Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5512, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951525

RESUMEN

Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.


Asunto(s)
Electrodos Implantados , Microelectrodos , Microglía , Ondas Ultrasónicas , Microglía/efectos de la radiación , Microglía/metabolismo , Animales , Masculino , Reacción a Cuerpo Extraño/prevención & control , Reacción a Cuerpo Extraño/etiología , Ratones , Corteza Cerebral/efectos de la radiación , Corteza Cerebral/citología , Interfaces Cerebro-Computador , Movimiento Celular/efectos de la radiación , Ratas
2.
J Alzheimers Dis ; 100(s1): S327-S339, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39058444

RESUMEN

Background: Space radiation was linked to neurological damage and behavioral deficits which raised concerns of increased degenerative risk on the brain and development of Alzheimer's disease following space travel. Objective: In this study, we investigated the effects of irradiation by 56Fe and 28Si in CRND8 mice, an Alzheimer's disease mouse model. Methods: Six-month-old CRND8 mice were exposed to whole body irradiation by 56Fe and 28Si at 0.5 Gy and 2 Gy doses. Behavior tests were administered 1-month to 3-months post-irradiation. Amyloid deposition and other pathological changes were analyzed 3-months and/or 6-months post-irradiation. Results: The Novel Object Recognition test showed some decline in 8-month-old mice compared to non-irradiated CRND8 mice. Male mice also showed a loss of freezing behavior in the fear conditioning contextual test following irradiation. Golgi staining revealed a loss of spines in hippocampal neurons after irradiation. Total amyloid immunohistochemistry showed a robust increase in 3-months post-irradiation 56Fe groups which became normalized to non-irradiated group by 6-months post-irradiation. However, 2 Gy 28Si caused a trend towards increased plaque load at 3-months post-irradiation which became significant at 6-months post irradiation only in male CRND8 mice. While 0.5 Gy Fe did not induce obvious changes in the total number of iba-1 positive microglia, more hippocampal microglia were found to express PCNA after 0.5 Gy Fe treatment, suggesting potential involvement of microglial dysfunction. Conclusions: Overall, our study provides new evidence of gender-specific and ion-dependent effects of space radiation on cognition and amyloid pathology in AD models.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Animales , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Masculino , Ratones , Irradiación Corporal Total/efectos adversos , Reconocimiento en Psicología/efectos de la radiación , Hipocampo/efectos de la radiación , Hipocampo/patología , Hipocampo/metabolismo , Miedo/efectos de la radiación , Miedo/psicología , Femenino , Ratones Transgénicos , Encéfalo/efectos de la radiación , Encéfalo/patología , Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Disfunción Cognitiva/metabolismo , Microglía/efectos de la radiación , Microglía/patología , Microglía/metabolismo , Radiación Cósmica/efectos adversos , Placa Amiloide/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo
3.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38831600

RESUMEN

PURPOSE: Radiation-induced brain injury, one of the side effects of cranial radiotherapy in tumour patients, usually results in durable and serious cognitive disorders. Microglia are important innate immune-effector cells in the central nervous system. However, the interaction between microglia and neurons in radiation-induced brain injury remains uncharacterised. METHODS AND MATERIALS: We established a microglia-neuron indirect co-culture model to assess the interaction between them. Microglia exposed to radiation were examined for pyroptosis using lactate dehydrogenase (LDH) release, Annexin V/PI staining, SYTOX staining and western blot. The role of nucleotide-binding oligomerisation domain-like receptor family pyrin domain containing 3 (NLRP3) was investigated in microglia exposed to radiation and in mouse radiation brain injury model through siRNA or inhibitor. Mini-mental state examination and cytokines in blood were performed in 23 patients who had experienced cranial irradiation. RESULTS: Microglia exerted neurotoxic features after radiation in the co-culture model. NLRP3 was up-regulated in microglia exposed to radiation, and then caspase-1 was activated. Thus, the gasdermin D protein was cleaved, and it triggered pyroptosis in microglia, which released inflammatory cytokines. Meanwhile, treatment with siRNA NLRP3 in vitro and NLRP3 inhibitor in vivo attenuated the damaged neuron cell and cognitive impairment, respectively. What is more, we found that the patients after radiation with higher IL-6 were observed to have a decreased MMSE score. CONCLUSIONS: These findings indicate that radiation-induced pyroptosis in microglia may promote radiation-induced brain injury via the secretion of neurotoxic cytokines. NLRP3 was evaluated as an important mediator in radiation-induced pyroptosis and a promising therapeutic target for radiation-induced brain injury.


