Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 335
Filter
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649213

ABSTRACT

Various neuromodulation approaches have been employed to alter neuronal spiking activity and thus regulate brain functions and alleviate neurological disorders. Infrared neural stimulation (INS) could be a potential approach for neuromodulation because it requires no tissue contact and possesses a high spatial resolution. However, the risk of overheating and an unclear mechanism hamper its application. Here we show that midinfrared stimulation (MIRS) with a specific wavelength exerts nonthermal, long-distance, and reversible modulatory effects on ion channel activity, neuronal signaling, and sensorimotor behavior. Patch-clamp recording from mouse neocortical pyramidal cells revealed that MIRS readily provides gain control over spiking activities, inhibiting spiking responses to weak inputs but enhancing those to strong inputs. MIRS also shortens action potential (AP) waveforms by accelerating its repolarization, through an increase in voltage-gated K+ (but not Na+) currents. Molecular dynamics simulations further revealed that MIRS-induced resonance vibration of -C=O bonds at the K+ channel ion selectivity filter contributes to the K+ current increase. Importantly, these effects are readily reversible and independent of temperature increase. At the behavioral level in larval zebrafish, MIRS modulates startle responses by sharply increasing the slope of the sensorimotor input-output curve. Therefore, MIRS represents a promising neuromodulation approach suitable for clinical application.


Subject(s)
Behavior, Animal/radiation effects , Infrared Rays , Neurons/metabolism , Signal Transduction/radiation effects , Synaptic Transmission/radiation effects , Zebrafish/metabolism , Action Potentials/radiation effects , Animals , Mice
2.
Neurobiol Dis ; 151: 105252, 2021 04.
Article in English | MEDLINE | ID: mdl-33418069

ABSTRACT

Galactic cosmic radiation (GCR), composed of highly energetic and fully ionized atomic nuclei, produces diverse deleterious effects on the body. In researching the neurological risks of GCR exposures, including during human spaceflight, various ground-based single-ion GCR irradiation paradigms induce differential disruptions of cellular activity and overall behavior. However, it remains less clear how irradiation comprising a mix of multiple ions, more accurately recapitulating the space GCR environment, impacts the central nervous system. We therefore examined how mixed-ion GCR irradiation (two similar 5-6 beam combinations of protons, helium, oxygen, silicon and iron ions) influenced neuronal connectivity, functional generation of activity within neural circuits and cognitive behavior in mice. In electrophysiological recordings we find that space-relevant doses of mixed-ion GCR preferentially alter hippocampal inhibitory neurotransmission and produce related disruptions in the local field potentials of hippocampal oscillations. Such underlying perturbation in hippocampal network activity correspond with perturbed learning, memory and anxiety behavior.


Subject(s)
Cosmic Radiation/adverse effects , Hippocampus/radiation effects , Synaptic Transmission/radiation effects , Animals , Behavior, Animal/radiation effects , Cognitive Dysfunction/etiology , Male , Mice , Mice, Inbred C57BL
3.
Neuron ; 109(4): 663-676.e5, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33333001

ABSTRACT

Neocortical pyramidal neurons regulate firing around a stable mean firing rate (FR) that can differ by orders of magnitude between neurons, but the factors that determine where individual neurons sit within this broad FR distribution are not understood. To access low- and high-FR neurons for ex vivo analysis, we used Ca2+- and UV-dependent photoconversion of CaMPARI2 in vivo to permanently label neurons according to mean FR. CaMPARI2 photoconversion was correlated with immediate early gene expression and higher FRs ex vivo and tracked the drop and rebound in ensemble mean FR induced by prolonged monocular deprivation. High-activity L4 pyramidal neurons had greater intrinsic excitability and recurrent excitatory synaptic strength, while E/I ratio, local output strength, and local connection probability were not different. Thus, in L4 pyramidal neurons (considered a single transcriptional cell type), a broad mean FR distribution is achieved through graded differences in both intrinsic and synaptic properties.


