Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 43(8): 4345-4362, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37934363

ABSTRACT

Neuromodulation through magnetic fields irradiation with ait® (AT-04), a device that irradiates a mixed alternating magnetic fields (2 kHz and 83.3 MHz), has been shown to have high efficacy for fibromyalgia and low back pain in our previous clinical trials. The aim of this study was to elucidate the underlying analgesic mechanism of the AT-04 using the partial sciatic nerve ligation (PSL) model as an animal model of neuropathic pain. AT-04 was applied to PSL model rats with hyperalgesia and its pain-improving effect was verified by examining mechanical allodynia using the von Frey method. The results demonstrated a significant improvement in hyperalgesia in PSL model rats. We also examined the involvement of descending pain modulatory systems in the analgesic effects of AT-04 using antagonism by serotonin and noradrenergic receptor antagonists. These antagonists significantly reduced the analgesic effect of AT-04 on pain in PSL model rats by approximately 50%. We also measured the amount of serotonin and noradrenaline in the spinal fluid of PSL model rats using microdialysis during AT-04 treatment. Both monoamines were significantly increased by magnetic fields irradiation with AT-04. Furthermore, we evaluated the involvement of opioid analgesia in the analgesic effects of AT-04 using naloxone, the main antagonist of the opioid receptor, and found that it significantly antagonized the effects by approximately 60%. Therefore, the analgesic effects of AT-04 in PSL model rats involve both the endogenous pain modulation systems, including the descending pain modulatory system and the opioid analgesic system.


Subject(s)
Analgesia , Neuralgia , Rats , Animals , Hyperalgesia/complications , Hyperalgesia/drug therapy , Analgesics, Opioid/therapeutic use , Serotonin , Pain Measurement , Neuralgia/drug therapy , Analgesics/pharmacology , Disease Models, Animal
2.
J Pain ; 22(6): 715-729, 2021 06.
Article in English | MEDLINE | ID: mdl-33465503

ABSTRACT

Opioid usage for pain therapy is limited by its undesirable clinical effects, including paradoxical hyperalgesia, also known as opioid-induced hyperalgesia (OIH). However, the mechanisms associated with the development and maintenance of OIH remain unclear. Here, we investigated the effect of serotonin inhibition by the 5-HT3 receptor antagonist, ondansetron (OND), as well as serotonin deprivation via its synthesis inhibitor para-chlorophenylalanine, on mouse OIH models, with particular focus on astrocyte activation. Co-administering of OND and morphine, in combination with serotonin depletion, inhibited mechanical hyperalgesia and astrocyte activation in the spinal dorsal horn of mouse OIH models. Although previous studies have suggested that activation of astrocytes in the spinal dorsal horn is essential for the development and maintenance of OIH, herein, treatment with carbenoxolone (CBX), a gap junction inhibitor that suppresses astrocyte activation, did not ameliorate mechanical hyperalgesia in mouse OIH models. These results indicate that serotonin in the spinal dorsal horn, and activation of the 5-HT3 receptor play essential roles in OIH induced by chronic morphine, while astrocyte activation in the spinal dorsal horn serves as a secondary effect of OIH. Our findings further suggest that serotonergic regulation in the spinal dorsal horn may be a therapeutic target of OIH. PERSPECTIVE: The current study revealed that the descending serotonergic pain-facilitatory system in the spinal dorsal horn is crucial in OIH, and that activation of astrocytes is a secondary phenotype of OIH. Our study offers new therapeutic targets for OIH and may help reduce inappropriate opioid use.


Subject(s)
Analgesics, Opioid/pharmacology , Astrocytes , Hyperalgesia , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Serotonin/metabolism , Spinal Cord Dorsal Horn , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Ondansetron/pharmacology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism
3.
Front Pharmacol ; 11: 580289, 2020.
Article in English | MEDLINE | ID: mdl-33328986

