1
|
Strohm AO, Oldfield S, Hernady E, Johnston CJ, Marples B, O'Banion MK, Majewska AK. Biological sex, microglial signaling pathways, and radiation exposure shape cortical proteomic profiles and behavior in mice. Brain Behav Immun Health 2025; 43:100911. [PMID: 39677060 PMCID: PMC11634995 DOI: 10.1016/j.bbih.2024.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Patients receiving cranial radiation therapy experience tissue damage and cognitive deficits that severely decrease their quality of life. Experiments in rodent models show that these adverse neurological effects are in part due to functional changes in microglia, the resident immune cells of the central nervous system. Increasing evidence suggests that experimental manipulation of microglial signaling can regulate radiation-induced changes in the brain and behavior. Furthermore, many studies show sex-dependent neurological effects of radiation exposure. Despite this, few studies have used both males and females to explore how sex and microglial function interact to influence radiation effects on the brain. Here, we used a system levels approach to examine how deficiencies in purinergic and fractalkine signaling, two important microglial signaling pathways, impact brain proteomic and behavioral profiles in irradiated and control male and female mice. We performed a comprehensive analysis of the cortical proteomes from irradiated and control C57BL/6J, P2Y12-/-, and CX3CR1-/- mice of both sexes using multiple bioinformatics methods. We identified distinct proteins and biological processes, as well as behavioral profiles, regulated by sex, genotype, radiation exposure, and their interactions. Disrupting microglial signaling, had the greatest impact on proteomic expression, with CX3CR1-/- mice showing the most distinct proteomic profile characterized by upregulation of CX3CL1. Surprisingly, radiation exposure caused relatively smaller proteomic changes in glial and synaptic proteins, including Rgs10, Crybb1, C1qa, and Hexb. While we observed some radiation effects on locomotor behavior, biological sex as well as loss of P2Y12 and CX3CR1 signaling had a stronger influence on locomotor outcomes in our model. Lastly, loss of P2Y12 and CX3CR1 strongly regulated exploratory behaviors. Overall, our findings provide novel insights into the molecular pathways and proteins that are linked to P2Y12 and CX3CR1 signaling, biological sex, radiation exposure, and their interactions.
Collapse
Affiliation(s)
- Alexandra O. Strohm
- Departments of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Sadie Oldfield
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Eric Hernady
- Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Carl J. Johnston
- Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian Marples
- Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M. Kerry O'Banion
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ania K. Majewska
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
2
|
Georgiou CJ, Brown MK, Cai Z, Alshafai L, Gao A, Rutka JT, Winnik MA, Reilly RM. Convection-enhanced delivery of [ 177Lu]Lu-labeled gold nanoparticles combined with anti-PD1 checkpoint immunotherapy improves the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors. Nucl Med Biol 2025; 140-141:108970. [PMID: 39571483 DOI: 10.1016/j.nucmedbio.2024.108970] [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: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Our objective was to study convection enhanced delivery (CED) of 177Lu-labeled metal chelating polymer (MCP) conjugated to gold nanoparticles ([177Lu]Lu-MCP-AuNP) alone or combined with anti-PD1 immune checkpoint inhibition (ICI) for improving the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors. METHODS C57BL/6J mice with GL261 tumors were treated with [177Lu]Lu-MCP-AuNP (0.8 or 2.7 MBq; 4 × 1011 AuNP) alone or combined with anti-PD1 antibodies (200 μg i.p. every 2 d × 3 doses). Control mice received normal saline, non-radioactive MCP-AuNP or anti-PD1 antibodies. Kaplan-Meier median survival was estimated. T-cell infiltration into the brain was probed by flow cytometry. Toxicity was assessed by monitoring body weight and cognitive function tests [Object Location Test (OLT) and Novel Object Recognition Test (NORT)] and T2-weighted MRI of the brain, overall health and ex vivo histopathological examination of the brain. RESULTS Treatment with [177Lu]Lu-MCP-AuNP (0.8 MBq) significantly increased median survival compared to MCP-AuNP (29 vs. 25 d; P = 0.007) or normal saline-treated mice (24 d; P < 0.001). Combining [177Lu]Lu-MCP-AuNP (0.8 MBq) with anti-PD1 antibodies increased median survival to 32 d (P < 0.0001 vs. normal saline). Increasing the mean amount of [177Lu]Lu-MCP-AuNP to 2.7 MBq and combining with anti-PD1 antibodies extended survival to at least 218 d in 5/9 mice. Increased CD8+ cytotoxic T-cells and decreased CD4+ helper T-cells were found in the brain vs. normal saline-treated mice. No weight loss (>20 %) was observed for treated or control mice. There was no change in cognitive function in mice treated with [177Lu]Lu-MCP-AuNP (0.8 MBq) alone or combined with anti-PD1 antibodies assessed by the OLT or NORT. T2-weighted MRI in mice treated with 2.7 MBq [177Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies revealed edema, gliosis and ex vacuo dilatation of the ventricle proximal to the site of infusion. Histopathological examination of the brain revealed dilatation of the ventricle and gliosis proximal to the site of infusion but no radiation necrosis. MRI and histological analysis did not reveal tumor in the brain of these mice. Mice treated with 2.7 MBq [177Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies did not demonstrate overall deleterious health effects. CONCLUSIONS We conclude that CED of [177Lu]Lu-MCP-AuNP combined with anti-PD1 checkpoint immunotherapy improved the survival of immunocompetent C67BL/6J mice with GL261 glioma tumors in the brain. Higher administered amounts of [177Lu]Lu-MCP-AuNP (2.7 MBq vs. 0.8 MBq) were most effective and yielded long-term survival. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This study demonstrates that combining a locally-infused radiation nanomedicine, [177Lu]Lu-MCP-AuNP and anti-PD1 checkpoint immunotherapy improved the survival of mice with glioma tumors in the brain. In the future, this treatment may be useful to treat residual tumor at the surgical margins in patients with GBM to prevent local recurrence and improve survival.
