Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.012
Filter
1.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38831600

ABSTRACT

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.


Subject(s)
Brain Injuries , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Pyroptosis/radiation effects , Pyroptosis/physiology , Microglia/metabolism , Microglia/radiation effects , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Mice , Humans , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/etiology , Male , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Coculture Techniques , Radiation Injuries/pathology , Radiation Injuries/metabolism , Female , Mice, Inbred C57BL , Middle Aged
2.
Sci Rep ; 14(1): 12274, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806540

ABSTRACT

Cranial irradiation used to control brain malignancies invariably leads to progressive and debilitating declines in cognition. Clinical efforts implementing hippocampal avoidance and NMDAR antagonism, have sought to minimize dose to radiosensitive neurogenic regions while normalizing excitatory/inhibitory (E/I) tone. Results of these trials have yielded only marginal benefits to cognition, prompting current studies to evaluate the potential of systemic extracellular vesicle (EV) therapy to restore neurocognitive functionality in the irradiated brain. Here we tested the hypothesis that EVs derived from inhibitory but not excitatory neuronal cultures would prove beneficial to cognition and associated pathology. Rats subjected to a clinically relevant, fractionated cranial irradiation paradigm were given multiple injections of either GABAergic- or glutamatergic-derived EV and subjected to behavioral testing. Rats treated with GABAergic but not glutamatergic EVs showed significant improvements on hippocampal- and cortical-dependent behavioral tasks. While each treatment enhanced levels of the neurotrophic factors BDNF and GDNF, only GABAergic EVs preserved granule cell neuron dendritic spine density. Additional studies conducted with GABAergic EVs, confirmed significant benefits on amygdala-dependent behavior and modest changes in synaptic plasticity as measured by long-term potentiation. These data point to a potentially more efficacious approach for resolving radiation-induced neurological deficits, possibly through a mechanism able to restore homeostatic E/I balance.


Subject(s)
Cranial Irradiation , Extracellular Vesicles , GABAergic Neurons , Animals , Extracellular Vesicles/metabolism , Rats , Cranial Irradiation/adverse effects , GABAergic Neurons/metabolism , GABAergic Neurons/radiation effects , Male , Hippocampus/radiation effects , Hippocampus/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neurons/radiation effects , Neurons/metabolism , Glutamic Acid/metabolism , Neuronal Plasticity/radiation effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Behavior, Animal/radiation effects
3.
Clin. transl. oncol. (Print) ; 25(11): 3165-3173, 11 nov. 2023. ilus, graf
Article in English | IBECS | ID: ibc-226841

ABSTRACT

Purpose Radiotherapy is a critical component of cancer treatment, along with surgery and chemotherapy. Approximately, 90% of cancer patients undergoing pelvic radiotherapy show gastrointestinal (GI) toxicity, including bloody diarrhea, and gastritis, most of which are associated with gut dysbiosis. In addition to the direct effect of radiation on the brain, pelvic irradiation can alter the gut microbiome, leading to inflammation and breakdown of the gut–blood barrier. This allows toxins and bacteria to enter the bloodstream and reach the brain. Probiotics have been proven to prevent GI toxicity by producing short-chain fatty acids and exopolysaccharides beneficial for protecting mucosal integrity and oxidative stress reduction in the intestine and also shown to be beneficial in brain health. Microbiota plays a significant role in maintaining gut and brain health, so it is important to study whether bacterial supplementation will help in maintaining the gut and brain structure after radiation exposure. Methods In the present study, male C57BL/6 mice were divided into control, radiation, probiotics, and probiotics + radiation groups. On the 7th day, animals in the radiation and probiotics + radiation groups received a single dose of 4 Gy to whole-body. Posttreatment, mice were sacrificed, and the intestine and brain tissues were excised for histological analysis to assess GI and neuronal damage. Results Radiation-induced damage to the villi height and mucosal thickness was mitigated by the probiotic treatment significantly (p < 0.01). Further, radiation-induced pyknotic cell numbers in the DG, CA2, and CA3 areas were substantially reduced with bacterial supplementation (p < 0.001). Similarly, probiotics reduced neuronal inflammation induced by radiation in the cortex, CA2, and DG region (p < 0.01) (AU)


