Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.962
Filter
1.
Brain Behav ; 14(7): e3610, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38945806

ABSTRACT

INTRODUCTION: Pregnant women may need to undergo non-obstetric surgery under general anesthesia owing to medical needs, and pregnant women frequently experience sleep disturbances during late gestation. Preclinical studies demonstrated that maternal isoflurane exposure (MISO) or maternal sleep deprivation (MSD) contributed to cognitive impairments in offspring. Research studies in mice have revealed that SD can aggravate isoflurane-induced cognitive deficits. However, it remains unclear whether MSD aggravates MISO-induced cognitive deficits in offspring. The purpose of this research was to explore the combined effects of MSD and MISO on offspring cognitive function and the role of neuroinflammation and synaptic function in the process of MSD + MISO. METHODS: Pregnant mice were exposed to 1.4% isoflurane by inhalation for 4 h on gestational day (GD) 14. Dams were then subjected to SD for 6 h (12:00-18:00 h) during GD15-21. At 3 months of age, the offspring mice were subjected to the Morris water maze test to assess cognitive function. Then the levels of inflammatory and anti-inflammatory markers and synaptic function-related proteins were assessed using molecular biology methods. RESULTS: The results of this study demonstrated that MISO led to cognitive dysfunction, an effect that was aggravated by MSD. In addition, MSD exacerbated the maternal isoflurane inhalation, leading to an enhancement in the expression levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha and a reduction in the hippocampal levels of IL-10, synaptophysin, post-synaptic density-95, growth-associated protein-43, and brain-derived neurotrophic factor. CONCLUSION: Our findings revealed that MSD aggravated the cognitive deficits induced by MISO in male offspring mice, and these results were associated with neuroinflammation and alternations in synaptic function.


Subject(s)
Anesthetics, Inhalation , Cognitive Dysfunction , Hippocampus , Isoflurane , Neuroinflammatory Diseases , Prenatal Exposure Delayed Effects , Sleep Deprivation , Animals , Isoflurane/adverse effects , Isoflurane/pharmacology , Isoflurane/administration & dosage , Female , Cognitive Dysfunction/etiology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/physiopathology , Pregnancy , Sleep Deprivation/complications , Sleep Deprivation/physiopathology , Mice , Hippocampus/metabolism , Hippocampus/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Synapses/drug effects , Male , Mice, Inbred C57BL , Maternal Deprivation , Brain-Derived Neurotrophic Factor/metabolism
2.
Open Vet J ; 14(5): 1251-1258, 2024 May.
Article in English | MEDLINE | ID: mdl-38938419

ABSTRACT

Background: The intramuscular (IM) administration of 7.5-10 mg/kg of alfaxalone produces anesthetic effects that enable endotracheal intubation with mild cardiorespiratory depression in dogs. However, the effects of IM co-administration of medetomidine, butorphanol, and alfaxalone on cardiorespiratory function under inhalation anesthesia have not been studied. Aim: To assess the cardiorespiratory function following the IM co-administration of 5 µg/kg of medetomidine, 0.3 mg/kg of butorphanol, and 2.5 mg/kg of alfaxalone (MBA) in dogs anesthetized with sevoflurane. Methods: Seven intact healthy Beagles (three males and four females, aged 3-6 years old and weighing 10.0-18.1 kg) anesthetized with a predetermined minimum alveolar concentration (MAC) of sevoflurane were included in this study. The baseline cardiorespiratory variable values were recorded using the thermodilution method with a pulmonary artery catheter after stabilization for 15 minutes at 1.3 times their individual sevoflurane MAC. The cardiorespiratory variables were measured again following the IM administration of MBA. Data are expressed as median [interquartile range] and compared with the corresponding baseline values using the Friedman test and Sheff's method. A p < 0.05 was considered statistically significant. Results: The intramuscular administration of MBA transiently decreased the cardiac index [baseline: 3.46 (3.18-3.69), 5 minutes: 1.67 (1.57-1.75) l/minute/m2 : p < 0.001], respiratory frequency, and arterial pH. In contrast, it increased the systemic vascular resistance index [baseline: 5,367 (3,589-6,617), 5 minutes:10,197 (9,955-15,005) dynes second/cm5/m2 : p = 0.0092], mean pulmonary arterial pressure, and arterial partial pressure of carbon dioxide. Conclusion: The intramuscular administration of MBA in dogs anesthetized with sevoflurane transiently decreased cardiac output due to vasoconstriction. Although spontaneous breathing was maintained, MBA administration resulted in respiratory acidosis due to hypoventilation. Thus, it is important to administer MBA with caution to dogs with insufficient cardiovascular function. In addition, ventilatory support is recommended.


