Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Elife ; 82019 11 18.
Article in English | MEDLINE | ID: mdl-31736463

ABSTRACT

Optogenetics allows manipulations of genetically and spatially defined neuronal populations with excellent temporal control. However, neurons are coupled with other neurons over multiple length scales, and the effects of localized manipulations thus spread beyond the targeted neurons. We benchmarked several optogenetic methods to inactivate small regions of neocortex. Optogenetic excitation of GABAergic neurons produced more effective inactivation than light-gated ion pumps. Transgenic mice expressing the light-dependent chloride channel GtACR1 produced the most potent inactivation. Generally, inactivation spread substantially beyond the photostimulation light, caused by strong coupling between cortical neurons. Over some range of light intensity, optogenetic excitation of inhibitory neurons reduced activity in these neurons, together with pyramidal neurons, a signature of inhibition-stabilized neural networks ('paradoxical effect'). The offset of optogenetic inactivation was followed by rebound excitation in a light dose-dependent manner, limiting temporal resolution. Our data offer guidance for the design of in vivo optogenetics experiments.


Subject(s)
GABAergic Neurons/radiation effects , Light Signal Transduction/genetics , Neocortex/radiation effects , Nerve Net/radiation effects , Pyramidal Cells/radiation effects , Somatosensory Cortex/radiation effects , Animals , Benchmarking , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Expression , Genes, Reporter , Light , Mice , Mice, Transgenic , Neocortex/cytology , Neocortex/metabolism , Nerve Net/cytology , Nerve Net/metabolism , Optogenetics/methods , Photic Stimulation , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Spatio-Temporal Analysis , Transgenes
2.
Opt Lett ; 43(15): 3802-3805, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30067683

ABSTRACT

Neocortical systems encode information in electrochemical spike timings, not just mean firing rates. Learning and memory in networks of spiking neurons is achieved by the precise timing of action potentials that induces synaptic strengthening (with excitation) or weakening (with inhibition). Inhibition should be incorporated into brain-inspired spike processing in the optical domain to enhance its information-processing capability. We demonstrate the simultaneous excitatory and inhibitory dynamics in an excitable (i.e., a pulsed) laser neuron, both numerically and experimentally. We investigate the bias strength effect, inhibitory strength effect, and excitatory and inhibitory input timing effect, based on the simulation platform of an integrated graphene excitable laser. We further corroborate these analyses with proof-of-principle experiments utilizing a fiber-based graphene excitable laser, where we introduce inhibition by directly modulating the gain of the laser. This technology may potentially open novel spike-processing functionality for future neuromorphic photonic systems.


Subject(s)
Electrophysiological Phenomena/radiation effects , Lasers , Models, Neurological , Neocortex/cytology , Neocortex/physiology , Neocortex/radiation effects , Neurons/cytology , Neurons/radiation effects , Time Factors
3.
Brain Res ; 1646: 543-550, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27369449

ABSTRACT

Diffusion within the extracellular and perivascular spaces of the brain plays an important role in biological processes, therapeutic delivery, and clearance mechanisms within the central nervous system. Recently, ultrasound has been used to enhance the dispersion of locally administered molecules and particles within the brain, but ultrasound-mediated effects on the brain parenchyma remain poorly understood. We combined an electron microscopy-based ultrastructural analysis with high-resolution tracking of non-adhesive nanoparticles in order to probe changes in the extracellular and perivascular spaces of the brain following a non-destructive pulsed ultrasound regimen known to alter diffusivity in other tissues. Freshly obtained rat brain neocortical slices underwent sham treatment or pulsed, low intensity ultrasound for 5min at 1MHz. Transmission electron microscopy revealed intact cells and blood vessels and evidence of enlarged spaces, particularly adjacent to blood vessels, in ultrasound-treated brain slices. Additionally, ultrasound significantly increased the diffusion rate of 100nm, 200nm, and 500nm nanoparticles that were injected into the brain slices, while 2000nm particles were unaffected. In ultrasound-treated slices, 91.6% of the 100nm particles, 20.7% of the 200nm particles, 13.8% of the 500nm particles, and 0% of the 2000nm particles exhibited diffusive motion. Thus, pulsed ultrasound can have meaningful structural effects on the brain extracellular and perivascular spaces without evidence of tissue disruption.


