Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Nat Methods ; 17(11): 1067, 2020 11.
Article in English | MEDLINE | ID: mdl-33057240
2.
Lab Chip ; 19(11): 2038-2048, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31094398

ABSTRACT

This paper presents the design, optimisation and fabrication of a mechanically robust 3D printed microfluidic device for the high time resolution online analysis of biomarkers in a microdialysate stream at microlitre per minute flow rates. The device consists of a microfluidic channel with secure low volume connections that easily integrates electrochemical biosensors for biomarkers such as glutamate, glucose and lactate. The optimisation process of the microfluidic channel fabrication, including for different types of 3D printer, is explained and the resulting improvement in sensor response time is quantified. The time resolution of the device is characterised by recording short lactate concentration pulses. The device is employed to record simultaneous glutamate, glucose and lactate concentration changes simulating the physiological response to spreading depolarisation events in cerebrospinal fluid dialysate. As a proof-of-concept study, the device is then used in the intensive care unit for online monitoring of a brain injury patient, demonstrating its capabilities for clinical monitoring.


Subject(s)
Brain/metabolism , Lab-On-A-Chip Devices , Microdialysis/instrumentation , Neurochemistry/instrumentation , Printing, Three-Dimensional , Biosensing Techniques , Brain/cytology , Equipment Design , Humans , Online Systems , Signal-To-Noise Ratio
3.
Methods Enzymol ; 603: 237-255, 2018.
Article in English | MEDLINE | ID: mdl-29673529

ABSTRACT

Anesthetic mechanisms that eliminate consciousness and perception of pain are products of the nervous system. Chemical approaches to the study of anesthetic mechanisms have the potential to serve as an ideal interface between basic and clinical neuroscience. There are disproportionately more basic neurochemical studies than clinical studies of anesthetic mechanisms. Even within neuroscience, the study of anesthetic mechanisms is sparse. The Society for Neuroscience hosts one of the world's largest and most vibrant scientific meetings, yet the content themes of that meeting do not include anesthesia. One goal of this chapter is to facilitate neurochemical studies of anesthetic mechanisms by outlining user-friendly descriptions of existing and emerging techniques. The introduction provides a context for chapter goals. The second portion of this chapter focuses on microdialysis methods that enable the humane acquisition of neurochemical samples from intact, behaving animals during anesthetic induction, maintenance, and emergence. No single neurotransmitter and no single brain region regulate the physiological and behavioral traits characteristic of any anesthetic state. This limitation is being addressed via application of new instrumentation and techniques in analytic chemistry. The final third of this chapter highlights selected omics approaches that are now being applied to the neurochemical study of anesthetic mechanisms. We hope that this brief chapter can stimulate basic and clinical metabolomic approaches aiming to elucidate the mechanisms of anesthetic action.


Subject(s)
Anesthesia, General/methods , Anesthetics/pharmacokinetics , Metabolome/physiology , Microdialysis/methods , Neurochemistry/methods , Neurotransmitter Agents/analysis , Animals , Brain/anatomy & histology , Brain/metabolism , Brain Chemistry/physiology , Brain Mapping , Chemistry Techniques, Analytical , Chromatography, High Pressure Liquid , Humans , Infusion Pumps , Limit of Detection , Microdialysis/instrumentation , Nerve Net/anatomy & histology , Nerve Net/physiology , Neurochemistry/instrumentation , Neurotransmitter Agents/metabolism , Rats , Stereotaxic Techniques , Wakefulness/physiology
4.
Chem Soc Rev ; 46(10): 2692-2704, 2017 May 22.
Article in English | MEDLINE | ID: mdl-28418061

ABSTRACT

Real-time in vivo analysis of neurochemical dynamics has great physiological and pathological implications for a full understanding of the brain. Self-powered electrochemical systems (SPESs) built on galvanic cell configurations bear the advantages of easy miniaturization for implantation and no interference to electric activities of neurons over traditional externally-powered electrochemical sensors for self-triggered in vivo analysis. However, this is still a new concept for in vivo neurochemical sensing with few implanted examples reported so far. This tutorial review summarizes the development of SPESs toward implantable applications from both principal and practical perspectives, ultimately aimed at providing a guide map to the future design of neurochemical sensors for in vivo analysis of brain chemistry.


