrimination index = 0.15; net reclassification index = 0.74). The ROC curve area was 0.66, emphasizing the discriminative value of nondosimetric variables. CONCLUSIONS Key patient variables critical to understanding brain necrosis post-RT were identified, aiding the study of dosimetric impacts and providing treatment confounders and moderators. This pipeline aims to enhance outcome assessments by revealing at-risk patients, offering a versatile tool for broader applications in RT to improve treatment personalization in different disease sites.
Collapse
Affiliation(s)
- Ibrahim Chamseddine
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Keyur Shah
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hoyeon Lee
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Felix Ehret
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité – Universitätsmedizin Berlin, Germany
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Alejandro Bertolet
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Helen A. Shih
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
13
|
Zhou L, Zhu J, Liu Y, Zhou P, Gu Y. Mechanisms of radiation-induced tissue damage and response. MedComm (Beijing) 2024; 5:e725. [PMID: 39309694 PMCID: PMC11413508 DOI: 10.1002/mco2.725] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Radiation-induced tissue injury (RITI) is the most common complication in clinical tumor radiotherapy. Due to the heterogeneity in the response of different tissues to radiation (IR), radiotherapy will cause different types and degrees of RITI, which greatly limits the clinical application of radiotherapy. Efforts are continuously ongoing to elucidate the molecular mechanism of RITI and develop corresponding prevention and treatment drugs for RITI. Single-cell sequencing (Sc-seq) has emerged as a powerful tool in uncovering the molecular mechanisms of RITI and for identifying potential prevention targets by enhancing our understanding of the complex intercellular relationships, facilitating the identification of novel cell phenotypes, and allowing for the assessment of cell heterogeneity and spatiotemporal developmental trajectories. Based on a comprehensive review of the molecular mechanisms of RITI, we analyzed the molecular mechanisms and regulatory networks of different types of RITI in combination with Sc-seq and summarized the targeted intervention pathways and therapeutic drugs for RITI. Deciphering the diverse mechanisms underlying RITI can shed light on its pathogenesis and unveil new therapeutic avenues to potentially facilitate the repair or regeneration of currently irreversible RITI. Furthermore, we discuss how personalized therapeutic strategies based on Sc-seq offer clinical promise in mitigating RITI.
Collapse
Affiliation(s)
- Lin Zhou
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Jiaojiao Zhu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Yuhao Liu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ping‐Kun Zhou
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Yongqing Gu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunanChina
- College of Life SciencesHebei UniversityBaodingChina
| |
Collapse
|
14
|
Lan J, Ren Y, Liu Y, Chen L, Liu J. A bibliometric analysis of radiation-induced brain injury: a research of the literature from 1998 to 2023. Discov Oncol 2024; 15:364. [PMID: 39172266 PMCID: PMC11341524 DOI: 10.1007/s12672-024-01223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RIBI) is a debilitating sequela after cranial radiotherapy. Research on the topic of RIBI has gradually entered the public eye, with more innovations and applications of evidence-based research and biological mechanism research in the field of that. This was the first bibliometric analysis on RIBI, assessing brain injury related to radiation articles that were published during 1998-2023, to provide an emerging theoretical basis for the future development of RIBI. METHODS Literature were obtained from the Web of Science Core Collection (WOSCC) from its inception to December 31, 2023. The column of publications, author details, affiliated institutions and countries, publication year, and keywords were also recorded. RESULTS A total of 2543 journal articles were selected. The annual publications on RIBI fluctuated within a certain range. Journal of Neuro-oncology was the most published journal and Radiation Oncology was the most impactful one. LIMOLI CL was the most prolific author with 37 articles and shared the highest h-index with BARNETT GH. The top one country and institutions were the USA and the University of California System, respectively. Clusters analysis of co-keywords demonstrated that the temporal research trends in this field primarily focused on imaging examination and therapy for RIBI. CONCLUSION This study collects, visualizes, and analyzes the literature within the field of RIBI over the last 25 years to map the development process, research frontiers and hotspots, and cutting-edge directions in clinical practice and mechanisms related to RIBI.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yifan Ren
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuyang Liu
- Department of Neurosurgery, The 920th Hospital of Joint Logistics Support Force, Kunming, 650032, Yunnan, China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Jialin Liu
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
15
|
Kiang JG, Cannon G, Singh VK. An Overview of Radiation Countermeasure Development in Radiation Research from 1954 to 2024. Radiat Res 2024; 202:420-431. [PMID: 38964743 PMCID: PMC11385179 DOI: 10.1667/rade-24-00036.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Preparation for medical responses to major radiation accidents, further driven by increases in the threat of nuclear warfare, has led to a pressing need to understand the underlying mechanisms of radiation injury (RI) alone or in combination with other trauma (combined injury, CI). The identification of these mechanisms suggests molecules and signaling pathways that can be targeted to develop radiation medical countermeasures. Thus far, the United States Food and Drug Administration (U.S. FDA) has approved seven countermeasures to mitigate hematopoietic acute radiation syndrome (H-ARS), but no drugs are available for prophylaxis and no agents have been approved to combat the other sub-syndromes of ARS, let alone delayed effects of acute radiation exposure or the effects of combined injury. From its inception, Radiation Research has significantly contributed to the understanding of the underlying mechanisms of radiation injury and combined injury, and to the development of radiation medical countermeasures for these indications through the publication of peer-reviewed research and review articles.
Collapse
Affiliation(s)
- Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Department of Pharmacology and Molecular Therapeutics, School of Medicine
- Department of Medicine, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Georgetta Cannon
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Vijay K Singh
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Department of Pharmacology and Molecular Therapeutics, School of Medicine
| |
Collapse
|
16
|
Gupta K, Perkerson RB, Parsons TM, Angom R, Amerna D, Burgess JD, Ren Y, McLean PJ, Mukhopadhyay D, Vibhute P, Wszolek ZK, Zubair AC, Quiñones-Hinojosa A, Kanekiyo T. Secretome from iPSC-derived MSCs exerts proangiogenic and immunosuppressive effects to alleviate radiation-induced vascular endothelial cell damage. Stem Cell Res Ther 2024; 15:230. [PMID: 39075600 PMCID: PMC11287895 DOI: 10.1186/s13287-024-03847-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Radiation therapy is the standard of care for central nervous system tumours. Despite the success of radiation therapy in reducing tumour mass, irradiation (IR)-induced vasculopathies and neuroinflammation contribute to late-delayed complications, neurodegeneration, and premature ageing in long-term cancer survivors. Mesenchymal stromal cells (MSCs) are adult stem cells that facilitate tissue integrity, homeostasis, and repair. Here, we investigated the potential of the iPSC-derived MSC (iMSC) secretome in immunomodulation and vasculature repair in response to radiation injury utilizing human cell lines. METHODS We generated iPSC-derived iMSC lines and evaluated the potential of their conditioned media (iMSC CM) to treat IR-induced injuries in human monocytes (THP1) and brain vascular endothelial cells (hCMEC/D3). We further assessed factors in the iMSC secretome, their modulation, and the molecular pathways they elicit. RESULTS Increasing doses of IR disturbed endothelial tube and spheroid formation in hCMEC/D3. When IR-injured hCMEC/D3 (IR ≤ 5 Gy) were treated with iMSC CM, endothelial cell viability, adherence, spheroid compactness, and proangiogenic sprout formation were significantly ameliorated, and IR-induced ROS levels were reduced. iMSC CM augmented tube formation in cocultures of hCMEC/D3 and iMSCs. Consistently, iMSC CM facilitated angiogenesis in a zebrafish model in vivo. Furthermore, iMSC CM suppressed IR-induced NFκB activation, TNF-α release, and ROS production in THP1 cells. Additionally, iMSC CM diminished NF-kB activation in THP1 cells cocultured with irradiated hCMEC/D3, iMSCs, or HMC3 microglial lines. The cytokine array revealed that iMSC CM contains the proangiogenic and immunosuppressive factors MCP1/CCL2, IL6, IL8/CXCL8, ANG (Angiogenin), GROα/CXCL1, and RANTES/CCL5. Common promoter regulatory elements were enriched in TF-binding motifs such as androgen receptor (ANDR) and GATA2. hCMEC/D3 phosphokinome profiling revealed increased expression of pro-survival factors, the PI3K/AKT/mTOR modulator PRAS40 and β-catenin in response to CM. The transcriptome analysis revealed increased expression of GATA2 in iMSCs and the enrichment of pathways involved in RNA metabolism, translation, mitochondrial respiration, DNA damage repair, and neurodevelopment. CONCLUSIONS The iMSC secretome is a comodulated composite of proangiogenic and immunosuppressive factors that has the potential to alleviate radiation-induced vascular endothelial cell damage and immune activation.
