Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
Turk J Med Sci ; 54(1): 1-15, 2024.
Article in English | MEDLINE | ID: mdl-38812654

ABSTRACT

Background/aim: This study aims to determine the possible embryotoxic effects of propofol on the cerebellum and spinal cord using fertile chicken eggs. Materials and methods: A total of 430 fertile eggs were divided into 5 groups: control, saline, 2.5 mg.kg-1, 12.5 mg.kg-1, and 37.5 mg.kg-1 propofol. Injections were made immediately before incubation via the air chamber. On the 15th, 18th, and 21st day of incubation, 6 embryos from each group were evaluated. Serial paraffin sections taken from the cerebellum and spinal cord were stained with hematoxylin-eosin, Kluver-Barrera, toluidine blue, and periodic acid-Schiff's reaction. The outer granular layer and total cortex thickness were measured, and the linear density of the Purkinje cells was determined. The ratios of the substantia grisea surface area to the total surface area of the spinal cord were calculated. The transverse and longitudinal diameters of the canalis centralis were also assessed. Results: No structural malformation was observed in any embryos examined macroscopically. No significant difference was observed between the groups in terms of development and histologic organization of the cerebellum and spinal cord. However, on the 15th, 18th, and 21st day, the outer granular layer (p < 0.001 for all days) and the total cortex thickness (p < 0.01, p < 0.001, and p < 0.001, respectively) decreased significantly in different propofol dose groups in varying degrees in the cerebellum. Similarly, in the spinal cord, there were significant changes in the ratios of the substantia grisea surface area to the total surface area (p < 0.01 and p < 0.001, respectively). Conclusion: It was concluded that the in-ovo-administered propofol given immediately before incubation has adverse effects on the developing cerebellum and spinal cord. Therefore, it is important for anesthesiologists always to remain vigilant when treating female patients of childbearing age.


Subject(s)
Cerebellum , Propofol , Spinal Cord , Animals , Propofol/toxicity , Propofol/administration & dosage , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/embryology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/embryology , Chick Embryo/drug effects , Anesthetics, Intravenous/toxicity , Anesthetics, Intravenous/administration & dosage
2.
Mol Neurobiol ; 61(1): 519-532, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37644280

ABSTRACT

Propofol is one of the most used intravenous anesthetic agents, which is widely used in clinical anesthesia induction and maintenance of pediatric patients. Exposure of the developing brain to propofol has been reported to lead to adverse brain changes, which in turn can induce persistent behavioral abnormalities in adulthood. However, the mechanisms by which propofol exposure in the developing brain induces cognitive impairment remain unclear. Here we report that repeated propofol exposure during the second postnatal week impairs spatial learning and memory in young mice. The reduced excitatory synaptic function and synaptogenesis in hippocampal CA1 neurons underlie this cognitive impairment. Propofol exposure specifically activates Toll-like receptor 4 (TLR4)-myeloid differentiation primary response protein 88 (MyD88)-NF-κB signaling pathway. TLR4 deficiency recues propofol exposure-induced synaptic function and cognitive deficits in young mice. Thus, we provide evidence that the activation of the TLR4-mediated pathway by propofol exposure may serve as a crucial trigger for the cognitive impairment in young adulthood caused by repeated exposure to propofol in the developing brain.


Subject(s)
Cognitive Dysfunction , Propofol , Animals , Mice , Anesthetics, Intravenous/toxicity , Cognition , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Neuronal Plasticity , Propofol/pharmacology , Toll-Like Receptor 4/metabolism
3.
Biomed Pharmacother ; 144: 112309, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34653761

ABSTRACT

Anesthetics are extensively used during cancer surgeries. The progression of cancer can be influenced by perioperative events such as exposure to general or local anesthesia. However, whether they inhibit cancer or act as a causative factor for metastasis and exert deleterious effects on cancer growth differs based on the type of cancer and the therapy administration. Recent experimental data suggested that many of the most commonly used anesthetics in surgical oncology, whether general or local agents, can alter gene expression and cause epigenetic changes via modulating miRNAs. miRNAs are single-stranded non-coding RNAs that regulate gene expression at various levels, and their dysregulation contributes to the pathogenesis of cancers. However, anesthetics via regulating miRNAs can concurrently target several effectors of cellular signaling pathways involved in cell differentiation, proliferation, and viability. This review summarized the current research about the effects of different anesthetics in regulating cancer, with a particular emphasis on the role of miRNAs. A significant number of studies conducted in this area of research illuminate the effects of anesthetics on the regulation of miRNA expression; therefore, we hope that a thorough understanding of the underlying mechanisms involved in the regulation of miRNA in the context of anesthesia-induced cancer regulation could help to define optimal anesthetic regimens and provide better perspectives for further studies.