Subject(s)
Brain Chemistry , Chemistry Techniques, Analytical/instrumentation , Electrochemical Techniques , Neurochemistry/instrumentation , Chemistry Techniques, Analytical/methods , Electrodes , Humans , Neurochemistry/methods
5.
Biosens Bioelectron ; 77: 62-8, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26386904

ABSTRACT

We report a novel single neural probe for real-time simultaneous monitoring of multiple neurochemicals and direct-current electrocorticography (DC-ECoG). A major advance of this probe is the inclusion of two iridium oxide reference electrodes to improve sensor accuracy. The ECoG reference electrode is identical to the ECoG recording electrodes to significantly improve DC stability, while the reference for electrochemical sensors has 10-fold lower polarization rate to minimize the small current-induced drift in the reference electrode potential. In vitro, the single probe selectively measured oxygen (r(2)=0.985 ± 0.01, concentration range=0-60 mmHg, limit of detection=0.4 ± 0.07 mmHg) and glucose (r(2)=0.989 ± 0.009, concentration range=0-4mM, limit of detection=31 ± 8 µM) in a linear fashion. The performance of the single probe was assessed in an in vivo needle prick model to mimic sequelae of traumatic brain injury. It successfully monitored the theoretically expected transient brain oxygen, glucose, and DC potential changes during the passage of spreading depolarization (SD) waves. We envision that the developed probe can be used to decipher the cause-effect relationships between multiple variables of brain pathophysiology with the high temporal and spatial resolutions that it provides.


Subject(s)
Action Potentials/physiology , Electrocorticography/instrumentation , Electrodes, Implanted , Microelectrodes , Nerve Tissue Proteins/metabolism , Parietal Lobe/physiology , Animals , Computer Systems , Conductometry/instrumentation , Equipment Design , Equipment Failure Analysis , Male , Neurochemistry/instrumentation , Parietal Lobe/cytology , Rats , Rats, Sprague-Dawley
6.
J Vis Exp ; (103)2015 09 05.
Article in English | MEDLINE | ID: mdl-26381747

ABSTRACT

Electrical activity in the brain is accompanied by significant ion fluxes across membranes, resulting in complex changes in the extracellular concentration of all major ions. As these ion shifts bear significant functional consequences, their quantitative determination is often required to understand the function and dysfunction of neural networks under physiological and pathophysiological conditions. In the present study, we demonstrate the fabrication and calibration of double-barreled ion-selective microelectrodes, which have proven to be excellent tools for such measurements in brain tissue. Moreover, so-called "concentric" ion-selective microelectrodes are also described, which, based on their different design, offer a far better temporal resolution of fast ion changes. We then show how these electrodes can be employed in acute brain slice preparations of the mouse hippocampus. Using double-barreled, potassium-selective microelectrodes, changes in the extracellular potassium concentration ([K+]o) in response to exogenous application of glutamate receptor agonists or during epileptiform activity are demonstrated. Furthermore, we illustrate the response characteristics of sodium-sensitive, double-barreled and concentric electrodes and compare their detection of changes in the extracellular sodium concentration ([Na+]o) evoked by bath or pressure application of drugs. These measurements show that while response amplitudes are similar, the concentric sodium microelectrodes display a superior signal-to-noise ratio and response time as compared to the double-barreled design. Generally, the demonstrated procedures will be easily transferable to measurement of other ions species, including pH or calcium, and will also be applicable to other preparations.


