Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2024

Open Access 01-12-2024 | Research

Cranial irradiation disrupts homeostatic microglial dynamic behavior

Authors: Alexandra O. Strohm, Carl Johnston, Eric Hernady, Brian Marples, M. Kerry O’Banion, Ania K. Majewska

Published in: Journal of Neuroinflammation | Issue 1/2024

Login to get access

Abstract

Cranial irradiation causes cognitive deficits that are in part mediated by microglia, the resident immune cells of the brain. Microglia are highly reactive, exhibiting changes in shape and morphology depending on the function they are performing. Additionally, microglia processes make dynamic, physical contacts with different components of their environment to monitor the functional state of the brain and promote plasticity. Though evidence suggests radiation perturbs homeostatic microglia functions, it is unknown how cranial irradiation impacts the dynamic behavior of microglia over time. Here, we paired in vivo two-photon microscopy with a transgenic mouse model that labels cortical microglia to follow these cells and determine how they change over time in cranial irradiated mice and their control littermates. We show that a single dose of 10 Gy cranial irradiation disrupts homeostatic cortical microglia dynamics during a 1-month time course. We found a lasting loss of microglial cells following cranial irradiation, coupled with a modest dysregulation of microglial soma displacement at earlier timepoints. The homogeneous distribution of microglia was maintained, suggesting microglia rearrange themselves to account for cell loss and maintain territorial organization following cranial irradiation. Furthermore, we found cranial irradiation reduced microglia coverage of the parenchyma and their surveillance capacity, without overtly changing morphology. Our results demonstrate that a single dose of radiation can induce changes in microglial behavior and function that could influence neurological health. These results set the foundation for future work examining how cranial irradiation impacts complex cellular dynamics in the brain which could contribute to the manifestation of cognitive deficits.
Appendix
Available only for authorised users
Literature
1.
go back to reference Pazzaglia S, et al. Neurocognitive decline following radiotherapy: mechanisms and therapeutic implications. Cancers (Basel). 2020;12(1):146.PubMedCrossRef Pazzaglia S, et al. Neurocognitive decline following radiotherapy: mechanisms and therapeutic implications. Cancers (Basel). 2020;12(1):146.PubMedCrossRef
2.
go back to reference Delaney G, et al. The role of radiotherapy in cancer treatment: estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer. 2005;104(6):1129–37.PubMedCrossRef Delaney G, et al. The role of radiotherapy in cancer treatment: estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer. 2005;104(6):1129–37.PubMedCrossRef
3.
go back to reference Turnquist C, Harris BT, Harris CC. Radiation-induced brain injury: current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv. 2020;2(1):vdaa057.PubMedPubMedCentral Turnquist C, Harris BT, Harris CC. Radiation-induced brain injury: current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv. 2020;2(1):vdaa057.PubMedPubMedCentral
4.
go back to reference Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov. 2013;12(7):526–42.PubMedPubMedCentralCrossRef Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov. 2013;12(7):526–42.PubMedPubMedCentralCrossRef
6.
go back to reference Lee YW, et al. Whole brain radiation-induced cognitive impairment: pathophysiological mechanisms and therapeutic targets. Biomol Ther (Seoul). 2012;20(4):357–70.PubMedCrossRef Lee YW, et al. Whole brain radiation-induced cognitive impairment: pathophysiological mechanisms and therapeutic targets. Biomol Ther (Seoul). 2012;20(4):357–70.PubMedCrossRef
7.
go back to reference Rapp SR, et al. Donepezil for irradiated brain tumor survivors: a phase III randomized placebo-controlled clinical trial. J Clin Oncol. 2015;33(15):1653–9.PubMedPubMedCentralCrossRef Rapp SR, et al. Donepezil for irradiated brain tumor survivors: a phase III randomized placebo-controlled clinical trial. J Clin Oncol. 2015;33(15):1653–9.PubMedPubMedCentralCrossRef
8.
go back to reference Nieder C, et al. Late radiation toxicity after whole brain radiotherapy: the influence of antiepileptic drugs. Am J Clin Oncol. 1999;22(6):573–9.PubMedCrossRef Nieder C, et al. Late radiation toxicity after whole brain radiotherapy: the influence of antiepileptic drugs. Am J Clin Oncol. 1999;22(6):573–9.PubMedCrossRef
10.