Subject(s)
Anesthetics, Inhalation , Butorphanol , Medetomidine , Pregnanediones , Sevoflurane , Animals , Sevoflurane/administration & dosage , Sevoflurane/pharmacology , Butorphanol/administration & dosage , Butorphanol/pharmacology , Medetomidine/administration & dosage , Medetomidine/pharmacology , Dogs/physiology , Pregnanediones/administration & dosage , Pregnanediones/pharmacology , Male , Female , Injections, Intramuscular/veterinary , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Heart Rate/drug effects , Blood Pressure/drug effects
3.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38928030

ABSTRACT

Disruption of any stage of iron homeostasis, including uptake, utilization, efflux, and storage, can cause progressive damage to peripheral organs. The health hazards associated with occupational exposure to inhalation anesthetics (IA) in combination with chronic iron overload are not well documented. This study aimed to investigate changes in the concentration of essential metals in the peripheral organs of rats after iron overload in combination with IA. The aim was also to determine how iron overload in combination with IA affects tissue metal homeostasis, hepcidin-ferritin levels, and MMP levels according to physiological, functional, and tissue features. According to the obtained results, iron accumulation was most pronounced in the liver (19×), spleen (6.7×), lungs (3.1×), and kidneys (2.5×) compared to control. Iron accumulation is associated with elevated heavy metal levels and impaired essential metal concentrations due to oxidative stress (OS). Notably, the use of IA increases the iron overload toxicity, especially after Isoflurane exposure. The results show that the regulation of iron homeostasis is based on the interaction of hepcidin, ferritin, and other proteins regulated by inflammation, OS, free iron levels, erythropoiesis, and hypoxia. Long-term exposure to IA and iron leads to the development of numerous adaptation mechanisms in response to toxicity, OS, and inflammation. These adaptive mechanisms of iron regulation lead to the inhibition of MMP activity and reduction of oxidative stress, protecting the organism from possible damage.


Subject(s)
Anesthetics, Inhalation , Hepcidins , Iron-Dextran Complex , Iron , Oxidative Stress , Animals , Rats , Hepcidins/metabolism , Oxidative Stress/drug effects , Iron/metabolism , Male , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/toxicity , Iron-Dextran Complex/administration & dosage , Iron-Dextran Complex/toxicity , Ferritins/metabolism , Iron Overload/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Rats, Wistar , Homeostasis/drug effects , Isoflurane/adverse effects
5.
BMC Anesthesiol ; 24(1): 200, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840092

ABSTRACT

BACKGROUND: The inhalational anesthetic isoflurane is commonly utilized in clinical practice, particularly in the field of pediatric anesthesia. Research has demonstrated its capacity to induce neuroinflammation and long-term behavioral disorders; however, the underlying mechanism remains unclear [1]. The cation-chloride cotransporters Na+-K+-2Cl--1 (NKCC1) and K+-2Cl--2 (KCC2) play a pivotal role in regulating neuronal responses to gamma-aminobutyric acid (GABA) [2]. Imbalances in NKCC1/KCC2 can disrupt GABA neurotransmission, potentially leading to neural circuit hyperexcitability and reduced inhibition following neonatal exposure to anesthesia [3]. Therefore, this study postulates that anesthetics have the potential to dysregulate NKCC1 and/or KCC2 during brain development. METHODS: We administered 1.5% isoflurane anesthesia to neonatal rats for a duration of 4 h at postnatal day 7 (PND7). Anxiety levels were assessed using the open field test at PND28, while cognitive function was evaluated using the Morris water maze test between PND31 and PND34. Protein levels of NKCC1, KCC2, BDNF, and phosphorylated ERK (P-ERK) in the hippocampus were measured through Western blotting analysis. Pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α were quantified using ELISA. RESULTS: We observed a decrease in locomotion trajectories within the central region and a significantly shorter total distance in the ISO group compared to CON pups, indicating that isoflurane induces anxiety-like behavior. In the Morris water maze (MWM) test, rats exposed to isoflurane exhibited prolonged escape latency onto the platform. Additionally, isoflurane administration resulted in reduced time spent crossing in the MWM experiment at PND34, suggesting long-term impairment of memory function. Furthermore, we found that isoflurane triggered activation of pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α; downregulated KCC2/BDNF/P-ERK expression; and increased the NKCC1/KCC2 ratio in the hippocampus of PND7 rats. Bumetadine (NKCC1 specific inhibitors) reversed cognitive damage and effective disorder induced by isoflurane in neonatal rats by inhibiting TNF-α activation, normalizing IL-6 and IL-1ß levels, restoring KCC2 expression levels as well as BDNF and ERK signaling pathways. Based on these findings, it can be speculated that BDNF, P-ERK, IL-1ß, IL-6 and TNF - α may act downstream of the NKCC1/KCC2 pathway. CONCLUSIONS: Our findings provide evidence that isoflurane administration in neonatal rats leads to persistent cognitive deficits through dysregulation of the Cation-Chloride Cotransporters NKCC1 and KCC2, BDNF, p-ERK proteins, as well as neuroinflammatory processes.


