Skip to main content
The British Journal of Radiology logoLink to The British Journal of Radiology
. 2020 Oct 8;93(1115):20200245. doi: 10.1259/bjr.20200245

Can a comparison of clinical and deep space irradiation scenarios shed light on the radiation response of the brain?

Charles Limoli 1,
PMCID: PMC8519643  PMID: 32970457

Abstract

Not surprisingly, our knowledge of the impact of radiation on the brain has evolved considerably. Decades of work have struggled with identifying the critical cellular targets in the brain, the latency of functional change and understanding how irradiation alters the balance between excitatory and inhibitory circuits. Radiation-induced cell kill following clinical fractionation paradigms pointed to both stromal and parenchymal targets but also defined an exquisite sensitivity of neurogenic populations of newly born cells in the brain. It became more and more apparent too, that acute (days) events transpiring after exposure were poorly prognostic of the late (months-years) waves of radiation injury believed to underlie neurocognitive deficits. Much of these gaps in knowledge persisted as NASA became interested in how exposure to much different radiation types, doses and dose rates that characterize the space radiation environment might impair central nervous system functionality, with possibly negative implications for deep space travel. Now emerging evidence from researchers engaged in clinical, translational and environmental radiation sciences have begun to fill these gaps and have uncovered some surprising similarities in the response of the brain to seemingly disparate exposure scenarios. This article highlights many of the commonalities between the vastly different irradiation paradigms that distinguish clinical treatments from occupational exposures in deep space.

Introduction

Exposure of the central nervous system (CNS) to multiple radiation types comes from a variety of sources, including environmental (background), occupational (nuclear workers), and from medical procedures (diagnostic and therapeutic). The most significant data regarding the radioresponse of the CNS have been derived from the clinical experience, where therapeutic treatments using cranial irradiation have been used to forestall primary and secondary CNS malignancies.1–5 Typical doses involve 60 Gy of photon-based (X-ray, gamma-ray) radiation modalities delivered in multiple small fractions aimed at minimizing normal tissue damage while eliciting certain levels of tumor growth delay if not control.1 More recently, hadron-based (charged particles generated in cyclotrons) therapies have been used to control head and neck and CNS tumors, and most often involve protons6,7 while outside the USA (largely in Germany and Japan), heavier ions (carbon) have been utilized.8,9 Charged particles of specified energy and mass can be directed such that energy deposited can be localized to the tumor, where the density of ionizations (Bragg peak) can be superimposed over the tumor volume while minimizing damage to the collateral normal tissue bed.10 Interestingly in space, a wider range of charged particles exist, that include protons and helium ions derived from the sun as well as minor contributions from heavier charged species ranging up to iron ions (atomic number Z ≤ 26) that define the isotropic field of Galactic Cosmic Rays (GCR).11 High atomic number (Z) and Energy (HZE) particles are charged nuclei in the GCR that possess an electrical charge greater than +2. The multiple fluences and energies of these HZE particles traveling at near relativistic speeds highlight the complexities of the radiation fields in space, and provide a launching point for our discussions highlighting the nuances of the CNS radiation response to the vastly disparate radiation exposure scenarios encountered terrestrially and in deep space.

What happens to your brain following clinical radiotherapy of brain tumors

Clinicians have known for decades that brain tumor survivors (adult and pediatric) suffer from progressive and debilitating cognitive impairments resulting from their cranial radiotherapy.3–5,12 These unintended normal tissue toxicities limit the dose that can be safely administered to the tumor bed, and severely compromise quality of life. For the treatment of glioblastoma multiforme (GBM), the most aggressive primary brain tumor, radiotherapy protocols are used in combination with temozolomide (TMZ). The use of TMZ as a concurrent and adjuvant chemotherapeutic agent, has been shown to increase both overall and progression-free survival in patients.2 Furthermore, GBM patients typically receive whole-brain fractionated x-irradiation to a total dose of 60 Gy delivered in 2 Gy fractions over 6 weeks.1,13 Such clinical radiation exposures have clearly been shown to induce cognitive impairments,4,14 and combined treatment using radiotherapy and TMZ have similarly been shown to elicit significant adverse neurocognitive side effects.2,13 Despite acknowledgment of the cognitive problems, interpretation of the literature is hampered by numerous confounding factors (e.g., differences in disease status, inter patient variation, treatment regimen, psychological reactions to diagnosis and treatment, baseline cognitive reserve and differences in test administration).15–18 In addition, it is difficult to delineate the specific brain regions most sensitive to radiation and cytotoxic drug exposure and thus the mechanisms underlying effects on cognitive function. While it is beyond the scope of the present manuscript to review the clinical literature, several comprehensive reviews have elaborated on the radiation response of the CNS and the many potential causes and consequences of radiation-induced cognitive dysfunction.3,19–23 Here, the focus will be on data derived from pre-clinical models, able to provide deeper mechanistic insight regarding the potential parallels between clinical and space radiation exposure of the CNS.

While the mechanisms underlying the unintended side effects of cranial radiation exposure remain to be completely elucidated, rodent studies using single dose and fractionated irradiation protocols, designed to approximate clinical treatments, have pointed to certain underlying mechanisms that characterize the CNS response to irradiation. Some insight has been provided by structural MRI studies, where white matter necrosis transpiring at protracted times after the cessation of treatments appears more dependent on total dose rather than a particular fractionation schedule.24 Neuroinflammation has also been shown to be a persistent problem, and in many instances can be linked to microglial activation.25–31 Adverse effects can result from a pro-inflammatory environment, where disruptions in synaptic transmission and secretion of growth factors may result.32,33 Astrocytes also play a role in neuropathological conditions, including neurodegeneration and neuroinflammation.34,35 In the context of brain injury, persistent modifications that characterize reactive astrogliosis have been described over acute and protracted post-irradiation time frames.25,36

Following cranial exposures (≤10 Gy) rodents exhibit a number neurocognitive decrements spanning multiple regions of the brain that persist (perhaps indefinitely) and over protracted post-irradiation times.37–40 Hippocampal and cortical-based deficits in learning and memory, along with the emergence of mood disorders are temporally coincident with changes in the structural plasticity of neurons and inflammation as documented after cosmic radiation exposures.39–41 Clinical irradiation paradigms elicit marked reductions in neurogenesis, dendritic complexity, spine density, and elevations in neuroinflammation that are hallmarks of the CNS radiation response.29,42–46 Data derived from both space and clinical irradiation scenarios suggest that compromised neurocognitive functionality goes in step with reductions in the structural complexity of neurons and increased inflammation in the brain. Faced with this reality, much of the work from my laboratory has focused on interventions able to ameliorate radiation-induced cognitive dysfunction, and identifying common mechanistic themes for resolving radiation-induced pathology in the brain.