Asunto(s)
Lesiones Encefálicas , Microglía , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Piroptosis/efectos de la radiación , Piroptosis/fisiología , Microglía/metabolismo , Microglía/efectos de la radiación , Microglía/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Ratones , Humanos , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Lesiones Encefálicas/etiología , Masculino , Neuronas/metabolismo , Neuronas/patología , Neuronas/efectos de la radiación , Técnicas de Cocultivo , Traumatismos por Radiación/patología , Traumatismos por Radiación/metabolismo , Femenino , Ratones Endogámicos C57BL , Persona de Mediana Edad
4.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892109

RESUMEN

Astronauts on exploratory missions will be exposed to galactic cosmic rays (GCR), which can induce neuroinflammation and oxidative stress (OS) and may increase the risk of neurodegenerative disease. As key regulators of inflammation and OS in the CNS, microglial cells may be involved in GCR-induced deficits, and therefore could be a target for neuroprotection. This study assessed the effects of exposure to helium (4He) and iron (56Fe) particles on inflammation and OS in microglia in vitro, to establish a model for testing countermeasure efficacy. Rat microglia were exposed to a single dose of 20 cGy (300 MeV/n) 4He or 2 Gy 56Fe (600 MeV/n), while the control cells were not exposed (0 cGy). Immediately following irradiation, fresh media was applied to the cells, and biomarkers of inflammation (cyclooxygenase-2 [COX-2], nitric oxide synthase [iNOS], phosphorylated IκB-α [pIκB-α], tumor necrosis factor-α [TNFα], and nitrite [NO2-]) and OS (NADPH oxidase [NOX2]) were assessed 24 h later using standard immunochemical techniques. Results showed that radiation did not increase levels of NO2- or protein levels of COX-2, iNOS, pIκB-α, TNFα, or NOX2 compared to non-irradiated control conditions in microglial cells (p > 0.05). Therefore, microglia in isolation may not be the primary cause of neuroinflammation and OS following exposures to helium or iron GCR particles.


Asunto(s)
Biomarcadores , Radiación Cósmica , Inflamación , Microglía , Estrés Oxidativo , Animales , Microglía/metabolismo , Microglía/efectos de la radiación , Radiación Cósmica/efectos adversos , Estrés Oxidativo/efectos de la radiación , Ratas , Inflamación/metabolismo , Inflamación/etiología , Biomarcadores/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Hierro/metabolismo , Ciclooxigenasa 2/metabolismo , Helio/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , NADPH Oxidasa 2/metabolismo
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(7): 159524, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38857757

RESUMEN

Neuroinflammation is a hallmark of several neurodegenerative disorders that has been extensively studied in recent years. Microglia, the primary immune cells of the central nervous system (CNS), are key players in this physiological process, demonstrating a remarkable adaptability in responding to various stimuli in the eye and the brain. Within the complex network of neuroinflammatory signals, the fatty acid N-ethanolamines, in particular N-arachidonylethanolamine (anandamide, AEA), emerged as crucial regulators of microglial activity under both physiological and pathological states. In this study, we interrogated for the first time the impact of the signaling of these bioactive lipids on microglial cell responses to a sub-lethal acute UVB radiation, a physical stressor responsible of microglia reactivity in either the retina or the brain. To this end, we developed an in vitro model using mouse microglial BV-2 cells. Upon 24 h of UVB exposure, BV-2 cells showed elevated oxidative stress markers and, cyclooxygenase (COX-2) expression, enhanced phagocytic and chemotactic activities, along with an altered immune profiling. Notably, UVB exposure led to a selective increase in expression and activity of fatty acid amide hydrolase (FAAH), the main enzyme responsible for degradation of fatty acid ethanolamides. Pharmacological FAAH inhibition via URB597 counteracted the effects of UVB exposure, decreasing tumor necrosis factor α (TNF-α) and nitric oxide (NO) release and reverting reactive oxidative species (ROS), interleukin-1ß (IL-1ß), and interleukin-10 (IL-10) levels to the control levels. Our findings support the potential of enhanced fatty acid amide signaling in mitigating UVB-induced cellular damage, paving the way to further exploration of these lipids in light-induced immune responses.


