Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069478

ABSTRACT

Exposure to radiofrequency electromagnetic fields (RF-EMFs) has increased rapidly in children, but information on the effects of RF-EMF exposure to the central nervous system in children is limited. In this study, pups and dams were exposed to whole-body RF-EMF at 4.0 W/kg specific absorption rate (SAR) for 5 h per day for 4 weeks (from postnatal day (P) 1 to P28). The effects of RF-EMF exposure on neurons were evaluated by using both pups' hippocampus and primary cultured hippocampal neurons. The total number of dendritic spines showed statistically significant decreases in the dentate gyrus (DG) but was not altered in the cornu ammonis (CA1) in hippocampal neurons. In particular, the number of mushroom-type dendritic spines showed statistically significant decreases in the CA1 and DG. The expression of glutamate receptors was decreased in mushroom-type dendritic spines in the CA1 and DG of hippocampal neurons following RF-EMF exposure. The expression of brain-derived neurotrophic factor (BDNF) in the CA1 and DG was significantly lower statistically in RF-EMF-exposed mice. The number of post-synaptic density protein 95 (PSD95) puncta gradually increased over time but was significantly decreased statistically at days in vitro (DIV) 5, 7, and 9 following RF-EMF exposure. Decreased BDNF expression was restricted to the soma and was not observed in neurites of hippocampal neurons following RF-EMF exposure. The length of neurite outgrowth and number of branches showed statistically significant decreases, but no changes in the soma size of hippocampal neurons were observed. Further, the memory index showed statistically significant decreases in RF-EMF-exposed mice, suggesting that decreased synaptic density following RF-EMF exposure at early developmental stages may affect memory function. Collectively, these data suggest that hindered neuronal outgrowth following RF-EMF exposure may decrease overall synaptic density during early neurite development of hippocampal neurons.


Subject(s)
Electromagnetic Fields/adverse effects , Neurites/radiation effects , Radio Waves/adverse effects , Animals , Animals, Newborn/physiology , Brain-Derived Neurotrophic Factor/metabolism , Female , Hippocampus/metabolism , Hippocampus/radiation effects , Male , Mice , Mice, Inbred ICR , Neurites/metabolism , Neurogenesis , Neuronal Outgrowth , Neurons/metabolism , Neurons/radiation effects , Synapses/metabolism , Synapses/radiation effects
2.
Int J Radiat Biol ; 97(7): 1032-1041, 2021.
Article in English | MEDLINE | ID: mdl-33970763

ABSTRACT

BACKGROUND: Long-term potentiation (LTP) is an important functional indicator for synaptic plasticity. Extremely low frequency electromagnetic fields (ELF-EMFs) are a physical means to regulate LTP, which induce induced currents. It is unknown whether induced current is the key factor when LTP is regulated by ELF-EMFs.New Method: A method is proposed for calculating the current value induced by ELF-EMFs. Then, a comparison of ELF-EMFs with current on the regulation of theta-burst or high-frequency stimulation (TBS/HFS)-LTP was performed. RESULTS: The LTP after ELF-EMFs and µA current regulation was significantly reduced. The regulatory effect of 0.1 µA current on LTP was similar with 100 Hz/2 mT ELF-EMFs, while 0.2 µA had a stronger regulatory effect than 200 Hz/2 mT on HFS-LTP.Comparison with Existing Methods: Most of the existing methods were used to calculate the induced current in human models, while we present a more accurate model for calculating the induced current induced by ELF-EMFs in the rat brain slices. CONCLUSIONS: This work indicated that µA current and ELF-EMFs stimulation reduced LTP. Also, we demonstrated that the regulatory effect of ELF-EMFs on LTP is not entirely deriving from the induced current, since its magnetic mechanism might have played a certain role.


Subject(s)
Electromagnetic Fields , Hippocampus/physiology , Hippocampus/radiation effects , Long-Term Potentiation/radiation effects , Synapses/radiation effects , Animals , Neuronal Plasticity/radiation effects , Rats
3.
Ultrastruct Pathol ; 44(4-6): 372-378, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33121293