Treatments for radiation-induced cognitive dysfunction

Despite the growing acknowledgment that cognitive outcome is a major criterion for assessing therapeutic outcome, cognitive dysfunction following cancer treatments remains an unmet medical need. To address this unresolved normal tissue complication, our lab was the first to pioneer stem cell and stem-cell derived Extracellular Vesicle (EV)-based strategies for the potential resolution of such normal tissue toxicities.38 Using several models of radiation-induced cognitive dysfunction, we have shown that intrahippocampal transplantation of several human stem cell sources were capable of ameliorating cognitive deficits following clinical irradiation paradigms.47–51 Transplanted stem cells were shown to engraft, adopt neural cell fates and functionally integrate into hippocampal circuitry.47 Longer term effects pointed to trophic support mechanisms. In the relative absence of engrafted cells, expression of the Activity Regulated Cytoskeleton-associated protein (ARC) known to facilitate synaptic transmission, was elevated in irradiated brains previously transplanted with Human Neural Stem Cells (hNSC).51 Importantly, neurocognitive benefits associated with stem cell grafting included a preservation of host neuronal morphology and an attenuation of neuroinflammation.

Success of these preclinical studies prompted efforts to circumvent certain limitations associated with cellular transplantation strategies, namely immune rejection and teratoma formation. To this end, we evaluated the therapeutic benefits of hippocampally grafted EV derived from human stem cell sources following cranial irradiation. In a proof of principal study, we found that hNSC-derived EV afforded similar neuroprotective properties as grafted stem cells, where neurocognitive benefits of EV were again associated with significant protection of host neuronal morphology and a reduction of neuroinflammation.52 More recent work has now extended these findings by demonstrating that systemic administration of hNSC-derived EV can resolve radiation-induced cognitive dysfunction and inflammation in wild-type mice through an miR-124-based mechanism.53 The similar protective benefits found after either stem cell or EV grafting suggested that strategies able to preserve neuronal structure, myelination, and limit inflammation in the brain would have a positive impact on multiple facets of cognition following CNS exposure to ionizing radiation.54–57 Indeed, related data from us and others have found that in general, interventions able to curtail oxidative and/or inflammatory signaling in the brainprovide an overall beneficial outcome for the CNS following radiation exposure.31,44,58–60

A new twist for reducing normal tissue toxicities in the irradiated brain: The “FLASH effect”

Data highlighted to this point indicate that radiation-induced toxicities in the brain involving changes to neuronal structure and inflammation seem to be contributory if not causal to resultant cognitive dysfunction. It therefore stands to reason that potentially any strategy able to preserve neuronal structure, myelination state and minimize inflammation in the irradiated brain may have long-lasting neurocognitive benefits. In this regard, FLASH radiotherapy (FLASH-RT) does precisely that, and represents a burgeoning radiation modality that implements ultra-high mean dose rates in excess of 100 Gy/s. The idea that dose rate could be an adjustable parameter for therapeutic gain has caught the field of radiation oncology by surprise, and its capability to spare normal tissue toxicities without compromising tumor treatments points to one of the more exciting developments in radiation biology in years.61 Recent reports from our group highlight the tremendous potential of this innovative radiation modality. When compared to conventional dose rate irradiation, FLASH-RT eliminated short- and long-lasting cognitive deficits (1–6 months post-exposure), minimized astrogliosis and the activation of microglia and preserved the structural integrity of mature neurons in the brain.62,63 Implications for radiation oncology are significant and suggest that if properly implemented, dose escalation to the tumor bed can be safely achieved while maintaining and/or reducing normal tissue toxicities to acceptable levels.64 While the benefits of FLASH-RT are only starting to be realized, such advantages are certainly not limited to the CNS, as normal tissue sparing has now been demonstrated in multiple tissues using several preclinical animal models65–69 including the first human subject.70

Space radiation

Despite the fact that clinical treatments involve much different types (low LET) and higher doses of ionizing radiation (~60 Gy) used to control CNS malignancies,1,2 it behooves an examination of NASA-related research investigating space radiation effects on CNS functionality. As highlighted below, research emerging over the last decade has revealed some surprising parallels regarding the response of the CNS to these disparate irradiation scenarios.

The Earth is protected by the magnetosphere, a shield that deflects charged particles and prevents them from reaching the surface of our planet. This protection is sacrificed during deep space travel where the highly energetic charged particles of the GCR can penetrate the hull of the spacecraft and bodies of the astronauts. HZE particles are characterized in part by their mass and Linear Energy Transfer (LET, expressed as keV/μm) a term used to distinguish the microdosimetric properties of specific charged species.11 HZE particles of higher LET have higher ionizing capacity, since they produce an increased density of ionized species per unit volume at a given dose. Thus, they produce more complex “clustered” cellular damage that challenges the repair and regenerative reserve of cells traversed by these damaging charged particles.

The realization that deep space radiation exposures are far below those used therapeutically or diagnostically in medical practice is also important, where total doses from all radiation types are accrued at dose rates of ~1 mGy/day or one-thousandth of a Gray.11 While dose rates are low, biological damage will depend on the nature and extent of specific ionizations incurred from all particle traversals, and the sum of these events over a projected round-trip transit to Mars is not expected to exceed a combined dose 0.5 Gy (depending of course on mission duration).71 Given this brief backdrop, we will now explore how terrestrial simulations of the space radiation environment have contributed to our knowledge regarding the space radiation response of the brain.