Subject(s)
Humans , Animals , Male , Mice , Probiotics/therapeutic use , Radiation-Protective Agents , Gastrointestinal Tract/radiation effects , Neurons/radiation effects , Inflammation/metabolism , Mice, Inbred C57BL , Disease Models, Animal
4.
Science ; 381(6657): 495, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37535725
5.
Adv Sci (Weinh) ; 10(25): e2301854, 2023 09.
Article in English | MEDLINE | ID: mdl-37386797

ABSTRACT

Optoelectronic biointerfaces have gained significant interest for wireless and electrical control of neurons. Three-dimentional (3D) pseudocapacitive nanomaterials with large surface areas and interconnected porous structures have great potential for optoelectronic biointerfaces that can fulfill the requirement of high electrode-electrolyte capacitance to effectively transduce light into stimulating ionic currents. In this study, the integration of 3D manganese dioxide (MnO2 ) nanoflowers into flexible optoelectronic biointerfaces for safe and efficient photostimulation of neurons is demonstrated. MnO2 nanoflowers are grown via chemical bath deposition on the return electrode, which has a MnO2 seed layer deposited via cyclic voltammetry. They facilitate a high interfacial capacitance (larger than 10 mF cm-2 ) and photogenerated charge density (over 20 µC cm-2 ) under low light intensity (1 mW mm-2 ). MnO2 nanoflowers induce safe capacitive currents with reversible Faradaic reactions and do not cause any toxicity on hippocampal neurons in vitro, making them a promising material for biointerfacing with electrogenic cells. Patch-clamp electrophysiology is recorded in the whole-cell configuration of hippocampal neurons, and the optoelectronic biointerfaces trigger repetitive and rapid firing of action potentials in response to light pulse trains. This study points out the potential of electrochemically-deposited 3D pseudocapacitive nanomaterials as a robust building block for optoelectronic control of neurons.


Subject(s)
Electrochemistry , Light , Manganese Compounds , Nanostructures , Neurons , Oxides , Action Potentials/radiation effects , Electric Capacitance , Electrochemistry/methods , Electrodes , Electrolytes/chemistry , Electrolytes/radiation effects , Electrophysiology , Hippocampus/cytology , Manganese Compounds/chemistry , Nanostructures/adverse effects , Nanostructures/chemistry , Nanostructures/radiation effects , Neurons/metabolism , Neurons/radiation effects , Oxides/chemistry , Patch-Clamp Techniques , Photic Stimulation , Wireless Technology , Humans , Animals , Rats
6.
Neurochem Int ; 162: 105463, 2023 01.
Article in English | MEDLINE | ID: mdl-36513311

ABSTRACT

NLRP3 inflammasome activation is implicated in irradiation-induced cognitive dysfunction. Alternate-day fasting (ADF) has been demonstrated to improve neuroinflammation as a non-pharmacological intervention. However, the exact mechanism and the anti-inflammatory effect in irradiation-induced cognitive dysfunction still need further in-depth study. The present study examined the effects of eight-week ADF on the cognitive functions of mice as well as inflammasome-mediated hippocampal neuronal loss following irradiation in mouse models of irradiation-induced cognitive deficits using seven-week-old male C57BL/6J mice. The behavioral results of novel place recognition and object recognition tasks revealed that ADF ameliorated cognitive functions in irradiation-induced cognitive dysfunction mice. ADF inhibited the expression of components of the NLRP3 inflammasome (NLRP3, ASC, and Cl.caspase-1), the downstream inflammatory factor (IL-1ß and IL-18), and apoptosis-related proteins (caspase-3) via western blotting. Furthermore, an increased number of neurons and activated astrocytes were observed in the hippocampus using immunohistochemistry and Sholl analysis, which was jointly confirmed by western blotting. According to our study, this is the first time we found that ADF improved cognitive dysfunction induced by irradiation, and the anti-inflammatory effect of ADF could be due to inhibition in NLRP3-mediated hippocampal neuronal loss by suppressing astrocyte activation.