Collapse
Affiliation(s)
| | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Laila Alshafai
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Joint Department of Medical Imaging, Division of Neuroradiology, Mount Sinai Hospital and University Health Network, Toronto, ON, Canada
| | - Andrew Gao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - James T Rutka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
3
|
Frechou MA, Martin SS, McDermott KD, Huaman EA, Gökhan Ş, Tomé WA, Coen-Cagli R, Gonçalves JT. Adult neurogenesis improves spatial information encoding in the mouse hippocampus. Nat Commun 2024; 15:6410. [PMID: 39080283 PMCID: PMC11289285 DOI: 10.1038/s41467-024-50699-x] [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: 12/15/2022] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Adult neurogenesis is a unique form of neuronal plasticity in which newly generated neurons are integrated into the adult dentate gyrus in a process that is modulated by environmental stimuli. Adult-born neurons can contribute to spatial memory, but it is unknown whether they alter neural representations of space in the hippocampus. Using in vivo two-photon calcium imaging, we find that male and female mice previously housed in an enriched environment, which triggers an increase in neurogenesis, have increased spatial information encoding in the dentate gyrus. Ablating adult neurogenesis blocks the effect of enrichment and lowers spatial information, as does the chemogenetic silencing of adult-born neurons. Both ablating neurogenesis and silencing adult-born neurons decreases the calcium activity of dentate gyrus neurons, resulting in a decreased amplitude of place-specific responses. These findings are in contrast with previous studies that suggested a predominantly inhibitory action for adult-born neurons. We propose that adult neurogenesis improves representations of space by increasing the gain of dentate gyrus neurons and thereby improving their ability to tune to spatial features. This mechanism may mediate the beneficial effects of environmental enrichment on spatial learning and memory.
Collapse
Affiliation(s)
- M Agustina Frechou
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Sunaina S Martin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychology, University of California San Diego, La Jolla, CA, USA
| | - Kelsey D McDermott
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evan A Huaman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wolfgang A Tomé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ruben Coen-Cagli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
Seitzman BA, Reynoso FJ, Mitchell TJ, Bice AR, Jarang A, Wang X, Mpoy C, Strong L, Rogers BE, Yuede CM, Rubin JB, Perkins SM, Bauer AQ. Functional network disorganization and cognitive decline following fractionated whole-brain radiation in mice. GeroScience 2024; 46:543-562. [PMID: 37749370 PMCID: PMC10828348 DOI: 10.1007/s11357-023-00944-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: 08/18/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Cognitive dysfunction following radiotherapy (RT) is one of the most common complications associated with RT delivered to the brain, but the precise mechanisms behind this dysfunction are not well understood, and to date, there are no preventative measures or effective treatments. To improve patient outcomes, a better understanding of the effects of radiation on the brain's functional systems is required. Functional magnetic resonance imaging (fMRI) has shown promise in this regard, however, compared to neural activity, hemodynamic measures of brain function are slow and indirect. Understanding how RT acutely and chronically affects functional brain organization requires more direct examination of temporally evolving neural dynamics as they relate to cerebral hemodynamics for bridging with human studies. In order to adequately study the underlying mechanisms of RT-induced cognitive dysfunction, the development of clinically mimetic RT protocols in animal models is needed. To address these challenges, we developed a fractionated whole-brain RT protocol (3Gy/day for 10 days) and applied longitudinal wide field optical imaging (WFOI) of neural and hemodynamic brain activity at 1, 2, and 3 months post RT. At each time point, mice were subject to repeated behavioral testing across a variety of sensorimotor and cognitive domains. Disruptions in cortical neuronal and hemodynamic activity observed 1 month post RT were significantly worsened by 3 months. While broad changes were observed in functional brain organization post RT, brain regions most impacted by RT occurred within those overlapping with the mouse default mode network and other association areas similar to prior reports in human subjects. Further, significant cognitive deficits were observed following tests of novel object investigation and responses to auditory and contextual cues after fear conditioning. Our results fill a much-needed gap in understanding the effects of whole-brain RT on systems level brain organization and how RT affects neuronal versus hemodynamic signaling in the cortex. Having established a clinically-relevant injury model, future studies can examine therapeutic interventions designed to reduce neuroinflammation-based injury following RT. Given the overlap of sequelae that occur following RT with and without chemotherapy, these tools can also be easily incorporated to examine chemotherapy-related cognitive impairment.
Collapse
Affiliation(s)
- Benjamin A Seitzman
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Francisco J Reynoso
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Timothy J Mitchell
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Annie R Bice
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, 660 S. Euclid Ave, Campus Box 8225, St. Louis, MO, 63110, USA
| | - Anmol Jarang
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, 660 S. Euclid Ave, Campus Box 8225, St. Louis, MO, 63110, USA
| | - Xiaodan Wang
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, 660 S. Euclid Ave, Campus Box 8225, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Lori Strong
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Buck E Rogers
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Carla M Yuede
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Stephanie M Perkins
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA.