Subject(s)
Extracellular Space/radiation effects , Neocortex/radiation effects , Ultrasonic Waves , Animals , Diffusion , Extracellular Space/metabolism , Nanoparticles/administration & dosage , Neocortex/blood supply , Neocortex/metabolism , Neocortex/ultrastructure , Rats , Rats, Sprague-Dawley
4.
J Neurophysiol ; 116(3): 1012-23, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27281749

ABSTRACT

Two-photon imaging and optogenetic stimulation rely on high illumination powers, particularly for state-of-the-art applications that target deeper structures, achieve faster measurements, or probe larger brain areas. However, little information is available on heating and resulting damage induced by high-power illumination in the brain. In the current study we used thermocouple probes and quantum dot nanothermometers to measure temperature changes induced by two-photon microscopy in the neocortex of awake and anaesthetized mice. We characterized heating as a function of wavelength, exposure time, and distance from the center of illumination. Although total power is highest near the surface of the brain, heating was most severe hundreds of micrometers below the focal plane, due to heat dissipation through the cranial window. Continuous illumination of a 1-mm(2) area produced a peak temperature increase of ∼1.8°C/100 mW. Continuous illumination with powers above 250 mW induced lasting damage, detected with immunohistochemistry against Iba1, glial fibrillary acidic protein, heat shock proteins, and activated caspase-3. Higher powers were usable in experiments with limited duty ratios, suggesting an approach to mitigate damage in high-power microscopy experiments.


Subject(s)
Body Temperature/radiation effects , Heating , Microscopy, Confocal/methods , Neocortex/physiology , Neocortex/radiation effects , Analysis of Variance , Animals , Body Temperature/physiology , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Female , Gene Expression Regulation/radiation effects , Glial Fibrillary Acidic Protein/metabolism , Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Models, Neurological , Photons , Thermometry , Wakefulness
5.
Lasers Med Sci ; 29(3): 1117-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24232863

ABSTRACT

It has been reported that laser light irradiation (LLI) could regulate neuronal activities in the forebrain, but little is known if and how LLI in the red wavelength range affects neuronal excitability. Here, we investigated the effects of a continuous diode laser at 660 nm on intrinsic membrane properties and excitability of presumed pyramidal neurons in the thalamocortical input layer (layer 3/4) and in layer 5 of mouse primary auditory cortex using the whole-cell patch-clamp recording technique. In layer 3/4 neurons, 660-nm laser irradiation (LLI-660) at 20 mW for 5 min gradually increased resting membrane potentials, which reached a plateau after irradiation. Concomitantly, LLI-660 decreased onset latency of first action potentials (spikes) without changing spike threshold or peak amplitude, but increased inter-spike interval of initial bursting spike doublets and their peak amplitude ratio. None of these changes was observed in layer 5 neurons. Instead, LLI-660 at 20 mW rapidly reduced spike width ~5 % within 1 min of irradiation onset. The magnitude of this reduction did not change during 5 or 10 min irradiation, and returned quickly to at least baseline levels after turning the LLI off. Decreasing laser power to 10 mW reduced spike width to a lesser extent, suggesting laser power dependence of this phenomenon. These data suggest that LLI-660 regulates different aspects of neuronal excitability in cortical neurons in a layer-dependent manner possibly by affecting different voltage-gated ion channels.


Subject(s)
Action Potentials/radiation effects , Lasers , Neocortex/physiology , Neocortex/radiation effects , Pyramidal Cells/physiology , Pyramidal Cells/radiation effects , Animals , Cell Membrane/radiation effects , Mice , Patch-Clamp Techniques
6.
Radiats Biol Radioecol ; 54(6): 582-8, 2014.
Article in Russian | MEDLINE | ID: mdl-25980284

ABSTRACT

The effect of a local exposure of rat heads to X-ray radiation at a dose of 200 Gy on the number of phospho- lipids and neutral lipids in the nuclear fraction ofneocortex neurons and glia has been investigated A decrease in the amount ofphosphatidylinositol and an increase in sphingomyelin in neuronal nuclei occurred 2 h after irradiation at the time of repair of locomotive disorders. The amount of phosphatidylcholine and phosphati- dylinositol dropped, and the amount of sphingomyelin and cholesterol increased in the nuclei ofglial cells of the neocortex. Sphingomyelin, phosphatidylinositol, phosphatidylcholine and cholesterol of neuronal nuclei are involved in the dynamics of the CNS syndrome in mammals. Radio resistance of the responses of lipid nuclei in mammals with the CNS syndrome has been shown and a possible role of lipids in the post-irradia- tion DNA repair has been suggested.