Subject(s)
Calcium/metabolism , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Neurons/metabolism , Pyramidal Cells/metabolism , Synaptic Transmission/physiology , Animals , Calcium/analysis , Excitatory Postsynaptic Potentials/radiation effects , Female , Inhibitory Postsynaptic Potentials/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/chemistry , Neurons/radiation effects , Pyramidal Cells/chemistry , Pyramidal Cells/radiation effects , Synaptic Transmission/radiation effects , Ultraviolet Rays
4.
Mol Cell Neurosci ; 109: 103563, 2020 12.
Article in English | MEDLINE | ID: mdl-33039519

ABSTRACT

Recent work demonstrated that sympathetic neurons innervate the skeletal muscle near the neuromuscular junction (NMJ), and muscle sympathectomy and sympathomimetic agents strongly influence motoneuron synaptic vesicle release ex vivo. Moreover, reports attest that the pontine nucleus locus coeruleus (LC) projects to preganglionic sympathetic neurons and regulates human mobility and skeletal muscle physiology. Thus, we hypothesized that peripheral and central sympathetic neurons projecting directly or indirectly to the skeletal muscle regulate NMJ transmission. The aim of this study was to define the specific neuronal groups in the peripheral and central nervous systems that account for such regulation in adult mice in vivo by using optogenetics and NMJ transmission recordings in 3-5-month-old, male and female ChR2(H134R/EYFP)/TH-Cre mice. After detecting ChR2(H134R)/EYFP fluorescence in the paravertebral ganglia and LC neurons, we tested whether optostimulating the plantar nerve near the lumbricalis muscle or LC neurons effectively modulates motor nerve terminal synaptic vesicle release in living mice. Nerve optostimulation increased motor synaptic vesicle release in vitro and in vivo, while the presynaptic adrenoceptor blockers propranolol (ß1/ß2) and atenolol (ß1) prevented this outcome. The effect is primarily presynaptic since miniature end-plate potential (MEPP) kinetics remained statistically unmodified after stimulation. In contrast, optostimulation of LC neurons did not regulate NMJ transmission. In summary, we conclude that postganglionic sympathetic neurons, but not LC neurons, increased NMJ transmission by acting on presynaptic ß1-adrenergic receptors in vivo.


Subject(s)
Locus Coeruleus/physiology , Motor Neurons/physiology , Neuromuscular Junction/physiology , Optogenetics/methods , Synaptic Transmission/physiology , Tibial Nerve/physiology , Animals , Channelrhodopsins/analysis , Channelrhodopsins/genetics , Dependovirus/physiology , Female , Ganglia, Sympathetic/physiology , Genes, Reporter , Green Fluorescent Proteins/analysis , Lasers , Light , Male , Mice , Mice, Transgenic , Miniature Postsynaptic Potentials/physiology , Motor Neurons/radiation effects , Mutation, Missense , Receptors, Adrenergic, beta-1/physiology , Recombinant Fusion Proteins/analysis , Sympathetic Fibers, Postganglionic/physiology , Synaptic Transmission/radiation effects , Tibial Nerve/radiation effects
5.
J Chem Neuroanat ; 106: 101784, 2020 07.
Article in English | MEDLINE | ID: mdl-32205214

ABSTRACT

Behavioral impairments are the most pragmatic outcome of long-term mobile uses but the underlying causes are still poorly understood. Therefore, the Aim of the present study to determine the possible mechanism of mobile induced behavioral alterations by observing redox status, cholinesterase activity, cellular, genotoxic damage and cognitive alterations in rat hippocampus. This study was carried out on 24 male Wistar rats, randomly divided into four groups (n = 6 in each group): group I consisted of sham-exposed (control) rats, group II-IV consisted of rats exposed to microwave radiation (900 MHz) at different time duration 1 h, 2 h, and 4 h respectively for 90 days. After 90 days of exposure, rats were assessing learning ability by using T-Maze. A significantly increased level of malondialdehyde (MDA) with concomitantly depleted levels of superoxide dismutase (SOD), catalase (CAT) and redox enzymes (GSH, GPX, GR, GST, G-6PDH) indicated an exposure of mobile emitted EMR induced oxidative stress by the depleted redox status of brain cells. The depletion in the acetylcholinesterase (AChE) level reveals altered neurotransmission in brain cells. Resultant cellular degeneration was also observed in the radiation-exposed hippocampus. Conclusively, the present study revealed that microwave radiation induces oxidative stress, depleted redox status, and causes DNA damage with the subsequent reduction in working memory in a time-dependent manner. This study provides insight over the associative reciprocity between redox status, cellular degeneration and reduced cholinergic activity, which presumably leads to the behavioral alterations following mobile emitted electromagnetic radiation.