Collapse
Affiliation(s)
- Kshama Gupta
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| | - Ralph B Perkerson
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Ramacharan Angom
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Prasanna Vibhute
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Abba C Zubair
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Department of Neurosurgery, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| |
Collapse
|
17
|
Wang Y, Tian J, Liu D, Li T, Mao Y, Zhu C. Microglia in radiation-induced brain injury: Cellular and molecular mechanisms and therapeutic potential. CNS Neurosci Ther 2024; 30:e14794. [PMID: 38867379 PMCID: PMC11168970 DOI: 10.1111/cns.14794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.
Collapse
Affiliation(s)
- Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Jiayu Tian
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Dandan Liu
- Department of Electrocardiogram, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Tao Li
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Yanna Mao
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Department of PediatricsInstitute of Neuroscience and Third Affiliated Hospital of Zhengzhou UniversityKangfuqian Street 7Zhengzhou450052None SelectedChina
- Center for Brain Repair and Rehabilitation, Department of Clinical NeuroscienceInstitute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgMedicinaregtan 11Göteborg40530Sweden
| |
Collapse
|
18
|
Bécam J, Ropars G, Dwiri FA, Brunaud C, Toutain J, Chazalviel L, Naveau M, Valable S, Bernaudin M, Touzani O, Pérès EA. Physical Activity Attenuates Brain Irradiation-Associated Skeletal Muscle Damage in the Rat. Int J Radiat Oncol Biol Phys 2024; 118:1081-1093. [PMID: 37866760 DOI: 10.1016/j.ijrobp.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/09/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
PURPOSE Radiation therapy for brain tumors increases patient survival. Nonetheless, side effects are increasingly reported such as cognitive deficits and fatigue. The etiology of fatigue remains poorly described. Our hypothesis is that the abscopal effects of radiation therapy on skeletal muscle may be involved in fatigue. The present study aims to assess the effect of brain irradiation on skeletal muscles and its relationship with fatigue and to analyze whether physical activity could counteract brain radiation-induced side effects. METHODS AND MATERIALS Adult Wistar rats were randomly distributed between 4 groups: control (CTL), irradiated (IR), nonirradiated with physical activity (PA), and irradiated with physical activity (IR+PA). IR rats were exposed to a whole-brain irradiation (WBI) of 30 Gy (3 × 10 Gy). Rats subjected to PA underwent sessions of running on a treadmill, 3 times/week for 6 months. The effects of WBI on muscles were evaluated by complementary approaches: behavioral tests (fatigue, locomotion activity), magnetic resonance imaging, and histologic analyses. RESULTS IR rats displayed a significant fatigue and a reduced locomotor activity at short term compared with the CTL group, which were attenuated with PA at 6 months after WBI. The IR rat's gastrocnemius mass decreased compared with CTL rats, which was reversed by physical activity at 14 days after WBI. Multiparametric magnetic resonance imaging of the skeletal muscle highlighted an alteration of the fiber organization in IR rats as demonstrated by a significant decrease of the mean diffusivity in the gastrocnemius at short term. Alteration of fibers was confirmed by histologic analyses: the number of type I fibers was decreased, whereas that of type IIa fibers was increased in IR animals but not in the IR+PA group. CONCLUSIONS The data show that WBI induces skeletal muscle damage, which is attenuated by PA. This muscle damage may explain, at least in part, the fatigue of patients treated with radiation therapy.
Collapse
Affiliation(s)
- Julie Bécam
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Gwenn Ropars
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Fatima-Azzahra Dwiri
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Carole Brunaud
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Jérôme Toutain
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Laurent Chazalviel
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Mikaël Naveau
- Université de Caen Normandie, CNRS, INSERM, CEA, Normandie Université, UAR3408/US50, Cyceron, GIP Cyceron, F-14000 Caen, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Myriam Bernaudin
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Omar Touzani
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France
| | - Elodie Anne Pérès
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, F-14000 Caen, France.
| |
Collapse
|
19
|
Shahriari A, Etemadrezaie H, Zabihyan S, Amirabadi A, Aalami AH. Alterations in hypothalamic-pituitary axis (HPA) hormones 6 months after cranial radiotherapy in adult patients with primary brain tumors outside the HPA region. Mol Biol Rep 2024; 51:373. [PMID: 38418676 DOI: 10.1007/s11033-024-09257-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Cranial radiotherapy is a common treatment for brain tumors, but it can affect the hypothalamic-pituitary (H-P) axis and lead to hormonal disorders. This study aimed to compare serum levels of HPA hormones before and after cranial radiation. MATERIALS AND METHODS This study involved 27 adult patients who underwent brain tumor resection before the initiation of radiotherapy, and none had metastatic brain tumors. All participants had the HPA within the radiation field, and their tumors were located in brain areas outside from the HPA. Serum levels of HPA hormones were recorded both before and 6 months after cranial radiotherapy. RESULTS A total of 27 adult patients, comprising 16 (59.3%) males and 11 (40.7%) females, with a mean age of 56.37 ± 11.38 years, were subjected to evaluation. Six months post-radiotherapy, serum levels of GH and TSH exhibited a significant decrease. Prior to radiotherapy, a substantial and direct correlation was observed between TSH and FSH (p = 0.005) as well as LH (p = 0.014). Additionally, a significant and direct relationship was noted between serum FSH and LH (p < 0.001) before radiotherapy. After radiotherapy, a significant and direct correlation persisted between TSH and FSH (p = 0.003) as well as LH (p = 0.005), along with a significant and direct relationship between serum FSH and LH (p < 0.001). Furthermore, a significant and direct association was identified between changes in serum GH levels and FSH (p = 0.04), as well as between serum LH and FSH (p < 0.001). CONCLUSION Reduced serum levels of HPA hormones are a significant complication of cranial radiotherapy and should be evaluated in follow-up assessments.
