1
|
Li G, Li H, Weng N, Liu C, Li X, Li Q, Bin L, Zhu K, Huang D, Liu J, Liu Y, Wang X. Preclinical monitoring of radiation-induced brain injury via GluCEST MRI and resting-state fMRI at 7 T: an exploratory study on MRI-guided OAR avoidance. Strahlenther Onkol 2025; 201:411-419. [PMID: 39259349 DOI: 10.1007/s00066-024-02292-w] [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: 11/08/2023] [Accepted: 07/30/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE To assess the value of glutamate chemical exchange saturation transfer (GluCEST) after whole-brain radiotherapy (WBRT) as an imaging marker of radiation-induced brain injury (RBI) and to preliminarily show the feasibility of multiparametric MRI-guided organ at risk (OAR) avoidance. METHODS Rats were divided into two groups: the control (CTRL) group (n = 9) and the RBI group (n = 9). The rats in the RBI group were irradiated with an X‑ray radiator and then subjected to a water maze experiment 4 weeks later. In combination with high-performance liquid chromatography (HPLC), we evaluated the value of GluCEST applied to glutamate changes for RBI and investigated the effect of such changes on glutamatergic neuronal function. RESULTS The average GluCEST values were markedly lower in the hippocampus and cerebral cortex. Positive correlations were observed between GluCEST values and regional homogeneity (ReHo) values in both the hippocampus and the cerebral cortex. HPLC showed a positive correlation with GluCEST values in the hippocampus. GluCEST values were positively correlated with spatial memory. CONCLUSION GluCEST MRI provides a visual assessment of glutamate changes in RBI rats for monitoring OAR cognitive toxicity reactions and may be used as a biomarker of OAR avoidance as well as metabolism to facilitate monitoring and intervention in radiation damage that occurs after radiotherapy.
Collapse
Affiliation(s)
- Guodong Li
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Hao Li
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Na Weng
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Caiyun Liu
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Xianglin Li
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Qinglong Li
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Li Bin
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Kai Zhu
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Danqi Huang
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Jia Liu
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China
| | - Yan Liu
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China.
| | - Xu Wang
- Department of Nuclear Medicine, Binzhou Medical University Hospital, School of Medical Imaging, Binzhou Medical University, Shandong, China.
| |
Collapse
|
2
|
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
|
3
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
4
|
El-Khatib SM, Vagadia AR, Le ACD, Baulch JE, Ng DQ, Du M, Johnston KG, Tan Z, Xu X, Chan A, Acharya MM. BDNF augmentation reverses cranial radiation therapy-induced cognitive decline and neurodegenerative consequences. Acta Neuropathol Commun 2024; 12:190. [PMID: 39696694 DOI: 10.1186/s40478-024-01906-9] [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: 09/01/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Cranial radiation therapy (RT) for brain cancers is often associated with the development of radiation-induced cognitive dysfunction (RICD). RICD significantly impacts the quality of life for cancer survivors, highlighting an unmet medical need. Previous human studies revealed a marked reduction in plasma brain-derived neurotrophic factor (BDNF) post-chronic chemotherapy, linking this decline to a substantial cognitive dysfunction among cancer survivors. Moreover, riluzole (RZ)-mediated increased BDNF in vivo in the chemotherapy-exposed mice reversed cognitive decline. RZ is an FDA-approved medication for ALS known to increase BDNF in vivo. In an effort to mitigate the detrimental effects of RT-induced BDNF decline in RICD, we tested the efficacy of RZ in a cranially irradiated (9 Gy) adult mouse model. Notably, RT-exposed mice exhibited significantly reduced hippocampal BDNF, accompanied by increased neuroinflammation, loss of neuronal plasticity-related immediate early gene product, cFos, and synaptic density. Spatial transcriptomic profiling comparing the RT + Vehicle with the RT + RZ group showed gene expression signatures of neuroprotection of hippocampal excitatory neurons post-RZ. RT-exposed mice performed poorly on learning and memory, and memory consolidation tasks. However, irradiated mice receiving RZ (13 mg/kg, drinking water) for 6-7 weeks showed a significant improvement in cognitive function compared to RT-exposed mice receiving vehicle. Dual-immunofluorescence staining, spatial transcriptomics, and biochemical assessment of RZ-treated irradiated brains demonstrated preservation of synaptic integrity and mature neuronal plasticity but not neurogenesis and reduced neuroinflammation concurrent with elevated BDNF levels and transcripts compared to vehicle-treated irradiated brains. In summary, oral administration of RZ represents a viable and translationally feasible neuroprotective approach against RICD.
Collapse
Affiliation(s)
- Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Arya R Vagadia
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA
| | - Mingyu Du
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA.
| |
Collapse
|
5
|
El-Khatib SM, Vagadia AR, Le ACD, Ng DQ, Baulch JE, Du M, Tan Z, Xu X, Chan A, Acharya MM. BDNF augmentation reverses cranial radiation therapy-induced cognitive decline and neurodegenerative consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614590. [PMID: 39386496 PMCID: PMC11463616 DOI: 10.1101/2024.09.23.614590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cranial radiation therapy (RT) for brain cancers is often associated with the development of radiation-induced cognitive dysfunction (RICD). RICD significantly impacts the quality of life for cancer survivors, highlighting an unmet medical need. Previous human studies revealed a marked reduction in plasma brain-derived neurotrophic factor (BDNF) post-chronic chemotherapy, linking this decline to a substantial cognitive dysfunction among cancer survivors. Moreover, riluzole (RZ)-mediated increased BDNF in vivo in the chemotherapy-exposed mice reversed cognitive decline. RZ is an FDA-approved medication for ALS known to increase BDNF in vivo . In an effort to mitigate the detrimental effects of RT-induced BDNF decline in RICD, we tested the efficacy of RZ in a cranially irradiated (9 Gy) adult mouse model. Notably, RT-exposed mice exhibited significantly reduced hippocampal BDNF, accompanied by increased neuroinflammation, loss of neuronal plasticity-related immediate early gene product, cFos, and synaptic density. Spatial transcriptomic profiling comparing the RT+Veh with the RT+RZ group showed gene expression signatures of neuroprotection of hippocampal excitatory neurons post-RZ. RT-exposed mice performed poorly on learning and memory, and memory consolidation tasks. However, irradiated mice receiving RZ (13 mg/kg, drinking water) for 6-7 weeks showed a significant improvement in cognitive function compared to RT-exposed mice receiving vehicle. Dual-immunofluorescence staining, spatial transcriptomics, and biochemical assessment of RZ-treated irradiated brains demonstrated preservation of synaptic integrity and neuronal plasticity but not neurogenesis and reduced neuroinflammation concurrent with elevated BDNF levels and transcripts compared to vehicle-treated irradiated brains. In summary, oral administration of RZ represents a viable and translationally feasible neuroprotective approach against RICD.
Collapse
|
6
|
Sun R, Xi K, Ji S, Song X, Xi D, Yin W, Shao Y, Gu W, Jiang J. TDP-43 was Involved in Radiation-induced Neuronal Damage and May Not Through the BDNF/TrkB Pathway. Radiat Res 2024; 201:240-251. [PMID: 38235539 DOI: 10.1667/rade-23-00168.1] [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: 08/03/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Cognitive dysfunction is the most common form of radiation-induced brain injury. TDP-43 is known to be associated with hippocampal degeneration and cognitive dysfunction, in this study we wanted to know if it also had an effect on radiation-induced hippocampus damage. At first, we found the expression of TDP-43 and p-TDP-43 was increased in the hippocampus of rats with radiation-induced cognitive dysfunction. Single-cell RNA-seq analysis of the rat hippocampus showed that TDP-43 was expressed in all cell types and was significantly upregulated in neuron cells after irradiation. Enrichment analysis of gene ontology (GO) functions and KEGG pathways showed that the differential expression genes in neuron after irradiation may be involved in synaptic plasticity. In vitro, the expression of TDP-43 was also increased in neuron cells after irradiation, while the expression of brain-derived neurotrophic factor (BDNF), TrkB, typical synaptic signature proteins (SYN, GAP43 and PSD95), β-tubulin and dendritic spines were decreased. In the irradiated neurons, the β-tubulin, dendritic and spines typical synaptic signature proteins had more severe damage in pcDNA3.1-TDP-43 plasmid transfections group, however, the damages were alleviated in the siRNA-TDP-43 plasmid transfections group. BDNF was highly expressed in the irradiated pcDNA3.1-TDP-43 plasmid transfections group, while its expression was decreased in the siRNA-TDP-43 group. The TrkB expression was significantly reduced in neurons after exposure to ionizing radiation, however, there was no significant correlation with TDP-43 expression. These data indicate that TDP-43 is involved in radiation-induced neuronal synaptic plasticity decline and developmental damage, furthermore, the BDNF/TrkB signaling pathway may not be involved in this process.