Subject(s)
Central Nervous System Diseases/metabolism , Lipid Metabolism/radiation effects , Neocortex/metabolism , Neurons/metabolism , Animals , Cell Nucleus/radiation effects , Central Nervous System Diseases/pathology , Cholesterol/metabolism , Neocortex/radiation effects , Neuroglia/metabolism , Neuroglia/radiation effects , Neurons/radiation effects , Phosphatidylinositols/metabolism , Rats , Sphingomyelins/metabolism , X-Rays
7.
PLoS One ; 10(4): e0123833, 2014.
Article in English | MEDLINE | ID: mdl-25919297

ABSTRACT

BACKGROUND AND OBJECTIVE: Accumulating evidence has shown that low-power laser irradiation (LLI) affects cell proliferation and survival, but little is known about LLI effects on neural stem/progenitor cells (NSPCs). Here we investigate whether transcranial 532 nm LLI affects NSPCs in adult murine neocortex and in neurospheres from embryonic mice. STUDY DESIGN/MATERIALS AND METHODS: We applied 532 nm LLI (Nd:YVO4, CW, 60 mW) on neocortical surface via cranium in adult mice and on cultured cells from embryonic mouse brains in vitro to investigate the proliferation and migration of NSPCs and Akt expression using immunohistochemical assays and Western blotting techniques. RESULTS: In vivo experiments demonstrated that 532 nm LLI significantly facilitated the migration of GABAergic NSPCs that were induced to proliferate in layer 1 by mild ischemia. In vitro experiments using GABAergic NSPCs derived from embryonic day 14 ganglionic eminence demonstrated that 532 nm LLI for 60 min promoted the migration of GAD67-immunopositive NSPCs with a significant increase of Akt expression. Meanwhile, the LLI induced proliferation, but not migration, of NSPCs that give rise to excitatory neurons. CONCLUSION: It is concluded that 532 nm LLI promoted the migration of GABAergic NSPCs into deeper layers of the neocortex in vivo by elevating Akt expression.


Subject(s)
GABAergic Neurons/physiology , GABAergic Neurons/radiation effects , Neocortex/cytology , Neural Stem Cells/physiology , Neural Stem Cells/radiation effects , Animals , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , GABAergic Neurons/cytology , Gene Expression Regulation/radiation effects , Low-Level Light Therapy/methods , Mice , Neocortex/embryology , Neocortex/radiation effects , Proto-Oncogene Proteins c-akt/metabolism
8.
Cold Spring Harb Protoc ; 2013(12): 1153-60, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298038

ABSTRACT

The ability to form targeted vascular occlusions in small vessels of the brain is an important technique for studying the microscopic basis of cerebral ischemia. We describe two complementary methods that enable targeted occlusion of any single blood vessel within the upper 500 µm of adult rodent neocortex. Our goal is to generate highly localized regions of ischemia by blocking penetrating arterioles and ascending venules, which are bottlenecks of flow in the cortical angioarchitecture. One method, termed photothrombosis, makes use of linear optical absorption by a photosensitizer, transiently circulated in the blood stream, to induce a clot in a surface or near-surface segment of a vessel. The second method, termed plasma-mediated ablation, makes use of nonlinear optical interactions, without the need to introduce an exogenous absorber, to induce clots in subsurface segments of penetrating vessels, as well as subsurface microvessels and capillaries. The choice of the method for occlusion of individual vessels depends on the location of the vessels being studied and the objectives of the study. Here we describe concurrent high resolution in vivo imaging and auxiliary laser setups, occlusion protocols, and post hoc histological procedures.