Collapse
Affiliation(s)
- Ali Shahriari
- Department of Internal Medicine, Faculty of Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Hamid Etemadrezaie
- Department of Neurosurgery, Ghaem Teaching Hospital, Mashhad University of Medical Sciences, Mashhad, Razavi Khorasan, Iran.
| | - Samira Zabihyan
- Department of Neurosurgery, Ghaem Teaching Hospital, Mashhad University of Medical Sciences, Mashhad, Razavi Khorasan, Iran
| | - Amir Amirabadi
- Department of Internal Medicine, Faculty of Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
- Innovative Medical Research Center, Faculty of Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amir Hossein Aalami
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA.
- Division of Nephrology and Hypertension, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA.
| |
Collapse
|
20
|
Asana Marican HT, Shen H. Dynamics of Chromosome Aberrations and Cell Death in Zebrafish Embryos Exposed to 137Cs Total-Body Irradiation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:2204-2213. [PMID: 38269402 DOI: 10.1021/acs.est.3c05389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Ionizing radiation exposure induces significant DNA damage and cell death in aquatic species. Accurate sensing and quantification play pivotal roles in environmental monitoring and surveillance. Zebrafish (Danio rerio) is a well-suited animal model for research into this aspect, especially with recent development of cytogenetic and transgenic tools. In this study, we present time-course studies of chromosome aberrations and cell death in zebrafish embryos exposed to 2 Gy 137Cs total-body irradiation. Using a cytogenetic approach, we quantified chromosome and chromatid aberrations in irradiated embryos at 6, 14, 20, and 24 h postirradiation. Metaphases with aberrations showed rapid declining kinetics, accompanied by incomplete karyotypes and irregular chromatin contents. Using an apoptosis-reporting transgenic zebrafish, we found increasing cell death along these time points, with the embryonic eyes and brain contributing the majority of the cell death volumes. We provide evidence that self-proliferating progenitor cells form the underlying linkage between the two kinetics and their positions define radiosensitive niches in zebrafish embryos. Our results provide detailed chromosome aberration and cell death dynamics in 137Cs-irradiated zebrafish embryos and unveil the appropriate timeline and tissue positions for accurate sensing and quantification of radiation-induced damages in zebrafish embryos.
Collapse
Affiliation(s)
- Halida Thanveer Asana Marican
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 Create Way, Singapore 138602, Singapore
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 Create Way, Singapore 138602, Singapore
| |
Collapse
|
21
|
Buczek D, Zaucha R, Jassem J. Neurotoxicity-sparing radiotherapy for brain metastases in breast cancer: a narrative review. Front Oncol 2024; 13:1215426. [PMID: 38370347 PMCID: PMC10869626 DOI: 10.3389/fonc.2023.1215426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 12/19/2023] [Indexed: 02/20/2024] Open
Abstract
Breast cancer brain metastasis (BCBM) has a devastating impact on patient survival, cognitive function and quality of life. Radiotherapy remains the standard management of BM but may result in considerable neurotoxicity. Herein, we describe the current knowledge on methods for reducing radiation-induced cognitive dysfunction in patients with BCBM. A better understanding of the biology and molecular underpinnings of BCBM, as well as more sophisticated prognostic models and individualized treatment approaches, have appeared to enable more effective neuroprotection. The therapeutic armamentarium has expanded from surgery and whole-brain radiotherapy to stereotactic radiosurgery, targeted therapies and immunotherapies, used sequentially or in combination. Advances in neuroimaging have allowed more accurate screening for intracranial metastases, precise targeting of intracranial lesions and the differentiation of the effects of treatment from disease progression. The availability of numerous treatment options for patients with BCBM and multidisciplinary approaches have led to personalized treatment and improved therapeutic outcomes. Ongoing studies may define the optimal sequencing of available and emerging treatment options for patients with BCBM.
Collapse
|
22
|
Wang X, Guo L, Qin T, Lai P, Jing Y, Zhang Z, Zhou G, Gao P, Ding G. Effects of X-ray cranial irradiation on metabolomics and intestinal flora in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115898. [PMID: 38171101 DOI: 10.1016/j.ecoenv.2023.115898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
Cranial radiotherapy is an important treatment for intracranial and head and neck tumors. To investigate the effects of cranial irradiation (C-irradiation) on gut microbiota and metabolomic profile, the feces, plasma and cerebral cortex were isolated after exposing mice to cranial X-ray irradiation at a dose rate of 2.33 Gy/min (5 Gy/d for 4 d consecutively). The gut microorganisms and metabolites were detected by 16 S rRNA gene sequencing method and LC-MS method, respectively. We found that compared with sham group, the gut microbiota composition changed at 2 W and 4 W after C-irradiation at the genus level. The fecal metabolomics showed that compared with Sham group, 44 and 66 differential metabolites were found to be annotated into metabolism pathways at 2 W and 4 W after C-irradiation, which were significantly enriched in the arginine and proline metabolism. Metabolome analysis of serum and cerebral cortex showed that, at 4 W after C-irradiation, the expression pattern of metabolites in serum samples of mice was similar to that of sham group, and the cerebral cortex metabolites of the two groups were completely separated. KEGG functional analysis showed that serum and brain tissue differential metabolites were respectively enriched in tryptophan metabolism, and arginine proline metabolism. The correlation analysis showed that the changes of gut microbiota genera were significantly correlated with the changes of metabolism, especially Helicobacter, which was significantly correlated with many different metabolites at 4 W after C-irradiation. These data suggested that C-irradiation could affect the gut microbiota and metabolism profile, even at relatively long times after C-irradiation.
Collapse
Affiliation(s)
- Xing Wang
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Ling Guo
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Tongzhou Qin
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Panpan Lai
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Yuntao Jing
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Zhaowen Zhang
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| | - Guiqiang Zhou
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China; Department of Labor and Environmental Hygiene, School of public health, Weifang Medical University, Weifang, China.
| | - Peng Gao
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China
| | - Guirong Ding
- Department of Radiation Protection Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China; Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Xi'an, China.
| |
Collapse
|
23
|
Wu B, Li S, Wang J, Wang J, Qiu W, Gao H. Bibliometric and visualization analysis of radiation brain injury from 2003 to 2023. Front Neurol 2024; 14:1275836. [PMID: 38298563 PMCID: PMC10828967 DOI: 10.3389/fneur.2023.1275836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/15/2023] [Indexed: 02/02/2024] Open
Abstract
Background Over the past two decades, the field of radiation brain injury has attracted the attention of an increasing number of brain scientists, particularly in the areas of molecular pathology and therapeutic approaches. Characterizing global collaboration networks and mapping development trends over the past 20 years is essential. Objective The aim of this paper is to examine significant issues and future directions while shedding light on collaboration and research status in the field of radiation brain injury. Methods Bibliometric studies were performed using CiteSpaceR-bibliometrix and VOSviewer software on papers regarding radiation brain injury that were published before November 2023 in the Web of Science Core Collection. Results In the final analysis, we found 4,913 records written in 1,219 publications by 21,529 authors from 5,007 institutions in 75 countries. There was a noticeable increase in publications in 2014 and 2021. The majority of records listed were produced by China, the United States, and other high-income countries. The largest nodes in each cluster of the collaboration network were Sun Yat-sen University, University of California-San Francisco, and the University of Toronto. Galldiks N, Barnett GH, Langen KJ and Kim JH are known to be core authors in the field. The top 3 keywords in that time frame are radiation, radiation necrosis, and radiation-therapy. Conclusions The objective and thorough bibliometric analysis also identifies current research hotspots and potential future paths, providing a retrospective perspective on RBI and offering useful advice to researchers choosing research topics. Future development directions include the integration of multi-omics methodologies and novel imaging techniques to improve RBI's diagnostic effectiveness and the search for new therapeutic targets.