| | - Adam Q Bauer
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, 660 S. Euclid Ave, Campus Box 8225, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
5
|
Castle J, Shaw G, Weller D, Fielder E, Egnuni T, Singh M, Skinner R, von Zglinicki T, Clifford SC, Short SC, Miwa S, Hicks D. In vivo modeling recapitulates radiotherapy delivery and late-effect profile for childhood medulloblastoma. Neurooncol Adv 2024; 6:vdae091. [PMID: 38946880 PMCID: PMC11212071 DOI: 10.1093/noajnl/vdae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Background Medulloblastoma (MB) is the most common malignant pediatric brain tumor, with 5-year survival rates > 70%. Cranial radiotherapy (CRT) to the whole brain, with posterior fossa boost (PFB), underpins treatment for non-infants; however, radiotherapeutic insult to the normal brain has deleterious consequences to neurocognitive and physical functioning, and causes accelerated aging/frailty. Approaches to ameliorate radiotherapy-induced late-effects are lacking and a paucity of appropriate model systems hinders their development. Methods We have developed a clinically relevant in vivo model system that recapitulates the radiotherapy dose, targeting, and developmental stage of childhood medulloblastoma. Consistent with human regimens, age-equivalent (postnatal days 35-37) male C57Bl/6J mice received computerized tomography image-guided CRT (human-equivalent 37.5 Gy EQD2, n = 12) ± PFB (human-equivalent 48.7 Gy EQD2, n = 12), via the small animal radiation research platform and were longitudinally assessed for > 12 months. Results CRT was well tolerated, independent of PFB receipt. Compared to a sham-irradiated group (n = 12), irradiated mice were significantly frailer following irradiation (frailty index; P = .0002) and had reduced physical functioning; time to fall from a rotating rod (rotarod; P = .026) and grip strength (P = .006) were significantly lower. Neurocognitive deficits were consistent with childhood MB survivors; irradiated mice displayed significantly worse working memory (Y-maze; P = .009) and exhibited spatial memory deficits (Barnes maze; P = .029). Receipt of PFB did not induce a more severe late-effect profile. Conclusions Our in vivo model mirrored childhood MB radiotherapy and recapitulated features observed in the late-effect profile of MB survivors. Our clinically relevant model will facilitate both the elucidation of novel/target mechanisms underpinning MB late effects and the development of novel interventions for their amelioration.
Collapse
Affiliation(s)
- Jemma Castle
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Gary Shaw
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Dominic Weller
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Teklu Egnuni
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Mankaran Singh
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Roderick Skinner
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Thomas von Zglinicki
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Susan C Short
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Satomi Miwa
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Debbie Hicks
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
6
|
Sorokina SS, Malkov AE, Rozanova OM, Smirnova EN, Shemyakov AE. Behavioral performance and microglial status in mice after moderate dose of proton irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023; 62:497-509. [PMID: 37794305 DOI: 10.1007/s00411-023-01044-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
Cognitive impairment is a remote effect of gamma radiation treatment of malignancies. The major part of the studies on the effect of proton irradiation (a promising alternative in the treatment of radio-resistant tumors and tumors located close to critical organs) on the cognitive abilities of laboratory animals and their relation to morphological changes in the brain is rather contradictory. The aim of this study was to investigate cognitive functions and the dynamics of changes in morphological parameters of hippocampal microglial cells after 7.5 Gy of proton irradiation. Two months after the cranial irradiation, 8- to 9-week-old male SHK mice were tested for total activity, spatial learning, as well as long- and short-term hippocampus-dependent memory. To estimate the morphological parameters of microglia, brain slices of control and irradiated animals each with different time after proton irradiation (24 h, 7 days, 1 month) were stained for microglial marker Iba-1. No changes in behavior or deficits in short-term and long-term hippocampus-dependent memory were found, but an impairment of episodic memory was observed. A change in the morphology of hippocampal microglial cells, which is characteristic of the transition of cells to an activated state, was detected. One day after proton exposure in the brain tissue, a slight decrease in cell density was observed, which was restored to the control level by the 30th day after treatment. The results obtained may be promising with regard to the future use of using high doses of protons per fraction in the irradiation of tumors.
Collapse
Affiliation(s)
- S S Sorokina
- Laboratory of Isotope Investigations, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia.
| | - A E Malkov
- Laboratory of Neurons Systematic Organization, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - O M Rozanova
- Laboratory of Cell Engineering, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - E N Smirnova
- Laboratory of Cell Engineering, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - A E Shemyakov
- Theranostics and Nuclear Medicine Laboratory, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| |
Collapse
|
7
|
Extracellular Vesicles and Cancer Therapy: Insights into the Role of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11061194. [PMID: 35740091 PMCID: PMC9228181 DOI: 10.3390/antiox11061194] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress plays a significant role in cancer development and cancer therapy, and is a major contributor to normal tissue injury. The unique characteristics of extracellular vesicles (EVs) have made them potentially useful as a diagnostic tool in that their molecular content indicates their cell of origin and their lipid membrane protects the content from enzymatic degradation. In addition to their possible use as a diagnostic tool, their role in how normal and diseased cells communicate is of high research interest. The most exciting area is the association of EVs, oxidative stress, and pathogenesis of numerous diseases. However, the relationship between oxidative stress and oxidative modifications of EVs is still unclear, which limits full understanding of the clinical potential of EVs. Here, we discuss how EVs, oxidative stress, and cancer therapy relate to one another; how oxidative stress can contribute to the generation of EVs; and how EVs’ contents reveal the presence of oxidative stress. We also point out the potential promise and limitations of using oxidatively modified EVs as biomarkers of cancer and tissue injury with a focus on pediatric oncology patients.
Collapse
|
8
|
Al Dahhan NZ, Cox E, Nieman BJ, Mabbott DJ. Cross-translational models of late-onset cognitive sequelae and their treatment in pediatric brain tumor survivors. Neuron 2022; 110:2215-2241. [PMID: 35523175 DOI: 10.1016/j.neuron.2022.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Pediatric brain tumor treatments have a high success rate, but survivors are at risk of cognitive sequelae that impact long-term quality of life. We summarize recent clinical and animal model research addressing pathogenesis or evaluating candidate interventions for treatment-induced cognitive sequelae. Assayed interventions encompass a broad range of approaches, including modifications to radiotherapy, modulation of immune response, prevention of treatment-induced cell loss or promotion of cell renewal, manipulation of neuronal signaling, and lifestyle/environmental adjustments. We further emphasize the potential of neuroimaging as a key component of cross-translation to contextualize laboratory research within broader clinical findings. This cross-translational approach has the potential to accelerate discovery to improve pediatric cancer survivors' long-term quality of life.