ABSTRACT

Acetaminophen is one of the most commonly used analgesic agents for treating acute and chronic pain. However, its metabolism is complex, and its analgesic mechanisms have not been completely understood. Previously, it was believed that acetaminophen induces analgesia by inhibiting cyclooxygenase enzymes; however, it has been considered recently that the main analgesic mechanism of acetaminophen is its metabolization to N-acylphenolamine (AM404), which then acts on the transient receptor potential vanilloid 1 (TRPV1) and cannabinoid 1 receptors in the brain. We also recently revealed that the acetaminophen metabolite AM404 directly induces analgesia via TRPV1 receptors on terminals of C-fibers in the spinal dorsal horn. It is known that, similar to the brain, the spinal dorsal horn is critical to pain pathways and modulates nociceptive transmission. Therefore, acetaminophen induces analgesia by acting not only on the brain but also the spinal cord. In addition, acetaminophen is not considered to possess any anti-inflammatory activity because of its weak inhibition of cyclooxygenase (COX). However, we also revealed that AM404 induces analgesia via TRPV1 receptors on the spinal dorsal horn in an inflammatory pain rat model, and these analgesic effects were stronger in the model than in naïve rats. The purpose of this review was to summarize the previous and new issues related to the analgesic mechanisms of acetaminophen. We believe that it will allow clinicians to consider new pain management techniques involving acetaminophen.

4.
J Physiol ; 597(13): 3441-3455, 2019 07.
Article in English | MEDLINE | ID: mdl-31087329

ABSTRACT

KEY POINTS: Neuropathic pain spreads spatially beyond the injured sites, and the mechanism underlying the spread has been attributed to inflammation occurring in the spinal cord. However, the spatial spread of spinal/cortical potentiation induced by conduction block of the peripheral nerves can be observed prior to inflammation. In the present study, we found that spreading potentiation and hypersensitivity acutely induced by unilateral hindpaw ischaemia are nitric oxide (NO)-dependent and that NO is produced by ischaemia and quickly diffuses within the spinal cord. We also found that NO production induced by ischaemia is not observed in the presence of an antagonist for group II metabotropic glutamate receptors (mGluRs) and that neuronal NO synthase-positive dorsal horn neurons express group II mGluRs. These results suggest strongly that NO-mediated spreading potentiation in the spinal cord is one of the trigger mechanisms for neuropathic pain. ABSTRACT: Cortical/spinal responses to hindpaw stimulation are bilaterally potentiated by unilateral hindpaw ischaemia in mice. We tested the hypothesis that hindpaw ischaemia produces nitric oxide (NO), which diffuses in the spinal cord to induce spatially spreading potentiation. Using flavoprotein fluorescence imaging, we confirmed that the spreading potentiation in hindpaw responses was induced during ischaemia in the non-stimulated hindpaw. This spreading potentiation was blocked by spinal application of l-NAME, an inhibitor of NO synthase (NOS). Furthermore, no spreading potentiation was observed in neural NOS (nNOS) knockout mice. Spinal application of an NO donor was enough to induce cortical potentiation and mechanical hypersensitivity. The spatial distribution of NO during unilateral hindpaw ischaemia was visualized using 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM). An increase in fluorescence derived from the complex of DAF-FM with NO was observed on the ischaemic side of the spinal cord. A similar but smaller increase was also observed on the contralateral side. Somatosensory potentiation after hindpaw ischaemia is known to be inhibited by spinal application of LY354740, an agonist of group II metabotropic glutamate receptors (mGluRs). We confirmed that the spinal DAF-FM fluorescence increases during hindpaw ischaemia were not observed in the presence of LY354740. We also confirmed that approximately half of the nNOS-positive neurons in the superficial laminae of the dorsal horn expressed mGluR2 mRNA. These results suggest that disinhibition of mGluR2 produces NO which in turn induces a spreading potentiation in a wide area of the spinal cord. Such spreading, along with the consequent non-specific potentiation in the spinal cord, may trigger neuropathic pain.


Subject(s)
Ischemia/metabolism , Neuralgia/metabolism , Nitric Oxide/metabolism , Spinal Cord/metabolism , Animals , Ischemia/drug therapy , Male , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Neuralgia/drug therapy , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Pain Measurement/methods , Receptors, Metabotropic Glutamate/metabolism , Spinal Cord/drug effects
5.
Neuroscience ; 408: 161-176, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30986437

ABSTRACT

Noradrenaline (NA) modulates the spinal motor networks for locomotion and facilitates neuroplasticity, possibly assisting neuronal network activation and neuroplasticity in the recovery phase of spinal cord injuries. However, neither the effects nor the mechanisms of NA on synaptic transmission and neuronal excitability in spinal ventral horn (VH) neurons are well characterized, especially in rats aged 7 postnatal days or older. To gain insight into NA regulation of VH neuronal activity, we used a whole-cell patch-clamp approach in late neonatal rats (postnatal day 7-15). In voltage-clamp recordings at -70 mV, NA increased the frequency and amplitude of excitatory postsynaptic currents via the activation of somatic α1- and ß-adrenoceptors of presynaptic neurons. Moreover, NA induced an inward current through the activation of postsynapticα1- and ß-adrenoceptors. At a holding potential of 0 mV, NA also increased frequency and amplitude of both GABAergic and glycinergic inhibitory postsynaptic currents via the activation of somatic adrenoceptors in presynaptic neurons. In current-clamp recordings, NA depolarized resting membrane potentials and increased the firing frequency of action potentials in VH neurons, indicating that it enhances the excitability of these neurons. Our findings provide new insights that establish NA-based pharmacological therapy as an effective method to activate neuronal networks of the spinal VH in the recovery phase of spinal cord injuries.