Asunto(s)
Amidohidrolasas , Microglía , Rayos Ultravioleta , Microglía/metabolismo , Microglía/efectos de los fármacos , Microglía/efectos de la radiación , Animales , Ratones , Amidohidrolasas/metabolismo , Amidohidrolasas/antagonistas & inhibidores , Rayos Ultravioleta/efectos adversos , Línea Celular , Carbamatos/farmacología , Benzamidas/farmacología , Estrés Oxidativo/efectos de los fármacos , Endocannabinoides/metabolismo , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/genética , Ácidos Araquidónicos/metabolismo , Ácidos Araquidónicos/farmacología , Interleucina-10/metabolismo , Alcamidas Poliinsaturadas
6.
J Neuroinflammation ; 21(1): 162, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915029

RESUMEN

Radiation retinopathy (RR) is a major side effect of ocular tumor treatment by plaque brachytherapy or proton beam therapy. RR manifests as delayed and progressive microvasculopathy, ischemia and macular edema, ultimately leading to vision loss, neovascular glaucoma, and, in extreme cases, secondary enucleation. Intravitreal anti-VEGF agents, steroids and laser photocoagulation have limited effects on RR. The role of retinal inflammation and its contribution to the microvascular damage occurring in RR remain incompletely understood. To explore cellular and vascular events after irradiation, we analyzed their time course at 1 week, 1 month and 6 months after rat eyes received 45 Gy X-beam photons. Müller glial cells, astrocytes and microglia were rapidly activated, and these markers of retinal inflammation persisted for 6 months after irradiation. This was accompanied by early cell death in the outer retina, which persisted at later time points, leading to retinal thinning. A delayed loss of small retinal capillaries and retinal hypoxia were observed after 6 months, indicating inner blood‒retinal barrier (BRB) alteration but without cell death in the inner retina. Moreover, activated microglial cells invaded the entire retina and surrounded retinal vessels, suggesting the role of inflammation in vascular alteration and in retinal cell death. Radiation also triggered early and persistent invasion of the retinal pigment epithelium by microglia and macrophages, contributing to outer BRB disruption. This study highlights the role of progressive and long-lasting inflammatory mechanisms in RR development and demonstrates the relevance of this rat model to investigate human pathology.


Asunto(s)
Modelos Animales de Enfermedad , Retina , Animales , Ratas , Retina/patología , Retina/efectos de la radiación , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Inflamación/patología , Inflamación/etiología , Traumatismos Experimentales por Radiación/patología , Traumatismos por Radiación/patología , Traumatismos por Radiación/etiología , Masculino , Microglía/efectos de la radiación , Microglía/patología
7.
CNS Neurosci Ther ; 30(6): e14794, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38867379

RESUMEN

BACKGROUND: Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS: This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS: Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS: The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.


Asunto(s)
Lesiones Encefálicas , Microglía , Traumatismos por Radiación , Microglía/efectos de la radiación , Microglía/metabolismo , Animales , Humanos , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/terapia , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Enfermedades Neuroinflamatorias/etiología
8.
J Neuroinflammation ; 21(1): 82, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570852

RESUMEN

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.


