Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 536
Filtrar
1.
Hum Exp Toxicol ; 43: 9603271231218707, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38487884

RESUMO

BACKGROUND: Bupivacaine (BUP), a long-acting local anesthetic, has been widely used in analgesia and anesthesia. However, evidence strongly suggests that excessive application of BUP may lead to neurotoxicity in neurons. Sphingosine kinase 2 (SPHK2) has been reported to exert neuroprotective effects. In this study, we intended to investigate the potential role and mechanism of SPHK2 in BUP-induced neurotoxicity in dorsal root ganglion (DRG) neurons. METHODS: DRG neurons were cultured with BUP to simulate BUP-induced neurotoxicity in vitro. CCK-8, LDH, and flow cytometry assays were performed to detect the viability, LDH activity, and apoptosis of DRG neurons. RT-qPCR and western blotting was applied to measure gene and protein expression. Levels. MeRIP-qPCR was applied for quantification of m6A modification. RIP-qPCR was used to analyze the interaction between SPHK2 and YTHDF1. RESULTS: SPHK2 expression significantly declined in DRG neurons upon exposure to BUP. BUP challenge substantially reduced the cell viability and increased the apoptosis rate in DRG neurons, which was partly abolished by SPHK2 upregulation. YTHDF1, an N6-methyladenosine (m6A) reader, promoted SPHK2 expression in BUP-treated DRG neurons in an m6A-dependent manner. YTHDF1 knockdown partly eliminated the increase in SPHK2 protein level and the protection against BUP-triggered neurotoxicity in DRG neurons mediated by SPHK2 overexpression. Moreover, SPHK2 activated the PI3K/AKT signaling to protect against BUP-induced cytotoxic effects on DRG neurons. CONCLUSIONS: In sum, YTHDF1-mediated SPHK2 upregulation ameliorated BUP-induced neurotoxicity in DRG neurons via promoting activation of the PI3K/AKT signaling pathway.


Assuntos
Bupivacaína , Síndromes Neurotóxicas , Fosfotransferases (Aceptor do Grupo Álcool) , Humanos , Bupivacaína/toxicidade , Regulação para Cima , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Síndromes Neurotóxicas/prevenção & controle , Apoptose , Proteínas de Ligação a RNA
2.
Toxicology ; 503: 153740, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38316350

RESUMO

Bupivacaine, a common amide local anesthetic, can provide effective analgesia or pain relief but can also cause neurotoxicity, which remains a mounting concern in clinic and animal care. However, the precise underlying mechanisms have not been fully elucidated. A natural compound, notoginsenoside R1 (NG-R1) has been reported to exhibit a neuroprotective role in stress conditions. In this study, we explored the function and mechanism of NG-R1 in alleviating bupivacaine-induced neurotoxicity in mouse hippocampal neuronal (HT-22) and mouse neuroblastoma (Neuro-2a) cell lines. Our results exhibited that NG-R1 treatment can significantly rescue the decline of cell survival induced by bupivacaine. Tunel staining and western blotting showed that NG-R1 could attenuate BPV­induced cell apoptosis. Besides, we focused on Mcl1 as a potential target as it showed opposite expression tendency in response to NG-R1 and bupivacaine exposure. Mcl1 knockdown blocked the inhibitory effect of NG-R1 on cell apoptosis against bupivacaine treatment. Intriguingly, we found that NG-R1 can upregulate Mcl1 transcription by activating Stat3 and promote its nuclear translocation. In addition, NG-R1 can also promote Jak1 phosphorylation and docking analysis provide a predicted model for interaction between NG-R1 and phosphorylated Jak1. Taken together, our results demonstrated that NG-R1 can attenuate bupivacaine induced neurotoxicity by activating Jak1/Stat3/Mcl1 pathway.