ABSTRACT

This study aimed to investigate ultrastructural synaptic alterations in rat hippocampus after in utero exposure to irradiation (IR) and postnatal exposure to hyperthermia (HT). There were four groups in each of the time points (3rd and 6th months). IR group: Pregnant rats were exposed to radiation on the 17th gestational day. HT group: Hyperthermia was applied to the rat pups on the 10th day after their birth. IR+HT group: Both IR and HT were applied at the same time periods. Control group: No IR or HT was applied. Rat pups were sacrificed after 3 and 6 months. Thin sections from the dentate gyrus (DG) and the CA3 of hippocampus were evaluated for synapse numbers by electron microscopy. Synapses were counted, and statistical analysis was performed. Abnormalities in myelin sheath, mossy terminals and neuropil were observed in the CA3 and DG of all groups. The synapses in the CA3 region were significantly increased in the IR-3rd month, IR-6th month, and IR+HT-3rd month groups vs control group. Synapses were significantly increased in the DG of HT-3rd month group. A trend for an increase in synapse numbers was seen in the CA3 and DG. Increased number of synapses in the rat hippocampus may be due to mossy fiber sprouting, possibly caused by in utero irradiation and/or postnatal hyperthermia.


Subject(s)
Hippocampus/ultrastructure , Hyperthermia/pathology , Prenatal Exposure Delayed Effects/pathology , Radiation Injuries, Experimental/pathology , Synapses/ultrastructure , Animals , Female , Hippocampus/pathology , Hippocampus/radiation effects , Pregnancy , Rats , Rats, Wistar , Synapses/pathology , Synapses/radiation effects
4.
Elife ; 92020 08 18.
Article in English | MEDLINE | ID: mdl-32808923

ABSTRACT

Microglia continuously monitor synapses, but active synaptic remodeling by microglia in mature healthy brains is rarely directly observed. We performed targeted photoablation of single synapses in mature transgenic mice expressing fluorescent labels in neurons and microglia. The photodamage focally increased the duration of microglia-neuron contacts, and dramatically exacerbated both the turnover of dendritic spines and presynaptic boutons as well as the generation of new filopodia originating from spine heads or boutons. The results of microglia depletion confirmed that elevated spine turnover and the generation of presynaptic filopodia are microglia-dependent processes.


Subject(s)
Microglia/radiation effects , Neuronal Plasticity/radiation effects , Synapses/radiation effects , Animals , Green Fluorescent Proteins/chemistry , Light , Luminescent Proteins/chemistry , Male , Mice , Mice, Transgenic , Microglia/physiology , Microscopy, Fluorescence, Multiphoton , Presynaptic Terminals/physiology , Presynaptic Terminals/radiation effects , Pseudopodia/physiology , Pseudopodia/radiation effects , Synapses/physiology , Red Fluorescent Protein
5.
Cell Rep ; 30(3): 630-641.e5, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968242

ABSTRACT

In the neocortex, synaptic inhibition shapes all forms of spontaneous and sensory evoked activity. Importantly, inhibitory transmission is highly plastic, but the functional role of inhibitory synaptic plasticity is unknown. In the mouse barrel cortex, activation of layer (L) 2/3 pyramidal neurons (PNs) elicits strong feedforward inhibition (FFI) onto L5 PNs. We find that FFI involving parvalbumin (PV)-expressing cells is strongly potentiated by postsynaptic PN burst firing. FFI plasticity modifies the PN excitation-to-inhibition (E/I) ratio, strongly modulates PN gain, and alters information transfer across cortical layers. Moreover, our LTPi-inducing protocol modifies firing of L5 PNs and alters the temporal association of PN spikes to γ-oscillations both in vitro and in vivo. All of these effects are captured by unbalancing the E/I ratio in a feedforward inhibition circuit model. Altogether, our results indicate that activity-dependent modulation of perisomatic inhibitory strength effectively influences the participation of single principal cortical neurons to cognition-relevant network activity.


Subject(s)
Neocortex/physiology , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Female , Gamma Rhythm/radiation effects , Light , Long-Term Potentiation/physiology , Long-Term Potentiation/radiation effects , Mice, Inbred C57BL , Models, Neurological , Neural Inhibition/radiation effects , Neuronal Plasticity/radiation effects , Pyramidal Cells/physiology , Pyramidal Cells/radiation effects , Synapses/radiation effects , Time Factors , gamma-Aminobutyric Acid/metabolism
6.
PLoS One ; 15(1): e0226797, 2020.
Article in English | MEDLINE | ID: mdl-31940316