Subject(s)
Brain/metabolism , Electrochemical Techniques/instrumentation , Microelectrodes , Neurochemistry/instrumentation , Potassium/analysis , Sodium/analysis , Animals , Brain Chemistry , CA1 Region, Hippocampal/chemistry , CA1 Region, Hippocampal/metabolism , Calcium/analysis , Calcium/metabolism , Electrochemical Techniques/methods , Extracellular Fluid/metabolism , Mice , Neurochemistry/methods , Potassium/metabolism , Sodium/metabolism
7.
Mol Neurobiol ; 48(2): 380-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23975638

ABSTRACT

The ability to rapidly detect neurotransmitter release has broad implications in the study of a variety of neurodegenerative diseases. Electrochemical detection methods using carbon nanofiber nanoelectrodes integrated into the Wireless Instantaneous Neurotransmitter Concentration Sensing System (WINCS) offer many important advantages including biocompatibility, selectivity, sensitivity, and rapid adsorption kinetics. Carbon nanofiber nanoelectrodes exhibit greater selectivity and sensitivity in the electrochemical detection of neurotransmitters compared to macroelectrodes and are able to resolve a ternary mixture of dopamine (DA), serotonin (5-HT), and ascorbic acid as well as to detect individual neurotransmitters in concentrations as low as 50 nM for DA and 100 nM for 5-HT using differential pulse voltammetry. Adsorption kinetics studies and isopropyl alcohol treatments modeled on previous studies on carbon fiber microelectrodes were conducted to investigate the analogous properties on carbon nanofiber electrodes using fast-scan cyclic voltammetry with WINCS and showed analogous results in carbon nanofiber electrodes compared with carbon fiber microelectrodes.


Subject(s)
Carbon/chemistry , Nanofibers/chemistry , Neurochemistry/instrumentation , Neurochemistry/methods , Neurotransmitter Agents/analysis , 2-Propanol/chemistry , Adsorption , Carbon Fiber , Dopamine/analysis , Electrodes , Kinetics , Nanofibers/ultrastructure
8.
Curr Opin Chem Biol ; 17(5): 860-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856056

ABSTRACT

Mapping chemical dynamics in the brain of live subjects is a challenging but highly rewarding goal because it allows neurotransmitter fluctuations to be related to behavior, drug effects, and disease states. A popular method for such measurements is microdialysis sampling coupled to analytical measurements. This method has become well-established for monitoring low molecular weight neurotransmitters, metabolites, and drugs, especially in pharmacological and pharmacokinetic studies. Recent technological developments which improve the temporal and spatial resolution of the methods will enable it to be used for studying behavior and small brain nuclei. Better assays allow monitoring more neurotransmitters simultaneously. Extension to analysis of aggregating proteins like amyloid ß is proving extremely useful for uncovering the roles of these molecules and how they contribute to neurodegenerative diseases.


Subject(s)
Microdialysis/methods , Neurochemistry/methods , Amyloid beta-Peptides/analysis , Animals , Brain Chemistry , Humans , Microdialysis/instrumentation , Neurochemistry/instrumentation , Neurotransmitter Agents/analysis
9.
J Neurosci Methods ; 196(2): 303-7, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21291912

ABSTRACT

The dopamine (DA) transporter (DAT) is a key regulator of dopaminergic signaling as it mediates the reuptake of extrasynaptic DA and thereby terminates dopaminergic signaling. Emerging evidence indicates that DAT function is influenced through interactions with other proteins. The current report describes a method to identify such interactions following DAT immunoprecipitation from a rat striatal synaptosomal preparation. This subcellular fraction was selected since DAT function is often determined ex vivo by measuring DA uptake in this preparation and few reports investigating DAT-protein interactions have utilized this preparation. Following SDS-PAGE and colloidal Coomassie staining, selected protein bands from a DAT-immunoprecipitate were excised, digested with trypsin, extracted, and analyzed by liquid chromatography tandem mass spectrometry (LC/MS/MS). From the analysis of the tryptic peptides, several proteins were identified including DAT, Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) ß, CaMKII δ, protein kinase C (PKC) ß, and PKC γ. Co-immunoprecipitation of PKC, CaMKII, and protein interacting with C kinase-1 with DAT was confirmed by Western blotting. Thus, the present study highlights a method to immunoprecipitate DAT and to identify co-immunoprecipitating proteins using LC/MS/MS and Western blotting. This method can be utilized to evaluate DAT protein-protein interactions but also to assess interactions involving other synaptic proteins. Ex vivo identification of protein-protein interactions will provide new insight into the function and regulation of a variety of synaptic, membrane-associated proteins, including DAT.