Collapse
Affiliation(s)
- Noor Z Al Dahhan
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Elizabeth Cox
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada; Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
9
|
Sukati S, Ho J, Chaiswing L, Sompol P, Pandit H, Wei W, Izumi T, Chen Q, Weiss H, Noel T, Bondada S, Allan Butterfield D, St. Clair DK. Extracellular vesicles released after cranial radiation: An insight into an early mechanism of brain injury. Brain Res 2022; 1782:147840. [PMID: 35183524 PMCID: PMC8918058 DOI: 10.1016/j.brainres.2022.147840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/16/2022]
Abstract
Cranial radiation is important for treating both primary brain tumors and brain metastases. A potential delayed side effect of cranial radiation is neurocognitive function decline. Early detection of CNS injury might prevent further neuronal damage. Extracellular vesicles (EVs) have emerged as a potential diagnostic tool because of their unique membranous characteristics and cargos. We investigated whether EVs can be an early indicator of CNS injury by giving C57BJ/6 mice 10 Gy cranial IR. EVs were isolated from sera to quantify: 1) number of EVs using nanoparticle tracking analysis (NTA); 2) Glial fibrillary acidic protein (GFAP), an astrocyte marker; and 3) protein-bound 4-hydroxy-2-nonenal (HNE) adducts, an oxidative damage marker. Brain tissues were prepared for immunohistochemistry staining and protein immunoblotting. The results demonstrate: 1) increased GFAP levels (p < 0.05) in EVs, but not brain tissue, in the IR group; and 2) increased HNE-bound protein adduction levels (p < 0.05). The results support using EVs as an early indicator of cancer therapy-induced neuronal injury.
Collapse
|
10
|
Zhang P, Yao L, Shan G, Chen Y. A model of radiation-induced temporomandibular joint damage in mice. Int J Radiat Biol 2022; 98:1645-1654. [PMID: 35467478 DOI: 10.1080/09553002.2022.2069298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 02/24/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE A small animal radiation research platform (SARRP) equipped with a miniature beam system, an image-guided positioning system, and a dose planning system was used to develop and evaluate a mouse model of radiation-induced temporomandibular damage. METHODS Left jaw disks of adult male C57BL/6 mice and C3H mice were targeted using the SARRP for image-guided irradiation. The total radiation dose was 75 Gy. Experiment 1 (Scoping study): Mice in the C57BL/6 mouse test and control groups were sacrificed at 1, 3, 6, 9, 12, 15, and 18 weeks after irradiation, whereas mice in the C3H test and control groups were sacrificed at 1, 3, 6, 9, and 12 weeks after irradiation. Experiment 2 (Full -scale validation study): Mice in the C57BL/6 mouse test and control groups were sacrificed at 1, 3 and 6 weeks after irradiation. Histopathological analysis of the temporomandibular skeletal muscle in each group was performed using hematoxylin and eosin (H&E) and Masson staining; the temporal mandibular bone was examined through H&E staining. RESULTS SARRP delivered the rated dose to the temporomandibular joints of C57BL/6 and C3H mice. C3H and C57BL/6 mice in the test group showed different degrees of osteocytic necrosis and osteoporosis at different time points. H&E staining of skeletal muscle tissue showed slight fibrosis in the C57BL/6 test at 3 and 6 weeks time point. CONCLUSION We established a model of radiation-induced damage in the temporomandibular joint of C57BL/6 mice and demonstrated that the observed physiological and histological changes correspond to radiation damage observed in humans. Furthermore, the SARRP can deliver precise radiation doses.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Radiology Physics, Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Lejing Yao
- Department of Radiology Physics, Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Guoping Shan
- Department of Radiology Physics, Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiology Oncology, Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
11
|
Abstract
Modeling of metastatic disease in animal models is a critical resource to study the complexity of this multi-step process in a relevant system. Available models of metastatic disease to the brain are still far from ideal but they allow to address specific aspects of the biology or mimic clinically relevant scenarios. We not only review experimental models and their potential improvements but also discuss specific answers that could be obtained from them on unsolved aspects of clinical management.
Collapse
Affiliation(s)
- Lauritz Miarka
- Brain Metastasis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
12
|
Kumar G, Dutta P, Parihar VK, Chamallamudi MR, Kumar N. Radiotherapy and Its Impact on the Nervous System of Cancer Survivors. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:374-385. [PMID: 32640964 DOI: 10.2174/1871527319666200708125741] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 12/12/2022]
Abstract
Radiotherapy is routinely used for the treatment of nearly all brain tumors, but it may lead to progressive and debilitating impairments of cognitive function. The growing evidence supports the fact that radiation exposure to CNS disrupts diverse cognitive functions including learning, memory, processing speed, attention and executive functions. The present review highlights the types of radiotherapy and the possible mechanisms of cognitive deficits and neurotoxicity following radiotherapy. The review summarizes the articles from Scopus, PubMed, and Web of science search engines. Radiation therapy uses high-powered x-rays, particles, or radioactive seeds to kill cancer cells, with minimal damage to healthy cells. While radiotherapy has yielded relative success in the treatment of cancer, patients are often plagued with unwanted and even debilitating side effects from the treatment, which can lead to dose reduction or even cessation of treatment. Little is known about the underlying mechanisms responsible for the development of these behavioral toxicities; however, neuroinflammation is widely considered as one of the major mechanisms responsible for radiotherapy-induced toxicities. The present study reviews the different types of radiotherapy available for the treatment of various types of cancers and their associated neurological complications. It also summarizes the doses of radiations used in the variety of radiotherapy, and their early and delayed side effects. Special emphasis is given to the effects of various types of radiations or late side effects on cognitive impairments.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Priyadarshini Dutta