Collapse
Affiliation(s)
- Rui Sun
- Department of Oncology Radiotherapy
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Kedi Xi
- Department of Oncology Radiotherapy
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | | | - Dan Xi
- Department of Oncology Radiotherapy
| | | | | | | | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
7
|
Nakkazi A, Forster D, Whitfield GA, Dyer DP, Dickie BR. A systematic review of normal tissue neurovascular unit damage following brain irradiation-Factors affecting damage severity and timing of effects. Neurooncol Adv 2024; 6:vdae098. [PMID: 39239570 PMCID: PMC11375288 DOI: 10.1093/noajnl/vdae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Background Radiotherapy is key in the treatment of primary and secondary brain tumors. However, normal tissue is inevitably irradiated, causing toxicity and contributing to cognitive dysfunction. The relative importance of vascular damage to cognitive decline is poorly understood. Here, we systematically review the evidence for radiation-induced damage to the entire neurovascular unit (NVU), particularly focusing on establishing the factors that influence damage severity, and timing and duration of vascular effects relative to effects on neural tissue. Methods Using PubMed and Web of Science, we searched preclinical and clinical literature published between January 1, 1970 and December 1, 2022 and evaluated factors influencing NVU damage severity and timing of NVU effects resulting from ionizing radiation. Results Seventy-two rodents, 4 canines, 1 rabbit, and 5 human studies met inclusion criteria. Radiation increased blood-brain barrier (BBB) permeability, reduced endothelial cell number and extracellular matrix proteoglycans, reduced tight junction proteins, upregulated cellular adhesion molecule expression, reduced activity of glucose and BBB efflux transporters and activated glial cells. In the brain parenchyma, increased metalloproteinases 2 and 9 levels, demyelination, cell death, and inhibited differentiation were observed. Effects on the vasculature and neural compartment were observed across acute, delayed, and late timepoints, and damage extent was higher with low linear energy transfer radiation, higher doses, lower dose rates, broader beams, and in the presence of a tumor. Conclusions Irradiation of normal brain tissue leads to widespread and varied impacts on the NVU. Data indicate that vascular damage is in most cases an early effect that does not quickly resolve. More studies are needed to confirm sequence of damages, and mechanisms that lead to cognitive dysfunction.
Collapse
Affiliation(s)
- Annet Nakkazi
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
- Faculty of Biology, Medicine, and Health, Division of Informatics, Imaging, and Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Duncan Forster
- Faculty of Biology, Medicine, and Health, Division of Informatics, Imaging, and Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Gillian A Whitfield
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - Douglas P Dyer
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
| | - Ben R Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
- Faculty of Biology, Medicine, and Health, Division of Informatics, Imaging, and Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Liu Z, Xu K, Pan S, Zhang N, Wang D, Chen Y, Zhao Y, Wang S, Li J, Tong X. Manganese-enhanced magnetic resonance assessment of changes in hippocampal neural function after the treatment of radiation-induced brain injury with bone marrow mesenchymal stem cells. Brain Res Bull 2023; 204:110795. [PMID: 37863438 DOI: 10.1016/j.brainresbull.2023.110795] [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: 07/20/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
The role of bone marrow mesenchymal stem cells (BMSCs) in treating radiation-induced brain injury (RIBI) is not completely understood, and assessment methods to directly characterize neurological function are lacking. In this study, we aimed to evaluate the effects of BMSCs treatment on changes in hippocampal neural function in Sprague-Dawley(SD) rats with RIBI, and to evaluate the therapeutic effect of BMSCs by manganese-enhanced magnetic resonance imaging (MEMRI). First, we assessed cognitive function after RIBI treatment with BMSCs using the Morris water maze. Next, we used MEMRI at two time points to observe the treatment effect and explore the correlation between MEMRI and cognitive function. Finally, we evaluated the expression of specific hippocampal neurofunctional proteins, the ultrastructure of hippocampal nerves, and the histological changes in the hippocampus. After BMSCs treatment of RIBI, cognitive dysfunction improved significantly, the expression of hippocampal neurofunctional proteins was increased, the integrity of the hippocampal neural structure was protected, and nerve cell survival was enhanced. The improvement in neurological function was successfully detected by MEMRI, and MEMRI was highly correlated with cognitive function and histological changes. These results suggest that BMSCs treatment of RIBI is an optional modality, and MEMRI can be used for treatment evaluation.
Collapse
Affiliation(s)
- Zhanhong Liu
- College of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Kaina Xu
- College of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Shichao Pan
- College of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Na Zhang
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Dapeng Wang
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Ying Chen
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Yaru Zhao
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Siqi Wang
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Jing Li
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China
| | - Xu Tong
- Department of Radiotherapy, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161006, China.
| |
Collapse
|
9
|
Liu X, Liu M, Liu H, Yuan H, Wang Y, Chen X, Li J, Qin X. Comprehensive brain tissue metabolomics and biological network technology to decipher the mechanism of hydrogen-rich water on Radiation-induced cognitive impairment in rats. BMC Mol Cell Biol 2023; 24:30. [PMID: 37752412 PMCID: PMC10523633 DOI: 10.1186/s12860-023-00491-4] [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: 01/17/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Hydrogen-rich water (HRW) has been shown to prevent cognitive impairment caused by ionizing radiation. This study aimed to investigate the pharmacological effects and mechanisms of HRW on ionizing radiation by coupling the brain metabolomics and biological target network methods. METHODS AND RESULTS HRW significantly improves the cognitive impairment in rats exposed to ionizing radiation. Based on metabolomics and biological network results, we identified 54 differential metabolites and 93 target genes. The KEGG pathway indicates that glutathione metabolism, ascorbic acid and aldehyde acid metabolism, pentose and glucuronic acid interconversion, and glycerophospholipid metabolism play important roles in ionizing radiation therapy. CONCLUSION Our study has systematically elucidated the molecular mechanism of HRW against ionizing radiation, which can be mediated by modulating targets, pathways and metabolite levels. This provides a new perspective for identifying the underlying pharmacological mechanism of HRW.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China
| | - Mengya Liu
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China
| | - Huan Liu
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China
| | - Hui Yuan
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China
| | - Yong Wang
- School of forensic medicine, Shanxi Medical University, Taiyuan, 030001, P. R. China
| | - Xiaoman Chen
- School of forensic medicine, Shanxi Medical University, Taiyuan, 030001, P. R. China
| | - Jianguo Li
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China
| | - Xiujun Qin
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, CAEA Center of Excellence on Nuclear Technology Applications for Non-Clinical Evaluation for Radiopharmaceutical, Shanxi Key Laboratory for Pharmaceutical Toxicology & Radiation Injury Pharmaceuticals, CNNC Key Laboratory for Radiotoxicology and Preclinical Assessment of Radiopharmaceuticals, Taiyuan, 030006, P. R. China.
| |
Collapse
|
10
|
Comparison of whole brain radiation therapy for synchronous brain metastases with irradiation protecting the hippocampus versus whole brain radiotherapy for sequential brain metastases to boost irradiation in the treatment of brain metastases from SCLC: study protocol for a randomized controlled trial. Trials 2022; 23:876. [PMID: 36242060 PMCID: PMC9569116 DOI: 10.1186/s13063-022-06826-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 10/05/2022] [Indexed: 12/04/2022] Open
Abstract
Background This study is in regard to the comparison of whole brain radiation therapy for synchronous brain metastases with irradiation protecting the hippocampus versus whole brain radiotherapy for sequential brain metastases to boost irradiation in the treatment of brain metastases from small cell lung cancer (SCLC). Therapeutically, they have notably varying dose distributions. Based on theoretical and model studies, it has long been speculated that these modes may result in different prognostic outcomes. We aim to assess the efficacy of tomotherapy in the treatment of SCLC brain metastases while protecting the key functional area, the hippocampus, and minimizing any neurocognitive impairments incurred by radiation. Methods This is a randomized, controlled, prospective study including 102 SCLC patients with brain metastases randomized (1:1) to the experimental (whole brain radiation therapy for synchronous brain metastases with irradiation to protect the hippocampus) or control (whole brain radiotherapy for sequential brain metastases to boost irradiation) group. The sample size is calculated through a single-sided test; 102 participants will be required for the main results to have statistical and clinical significance. We aim to provide clinical trial data support for better prognostic treatment options in patients with SCLC and brain metastases. The clinical trial data include both the primary and secondary outcomes; the primary outcome is the intracranial progression-free survival time after the new technology application. The secondary study outcomes include the assessment of neurological function, the quality of life, and the overall survival rate. Follow-up consultations will be conducted every 2 months. After the final patient completes follow-up, the Statistical Product and Service Solutions software will be used for scientific and rigorous data analysis. Version 1.0 of the protocol was implemented on January 1, 2021; the recruitment process for this clinical trial commenced on April 1, 2021, and will end on March 31, 2024. Discussion The study will provide high-quality clinical evidence to support the efficacy and safety of whole brain radiation therapy for synchronous brain metastases with dose irradiation protecting the hippocampus versus whole brain radiotherapy for sequential brain metastases with push volume irradiation for the treatment of patients who have lung cancer as well as brain metastases. This has not been previously reported. Trial registration This trial is registered with the Chinese Clinical Trial Registry (ChiCTR1900027539; November 17, 2019) (URL: https://www.chictr.org.cn/hvshowproject.aspx?id=20515).
Collapse
|
11
|
De Stefano I, Leonardi S, Casciati A, Pasquali E, Giardullo P, Antonelli F, Novelli F, Babini G, Tanori M, Tanno B, Saran A, Mancusoa M, Pazzaglia S. Contribution of Genetic Background to the Radiation Risk for Cancer and Non-Cancer Diseases in Ptch1+/- Mice. Radiat Res 2022; 197:43-56. [PMID: 33857285 DOI: 10.1667/rade-20-00247.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/19/2021] [Indexed: 11/03/2022]
Abstract
Experimental mouse studies are important to gain a comprehensive, quantitative and mechanistic understanding of the biological factors that modify individual risk of radiation-induced health effects, including age at exposure, dose, dose rate, organ/tissue specificity and genetic factors. In this study, neonatal Ptch1+/- mice bred on CD1 and C57Bl/6 background received whole-body irradiation at postnatal day 2. This time point represents a critical phase in the development of the eye lens, cerebellum and dentate gyrus (DG), when they are also particularly susceptible to radiation effects. Irradiation was performed with γ rays (60Co) at doses of 0.5, 1 and 2 Gy, delivered at 0.3 Gy/min or 0.063 Gy/min. Wild-type and mutant mice were monitored for survival, lens opacity, medulloblastoma (MB) and neurogenesis defects. We identified an inverse genetic background-driven relationship between the radiosensitivity to induction of lens opacity and MB and that to neurogenesis deficit in Ptch1+/- mutants. In fact, high incidence of radiation-induced cataract and MB were observed in Ptch1+/-/CD1 mutants that instead showed no consequence of radiation exposure on neurogenesis. On the contrary, no induction of radiogenic cataract and MB was reported in Ptch1+/-/C57Bl/6 mice that were instead susceptible to induction of neurogenesis defects. Compared to Ptch1+/-/CD1, the cerebellum of Ptch1+/-/C57Bl/6 mice showed increased radiosensitivity to apoptosis, suggesting that differences in processing radiation-induced DNA damage may underlie the opposite strain-related radiosensitivity to cancer and non-cancer pathologies. Altogether, our results showed lack of dose-rate-related effects and marked influence of genetic background on the radiosensitivity of Ptch1+/-mice, supporting a major contribution of individual sensitivity to radiation risk in the population.