ABSTRACT

Analysis of neuronal compartments has revealed many state-dependent changes in geometry but establishing synapse-specific mechanisms at the nanoscale has proven elusive. We co-expressed channelrhodopsin2-GFP and mAPEX2 in a subset of hippocampal CA3 neurons and used trains of light to induce late-phase long-term potentiation (L-LTP) in area CA1. L-LTP was shown to be specific to the labeled axons by severing CA3 inputs, which prevented back-propagating recruitment of unlabeled axons. Membrane-associated mAPEX2 tolerated microwave-enhanced chemical fixation and drove tyramide signal amplification to deposit Alexa Fluor dyes in the light-activated axons. Subsequent post-embedding immunogold labeling resulted in outstanding ultrastructure and clear distinctions between labeled (activated), and unlabeled axons without obscuring subcellular organelles. The gold-labeled axons in potentiated slices were reconstructed through serial section electron microscopy; presynaptic vesicles and other constituents could be quantified unambiguously. The genetic specification, reliable physiology, and compatibility with established methods for ultrastructural preservation make this an ideal approach to link synapse ultrastructure and function in intact circuits.


Subject(s)
Axons/radiation effects , Axons/ultrastructure , Light , Long-Term Potentiation/radiation effects , Optogenetics , Animals , Axons/metabolism , Axons/physiology , Rats , Synapses/metabolism , Synapses/radiation effects
7.
Radiat Res ; 192(3): 345-351, 2019 09.
Article in English | MEDLINE | ID: mdl-31295088

ABSTRACT

Stem cell transplantation is thought to be an effective method for radiation-induced cognitive dysfunction. However, there have been few studies performed to determine whether transplanted stem cells can integrate into hippocampus circuits. Brain-derived neurotrophic factor (BDNF) plays a critical role in brain development. Therefore, we investigated the differentiation and integration of brain-derived neurotrophic factor overexpressing neural stem cells (NSCs). We observed that these transplanted cells migrated to the subgranular zone of irradiated rats at 4 weeks after transplantation. However, control neural stem cells were disordered, distributing in the irradiated hippocampus, and showed greater astroglia differentiation tendency. Retrograde monosynaptic tracing showed that neurons derived from transplanted brain-derived neurotrophic factor overexpressing neural stem cells integrated into the circuit better than those from control cells. Brain-derived neurotrophic factor overexpressing neural stem cells s promoted the expression of brain-derived neurotrophic factor and nerve growth factor and reduced the number of activated microglia caused by radiation. Transplanted brain-derived neurotrophic factor overexpressing neural stem cells failed to improve radiation-induced cognitive dysfunction. These results indicate that brain-derived neurotrophic factor overexpressing neural stem cells suffered less from changed microenvironment after irradiation and possessed the ability to improve the host niche. Neurons derived from Brain-derived neurotrophic factor overexpressing neural stem cells showed the integration potency in the irradiated hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Hippocampus/cytology , Hippocampus/radiation effects , Nerve Net/cytology , Neural Stem Cells/transplantation , Neurons/cytology , Stem Cell Transplantation , Animals , Cell Differentiation/radiation effects , Cognition/radiation effects , Gene Expression , Hippocampus/physiology , Male , Nerve Net/radiation effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/radiation effects , Rats , Rats, Sprague-Dawley , Synapses/radiation effects
8.
J Biophotonics ; 12(11): e201900145, 2019 11.
Article in English | MEDLINE | ID: mdl-31240853

ABSTRACT

A certain degree of noise can cause hearing problems without a permanent change in the hearing threshold, which is called hidden hearing loss and results from partial loss of auditory synapses. Photobiomodulation (PBM) enhances neural growth and connections in the peripheral nervous systems. In this study, we assessed whether PBM could rescue cochlear synaptopathy after acoustic overexposure in rat. PBM was performed for 7 days after noise exposure. The auditory brainstem responses (ABRs) were acquired before and after noise exposure using a tone and a paired-click stimulus. Auditory response to paired click sound with short time interval was performed to evaluate auditory temporal processing ability. In the result, hearing threshold recovered 2 weeks after noise exposure in both groups. Peak wave 1 amplitude of the ABR and ABR recovery threshold did not recover in the noise only group, whereas it fully recovered in the noise + PBM group. The number of synaptic ribbons was significantly different in the control and noise only groups, while there was no difference between the control and noise + PBM group. These results indicate that PBM rescued peak wave 1 amplitude and maintained the auditory temporal processing ability resulting from a loss of synaptic ribbons after acoustic overexposure.