go back to reference Feng X, et al. Colony-stimulating factor 1 receptor blockade prevents fractionated whole-brain irradiation-induced memory deficits. J Neuroinflamm. 2016;13(1):215.CrossRef Feng X, et al. Colony-stimulating factor 1 receptor blockade prevents fractionated whole-brain irradiation-induced memory deficits. J Neuroinflamm. 2016;13(1):215.CrossRef
11.
go back to reference Bhat K, et al. 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine treatment after brain irradiation preserves cognitive function in mice. Neuro Oncol. 2020;22(10):1484–94.PubMedPubMedCentralCrossRef Bhat K, et al. 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine treatment after brain irradiation preserves cognitive function in mice. Neuro Oncol. 2020;22(10):1484–94.PubMedPubMedCentralCrossRef
13.
go back to reference Allen BD, et al. Mitigation of helium irradiation-induced brain injury by microglia depletion. J Neuroinflamm. 2020;17(1):159.CrossRef Allen BD, et al. Mitigation of helium irradiation-induced brain injury by microglia depletion. J Neuroinflamm. 2020;17(1):159.CrossRef
14.
go back to reference Markarian M, et al. Glia-selective deletion of complement C1q prevents radiation-induced cognitive deficits and neuroinflammation. Cancer Res. 2021;81(7):1732–44.PubMedCrossRef Markarian M, et al. Glia-selective deletion of complement C1q prevents radiation-induced cognitive deficits and neuroinflammation. Cancer Res. 2021;81(7):1732–44.PubMedCrossRef
15.
17.
go back to reference Krukowski K, et al. Temporary microglia-depletion after cosmic radiation modifies phagocytic activity and prevents cognitive deficits. Sci Rep. 2018;8(1):7857.PubMedPubMedCentralCrossRef Krukowski K, et al. Temporary microglia-depletion after cosmic radiation modifies phagocytic activity and prevents cognitive deficits. Sci Rep. 2018;8(1):7857.PubMedPubMedCentralCrossRef
18.
go back to reference Jenrow KA, et al. Selective inhibition of microglia-mediated neuroinflammation mitigates radiation-induced cognitive impairment. Radiat Res. 2013;179(5):549–56.PubMedPubMedCentralCrossRef Jenrow KA, et al. Selective inhibition of microglia-mediated neuroinflammation mitigates radiation-induced cognitive impairment. Radiat Res. 2013;179(5):549–56.PubMedPubMedCentralCrossRef
19.
go back to reference Kalm M, et al. C3 deficiency ameliorates the negative effects of irradiation of the young brain on hippocampal development and learning. Oncotarget. 2016;7(15):19382–94.PubMedPubMedCentralCrossRef Kalm M, et al. C3 deficiency ameliorates the negative effects of irradiation of the young brain on hippocampal development and learning. Oncotarget. 2016;7(15):19382–94.PubMedPubMedCentralCrossRef
20.
go back to reference Han W, et al. Cranial irradiation induces transient microglia accumulation, followed by long-lasting inflammation and loss of microglia. Oncotarget. 2016;7(50):82305–23.PubMedPubMedCentralCrossRef Han W, et al. Cranial irradiation induces transient microglia accumulation, followed by long-lasting inflammation and loss of microglia. Oncotarget. 2016;7(50):82305–23.PubMedPubMedCentralCrossRef
21.
go back to reference Constanzo J, et al. Brain irradiation leads to persistent neuroinflammation and long-term neurocognitive dysfunction in a region-specific manner. Prog Neuropsychopharmacol Biol Psychiatry. 2020;102: 109954.PubMedCrossRef Constanzo J, et al. Brain irradiation leads to persistent neuroinflammation and long-term neurocognitive dysfunction in a region-specific manner. Prog Neuropsychopharmacol Biol Psychiatry. 2020;102: 109954.PubMedCrossRef
22.
go back to reference Osman AM, et al. Radiation triggers a dynamic sequence of transient microglial alterations in juvenile brain. Cell Rep. 2020;31(9): 107699.PubMedCrossRef Osman AM, et al. Radiation triggers a dynamic sequence of transient microglial alterations in juvenile brain. Cell Rep. 2020;31(9): 107699.PubMedCrossRef
23.