Collapse
Affiliation(s)
- I De Stefano
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - S Leonardi
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - A Casciati
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - E Pasquali
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - P Giardullo
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - F Antonelli
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - F Novelli
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - G Babini
- Department of Physics, University of Pavia, Pavia, Italy
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - M Tanori
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - B Tanno
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - A Saran
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - M Mancusoa
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - S Pazzaglia
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| |
Collapse
|
12
|
Yang S, Fu ZZ, Zhang YQ, Fu BH, Dong L. The G to A transformation of rs4702 polymorphism in 3'UTR of FURIN reduced the risk of radiotherapy-induced cognitive impairment in glioma patients. J Cell Mol Med 2021; 26:684-692. [PMID: 34953024 PMCID: PMC8817137 DOI: 10.1111/jcmm.17074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/14/2021] [Accepted: 10/24/2021] [Indexed: 11/28/2022] Open
Abstract
The G allele of rs4702 polymorphism has been reported to reduce the production of mature BDNF and FURIN, both of which were closely associated with cognitive functions. Real‐time PCR, ELISA and luciferase assay were performed to explore the interactions between miR‐338‐3p, FURIN and BDNF. T‐RFLP was used to assess the intestinal flora in the stool samples of glioma patients after radiotherapy. We grouped the 106 glioma patients recruited according to the rs4702 polymorphism. The results showed no obvious correlation between rs4702 polymorphism and the expression of miR‐338‐3p. However, rs4702‐A was associated with increased expression of FURIN and BDNF in the serum and PBMC of glioma patients after radiotherapy. Besides, the study found that rs4702‐A was remarkably associated with increased enterotype I and decreased enterotype III in the stool of glioma patients after radiotherapy. Rs4702‐A was also proved to be closely associated with increased MMSE, role functioning and social functioning at three months after radiotherapy. Furthermore, miR‐338‐3p repressed the expression of FURIN‐G. Compared with G allele, the presence of A allele of rs4702 polymorphism in FURIN could obstruct the suppressive effect of miR‐338‐3p upon the expression of FURIN and BDNF in intestinal flora. Therefore, the carriers of A allele will be challenged with less risk of radiotherapy‐induced cognitive impairment.
Collapse
Affiliation(s)
- Sen Yang
- Department of Radiotherapy (Ward II), Qinhuangdao First Hospital, Qinhuangdao, China
| | - Zhan-Zhao Fu
- Department of Radiotherapy (Ward II), Qinhuangdao First Hospital, Qinhuangdao, China
| | - Yan-Qiu Zhang
- Department of Radiotherapy (Ward II), Qinhuangdao First Hospital, Qinhuangdao, China
| | - Bao-Hong Fu
- Department of Radiotherapy (Ward II), Qinhuangdao First Hospital, Qinhuangdao, China
| | - Lixin Dong
- Department of Radiotherapy (Ward II), Qinhuangdao First Hospital, Qinhuangdao, China
| |
Collapse
|
13
|
Can Dexmedetomidine Be Effective in the Protection of Radiotherapy-Induced Brain Damage in the Rat? Neurotox Res 2021; 39:1338-1351. [PMID: 34057703 DOI: 10.1007/s12640-021-00379-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/10/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
Approximately 7 million people are reported to be undergoing radiotherapy (RT) at any one time in the world. However, it is still not possible to prevent damage to secondary organs that are off-target. This study, therefore, investigated the potential adverse effects of RT on the brain, using cognitive, histopathological, and biochemical methods, and the counteractive effect of the α2-adrenergic receptor agonist dexmedetomidine. Thirty-two male Sprague Dawley rats aged 5-6 months were randomly allocated into four groups: untreated control, and RT, RT + dexmedetomidine-100, and RT + dexmedetomidine-200-treated groups. The passive avoidance test was applied to all groups. The RT groups received total body X-ray irradiation as a single dose of 8 Gy. The rats were sacrificed 24 h after X-ray irradiation, and following the application of the passive avoidance test. The brain tissues were subjected to histological and biochemical evaluation. No statistically significant difference was found between the control and RT groups in terms of passive avoidance outcomes and 8-hydroxy-2'- deoxyguanosine (8-OHdG) positivity. In contrast, a significant increase in tissue MDA and GSH levels and positivity for TUNEL, TNF-α, and nNOS was observed between the control and the irradiation groups (p < 0.05). A significant decrease in these values was observed in the groups receiving dexmedetomidine. Compared with the control group, gradual elevation was determined in GSH levels in the RT group, followed by the RT + dexmedetomidine-100 and RT + dexmedetomidine-200 groups. Dexmedetomidine may be beneficial in countering the adverse effects of RT in the cerebral and hippocampal regions.
Collapse
|
14
|
Behl T, Kaur G, Sehgal A, Zengin G, Singh S, Ahmadi A, Bungau S. Flavonoids, the Family of Plant-derived Antioxidants making inroads into Novel Therapeutic Design against IR-induced Oxidative Stress in Parkinson's Disease. Curr Neuropharmacol 2021; 20:324-343. [PMID: 34030619 PMCID: PMC9413797 DOI: 10.2174/1570159x19666210524152817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/17/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022] Open
Abstract
Background: Ionizing radiation from telluric sources is unceasingly an unprotected pitfall to humans. Thus, the foremost contributors to human exposure are global and medical radiations. Various evidences assembled during preceding years reveal the pertinent role of ionizing radiation-induced oxidative stress in the progression of neurodegenerative insults, such as Parkinson’s disease, which have been contributing to increased proliferation and generation of reactive oxygen species. Objective: This review delineates the role of ionizing radiation-induced oxidative stress in Parkinson’s disease and proposes novel therapeutic interventions of flavonoid family, offering effective management and slowing down the progression of Parkinson’s disease. Methods: Published papers were searched in MEDLINE, PubMed, etc., published to date for in-depth database collection. Results: The oxidative damage may harm the non-targeted cells. It can also modulate the functions of the central nervous system, such as protein misfolding, mitochondria dysfunction, increased levels of oxidized lipids, and dopaminergic cell death, which accelerate the progression of Parkinson’s disease at the molecular, cellular, or tissue levels. In Parkinson’s disease, reactive oxygen species exacerbate the production of nitric oxides and superoxides by activated microglia, rendering death of dopaminergic neuronal cell through different mechanisms. Conclusion: Rising interest has extensively engrossed in the clinical trial designs based on the plant-derived family of antioxidants. They are known to exert multifarious impact on neuroprotection via directly suppressing ionizing radiation-induced oxidative stress and reactive oxygen species production or indirectly increasing the dopamine levels and activating the glial cells.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Gagandeep Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari. Iran
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| |
Collapse
|
15
|
Out-of-Field Hippocampus from Partial-Body Irradiated Mice Displays Changes in Multi-Omics Profile and Defects in Neurogenesis. Int J Mol Sci 2021; 22:ijms22084290. [PMID: 33924260 PMCID: PMC8074756 DOI: 10.3390/ijms22084290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
The brain undergoes ionizing radiation exposure in many clinical situations, particularly during radiotherapy for brain tumors. The critical role of the hippocampus in the pathogenesis of radiation-induced neurocognitive dysfunction is well recognized. The goal of this study is to test the potential contribution of non-targeted effects in the detrimental response of the hippocampus to irradiation and to elucidate the mechanisms involved. C57Bl/6 mice were whole body (WBI) or partial body (PBI) irradiated with 0.1 or 2.0 Gy of X-rays or sham irradiated. PBI consisted of the exposure of the lower third of the mouse body, whilst the upper two thirds were shielded. Hippocampi were collected 15 days or 6 months post-irradiation and a multi-omics approach was adopted to assess the molecular changes in non-coding RNAs, proteins and metabolic levels, as well as histological changes in the rate of hippocampal neurogenesis. Notably, at 2.0 Gy the pattern of early molecular and histopathological changes induced in the hippocampus at 15 days following PBI were similar in quality and quantity to the effects induced by WBI, thus providing a proof of principle of the existence of out-of-target radiation response in the hippocampus of conventional mice. We detected major alterations in DAG/IP3 and TGF-β signaling pathways as well as in the expression of proteins involved in the regulation of long-term neuronal synaptic plasticity and synapse organization, coupled with defects in neural stem cells self-renewal in the hippocampal dentate gyrus. However, compared to the persistence of the WBI effects, most of the PBI effects were only transient and tended to decrease at 6 months post-irradiation, indicating important mechanistic difference. On the contrary, at low dose we identified a progressive accumulation of molecular defects that tended to manifest at later post-irradiation times. These data, indicating that both targeted and non-targeted radiation effects might contribute to the pathogenesis of hippocampal radiation-damage, have general implications for human health.