Subject(s)
Blood Vessels/radiation effects , Hemostatic Techniques , Light , Neocortex/radiation effects , Animals , Photosensitizing Agents/radiation effects , Rodentia
9.
DNA Repair (Amst) ; 12(8): 645-55, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23683352

ABSTRACT

MCPH1 encodes BRCT-containing protein MCPH1/Microcephalin/BRIT1, mutations of which in humans cause autosomal recessive disorder primary microcephaly type 1 (MCPH1), characterized by a congenital reduction of brain size particularly in the cerebral cortex. We have shown previously that a deletion of Mcph1 in mice results in microcephaly because of a premature switch from symmetric to asymmetric division of the neuroprogenitors, which is regulated by MCPH1's function in the centrosome. Because MCPH1 has been implicated in ATM and ATR-mediated DNA damage response (DDR) and defective DDR is often associated with neurodevelopmental diseases, we wonder whether the DDR-related function of MCPH1 prevents microcephaly. Here, we show that a deletion of Mcph1 results in a specific reduction of the cerebral cortex at birth, which is persistent through life. Due to an effect on premature neurogenic production, Mcph1-deficient progenitors give rise to a high level of early-born neurons that form deep layers (IV-VI), while generate less late-born neurons that form a thinner outer layer (II-III) of the cortex. However, neuronal migration seems to be unaffected by Mcph1 deletion. Ionizing radiation (IR) induces a massive apoptosis in the Mcph1-null neocortex and also embryonic lethality. Finally, Mcph1 deletion compromises homologous recombination repair and increases genomic instability. Altogether, our data suggest that MCPH1 ensures proper neuroprogenitor expansion and differentiation not only through its function in the centrosome, but also in the DDR.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Damage , Microcephaly/genetics , Animals , Apoptosis/radiation effects , Cell Cycle Proteins , Cell Differentiation , Centrosome/metabolism , Centrosome/pathology , Chromosomal Proteins, Non-Histone/metabolism , Cytoskeletal Proteins , DNA Repair , Disease Models, Animal , Gene Deletion , Gene Knockout Techniques , Genomic Instability , Mice , Microcephaly/embryology , Microcephaly/pathology , Neocortex/embryology , Neocortex/pathology , Neocortex/radiation effects , Neurons/cytology , Neurons/pathology , Radiation, Ionizing , Recombination, Genetic
10.
Klin Khir ; (12): 55-60, 2012 Dec.
Article in Ukrainian | MEDLINE | ID: mdl-23610823

ABSTRACT

There were studied in experimental investigations the changes of dophamin synthesis in culture of neurons from middle brain (MB) in a newborn rats as well as in the water-ion metabolism in tissues of the rabbits big brain hemispheres and ultrastructure changes in the rabbits synaptic apparatus of the neocortex and MB neurons under the magnet-laser influence (MLI). The signs of intensive synthesis and transport of dophamin, changes of quantitative indices of water-ion metabolism as well as ultrastructural components in synaptic apparatus of neurons have had witnessed about activation of the neuromediator and water-ion metabolism and the MLI. All the abovementioned substantiates the possibility of MLI application in neurosurgery, neurology, traumatology in states, which are accompanied by disorders of the neuromediator and water-ion metabolism for prophylaxis of possible complications and the patients' quality of life improvement.


Subject(s)
Dopamine/biosynthesis , Infrared Rays , Lasers , Magnetic Fields , Neurons/metabolism , Neurons/radiation effects , Animals , Animals, Newborn , Cells, Cultured , Ion Transport/radiation effects , Male , Microscopy, Electron , Microscopy, Fluorescence , Neocortex/metabolism , Neocortex/radiation effects , Neocortex/ultrastructure , Neurons/ultrastructure , Potassium/metabolism , Rabbits , Rats , Rats, Wistar , Sodium/metabolism , Synapses/metabolism , Synaptic Transmission/radiation effects , Water/metabolism
11.
Radiats Biol Radioecol ; 51(6): 689-93, 2011.
Article in Russian | MEDLINE | ID: mdl-22384719

ABSTRACT

X-ray irradiation at a dose of 200 Gy with local exposure of the rat head induced the change of the lipid content in the neocortex tissue. The amount of phosphatidylinositol was decreased, the amount of free fatty acids, diglycerols, sphingomyelin was increased, and the amount of cholesterol had a growth trend in 2 h after X-ray exposition. The results testify in favor of participation of phosphatidylinositol- and sphingomyelin-relating signal systems and cholesterol in early stages of the cerebral radiation syndrome. We suggest that the change of the lipid content in early periods after the effect of a super-high dose of X-ray irradiation indicates the lipid dependence in the elimination of motion damages and the restoration of the functions of nerve cells. Effects on the lipid metabolism in the nerve tissue are promising for correcting the cerebral radiation syndrome.