What happens to your brain when you travel to mars: The CNS response to space radiation

Our understanding of the CNS response to charged particle exposures that typify the space radiation environment have made tremendous strides over this last decade. This progress has come in large part, through the efforts of NASA-funded investigators carrying our ground-based simulations of the radiation fields in space at the NASA

Table 1.

Definition of the Full GCR Simulation (In order of delivery)

Ion Energy (MeV)
H 1000
He 1000
Si 600
H 20
H 23
He 20
He 23
Ti 1000
He 27
He 32
H 27
H 32
H 37
H 43
He 37
He 43
O 350
He 50
He 59
H 50
H 59
H 69
H 80
He 69
He 80
C 1000
He 100
H 100
H 150
He 150
Fe 600
He 250
H 250

Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory (BNL). Here, investigators expose a variety of biological samples to selected beams and scrutinize the consequences of such exposures at various times post-irradiation. For the CNS, rodent models have proven invaluable, as the need to assess neurobiological outcomes in an intact brain are an absolute necessity. In this light, mice and rats exposed to low doses (≤0.5 Gy) of single beams or combinations of multiple beams including protons, helium ions, and various HZE particles have revealed marked neurocognitive decrements that persist for months to years after exposure.72–74 Over the past few years, NASA and the team at the NSRL have developed a 33-beam complex GCR simulation that can sequentially deliver a complex spectrum of space radiations (Table). Investigators having access to this advanced GCR simulation have been able to expose their samples to pre-selected total doses (≤50 cGy) over a chronic 6 day/week, 4-week interval (2.08 cGy/day), and compare data to that obtained when the same total dose and sequential ion combination is delivered in a single day (i.e., 2 h). Results derived from these ongoing experiments will undoubtedly shed further light on the multifaceted consequences of space radiation exposure on CNS functionality.

Armed with a battery of behavioral tasks able to interrogate multiple cognitive domains, research has clearly identified space radiation-induced impairments in learning and memory that suggest astronauts engaged in deep space travel are at an elevated risk for manifesting mission critical performance decrements.75–78 Past work with rodents has shown that whole-body exposure to charged particles can disrupt behavioral performance that can be linked to impairments in the hippocampus,76,79–81 amygdala,73 basal forebrain,75 mPFC,72,73,75,82 and other regions.74,83,84 Furthermore, individual animals subjected to charged particles often exhibit deficits in multiple behavioral paradigms, and many of these decrements transpire over 6–52 weeks after exposure and are temporally coincident with a marked structural plasticity of neurons and glia and elevated neuroinflammation.72–74 This realization has triggered a surge of research designed to evaluate further the potential CNS health risks associated with such exposures. While rodent-based studies have clearly uncovered many intriguing mechanisms impacting space radiation-induced cognitive dysfunction, disruptions to the integrity of mature neuronal structure and chronic neuroinflammation represent two salient features shared in common with clinical exposure paradigms.

Studies have shown that space relevant doses of various charged particles elicit a robust and persistent deterioration of dendritic structure.72,73,80,85 These changes have now been documented in various types of neurons throughout the brain and include reductions in dendritic arborization and overall dendritic complexity that point to the capability of charged particles to significantly compromise neuronal morphology over extended times. Coincident with these more macroscopic alterations to neuronal structure are more microscopic changes to the dendritic spines.72,73 Dendritic spines are the structural correlates of learning and memory and contain the synaptic machinery that mediates neurotransmission. Space radiation exposure elicits significant reductions in dendritic spine density, essentially stripping these structures off the dendritic shaft. Individual animals showing the greatest reductions in dendritic spine density routinely exhibit the poorest performance on select behavioral tasks, suggesting a defined structure function relationship between neuronal structure and cognition.73 At higher resolution, electron micrographs reveal that similar charged particle exposures lead to significant reductions in axonal myelination,86 implicating the capability of space radiation exposure to compromise conduction velocity and neurotransmission at multiple levels.

Superimposed on the structural alterations detailed above, are indications of a persistent inflammation in the brain that serves to perpetuate the signature of radiation injury over extended post-irradiation times.73,74 Multiple reports have now shown that space relevant exposures elicit a persistent neuroinflammation involving significant increases in activated microglia, the innate immune cells of the brain.73,74,87–89 Chronic increases in activated microglia can trigger inflammatory cascades in the irradiated brain that can elicit signaling changes to disrupt the balance between excitatory and inhibitory neurotransmission. Microglia also play active roles in re-shaping the synaptic landscape, where they can prune dendritic arbors and spines to remodel the connectivity of the irradiated brain.90–92 Studies showing that microglial elimination can restore neurocognitive function after space and clinical radiation exposures support this idea,59,93 and point to importance of neuroinflammation in dictating the long-term radiation response of the brain.

Interestingly, our group has recently completed studies undertaken to simulate chronic, space relevant low dose (≤0.20 Gy) and low dose rate (1 mGy/day) exposures using neutrons and photons derived from a 252Cf source.94 Results have shown that rodents develop significant cognitive decrements and reductions in Long-Term Potentiation (LTP), a form of synaptic plasticity that facilitates learning and memory.94 These new findings point to the marked susceptibility of the CNS, where critical functional outcomes show surprising sensitivity to low dose and low dose rate radiation exposures. While these data sets portend several potential problems associated with radiation exposure in space, this is not an insurmountable problem or a deal breaker for space travel.95,96 Advanced shielding and biological interventions designed to combat the adverse effects of space radiation exposure on the brain are areas of active investigation.