Collapse
|
16
|
Zhang J, Zhang Y, Xu M, Miao Z, Tian Y. Inhibition of the CDK5/caspase-3 Pathway by p5-TAT Protects Hippocampal Neurogenesis and Alleviates Radiation-induced Cognitive Dysfunction. Neuroscience 2021; 463:204-215. [PMID: 33838288 DOI: 10.1016/j.neuroscience.2021.03.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 11/17/2022]
Abstract
Radiation-induced cognitive dysfunction is a common complication associated with cranial radiation therapy. Inhibition of hippocampal neurogenesis and proliferation plays a critical role in this complication. Relieving hippocampal apoptosis may significantly protect hippocampal neurogenesis and proliferation. Previous studies have demonstrated that hyperactivity of cyclin-dependent kinase 5 (Cdk5) is closely related to apoptosis. The exact molecular changes and function of Cdk5 in radiation-induced cognitive dysfunction are still not clear. Whether inhibition of Cdk5 and the relevant caspase-3 could improve hippocampal neurogenesis and ameliorate radiation-induced cognitive dysfunction needs further exploration. We hypothesized that inhibition of the Cdk5/caspase-3 pathway by p5-TAT could protect hippocampal neurogenesis and alleviate radiation-induced cognitive dysfunction. In our study, we reported that radiation induced hyperactivity of Cdk5 accompanied by elevation of the levels of cleaved caspase-3, a marker of neuronal apoptosis. Inhibition of hippocampal neurogenesis and proliferation as well as cognitive dysfunction was also observed. p5-TAT, a specific inhibitor of Cdk5, decreased the overactivation of Cdk5 without affecting the levels of Cdk5 activators. Additionally, this treatment suppressed the expression of cleaved caspase-3. We further demonstrated that p5-TAT treatment reduced hippocampal dysfunction and improved behavioral performance. Therefore, Cdk5 inhibition by the small peptide p5-TAT is a promising therapeutic strategy for radiation-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Junjun Zhang
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, China; Institute of Radiotherapy and Oncology, Soochow University, China; Suzhou Key Laboratory for Radiation Oncology, China
| | - Yujuan Zhang
- Experiment Center, Medicine College of Soochow University, Suzhou City, China
| | - Meiling Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, China; Institute of Radiotherapy and Oncology, Soochow University, China; Suzhou Key Laboratory for Radiation Oncology, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, China.
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, China; Institute of Radiotherapy and Oncology, Soochow University, China; Suzhou Key Laboratory for Radiation Oncology, China.
| |
Collapse
|
17
|
Saw G, Tang FR. Epigenetic Regulation of the Hippocampus, with Special Reference to Radiation Exposure. Int J Mol Sci 2020; 21:ijms21249514. [PMID: 33327654 PMCID: PMC7765140 DOI: 10.3390/ijms21249514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 01/28/2023] Open
Abstract
The hippocampus is crucial in learning, memory and emotion processing, and is involved in the development of different neurological and neuropsychological disorders. Several epigenetic factors, including DNA methylation, histone modifications and non-coding RNAs, have been shown to regulate the development and function of the hippocampus, and the alteration of epigenetic regulation may play important roles in the development of neurocognitive and neurodegenerative diseases. This review summarizes the epigenetic modifications of various cell types and processes within the hippocampus and their resulting effects on cognition, memory and overall hippocampal function. In addition, the effects of exposure to radiation that may induce a myriad of epigenetic changes in the hippocampus are reviewed. By assessing and evaluating the current literature, we hope to prompt a more thorough understanding of the molecular mechanisms that underlie radiation-induced epigenetic changes, an area which can be further explored.
Collapse
|
18
|
Ji S, Wu H, Ding X, Chen Q, Jin X, Yu J, Yang M. Increased hippocampal TrkA expression ameliorates cranial radiation‑induced neurogenesis impairment and cognitive deficit via PI3K/AKT signaling. Oncol Rep 2020; 44:2527-2536. [PMID: 33125501 PMCID: PMC7640353 DOI: 10.3892/or.2020.7782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 09/01/2020] [Indexed: 01/13/2023] Open
Abstract
Cognitive deficit is one of the most serious complications of cranial radiotherapy of head and neck cancers. However, the underlying mechanism of this cognitive impairment remains unclear. In the present study, the role of tropomyosin receptor kinase A (TrkA) and its ligand neurotrophin nerve growth factor (NGF) were investigated following whole‑brain irradiation (WBI). Young male Sprague‑Dawley rats underwent WBI at a single dose of 10 Gy. WBI was determined to result in notable memory decline and substantial neurogenesis impairment in the hippocampus 3 months post‑irradiation. Compared with the control group, TrkA protein expression was greater in irradiated rats 1 week after WBI, which then decreased significantly by the 3‑month time‑point. However, no difference in NGF expression was observed from 1 day to 3 months post‑WBI. Overexpression of hippocampal TrkA in rats using adeno‑associated virus ameliorated memory decline induced by irradiation. Additionally, upregulating TrkA expression rescued irradiation‑induced hippocampal precursor cell proliferation and promoted neurogenesis. PI3K, Akt and ERK1/2 phosphorylation were also revealed to be significantly inhibited by WBI, which was ameliorated by TrkA overexpression. Findings of the present study indicated that the TrkA‑dependent signaling pathway may serve a critical role in radiotherapy‑induced cognitive deficit and impairments in neurogenesis.
Collapse
Affiliation(s)
- Shengjun Ji
- Clinical College, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
- Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Haohao Wu
- Department of Radiotherapy and Oncology, Yancheng No. 1 People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Xin Ding
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Qingqing Chen
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Xing Jin
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jinming Yu
- Clinical College, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ming Yang
- Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
19
|
Raber J, Fuentes Anaya A, Torres ERS, Lee J, Boutros S, Grygoryev D, Hammer A, Kasschau KD, Sharpton TJ, Turker MS, Kronenberg A. Effects of Six Sequential Charged Particle Beams on Behavioral and Cognitive Performance in B6D2F1 Female and Male Mice. Front Physiol 2020; 11:959. [PMID: 32982769 PMCID: PMC7485338 DOI: 10.3389/fphys.2020.00959] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
The radiation environment astronauts are exposed to in deep space includes galactic cosmic radiation (GCR) with different proportions of all naturally occurring ions. To assist NASA with assessment of risk to the brain following exposure to a mixture of ions broadly representative of the GCR, we assessed the behavioral and cognitive performance of female and male C57BL/6J × DBA2/J F1 (B6D2F1) mice two months following rapidly delivered, sequential 6 beam irradiation with protons (1 GeV, LET = 0.24 keV, 50%), 4He ions (250 MeV/n, LET = 1.6 keV/μm, 20%), 16O ions (250 MeV/n, LET = 25 keV/μm 7.5%), 28Si ions (263 MeV/n, LET = 78 keV/μm, 7.5%), 48Ti ions (1 GeV/n, LET = 107 keV/μm, 7.5%), and 56Fe ions (1 GeV/n, LET = 151 keV/μm, 7.5%) at 0, 25, 50, or 200 cGy) at 4-6 months of age. When the activity over 3 days of open field habituation was analyzed in female mice, those irradiated with 50 cGy moved less and spent less time in the center than sham-irradiated mice. Sham-irradiated female mice and those irradiated with 25 cGy showed object recognition. However, female mice exposed to 50 or 200 cGy did not show object recognition. When fear memory was assessed in passive avoidance tests, sham-irradiated mice and mice irradiated with 25 cGy showed memory retention while mice exposed to 50 or 200 cGy did not. The effects of radiation passive avoidance memory retention were not sex-dependent. There was no effect of radiation on depressive-like behavior in the forced swim test. There was a trend toward an effect of radiation on BDNF levels in the cortex of males, but not for females, with higher levels in male mice irradiated with 50 cGy than sham-irradiated. Finally, sequential 6-ion irradiation impacted the composition of the gut microbiome in a sex-dependent fashion. Taxa were uncovered whose relative abundance in the gut was associated with the radiation dose received. Thus, exposure to sequential six-beam irradiation significantly affects behavioral and cognitive performance and the gut microbiome.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Andrea Fuentes Anaya
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Eileen Ruth S. Torres
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joanne Lee
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Sydney Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Dmytro Grygoryev
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States
| | - Austin Hammer
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Kristin D. Kasschau
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
- Department of Statistics, Oregon State University, Corvallis, OR, United States
| | - Mitchell S. Turker
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
20
|
Wang Z, Zhong XL, Xu Y, He J, Liu ZH, Nai AT, Niu L, Luo SS, Yang H, Zeng JY, He SY, Chen X, Wan W, Cao WY. Irradiation increases brain-derived neurotrophic factor precursor signaling in the mouse hippocampus. Neurobiol Learn Mem 2020; 171:107186. [PMID: 32084558 DOI: 10.1016/j.nlm.2020.107186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Zhen Wang
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Xiao Lin Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001 Hengyang, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Jie He
- Department of Pathology, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Zheng Hai Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Ai Tao Nai
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, 421001 Hengyang, Hunan, China
| | - Lei Niu
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Shi Shi Luo
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Hui Yang
- Department of Pathology, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Jia Yu Zeng
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Shu Ya He
- School of Public Health, University of South China, 421001 Hengyang, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Wei Wan
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China; Key Laboratory of Brain Science Research & Transformation In Tropical Environment of Hainan Province, Hainan Medical University, 571199 Haikou, China.
| | - Wen Yu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China.