Subject(s)
Acoustics , Cochlea/radiation effects , Lasers, Semiconductor , Low-Level Light Therapy , Synapses/pathology , Synapses/radiation effects , Animals , Auditory Threshold/radiation effects , Hearing/physiology , Hearing/radiation effects , Male , Noise/adverse effects , Rats , Synapses/physiology
9.
Nat Cell Biol ; 21(6): 768-777, 2019 06.
Article in English | MEDLINE | ID: mdl-31061466

ABSTRACT

Controlling cellular processes with light can help elucidate their underlying mechanisms. Here we present zapalog, a small-molecule dimerizer that undergoes photolysis when exposed to blue light. Zapalog dimerizes any two proteins tagged with the FKBP and DHFR domains until exposure to light causes its photolysis. Dimerization can be repeatedly restored with uncleaved zapalog. We implement this method to investigate mitochondrial motility and positioning in cultured neurons. Using zapalog, we tether mitochondria to constitutively active kinesin motors, forcing them down the axon towards microtubule (+) ends until their instantaneous release via blue light, which results in full restoration of their endogenous motility. We find that one-third of stationary mitochondria cannot be pulled away from their position and that these firmly anchored mitochondria preferentially localize to VGLUT1-positive presynapses. Furthermore, inhibition of actin polymerization with latrunculin A reduces this firmly anchored pool. On release from exogenous motors, mitochondria are preferentially recaptured at presynapses.


Subject(s)
Axons/metabolism , Mitochondria/genetics , Photolysis , Protein Multimerization/radiation effects , Actins/antagonists & inhibitors , Animals , Axons/chemistry , Axons/radiation effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , COS Cells , Chlorocebus aethiops , Kinesins/chemistry , Light , Microtubules/genetics , Microtubules/radiation effects , Mitochondria/chemistry , Mitochondria/radiation effects , Neurons/chemistry , Neurons/radiation effects , Polymerization/drug effects , Protein Domains/genetics , Protein Domains/radiation effects , Protein Multimerization/genetics , Synapses/chemistry , Synapses/genetics , Synapses/radiation effects , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/genetics , Thiazolidines/pharmacology , Vesicular Glutamate Transport Protein 1/genetics
10.
Biomaterials ; 199: 63-75, 2019 04.
Article in English | MEDLINE | ID: mdl-30738336

ABSTRACT

Tissue specific extracellular matrices (ECM) provide structural support and enable access to molecular signals and metabolites, which are essential for directing stem cell renewal and differentiation. To mimic this phenomenon in vitro, tissue decellularisation approaches have been developed, resulting in the generation of natural ECM scaffolds that have comparable physical and biochemical properties of the natural tissues and are currently gaining traction in tissue engineering and regenerative therapies due to the ease of standardised production, and constant availability. In this manuscript we report the successful generation of decellularised ECM-derived peptides from neural retina (decel NR) and retinal pigment epithelium (decel RPE), and their impact on differentiation of human pluripotent stem cells (hPSCs) to retinal organoids. We show that culture media supplementation with decel RPE and RPE-conditioned media (CM RPE) significantly increases the generation of rod photoreceptors, whilst addition of decel NR and decel RPE significantly enhances ribbon synapse marker expression and the light responsiveness of retinal organoids. Photoreceptor maturation, formation of correct synapses between retinal cells and recording of robust light responses from hPSC-derived retinal organoids remain unresolved challenges for the field of regenerative medicine. Enhanced rod photoreceptor differentiation, synaptogenesis and light response in response to addition of decellularised matrices from RPE and neural retina as shown herein provide a novel and substantial advance in generation of retinal organoids for drug screening, tissue engineering and regenerative medicine.


Subject(s)
Biomarkers/metabolism , Extracellular Matrix/chemistry , Light , Organoids/cytology , Peptides/pharmacology , Pluripotent Stem Cells/cytology , Retinal Pigment Epithelium/metabolism , Synapses/metabolism , Adult , Animals , Cattle , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/radiation effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/radiation effects , Human Embryonic Stem Cells/ultrastructure , Humans , Organoids/drug effects , Organoids/radiation effects , Organoids/ultrastructure , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Photoreceptor Cells, Vertebrate/ultrastructure , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/radiation effects , Synapses/drug effects , Synapses/radiation effects
11.
J Biophotonics ; 12(6): e201800359, 2019 06.
Article in English | MEDLINE | ID: mdl-30652418