Subject(s)
Cognitive Dysfunction , Hippocampus , Intermittent Fasting , Radiation Injuries , Animals , Male , Mice , Apoptosis Regulatory Proteins/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Hippocampus/pathology , Hippocampus/radiation effects , Inflammasomes/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Neuroinflammatory Diseases/therapy , Neurons/pathology , Neurons/radiation effects , Radiotherapy/adverse effects
7.
Sci Rep ; 12(1): 3506, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241689

ABSTRACT

Exposures to radiofrequency electromagnetic fields (RF-EMFs, 100 kHz to 6 GHz) have been associated with both positive and negative effects on cognitive behavior. To elucidate the mechanism of RF-EMF interaction, a few studies have examined its impact on neuronal activity and synaptic plasticity. However, there is still a need for additional basic research that further our understanding of the underlying mechanisms of RF-EMFs on the neuronal system. The present study investigated changes in neuronal activity and synaptic transmission following a 60-min exposure to 3.0 GHz RF-EMF at a low dose (specific absorption rate (SAR) < 1 W/kg). We showed that RF-EMF exposure decreased the amplitude of action potential (AP), depolarized neuronal resting membrane potential (MP), and increased neuronal excitability and synaptic transmission in cultured primary hippocampal neurons (PHNs). The results show that RF-EMF exposure can alter neuronal activity and highlight that more investigations should be performed to fully explore the RF-EMF effects and mechanisms.


Subject(s)
Electromagnetic Fields , Hippocampus , Neurons , Electromagnetic Fields/adverse effects , Hippocampus/radiation effects , Neurons/radiation effects , Radio Waves/adverse effects
8.
Nature ; 602(7897): 461-467, 2022 02.
Article in English | MEDLINE | ID: mdl-35140401

ABSTRACT

Visual cortical neurons encode the position and motion direction of specific stimuli retrospectively, without any locomotion or task demand1. The hippocampus, which is a part of the visual system, is hypothesized to require self-motion or a cognitive task to generate allocentric spatial selectivity that is scalar, abstract2,3 and prospective4-7. Here we measured rodent hippocampal selectivity to a moving bar of light in a body-fixed rat to bridge these seeming disparities. About 70% of dorsal CA1 neurons showed stable activity modulation as a function of the angular position of the bar, independent of behaviour and rewards. One-third of tuned cells also encoded the direction of revolution. In other experiments, neurons encoded the distance of the bar, with preference for approaching motion. Collectively, these demonstrate visually evoked vectorial selectivity (VEVS). Unlike place cells, VEVS was retrospective. Changes in the visual stimulus or its predictability did not cause remapping but only caused gradual changes. Most VEVS-tuned neurons behaved like place cells during spatial exploration and the two selectivities were correlated. Thus, VEVS could form the basic building block of hippocampal activity. When combined with self-motion, reward or multisensory stimuli8, it can generate the complexity of prospective representations including allocentric space9, time10,11 and episodes12.


Subject(s)
Hippocampus , Light , Space Perception , Spatial Processing , Visual Cortex , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/radiation effects , Hippocampus/cytology , Hippocampus/physiology , Hippocampus/radiation effects , Neurons/physiology , Neurons/radiation effects , Rats , Visual Cortex/cytology , Visual Cortex/physiology
9.
Clin Neurophysiol ; 135: 51-73, 2022 03.
Article in English | MEDLINE | ID: mdl-35033772