Assuntos
Ginsenosídeos , Síndromes Neurotóxicas , Camundongos , Animais , Bupivacaína/toxicidade , Ginsenosídeos/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Síndromes Neurotóxicas/metabolismo , Linhagem Celular , Apoptose
3.
Genes Genomics ; 46(1): 149-160, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37523128

RESUMO

BACKGROUND: Bupivacaine, a common local anesthetic, can cause neurotoxicity and permanent neurological disorders. Crocin has been widely reported as a potential neuroprotective agent in neural injury models. OBJECTIVE: The aim of this study was to investigate the role and regulatory mechanism of crocin underlying bupivacaine-induced neurotoxicity. METHOD: Human neuroblastoma SH-SY5Y cells were treated with bupivacaine and/or crocin for 24 h, followed by detecting cell viability using 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay. The effect of crocin or bupivacaine on SH-SY5Y cell proliferation was measured by Ki67 immunofluorescence assay. The levels of apoptosis-related proteins and the markers in the PI3K/Akt signaling pathway were examined using western blot analysis. The activities of caspase 3, catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were tested using respective commercial assay kits. Flow cytometry analysis was executed for detecting SH-SY5Y cell apoptosis. RESULT: Crocin attenuated bupivacaine-induced neurotoxicity in SH-SY5Y cells. Meanwhile, crocin inhibited SH-SY5Y cell apoptosis induced by bupivacaine via repressing the activity of caspase-3, reducing Bax expression, and elevating Bcl-2 expression. Moreover, crocin mitigated oxidative stress in SH-SY5Y cells by increasing the content of CAT, SOD, GSH-Px and reducing the content of MDA. Additionally, crocin protected against bupivacaine-induced dephosphorylation of Akt and GSK-3ß. The protective effects of crocin against bupivacaine-induced neurotoxicity in SH-SY5Y cells were counteracted by the Akt inhibitor. CONCLUSION: These results suggested that crocin may exert a neuroprotective function by promoting cell proliferation and suppressing apoptosis and oxidative stress in SH-SY5Y cells. Thus, crocin might become a promising drug for the treatment of bupivacaine-induced neurotoxicity.


Assuntos
Carotenoides , Neuroblastoma , Proteínas Proto-Oncogênicas c-akt , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Bupivacaína/toxicidade , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicogênio Sintase Quinase 3 beta/farmacologia , Linhagem Celular Tumoral , Transdução de Sinais , Superóxido Dismutase/metabolismo
4.
Neuropharmacology ; 245: 109814, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38104768

RESUMO

Current evidence suggests that hyperactivated or impaired autophagy can lead to neuronal death. The effect of local anesthetics on painful diabetic neuropathy (PDN) and the role of autophagy in the above pathological process remain unclear, warranting further studies. So, PDN models were established by assessing the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) in leptin gene-mutation (db/db) mice. Wild type (WT) and PDN mice received intrathecal 0.75% bupivacaine or/with intraperitoneal drug treatment (rapamycin or bafilomycin A1). Subsequently, the PWT and PWL were measured to assess hyperalgesia at 6 h, 24 h, 30 h, and 48 h after intrathecal bupivacaine. Also, sensory nerve conduction velocity (SNCV) and motor nerve conduction velocity (MNCV) were measured before and 48 h after intrathecal bupivacaine treatment. The spinal cord tissue of L4-L6 segments and serum were harvested to evaluate the change of autophagy, oxidative stress, oxidative injury, and apoptosis. We found that bupivacaine induced the activation of autophagy but did not affect the pain threshold, SNCV and MNCV in WT mice at predefined time points. Conversely, bupivacaine lowered autophagosome generation and degradation, slowed SNCV and aggravated spinal dorsal horn neuron oxidative injury and hyperalgesia in PDN mice. The autophagy activator (rapamycin) could decrease spinal dorsal horn neuron oxidative injury, alleviate the alterations in SNCV and hyperalgesia in bupivacaine-treated PDN mice. Meanwhile, the autophagy inhibitor (bafilomycin A1) could exacerbate spinal dorsal horn neuron oxidative injury, the alterations in SNCV and hyperalgesia in bupivacaine-treated PDN mice. Our results showed that bupivacaine could induce defective autophagy, slowed SNCV and aggravate spinal dorsal horn neuron oxidative injury and hyperalgesia in PDN mice. Restoring autophagy may represent a potential therapeutic approach against nerve injury in PDN patients with local anesthesia and analgesia.