Subject(s)
Anesthetics, Inhalation , Animals, Newborn , Isoflurane , K Cl- Cotransporters , Solute Carrier Family 12, Member 2 , Symporters , Animals , Isoflurane/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Symporters/metabolism , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/adverse effects , Rats , Mice , Rats, Sprague-Dawley , Male , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/metabolism , Female , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism
6.
CNS Neurosci Ther ; 30(6): e14782, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38828651

ABSTRACT

BACKGROUND: The thalamus system plays critical roles in the regulation of reversible unconsciousness induced by general anesthetics, especially the arousal stage of general anesthesia (GA). But the function of thalamus in GA-induced loss of consciousness (LOC) is little known. The thalamic reticular nucleus (TRN) is the only GABAergic neurons-composed nucleus in the thalamus, which is composed of parvalbumin (PV) and somatostatin (SST)-expressing GABAergic neurons. The anterior sector of TRN (aTRN) is indicated to participate in the induction of anesthesia, but the roles remain unclear. This study aimed to reveal the role of the aTRN in propofol and isoflurane anesthesia. METHODS: We first set up c-Fos straining to monitor the activity variation of aTRNPV and aTRNSST neurons during propofol and isoflurane anesthesia. Subsequently, optogenetic tools were utilized to activate aTRNPV and aTRNSST neurons to elucidate the roles of aTRNPV and aTRNSST neurons in propofol and isoflurane anesthesia. Electroencephalogram (EEG) recordings and behavioral tests were recorded and analyzed. Lastly, chemogenetic activation of the aTRNPV neurons was applied to confirm the function of the aTRN neurons in propofol and isoflurane anesthesia. RESULTS: c-Fos straining showed that both aTRNPV and aTRNSST neurons are activated during the LOC period of propofol and isoflurane anesthesia. Optogenetic activation of aTRNPV and aTRNSST neurons promoted isoflurane induction and delayed the recovery of consciousness (ROC) after propofol and isoflurane anesthesia, meanwhile chemogenetic activation of the aTRNPV neurons displayed the similar effects. Moreover, optogenetic and chemogenetic activation of the aTRN neurons resulted in the accumulated burst suppression ratio (BSR) during propofol and isoflurane GA, although they represented different effects on the power distribution of EEG frequency. CONCLUSION: Our findings reveal that the aTRN GABAergic neurons play a critical role in promoting the induction of propofol- and isoflurane-mediated GA.


Subject(s)
Anesthesia, General , Consciousness , GABAergic Neurons , Isoflurane , Propofol , Propofol/pharmacology , Isoflurane/pharmacology , Animals , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Mice , Consciousness/drug effects , Consciousness/physiology , Male , Electroencephalography , Anesthetics, Inhalation/pharmacology , Anterior Thalamic Nuclei/drug effects , Anterior Thalamic Nuclei/physiology , Mice, Inbred C57BL , Mice, Transgenic , Anesthetics, Intravenous/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Optogenetics
7.
Trials ; 25(1): 362, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38840210

ABSTRACT

BACKGROUND: Flash visual evoked potentials (FVEPs) are a reliable method for protecting visual function during spine surgery in prone position. However, the popularization and application of FVEPs remain limited due to the unclear influence of various anesthetics on FVEPs. Exploring the effects of anesthetic drugs on FVEP and establishing appropriate anesthesia maintenance methods are particularly important for promoting and applying FVEP. According to the conventional concept, inhaled narcotic drugs significantly affect the success of FVEP monitoring, FVEP extraction, and interpretation. Nonetheless, our previous study demonstrated that sevoflurane-propofol balanced anesthesia was a practicable regimen for FVEPs. Desflurane is widely used in general anesthesia for its rapid recovery properties. As the effect of desflurane on FVEP remains unclear, this trial will investigate the effect of different inhaled concentrations of desflurane anesthesia on amplitude of FVEPs during spine surgery, aiming to identify more feasible anesthesia schemes for the clinical application of FVEP. METHODS/ DESIGN: A total of 70 patients undergoing elective spinal surgery will be enrolled in this prospective, randomized controlled, open-label, patient-assessor-blinded, superiority trial and randomly assigned to the low inhaled concentration of desflurane group (LD group) maintained with desflurane-propofolremifentanil-balanced anesthesia or high inhaled concentration of desflurane group (HD group) maintained with desflurane-remifentanil anesthesia maintenance group at a ratio of 1:1. All patients will be monitored for intraoperative FVEPs, and the baseline will be measured half an hour after induction under total intravenous anesthesia (TIVA). After that, patients will receive 0.5 minimum alveolar concentration (MAC) of desflurane combined with propofol and remifentanil for anesthesia maintenance in the LD group, while 0.7-1.0 MAC of desflurane and remifentanil will be maintained in the HD group. The primary outcome is the N75-P100 amplitude 1 h after the induction of anesthesia. We intend to use the dual measure evaluation, dual data entry, and statistical analysis by double trained assessors to ensure the reliability and accuracy of the results. DISCUSSION: This randomized controlled trial aims to explore the superiority effect of low inhaled concentration of desflurane combined with propofolremifentanil-balanced anesthesia versus high inhaled concentration of desflurane combined with remifentanil anesthesia on amplitude of FVEPs. The study is meant to be published in a peer-reviewed journal and might guide the anesthetic regimen for FVEPs. The conclusion is expected to provide high-quality evidence for the effect of desflurane on FVEPs and aim to explore more feasible anesthesia schemes for the clinical application of FVEPs and visual function protection. TRIAL REGISTRATION: This study was registered on clinicaltrials.gov on July 15, 2022. CLINICALTRIALS: gov Identifier: NCT05465330.