Subject(s)
Cholesterol/metabolism , Fatty Acids, Nonesterified/metabolism , Neocortex/radiation effects , Phosphatidylinositols/metabolism , Sphingomyelins/metabolism , Animals , Head/radiation effects , Lipid Metabolism/radiation effects , Male , Maximum Tolerated Dose , Neurons/metabolism , Neurons/radiation effects , Radiation, Ionizing , Rats , Rats, Wistar
12.
Restor Neurol Neurosci ; 28(4): 399-417, 2010.
Article in English | MEDLINE | ID: mdl-20714065

ABSTRACT

Transcranial magnetic stimulation (TMS) can be used in two different ways to manipulate cortical information processing, either by applying a single pulse around the time point of expected task processing or by persistently shifting cortical excitability by repetitive stimulation (rTMS). Single pulses applied when specific cortical processing takes place always impair cortical function due to increased noise or enhanced inhibition, both resulting in decreased signal-to-noise ratio, while repetitive stimulation may allow to weaken or improve cortical processing depending on the type of stimulation. The opposite effects of low- ( approximately 1 Hz) and high-frequency rTMS (5-20 Hz), as well as the opposing effects of continuous versus intermittent theta-burst trains, lowering or raising cortical excitability respectively, have mainly been attributed to synaptic plasticity. As reviewed in this article, in a series of electrophysiological, immunohistochemical and molecular-biological animal experiments we obtained evidence for modulation of inhibitory cortical activity as a further reason of changing cortical excitability following rTMS.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/radiation effects , Transcranial Magnetic Stimulation , Animals , Brain Chemistry/radiation effects , Cats , Evoked Potentials, Motor/physiology , Evoked Potentials, Visual/physiology , Motor Cortex/physiology , Neocortex/cytology , Neocortex/radiation effects , Nerve Tissue Proteins/biosynthesis , Rats , Visual Cortex/cytology , Visual Cortex/radiation effects
13.
Mol Cell Neurosci ; 45(2): 180-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20599617

ABSTRACT

A considerable number of cells expressing typical immature neuronal markers including doublecortin (DCX+) are present around layer II in the cerebral cortex of young and adult guinea pigs and other larger mammals, and their origin and biological implication await further characterization. We show here in young adult guinea pigs that these DCX+ cells are accompanied by in situ cell division around the superficial cortical layers mostly in layer I, but they co-express proliferating cell nuclear antigen (PCNA) and an early neuronal fate determining factor, PAX6. A small number of these DCX+ cells also colocalize with BrdU following administration of this mitotic indicator. Cranial X-ray irradiation causes a decline of DCX+ cells around layer II, and novel environmental exploration induces c-Fos expression among these cells in several neocortical areas. Together, these data are compatible with a notion that DCX+ cortical neurons around layer II might derive from proliferable neuronal precursors around layer I in young adult guinea pig cerebrum, and that these cells might be modulated by experience under physiological conditions.


Subject(s)
Cerebrum/physiology , Neocortex/physiology , Neurogenesis , Animals , Cell Division , Cerebrum/cytology , Cerebrum/radiation effects , Doublecortin Domain Proteins , Eye Proteins/analysis , Guinea Pigs , Homeodomain Proteins/analysis , Microtubule-Associated Proteins/metabolism , Neocortex/cytology , Neocortex/radiation effects , Nerve Tissue Proteins/analysis , Neuropeptides/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/analysis , Proliferating Cell Nuclear Antigen/analysis , Proto-Oncogene Proteins c-fos/analysis , Repressor Proteins/analysis
14.
Neurosci Lett ; 473(1): 52-5, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20156525

ABSTRACT

The exposure of primary rat neocortical astroglial cell cultures to acute electromagnetic fields (EMF) in the microwave range was studied. Differentiated astroglial cell cultures at 14 days in vitro were exposed for 5, 10, or 20min to either 900MHz continuous waves or 900MHz waves modulated in amplitude at 50Hz using a sinusoidal waveform and 100% modulation index. The strength of the electric field (rms value) at the sample position was 10V/m. No change in cellular viability evaluated by MTT test and lactate dehydrogenase release was observed. A significant increase in ROS levels and DNA fragmentation was found only after exposure of the astrocytes to modulated EMF for 20min. No evident effects were detected when shorter time intervals or continuous waves were used. The irradiation conditions allowed the exclusion of any possible thermal effect. Our data demonstrate, for the first time, that even acute exposure to low intensity EMF induces ROS production and DNA fragmentation in astrocytes in primary cultures, which also represent the principal target of modulated EMF. Our findings also suggest the hypothesis that the effects could be due to hyperstimulation of the glutamate receptors, which play a crucial role in acute and chronic brain damage. Furthermore, the results show the importance of the amplitude modulation in the interaction between EMF and neocortical astrocytes.


