Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 19478, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945922

ABSTRACT

Studying animal models furthers our understanding of Parkinson's disease (PD) pathophysiology by providing tools to investigate detailed molecular, cellular and circuit functions. Different versions of the neurotoxin-based 6-hydroxydopamine (6-OHDA) model of PD have been widely used in rats. However, these models typically assess the result of extensive and definitive dopaminergic lesions that reflect a late stage of PD, leading to a paucity of studies and a consequential gap of knowledge regarding initial stages, in which early interventions would be possible. Additionally, the better availability of genetic tools increasingly shifts the focus of research from rats to mice, but few mouse PD models are available yet. To address these, we characterize here the behavioral, neuronal and ultrastructural features of a graded-dose unilateral, single-injection, striatal 6-OHDA model in mice, focusing on early-stage changes within the first two weeks of lesion induction. We observed early onset, dose-dependent impairments of overall locomotion without substantial deterioration of motor coordination. In accordance, histological evaluation demonstrated a partial, dose-dependent loss of dopaminergic neurons of substantia nigra pars compacta (SNc). Furthermore, electron microscopic analysis revealed degenerative ultrastructural changes in SNc dopaminergic neurons. Our results show that mild ultrastructural and cellular degradation of dopaminergic neurons of the SNc can lead to certain motor deficits shortly after unilateral striatal lesions, suggesting that a unilateral dose-dependent intrastriatal 6-OHDA lesion protocol can serve as a successful model of the early stages of Parkinson's disease in mice.


Subject(s)
Parkinson Disease , Rats , Mice , Animals , Parkinson Disease/etiology , Parkinson Disease/pathology , Oxidopamine/pharmacology , Pars Compacta/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Disease Models, Animal , Substantia Nigra/metabolism
2.
Sci Rep ; 11(1): 9775, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963220

ABSTRACT

Infrared neural stimulation is a promising tool for stimulating the brain because it can be used to excite with high spatial precision without the need of delivering or inserting any exogenous agent into the tissue. Very few studies have explored its use in the brain, as most investigations have focused on sensory or motor nerve stimulation. Using intravital calcium imaging with the genetically encoded calcium indicator GCaMP6f, here we show that the application of infrared neural stimulation induces intracellular calcium signals in Layer 2/3 neurons in mouse cortex in vivo. The number of neurons exhibiting infrared-induced calcium response as well as the amplitude of those signals are shown to be both increasing with the energy density applied. By studying as well the spatial extent of the stimulation, we show that reproducibility of the stimulation is achieved mainly in the central part of the infrared beam path. Stimulating in vivo at such a degree of precision and without any exogenous chromophores enables multiple applications, from mapping the brain's connectome to applications in systems neuroscience and the development of new therapeutic tools for investigating the pathological brain.


Subject(s)
Calcium Signaling , Calcium/metabolism , Evoked Potentials/physiology , Imaging, Three-Dimensional , Neurons/physiology , Photons , Visual Cortex/cytology , Animals , Infrared Rays , Mice, Inbred C57BL , Neurons/metabolism
3.
Neuroscience ; 435: 95-111, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32222556

ABSTRACT

The extent of the networks that control the genesis and modulation of hippocampal sharp-wave ripples (SPW-Rs), which are involved in memory consolidation, remains incompletely understood. Here, we performed a detailed in vivo analysis of single cell firing in the lateral supramammillary nucleus (lSuM) during theta and slow oscillations, including SPW-Rs, in anesthetized rats. We classified neurons as SPW-R-active and SPW-R-unchanged according to whether or not they increased their firing during SPW-Rs. We show that lSuM SPW-R-active neurons increase their firing prior to SPW-Rs peak power and prior to hippocampal excitatory cell activation. Moreover, lSuM SPW-R-active neurons show increased firing activity during theta and slow oscillations as compared to unchanged neurons. These results suggest that a sub-population of lSuM neurons can interact with the hippocampus during SPW-Rs, raising the possibility that the lSuM may modulate memory consolidation.