Subject(s)
Action Potentials/drug effects , Anterior Horn Cells/drug effects , Norepinephrine/pharmacology , Spinal Cord/drug effects , Synaptic Transmission/drug effects , Animals , Membrane Potentials/drug effects , Patch-Clamp Techniques , Rats
6.
Br J Anaesth ; 123(2): e215-e225, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31030988

ABSTRACT

BACKGROUND: The neurosteroid dehydroepiandrosterone sulphate (DHEAS) activates the sigma-1 receptor, inhibits gamma-aminobutyric acid A (GABAA) and glycine receptors, and induces hyperalgesic effects. Although its effects have been studied in various tissues of the nervous system, its synaptic mechanisms in nociceptive pathways remain to be elucidated. METHODS: The threshold of mechanical hypersensitivity and spontaneous pain behaviour was assessed using the von Frey test in adult male Wistar rats after intrathecal administration of DHEAS. We also investigated the effects of DHEAS on synaptic transmission in the spinal dorsal horn using slice patch-clamp electrophysiology. RESULTS: Intrathecally administered DHEAS elicited dose-dependent mechanical hyperalgesia and spontaneous pain behaviours (withdrawal threshold: saline; 51.0 [20.1] g, 3 µg DHEAS; 14.0 [7.8] g, P<0.01, 10 µg DHEAS; 6.9 [5.2] g, 15 min after administration, P<0.001). DHEAS at 100 µM increased the frequency of miniature postsynaptic currents in the rat dorsal spinal horn; this increase was extracellular Ca2+-dependent but not sigma-1 and N-methyl-d-aspartate receptor-dependent. DHEAS suppressed the frequency of miniature inhibitory postsynaptic currents in a GABAA receptor- and sigma-1 receptor-dependent manner. CONCLUSIONS: These results suggest that DHEAS participates in the pathophysiology of nociceptive synaptic transmission in the spinal cord by potentiation of glutamate release and inhibition of the GABAA receptor.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , Pain/physiopathology , Spinal Cord Dorsal Horn/physiopathology , Animals , Disease Models, Animal , Male , Patch-Clamp Techniques , Rats , Rats, Wistar , Synaptic Transmission/physiology
7.
Biochem Biophys Res Commun ; 500(4): 897-901, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29702090

ABSTRACT

ß-alanine is a structural analog of glycine and γ-aminobutyric acid (GABA) and is thought to be involved in the modulation of nociceptive information at the spinal cord. However, it is not known whether ß-alanine exerts its effect in substantia gelatinosa (SG) neurons of the spinal dorsal horn, where glycine and GABA play an important role in regulating nociceptive transmission from the periphery. Here, we investigated the effects of ß-alanine on inhibitory synaptic transmission in adult rat SG neurons using whole-cell patch-clamp. ß-alanine dose-dependently induced outward currents in SG neurons. Current-voltage plots revealed a reversal potential at approximately -70 mV, which was close to the equilibrium potential of Cl-. Pharmacological analysis revealed that ß-alanine activates glycine receptors, but not GABAA receptors. These results suggest that ß-alanine hyperpolarizes the membrane potential of SG neurons by activating Cl- channels through glycine receptors. Our findings raise the possibility that ß-alanine may modulate pain sensation through glycine receptors.