Subject(s)
Astrocytes/radiation effects , DNA Fragmentation/radiation effects , Microwaves , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Electromagnetic Fields , Neocortex/cytology , Neocortex/metabolism , Neocortex/radiation effects , Rats , Rats, Wistar
15.
Nature ; 463(7277): 98-102, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20054397

ABSTRACT

The ability to silence the activity of genetically specified neurons in a temporally precise fashion would provide the opportunity to investigate the causal role of specific cell classes in neural computations, behaviours and pathologies. Here we show that members of the class of light-driven outward proton pumps can mediate powerful, safe, multiple-colour silencing of neural activity. The gene archaerhodopsin-3 (Arch) from Halorubrum sodomense enables near-100% silencing of neurons in the awake brain when virally expressed in the mouse cortex and illuminated with yellow light. Arch mediates currents of several hundred picoamps at low light powers, and supports neural silencing currents approaching 900 pA at light powers easily achievable in vivo. Furthermore, Arch spontaneously recovers from light-dependent inactivation, unlike light-driven chloride pumps that enter long-lasting inactive states in response to light. These properties of Arch are appropriate to mediate the optical silencing of significant brain volumes over behaviourally relevant timescales. Arch function in neurons is well tolerated because pH excursions created by Arch illumination are minimized by self-limiting mechanisms to levels comparable to those mediated by channelrhodopsins or natural spike firing. To highlight how proton pump ecological and genomic diversity may support new innovation, we show that the blue-green light-drivable proton pump from the fungus Leptosphaeria maculans (Mac) can, when expressed in neurons, enable neural silencing by blue light, thus enabling alongside other developed reagents the potential for independent silencing of two neural populations by blue versus red light. Light-driven proton pumps thus represent a high-performance and extremely versatile class of 'optogenetic' voltage and ion modulator, which will broadly enable new neuroscientific, biological, neurological and psychiatric investigations.


Subject(s)
Genetic Engineering/methods , Neurons/metabolism , Neurons/radiation effects , Proton Pumps/metabolism , Proton Pumps/radiation effects , Action Potentials/radiation effects , Animals , Ascomycota/metabolism , Ascomycota/radiation effects , Color , Electric Conductivity , Euryarchaeota/metabolism , Euryarchaeota/radiation effects , Hydrogen-Ion Concentration , Mice , Molecular Sequence Data , Neocortex/cytology , Neocortex/physiology , Neocortex/radiation effects , Proton Pumps/classification , Proton Pumps/genetics , Rhodopsins, Microbial/antagonists & inhibitors , Rhodopsins, Microbial/genetics , Rhodopsins, Microbial/metabolism , Rhodopsins, Microbial/radiation effects , Wakefulness
16.
Brain Res ; 1311: 189-96, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19879861

ABSTRACT

Increasing evidence indicates that oxidative stress may be involved in the adverse effects of radiofrequency (RF) radiation on the brain. Because mitochondrial DNA (mtDNA) defects are closely associated with various nervous system diseases and mtDNA is particularly susceptible to oxidative stress, the purpose of this study was to determine whether radiofrequency radiation can cause oxidative damage to mtDNA. In this study, we exposed primary cultured cortical neurons to pulsed RF electromagnetic fields at a frequency of 1800 MHz modulated by 217 Hz at an average special absorption rate (SAR) of 2 W/kg. At 24 h after exposure, we found that RF radiation induced a significant increase in the levels of 8-hydroxyguanine (8-OHdG), a common biomarker of DNA oxidative damage, in the mitochondria of neurons. Concomitant with this finding, the copy number of mtDNA and the levels of mitochondrial RNA (mtRNA) transcripts showed an obvious reduction after RF exposure. Each of these mtDNA disturbances could be reversed by pretreatment with melatonin, which is known to be an efficient antioxidant in the brain. Together, these results suggested that 1800 MHz RF radiation could cause oxidative damage to mtDNA in primary cultured neurons. Oxidative damage to mtDNA may account for the neurotoxicity of RF radiation in the brain.