ABSTRACT

Transcranial ultrasound stimulation (TUS) holds great potential as a tool to alter neural circuits non-invasively in both animals and humans. In contrast to established non-invasive brain stimulation methods, ultrasonic waves can be focused on both cortical and deep brain targets with the unprecedented spatial resolution as small as a few cubic millimeters. This focusing allows exclusive targeting of small subcortical structures, previously accessible only by invasive deep brain stimulation devices. The neuromodulatory effects of TUS are likely derived from the kinetic interaction of the ultrasound waves with neuronal membranes and their constitutive mechanosensitive ion channels, to produce short term and long-lasting changes in neuronal excitability and spontaneous firing rate. After decades of mechanistic and safety investigation, the technique has finally come of age, and an increasing number of human TUS studies are expected. Given its excellent compatibility with non-invasive brain mapping techniques, such as electroencephalography (EEG) and functional magnetic resonance imaging (fMRI), as well as neuromodulatory techniques, such as transcranial magnetic stimulation (TMS), systemic TUS effects can readily be assessed in both basic and clinical research. In this review, we present the fundamentals of TUS for a broader audience. We provide up-to-date information on the physical and neurophysiological mechanisms of TUS, available readouts for its neural and behavioral effects, insights gained from animal models and human studies, potential clinical applications, and safety considerations. Moreover, we discuss the indirect effects of TUS on the nervous system through peripheral co-stimulation and how these confounding factors can be mitigated by proper control conditions.


Subject(s)
Brain/physiology , Evoked Potentials , Neuronal Plasticity , Ultrasonography, Interventional/methods , Animals , Brain/cytology , Humans , Neurons/metabolism , Neurons/physiology , Neurons/radiation effects , Ultrasonic Waves
10.
Nat Commun ; 13(1): 417, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058427

ABSTRACT

Photoactivatable drugs targeting ligand-gated ion channels open up new opportunities for light-guided therapeutic interventions. Photoactivable toxins targeting ion channels have the potential to control excitable cell activities with low invasiveness and high spatiotemporal precision. As proof-of-concept, we develop HwTxIV-Nvoc, a UV light-cleavable and photoactivatable peptide that targets voltage-gated sodium (NaV) channels and validate its activity in vitro in HEK293 cells, ex vivo in brain slices and in vivo on mice neuromuscular junctions. We find that HwTxIV-Nvoc enables precise spatiotemporal control of neuronal NaV channel function under all conditions tested. By creating multiple photoactivatable toxins, we demonstrate the broad applicability of this toxin-photoactivation technology.


Subject(s)
Light , Peptides/toxicity , Toxins, Biological/toxicity , Voltage-Gated Sodium Channels/metabolism , Amino Acid Sequence , Animals , Brain/physiology , HEK293 Cells , Humans , Ion Channel Gating/radiation effects , Mice, Inbred C57BL , Neurons/physiology , Neurons/radiation effects , Peptides/chemical synthesis , Peptides/chemistry , Protein Engineering , Time Factors , Ultraviolet Rays , Zebrafish
11.
Front Public Health ; 9: 771508, 2021.
Article in English | MEDLINE | ID: mdl-34881219