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA 92697- 2695, United States
| | - Mallikarjuna R Chamallamudi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Nitesh Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| |
Collapse
|
13
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
14
|
Gorbunov NV, Kiang JG. Brain Damage and Patterns of Neurovascular Disorder after Ionizing Irradiation. Complications in Radiotherapy and Radiation Combined Injury. Radiat Res 2021; 196:1-16. [PMID: 33979447 PMCID: PMC8297540 DOI: 10.1667/rade-20-00147.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 04/02/2021] [Indexed: 12/31/2022]
Abstract
Exposure to ionizing radiation, mechanical trauma, toxic chemicals or infections, or combinations thereof (i.e., combined injury) can induce organic injury to brain tissues, the structural disarrangement of interactive networks of neurovascular and glial cells, as well as on arrays of the paracrine and systemic destruction. This leads to subsequent decline in cognitive capacity and decompensation of mental health. There is an ongoing need for improvement in mitigating and treating radiation- or combined injury-induced brain injury. Cranial irradiation per se can cause a multifactorial encephalopathy that occurs in a radiation dose- and time-dependent manner due to differences in radiosensitivity among the various constituents of brain parenchyma and vasculature. Of particular concern are the radiosensitivity and inflammation susceptibility of: 1. the neurogenic and oligodendrogenic niches in the subependymal and hippocampal domains; and 2. the microvascular endothelium. Thus, cranial or total-body irradiation can cause a plethora of biochemical and cellular disorders in brain tissues, including: 1. decline in neurogenesis and oligodendrogenesis; 2. impairment of the blood-brain barrier; and 3. ablation of vascular capillary. These changes, along with cerebrovascular inflammation, underlie different stages of encephalopathy, from the early protracted stage to the late delayed stage. It is evident that ionizing radiation combined with other traumatic insults such as penetrating wound, burn, blast, systemic infection and chemotherapy, among others, can exacerbate the radiation sequelae (and vice versa) with increasing severity of neurogenic and microvascular patterns of radiation brain damage.
Collapse
Affiliation(s)
| | - Juliann G. Kiang
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
15
|
Unexpected Benefits of Multiport Synchrotron Microbeam Radiation Therapy for Brain Tumors. Cancers (Basel) 2021; 13:cancers13050936. [PMID: 33668110 PMCID: PMC7956531 DOI: 10.3390/cancers13050936] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary We unveiled the potential of an innovative irradiation technique that ablates brain cancer while sparing normal tissues. Spatially fractionating the incident beam into arrays of micrometer-wide beamlets of X-rays (MRT for Microbeam Radiation Therapy) has led to significantly increased survival and tumor control in preclinical studies. Multiport MRT versus conventional irradiations, for the same background continuous dose, resulted in unexpectedly high equivalent biological effects in rats that have not been achieved with any other radiotherapeutic method. These hallmarks of multiport MRT, i.e., minimal impact on normal tissues and exceptional tumor control, may promote this method towards clinical applications, possibly increasing survival and improving long-term outcomes in neuro-oncology patients. Abstract Delivery of high-radiation doses to brain tumors via multiple arrays of synchrotron X-ray microbeams permits huge therapeutic advantages. Brain tumor (9LGS)-bearing and normal rats were irradiated using a conventional, homogeneous Broad Beam (BB), or Microbeam Radiation Therapy (MRT), then studied by behavioral tests, MRI, and histopathology. A valley dose of 10 Gy deposited between microbeams, delivered by a single port, improved tumor control and median survival time of tumor-bearing rats better than a BB isodose. An increased number of ports and an accumulated valley dose maintained at 10 Gy delayed tumor growth and improved survival. Histopathologically, cell death, vascular damage, and inflammatory response increased in tumors. At identical valley isodose, each additional MRT port extended survival, resulting in an exponential correlation between port numbers and animal lifespan (r2 = 0.9928). A 10 Gy valley dose, in MRT mode, delivered through 5 ports, achieved the same survival as a 25 Gy BB irradiation because of tumor dose hot spots created by intersecting microbeams. Conversely, normal tissue damage remained minimal in all the single converging extratumoral arrays. Multiport MRT reached exceptional ~2.5-fold biological equivalent tumor doses. The unique normal tissue sparing and therapeutic index are eminent prerequisites for clinical translation.
Collapse
|
16
|
Voshart DC, Wiedemann J, van Luijk P, Barazzuol L. Regional Responses in Radiation-Induced Normal Tissue Damage. Cancers (Basel) 2021; 13:cancers13030367. [PMID: 33498403 PMCID: PMC7864176 DOI: 10.3390/cancers13030367] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Normal tissue side effects remain a major concern in radiotherapy. The improved precision of radiation dose delivery of recent technological developments in radiotherapy has the potential to reduce the radiation dose to organ regions that contribute the most to the development of side effects. This review discusses the contribution of regional variation in radiation responses in several organs. In the brain, various regions were found to contribute to radiation-induced neurocognitive dysfunction. In the parotid gland, the region containing the major ducts was found to be critical in hyposalivation. The heart and lung were each found to exhibit regional responses while also mutually affecting each other's response to radiation. Sub-structures critical for the development of side effects were identified in the pancreas and bladder. The presence of these regional responses is based on a non-uniform distribution of target cells or sub-structures critical for organ function. These characteristics are common to most organs in the body and we therefore hypothesize that regional responses in radiation-induced normal tissue damage may be a shared occurrence. Further investigations will offer new opportunities to reduce normal tissue side effects of radiotherapy using modern and high-precision technologies.