Subject(s)
Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Anesthetics, Local/pharmacology , MicroRNAs/metabolism , Neoplasms/drug therapy , Propofol/pharmacology , Anesthetics, Inhalation/toxicity , Anesthetics, Intravenous/toxicity , Anesthetics, Local/toxicity , Animals , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway
4.
Neurotox Res ; 39(5): 1449-1458, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34216363

ABSTRACT

It has been widely reported that severe neurotoxicity can be induced by the application of propofol, which is closely related to the disruption of the blood-brain barrier (BBB) induced by inflammation and injury in the human brain microvascular endothelial cells (HBMVECs). Benzbromarone is a classic anti-gout agent that has been recently reported to exert anti-inflammatory and anti-oxidative stress effects. In the present study, we aim to investigate the protective property of Benzbromarone against propofol-induced injury on HBMVECs and the underlying mechanism. CCK8 assay was used to detect the cell viability of treated HBMVECs. Oxidative stress in HBMVECs was evaluated by measuring the levels of MDA and mitochondrial ROS. ELISA and qRT-PCR assay were used to determine the production of IL-1ß, IL-8, MCP-1, ICAM-1, and VCAM-1 by treated HBMVECs. Calcein-AM staining was utilized to evaluate the attachment of U937 monocytes to HBMVECs. The expression level of Egr-1 was determined by qRT-PCR and Western blot assay. Firstly, the decreased cell viability of HBMVECs induced by propofol was significantly elevated by treatment with Benzbromarone. The increased levels of MDA and mitochondrial ROS induced by propofol were dramatically suppressed by Benzbromarone. Secondly, the excessive production of inflammatory factors (IL-1ß, IL-8, and MCP-1) and adhesion molecules (ICAM-1 and VCAM-1) triggered by propofol was pronouncedly inhibited by Benzbromarone. Benzbromarone ameliorated propofol-induced attachment of U937 monocytes to HBMVECs. Lastly, Benzbromarone downregulated propofol-induced expression of the transcriptional factor Egr-1 in HBMVECs. Benzbromarone protected against propofol-induced inflammation and injury through suppressing Egr-1 in human brain vascular endothelial cells.


Subject(s)
Benzbromarone/pharmacology , Brain/drug effects , Endothelial Cells/drug effects , Microvessels/drug effects , Neuroprotective Agents/pharmacology , Propofol/toxicity , Anesthetics, Intravenous/toxicity , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/cytology , Brain/pathology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Endothelial Cells/pathology , Humans , Microvessels/cytology , Microvessels/pathology
5.
Br J Anaesth ; 126(6): 1141-1156, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33641936

ABSTRACT

BACKGROUND: Both animal and retrospective human studies have linked extended and repeated general anaesthesia during early development with cognitive and behavioural deficits later in life. However, the neuronal circuit mechanisms underlying this anaesthesia-induced behavioural impairment are poorly understood. METHODS: Neonatal mice were administered one or three doses of propofol, a commonly used i.v. general anaesthetic, over Postnatal days 7-11. Control mice received Intralipid® vehicle injections. At 4 months of age, the mice were subjected to a series of behavioural tests, including motor learning. During the process of motor learning, calcium activity of pyramidal neurones and three classes of inhibitory interneurones in the primary motor cortex were examined in vivo using two-photon microscopy. RESULTS: Repeated, but not a single, exposure of neonatal mice to propofol i.p. caused motor learning impairment in adulthood, which was accompanied by a reduction of pyramidal neurone number and activity in the motor cortex. The activity of local inhibitory interneurone networks was also altered: somatostatin-expressing and parvalbumin-expressing interneurones were hypoactive, whereas vasoactive intestinal peptide-expressing interneurones were hyperactive when the mice were performing a motor learning task. Administration of low-dose pentylenetetrazol to attenuate γ-aminobutyric acid A receptor-mediated inhibition or CX546 to potentiate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-subtype glutamate receptor function during emergence from anaesthesia ameliorated neuronal dysfunction in the cortex and prevented long-term behavioural deficits. CONCLUSIONS: Repeated exposure of neonatal mice to propofol anaesthesia during early development causes cortical circuit dysfunction and behavioural impairments in later life. Potentiation of neuronal activity during recovery from anaesthesia reduces these adverse effects of early-life anaesthesia.


Subject(s)
Anesthetics, Intravenous/toxicity , Behavior, Animal/drug effects , Maze Learning/drug effects , Motor Activity/drug effects , Motor Cortex/drug effects , Neurotoxicity Syndromes/etiology , Propofol/toxicity , Animals , Animals, Newborn , Calcium Signaling/drug effects , Elevated Plus Maze Test , Excitatory Amino Acid Agonists/pharmacology , GABA Antagonists/pharmacology , Interneurons/drug effects , Interneurons/metabolism , Mice, Transgenic , Motor Cortex/metabolism , Motor Cortex/physiopathology , Neural Inhibition/drug effects , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/prevention & control , Neurotoxicity Syndromes/psychology , Open Field Test/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Social Behavior
6.
Neurosci Lett ; 740: 135461, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33115643