Asunto(s)
Encéfalo , Microglía , Ratones , Animales , Microglía/efectos de la radiación , Ratones Transgénicos , Modelos Animales de Enfermedad , Irradiación Craneana/efectos adversos
9.
Cell Prolif ; 57(6): e13606, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38454614

RESUMEN

Glioblastoma (GBM), a WHO grade IV glioma, is a malignant primary brain tumour for which combination of surgery, chemotherapy and radiotherapy is the first-line approach despite adverse effects. Tumour microenvironment (TME) is characterized by an interplay of cells and soluble factors holding a critical role in neoplastic development. Significant pathophysiological changes have been found in GBM TME, such as glia activation and oxidative stress. Microglia play a crucial role in favouring GBM growth, representing target cells of immune escape mechanisms. Our study aims at analysing radiation-induced effects in modulating intercellular communication and identifying the basis of protective mechanisms in radiation-naïve GBM cells. Tumour cells were treated with conditioned media (CM) derived from 0, 2 or 15 Gy irradiated GBM cells or 0, 2 or 15 Gy irradiated human microglia. We demonstrated that irradiated microglia promote an increase of GBM cell lines proliferation through paracrine signalling. On the contrary, irradiated GBM-derived CM affect viability, triggering cell death mechanisms. In addition, we investigated whether these processes involve mitochondrial mass, fitness and oxidative phosphorylation and how GBM cells respond at these induced alterations. Our study suggests that off-target radiotherapy modulates microglia to support GBM proliferation and induce metabolic modifications.


Asunto(s)
Neoplasias Encefálicas , Proliferación Celular , Glioblastoma , Microglía , Microambiente Tumoral , Humanos , Glioblastoma/radioterapia , Glioblastoma/patología , Glioblastoma/metabolismo , Microglía/metabolismo , Microglía/patología , Microglía/efectos de la radiación , Proliferación Celular/efectos de la radiación , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Microambiente Tumoral/efectos de la radiación , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/metabolismo , Supervivencia Celular/efectos de la radiación , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación
10.
Int J Radiat Oncol Biol Phys ; 119(4): 1248-1260, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38364947

RESUMEN

PURPOSE: Diffuse midline glioma (DMG) is a fatal tumor traditionally treated with radiation therapy (RT) and previously characterized as having a noninflammatory tumor immune microenvironment (TIME). FLASH is a novel RT technique using ultra-high dose rate that is associated with decreased toxicity and effective tumor control. However, the effect of FLASH and conventional (CONV) RT on the DMG TIME has not yet been explored. METHODS AND MATERIALS: Here, we performed single-cell RNA sequencing (scRNA-seq) and flow cytometry on immune cells isolated from an orthotopic syngeneic murine model of brainstem DMG after the use of FLASH (90 Gy/sec) or CONV (2 Gy/min) dose-rate RT and compared to unirradiated tumor (SHAM). RESULTS: At day 4 post-RT, FLASH exerted similar effects as CONV in the predominant microglial (MG) population, including the presence of two activated subtypes. However, at day 10 post-RT, we observed a significant increase in the type 1 interferon α/ß receptor (IFNAR+) in MG in CONV and SHAM compared to FLASH. In the non-resident myeloid clusters of macrophages (MACs) and dendritic cells (DCs), we found increased type 1 interferon (IFN1) pathway enrichment for CONV compared to FLASH and SHAM by scRNA-seq. We observed this trend by flow cytometry at day 4 post-RT in IFNAR+ MACs and DCs, which equalized by day 10 post-RT. DMG control and murine survival were equivalent between RT dose rates. CONCLUSIONS: Our work is the first to map CONV and FLASH immune alterations of the DMG TIME with single-cell resolution. Although DMG tumor control and survival were similar between CONV and FLASH, we found that changes in immune compartments differed over time. Importantly, although both RT modalities increased IFN1, we found that the timing of this response was cell-type and dose-rate dependent. These temporal differences, particularly in the context of tumor control, warrant further study.