Collapse
Affiliation(s)
- Daniëlle C. Voshart
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Julia Wiedemann
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Peter van Luijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| |
Collapse
|
17
|
Franco-Pérez J, Montes S, Sánchez-Hernández J, Ballesteros-Zebadúa P. Whole-brain irradiation differentially modifies neurotransmitters levels and receptors in the hypothalamus and the prefrontal cortex. Radiat Oncol 2020; 15:269. [PMID: 33228731 PMCID: PMC7684903 DOI: 10.1186/s13014-020-01716-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/13/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Whole-brain radiotherapy is a primary treatment for brain tumors and brain metastasis, but it also induces long-term undesired effects. Since cognitive impairment can occur, research on the etiology of secondary effects has focused on the hippocampus. Often overlooked, the hypothalamus controls critical homeostatic functions, some of which are also susceptible after whole-brain radiotherapy. Therefore, using whole-brain irradiation (WBI) in a rat model, we measured neurotransmitters and receptors in the hypothalamus. The prefrontal cortex and brainstem were also analyzed since they are highly connected to the hypothalamus and its regulatory processes. METHODS Male Wistar rats were exposed to WBI with 11 Gy (Biologically Effective Dose = 72 Gy). After 1 month, we evaluated changes in gamma-aminobutyric acid (GABA), glycine, taurine, aspartate, glutamate, and glutamine in the hypothalamus, prefrontal cortex, and brainstem according to an HPLC method. Ratios of Glutamate/GABA and Glutamine/Glutamate were calculated. Through Western Blott analysis, we measured the expression of GABAa and GABAb receptors, and NR1 and NR2A subunits of NMDA receptors. Changes were analyzed comparing results with sham controls using the non-parametric Mann-Whitney U test (p < 0.05). RESULTS WBI with 11 Gy induced significantly lower levels of GABA, glycine, taurine, aspartate, and GABAa receptor in the hypothalamus. Also, in the hypothalamus, a higher Glutamate/GABA ratio was found after irradiation. In the prefrontal cortex, WBI induced significant increases of glutamine and glutamate, Glutamine/Glutamate ratio, and increased expression of both GABAa receptor and NMDA receptor NR1 subunit. The brainstem showed no statistically significant changes after irradiation. CONCLUSION Our findings confirm that WBI can affect rat brain regions differently and opens new avenues for study. After 1 month, WBI decreases inhibitory neurotransmitters and receptors in the hypothalamus and, conversely, increases excitatory neurotransmitters and receptors in the prefrontal cortex. Increments in Glutamate/GABA in the hypothalamus and Glutamine/Glutamate in the frontal cortex indicate a neurochemical imbalance. Found changes could be related to several reported radiotherapy secondary effects, suggesting new prospects for therapeutic targets.
Collapse
Affiliation(s)
- Javier Franco-Pérez
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Sergio Montes
- Laboratory of Neurochemistry, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Josué Sánchez-Hernández
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Paola Ballesteros-Zebadúa
- Laboratory of Medical Physics, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico.
| |
Collapse
|
18
|
Constanzo J, Midavaine É, Fouquet J, Lepage M, Descoteaux M, Kirby K, Tremblay L, Masson-Côté L, Geha S, Longpré JM, Paquette B, Sarret P. Brain irradiation leads to persistent neuroinflammation and long-term neurocognitive dysfunction in a region-specific manner. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109954. [PMID: 32360786 DOI: 10.1016/j.pnpbp.2020.109954] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 01/04/2023]
Abstract
Long-term cognitive deficits are observed after treatment of brain tumors or metastases by radiotherapy. Treatment optimization thus requires a better understanding of the effects of radiotherapy on specific brain regions, according to their sensitivity and interconnectivity. In the present study, behavioral tests supported by immunohistology and magnetic resonance imaging provided a consistent picture of the persistent neurocognitive decline and neuroinflammation after the onset of irradiation-induced necrosis in the right primary somatosensory cortex of Fischer rats. Necrosis surrounded by neovascularization was first detected 54 days after irradiation and then spread to 110 days in the primary motor cortex, primary somatosensory region, striatum and right ventricle, resulting in fiber bundle disruption and demyelination in the corpus callosum of the right hemisphere. These structural damages translated into selective behavioral changes including spatial memory loss, disinhibition of anxiety-like behaviors, hyperactivity and pain hypersensitivity, but no significant alteration in motor coordination and grip strength abilities. Concomitantly, activated microglia and reactive astrocytes, accompanied by infiltration of leukocytes (CD45+) and T-cells (CD3+) cooperated to shape the neuroinflammation response. Overall, our study suggests that the slow and gradual onset of cellular damage would allow adaptation in brain regions that are susceptible to neuronal plasticity; while other cerebral structures that do not have this capacity would be more affected. The planning of radiotherapy, adjusted to the sensitivity and adaptability of brain structures, could therefore preserve certain neurocognitive functions; while higher doses of radiation could be delivered to brain areas that can better adapt to this treatment. In addition, strategies to block early post-radiation events need to be explored to prevent the development of long-term cognitive dysfunction.
Collapse
Affiliation(s)
- Julie Constanzo
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Élora Midavaine
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Jérémie Fouquet
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Martin Lepage
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Maxime Descoteaux
- Computer Science Department, Université de Sherbrooke, Sherbrooke, Québec J1K 2R1, Canada
| | - Karyn Kirby
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Luc Tremblay
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Laurence Masson-Côté
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Service of Radiation Oncology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Benoit Paquette
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
19
|
Dos Santos M, Kereselidze D, Gloaguen C, Benadjaoud MA, Tack K, Lestaevel P, Durand C. Development of whole brain versus targeted dentate gyrus irradiation model to explain low to moderate doses of exposure effects in mice. Sci Rep 2018; 8:17262. [PMID: 30467388 PMCID: PMC6250717 DOI: 10.1038/s41598-018-35579-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/16/2018] [Indexed: 12/23/2022] Open
Abstract
Evaluation of the consequences of low to moderate doses of ionizing radiation (IR) remains a societal challenge, especially for children exposed to CT scans. Appropriate experimental models are needed to improve scientific understanding of how exposure of the postnatal brain to IR affects behavioral functions and their related pathophysiological mechanisms, considering brain complex functional organization. In the brain, the dorsal and ventral hippocampal dentate gyrus can be involved in distinct major behavioral functions. To study the long term behavioral effects of brain exposure at low to moderate doses of IR (doses range 0.25–1 Gy), we developed three new experimental models in 10-day-old mice: a model of brain irradiation and two targeted irradiation models of the dorsal and ventral dentate gyrus. We used the technological properties of the SARRP coupled with MR imaging. Our irradiation strategy has been twofold endorsed. The millimetric ballistic specificity of our models was first validated by measuring gamma-H2AX increase after irradiation. We then demonstrated higher anxiety/depressive-like behavior, preferentially mediate by the ventral part of the dentate gyrus, in mice after brain and ventral dentate gyrus IR exposure. This work provides new tools to enhance scientific understanding of how to protect children exposed to IR.