Summary and perspectives

The response of the CNS to either clinical or cosmic irradiation scenarios involves a complex and multifaceted series of events that never completely resolve and invariably compromise the functional connectivity of neural circuits and synaptic transmission. While much is now known regarding the radioresponse of the CNS, much remains to be resolved, including the identity of critical cellular and subcellular targets that exhibit differential radiosensitivities. Elucidating meaningful parallels between clinical and space radiation-induced CNS responses is confounded further by the paucity of human data regarding exposure to combined spaceflight stressors. A longitudinal MRI study of astronauts pre- and post-flight uncovered volume changes in various regions of the brain97 and the highly publicized NASA twin study found that compared to his terrestrial bound brother, spaceflight compromised certain indices of cognitive performance.98 These recent studies point to spaceflight-induced changes to the brain, but in each instance, it remains difficult to attribute such changes to the effects of radiation exposure alone.

Our capability to intervene on the adverse cascade of signaling events triggered by irradiation will ultimately depend on a more thorough understanding of how ionizing radiation damages and disrupts the delicate balance between excitatory and inhibitory neurotransmission. Protecting the brain against the structural degradation of neurons and the ensuing cascades of oxidative and inflammatory processes triggered by irradiation seem critical to preserve CNS functionality. In support of this rationale, three distinct interventions were highlighted that have been shown to be neuroprotective in the irradiated rodent brain, namely, 1) cranial grafting of human stem cells, 2) cranial grafting and systemic injection of human stem cell-derived EV, and 3) FLASH irradiation. Noteworthy here was that for each of these treatments, neurocognitive sparing was temporally coincident with reductions in neuroinflammation and a preservation of mature neuronal morphology. Thus, it stands to reason, that potential treatments targeting the adverse neurocognitive effects of cranial radiotherapy in humans should, in part, consider agents capable of impacting pathways known to minimize inflammation and protect neuronal structure. Similar logic would apply to the protection of astronauts exposed to the deep space radiation environment. For long-term human health, the brain is likely the last frontier, and protection of our neural circuitry must be a priority for those trying to resolve the neurocognitive side effects resulting from brain tumor radiotherapy and for those destined to engage in deep space travel.