| |
Collapse
|
21
|
Gan L, Guo M, Si J, Zhang J, Liu Z, Zhao J, Wang F, Yan J, Li H, Zhang H. Protective effects of phenformin on zebrafish embryonic neurodevelopmental toxicity induced by X-ray radiation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:4202-4210. [DOI: 10.1080/21691401.2019.1687505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Lu Gan
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Menghuan Guo
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jing Si
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinhua Zhang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Liu
- College of Chemical Engineering, Northwest Minzu University, Lanzhou, China
| | - Jin Zhao
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Fang Wang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Junfang Yan
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hongyan Li
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hong Zhang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
Liu M, Yuan H, Yin J, Wang R, Song J, Hu B, Li J, Qin X. Effect of Hydrogen-Rich Water on Radiation-Induced Cognitive Dysfunction in Rats. Radiat Res 2019; 193:16-23. [PMID: 31634054 DOI: 10.1667/rr15464.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The goal of this work was to determine whether hydrogen-rich water (HRW) could attenuate radiation-induced cognitive dysfunction in rats and to explore the underlying mechanisms. Rats received 30 Gy whole-brain irradiation using a 6-MeV electron beam. Either purified water or HRW (0.8-0.9 ppm) was administrated at 10 min prior to irradiation, as well as a daily HRW treatment after irradiation for 30 consecutive days. The Morris water maze was used to test spatial memory in the rats. The concentration of glutathione (GSH), malondialdehyde (MDA), 8-hydroxydeoxyguanosine (8-OHdG) and the super-oxidedismutase (SOD) activity in cerebral cortex, as well as brain-derived neurotrophic factor (BDNF) level in serum, were measured. Immunofluorescence staining was adopted to detect proliferating cells. The expression of BDNF-TrkB pathway-related genes and proteins were detected using qRT-PCR and Western blot. Models of cognitive dysfunction were successfully established using a 30 Gy dose of ionizing radiation. Compared to the radiation treated group, the radiation-HRW treated group showed significantly decreased escape latency (P < 0.05), but increased retention time, swimming distance of original platform quadrant (P < 0.05) and number of platform crossings (P < 0.05). Furthermore, the SOD, GSH (P < 0.05) and BDNF (P < 0.05) levels in the radiation-HRW treated group were higher compared to the radiation treated group. The MDA and 8-OHdG levels (P < 0.05) were decreased in the radiation-HRW treated group when compared to the radiation treated group. Additionally, treatment with HRW increased the number of BrdU+NeuN+ cells in the radiation treated group. The mRNA and protein levels of BDNF and TrkB (P < 0.05) in radiation-HRW treated group was higher than that in the radiation treated group. Collectively, our study indicates that HRW has a protective effect on radiation-induced cognitive dysfunction, and that the possible mechanisms mainly involve anti-oxidative and anti-inflammatory reactions, and its protection of newborn neurons by regulating the BDNF-TrkB signaling pathway.
Collapse
Affiliation(s)
- Mengya Liu
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Hui Yuan
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Jingjing Yin
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Ruoqi Wang
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Jianbo Song
- Department of Radiotherapy, Shanxi Dayi Hospital, Taiyuan 030032, P. R. China
| | - Bo Hu
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Jianguo Li
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| | - Xiujun Qin
- Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, P.R. China
| |
Collapse
|
23
|
Metzdorf J, Hobloss Z, Schlevogt S, Ayzenberg I, Stahlke S, Pedreiturria X, Haupeltshofer S, Gold R, Tönges L, Kleiter I. Fingolimod for Irradiation-Induced Neurodegeneration. Front Neurosci 2019; 13:699. [PMID: 31354410 PMCID: PMC6633210 DOI: 10.3389/fnins.2019.00699] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/20/2019] [Indexed: 01/10/2023] Open
Abstract
Background Cranial irradiation is a common therapy for the treatment of brain tumors, but unfortunately patients suffer from side effects, particularly cognitive impairment, caused by neurodegenerative and neuroinflammatory mechanisms. Finding a therapeutic agent protecting hippocampal neurons would be beneficial. Fingolimod (FTY720), a sphingosine-1-phosphate receptor modulator approved for multiple sclerosis, is an immunosuppressant and known to enhance proliferation and differentiation of neuronal precursor cells (NPCs). Objectives To investigate whether pre-treatment with FTY720 protects NPCs in vitro and in vivo from irradiation-induced damage. Methods Neuronal precursor cells were isolated from E13 C57BL/6 wildtype mice, treated at day 0 of differentiation with FTY720 and irradiated on day 6 with 1 Gy. NPCs were analyzed for markers of cell death (PI, caspase-3), proliferation (Ki67), and differentiation (DCX, βIII-tubulin). Adult C57BL/6 wildtype mice were treated with FTY720 (1 mg/kg) and received a single dose of 6 Gy cranial irradiation at day 7. Using immunohistochemistry, we analyzed DCX and BrdU as markers of neurogenesis and Iba1, GFAP, and CD3 to visualize inflammation in the dentate gyrus (DG) and the subventricular zone (SVZ). B6(Cg)-Tyrc-2J/J DCX-luc reporter mice were used for bioluminescence imaging to evaluate the effect of FTY720 on neurogenesis in the DG and the spinal cord of naïve mice. Results FTY720 protected NPCs against irradiation induced cell death in vitro. Treatment with FTY720 dose-dependently reduced the number of PI+ cells 24 and 96 h after irradiation without effecting proliferation or neuronal differentiation. In vivo treatment resulted in a significant survival of DCX+ neurons in the DG and the SVZ 4 weeks after irradiation as well as a slight increase of proliferating cells. FTY720 inhibited microglia activation 24 h after X-ray exposure in the DG, while astrocyte activation was unaffected and no lymphocyte infiltrations were found. In naïve mice, FTY720 treatment for 4 weeks had no effect on neurogenesis. Conclusion FTY720 treatment of NPCs prior to X-ray exposure and of mice prior to cranial irradiation is neuroprotective. No effects on neurogenesis were found.
Collapse
Affiliation(s)
- Judith Metzdorf
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Zaynab Hobloss
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Sibylle Schlevogt
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Ilya Ayzenberg
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany.,Department of Neurology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sarah Stahlke
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | | | | | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany.,Marianne-Strauß-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke, Berg, Germany
| |
Collapse
|
24
|
Jiang A, Sun W, Zhao F, Wu Z, Shang D, Yu Q, Wang S, Zhu J, Yang F, Yuan S. Dosimetric evaluation of four whole brain radiation therapy approaches with hippocampus and inner ear avoidance and simultaneous integrated boost for limited brain metastases. Radiat Oncol 2019; 14:46. [PMID: 30876444 PMCID: PMC6419811 DOI: 10.1186/s13014-019-1255-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/07/2019] [Indexed: 01/31/2023] Open
Abstract
AIMS To perform a dosimetric evaluation of four different simultaneous integrated boost whole brain radiotherapy modalities with hippocampus and inner ear avoidance in the treatment of limited brain metastases. METHODS Computed tomography/magnetic resonance imaging data of 10 patients with limited (1-5) brain metastases were used to replan step-and-shoot intensity-modulated radiotherapy (sIMRT), dynamic intensity-modulated radiation therapy (dIMRT), volumetric-modulated arc therapy (VMAT), and helical tomotherapy (Tomo). The prescribed doses of 40-50 Gy in 10 fractions and 30 Gy in 10 fractions were simultaneously delivered to the metastatic lesions and the whole-brain volume, respectively. The hippocampal dose met the RTOG 0933 criteria for hippocampal avoidance (Dmax ≤17 Gy, D100% ≤10 Gy). The inner ear dose was restrained to Dmean ≤15 Gy. Target coverage (TC), homogeneity index (HI), conformity index (CI), maximum dose (Dmax), minimum dose (Dmin) and dose to organs at risk (OARs) were compared. RESULTS All plans met the indicated dose restrictions. The mean percentage of planning target volume of metastases (PTVmets) coverage ranged from 97.1 to 99.4%. For planning target volume of brain (PTVbrain), Tomo provided the lowest average D2% (37.5 ± 2.8 Gy), the highest average D98% (25.2 ± 2.0 Gy), and the best TC (92.6% ± 2.1%) and CI (0.79 ± 0.06). The two fixed gantry IMRT modalities (step and shot, dynamic) provided similar PTVbrain dose homogeneity (both 0.76). Significant differences across the four approaches were observed for the maximum and minimum doses to the hippocampus and the maximum doses to the eyes, lens and optic nerves. CONCLUSION All four radiotherapy modalities produced acceptable treatment plans with good avoidance of the hippocampus and inner ear. Tomo obtained satisfactory PTVbrain coverage and the best homogeneity index. TRIAL REGISTRATION Clinicaltrials.gov, NCT03414944 . Registered 29 January 2018.
Collapse
Affiliation(s)
- Aijun Jiang
- Shandong University, Jinan, 250117, Shandong, China.,Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Weipeng Sun
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Fen Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Zhenxuan Wu
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Dongping Shang
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Qingxi Yu
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Suzhen Wang
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Jian Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Fengchang Yang
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital affiliated to Shandong University, 440 Jiyan Road, Jinan, 250117, Shandong, China.