24.
26.
go back to reference Zhang D, et al. Cranial irradiation induces axon initial segment dysfunction and neuronal injury in the prefrontal cortex and impairs hippocampal coupling. Neurooncol Adv. 2020;2(1):vdaa058.PubMedPubMedCentral Zhang D, et al. Cranial irradiation induces axon initial segment dysfunction and neuronal injury in the prefrontal cortex and impairs hippocampal coupling. Neurooncol Adv. 2020;2(1):vdaa058.PubMedPubMedCentral
28.
go back to reference Lee H-J, et al. Profiling of gene expression in the brain associated with anxiety-related behaviors in the chronic phase following cranial irradiation. Sci Rep. 2022;12(1):13162.PubMedPubMedCentralCrossRef Lee H-J, et al. Profiling of gene expression in the brain associated with anxiety-related behaviors in the chronic phase following cranial irradiation. Sci Rep. 2022;12(1):13162.PubMedPubMedCentralCrossRef
29.
go back to reference Franco-Perez J, et al. Whole-brain irradiation differentially modifies neurotransmitters levels and receptors in the hypothalamus and the prefrontal cortex. Radiat Oncol. 2020;15(1):269.PubMedPubMedCentralCrossRef Franco-Perez J, et al. Whole-brain irradiation differentially modifies neurotransmitters levels and receptors in the hypothalamus and the prefrontal cortex. Radiat Oncol. 2020;15(1):269.PubMedPubMedCentralCrossRef
30.
go back to reference Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308(5726):1314–8.PubMedCrossRef Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308(5726):1314–8.PubMedCrossRef
31.
32.
go back to reference Wake H, et al. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J Neurosci. 2009;29(13):3974–80.PubMedPubMedCentralCrossRef Wake H, et al. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J Neurosci. 2009;29(13):3974–80.PubMedPubMedCentralCrossRef
33.
34.
go back to reference Paolicelli RC, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048):1456–8.PubMedCrossRef Paolicelli RC, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048):1456–8.PubMedCrossRef
36.
go back to reference Damani MR, et al. Age-related alterations in the dynamic behavior of microglia. Aging Cell. 2011;10(2):263–76.PubMedCrossRef Damani MR, et al. Age-related alterations in the dynamic behavior of microglia. Aging Cell. 2011;10(2):263–76.PubMedCrossRef
37.
go back to reference Davies DS, et al. Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer’s disease. Brain Pathol. 2017;27(6):795–808.PubMedCrossRef Davies DS, et al. Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer’s disease. Brain Pathol. 2017;27(6):795–808.PubMedCrossRef
38.
go back to reference Hefendehl JK, et al. Homeostatic and injury-induced microglia behavior in the aging brain. Aging Cell. 2014;13(1):60–9.PubMedCrossRef Hefendehl JK, et al. Homeostatic and injury-induced microglia behavior in the aging brain. Aging Cell. 2014;13(1):60–9.PubMedCrossRef
40.
go back to reference Gyoneva S, et al. Altered motility of plaque-associated microglia in a model of Alzheimer’s disease. Neuroscience. 2016;330:410–20.PubMedCrossRef Gyoneva S, et al. Altered motility of plaque-associated microglia in a model of Alzheimer’s disease. Neuroscience. 2016;330:410–20.PubMedCrossRef
41.
go back to reference Mendes MS, et al. The role of P2Y12 in the kinetics of microglial self-renewal and maturation in the adult visual cortex in vivo. Elife. 2021;10: e61173.PubMedPubMedCentralCrossRef Mendes MS, et al. The role of P2Y12 in the kinetics of microglial self-renewal and maturation in the adult visual cortex in vivo. Elife. 2021;10: e61173.PubMedPubMedCentralCrossRef
43.
go back to reference Fuger P, et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat Neurosci. 2017;20(10):1371–6.PubMedCrossRef Fuger P, et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat Neurosci. 2017;20(10):1371–6.PubMedCrossRef
47.