Asunto(s)
Glioma , Microglía , Animales , Glioma/radioterapia , Glioma/inmunología , Glioma/patología , Ratones , Microglía/efectos de la radiación , Microglía/inmunología , Microambiente Tumoral/inmunología , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Receptor de Interferón alfa y beta/genética , Ratones Endogámicos C57BL , Análisis de la Célula Individual/métodos , Células Dendríticas/inmunología , Células Dendríticas/efectos de la radiación , Macrófagos/inmunología
11.
Int J Radiat Oncol Biol Phys ; 119(3): 912-923, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38142839

RESUMEN

PURPOSE: Cranial irradiation induces healthy tissue damage that can lead to neurocognitive complications, negatively affecting patient quality of life. One damage indicator associated with cognitive impairment is loss of neuronal spine density. We previously demonstrated that irradiation-mediated spine loss is microglial complement receptor 3 (CR3) and sex dependent. We hypothesized that these changes are associated with late-delayed cognitive deficits and amenable to pharmacologic intervention. METHODS AND MATERIALS: Our model of cranial irradiation (acute, 10 Gy gamma) used male and female CR3-wild type and CR3-deficient Thy-1 YFP mice of C57BL/6 background. Forty-five days after irradiation and behavioral testing, we quantified spine density and markers of microglial reactivity in the hippocampal dentate gyrus. In a separate experiment, male Thy-1 YFP C57BL/6 mice were treated with leukadherin-1, a modulator of CR3 function. RESULTS: We found that male mice demonstrate irradiation-mediated spine loss and cognitive deficits but that female and CR3 knockout mice do not. These changes were associated with greater reactivity of microglia in male mice. Pharmacologic manipulation of CR3 with LA1 prevented spine loss and cognitive deficits in irradiated male mice. CONCLUSIONS: This work improves our understanding of irradiation-mediated mechanisms and sex dependent responses and may help identify novel therapeutics to reduce irradiation-induced cognitive decline and improve patient quality of life.


Asunto(s)
Disfunción Cognitiva , Irradiación Craneana , Espinas Dendríticas , Ratones Endogámicos C57BL , Microglía , Animales , Masculino , Femenino , Ratones , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/efectos de la radiación , Irradiación Craneana/efectos adversos , Microglía/efectos de los fármacos , Microglía/efectos de la radiación , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Antígeno de Macrófago-1/metabolismo , Ratones Noqueados , Giro Dentado/efectos de los fármacos , Giro Dentado/efectos de la radiación , Factores Sexuales , Compuestos Orgánicos
12.
Radiat Environ Biophys ; 62(4): 497-509, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37794305

RESUMEN

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.


Asunto(s)
Neoplasias , Protones , Ratones , Masculino , Animales , Microglía/patología , Microglía/efectos de la radiación , Radiación Ionizante , Encéfalo/efectos de la radiación , Neoplasias/patología , Ratones Endogámicos C57BL
13.
Cell Mol Neurobiol ; 43(3): 1369-1384, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35864429

RESUMEN

Seasonal changes in peripheral inflammation are well documented in both humans and animal models, but seasonal changes in neuroinflammation, especially the impact of seasonal lighting environment on neuroinflammation remain unclear. To address this question, the present study examined the effects of environmental lighting conditions on neuroinflammation in a diurnal rodent model, Nile grass rats (Arvicanthis niloticus). Male and female grass rats were housed in either bright (brLD) or dim (dimLD) light during the day to simulate a summer or winter light condition, respectively. After 4 weeks, microglia markers Iba-1 and CD11b, as well as pro-inflammatory cytokines TNF-α and IL-6, were examined in the anterior cingulate cortex (ACC), basolateral amygdala (BLA), and dorsal hippocampus (dHipp). The results revealed that winter-like dim light during the day leads to indicators of increased neuroinflammation in a brain site- and sex-specific manner. Specifically, relatively few changes in the neuroinflammatory markers were observed in the ACC, while numerous changes were found in the BLA and dHipp. In the BLA, winter-like dimLD resulted in hyper-ramified microglia morphology and increased expression of the pro-inflammatory cytokine IL-6, but only in males. In the dHipp, dimLD led to a higher number and hyper-ramified morphology of microglia as well as increased expression of CD11b and TNF-α, but only in females. Neuroinflammatory state is thus influenced by environmental light, differently in males and females, and could play a role in sex differences in the prevalence and symptoms of psychiatric or neurological disorders that are influenced by season or other environmental light conditions. Diurnal Nile grass rats were housed under bright or dim light during the day for 4 weeks, simulating seasonal fluctuations in daytime lighting environment. Dim light housing resulted in hyper-ramified morphology of microglia (scale bar, 15 µm) and altered expression of pro-inflammatory cytokines (TNF-α) in a sex- and brain region-specific manner.