Collapse
Affiliation(s)
- M Dos Santos
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department of RAdiobiology and regenerative MEDicine (SERAMED), Laboratory of Radiobiology of Accidental exposures (LRAcc), Fontenay-aux-Roses, France
| | - D Kereselidze
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - C Gloaguen
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - M A Benadjaoud
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department of RAdiobiology and regenerative MEDicine (SERAMED), Fontenay-aux-Roses, France
| | - K Tack
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - P Lestaevel
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - C Durand
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France.
| |
Collapse
|
20
|
Abstract
OBJECTIVES To address the estimated rates of incidence, potential underlying etiologies, and cognitive domains affected from diagnosis and treatment. To describe potential cognitive function interventions. DATA SOURCES PubMed. CONCLUSION Adults with gliomas report that the most distressing, persistent, and greatest negative impact on their lives relates to the cognitive impairment they experience. However, there are several potential interventions that may prevent cognitive decline during treatment or maintain cognitive function long term. IMPLICATIONS FOR NURSING PRACTICE Awareness of cognitive sequela that adults with gliomas face can lead to early identification, full neurocognitive profiling, and implementation of evidence-based interventions for those experiencing cognitive impairments following cancer treatment.
Collapse
|
21
|
Results of the radiation dose of head, body and tail of hippocampus in nasopharyngeal carcinoma patients treated with intensity modulated radiotherapy. Sci Rep 2018; 8:5595. [PMID: 29618828 PMCID: PMC5884782 DOI: 10.1038/s41598-018-23127-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/06/2018] [Indexed: 01/29/2023] Open
Abstract
This study is to analyze the radiation dose of head, body and tail of hippocampus (HC) of nasopharyngeal carcinoma (NPC) patients treated with intensity modulated radiotherapy (IMRT). Evaluate cognitive function of patients with Wechsler adult intelligence scale-Chinese revised (WAIS-CR). HC were segmented into HC head (HH), HC body (HB) and HC tail (HT) and the indexes were then analyzed. WAIS-CR was tested before and 3months after radiotherapy. The mean radiation dose of left and right HC was (1147 ± 976)cGy, (1011 ± 602)cGy respectively. The radiation dose and the volume exposed in different dose of HH, HB and HT decreased in turn. For 5 patients, before and after radiotherapy, the regular-order score was 8.60 ± 1.34, 8.0 ± 1.00 (P = 0.43), while the reverse-order score was 5.80 ± 0.84, 5.20 ± 0.84 (P = 0.07). The radiation dose of HH, HB and HT was different, and the radiation dose of HH was the highest, which should be emphasized especially.
Collapse
|
22
|
Montay-Gruel P, Petersson K, Jaccard M, Boivin G, Germond JF, Petit B, Doenlen R, Favaudon V, Bochud F, Bailat C, Bourhis J, Vozenin MC. Irradiation in a flash: Unique sparing of memory in mice after whole brain irradiation with dose rates above 100Gy/s. Radiother Oncol 2017; 124:365-369. [PMID: 28545957 DOI: 10.1016/j.radonc.2017.05.003] [Citation(s) in RCA: 440] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/13/2017] [Accepted: 05/04/2017] [Indexed: 10/19/2022]
Abstract
This study shows for the first time that normal brain tissue toxicities after WBI can be reduced with increased dose rate. Spatial memory is preserved after WBI with mean dose rates above 100Gy/s, whereas 10Gy WBI at a conventional radiotherapy dose rate (0.1Gy/s) totally impairs spatial memory.
Collapse
Affiliation(s)
- Pierre Montay-Gruel
- Department of Radiation Oncology/DO/CHUV, Lausanne University Hospital, Switzerland; Institut Curie, INSERM U1021/CNRS UMR3347, Université Paris-Saclay, Orsay, France
| | | | - Maud Jaccard
- Institute of Radiation Physics (IRA), Lausanne University Hospital, Switzerland
| | - Gaël Boivin
- Department of Radiation Oncology/DO/CHUV, Lausanne University Hospital, Switzerland
| | | | - Benoit Petit
- Department of Radiation Oncology/DO/CHUV, Lausanne University Hospital, Switzerland
| | - Raphaël Doenlen
- Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland
| | - Vincent Favaudon
- Institut Curie, INSERM U1021/CNRS UMR3347, Université Paris-Saclay, Orsay, France
| | - François Bochud
- Institute of Radiation Physics (IRA), Lausanne University Hospital, Switzerland
| | - Claude Bailat
- Institute of Radiation Physics (IRA), Lausanne University Hospital, Switzerland
| | - Jean Bourhis
- Department of Radiation Oncology/DO/CHUV, Lausanne University Hospital, Switzerland
| | | |
Collapse
|
23
|
Lumniczky K, Szatmári T, Sáfrány G. Ionizing Radiation-Induced Immune and Inflammatory Reactions in the Brain. Front Immunol 2017; 8:517. [PMID: 28529513 PMCID: PMC5418235 DOI: 10.3389/fimmu.2017.00517] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/18/2017] [Indexed: 01/17/2023] Open
Abstract
Radiation-induced late brain injury consisting of vascular abnormalities, demyelination, white matter necrosis, and cognitive impairment has been described in patients subjected to cranial radiotherapy for brain tumors. Accumulating evidence suggests that various degrees of cognitive deficit can develop after much lower doses of ionizing radiation, as well. The pathophysiological mechanisms underlying these alterations are not elucidated so far. A permanent deficit in neurogenesis, chronic microvascular alterations, and blood–brain barrier dysfunctionality are considered among the main causative factors. Chronic neuroinflammation and altered immune reactions in the brain, which are inherent complications of brain irradiation, have also been directly implicated in the development of cognitive decline after radiation. This review aims to give a comprehensive overview on radiation-induced immune alterations and inflammatory reactions in the brain and summarizes how these processes can influence cognitive performance. The available data on the risk of low-dose radiation exposure in the development of cognitive impairment and the underlying mechanisms are also discussed.