| |
Collapse
|
25
|
Raber J, Yamazaki J, Torres ERS, Kirchoff N, Stagaman K, Sharpton T, Turker MS, Kronenberg A. Combined Effects of Three High-Energy Charged Particle Beams Important for Space Flight on Brain, Behavioral and Cognitive Endpoints in B6D2F1 Female and Male Mice. Front Physiol 2019; 10:179. [PMID: 30914962 PMCID: PMC6422905 DOI: 10.3389/fphys.2019.00179] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/13/2019] [Indexed: 12/30/2022] Open
Abstract
The radiation environment in deep space includes the galactic cosmic radiation with different proportions of all naturally occurring ions from protons to uranium. Most experimental animal studies for assessing the biological effects of charged particles have involved acute dose delivery for single ions and/or fractionated exposure protocols. Here, we assessed the behavioral and cognitive performance of female and male C57BL/6J × DBA2/J F1 (B6D2F1) mice 2 months following rapidly delivered, sequential irradiation with protons (1 GeV, 60%), 16O (250 MeV/n, 20%), and 28Si (263 MeV/n, 20%) at 0, 25, 50, or 200 cGy at 4-6 months of age. Cortical BDNF, CD68, and MAP-2 levels were analyzed 3 months after irradiation or sham irradiation. During the dark period, male mice irradiated with 50 cGy showed higher activity levels in the home cage than sham-irradiated mice. Mice irradiated with 50 cGy also showed increased depressive behavior in the forced swim test. When cognitive performance was assessed, sham-irradiated mice of both sexes and mice irradiated with 25 cGy showed normal responses to object recognition and novel object exploration. However, object recognition was impaired in female and male mice irradiated with 50 or 200 cGy. For cortical levels of the neurotrophic factor BDNF and the marker of microglial activation CD68, there were sex × radiation interactions. In females, but not males, there were increased CD68 levels following irradiation. In males, but not females, there were reduced BDNF levels following irradiation. A significant positive correlation between BDNF and CD68 levels was observed, suggesting a role for activated microglia in the alterations in BDNF levels. Finally, sequential beam irradiation impacted the diversity and composition of the gut microbiome. These included dose-dependent impacts and alterations to the relative abundance of several gut genera, such as Butyricicoccus and Lachnospiraceae. Thus, exposure to rapidly delivered sequential proton, 16O ion, and 28Si ion irradiation significantly affects behavioral and cognitive performance, cortical levels of CD68 and BDNF in a sex-dependent fashion, and the gut microbiome.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Department of Neurology, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States.,Department of Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Joy Yamazaki
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Eileen Ruth S Torres
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Nicole Kirchoff
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Keaton Stagaman
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Thomas Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, United States.,Department of Statistics, Oregon State University, Corvallis, OR, United States
| | - Mitchell S Turker
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States.,Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
26
|
Zhang L, Ding X, Wu Z, Wang M, Tian M. Curcumin alleviates pain and improves cognitive impairment in a rat model of cobra venom-induced trigeminal neuralgia. J Pain Res 2018; 11:1095-1104. [PMID: 29950886 PMCID: PMC6016256 DOI: 10.2147/jpr.s162668] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Cognitive impairment is a common complication in patients with chronic neuropathic pain, without effective therapy. Recent works have indicated that curcumin (Cur) possesses antinociceptive and neuroprotective potentials, suggesting its possible effectiveness for the treatment of this complication. Objective The aim of this study was to explore the effects of Cur on pain behaviors and cognitive impairment in rats with cobra venom-induced trigeminal neuralgia (TN). Design This is a randomized, controlled experiment. Setting This study was conducted at the Experimental Animal Center, Beijing Friendship Hospital, Capital Medical University. Subjects A total of 40 adult male Sprague Dawley rats were used in this study. Methods A cobra venom solution was injected into the sheath of infraorbital nerve. Cur was administered intragastrically at 45 mg/kg twice daily for 28 successive days from postoperative day 15. Mechanical allodynia was evaluated using von Frey filaments. Free behaviors were observed using video recording. Cognitive capacity was tested using the Morris water maze. Both morphology and ultrastructure of the CA1 hippocampal region were visualized using hematoxylin and eosin (HE) staining and transmission electron microscopy, respectively. Results Cur treatment reduced mechanical allodynia and face-grooming activities but increased exploratory activities and improved spatial learning and memory deficits. Microscopic examination revealed nucleus pyknosis, swollen organelles, and decreased synapse density in the CA1 hippocampal region after cobra venom injection. However, chronic Cur treatment reversed damage to hippocampal neurons and synapses. Conclusion Cur can alleviate pain, improve spatial learning and memory deficits, and restore the damage to hippocampal neurons and synapses in cobra venom-induced TN rats. Cur may be useful as an adjuvant to treat chronic neuropathic pain-induced cognitive deficits.
Collapse
Affiliation(s)
- Li Zhang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050
| | - Xinli Ding
- Department of Rehabilitation Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021
| | - Zhe Wu
- Department of Anesthesiology, Pain Medicine and Critical Care Medicine, Aviation General Hospital of China Medical University, Beijing 100012, China
| | - Min Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050
| | - Ming Tian
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050
| |
Collapse
|
27
|
Detrimental Effects of Helium Ion Irradiation on Cognitive Performance and Cortical Levels of MAP-2 in B6D2F1 Mice. Int J Mol Sci 2018; 19:ijms19041247. [PMID: 29677125 PMCID: PMC5979430 DOI: 10.3390/ijms19041247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 11/17/2022] Open
Abstract
The space radiation environment includes helium (⁴He) ions that may impact brain function. As little is known about the effects of exposures to ⁴He ions on the brain, we assessed the behavioral and cognitive performance of C57BL/6J × DBA2/J F1 (B6D2F1) mice three months following irradiation with ⁴He ions (250 MeV/n; linear energy transfer (LET) = 1.6 keV/μm; 0, 21, 42 or 168 cGy). Sham-irradiated mice and mice irradiated with 21 or 168 cGy showed novel object recognition, but mice irradiated with 42 cGy did not. In the passive avoidance test, mice received a slight foot shock in a dark compartment, and latency to re-enter that compartment was assessed 24 h later. Sham-irradiated mice and mice irradiated with 21 or 42 cGy showed a higher latency on Day 2 than Day 1, but the latency to enter the dark compartment in mice irradiated with 168 cGy was comparable on both days. ⁴He ion irradiation, at 42 and 168 cGy, reduced the levels of the dendritic marker microtubule-associated protein-2 (MAP-2) in the cortex. There was an effect of radiation on apolipoprotein E (apoE) levels in the hippocampus and cortex, with higher apoE levels in mice irradiated at 42 cGy than 168 cGy and a trend towards higher apoE levels in mice irradiated at 21 than 168 cGy. In addition, in the hippocampus, there was a trend towards a negative correlation between MAP-2 and apoE levels. While reduced levels of MAP-2 in the cortex might have contributed to the altered performance in the passive avoidance test, it does not seem sufficient to do so. The higher hippocampal and cortical apoE levels in mice irradiated at 42 than 168 cGy might have served as a compensatory protective response preserving their passive avoidance memory. Thus, there were no alterations in behavioral performance in the open filed or depressive-like behavior in the forced swim test, while cognitive impairments were seen in the object recognition and passive avoidance tests, but not in the contextual or cued fear conditioning tests. Taken together, the results indicate that some aspects of cognitive performance are altered in male mice exposed to ⁴He ions, but that the response is task-dependent. Furthermore, the sensitive doses can vary within each task in a non-linear fashion. This highlights the importance of assessing the cognitive and behavioral effects of charged particle exposure with a variety of assays and at multiple doses, given the possibility that lower doses may be more damaging due to the absence of induced compensatory mechanisms at higher doses.
Collapse
|
28
|
Ding X, Wu HH, Ji SJ, Cai S, Dai PW, Xu ML, Zhang JJ, Zhang QX, Tian Y, Ma QH. The p75 neurotrophin receptor regulates cranial irradiation-induced hippocampus-dependent cognitive dysfunction. Oncotarget 2018; 8:40544-40557. [PMID: 28380447 PMCID: PMC5522261 DOI: 10.18632/oncotarget.16492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/22/2017] [Indexed: 11/28/2022] Open
Abstract
Cognitive deficits, characterized by progressive problems with hippocampus-dependent learning, memory and spatial processing, are the most serious complication of cranial irradiation. However, the underlying mechanisms remain obscure. The p75 neurotrophin receptor (p75NTR) is involved in a diverse arrays of cellular responses, including neurite outgrowth, neurogenesis, and negative regulation of spine density, which are associated with various neurological disorders. In this study, male Sprague-Dawley (SD) rats received 10 Gy cranial irradiation. Then, we evaluated the expression of p75NTR in the hippocampus after cranial irradiation and explored its potential role in radiation-induced synaptic dysfunction and memory deficits. We found that the expression of p75NTR was significantly increased in the irradiated rat hippocampus. Knockdown of p75NTR by intrahippocampal infusion of AAV8-shp75 ameliorated dendritic spine abnormalities, and restored synapse-related protein levels, thus preventing memory deficits, likely through normalization the phosphor-AKT activity. Moreover, viral-mediated overexpression of p75NTR in the normal hippocampus reproduced learning and memory deficits. Overall, this study demonstrates that p75NTR is an important mediator of irradiation-induced cognitive deficits by regulating dendritic development and synapse structure.
Collapse
Affiliation(s)
- Xin Ding
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Hao-Hao Wu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Sheng-Jun Ji
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Radiotherapy and Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Shang Cai
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Pei-Wen Dai
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Mei-Ling Xu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Jun-Jun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Qi-Xian Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Ye Tian
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
29
|
Chen BH, Park JH, Lee TK, Song M, Kim H, Lee JC, Kim YM, Lee CH, Hwang IK, Kang IJ, Yan BC, Won MH, Ahn JH. Melatonin attenuates scopolamine-induced cognitive impairment via protecting against demyelination through BDNF-TrkB signaling in the mouse dentate gyrus. Chem Biol Interact 2018; 285:8-13. [DOI: 10.1016/j.cbi.2018.02.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/07/2018] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
|
30
|
Ji S, Ding X, Ji J, Wu H, Sun R, Li X, Zhang L, Tian Y. Cranial irradiation inhibits hippocampal neurogenesis via DNMT1 and DNMT3A. Oncol Lett 2018; 15:2899-2904. [PMID: 29435016 PMCID: PMC5778827 DOI: 10.3892/ol.2017.7643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
Impairment of neurogenesis in the hippocampus following whole-brain irradiation is the most important mechanism of radiation-induced cognitive dysfunction. However, the underlying mechanism remains obscure, meaning an ideal therapeutic target has not been identified. Evidence indicates that DNA methylation in neurons regulates synaptic plasticity and neuronal network activity. In the present study, the expression of DNA methyltransferases (DNMTs) in the hippocampus was analyzed to investigate their potential function in radiation-induced neurogenesis impairment. Sprague-Dawley rats were used throughout the present study, apportioned to the following groups: Control, radiation only, zebularine (a DNMT inhibitor) only, and radiation and zebularine together. Immunofluorescence staining revealed that radiation inhibited cellular proliferation and dendritic growth within new neurons of the hippocampus. In addition, western blot analysis demonstrated lower expression levels of DNMT1 and DNMT3A protein following radiation treatment compared with that in the non-irradiated control. Furthermore, compared with the radiation-only group, the radiation and zebularine group had significantly lower cell proliferative abilities, dendritic growth, and DNMT1 and DNMT3A protein levels. The results of the present study indicated that DNMT1 and DNMT3A may be involved in the pathogenesis of whole-brain radiation-induced neurogenesis impairment.