ABSTRACT

Background: With the global popularity of communication devices such as mobile phones, there are increasing concerns regarding the effect of radiofrequency electromagnetic radiation (RF-EMR) on the brain, one of the most important organs sensitive to RF-EMR exposure at 1,800 MHz. However, the effects of RF-EMR exposure on neuronal cells are unclear. Neurite outgrowth plays a critical role in brain development, therefore, determining the effects of 1,800 MHz RF-EMR exposure on neurite outgrowth is important for exploring its effects on brain development. Objectives: We aimed to investigate the effects of 1,800 MHz RF-EMR exposure for 48 h on neurite outgrowth in neuronal cells and to explore the associated role of the Rap1 signaling pathway. Material and Methods: Primary hippocampal neurons from C57BL/6 mice and Neuro2a cells were exposed to 1,800 MHz RF-EMR at a specific absorption rate (SAR) value of 4 W/kg for 48 h. CCK-8 assays were used to determine the cell viability after 24, 48, and 72 h of irradiation. Neurite outgrowth of primary hippocampal neurons (DIV 2) and Neuro2a cells was observed with a 20 × optical microscope and recognized by ImageJ software. Rap1a and Rap1b gene expressions were detected by real-time quantitative PCR. Rap1, Rap1a, Rap1b, Rap1GAP, and p-MEK1/2 protein expressions were detected by western blot. Rap1-GTP expression was detected by immunoprecipitation. The role of Rap1-GTP was assessed by transfecting a constitutively active mutant plasmid (Rap1-Gly_Val-GFP) into Neuro2a cells. Results: Exposure to 1,800 MHz RF-EMR for 24, 48, and 72 h at 4 W/kg did not influence cell viability. The neurite length, primary and secondary neurite numbers, and branch points of primary mouse hippocampal neurons were significantly impaired by 48-h RF-EMR exposure. The neurite-bearing cell percentage and neurite length of Neuro2a cells were also inhibited by 48-h RF-EMR exposure. Rap1 activity was inhibited by 48-h RF-EMR with no detectable alteration in either gene or protein expression of Rap1. The protein expression of Rap1GAP increased after 48-h RF-EMR exposure, while the expression of p-MEK1/2 protein decreased. Overexpression of constitutively active Rap1 reversed the decrease in Rap1-GTP and the neurite outgrowth impairment in Neuro2a cells induced by 1,800 MHz RF-EMR exposure for 48 h. Conclusion: Rap1 activity and related signaling pathways are involved in the disturbance of neurite outgrowth induced by 48-h 1,800 MHz RF-EMR exposure. The effects of RF-EMR exposure on neuronal development in infants and children deserve greater focus.


Subject(s)
Hippocampus , Neurons , Animals , Electromagnetic Radiation , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Hippocampus/metabolism , Hippocampus/radiation effects , Humans , Mice , Mice, Inbred C57BL , Neuronal Outgrowth , Neurons/metabolism , Neurons/radiation effects
13.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830484

ABSTRACT

Radiation therapy represents one of the primary treatment modalities for primary and metastatic brain tumors. Although recent advances in radiation techniques, that allow the delivery of higher radiation doses to the target volume, reduce the toxicity to normal tissues, long-term neurocognitive decline is still a detrimental factor significantly affecting quality of life, particularly in pediatric patients. This imposes the need for the development of prevention strategies. Based on recent evidence, showing that manipulation of the Shh pathway carries therapeutic potential for brain repair and functional recovery after injury, here we evaluate how radiation-induced hippocampal alterations are modulated by the constitutive activation of the Shh signaling pathway in Patched 1 heterozygous mice (Ptch1+/-). Our results show, for the first time, an overall protective effect of constitutive Shh pathway activation on hippocampal radiation injury. This activation, through modulation of the proneural gene network, leads to a long-term reduction of hippocampal deficits in the stem cell and new neuron compartments and to the mitigation of radio-induced astrogliosis, despite some behavioral alterations still being detected in Ptch1+/- mice. A better understanding of the pathogenic mechanisms responsible for the neural decline following irradiation is essential for identifying prevention measures to contain the harmful consequences of irradiation. Our data have important translational implications as they suggest a role for Shh pathway manipulation to provide the therapeutic possibility of improving brain repair and functional recovery after radio-induced injury.


Subject(s)
Hedgehog Proteins/genetics , Hippocampus/radiation effects , Neurogenesis/genetics , Patched-1 Receptor/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Gene Regulatory Networks/radiation effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Knockout , Neurogenesis/radiation effects , Neurons/metabolism , Neurons/radiation effects , Quality of Life , Radiation, Ionizing , Signal Transduction/radiation effects
14.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830365

ABSTRACT

BACKGROUND: Cerebral ischemia, a common cerebrovascular disease, is one of the great threats to human health and new targets for stroke therapy are needed. The transcriptional activity in the cell is regulated by epigenetic processes such as DNA methylation/demethylation, acetylation/deacetylation, histone methylation, etc. Changes in DNA methylation after ischemia can have both neuroprotective and neurotoxic effects depending on the degree of ischemia damage, the time elapsed after injury, and the site of methylation. METHODS: In this study, we investigated the changes in the expression and intracellular localization of DNA methyltransferase DNMT1, histone methyltransferases SUV39H1, and G9a in penumbra neurons and astrocytes at 4 and 24 h after stroke in the rat cerebral cortex using photothrombotic stroke (PTS) model. Methods of immunofluorescence microscopy analysis, apoptosis analysis, and immunoblotting were used. Additionally, we have studied the effect of DNMT1 and G9a inhibitors on the volume of PTS-induced infarction and apoptosis of penumbra cells in the cortex of mice after PTS. RESULTS: This study has shown that the level of DNMT1 increased in the nuclear and cytoplasmic fractions of the penumbra tissue at 24 h after PTS. Inhibition of DNMT1 by 5-aza-2'-deoxycytidine protected cells of PTS-induced penumbra from apoptosis. An increase in the level of SUV39H1 in the penumbra was found at 24 h after PTS and G9a was overexpressed at 4 and 24 h after PTS. G9a inhibitors A-366 and BIX01294 protected penumbra cells from apoptosis and reduced the volume of PTS-induced cerebral infarction. CONCLUSION: Thus, the data obtained show that DNA methyltransferase DNMT1 and histone methyltransferase G9a can be potential protein targets in ischemic penumbra cells, and their inhibitors are potential neuroprotective agents capable of protecting penumbra cells from postischemic damage to the cerebral cortex.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , Histone-Lysine N-Methyltransferase/genetics , Methyltransferases/genetics , Neurons/metabolism , Repressor Proteins/genetics , Stroke/genetics , Animals , Astrocytes/metabolism , Astrocytes/radiation effects , Cerebral Cortex/metabolism , Cerebral Cortex/radiation effects , DNA Methylation/radiation effects , Disease Models, Animal , Gene Expression Regulation, Enzymologic/radiation effects , Humans , Light , Mice , Neurons/pathology , Neurons/radiation effects , Rats , Stroke/pathology , Stroke/therapy
15.
Nat Commun ; 12(1): 5662, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580314

ABSTRACT

Using multi-color visible lights for independent optogenetic manipulation of multiple neuronal populations offers the ability for sophisticated brain functions and behavior dissection. To mitigate invasive fiber insertion, infrared light excitable upconversion nanoparticles (UCNPs) with deep tissue penetration have been implemented in optogenetics. However, due to the chromatic crosstalk induced by the multiple emission peaks, conventional UCNPs or their mixture cannot independently activate multiple targeted neuronal populations. Here, we report NIR multi-color optogenetics by the well-designed trichromatic UCNPs with excitation-specific luminescence. The blue, green and red color emissions can be separately tuned by switching excitation wavelength to match respective spectral profiles of optogenetic proteins ChR2, C1V1 and ChrimsonR, which enables selective activation of three distinct neuronal populations. Such stimulation with tunable intensity can not only activate distinct neuronal populations selectively, but also achieve transcranial selective modulation of the motion behavior of awake-mice, which opens up a possibility of multi-color upconversion optogenetics.


Subject(s)
Brain/physiology , Deep Brain Stimulation/methods , Infrared Rays , Nanoparticles/radiation effects , Optogenetics/methods , Animals , Brain/cytology , Brain/radiation effects , Color , Male , Mice , Microscopy, Electron, Transmission , Models, Animal , Movement/physiology , Neurons/physiology , Neurons/radiation effects , Patch-Clamp Techniques , Single Molecule Imaging/methods , Stereotaxic Techniques
16.
Adv Sci (Weinh) ; 8(21): e2101934, 2021 11.
Article in English | MEDLINE | ID: mdl-34546652