ABSTRACT

Neonatal hypoxia-ischemia (HI) injury caused by oxygen deprivation is the most common cause of mortality and severe neurologic deficits in neonates. The present work evaluated the preventative effect of photobiomodulation (PBM) preconditioning, and its underlying mechanism of action on brain damage in an HI model in neonatal rats. According to the optimal time response of ATP levels in brain samples removed from normal rats, a PBM preconditioning (PBM-P) regimen (808 nm CW laser, 1 cm2 spot, 100 mW/cm2 , 12 J/cm2 ) was delivered to the scalp 6 hours before HI. PBM-P significantly attenuated cognitive impairment, volume shrinkage in the brain, neuron loss, dendritic and synaptic injury after HI. Further mechanistic investigation found that PBM-P could restore HI-induced mitochondrial dynamics and inhibit mitochondrial fragmentation, followed by a robust suppression of cytochrome c release, and prevention of neuronal apoptosis by inhibition of caspase activation. Our work suggests that PBM-P can attenuate HI-induced brain injury by maintaining mitochondrial dynamics and inhibiting the mitochondrial apoptotic pathway.


Subject(s)
Cognitive Dysfunction/complications , Cognitive Dysfunction/prevention & control , Hypoxia-Ischemia, Brain/complications , Low-Level Light Therapy , Animals , Animals, Newborn , Apoptosis/radiation effects , Behavior, Animal/radiation effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cytochromes c/metabolism , Dendritic Cells/pathology , Dendritic Cells/radiation effects , Disease Models, Animal , Female , Male , Mitochondrial Dynamics/radiation effects , Neurons/pathology , Neurons/radiation effects , Rats , Rats, Sprague-Dawley , Synapses/pathology , Synapses/radiation effects
12.
Mol Neurobiol ; 56(5): 3341-3355, 2019 May.
Article in English | MEDLINE | ID: mdl-30120733

ABSTRACT

Tau oligomers are emerging as a key contributor to the synaptic dysfunction that drives cognitive decline associated with the clinical manifestation and progression of Alzheimer's disease (AD). Accordingly, there is ample consensus that interventions that target tau oligomers may slow or halt the progression of AD. With this ultimate goal in mind, in the present study, we investigated tau oligomer accumulation and its synaptic and behavioral consequences after an in vivo treatment with near infrared (NIR) light (600-1000 nm) in two transgenic mouse models, overexpressing human tau either alone (hTau mice) or in combination with amyloid beta (3xTgAD mice). We found that a 4-week exposure to NIR light (90 s/day/5 days a week) significantly reduced levels of endogenous total and oligomeric tau in both synaptosomes and total protein extracts from the hippocampus and cortex of hTau mice and improved deteriorating memory function. Similar results were observed in the 3xTgAD mice, which further displayed reduced synaptic Aß after NIR light treatment. On the other hand, ex vivo binding of tau oligomers in isolated synaptosomes as well as tau oligomer-induced depression of long-term potentiation (LTP) in hippocampal slices from NIR light-treated wt mice were unaffected. Finally, levels of proteins critically involved in two mechanisms associated with clearance of misfolded tau, inducible HSP70 and autophagy, were upregulated in NIR light treated mice. Collectively, these results show that NIR light decreases levels of endogenous toxic tau oligomers and alleviate associated memory deficits, thus furthering the development of NIR light as a possible therapeutic for AD.


Subject(s)
Infrared Rays , Protein Multimerization , Synapses/metabolism , Synapses/radiation effects , Tauopathies/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Female , HSP70 Heat-Shock Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Long-Term Potentiation , Male , Memory , Mice, Transgenic , Protein Multimerization/radiation effects , Synaptosomes/metabolism , Synaptosomes/radiation effects , Tauopathies/pathology , Tauopathies/physiopathology , Up-Regulation
13.
J Comp Neurol ; 526(17): 2845-2855, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30198564

ABSTRACT

High-energy charged particles are considered particularly hazardous components of the space radiation environment. Such particles include fully ionized energetic nuclei of helium, silicon, and oxygen, among others. Exposure to charged particles causes reactive oxygen species production, which has been shown to result in neuronal dysfunction and myelin degeneration. Here we demonstrate that mice exposed to high-energy charged particles exhibited alterations in dendritic spine density in the hippocampus, with a significant decrease of thin spines in mice exposed to helium, oxygen, and silicon, compared to sham-irradiated controls. Electron microscopy confirmed these findings and revealed a significant decrease in overall synapse density and in nonperforated synapse density, with helium and silicon exhibiting more detrimental effects than oxygen. Degeneration of myelin was also evident in exposed mice with significant changes in the percentage of myelinated axons and g-ratios. Our data demonstrate that exposure to all types of high-energy charged particles have a detrimental effect, with helium and silicon having more synaptotoxic effects than oxygen. These results have important implications for the integrity of the central nervous system and the cognitive health of astronauts after prolonged periods of space exploration.