Collapse
Affiliation(s)
- Baofang Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Shaojie Li
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Jian Wang
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiayin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Weizhi Qiu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| |
Collapse
|
24
|
Lojek NM, Williams VA, Rogers AM, Sajo E, Black BJ, Ghezzi CE. A 3D In Vitro Cortical Tissue Model Based on Dense Collagen to Study the Effects of Gamma Radiation on Neuronal Function. Adv Healthc Mater 2024; 13:e2301123. [PMID: 37921265 PMCID: PMC11468710 DOI: 10.1002/adhm.202301123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/14/2023] [Indexed: 11/04/2023]
Abstract
Studies on gamma radiation-induced injury have long been focused on hematopoietic, gastrointestinal, and cardiovascular systems, yet little is known about the effects of gamma radiation on the function of human cortical tissue. The challenge in studying radiation-induced cortical injury is, in part, due to a lack of human tissue models and physiologically relevant readouts. Here, a physiologically relevant 3D collagen-based cortical tissue model (CTM) is developed for studying the functional response of human iPSC-derived neurons and astrocytes to a sub-lethal radiation exposure (5 Gy). Cytotoxicity, DNA damage, morphology, and extracellular electrophysiology are quantified. It is reported that 5 Gy exposure significantly increases cytotoxicity, DNA damage, and astrocyte reactivity while significantly decreasing neurite length and neuronal network activity. Additionally, it is found that clinically deployed radioprotectant amifostine ameliorates the DNA damage, cytotoxicity, and astrocyte reactivity. The CTM provides a critical experimental platform to understand cell-level mechanisms by which gamma radiation (GR) affects human cortical tissue and to screen prospective radioprotectant compounds.
Collapse
Affiliation(s)
- Neal M. Lojek
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Victoria A. Williams
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Andrew M. Rogers
- Department of Physics and Applied PhysicsUniversity of Massachusetts LowellLowellMA01854USA
| | - Erno Sajo
- Department of Physics and Applied PhysicsUniversity of Massachusetts LowellLowellMA01854USA
| | - Bryan J. Black
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Chiara E. Ghezzi
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| |
Collapse
|
25
|
Kiang JG, Cannon G, Olson MG, Zhai M, Woods AK, Xu F, Lin B, Li X, Hull L, Jiang S, Xiao M. Ciprofloxacin and pegylated G-CSF combined therapy mitigates brain hemorrhage and mortality induced by ionizing irradiation. Front Public Health 2023; 11:1268325. [PMID: 38162617 PMCID: PMC10756649 DOI: 10.3389/fpubh.2023.1268325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Brain hemorrhage was found between 13 and 16 days after acute whole-body 9.5 Gy 60Co-γ irradiation (IR). This study tested countermeasures mitigating brain hemorrhage and increasing survival from IR. Previously, we found that pegylated G-CSF therapy (PEG) (i.e., Neulasta®, an FDA-approved drug) improved survival post-IR by 20-40%. This study investigated whether Ciprofloxacin (CIP) could enhance PEG-induced survival and whether IR-induced brain hemorrhage could be mitigated by PEG alone or combined with CIP. Methods B6D2F1 female mice were exposed to 60Co-γ-radiation. CIP was fed to mice for 21 days. PEG was injected on days 1, 8, and 15. 30-day survival and weight loss were studied in mice treated with vehicles, CIP, PEG, or PEG + CIP. For the early time point study, blood and sternums on days 2, 4, 9, and 15 and brains on day 15 post-IR were collected. Platelet numbers, brain hemorrhage, and histopathology were analyzed. The cerebellum/pons/medulla oblongata were detected with glial fibrillary acidic protein (GFAP), p53, p16, interleukin-18 (IL-18), ICAM1, Claudin 2, ZO-1, and complement protein 3 (C3). Results CIP + PEG enhanced survival after IR by 85% vs. the 30% improvement by PEG alone. IR depleted platelets, which was mitigated by PEG or CIP + PEG. Brain hemorrhage, both surface and intracranial, was observed, whereas the sham mice displayed no hemorrhage. CIP or CIP + PEG significantly mitigated brain hemorrhage. IR reduced GFAP levels that were recovered by CIP or CIP + PEG, but not by PEG alone. IR increased IL-18 levels on day 4 only, which was inhibited by CIP alone, PEG alone, or PEG + CIP. IR increased C3 on day 4 and day 15 and that coincided with the occurrence of brain hemorrhage on day 15. IR increased phosphorylated p53 and p53 levels, which was mitigated by CIP, PEG or PEG + CIP. P16, Claudin 2, and ZO-1 were not altered; ICAM1 was increased. Discussion CIP + PEG enhanced survival post-IR more than PEG alone. The Concurrence of brain hemorrhage, C3 increases and p53 activation post-IR suggests their involvement in the IR-induced brain impairment. CIP + PEG effectively mitigated the brain lesions, suggesting effectiveness of CIP + PEG therapy for treating the IR-induced brain hemorrhage by recovering GFAP and platelets and reducing C3 and p53.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgetta Cannon
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Matthew G. Olson
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Min Zhai
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Akeylah K. Woods
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Feng Xu
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Bin Lin
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Xianghong Li
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Lisa Hull
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Suping Jiang
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Mang Xiao
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| |
Collapse
|
26
|
Makranz C, Lubotzky A, Zemmour H, Shemer R, Glaser B, Cohen J, Maoz M, Sapir E, Wygoda M, Peretz T, Weizman N, Feldman J, Abrams RA, Lossos A, Dor Y, Zick A. Short report: Plasma based biomarkers detect radiation induced brain injury in cancer patients treated for brain metastasis: A pilot study. PLoS One 2023; 18:e0285646. [PMID: 38015964 PMCID: PMC10684068 DOI: 10.1371/journal.pone.0285646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/15/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Radiotherapy has an important role in the treatment of brain metastases but carries risk of short and/or long-term toxicity, termed radiation-induced brain injury (RBI). As the diagnosis of RBI is crucial for correct patient management, there is an unmet need for reliable biomarkers for RBI. The aim of this proof-of concept study is to determine the utility of brain-derived circulating free DNA (BncfDNA), identified by specific methylation patterns for neurons, astrocytes, and oligodendrocytes, as biomarkers brain injury induced by radiotherapy. METHODS Twenty-four patients with brain metastases were monitored clinically and radiologically before, during and after brain radiotherapy, and blood for BncfDNA analysis (98 samples) was concurrently collected. Sixteen patients were treated with whole brain radiotherapy and eight patients with stereotactic radiosurgery. RESULTS During follow-up nine RBI events were detected, and all correlated with significant increase in BncfDNA levels compared to baseline. Additionally, resolution of RBI correlated with a decrease in BncfDNA. Changes in BncfDNA were independent of tumor response. CONCLUSIONS Elevated BncfDNA levels reflects brain cell injury incurred by radiotherapy. further research is needed to establish BncfDNA as a novel plasma-based biomarker for brain injury induced by radiotherapy.