Assuntos
Diabetes Mellitus , Neuropatias Diabéticas , Macrolídeos , Ratos , Camundongos , Humanos , Animais , Hiperalgesia/metabolismo , Ratos Sprague-Dawley , Neuropatias Diabéticas/induzido quimicamente , Neuropatias Diabéticas/tratamento farmacológico , Neuropatias Diabéticas/metabolismo , Bupivacaína/toxicidade , Sirolimo , Autofagia
5.
Sud Med Ekspert ; 66(4): 62-66, 2023.
Artigo em Russo | MEDLINE | ID: mdl-37496485

RESUMO

THE AIM OF THE STUDY: Was to assess the lidocaine and bupivacaine systemic toxicity in forensic medical practice. The number of patients' clinical observations equal three with local anesthetic systemic toxicity (LAST) from the practice of forensic medical experts were studied, and a search of scientific publications for the last 5 years in PubMed database was conducted. The amount of publications, describing cases with LAST, equal four were selected. Differential diagnostic features between LAST and anaphylaxis were considered. The literature data about relationship between lidocaine's concentration in the blood serum and clinical features are shown. The forensic medical assessment of LAST is proposed.


Assuntos
Bupivacaína , Lidocaína , Humanos , Bupivacaína/toxicidade , Anestésicos Locais/toxicidade
6.
Pharmacol Res Perspect ; 11(4): e01116, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37470146

RESUMO

This study was aimed at examining the anesthetic effects and spinal cord injuries in the rats by intrathecal injection of levobupivacaine at different concentrations. Rats with successful intrathecal cannulation were selected and randomly divided into six groups (n = 72), and administered 0.1 mL of 0.125%, 0.25%, 0.5%, or 0.75% levobupivacaine, saline or 5% lidocaine via intrathecal catheters. The potency of levobupivacaine was evaluated by walking behavior. To identify the motor and sensory function, walking behavior and paw withdrawal thresholds (PWTs) were measured once a day. After 7 days, the L4-5 spinal cord segments were removed for histological examination. The onset time of 0.125% levobupivacaine intrathecal injection was 70.0 ± 8.9 s, and the maintenance time was 9.5 ± 1.8 min. The onset time of 0.75% levobupivacaine intrathecal injection was significantly shortened to 31.0 ± 5.5 s, and the maintenance time was significantly extended to 31.3 ± 5.4 min. The severe injury was observed in the 5% lidocaine group, while milder injury was observed in the 0.75% levobupivacaine group. The damage in the 0.5% levobupivacaine group was mild, and there were no histological abnormalities in the 0.125%, 0.25% levobupivacaine and saline groups. The neurotoxicity of intrathecally administered levobupivacaine was concentration dependent. In addition, higher concentrations of levobupivacaine were associated with shorter onset and longer maintenance times. The clinical concentration of levobupivacaine should not exceed 0.5% to avoid potential damage.


Assuntos
Anestésicos Locais , Bupivacaína , Animais , Ratos , Anestésicos Locais/toxicidade , Bupivacaína/toxicidade , Injeções Espinhais , Levobupivacaína , Lidocaína/toxicidade
7.
Cell Mol Biol (Noisy-le-grand) ; 69(2): 84-89, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-37224040