Subject(s)
Anesthetics, Inhalation , Desflurane , Evoked Potentials, Visual , Intraoperative Neurophysiological Monitoring , Randomized Controlled Trials as Topic , Remifentanil , Spine , Humans , Desflurane/administration & dosage , Evoked Potentials, Visual/drug effects , Anesthetics, Inhalation/administration & dosage , Prospective Studies , Spine/surgery , Middle Aged , Intraoperative Neurophysiological Monitoring/methods , Adult , Male , Remifentanil/administration & dosage , Female , Propofol/administration & dosage , Young Adult , Aged , Anesthetics, Intravenous/administration & dosage , Adolescent , Time Factors , Orthopedic Procedures , Photic Stimulation
8.
J Zoo Wildl Med ; 55(2): 424-429, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38875198

ABSTRACT

The marbled crayfish (Procambarus virginalis) is a parthenogenetic invasive species across much of the world, and when found, euthanasia is often recommended to reduce spread to naïve ecosystems. Euthanasia recommendations in crustaceans includes a two-step method: first to produce nonresponsiveness and then to destroy central nervous tissue. Minimal data exist on adequate anesthetic or immobilization methods for crayfish. A population of 90 marbled crayfish was scheduled for euthanasia due to invasive species concerns. The population was divided into six treatment groups to evaluate whether immersion in emulsified isoflurane or propofol solutions could produce nonresponsiveness. Each group was exposed to one of six treatments for 1 h: isoflurane emulsified at 0.1%, 0.5%, 2%, 5%, and 15% or propofol at 10 mg/L and then increased to 100 mg/L. Crayfish from all treatment groups were moved to nonmedicated water after completion of 1 h and observed for an additional 4 h. All crayfish treated with isoflurane showed lack of a righting reflex at 5 min and loss of movement after 30 min. By 240 min (4 h), none of the crayfish from the isoflurane treatment groups regained movement. None of the crayfish in the propofol treatment achieved loss of reflexes or responsiveness, and all remained normal upon return to nonmedicated water. Isoflurane emulsified in water produces nonresponsiveness that is appropriate for the first step of euthanasia, while propofol was insufficient at these treatment doses.


Subject(s)
Astacoidea , Euthanasia, Animal , Isoflurane , Propofol , Animals , Astacoidea/drug effects , Isoflurane/administration & dosage , Isoflurane/pharmacology , Propofol/pharmacology , Propofol/administration & dosage , Euthanasia, Animal/methods , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Immersion , Dose-Response Relationship, Drug
9.
Biomed Pharmacother ; 176: 116937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38870632

ABSTRACT

The advent of general anesthesia (GA) has significant implications for clinical practice. However, the exact mechanisms underlying GA-induced transitions in consciousness remain elusive. Given some similarities between GA and sleep, the sleep-arousal neural nuclei and circuits involved in sleep-arousal, including the 5-HTergic system, could be implicated in GA. Herein, we utilized pharmacology, optogenetics, chemogenetics, fiber photometry, and retrograde tracing to demonstrate that both endogenous and exogenous activation of the 5-HTergic neural circuit between the dorsal raphe nucleus (DR) and basolateral amygdala (BLA) promotes arousal and facilitates recovery of consciousness from sevoflurane anesthesia. Notably, the 5-HT1A receptor within this pathway holds a pivotal role. Our findings will be conducive to substantially expanding our comprehension of the neural circuit mechanisms underlying sevoflurane anesthesia and provide a potential target for modulating consciousness, ultimately leading to a reduction in anesthetic dose requirements and side effects.


Subject(s)
Anesthetics, Inhalation , Basolateral Nuclear Complex , Consciousness , Dorsal Raphe Nucleus , Sevoflurane , Sevoflurane/pharmacology , Animals , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Consciousness/drug effects , Anesthetics, Inhalation/pharmacology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/physiology , Male , Mice , Mice, Inbred C57BL , Serotonin/metabolism , Neural Pathways/drug effects , Neural Pathways/physiology , Receptor, Serotonin, 5-HT1A/metabolism , Optogenetics
10.
BMC Anesthesiol ; 24(1): 207, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872117

ABSTRACT

BACKGROUND: Intra-operative anaesthesia management should be optimised to reduce the occurrence of postoperative nausea and vomiting in high-risk patients; however, a single intervention may not effectively reduce postoperative nausea and vomiting in such patients. This study assessed the effect of an optimised anaesthetic protocol versus a conventional one on postoperative nausea and vomiting in patients who underwent laparoscopic sleeve gastrectomy. METHODS: A single-centre randomised trial was conducted at Peking University Shenzhen Hospital from June 2021 to December 2022. Among 168 patients who underwent laparoscopic sleeve gastrectomy, 116 qualified, and 103 completed the study with available data. Patients were categorized into the conventional group (received sevoflurane and standard fluids) and the optimised group (underwent propofol-based anaesthesia and was administered goal-directed fluids). The primary endpoints were postoperative nausea and vomiting incidence and severity within 24 h. RESULTS: Postoperative nausea and vomiting assessment at 0-3 h post-surgery revealed no significant differences between groups. However, at 3-24 h, the optimised anaesthetic protocol group showed lower postoperative nausea and vomiting incidence and severity than those of the conventional group (P = 0.005). In the conventional group, 20 (37.04%) patients experienced moderate-to-severe postoperative nausea and vomiting, compared to six (12.25%) patients in the optimised group (odds ratio = 0.237; 95% CI = 0.086, 0.656; P = 0.006). No significant differences were noted in antiemetic treatment, moderate-to-severe pain incidence, anaesthesia recovery, post-anaesthetic care unit stay, or postoperative duration between the groups. While the total intra-operative infusion volumes were comparable, the optimised group had a significantly higher colloidal infusion volume (500 mL vs. 0 mL, P = 0.014) than that of the conventional group. CONCLUSIONS: The incidence and severity of postoperative nausea and vomiting 3-24 h postoperatively in patients who underwent laparoscopic sleeve gastrectomy were significantly lower with propofol-based total intravenous anaesthesia and goal-directed fluid therapy than with sevoflurane anaesthesia and traditional fluid management. Total intravenous anaesthesia is an effective multimodal antiemetic strategy for bariatric surgery. TRIAL REGISTRATION: This trial was registered with the Chinese Clinical Trial Registry (ChiCTR-TRC- 2,100,046,534, registration date: 21 May 2021).


Subject(s)
Gastrectomy , Laparoscopy , Postoperative Nausea and Vomiting , Propofol , Sevoflurane , Humans , Postoperative Nausea and Vomiting/prevention & control , Postoperative Nausea and Vomiting/epidemiology , Male , Female , Laparoscopy/methods , Gastrectomy/methods , Gastrectomy/adverse effects , Adult , Propofol/administration & dosage , Sevoflurane/administration & dosage , Middle Aged , Anesthetics, Intravenous/administration & dosage , Anesthetics, Inhalation/administration & dosage , Anesthesia/methods
11.
J Physiol Sci ; 74(1): 33, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867187

ABSTRACT

Hibernation and torpor are not passive responses caused by external temperature drops and fasting but are active brain functions that lower body temperature. A population of neurons in the preoptic area was recently identified as such active torpor-regulating neurons. We hypothesized that the other hypothermia-inducing maneuvers would also activate these neurons. To test our hypothesis, we first refined the previous observations, examined the brain regions explicitly activated during the falling phase of body temperature using c-Fos expression, and confirmed the preoptic area. Next, we observed long-lasting hypothermia by reactivating torpor-tagged Gq-expressing neurons using the activity tagging and DREADD systems. Finally, we found that about 40-60% of torpor-tagged neurons were activated by succeeding isoflurane anesthesia and by icv administration of an adenosine A1 agonist. Isoflurane-induced and central adenosine-induced hypothermia is, at least in part, an active process mediated by the torpor-regulating neurons in the preoptic area.


Subject(s)
Adenosine , Isoflurane , Neurons , Preoptic Area , Animals , Preoptic Area/drug effects , Preoptic Area/metabolism , Isoflurane/pharmacology , Isoflurane/administration & dosage , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Body Temperature/drug effects , Body Temperature/physiology , Hypothermia/chemically induced , Hypothermia/metabolism , Torpor/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism
12.
Curr Opin Anaesthesiol ; 37(4): 379-383, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38842001

ABSTRACT

PURPOSE OF REVIEW: The environmental impact of anesthesia far exceeds that of other medical specialties due to our use of inhaled anesthetic agents (which are potent greenhouse gases) and many intravenous medications. RECENT FINDINGS: Calls for reducing the carbon footprint of anesthesia are ubiquitous in the anesthesia societies of developed nations and are appearing in proposed changes for hospital accreditation and funding in the United States. The body of research on atmospheric, land and water impacts of anesthetic pharmaceuticals is growing and generally reinforces existing recommendations to reduce the greenhouse gas emissions of anesthesia care. SUMMARY: The environmental impact of anesthesia care should factor into our clinical decisions. The onus is on clinicians to safely care for our patients in ways that contribute the least harm to the environment. Intravenous anesthesia and regional techniques have less environmental impact than the use of inhaled agents; efforts to reduce and properly dispose of pharmaceutical waste are central to reducing environmental burden; desflurane should not be used; nitrous oxide should be avoided except where clinically necessary; central nitrous pipelines should be abandoned; low fresh gas flows should be utilized whenever inhaled agents are used.


Subject(s)
Anesthetics, Inhalation , Humans , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/administration & dosage , Anesthetics/adverse effects , Anesthetics/administration & dosage , Carbon Footprint , Environment , Greenhouse Gases/adverse effects , Greenhouse Gases/analysis
13.
Exp Biol Med (Maywood) ; 249: 10037, 2024.
Article in English | MEDLINE | ID: mdl-38854792

ABSTRACT

In-ovo imaging using avian eggs has been described as a potential alternative to animal testing using rodents. However, imaging studies are hampered by embryonal motion producing artifacts. This study aims at systematically comparing isoflurane, desflurane and sevoflurane in three different concentrations in ostrich embryos. Biomagnetic signals of ostrich embryos were recorded analyzing cardiac action and motion. Ten groups comprising eight ostrich embryos each were investigated: Control, isoflurane (2%, 4%, and 6%), desflurane (6%, 12%, and 18%) and sevoflurane (3%, 5%, and 8%). Each ostrich egg was exposed to the same narcotic gas and concentration on development day (DD) 31 and 34. Narcotic gas exposure was upheld for 90 min and embryos were monitored for additional 75 min. Toxicity was evaluated by verifying embryo viability 24 h after the experiments. Initial heart rate of mean 148 beats/min (DD 31) and 136 beats/min (DD 34) decreased over time by 44-48 beats/minute. No significant differences were observed between groups. All narcotic gases led to distinct movement reduction after mean 8 min. Embryos exposed to desflurane 6% showed residual movements. Isoflurane 6% and sevoflurane 8% produced motion-free time intervals of mean 70 min after discontinuation of narcotic gas exposure. Only one embryo death occurred after narcotic gas exposure with desflurane 6%. This study shows that isoflurane, desflurane and sevoflurane are suitable for ostrich embryo immobilization, which is a prerequisite for motion-artifact free imaging. Application of isoflurane 6% and sevoflurane 8% is a) safe as no embryonal deaths occurred after exposure and b) effective as immobilization was observed for approx. 70 min after the end of narcotic gas exposure. These results should be interpreted with caution regarding transferability to other avian species as differences in embryo size and incubation duration exist.


Subject(s)
Desflurane , Embryo, Nonmammalian , Isoflurane , Struthioniformes , Animals , Struthioniformes/embryology , Embryo, Nonmammalian/drug effects , Anesthetics, Inhalation , Sevoflurane/adverse effects , Sevoflurane/pharmacology , Narcotics/toxicity , Immobilization
14.
Anesth Analg ; 139(1): 114-123, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38885399

ABSTRACT

BACKGROUND: Many studies have suggested that volatile anesthetic use may improve postoperative outcomes after cardiac surgery compared to total intravenous anesthesia (TIVA) owing to its potential cardioprotective effect. However, the results were inconclusive, and few studies have included patients undergoing heart valve surgery. METHODS: This nationwide population-based study included all adult patients who underwent heart valve surgery between 2010 and 2019 in Korea based on data from a health insurance claim database. Patients were divided based on the use of volatile anesthetics: the volatile anesthetics or TIVA groups. After stabilized inverse probability of treatment weighting (IPTW), the association between the use of volatile anesthetics and the risk of cumulative 1-year all-cause mortality (the primary outcome) and cumulative long-term (beyond 1 year) mortality were assessed using Cox regression analysis. RESULTS: Of the 30,755 patients included in this study, the overall incidence of 1-year mortality was 8.5%. After stabilized IPTW, the risk of cumulative 1-year mortality did not differ in the volatile anesthetics group compared to the TIVA group (hazard ratio, 0.98; 95% confidence interval, 0.90-1.07; P = .602), nor did the risk of cumulative long-term mortality (hazard ratio, 0.98; 95% confidence interval, 0.93-1.04; P = .579) at a median (interquartile range) follow-up duration of 4.8 (2.6-7.6) years. CONCLUSIONS: Compared with TIVA, volatile anesthetic use was not associated with reduced postoperative mortality risk in patients undergoing heart valve surgery. Our findings indicate that the use of volatile anesthetics does not have a significant impact on mortality after heart valve surgery. Therefore, the choice of anesthesia type can be based on the anesthesiologists' or institutional preference and experience.


Subject(s)
Anesthesia, Intravenous , Anesthetics, Inhalation , Heart Valves , Humans , Male , Female , Middle Aged , Anesthesia, Intravenous/adverse effects , Anesthesia, Intravenous/mortality , Aged , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/adverse effects , Republic of Korea/epidemiology , Heart Valves/surgery , Adult , Cardiac Surgical Procedures/mortality , Cardiac Surgical Procedures/adverse effects , Treatment Outcome , Retrospective Studies , Databases, Factual , Risk Factors , Postoperative Complications/mortality , Postoperative Complications/prevention & control , Postoperative Complications/epidemiology , Anesthesia, Inhalation/adverse effects , Anesthesia, Inhalation/mortality , Time Factors
15.
J Toxicol Sci ; 49(6): 269-279, 2024.
Article in English | MEDLINE | ID: mdl-38825486

ABSTRACT

Although morphine has been used for treatment-resistant dyspnea in end-stage heart failure patients, information on its cardiovascular safety profile remains limited. Morphine was intravenously administered to halothane-anesthetized dogs (n=4) in doses of 0.1, 1 and 10 mg/kg/10 min with 20 min of observation period. The low and middle doses attained therapeutic (0.13 µg/mL) and supratherapeutic (0.97 µg/mL) plasma concentrations, respectively. The low dose hardly altered any of the cardiovascular variables except that the QT interval was prolonged for 10-15 min after its start of infusion. The middle dose reduced the preload and afterload to the left ventricle for 5-15 min, then decreased the left ventricular contractility and mean blood pressure for 10-30 min, and finally suppressed the heart rate for 15-30 min. Moreover, the middle dose gradually but progressively prolonged the atrioventricular conduction time, QT interval/QTcV, ventricular late repolarization period and ventricular effective refractory period without altering the intraventricular conduction time, ventricular early repolarization period or terminal repolarization period. A reverse-frequency-dependent delay of ventricular repolarization was confirmed. The high dose induced cardiohemodynamic collapse mainly due to vasodilation in the initial 2 animals by 1.9 and 3.3 min after its start of infusion, respectively, which needed circulatory support to treat. The high dose was not tested further in the remaining 2 animals. Thus, intravenously administered morphine exerts a rapidly appearing vasodilator action followed by slowly developing cardiosuppressive effects. Morphine can delay the ventricular repolarization possibly through IKr inhibition in vivo, but its potential to develop torsade de pointes will be small.