Asunto(s)
Encéfalo , Iluminación , Microglía , Enfermedades Neuroinflamatorias , Enfermedades Neuroinflamatorias/etiología , Murinae , Modelos Animales , Masculino , Femenino , Animales , Encéfalo/fisiopatología , Encéfalo/efectos de la radiación , Antígeno CD11b/análisis , Antígeno CD11b/genética , Biomarcadores/análisis , Regulación de la Expresión Génica/efectos de la radiación , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/genética , Interleucina-6/análisis , Interleucina-6/genética , Factores Sexuales , Microglía/metabolismo , Microglía/efectos de la radiación
14.
J Neuroinflammation ; 18(1): 256, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34740378

RESUMEN

BACKGROUND: Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS: Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS: PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION: Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.


Asunto(s)
Astrocitos/efectos de la radiación , Terapia por Luz de Baja Intensidad , Microglía/efectos de la radiación , Recuperación de la Función/efectos de la radiación , Traumatismos de la Médula Espinal/patología , Animales , Astrocitos/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 2/efectos de la radiación , Lipocalina 2/metabolismo , Lipocalina 2/efectos de la radiación , Masculino , Microglía/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/efectos de la radiación , Transducción de Señal/efectos de la radiación , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba
15.
Cells ; 10(9)2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34572030

RESUMEN

Microglia, the innate immune cells of the central nervous system, play a pivotal role in the modulation of neuroinflammation. Neuroinflammation has been implicated in many diseases of the CNS, including Alzheimer's disease and Parkinson's disease. It is well documented that microglial activation, initiated by a variety of stressors, can trigger a potentially destructive neuroinflammatory response via the release of pro-inflammatory molecules, and reactive oxygen and nitrogen species. However, the potential anti-inflammatory and neuroprotective effects that microglia are also thought to exhibit have been under-investigated. The application of ionising radiation at different doses and dose schedules may reveal novel methods for the control of microglial response to stressors, potentially highlighting avenues for treatment of neuroinflammation associated CNS disorders, such as Alzheimer's disease and Parkinson's disease. There remains a need to characterise the response of microglia to radiation, particularly low dose ionising radiation.


Asunto(s)
Mediadores de Inflamación/metabolismo , Microglía/efectos de la radiación , Enfermedades Neurodegenerativas/radioterapia , Neuroinmunomodulación/efectos de la radiación , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Relación Dosis-Respuesta en la Radiación , Humanos , Inmunidad Innata/efectos de la radiación , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Estrés Nitrosativo/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Fenotipo , Receptores de GABA/metabolismo
16.
Int J Mol Sci ; 22(18)2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-34575905

RESUMEN

BACKGROUND: In adult rats we study the short- and long-term effects of focal blue light-emitting diode (LED)-induced phototoxicity (LIP) on retinal thickness and Iba-1+ activation. METHODS: The left eyes of previously dark-adapted Sprague Dawley (SD) rats were photoexposed to a blue LED (20 s, 200 lux). In vivo longitudinal monitoring of retinal thickness, fundus images, and optical retinal sections was performed from 1 to 30 days (d) after LIP with SD-OCT. Ex vivo, we analysed the population of S-cone and Iba-1+ cells within a predetermined fixed-size circular area (PCA) centred on the lesion. RESULTS: LIP resulted in a circular focal lesion readily identifiable in vivo by fundus examination, which showed within the PCAs a progressive thinning of the outer retinal layer, and a diminution of the S-cone population to 19% by 30 d. In parallel to S-cone loss, activated Iba-1+ cells delineated the lesioned area and acquired an ameboid morphology with peak expression at 3 d after LIP. Iba-1+ cells adopted a more relaxed-branched morphology at 7 d and by 14-30 d their morphology was fully branched. CONCLUSION: LIP caused a progressive reduction of the outer retina with loss of S cones and a parallel dynamic activation of microglial cells in the lesioned area.