Subject(s)
Acetylcholinesterase/metabolism , Brain/radiation effects , Electromagnetic Radiation , Memory, Short-Term/radiation effects , Oxidation-Reduction , Oxidative Stress/radiation effects , Animals , Brain/metabolism , Catalase/metabolism , DNA Damage/radiation effects , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects
6.
Front Immunol ; 11: 614509, 2020.
Article in English | MEDLINE | ID: mdl-33391287

ABSTRACT

Systemic inflammation is associated with alterations in complex brain functions such as learning and memory. However, diagnostic approaches to functionally assess and quantify inflammation-associated alterations in synaptic plasticity are not well-established. In previous work, we demonstrated that bacterial lipopolysaccharide (LPS)-induced systemic inflammation alters the ability of hippocampal neurons to express synaptic plasticity, i.e., the long-term potentiation (LTP) of excitatory neurotransmission. Here, we tested whether synaptic plasticity induced by repetitive magnetic stimulation (rMS), a non-invasive brain stimulation technique used in clinical practice, is affected by LPS-induced inflammation. Specifically, we explored brain tissue cultures to learn more about the direct effects of LPS on neural tissue, and we tested for the plasticity-restoring effects of the anti-inflammatory cytokine interleukin 10 (IL10). As shown previously, 10 Hz repetitive magnetic stimulation (rMS) of organotypic entorhino-hippocampal tissue cultures induced a robust increase in excitatory neurotransmission onto CA1 pyramidal neurons. Furthermore, LPS-treated tissue cultures did not express rMS-induced synaptic plasticity. Live-cell microscopy in tissue cultures prepared from a novel transgenic reporter mouse line [C57BL/6-Tg(TNFa-eGFP)] confirms that ex vivo LPS administration triggers microglial tumor necrosis factor alpha (TNFα) expression, which is ameliorated in the presence of IL10. Consistent with this observation, IL10 hampers the LPS-induced increase in TNFα, IL6, IL1ß, and IFNγ and restores the ability of neurons to express rMS-induced synaptic plasticity in the presence of LPS. These findings establish organotypic tissue cultures as a suitable model for studying inflammation-induced alterations in synaptic plasticity, thus providing a biological basis for the diagnostic use of transcranial magnetic stimulation in the context of brain inflammation.


Subject(s)
Hippocampus/physiology , Interleukin-10/pharmacology , Neuronal Plasticity/physiology , Neurons/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Genes, Reporter , Hippocampus/metabolism , Hippocampus/radiation effects , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/radiation effects , Neurons/metabolism , Organoids , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , Transcranial Magnetic Stimulation
7.
PLoS One ; 14(11): e0224846, 2019.
Article in English | MEDLINE | ID: mdl-31710637

ABSTRACT

Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60-70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100-170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.


Subject(s)
Human Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Optogenetics , Animals , Astrocytes/cytology , Astrocytes/radiation effects , Axons/metabolism , Axons/radiation effects , Cell Differentiation/radiation effects , Cell Lineage/radiation effects , Cell Survival/radiation effects , Channelrhodopsins/metabolism , Human Embryonic Stem Cells/radiation effects , Humans , Lentivirus/metabolism , Light , Mice, Nude , Motor Cortex/metabolism , Neural Stem Cells/radiation effects , Neuronal Plasticity/radiation effects , Neurons/radiation effects , Neurotransmitter Agents/metabolism , Phenotype , Photic Stimulation , Rats, Nude , Synaptic Transmission/radiation effects
8.
PLoS One ; 14(8): e0218818, 2019.
Article in English | MEDLINE | ID: mdl-31461464

ABSTRACT

Although it is well established that the vertebrate retina contains endogenous circadian clocks that regulate retinal physiology and function during day and night, the processes that the clocks affect and the means by which the clocks control these processes remain unresolved. We previously demonstrated that a circadian clock in the goldfish retina regulates rod-cone electrical coupling so that coupling is weak during the day and robust at night. The increase in rod-cone coupling at night introduces rod signals into cones so that the light responses of both cones and cone horizontal cells, which are post-synaptic to cones, become dominated by rod input. By comparing the light responses of cones, cone horizontal cells and rod horizontal cells, which are post-synaptic to rods, under dark-adapted conditions during day and night, we determined whether the daily changes in the strength of rod-cone coupling could account entirely for rhythmic changes in the light response properties of cones and cone horizontal cells. We report that although some aspects of the day/night changes in cone and cone horizontal cell light responses, such as response threshold and spectral tuning, are consistent with modulation of rod-cone coupling, other properties cannot be solely explained by this phenomenon. Specifically, we found that at night compared to the day the time course of spectrally-isolated cone photoresponses was slower, cone-to-cone horizontal cell synaptic transfer was highly non-linear and of lower gain, and the delay in cone-to-cone horizontal cell synaptic transmission was longer. However, under bright light-adapted conditions in both day and night, cone-to-cone horizontal cell synaptic transfer was linear and of high gain, and no additional delay was observed at the cone-to-cone horizontal cell synapse. These findings suggest that in addition to controlling rod-cone coupling, retinal clocks shape the light responses of cone horizontal cells by modulating cone-to-cone horizontal cell synaptic transmission.