Subject(s)
Brain Chemistry/physiology , Dopamine Plasma Membrane Transport Proteins/metabolism , Neurochemistry/methods , Presynaptic Terminals/metabolism , Protein Interaction Mapping/methods , Proteomics/methods , Synaptosomes/metabolism , Animals , Male , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurochemistry/instrumentation , Presynaptic Terminals/chemistry , Proteomics/instrumentation , Rats , Rats, Sprague-Dawley , Synaptosomes/chemistry
10.
J Neurosci Methods ; 189(1): 14-22, 2010 May 30.
Article in English | MEDLINE | ID: mdl-20298719

ABSTRACT

Quantification of neurotransmitter transport dynamics is hindered by a lack of sufficient tools to directly monitor bioactive flux under physiological conditions. Traditional techniques for studying neurotransmitter release/uptake require inferences from non-selective electrical recordings, are invasive/destructive, and/or suffer from poor temporal resolution. Recent advances in electrochemical biosensors have enhanced in vitro and in vivo detection of neurotransmitter concentration under physiological/pathophysiological conditions. The use of enzymatic biosensors with performance enhancing materials (e.g., carbon nanotubes) has been a major focus for many of these advances. However, these techniques are not used as mainstream neuroscience research tools, due to relatively low sensitivity, excessive drift/noise, low signal-to-noise ratio, and inability to quantify rapid neurochemical kinetics during synaptic transmission. A sensing technique known as self-referencing overcomes many of these problems, and allows non-invasive quantification of biophysical transport. This work presents a self-referencing CNT modified glutamate oxidase biosensor for monitoring glutamate flux near neural/neuronal cells. Concentration of basal glutamate was similar to other in vivo and in vitro measurements. The biosensor was used in self-referencing (oscillating) mode to measure net glutamate flux near neural cells during electrical stimulation. Prior to stimulation, the average influx was 33.9+/-6.4 fmol cm(-2)s(-1)). Glutamate efflux took place immediately following stimulation, and was always followed by uptake in the 50-150 fmol cm(-2)s(-1) range. Uptake was inhibited using threo-beta-benzyloxyaspartate, and average surface flux in replicate cells (1.1+/-7.4 fmol cm(-2)s(-1)) was significantly lower than uninhibited cells. The technique is extremely valuable for studying neuropathological conditions related to neurotransmission under dynamic physiological conditions.


Subject(s)
Biosensing Techniques/instrumentation , Brain Chemistry/physiology , Electrophysiology/instrumentation , Glutamic Acid/metabolism , Neurochemistry/instrumentation , Neurons/metabolism , Animals , Aspartic Acid/pharmacology , Biological Transport, Active/physiology , Biosensing Techniques/methods , Cells, Cultured , Electric Stimulation , Electrophysiology/methods , Glutamic Acid/analysis , Mice , Neurochemistry/methods , Oxidoreductases/chemistry , Reaction Time/physiology , Synaptic Transmission/physiology , Time Factors
11.
J Neurosci Methods ; 180(2): 278-89, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19464518

ABSTRACT

Deep brain stimulation (DBS) is an effective symptomatic treatment in Parkinson's disease. High frequency stimulation (HFS) of the subthalamic nucleus elicits neurotransmitter release in multiple nuclei. Therefore, we tested the hypothesis that neurotransmitter release during HFS may be used to provide feedback control of the intensity and pattern of HFS. We studied the dynamic relationship between extracellular glutamate levels and HFS in and around the STN in anesthetized rats. We used a pseudorandom binary sequence (PRBS) of stimulation in the STN, the independent forcing function, while measuring extracellular glutamate in the same nucleus, the dependent variable. The PRBS consisted of 90 s periods during which stimulation (100 microA, 150Hz, 10% duty cycle) was either off or on. The stimulation and extracellular glutamate levels were fitted using an autoregressive exogenous model (ARX) to determine the transfer function between HFS and the extracellular glutamate concentration in the STN. The ARX model fit the dynamics of extracellular glutamate levels well (correlation coefficients ranged from 0.74 to 0.99; n=11). The transfer function accurately predicted extracellular glutamate levels in the STN even when the pattern of HFS was modified. We used the transfer function to develop a feedback controlled stimulation algorithm. Feedback controlled HFS maintained extracellular glutamate concentrations at any predefined level, but only intermittent HFS was required. We conclude that the transfer function between HFS and neurotransmitter levels in the brain can be used to design DBS protocols that generate specific temporal patterns of glutamate release in the STN.