Subject(s)
Anesthetics, Inhalation , Halothane , Heart Rate , Morphine , Animals , Dogs , Morphine/administration & dosage , Heart Rate/drug effects , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacokinetics , Male , Toxicokinetics , Dose-Response Relationship, Drug , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Blood Pressure/drug effects , Electrocardiography/drug effects , Female , Infusions, Intravenous , Vasodilation/drug effects , Electrophysiological Phenomena/drug effects
17.
Sci Rep ; 14(1): 14060, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890405

ABSTRACT

Isoflurane anesthesia (IA) partially compensates NREM sleep (NREMS) and not REM sleep (REMS) requirement, eliciting post-anesthetic REMS rebound. Sleep deprivation triggers compensatory NREMS rebounds and REMS rebounds during recovery sleep as a result of the body's homeostatic mechanisms. A combination of sleep deprivation and isoflurane anesthesia is common in clinical settings, especially prior to surgeries. This study investigates the effects of pre-anesthetic sleep deprivation on post-anesthetic sleep-wake architecture. The effects of isoflurane exposure (90 min) alone were compared with the effects of isoflurane exposure preceded by experimental sleep deprivation (6 h, gentle handling) on recovery sleep in adult mice by studying the architecture of post-anesthetic sleep for 3 consecutive post-anesthetic days. Effects of isoflurane anesthesia on recovery sleep developed only during the first dark period after anesthesia, the active phase in mice. During this time, mice irrespective of preceding sleep pressure, showed NREMS and REMS rebound and decreased wakefulness during recovery sleep. Additionally, sleep deprivation prior to isoflurane treatment caused a persistent reduction of theta power during post-anesthetic REMS at least for 3 post-anesthetic days. We showed that isoflurane causes NREMS rebound during recovery sleep which suggests that isoflurane may not fully compensate for natural NREMS. The study also reveals that isoflurane exposure preceded by sleep deprivation caused a persistent disruption of REMS quality. We suggest that preoperative sleep deprivation may impair postoperative recovery through lasting disruption in sleep quality.


Subject(s)
Anesthetics, Inhalation , Isoflurane , Sleep Deprivation , Sleep, REM , Wakefulness , Isoflurane/adverse effects , Isoflurane/pharmacology , Animals , Sleep Deprivation/physiopathology , Mice , Male , Anesthetics, Inhalation/adverse effects , Sleep, REM/drug effects , Wakefulness/drug effects , Wakefulness/physiology , Mice, Inbred C57BL , Electroencephalography , Sleep/drug effects , Sleep/physiology , Anesthesia/adverse effects
18.
J Cardiothorac Surg ; 19(1): 394, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937812

ABSTRACT

OBJECTIVE: Postoperative cognitive dysfunction (POCD) is a serious surgical complication. We assessed the different POCD incidences between anesthesia using sevoflurane and sevoflurane combined with dexmedetomidine, with propofol-based sedation in elderly patients who underwent a thoracic surgical procedure. METHODS: A total of 90 patients aged 65 to 80 years old who underwent a thoracic surgical procedure at our hospital and 15 nonsurgical participants as controls, were enrolled in this study. Patients were divided in a randomized 1:1:1 ratio into 3 groups. All participants were randomized into a trial with three anesthesia groups (P, PS, PSD) or a control group (C) of healthy matches. All trial groups received distinct anesthetic combinations during surgery, while controls mirrored patient criteria.Group P (propofol and remifentanil were maintained during the surgery), Group PS (propofol, remifentanil, and sevoflurane were maintained during the surgery), and Group PSD (propofol, remifentanil, sevoflurane, and dexmedetomidine were maintained during the surgery).All participants were rated using a series of cognitive assessment scales before and three days after surgery. All participants were interviewed over the telephone, 7 days, 30 days, and 90 days postoperatively. RESULTS: POCD incidences in the PSD (combined anesthetization with propofol, sevoflurane, and dexmedetomidine) group was significantly lower than that in the PS (combined anesthetization with propofol and sevoflurane) group, 1 day post-surgery (10.0% vs. 40.0%, P = 0.008), and the results were consistent at 3 days post-surgery. When the patients were assessed 7 days, 30 days, and 90 days postoperatively, there was no significant difference in POCD incidence among the three groups. Multivariate logistic regression analysis of POCD one day after surgery showed that education level was negatively correlated with incidence of POCD (P = 0.018) and single lung ventilation time was positively correlated with incidence of POCD (P = 0.001). CONCLUSION: For elderly patients who underwent a thoracic surgical procedure, dexmedetomidine sedation shows an obvious advantage on improving short-term POCD incidence, which is caused by sevoflurane.