Subject(s)
Circadian Clocks/physiology , Goldfish , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synaptic Transmission/physiology , Animals , Circadian Clocks/radiation effects , Light , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Horizontal Cells/radiation effects , Synaptic Transmission/radiation effects
9.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31383727

ABSTRACT

As NASA prepares for a mission to Mars, concerns regarding the health risks associated with deep space radiation exposure have emerged. Until now, the impacts of such exposures have only been studied in animals after acute exposures, using dose rates ∼1.5×105 higher than those actually encountered in space. Using a new, low dose-rate neutron irradiation facility, we have uncovered that realistic, low dose-rate exposures produce serious neurocognitive complications associated with impaired neurotransmission. Chronic (6 month) low-dose (18 cGy) and dose rate (1 mGy/d) exposures of mice to a mixed field of neutrons and photons result in diminished hippocampal neuronal excitability and disrupted hippocampal and cortical long-term potentiation. Furthermore, mice displayed severe impairments in learning and memory, and the emergence of distress behaviors. Behavioral analyses showed an alarming increase in risk associated with these realistic simulations, revealing for the first time, some unexpected potential problems associated with deep space travel on all levels of neurological function.


Subject(s)
Cognition/radiation effects , Cosmic Radiation/adverse effects , Hippocampus/radiation effects , Neutrons/adverse effects , Photons/adverse effects , Synaptic Transmission/radiation effects , Animals , Anxiety/etiology , Depression/etiology , Extinction, Psychological/radiation effects , Male , Memory/radiation effects , Mice, Inbred C57BL , Neurons/radiation effects , Social Behavior
10.
Curr Opin Struct Biol ; 57: 23-30, 2019 08.
Article in English | MEDLINE | ID: mdl-30825844

ABSTRACT

Chemical and electrical signaling at the synapse is a dynamic process that is crucial to neurotransmission and pathology. Traditional pharmacotherapy has found countless applications in both academic labs and the clinic; however, diffusible drugs lack spatial and temporal precision when employed in heterogeneous tissues such as the brain. In the field of photopharmacology, chemical attachment of a synthetic photoswitch to a bioactive ligand allows cellular signaling to be controlled with light. Azobenzenes have remained the go-to photoswitch for biological applications due to their tunable photophysical properties, and can be leveraged to achieve reversible optical control of numerous receptors and ion channels. Here, we discuss the most recent advances in photopharmacology which will improve the use of azobenzene-based probes for neuroscience applications.


Subject(s)
Azo Compounds , Drug Design , Molecular Probes , Synaptic Transmission , Azo Compounds/metabolism , Light , Molecular Probes/metabolism , Signal Transduction/radiation effects , Synaptic Transmission/radiation effects
11.
Cell Rep ; 20(9): 2026-2043, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854356

ABSTRACT

DNA damage causally contributes to aging and age-related diseases. Mutations in nucleotide excision repair (NER) genes cause highly complex congenital syndromes characterized by growth retardation, cancer susceptibility, and accelerated aging in humans. Orthologous mutations in Caenorhabditis elegans lead to growth delay, genome instability, and accelerated functional decline, thus allowing investigation of the consequences of persistent DNA damage during development and aging in a simple metazoan model. Here, we conducted proteome, lipidome, and phosphoproteome analysis of NER-deficient animals in response to UV treatment to gain comprehensive insights into the full range of physiological adaptations to unrepaired DNA damage. We derive metabolic changes indicative of a tissue maintenance program and implicate an autophagy-mediated proteostatic response. We assign central roles for the insulin-, EGF-, and AMPK-like signaling pathways in orchestrating the adaptive response to DNA damage. Our results provide insights into the DNA damage responses in the organismal context.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Cellular Reprogramming , DNA Damage , Active Transport, Cell Nucleus/radiation effects , Aging/metabolism , Animals , Caenorhabditis elegans/radiation effects , Caenorhabditis elegans Proteins/metabolism , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cellular Reprogramming/radiation effects , DNA Repair/radiation effects , Down-Regulation/radiation effects , Ion Transport/radiation effects , Lipid Metabolism/radiation effects , Mutation/genetics , Phosphoproteins/metabolism , Proteolysis/radiation effects , Proteome/metabolism , Proteomics , Starvation/metabolism , Synaptic Transmission/radiation effects , Transcriptome/genetics , Transcriptome/radiation effects , Ultraviolet Rays , Up-Regulation/radiation effects
12.
Sci Rep ; 7(1): 3275, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28607402