Collapse
Affiliation(s)
- Chen Makranz
- Department of Neurology and Oncology, The Gaffin Center for Neurooncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Asael Lubotzky
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Division of Neurology and Department of Molecular Genetics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Hai Zemmour
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Jonathan Cohen
- Department of Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Myriam Maoz
- Department of Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eli Sapir
- Department of Radiation Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Radiation Oncology Institute, Samson Assuta Ashdod University Hospital, Ben Gurion University, Ashdod, Israel
| | - Marc Wygoda
- Department of Radiation Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamar Peretz
- Department of Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Noam Weizman
- Department of Radiation Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jon Feldman
- Department of Radiation Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ross A. Abrams
- Department of Radiation Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander Lossos
- Department of Neurology and Oncology, The Gaffin Center for Neurooncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Aviad Zick
- Department of Oncology, Sharett Institute for Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
27
|
Sun LWH, Asana Marican HT, Beh LK, Shen H. Imaging the radioprotective effect of amifostine in the developing brain using an apoptosis-reporting transgenic zebrafish. Int J Radiat Biol 2023; 100:433-444. [PMID: 37922446 DOI: 10.1080/09553002.2023.2280011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE Normal tissue radioprotectants alleviate radiation-induced damages and preserve critical organ functions. Investigating their efficacy in vivo remains challenging, especially in enclosed organs like the brain. An animal model that enables direct visualization of radiation-induced apoptosis while possessing the structural complexity of a vertebrate brain facilitates these studies in a precise and effective manner. MATERIALS AND METHODS We employed a secA5 transgenic zebrafish expressing secreted Annexin V fused with a yellow fluorescent protein to visualize radiation-induced apoptosis in vivo. We developed a semi-automated imaging method for standardized acquisition of apoptosis signals in batches of zebrafish larvae. Using these approaches, we studied the protective effect of amifostine (WR-2721) in the irradiated zebrafish larval brain. RESULTS Upon 2 Gy total-body 137Cs irradiation, increased apoptosis could be visualized at high resolution in the secA5 brain at 2, 24, and 48 hour post irradiation (hpi). Amifostine treatment (4 mM) during irradiation reduced apoptosis significantly at 24 hpi and preserved Wnt active cells in the larval brain. When the 2 Gy irradiation was delivered in combination with cisplatin treatment (0.1 mM), the radioprotective effect of amifostine was also observed. CONCLUSIONS Our study reveals the radioprotective effect of amifostine in the developing zebrafish larval brain, and highlights the utility of secA5 transgenic zebrafish as a novel system for investigating normal tissue radioprotectants in vivo.
Collapse
Affiliation(s)
- Lucas W H Sun
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | | | - Lih Khiang Beh
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
28
|
Yang T, Du X, Xu L. Radioprotective effect of Ginkgolide B on brain: the mediating role of DCC/MST1 signaling. Int J Radiat Biol 2023; 100:371-384. [PMID: 37934907 DOI: 10.1080/09553002.2023.2281515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/24/2023] [Indexed: 11/09/2023]
Abstract
PURPOSE The risk of brain exposure to ionizing radiation increases gradually due to the extensive application of nuclear technology in medical, industrial, and aerospace fields. Radiation-induced brain injury (RBI) is highly likely to cause a wide range of neurological complications, including schizophrenia, Alzheimer's disease (AD), depression. Ginkgolide B (GB) is one of the effective active components extracted from ginkgo biloba leaves, exerts protective effects on CNS, which is involved in the regulation of the Hippo signaling pathway. MST1, as one of the core kinases of the Hippo pathway, participated in regulating cell proliferation, differentiation, and apoptosis. However, it remains unclear whether GB attenuates radiation brain injury (RBI) and whether the radioprotective effect of GB refers to MST1 signaling. Hence, our study aimed to explore the radiation protection effect and the potential mechanism of GB. MATERIALS AND METHODS C57BL/6 mice were stimulated with an X-ray (20 Gy) to establish an RBI model. Then, morris water maze test (MWM) and step-down passive avoidance test (SDPAT) were used to assess the learning and memory function of mice. The open field test (OFT), tail suspension test (TST), and forced swimming test (FST) were used to assess changes in locomotor activity and hopelessness. Besides, X-ray-stimulated SH-SY5Y cells were used to verify the radioprotective effect of GB. Immunofluorescence double staining, Dihydroethidium (DHE), western blot, and flow cytometry were used to explore the role of DCC/MST1 signaling in RBI. RESULTS In this study, X-ray-treated mice exhibited cognitive impairment and depression-like behavior, which was ameliorated by GB treatment. GB also reduced the ROS production and the number of TUNEL-positive cells in the hippocampus. Moreover, GB increased the protein levels of p-AKT and Bcl2, while decreased the protein levels of MST1, p-p38, p-JNK, cleaved-caspase-3 and Bax both in vivo and in vitro. Additionally, exogenous Netrin-1 alleviated X-ray-induced ROS production and apoptosis, whereas knockout of Netrin-1 receptor DCC abolished the protective effect of GB. CONCLUSION Oxidative stress and MST1-mediated neuronal apoptosis participated in radiation-induced cognitive impairment and depression-like behaviors, and modulation of DCC by GB was an effective intervention against RBI.
Collapse
Affiliation(s)
- Tao Yang
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, Jiangsu, China
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiao Du
- Division of Clinical Pharmacy, Department of Pharmacy, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Lixing Xu
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
29
|
Choi DH, Oh D, Na K, Kim H, Choi D, Jung YH, Ahn J, Kim J, Kim CH, Chung S. Radiation induces acute and subacute vascular regression in a three-dimensional microvasculature model. Front Oncol 2023; 13:1252014. [PMID: 37909014 PMCID: PMC10613678 DOI: 10.3389/fonc.2023.1252014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/28/2023] [Indexed: 11/02/2023] Open
Abstract
Radiation treatment is one of the most frequently used therapies in patients with cancer, employed in approximately half of all patients. However, the use of radiation therapy is limited by acute or chronic adverse effects and the failure to consider the tumor microenvironment. Blood vessels substantially contribute to radiation responses in both normal and tumor tissues. The present study employed a three-dimensional (3D) microvasculature-on-a-chip that mimics physiological blood vessels to determine the effect of radiation on blood vessels. This model represents radiation-induced pathophysiological effects on blood vessels in terms of cellular damage and structural and functional changes. DNA double-strand breaks (DSBs), apoptosis, and cell viability indicate cellular damage. Radiation-induced damage leads to a reduction in vascular structures, such as vascular area, branch length, branch number, junction number, and branch diameter; this phenomenon occurs in the mature vascular network and during neovascularization. Additionally, vasculature regression was demonstrated by staining the basement membrane and microfilaments. Radiation exposure could increase the blockage and permeability of the vascular network, indicating that radiation alters the function of blood vessels. Radiation suppressed blood vessel recovery and induced a loss of angiogenic ability, resulting in a network of irradiated vessels that failed to recover, deteriorating gradually. These findings demonstrate that this model is valuable for assessing radiation-induced vascular dysfunction and acute and chronic effects and can potentially improve radiotherapy efficiency.