Subject(s)
Elementary Particles , Myelin Sheath/radiation effects , Synapses/radiation effects , Animals , Axons/radiation effects , Axons/ultrastructure , Dendritic Spines/radiation effects , Exploratory Behavior/radiation effects , Helium , Hippocampus/cytology , Hippocampus/radiation effects , Male , Mice , Mice, Inbred C57BL , Myelin Sheath/ultrastructure , Oxygen , Silicon , Synapses/ultrastructure
14.
Brain Behav Immun ; 74: 106-120, 2018 11.
Article in English | MEDLINE | ID: mdl-30107198

ABSTRACT

Interplanetary exploration will be humankind's most ambitious expedition and the journey required to do so, is as intimidating as it is intrepid. One major obstacle for successful deep space travel is the possible negative effects of galactic cosmic radiation (GCR) exposure. Here, we investigate for the first time how combined GCR impacts long-term behavioral and cellular responses in male and female mice. We find that a single exposure to simulated GCR induces long-term cognitive and behavioral deficits only in the male cohorts. GCR exposed male animals have diminished social interaction, increased anxiety-like phenotype and impaired recognition memory. Remarkably, we find that the female cohorts did not display any cognitive or behavioral deficits after GCR exposure. Mechanistically, the maladaptive behavioral responses observed only in the male cohorts correspond with microglia activation and synaptic loss in the hippocampus, a brain region involved in the cognitive domains reported here. Furthermore, we measured reductions in AMPA expressing synaptic terminals in the hippocampus. No changes in any of the molecular markers measured here are observed in the females. Taken together these findings suggest that GCR exposure can regulate microglia activity and alter synaptic architecture, which in turn leads to a range of cognitive alterations in a sex dependent manner. These results identify sex-dependent differences in behavioral and cognitive domains revealing promising cellular and molecular intervention targets to reduce GCR-induced chronic cognitive deficits thereby boosting chances of success for humans in deep space missions such as the upcoming Mars voyage.


Subject(s)
Behavior, Animal/radiation effects , Cosmic Radiation/adverse effects , Sex Factors , Animals , Cognitive Dysfunction/physiopathology , Female , Male , Mice , Mice, Inbred C57BL , Microglia/radiation effects , Models, Animal , Space Flight , Synapses/radiation effects
15.
Brain Res ; 1679: 134-143, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29180226

ABSTRACT

The popularization of microwave raised concerns about its influence on health including cognitive function which is associated greatly with dendritic spines plasticity. SNK-SPAR is a molecular pathway for neuronal homeostatic plasticity during chronically elevated activity. In this study, Wistar rats were exposed to microwaves (30 mW/cm2 for 6 min, 3 times/week for 6 weeks). Spatial learning and memory function, distribution of dendritic spines, ultrastructure of the neurons and their dendritic spines in hippocampus as well as the related critical molecules of SNK-SPAR pathway were examined at different time points after microwave exposure. There was deficiency in spatial learning and memory in rats, loss of spines in granule cells and shrinkage of mature spines in pyramidal cells, accompanied with alteration of ultrastructure of hippocampus neurons. After exposure to 30 mW/cm2 microwave radiation, the up-regulated SNK induced decrease of SPAR and PSD-95, which was thought to cause the changes mentioned above. In conclusion, the microwave radiation led to shrinkage and even loss of dendritic spines in hippocampus by SNK-SPAR pathway, resulting in the cognitive impairments.


Subject(s)
Dendritic Spines/radiation effects , GTPase-Activating Proteins/metabolism , Hippocampus/cytology , Microwaves/adverse effects , Neurons/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/radiation effects , Animals , Dendritic Spines/ultrastructure , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/ultrastructure , Hippocampus/radiation effects , Male , Maze Learning/radiation effects , Microscopy, Electron, Transmission , Neurons/radiation effects , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Silver Staining , Synapses/metabolism , Synapses/radiation effects , Synapses/ultrastructure , Time Factors , Up-Regulation/radiation effects
16.
Neuro Oncol ; 20(5): 655-665, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29112734

ABSTRACT

Background: Memantine has shown clinical utility in preventing radiation-induced cognitive impairment, but the mechanisms underlying its protective effects remain unknown. We hypothesized that abnormal glutamate signaling causes radiation-induced abnormalities in neuronal structure and that memantine prevents synaptic toxicity. Methods: Hippocampal cultures expressing enhanced green fluorescent protein were irradiated or sham-treated and their dendritic spine morphology assessed at acute (minutes) and later (days) times using high-resolution confocal microscopy. Excitatory synapses, defined by co-localization of the pre- and postsynaptic markers vesicular glutamate transporter 1 and postsynaptic density protein 95, were also analyzed. Neurons were pretreated with vehicle, the N-methyl-d-aspartate-type glutamate receptor antagonist memantine, or the glutamate scavenger glutamate pyruvate transaminase to assess glutamate signaling. For animal studies, Thy-1-YFP mice were treated with whole-brain radiotherapy or sham with or without memantine. Results: Unlike previously reported long-term losses of dendritic spines, we found that the acute response to radiation is an initial increase in spines and excitatory synapses followed by a decrease in spine/synapse density with altered spine dynamics. Memantine pre-administration prevented this radiation-induced synaptic remodeling. Conclusion: These results demonstrate that radiation causes rapid, dynamic changes in synaptic structural plasticity, implicate abnormal glutamate signaling in cognitive dysfunction following brain irradiation, and describe a protective mechanism of memantine.


Subject(s)
Abnormalities, Radiation-Induced/prevention & control , Dendritic Spines/drug effects , Gamma Rays/adverse effects , Hippocampus/drug effects , Memantine/pharmacology , Synapses/drug effects , Abnormalities, Radiation-Induced/etiology , Abnormalities, Radiation-Induced/pathology , Animals , Cells, Cultured , Dendritic Spines/pathology , Dendritic Spines/radiation effects , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/pathology , Hippocampus/radiation effects , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Synapses/radiation effects
17.
Radiat Res ; 189(1): 53-63, 2018 01.
Article in English | MEDLINE | ID: mdl-29136391

ABSTRACT

Radiation from galactic cosmic rays (GCR) poses a significant health risk for deep-space flight crews. GCR are unique in their extremely high-energy particles. With current spacecraft shielding technology, some of the predominant particles astronauts would be exposed to are 1H + 16O. Radiation has been shown to cause cognitive deficits in mice. The hippocampus plays a key role in memory and cognitive tasks; it receives information from the cortex, undergoes dendritic-dependent processing and then relays information back to the cortex. In this study, we investigated the effects of combined 1H + 16O irradiation on cognition and dendritic structures in the hippocampus of adult male mice three months postirradiation. Six-month-old male C57BL/6 mice were irradiated first with 1H (0.5 Gy, 150 MeV/n) and 1 h later with 16O (0.1 Gy, 600 MeV/n) at the NASA Space Radiation Laboratory (Upton, NY). Three months after irradiation, animals were tested for hippocampus-dependent cognitive performance using the Y-maze. Upon sacrifice, molecular and morphological assessments were performed on hippocampal tissues. During Y-maze testing, the irradiated mice failed to distinguish the novel arm, spending approximately the same amount of time in all three arms during the retention trial relative to sham-treated controls. Irradiated animals also showed changes in expression of glutamate receptor subunits and synaptic density-associated proteins. 1H + 16O radiation compromised dendritic morphology in the cornu ammonis 1 and dentate gyrus within the hippocampus. These data indicate cognitive injuries due to 1H + 16O at three months postirradiation.