Subject(s)
Hippocampus , Memory Consolidation , Animals , Hypothalamus, Posterior , Neurons , Rats
4.
J Vis Exp ; (147)2019 05 16.
Article in English | MEDLINE | ID: mdl-31157762

ABSTRACT

Epilepsy is a group of neurological disorders which affects millions of people worldwide. Although treatment with medication is helpful in 70% of the cases, serious side effects affect the quality of life of patients. Moreover, a high percentage of epileptic patients are drug resistant; in their case, neurosurgery or neurostimulation are necessary. Therefore, the major goal of epilepsy research is to discover new therapies which are either capable of curing epilepsy without side effects or preventing recurrent seizures in drug-resistant patients. Neuroengineering provides new approaches by using novel strategies and technologies to find better solutions to cure epileptic patients at risk. As a demonstration of a novel experimental protocol in an acute mouse model of epilepsy, a direct in situ electrophoretic drug delivery system is used. Namely, a neural probe incorporating a microfluidic ion pump (µFIP) for on-demand drug delivery and simultaneous recording of local neural activity is implanted and demonstrated to be capable of controlling 4-aminopyridine-induced (4AP-induced) seizure-like event (SLE) activity. The γ-aminobutyric acid (GABA) concentration is kept in the physiological range by the precise control of GABA delivery to reach an antiepileptic effect in the seizure focus but not to cause overinhibition-induced rebound bursts. The method allows both the detection of pathological activity and intervention to stop seizures by delivering inhibitory neurotransmitters directly to the epileptic focus with precise spatiotemporal control. As a result of the developments to the experimental method, SLEs can be induced in a highly localized manner that allows seizure control by the precisely tuned GABA delivery at the seizure onset.


Subject(s)
Electrophoresis , Epilepsy/drug therapy , Seizures/prevention & control , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/therapeutic use , 4-Aminopyridine , Animals , Brain/diagnostic imaging , Brain/pathology , Craniotomy , Disease Models, Animal , Humans , Male , Mice , Microfluidics , Seizures/chemically induced
5.
Adv Biosyst ; 3(2): e1800270, 2019 02.
Article in English | MEDLINE | ID: mdl-32627377

ABSTRACT

The challenge of treating neurological disorders has motivated the development of implantable devices that can deliver treatment when and where it is needed. This study presents a novel brain implant capable of electrophoretically delivering drugs and recording local neural activity on the surface of the brain. The drug delivery is made possible by the integration of a microfluidic ion pump (µFIP) into a conformable electrocorticography (ECoG) device with recording cites embedded next to the drug delivery outlets. The µFIP ECoG device can deliver a high capacity of several biologically important cationic species on demand. The therapeutic potential of the device is demonstrated by using it to deliver neurotransmitters in a rodent model while simultaneously recording local neural activity. These developments represent a significant step forward for cortical drug-delivery systems.


Subject(s)
Brain , Drug Delivery Systems/instrumentation , Electrocorticography/instrumentation , Microfluidic Analytical Techniques/instrumentation , Animals , Brain/drug effects , Brain/physiology , Electrocorticography/methods , Electrophoresis/instrumentation , Equipment Design , Male , Mice , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/pharmacology
6.
Sci Adv ; 4(8): eaau1291, 2018 08.
Article in English | MEDLINE | ID: mdl-30167463

ABSTRACT

The persistence of intractable neurological disorders necessitates novel therapeutic solutions. We demonstrate the utility of direct in situ electrophoretic drug delivery to treat neurological disorders. We present a neural probe incorporating a microfluidic ion pump (µFIP) for on-demand drug delivery and electrodes for recording local neural activity. The µFIP works by electrophoretically pumping ions across an ion exchange membrane and thereby delivers only the drug of interest and not the solvent. This "dry" delivery enables precise drug release into the brain region with negligible local pressure increase. The therapeutic potential of the µFIP probe is tested in a rodent model of epilepsy. The µFIP probe can detect pathological activity and then intervene to stop seizures by delivering inhibitory neurotransmitters directly to the seizure source. We anticipate that further tailored engineering of the µFIP platform will enable additional applications in neural interfacing and the treatment of neurological disorders.


Subject(s)
Drug Delivery Systems , GABA Agents/administration & dosage , Microfluidics/methods , Seizures/prevention & control , gamma-Aminobutyric Acid/administration & dosage , Animals , Mice
7.
J Neural Eng ; 15(6): 065001, 2018 12.
Article in English | MEDLINE | ID: mdl-30132444

ABSTRACT

OBJECTIVE: Neural electrophysiology is often conducted with traditional, rigid depth probes. The mechanical mismatch between these probes and soft brain tissue is unfavorable for tissue interfacing. Making probes compliant can improve biocompatibility, but as a consequence, they become more difficult to insert into the brain. Therefore, new methods for inserting compliant neural probes must be developed. APPROACH: Here, we present a new bioresorbable shuttle based on the hydrolytically degradable poly(vinyl alcohol) (PVA) and poly(lactic-co-glycolic acid) (PLGA). We show how to fabricate the PVA/PLGA shuttles on flexible and thin parylene probes. The method consists of PDMS molding used to fabricate a PVA shuttle aligned with the probe and to also impart a sharp tip necessary for piercing brain tissue. The PVA shuttle is then dip-coated with PLGA to create a bi-layered shuttle. MAIN RESULTS: While single layered PVA shuttles are able to penetrate agarose brain models, only limited depths were achieved and repositioning was not possible due to the fast degradation. We demonstrate that a bilayered approach incorporating a slower dissolving PLGA layer prolongs degradation and enables facile insertion for at least several millimeters depth. Impedances of electrodes before and after shuttle preparation were characterized and showed that careful deposition of PLGA is required to maintain low impedance. Facilitated by the shuttles, compliant parylene probes were also successfully implanted into anaesthetized mice and enabled the recording of high quality local field potentials. SIGNIFICANCE: This work thereby presents a solution towards addressing a key challenge of implanting compliant neural probes using a two polymer system. PVA and PLGA are polymers with properties ideal for translation: commercially available, biocompatible with FDA-approved uses and bioresorbable. By presenting new ways to implant compliant neural probes, we can begin to fully evaluate their chronic biocompatibility and performance compared to traditional, rigid electronics.


Subject(s)
Biocompatible Materials , Electrodes, Implanted , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polyvinyl Alcohol/chemistry , Absorbable Implants , Animals , Brain , Electric Impedance , Male , Mice , Mice, Inbred C57BL
8.
eNeuro ; 5(6)2018.
Article in English | MEDLINE | ID: mdl-30783610

ABSTRACT

Transparent and flexible materials are attractive for a wide range of emerging bioelectronic applications. These include neural interfacing devices for both recording and stimulation, where low electrochemical electrode impedance is valuable. Here the conducting polymer poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) is used to fabricate electrodes that are small enough to allow unencumbered optical access for imaging a large cell population with two-photon (2P) microscopy, yet provide low impedance for simultaneous high quality recordings of neural activity in vivo. To demonstrate this, pathophysiological activity was induced in the mouse cortex using 4-aminopyridine (4AP), and the resulting electrical activity was detected with the PEDOT:PSS-based probe while imaging calcium activity directly below the probe area. The induced calcium activity of the neuronal network as measured by the fluorescence change in the cells correlated well with the electrophysiological recordings from the cortical grid of PEDOT:PSS microelectrodes. Our approach provides a valuable vehicle for complementing classical high temporal resolution electrophysiological analysis with optical imaging.


Subject(s)
Brain/physiology , Electrodes, Implanted , Electrophysiology/instrumentation , Nerve Net/physiology , Neuroimaging/instrumentation , Animals , Electrophysiology/methods , Male , Mice , Mice, Transgenic , Neuroimaging/methods
9.
Neuron ; 82(6): 1367-79, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24945776

ABSTRACT

Sleep spindles are major transient oscillations of the mammalian brain. Spindles are generated in the thalamus; however, what determines their duration is presently unclear. Here, we measured somatic activity of excitatory thalamocortical (TC) cells together with axonal activity of reciprocally coupled inhibitory reticular thalamic cells (nRTs) and quantified cycle-by-cycle alterations in their firing in vivo. We found that spindles with different durations were paralleled by distinct nRT activity, and nRT firing sharply dropped before the termination of all spindles. Both initial nRT and TC activity was correlated with spindle length, but nRT correlation was more robust. Analysis of spindles evoked by optogenetic activation of nRT showed that spindle probability, but not spindle length, was determined by the strength of the light stimulus. Our data indicate that during natural sleep a dynamically fluctuating thalamocortical network controls the duration of sleep spindles via the major inhibitory element of the circuits, the nRT.


Subject(s)
Cerebral Cortex/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Sleep/physiology , Thalamus/physiology , Animals , Electroencephalography/methods , Male , Mice, 129 Strain , Mice, Transgenic , Rats , Rats, Wistar , Time Factors
10.
J Neurosci ; 34(21): 7137-47, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849349

ABSTRACT

GABA-A receptors (GABA-ARs) are typically expressed at synaptic or nonsynaptic sites mediating phasic and tonic inhibition, respectively. These two forms of inhibition conjointly control various network oscillations. To disentangle their roles in thalamocortical rhythms, we focally deleted synaptic, γ2 subunit-containing GABA-ARs in the thalamus using viral intervention in mice. After successful removal of γ2 subunit clusters, spontaneous and evoked GABAergic synaptic currents disappeared in thalamocortical cells when the presynaptic, reticular thalamic (nRT) neurons fired in tonic mode. However, when nRT cells fired in burst mode, slow phasic GABA-AR-mediated events persisted, indicating a dynamic, burst-specific recruitment of nonsynaptic GABA-ARs. In vivo, removal of synaptic GABA-ARs reduced the firing of individual thalamocortical cells but did not abolish slow oscillations or sleep spindles. We conclude that nonsynaptic GABA-ARs are recruited in a phasic manner specifically during burst firing of nRT cells and provide sufficient GABA-AR activation to control major thalamocortical oscillations.


Subject(s)
Cerebral Cortex/physiology , Neural Inhibition/physiology , Neurons/physiology , Receptors, GABA-A/metabolism , Thalamus/physiology , Animals , Dependovirus/genetics , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyridazines/pharmacology , Receptors, GABA-A/genetics , Synapses/drug effects , Synapses/genetics , Vesicular Glutamate Transport Protein 2/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Brain Res Bull ; 97: 16-23, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23707857

ABSTRACT

Pharmacological and functional data suggest the existence of uridine (Urd) receptors in the central nervous system (CNS). In the present study, simultaneous extracellular single unit recording and microiontophoretic injection of the pyrimidine nucleoside Urd was used to provide evidence for the presence of Urd-sensitive neurons in the thalamus and the cerebral cortex of Long Evans rats. Twenty-two neurons in the thalamus (24% of recorded neurons) and 17 neurons in the cortex (55%) responded to the direct iontophoresis of Urd. The majority of Urd-sensitive neurons in the thalamus and cortex (82% and 59%, respectively) increased their firing rate in response to Urd. In contrary, adenosine (Ado) and uridine 5'-triphosphate (UTP) decreased the firing rate of all responding neurons in the thalamus, and the majority of responding neurons in the cortex (83% and 87%, respectively). Functional relevance of Urd-sensitive neurons was investigated in spontaneously epileptic freely moving Long Evans and Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats. Intraperitoneal (i.p.) injection of 500mg/kg Urd decreased epileptic activity (210-270min after injection) in both rat strains. Intraperitoneal administration of 1000mg/kg Urd decreased the number of spike-wave discharges (SWDs) between 150-270min and 90-270min in Long Evans and WAG/Rij rats, respectively. The effect of Urd was long-lasting in both rat strains as the higher dose significantly decreased the number of SWDs even 24h after Urd injection. The present results suggest that Urd-sensitive neurons in the thalamus and the cerebral cortex may play a role in the antiepileptic action of Urd possibly via modulation of thalamocortical neuronal circuits.


Subject(s)
Anticonvulsants/pharmacology , Neural Inhibition , Neurons/drug effects , Uridine/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Epilepsy, Absence/physiopathology , Male , Neurons/physiology , Rats , Rats, Long-Evans , Rats, Wistar , Thalamus/drug effects , Thalamus/physiology
12.
J Neurosci ; 31(2): 607-17, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21228169

ABSTRACT

The exact timing of cortical afferent activity is instrumental for the correct coding and retrieval of internal and external stimuli. Thalamocortical inputs represent the most significant subcortical pathway to the cortex, but the precise timing and temporal variability of thalamocortical activity is not known. To examine this question, we studied the phase of thalamic action potentials relative to cortical oscillations and established correlations among phase, the nuclear location of the thalamocortical neurons, and the frequency of cortical activity. The phase of thalamic action potentials depended on the exact frequency of the slow cortical oscillation both on long (minutes) and short (single wave) time scales. Faster waves were accompanied by phase advancement in both cases. Thalamocortical neurons located in different nuclei fired at significantly different phases of the slow waves but were active at a similar phase of spindle oscillations. Different thalamic nuclei displayed distinct burst patterns. Bursts with a higher number of action potentials displayed progressive phase advancement in a nucleus-specific manner. Thalamic neurons located along nuclear borders were characterized by mixed burst and phase properties. Our data demonstrate that the temporal relationship between cortical and thalamic activity is not fixed but displays dynamic changes during oscillatory activity. The timing depends on the precise location and exact activity of thalamocortical cells and the ongoing cortical network pattern. This variability of thalamic output and its coupling to cortical activity can enable thalamocortical neurons to actively participate in the coding and retrieval of cortical signals.


Subject(s)
Cerebral Cortex/physiology , Thalamus/physiology , Action Potentials , Animals , Male , Neurons/physiology , Periodicity , Rats , Rats, Wistar
13.
J Neurosci ; 28(46): 11848-61, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19005050

ABSTRACT

Diverse sources of GABAergic inhibition are a major feature of cortical networks, but distinct inhibitory input systems have not been systematically characterized in the thalamus. Here, we contrasted the properties of two independent GABAergic pathways in the posterior thalamic nucleus of rat, one input from the reticular thalamic nucleus (nRT), and one "extrareticular" input from the anterior pretectal nucleus (APT). The vast majority of nRT-thalamic terminals formed single synapses per postsynaptic target and innervated thin distal dendrites of relay cells. In contrast, single APT-thalamic terminals formed synaptic contacts exclusively via multiple, closely spaced synapses on thick relay cell dendrites. Quantal analysis demonstrated that the two inputs displayed comparable quantal amplitudes, release probabilities, and multiple release sites. The morphological and physiological data together indicated multiple, single-site contacts for nRT and multisite contacts for APT axons. The contrasting synaptic arrangements of the two pathways were paralleled by different short-term plasticities. The multisite APT-thalamic pathway showed larger charge transfer during 50-100 Hz stimulation compared with the nRT pathway and a greater persistent inhibition accruing during stimulation trains. Our results demonstrate that the two inhibitory systems are morpho-functionally distinct and suggest and that multisite GABAergic terminals are tailored for maintained synaptic inhibition even at high presynaptic firing rates. These data explain the efficacy of extrareticular inhibition in timing relay cell activity in sensory and motor thalamic nuclei. Finally, based on the classic nomenclature and the difference between reticular and extrareticular terminals, we define a novel, multisite GABAergic terminal type (F3) in the thalamus.


Subject(s)
Intralaminar Thalamic Nuclei/metabolism , Posterior Thalamic Nuclei/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Afferent Pathways/metabolism , Afferent Pathways/ultrastructure , Animals , Dendrites/metabolism , Dendrites/ultrastructure , Electric Stimulation , Inhibitory Postsynaptic Potentials/physiology , Intralaminar Thalamic Nuclei/ultrastructure , Male , Microscopy, Immunoelectron , Neural Inhibition/physiology , Posterior Thalamic Nuclei/ultrastructure , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Superior Colliculi/metabolism , Superior Colliculi/ultrastructure , Synaptic Transmission/physiology
14.
J Comp Neurol ; 506(1): 122-40, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17990275