RESUMO

This study was to explore the application value of chromosome ten (PTEN) - phosphatidylinositol 3-kinase (PI3K) - protein kinase B (AKT) signaling pathway in the treatment of Bupivacaine toxicity to neuronal cells under the regulation of fat emulsion. Neurons in the hippocampus of newborn rats were treated with Bupivacaine and fat emulsion and divided into five groups. The activity and action potential of neurons in each group were measured and Nissl's staining was performed. The results showed that the neuron activity of Bupivacaine group (42.36 ± 5.48%), Bupivacaine + fat emulsion group (70.23 ± 3.66%), and Bupivacaine + fat emulsion + PTEN/PI3K/AKT inhibitor group (79.28 ± 5.14%) was lower than that of the blank group (99.95 ± 3.42%). The duration of action potential in Bupivacaine group was increased (5.19 ± 0.48ms) and the frequency of action potential was decreased (13.87 ± 1.95) compared with the blank group (2.44 ± 0.37ms, 19.59 ± 2.14). The duration of the fat emulsion group (2.39 ± 0.39ms, 19.76 ± 2.05), Bupivacaine + fat emulsion group (2.88 ± 0.52ms, 18.53 ± 1.66), and Bupivacaine + fat emulsion + PTEN/PI3K/AKT inhibitor group (3.43 ± 0.69ms, 17.57 ± 1.58) was decreased, but the number of times increased (P < 0.05). In short, the fat emulsion can reverse the toxic effects of Bupivacaine on rat hippocampal neurons by regulating the PTEN/PI3K/AKT signaling pathway. This study provided a reference for the clinical treatment of the neurotoxicity of Bupivacaine.


Assuntos
Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Animais , Ratos , Fosfatidilinositol 3-Quinase , Emulsões/farmacologia , Transdução de Sinais , Bupivacaína/toxicidade , Inibidores de Fosfoinositídeo-3 Quinase , PTEN Fosfo-Hidrolase
8.
Curr Neuropharmacol ; 21(10): 2134-2150, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37021417

RESUMO

OBJECTIVE: Severe neurologic complications after spinal anesthesia are rare but highly distressing, especially in pregnant women. Bupivacaine is widely used in spinal anesthesia, but its neurotoxic effects have gained attention. METHODS: Furthermore, the etiology of bupivacaine-mediated neurotoxicity in obstetric patients remains unclear. Female C57BL/6 mice were intrathecally injected with 0.75% bupivacaine on the 18th day of pregnancy. We used immunohistochemistry to examine DNA damage after bupivacaine treatment in pregnant mice and measured γ-H2AX (Ser139) and 8-OHdG in the spinal cord. A PARP-1 inhibitor (PJ34) and autophagy inhibitor (3-MA) were administered with bupivacaine in pregnant mice. Parp-1flox/flox mice were crossed with Nes-Cre transgenic mice to obtain neuronal conditional knockdown mice. Then, LC3B and P62 staining were performed to evaluate autophagic flux in the spinal cords of pregnant wild-type (WT) and Parp-1-/- mice. We performed transmission electron microscopy (TEM) to evaluate autophagosomes. RESULTS: The present study showed that oxidative stress-mediated DNA damage and neuronal injury were increased after bupivacaine treatment in the spinal cords of pregnant mice. Moreover, PARP-1 was significantly activated, and autophagic flux was disrupted. Further studies revealed that PARP-1 knockdown and autophagy inhibitors could alleviate bupivacaine-mediated neurotoxicity in pregnant mice. CONCLUSION: Bupivacaine may cause neuronal DNA damage and PARP-1 activation in pregnant mice. PARP-1 further obstructed autophagic flux and ultimately led to neurotoxicity.


Assuntos
Anestésicos Locais , Bupivacaína , Animais , Feminino , Humanos , Camundongos , Gravidez , Anestésicos Locais/toxicidade , Apoptose , Autofagia , Bupivacaína/toxicidade , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia
9.
Kaohsiung J Med Sci ; 39(7): 710-717, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37078471

RESUMO

Kaempferol (KA), a widely recognized anti-oxidation and anti-inflammation agent, has been reported to have neuroprotective effects. This work aimed to investigate whether KA protects mouse dorsal root ganglia (DRG) neurons against bupivacaine (BU)-stimulated neurotoxicity and explore the underlying mechanisms. In this study, BU treatment suppressed DRG neuron viability and promoted LDH leakage, which was partially abated by KA. Besides, BU-triggered DRG neuron apoptosis, and changes in Bax and Bcl-2 levels were attenuated by KA treatment. In addition, pretreatment with KA substantially reduced interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α levels in BU-treated DRG neurons. In addition, KA administration abrogated BU-induced decline in CAT, SOD, and GSH-Px levels, as well as the increase in the malondialdehyde level. Interestingly, we found that KA significantly attenuated BU-induced TNF receptor-associated factor 6 (TRAF6) upregulation as well as NF-κB activation. Furthermore, oe-TRAF6-mediated TRAF6 overexpression promoted NF-κB activation and partly abolished KA-induced protection against BU-triggered neurotoxic effects on DRG neurons. Our results revealed that KA mitigated BU-induced neurotoxic effects on DRG neurons by deactivating the TRAF6/NF-κB signaling.