Asunto(s)
Luz , Retina/patología , Retina/efectos de la radiación , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Microglía/metabolismo , Microglía/patología , Microglía/efectos de la radiación , Ratas , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/etiología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Factores de Tiempo , Tomografía de Coherencia Óptica
17.
J Radiat Res ; 62(5): 793-803, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34062561

RESUMEN

The objective of the study was to describe cellular and molecular markers of radioprotection by anisomycin, focusing on the changes in rat brain tissue. Two-month-old Wistar rats were exposed to a 60Co radiation source at a dose of 6 Gy, with or without radioprotection with anisomycin (150 mg/kg) administered subcutaneously 30 min before or 3 or 6 h after irradiation. Survivors were analyzed 30 days after treatment. Astroglial and microglial responses were investigated based on the expression of glial markers assessed with immunohistochemistry, and quantitative changes in brain biomolecules were investigated by Raman microspectroscopy. In addition, blood plasma levels of pro-inflammatory (interleukin 6 and tumor necrosis factor α) and anti-inflammatory (interleukin 10) cytokines were assessed. We found that application of anisomycin either before or after irradiation significantly decreased the expression of the microglial marker Iba-1. We also found an increased intensity of Raman spectral bands related to nucleic acids, as well as an increased level of cytokines when anisomycin was applied after irradiation. This suggests that the radioprotective effects of anisomycin are by decreasing Iba-1 expression and stabilizing genetic material by increasing the level of nucleic acids.


Asunto(s)
Anisomicina/uso terapéutico , Encéfalo/efectos de la radiación , Irradiación Craneana/efectos adversos , Rayos gamma/efectos adversos , Traumatismos Experimentales por Radiación/metabolismo , Protectores contra Radiación/uso terapéutico , Animales , Anisomicina/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/efectos de la radiación , Encéfalo/efectos de los fármacos , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Radioisótopos de Cobalto , Citocinas/sangre , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Microglía/efectos de los fármacos , Microglía/efectos de la radiación , Ácidos Nucleicos/metabolismo , Premedicación , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Ratas , Ratas Wistar
18.
Neurosci Lett ; 741: 135462, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33259927

RESUMEN

Microglia are the resident immune cells of the central nervous system (CNS). In physiological conditions, microglia contribute to maintaining brain homeostasis by scanning the surrounding parenchyma and acting as scavenger cells. Following different insults to the CNS, microglia turn into a "reactive" state characterized by the production of inflammatory mediators that promote tissue repair to restore homeostasis. Brain insults such as traumatic brain injury, therapeutic brain irradiation and galactic cosmic ray exposure are associated with chronic microglia activation. Chronic microglia activation contributes to injury-related impairments in cognitive functions. Microglia depletion achieved either by pharmacological or genetic techniques represents not only a useful tool for more extensive investigations of microglia roles, but also a potential therapeutic approach to ameliorate or prevent cognitive dysfunctions following brain injury.


Asunto(s)
Lesiones Encefálicas/inmunología , Lesiones Encefálicas/psicología , Cognición/fisiología , Radiación Cósmica/efectos adversos , Microglía/inmunología , Microglía/efectos de la radiación , Animales , Lesiones Encefálicas/etiología , Cognición/efectos de la radiación , Encefalitis/etiología , Encefalitis/inmunología , Humanos
19.
J Neuroinflammation ; 17(1): 321, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33109221