ABSTRACT

Novel clinical treatments to target peripheral nerves are being developed which primarily use electrical current. Recently, infrared (IR) light was shown to inhibit peripheral nerves with high spatial and temporal specificity. Here, for the first time, we demonstrate that IR can selectively and reversibly inhibit small-diameter axons at lower radiant exposures than large-diameter axons. We provide a mathematical rationale, and then demonstrate it experimentally in individual axons of identified neurons in the marine mollusk Aplysia californica, and in axons within the vagus nerve of a mammal, the musk shrew Suncus murinus. The ability to selectively, rapidly, and reversibly control small-diameter sensory fibers may have many applications, both for the analysis of physiology, and for treating diseases of the peripheral nervous system, such as chronic nausea, vomiting, pain, and hypertension. Moreover, the mathematical analysis of how IR affects the nerve could apply to other techniques for controlling peripheral nerve signaling.


Subject(s)
Axons/physiology , Axons/radiation effects , Infrared Rays , Animals , Aplysia , Electrophysiological Phenomena/radiation effects , Infrared Rays/adverse effects , Male , Neurons/physiology , Neurons/radiation effects , Synaptic Transmission/radiation effects , Vagus Nerve
13.
PLoS Comput Biol ; 12(11): e1005206, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27893786

ABSTRACT

Transcranial brain stimulation and evidence of ephaptic coupling have recently sparked strong interests in understanding the effects of weak electric fields on the dynamics of brain networks and of coupled populations of neurons. The collective dynamics of large neuronal populations can be efficiently studied using single-compartment (point) model neurons of the integrate-and-fire (IF) type as their elements. These models, however, lack the dendritic morphology required to biophysically describe the effect of an extracellular electric field on the neuronal membrane voltage. Here, we extend the IF point neuron models to accurately reflect morphology dependent electric field effects extracted from a canonical spatial "ball-and-stick" (BS) neuron model. Even in the absence of an extracellular field, neuronal morphology by itself strongly affects the cellular response properties. We, therefore, derive additional components for leaky and nonlinear IF neuron models to reproduce the subthreshold voltage and spiking dynamics of the BS model exposed to both fluctuating somatic and dendritic inputs and an extracellular electric field. We show that an oscillatory electric field causes spike rate resonance, or equivalently, pronounced spike to field coherence. Its resonance frequency depends on the location of the synaptic background inputs. For somatic inputs the resonance appears in the beta and gamma frequency range, whereas for distal dendritic inputs it is shifted to even higher frequencies. Irrespective of an external electric field, the presence of a dendritic cable attenuates the subthreshold response at the soma to slowly-varying somatic inputs while implementing a low-pass filter for distal dendritic inputs. Our point neuron model extension is straightforward to implement and is computationally much more efficient compared to the original BS model. It is well suited for studying the dynamics of large populations of neurons with heterogeneous dendritic morphology with (and without) the influence of weak external electric fields.


Subject(s)
Action Potentials/physiology , Electromagnetic Fields , Membrane Potentials/physiology , Models, Neurological , Neurons/physiology , Synaptic Transmission/physiology , Action Potentials/radiation effects , Animals , Computer Simulation , Dendrites/drug effects , Dendrites/physiology , Humans , Membrane Potentials/radiation effects , Neurons/radiation effects , Radiation Dosage , Synaptic Transmission/radiation effects
14.
Lab Chip ; 16(9): 1684-90, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27053163

ABSTRACT

In this study, we demonstrate a novel platform for optical stimulation of neural circuits combined with a microfluidic culture method and microelectrode array measurements. Neuron-on-a-chip was designed and fabricated to isolate axons without a soma or dendrite. Thus, it is readily able to manipulate the neuronal alignment and to investigate the neuronal activity at the locations we want to observe. We adapted the optical stimulation technique to the arranged neurons to generate the neuronal signals in a non-invasive fashion. A blue light-emitting diode and a femtosecond laser with 780 nm center wavelength were used for neuronal activation and the corresponding neuronal signals were measured by MEAs at the same time. We found that one-photon light via caged glutamate provoked periodic spiking. In contrast, the femtosecond pulse irradiation generated repetitive firing at constant rates. Response times of one-photon and two-photon stimulation were around 200 ms and 50 ms, respectively. We also quantified neural responses, by varying optical parameters such as exposure time and irradiation power.