Subject(s)
Hippocampus/physiology , Hippocampus/radiation effects , Hydrogen/adverse effects , Memory, Short-Term/radiation effects , Oxygen/adverse effects , Animals , Behavior, Animal/drug effects , Cosmic Radiation/adverse effects , Gene Expression Regulation/radiation effects , Male , Mice , Mice, Inbred C57BL , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Synapses/radiation effects
18.
Sci Rep ; 7(1): 15574, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29138442

ABSTRACT

The skin senses external environment, including ultraviolet light (UV). Hippocampus is a brain region that is responsible for memory and emotion. However, changes in hippocampus by UV irradiation to the skin have not been studied. In this study, after 2 weeks of UV irradiation to the mouse skin, we examined molecular changes related to cognitive functions in the hippocampus and activation of the hypothalamic-pituitary-adrenal (HPA) axis. UV exposure to the skin decreased doublecortin-positive immature neurons and synaptic proteins, including N-methyl-D-aspartate receptor 2 A and postsynaptic density protein-95, in the hippocampus. Moreover, we observed that UV irradiation to the skin down-regulated brain-derived neurotrophic factor expression and ERK signaling in the hippocampus, which are known to modulate neurogenesis and synaptic plasticity. The cutaneous and central HPA axes were activated by UV, which resulted in significant increases in serum levels of corticosterone. Subsequently, UV irradiation to the skin activated the glucocorticoid-signaling pathway in the hippocampal dentate gyrus. Interestingly, after 6 weeks of UV irradiation, mice showed depression-like behavior in the tail suspension test. Taken together, our data suggest that repeated UV exposure through the skin may negatively affect hippocampal neurogenesis and synaptic plasticity along with HPA axis activation.


Subject(s)
Depressive Disorder/genetics , Disks Large Homolog 4 Protein/genetics , Neurogenesis/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Brain-Derived Neurotrophic Factor/blood , Corticosterone/blood , Depressive Disorder/blood , Depressive Disorder/physiopathology , Gene Expression Regulation/radiation effects , Hypothalamo-Hypophyseal System/metabolism , MAP Kinase Signaling System/radiation effects , Male , Mice , Neuronal Plasticity/radiation effects , Pituitary-Adrenal System/metabolism , Skin/metabolism , Skin/radiation effects , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Synapses/metabolism , Synapses/radiation effects , Temporal Lobe/physiopathology , Ultraviolet Rays
19.
Nat Commun ; 8(1): 1220, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089553

ABSTRACT

Vision in dim light depends on synapses between rods and rod bipolar cells (RBCs). Here, we find that these synapses exist in multiple configurations, in which single release sites of rods are apposed by one to three postsynaptic densities (PSDs). Single RBCs often form multiple PSDs with one rod; and neighboring RBCs share ~13% of their inputs. Rod-RBC synapses develop while ~7% of RBCs undergo programmed cell death (PCD). Although PCD is common throughout the nervous system, its influences on circuit development and function are not well understood. We generate mice in which ~53 and ~93% of RBCs, respectively, are removed during development. In these mice, dendrites of the remaining RBCs expand in graded fashion independent of light-evoked input. As RBC dendrites expand, they form fewer multi-PSD contacts with rods. Electrophysiological recordings indicate that this homeostatic co-regulation of neurite and synapse development preserves retinal function in dim light.


Subject(s)
Homeostasis , Neuronal Plasticity/physiology , Synapses/physiology , Visual Pathways/physiology , Animals , Axons/metabolism , Cell Count , Dendrites/physiology , Dendrites/radiation effects , Homeostasis/radiation effects , Light , Mice, Transgenic , Neuronal Plasticity/radiation effects , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/radiation effects , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/radiation effects , Synapses/radiation effects , Visual Pathways/radiation effects
20.
Sci Rep ; 7(1): 13802, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062068

ABSTRACT

Electrical stimulation is an important tool in neuroscience research and clinically. In the retina, extensive work has revealed how the retinal ganglion cells respond to extracellular electrical stimulation. But little is known about the responses of other neuronal types, and more generally, how the network responds to stimulation. We conducted a survey of electrically evoked responses, over a range of pulse amplitudes and pulse widths, for 21 cell types spanning the inner two layers of the rabbit retina. It revealed: (i) the evoked responses of some neurons were charge insensitive; (ii) pulse-width sensitivity varied between cell types, allowing preferential recruitment of cell types; and (iii) 10-20 Hz damped oscillations across retinal layers. These oscillations were generated by reciprocal excitatory / inhibitory synapses, at locations as early as the cone-horizontal-cell synapses. These results illustrate at cellular resolution how a network responds to extracellular stimulation, and could inform the development of bioelectronic implants for treating blindness.


Subject(s)
Electric Stimulation/methods , Evoked Potentials, Visual/radiation effects , Neurons/physiology , Retinal Ganglion Cells/physiology , Synapses/physiology , Animals , Cells, Cultured , Neurons/cytology , Neurons/radiation effects , Rabbits , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/radiation effects , Synapses/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...