Subject(s)
DNA Damage/radiation effects , DNA, Mitochondrial/radiation effects , Mitochondria/radiation effects , Neurons/radiation effects , Oxidative Stress/radiation effects , Radio Waves/adverse effects , Animals , Antioxidants/pharmacology , Cells, Cultured , DNA Damage/drug effects , DNA, Mitochondrial/drug effects , Electromagnetic Fields/adverse effects , Gene Dosage/drug effects , Gene Dosage/radiation effects , Guanine/analogs & derivatives , Guanine/metabolism , Melatonin/pharmacology , Mitochondria/drug effects , Mitochondria/physiology , Neocortex/drug effects , Neocortex/physiology , Neocortex/radiation effects , Neurons/drug effects , Neurons/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/radiation effects , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects
17.
Radiats Biol Radioecol ; 48(1): 76-81, 2008.
Article in Russian | MEDLINE | ID: mdl-18666582

ABSTRACT

Lipid content of tissue and of fraction of microsomes in neocortex of Wistar rats was studies under artificial hypothermia, after X-ray irradiation in dose 8 Gy under conditions of normothermia and artificial hypothermia in 48 h. The condition of artificial hypothermia get by cooling of rats to 15-18 degrees C. It was shown, that in fraction of microsomes of hypothermia rats the content of phosphatidylinositol was decreased, and in 48 h after cooling of rats the amount of protein, total and individual phospholipids was increased. The lipid content in tissue and in fraction of microsomes of rats, which were irradiated in normotermia, had no changes after 48 h. In fraction of microsomes of rats, which were irradiated after hypothermia, the amount of protein, total phospholipids, sphingomyelin, phosphatidylcholine and phosphatidylserine is increased trustworthy. Thus, we think, that radioprotective effect of hypotermia may be connected with the accumulation of proteins and of phospholipids in the endoplasmic reticulum membranes of neocortex.


Subject(s)
Hypothermia, Induced , Lipid Metabolism/radiation effects , Neocortex/radiation effects , Radiation Tolerance/physiology , Animals , Male , Microsomes/metabolism , Neocortex/metabolism , Phosphatidylinositols/metabolism , Phospholipids/metabolism , Rats , Rats, Wistar , Time Factors , X-Rays
18.
Epilepsy Res ; 73(1): 53-64, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17011168

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by multi-organ pathologies. Most TSC patients exhibit seizures, usually starting in early childhood. The neuropathological hallmarks of the disease - cortical tubers, containing cytopathological neuronal and glial cell types - appear to be the source of seizure initiation. However, the contribution of these aberrant cell populations to TSC-associated epilepsies is not fully understood. To gain further insight, investigators have attempted to generate animal models with TSC-like brain abnormalities. In the current study, we focused on the Eker rat, in which there is a spontaneous mutation of the TSC2 gene (TSC2+/-). We attempted to exacerbate TSC-like brain pathologies with a "second-hit" strategy - exposing young pups to ionizing irradiation of different intensities, and at different developmental timepoints (between E18 and P6). We found that the frequency of occurrence of dysmorphic neurons and giant astrocytes was strongly dependent on irradiation dose, and weakly dependent on timing of irradiation in Eker rats, but not in irradiated normal controls. The frequency of TSC-like pathology was progressive; there were many more abnormal cells at 3 months compared to 1 month post-irradiation. Measures of seizure propensity (flurothyl seizure latency) and brain excitability (paired-pulse and post-tetanic stimulation studies in vitro), however, showed no functional changes associated with the appearance of TSC-like cellular abnormalities in irradiated Eker rats.


Subject(s)
Cerebral Cortex/radiation effects , Epilepsy/pathology , Seizures/pathology , Tuberous Sclerosis/pathology , Tumor Suppressor Proteins/genetics , Animals , Animals, Newborn , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Radiation , Electroencephalography , Epilepsy/etiology , Mutation , Neocortex/pathology , Neocortex/radiation effects , Rats , Rats, Mutant Strains , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/radiation effects , Video Recording , Whole-Body Irradiation
19.
J Neurophysiol ; 97(3): 1887-902, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17151229