Subject(s)
Cell Culture Techniques/instrumentation , Hippocampus/radiation effects , Lab-On-A-Chip Devices , Neurons/radiation effects , Photic Stimulation , Single-Cell Analysis/instrumentation , Synaptic Transmission/radiation effects , Animals , Cell Separation/instrumentation , Cells, Cultured , Dimethylpolysiloxanes/chemistry , Dose-Response Relationship, Radiation , Embryo, Mammalian/cytology , Equipment Design , Hippocampus/cytology , Hippocampus/physiology , Image Processing, Computer-Assisted , Microarray Analysis/instrumentation , Microscopy, Phase-Contrast , Neurons/cytology , Neurons/physiology , Photons , Rats, Sprague-Dawley , Reproducibility of Results
15.
J Proteomics ; 140: 24-36, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27020882

ABSTRACT

UNLABELLED: Recent advances in the field of biodosimetry have shown that the response of biological systems to ionizing radiation is complex and depends on the type and dose of radiation, the tissue(s) exposed, and the time lapsed after exposure. The biological effects of low dose radiation on learning and memory are not well understood. An ion mobility-enhanced data-independent acquisition (MS(E)) approach in conjunction with the ISOQuant software tool was utilized for label-free quantification of hippocampal proteins with the goal of determining protein alteration associated with low-dose whole body ionizing radiation (X-rays, 1Gy) of 5.5-month-old male C57BL/6J mice post contextual fear conditioning training. Global proteome analysis revealed deregulation of 73 proteins (out of 399 proteins). Deregulated proteins indicated adverse effects of irradiation on myelination and perturbation of energy metabolism pathways involving a shift from the TCA cycle to glutamate oxidation. Our findings also indicate that proteins associated with synaptic activity, including vesicle recycling and neurotransmission, were altered in the irradiated mice. The elevated LTP and decreased LTD suggest improved synaptic transmission and enhanced efficiency of neurotransmitter release which would be consistent with the observed comparable contextual fear memory performance of the mice following post-training whole body or sham-irradiation. SIGNIFICANCE: This study is significant because the biological consequences of low dose radiation on learning and memory are complex and not yet well understood. We conducted a IMS-enhanced MS(E)-based label-free quantitative proteomic analysis of hippocampal tissue with the goal of determining protein alteration associated with low-dose whole body ionizing radiation (X-ray, 1Gy) of 5.5-month-old male C57BL/6J mice post contextual fear conditioning training. The IMS-enhanced MS(E) approach in conjunction with ISOQuant software was robust and accurate with low median CV values of 0.99% for the technical replicates of samples from both the sham and irradiated group. The biological variance was as low as 1.61% for the sham group and 1.31% for the irradiated group. The applied data generation and processing workflow allowed the quantitative evaluation of 399 proteins. The current proteomic analysis indicates that myelination is sensitive to low dose radiation. The observed protein level changes imply modulation of energy metabolism pathways in the radiation exposed group, specifically changes in protein abundance levels suggest a shift from TCA cycle to glutamate oxidation to satisfy energy demands. Most significantly, our study reveals deregulation of proteins involved in processes that govern synaptic activity including enhanced synaptic vesicle cycling, and altered long-term potentiation (LTP) and depression (LTD). An elevated LTP and decreased LTD suggest improved synaptic transmission and enhanced efficiency of neurotransmitter release which is consistent with the observed comparable contextual fear memory performance of the mice following post-training whole body or sham-irradiation. Overall, our results underscore the importance of low dose radiation experiments for illuminating the sensitivity of biochemical pathways to radiation, and the modulation of potential repair and compensatory response mechanisms. This kind of studies and associated findings may ultimately lead to the design of strategies for ameliorating hippocampal and CNS injury following radiation exposure as part of medical therapies or as a consequence of occupational hazards.