ABSTRACT

BACKGROUND: Elderly patients receive propofol at regular intervals for sedation during gastrointestinal endoscopy. However, the link between cognition and intermittent propofol exposure remains unclear. Thus, we used aged rats to investigate the effect of propofol on cognition. METHODS: The study included two parts. In the first part, aged (18-20 months old) male Sprague-Dawley rats underwent intermittent intraperitoneal injection of propofol (200 mg/kg) or intralipid, every 9 days or once a day. In the second part, some aged rats received intraperitoneal injection of Bay 11-7082 (1 mg/kg), a specific inhibitor of NF-κB, 30 min before propofol injection. Memory tests were performed to evaluate cognition 24 h after the entire treatment. The hippocampal neuronal damage was assessed by TUNEL staining. The hippocampal levels of p-NF-κB p65, NLRP3, caspase-1 p20, and cleaved caspase-3 were detected by western blotting. The hippocampal and serum levels of IL-1ß, IL-6, and TNF-α were evaluated using ELISA. RESULTS: There were no differences in the behavioral tests, hippocampal neuronal damage, and neuroinflammation between groups given intralipid and propofol treatment every 9 days. However, repeated propofol treatment once a day promoted activation of NF-κB and the NLRP3 inflammasome, inducing cognitive impairment and neuroinflammation. Interestingly, pretreatment with Bay-11-7082 not only inhibited NF-κB/NLRP3 inflammasome activation, but also attenuated neuronal damage and cognitive dysfunction in aged rats exposed to daily propofol treatment. CONCLUSIONS: Intermittent propofol treatment every 9 days may be safe for aged rats. However, propofol treatment once a day could impair the cognition of aged rats, partly through the activation of the NF-κB pathway and NLRP3 inflammasome, which may be a potential targets for the treatment of cognitive impairment in elderly patients.


Subject(s)
Anesthetics, Intravenous/toxicity , Cognition Disorders/chemically induced , Inflammasomes/drug effects , NF-kappa B/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neurons/pathology , Propofol/toxicity , Aging/psychology , Animals , Cognition/drug effects , Cognition Disorders/psychology , Conditioning, Operant/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Memory/drug effects , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
7.
Biomarkers ; 25(8): 685-692, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33032457

ABSTRACT

PURPOSE: Propofol, an aesthetic agent in paediatric patients, results in neurotoxicity in the developing neurons. To reduce side effects of propofol, the protective role of miR-455-3p (microRNA-455-3p) in developing rat brain was investigated. MATERIALS AND METHODS: Primary hippocampal neurons were isolated from postnatal day 1 or 2 SD (Sprague-Dawley) rats. The neurons were exposed to various concentrations of propofol (0, 10, 30, or 50 µM) for 6 h. Propofol-induced cell viability was assessed by MTT assay, expression levels of miR-455-3p and EphA4 (erythropoietin-producing hepatocellular A4) in propofol-induced neurons were determined using qRT-PCR and western blot, respectively. Binding ability between miR-455-3p and EphA4 was predicted, and then validated by luciferase reporter assay. Neurons expressing miR-455-3p mimics, were treated with 50 µM propofol for 6 h, and apoptosis status was evaluated by flow cytometry. RESULTS: Exposure to propofol significantly decreased cell viability of developing neurons isolated from neonatal rats. Propofol decreased miR-455-3p expression, while increased EphA4 level in the neurons. miR-455-3p mimics increased propofol-induced reduce in cell viability, and attenuated propofol-induced cell apoptosis of neurons. MiR-455-3p could target EphA4, and decreased expression of EphA4 in neurons exposure to propofol. EphA4 knockdown counteracted with the promotive effects of propofol on cell viability and apoptosis of neurons. CONCLUSION: Propofol treatment induces neurotoxicity and suppresses miR-455-3p levels in the developing hippocampal neurons. However, miR-455-3p could alleviate such neurotoxicity by reducing EphA4 expression, provided new insights into miR-455-3p as novel therapeutic target to prevent propofol-induced damages from bench to clinic.


Subject(s)
Anesthetics, Intravenous/toxicity , Hippocampus/drug effects , MicroRNAs/metabolism , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Propofol/toxicity , Receptor, EphA4/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Hippocampus/metabolism , Hippocampus/pathology , MicroRNAs/genetics , Neurons/metabolism , Neurons/pathology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Rats, Sprague-Dawley , Receptor, EphA4/genetics
8.
Eur J Pharmacol ; 884: 173303, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32681942