REFERENCES

  • 1.Cabrera AR, Kirkpatrick JP, Fiveash JB, Shih HA, Koay EJ, Lutz S, et al. Radiation therapy for glioblastoma: Executive summary of an American Society for radiation oncology evidence-based clinical practice guideline. Pract Radiat Oncol 2016; 6: 217–25. doi: 10.1016/j.prro.2016.03.007 [DOI] [PubMed] [Google Scholar]
  • 2.Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009; 10: 459–66. doi: 10.1016/S1470-2045(09)70025-7 [DOI] [PubMed] [Google Scholar]
  • 3.Makale MT, McDonald CR, Hattangadi-Gluth JA, Kesari S. Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours. Nat Rev Neurol 2017; 13: 52–64. doi: 10.1038/nrneurol.2016.185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Butler JM, Rapp SR, Shaw EG. Managing the cognitive effects of brain tumor radiation therapy. Curr Treat Options Oncol 2006; 7: 517–23. doi: 10.1007/s11864-006-0026-5 [DOI] [PubMed] [Google Scholar]
  • 5.Meyers CA. Neurocognitive dysfunction in cancer patients. Oncology 2000; 14: 75–9discussion 79, 81-2, 85. [PubMed] [Google Scholar]
  • 6.Gridley DS, Grover RS, Loredo LN, Wroe AJ, Slater JD. Proton-beam therapy for tumors of the CNS. Expert Rev Neurother 2010; 10: 319–30. doi: 10.1586/ern.09.150 [DOI] [PubMed] [Google Scholar]
  • 7.Mohan R, Bortfeld T. Proton therapy: clinical gains through current and future treatment programs. Front Radiat Ther Oncol 2011; 43: 440–64. doi: 10.1159/000322509 [DOI] [PubMed] [Google Scholar]
  • 8.Miyawaki D, Murakami M, Demizu Y, Sasaki R, Niwa Y, Terashima K, et al. Brain injury after proton therapy or carbon ion therapy for head-and-neck cancer and skull base tumors. Int J Radiat Oncol Biol Phys 2009; 75: 378–84. doi: 10.1016/j.ijrobp.2008.12.092 [DOI] [PubMed] [Google Scholar]
  • 9.Seidensaal K, Harrabi SB, Uhl M, Debus J. Re-Irradiation with protons or heavy ions with focus on head and neck, skull base and brain malignancies. Br J Radiol 2020; 93: 20190516. doi: 10.1259/bjr.20190516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Paganetti H. Relative biological effectiveness (RBE) values for proton beam therapy. variations as a function of biological endpoint, dose, and linear energy transfer. Phys Med Biol 2014; 59: R419–72. doi: 10.1088/0031-9155/59/22/R419 [DOI] [PubMed] [Google Scholar]
  • 11.Nelson GA. Space radiation and human exposures, a primer. Radiat Res 2016; 185: 349–58. doi: 10.1667/RR14311.1 [DOI] [PubMed] [Google Scholar]
  • 12.King AA, Seidel K, Di C, Leisenring WM, Perkins SM, Krull KR, et al. Long-Term neurologic health and psychosocial function of adult survivors of childhood medulloblastoma/PNET: a report from the childhood cancer Survivor study. Neuro Oncol 2017; 19: 689–98. doi: 10.1093/neuonc/now242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352: 987–96. doi: 10.1056/NEJMoa043330 [DOI] [PubMed] [Google Scholar]
  • 14.Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol 2006; 24: 1305–9. doi: 10.1200/JCO.2005.04.6086 [DOI] [PubMed] [Google Scholar]
  • 15.Myers JS. Chemotherapy-Related cognitive impairment. Clin J Oncol Nurs 2009; 13: 413–21. doi: 10.1188/09.CJON.413-421 [DOI] [PubMed] [Google Scholar]
  • 16.Vardy J, Rourke S, Tannock IF. Evaluation of cognitive function associated with chemotherapy: a review of published studies and recommendations for future research. J Clin Oncol 2007; 25: 2455–63. doi: 10.1200/JCO.2006.08.1604 [DOI] [PubMed] [Google Scholar]
  • 17.Vardy J, Wefel JS, Ahles T, Tannock IF, Schagen SB. Cancer and cancer-therapy related cognitive dysfunction: an international perspective from the Venice cognitive workshop. Ann Oncol 2008; 19: 623–9. doi: 10.1093/annonc/mdm500 [DOI] [PubMed] [Google Scholar]
  • 18.Wefel JS, Vardy J, Ahles T, Schagen SB. International cognition and cancer Task force recommendations to harmonise studies of cognitive function in patients with cancer. Lancet Oncol 2011; 12: 703–8. doi: 10.1016/S1470-2045(10)70294-1 [DOI] [PubMed] [Google Scholar]
  • 19.Greene-Schloesser D, Moore E, Robbins ME. Molecular pathways: radiation-induced cognitive impairment. Clin Cancer Res 2013; 19: 2294–300. doi: 10.1158/1078-0432.CCR-11-2903 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Greene-Schloesser D, Robbins ME, Peiffer AM, Shaw EG, Wheeler KT, Chan MD. Radiation-Induced brain injury: a review. Front Oncol 2012; 2: 73. doi: 10.3389/fonc.2012.00073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tofilon PJ, Fike JR. The radioresponse of the central nervous system: a dynamic process. Radiat Res 2000; 153: 357–70. doi: 10.1667/0033-7587(2000)153[0357:TROTCN]2.0.CO;2 [DOI] [PubMed] [Google Scholar]
  • 22.Walker AJ, Ruzevick J, Malayeri AA, Rigamonti D, Lim M, Redmond KJ, et al. Postradiation imaging changes in the CNS: how can we differentiate between treatment effect and disease progression? Future Oncol 2014; 10: 1277–97. doi: 10.2217/fon.13.271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kim JH, Jenrow KA, Brown SL. Mechanisms of radiation-induced normal tissue toxicity and implications for future clinical trials. Radiat Oncol J 2014; 32: 103–15. doi: 10.3857/roj.2014.32.3.103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jiang X, Yuan L, Engelbach JA, Cates J, Perez-Torres CJ, Gao F, et al. A Gamma-Knife-Enabled mouse model of cerebral Single-Hemisphere delayed radiation necrosis. PLoS One 2015; 10: e0139596. doi: 10.1371/journal.pone.0139596 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chiang CS, Hong JH, Stalder A, Sun JR, Withers HR, McBride WH. Delayed molecular responses to brain irradiation. Int J Radiat Biol 1997; 72: 45–53. doi: 10.1080/095530097143527 [DOI] [PubMed] [Google Scholar]
  • 26.Limoli CL, Giedzinski E, Rola R, Otsuka S, Palmer TD, Fike JR. Radiation response of neural precursor cells: linking cellular sensitivity to cell cycle checkpoints, apoptosis and oxidative stress. Radiat Res 2004; 161: 17–27. doi: 10.1667/RR3112 [DOI] [PubMed] [Google Scholar]
  • 27.Lan ML, Acharya MM, Tran KK, Bahari-Kashani J, Patel NH, Strnadel J, et al. Characterizing the radioresponse of pluripotent and multipotent human stem cells. PLoS One 2012; 7: e50048. doi: 10.1371/journal.pone.0050048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Acharya MM, Lan ML, Kan VH, Patel NH, Giedzinski E, Tseng BP, et al. Consequences of ionizing radiation-induced damage in human neural stem cells. Free Radic Biol Med 2010; 49: 1846–55. doi: 10.1016/j.freeradbiomed.2010.08.021 [DOI] [PubMed] [Google Scholar]
  • 29.Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR. Extreme sensitivity of adult neurogenesis to low doses of x-irradiation. Cancer Res 2003; 63: 4021–7. [PubMed] [Google Scholar]
  • 30.Parihar VK, Acharya MM, Roa DE, Bosch O, Christie L-A, Limoli CL. Defining functional changes in the brain caused by targeted stereotaxic radiosurgery. Transl Cancer Res 2014; 3: 124–37. doi: 10.3978/j.issn.2218-676X.2013.06.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jenrow KA, Brown SL, Lapanowski K, Naei H, Kolozsvary A, Kim JH. Selective inhibition of microglia-mediated neuroinflammation mitigates radiation-induced cognitive impairment. Radiat Res 2013; 179: 549–56. doi: 10.1667/RR3026.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bushong EA, Martone ME, Jones YZ, Ellisman MH. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains. J Neurosci 2002; 22: 183–92. doi: 10.1523/JNEUROSCI.22-01-00183.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Araque A, Carmignoto G, Haydon PG, Oliet SHR, Robitaille R, Volterra A. Gliotransmitters travel in time and space. Neuron 2014; 81: 728–39. doi: 10.1016/j.neuron.2014.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017; 541: 481–7. doi: 10.1038/nature21029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mciver SR, Faideau M, Haydon PG. Neural-Immune interactions in brain function and alcohol related disorders. New York: Springer Science+Business Media 2012;. [Google Scholar]
  • 36.Hong JH, Chiang CS, Campbell IL, Sun JR, Withers HR, McBride WH. Induction of acute phase gene expression by brain irradiation. Int J Radiat Oncol Biol Phys 1995; 33: 619–26. doi: 10.1016/0360-3016(95)00279-8 [DOI] [PubMed] [Google Scholar]
  • 37.Parihar VK, Acharya MM, Roa DE, Bosch O, Christie L-A, Limoli CL. Defining functional changes in the brain caused by targeted stereotaxic radiosurgery. Transl Cancer Res 2014; 3: 124–37. doi: 10.3978/j.issn.2218-676X.2013.06.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Acharya MM, Christie L-A, Lan ML, Donovan PJ, Cotman CW, Fike JR, et al. Rescue of radiation-induced cognitive impairment through cranial transplantation of human embryonic stem cells. Proc Natl Acad Sci U S A 2009; 106: 19150–5. doi: 10.1073/pnas.0909293106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Acharya MM, Martirosian V, Christie L-A, Limoli CL. Long-Term cognitive effects of human stem cell transplantation in the irradiated brain. Int J Radiat Biol 2014; 90: 816–20. doi: 10.3109/09553002.2014.927934 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Acharya MM, Martirosian V, Christie L-A, Riparip L, Strnadel J, Parihar VK, et al. Defining the optimal window for cranial transplantation of human induced pluripotent stem cell-derived cells to ameliorate radiation-induced cognitive impairment. Stem Cells Transl Med 2015; 4: 74–83. doi: 10.5966/sctm.2014-0063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Smith SM, Giedzinski E, Angulo MC, Lui T, Lu C, Park AL, et al. Functional equivalence of stem cell and stem cell-derived extracellular vesicle transplantation to repair the irradiated brain. Stem Cells Transl Med 2020; 9: 93-105. doi: 10.1002/sctm.18-0227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chakraborti A, Allen A, Allen B, Rosi S, Fike JR. Cranial irradiation alters dendritic spine density and morphology in the hippocampus. PLoS One 2012; 7: e40844. doi: 10.1371/journal.pone.0040844 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Belarbi K, Jopson T, Arellano C, Fike JR, Rosi S. Ccr2 deficiency prevents neuronal dysfunction and cognitive impairments induced by cranial irradiation. Cancer Res 2013; 73: 1201–10. doi: 10.1158/0008-5472.CAN-12-2989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003; 302: 1760–5. doi: 10.1126/science.1088417 [DOI] [PubMed] [Google Scholar]
  • 45.Schaue D, Micewicz ED, Ratikan JA, Xie MW, Cheng G, McBride WH. Radiation and inflammation. Semin Radiat Oncol 2015; 25: 4–10. doi: 10.1016/j.semradonc.2014.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hinkle JJ, Olschowka JA, Love TM, Williams JP, O'Banion MK. Cranial irradiation mediated spine loss is sex-specific and complement receptor-3 dependent in male mice. Sci Rep 2019; 9: 18899. doi: 10.1038/s41598-019-55366-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Acharya MM, Christie L-A, Lan ML, Giedzinski E, Fike JR, Rosi S, et al. Human neural stem cell transplantation ameliorates radiation-induced cognitive dysfunction. Cancer Res 2011; 71: 4834–45. doi: 10.1158/0008-5472.CAN-11-0027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Acharya MM, Christie L-A, Lan ML, Limoli CL. Comparing the functional consequences of human stem cell transplantation in the irradiated rat brain. Cell Transplant 2013; 22: 55–64. doi: 10.3727/096368912X640565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Acharya MM, Martirosian V, Christie L-A, Riparip L, Strnadel J, Parihar VK, et al. Defining the optimal window for cranial transplantation of human induced pluripotent stem cell-derived cells to ameliorate radiation-induced cognitive impairment. Stem Cells Transl Med 2015; 4: 74–83. doi: 10.5966/sctm.2014-0063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Acharya MM, Martirosian V, Parihar VK, Christie LA, Limoli CL. Analyzing the translational potential of transplanted iPS-Derived human neural stem cells for treating radiation-induced cognitive dysfunction. Cell Transplantation 2013; 22: 893–93.28845689 [Google Scholar]