Subject(s)
Hippocampus/radiation effects , Proteome/analysis , Radiation, Ionizing , Animals , Behavior, Animal , Fear/radiation effects , Gene Expression Regulation/radiation effects , Hippocampus/chemistry , Long-Term Potentiation/radiation effects , Male , Memory/radiation effects , Mice , Mice, Inbred C57BL , Proteome/radiation effects , Proteomics/methods , Synaptic Transmission/radiation effects
16.
Neuroendocrinology ; 103(6): 678-92, 2016.
Article in English | MEDLINE | ID: mdl-26562443

ABSTRACT

BACKGROUND/AIMS: Photoperiod is a major environmental cue in temperate-zone birds which synchronizes breeding with the time of year that offers the optimal environment for offspring survival. Despite continued long photoperiods, these birds eventually become refractory to the stimulating photoperiod and their reproductive systems regress. In this study, we characterized the role of γ-aminobutyric acid (GABA)ergic neurotransmission in modulating the response of the premammillary nucleus (PMM) to a gonad stimulatory photoperiod and the onset of photorefractoriness. METHODS AND RESULTS: Bilateral ablation of the PMM blocked the light-induced neuroendocrine response from occurring in photosensitive turkeys. Microarray analyses revealed an increase in GABAergic activity in the PMM of photorefractory birds as opposed to photosensitive ones, and this enhanced GABAergic activity appeared to inhibit the photoperiodic signal. Additionally, GABAA and GABAB receptors were expressed by dopamine-melatonin neurons in the PMM, and the administration of the GABA receptor agonist baclofen blocked the photoperiodic reproductive neuroendocrine responses. CONCLUSIONS: Consistent with the present findings, we propose that the long-sought-after mechanism underlying photorefractoriness is linked to the inhibitory actions of GABA. We suggest that (1) GABAergic interference with photoperiodic entrainment in the PMM initiates the photorefractory state and terminates the annual breeding season in temperate-zone birds, and (2) the PMM is a site of photoreception and photorefractoriness that controls the initiation and termination of avian reproductive seasonality.


Subject(s)
Hypothalamus, Posterior/injuries , Light , Photoperiod , Reproduction/physiology , Seasons , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Dopamine/metabolism , Female , GABA Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Glutamate Decarboxylase/metabolism , Hypothalamus, Posterior/cytology , Melatonin/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, GABA/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects , Turkey
17.
J Proteome Res ; 14(11): 4674-86, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26420666

ABSTRACT

Recent epidemiological data indicate that radiation doses as low as those used in computer tomography may result in long-term neurocognitive side effects. The aim of this study was to elucidate long-term molecular alterations related to memory formation in the brain after low and moderate doses of γ radiation. Female C57BL/6J mice were irradiated on postnatal day 10 with total body doses of 0.1, 0.5, or 2.0 Gy; the control group was sham-irradiated. The proteome analysis of hippocampus, cortex, and synaptosomes isolated from these brain regions indicated changes in ephrin-related, RhoGDI, and axonal guidance signaling. Immunoblotting and miRNA-quantification demonstrated an imbalance in the synapse morphology-related Rac1-Cofilin pathway and long-term potentiation-related cAMP response element-binding protein (CREB) signaling. Proteome profiling also showed impaired oxidative phosphorylation, especially in the synaptic mitochondria. This was accompanied by an early (4 weeks) reduction of mitochondrial respiration capacity in the hippocampus. Although the respiratory capacity was restored by 24 weeks, the number of deregulated mitochondrial complex proteins was increased at this time. All observed changes were significant at doses of 0.5 and 2.0 Gy but not at 0.1 Gy. This study strongly suggests that ionizing radiation at the neonatal state triggers persistent proteomic alterations associated with synaptic impairment.


Subject(s)
Cerebral Cortex/radiation effects , Gamma Rays/adverse effects , Hippocampus/radiation effects , Long-Term Potentiation/radiation effects , Proteome/genetics , Synaptic Transmission/radiation effects , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Animals, Newborn , Axons/metabolism , Axons/radiation effects , Axons/ultrastructure , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Ephrins/genetics , Ephrins/metabolism , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Memory/drug effects , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondria/radiation effects , Neuropeptides/genetics , Neuropeptides/metabolism , Oxidative Phosphorylation/radiation effects , Proteome/metabolism , Synaptosomes/metabolism , Synaptosomes/radiation effects , Whole-Body Irradiation , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/genetics , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism
18.
Pathobiology ; 82(5): 181-94, 2015.
Article in English | MEDLINE | ID: mdl-26337368