Subject(s)
Algorithms , Deep Brain Stimulation/methods , Feedback/physiology , Glutamic Acid/metabolism , Neurochemistry/methods , Subthalamic Nucleus/metabolism , Animals , Biosensing Techniques , Computer Simulation , Deep Brain Stimulation/instrumentation , Extracellular Fluid/metabolism , Neurochemistry/instrumentation , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Signal Processing, Computer-Assisted , Software , Synaptic Transmission/physiology
12.
J Neurosci Methods ; 177(2): 294-302, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19013195

ABSTRACT

Fast synaptic transmission is mediated by post-synaptic ligand-gated ion channels (LGICs) transiently activated by neurotransmitter released from pre-synaptic vesicles. Although disruption of synaptic transmission has been implicated in numerous neurological and psychiatric disorders, effective and practical methods for studying LGICs in vitro under synaptically relevant conditions are unavailable. Here, we describe a novel microfluidic approach to solution switching that allows for precise temporal control over the neurotransmitter transient while substantially increasing experimental throughput, flexibility, reproducibility, and cost-effectiveness. When this system was used to apply ultra-brief ( approximately 400micros) GABA pulses to recombinant GABA(A) receptors, members of the cys-loop family of LGICs, the resulting currents resembled hippocampal inhibitory post-synaptic currents (IPSCs) and differed from currents evoked by longer, conventional pulses, illustrating the importance of evaluating LGICs on a synaptic timescale. This methodology should therefore allow the effects of disease-causing mutations and allosteric modulators to be evaluated in vitro under physiologically relevant conditions.


Subject(s)
Drug Delivery Systems/methods , Electrophysiology/methods , Microfluidic Analytical Techniques/methods , Neurotransmitter Agents/metabolism , Synaptic Transmission/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Cell Line , Drug Delivery Systems/instrumentation , Electronics, Medical/instrumentation , Electronics, Medical/methods , Electrophysiology/instrumentation , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Microfluidic Analytical Techniques/instrumentation , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurochemistry/instrumentation , Neurochemistry/methods , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Synaptic Transmission/drug effects , Time Factors
13.
Prog Brain Res ; 180: 127-39, 2009.
Article in English | MEDLINE | ID: mdl-20302832

ABSTRACT

Although neuromodulation with implanted brain electrodes (deep brain stimulation, DBS) has been increasingly effective in treating many patients with movement disorders (e.g., advanced Parkinson's disease) over the past 20 years, the techniques have changed little for more than 50 years. After summarizing the current state of DBS, this chapter considers (1) the advances being offered by computational analysis techniques as well as (2) the benefits of monitoring and modulating brain chemical activity in addition to brain electrical activity. A review of the current state of sensory neuroprostheses follows, with consideration of emerging data on the optimal configuration of micron-sized retinal prostheses as well as on the optimal site for stimulation of cells in the retina. Very recent findings on nanotechniques to enhance charge transfer from prosthesis to cell (neuronal or glial), that is, enhancement of the neural-electrical interface, are then reviewed. The final section summarizes areas of potential cross-fertilization between those centers developing sensory neuroprostheses and those centers developing nanotechniques for DBS.