go back to reference Liu B, et al. Space-like (56)Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer’s-like transgenic mice. Sci Rep. 2019;9(1):12118.PubMedPubMedCentralCrossRef Liu B, et al. Space-like (56)Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer’s-like transgenic mice. Sci Rep. 2019;9(1):12118.PubMedPubMedCentralCrossRef
51.
go back to reference Ziebell JM, et al. Rod microglia: elongation, alignment, and coupling to form trains across the somatosensory cortex after experimental diffuse brain injury. J Neuroinflamm. 2012;9:247.CrossRef Ziebell JM, et al. Rod microglia: elongation, alignment, and coupling to form trains across the somatosensory cortex after experimental diffuse brain injury. J Neuroinflamm. 2012;9:247.CrossRef
52.
go back to reference Savage JC, et al. Microglial physiological properties and interactions with synapses are altered at presymptomatic stages in a mouse model of Huntington’s disease pathology. J Neuroinflamm. 2020;17(1):98.CrossRef Savage JC, et al. Microglial physiological properties and interactions with synapses are altered at presymptomatic stages in a mouse model of Huntington’s disease pathology. J Neuroinflamm. 2020;17(1):98.CrossRef
53.
go back to reference Bordeleau M, et al. Maternal high-fat diet in mice induces cerebrovascular, microglial and long-term behavioural alterations in offspring. Commun Biol. 2022;5(1):26.PubMedPubMedCentralCrossRef Bordeleau M, et al. Maternal high-fat diet in mice induces cerebrovascular, microglial and long-term behavioural alterations in offspring. Commun Biol. 2022;5(1):26.PubMedPubMedCentralCrossRef
55.
57.
go back to reference Son Y, et al. Hippocampal dysfunction during the chronic phase following a single exposure to cranial irradiation. Exp Neurol. 2014;254:134–44.PubMedCrossRef Son Y, et al. Hippocampal dysfunction during the chronic phase following a single exposure to cranial irradiation. Exp Neurol. 2014;254:134–44.PubMedCrossRef
58.
60.
go back to reference Kluge MG, et al. Spatiotemporal analysis of impaired microglia process movement at sites of secondary neurodegeneration post-stroke. J Cereb Blood Flow Metab. 2019;39(12):2456–70.PubMedCrossRef Kluge MG, et al. Spatiotemporal analysis of impaired microglia process movement at sites of secondary neurodegeneration post-stroke. J Cereb Blood Flow Metab. 2019;39(12):2456–70.PubMedCrossRef
61.
62.
go back to reference Moravan MJ, et al. Cranial irradiation leads to acute and persistent neuroinflammation with delayed increases in T-cell infiltration and CD11c expression in C57BL/6 mouse brain. Radiat Res. 2011;176(4):459–73.PubMedPubMedCentralCrossRef Moravan MJ, et al. Cranial irradiation leads to acute and persistent neuroinflammation with delayed increases in T-cell infiltration and CD11c expression in C57BL/6 mouse brain. Radiat Res. 2011;176(4):459–73.PubMedPubMedCentralCrossRef
63.
go back to reference Monje ML, et al. Irradiation induces neural precursor-cell dysfunction. Nat Med. 2002;8(9):955–62.PubMedCrossRef Monje ML, et al. Irradiation induces neural precursor-cell dysfunction. Nat Med. 2002;8(9):955–62.PubMedCrossRef
65.
go back to reference Kalambogias J, et al. Development and sensory experience dependent regulation of microglia in barrel cortex. J Comp Neurol. 2020;528(4):559–73.PubMedCrossRef Kalambogias J, et al. Development and sensory experience dependent regulation of microglia in barrel cortex. J Comp Neurol. 2020;528(4):559–73.PubMedCrossRef
67.
68.