ABSTRACT

Propofol, most frequently used as a general anesthetic due to its versatility and short-acting characteristics, is thought to exert its anesthetic actions via GABAA receptors; however, the precise mechanisms of its adverse action including angialgia remain unclear. We examined the propofol-induced elevation of intracellular calcium and morphological changes in intracellular organelles using SHSY-5Y neuroblastoma cells, COS-7 cells, HEK293 cells, and HUVECs loaded with fluorescent dyes for live imaging. Although propofol (>50 µM) increased intracellular calcium in a dose-dependent manner in these cells, it was not influenced by the elimination of extracellular calcium. The calcium elevation was abolished when intracellular or intraendoplasmic reticulum (ER) calcium was depleted by BAPTA-AM or thapsigargin, respectively, suggesting that calcium was mobilized from the ER. Studies using U-73122, xestospongin C, and dantrolene revealed that propofol-induced calcium elevation was not mediated by G-protein coupled receptors, IP3 receptors, or ryanodine receptors. We performed live imaging of the ER, mitochondria and Golgi apparatus during propofol stimulation using fluorescent dyes. Concomitant with the calcium elevation, the structure of the ER and mitochondria was fragmented and aggregated, and these changes were not reversed during the observation period, suggesting that propofol-induced calcium elevation occurs due to calcium leakage from these organelles. Although the concentration of propofol used in this experiment was greater than that used clinically (30 µM), it is possible that the concentration exceeds 30 µM at the site where propofol is injected, leading the idea that these phenomena might relate to the various propofol-induced adverse effects including angialgia.


Subject(s)
Anesthetics, Intravenous/toxicity , Calcium Signaling/drug effects , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Mitochondria/drug effects , Propofol/toxicity , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Time Factors
9.
J Appl Toxicol ; 40(6): 855-863, 2020 06.
Article in English | MEDLINE | ID: mdl-31998977

ABSTRACT

Prenatal propofol exposure induced neurotoxicity in the developing brains and led to persistent learning deficits in the offspring. Our goal was to use zebrafish to explore whether the decline in learning and memory was correlated with inhibition of neuronal growth after propofol exposure. Zebrafish embryos at 6 hours postfertilization (hpf) were exposed to control or 1, 2 or 4 µg/mL propofol until 48 hpf. Spontaneous locomotor activity and swimming behavior in response to dark-to-light photoperiod stimulation were studied in zebrafish larvae at 6 days postfertilization (dpf). The adaptability to repeated stimulation was used to indicate learning and memory function of larvae. Transgenic NBT line zebrafish was used to quantitate the effect of propofol on motor neuronal growth of embryos in vivo. Six dpf transgenic zebrafish larvae went through photoperiod stimulation after their neuronal length had been analyzed during the embryonic period. Our data indicate that embryonic exposure to 1, 2 and 4 µg/mL propofol had no adverse effect on spontaneous movement in zebrafish larvae, but 2 and 4 µg/mL propofol significantly impaired the learning and memory function of larvae. Moreover, propofol significantly inhibited axonal growth of motor neurons during the embryonic stage, which was correlated with learning and memory deficiency in larvae. Our findings demonstrate that the neuronal growth was correlated with learning and memory function, indicating the relevance of zebrafish as a new model to explore the mechanisms through which propofol induces long-term learning and memory impairment.


Subject(s)
Anesthetics, Intravenous/toxicity , Axons/drug effects , Behavior, Animal/drug effects , Embryo, Nonmammalian/drug effects , Locomotion/drug effects , Propofol/toxicity , Animals , Animals, Genetically Modified , Axons/pathology , Learning/drug effects , Memory/drug effects , Photoperiod , Risk Assessment , Swimming , Zebrafish/embryology , Zebrafish/genetics
10.
Anesth Analg ; 130(1): e1-e4, 2020 01.
Article in English | MEDLINE | ID: mdl-30198930

ABSTRACT

The understanding of anesthetic side effects on the heart has been hindered by the lack of sophisticated clinical models. Using micropatterned human-induced pluripotent stem cell-derived cardiomyocytes, we obtained cardiac muscle depressant profiles for propofol, etomidate, and our newly identified anesthetic compound KSEB01-S2. Propofol was the strongest depressant among the 3 compounds tested, exhibiting the largest decrease in contraction velocity, depression rate, and beating frequency. Interestingly, KSEB01-S2 behaved similarly to etomidate, suggesting a better cardiac safety profile. Our results provide a proof-of-concept for using human-induced pluripotent stem cell-derived cardiomyocytes as an in vitro platform for future drug design.


Subject(s)
Anesthetics, Intravenous/toxicity , Etomidate/toxicity , Heart Diseases/chemically induced , Heart Rate/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Propofol/toxicity , Adult , Cardiotoxicity , Cell Line , Female , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Myocytes, Cardiac/pathology , Proof of Concept Study , Risk Assessment , Time Factors , Young Adult
11.
Neurosci Lett ; 716: 134647, 2020 01 18.
Article in English | MEDLINE | ID: mdl-31765729

ABSTRACT

BACKGROUND: This study was to investigate the neuroprotective effect of erythropoietin (EPO) on hippocampal neuronal cell injury in developing rats. METHODS: The hippocampal neurons cells were obtained from SD rats aged 10 days and divided into control, propofol, EPO, and propofol + erythropoietin (E + P) groups. Cell proliferation and apoptosis were measured by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Ki-67 immunofluorescence, and flow cytometry, respectively. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, IL-4 and IL-10 were detected by enzyme-linked immunosorbent assay (ELISA). Cellular immunohistochemistry was utilized to detect the expression of proliferating cell nuclear antigen (PCNA), nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3). Quantitative real time polymerase chain reaction (qRT-PCR) and western blot were used to detect the expression of Bax, Bcl-2, Caspase-3, toll-like receptor 4 (TLR4) and p65. Furthermore, TLR4 antagonist (TAK-242) and activator (LPS) were used to study the relationship between EPO and TLR4. RESULTS: Propofol treatment caused morphological and structural damage of hippocampal neurons. However, EPO significantly improved this damage, enhanced cell proliferation, decreased apoptosis and pro-inflammatory factor content, up-regulated the expression of Ki-67, PCNA, Bcl-2, NGF, BDNF and NT-3, as well as decreased the expression of Bax, Caspase-3, TLR4 and p65 (p < 0.05). After TAK-242 or LPS treatment, it showed similar results in propofol + TAK-242 (T + P) group and E + P group. CONCLUSION: Erythropoietin could attenuate propofol-induced hippocampal neuronal cell injury in developing rats, which may be related to inhibit TLR4 expression.


Subject(s)
Anesthetics, Intravenous/toxicity , Erythropoietin/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Propofol/toxicity , Toll-Like Receptor 4/drug effects , Animals , Cells, Cultured , Female , Hippocampus/metabolism , Male , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism
12.
J Appl Toxicol ; 40(5): 679-690, 2020 05.
Article in English | MEDLINE | ID: mdl-31867768

ABSTRACT

ET-26 hydrochloride (ET-26HCl) is a novel etomidate analogue, approved for clinical trials, which has an effective sedative-hypnotic effect, a stable myocardial performance, and milder adrenocortical suppression than etomidate in rats and beagle dogs. Additionally, ET-26HCl showed similar hemodynamic stability as etomidate in the rat uncontrolled hemorrhagic shock model. Furthermore, ET-26HCl, in the rat lipopolysaccharide-induced sepsis model, was found to have a higher survival rate, a lower inflammatory reaction, and less organ injury. In the present study, we measured the potential adverse effects of ET-26HCl in beagle dogs in accordance with the Guidance on single- and repeated-dose toxicity published by the China Food and Drug Administration. In toxicity studies, single and repeated (14 days) intravenous doses of up to 16 mg/kg were well tolerated, with only pharmacologically related clinical signs seen in both studies. Thus, the no-observed-adverse-effect level (NOAEL) of ET-26HCl was found at 16 mg/kg/day. Toxicokinetic examination demonstrated that ET-26HCl showed a dose-dependent increase to exposure, no gender difference, and no evidence of accumulation. These results provide useful information for guiding a phase I clinical trial of ET-26HCl in healthy volunteers.


Subject(s)
Anesthetics, Intravenous/toxicity , Etomidate/analogs & derivatives , Toxicity Tests , Administration, Intravenous , Anesthetics, Intravenous/administration & dosage , Animals , Dogs , Etomidate/administration & dosage , Etomidate/toxicity , Female , Male , No-Observed-Adverse-Effect Level , Risk Assessment , Toxicokinetics
13.
Bioengineered ; 10(1): 493-500, 2019 12.
Article in English | MEDLINE | ID: mdl-31656124

ABSTRACT

We aimed to evaluate the neurotoxicity and mechanisms of anesthetics propofol in hESC-derived neurons. Cell apoptosis in hESC-derived neurons' exposure to 4, 10 and 20 µg/mL propofol for 6 h was assessed using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick end labeling (TUNEL) staining and microRNA-665 (miR-665) expression was assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR). miR-665 was overexpressed and knocked down using a miR-665 mimic and anti-665 transfection, respectively. The results showed that hESCs exposed to propofol showed a dose-dependent cell apoptosis, followed by the upregulation of miR-665 expression. Overexpression of miR-665 increased propofol-induced apoptosis in hESC cells. And targeting miR-665 decreased propofol-induced cell apoptosis in hESC cells. These data suggest that propofol induces cell death in hESC-derived neurons and the propofol-induced cell apoptosis may occur via miR-665-dependent mechanism.


Subject(s)
Anesthetics, Intravenous/toxicity , Apoptosis/drug effects , MicroRNAs/metabolism , Neurons/drug effects , Propofol/toxicity , Stem Cells/drug effects , Animals , Cells, Cultured , Humans , Mice , MicroRNAs/genetics , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism
14.
Neurotoxicology ; 74: 67-73, 2019 09.
Article in English | MEDLINE | ID: mdl-31085211

ABSTRACT

BACKGROUND: Identifying chemicals with narcotic potency is an important aspect of assessing the safety of consumer products that may be accidentally ingested. A rapid and efficient assay of narcotic potency is desired for assessing chemicals with such suspected activity. OBJECTIVES: This purpose of this research was to develop a non-mammalian vertebrate, high throughput, neurobehavioral method to assess the narcotic potency of chemicals using larval zebrafish. METHODS: Larval zebrafish were acutely exposed to chemicals beginning at 5 days post fertilization (5 dpf). Locomotor activity, elicited by regular, periodic photostimulation, was quantified using a video tracking apparatus. Narcotic potency was determined as the molar concentration at which photostimulated locomotor activity was reduced by 50% (IC50). Toxicity was assessed based on observations of morbidity or mortality. Recovery was assessed following removal of test material by serial dilution and reassessment of photostimulated behavior 24 hr later (6 dpf). RESULTS: A total of 21 chemicals were assessed. Etomidate, a human narcotic analgesic agent, was used as a reference material. Investigating a series of eleven linear, primary alcohols (C6 to C16), a relationship between narcotic potency and carbon number was observed; narcotic potency increased with carbon number up to C12, consistent with historical studies. For a set of technical grade surfactants, nonionic surfactants (i.e., alcohol ethoxylates) were observed to be narcotic agents while anionic surfactants produced evidence of reduced locomotor activity only in combination with toxicity. Of the solvents evaluated, only ethanol exhibited narcotic activity with an IC50 of 261 mM and was the least potent of the chemicals investigated. Etomidate was the most potent material evaluated with an IC50 of 0.39 µM. CONCLUSIONS: The larval zebrafish neurobehavioral assay provides a method capable of estimating the narcotic potency of chemicals and can identify if toxicity contributes to observed neurobehavioral effects in the test organism.


Subject(s)
Behavior, Animal/drug effects , Larva/drug effects , Narcotics/pharmacology , Zebrafish , Alcohols/chemistry , Alcohols/toxicity , Anesthetics, Intravenous/toxicity , Animals , Embryonic Development/drug effects , Etomidate/toxicity , Motor Activity/drug effects , Narcotics/toxicity , Photic Stimulation , Solvents/toxicity , Structure-Activity Relationship , Surface-Active Agents/toxicity
15.
J Alzheimers Dis ; 67(1): 137-147, 2019.
Article in English | MEDLINE | ID: mdl-30636740

ABSTRACT

BACKGROUND: Disruption of intracellular Ca2+ homeostasis and associated autophagy dysfunction contribute to neuropathology in Alzheimer's disease (AD). OBJECTIVE: To study the effects of propofol on cell viability via its effects on intracellular Ca2+ homeostasis, and the impact of autophagy, in a neuronal model of presenilin-mutated familial AD (FAD). METHODS: We treated PC12 cells, stably transfected with either mutated presenilin-1 (L286V) or wild type (WT) controls, with propofol at different doses and durations, in the presence or absence of extracellular Ca2+, antagonists of inositol trisphosphate receptors (InsP3R, xestospongin C) and/or ryanodine receptors (RYR, dantrolene), or an inhibitor of autophagy flux (Bafilomycin). We determined cell viability, cytosolic Ca2+ concentrations ([Ca2+]c), vATPase protein expression, and lysosomal acidification. RESULTS: The propofol dose- and time-dependently decreased cell viability significantly more in L286V than WT cells, especially at the pharmacological dose (>50µM), and together with bafilomycin (40 nM). Clinically used concentrations of propofol (<20µM) tended to increase cell viability. Propofol significantly increased [Ca2+]c more in L286V than in WT cells, which was associated with decrease of vATPase expression and localization to the lysosome. Both toxicity and increased Ca2+ levels were ameliorated by inhibiting InsP3R/RYR. However, the combined inhibition of both receptors paradoxically increased [Ca2+]c, by inducing Ca2+ influx from the extracellular space, causing greater cytotoxicity. CONCLUSION: Impairment in autophagy function acts to deteriorate cell death induced by propofol in FAD neuronal cells. Cell death is ameliorated by either RYR or InsP3R antagonists on their own, but not when both are co-administered.