Subject(s)
Deep Brain Stimulation/methods , Nanotechnology/methods , Neurochemistry/methods , Prostheses and Implants/standards , Signal Processing, Computer-Assisted , Deep Brain Stimulation/instrumentation , Deep Brain Stimulation/trends , Electrodes, Implanted/trends , Electrophysiology/instrumentation , Electrophysiology/methods , Electrophysiology/trends , Humans , Monitoring, Physiologic/instrumentation , Monitoring, Physiologic/methods , Nanotechnology/instrumentation , Nanotechnology/trends , Neurochemistry/instrumentation , Neurochemistry/trends , Prostheses and Implants/trends , Prosthesis Implantation/methods , Prosthesis Implantation/trends , User-Computer Interface
14.
Nat Rev Neurosci ; 9(10): 779-88, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18784656

ABSTRACT

High-content analysis (HCA) combines automated microscopy and automated image analysis to quantify complex cellular anatomy and biochemistry objectively, accurately and quickly. High-content assays that are applicable to neuroscience include those that can quantify various aspects of dendritic trees, protein aggregation, transcription factor translocation, neurotransmitter receptor internalization, neuron and synapse number, cell migration, proliferation and apoptosis. The data that are generated by HCA are rich and multiplexed. HCA thus provides a powerful high-throughput tool for neuroscientists.


Subject(s)
Image Cytometry/methods , Microscopy/methods , Neurosciences/methods , Automation/instrumentation , Automation/methods , Cell Shape/physiology , Computational Biology/instrumentation , Computational Biology/methods , Computational Biology/trends , Image Cytometry/instrumentation , Image Cytometry/trends , Microscopy/instrumentation , Microscopy/trends , Neurochemistry/instrumentation , Neurochemistry/methods , Neurochemistry/trends , Neurons/cytology , Neurosciences/instrumentation , Neurosciences/trends , Software/trends
15.
J Neurosci Methods ; 174(1): 62-70, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18692090

ABSTRACT

Implantable microfabricated microelectrode arrays represent a versatile and powerful tool to record electrophysiological activity across multiple spatial locations in the brain. Spikes and field potentials, however, correspond to only a fraction of the physiological information available at the neural interface. In urethane-anesthetized rats, microfabricated microelectrode arrays were implanted acutely for simultaneous recording of striatal local field potentials, spikes, and electrically evoked dopamine overflow on the same spatiotemporal scale. During these multi-modal recordings we observed (1) that the amperometric method used to detect dopamine did not significantly influence electrophysiological activity, (2) that electrical stimulation in the medial forebrain bundle (MFB) region resulted in electrochemically transduced dopamine transients in the striatum that were spatially heterogeneous within at least 200 microm, and (3) following MFB stimulation, dopamine levels and electrophysiological activity within the striatum exhibited similar temporal profiles. These neural probes are capable of incorporating customized microelectrode geometries and configurations, which may be useful for examining specific spatiotemporal relationships between electrical and chemical signaling in the brain.


Subject(s)
Action Potentials/physiology , Dopamine/analysis , Dopamine/metabolism , Electrophysiology/instrumentation , Neurochemistry/instrumentation , Neurophysiology/instrumentation , Anesthetics/pharmacology , Animals , Brain/anatomy & histology , Brain/metabolism , Corpus Striatum/metabolism , Electric Stimulation/instrumentation , Electric Stimulation/methods , Electrodes, Implanted/standards , Electrophysiology/methods , Male , Medial Forebrain Bundle/physiology , Microelectrodes/standards , Neurochemistry/methods , Neurons/physiology , Neurophysiology/methods , Rats , Rats, Sprague-Dawley , Urethane/pharmacology
16.
Neurol Res ; 30(7): 661-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18631429

ABSTRACT

BACKGROUND: Microdialysis is a technique to monitor extracellular changes in living tissue. Substances present in the extracellular space, such as neurotransmitters and metabolites transported between cells and capillaries in the extracellular fluid (ECF), are major object. RESULTS: Since its introduction to the research of the nervous system, microdialysis has become a popular method for the measurements of brain chemistry and greatly affected in the fields of neuropharmacology, neuroanatomy and neurophysiology. Most of published papers using microdialysis have focused on the area of neuroscience, recently more biomedical application. CONCLUSION: In this review, we focused on cerebral microdialysis as a monitoring tool for physiologic and pathophysiologic changes in chemical processes in the brain. Then we presented the principle and various applications of cerebral microdialysis.