Collapse
Affiliation(s)
- Shengjun Ji
- Cancer Center, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215001, P.R. China
| | - Xin Ding
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jiang Ji
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Haohao Wu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Rui Sun
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaoyang Li
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Liyuan Zhang
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
31
|
Consales C, Cirotti C, Filomeni G, Panatta M, Butera A, Merla C, Lopresto V, Pinto R, Marino C, Benassi B. Fifty-Hertz Magnetic Field Affects the Epigenetic Modulation of the miR-34b/c in Neuronal Cells. Mol Neurobiol 2017; 55:5698-5714. [PMID: 29039021 DOI: 10.1007/s12035-017-0791-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/24/2022]
Abstract
The exposure to extremely low-frequency magnetic fields (ELF-MFs) has been associated to increased risk of neurodegenerative diseases, although the underlying molecular mechanisms are still undefined. Since epigenetic modulation has been recently encountered among the key events leading to neuronal degeneration, we here aimed at assessing if the control of gene expression mediated by miRNAs, namely miRs-34, has any roles in driving neuronal cell response to 50-Hz (1 mT) magnetic field in vitro. We demonstrate that ELF-MFs drive an early reduction of the expression level of miR-34b and miR-34c in SH-SY5Y human neuroblastoma cells, as well as in mouse primary cortical neurons, by affecting the transcription of the common pri-miR-34. This modulation is not p53 dependent, but attributable to the hyper-methylation of the CpG island mapping within the miR-34b/c promoter. Incubation with N-acetyl-l-cysteine or glutathione ethyl-ester fails to restore miR-34b/c expression, suggesting that miRs-34 are not responsive to ELF-MF-induced oxidative stress. By contrast, we show that miRs-34 control reactive oxygen species production and affect mitochondrial oxidative stress triggered by ELF-MFs, likely by modulating mitochondria-related miR-34 targets identified by in silico analysis. We finally demonstrate that ELF-MFs alter the expression of the α-synuclein, which is specifically stimulated upon ELF-MFs exposure via both direct miR-34 targeting and oxidative stress. Altogether, our data highlight the potential of the ELF-MFs to tune redox homeostasis and epigenetic control of gene expression in vitro and shed light on the possible mechanism(s) producing detrimental effects and predisposing neurons to degeneration.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| | - Claudia Cirotti
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.,Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Martina Panatta
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.,Vectorology and Anticancer Therapies, UMR 8203, CNRS, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
32
|
Kang S, Son Y, Lee S, Kim J, Kim JC, Kim JS, Jung U, Kim SH, Yang M, Moon C. Changes in epigenetic markers, DNMT1 and HDAC1/2, in the adult mouse hippocampus after cranial irradiation. Neurosci Lett 2017; 657:113-119. [DOI: 10.1016/j.neulet.2017.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 07/25/2017] [Accepted: 08/01/2017] [Indexed: 01/30/2023]
|
33
|
CNS bioavailability and radiation protection of normal hippocampal neurogenesis by a lipophilic Mn porphyrin-based superoxide dismutase mimic, MnTnBuOE-2-PyP 5. Redox Biol 2017; 12:864-871. [PMID: 28454069 PMCID: PMC5407575 DOI: 10.1016/j.redox.2017.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
Although radiation therapy can be effective against cancer, potential damage to normal tissues limits the amount that can be safely administered. In central nervous system (CNS), radiation damage to normal tissues is presented, in part, as suppressed hippocampal neurogenesis and impaired cognitive functions. Mn porphyrin (MnP)-based redox active drugs have demonstrated differential effects on cancer and normal tissues in experimental animals that lead to protection of normal tissues and radio- and chemo-sensitization of cancers. To test the efficacy of MnPs in CNS radioprotection, we first examined the tissue levels of three different MnPs – MnTE-2-PyP5+(MnE), MnTnHex-2-PyP5+(MnHex), and MnTnBuOE-2-PyP5+(MnBuOE). Nanomolar concentrations of MnHex and MnBuOE were detected in various brain regions after daily subcutaneous administration, and MnBuOE was well tolerated at a daily dose of 3 mg/kg. Administration of MnBuOE for one week before cranial irradiation and continued for one week afterwards supported production and long-term survival of newborn neurons in the hippocampal dentate gyrus. MnP-driven S-glutathionylation in cortex and hippocampus showed differential responses to MnP administration and radiation in these two brain regions. A better understanding of how preserved hippocampal neurogenesis correlates with cognitive functions following cranial irradiation will be helpful in designing better MnP-based radioprotection strategies. Bioavailability of MnPs in individual brain regions were determined by LC-MS/MS. CNS MnBuOE and MnHex levels were between 15 and 160 nM after daily administration. MnBuOE administration ameliorated radiation effects on hippocampal neurogenesis. MnBuOE preserved categories E&F Dcx+ neurons after cranial irradiation. MnBuOE and irradiation lead to changes in protein S-glutathionylation in the CNS.
Collapse
|
34
|
Identification of molecular characteristics induced by radiotherapy in rectal cancer based on microarray data. Oncol Lett 2017; 13:2777-2783. [PMID: 28454466 DOI: 10.3892/ol.2017.5750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/06/2016] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to reveal the molecular characteristics induced by radiotherapy in rectal cancer at the transcriptome level. Microarray data (ID, GSE26027) downloaded from the Gene Expression Omnibus database were re-analyzed to identify differentially expressed genes (DEGs) between rectal cancer tissues during and prior to radiotherapy. The DEGs were then inputted into the database for annotation, visualization and integrated discovery, an online tool to perform enrichment analyses, and into the search tool for the retrieval of interacting genes/proteins database to identify protein-protein interactions (PPIs). Subsequently, a PPI network was constructed, which was screened for densely connected modules. Furthermore, protein domain enrichment analysis was performed. In total, 690 DEGs, including 179 upregulated and 511 downregulated DEGs, were found in rectal cancer tissues during and prior to radiotherapy. The upregulated DEGs were significantly enriched in 'positive regulation of transport' and 'regulation of cardiac muscle contraction', while the downregulated DEGs were most markedly enriched in 'cell migration', 'cell-cell signaling', 'extracellular matrix organization' and 'blood vessel development', including prostaglandin-endoperoxide synthase 2, transforming growth factor β-induced, 68 kDa endothelin receptor type A, brain-derived neurotrophic factor, TIMP metallopeptidase inhibitor 1, and serpin family E member 1, which were the top 6 hub nodes in the PPI network. Furthermore, 2 protein domains were significantly enriched by PPI modules, including: The collagen triple helix repeat (CTHR) family members collagen type (COL) 5A2, COL9A3, COL6A3, COL21A1, COL5A3, COL11A1, COL7A1 and CTHR-containing-1; and the olfactory receptor family (OR) members OR7E24, OR7A17, OR6A2, OR1F1, OR10H3 and OR7A10. A total of 7 upregulated DEGs were characterized as tumor suppressor genes, and 8 downregulated DEGs were characterized as oncogenes. The biological processes or protein domains enriched by upregulated or downregulated DEGs may improve the understanding of molecular characteristics in response to radiotherapy.
Collapse
|
35
|
Correa DD, Satagopan J, Cheung K, Arora AK, Kryza-Lacombe M, Xu Y, Karimi S, Lyo J, DeAngelis LM, Orlow I. COMT, BDNF, and DTNBP1 polymorphisms and cognitive functions in patients with brain tumors. Neuro Oncol 2016; 18:1425-33. [PMID: 27091610 PMCID: PMC5035520 DOI: 10.1093/neuonc/now057] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/11/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Cognitive dysfunction is common among patients with brain tumors and can be associated with the disease and treatment with radiotherapy and chemotherapy. However, little is known about genetic risk factors that may moderate the vulnerability for developing cognitive dysfunction. In this study, we examined the association of single nucleotide polymorphisms (SNPs) in the catechol-O-methyl transferase (COMT), brain-derived neurotrophic factor (BDNF), and dystrobrevin-binding protein 1 (DTNBP1) genes with cognitive functions and neuroimaging outcomes in patients with brain tumors. METHODS One hundred and fifty patients with brain tumors completed neuropsychological tests of attention, executive functions, and memory and were genotyped for polymorphisms in the COMT, BDNF, and DTNBP1 genes. Ratings of white matter (WM) abnormalities on magnetic resonance imaging scans were performed. RESULTS Multivariate regression shrinkage analyses, adjusted for age, education, treatment type, time since treatment completion, and tumor location, indicated a significant association between the COMT SNP rs4680 (Val158Met) and memory with lower scores in delayed recall (P < .01) among homozygotes (valine/valine). Additional COMT, BDNF and DTNBP1 SNPs were significantly associated with attention, executive functions, and memory scores. CONCLUSION This is the first study to suggest that known and newly described polymorphisms in genes associated with executive and memory functions in healthy individuals and other clinical populations may modulate cognitive outcome in patients with brain tumors.