ABSTRACT

Recent studies have highlighted the important role of the postsynaptic NMDAR-PSD95-CaMKII pathway for synaptic transmission and related neuronal injury. Here, we tested changes in the components of this pathway upon microwave-induced neuronal structure and function impairments. Ultrastructural and functional changes were induced in hippocampal neurons of rats and in PC12 cells exposed to microwave radiation. We detected abnormal protein and mRNA expression, as well as posttranslational modifications in the NMDAR-PSD95-CaMKII pathway and its associated components, such as synapsin I, following microwave radiation exposure of rats and PC12 cells. Thus, microwave radiation may induce neuronal injury via changes in the molecular organization of postsynaptic density and modulation of the biochemical cascade that potentiates synaptic transmission.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/radiation effects , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microwaves/adverse effects , Neurons/radiation effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disks Large Homolog 4 Protein , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/ultrastructure , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Post-Synaptic Density/radiation effects , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/radiation effects , Rats , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction , Synaptic Transmission/radiation effects
19.
PLoS Comput Biol ; 11(7): e1004304, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132103

ABSTRACT

Motion tracking is a challenge the visual system has to solve by reading out the retinal population. It is still unclear how the information from different neurons can be combined together to estimate the position of an object. Here we recorded a large population of ganglion cells in a dense patch of salamander and guinea pig retinas while displaying a bar moving diffusively. We show that the bar's position can be reconstructed from retinal activity with a precision in the hyperacuity regime using a linear decoder acting on 100+ cells. We then took advantage of this unprecedented precision to explore the spatial structure of the retina's population code. The classical view would have suggested that the firing rates of the cells form a moving hill of activity tracking the bar's position. Instead, we found that most ganglion cells in the salamander fired sparsely and idiosyncratically, so that their neural image did not track the bar. Furthermore, ganglion cell activity spanned an area much larger than predicted by their receptive fields, with cells coding for motion far in their surround. As a result, population redundancy was high, and we could find multiple, disjoint subsets of neurons that encoded the trajectory with high precision. This organization allows for diverse collections of ganglion cells to represent high-accuracy motion information in a form easily read out by downstream neural circuits.


Subject(s)
Action Potentials/physiology , Models, Neurological , Motion Perception/physiology , Nerve Net/physiology , Retinal Ganglion Cells/physiology , Vision, Ocular/physiology , Action Potentials/radiation effects , Animals , Computer Simulation , Guinea Pigs , Light , Motion Perception/radiation effects , Nerve Net/radiation effects , Photic Stimulation/methods , Retinal Ganglion Cells/radiation effects , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , Urodela , Vision, Ocular/radiation effects
20.
Radiat Res ; 183(2): 208-18, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25621896

ABSTRACT

High-energy protons constitute at least 85% of the fluence of energetic ions in interplanetary space. Although protons are only sparsely ionizing compared to higher atomic mass ions, they nevertheless significantly contribute to the delivered dose received by astronauts that can potentially affect central nervous system function at high fluence, especially during prolonged deep space missions such as to Mars. Here we report on the long-term effects of 1 Gy proton irradiation on electrophysiological properties of CA1 pyramidal neurons in the mouse hippocampus. The hippocampus is a key structure for the formation of long-term episodic memory, for spatial orientation and for information processing in a number of other cognitive tasks. CA1 pyramidal neurons form the last and critical relay point in the trisynaptic circuit of the hippocampal principal neurons through which information is processed before being transferred to other brain areas. Proper functioning of CA1 pyramidal neurons is crucial for hippocampus-dependent tasks. Using the patch-clamp technique to evaluate chronic effects of 1 Gy proton irradiation on CA1 pyramidal neurons, we found that the intrinsic membrane properties of CA1 pyramidal neurons were chronically altered at 3 months postirradiation, resulting in a hyperpolarization of the resting membrane potential (VRMP) and a decrease in input resistance (Rin). These small but significant alterations in intrinsic properties decreased the excitability of CA1 pyramidal neurons, and had a dramatic impact on network function in a computational model of the CA1 microcircuit. We also found that proton-radiation exposure upregulated the persistent Na(+) current (INaP) and increased the rate of miniature excitatory postsynaptic currents (mEPSCs). Both the INaP and the heightened rate of mEPSCs contribute to neuronal depolarization and excitation, and at least in part, could compensate for the reduced excitability resulting from the radiation effects on the VRMP and the Rin. These results show long-term alterations in the intrinsic properties of CA1 pyramidal cells after realistic, low-dose proton irradiation.


Subject(s)
CA1 Region, Hippocampal/physiology , Membrane Potentials/physiology , Models, Neurological , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Adaptation, Physiological/physiology , Adaptation, Physiological/radiation effects , Animals , CA1 Region, Hippocampal/radiation effects , Computer Simulation , Dose-Response Relationship, Radiation , Male , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Neuronal Plasticity/radiation effects , Protons , Radiation Dosage , Synapses/radiation effects , Synaptic Transmission/radiation effects , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...