Subject(s)
Nociception/drug effects , Posterior Horn Cells/drug effects , Receptors, Glycine/metabolism , Substantia Gelatinosa/drug effects , Synaptic Transmission/drug effects , beta-Alanine/pharmacology , Animals , Glycine/metabolism , Glycine/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nociception/physiology , Patch-Clamp Techniques , Posterior Horn Cells/cytology , Posterior Horn Cells/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, Glycine/agonists , Substantia Gelatinosa/cytology , Substantia Gelatinosa/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
8.
Brain Res ; 1682: 24-35, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29294349

ABSTRACT

We used a multimodal approach to evaluate the effects of edaravone in a rat model of spinal cord injury (SCI). SCI was induced by extradural compression of thoracic spinal cord. In experiment 1, 30 min prior to compression, rats received a 3 mg/kg intravenous bolus of edaravone followed by a maintenance infusion of 1 (low-dose), 3 (moderate-dose), or 10 (high-dose) mg/kg/h edaravone. Although both moderate- and high-dose edaravone regimens promoted recovery of spinal motor-evoked potentials (MEPs) at 2 h post-SCI, the effect of the moderate dose was more pronounced. In experiment 2, moderate-dose edaravone was administered 30 min prior to compression, at the start of compression, or 10 min after decompression. Although both preemptive and coincident administration resulted in significantly improved spinal MEPs at 2 h post-SCI, the effect of preemptive administration was more pronounced. A moderate dose of edaravone resulted in significant attenuation of lipid peroxidation, as evidenced by lower concentrations of the free radical malonyldialdehyde in the spinal cord 3 h post-SCI. Malonyldialdehyde levels in the high-dose edaravone group were not reduced. Both moderate- and high-dose edaravone resulted in significant functional improvements, evidenced by better Basso-Beattie-Bresnahan (BBB) scores and better performance on an inclined plane during an 8 week period post-SCI. Both moderate- and high-dose edaravone significantly attenuated neuronal loss in the spinal cord at 8 weeks post-SCI, as evidenced by quantitative immunohistochemical analysis of NeuN-positive cells. In conclusion, early administration of a moderate dose of edaravone minimized the negative consequences of SCI and facilitated functional recovery.


Subject(s)
Antipyrine/analogs & derivatives , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , 3,4-Methylenedioxyamphetamine/metabolism , Analysis of Variance , Animals , Antipyrine/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Edaravone , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Lipid Peroxidation/drug effects , Male , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Recovery of Function/drug effects , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/physiopathology , Time Factors
9.
10.
Anesthesiology ; 127(2): 355-371, 2017 08.
Article in English | MEDLINE | ID: mdl-28542001

ABSTRACT

BACKGROUND: The widely used analgesic acetaminophen is metabolized to N-acylphenolamine, which induces analgesia by acting directly on transient receptor potential vanilloid 1 or cannabinoid 1 receptors in the brain. Although these receptors are also abundant in the spinal cord, no previous studies have reported analgesic effects of acetaminophen or N-acylphenolamine mediated by the spinal cord dorsal horn. We hypothesized that clinical doses of acetaminophen induce analgesia via these spinal mechanisms. METHODS: We assessed our hypothesis in a rat model using behavioral measures. We also used in vivo and in vitro whole cell patch-clamp recordings of dorsal horn neurons to assess excitatory synaptic transmission. RESULTS: Intravenous acetaminophen decreased peripheral pinch-induced excitatory responses in the dorsal horn (53.1 ± 20.7% of control; n = 10; P < 0.01), while direct application of acetaminophen to the dorsal horn did not reduce these responses. Direct application of N-acylphenolamine decreased the amplitudes of monosynaptic excitatory postsynaptic currents evoked by C-fiber stimulation (control, 462.5 ± 197.5 pA; N-acylphenolamine, 272.5 ± 134.5 pA; n = 10; P = 0.022) but not those evoked by stimulation of Aδ-fibers. These phenomena were mediated by transient receptor potential vanilloid 1 receptors, but not cannabinoid 1 receptors. The analgesic effects of acetaminophen and N-acylphenolamine were stronger in rats experiencing an inflammatory pain model compared to naïve rats. CONCLUSIONS: Our results suggest that the acetaminophen metabolite N-acylphenolamine induces analgesia directly via transient receptor potential vanilloid 1 receptors expressed on central terminals of C-fibers in the spinal dorsal horn and leads to conduction block, shunt currents, and desensitization of these fibers.


Subject(s)
Acetaminophen/pharmacology , Analgesia/methods , Neurons, Afferent/drug effects , Pain/prevention & control , Spinal Cord Dorsal Horn/drug effects , TRPV Cation Channels/drug effects , Analgesics, Non-Narcotic/pharmacology , Animals , Disease Models, Animal , Male , Nerve Fibers, Unmyelinated/drug effects , Rats , Rats, Wistar , TRPV Cation Channels/genetics
11.
PLoS One ; 12(3): e0173622, 2017.
Article in English | MEDLINE | ID: mdl-28282425

ABSTRACT

BACKGROUND: We recently reported that tranexamic acid (TXA) evokes pain in rats by inhibiting γ-aminobutyric acid and glycine receptors on neurons in the spinal dorsal horn. Although TXA is commonly used to reduce perioperative blood loss during various surgeries, its potential to induce intraoperative nociception, thereby increasing the need for more analgesics during surgery, has not been investigated. Therefore, this study aimed to investigate whether TXA evokes pain and increases the need for a higher infusion rate of remifentanil in patients undergoing surgery for adolescent idiopathic scoliosis (AIS). METHODS: Data were collected from patients with AIS who underwent posterior spinal fusion surgery from January 2008 to December 2015. All surgical procedures were performed under total intravenous anesthesia with propofol and remifentanil, by the same team of orthopedic surgeons and anesthesiologists at a single institution. Patients in the TXA group were administered TXA (loading and maintenance doses, 1000 mg and 100 mg/h) whereas those in the control group were not. Our primary outcome was the infusion rate of the intraoperative opioid analgesic remifentanil. RESULTS: The final analysis was based on data collected from 33 and 30 patients in the control and TXA groups, respectively. No differences were observed in the demographic data or the hemodynamic parameters between the two groups of patients. In the TXA group, the durations of surgery and anesthesia were shorter, intravascular fluid volume and total blood loss were lower, and the doses of fentanyl and ketamine administered were higher than they were in the control group (P < 0.05 for all). The mean infusion rate of intraoperative remifentanil was significantly higher in the TXA group than in the control group (control group: 0.23 ± 0.04 µg/kg/min; TXA group: 0.28 ± 0.12 µg/kg/min; P = 0.014). CONCLUSIONS: Patients who received TXA during the AIS surgery required a higher infusion rate of remifentanil, indicating that TXA evoked pain during the surgery.


Subject(s)
Blood Loss, Surgical/prevention & control , Nociception/drug effects , Pain , Scoliosis , Spinal Fusion , Tranexamic Acid , Adolescent , Animals , Female , Humans , Male , Pain/chemically induced , Pain/physiopathology , Pain/surgery , Rats , Scoliosis/physiopathology , Scoliosis/surgery , Tranexamic Acid/administration & dosage , Tranexamic Acid/adverse effects
12.
Neuroscience ; 331: 206-20, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27343829

ABSTRACT

Hydrogen peroxide (H2O2), a reactive oxygen species, is an important signaling molecule for synaptic and neuronal activity in the central nervous system; it is produced excessively in brain ischemia and spinal cord injury. Although H2O2-mediated modulations of synaptic transmission have been reported in ventral horn (VH) neurons of the rat spinal cord, the effects of H2O2 on neuronal excitability and membrane properties remain poorly understood. Accordingly, the present study investigated such effects using a whole-cell patch-clamp technique. The bath-application of H2O2 decreased neuronal excitability accompanied by decreased input resistance, firing frequency, and action potential amplitude and by increased rheobase. These H2O2-mediated changes were induced by activation of extrasynaptic, but not synaptic, GABAA receptors. Indeed, GABAergic tonic currents were enhanced by H2O2. On the other hand, the amplitude of medium and slow afterhyperpolarization (mAHP and sAHP), which plays important roles in controlling neuronal excitability and is mediated by small-conductance calcium-activated potassium (SK) channels, was significantly decreased by H2O2. When extrasynaptic GABAA receptors were completely blocked, these decreases of mAHP and sAHP persisted, and H2O2 increased excitability, suggesting that H2O2 per se might have the potential to increase neuronal excitability via decreased SK channel conductance. These findings indicate that activating extrasynaptic GABAA receptors or SK channels may attenuate acute neuronal damage caused by H2O2-induced hyperexcitability and therefore represent a novel therapeutic target for the prevention and treatment of H2O2-induced motor neuron disorders.


Subject(s)
Anterior Horn Cells/physiology , Hydrogen Peroxide/metabolism , Membrane Potentials/physiology , Animals , Anterior Horn Cells/drug effects , Bicuculline/pharmacology , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Membrane Potentials/drug effects , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, Glycine/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Strychnine/pharmacology , Synapses/drug effects , Synapses/metabolism , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
13.
J Clin Anesth ; 31: 193-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27185708

ABSTRACT

We describe a very rare case of an indwelling central venous catheter (CVC) through the left internal jugular vein that perforated the superior vena cava (SVC) wall postoperatively, although the CVC was placed in the appropriate position preoperatively. Three days after CVC insertion, a chest radiograph showed that the CVC tip had moved from the lower SVC to the upper SVC. Five days after the insertion, computed tomography showed SVC perforation and the resulting hydrothorax. In cases of CVC insertion through the left side, the CVC tip should not be placed in the upper SVC (zone B). Considering individual clinical factors and the indwelling period for the CVC, the left innominate vein (zone C) may be a suitable site for the left-sided CVC tip to reduce the risk of SVC perforation.


Subject(s)
Catheterization, Central Venous/adverse effects , Central Venous Catheters/adverse effects , Jugular Veins/diagnostic imaging , Ovarian Neoplasms/surgery , Vena Cava, Superior/injuries , Fatal Outcome , Female , Humans , Hydrothorax/etiology , Tomography, X-Ray Computed , Vena Cava, Superior/diagnostic imaging
14.
Sci Rep ; 6: 26253, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27188335

ABSTRACT

Intravenous lidocaine administration produces an analgesic effect in various pain states, such as neuropathic and acute pain, although the underlying mechanisms remains unclear. Here, we hypothesized that intravenous lidocaine acts on spinal cord neurons and induces analgesia in acute pain. We therefore examined the action of intravenous lidocaine in the spinal cord using the in vivo patch-clamp technique. We first investigated the effects of intravenous lidocaine using behavioural measures in rats. We then performed in vivo patch-clamp recording from spinal substantia gelatinosa (SG) neurons. Intravenous lidocaine had a dose-dependent analgesic effect on the withdrawal response to noxious mechanical stimuli. In the electrophysiological experiments, intravenous lidocaine inhibited the excitatory postsynaptic currents (EPSCs) evoked by noxious pinch stimuli. Intravenous lidocaine also decreased the frequency, but did not change the amplitude, of both spontaneous and miniature EPSCs. However, it did not affect inhibitory postsynaptic currents. Furthermore, intravenous lidocaine induced outward currents in SG neurons. Intravenous lidocaine inhibits glutamate release from presynaptic terminals in spinal SG neurons. Concomitantly, it hyperpolarizes postsynaptic neurons by shifting the membrane potential. This decrease in the excitability of spinal dorsal horn neurons may be a possible mechanism for the analgesic action of intravenous lidocaine in acute pain.


Subject(s)
Analgesics/pharmacology , Lidocaine/pharmacology , Posterior Horn Cells/drug effects , Administration, Intravenous , Analgesics/administration & dosage , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Lidocaine/administration & dosage , Male , Membrane Potentials/drug effects , Pain Measurement/drug effects , Patch-Clamp Techniques , Posterior Horn Cells/physiology , Rats, Wistar , Synaptic Transmission/physiology
15.
J Physiol ; 594(1): 115-34, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26510999

ABSTRACT

KEY POINTS: Excessive production of reactive oxygen species (ROS) is implicated in many central nervous system disorders; however, the physiological role of ROS in spinal ventral horn (VH) neurons remains poorly understood. We investigated how pathological levels of H2O2, an abundant ROS, regulate synaptic transmission in VH neurons of rats using a whole-cell patch clamp approach. H2O2 increased the release of glutamate and GABA from presynaptic terminals. The increase in glutamate release involved N-type voltage-gated calcium channels (VGCCs), ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3 Rs); the increase in GABA release, which inhibited glutamatergic transmission, involved IP3 R. Inhibiting N-type VGCCs and RyRs attenuates excitotoxicity resulting from increased glutamatergic activity while preserving the neuroprotective effects of GABA, and may represent a novel strategy for treating H2O2-induced motor neuron disorders resulting from trauma or ischaemia-reperfusion injury. Excessive production of reactive oxygen species (ROS) is a critical component of the cellular and molecular pathophysiology of many central nervous system (CNS) disorders, including trauma, ischaemia-reperfusion injury, and neurodegenerative diseases. Hydrogen peroxide (H2O2), an abundant ROS, modulates synaptic transmission and contributes to neuronal damage in the CNS; however, the pathophysiological role of H2O2 in spinal cord ventral horn (VH) neurons remains poorly understood, despite reports that these neurons are highly vulnerable to oxidative stress and ischaemia. This was investigated in the present study using a whole-cell patch clamp approach in rats. We found that exogenous application of H2O2 increased the release of glutamate from excitatory presynaptic terminals and γ-aminobutyric acid (GABA) from inhibitory presynaptic terminals. The increase of glutamate release was induced in part by an increase in Ca(2+) influx through N-type voltage-gated calcium channels (VGCCs) as well as by ryanodine receptor (RyR)- and inositol trisphosphate receptor-mediated Ca(2+) release from the endoplasmic reticulum (ER). In inhibitory presynaptic neurons, increased IP3 R-mediated Ca(2+) release from the ER increased GABAergic transmission, which served to rescue VH neurons from excessive release of glutamate from presynaptic terminals. These findings indicate that inhibiting N-type VGCCs or RyRs may attenuate excitotoxicity resulting from increased glutamatergic activity while preserving the neuroprotective effects of GABA, and may therefore represent a novel and targeted strategy for preventing and treating H2O2-induced motor neuron disorders.


Subject(s)
Anterior Horn Cells/drug effects , Hydrogen Peroxide/pharmacology , Synaptic Potentials , Animals , Anterior Horn Cells/metabolism , Anterior Horn Cells/physiology , Calcium Signaling , Female , Glutamic Acid/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Voltage-Gated Sodium Channels/metabolism , gamma-Aminobutyric Acid/metabolism
16.
Sci Rep ; 5: 13458, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26293582

ABSTRACT

Tranexamic acid (TXA) is an antifibrinolytic agent widely used to reduce blood loss during surgery. However, a serious adverse effect of TXA is seizure due to inhibition of γ-aminobutyric acid (GABA) and glycine receptors in cortical neurons. These receptors are also present in the spinal cord, and antagonism of these receptors in spinal dorsal horn neurons produces pain-related phenomena, such as allodynia and hyperalgesia, in experimental animals. Moreover, some patients who are injected intrathecally with TXA develop severe back pain. However, the effect of TXA on spinal dorsal horn neurons remain poorly understood. Here, we investigated the effects of TXA by using behavioral measures in rats and found that TXA produces behaviors indicative of spontaneous pain and mechanical allodynia. We then performed whole-cell patch-clamp experiments that showed that TXA inhibits GABAA and glycine receptors in spinal dorsal horn neurons. Finally, we also showed that TXA facilitates activation of the extracellular signal-regulated kinase in the spinal cord. These results indicated that TXA produces pain by inhibiting GABAA and glycine receptors in the spinal dorsal horn.


Subject(s)
Pain/chemically induced , Pain/physiopathology , Posterior Horn Cells/pathology , Tranexamic Acid/adverse effects , Animals , Behavior, Animal , Biomarkers/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Glycine/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Inhibitory Postsynaptic Potentials , Injections, Intraperitoneal , Injections, Spinal , Male , Models, Biological , Nociception/drug effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/enzymology , Rats, Wistar , Tranexamic Acid/administration & dosage , gamma-Aminobutyric Acid/pharmacology
17.
Sci Rep ; 5: 11191, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26165560

ABSTRACT

Transient ischemia produces postischemic tingling sensation. Ischemia also produces nerve conduction block that may modulate spinal neural circuits. In the present study, reduced mechanical thresholds for hindpaw-withdrawal reflex were found in mice after transient hindpaw ischemia, which was produced by a high pressure applied around the hindpaw for 30 min. The reduction in the threshold was blocked by spinal application of LY354740, a specific agonist of group II metabotropic glutamate receptors. Neural activities in the spinal cord and the primary somatosensory cortex (S1) were investigated using activity-dependent changes in endogenous fluorescence derived from mitochondrial flavoproteins. Ischemic treatment induced potentiation of the ipsilateral spinal and contralateral S1 responses to hindpaw stimulation. Both types of potentiation were blocked by spinal application of LY354740. The contralateral S1 responses, abolished by lesioning the ipsilateral dorsal column, reappeared after ischemic treatment, indicating that postischemic tingling sensation reflects a sensory modality shift from tactile sensation to nociception in the spinal cord. Changes in neural responses were investigated during ischemic treatment in the contralateral spinal cord and the ipsilateral S1. Potentiation already appeared during ischemic treatment for 30 min. The present findings suggest that the postischemic potentiation shares spinal mechanisms, at least in part, with neuropathic pain.


Subject(s)
Ischemia/pathology , Spinal Cord/physiopathology , Animals , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Ischemia/drug therapy , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Optical Imaging , Pressure , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiopathology , Spinal Cord/drug effects , Thigh/blood supply
18.
Masui ; 63(8): 866-71, 2014 Aug.
Article in Japanese | MEDLINE | ID: mdl-25199319

ABSTRACT

BACKGROUND: There is an increasing number of patients scheduled for abdominal aortic aneurysm resection in whom epidural anesthesia cannot be performed because of concomitant antiplatelet/anticoagulant therapy. Instead of epidural anesthesia for postoperative analgesia in such patients it is possible to use repeated bilateral subcostal transversus abdominis plane (TAP) blocks. METHODS: Four patients receiving antiplatelet/anticoagulant therapy for abdominal aortic aneurysm resection under general anesthesia were studied. After the completion of surgery and before emergence from anesthesia 18-gauge intravenous catheters were inserted bilaterally into subcostal TAP and 100 ml (50 ml on each side) of 0.2% lidocaine with 1/500,000 epinephrine were injected via the catheters twice daily until the second postoperative day. Pain intensity was assessed using a 0-10 numerical rating scale at rest and during movement, before and after each block. RESULTS: Numerical pain ratings at rest and during movement decreased after each block, and good analgesia was obtained. No complications such as nausea, vomiting or infection were observed in the postoperative period. CONCLUSIONS: These findings suggest that repeated bilateral subcostal TAP blocks with 0.2% lidocaine performed via 18-gauge intravenous catheters provide good postoperative analgesia after abdominal aortic aneurysm resection.


Subject(s)
Abdominal Muscles/innervation , Analgesia/methods , Anesthetics, Local/administration & dosage , Aortic Aneurysm, Abdominal/surgery , Lidocaine/administration & dosage , Nerve Block/methods , Pain, Postoperative/drug therapy , Vascular Access Devices , Aged , Epinephrine/administration & dosage , Humans , Male , Postoperative Care , Treatment Outcome
19.
J Anesth ; 28(6): 839-45, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24748402

ABSTRACT

PURPOSE: Opioid-induced spastic paraplegia after transient spinal cord ischemia during aortic surgery has been reported. Opioids modulate neurotransmission through mu (µ) opioid receptors (MORs) in the spinal ventral horn. However, their effects during ischemic insult are not understood. METHODS: The effects of the selective µ agonist [D-Ala(2),-N-Me-Phe(4), Gly(5)-ol]enkephalin (DAMGO) on ischemia-induced agonal currents were examined in the spinal lamina IX neurons of neonatal rats by using the whole-cell patch-clamp technique. Ischemia was simulated in vitro by oxygen/glucose deprivation. RESULTS: DAMGO (1 µM) produced outward currents in ~60% of spinal lamina IX neurons at a holding potential of -70 mV. Superfusion with ischemia-simulating medium elicited an agonal current. The latency was 457 ± 18 s. Despite its neuromodulatory effects, DAMGO did not significantly change the latencies of the agonal currents with (440 ± 23 s) or without (454 ± 33 s) DAMGO-induced currents. CONCLUSION: Activation of MORs does not influence ongoing ischemia-induced neuronal death. Our findings indicate that MOR agonist administration should be suitable as an anesthetic during aortic surgery.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Receptors, Opioid, mu/agonists , Spinal Cord Ischemia/physiopathology , Animals , Animals, Newborn , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Synaptic Transmission/drug effects
20.
Springerplus ; 2: 487, 2013.
Article in English | MEDLINE | ID: mdl-24102044

ABSTRACT

We report the anesthetic management of a patient scheduled for tumor resection with a giant ovarian tumor containing 83 l of fluid. A 59-year-old woman [height 154 cm; weight 146 kg (ideal: 52 kg)] with a giant ovarian tumor was scheduled for tumor resection. Her preoperative abdominal circumference was 194 cm, which made supine positioning difficult. The thoracoabdominal computed tomography revealed a right giant cystic ovarian tumor with an estimated mass of 100 kg. Evidence of malignant tumor was not observed. In the operation room, she was intubated using a video laryngoscope (Airway Scope®, Hoya, Tokyo, Japan) in a semirecumbent position under conscious sedation. Following general anesthesia, the tumor fluid was gradually aspirated at a rate of 500 ml/min, and during this procedure, spontaneous respiration was preserved with pressure support ventilation. After the fluid was drained, the tumor was resected in a supine position. There were no major perioperative complications in hemodynamic and respiratory status, such as supine hypotensive syndrome or re-expansion pulmonary edema. Her weight decreased to 50 kg postoperatively. Maintenance of spontaneous respiration and slow aspiration of the tumor fluid prevented respiratory and hemodynamic failure and resulted in safe anesthesia management during giant ovarian tumor resection.

SELECTION OF CITATIONS
SEARCH DETAIL
...