69.
go back to reference Seibert TM, et al. Cerebral cortex regions selectively vulnerable to radiation dose-dependent atrophy. Int J Radiat Oncol Biol Phys. 2017;97(5):910–8.PubMedPubMedCentralCrossRef Seibert TM, et al. Cerebral cortex regions selectively vulnerable to radiation dose-dependent atrophy. Int J Radiat Oncol Biol Phys. 2017;97(5):910–8.PubMedPubMedCentralCrossRef
70.
go back to reference Nagtegaal SHJ, et al. Changes in cortical thickness and volume after cranial radiation treatment: a systematic review. Radiother Oncol. 2019;135:33–42.PubMedCrossRef Nagtegaal SHJ, et al. Changes in cortical thickness and volume after cranial radiation treatment: a systematic review. Radiother Oncol. 2019;135:33–42.PubMedCrossRef
71.
go back to reference Lin J, et al. Radiation-induced abnormal cortical thickness in patients with nasopharyngeal carcinoma after radiotherapy. Neuroimage Clin. 2017;14:610–21.PubMedPubMedCentralCrossRef Lin J, et al. Radiation-induced abnormal cortical thickness in patients with nasopharyngeal carcinoma after radiotherapy. Neuroimage Clin. 2017;14:610–21.PubMedPubMedCentralCrossRef
72.
go back to reference Yabluchanskiy A, et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience. 2020;42(2):409–28.PubMedPubMedCentralCrossRef Yabluchanskiy A, et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience. 2020;42(2):409–28.PubMedPubMedCentralCrossRef
73.
go back to reference Li B, et al. Measurements of cerebral microvascular blood flow, oxygenation, and morphology in a mouse model of whole-brain irradiation-induced cognitive impairment by two-photon microscopy and optical coherence tomography: evidence for microvascular injury in the cerebral white matter. Geroscience. 2023;45(3):1491–510.PubMedPubMedCentralCrossRef Li B, et al. Measurements of cerebral microvascular blood flow, oxygenation, and morphology in a mouse model of whole-brain irradiation-induced cognitive impairment by two-photon microscopy and optical coherence tomography: evidence for microvascular injury in the cerebral white matter. Geroscience. 2023;45(3):1491–510.PubMedPubMedCentralCrossRef
74.
go back to reference Ungvari Z, et al. Cerebromicrovascular dysfunction predicts cognitive decline and gait abnormalities in a mouse model of whole brain irradiation-induced accelerated brain senescence. Geroscience. 2017;39(1):33–42.PubMedPubMedCentralCrossRef Ungvari Z, et al. Cerebromicrovascular dysfunction predicts cognitive decline and gait abnormalities in a mouse model of whole brain irradiation-induced accelerated brain senescence. Geroscience. 2017;39(1):33–42.PubMedPubMedCentralCrossRef
75.
go back to reference Hwang SY, et al. Ionizing radiation induces astrocyte gliosis through microglia activation. Neurobiol Dis. 2006;21(3):457–67.PubMedCrossRef Hwang SY, et al. Ionizing radiation induces astrocyte gliosis through microglia activation. Neurobiol Dis. 2006;21(3):457–67.PubMedCrossRef
76.
go back to reference Masuda T, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566(7744):388–92.PubMedCrossRef Masuda T, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566(7744):388–92.PubMedCrossRef
77.
go back to reference Wang J, et al. Neuroprotective effect of fractalkine on radiation-induced brain injury through promoting the M2 polarization of microglia. Mol Neurobiol. 2021;58(3):1074–87.PubMedCrossRef Wang J, et al. Neuroprotective effect of fractalkine on radiation-induced brain injury through promoting the M2 polarization of microglia. Mol Neurobiol. 2021;58(3):1074–87.PubMedCrossRef
78.
go back to reference Masuda T, et al. Novel Hexb-based tools for studying microglia in the CNS. Nat Immunol. 2020;21(7):802–15.PubMedCrossRef Masuda T, et al. Novel Hexb-based tools for studying microglia in the CNS. Nat Immunol. 2020;21(7):802–15.PubMedCrossRef
79.
go back to reference Hirasawa T, et al. Visualization of microglia in living tissues using Iba1-EGFP transgenic mice. J Neurosci Res. 2005;81(3):357–62.PubMedCrossRef Hirasawa T, et al. Visualization of microglia in living tissues using Iba1-EGFP transgenic mice. J Neurosci Res. 2005;81(3):357–62.PubMedCrossRef
80.
go back to reference Moravan MJ, et al. Brain radiation injury leads to a dose- and time-dependent recruitment of peripheral myeloid cells that depends on CCR2 signaling. J Neuroinflamm. 2016;13:30.CrossRef Moravan MJ, et al. Brain radiation injury leads to a dose- and time-dependent recruitment of peripheral myeloid cells that depends on CCR2 signaling. J Neuroinflamm. 2016;13:30.CrossRef
81.
go back to reference Lee SW, et al. Absence of CCL2 is sufficient to restore hippocampal neurogenesis following cranial irradiation. Brain Behav Immun. 2013;30:33–44.PubMedCrossRef Lee SW, et al. Absence of CCL2 is sufficient to restore hippocampal neurogenesis following cranial irradiation. Brain Behav Immun. 2013;30:33–44.PubMedCrossRef
82.
go back to reference Hanamsagar R, et al. Generation of a microglial developmental index in mice and in humans reveals a sex difference in maturation and immune reactivity. Glia. 2017;65(9):1504–20.PubMedPubMedCentralCrossRef Hanamsagar R, et al. Generation of a microglial developmental index in mice and in humans reveals a sex difference in maturation and immune reactivity. Glia. 2017;65(9):1504–20.PubMedPubMedCentralCrossRef
83.
go back to reference Han J, et al. Uncovering sex differences of rodent microglia. J Neuroinflamm. 2021;18(1):74.CrossRef Han J, et al. Uncovering sex differences of rodent microglia. J Neuroinflamm. 2021;18(1):74.CrossRef
84.
go back to reference Roughton K, Kalm M, Blomgren K. Sex-dependent differences in behavior and hippocampal neurogenesis after irradiation to the young mouse brain. Eur J Neurosci. 2012;36(6):2763–72.PubMedCrossRef Roughton K, Kalm M, Blomgren K. Sex-dependent differences in behavior and hippocampal neurogenesis after irradiation to the young mouse brain. Eur J Neurosci. 2012;36(6):2763–72.PubMedCrossRef
85.
go back to reference Raber J, et al. Radiation-induced cognitive impairments are associated with changes in indicators of hippocampal neurogenesis. Radiat Res. 2004;162(1):39–47.PubMedCrossRef Raber J, et al. Radiation-induced cognitive impairments are associated with changes in indicators of hippocampal neurogenesis. Radiat Res. 2004;162(1):39–47.PubMedCrossRef
86.
go back to reference Rola R, et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol. 2004;188(2):316–30.PubMedCrossRef Rola R, et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol. 2004;188(2):316–30.PubMedCrossRef
87.
go back to reference Jung S, et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol. 2000;20(11):4106–14.PubMedPubMedCentralCrossRef Jung S, et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol. 2000;20(11):4106–14.PubMedPubMedCentralCrossRef
89.
go back to reference Stowell RD, Majewska AK. Acute ethanol exposure rapidly alters cerebellar and cortical microglial physiology. Eur J Neurosci. 2021;54(5):5834–43.PubMedCrossRef Stowell RD, Majewska AK. Acute ethanol exposure rapidly alters cerebellar and cortical microglial physiology. Eur J Neurosci. 2021;54(5):5834–43.PubMedCrossRef
90.
go back to reference Stowell RD, et al. Noradrenergic signaling in the wakeful state inhibits microglial surveillance and synaptic plasticity in the mouse visual cortex. Nat Neurosci. 2019;22(11):1782–92.PubMedPubMedCentralCrossRef Stowell RD, et al. Noradrenergic signaling in the wakeful state inhibits microglial surveillance and synaptic plasticity in the mouse visual cortex. Nat Neurosci. 2019;22(11):1782–92.PubMedPubMedCentralCrossRef
Metadata
Title
Cranial irradiation disrupts homeostatic microglial dynamic behavior
Authors
Alexandra O. Strohm
Carl Johnston
Eric Hernady
Brian Marples
M. Kerry O’Banion
Ania K. Majewska
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2024
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-024-03073-z

Other articles of this Issue 1/2024

Journal of Neuroinflammation 1/2024 Go to the issue