Collapse
Affiliation(s)
- Dong-Hee Choi
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Dongwoo Oh
- Korea University-Korea institute of Science and Technology (KU-KIST) Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Kyuhwan Na
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Hyunho Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Dongjin Choi
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Yong Hun Jung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Jinchul Ahn
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Jaehoon Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Chun-Ho Kim
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- Korea University-Korea institute of Science and Technology (KU-KIST) Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
30
|
Xiao M, Li X, Wang L, Lin B, Zhai M, Hull L, Zizzo A, Cui W, Kiang JG. Skin Wound following Irradiation Aggravates Radiation-Induced Brain Injury in a Mouse Model. Int J Mol Sci 2023; 24:10701. [PMID: 37445879 DOI: 10.3390/ijms241310701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radiation injury- and radiation combined with skin injury-induced inflammatory responses in the mouse brain were evaluated in this study. Female B6D2F1/J mice were subjected to a sham, a skin wound (SW), 9.5 Gy 60Co total-body gamma irradiation (RI), or 9.5 Gy RI combined with a skin puncture wound (RCI). Survival, body weight, and wound healing were tracked for 30 days, and mouse brain samples were collected on day 30 after SW, RI, RCI, and the sham control. Our results showed that RCI caused more severe animal death and body weight loss compared with RI, and skin wound healing was significantly delayed by RCI compared to SW. RCI and RI increased the chemokines Eotaxin, IP-10, MIG, 6Ckine/Exodus2, MCP-5, and TIMP-1 in the brain compared to SW and the sham control mice, and the Western blot results showed that IP-10 and p21 were significantly upregulated in brain cells post-RI or -RCI. RI and RCI activated both astrocytes and endothelial cells in the mouse brain, subsequently inducing blood-brain barrier (BBB) leakage, as shown by the increased ICAM1 and GFAP proteins in the brain and GFAP in the serum. The Doublecortin (DCX) protein, the "gold standard" for measuring neurogenesis, was significantly downregulated by RI and RCI compared with the sham group. Furthermore, RI and RCI decreased the expression of the neural stem cell marker E-cadherin, the intermediate progenitor marker MASH1, the immature neuron cell marker NeuroD1, and the mature neuron cell marker NeuN, indicating neural cell damage in all development stages after RI and RCI. Immunohistochemistry (IHC) staining further confirmed the significant loss of neural cells in RCI. Our data demonstrated that RI and RCI induced brain injury through inflammatory pathways, and RCI exacerbated neural cell damage more than RI.
Collapse
Affiliation(s)
- Mang Xiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Xianghong Li
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Li Wang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Bin Lin
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Min Zhai
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Lisa Hull
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Alex Zizzo
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Wanchang Cui
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
31
|
Rübe CE, Raid S, Palm J, Rübe C. Radiation-Induced Brain Injury: Age Dependency of Neurocognitive Dysfunction Following Radiotherapy. Cancers (Basel) 2023; 15:cancers15112999. [PMID: 37296960 DOI: 10.3390/cancers15112999] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Cranial radiotherapy is a known risk factor for neurocognitive impairment in cancer survivors. Although radiation-induced cognitive dysfunction is observed in patients of all ages, children seem to be more vulnerable than adults to suffering age-related deficits in neurocognitive skills. So far, the underlying mechanisms by which IR negatively influences brain functions as well as the reasons for the profound age dependency are still insufficiently known. We performed a comprehensive Pubmed-based literature search to identify original research articles that reported on age dependency of neurocognitive dysfunction following cranial IR exposure. Numerous clinical trials in childhood cancer survivors indicate that the severity of radiation-induced cognitive dysfunction is clearly dependent on age at IR exposure. These clinical findings were related to the current state of experimental research providing important insights into the age dependency of radiation-induced brain injury and the development of neurocognitive impairment. Research in pre-clinical rodent models demonstrates age-dependent effects of IR exposure on hippocampal neurogenesis, radiation-induced neurovascular damage and neuroinflammation.
Collapse
Affiliation(s)
- Claudia E Rübe
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Silvia Raid
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Jan Palm
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Christian Rübe
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| |
Collapse
|
32
|
Wu Q, Fang Y, Huang X, Zheng F, Ma S, Zhang X, Han T, Gao H, Shen B. Role of Orai3-Mediated Store-Operated Calcium Entry in Radiation-Induced Brain Microvascular Endothelial Cell Injury. Int J Mol Sci 2023; 24:ijms24076818. [PMID: 37047790 PMCID: PMC10095176 DOI: 10.3390/ijms24076818] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 04/14/2023] Open
Abstract
Radiation-induced brain injury is a serious complication with complex pathogenesis that may accompany radiotherapy of head and neck tumors. Although studies have shown that calcium (Ca2+) signaling may be involved in the occurrence and development of radiation-induced brain injury, the underlying molecular mechanisms are not well understood. In this study, we used real-time quantitative polymerase chain reaction and Western blotting assays to verify our previous finding using next-generation sequencing that the mRNA and protein expression levels of Orai3 in rat brain microvascular endothelial cells (rBMECs) increased after X-ray irradiation. We next explored the role of Orai3 and Orai3-mediated store-operated Ca2+ entry (SOCE) in radiation-induced brain injury. Primary cultured rBMECs derived from wild-type and Orai3 knockout (Orai3(-/-)) Sprague-Dawley rats were used for in vitro experiments. Orai3-mediated SOCE was significantly increased in rBMECs after X-ray irradiation. However, X-ray irradiation-induced SOCE increase was markedly reduced in Orai3 knockout rBMECs, and the percentage of BTP2 (a nonselective inhibitor of Orai channels)-inhibited SOCE was significantly decreased in Orai3 knockout rBMECs. Functional studies indicated that X-ray irradiation decreased rBMEC proliferation, migration, and tube formation (a model for assessing angiogenesis) but increased rBMEC apoptosis, all of which were ameliorated by BTP2. In addition, occurrences of all four functional deficits were suppressed in X-ray irradiation-exposed rBMECs derived from Orai3(-/-) rats. Cerebrovascular damage caused by whole-brain X-ray irradiation was much less in Orai3(-/-) rats than in wild-type rats. These findings provide evidence that Orai3-mediated SOCE plays an important role in radiation-induced rBMEC damage and brain injury and suggest that Orai3 may warrant development as a potential therapeutic target for reducing or preventing radiation-induced brain injury.