Collapse
Affiliation(s)
- Katalin Lumniczky
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Tünde Szatmári
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Géza Sáfrány
- Division of Radiation Medicine, National Public Health Centre, National Research Directorate for Radiobiology and Radiohygiene, Budapest, Hungary
| |
Collapse
|
24
|
Sweet TB, Hurley SD, Wu MD, Olschowka JA, Williams JP, O'Banion MK. Neurogenic Effects of Low-Dose Whole-Body HZE (Fe) Ion and Gamma Irradiation. Radiat Res 2016; 186:614-623. [PMID: 27905869 DOI: 10.1667/rr14530.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Understanding the dose-toxicity profile of radiation is critical when evaluating potential health risks associated with natural and man-made sources in our environment. The purpose of this study was to evaluate the effects of low-dose whole-body high-energy charged (HZE) iron (Fe) ions and low-energy gamma exposure on proliferation and differentiation of adult-born neurons within the dentate gyrus of the hippocampus, cells deemed to play a critical role in memory regulation. To determine the dose-response characteristics of the brain to whole-body Fe-ion vs. gamma-radiation exposure, C57BL/6J mice were irradiated with 1 GeV/n Fe ions or a static 137Cs source (0.662 MeV) at doses ranging from 0 to 300 cGy. The neurogenesis was analyzed at 48 h and one month postirradiation. These experiments revealed that whole-body exposure to either Fe ions or gamma radiation leads to: 1. An acute decrease in cell division within the dentate gyrus of the hippocampus, detected at doses as low as 30 and 100 cGy for Fe ions and gamma radiation, respectively; and 2. A reduction in newly differentiated neurons (DCX immunoreactivity) at one month postirradiation, with significant decreases detected at doses as low as 100 cGy for both Fe ions and gamma rays. The data presented here contribute to our understanding of brain responses to whole-body Fe ions and gamma rays and may help inform health-risk evaluations related to systemic exposure during a medical or radiologic/nuclear event or as a result of prolonged space travel.
Collapse
Affiliation(s)
- Tara B Sweet
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sean D Hurley
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Michael D Wu
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - John A Olschowka
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,c Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - M Kerry O'Banion
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,d Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
25
|
Zhao R, Kong W, Shang J, Zhe H, Wang YY. Hippocampal-Sparing Whole-Brain Radiotherapy for Lung Cancer. Clin Lung Cancer 2016; 18:127-131. [PMID: 28340924 DOI: 10.1016/j.cllc.2016.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/31/2016] [Accepted: 09/06/2016] [Indexed: 11/29/2022]
Abstract
Brain metastases occur in 20% to 40% of lung cancer patients. Whole-brain radiotherapy (WBRT) has long been considered the treatment of choice for many patients with lung cancer, because of its wide availability, ease of delivery, and effectiveness in prolonging survival. However, WBRT is also associated with several side effects, such as decline in memory and other cognitive functions. There exists significant preclinical and clinical evidence that radiation-induced injury to the hippocampus correlates with neurocognitive decline of patients who receive WBRT. Technological advances in treatment planning and delivery facilitate the use of hippocampal-sparing (HS) WBRT as prophylactic cranial irradiation or the primary treatment modality for lung cancer patients with brain metastases. In this review, we provide a detailed and comprehensive discussion of the safety profile, techniques for hippocampus-sparing, and the clinical evidence of HS-WBRT for lung cancer patients.
Collapse
Affiliation(s)
- Ren Zhao
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, and Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wei Kong
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, and Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jun Shang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, and Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hong Zhe
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, and Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yan-Yang Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, and Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
26
|
Tomé WA, Gökhan Ş, Gulinello ME, Brodin NP, Heard J, Mehler MF, Guha C. Hippocampal-dependent neurocognitive impairment following cranial irradiation observed in pre-clinical models: current knowledge and possible future directions. Br J Radiol 2015; 89:20150762. [PMID: 26514377 DOI: 10.1259/bjr.20150762] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
We reviewed the literature for studies pertaining to impaired adult neurogenesis leading to neurocognitive impairment following cranial irradiation in rodent models. This compendium was compared with respect to radiation dose, converted to equivalent dose in 2 Gy fractions (EQD2) to allow for direct comparison between studies. The effects of differences between animal species and the dependence on animal age as well as for time after irradiation were also considered. One of the major sites of de novo adult neurogenesis is the hippocampus, and as such, this review also focuses on assessing evidence related to the expression and potential effects of inflammatory cytokines on neural stem cells in the subgranular zone of the dentate gyrus and whether this correlates with neurocognitive impairment. This review also discusses potential strategies to mitigate the detrimental effects on neurogenesis and neurocognition resulting from cranial irradiation, and how the rationale for these strategies compares with the current outcome of pre-clinical studies.
Collapse
Affiliation(s)
- Wolfgang A Tomé
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA.,3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- 3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria E Gulinello
- 4 Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - N Patrik Brodin
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - John Heard
- 2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Mark F Mehler
- 3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.,4 Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,5 Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chandan Guha
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|