Collapse
Affiliation(s)
- Denise D Correa
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Jaya Satagopan
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Kenneth Cheung
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Arshi K Arora
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Maria Kryza-Lacombe
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Youming Xu
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Sasan Karimi
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - John Lyo
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| | - Irene Orlow
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (D.D.C., M.K.-L., L.M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (J.S., K.C., A.K.A., Y.X., I.O.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (S.K., J.L.); Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York (D.D.C., L.M.D.)
| |
Collapse
|
36
|
Betlazar C, Middleton RJ, Banati RB, Liu GJ. The impact of high and low dose ionising radiation on the central nervous system. Redox Biol 2016; 9:144-156. [PMID: 27544883 PMCID: PMC4993858 DOI: 10.1016/j.redox.2016.08.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Responses of the central nervous system (CNS) to stressors and injuries, such as ionising radiation, are modulated by the concomitant responses of the brains innate immune effector cells, microglia. Exposure to high doses of ionising radiation in brain tissue leads to the expression and release of biochemical mediators of ‘neuroinflammation’, such as pro-inflammatory cytokines and reactive oxygen species (ROS), leading to tissue destruction. Contrastingly, low dose ionising radiation may reduce vulnerability to subsequent exposure of ionising radiation, largely through the stimulation of adaptive responses, such as antioxidant defences. These disparate responses may be reflective of non-linear differential microglial activation at low and high doses, manifesting as an anti-inflammatory or pro-inflammatory functional state. Biomarkers of pathology in the brain, such as the mitochondrial Translocator Protein 18 kDa (TSPO), have facilitated in vivo characterisation of microglial activation and ‘neuroinflammation’ in many pathological states of the CNS, though the exact function of TSPO in these responses remains elusive. Based on the known responsiveness of TSPO expression to a wide range of noxious stimuli, we discuss TSPO as a potential biomarker of radiation-induced effects. Ionising radiation can modulate responses of microglial cells in the CNS. High doses can induce ROS formation, oxidative stress and neuroinflammation. Low doses can mitigate tissue damage via antioxidant defences. TSPO as a potential biomarker and modulator of radiation induced effects in the CNS. Non-linear differential microglial activation to high and low doses is proposed.
Collapse
Affiliation(s)
- Calina Betlazar
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia
| | - Ryan J Middleton
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - Richard B Banati
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia.
| | - Guo-Jun Liu
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia.
| |
Collapse
|
37
|
Hayase T. Putative Epigenetic Involvement of the Endocannabinoid System in Anxiety- and Depression-Related Behaviors Caused by Nicotine as a Stressor. PLoS One 2016; 11:e0158950. [PMID: 27404492 PMCID: PMC4942073 DOI: 10.1371/journal.pone.0158950] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 06/26/2016] [Indexed: 01/01/2023] Open
Abstract
Like various stressors, the addictive use of nicotine (NC) is associated with emotional symptoms such as anxiety and depression, although the underlying mechanisms have not yet been fully elucidated due to the complicated involvement of target neurotransmitter systems. In the elicitation of these emotional symptoms, the fundamental involvement of epigenetic mechanisms such as histone acetylation has recently been suggested. Furthermore, among the interacting neurotransmitter systems implicated in the effects of NC and stressors, the endocannabinoid (ECB) system is considered to contribute indispensably to anxiety and depression. In the present study, the epigenetic involvement of histone acetylation induced by histone deacetylase (HDAC) inhibitors was investigated in anxiety- and depression-related behavioral alterations caused by NC and/or immobilization stress (IM). Moreover, based on the contributing roles of the ECB system, the interacting influence of ECB ligands on the effects of HDAC inhibitors was evaluated in order to examine epigenetic therapeutic interventions. Anxiety-like (elevated plus-maze test) and depression-like (forced swimming test) behaviors, which were observed in mice treated with repeated (4 days) NC (subcutaneous 0.8 mg/kg) and/or IM (10 min), were blocked by the HDAC inhibitors sodium butyrate (SB) and valproic acid (VA). The cannabinoid type 1 (CB1) agonist ACPA (arachidonylcyclopropylamide; AC) also antagonized these behaviors. Conversely, the CB1 antagonist SR 141716A (SR), which counteracted the effects of AC, attenuated the anxiolytic-like effects of the HDAC inhibitors commonly in the NC and/or IM groups. SR also attenuated the antidepressant-like effects of the HDAC inhibitors, most notably in the IM group. From these results, the combined involvement of histone acetylation and ECB system was shown in anxiety- and depression-related behaviors. In the NC treatment groups, the limited influence of SR against the HDAC inhibitor-induced antidepressant-like effects may reflect the characteristic involvement of histone acetylation within the NC-related neurotransmitter systems other than the ECB system.
Collapse
Affiliation(s)
- Tamaki Hayase
- Department of Legal Medicine, Kyoto University, Kyoto 606–8501, Japan
- * E-mail:
| |
Collapse
|
38
|
Sun R, Zhang LY, Chen LS, Tian Y. Long-term outcome of changes in cognitive function of young rats after various/different doses of whole brain irradiation. Neurol Res 2016; 38:647-54. [PMID: 27238733 DOI: 10.1080/01616412.2016.1188483] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To characterize the early delayed and late-delayed cognitive dysfunction induced by various doses of whole brain irradiation in young rats. METHODS One-month-old Sprague-Dawley male rats were divided randomly into the 0 (control), 0 (anesthesia control), 2, 10, 20, and 30-Gy groups. Each group was then subdivided into 4 groups according to the experimental intervals: 1, 2, 3, and 6 months after radiation. Rats were irradiated using a 4-MeV electron beam, which was generated by a linear accelerator. Sequential behavioral tests, including open field, novel location and novel object recognition and Morris water maze were performed after radiation. Changes in gross neurological symptoms, body weight, topical skin response, and histopathology were observed. RESULTS In the open field test, there were no radiation-induced alterations found. In the novel location and novel object recognition tests, rats of the 20-Gy group spent less time exploring the novel object and novel location 3 months after irradiation. During the place navigation test, the spatial working memory of the 30 and 20-Gy irradiated rats were impaired from 1 to 2 months after irradiation, respectively. In the spatial probe test, the 20 and 30-Gy irradiated rats spent less time in the critical region compared to control rats at 3 and 6 months post-irradiation. Morphological changes, including edema, vascular dilation, focal necrosis, demyelination, and adjacent reactive gliosis were observed in the 30-Gy irradiation group. CONCLUSION More than 20 Gy of whole brain irradiation dose can cause significant cognitive dysfunction in young rats.
Collapse
Affiliation(s)
- Rui Sun
- a Department of Radiotherapy & Oncology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Li-Yuan Zhang
- a Department of Radiotherapy & Oncology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Lie-Song Chen
- a Department of Radiotherapy & Oncology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Ye Tian
- a Department of Radiotherapy & Oncology , The Second Affiliated Hospital of Soochow University , Suzhou , China.,b Institute of Radiotherapy & Oncology , Soochow University , Suzhou , China.,c Suzhou Key Laboratory for Radiation Oncology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| |
Collapse
|
39
|
Yang L, Yang J, Li G, Li Y, Wu R, Cheng J, Tang Y. Pathophysiological Responses in Rat and Mouse Models of Radiation-Induced Brain Injury. Mol Neurobiol 2016; 54:1022-1032. [PMID: 26797684 PMCID: PMC5310567 DOI: 10.1007/s12035-015-9628-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022]
Abstract
The brain is the major dose-limiting organ in patients undergoing radiotherapy for assorted conditions. Radiation-induced brain injury is common and mainly occurs in patients receiving radiotherapy for malignant head and neck tumors, arteriovenous malformations, or lung cancer-derived brain metastases. Nevertheless, the underlying mechanisms of radiation-induced brain injury are largely unknown. Although many treatment strategies are employed for affected individuals, the effects remain suboptimal. Accordingly, animal models are extremely important for elucidating pathogenic radiation-associated mechanisms and for developing more efficacious therapies. So far, models employing various animal species with different radiation dosages and fractions have been introduced to investigate the prevention, mechanisms, early detection, and management of radiation-induced brain injury. However, these models all have limitations, and none are widely accepted. This review summarizes the animal models currently set forth for studies of radiation-induced brain injury, especially rat and mouse, as well as radiation dosages, dose fractionation, and secondary pathophysiological responses.
Collapse
Affiliation(s)
- Lianhong Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jianhua Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China
| | - Guoqian Li
- Department of Neurology, Fujian Provincical Quanzhou First Hospital, Quanzhou, Fujian Province, China
| | - Yi Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China
| | - Rong Wu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China. .,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, 510120, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
40
|
Cacan E, Greer SF, Garnett-Benson C. Radiation-induced modulation of immunogenic genes in tumor cells is regulated by both histone deacetylases and DNA methyltransferases. Int J Oncol 2015; 47:2264-75. [PMID: 26458736 DOI: 10.3892/ijo.2015.3192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/16/2015] [Indexed: 11/06/2022] Open
Abstract
Radiation treatment is a pivotal therapy for several cancer types, including colorectal cancer. It has been shown that sublethal doses of radiation modulate gene expression, making tumor cells more susceptible to T-cell-mediated immune attack. We have recently shown that low dose radiation enhances expression of multiple death receptors (Fas, DR4 and DR5) and co-stimulatory molecules (4-1BBL and OX-40L) in colorectal cancer (CRC) cells; however, it is unclear how ionizing radiation (IR) enhances expression of these molecules mechanistically. In the present study, we elucidate the molecular mechanisms by which radiation controls expression of these molecules in CRC. Here we report that, enhanced expression of these genes following radiation treatment of CRC cells is due, in part, to changes in DNA methylation and histone acetylation. We observed that radiation (5 Gy) significantly increased histone acetylation at the promoter regions of 4-1BBL, Fas and DR5 but not OX-40L. However, radiation did not induce changes in the global levels of acetylated histone H3 suggesting specificity of IR-induced changes. Furthermore, evaluation of epigenetic controlling enzymes revealed that IR did not alter overall cellular levels of HDACs (HDAC1, HDAC2 or HDAC3) or DNMTs (DNMT1, DNMT3a, or DNMT3b). Instead, radiation decreased binding of HDAC2 and HDAC3 at the promoter regions of Fas and 4-1BBL, respectively. Radiation also resulted in reduced DNMT1 at both the Fas and 4-1BBL promoter regions but not a control gene. We conclude that single dose radiation can influence the expression of immune response relevant genes in colorectal tumor cells by altering the binding of epigenetic enzymes, and modulating histone acetylation, at specific gene promoters.
Collapse
Affiliation(s)
- Ercan Cacan
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Susanna F Greer
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | |
Collapse
|