ABSTRACT

Approximately 30% of epilepsy patients suffer from drug-resistant epilepsy. Direct electrical stimulation of the epileptogenic zone is a potential new treatment modality for this devastating disease. In this study, we investigated the effect of two electrical stimulation paradigms, sustained low-frequency stimulation and short trains of high-frequency stimulation, on epileptiform discharges in neocortical brain slices treated with either bicuculline or magnesium-free extracellular solution. Sustained low-frequency stimulation (5-30 min of 0.1- to 5-Hz stimulation) prevented both interictal-like discharges and seizure-like events in an intensity-, frequency-, and distance-dependent manner. Short trains of high-frequency stimulation (1-5 s of 25- to 200-Hz stimulation) prematurely terminated seizure-like events in a frequency-, intensity-, and duration-dependent manner. Roughly one half the seizures terminated within the 100-Hz stimulation train (P < 0.01 compared with control), whereas the remaining seizures were significantly shortened by 53 +/- 21% (P < 0.01). Regarding the cellular mechanisms underlying the antiepileptic effects of electrical stimulation, both low- and high-frequency stimulation markedly depressed excitatory postsynaptic potentials (EPSPs). The EPSP amplitude decreased by 75 +/- 3% after 10-min, 1-Hz stimulation and by 86 +/- 6% after 1-s, 100-Hz stimulation. Moreover, partial pharmacological blockade of ionotropic glutamate receptors was sufficient to suppress epileptiform discharges and enhance the antiepileptic effects of stimulation. In conclusion, this study showed that both low- and high-frequency electrical stimulation possessed antiepileptic effects in the neocortex in vitro, established the parameters determining the antiepileptic efficacy of both stimulation paradigms, and suggested that the antiepileptic effects of stimulation were mediated mostly by short-term synaptic depression of excitatory neurotransmission.


Subject(s)
Action Potentials/radiation effects , Electric Stimulation/methods , Epilepsy/therapy , Neocortex/radiation effects , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Analysis of Variance , Animals , Animals, Newborn , Bicuculline/pharmacology , Dose-Response Relationship, Radiation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/radiation effects , GABA Antagonists/pharmacology , In Vitro Techniques , Neocortex/drug effects , Rats , Rats, Wistar , Time Factors
20.
Brain Res Bull ; 68(5): 361-73, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16377444

ABSTRACT

The autoinhibitory control of electrically evoked release of [3H]-dopamine and the properties of that induced by nicotinic receptor (nAChR) stimulation were studied in slices of the human neocortex. In both models [3H]-dopamine release was action potential-induced and exocytotic. The selective dopamine D2 receptor agonist (-)-quinpirole reduced electrically evoked release of [3H]-dopamine, yielding IC50 and I(max) values of 23 nM and 76%, respectively. Also, the effects of several other subtype-selective dopamine receptor ligands confirmed that the terminal dopamine autoreceptor belongs to the D2 subtype. The autoinhibitory feedback control was slightly operative under stimulation conditions of 90 pulses and 3 Hz, with a biophase concentration of endogenous dopamine of 3.6 nM, and was enhanced under blockade of dopamine reuptake. [3H]-dopamine release evoked in an identical manner in mouse neocortical slices was not inhibited by (-)-quinpirole, suggesting the absence of dopamine autoreceptors in this tissue and underlining an important species difference. Also, nAChR stimulation-induced release of [3H]-dopamine revealed a species difference: [3H]-dopamine release was evoked in human, but not in rat neocortical slices. The nAChRs inducing [3H]-dopamine release most probably belong to the alpha3/beta2subtype, according to the potencies and efficacies of subtype-selective nAChR ligands. Part of these receptors may be located on glutamatergic neurons.


Subject(s)
Autoreceptors/physiology , Dopamine/metabolism , Neocortex/metabolism , Receptors, Nicotinic/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Adolescent , Adult , Aged , Alkaloids/pharmacology , Analysis of Variance , Animals , Azocines/pharmacology , Calcium/pharmacology , Child , Child, Preschool , Domperidone/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Feedback/drug effects , Female , Fluvoxamine/pharmacology , Humans , In Vitro Techniques , Isoxazoles/pharmacology , Male , Maprotiline/analogs & derivatives , Maprotiline/pharmacology , Mice , Middle Aged , Neocortex/drug effects , Neocortex/radiation effects , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Potassium/pharmacology , Pyrrolidines/pharmacology , Quinolizines/pharmacology , Rats , Sulpiride/pharmacology , Time Factors , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL