1
|
Fleiner AS, Kolnier D, Hagger-Vaughan N, Ræder J, Storm JF. Effects of ketamine and propofol on muscarinic plateau potentials in rat neocortical pyramidal cells. PLoS One 2025; 20:e0316262. [PMID: 39746093 PMCID: PMC11695037 DOI: 10.1371/journal.pone.0316262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Propofol and ketamine are widely used general anaesthetics, but have different effects on consciousness: propofol gives a deeply unconscious state, with little or no dream reports, whereas vivid dreams are often reported after ketamine anaesthesia. Ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist, while propofol is a γ-aminobutyric-acid (GABAA) receptor positive allosteric modulator, but these mechanisms do not fully explain how these drugs alter consciousness. Most previous in vitro studies of cellular mechanisms of anaesthetics have used brain slices or neurons in a nearly "comatose" state, because no "arousing" neuromodulators were added. Here we tested mechanisms of anaesthetics in rat medial prefrontal cortex (mPFC) slices after bath-applying the cholinergic agonist muscarine to partly mimic an "aroused-like" state, using whole-cell patch-clamp recordings from layer 2/3 pyramidal cells (L2/3PCs). According to leading theories of access consciousness and working memory, L2/3PCs are particularly important for these cognitive functions. We found that muscarine induced long-lasting depolarising plateau potentials (PPs) and spiking following brief depolarising current injections in the L2/3PCs. After 2 hours of pre-incubation with ketamine or propofol, the muscarine-induced PPs were altered in seemingly different ways: 3 μM propofol reduced the PPs and (significantly) spiking, whereas 20 μM ketamine seemed to enhance PPs and spiking (non-significantly). Brief wash-in of these drug concentrations failed to induce such effects, probably due to insufficient equilibration by diffusion in the slices. In contrast, pre-incubation with a high dose (100 μM) of ketamine suppressed the PPs and spiking. We discuss whether the apparently different effects on PPs may possibly be related to contrasting clinical effects: ketamine causing atypical anaesthesia with vivid, "psychedelic" dreaming while propofol causes less dreaming.
Collapse
Affiliation(s)
- Anne S. Fleiner
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Daniel Kolnier
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Nicholas Hagger-Vaughan
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Johan Ræder
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Johan F. Storm
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Lindquist BE, Timbie C, Voskobiynyk Y, Paz JT. Thalamocortical circuits in generalized epilepsy: Pathophysiologic mechanisms and therapeutic targets. Neurobiol Dis 2023; 181:106094. [PMID: 36990364 PMCID: PMC10192143 DOI: 10.1016/j.nbd.2023.106094] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/02/2023] [Accepted: 03/19/2023] [Indexed: 03/29/2023] Open
Abstract
Generalized epilepsy affects 24 million people globally; at least 25% of cases remain medically refractory. The thalamus, with widespread connections throughout the brain, plays a critical role in generalized epilepsy. The intrinsic properties of thalamic neurons and the synaptic connections between populations of neurons in the nucleus reticularis thalami and thalamocortical relay nuclei help generate different firing patterns that influence brain states. In particular, transitions from tonic firing to highly synchronized burst firing mode in thalamic neurons can cause seizures that rapidly generalize and cause altered awareness and unconsciousness. Here, we review the most recent advances in our understanding of how thalamic activity is regulated and discuss the gaps in our understanding of the mechanisms of generalized epilepsy syndromes. Elucidating the role of the thalamus in generalized epilepsy syndromes may lead to new opportunities to better treat pharmaco-resistant generalized epilepsy by thalamic modulation and dietary therapy.
Collapse
Affiliation(s)
- Britta E Lindquist
- UCSF Department of Neurology, Division of Neurocritical Care, United States of America; UCSF Department of Neurology, Division of Pediatric Epilepsy, United States of America; UCSF Department of Neurology, United States of America
| | - Clare Timbie
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, Division of Pediatric Epilepsy, United States of America; UCSF Department of Neurology, United States of America
| | - Yuliya Voskobiynyk
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, United States of America
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, United States of America; Kavli Institute for Fundamental Neuroscience, UCSF, United States of America.
| |
Collapse
|
3
|
Sedative Properties of Dexmedetomidine Are Mediated Independently from Native Thalamic Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel Function at Clinically Relevant Concentrations. Int J Mol Sci 2022; 24:ijms24010519. [PMID: 36613961 PMCID: PMC9820684 DOI: 10.3390/ijms24010519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Dexmedetomidine is a selective α2-adrenoceptor agonist and appears to disinhibit endogenous sleep-promoting pathways, as well as to attenuate noradrenergic excitation. Recent evidence suggests that dexmedetomidine might also directly inhibit hyperpolarization-activated cyclic-nucleotide gated (HCN) channels. We analyzed the effects of dexmedetomidine on native HCN channel function in thalamocortical relay neurons of the ventrobasal complex of the thalamus from mice, performing whole-cell patch-clamp recordings. Over a clinically relevant range of concentrations (1-10 µM), the effects of dexmedetomidine were modest. At a concentration of 10 µM, dexmedetomidine significantly reduced maximal Ih amplitude (relative reduction: 0.86 [0.78-0.91], n = 10, and p = 0.021), yet changes to the half-maximal activation potential V1/2 occurred exclusively in the presence of the very high concentration of 100 µM (-4,7 [-7.5--4.0] mV, n = 10, and p = 0.009). Coincidentally, only the very high concentration of 100 µM induced a significant deceleration of the fast component of the HCN activation time course (τfast: +135.1 [+64.7-+151.3] ms, n = 10, and p = 0.002). With the exception of significantly increasing the membrane input resistance (starting at 10 µM), dexmedetomidine did not affect biophysical membrane properties and HCN channel-mediated parameters of neuronal excitability. Hence, the sedative qualities of dexmedetomidine and its effect on the thalamocortical network are not decisively shaped by direct inhibition of HCN channel function.
Collapse
|
4
|
Liu Y, Chen B, Cai Y, Han Y, Xia Y, Li N, Fan B, Yuan T, Jiang J, Gao PO, Yu W, Jiao Y, Li W. Activation of anterior thalamic reticular nucleus GABAergic neurons promotes arousal from propofol anesthesia in mice. Acta Biochim Biophys Sin (Shanghai) 2021; 53:883-892. [PMID: 33929026 DOI: 10.1093/abbs/gmab056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Indexed: 11/14/2022] Open
Abstract
Propofol is widely used for the induction and maintenance of anesthesia, which causes a rapid loss of consciousness. However, the mechanisms underlying the hypnosis effect of propofol are still not fully understood. The thalamic reticular nucleus (TRN) is crucial for regulating wakefulness, sleep rhythm generation, and sleep stability, while the role of TRN in the process of propofol-induced anesthesia is still unknown. Here, we investigated the function of the anterior TRN in propofol general anesthesia. Our results demonstrated that the neural activity of anterior TRN is suppressed during propofol anesthesia, whereas it is robustly activated from anesthesia by recording the calcium signals using fiber photometry technology. The results showed that the activation of anterior TRN neurons by chemogenetic and optogenetic methods shortens the emergency time without changing the induction time. Conversely, chemogenetic or optogenetic inhibition of the TRN neurons leads to a delay in the recovery time. Our study showed that anterior TRN is crucial for behavioral arousal without affecting the induction time of propofol anesthesia.
Collapse
Affiliation(s)
- Yanjun Liu
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Bing Chen
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Yirong Cai
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Yuan Han
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Ying Xia
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Nanqi Li
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Bingqian Fan
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Tianjie Yuan
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Junli Jiang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - P o Gao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Wenxian Li
- Department of Anesthesiology, Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| |
Collapse
|
5
|
Kadam M, Perveen S, Kushwah N, Prasad D, Panjwani U, Kumar B, Khan N. Elucidating the role of hypoxia/reoxygenation in hippocampus-dependent memory impairment: do SK channels play role? Exp Brain Res 2021; 239:1747-1763. [PMID: 33779792 DOI: 10.1007/s00221-021-06095-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
Professionals and mountaineers often face the problem of reperfusion injury due to re-oxygenation, upon their return to sea-level after sojourn at high altitude. Small conductance calcium-activated potassium channels (SK channels) have a role in regulating hippocampal synaptic plasticity. However, the role of SK channels under hypoxia-reoxygenation (H/R) is unknown. The present study hypothesized that SK channels play a significant role in H/R induced cognitive dysfunction. Sprague-Dawley rats were exposed to simulated HH (25,000 ft) continuously for 7 days followed by reoxygenation periods 3, 6, 24, 48, 72 and 120 h. It was observed that H/R exposure caused impairment in spatial memory as indicated by increased latency (p < 0.001) and pathlength (p < 0.001). The SK1 channel expression increased upon HH exposure (102.89 ± 7.055), which abrogated upon reoxygenation. HH exposure results in an increase in SK2 (CA3, 297.67 ± 6.69) and SK3 (CA1, 246 ± 5.13) channels which continued to increase gradually upon reoxygenation. The number of pyknotic cells (24 ± 2.03) (p < 0.01) and the expression of caspase-3 increased with HH exposure, which continued in the reoxygenation group (177.795 ± 1.264). Similar pattern was observed in lipid peroxidation (p < 0.001), LDH activity (p < 0.001) and ROS production (p < 0.001). A positive correlation of memory, cell death and oxidative stress indicates that H/R exposure increases oxidative stress coupled with SK channel expression, which may play a role in H/R-induced cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Manisha Kadam
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Saba Perveen
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Neetu Kushwah
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Dipti Prasad
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Usha Panjwani
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Bhuvnesh Kumar
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Nilofar Khan
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
6
|
Schwerin S, Kopp C, Pircher E, Schneider G, Kreuzer M, Haseneder R, Kratzer S. Attenuation of Native Hyperpolarization-Activated, Cyclic Nucleotide-Gated Channel Function by the Volatile Anesthetic Sevoflurane in Mouse Thalamocortical Relay Neurons. Front Cell Neurosci 2021; 14:606687. [PMID: 33551750 PMCID: PMC7858256 DOI: 10.3389/fncel.2020.606687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/18/2020] [Indexed: 11/24/2022] Open
Abstract
As thalamocortical relay neurons are ascribed a crucial role in signal propagation and information processing, they have attracted considerable attention as potential targets for anesthetic modulation. In this study, we analyzed the effects of different concentrations of sevoflurane on the excitability of thalamocortical relay neurons and hyperpolarization-activated, cyclic-nucleotide gated (HCN) channels, which play a decisive role in regulating membrane properties and rhythmic oscillatory activity. The effects of sevoflurane on single-cell excitability and native HCN channels were investigated in acutely prepared brain slices from adult wild-type mice with the whole-cell patch-clamp technique, using voltage-clamp and current-clamp protocols. Sevoflurane dose-dependently depressed membrane biophysics and HCN-mediated parameters of neuronal excitability. Respective half-maximal inhibitory and effective concentrations ranged between 0.30 (95% CI, 0.18–0.50) mM and 0.88 (95% CI, 0.40–2.20) mM. We witnessed a pronounced reduction of HCN dependent Ih current amplitude starting at a concentration of 0.45 mM [relative change at −133 mV; 0.45 mM sevoflurane: 0.85 (interquartile range, 0.79–0.92), n = 12, p = 0.011; 1.47 mM sevoflurane: 0.37 (interquartile range, 0.34–0.62), n = 5, p < 0.001] with a half-maximal inhibitory concentration of 0.88 (95% CI, 0.40–2.20) mM. In contrast, effects on voltage-dependent channel gating were modest with significant changes only occurring at 1.47 mM [absolute change of half-maximal activation potential; 1.47 mM: −7.2 (interquartile range, −10.3 to −5.8) mV, n = 5, p = 0.020]. In this study, we demonstrate that sevoflurane inhibits the excitability of thalamocortical relay neurons in a concentration-dependent manner within a clinically relevant range. Especially concerning its effects on native HCN channel function, our findings indicate substance-specific differences in comparison to other anesthetic agents. Considering the importance of HCN channels, the observed effects might mechanistically contribute to the hypnotic properties of sevoflurane.
Collapse
Affiliation(s)
- Stefan Schwerin
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Claudia Kopp
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Elisabeth Pircher
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Rainer Haseneder
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| | - Stephan Kratzer
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich School of Medicine, Munich, Germany
| |
Collapse
|
7
|
Zhang Y, Fu B, Liu C, Yu S, Luo T, Zhang L, Zhou W, Yu T. Activation of noradrenergic terminals in the reticular thalamus delays arousal from propofol anesthesia in mice. FASEB J 2019; 33:7252-7260. [DOI: 10.1096/fj.201802164rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Yu Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityGuizhouChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityGuizhouChina
| | - Bao Fu
- Department of Critical Care MedicineAffiliated Hospital of Zunyi Medical UniversityGuizhouChina
| | - Chengxi Liu
- Guizhou Key Laboratory of Brain ScienceZunyi Medical UniversityGuizhouChina
| | - Shouyang Yu
- Guizhou Key Laboratory of Brain ScienceZunyi Medical UniversityGuizhouChina
| | - Tianyuan Luo
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityGuizhouChina
| | - Lin Zhang
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityGuizhouChina
| | - Wenjing Zhou
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityGuizhouChina
| | - Tian Yu
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityGuizhouChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityGuizhouChina
| |
Collapse
|
8
|
Regulatory Effect of General Anesthetics on Activity of Potassium Channels. Neurosci Bull 2018; 34:887-900. [PMID: 29948841 PMCID: PMC6129254 DOI: 10.1007/s12264-018-0239-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
General anesthesia is an unconscious state induced by anesthetics for surgery. The molecular targets and cellular mechanisms of general anesthetics in the mammalian nervous system have been investigated during past decades. In recent years, K+ channels have been identified as important targets of both volatile and intravenous anesthetics. This review covers achievements that have been made both on the regulatory effect of general anesthetics on the activity of K+ channels and their underlying mechanisms. Advances in research on the modulation of K+ channels by general anesthetics are summarized and categorized according to four large K+ channel families based on their amino-acid sequence homology. In addition, research achievements on the roles of K+ channels in general anesthesia in vivo, especially with regard to studies using mice with K+ channel knockout, are particularly emphasized.
Collapse
|
9
|
Kratzer S, Mattusch C, Garcia PS, Schmid S, Kochs E, Rammes G, Schneider G, Kreuzer M, Haseneder R. Propofol and Sevoflurane Differentially Modulate Cortical Depolarization following Electric Stimulation of the Ventrobasal Thalamus. Front Comput Neurosci 2017; 11:109. [PMID: 29321737 PMCID: PMC5732174 DOI: 10.3389/fncom.2017.00109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/13/2017] [Indexed: 01/14/2023] Open
Abstract
The neuronal mechanisms how anesthetics lead to loss of consciousness are unclear. Thalamocortical interactions are crucially involved in conscious perception; hence the thalamocortical network might be a promising target for anesthetic modulation of neuronal information pertaining to arousal and waking behavior. General anesthetics affect the neurophysiology of the thalamus and the cortex but the exact mechanisms of how anesthetics interfere with processing thalamocortical information remain to be elucidated. Here we investigated the effect of the anesthetic agents sevoflurane and propofol on thalamocortical network activity in vitro. We used voltage-sensitive dye imaging techniques to analyze the cortical depolarization in response to stimulation of the thalamic ventrobasal nucleus in brain slices from mice. Exposure to sevoflurane globally decreased cortical depolarization in a dose-dependent manner. Sevoflurane reduced the intensity and extent of cortical depolarization and delayed thalamocortical signal propagation. In contrast, propofol neither affected area nor amplitude of cortical depolarization. However, propofol exposure resulted in regional changes in spatial distribution of maximum fluorescence intensity in deep regions of the cortex. In summary, our experiments revealed substance-specific effects on the thalamocortical network. Functional changes of the neuronal network are known to be pivotally involved in the anesthetic-induced loss of consciousness. Our findings provide further evidence that the mechanisms of anesthetic-mediated loss of consciousness are drug- and pathway-specific.
Collapse
Affiliation(s)
- Stephan Kratzer
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Corinna Mattusch
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Paul S Garcia
- Department of Anesthesiology, Emory University, Atlanta, GA, United States
- Research Service, Atlanta VA Medical Center, Atlanta, GA, United States
| | - Sebastian Schmid
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Eberhard Kochs
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gerhard Rammes
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gerhard Schneider
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Matthias Kreuzer
- Department of Anesthesiology, Emory University, Atlanta, GA, United States
- Research Service, Atlanta VA Medical Center, Atlanta, GA, United States
| | - Rainer Haseneder
- Department of Anesthesiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
10
|
Hashemi M, Hutt A, Hight D, Sleigh J. Anesthetic action on the transmission delay between cortex and thalamus explains the beta-buzz observed under propofol anesthesia. PLoS One 2017; 12:e0179286. [PMID: 28622355 PMCID: PMC5473556 DOI: 10.1371/journal.pone.0179286] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 05/26/2017] [Indexed: 11/18/2022] Open
Abstract
In recent years, more and more surgeries under general anesthesia have been performed with the assistance of electroencephalogram (EEG) monitors. An increase in anesthetic concentration leads to characteristic changes in the power spectra of the EEG. Although tracking the anesthetic-induced changes in EEG rhythms can be employed to estimate the depth of anesthesia, their precise underlying mechanisms are still unknown. A prominent feature in the EEG of some patients is the emergence of a strong power peak in the β-frequency band, which moves to the α-frequency band while increasing the anesthetic concentration. This feature is called the beta-buzz. In the present study, we use a thalamo-cortical neural population feedback model to reproduce observed characteristic features in frontal EEG power obtained experimentally during propofol general anesthesia, such as this beta-buzz. First, we find that the spectral power peak in the α- and δ-frequency ranges depend on the decay rate constant of excitatory and inhibitory synapses, but the anesthetic action on synapses does not explain the beta-buzz. Moreover, considering the action of propofol on the transmission delay between cortex and thalamus, the model reveals that the beta-buzz may result from a prolongation of the transmission delay by increasing propofol concentration. A corresponding relationship between transmission delay and anesthetic blood concentration is derived. Finally, an analytical stability study demonstrates that increasing propofol concentration moves the systems resting state towards its stability threshold.
Collapse
Affiliation(s)
- Meysam Hashemi
- INRIA Grand Est - Nancy, Team NEUROSYS, Villers-lès-Nancy, France
- CNRS, Loria, UMR nō 7503, Vandoeuvre-lès-Nancy, France
- Université de Lorraine, Loria, UMR nō 7503, Vandoeuvre-lès-Nancy, France
- Aix Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille, France
| | - Axel Hutt
- German Meteorology Service, Offenbach am Main, Germany
- Department of Mathematics and Statistics, University of Reading, Reading, United Kingdom
| | | | | |
Collapse
|
11
|
Kaneko K, Koyanagi Y, Oi Y, Kobayashi M. Propofol-induced spike firing suppression is more pronounced in pyramidal neurons than in fast-spiking neurons in the rat insular cortex. Neuroscience 2016; 339:548-560. [DOI: 10.1016/j.neuroscience.2016.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/15/2016] [Accepted: 10/04/2016] [Indexed: 11/15/2022]
|
12
|
Wells MF, Wimmer RD, Schmitt LI, Feng G, Halassa MM. Thalamic reticular impairment underlies attention deficit in Ptchd1(Y/-) mice. Nature 2016; 532:58-63. [PMID: 27007844 PMCID: PMC4875756 DOI: 10.1038/nature17427] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/17/2016] [Indexed: 12/19/2022]
Abstract
Developmental disabilities, including attention-deficit hyperactivity disorder (ADHD), intellectual disability (ID), and autism spectrum disorders (ASD), affect 1 in 6 children in the United States. Recently, PTCHD1 (Patched-domain containing protein 1) gene mutations have been found in ~1% of patients with ID and ASD. PTCHD1 deletion patients show symptoms of ADHD, sleep disruption, hypotonia, aggression, ASD, and ID. Although PTCHD1 is likely critical for normal development, the connection between its deletion and the ensuing behavioral defects is poorly understood. Here, we report that during early postnatal development, mouse Ptchd1 is selectively expressed in the thalamic reticular nucleus (TRN), a group of GABAergic neurons that regulate thalamo-cortical transmission, sleep rhythms, and attention. Ptchd1 deletion attenuates TRN activity by reducing calcium-dependent potassium currents (SK). Restricted TRN deletion of Ptchd1 leads to attention deficits and hyperactivity, both of which are rescued by pharmacological augmentation of SK channels. Global Ptchd1 deletion recapitulates learning impairment, hyper-aggression, and motor defects, all of which are insensitive to SK pharmacological targeting and not found in the TRN-restricted deletion mouse. This study maps clinically-relevant behavioral phenotypes onto TRN dysfunction in a human disease model, while also identifying molecular and circuit targets for intervention.
Collapse
Affiliation(s)
- Michael F Wells
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA.,McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ralf D Wimmer
- Neuroscience Institute, New York University Langone Medical Center, New York, New York 10016, USA.,Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, New York 10016, USA
| | - L Ian Schmitt
- Neuroscience Institute, New York University Langone Medical Center, New York, New York 10016, USA.,Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, New York 10016, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Michael M Halassa
- Neuroscience Institute, New York University Langone Medical Center, New York, New York 10016, USA.,Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, New York 10016, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, New York 10016, USA.,Center for Neural Science, New York University, New York, New York 1003, USA
| |
Collapse
|
13
|
Heuermann RJ, Jaramillo TC, Ying SW, Suter BA, Lyman KA, Han Y, Lewis AS, Hampton TG, Shepherd GMG, Goldstein PA, Chetkovich DM. Reduction of thalamic and cortical Ih by deletion of TRIP8b produces a mouse model of human absence epilepsy. Neurobiol Dis 2016; 85:81-92. [PMID: 26459112 PMCID: PMC4688217 DOI: 10.1016/j.nbd.2015.10.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/22/2015] [Accepted: 10/08/2015] [Indexed: 12/19/2022] Open
Abstract
Absence seizures occur in several types of human epilepsy and result from widespread, synchronous feedback between the cortex and thalamus that produces brief episodes of loss of consciousness. Genetic rodent models have been invaluable for investigating the pathophysiological basis of these seizures. Here, we identify tetratricopeptide-containing Rab8b-interacting protein (TRIP8b) knockout mice as a new model of absence epilepsy, featuring spontaneous spike-wave discharges on electroencephalography (EEG) that are the electrographic hallmark of absence seizures. TRIP8b is an auxiliary subunit of the hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels, which have previously been implicated in the pathogenesis of absence seizures. In contrast to mice lacking the pore-forming HCN channel subunit HCN2, TRIP8b knockout mice exhibited normal cardiac and motor function and a less severe seizure phenotype. Evaluating the circuit that underlies absence seizures, we found that TRIP8b knockout mice had significantly reduced HCN channel expression and function in thalamic-projecting cortical layer 5b neurons and thalamic relay neurons, but preserved function in inhibitory neurons of the reticular thalamic nucleus. Our results expand the known roles of TRIP8b and provide new insight into the region-specific functions of TRIP8b and HCN channels in constraining cortico-thalamo-cortical excitability.
Collapse
Affiliation(s)
- Robert J Heuermann
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Thomas C Jaramillo
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Shui-Wang Ying
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Ave., Room A-1050, New York, New York 10021, USA.
| | - Benjamin A Suter
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Kyle A Lyman
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Ye Han
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Alan S Lewis
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Thomas G Hampton
- Mouse Specifics, Inc., 2 Central Street, Level 1 Suite 1, Framingham, MA 01701, USA.
| | - Gordon M G Shepherd
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| | - Peter A Goldstein
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Ave., Room A-1050, New York, New York 10021, USA.
| | - Dane M Chetkovich
- Davee Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave., Ward Building, Room 10-201, Chicago, IL 60611, USA.
| |
Collapse
|
14
|
Retinoic Acid-Mediated Regulation of GLI3 Enables Efficient Motoneuron Derivation from Human ESCs in the Absence of Extrinsic SHH Activation. J Neurosci 2015; 35:11462-81. [PMID: 26290227 DOI: 10.1523/jneurosci.3046-14.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The derivation of somatic motoneurons (MNs) from ES cells (ESCs) after exposure to sonic hedgehog (SHH) and retinoic acid (RA) is one of the best defined, directed differentiation strategies to specify fate in pluripotent lineages. In mouse ESCs, MN yield is particularly high after RA + SHH treatment, whereas human ESC (hESC) protocols have been generally less efficient. In an effort to optimize yield, we observe that functional MNs can be derived from hESCs at high efficiencies if treated with patterning molecules at very early differentiation steps before neural induction. Remarkably, under these conditions, equal numbers of human MNs were obtained in the presence or absence of SHH exposure. Using pharmacological and genetic strategies, we demonstrate that early RA treatment directs MN differentiation independently of extrinsic SHH activation by suppressing the induction of GLI3. We further demonstrate that neural induction triggers a switch from a poised to an active chromatin state at GLI3. Early RA treatment prevents this switch by direct binding of the RA receptor at the GLI3 promoter. Furthermore, GLI3 knock-out hESCs can bypass the requirement for early RA patterning to yield MNs efficiently. Our data demonstrate that RA-mediated suppression of GLI3 is sufficient to generate MNs in an SHH-independent manner and that temporal changes in exposure to patterning factors such as RA affect chromatin state and competency of hESC-derived lineages to adopt specific neuronal fates. Finally, our work presents a streamlined platform for the highly efficient derivation of human MNs from ESCs and induced pluripotent stem cells. SIGNIFICANCE STATEMENT Our study presents a rapid and efficient protocol to generate human motoneurons from embryonic and induced pluripotent stem cells. Surprisingly, and in contrast to previous work, motoneurons are generated in the presence of retinoic acid but in the absence of factors that activate sonic hedgehog signaling. We show that early exposure to retinoic acid modulates the chromatin state of cells to be permissive for motoneuron generation and directly suppresses the induction of GLI3, a negative regulator of SHH signaling. Therefore, our data point to a novel mechanism by which retinoic acid exposure can bypass the requirement for extrinsic SHH treatment during motoneuron induction.
Collapse
|
15
|
How the cortico-thalamic feedback affects the EEG power spectrum over frontal and occipital regions during propofol-induced sedation. J Comput Neurosci 2015; 39:155-79. [PMID: 26256583 DOI: 10.1007/s10827-015-0569-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/05/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022]
Abstract
Increasing concentrations of the anaesthetic agent propofol initially induces sedation before achieving full general anaesthesia. During this state of anaesthesia, the observed specific changes in electroencephalographic (EEG) rhythms comprise increased activity in the δ- (0.5-4 Hz) and α- (8-13 Hz) frequency bands over the frontal region, but increased δ- and decreased α-activity over the occipital region. It is known that the cortex, the thalamus, and the thalamo-cortical feedback loop contribute to some degree to the propofol-induced changes in the EEG power spectrum. However the precise role of each structure to the dynamics of the EEG is unknown. In this paper we apply a thalamo-cortical neuronal population model to reproduce the power spectrum changes in EEG during propofol-induced anaesthesia sedation. The model reproduces the power spectrum features observed experimentally both in frontal and occipital electrodes. Moreover, a detailed analysis of the model indicates the importance of multiple resting states in brain activity. The work suggests that the α-activity originates from the cortico-thalamic relay interaction, whereas the emergence of δ-activity results from the full cortico-reticular-relay-cortical feedback loop with a prominent enforced thalamic reticular-relay interaction. This model suggests an important role for synaptic GABAergic receptors at relay neurons and, more generally, for the thalamus in the generation of both the δ- and the α- EEG patterns that are seen during propofol anaesthesia sedation.
Collapse
|
16
|
Amico E, Gomez F, Di Perri C, Vanhaudenhuyse A, Lesenfants D, Boveroux P, Bonhomme V, Brichant JF, Marinazzo D, Laureys S. Posterior cingulate cortex-related co-activation patterns: a resting state FMRI study in propofol-induced loss of consciousness. PLoS One 2014; 9:e100012. [PMID: 24979748 PMCID: PMC4076184 DOI: 10.1371/journal.pone.0100012] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/21/2014] [Indexed: 11/25/2022] Open
Abstract
Background Recent studies have been shown that functional connectivity of cerebral areas is not a static phenomenon, but exhibits spontaneous fluctuations over time. There is evidence that fluctuating connectivity is an intrinsic phenomenon of brain dynamics that persists during anesthesia. Lately, point process analysis applied on functional data has revealed that much of the information regarding brain connectivity is contained in a fraction of critical time points of a resting state dataset. In the present study we want to extend this methodology for the investigation of resting state fMRI spatial pattern changes during propofol-induced modulation of consciousness, with the aim of extracting new insights on brain networks consciousness-dependent fluctuations. Methods Resting-state fMRI volumes on 18 healthy subjects were acquired in four clinical states during propofol injection: wakefulness, sedation, unconsciousness, and recovery. The dataset was reduced to a spatio-temporal point process by selecting time points in the Posterior Cingulate Cortex (PCC) at which the signal is higher than a given threshold (i.e., BOLD intensity above 1 standard deviation). Spatial clustering on the PCC time frames extracted was then performed (number of clusters = 8), to obtain 8 different PCC co-activation patterns (CAPs) for each level of consciousness. Results The current analysis shows that the core of the PCC-CAPs throughout consciousness modulation seems to be preserved. Nonetheless, this methodology enables to differentiate region-specific propofol-induced reductions in PCC-CAPs, some of them already present in the functional connectivity literature (e.g., disconnections of the prefrontal cortex, thalamus, auditory cortex), some others new (e.g., reduced co-activation in motor cortex and visual area). Conclusion In conclusion, our results indicate that the employed methodology can help in improving and refining the characterization of local functional changes in the brain associated to propofol-induced modulation of consciousness.
Collapse
Affiliation(s)
- Enrico Amico
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
- Faculty of Psychology and Educational Sciences, Department of Data Analysis, Ghent University, Ghent, Belgium
- * E-mail:
| | - Francisco Gomez
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
| | - Carol Di Perri
- Department of Neuroradiology, National Neurological Institute C. Mondino, Pavia, Italy
| | - Audrey Vanhaudenhuyse
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
- Department of Algology and Palliative Care, CHU Sart Tilman Hospital, University of Liège, Liège, Belgium
| | - Damien Lesenfants
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
| | - Pierre Boveroux
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
- Department of Anesthesia and Intensive Care Medicine, CHU Sart Tilman Hospital, University of Liège, Liège, Belgium
| | - Vincent Bonhomme
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
- Department of Anesthesia and Intensive Care Medicine, CHU Sart Tilman Hospital, University of Liège, Liège, Belgium
- Department of Anesthesia and Intensive Care Medicine, CHR Citadelle, University of Liège, Liège, Belgium
| | - Jean-François Brichant
- Department of Anesthesia and Intensive Care Medicine, CHU Sart Tilman Hospital, University of Liège, Liège, Belgium
| | - Daniele Marinazzo
- Faculty of Psychology and Educational Sciences, Department of Data Analysis, Ghent University, Ghent, Belgium
| | - Steven Laureys
- Coma Science Group, Cyclotron Research Centre, University of Liège, Liège, Belgium
- Department of Neurology, University of Liège, Liège, Belgium
| |
Collapse
|
17
|
Herd MB, Lambert JJ, Belelli D. The general anaesthetic etomidate inhibits the excitability of mouse thalamocortical relay neurons by modulating multiple modes of GABAA receptor-mediated inhibition. Eur J Neurosci 2014; 40:2487-501. [PMID: 24773078 PMCID: PMC4215602 DOI: 10.1111/ejn.12601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/12/2014] [Accepted: 03/26/2014] [Indexed: 12/12/2022]
Abstract
Modulation of thalamocortical (TC) relay neuron function has been implicated in the sedative and hypnotic effects of general anaesthetics. Inhibition of TC neurons is mediated predominantly by a combination of phasic and tonic inhibition, together with a recently described ‘spillover’ mode of inhibition, generated by the dynamic recruitment of extrasynaptic γ-aminobutyric acid (GABA)A receptors (GABAARs). Previous studies demonstrated that the intravenous anaesthetic etomidate enhances tonic and phasic inhibition in TC relay neurons, but it is not known how etomidate may influence spillover inhibition. Moreover, it is unclear how etomidate influences the excitability of TC neurons. Thus, to investigate the relative contribution of synaptic (α1β2γ2) and extrasynaptic (α4β2δ) GABAARs to the thalamic effects of etomidate, we performed whole-cell recordings from mouse TC neurons lacking synaptic (α10/0) or extrasynaptic (δ0/0) GABAARs. Etomidate (3 μm) significantly inhibited action-potential discharge in a manner that was dependent on facilitation of both synaptic and extrasynaptic GABAARs, although enhanced tonic inhibition was dominant in this respect. Additionally, phasic inhibition evoked by stimulation of the nucleus reticularis exhibited a spillover component mediated by δ-GABAARs, which was significantly prolonged in the presence of etomidate. Thus, etomidate greatly enhanced the transient suppression of TC spike trains by evoked inhibitory postsynaptic potentials. Collectively, these results suggest that the deactivation of thalamus observed during etomidate-induced anaesthesia involves potentiation of tonic and phasic inhibition, and implicate amplification of spillover inhibition as a novel mechanism to regulate the gating of sensory information through the thalamus during anaesthetic states.
Collapse
Affiliation(s)
- Murray B Herd
- Division of Neuroscience, Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | | | | |
Collapse
|
18
|
Lafaille FG, Pessach IM, Zhang SY, Ciancanelli MJ, Herman M, Abhyankar A, Ying SW, Keros S, Goldstein PA, Mostoslavsky G, Ordovas-Montanes J, Jouanguy E, Plancoulaine S, Tu E, Elkabetz Y, Al-Muhsen S, Tardieu M, Schlaeger TM, Daley GQ, Abel L, Casanova JL, Studer L, Notarangelo LD. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012; 491:769-73. [PMID: 23103873 PMCID: PMC3527075 DOI: 10.1038/nature11583] [Citation(s) in RCA: 262] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 09/12/2012] [Indexed: 12/14/2022]
Abstract
In the course of primary infection with herpes simplex virus 1 (HSV-1), children with inborn errors of toll-like receptor 3 (TLR3) immunity are prone to HSV-1 encephalitis (HSE). We tested the hypothesis that the pathogenesis of HSE involves non-haematopoietic CNS-resident cells. We derived induced pluripotent stem cells (iPSCs) from the dermal fibroblasts of TLR3- and UNC-93B-deficient patients and from controls. These iPSCs were differentiated into highly purified populations of neural stem cells (NSCs), neurons, astrocytes and oligodendrocytes. The induction of interferon-β (IFN-β) and/or IFN-λ1 in response to stimulation by the dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) was dependent on TLR3 and UNC-93B in all cells tested. However, the induction of IFN-β and IFN-λ1 in response to HSV-1 infection was impaired selectively in UNC-93B-deficient neurons and oligodendrocytes. These cells were also much more susceptible to HSV-1 infection than control cells, whereas UNC-93B-deficient NSCs and astrocytes were not. TLR3-deficient neurons were also found to be susceptible to HSV-1 infection. The rescue of UNC-93B- and TLR3-deficient cells with the corresponding wild-type allele showed that the genetic defect was the cause of the poly(I:C) and HSV-1 phenotypes. The viral infection phenotype was rescued further by treatment with exogenous IFN-α or IFN-β ( IFN-α/β) but not IFN-λ1. Thus, impaired TLR3- and UNC-93B-dependent IFN-α/β intrinsic immunity to HSV-1 in the CNS, in neurons and oligodendrocytes in particular, may underlie the pathogenesis of HSE in children with TLR3-pathway deficiencies.
Collapse
Affiliation(s)
- Fabien G Lafaille
- Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ying SW, Kanda VA, Hu Z, Purtell K, King EC, Abbott GW, Goldstein PA. Targeted deletion of Kcne2 impairs HCN channel function in mouse thalamocortical circuits. PLoS One 2012; 7:e42756. [PMID: 22880098 PMCID: PMC3411840 DOI: 10.1371/journal.pone.0042756] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/12/2012] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels generate the pacemaking current, I(h), which regulates neuronal excitability, burst firing activity, rhythmogenesis, and synaptic integration. The physiological consequence of HCN activation depends on regulation of channel gating by endogenous modulators and stabilization of the channel complex formed by principal and ancillary subunits. KCNE2 is a voltage-gated potassium channel ancillary subunit that also regulates heterologously expressed HCN channels; whether KCNE2 regulates neuronal HCN channel function is unknown. METHODOLOGY/PRINCIPAL FINDINGS We investigated the effects of Kcne2 gene deletion on I(h) properties and excitability in ventrobasal (VB) and cortical layer 6 pyramidal neurons using brain slices prepared from Kcne2(+/+) and Kcne2(-/-) mice. Kcne2 deletion shifted the voltage-dependence of I(h) activation to more hyperpolarized potentials, slowed gating kinetics, and decreased I(h) density. Kcne2 deletion was associated with a reduction in whole-brain expression of both HCN1 and HCN2 (but not HCN4), although co-immunoprecipitation from whole-brain lysates failed to detect interaction of KCNE2 with HCN1 or 2. Kcne2 deletion also increased input resistance and temporal summation of subthreshold voltage responses; this increased intrinsic excitability enhanced burst firing in response to 4-aminopyridine. Burst duration increased in corticothalamic, but not thalamocortical, neurons, suggesting enhanced cortical excitatory input to the thalamus; such augmented excitability did not result from changes in glutamate release machinery since miniature EPSC frequency was unaltered in Kcne2(-/-) neurons. CONCLUSIONS/SIGNIFICANCE Loss of KCNE2 leads to downregulation of HCN channel function associated with increased excitability in neurons in the cortico-thalamo-cortical loop. Such findings further our understanding of the normal physiology of brain circuitry critically involved in cognition and have implications for our understanding of various disorders of consciousness.
Collapse
Affiliation(s)
- Shui-Wang Ying
- Department of Anesthesiology, Weill Cornell Medical College, New York, New York, United States of America
| | - Vikram A. Kanda
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, United States of America
| | - Zhaoyang Hu
- Departments of Pharmacology, and Physiology and Biophysics, University of California Irvine, Irvine, California, United States of America
| | - Kerry Purtell
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, United States of America
| | - Elizabeth C. King
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, United States of America
| | - Geoffrey W. Abbott
- Departments of Pharmacology, and Physiology and Biophysics, University of California Irvine, Irvine, California, United States of America
| | - Peter A. Goldstein
- Department of Anesthesiology, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
20
|
Affiliation(s)
- John P. Adelman
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239;
| | - James Maylie
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon 97239;
| | - Pankaj Sah
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, 4072, Australia;
| |
Collapse
|
21
|
Ying SW, Tibbs GR, Picollo A, Abbas SY, Sanford RL, Accardi A, Hofmann F, Ludwig A, Goldstein PA. PIP2-mediated HCN3 channel gating is crucial for rhythmic burst firing in thalamic intergeniculate leaflet neurons. J Neurosci 2011; 31:10412-23. [PMID: 21753018 PMCID: PMC6623048 DOI: 10.1523/jneurosci.0021-11.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 04/21/2011] [Accepted: 05/30/2011] [Indexed: 01/26/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels generate a pacemaking current, I(h), which regulates neuronal excitability and oscillatory activity in the brain. Although all four HCN isoforms are expressed in the brain, the functional contribution of HCN3 is unknown. Using immunohistochemistry, confocal microscopy, and whole-cell patch-clamp recording techniques, we investigated HCN3 function in thalamic intergeniculate leaflet (IGL) neurons, as HCN3 is reportedly preferentially expressed in these cells. We observed that I(h) recorded from IGL, but not ventral geniculate nucleus, neurons in HCN2(+/+) mice and rats activated slowly and were cAMP insensitive, which are hallmarks of HCN3 channels. We also observed strong immunolabeling for HCN3, with no labeling for HCN1 and HCN4, and only very weak labeling for HCN2. Deletion of HCN2 did not alter I(h) characteristics in mouse IGL neurons. These data together indicate that the HCN3 channel isoform generated I(h) in IGL neurons. Intracellular phosphatidylinositol-4,5-bisphosphate (PIP(2)) shifted I(h) activation to more depolarized potentials and accelerated activation kinetics. Upregulation of HCN3 function by PIP(2) augmented low-threshold burst firing and spontaneous oscillations; conversely, depletion of PIP(2) or pharmacologic block of I(h) resulted in a profound inhibition of excitability. The results indicate that functional expression of HCN3 channels in IGL neurons is crucial for intrinsic excitability and rhythmic burst firing, and PIP(2) serves as a powerful modulator of I(h)-dependent properties via an effect on HCN3 channel gating. Since the IGL is a major input to the suprachiasmatic nucleus, regulation of pacemaking function by PIP(2) in the IGL may influence sleep and circadian rhythms.
Collapse
Affiliation(s)
- Shui-Wang Ying
- Department of Anesthesiology, Weill Cornell Medical College, New York, New York 10065, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hutt A, Longtin A. Effects of the anesthetic agent propofol on neural populations. Cogn Neurodyn 2010; 4:37-59. [PMID: 19768579 PMCID: PMC2837528 DOI: 10.1007/s11571-009-9092-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 08/29/2009] [Accepted: 08/31/2009] [Indexed: 11/30/2022] Open
Abstract
The neuronal mechanisms of general anesthesia are still poorly understood. Besides several characteristic features of anesthesia observed in experiments, a prominent effect is the bi-phasic change of power in the observed electroencephalogram (EEG), i.e. the initial increase and subsequent decrease of the EEG-power in several frequency bands while increasing the concentration of the anaesthetic agent. The present work aims to derive analytical conditions for this bi-phasic spectral behavior by the study of a neural population model. This model describes mathematically the effective membrane potential and involves excitatory and inhibitory synapses, excitatory and inhibitory cells, nonlocal spatial interactions and a finite axonal conduction speed. The work derives conditions for synaptic time constants based on experimental results and gives conditions on the resting state stability. Further the power spectrum of Local Field Potentials and EEG generated by the neural activity is derived analytically and allow for the detailed study of bi-spectral power changes. We find bi-phasic power changes both in monostable and bistable system regime, affirming the omnipresence of bi-spectral power changes in anesthesia. Further the work gives conditions for the strong increase of power in the δ-frequency band for large propofol concentrations as observed in experiments.
Collapse
Affiliation(s)
- Axel Hutt
- INRIA CR Nancy - Grand Est, CS20101, 54603 Villers-ls-Nancy Cedex, France
| | - Andre Longtin
- Department of Physics, University of Ottawa, 150 Louis Pasteur, Ottawa, ON K1N-6N5 Canada
| |
Collapse
|
23
|
Souza A, Castelo T, Queiroz J, Barros I, Paula V, Oliveira M, Silva A. Evaluation of anesthetic protocol for the collection of semen from captive collared peccaries (Tayassu tajacu) by electroejaculation. Anim Reprod Sci 2009; 116:370-5. [DOI: 10.1016/j.anireprosci.2009.02.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 02/19/2009] [Accepted: 02/27/2009] [Indexed: 10/21/2022]
|
24
|
Jin YH, Zhang Z, Mendelowitz D, Andresen MC. Presynaptic actions of propofol enhance inhibitory synaptic transmission in isolated solitary tract nucleus neurons. Brain Res 2009; 1286:75-83. [PMID: 19559683 DOI: 10.1016/j.brainres.2009.06.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 06/17/2009] [Accepted: 06/18/2009] [Indexed: 10/20/2022]
Abstract
General anesthetics variably enhance inhibitory synaptic transmission that relies on (-aminobutyric acid (GABA) and GABAA receptor function with distinct differences across brain regions. Activation of "extra-synaptic" GABAA receptors produces a tonic current considered the most sensitive target for general anesthetics, particularly in forebrain neurons. To evaluate the contribution of poor drug access to neurons in slices, we tested the intravenous anesthetic propofol in mechanically isolated neurons from the solitary tract nucleus (NTS). Setting chloride concentrations to ECl=-29 mV made GABA currents inward at holding potentials of -60 mV. Propofol triggered pronounced but slowly-developing tonic currents that reversed with 5 min washing. Effective concentrations in isolated cells were lower than in slices and propofol enhanced phasic IPSCs more potently than tonic currents (1 microM increased phasic decay-time constant vs. >3 microM tonic currents). Propofol increased IPSC frequency (>3 microM), a presynaptic action. Bicuculline blocked all propofol actions. Gabazine blocked only phasic IPSCs. IPSCs persisted in TTX and/or cadmium but these agents prevented propofol-induced increases in IPSC frequency. Furosemide (>1 mM) reversibly blocked propofol-evoked IPSC frequency changes without altering waveforms. We conclude that presynaptic actions of propofol depend on a depolarizing chloride gradient across presynaptic inhibitory terminals. Our results in isolated neurons indicate that propofol pharmacokinetics intrinsically trigger the tonic currents slowly and the time course is not related to slow permeation or delivery. Unlike forebrain, phasic NTS GABAA receptors are more sensitive to propofol than tonic receptors but that presynaptic GABAA receptor mechanisms regulate GABA release.
Collapse
Affiliation(s)
- Young-Ho Jin
- Department of Physiology, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | |
Collapse
|
25
|
Ying SW, Werner DF, Homanics GE, Harrison NL, Goldstein PA. Isoflurane modulates excitability in the mouse thalamus via GABA-dependent and GABA-independent mechanisms. Neuropharmacology 2008; 56:438-47. [PMID: 18948126 DOI: 10.1016/j.neuropharm.2008.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 09/17/2008] [Accepted: 09/22/2008] [Indexed: 11/29/2022]
Abstract
GABAergic neurons in the reticular thalamic nucleus (RTN) synapse onto thalamocortical neurons in the ventrobasal (VB) thalamus, and this reticulo-thalamocortical pathway is considered an anatomic target for general anesthetic-induced unconsciousness. A mutant mouse was engineered to harbor two amino acid substitutions (S270H, L277A) in the GABA(A) receptor (GABA(A)-R) alpha1 subunit; this mutation abolished sensitivity to the volatile anesthetic isoflurane in recombinant GABA(A)-Rs, and reduced in vivo sensitivity to isoflurane in the loss-of-righting-reflex assay. We examined the effects of the double mutation on GABA(A)-R-mediated synaptic currents and isoflurane sensitivity by recording from thalamic neurons in brain slices. The double mutation accelerated the decay, and decreased the (1/2) width of, evoked inhibitory postsynaptic currents (eIPSCs) in VB neurons and attenuated isoflurane-induced prolongation of the eIPSC. The hypnotic zolpidem, a selective modulator of GABA(A)-Rs containing the alpha1 subunit, prolonged eIPSC duration regardless of genotype, indicating that mutant mice incorporate alpha1 subunit-containing GABA(A)-Rs into synapses. In RTN neurons, which lack the alpha1 subunit, eIPSC duration was longer than in VB, regardless of genotype. Isoflurane reduced the efficacy of GABAergic transmission from RTN to VB, independent of genotype, suggesting a presynaptic action in RTN neurons. Consistent with this observation, isoflurane inhibited both tonic action potential and rebound burst firing in the presence of GABA(A)-R blockade. The suppressed excitability in RTN neurons is likely mediated by isoflurane-enhanced Ba(2+)-sensitive, but 4-aminopyridine-insenstive, potassium conductances. We conclude that isoflurane enhances inhibition of thalamic neurons in VB via GABA(A)-R-dependent, but in RTN via GABA(A)-R-independent, mechanisms.
Collapse
Affiliation(s)
- Shui-Wang Ying
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Cornell Medical College, 1300 York Avenue, Room A-1050, New York, NY 10065, United States
| | | | | | | | | |
Collapse
|
26
|
The mechanisms of propofol-induced block on ion currents in differentiated H9c2 cardiac cells. Eur J Pharmacol 2008; 590:93-8. [DOI: 10.1016/j.ejphar.2008.05.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 05/09/2008] [Accepted: 05/22/2008] [Indexed: 11/20/2022]
|
27
|
Ying SW, Jia F, Abbas SY, Hofmann F, Ludwig A, Goldstein PA. Dendritic HCN2 channels constrain glutamate-driven excitability in reticular thalamic neurons. J Neurosci 2007; 27:8719-32. [PMID: 17687049 PMCID: PMC6672930 DOI: 10.1523/jneurosci.1630-07.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 06/27/2007] [Accepted: 06/27/2007] [Indexed: 01/07/2023] Open
Abstract
Hyperpolarization activated cyclic nucleotide (HCN) gated channels conduct a current, I(h); how I(h) influences excitability and spike firing depends primarily on channel distribution in subcellular compartments. For example, dendritic expression of HCN1 normalizes somatic voltage responses and spike output in hippocampal and cortical neurons. We reported previously that HCN2 is predominantly expressed in dendritic spines in reticular thalamic nucleus (RTN) neurons, but the functional impact of such nonsomatic HCN2 expression remains unknown. We examined the role of HCN2 expression in regulating RTN excitability and GABAergic output from RTN to thalamocortical relay neurons using wild-type and HCN2 knock-out mice. Pharmacological blockade of I(h) significantly increased spike firing in RTN neurons and large spontaneous IPSC frequency in relay neurons; conversely, pharmacological enhancement of HCN channel function decreased spontaneous IPSC frequency. HCN2 deletion abolished I(h) in RTN neurons and significantly decreased sensitivity to 8-bromo-cAMP and lamotrigine. Recapitulating the effects of I(h) block, HCN2 deletion increased both temporal summation of EPSPs in RTN neurons as well as GABAergic output to postsynaptic relay neurons. The enhanced excitability of RTN neurons after I(h) block required activation of ionotropic glutamate receptors; consistent with this was the colocalization of HCN2 and glutamate receptor 4 subunit immunoreactivities in dendritic spines of RTN neurons. The results indicate that, in mouse RTN neurons, HCN2 is the primary functional isoform underlying I(h) and expression of HCN2 constrains excitatory synaptic integration.
Collapse
Affiliation(s)
- Shui-Wang Ying
- C. V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College, Cornell University, New York, New York 10021
| | - Fan Jia
- C. V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College, Cornell University, New York, New York 10021
| | - Syed Y. Abbas
- C. V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College, Cornell University, New York, New York 10021
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, 80802 München, Germany, and
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Peter A. Goldstein
- C. V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College, Cornell University, New York, New York 10021
| |
Collapse
|
28
|
Sukhotinsky I, Reiner K, Govrin-Lippmann R, Belenky M, Lu J, Hopkins DA, Saper CB, Devor M. Projections from the mesopontine tegmental anesthesia area to regions involved in pain modulation. J Chem Neuroanat 2006; 32:159-78. [PMID: 17049433 DOI: 10.1016/j.jchemneu.2006.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 08/25/2006] [Accepted: 08/30/2006] [Indexed: 11/19/2022]
Abstract
Pentobarbital microinjected into a restricted locus in the upper brainstem induces a general anesthesia-like state characterized by atonia, loss of consciousness, and pain suppression as assessed by loss of nocifensive response to noxious stimuli. This locus is the mesopontine tegmental anesthesia area (MPTA). Although anesthetic agents directly influence spinal cord nociceptive processing, antinociception during intracerebral microinjection indicates that they can also act supraspinally. Using neuroanatomical tracing methods we show that the MPTA has multiple descending projections to brainstem and spinal areas associated with pain modulation. Most prominent is a massive projection to the rostromedial medulla, a nodal region for descending pain modulation. Together with the periaqueductal gray (PAG), the MPTA is the major mesopontine input to this region. Less dense projections target the PAG, the locus coeruleus and pericoerulear areas, and dorsal and ventral reticular nuclei of the caudal medulla. The MPTA also has modest direct projections to the trigeminal nuclear complex and to superficial layers of the dorsal horn. Double anterograde and retrograde labeling at the light and electron microscopic levels shows that MPTA neurons with descending projections synapse directly on spinally projecting cells of rostromedial medulla. The prominence of the MPTA's projection to the rostromedial medulla suggests that, like the PAG, it may exert antinociceptive actions via this bulbospinal relay.
Collapse
Affiliation(s)
- I Sukhotinsky
- Department of Cell and Animal Biology, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abbas SY, Ying SW, Goldstein PA. Compartmental distribution of hyperpolarization-activated cyclic-nucleotide-gated channel 2 and hyperpolarization-activated cyclic-nucleotide-gated channel 4 in thalamic reticular and thalamocortical relay neurons. Neuroscience 2006; 141:1811-25. [PMID: 16806719 DOI: 10.1016/j.neuroscience.2006.05.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 04/27/2006] [Accepted: 05/16/2006] [Indexed: 01/07/2023]
Abstract
Hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels conduct a monovalent cationic current, I(h), which contributes to the electrophysiological properties of neurons and regulates thalamic oscillations in circuits containing the glutamatergic ventrobasal complex (VB) and GABAergic reticular thalamic nucleus (RTN). Four distinct HCN channel isoforms (HCN1-4) have been identified, and mRNAs and proteins for HCN channels have been detected in the RTN and VB, with HCN2 and HCN4 being the predominant isoforms. RTN and VB neurons have distinct electrophysiological properties, and those differences may reflect variable compartmental distribution of HCN channels. Whole cell patch clamp recordings from thalamic neurons in brain slices obtained from C57/Bl6 mice demonstrate that I(h) is much smaller in RTN than in VB neurons although the time constants for I(h) current activation are very similar. To study the compartmental distribution of the underlying channels, we performed qualitative and quantitative examination of HCN2 and HCN4 expression using fluorescent immunohistochemistry and confocal microscopy. HCN2-immunoreactivity (IR) on the somata of RTN neurons was approximately 10-fold less than that seen in VB neurons while HCN4-IR was detected on the somata of RTN and VB neurons to an equal degree. HCN2-IR in RTN and VB did not overlap with synaptophysin-IR, but strongly colocalized with cortactin-IR, indicating that HCN2 was not present in axon terminals but was present in dendritic spines. Although HCN2-IR in spines was more pronounced in VB than in RTN, the ratio of spinous to somatic expression in RTN was dramatically higher than that in VB, strongly suggesting that HCN2-IR in RTN is principally located in sites distal to the soma. In contrast, HCN4-IR did not colocalize with either synaptophysin or cortactin. The colocalization of HCN2-IR with HCN4-IR was greater in VB than in RTN. The results suggest that the distinct compartmental distribution of HCN2 channels in RTN and VB neurons contributes to the profound differences in the I(h)-dependent properties of these cells.
Collapse
Affiliation(s)
- S Y Abbas
- C. V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Avenue, Room A-1050, New York, NY 10021, USA
| | | | | |
Collapse
|
30
|
Smith PH, Bartlett EL, Kowalkowski A. Unique combination of anatomy and physiology in cells of the rat paralaminar thalamic nuclei adjacent to the medial geniculate body. J Comp Neurol 2006; 496:314-34. [PMID: 16566009 PMCID: PMC2943380 DOI: 10.1002/cne.20913] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The medial geniculate body (MGB) has three major subdivisions, ventral (MGV), dorsal (MGD), and medial (MGM). MGM is linked with paralaminar nuclei that are situated medial and ventral to MGV/MGD. Paralaminar nuclei have unique inputs and outputs compared with MGV and MGD and have been linked to circuitry underlying some important functional roles. We recorded intracellularly from cells in the paralaminar nuclei in vitro. We found that they possess an unusual combination of anatomical and physiological features compared with those reported for "standard" thalamic neurons seen in the MGV/MGD and elsewhere in the thalamus. Compared with MGV/MGD neurons, anatomically, 1) paralaminar cell dendrites can be long, branch sparingly, and encompass a much larger area; 2) their dendrites may be smooth but can have well defined spines; and 3) their axons can have collaterals that branch locally within the same or nearby paralaminar nuclei. When compared with MGV/MGD neurons, physiologically, 1) their spikes are larger in amplitude and can be shorter in duration; 2) their spikes can have dual afterhyperpolarizations with fast and slow components; and 3) they can have a reduction or complete absence of the low-threshold, voltage-sensitive calcium conductance that reduces or eliminates the voltage-dependent burst response. We also recorded from cells in the parafascicular nucleus, a nucleus of the posterior intralaminar nuclear group, because they have unusual anatomical features that are similar to those of some of our paralaminar cells. As with the labeled paralaminar cells, parafascicular cells had physiological features distinguishing them from typical thalamic neurons.
Collapse
Affiliation(s)
- Philip H Smith
- Department of Anatomy, University of Wisconsin, Medical School-Madison, 53706, USA.
| | | | | |
Collapse
|
31
|
Ying SW, Abbas SY, Harrison NL, Goldstein PA. Propofol block of I(h) contributes to the suppression of neuronal excitability and rhythmic burst firing in thalamocortical neurons. Eur J Neurosci 2006; 23:465-80. [PMID: 16420453 DOI: 10.1111/j.1460-9568.2005.04587.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although the depressant effects of the general anesthetic propofol on thalamocortical relay neurons clearly involve gamma-aminobutyric acid (GABA)(A) receptors, other mechanisms may be involved. The hyperpolarization-activated cation current (I(h)) regulates excitability and rhythmic firing in thalamocortical relay neurons in the ventrobasal (VB) complex of the thalamus. Here we investigated the effects of propofol on I(h)-related function in vitro and in vivo. In whole-cell current-clamp recordings from VB neurons in mouse (P23-35) brain slices, propofol markedly reduced the voltage sag and low-threshold rebound excitation that are characteristic of the activation of I(h). In whole-cell voltage-clamp recordings, propofol suppressed the I(h) conductance and slowed the kinetics of activation. The block of I(h) by propofol was associated with decreased regularity and frequency of delta-oscillations in VB neurons. The principal source of the I(h) current in these neurons is the hyperpolarization-activated cyclic nucleotide-gated (HCN) type 2 channel. In human embryonic kidney (HEK)293 cells expressing recombinant mouse HCN2 channels, propofol decreased I(h) and slowed the rate of channel activation. We also investigated whether propofol might have persistent effects on thalamic excitability in the mouse. Three hours following an injection of propofol sufficient to produce loss-of-righting reflex in mice (P35), I(h) was decreased, and this was accompanied by a corresponding decrease in HCN2 and HCN4 immunoreactivity in thalamocortical neurons in vivo. These results suggest that suppression of I(h) may contribute to the inhibition of thalamocortical activity during propofol anesthesia. Longer-term effects represent a novel form of propofol-mediated regulation of I(h).
Collapse
Affiliation(s)
- Shui-Wang Ying
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
32
|
Jia F, Pignataro L, Schofield CM, Yue M, Harrison NL, Goldstein PA. An Extrasynaptic GABAA Receptor Mediates Tonic Inhibition in Thalamic VB Neurons. J Neurophysiol 2005; 94:4491-501. [PMID: 16162835 DOI: 10.1152/jn.00421.2005] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Whole cell patch-clamp recordings were obtained from thalamic ventrobasal (VB) and reticular (RTN) neurons in mouse brain slices. A bicuculline-sensitive tonic current was observed in VB, but not in RTN, neurons; this current was increased by the GABAA receptor agonist 4,5,6,7-tetrahydroisothiazolo-[5,4-c]pyridine-3-ol (THIP; 0.1 μM) and decreased by Zn2+ (50 μM) but was unaffected by zolpidem (0.3 μM) or midazolam (0.2 μM). The pharmacological profile of the tonic current is consistent with its generation by activation of GABAA receptors that do not contain the α1 or γ2 subunits. GABAA receptors expressed in HEK 293 cells that contained α4β2δ subunits showed higher sensitivity to THIP (gaboxadol) and GABA than did receptors made up from α1β2δ, α4β2γ2s, or α1β2γ2s subunits. Western blot analysis revealed that there is little, if any, α3 or α5 subunit protein in VB. In addition, co-immunoprecipitation studies showed that antibodies to the δ subunit could precipitate α4, but not α1 subunit protein. Confocal microscopy of thalamic neurons grown in culture confirmed that α4 and δ subunits are extensively co-localized with one another and are found predominantly, but not exclusively, at extrasynaptic sites. We conclude that thalamic VB neurons express extrasynaptic GABAA receptors that are highly sensitive to GABA and THIP and that these receptors are most likely made up of α4β2δ subunits. In view of the critical role of thalamic neurons in the generation of oscillatory activity associated with sleep, these receptors may represent a principal site of action for the novel hypnotic agent gaboxadol.
Collapse
Affiliation(s)
- Fan Jia
- CV Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College, Cornell University, New York, NY 10021, USA,
| | | | | | | | | | | |
Collapse
|
33
|
Chen X, Shu S, Bayliss DA. Suppression of ih contributes to propofol-induced inhibition of mouse cortical pyramidal neurons. J Neurophysiol 2005; 94:3872-83. [PMID: 16093340 DOI: 10.1152/jn.00389.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The contributions of the hyperpolarization-activated current, I(h), to generation of rhythmic activities are well described for various central neurons, particularly in thalamocortical circuits. In the present study, we investigated effects of a general anesthetic, propofol, on native I(h) in neurons of thalamus and cortex and on the corresponding cloned HCN channel subunits. Whole cell voltage-clamp recordings from mouse brain slices identified neuronal I(h) currents with fast activation kinetics in neocortical pyramidal neurons and with slower kinetics in thalamocortical relay cells. Propofol inhibited the fast-activating I(h) in cortical neurons at a clinically relevant concentration (5 microM); inhibition of I(h) involved a hyperpolarizing shift in half-activation voltage (DeltaV1/2 approximately -9 mV) and a decrease in maximal available current (approximately 36% inhibition, measured at -120 mV). With the slower form of I(h) expressed in thalamocortical neurons, propofol had no effect on current activation or amplitude. In heterologous expression systems, 5 muM propofol caused a large shift in V1/2 and decrease in current amplitude in homomeric HCN1 and linked heteromeric HCN1-HCN2 channels, both of which activate with fast kinetics but did not affect V1/2 or current amplitude of slowly activating homomeric HCN2 channels. With GABA(A) and glycine receptor channels blocked, propofol caused membrane hyperpolarization and suppressed action potential discharge in cortical neurons; these effects were occluded by the I(h) blocker, ZD-7288. In summary, these data indicate that propofol selectively inhibits HCN channels containing HCN1 subunits, such as those that mediate I(h) in cortical pyramidal neurons-and they suggest that anesthetic actions of propofol may involve inhibition of cortical neurons and perhaps other HCN1-expressing cells.
Collapse
Affiliation(s)
- Xiangdong Chen
- Department of Pharmacology, University of Virginia, Charlottesville, 22908-0735, USA.
| | | | | |
Collapse
|
34
|
Ying SW, Goldstein PA. Propofol suppresses synaptic responsiveness of somatosensory relay neurons to excitatory input by potentiating GABA(A) receptor chloride channels. Mol Pain 2005; 1:2. [PMID: 15813991 PMCID: PMC1074352 DOI: 10.1186/1744-8069-1-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 01/14/2005] [Indexed: 11/23/2022] Open
Abstract
Propofol is a widely used intravenous general anesthetic. Propofol-induced unconsciousness in humans is associated with inhibition of thalamic activity evoked by somatosensory stimuli. However, the cellular mechanisms underlying the effects of propofol in thalamic circuits are largely unknown. We investigated the influence of propofol on synaptic responsiveness of thalamocortical relay neurons in the ventrobasal complex (VB) to excitatory input in mouse brain slices, using both current- and voltage-clamp recording techniques. Excitatory responses including EPSP temporal summation and action potential firing were evoked in VB neurons by electrical stimulation of corticothalamic fibers or pharmacological activation of glutamate receptors. Propofol (0.6 - 3 microM) suppressed temporal summation and spike firing in a concentration-dependent manner. The thalamocortical suppression was accompanied by a marked decrease in both EPSP amplitude and input resistance, indicating that a shunting mechanism was involved. The propofol-mediated thalamocortical suppression could be blocked by a GABAA receptor antagonist or chloride channel blocker, suggesting that postsynaptic GABAA receptors in VB neurons were involved in the shunting inhibition. GABAA receptor-mediated inhibitory postsynaptic currents (IPSCs) were evoked in VB neurons by electrical stimulation of the reticular thalamic nucleus. Propofol markedly increased amplitude, decay time, and charge transfer of GABAA IPSCs. The results demonstrated that shunting inhibition of thalamic somatosensory relay neurons by propofol at clinically relevant concentrations is primarily mediated through the potentiation of the GABAA receptor chloride channel-mediated conductance, and such inhibition may contribute to the impaired thalamic responses to sensory stimuli seen during propofol-induced anesthesia.
Collapse
Affiliation(s)
- Shui-Wang Ying
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Avenue, Room A-1050, New York, NY 10021, USA
| | - Peter A Goldstein
- C.V. Starr Laboratory for Molecular Neuropharmacology, Department of Anesthesiology, Weill Medical College of Cornell University, 1300 York Avenue, Room A-1050, New York, NY 10021, USA
| |
Collapse
|