Subject(s)
Brain/metabolism , Extracellular Fluid/chemistry , Microdialysis/methods , Neurochemistry/methods , Neurosciences/methods , Animals , Catheterization/instrumentation , Diffusion , Electrochemistry , Electrodes/standards , Humans , Microdialysis/instrumentation , Neurochemistry/instrumentation , Neurosciences/instrumentation , Neurotransmitter Agents/analysis , Neurotransmitter Agents/metabolism
17.
J Neurosci Methods ; 172(2): 215-9, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18555535

ABSTRACT

Creatine (Cr) is an amino acid, which upon phosphorylation is utilized as an energy reservoir in cells with high-energy demand. The ongoing catabolism of creatine to creatinine requires a permanent creatine replenishment into the cells. Because neurons themselves cannot synthesize creatine, they have to take it up via the creatine transporter (CrT). Thus, the concentration of intracellular Cr available for the Cr/PCr shuttle system depends on the expression level of CrT protein. The proton magnetic resonance spectroscopy (MRS) creatine peak (total creatine=tCr) constitutes of two metabolites, namely Cr and phosphocreatine (PCr). We have quantified the level of CrT protein expression with western blotting and compared it to tCr content as estimated by in vitro MRS in Sprague-Dawley rats. Under the assumption of hemispheric symmetry, we took identical samples from left and right hemisphere, which were used for in vitro MRS (tCr) and for western blotting (CrT), respectively. Altogether, it was possible to take 90 corresponding brain samples from 31 animals. A Pearson linear regression analysis for CrT and tCr revealed p<0.0001, explaining 14% of the variance. Since MR-detectable alterations of tCr in the human brain are widespread (e.g. in most major psychiatric disorders proton MRS detectable tCr alterations have been described as regionally and usually state dependent) it is stringent to elucidate their meaning. An influence of tCr on the brain's energy regulating system seems plausible.


Subject(s)
Brain/metabolism , Creatine/metabolism , Energy Metabolism/physiology , Magnetic Resonance Spectroscopy/methods , Membrane Transport Proteins/metabolism , Neurons/metabolism , Animals , Blotting, Western , Brain Chemistry/physiology , Cerebrum/metabolism , Cerebrum/physiopathology , Creatine/analysis , Creatinine/metabolism , Membrane Transport Proteins/analysis , Neurochemistry/instrumentation , Neurochemistry/methods , Phosphocreatine/analysis , Phosphocreatine/metabolism , Protons , Rats , Rats, Sprague-Dawley
18.
Neurochem Int ; 52(6): 1076-85, 2008 May.
Article in English | MEDLINE | ID: mdl-18164517

ABSTRACT

It is known that fibroblast growth factor-1 (FGF1) lacking a conventional signal peptide sequence shows non-classical release independent of the endoplasmic reticulum-Golgi system. Recent studies reveal that FGF1 is co-released with S100A13, a Ca2+-binding protein that acts as an extracellular cargo molecule. Although both FGF1 and S100A13 are Cu2+-binding proteins, the role of Cu2+, as well as that of Ca2+, in non-classical release, remains to be clarified. In the present study we examined the requirements of both metal ions for the interaction between these two proteins. The addition of Ca2+ significantly increased the ka value, while decreasing the KD value, for the interaction between Strep-tagII-S100A13 and GST-FGF1; both values were obtained by use of a quartz crystal microbalance, a real-time mass-measuring device. The EC50 of Ca2+ to enhance the interaction was 10.11 microM. Although the addition of Cu2+ alone had no effect, it caused a marked potentiation of the Ca2+-enhanced interaction. The EC50 of Cu2+ for the potentiation was 50.45 nM. On the other hand, the EC50 of Ca2+ and the KD values were decreased from 11.69 to 2.07 microM and 0.75 to 0.38 x 10(-7)M, respectively, by the addition of 200 nM Cu2+. The Cu2+-induced potentiation of this interaction was abolished by amlexanox, which inhibits non-classical release of FGF1. All of these findings suggest that synergistic effects of Ca2+ and Cu2+ play a key role in the interaction between FGF1 and S100A13, which is the initial step in non-classical release of FGF1.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Copper/metabolism , Fibroblast Growth Factor 1/metabolism , S100 Proteins/metabolism , Aminopyridines/pharmacology , Animals , Anti-Allergic Agents/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Biological Assay , Calcium/pharmacology , Calcium Signaling/drug effects , Copper/pharmacology , Dose-Response Relationship, Drug , Humans , Kinetics , Molecular Weight , Neurochemistry/instrumentation , Neurochemistry/methods , Quartz
19.
J Neurosci Methods ; 169(1): 135-40, 2008 Mar 30.
Article in English | MEDLINE | ID: mdl-18201768