ABSTRACT

The anterior pretectal nucleus (APT) and the zona incerta (ZI) are diencephalic nuclei that exert a strong inhibitory influence selectively in higher order thalamic relays. The APT is also known to project to the ZI as well as the thalamus, but anatomical details of the APT-ZI projection have not been described. In the present study, the efferent pathways of the APT were examined in the APT-ZI-thalamus network by using anterograde and retrograde tracing in combination with pre- and postembedding immunocytochemical stainings and in situ hybridization. The vast majority of APT fibers selectively innervated the parvalbumin-positive, ventral part of the ZI, which contains ZI neurons with axons projecting to higher order thalamic nuclei. The APT-ZI pathway consisted of both gamma-aminobutyric acid (GABA)-negative and GABA-positive components; 38.2% of the terminals in the ZI contained GABA, and 8.6% of the projecting somata in the APT were glutamic acid decarboxylase 67 (GAD67) mRNA positive. The combination of parvalbumin immunostaining with retrograde tracing showed that strongly and weakly parvalbumin-positive as well as parvalbumin-negative neurons were all among the population of APT cells projecting to the ZI. Similar heterogeneity was found among the APT cells projecting to the thalamus. Double retrograde tracing from higher order thalamic nuclei and their topographically matched ZI regions revealed hardly any APT neuron with dual projections. Our data suggest that both ZI and the higher order thalamic relays are innervated by distinct, physiologically heterogeneous APT neurons. These various efferent pathways probably interact via the rich recurrent collaterals of the projecting APT cells. Therefore, the powerful, GABAergic APT and ZI outputs to the thalamus are apparently co-modulated in a synergistic manner via dual excitatory and inhibitory APT-ZI connections.


Subject(s)
Rats, Wistar/anatomy & histology , Subthalamus/cytology , Superior Colliculi/cytology , Thalamic Nuclei/cytology , Animals , Biotin/analogs & derivatives , Dextrans , Glutamate Decarboxylase/metabolism , Male , Microscopy, Electron , Neural Pathways , Neurons/metabolism , Neurons/ultrastructure , Nociceptors/physiology , Phytohemagglutinins , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , gamma-Aminobutyric Acid/metabolism
15.
J Neurosci ; 27(7): 1670-81, 2007 Feb 14.
Article in English | MEDLINE | ID: mdl-17301175

ABSTRACT

The zona incerta (ZI) is at the crossroad of almost all major ascending and descending fiber tracts and targets numerous brain centers from the thalamus to the spinal cord. Effective ascending drive of ZI cells has been described, but the role of descending cortical signals in patterning ZI activity is unknown. Cortical control over ZI function was examined during slow cortical waves (1-3 Hz), paroxysmal high-voltage spindles (HVSs), and 5-9 Hz oscillations in anesthetized rats. In all conditions, rhythmic cortical activity significantly altered the firing pattern of ZI neurons recorded extracellularly and labeled with the juxtacellular method. During slow oscillations, the majority of ZI neurons became synchronized to the depth-negative phase ("up state") of the cortical waves to a degree comparable to thalamocortical neurons. During HVSs, ZI cells displayed highly rhythmic activity in tight synchrony with the cortical oscillations. ZI neurons responded to short epochs of cortical 5-9 Hz oscillations, with a change in the interspike interval distribution and with an increase in spectral density in the 5-9 Hz band as measured by wavelet analysis. Morphological reconstruction revealed that most ZI cells have mediolaterally extensive dendritic trees and very long dendritic segments. Cortical terminals established asymmetrical synapses on ZI cells with very long active zones. These data suggest efficient integration of widespread cortical signals by single ZI neurons and strong cortical drive. We propose that the efferent GABAergic signal of ZI neurons patterned by the cortical activity can play a critical role in synchronizing thalamocortical and brainstem rhythms.


Subject(s)
Cerebral Cortex/physiology , Neural Pathways/physiology , Subthalamus/physiology , Action Potentials/drug effects , Action Potentials/physiology , Anesthesia/methods , Animals , Brain Mapping , Cerebral Cortex/drug effects , Cortical Synchronization , Electroencephalography , Imaging, Three-Dimensional , Microscopy, Electron, Transmission , Neural Pathways/drug effects , Neurons/physiology , Neurons/ultrastructure , Rats , Subthalamus/cytology , Subthalamus/drug effects , Urethane/pharmacology
16.
J Physiol ; 574(Pt 1): 209-27, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16627566

ABSTRACT

Absence-related spike-and-wave discharges (SWDs) occur in the thalamocortical system during quiet wakefulness or drowsiness. In feline generalized penicillin epilepsy, SWDs develop from sleep spindles. In contrast, in genetic absence epilepsy rats from Strasbourg (GAERS), SWDs develop from wake-related 5-9 Hz oscillations, which are distinct from spindle oscillations (7-15 Hz). Since these two oscillation types share common frequency bands and may contribute to SWD genesis, it is important to compare their thalamic cellular mechanisms. Under neuroleptic analgesia, in GAERS and control non-epileptic rats barbiturates abolished both SWDs and 5-9 Hz oscillations but increased the incidence of spindle-like oscillations. Within the thalamocortical circuit 5-9 Hz oscillations occurred more coherently than spindle-like oscillations. Intracellular events associated with 5-9 Hz and spindle-like oscillations were distinctively different in both thalamic relay and reticular neurons. In both cell types, SWDs and 5-9 Hz oscillations emerged from a significantly more depolarized membrane potential than spindle-like oscillations. In relay neurons, 5-9 Hz oscillations were mainly characterized by a rhythmic depolarization, which occurred during a tonic hyperpolarization and which could trigger an apparent low-threshold Ca2+ potential, whereas spindle-like oscillations were characterized by a rhythmic hyperpolarization. In reticular cells, SWDs and 5-9 Hz oscillations occurred during a tonic hyperpolarization, whereas spindle-like oscillations occurred during a long-lasting depolarizing envelope. The difference in the intracellular events between 5-9 Hz and spindle-like oscillations and similarities between 5-9 Hz oscillations and SWDs indicate that in GAERS, 5-9 Hz oscillations are more pro-epileptogenic than spindle-like oscillations. In conclusion, the present study strongly supports the hypothesis that SWDs in GAERS are generated by a wake-related corticothalamic resonance, and not by sleep-related, hypersynchronous, spindle-like activity originating in the thalamus.


Subject(s)
Action Potentials , Biological Clocks , Cerebral Cortex/physiopathology , Epilepsy/physiopathology , Neural Pathways/physiopathology , Neurons , Sleep , Animals , Electroencephalography/methods , Male , Rats , Rats, Wistar
17.
Neuron ; 45(6): 929-40, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15797553

ABSTRACT

GABAergic signaling is central to the function of the thalamus and has been traditionally attributed primarily to the nucleus reticularis thalami (nRT). Here we present a GABAergic pathway, distinct from the nRT, that exerts a powerful inhibitory effect selectively in higher-order thalamic relays of the rat. Axons originating in the anterior pretectal nucleus (APT) innervated the proximal dendrites of relay cells via large GABAergic terminals with multiple release sites. Stimulation of the APT in an in vitro slice preparation revealed a GABA(A) receptor-mediated, monosynaptic IPSC in relay cells. Activation of presumed single APT fibers induced rebound burst firing in relay cells. Different APT neurons recorded in vivo displayed fast bursting, tonic, or rhythmic firing. Our data suggest that selective extrareticular GABAergic control of relay cell activity will result in effective, state-dependent gating of thalamocortical information transfer in higher-order but not in first-order relays.


Subject(s)
Afferent Pathways/physiology , Biotin/analogs & derivatives , Mesencephalon/physiology , Neural Inhibition/physiology , Synaptic Transmission/physiology , Thalamus/physiology , gamma-Aminobutyric Acid/metabolism , Action Potentials/physiology , Afferent Pathways/ultrastructure , Animals , Cell Shape/physiology , Dendrites/physiology , Dendrites/ultrastructure , Dextrans , Electric Stimulation , Immunohistochemistry , Male , Mesencephalon/ultrastructure , Microscopy, Electron, Transmission , Organ Culture Techniques , Parvalbumins/metabolism , Phytohemagglutinins , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Thalamus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...