Subject(s)
Dexmedetomidine , Postoperative Cognitive Complications , Propofol , Sevoflurane , Thoracic Surgical Procedures , Humans , Aged , Male , Female , Thoracic Surgical Procedures/adverse effects , Thoracic Surgical Procedures/methods , Postoperative Cognitive Complications/prevention & control , Postoperative Cognitive Complications/epidemiology , Postoperative Cognitive Complications/etiology , Double-Blind Method , Sevoflurane/administration & dosage , Sevoflurane/adverse effects , Aged, 80 and over , Dexmedetomidine/therapeutic use , Dexmedetomidine/administration & dosage , Propofol/adverse effects , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/administration & dosage , Cognition/drug effects , Incidence , Remifentanil/administration & dosage , Anesthetics, Intravenous/adverse effects
19.
Brain Stimul ; 17(3): 687-697, 2024.
Article in English | MEDLINE | ID: mdl-38821397

ABSTRACT

BACKGROUND: Dopaminergic neurons in the ventral tegmental area (VTA) are crucially involved in regulating arousal, making them a potential target for reversing general anesthesia. Electrical deep brain stimulation (DBS) of the VTA restores consciousness in animals anesthetized with drugs that primarily enhance GABAA receptors. However, it is unknown if VTA DBS restores consciousness in animals anesthetized with drugs that target other receptors. OBJECTIVE: To evaluate the efficacy of VTA DBS in restoring consciousness after exposure to four anesthetics with distinct receptor targets. METHODS: Sixteen adult Sprague-Dawley rats (8 female, 8 male) with bipolar electrodes implanted in the VTA were exposed to dexmedetomidine, fentanyl, ketamine, or sevoflurane to produce loss of righting, a proxy for unconsciousness. After receiving the dopamine D1 receptor antagonist, SCH-23390, or saline (vehicle), DBS was initiated at 30 µA and increased by 10 µA until reaching a maximum of 100 µA. The current that evoked behavioral arousal and restored righting was recorded for each anesthetic and compared across drug (saline/SCH-23390) condition. Electroencephalogram, heart rate and pulse oximetry were recorded continuously. RESULTS: VTA DBS restored righting after sevoflurane, dexmedetomidine, and fentanyl-induced unconsciousness, but not ketamine-induced unconsciousness. D1 receptor antagonism diminished the efficacy of VTA stimulation following sevoflurane and fentanyl, but not dexmedetomidine. CONCLUSIONS: Electrical DBS of the VTA restores consciousness in animals anesthetized with mechanistically distinct drugs, excluding ketamine. The involvement of the D1 receptor in mediating this effect is anesthetic-specific.


Subject(s)
Deep Brain Stimulation , Dexmedetomidine , Fentanyl , Rats, Sprague-Dawley , Sevoflurane , Unconsciousness , Ventral Tegmental Area , Animals , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Sevoflurane/pharmacology , Dexmedetomidine/pharmacology , Male , Fentanyl/pharmacology , Rats , Female , Unconsciousness/chemically induced , Unconsciousness/therapy , Consciousness/drug effects , Consciousness/physiology , Ketamine/pharmacology , Anesthetics, Inhalation/pharmacology
20.
Braz J Med Biol Res ; 57: e13437, 2024.
Article in English | MEDLINE | ID: mdl-38808889

ABSTRACT

Clinical studies have found that neonatal sevoflurane exposure can increase the risk of cognitive dysfunction. However, recent studies have found that it can exhibit neuroprotective effects in some situations. In this study, we aimed to explore the effects of sevoflurane neonatal exposure in rats. A total of 144 rat pups (72 males and 72 females) were assigned to six groups and separately according to sevoflurane exposure of different times on the seventh day after birth. Blood gas analysis and western blot detection in the hippocampus were conducted after exposure. The Morris water maze test was conducted on the 32nd to 38th days after birth. The expression of PSD95 and synaptophysin in the hippocampus was detected after the Morris water maze test. We found that neonatal exposure to sevoflurane promoted apoptosis in the hippocampus, and Bax and caspase-3 were increased in a dose-dependent manner. The 2-h exposure had the greatest effects on cognitive dysfunction. However, with the extension of exposure time to 6 h, the effects on cognitive function were partly compensated. In addition, sevoflurane exposure decreased synaptogenesis in the hippocampus. However, as the exposure time was extended, the suppression of synaptogenesis was attenuated. In conclusion, neonatal sevoflurane exposure exhibited duration-dependent effects on cognitive function via Bax-caspase-3-dependent apoptosis and bidirectional effects on synaptogenesis in rats.


Subject(s)
Animals, Newborn , Cognition , Hippocampus , Sevoflurane , Sevoflurane/pharmacology , Animals , Female , Male , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Cognition/drug effects , Time Factors , Maze Learning/drug effects , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/adverse effects , Apoptosis/drug effects , Sex Factors , Rats, Sprague-Dawley , Methyl Ethers/pharmacology , Blotting, Western , Blood Gas Analysis , Cognitive Dysfunction/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...