  • 51.Acharya MM, Rosi S, Jopson T, Limoli CL. Human neural stem cell transplantation provides long-term restoration of neuronal plasticity in the irradiated hippocampus. Cell Transplant 2015; 24: 691–702. doi: 10.3727/096368914X684600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Baulch JE, Acharya MM, Allen BD, Ru N, Chmielewski NN, Martirosian V, et al. Cranial grafting of stem cell-derived microvesicles improves cognition and reduces neuropathology in the irradiated brain. Proc Natl Acad Sci U S A 2016; 113: 4836–41. doi: 10.1073/pnas.1521668113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Leavitt RJ, Acharya MM, Baulch JE, Limoli CL. Extracellular vesicle-derived miR-124 resolves radiation-induced brain injury. Cancer Res 2020;: canres.1599.202019 Aug 2020. doi: 10.1158/0008-5472.CAN-20-1599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Piao J, Major T, Auyeung G, Policarpio E, Menon J, Droms L, et al. Human embryonic stem cell-derived oligodendrocyte progenitors remyelinate the brain and rescue behavioral deficits following radiation. Cell Stem Cell 2015; 16: 198–210. doi: 10.1016/j.stem.2015.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Smith SM, Limoli CL. Stem cell therapies for the resolution of radiation injury to the brain. Curr Stem Cell Rep 2017; 3: 342–7. doi: 10.1007/s40778-017-0105-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Smith SM, Giedzinski E, Angulo MC, Lui T, Lu C, Park AL, et al. Functional equivalence of stem cell and stem cell-derived extracellular vesicle transplantation to repair the irradiated brain. Stem Cells Transl Med 2020; 9: 93–105. doi: 10.1002/sctm.18-0227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Leavitt RJ, Limoli CL, Baulch JE. miRNA-based therapeutic potential of stem cell-derived extracellular vesicles: a safe cell-free treatment to ameliorate radiation-induced brain injury. Int J Radiat Biol 2019; 95: 427–35. doi: 10.1080/09553002.2018.1522012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Acharya MM, Baulch JE, Lusardi TA, Allen BD, Chmielewski NN, Baddour AAD, et al. Adenosine kinase inhibition protects against cranial radiation-induced cognitive dysfunction. Front Mol Neurosci 2016; 9: 42. doi: 10.3389/fnmol.2016.00042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Acharya MM, Green KN, Allen BD, Najafi AR, Syage A, Minasyan H, et al. Elimination of microglia improves cognitive function following cranial irradiation. Sci Rep 2016; 6: 31545. doi: 10.1038/srep31545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Allen BD, Acharya MM, Lu C, Giedzinski E, Chmielewski NN, Quach D, et al. Remediation of radiation-induced cognitive dysfunction through oral administration of the neuroprotective compound NSI-189. Radiat Res 2018; 189: 345–53. doi: 10.1667/RR14879.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Harrington KJ. Ultrahigh dose-rate radiotherapy: next steps for FLASH-RT. Clin Cancer Res 2019; 25: 3–5. doi: 10.1158/1078-0432.CCR-18-1796 [DOI] [PubMed] [Google Scholar]
  • 62.Montay-Gruel P, Acharya MM, Petersson K, Alikhani L, Yakkala C, Allen BD, et al. Long-Term neurocognitive benefits of flash radiotherapy driven by reduced reactive oxygen species. Proc Natl Acad Sci U S A 2019; 116: 10943–51. doi: 10.1073/pnas.1901777116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alaghband Y, Cheeks SN, Allen BD, Montay-Gruel P, Doan N-L, Petit B, et al. Neuroprotection of radiosensitive juvenile mice by ultra-high dose rate flash irradiation. Cancers 2020; 12: 167124 06 2020. doi: 10.3390/cancers12061671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vozenin M-C, Hendry JH, Limoli CL. Biological benefits of ultra-high dose rate flash radiotherapy: sleeping Beauty Awoken. Clin Oncol 2019; 31: 407–15. doi: 10.1016/j.clon.2019.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fouillade C, Curras-Alonso S, Giuranno L, Quelennec E, Heinrich S, Bonnet-Boissinot S, et al. Flash irradiation spares lung progenitor cells and limits the incidence of Radio-induced senescence. Clin Cancer Res 2020; 26: 1497–506. doi: 10.1158/1078-0432.CCR-19-1440 [DOI] [PubMed] [Google Scholar]
  • 66.Favaudon V, Caplier L, Monceau V, Pouzoulet F, Sayarath M, Fouillade C, et al. Ultrahigh dose-rate flash irradiation increases the differential response between normal and tumor tissue in mice. Sci Transl Med 2014; 6: 245ra936(245):245ra93. doi: 10.1126/scitranslmed.3008973 [DOI] [PubMed] [Google Scholar]
  • 67.Vozenin M-C, De Fornel P, Petersson K, Favaudon V, Jaccard M, Germond J-F, et al. The advantage of flash radiotherapy confirmed in mini-pig and Cat-cancer patients. Clin Cancer Res 2019; 25: 35–42. doi: 10.1158/1078-0432.CCR-17-3375 [DOI] [PubMed] [Google Scholar]
  • 68.Montay-Gruel P, Meziani L, Yakkala C, Vozenin M-C. Expanding the therapeutic index of radiation therapy by normal tissue protection. Br J Radiol 2019; 92: 20180008. doi: 10.1259/bjr.20180008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Montay-Gruel P, Petersson K, Jaccard M, Boivin G, Germond J-F, Petit B, et al. Irradiation in a flash: unique sparing of memory in mice after whole brain irradiation with dose rates above 100Gy/s. Radiother Oncol 2017; 124: 365–9. doi: 10.1016/j.radonc.2017.05.003 [DOI] [PubMed] [Google Scholar]
  • 70.Bourhis J, Sozzi WJ, Jorge PG, Gaide O, Bailat C, Duclos F, et al. Treatment of a first patient with FLASH-radiotherapy. Radiother Oncol 2019; 139: 18–22. doi: 10.1016/j.radonc.2019.06.019 [DOI] [PubMed] [Google Scholar]
  • 71.Zeitlin C, Hassler DM, Cucinotta FA, Ehresmann B, Wimmer-Schweingruber RF, Brinza DE, et al. Measurements of energetic particle radiation in transit to Mars on the Mars science laboratory. Science 2013; 340: 1080–4. doi: 10.1126/science.1235989 [DOI] [PubMed] [Google Scholar]
  • 72.Parihar VK, Allen B, Tran KK, Macaraeg TG, Chu EM, Kwok SF, et al. What happens to your brain on the way to Mars. Sci Adv 2015; 1: e1400256. doi: 10.1126/sciadv.1400256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Parihar VK, Allen BD, Caressi C, Kwok S, Chu E, Tran KK, et al. Cosmic radiation exposure and persistent cognitive dysfunction. Sci Rep 2016; 6: 34774. doi: 10.1038/srep34774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Parihar VK, Maroso M, Syage A, Allen BD, Angulo MC, Soltesz I, et al. Persistent nature of alterations in cognition and neuronal circuit excitability after exposure to simulated COSMIC radiation in mice. Exp Neurol 2018; 305: 44–55. doi: 10.1016/j.expneurol.2018.03.009 [DOI] [PubMed] [Google Scholar]