RESUMEN

BACKGROUND: Microglia have been implicated in the pathogenesis of radiation-induced brain injury (RIBI), which severely influences the quality of life during long-term survival. Recently, irradiated microglia were speculated to present an aging-like phenotype. Long noncoding RNAs (lncRNAs) have been recognized to regulate a wide spectrum of biological processes, including senescence; however, their potential role in irradiated microglia remains largely uncharacterized. METHODS: We used bioinformatics and experimental methods to identify and analyze the senescence phenotype of irradiated microglia. Western blotting, enzyme-linked immunosorbent assays, immunofluorescence, and quantitative real-time reverse transcription-polymerase chain reaction were performed to clarify the relationship between the radiation-induced differentially expressed lncRNAs (RILs) and the distinctive molecular features of senescence in irradiated microglia. RESULTS: We found that the senescence of microglia could be induced using ionizing radiation (IR). A mutual regulation mode existed between RILs and three main features of the senescence phenotype in irradiated microglia: inflammation, the DNA damage response (DDR), and metabolism. Specifically, for inflammation, the expression of two selected RILs (ENSMUST00000190863 and ENSMUST00000130679) was dependent on the major inflammatory signaling pathways of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK). The two RILs modulated the activation of NF-κB/MAPK signaling and subsequent inflammatory cytokine secretion. For the DDR, differential severity of DNA damage altered the expression profiles of RILs. The selected RIL, ENSMUST00000130679, promoted the DDR. For metabolism, blockade of sterol regulatory element-binding protein-mediated lipogenesis attenuated the fold-change of several RILs induced by IR. CONCLUSIONS: Our findings revealed that certain RILs interacted with senescence in irradiated microglia. RILs actively participated in the regulation of senescence features, suggesting that RILs could be promising intervention targets to treat RIBI.


Asunto(s)
Senescencia Celular/efectos de la radiación , Microglía/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal/efectos de la radiación , Animales , Línea Celular , Ratones , Microglía/efectos de la radiación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Radiación Ionizante
20.
Cancer Res ; 80(19): 4266-4277, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32816912

RESUMEN

Radiation-induced cognitive dysfunction (RICD) is a progressive and debilitating health issue facing patients following cranial radiotherapy to control central nervous system cancers. There has been some success treating RICD in rodents using human neural stem cell (hNSC) transplantation, but the procedure is invasive, requires immunosuppression, and could cause other complications such as teratoma formation. Extracellular vesicles (EV) are nanoscale membrane-bound structures that contain biological contents including mRNA, miRNA, proteins, and lipids that can be readily isolated from conditioned culture media. It has been previously shown that hNSC-derived EV resolves RICD following cranial irradiation using an immunocompromised rodent model. Here, we use immunocompetent wild-type mice to show that hNSC-derived EV treatment administered either intravenously via retro-orbital vein injection or via intracranial transplantation can ameliorate cognitive deficits following 9 Gy head-only irradiation. Cognitive function assessed on the novel place recognition, novel object recognition, and temporal order tasks was not only improved at early (5 weeks) but also at delayed (6 months) postirradiation times with just a single EV treatment. Improved behavioral outcomes were also associated with reduced neuroinflammation as measured by a reduction in activated microglia. To identify the mechanism of action, analysis of EV cargo implicated miRNA (miR-124) as a potential candidate in the mitigation of RICD. Furthermore, viral vector-mediated overexpression of miR-124 in the irradiated brain ameliorated RICD and reduced microglial activation. Our findings demonstrate for the first time that systemic administration of hNSC-derived EV abrogates RICD and neuroinflammation in cranially irradiated wild-type rodents through a mechanism involving miR-124. SIGNIFICANCE: Radiation-induced neurocognitive decrements in immunocompetent mice can be resolved by systemic delivery of hNSC-derived EVs involving a mechanism dependent on expression of miR-124.


Asunto(s)
Encéfalo/efectos de la radiación , Vesículas Extracelulares/genética , MicroARNs/farmacología , Células-Madre Neurales/citología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Encéfalo/efectos de los fármacos , Lesiones Encefálicas , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Vesículas Extracelulares/trasplante , Hipocampo/efectos de los fármacos , Hipocampo/efectos de la radiación , Humanos , Inyecciones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/aislamiento & purificación , Microglía/efectos de los fármacos , Microglía/efectos de la radiación , Células-Madre Neurales/fisiología , Traumatismos Experimentales por Radiación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA