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
|
Chen Y, Yu T, Jiang J. Effects of propofol on the electrophysiological properties of glutamatergic neurons in the ventrolateral medulla of mice. BMC Anesthesiol 2024; 24:432. [PMID: 39604849 PMCID: PMC11600619 DOI: 10.1186/s12871-024-02813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Propofol, a commonly used intravenous anesthetic, is associated with various respiratory adverse events, most notably different degrees of respiratory depression, which pose significant concerns for patient safety. Respiration is a fundamental behavior, with the initiation of breathing in mammals dependent on neuronal activity in the lower brainstem. Previous studies have suggested that propofol-induced respiratory depression might be associated with glutamatergic neurons in the pre-Bötzinger complex (preBötC), though the precise mechanisms are not well understood. In this study, we classify glutamatergic neurons in the brainstem preBötC using whole-cell patch-clamp techniques and investigate the effects of propofol on the electrophysiological properties of these neurons. Our findings aim to shed light on the mechanisms of propofol-induced respiratory depression and provide new experimental insights. METHODS We first employed electrophysiological techniques to classify glutamatergic neurons within the preBötC as Type-1 or Type-2. Following this classification, we applied varying concentrations of propofol through bath application to examine its effects on the electrophysiological properties of each type of glutamatergic neuron. RESULTS We found that Type-1 neurons exhibited a longer latency in excitation, while Type-2 neurons did not show this delayed excitation. On this basis, we further observed that bath application of propofol at concentrations of 5 μM and 10 μM shortened the latency period of Type-1 glutamatergic neurons but did not affect the latency period of Type-2 glutamatergic neurons. CONCLUSION Our study focuses on the glutamatergic neurons in the preBötC of adult mice. It introduces a novel method for classifying these neurons and reveals how propofol affects the activity of the two different types of glutamatergic neurons within the preBötC. These findings contribute to understanding the cellular basis of propofol-induced respiratory depression.
Collapse
Affiliation(s)
- Ya Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China.
| | - Junli Jiang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
3
|
Delgado-Ramírez M, López-Serrano AL, Rodríguez-Menchaca AA. Inhibition of Kv2.1 potassium channels by the antidepressant drug sertraline. Eur J Pharmacol 2024; 970:176487. [PMID: 38458411 DOI: 10.1016/j.ejphar.2024.176487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Sertraline is a commonly used antidepressant of the selective serotonin reuptake inhibitors (SSRIs) class. In this study, we have used the patch-clamp technique to assess the effects of sertraline on Kv2.1 channels heterologously expressed in HEK-293 cells and on the voltage-gated potassium currents (IKv) of Neuro 2a cells, which are predominantly mediated by Kv2.1 channels. Our results reveal that sertraline inhibits Kv2.1 channels in a concentration-dependent manner. The sertraline-induced inhibition was not voltage-dependent and did not require the channels to be open. The kinetics of activation and deactivation were accelerated and decelerated, respectively, by sertraline. Moreover, the inhibition by this drug was use-dependent. Notably, sertraline significantly modified the inactivation mechanism of Kv2.1 channels; the steady-state inactivation was shifted to hyperpolarized potentials, the closed-state inactivation was enhanced and accelerated, and the recovery from inactivation was slowed, suggesting that this is the main mechanism by which sertraline inhibits Kv2.1 channels. Overall, this study provides novel insights into the pharmacological actions of sertraline on Kv2.1 channels, shedding light on the intricate interaction between SSRIs and ion channel function.
Collapse
Affiliation(s)
- Mayra Delgado-Ramírez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78210, Mexico.
| | - Ana Laura López-Serrano
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78210, Mexico
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78210, Mexico
| |
Collapse
|
4
|
Mitchell SJ, Phillips GD, Tench B, Li Y, Belelli D, Martin SJ, Swinny JD, Kelly L, Atack JR, Paradowski M, Lambert JJ. Neurosteroid Modulation of Synaptic and Extrasynaptic GABA A Receptors of the Mouse Nucleus Accumbens. Biomolecules 2024; 14:460. [PMID: 38672476 PMCID: PMC11048561 DOI: 10.3390/biom14040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
The recent approval of formulations of the endogenous neurosteroid allopregnanolone (brexanolone) and the synthetic neuroactive steroid SAGE-217 (zuranolone) to treat postpartum depression (PPD) has encouraged further research to elucidate why these potent enhancers of GABAAR function are clinically effective in this condition. Dopaminergic projections from the ventral tegmental area (VTA) to the nucleus accumbens are associated with reward/motivation and brain imaging studies report that individuals with PPD show reduced activity of this pathway in response to reward and infant engagement. However, the influence of neurosteroids on GABA-ergic transmission in the nucleus accumbens has received limited attention. Here, we investigate, in the medium spiny neurons (MSNs) of the mouse nucleus accumbens core, the effect of allopregnanolone, SAGE-217 and other endogenous and synthetic steroids of interest on fast phasic and tonic inhibition mediated by synaptic (α1/2βγ2) and extrasynaptic (α4βδ) GABAARs, respectively. We present evidence suggesting the resident tonic current results from the spontaneous opening of δ-GABAARs, where the steroid-enhanced tonic current is GABA-dependent. Furthermore, we demonstrate local neurosteroid synthesis in the accumbal slice preparation and reveal that GABA-ergic neurotransmission of MSNs is influenced by an endogenous neurosteroid tone. Given the dramatic fluctuations in allopregnanolone levels during pregnancy and postpartum, this neurosteroid-mediated local fine-tuning of GABAergic transmission in the MSNs will probably be perturbed.
Collapse
Affiliation(s)
- Scott J. Mitchell
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Grant D. Phillips
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Becks Tench
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Yunkai Li
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Delia Belelli
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Stephen J. Martin
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| | - Jerome D. Swinny
- School of Pharmacy & Biomedical Sciences, St. Michael’s Building, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, UK; (J.D.S.); (L.K.)
| | - Louise Kelly
- School of Pharmacy & Biomedical Sciences, St. Michael’s Building, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, UK; (J.D.S.); (L.K.)
| | - John R. Atack
- Main Building, Medicines Discovery Institute, Park Place, Cardiff University, Cardiff, CF10 3AT, UK; (J.R.A.); (M.P.)
| | - Michael Paradowski
- Main Building, Medicines Discovery Institute, Park Place, Cardiff University, Cardiff, CF10 3AT, UK; (J.R.A.); (M.P.)
| | - Jeremy J. Lambert
- Division of Cellular & Systems Medicine, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1 5HL, UK; (S.J.M.); (G.D.P.); (B.T.); (Y.L.); (D.B.); (S.J.M.)
| |
Collapse
|
5
|
Friese MB, Gujral TS, Palanisamy A, Hemmer B, Culley DJ, Crosby G. Anesthetics inhibit phosphorylation of the ribosomal protein S6 in mouse cultured cortical cells and developing brain. Front Aging Neurosci 2023; 15:1060186. [PMID: 37261265 PMCID: PMC10229047 DOI: 10.3389/fnagi.2023.1060186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 04/17/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction The development and maintenance of neural circuits is highly sensitive to neural activity. General anesthetics have profound effects on neural activity and, as such, there is concern that these agents may alter cellular integrity and interfere with brain wiring, such as when exposure occurs during the vulnerable period of brain development. Under those conditions, exposure to anesthetics in clinical use today causes changes in synaptic strength and number, widespread apoptosis, and long-lasting cognitive impairment in a variety of animal models. Remarkably, most anesthetics produce these effects despite having differing receptor mechanisms of action. We hypothesized that anesthetic agents mediate these effects by inducing a shared signaling pathway. Methods We exposed cultured cortical cells to propofol, etomidate, or dexmedetomidine and assessed the protein levels of dozens of signaling molecules and post-translational modifications using reverse phase protein arrays. To probe the role of neural activity, we performed separate control experiments to alter neural activity with non-anesthetics. Having identified anesthetic-induced changes in vitro, we investigated expression of the target proteins in the cortex of sevoflurane anesthetized postnatal day 7 mice by Western blotting. Results All the anesthetic agents tested in vitro reduced phosphorylation of the ribosomal protein S6, an important member of the mTOR signaling pathway. We found a comparable decrease in cortical S6 phosphorylation by Western blotting in sevoflurane anesthetized neonatal mice. Using a systems approach, we determined that propofol, etomidate, dexmedetomidine, and APV/TTX all similarly modulate a signaling module that includes pS6 and other cell mediators of the mTOR-signaling pathway. Discussion Reduction in S6 phosphorylation and subsequent suppression of the mTOR pathway may be a common and novel signaling event that mediates the impact of general anesthetics on neural circuit development.
Collapse
Affiliation(s)
- Matthew B Friese
- Laboratory for Aging Neuroscience, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Taranjit S Gujral
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Arvind Palanisamy
- Laboratory for Aging Neuroscience, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Brittany Hemmer
- Laboratory for Aging Neuroscience, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Deborah J Culley
- Laboratory for Aging Neuroscience, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Gregory Crosby
- Laboratory for Aging Neuroscience, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
6
|
McKinstry-Wu AR, Wasilczuk AZ, Dailey WP, Eckenhoff RG, Kelz MB. In Vivo Photoadduction of Anesthetic Ligands in Mouse Brain Markedly Extends Sedation and Hypnosis. J Neurosci 2023; 43:2338-2348. [PMID: 36849414 PMCID: PMC10072292 DOI: 10.1523/jneurosci.1884-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
Photoaffinity ligands are best known as tools used to identify the specific binding sites of drugs to their molecular targets. However, photoaffinity ligands have the potential to further define critical neuroanatomic targets of drug action. In the brains of WT male mice, we demonstrate the feasibility of using photoaffinity ligands in vivo to prolong anesthesia via targeted yet spatially restricted photoadduction of azi-m-propofol (aziPm), a photoreactive analog of the general anesthetic propofol. Systemic administration of aziPm with bilateral near-ultraviolet photoadduction in the rostral pons, at the border of the parabrachial nucleus and locus coeruleus, produced a 20-fold increase in the duration of sedative and hypnotic effects compared with control mice without UV illumination. Photoadduction that missed the parabrachial-coerulean complex also failed to extend the sedative or hypnotic actions of aziPm and was indistinguishable from nonadducted controls. Paralleling the prolonged behavioral and EEG consequences of on target in vivo photoadduction, we conducted electrophysiologic recordings in rostral pontine brain slices. Using neurons within the locus coeruleus to further highlight the cellular consequences of irreversible aziPm binding, we demonstrate transient slowing of spontaneous action potentials with a brief bath application of aziPm that becomes irreversible on photoadduction. Together, these findings suggest that photochemistry-based strategies are a viable new approach for probing CNS physiology and pathophysiology.SIGNIFICANCE STATEMENT Photoaffinity ligands are drugs capable of light-induced irreversible binding, which have unexploited potential to identify the neuroanatomic sites of drug action. We systemically administer a centrally acting anesthetic photoaffinity ligand in mice, conduct localized photoillumination within the brain to covalently adduct the drug at its in vivo sites of action, and successfully enrich irreversible drug binding within a restricted 250 µm radius. When photoadduction encompassed the pontine parabrachial-coerulean complex, anesthetic sedation and hypnosis was prolonged 20-fold, thus illustrating the power of in vivo photochemistry to help unravel neuronal mechanisms of drug action.
Collapse
Affiliation(s)
- Andrew R McKinstry-Wu
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - William P Dailey
- Department of Chemistry, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania 19104
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Mahoney Institute for Neurosciences, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| |
Collapse
|
7
|
Abstract
BACKGROUND The clinical actions of sugammadex have been well studied, but the detailed molecular mechanism of the drug encapsulation process has not been systematically documented. The hypothesis was that sugammadex would attract rocuronium and vecuronium via interaction with the sugammadex side-chain "tentacles," as previously suggested. METHODS Computational molecular dynamics simulations were done to investigate docking of sugammadex with rocuronium and vecuronium. To validate these methods, strength of binding was assessed between sugammadex and a heterogeneous group of nine other drugs, the binding affinities of which have been experimentally determined. These observations hinted that high concentrations of unbound sugammadex could bind to propofol, potentially altering its pharmacokinetic profile. This was tested experimentally in in vitro cortical slices. RESULTS Sugammadex encapsulation of rocuronium involved a sequential progression down a series of metastable states. After initially binding beside the sugammadex molecule (mean ± SD center-of-mass distance = 1.17 ± 0.13 nm), rocuronium then moved to the opposite side to that hypothesized, where it optimally aligned with the 16 hydroxyl groups (distance, 0.82 ± 0.04 nm) before entering the sugammadex cavity to achieve energetically stable encapsulation by approximately 120 ns (distance, 0.35 ± 0.12 nm). Vecuronium formed fewer hydrogen bonds with sugammadex than did rocuronium; hence, it was less avidly bound. For the other molecules, the computational results showed good agreement with the available experimental data, showing a clear bilogarithmic relation between the relative binding free energy and the association constant (R2 = 0.98). Weaker binding was manifest by periodic unbinding. The brain slice results confirmed the presence of a weak propofol-sugammadex interaction. CONCLUSIONS Computational simulations demonstrate the dynamics of neuromuscular blocking drug encapsulation by sugammadex occurring from the opposite direction to that hypothesized and also how high concentrations of unbound sugammadex can potentially weakly bind to other drugs given during general anesthesia. EDITOR’S PERSPECTIVE
Collapse
|
8
|
Burbanks A, Cerasuolo M, Ronca R, Turner L. A hybrid spatiotemporal model of PCa dynamics and insights into optimal therapeutic strategies. Math Biosci 2023; 355:108940. [PMID: 36400316 DOI: 10.1016/j.mbs.2022.108940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
Using a hybrid cellular automaton with stochastic elements, we investigate the effectiveness of multiple drug therapies on prostate cancer (PCa) growth. The ability of Androgen Deprivation Therapy to reduce PCa growth represents a milestone in prostate cancer treatment, nonetheless most patients eventually become refractory and develop castration-resistant prostate cancer. In recent years, a "second generation" drug called enzalutamide has been used to treat advanced PCa, or patients already exposed to chemotherapy that stopped responding to it. However, tumour resistance to enzalutamide is not well understood, and in this context, preclinical models and in silico experiments (numerical simulations) are key to understanding the mechanisms of resistance and to assessing therapeutic settings that may delay or prevent the onset of resistance. In our mathematical system, we incorporate cell phenotype switching to model the development of increased drug resistance, and consider the effect of the micro-environment dynamics on necrosis and apoptosis of the tumour cells. The therapeutic strategies that we explore include using a single drug (enzalutamide), and drug combinations (enzalutamide and everolimus or cabazitaxel) with different treatment schedules. Our results highlight the effectiveness of alternating therapies, especially alternating enzalutamide and cabazitaxel over a year, and a comparison is made with data taken from TRAMP mice to verify our findings.
Collapse
Affiliation(s)
- Andrew Burbanks
- School of Mathematics and Physics, University of Portsmouth, Lion Gate Building, Lion Terrace, Portsmouth, PO1 3HF, Hampshire, United Kingdom
| | - Marianna Cerasuolo
- School of Mathematics and Physics, University of Portsmouth, Lion Gate Building, Lion Terrace, Portsmouth, PO1 3HF, Hampshire, United Kingdom.
| | - Roberto Ronca
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Leo Turner
- School of Mathematics and Physics, University of Portsmouth, Lion Gate Building, Lion Terrace, Portsmouth, PO1 3HF, Hampshire, United Kingdom
| |
Collapse
|
9
|
The general anaesthetic propofol prevents cerebrocortical potentiation in neocortical mouse brain slices. Brain Res 2022; 1792:148018. [PMID: 35850186 DOI: 10.1016/j.brainres.2022.148018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Propofol is well known to cause amnesia independent of its sedative effect. Memory consolidation processes in the hippocampus have been proposed as a target - however the neural substrates for propofol's amnesic actions remain understudied and poorly described. In particular, the potential role of the cerebral cortex has not been investigated. As an in vitro experimental model of cortical memory consolidation, potentiated cerebral cortex evoked responses were generated in mouse neocortical slices using high frequency (20 Hz) stimulation to layer IV cortical grey matter or subcortical white matter. In separate experiments, slices were pretreated with propofol at two concentrations, 2 µg/mL and 4 µg/mL, to determine the effect of clinically relevant propofol levels on the potentiation response. Only grey matter stimulation induced a significant and lasting increase in cortical evoked potential amplitude in the drug-free condition. Propofol at 2 µg/mL completely inhibited cortical evoked response potentiation, while the 4 µg/mL concentration caused a small but significant depressant effect consequent to the high frequency stimulation. These findings support the hypothesis that propofol disrupts memory consolidation and actively facilitates memory decay in the cerebral cortex. The results further highlight the importance of the cerebral cortex in the early phase of long term memory consolidation.
Collapse
|
10
|
Midazolam at low nanomolar concentrations affects long-term potentiation and synaptic transmission predominantly via the α1-GABAA receptor subunit in mice. Anesthesiology 2022; 136:954-969. [PMID: 35285894 DOI: 10.1097/aln.0000000000004202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Midazolam amplifies synaptic inhibition via different GABAA receptor subtypes defined by the presence of α1, α2, α3 or α5-subunits in the channel complex. Midazolam blocks long-term potentiation and produces postoperative amnesia. The aims of this study were to identify the GABAA receptor subtypes targeted by midazolam responsible for affecting CA1-long-term potentiation and synaptic inhibition in neocortical neurons. METHODS The effects of midazolam on hippocampal CA1-long-term potentiation were studied in acutely prepared brain slices of male and female mice. Positive allosteric modulation on GABAA receptor-mediated miniature inhibitory postsynaptic currents was investigated in organotypic slice cultures of the mouse neocortex. In both experiments, wild-type mice and GABAA receptor knock-in mouse lines were compared in which α1-, α5-, α1/2/3-, α1/3/5- and α2/3/5-GABAA receptor subtypes had been rendered benzodiazepine-insensitive. RESULTS Midazolam 10nM completely blocked long-term potentiation (midazolam mean±SD 98±11%, n=14/8 (slices/mice) vs. control 156±19%, n=20/12; p<0.001). Experiments in slices of α1-, α5-, α1/2/3-, α1/3/5- and α2/3/5-knock-in mice revealed a dominant role for the α1-GABAA receptor subtype in the long-term potentiation suppressing effect.In slices from wild-type mice, midazolam increased (mean±SD) charge transfer of miniature synaptic events concentration-dependently, 50nM: 172±71% (n=10/6) vs. 500nM: 236±54% (n=6/6), p=0.041. In α2/3/5-knock-in mice, charge transfer of miniature synaptic events did not further enhance when applying 500nM midazolam, 50nM: 171±62% (n=8/6) vs. 500nM: 175±62% (n=6/6), p=0.454) indicating two different binding affinities for midazolam to α2/3/5- and α1-subunits. CONCLUSIONS These results demonstrate a predominant role of α1-GABAA receptors in the actions of midazolam at low nanomolar concentrations. At higher concentrations, midazolam also enhances other GABAA receptor subtypes. α1-GABAA receptors may already contribute at sedative doses to the phenomenon of postoperative amnesia that has been reported after midazolam administration.
Collapse
|
11
|
Jang G, MacIver MB. Ketamine Produces a Long-Lasting Enhancement of CA1 Neuron Excitability. Int J Mol Sci 2021; 22:ijms22158091. [PMID: 34360854 PMCID: PMC8347661 DOI: 10.3390/ijms22158091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
Ketamine is a clinical anesthetic and antidepressant. Although ketamine is a known NMDA receptor antagonist, the mechanisms contributing to antidepression are unclear. This present study examined the loci and duration of ketamine’s actions, and the involvement of NMDA receptors. Local field potentials were recorded from the CA1 region of mouse hippocampal slices. Ketamine was tested at antidepressant and anesthetic concentrations. Effects of NMDA receptor antagonists APV and MK-801, GABA receptor antagonist bicuculline, and a potassium channel blocker TEA were also studied. Ketamine decreased population spike amplitudes during application, but a long-lasting increase in amplitudes was seen during washout. Bicuculline reversed the acute effects of ketamine, but the washout increase was not altered. This long-term increase was statistically significant, sustained for >2 h, and involved postsynaptic mechanisms. A similar effect was produced by MK-801, but was only partially evident with APV, demonstrating the importance of the NMDA receptor ion channel block. TEA also produced a lasting excitability increase, indicating a possible involvement of potassium channel block. This is this first report of a long-lasting increase in excitability following ketamine exposure. These results support a growing literature that increased GABA inhibition contributes to ketamine anesthesia, while increased excitatory transmission contributes to its antidepressant effects.
Collapse
|
12
|
Geiger Z, VanVeller B, Lopez Z, Harrata AK, Battani K, Wegman-Points L, Yuan LL. Determination of Diffusion Kinetics of Ketamine in Brain Tissue: Implications for in vitro Mechanistic Studies of Drug Actions. Front Neurosci 2021; 15:678978. [PMID: 34276289 PMCID: PMC8280316 DOI: 10.3389/fnins.2021.678978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/10/2021] [Indexed: 01/26/2023] Open
Abstract
Ketamine has been in use for over 50 years as a general anesthetic, acting primarily through blockade of N-methyl-D-aspartate receptors in the brain. Recent studies have demonstrated that ketamine also acts as a potent and rapid-acting antidepressant when administered at sub-anesthetic doses. However, the precise mechanism behind this effect remains unclear. We examined the diffusion properties of ketamine in brain tissue to determine their effects in in vitro studies related to the antidepressant action of ketamine. Brain slices from adult mice were exposed to artificial cerebrospinal fluid (aCSF) containing ∼17 μM ketamine HCl for varying amounts of time. The amount of ketamine within each slice was then measured by tandem high-performance liquid chromatography - mass spectrometry to characterize the diffusion of ketamine into brain tissue over time. We successfully modeled the diffusion of ketamine into brain tissue using a mono-exponential function with a time constant of τ = 6.59 min. This curve was then compared to a one-dimensional model of diffusion yielding a diffusion coefficient of approximately 0.12 cm2⋅s-1 for ketamine diffusing into brain tissue. The brain:aCSF partition coefficient for ketamine was determined to be approximately 2.76. Our results suggest that the diffusion properties of ketamine have a significant effect on drug concentrations achieved within brain tissue during in vitro experiments. This information is vital to determine the ketamine concentration necessary for in vitro slice preparation to accurately reflect in vivo doses responsible for its antidepressant actions.
Collapse
Affiliation(s)
- Zachary Geiger
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Des Moines University, Des Moines, IA, United States
| | - Brett VanVeller
- Department of Chemistry, Iowa State University, Ames, IA, United States
| | - Zarin Lopez
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Des Moines University, Des Moines, IA, United States
| | - Abdel K. Harrata
- Department of Chemistry, Iowa State University, Ames, IA, United States
| | - Kathryn Battani
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Des Moines University, Des Moines, IA, United States
| | - Lauren Wegman-Points
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Des Moines University, Des Moines, IA, United States
| | - Li-Lian Yuan
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Des Moines University, Des Moines, IA, United States,*Correspondence: Li-Lian Yuan,
| |
Collapse
|
13
|
Voss LJ, Garcia V. Electrophysiological field potential identification of an intact GABAergic system in mouse cortical slices. Brain Res 2021; 1756:147295. [PMID: 33516817 DOI: 10.1016/j.brainres.2021.147295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/10/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
In brain slice experiments there's currently no validated electrophysiological method for differentiating viability between GABAergic and glutamatergic cell populations. Here we investigated the neurophysiology of high frequency field potential activity - and its utility for probing the functional state of the GABAergic system in brain slices. Field potentials were recorded from mouse cortical slices exposed to 50 mM potassium ("elevated-K") and the induced high frequency (>20 Hz) response characterized pharmacologically. The elevated-K responses were also related to the high frequency activity imbedded in no-magnesium seizure-like events (SLE) from the same slices. The elevated-K response, comprising a transient burst of high frequency activity, was strongly GABAA-dependent. The size of the high frequency response was reduced by 71% (p = 0.001) by picrotoxin, but not significantly attenuated by either APV or CNQX. High frequency activity embedded in no-magnesium SLEs correlated with the elevated-K response. The success rate for generating an elevated-K response - and high frequency SLE activity - declined rapidly with increasing time since slicing. These findings support the hypothesis that in cortical slices, a functioning synaptic GABAergic system is evidenced by a strong high frequency component to no-magnesium SLE activity - and that the integrity of the GABAergic system degrades quicker than the excitatory glutamatergic system in this preparation.
Collapse
Affiliation(s)
- Logan J Voss
- Anaesthesia Department, Waikato District Health Board, Hamilton, New Zealand.
| | | |
Collapse
|
14
|
Drexler B, Grenz J, Grasshoff C, Antkowiak B. Allopregnanolone Enhances GABAergic Inhibition in Spinal Motor Networks. Int J Mol Sci 2020; 21:ijms21197399. [PMID: 33036451 PMCID: PMC7582554 DOI: 10.3390/ijms21197399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022] Open
Abstract
The neurosteroid allopregnanolone (ALLO) causes unconsciousness by allosteric modulation of γ-aminobutyric acid type A (GABAA) receptors, but its actions on the spinal motor networks are unknown. We are therefore testing the hypothesis that ALLO attenuates the action potential firing of spinal interneurons and motoneurons predominantly via enhancing tonic, but not synaptic GABAergic inhibition. We used video microscopy to assess motoneuron-evoked muscle activity in organotypic slice cultures prepared from the spinal cord and muscle tissue. Furthermore, we monitored GABAA receptor-mediated currents by performing whole-cell voltage-clamp recordings. We found that ALLO (100 nM) reduced the action potential firing of spinal interneurons by 27% and that of α-motoneurons by 33%. The inhibitory effects of the combination of propofol (1 µM) and ALLO on motoneuron-induced muscle contractions were additive. Moreover, ALLO evoked a tonic, GABAA receptor-mediated current (amplitude: 41 pA), without increasing phasic GABAergic transmission. Since we previously showed that at a clinically relevant concentration of 1 µM propofol enhanced phasic, but not tonic GABAergic inhibition, we conclude that ALLO and propofol target distinct subpopulations of GABAA receptors. These findings provide first evidence that the combined application of ALLO and propofol may help to reduce intraoperative movements and undesired side effects that are frequently observed under total intravenous anesthesia.
Collapse
|
15
|
Murphy CA, Raz A, Grady SM, Banks MI. Optogenetic Activation of Afferent Pathways in Brain Slices and Modulation of Responses by Volatile Anesthetics. J Vis Exp 2020. [PMID: 32773759 DOI: 10.3791/61333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Anesthetics influence consciousness in part via their actions on thalamocortical circuits. However, the extent to which volatile anesthetics affect distinct cellular and network components of these circuits remains unclear. Ex vivo brain slices provide a means by which investigators may probe discrete components of complex networks and disentangle potential mechanisms underlying the effects of volatile anesthetics on evoked responses. To isolate potential cell type- and pathway-specific drug effects in brain slices, investigators must be able to independently activate afferent fiber pathways, identify non-overlapping populations of cells, and apply volatile anesthetics to the tissue in aqueous solution. In this protocol, methods to measure optogenetically-evoked responses to two independent afferent pathways to neocortex in ex vivo brain slices are described. Extracellular responses are recorded to assay network activity and targeted whole-cell patch clamp recordings are conducted in somatostatin- and parvalbumin-positive interneurons. Delivery of physiologically relevant concentrations of isoflurane via artificial cerebral spinal fluid to modulate cellular and network responses is described.
Collapse
Affiliation(s)
| | - Aeyal Raz
- Department of Anesthesiology, University of Wisconsin; Department of Anesthesiology, Rambam Health Care Campus, the Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology
| | - Sean M Grady
- Department of Anesthesiology, University of Wisconsin
| | | |
Collapse
|
16
|
A Metabolic Mechanism for Anaesthetic Suppression of Cortical Synaptic Function in Mouse Brain Slices-A Pilot Investigation. Int J Mol Sci 2020; 21:ijms21134703. [PMID: 32630300 PMCID: PMC7370287 DOI: 10.3390/ijms21134703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Regulation of synaptically located ionotropic receptors is thought to be the main mechanism by which anaesthetics cause unconsciousness. An alternative explanation, which has received much less attention, is that of primary anaesthetic disruption of brain metabolism via suppression of mitochondrial proteins. In this pilot study in mouse cortical slices, we investigated the effect of disrupting cellular metabolism on tissue oxygen handling and cortical population seizure-like event (SLE) activity, using the mitochondrial complex I inhibitor rotenone, and compared this to the effects of the general anaesthetics sevoflurane, propofol and ketamine. Rotenone caused an increase in tissue oxygen (98 mmHg to 157 mmHg (p < 0.01)) before any measurable change in SLE activity. Thereafter, tissue oxygen continued to increase and was accompanied by a significant and prolonged reduction in SLE root mean square (RMS) activity (baseline RMS of 1.7 to 0.7 µV, p < 0.001) and SLE frequency (baseline 4.2 to 0.4 events/min, p = 0.001). This temporal sequence of effects was replicated by all three anaesthetic drugs. In conclusion, anaesthetics with differing synaptic receptor mechanisms all effect changes in tissue oxygen handling and cortical network activity, consistent with a common inhibitory effect on mitochondrial function. The temporal sequence suggests that the observed synaptic depression—as seen in anaesthesia—may be secondary to a reduction in cellular metabolic capacity.
Collapse
|
17
|
Buntschu S, Tscherter A, Heidemann M, Streit J. Critical Components for Spontaneous Activity and Rhythm Generation in Spinal Cord Circuits in Culture. Front Cell Neurosci 2020; 14:81. [PMID: 32410961 PMCID: PMC7198714 DOI: 10.3389/fncel.2020.00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/19/2020] [Indexed: 11/23/2022] Open
Abstract
Neuronal excitability contributes to rhythm generation in central pattern generating networks (CPGs). In spinal cord CPGs, such intrinsic excitability partly relies on persistent sodium currents (INaP), whereas respiratory CPGs additionally depend on calcium-activated cation currents (ICAN). Here, we investigated the contributions of INaP and ICAN to spontaneous rhythm generation in neuronal networks of the spinal cord and whether they mainly involve Hb9 neurons. We used cultures of ventral and transverse slices from the E13-14 embryonic rodent lumbar spinal cord on multielectrode arrays (MEAs). All cultures showed spontaneous bursts of network activity. Blocking synaptic excitation with the AMPA receptor antagonist CNQX suppressed spontaneous network bursts and left asynchronous intrinsic activity at about 30% of the electrodes. Such intrinsic activity was completely blocked at all electrodes by both the INaP blocker riluzole as well as by the ICAN blocker flufenamic acid (FFA) and the more specific TRPM4 channel antagonist 9-phenanthrol. All three antagonists also suppressed spontaneous bursting completely and strongly reduced stimulus-evoked bursts. Also, FFA reduced repetitive spiking that was induced in single neurons by injection of depolarizing current pulses to few spikes. Other antagonists of unspecific cation currents or calcium currents had no suppressing effects on either intrinsic activity (gadolinium chloride) or spontaneous bursting (the TRPC channel antagonists clemizole and ML204 and the T channel antagonist TTA-P2). Combined patch-clamp and MEA recordings showed that Hb9 interneurons were activated by network bursts but could not initiate them. Together these findings suggest that both INaP through Na+-channels and ICAN through putative TRPM4 channels contribute to spontaneous intrinsic and repetitive spiking in spinal cord neurons and thereby to the generation of network bursts.
Collapse
Affiliation(s)
| | | | | | - Jürg Streit
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Drexler B, Seeger T, Worek F, Thiermann H, Antkowiak B, Grasshoff C. Impact of soman and acetylcholine on the effects of propofol in cultured cortical networks. Toxicol Lett 2020; 322:98-103. [PMID: 31954869 DOI: 10.1016/j.toxlet.2020.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 11/29/2022]
Abstract
Patients intoxicated with organophosphorous compounds may need general anaesthesia to enable mechanical ventilation or for control of epileptiform seizures. It is well known that cholinergic overstimulation attenuates the efficacy of general anaesthetics to reduce spontaneous network activity in the cortex. However, it is not clear how propofol, the most frequently used intravenous anaesthetic today, is affected. Here, we investigated the effects of cholinergic overstimulation induced by soman and acetylcholine on the ability of propofol to depress spontaneous action potential activity in organotypic cortical slices measured by extracellular voltage recordings. Cholinergic overstimulation by co-application of soman and acetylcholine (10 μM each) did not reduce the relative inhibition of propofol (1.0 μM; mean normalized action potential firing rate 0.49 ± 0.06 of control condition, p < 0.001, Wilcoxon signed rank test) but clearly reduced its efficacy. Co-application of atropine (10 nM) did not improve the efficacy. Propofol preserved its relative inhibitory potential but did not produce a degree of neuronal depression which can be expected to assure hypnosis in humans. Since a combination with atropine did not improve its efficacy, an increase in dosage will probably be necessary when propofol is used in victims suffering from organophosphorous intoxication.
Collapse
Affiliation(s)
- Berthold Drexler
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany.
| | - Thomas Seeger
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Bernd Antkowiak
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany; Werner Reichardt Center for Integrative Neuroscience, Eberhard-Karls-University, Tuebingen, Germany.
| | - Christian Grasshoff
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany.
| |
Collapse
|
19
|
Tesic V, Joksimovic SM, Quillinan N, Krishnan K, Covey DF, Todorovic SM, Jevtovic-Todorovic V. Neuroactive steroids alphaxalone and CDNC24 are effective hypnotics and potentiators of GABA A currents, but are not neurotoxic to the developing rat brain. Br J Anaesth 2020; 124:603-613. [PMID: 32151384 DOI: 10.1016/j.bja.2020.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 01/01/2020] [Accepted: 01/20/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The most currently used general anaesthetics are potent potentiators of γ-aminobutyric acid A (GABAA) receptors and are invariably neurotoxic during the early stages of brain development in preclinical animal models. As causality between GABAA potentiation and anaesthetic-induced developmental neurotoxicity has not been established, the question remains whether GABAergic activity is crucial for promoting/enhancing neurotoxicity. Using the neurosteroid analogue, (3α,5α)-3-hydroxy-13,24-cyclo-18,21-dinorchol-22-en-24-ol (CDNC24), which potentiates recombinant GABAA receptors, we examined whether this potentiation is the driving force in inducing neurotoxicity during development. METHODS The neurotoxic potential of CDNC24 was examined vis-à-vis propofol (2,6-diisopropylphenol) and alphaxalone (5α-pregnan-3α-ol-11,20-dione) at the peak of rat synaptogenesis. In addition to the morphological neurotoxicity studies of the subiculum and medial prefrontal cortex (mPFC), we assessed the extra-, pre-, and postsynaptic effects of these agents on GABAergic neurotransmission in acute subicular slices from rat pups. RESULTS CDNC24, like alphaxalone and propofol, caused dose-dependent hypnosis in vivo, with a higher therapeutic index. CDNC24 and alphaxalone, unlike propofol, did not cause developmental neuroapoptosis in the subiculum and mPFC. Propofol potentiated post- and extrasynaptic GABAA currents as evidenced by increased spontaneous inhibitory postsynaptic current (sIPSC) decay time and prominent tonic currents, respectively. CDNC24 and alphaxalone had a similar postsynaptic effect, but also displayed a strong presynaptic effect as evidenced by decreased frequency of sIPSCs and induced moderate tonic currents. CONCLUSIONS The lack of neurotoxicity of CDNC24 and alphaxalone may be at least partly related to suppression of presynaptic GABA release in the developing brain.
Collapse
Affiliation(s)
- Vesna Tesic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Srdjan M Joksimovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathiresan Krishnan
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, MO, USA
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
20
|
Kreuzer M, García PS, Brucklacher-Waldert V, Claassen R, Schneider G, Antkowiak B, Drexler B. Diazepam and ethanol differently modulate neuronal activity in organotypic cortical cultures. BMC Neurosci 2019; 20:58. [PMID: 31823754 PMCID: PMC6902402 DOI: 10.1186/s12868-019-0540-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/24/2019] [Indexed: 12/17/2022] Open
Abstract
Background The pharmacodynamic results of diazepam and ethanol administration are similar, in that each can mediate amnestic and sedative-hypnotic effects. Although each of these molecules effectively reduce the activity of central neurons, diazepam does so through modulation of a more specific set of receptor targets (GABAA receptors containing a γ-subunit), while alcohol is less selective in its receptor bioactivity. Our investigation focuses on divergent actions of diazepam and ethanol on the firing patterns of cultured cortical neurons. Method We used electrophysiological recordings from organotypic slice cultures derived from Sprague–Dawley rat neocortex. We exposed these cultures to either diazepam (15 and 30 µM, n = 7) or ethanol (30 and 60 mM, n = 11) and recorded the electrical activity at baseline and experimental conditions. For analysis, we extracted the episodes of spontaneous activity, i.e., cortical up-states. After separation of action potential and local field potential (LFP) activity, we looked at differences in the number of action potentials, in the spectral power of the LFP, as well as in the coupling between action potential and LFP phase. Results While both substances seem to decrease neocortical action potential firing in a not significantly different (p = 0.659, Mann–Whitney U) fashion, diazepam increases the spectral power of the up-state without significantly impacting the spectral composition, whereas ethanol does not significantly change the spectral power but the oscillatory architecture of the up-state as revealed by the Friedman test with Bonferroni correction (p < 0.05). Further, the action potential to LFP-phase coupling reveals a synchronizing effect of diazepam for a wide frequency range and a narrow-band de-synchronizing effect for ethanol (p < 0.05, Kolmogorov–Smirnov test). Conclusion Diazepam and ethanol, induce specific patterns of network depressant actions. Diazepam induces cortical network inhibition and increased synchronicity via gamma subunit containing GABAA receptors. Ethanol also induces cortical network inhibition, but without an increase in synchronicity via a wider span of molecular targets.
Collapse
Affiliation(s)
- Matthias Kreuzer
- Department of Anesthesiology and Intensive Care, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Paul S García
- Department of Anesthesiology, Neuroanesthesia Division, Columbia University Medical Center, New York Presbyterian Hospital, New York, USA
| | - Verena Brucklacher-Waldert
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Horizon Discovery, 8100 Cambridge Research Park, Waterbeach, Cambridge, CB25 9TL, UK
| | - Rebecca Claassen
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Psychiatrie-Zentrum Linthgebiet, Standort Rapperswil, Untere Bahnhofstrasse 11, 8640, Rapperswil, Switzerland
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Bernd Antkowiak
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Werner Reichardt Center for Integrative Neuroscience, Tübingen, Germany
| | - Berthold Drexler
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
21
|
Cayla NS, Dagne BA, Wu Y, Lu Y, Rodriguez L, Davies DL, Gross ER, Heifets BD, Davies MF, MacIver MB, Bertaccini EJ. A newly developed anesthetic based on a unique chemical core. Proc Natl Acad Sci U S A 2019; 116:15706-15715. [PMID: 31308218 PMCID: PMC6681746 DOI: 10.1073/pnas.1822076116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Intravenous anesthetic agents are associated with cardiovascular instability and poorly tolerated in patients with cardiovascular disease, trauma, or acute systemic illness. We hypothesized that a new class of intravenous (IV) anesthetic molecules that is highly selective for the slow type of γ-aminobutyric acid type A receptor (GABAAR) could have potent anesthetic efficacy with limited cardiovascular effects. Through in silico screening using our GABAAR model, we identified a class of lead compounds that are N-arylpyrrole derivatives. Electrophysiological analyses using both an in vitro expression system and intact rodent hippocampal brain slice recordings demonstrate a GABAAR-mediated mechanism. In vivo experiments also demonstrate overt anesthetic activity in both tadpoles and rats with a potency slightly greater than that of propofol. Unlike the clinically approved GABAergic anesthetic etomidate, the chemical structure of our N-arylpyrrole derivative is devoid of the chemical moieties producing adrenal suppression. Our class of compounds also shows minimal to no suppression of blood pressure, in marked contrast to the hemodynamic effects of propofol. These compounds are derived from chemical structures not previously associated with anesthesia and demonstrate that selective targeting of GABAAR-slow subtypes may eliminate the hemodynamic side effects associated with conventional IV anesthetics.
Collapse
Affiliation(s)
- Noëlie S Cayla
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Beza A Dagne
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Yun Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Yao Lu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Larry Rodriguez
- Department of Molecular Pharmacology and Toxicology, University of Southern California School of Pharmacy, Los Angeles, CA 90089
| | - Daryl L Davies
- Department of Molecular Pharmacology and Toxicology, University of Southern California School of Pharmacy, Los Angeles, CA 90089
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - M Frances Davies
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Department of Anesthesia, Palo Alto VA Health Care System, Palo Alto, CA 94304
| | - M Bruce MacIver
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Edward J Bertaccini
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305;
- Department of Anesthesia, Palo Alto VA Health Care System, Palo Alto, CA 94304
| |
Collapse
|
22
|
Bonifazi A, Yano H, Guerrero AM, Kumar V, Hoffman AF, Lupica CR, Shi L, Newman AH. Novel and Potent Dopamine D 2 Receptor Go-Protein Biased Agonists. ACS Pharmacol Transl Sci 2019; 2:52-65. [PMID: 30775693 PMCID: PMC6371206 DOI: 10.1021/acsptsci.8b00060] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 12/18/2022]
Abstract
![]()
The
discovery of functionally biased and physiologically beneficial
ligands directed toward G-protein coupled receptors (GPCRs) has provided
the impetus to design dopamine D2 receptor (D2R) targeted molecules that may be therapeutically advantageous for
the treatment of certain neuropsychiatric or basal ganglia related
disorders. Here we describe the synthesis of a novel series of D2R agonists linking the D2R unbiased agonist sumanirole
with privileged secondary molecular fragments. The resulting ligands
demonstrate improved D2R affinity and selectivity over
sumanirole. Extensive in vitro functional studies
and bias factor analysis led to the identification of a novel class
of highly potent Go-protein biased full D2R agonists with
more than 10-fold and 1000-fold bias selectivity toward activation
of specific G-protein subtypes and β-arrestin, respectively.
Intracellular electrophysiological recordings from midbrain dopamine
neurons demonstrated that Go-protein selective agonists can elicit
prolonged ligand-induced GIRK activity via D2Rs, which
may be beneficial in the treatment of dyskinesias associated with
dopamine system dysfunction.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Hideaki Yano
- Computational Chemistry and Molecular Biophysics Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Adrian M Guerrero
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Vivek Kumar
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Alexander F Hoffman
- Electrophysiology Research Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Carl R Lupica
- Electrophysiology Research Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
23
|
Neumann E, Rudolph U, Knutson DE, Li G, Cook JM, Hentschke H, Antkowiak B, Drexler B. Zolpidem Activation of Alpha 1-Containing GABA A Receptors Selectively Inhibits High Frequency Action Potential Firing of Cortical Neurons. Front Pharmacol 2019; 9:1523. [PMID: 30687091 PMCID: PMC6333667 DOI: 10.3389/fphar.2018.01523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 12/12/2018] [Indexed: 11/13/2022] Open
Abstract
Introduction: High frequency neuronal activity in the cerebral cortex can be induced by noxious stimulation during surgery, brain injury or poisoning. In this scenario, it is essential to block cortical hyperactivity to protect the brain against damage, e.g., by using drugs that act as positive allosteric modulators at GABAA receptors. Yet, cortical neurons express multiple, functionally distinct GABAA receptor subtypes. Currently there is a lack of knowledge which GABAA receptor subtypes would be a good pharmacological target to reduce extensive cortical activity. Methods: Spontaneous action potential activity was monitored by performing extracellular recordings from organotypic neocortical slice cultures of wild type and GABAAR-α1(H101R) mutant mice. Phases of high neuronal activity were characterized using peri-event time histograms. Drug effects on within-up state firing rates were quantified via Hedges' g. Results: We quantified the effects of zolpidem, a positive modulator of GABAA receptors harboring α1-subunits, and the experimental benzodiazepine SH-053-2'F-S-CH3, which preferably acts at α2/3/5- but spares α1-subunits. Both agents decreased spontaneous action potential activity but altered the firing patterns in different ways. Zolpidem reduced action potential firing during highly active network states. This action was abolished by flumazenil, suggesting that it was mediated by benzodiazepine-sensitive GABAA receptors. SH-053-2'F-S-CH3 also attenuated neuronal activity, but unlike zolpidem, failed to reduce high frequency firing. To confirm that zolpidem actions were indeed mediated via α1-dependent actions, it was evaluated in slices from wild type and α(H101R) knock-in mice. Inhibition of high frequency action potential firing was observed in slices from wild type but not mutant mice. Conclusion: Our results suggest that during episodes of scarce and high neuronal activity action potential firing of cortical neurons is controlled by different GABAA receptor subtypes. Exaggerated firing of cortical neurons is reduced by positive modulation of α1-, but not α2/3/5-subunit containing GABAA receptors.
Collapse
Affiliation(s)
- Elena Neumann
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Daniel E Knutson
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Guanguan Li
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Harald Hentschke
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Bernd Antkowiak
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care, Eberhard Karls Universität Tübingen, Tübingen, Germany.,Werner Reichardt Center for Integrative Neuroscience, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Berthold Drexler
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
24
|
Wang JB, Aryal M, Zhong Q, Vyas DB, Airan RD. Noninvasive Ultrasonic Drug Uncaging Maps Whole-Brain Functional Networks. Neuron 2018; 100:728-738.e7. [PMID: 30408444 PMCID: PMC6274638 DOI: 10.1016/j.neuron.2018.10.042] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/13/2018] [Accepted: 10/24/2018] [Indexed: 01/06/2023]
Abstract
Being able to noninvasively modulate brain activity, where and when an experimenter desires, with an immediate path toward human translation is a long-standing goal for neuroscience. To enable robust perturbation of brain activity while leveraging the ability of focused ultrasound to deliver energy to any point of the brain noninvasively, we have developed biocompatible and clinically translatable nanoparticles that allow ultrasound-induced uncaging of neuromodulatory drugs. Utilizing the anesthetic propofol, together with electrophysiological and imaging assays, we show that the neuromodulatory effect of ultrasonic drug uncaging is limited spatially and temporally by the size of the ultrasound focus, the sonication timing, and the pharmacokinetics of the uncaged drug. Moreover, we see secondary effects in brain regions anatomically distinct from and functionally connected to the sonicated region, indicating that ultrasonic drug uncaging could noninvasively map the changes in functional network connectivity associated with pharmacologic action at a particular brain target.
Collapse
Affiliation(s)
- Jeffrey B Wang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Muna Aryal
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Qian Zhong
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Daivik B Vyas
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA
| | - Raag D Airan
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Aminophylline and Ephedrine, but Not Flumazenil, Inhibit the Activity of the Excitatory Amino Acid Transporter 3 Expressed in Xenopus Oocytes and Reverse the Increased Activity by Propofol. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6817932. [PMID: 29888272 PMCID: PMC5985076 DOI: 10.1155/2018/6817932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 11/18/2022]
Abstract
We investigated the effects of flumazenil, aminophylline, and ephedrine on the excitatory amino acid transporter type 3 (EAAT3) activity and the interaction with propofol. EAAT3 was expressed in the Xenopus oocytes. L-Glutamate-induced membrane currents were measured using the two-electrode voltage clamp at various drug concentrations. Oocytes were preincubated with protein kinase C- (PKC-) activator, or inhibitor, and phosphatidylinositol 3-kinase (PI3K) inhibitor. To study the interaction with propofol, oocytes were exposed to propofol, propofol + aminophylline, or ephedrine. Aminophylline and ephedrine significantly decreased EAAT3 activity. Aminophylline (95 μM) and ephedrine (1.19 μM) significantly decreased Vmax, but not Km of EAAT3, for glutamate. The phorbol 12-myristate-13-acetate-induced increase in EAAT3 activity was abolished by aminophylline or ephedrine. The decreased EAAT3 activities by PKC inhibitors (staurosporine, chelerythrine) and PI3K inhibitor (wortmannin) were not significantly different from those by aminophylline or ephedrine, as well as those by PKC inhibitors or PI3K inhibitor + aminophylline or ephedrine. The enhanced EAAT3 activities induced by propofol were significantly abolished by aminophylline or ephedrine. Aminophylline and ephedrine inhibit EAAT3 activity via PKC and PI3K pathways and abolish the increased EAAT3 activity by propofol. Our results indicate a novel site of action for aminophylline and ephedrine.
Collapse
|
26
|
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
|
27
|
Differential depression of neuronal network activity by midazolam and its main metabolite 1-hydroxymidazolam in cultured neocortical slices. Sci Rep 2017; 7:3503. [PMID: 28615640 PMCID: PMC5471240 DOI: 10.1038/s41598-017-03154-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 04/25/2017] [Indexed: 11/12/2022] Open
Abstract
The benzodiazepine midazolam is widely used in critical care medicine. Midazolam has a clinically active metabolite, 1-hydroxymidazolam. The contribution of 1-hydroxymidazolam to the effects of midazolam is controversial. The aim of the current study was to compare the actions of midazolam and 1-hydroxymidazolam on network activity of cortical neurons. Midazolam depressed neuronal activity at a low concentration of 5 nM. When midazolam concentration was increased, it depressed neuronal discharge rates in a biphasic manner. In comparison, 1-hydroxymidazolam did not depress the cortical network activity at low nanomolar concentrations. Higher concentrations of 1-hydroxymidazolam consistently inhibited neuronal activity. Moreover, midazolam shortened cortical up states at low, but not at high concentrations, while the opposite effect was observed with 1-hydroxymidazolam. The network depressant action of midazolam at low concentrations was absent in slices from GABAA receptor α1(H101R)mutant mice. The α1(H101R)mutation renders α1-subunit containing GABAA receptors insensitive towards benzodiazepines. This GABAA receptor subtype is thought to mediate sedation. As midazolam is more potent than its metabolite 1-hydroxymidazolam, the major clinical effects are thus likely caused by midazolam itself. However, 1-hydroxymidazolam could add to the effects of midazolam, especially after the application of high doses of midazolam, and in case of impaired drug metabolism.
Collapse
|
28
|
Puskarjov M, Fiumelli H, Briner A, Bodogan T, Demeter K, Lacoh CM, Mavrovic M, Blaesse P, Kaila K, Vutskits L. K-Cl Cotransporter 2-mediated Cl- Extrusion Determines Developmental Stage-dependent Impact of Propofol Anesthesia on Dendritic Spines. Anesthesiology 2017; 126:855-867. [PMID: 28301408 DOI: 10.1097/aln.0000000000001587] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND General anesthetics potentiating γ-aminobutyric acid (GABA)-mediated signaling are known to induce a persistent decrement in excitatory synapse number in the cerebral cortex when applied during early postnatal development, while an opposite action is produced at later stages. Here, the authors test the hypothesis that the effect of general anesthetics on synaptogenesis depends upon the efficacy of GABA receptor type A (GABAA)-mediated inhibition controlled by the developmental up-regulation of the potassium-chloride (K-Cl) cotransporter 2 (KCC2). METHODS In utero electroporation of KCC2 was used to prematurely increase the efficacy of (GABAA)-mediated inhibition in layer 2/3 pyramidal neurons in the immature rat somatosensory cortex. Parallel experiments with expression of the inward-rectifier potassium channel Kir2.1 were done to reduce intrinsic neuronal excitability. The effects of these genetic manipulations (n = 3 to 4 animals per experimental group) were evaluated using iontophoretic injection of Lucifer Yellow (n = 8 to 12 cells per animal). The total number of spines analyzed per group ranged between 907 and 3,371. RESULTS The authors found a robust effect of the developmental up-regulation of KCC2-mediated Cl transport on the age-dependent action of propofol on dendritic spines. Premature expression of KCC2, unlike expression of a transport-inactive KCC2 variant, prevented a propofol-induced decrease in spine density. In line with a reduction in neuronal excitability, the above result was qualitatively replicated by overexpression of Kir2.1. CONCLUSIONS The KCC2-dependent developmental increase in the efficacy of GABAA-mediated inhibition is a major determinant of the age-dependent actions of propofol on dendritic spinogenesis.
Collapse
Affiliation(s)
- Martin Puskarjov
- From the Department of Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland (M.P., M.M., P.B., K.K.); Division of Biological and Environmental Sciences and Technology, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia (H.F.); Department of Anesthesiology, Pharmacology and Intensive Care, University Hospital of Geneva, Geneva, Switzerland (A.B., T.B., C.-M.L., L.V.); Department of Fundamental Neurosciences, University of Geneva Medical School, Geneva, Switzerland (A.B., T.B., K.D., L.V.); and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (K.D.). Current position: Institute of Physiology I, Westfälische Wilhelms-University Münster, Münster, Germany (P.B.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kaur J, Flores Gutiérrez J, Nistri A. Neuroprotective effect of propofol against excitotoxic injury to locomotor networks of the rat spinal cord in vitro. Eur J Neurosci 2016; 44:2418-2430. [PMID: 27468970 DOI: 10.1111/ejn.13353] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/11/2016] [Indexed: 01/09/2023]
Abstract
Although neuroprotection to contain the initial damage of spinal cord injury (SCI) is difficult, multicentre studies show that early neurosurgery under general anaesthesia confers positive benefits. An interesting hypothesis is that the general anaesthetic itself might largely contribute to neuroprotection, although in vivo clinical settings hamper studying this possibility directly. To further test neuroprotective effects of a widely used general anaesthetic, we studied if propofol could change the outcome of a rat isolated spinal cord SCI model involving excitotoxicity evoked by 1 h application of kainate with delayed consequences on neurons and locomotor network activity. Propofol (5 μm; 4-8 h) enhanced responses to GABA and depressed those to NMDA together with decrease in polysynaptic reflexes that partly recovered after 1 day washout. Fictive locomotion induced by dorsal root stimuli or NMDA and serotonin was weaker the day after propofol application. Kainate elicited a significant loss of spinal neurons, especially motoneurons, whose number was halved. When propofol was applied for 4-8 h after kainate washout, strong neuroprotection was observed in all spinal areas, including attenuation of motoneuron loss. Although propofol had minimal impact on recovery of electrophysiological characteristics 24 h later, it did not further depress network activity. A significant improvement in disinhibited burst periodicity suggested potential to ameliorate neuronal excitability in analogy to histological data. Functional recovery of locomotor networks perhaps required longer time due to the combined action of excitotoxicity and anaesthetic depression at 24 h. These results suggest propofol could confer good neuroprotection to spinal circuits during experimental SCI.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Neuroscience Department, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Javier Flores Gutiérrez
- Neuroscience Department, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Andrea Nistri
- Neuroscience Department, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy. .,SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione, Udine, Italy.
| |
Collapse
|
30
|
Humble SR. Neurosteroids are reduced in diabetic neuropathy and may be associated with the development of neuropathic pain. F1000Res 2016; 5:1923. [PMID: 28357038 PMCID: PMC5345788 DOI: 10.12688/f1000research.9034.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2016] [Indexed: 01/10/2023] Open
Abstract
Introduction: Peripheral and central sensitisation are implicated in the development of neuropathic pain. Hypersensitivity of pain pathway neurons has been described in animal models of diabetic neuropathy, which is postulated to be related to an imbalance between inhibitory and excitatory signals within the spinal cord. GABAergic neurons within the pain pathway are vital for the transmission of painful stimuli to higher centres. A developmental change in the rate of exponential decay of GABAergic synaptic events has been observed in other types of neurons and this may be associated with fluctuations in endogenous neurosteroid tone. Methods: The whole-cell patch-clamp technique was used on slices of neural tissue. Electrophysiological recordings were obtained from wild type mice between the ages of 6 and 80 days in the spinal cord, the nucleus reticularis of the thalamus and the cerebral cortex. Recordings were also obtained from mice with diabetic neuropathy (ob/ob and db/db) between the ages of 60 and 80 days. Behavioural experiments were performed to examine mechanical and thermal nociception. Results: Electrophysiological recordings from cortical pain pathway neurons from mature type-2 diabetic mice revealed that the endogenous neurosteroid tone is reduced compared to control. However, selected neurosteroid compounds had a more pronounced effect on the GABA
A receptors of these diabetic mice. ob/ob mice exhibit mechanical hyperalgesia and allodynia, which was reduced by neurosteroids applied exogenously. Conclusions: The reduced endogenous neurosteroid tone in ob/ob mice may be linked to their hypersensitivity. Neurosteroids may exert analgesic effects in pathological pain states by attempting to restore the physiological GABAergic inhibitory tone.
Collapse
Affiliation(s)
- Stephen R Humble
- Department of Anaesthetics and Pain Management, Charing Cross Hospital, Imperial College NHS Healthcare Trust London, London, W6 8RF, UK
| |
Collapse
|
31
|
Sleep Homeostasis and General Anesthesia: Are Fruit Flies Well Rested after Emergence from Propofol? Anesthesiology 2016; 124:404-16. [PMID: 26556728 DOI: 10.1097/aln.0000000000000939] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Shared neurophysiologic features between sleep and anesthetic-induced hypnosis indicate a potential overlap in neuronal circuitry underlying both states. Previous studies in rodents indicate that preexisting sleep debt discharges under propofol anesthesia. The authors explored the hypothesis that propofol anesthesia also dispels sleep pressure in the fruit fly. To the authors' knowledge, this constitutes the first time propofol has been tested in the genetically tractable model, Drosophila melanogaster. METHODS Daily sleep was measured in Drosophila by using a standard locomotor activity assay. Propofol was administered by transferring flies onto food containing various doses of propofol or equivalent concentrations of vehicle. High-performance liquid chromatography was used to measure the tissue concentrations of ingested propofol. To determine whether propofol anesthesia substitutes for natural sleep, the flies were subjected to 10-h sleep deprivation (SD), followed by 6-h propofol exposure, and monitored for subsequent sleep. RESULTS Oral propofol treatment causes anesthesia in flies as indicated by a dose-dependent reduction in locomotor activity (n = 11 to 41 flies from each group) and increased arousal threshold (n = 79 to 137). Recovery sleep in flies fed propofol after SD was delayed until after flies had emerged from anesthesia (n = 30 to 48). SD was also associated with a significant increase in mortality in propofol-fed flies (n = 44 to 46). CONCLUSIONS Together, these data indicate that fruit flies are effectively anesthetized by ingestion of propofol and suggest that homologous molecular and neuronal targets of propofol are conserved in Drosophila. However, behavioral measurements indicate that propofol anesthesia does not satisfy the homeostatic need for sleep and may compromise the restorative properties of sleep.
Collapse
|
32
|
Propofol postsynaptically suppresses stellate neuron excitability in the entorhinal cortex by influencing the HCN and TREK-2 channels. Neurosci Lett 2016; 619:54-9. [DOI: 10.1016/j.neulet.2016.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 11/22/2022]
|
33
|
Weimer I, Worek F, Seeger T, Thiermann H, Grasshoff C, Antkowiak B, Balk M. Anesthetic actions of thiopental remain largely unaffected during cholinergic overstimulation in cultured cortical networks. Toxicol Lett 2016; 244:129-135. [PMID: 26325215 DOI: 10.1016/j.toxlet.2015.08.1106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/28/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
In case of military or terrorist use of organophosphorus (OP) compounds victims are likely to suffer from not only intoxication but physical trauma as well. Appropriate emergency care may therefore include general anesthesia to allow life-saving surgical intervention. Since there is evidence that drug potency and efficacy of several anesthetics are attenuated by high concentrations of acetylcholine in the CNS, this study was designed to evaluate the anesthetic actions of thiopental during cholinergic overstimulation. Making use of organotypic slice cultures derived from the mouse neocortex, drug effects were assessed by extracellular voltage recordings of network activity at basal cholinergic tone and during simulated cholinergic crisis (high cholinergic tone). The latter was achieved by inhibition of acetylcholinesterases via soman and an ambient acetylcholine concentration of 10μM. The induction of cholinergic crisis in vitro increased the network activity of cortical neurons significantly. Surprisingly, differences in network activity between basal and high cholinergic tone became less pronounced with rising concentrations of thiopental and drug potency and efficacy were almost equivalent. These results clearly distinguish thiopental from previously tested general anesthetics and make it a promising candidate for in vivo studies to identify suitable anesthetics for victims of OP intoxication.
Collapse
Affiliation(s)
- Isabel Weimer
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany; Department of Anaesthesiology, Experimental Anaesthesiology Section, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany.
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Thomas Seeger
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Christian Grasshoff
- Department of Anaesthesiology, Experimental Anaesthesiology Section, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany
| | - Bernd Antkowiak
- Department of Anaesthesiology, Experimental Anaesthesiology Section, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany; Werner-Reichardt-Centre for Integrative Neuroscience, Eberhard-Karls-University, 72076 Tuebingen, Germany
| | - Monika Balk
- Department of Anaesthesiology, Experimental Anaesthesiology Section, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany
| |
Collapse
|
34
|
Brown AR, Mitchell SJ, Peden DR, Herd MB, Seifi M, Swinny JD, Belelli D, Lambert JJ. During postnatal development endogenous neurosteroids influence GABA-ergic neurotransmission of mouse cortical neurons. Neuropharmacology 2015; 103:163-73. [PMID: 26626485 PMCID: PMC4764649 DOI: 10.1016/j.neuropharm.2015.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 11/11/2015] [Accepted: 11/22/2015] [Indexed: 12/11/2022]
Abstract
As neuronal development progresses, GABAergic synaptic transmission undergoes a defined program of reconfiguration. For example, GABAA receptor (GABAAR)-mediated synaptic currents, (miniature inhibitory postsynaptic currents; mIPSCs), which initially exhibit a relatively slow decay phase, become progressively reduced in duration, thereby supporting the temporal resolution required for mature network activity. Here we report that during postnatal development of cortical layer 2/3 pyramidal neurons, GABAAR-mediated phasic inhibition is influenced by a resident neurosteroid tone, which wanes in the second postnatal week, resulting in the brief phasic events characteristic of mature neuronal signalling. Treatment of cortical slices with the immediate precursor of 5α-pregnan-3α-ol-20-one (5α3α), the GABAAR-inactive 5α-dihydroprogesterone, (5α-DHP), greatly prolonged the mIPSCs of P20 pyramidal neurons, demonstrating these more mature neurons retain the capacity to synthesize GABAAR-active neurosteroids, but now lack the endogenous steroid substrate. Previously, such developmental plasticity of phasic inhibition was ascribed to the expression of synaptic GABAARs incorporating the α1 subunit. However, the duration of mIPSCs recorded from L2/3 cortical neurons derived from α1 subunit deleted mice, were similarly under the developmental influence of a neurosteroid tone. In addition to principal cells, synaptic GABAARs of L2/3 interneurons were modulated by native neurosteroids in a development-dependent manner. In summary, local neurosteroids influence synaptic transmission during a crucial period of cortical neurodevelopment, findings which may be of importance for establishing normal network connectivity. Upon postnatal maturation GABAA receptor synaptic inhibition is reduced in duration. Reduced synthesis of local neurosteroids contributes to this cortical plasticity. The study reveals a potent mechanism to locally regulate cortical neuron activity.
Collapse
Affiliation(s)
- Adam R Brown
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Scott J Mitchell
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Dianne R Peden
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Murray B Herd
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Mohsen Seifi
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee, UK.
| |
Collapse
|
35
|
Wakita M, Kotani N, Akaike N. Effects of propofol on glycinergic neurotransmission in a single spinal nerve synapse preparation. Brain Res 2015; 1631:147-56. [PMID: 26616339 DOI: 10.1016/j.brainres.2015.11.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 12/30/2022]
Abstract
The effects of the intravenous anesthetic, propofol, on glycinergic transmission and on glycine receptor-mediated whole-cell currents (IGly) were examined in the substantia gelatinosa (SG) neuronal cell body, mechanically dissociated from the rat spinal cord. This "synaptic bouton" preparation, which retains functional native nerve endings, allowed us to evaluate glycinergic inhibitory postsynaptic currents (IPSCs) and whole-cell currents in a preparation in which experimental solution could rapidly access synaptic terminals. Synaptic IPSCs were measured as spontaneous (s) and evoked (e) IPSCs. The eIPSCs were elicited by applying paired-pulse focal electrical stimulation, while IGly was evoked by a bath application of glycine. A concentration-dependent enhancement of IGly was observed for ≥10µM propofol. Propofol (≥3µM) significantly increased the frequency of sIPSCs and prolonged the decay time without altering the current amplitude. However, propofol (≥3µM) also significantly increased the mean amplitude of eIPSCs and decreased the failure rate (Rf). A decrease in the paired-pulse ratio (PPR) was noted at higher concentrations (≥10µM). The decay time of eIPSCs was prolonged only at the maximum concentration tested (30µM). Propofol thus acts at both presynaptic glycine release machinery and postsynaptic glycine receptors. At clinically relevant concentrations (<1μM) there was no effect on IGly, sIPSCs or eIPSCs suggesting that at anesthetic doses propofol does not affect inhibitory glycinergic synapses in the spinal cord.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, JyuryoGroup, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya 343-0821, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, JyuryoGroup, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya 343-0821, Japan; Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
36
|
Zhou CH, Zhu YZ, Zhao PP, Xu CM, Zhang MX, Huang H, Li J, Liu L, Wu YQ. Propofol Inhibits Lipopolysaccharide-Induced Inflammatory Responses in Spinal Astrocytes via the Toll-Like Receptor 4/MyD88-Dependent Nuclear Factor-κB, Extracellular Signal-Regulated Protein Kinases1/2, and p38 Mitogen-Activated Protein Kinase Pathways. Anesth Analg 2015; 120:1361-8. [PMID: 25695672 DOI: 10.1213/ane.0000000000000645] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In this study, we investigated the effect of propofol, a commonly used IV anesthetic, on lipopolysaccharide (LPS)-induced inflammatory responses in astrocytes and explored the molecular mechanisms by which it occurs. METHODS Astrocytes were stimulated with LPS (1.0 μg/mL) in the absence and presence of different concentrations of propofol. The expression of astrocyte marker glial fibrillary acidic protein (GFAP) in astrocytes was detected using immunofluorescence staining and Western blot analysis. The levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α were measured using an enzyme-linked immunosorbent assay. The mRNA level of Toll-like receptor 4 (TLR4) was determined by semiquantitative reverse transcriptase-polymerase chain reaction. The protein expressions of TLR4, myeloid differentiation factor 88 (MyD88), p- extracellular signal-regulated protein kinases (ERK)1/2, p-c-Jun N-terminal kinase, p-p38 mitogen-activated protein kinase (MAPK), p-I-κBα, I-κBα, and p-nuclear factor-κB (NF-κB)p65 were detected by Western blot. RESULTS Our results show that after stimulation with LPS, the levels of IL-1β, IL-6, and tumor necrosis factor-α and the expression of GFAP in astrocytes were up-regulated significantly. In addition, the expression of TLR4, MyD88, p-ERK1/2, p-c-Jun N-terminal kinase, p-p38 MAPK, and p-NF-κBp65 increased, whereas the expression of total I-κBα decreased upon stimulation with LPS. Propofol (10 μM) reduced the secretion of proinflammatory cytokines, inhibited the expressions of GFAP, TLR4, MyD88, p-ERK1/2, p-p38 MAPK, and p-NF-κBp65 in astrocytes challenged with LPS. CONCLUSIONS In the present study, propofol 10 μM but not lower clinically relevant or higher supra-clinical concentrations attenuated LPS-induced astrocyte activation and subsequent inflammatory responses by inhibiting the TLR4/MyD88-dependent NF-κB, ERK1/2, and p38 MAPK pathways.
Collapse
Affiliation(s)
- Cheng-Hua Zhou
- From the *Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou, PR China; †Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, PR China; and ‡Department of Anesthetic Pharmacology, Xuzhou Medical College, Xuzhou, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kashiwagi M, Osaka Y, Onimaru H, Takeda J. Optical imaging of propofol-induced central respiratory depression in medulla-spinal cord preparations from newborn rats. Clin Exp Pharmacol Physiol 2015; 38:186-91. [PMID: 21251047 DOI: 10.1111/j.1440-1681.2011.05480.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
1. Propofol (2,6-diisopropylphenol) is an intravenous anaesthetic used for the induction and maintenance of general anaesthesia; it also potently and dose-dependently depresses respiration. The aim of the present study was to analyse propofol-induced changes in spatiotemporal patterns of inspiratory-related neural activity and to investigate the involvement of the GABAA receptor by using an optical imaging technique. 2. The brain stems and spinal cords of 0-1-day-old Wistar rats were isolated and stained using a fluorescent voltage-sensitive dye. Neuronal activity in the preparation was detected using an optical recording apparatus containing a charge-coupled device (CCD)-based camera. 3. Bath-applied propofol (7.5 μmol/L) decreased the C4 burst rate to 45.9% of baseline. Although optical signals corresponding to membrane depolarization during the pre-inspiratory phase in the parafacial region of the ventral medulla decreased to 28.7% of baseline following propofol application, those during the inspiratory phase in the caudal part of the rostral ventrolateral medulla did not. 4. The inhibitory effect of bath-applied propofol was reversed by 2 μmol/L bicuculline. 5. Changes in optical signals corresponding to the population activity of pre-inspiratory neurons were parallel to changes in the C4 burst rate. 6. The results suggest that propofol decreases the inspiratory burst rate by reducing the activity of pre-inspiratory neurons and that GABAA receptor activation plays a role in propofol-induced central respiratory depression. These results are consistent with those of previous electrophysiological studies.
Collapse
Affiliation(s)
- Masanori Kashiwagi
- Department of Anaesthesiology, Kitasato Institute Hospital, Kitasato UniversityDepartment of Physiology, Showa University School of MedicineDepartment of Anaesthesiology, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | |
Collapse
|
38
|
Woll KA, Weiser BP, Liang Q, Meng T, McKinstry-Wu A, Pinch B, Dailey WP, Gao WD, Covarrubias M, Eckenhoff RG. Role for the propofol hydroxyl in anesthetic protein target molecular recognition. ACS Chem Neurosci 2015; 6:927-35. [PMID: 25799399 DOI: 10.1021/acschemneuro.5b00078] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Propofol is a widely used intravenous general anesthetic. We synthesized 2-fluoro-1,3-diisopropylbenzene, a compound that we call "fropofol", to directly assess the significance of the propofol 1-hydroxyl for pharmacologically relevant molecular recognition in vitro and for anesthetic efficacy in vivo. Compared to propofol, fropofol had a similar molecular volume and only a small increase in hydrophobicity. Isothermal titration calorimetry and competition assays revealed that fropofol had higher affinity for a protein site governed largely by van der Waals interactions. Within another protein model containing hydrogen bond interactions, propofol demonstrated higher affinity. In vivo, fropofol demonstrated no anesthetic efficacy, but at high concentrations produced excitatory activity in tadpoles and mice; fropofol also antagonized propofol-induced hypnosis. In a propofol protein target that contributes to hypnosis, α1β2γ2L GABAA receptors, fropofol demonstrated no significant effect alone or on propofol positive allosteric modulation of the ion channel, suggesting an additional requirement for the 1-hydroxyl within synaptic GABAA receptor site(s). However, fropofol caused similar adverse cardiovascular effects as propofol by a dose-dependent depression of myocardial contractility. Our results directly implicate the propofol 1-hydroxyl as contributing to molecular recognition within protein targets leading to hypnosis, but not necessarily within protein targets leading to side effects of the drug.
Collapse
Affiliation(s)
| | | | - Qiansheng Liang
- Department
of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 900 Walnut Street, JHN 417, Philadelphia, Pennsylvania 19107, United States
| | - Tao Meng
- Department of Anesthesiology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012 P. R. China
- Department of Anesthesiology
and Critical Care Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, United States
| | | | - Benika Pinch
- Department of Chemistry, University of Pennsylvania School of Arts and Sciences, 231 S. 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - William P. Dailey
- Department of Chemistry, University of Pennsylvania School of Arts and Sciences, 231 S. 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Wei Dong Gao
- Department of Anesthesiology
and Critical Care Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, United States
| | - Manuel Covarrubias
- Department
of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 900 Walnut Street, JHN 417, Philadelphia, Pennsylvania 19107, United States
| | | |
Collapse
|
39
|
Brown AR, Herd MB, Belelli D, Lambert JJ. Developmentally regulated neurosteroid synthesis enhances GABAergic neurotransmission in mouse thalamocortical neurones. J Physiol 2014; 593:267-84. [PMID: 25556800 DOI: 10.1113/jphysiol.2014.280263] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/25/2014] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS During neuronal development synaptic events mediated by GABAA receptors are progressively reduced in their duration, allowing for rapid and precise network function. Here we focused on ventrobasal thalamocortical neurones, which contribute to behaviourally relevant oscillations between thalamus and cortex. We demonstrate that the developmental decrease in the duration of inhibitory phasic events results predominantly from a precisely timed loss of locally produced neurosteroids, which act as positive allosteric modulators of the GABAA receptor. The mature thalamus retains the ability to synthesise neurosteroids, thus preserving the capacity to enhance both phasic and tonic inhibition, mediated by synaptic and extrasynaptic GABAA receptors, respectively, in physiological and pathophysiological scenarios associated with perturbed neurosteroid levels. Our data establish a potent, endogenous mechanism to locally regulate the GABAA receptor function and thereby influence thalamocortical activity. During brain development the duration of miniature inhibitory postsynaptic currents (mIPSCs) mediated by GABAA receptors (GABAA Rs) progressively reduces, to accommodate the temporal demands required for precise network activity. Conventionally, this synaptic plasticity results from GABAA R subunit reorganisation. In particular, in certain developing neurones synaptic α2-GABAA Rs are replaced by α1-GABAA Rs. However, in thalamocortical neurones of the mouse ventrobasal (VB) thalamus, the major alteration to mIPSC kinetics occurs on postnatal (P) day 10, some days prior to the GABAA R isoform change. Here, whole-cell voltage-clamp recordings from VB neurones of mouse thalamic slices revealed that early in postnatal development (P7-P8), the mIPSC duration is prolonged by local neurosteroids acting in a paracrine or autocrine manner to enhance GABAA R function. However, by P10, this neurosteroid 'tone' rapidly dissipates, thereby producing brief mIPSCs. This plasticity results from a lack of steroid substrate as pre-treatment of mature thalamic slices (P20-24) with the GABAA R-inactive precursor 5α-dihydroprogesterone (5α-DHP) resulted in markedly prolonged mIPSCs and a greatly enhanced tonic conductance, mediated by synaptic and extrasynaptic GABAA Rs, respectively. In summary, endogenous neurosteroids profoundly influence GABAergic neurotransmission in developing VB neurones and govern a transition from slow to fast phasic synaptic events. Furthermore, the retained capacity for steroidogenesis in the mature thalamus raises the prospect that certain physiological or pathophysiological conditions may trigger neurosteroid neosynthesis, thereby providing a local mechanism for fine-tuning neuronal excitability.
Collapse
Affiliation(s)
- Adam R Brown
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, DD1 9SY, UK
| | | | | | | |
Collapse
|
40
|
Abstract
BACKGROUND Anesthetics enhance γ-aminobutyric acid (GABA)-mediated inhibition in the central nervous system. Different agents have been shown to act on tonic versus synaptic GABA receptors to different degrees, but it remains unknown whether different forms of synaptic inhibition are also differentially engaged. With this in mind, we tested the hypothesis that different types of GABA-mediated synapses exhibit different anesthetic sensitivities. The present study compared effects produced by isoflurane, halothane, pentobarbital, thiopental, and propofol on paired-pulse GABAA receptor-mediated synaptic inhibition. Effects on glutamate-mediated facilitation were also studied. METHODS Synaptic responses were measured in rat hippocampal brain slices. Orthodromic paired-pulse stimulation was used to assess anesthetic effects on either glutamate-mediated excitatory inputs or GABA-mediated inhibitory inputs to CA1 neurons. Antidromic stimulation was used to assess anesthetic effects on CA1 background excitability. Agents were studied at equieffective concentrations for population spike depression to compare their relative degree of effect on synaptic inhibition. RESULTS Differing degrees of anesthetic effect on paired-pulse facilitation at excitatory glutamate synapses were evident, and blocking GABA inhibition revealed a previously unseen presynaptic action for pentobarbital. Although all 5 anesthetics depressed synaptically evoked excitation of CA1 neurons, the involvement of enhanced GABA-mediated inhibition differed considerably among agents. Single-pulse inhibition was enhanced by propofol, thiopental, and pentobarbital, but only marginally by halothane and isoflurane. In contrast, isoflurane enhanced paired-pulse inhibition strongly, as did thiopental, but propofol, pentobarbital, and halothane were less effective. CONCLUSIONS These observations support the idea that different GABA synapses use receptors with differing subunit compositions and that anesthetics exhibit differing degrees of selectivity for these receptors. The differing anesthetic sensitivities seen in the present study, at glutamate and GABA synapses, help explain the unique behavioral/clinical profiles produced by different classes of anesthetics and indicate that there are selective targets for new agent development.
Collapse
Affiliation(s)
- M Bruce MacIver
- From the Department of Anesthesia, Stanford School of Medicine, Palo Alto, California
| |
Collapse
|
41
|
Eckle VS, Rudolph U, Antkowiak B, Grasshoff C. Propofol modulates phasic and tonic GABAergic currents in spinal ventral horn interneurones. Br J Anaesth 2014; 114:491-8. [PMID: 25150989 DOI: 10.1093/bja/aeu269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Surgical interventions like skin incisions trigger withdrawal reflexes which require motor neurones and local circuit interneurones in the spinal ventral horn. This region plays a key role in mediating immobilizing properties of the GABAergic anaesthetic propofol. However, it is unclear how propofol modulates GABA(A) receptors in the spinal ventral horn and whether tonic or phasic inhibition is involved. METHODS Organotypic spinal cord tissue slices were prepared from mice. Whole-cell recordings were performed for quantifying effects of propofol on GABA(A) receptor-mediated phasic transmission and tonic conductance. RESULTS Propofol increased GABAergic phasic transmission by a prolongation of the decay time constant in a concentration-dependent manner. The amount of the charge transferred per inhibitory post-synaptic current, described by the area under the curve, was significantly augmented by 1 µM propofol (P<0.01). A GABA(A) receptor-mediated tonic current was not induced by 1 µM propofol but at a concentration of 5 µM (P<0.05). CONCLUSIONS Propofol depresses ventral horn interneurones predominantly by phasic rather than by tonic GABA(A) receptor-mediated inhibition. However, the present results suggest that the involvement of a tonic inhibition might contribute to the efficacy of propofol to depress nociceptive reflexes at high concentrations of the anaesthetic.
Collapse
Affiliation(s)
- V S Eckle
- Experimental Anaesthesiology Section, Department of Anaesthesiology and Intensive Care, Eberhard-Karls-University, Tübingen, Germany
| | - U Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - B Antkowiak
- Experimental Anaesthesiology Section, Department of Anaesthesiology and Intensive Care, Eberhard-Karls-University, Tübingen, Germany
| | - C Grasshoff
- Experimental Anaesthesiology Section, Department of Anaesthesiology and Intensive Care, Eberhard-Karls-University, Tübingen, Germany
| |
Collapse
|
42
|
Dmitriev RI, Kondrashina AV, Koren K, Klimant I, Zhdanov AV, Pakan JMP, McDermott KW, Papkovsky DB. Small molecule phosphorescent probes for O2imaging in 3D tissue models. Biomater Sci 2014; 2:853-866. [DOI: 10.1039/c3bm60272a] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PtPFPP-carbohydrate conjugates are promising O2probes for 3D PLIM imaging of live spheroids and brain explants.
Collapse
Affiliation(s)
| | | | - Klaus Koren
- Institute of Analytical Chemistry and Food Chemistry
- Graz University of Technology
- 8010 Graz, Austria
| | - Ingo Klimant
- Institute of Analytical Chemistry and Food Chemistry
- Graz University of Technology
- 8010 Graz, Austria
| | | | | | | | | |
Collapse
|
43
|
Coman D, Sanganahalli BG, Cheng D, McCarthy T, Rothman DL, Hyder F. Mapping phosphorylation rate of fluoro-deoxy-glucose in rat brain by (19)F chemical shift imaging. Magn Reson Imaging 2013; 32:305-13. [PMID: 24581725 DOI: 10.1016/j.mri.2013.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 07/05/2013] [Accepted: 10/03/2013] [Indexed: 10/25/2022]
Abstract
(19)F magnetic resonance spectroscopy (MRS) studies of 2-fluoro-2-deoxy-d-glucose (FDG) and 2-fluoro-2-deoxy-d-glucose-6-phosphate (FDG-6P) can be used for directly assessing total glucose metabolism in vivo. To date, (19)F MRS measurements of FDG phosphorylation in the brain have either been achieved ex vivo from extracted tissue or in vivo by unusually long acquisition times. Electrophysiological and functional magnetic resonance imaging (fMRI) measurements indicate that FDG doses up to 500 mg/kg can be tolerated with minimal side effects on cerebral physiology and evoked fMRI-BOLD responses to forepaw stimulation. In halothane-anesthetized rats, we report localized in vivo detection and separation of FDG and FDG-6P MRS signals with (19)F 2D chemical shift imaging (CSI) at 11.7 T. A metabolic model based on reversible transport between plasma and brain tissue, which included a non-saturable plasma to tissue component, was used to calculate spatial distribution of FDG and FDG-6P concentrations in rat brain. In addition, spatial distribution of rate constants and metabolic fluxes of FDG to FDG-6P conversion were estimated. Mapping the rate of FDG to FDG-6P conversion by (19)F CSI provides an MR methodology that could impact other in vivo applications such as characterization of tumor pathophysiology.
Collapse
Affiliation(s)
- Daniel Coman
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA; Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA; Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA.
| | - Basavaraju G Sanganahalli
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA; Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA; Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
| | - David Cheng
- Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
| | | | - Douglas L Rothman
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA; Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA; Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA; Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA; Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
44
|
Drexler B, Thiermann H, Antkowiak B, Grasshoff C. Effects of succinylcholine in an organotypic spinal cord-skeletal muscle coculture of embryonic mice. Chem Biol Interact 2013; 206:555-60. [DOI: 10.1016/j.cbi.2013.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/20/2013] [Accepted: 06/28/2013] [Indexed: 10/26/2022]
|
45
|
Alf MF, Duarte JM, Schibli R, Gruetter R, Krämer SD. Brain Glucose Transport and Phosphorylation Under Acute Insulin-Induced Hypoglycemia in Mice: An 18F-FDG PET Study. J Nucl Med 2013; 54:2153-60. [DOI: 10.2967/jnumed.113.122812] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
46
|
Wakita M, Kotani N, Nonaka K, Shin MC, Akaike N. Effects of propofol on GABAergic and glutamatergic transmission in isolated hippocampal single nerve-synapse preparations. Eur J Pharmacol 2013; 718:63-73. [DOI: 10.1016/j.ejphar.2013.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023]
|
47
|
Liu YW, Zuo W, Ye JH. Propofol stimulates noradrenalin-inhibited neurons in the ventrolateral preoptic nucleus by reducing GABAergic inhibition. Anesth Analg 2013; 117:358-63. [PMID: 23780420 DOI: 10.1213/ane.0b013e318297366e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The cellular mechanisms underlying the sedative effect of general anesthetics are not completely understood. Accumulating evidence indicates that the ventrolateral preoptic area (VLPO) of the hypothalamus plays a critical role. The VLPO contains 2 major types of neurons, the noradrenalin-inhibited GABAergic projecting neurons (NA(-) neurons) and the noradrenalin-excited interneurons (NA(+) neurons) which are probably also γ-aminobutyric acid (GABA)-containing neurons. Our previous work suggests that NA(-) neurons are normally under the inhibitory control of NA(+) neurons. Previous studies also show that GABAergic agents including propofol activate GABAergic projecting neurons in the VLPO, which is believed to lead to the inhibition of the arousal-producing nuclei in the tuberomammillary nucleus and sedation. However, how propofol activates VLPO neurons remains unclear. We explored the possibility that propofol activates NA(-) neurons indirectly, by inhibiting GABAergic transmission including those from VLPO NA(+) neurons. METHODS Electrophysiological activities were recorded from VLPO cells in acute brain slices of rats. RESULTS Propofol facilitates the discharges of NA(-) neurons and reduces the frequency, but not the amplitude of spontaneous GABAergic inhibitory postsynaptic currents in NA(-) neurons. Conversely, propofol suppressed the discharges of NA(+) neurons. CONCLUSION Propofol excites VLPO NA(-) neurons by reducing GABAergic transmission, at least in part by inhibiting VLPO NA(+) neurons. This may be a critical mechanism contributing to propofol-induced sedation.
Collapse
Affiliation(s)
- Yu-Wei Liu
- Department of Anaesthesiology, UMDNJ, New Jersey Medical School, 185 South Orange Ave., Newark, NJ 07103, USA
| | | | | |
Collapse
|
48
|
Drexler B, Kreuzer M, Jordan D, Antkowiak B, Schneider G. Sevoflurane-induced loss of consciousness is paralleled by a prominent modification of neural activity during cortical down-states. Neurosci Lett 2013; 548:149-54. [PMID: 23721783 DOI: 10.1016/j.neulet.2013.05.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/12/2013] [Accepted: 05/15/2013] [Indexed: 11/25/2022]
Abstract
Networks of neocortical neurons display a bistable activity pattern characterised by phases of high frequency action potential firing, so called up-states, and episodes of low discharge activity (down-states). We hypothesised that during down-states neocortical neurons are vulnerable to anaesthetic agents. To tackle this issue, it is necessary to identify analytical methods, which are sufficiently sensitive for resolving anaesthetic effects during phases of scarce neuronal activity. The local field potential was recorded in organotypic cultures (OTC) from rat neocortex under control conditions and in the presence of increasing concentrations of sevoflurane by extracellular electrodes. Epochs from down-states were cut from the local field potential and analysed using power spectrum density as well as non-linear parameters approximate entropy (ApEn) and order recurrence rate (ORR). ApEn and ORR proved to be suitable tools for analysing the actions of volatile anaesthetics on cortical down-states. During these phases of low neuronal activity, sevoflurane caused prominent changes in the local field potential. Time series analysis using ApEn showed a reduction of signal predictability in the presence of sevoflurane. Furthermore, the ORR displayed an abrupt decrease at sevoflurane concentrations corresponding to loss of consciousness in vivo, indicating a drug-induced decrease in the signal to noise ratio. The actions of volatile anaesthetics on cortical down-states have been neglected so far, perhaps due to the lack of suitable analysis tools. In the current in vitro study the non-linear parameters ApEn and ORR are introduced to characterise volatile anaesthetics actions. Sevoflurane alters cortical down-states as indicated by non-linear parameter analysis of local field potential recording from cultured neuronal networks. ORR even displays an abrupt change, i.e., a step-like behaviour indicating an increased signal complexity at concentrations of sevoflurane corresponding to loss of consciousness in humans.
Collapse
Affiliation(s)
- Berthold Drexler
- Section of Experimental Anaesthesiology, University of Tuebingen, Germany
| | | | | | | | | |
Collapse
|
49
|
Drexler B, Zinser S, Huang S, Poe MM, Rudolph U, Cook JM, Antkowiak B. Enhancing the function of alpha5-subunit-containing GABAA receptors promotes action potential firing of neocortical neurons during up-states. Eur J Pharmacol 2013; 703:18-24. [PMID: 23380687 DOI: 10.1016/j.ejphar.2013.01.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 01/12/2013] [Accepted: 01/16/2013] [Indexed: 11/27/2022]
Abstract
Neocortical neurons mediate the sedative and anticonvulsant properties of benzodiazepines. These agents enhance synaptic inhibition via positive modulation of γ-aminobutyric acid (GABAA) receptors harboring α1-, α2-, α3- or α5-protein subunits. Benzodiazepine-sensitive GABAA receptors containing the α5-subunit are abundant in the neocortex, but their impact in controlling neuronal firing patterns is unknown. Here we studied how the discharge rates of cortical neurons are modified by a positive (SH-053-2'F-R-CH3) and a negative (L 655,708) α5-subunit-preferring allosteric modulator in comparison to diazepam, the classical non-selective benzodiazepine. Drug actions were characterized in slice cultures from wild-type and α5(H105R) knock-in mice by performing extracellular multi-unit-recordings. In knock-in mice, receptors containing the α5 subunit are insensitive to benzodiazepines. The non-selective positive allosteric modulator diazepam decreased the discharge rates of neocortical neurons during episodes of ongoing neuronal activity (up states). In contrast to diazepam, the α5-preferring positive modulator SH-053-2'F-R-CH3 accelerated action potential firing during up states. This promoting action was absent in slices from α5(H105R) mice, confirming that it is mediated by the α5-subunit. Consistent with these observations, the negative α5-selective modulator L 655,708 inhibited up state action potential activity in slices from wild-type mice. The opposing actions of diazepam and SH-053-2'F-R-CH3, which both enhance GABAA receptor function but differ in subtype-selectivity, uncovers contrasting roles of GABAA receptor subtypes in controlling the firing rates of cortical neurons. These findings may have important implications for the design of novel anaesthetic and anticonvulsant benzodiazepines displaying an improved efficacy and fewer side effects.
Collapse
Affiliation(s)
- Berthold Drexler
- Experimental Anaesthesiology Section, Department of Anaesthesiology and Intensive Care Medicine, Eberhard-Karls-University, 72072 Tuebingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
50
|
Loryan I, Fridén M, Hammarlund-Udenaes M. The brain slice method for studying drug distribution in the CNS. Fluids Barriers CNS 2013; 10:6. [PMID: 23336814 PMCID: PMC3602653 DOI: 10.1186/2045-8118-10-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/02/2013] [Indexed: 11/22/2022] Open
Abstract
The high-throughput brain slice method is a precise and robust technique for estimating the overall uptake of drugs into brain tissue through determination of the unbound volume of distribution in the brain (Vu,brain; ml·g brain-1). Vu,brain describes the relationship between the total drug concentration in the brain and the concentration of unbound drug in the brain interstitial fluid, regardless of blood–brain barrier function. The brain slice method is more physiologically based than the brain homogenate method with respect to the assessment of drug distribution in the brain because the cell-cell interactions, pH gradients and active transport systems are all conserved. The method provides information that is directly relevant to issues such as nonspecific binding to brain tissue, lysosomal trapping, and active uptake into the cells. For these reasons, the brain slice method is recommended for estimation of target-site pharmacokinetics in the early drug discovery process and fundamental pharmacological studies. This article provides a detailed protocol for the rat and mouse brain slice methods, with the aim of enabling simple, cost-effective profiling of compounds with diverse physicochemical properties. The procedure for assessing the viability of the brain slices after the 5 h incubation period is also described. The results are interpreted for a set of compounds covering a wide range of physicochemical properties and various pharmacological targets. Application of the method for evaluating the unbound intracellular-to-extracellular concentration ratio (Kp,uu,cell) and the unbound brain-to-plasma concentration ratio (Kp,uu,brain) is discussed.
Collapse
Affiliation(s)
- Irena Loryan
- Department of Pharmaceutical Biosciences, Translational PKPD Research Group, Uppsala University, Associate member of SciLife Lab, Box 591, Uppsala, SE-75124, Sweden.
| | | | | |
Collapse
|