ABSTRACT

Ultrasound is a promising new modality for non-invasive neuromodulation. Applied transcranially, it can be focused down to the millimeter or centimeter range. The ability to improve the treatment's spatial resolution to a targeted brain region could help to improve its effectiveness, depending upon the application. The present paper details a neurostimulation scheme using gas-filled nanostructures, gas vesicles (GVs), as actuators for improving the efficacy and precision of ultrasound stimuli. Sonicated primary neurons display dose-dependent, repeatable Ca2+ responses, closely synced to stimuli, and increased nuclear expression of the activation marker c-Fos in the presence of GVs. GV-mediated ultrasound triggered rapid and reversible Ca2+ responses in vivo and could selectively evoke neuronal activation in a deep-seated brain region. Further investigation indicate that mechanosensitive ion channels are important mediators of this effect. GVs themselves and the treatment scheme are also found not to induce significant cytotoxicity, apoptosis, or membrane poration in treated cells. Altogether, this study demonstrates a simple and effective method to achieve enhanced and better-targeted neurostimulation with non-invasive low-intensity ultrasound.


Subject(s)
Nanostructures/chemistry , Ultrasonic Waves , Unilamellar Liposomes/chemistry , Ventral Tegmental Area/metabolism , Anabaena/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Gases/chemistry , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Neurons/radiation effects , Rats , Unilamellar Liposomes/metabolism , Ventral Tegmental Area/pathology , Ventral Tegmental Area/radiation effects
17.
Biomed Res Int ; 2021: 2237370, 2021.
Article in English | MEDLINE | ID: mdl-34527734

ABSTRACT

Currently, the impact of electromagnetic field (EMF) exposure on the nervous system is an increasingly arousing public concern. The present study was designed to explore the effects of continuous long-term exposure to L-band high-power microwave (L-HPM) on brain function and related mechanisms. Forty-eight male Institute of Cancer Research (ICR) mice were exposed to L-HPM at various power densities (0.5, 1.0, and 1.5 W/m2) and the brain function was examined at different time periods after exposure. The morphology of the brain was examined by hematoxylin-eosin (HE) and deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining. Furthermore, cholinergic markers, oxidative stress markers, and the expression of c-fos were evaluated to identify a "potential" mechanism. The results showed that exposure to L-HPM at 1.5 W/m2 can cause generalized injuries in the hippocampus (CA1 and CA3) and cerebral cortex (the first somatosensory cortex) of mice, including cell apoptosis, cholinergic dysfunction, and oxidative damage. Moreover, the deleterious effects were closely related to the power density and exposure time, indicating that long-term and high-power density exposure may be detrimental to the nervous system.


Subject(s)
Brain/radiation effects , Cognition/radiation effects , Microwaves/adverse effects , Acetylcholinesterase , Animals , Apoptosis/physiology , Brain/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/radiation effects , China , Choline O-Acetyltransferase , Electromagnetic Fields/adverse effects , Hippocampus/metabolism , Hippocampus/radiation effects , Male , Mice , Mice, Inbred ICR , Neurons/metabolism , Neurons/radiation effects , Oxidative Stress/physiology , Proto-Oncogene Proteins c-fos/metabolism , Superoxide Dismutase-1
18.
Cells ; 10(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34359834

ABSTRACT

Astrocytes act as neural stem cells (NSCs) that have the potential to self-renew and differentiate into other neuronal cells. The protein expression of these astrocytes depends on the stage of differentiation, showing sequential expression of multiple proteins such as octamer-binding transcription factor 4 (Oct4), nestin, glial fibrillary acidic protein (GFAP), and aldehyde dehydrogenase 1 family member L1 (aldh1L1). Photobiomodulation (PBM) affects cell apoptosis, proliferation, migration, and adhesion. We hypothesized that astrocyte proliferation and differentiation would be modulated by PBM. We used an optimized astrocyte culture method and a 660-nanometer light-emitting diode (LED) to enhance the biological actions of many kinds of cells. We determined that the 660-nanometer LED promoted the biological actions of cultured astrocytes by increasing the reactive oxygen species levels. The overall viability of the cultured cells, which included various cells other than astrocytes, did not change after LED exposure; however, astrocyte-specific proliferation was observed by the increased co-expression of GFAP and bromodeoxyuridine (BrdU)/Ki67. Furthermore, the 660-nanometer LED provides evidence of differentiation, as shown by the decreased Oct4 and GFAP co-expression and increased nestin and aldh1L1 expression. These results demonstrate that a 660-nanometer LED can modify astrocyte proliferation, which suggests the efficacy of the therapeutic application of LED in various pathological states of the central nervous system.


Subject(s)
Astrocytes/radiation effects , Cell Proliferation/radiation effects , Gene Expression/radiation effects , Neurons/radiation effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cell Adhesion/radiation effects , Cell Differentiation/radiation effects , Cell Movement/radiation effects , Coculture Techniques , Embryo, Mammalian , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Lasers, Semiconductor , Light , Nestin/genetics , Nestin/metabolism , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism
19.
Neuron ; 109(20): 3268-3282.e6, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34416169

ABSTRACT

The suprachiasmatic nucleus (SCN) is the master circadian pacemaker in mammals and is entrained by environmental light. However, the molecular basis of the response of the SCN to light is not fully understood. We used RNA/chromatin immunoprecipitation/single-nucleus sequencing with circadian behavioral assays to identify mouse SCN cell types and explore their responses to light. We identified three peptidergic cell types that responded to light in the SCN: arginine vasopressin (AVP), vasoactive intestinal peptide (VIP), and cholecystokinin (CCK). In each cell type, light-responsive subgroups were enriched for expression of neuronal Per-Arnt-Sim (PAS) domain protein 4 (NPAS4) target genes. Further, mice lacking Npas4 had a longer circadian period under constant conditions, a damped phase response curve to light, and reduced light-induced gene expression in the SCN. Our data indicate that NPAS4 is necessary for normal transcriptional responses to light in the SCN and critical for photic phase-shifting of circadian behavior.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Circadian Rhythm/genetics , Light , Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Arginine Vasopressin/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cholecystokinin/metabolism , Chromatin Immunoprecipitation , Circadian Rhythm/radiation effects , Gene Expression Profiling , Mice , Mice, Knockout , Neurons/radiation effects , Sequence Analysis, RNA , Single-Cell Analysis , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/radiation effects , Vasoactive Intestinal Peptide/metabolism
20.
Cell Rep ; 36(1): 109313, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233180

ABSTRACT

Perineuronal nets (PNNs), components of the extracellular matrix, preferentially coat parvalbumin-positive interneurons and constrain critical-period plasticity in the adult cerebral cortex. Current strategies to remove PNN are long-lasting, invasive, and trigger neuropsychiatric symptoms. Here, we apply repeated anesthetic ketamine as a method with minimal behavioral effect. We find that this paradigm strongly reduces PNN coating in the healthy adult brain and promotes juvenile-like plasticity. Microglia are critically involved in PNN loss because they engage with parvalbumin-positive neurons in their defined cortical layer. We identify external 60-Hz light-flickering entrainment to recapitulate microglia-mediated PNN removal. Importantly, 40-Hz frequency, which is known to remove amyloid plaques, does not induce PNN loss, suggesting microglia might functionally tune to distinct brain frequencies. Thus, our 60-Hz light-entrainment strategy provides an alternative form of PNN intervention in the healthy adult brain.


Subject(s)
Anesthetics/pharmacology , Brain/physiology , Brain/radiation effects , Ketamine/pharmacology , Light , Nerve Net/physiology , Neurons/physiology , Neurons/radiation effects , Aging/physiology , Animals , Brain/drug effects , Female , Mice, Inbred C57BL , Microglia , Nerve Net/drug effects , Nerve Net/radiation effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Neurons/drug effects , Parvalbumins/metabolism , Photic Stimulation
SELECTION OF CITATIONS
SEARCH DETAIL
...