Collapse
Affiliation(s)
- Qibing Wu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yang Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiaoyu Huang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Fan Zheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Shaobo Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinchen Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Tingting Han
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Huiwen Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
33
|
Sun LWH, Asana Marican HT, Shen H. In Vivo Imaging of Radiation-Induced Apoptosis at Single-Cell Resolution in Transgenic Zebrafish Embryos. Radiat Res 2023; 199:229-239. [PMID: 36745564 DOI: 10.1667/rade-22-00174.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
Among the various types of cell death induced by ionizing radiation, apoptosis is a highly regulated and well-characterized form. Investigating radiation-induced apoptosis in an intact organism offers advantages in capturing the dynamics of apoptosis under preserved physiology, although high resolution imaging remains challenging. Owing to their optical transparency and genetic amenability, zebrafish is an ideal animal model for research into this aspect. In this study, we present a secA5 transgenic zebrafish expressing genetically encoded secreted ANNEXIN V fused with mVenus, a yellow fluorescent protein that enables reporting of radiation-induced apoptosis. Using in vivo imaging approach, we show that after 2 Gy total-body irradiation, apoptosis could be visualized at single-cell resolution in different cell types throughout the embryo. Elevated apoptosis could be imaged and quantified in the neuroepithelium of the embryonic brain, as well as the proliferative zone and parenchyma of the larval brain. In addition, clearance of apoptotic cells by microglia, the professional phagocytes residing in the brain, could be imaged at single-cell resolution in irradiated larvae. These results establish transgenic secA5 zebrafish as a useful and versatile in vivo system for investigating the dynamic process of radiation-induced apoptosis.
Collapse
Affiliation(s)
| | | | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
34
|
Muresanu DF, Sharma A, Tian ZR, Lafuente JV, Nozari A, Feng L, Buzoianu AD, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin with Mesenchymal Stem Cells Attenuates Heat Stress-Induced Exacerbation of Neuropathology Following Brain Blast Injury. ADVANCES IN NEUROBIOLOGY 2023; 32:231-270. [PMID: 37480463 DOI: 10.1007/978-3-031-32997-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Blast brain injury (bBI) following explosive detonations in warfare is one of the prominent causes of multidimensional insults to the central nervous and other vital organs injury. Several military personnel suffered from bBI during the Middle East conflict at hot environment. The bBI largely occurs due to pressure waves, generation of heat together with release of shrapnel and gun powders explosion with penetrating and/or impact head trauma causing multiple brain damage. As a result, bBI-induced secondary injury causes breakdown of the blood-brain barrier (BBB) and edema formation that further results in neuronal, glial and axonal injuries. Previously, we reported endocrine imbalance and influence of diabetes on bBI-induced brain pathology that was significantly attenuated by nanowired delivery of cerebrolysin in model experiments. Cerebrolysin is a balanced composition of several neurotrophic factors, and active peptide fragment is capable of neuroprotection in several neurological insults. Exposure to heat stress alone causes BBB damage, edema formation and brain pathology. Thus, it is quite likely that hot environment further exacerbates the consequences of bBI. Thus, novel therapeutic strategies using nanodelivery of stem cell and cerebrolysin may further enhance superior neuroprotection in bBI at hot environment. Our observations are the first to show that combined nanowired delivery of mesenchymal stem cells (MSCs) and cerebrolysin significantly attenuated exacerbation of bBI in hot environment and induced superior neuroprotection, not reported earlier. The possible mechanisms of neuroprotection with MSCs and cerebrolysin in bBI are discussed in the light of current literature.
Collapse
Affiliation(s)
- Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
35
|
Diehl CD, Rosenkranz E, Schwendner M, Mißlbeck M, Sollmann N, Ille S, Meyer B, Combs SE, Krieg SM. Dose Reduction to Motor Structures in Adjuvant Fractionated Stereotactic Radiotherapy of Brain Metastases: nTMS-Derived DTI-Based Motor Fiber Tracking in Treatment Planning. Cancers (Basel) 2022; 15:cancers15010282. [PMID: 36612277 PMCID: PMC9818359 DOI: 10.3390/cancers15010282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/07/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
Background: Resection of brain metastases (BM) close to motor structures is challenging for treatment. Navigated transcranial magnetic stimulation (nTMS) motor mapping, combined with diffusion tensor imaging (DTI)-based fiber tracking (DTI-FTmot.TMS), is a valuable tool in neurosurgery to preserve motor function. This study aimed to assess the practicability of DTI-FTmot.TMS for local adjuvant radiotherapy (RT) planning of BM. Methods: Presurgically generated DTI-FTmot.TMS-based corticospinal tract (CST) reconstructions (FTmot.TMS) of 24 patients with 25 BM resected during later surgery were incorporated into the RT planning system. Completed fractionated stereotactic intensity-modulated RT (IMRT) plans were retrospectively analyzed and adapted to preserve FTmot.TMS. Results: In regular plans, mean dose (Dmean) of complete FTmot.TMS was 5.2 ± 2.4 Gy. Regarding planning risk volume (PRV-FTTMS) portions outside of the planning target volume (PTV) within the 17.5 Gy (50%) isodose line, the DTI-FTmot.TMS Dmean was significantly reduced by 33.0% (range, 5.9−57.6%) from 23.4 ± 3.3 Gy to 15.9 ± 4.7 Gy (p < 0.001). There was no significant decline in the effective treatment dose, with PTV Dmean 35.6 ± 0.9 Gy vs. 36.0 ± 1.2 Gy (p = 0.063) after adaption. Conclusions: The DTI-FTmot.TMS-based CST reconstructions could be implemented in adjuvant IMRT planning of BM. A significant dose reduction regarding motor structures within critical dose levels seems possible.
Collapse
Affiliation(s)
- Christian D. Diehl
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), DKTK Partner Site, 81675 Munich, Germany
- Correspondence:
| | - Enrike Rosenkranz
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Maximilian Schwendner
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Martin Mißlbeck
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Nico Sollmann
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, 89081 Ulm, Germany
| | - Sebastian Ille
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), DKTK Partner Site, 81675 Munich, Germany
| | - Sandro M. Krieg
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| |
Collapse
|
36
|
Vitulli F, Spennato P, Cicala D, Mirone G, Scala MR, Cinalli G. Acute ischemic stroke secondary to ventriculoperitoneal shunt dysfunction in a child with Moyamoya syndrome. Surg Neurol Int 2022; 13:306. [PMID: 35928308 PMCID: PMC9345112 DOI: 10.25259/sni_434_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/29/2022] [Indexed: 11/04/2022] Open
Abstract
Background:
Patients with brain vascular disease and hydrocephalus may be predisposed to acute ischemic stroke in case of shunt dysfunction and subsequent increased intracranial pression. Patients with brain tumor may develop hydrocephalus as a consequence of obstruction of cerebrospinal fluid pathways and radiation-induced moyamoya syndrome secondary (RIMS) to radiotherapy (RT).
Case Description:
A 15-year-old male patient, affected by hydrocephalus and RIMS, presented acute cerebral ischemia after an episode of shunt malfunction. The shunt was promptly revised and the areas of ischemia visible at magnetic resonance imaging significantly decreased.
Conclusion:
Children who receive RT for brain tumor, particularly if the circle of Willis region is involved, require close surveillance for the development of vasculopathy and consequent stroke. This surveillance must be even tighter if the patient has been treated with ventricular shunt for the possible synergistic interaction between the two causes on reducing cerebral perfusion and increasing the risk of acute ischemic events.