Subject(s)
Alzheimer Disease/genetics , Anesthetics, Intravenous/toxicity , Autophagy/genetics , Calcium Metabolism Disorders/genetics , Calcium Metabolism Disorders/pathology , Neurons/drug effects , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Presenilin-1/genetics , Propofol/toxicity , Adenosine Triphosphatases/biosynthesis , Animals , Calcium Metabolism Disorders/metabolism , Humans , Neurotoxicity Syndromes/metabolism , PC12 Cells , Rats , Ryanodine Receptor Calcium Release Channel/drug effects
16.
Anesth Analg ; 129(2): 608-617, 2019 08.
Article in English | MEDLINE | ID: mdl-30303867

ABSTRACT

BACKGROUND: Propofol can cause degeneration of developing brain cells and subsequent long-term learning or memory impairment. However, at the early stage of embryonic development, the molecular mechanism of propofol-induced inhibition in neural stem cells (NSCs) neurogenesis is still unclear. The aim of this study was to determine the role of propofol in NSCs neurogenesis and, more importantly, to explore the underlying mechanism. METHODS: First, a single intraperitoneal injection of propofol was performed in pregnant mice, and 6 hours after administration of propofol, the hippocampus RNA and the protein of the embryos' brains was extracted to analyze the expression of neuron-specific markers. Second, the primary NSCs were isolated from the hippocampus of mouse embryonic brain and then treated with propofol for cell viability, immunostaining, and transwell assays; more importantly, we performed RNA sequencing (RNA-seq) and q-reverse transcription polymerase chain reaction assays to identify genes regulated by propofol; the Western blot, small interfering RNA (SiRNA), and luciferase reporter assays were used to study the effects of propofol on calmodulin-dependent protein kinase (CaMk) II/5' adenosine monophosphate-activated protein kinase (AMPK)/activating transcription factor 5 (ATF5) signaling pathway. RESULTS: Our results indicated that propofol treatment could inhibit the proliferation, migration, and differentiation of NSCs. The results of RNA-seq assays showed that propofol treatment resulted in downregulation of a group of Ca-dependent genes. The following mechanism studies showed that propofol regulates the proliferation, differentiation, and migration of NSCs through the CaMkII/phosphorylation of serine at amino acid position 485 (pS485)/AMPK/ATF5 signaling pathway. CONCLUSIONS: The results from study demonstrated that propofol inhibits the proliferation, differentiation, and migration of NSCs, and these effects are partially mediated by CaMkII/pS485/AMPK/ATF5 signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Activating Transcription Factors/metabolism , Anesthetics, Intravenous/toxicity , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Proliferation/drug effects , Hippocampus/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Propofol/toxicity , AMP-Activated Protein Kinases/genetics , Activating Transcription Factors/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Movement/drug effects , Cells, Cultured , Gene Expression Regulation , Hippocampus/enzymology , Hippocampus/pathology , Mice, Inbred C57BL , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Signal Transduction
17.
Neurotoxicology ; 69: 320-329, 2018 12.
Article in English | MEDLINE | ID: mdl-30107222

ABSTRACT

Anesthetic-induced neurotoxicity in the developing brain is a concern. This neurotoxicity is closely related to anesthetic exposure time, dose, and developmental stages. Using calcium imaging and morphological examinations in vitro, we sought to determine whether intravenous anesthetic-induced direct neurotoxicity varies according to different stages of the days in vitro (DIV) of neurons in primary culture. Cortical neurons from E17 Wistar rats were prepared. On DIV 3, 7, and 13, cells were exposed to the intravenous anesthetics thiopental sodium (TPS), midazolam (MDZ), or propofol (PPF), to investigate direct neurotoxicity using morphological experiments. Furthermore, using calcium imaging, the anesthetic-induced intracellular calcium concentration ([Ca2+]i) elevation was monitored in cells on DIV 4, 8, and 13. All anesthetics elicited significant [Ca2+]i increases on DIV 4. While TPS (100 µM) and MDZ (10 µM) did not alter neuronal death, PPF (10 µM and 100 µM) decreased the survival ratio (SR) significantly. On DIV 8, TPS and MDZ did not elicit [Ca2+]i elevation or SR decrease, while PPF still induced [Ca2+]i elevation (both at 10 µM and 100 µM) and significant SR decrease at 100 µM (0.76 ± 0.03; P < 0.05), but not at 10 µM (0.91 ± 0.03). Such anesthetic-induced [Ca2+]i elevation and SR decrease were not observed on DIV 13-14 for any of the anesthetic drugs. Our study indicates that more caution may be exercised when using PPF compared to TPS or MDZ during development.


Subject(s)
Aging/metabolism , Anesthetics, Intravenous/toxicity , Calcium/physiology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Aging/drug effects , Anesthetics, Intravenous/administration & dosage , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Female , Pregnancy , Rats , Rats, Wistar
18.
Neurosci Lett ; 675: 152-159, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29578002