  • 75.Britten RA, Davis LK, Jewell JS, Miller VD, Hadley MM, Sanford LD, et al. Exposure to mission relevant doses of 1 GeV/Nucleon (56)Fe particles leads to impairment of attentional set-shifting performance in socially mature rats. Radiat Res 2014; 182: 292–8. doi: 10.1667/RR3766.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Britten RA, Jewell JS, Miller VD, Davis LK, Hadley MM, Wyrobek AJ. Impaired Spatial Memory Performance in Adult Wistar Rats Exposed to Low (5-20 cGy) Doses of 1 GeV/n (56)Fe Particles. Radiat Res 2016; 185: 332–7. doi: 10.1667/RR14120.1 [DOI] [PubMed] [Google Scholar]
  • 77.Britten RA, Miller VD, Hadley MM, Jewell JS, Macadat E, hippocampus- Pin. Performance in hippocampus- and PFC-dependent cognitive domains are not concomitantly impaired in rats exposed to 20cGy of 1GeV/n 56Fe particles. Life Sci Space Res 2016; 10: 17–22. doi: 10.1016/j.lssr.2016.06.005 [DOI] [PubMed] [Google Scholar]
  • 78.Jewell JS, Duncan VD, Fesshaye A, Tondin A, Macadat E, Britten RA. Exposure to ≤15 cGy of 600 MeV/n 56Fe Particles Impairs Rule Acquisition but not Long-Term Memory in the Attentional Set-Shifting Assay. Radiat Res 2018; 190: 565–75. doi: 10.1667/RR15085.1 [DOI] [PubMed] [Google Scholar]
  • 79.Cacao E, Parihar VK, Limoli CL, Cucinotta FA. Stochastic modeling of radiation-induced dendritic damage on in silico mouse hippocampal neurons. Sci Rep 2018; 8: 5494. doi: 10.1038/s41598-018-23855-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Carr H, Alexander TC, Groves T, Kiffer F, Wang J, Price E, et al. Early effects of 16O radiation on neuronal morphology and cognition in a murine model. Life Sci Space Res 2018; 17: 63–73. doi: 10.1016/j.lssr.2018.03.001 [DOI] [PubMed] [Google Scholar]
  • 81.Whoolery CW, Walker AK, Richardson DR, Lucero MJ, Reynolds RP, Beddow DH, et al. Whole-Body Exposure to 28Si-Radiation Dose-Dependently Disrupts Dentate Gyrus Neurogenesis and Proliferation in the Short Term and New Neuron Survival and Contextual Fear Conditioning in the Long Term. Radiat Res 2017; 188: 612–31. doi: 10.1667/RR14797.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Britten RA, Duncan VD, Fesshaye A, Rudobeck E, Nelson GA, Vlkolinsky R. Altered Cognitive Flexibility and Synaptic Plasticity in the Rat Prefrontal Cortex after Exposure to Low (≤15 cGy) Doses of 28Si Radiation. Radiat Res 2020; 193: 223–35. doi: 10.1667/RR15458.1 [DOI] [PubMed] [Google Scholar]
  • 83.Davis CM, DeCicco-Skinner KL, Hienz RD. Deficits in sustained attention and changes in dopaminergic protein levels following exposure to proton radiation are related to basal dopaminergic function. PLoS One 2015; 10: e0144556. doi: 10.1371/journal.pone.0144556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Davis CM, DeCicco-Skinner KL, Roma PG, Hienz RD. Individual differences in attentional deficits and dopaminergic protein levels following exposure to proton radiation. Radiat Res 2014; 181: 258–71. doi: 10.1667/RR13359.1 [DOI] [PubMed] [Google Scholar]
  • 85.Allen AR, Raber J, Chakraborti A, Sharma S, Fike JR. 56)Fe Irradiation Alters Spine Density and Dendritic Complexity in the Mouse Hippocampus. Radiat Res 2015; 184: 586–94. doi: 10.1667/RR14103.1 [DOI] [PubMed] [Google Scholar]
  • 86.Dickstein DL, Talty R, Bresnahan E, Varghese M, Perry B, Janssen WGM, et al. Alterations in synaptic density and myelination in response to exposure to high-energy charged particles. J Comp Neurol 2018; 526: 2845–55. doi: 10.1002/cne.24530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Rola R, Fishman K, Baure J, Rosi S, Lamborn KR, Obenaus A, et al. Hippocampal neurogenesis and neuroinflammation after cranial irradiation with (56)Fe particles. Radiat Res 2008; 169: 626–32. doi: 10.1667/RR1263.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rola R, Sarkissian V, Obenaus A, Nelson GA, Otsuka S, Limoli CL, et al. High-LET radiation induces inflammation and persistent changes in markers of hippocampal neurogenesis. Radiat Res 2005; 164(4 Pt 2): 556–60. doi: 10.1667/RR3412.1 [DOI] [PubMed] [Google Scholar]
  • 89.Tseng BP, Giedzinski E, Izadi A, Suarez T, Lan ML, Tran KK, et al. Functional consequences of radiation-induced oxidative stress in cultured neural stem cells and the brain exposed to charged particle irradiation. Antioxid Redox Signal 2014; 20: 1410–22. doi: 10.1089/ars.2012.5134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011; 333: 1456–8. doi: 10.1126/science.1202529 [DOI] [PubMed] [Google Scholar]
  • 91.Ekdahl CT. Microglial activation - tuning and pruning adult neurogenesis. Front Pharmacol 2012; 3: 41. doi: 10.3389/fphar.2012.00041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hong S, Dissing-Olesen L, Stevens B. New insights on the role of microglia in synaptic pruning in health and disease. Curr Opin Neurobiol 2016; 36: 128–34. doi: 10.1016/j.conb.2015.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Krukowski K, Feng X, Paladini MS, Chou A, Sacramento K, Grue K, et al. Temporary microglia-depletion after COSMIC radiation modifies phagocytic activity and prevents cognitive deficits. Sci Rep 2018; 8: 7857. doi: 10.1038/s41598-018-26039-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Acharya MM, Baulch JE, Klein PM, Baddour AAD, Apodaca LA, Kramár EA, et al. New concerns for neurocognitive function during deep space exposures to chronic, low dose-rate, neutron radiation. eNeuro 2019; 6: ENEURO.0094-19.201922 08 2019. doi: 10.1523/ENEURO.0094-19.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Limoli C. Your brain on Mars. Radiat Res 2015; 184: 1–2. doi: 10.1667/RR14143.1 [DOI] [PubMed] [Google Scholar]
  • 96.Limoli CL. Deep-Space deal breaker. Sci Am 2017; 316: 54–9. doi: 10.1038/scientificamerican0217-54 [DOI] [PubMed] [Google Scholar]
  • 97.Roberts DR, Albrecht MH, Collins HR, Asemani D, Chatterjee AR, Spampinato MV, et al. Effects of spaceflight on astronaut brain structure as indicated on MRI. N Engl J Med 2017; 377: 1746–53. doi: 10.1056/NEJMoa1705129 [DOI] [PubMed] [Google Scholar]
  • 98.Garrett-Bakelman FE, Darshi M, Green SJ, Gur RC, Lin L, Macias BR, et al. The NASA twins study: a multidimensional analysis of a year-long human spaceflight. Science 2019; 36412 04 2019. doi: 10.1126/science.aau8650 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The British Journal of Radiology are provided here courtesy of Oxford University Press

RESOURCES