ABSTRACT

Deficiency of docosahexaenoic acid (DHA) and other omega-3 (omega3) fatty acids may constitute an alterable risk factor for Alzheimer's disease (AD). Mechanisms of potential involvement of DHA in the disease process have been postulated primarily from studies in vitro and in mouse models of AD. Information on the fatty acid profile of the brain in AD itself is limited and in some respects contradictory. Interpretation of the findings is complicated by the diversity of methods used in previous studies and a lack of information as to the effect of post-mortem delay on the results. Here we report the development of a simple and highly reproducible method that enables relatively high-throughput measurement of the fatty acid composition in samples of brain tissue and using this method we have demonstrated that there is no significant change in fatty acid composition under conditions designed to model post-mortem delay of up to 3 days at 4 degrees C (or even at room temperature). The development of this method and the observation that delay of up to 3 days has no effect on fatty acid content will facilitate further studies of fatty acid composition on large cohorts of post-mortem brains.


Subject(s)
Body Temperature/physiology , Brain Chemistry/physiology , Fatty Acids/analysis , Lipid Metabolism/physiology , Neurochemistry/methods , Postmortem Changes , Biological Assay/instrumentation , Biological Assay/methods , Chromatography/instrumentation , Chromatography/methods , Docosahexaenoic Acids/analysis , Docosahexaenoic Acids/metabolism , Fatty Acids/metabolism , Humans , Methylation , Neurochemistry/instrumentation , Time Factors
20.
Neurosci Res ; 60(1): 120-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17983679

ABSTRACT

In vivo voltammetry is a valuable technique for rapid measurement of dopamine in the brain of freely behaving rats. Using a conventional voltammetry system, however, behavioural freedom is restricted by cables connecting the head assembly to the measurement system. To overcome these difficulties, we developed a wireless voltammetry system utilizing radio waves. This system consisted of a potentiostat and transmitter system that was mounted on the back of the rat, and a receiver and analysis system. A single-step pulse (100-250 mV) was applied at 4 Hz after an activation pulse to a carbon fibre recording electrode (diameter: 7 microm). Measurement of dopamine (detection limit: 2.7 x 10(-7)M) was demonstrated in vitro. In vivo experiment was performed at least 1 week after the recording electrode was implanted in the rat striatum. Administration of 2-phenylethylamine to rats increased dopamine signal current, which was consistent with the result in the microdialysis measurement. During a resident-intruder test, dopamine signal current in a resident rat increased upon introduction of an intruder rat. These results show that the present wireless system is useful for a long-term measurement of dopamine in behaving rats.


Subject(s)
Brain Chemistry/physiology , Corpus Striatum/metabolism , Dopamine/analysis , Electrochemistry/instrumentation , Neurochemistry/methods , Telemetry/instrumentation , Anesthesia/methods , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain Chemistry/drug effects , Corpus Striatum/drug effects , Dopamine/metabolism , Electric Stimulation , Electrochemistry/methods , Electrodes, Implanted/standards , Electrophysiology/instrumentation , Electrophysiology/methods , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Male , Microdialysis/instrumentation , Microdialysis/methods , Neurochemistry/instrumentation , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , Rats , Reaction Time/drug effects , Reaction Time/physiology , Social Behavior , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors , Wakefulness/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...