Collapse
Affiliation(s)
- Francesca Vitulli
- Department of Neurosciences, Neurosurgery Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, Federico II University of Naples, Naples, Italy
| | - Pietro Spennato
- Department of Neurosciences, Neurosurgery Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
| | - Domenico Cicala
- Department of Neurosciences, Neuroradiology Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
| | - Giuseppe Mirone
- Department of Neurosciences, Neurosurgery Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
| | - Maria Rosaria Scala
- Department of Neurosciences, Neurosurgery Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, Federico II University of Naples, Naples, Italy
| | - Giuseppe Cinalli
- Department of Neurosciences, Neurosurgery Unit, AORN Santobono-Pausilipon Children’s Hospital, Naples, Italy
| |
Collapse
|
37
|
Iqubal A, Iqubal MK, Sharma S, Wasim M, Alfaleh MA, Md S, Baboota S, Ali J, Haque SE. Pathogenic mechanisms and therapeutic promise of phytochemicals and nanocarriers based drug delivery against radiotherapy-induced neurotoxic manifestations. Drug Deliv 2022; 29:1492-1511. [PMID: 35543534 PMCID: PMC9103628 DOI: 10.1080/10717544.2022.2064562] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Radiotherapy is one of the extensively used therapeutic modalities in glioblastoma and other types of cancers. Radiotherapy is either used as a first-line approach or combined with pharmacotherapy or surgery to manage and treat cancer. Although the use of radiotherapy significantly increased the survival time of patients, but its use has been reported with marked neuroinflammation and cognitive dysfunction that eventually reduced the quality of life of patients. Based on the preclinical and clinical investigations, the profound role of increased oxidative stress, nuclear translocation of NF-kB, production of proinflammatory cytokines such as TNF-α, IL-6, IL-β, increased level of MMPs, increased apoptosis, reduced angiogenesis, neurogenesis, and histological aberrations in CA1, CA2, CA3 and DG region of the hippocampus have been reported. Various pharmacotherapeutic drugs are being used as an adjuvant to counteract this neurotoxic manifestation. Still, most of these drugs suffer from systemic adverse effect, causes interference to ongoing chemotherapy, and exhibit pharmacokinetic limitations in crossing the blood-brain barrier. Therefore, various phytoconstituents, their nano carrier-based drug delivery systems and miRNAs have been explored to overcome the aforementioned limitations. The present review is focused on the mechanism and evidence of radiotherapy-induced neuroinflammation and cognitive dysfunction, pathological and molecular changes in the brain homeostasis, available adjuvants, their limitations. Additionally, the potential role and mechanism of neuroprotection of various nanocarrier based natural products and miRNAs have been discussed.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd, Gurugram, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohamed A Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
38
|
Diehl C, Rosenkranz E, Mißlbeck M, Schwendner M, Sollmann N, Ille S, Meyer B, Combs S, Bernhardt D, Krieg S. nTMS-derived DTI-based motor fiber tracking in radiotherapy treatment planning of high-grade gliomas for avoidance of motor structures. Radiother Oncol 2022; 171:189-197. [DOI: 10.1016/j.radonc.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
|
39
|
Pospelova M, Krasnikova V, Fionik O, Alekseeva T, Samochernykh K, Ivanova N, Trofimov N, Vavilova T, Vasilieva E, Topuzova M, Chaykovskaya A, Makhanova A, Mikhalicheva A, Bukkieva T, Restor K, Combs S, Shevtsov M. Potential Molecular Biomarkers of Central Nervous System Damage in Breast Cancer Survivors. J Clin Med 2022; 11:jcm11051215. [PMID: 35268306 PMCID: PMC8911416 DOI: 10.3390/jcm11051215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Damage of the central nervous system (CNS), manifested by cognitive impairment, occurs in 80% of women with breast cancer (BC) as a complication of surgical treatment and radiochemotherapy. In this study, the levels of ICAM-1, PECAM-1, NSE, and anti-NR-2 antibodies which are associated with the damage of the CNS and the endothelium were measured in the blood by ELISA as potential biomarkers that might reflect pathogenetic mechanisms in these patients. A total of 102 patients enrolled in this single-center trial were divided into four groups: (1) 26 patients after breast cancer treatment, (2) 21 patients with chronic brain ischemia (CBI) and asymptomatic carotid stenosis (ICA stenosis) (CBI + ICA stenosis), (3) 35 patients with CBI but without asymptomatic carotid stenosis, and (4) 20 healthy female volunteers (control group). Intergroup analysis demonstrated that in the group of patients following BC treatment there was a significant increase of ICAM-1 (mean difference: −368.56, 95% CI −450.30 to −286.69, p < 0.001) and PECAM-1 (mean difference: −47.75, 95% CI −68.73 to −26.77, p < 0.001) molecules, as compared to the group of healthy volunteers. Additionally, a decrease of anti-NR-2 antibodies (mean difference: 0.89, 95% CI 0.41 to 1.48, p < 0.001) was detected. The intergroup comparison revealed comparable levels of ICAM-1 (mean difference: −33.58, 95% CI −58.10 to 125.26, p = 0.76), PECAM-1 (mean difference: −5.03, 95% CI −29.93 to 19.87, p = 0.95), as well as anti-NR-2 antibodies (mean difference: −0.05, 95% CI −0.26 to 0.16, p = 0.93) in patients after BC treatment and in patients with CBI + ICA stenosis. The NSE level in the group CBI + ICA stenosis was significantly higher than in women following BC treatment (mean difference: −43.64, 95% CI 3.31 to −83.99, p = 0.03). Comparable levels of ICAM-1 were also detected in patients after BC treatment and in the group of CBI (mean difference: −21.28, 95% CI −111.03 to 68.48, p = 0.92). The level of PECAM-1 molecules in patients after BC treatment was also comparable to group of CBI (mean difference: −13.68, 95% CI −35.51 to 8.15, p = 0.35). In conclusion, among other mechanisms, endothelial dysfunction might play a role in the damage of the CNS in breast cancer survivors.
Collapse
Affiliation(s)
- Maria Pospelova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Varvara Krasnikova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Olga Fionik
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Tatyana Alekseeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Nataliya Ivanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Nikita Trofimov
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Tatyana Vavilova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Elena Vasilieva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Maria Topuzova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Alexandra Chaykovskaya
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Albina Makhanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Anna Mikhalicheva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Tatyana Bukkieva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
| | - Kenneth Restor
- Nursing Programme, University of St. Francis, Joliet, IL 60435, USA;
| | - Stephanie Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technishe Universität München (TUM), 81675 Munich, Germany;
| | - Maxim Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (M.P.); (V.K.); (O.F.); (T.A.); (K.S.); (N.I.); (N.T.); (T.V.); (E.V.); (M.T.); (A.C.); (A.M.); (A.M.); (T.B.)
- Department of Radiation Oncology, Klinikum rechts der Isar, Technishe Universität München (TUM), 81675 Munich, Germany;
- National Center for Neurosurgery, Nur-Sultan 010000, Kazakhstan
- Correspondence: ; Tel.: +49-173-1488882
| |
Collapse
|