ABSTRACT

AIM: To investigate the protective effect of microRNA-34a (miR-34a) on propofol-induced neurotoxicity and cognitive dysfunction. METHODS: After SH-SY5Y cells were treated with propofol to induce neurotoxicity, microRNA-34a mimics and inhibitors were transfected into the cells. The expression of apoptosis-related genes and the proteins were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot. Sprague-Dawley (SD) rats received intraperitoneal injections of propofol, and were treated with microRNA-34a mimics and lentivirus-mediated microRNA-34a inhibitors. The Morris water maze (MWM) test was used to detect changes in motor function. RESULTS: Propofol anesthesia had an adverse effect on cell survival due to the increased expression of apoptosis-related genes such as cleaved caspase-3/8 and Bax, which was accompanied by reduced expression of ERK1/2, pERK1/2, and phosphorylated NF-kappaB p65 both in vivo and in vitro. Unexpectedly, microRNA-34a was upregulated after propofol treatment, and the inhibitors protected the SH-SY5Y cells from propofol-induced apoptosis. The microRNA-34a inhibitor suppressed the apoptosis-induced effects of propofol. This protection may have been partly diminished by PD98059, a MAPK kinase inhibitor. MicroRNA-34a inhibited or reverted the reduced expression of ERK1/2 and upregulated the expression of p-CREB significantly and specifically. Additionally, the microRNA inhibitors improved the learning and memory functions of animals suffering from neurologic impairment due to propofol treatment and reduced cell apoptosis in the hippocampus. CONCLUSION: microRNA-34a could improve anesthesia-induced cognitive dysfunction by suppressing cell apoptosis and recovering the expression of genes associated with the MAPK/ERK signaling pathway.


Subject(s)
Anesthetics, Intravenous/toxicity , Apoptosis/drug effects , MAP Kinase Signaling System , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neuroprotective Agents/metabolism , Propofol/toxicity , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Motor Activity , Rats, Sprague-Dawley
19.
PLoS One ; 13(2): e0192796, 2018.
Article in English | MEDLINE | ID: mdl-29447230

ABSTRACT

The intravenous anesthetic propofol (2,6-diisopropylphenol) has been used for the induction and maintenance of anesthesia and sedation in critical patient care. However, the rare but severe complication propofol infusion syndrome (PRIS) can occur, especially in patients receiving high doses of propofol for prolonged periods. In vivo and in vitro evidence suggests that the propofol toxicity is related to the impaired mitochondrial function. However, underlying molecular mechanisms remain unknown. Therefore, we investigated effects of propofol on cell metabolism and death using a series of established cell lines of various origins, including neurons, myocytes, and trans-mitochondrial cybrids, with defined mitochondrial DNA deficits. We demonstrated that supraclinical concentrations of propofol in not less than 50 µM disturbed the mitochondrial function and induced a metabolic switch, from oxidative phosphorylation to glycolysis, by targeting mitochondrial complexes I, II and III. This disturbance in mitochondrial electron transport caused the generation of reactive oxygen species, resulting in apoptosis. We also found that a predisposition to mitochondrial dysfunction, caused by a genetic mutation or pharmacological suppression of the electron transport chain by biguanides such as metformin and phenformin, promoted propofol-induced caspase activation and cell death induced by clinical relevant concentrations of propofol in not more than 25 µM. With further experiments with appropriate in vivo model, it is possible that the processes to constitute the molecular basis of PRIS are identified.


Subject(s)
Anesthetics, Intravenous/toxicity , Cell Death/drug effects , Electron Transport/drug effects , Glycolysis/drug effects , Mitochondria/drug effects , Propofol/toxicity , Animals , Caspases/metabolism , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Electron Transport/physiology , Glycolysis/physiology , HeLa Cells , Humans , Hypoglycemic Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Metformin/pharmacology , Mice , Mitochondria/metabolism , Muscle Cells/drug effects , Muscle Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Reactive Oxygen Species/metabolism , Time Factors
20.
Anesth Analg ; 126(4): 1377-1380, 2018 04.
Article in English | MEDLINE | ID: mdl-29077609

ABSTRACT

Remote ischemic preconditioning (RIPC) seems to be a promising cardioprotective strategy with contradictive clinical data suggesting the anesthetic regimen influencing the favorable impact of RIPC. This study aimed to investigate whether cardio protection by RIPC is abolished by anesthetic regimens. Male Wistar rats were randomized to 6 groups. Anesthesia was either maintained by pentobarbital (Pento) alone or a combination of sevoflurane (Sevo) and remifentanil or propofol (Prop) and remifentanil in combination with and without RIPC. RIPC reduced infarct size in Pento- and Sevo-anesthetized rats (Pento-RIPC: 30% ± 9% versus Pento-control [Con]: 65% ± 6%, P < .001; Sevo-RIPC: 31% ± 6% versus Sevo-Con: 61% ± 8%, P < .001), but RIPC did not initiate cardio protection in Prop-anesthetized animals (Prop-RIPC: 59% ± 6% versus Prop-Con: 59% ± 8%, P = 1.000). Cardio protection by RIPC is abolished by Prop.


Subject(s)
Anesthetics, Intravenous/pharmacology , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Propofol/pharmacology , Analgesics, Opioid/pharmacology , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/toxicity , Animals , Disease Models, Animal , Hypnotics and Sedatives/pharmacology , Male , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Pentobarbital/pharmacology , Propofol/toxicity , Rats, Wistar , Remifentanil/pharmacology , Sevoflurane/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...