Assuntos
NF-kappa B , Fator 6 Associado a Receptor de TNF , Camundongos , Animais , NF-kappa B/genética , NF-kappa B/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Bupivacaína/toxicidade , Gânglios Espinais/metabolismo , Quempferóis/farmacologia , Neurônios , Interleucina-6/genética , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Biomed Res Int ; 2023: 1176232, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36865484

RESUMO

Bupivacaine (BUP) may cause neurotoxic effects after spinal anesthesia. Resveratrol (RSV), a natural agonist of Silent information regulator 1 (SIRT1), protects various tissues and organs from damage by regulating endoplasmic reticulum (ER) stress. The aim of this study is to explore whether RSV could alleviate the neurotoxicity induced by bupivacaine via regulating ER stress. We established a model of bupivacaine-induced spinal neurotoxicity in rats using intrathecal injection of 5% bupivacaine. The protective effect of RSV was evaluated by injecting intrathecally with 30 µg/µL RSV in total of 10 µL per day for 4 consecutive days. On day 3 after bupivacaine administration, tail-flick latency (TFL) tests and the Basso, Beattie, and Bresnahan (BBB) locomotor scores were assessed to neurological function, and the lumbar enlargement of the spinal cord was obtained. H&E and Nissl staining were used to evaluate the histomorphological changes and the number of survival neurons. TUNEL staining was conducted to determine apoptotic cells. The expression of proteins was detected by IHC, immunofluorescence, and western blot. The mRNA level of SIRT1 was determined by RT-PCR. Bupivacaine caused spinal cord neurotoxicity by inducing cell apoptosis and triggering ER stress. RSV treatment promoted the recovery of neurological dysfunction after bupivacaine administration by suppressing neuronal apoptosis and ER stress. Furthermore, RSV upregulated SIRT1 expression and inhibited PERK signaling pathway activation. In summary, resveratrol suppresses bupivacaine-induced spinal neurotoxicity in rats by inhibiting endoplasmic reticulum stress via SIRT1 modulation.


Assuntos
Síndromes Neurotóxicas , Sirtuína 1 , Animais , Ratos , Resveratrol/farmacologia , Medula Espinal , Bupivacaína/toxicidade , Estresse do Retículo Endoplasmático
11.
Korean J Anesthesiol ; 76(3): 171-182, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36704816

RESUMO

Currently, lipid emulsion (LE) is widely used to treat local anesthetic systemic toxicity (LAST). LE also ameliorates intractable cardiovascular collapse caused by lipid-soluble non-local anesthetic drug toxicity. This review aims to provide the underlying mechanism of LE resuscitation in drug toxicity (including LAST) and a detailed description of LE treatment and to discuss further research directions. We searched for relevant articles using the following keywords: "local anesthetic systemic toxicity or LAST or toxicity or intoxication or poisoning" and "Intralipid or lipid emulsion". The underlying mechanisms of LE treatment can be classified into indirect and direct effects. One indirect effect known as the lipid shuttle is a commonly accepted mechanism of LE treatment. The lipid shuttle involves the absorption of highly lipid-soluble drugs (e.g., bupivacaine) from the heart and brain through the lipid phase, which are then delivered to the muscle, adipose tissue, and liver for storage and detoxification. The direct effects include inotropic effects, fatty acid supply, attenuation of mitochondrial dysfunction, glycogen synthase kinase-3ß phosphorylation, and inhibition of nitric oxide. These mechanisms appear to act synergistically to treat drug toxicity. The recommended protocol for LE treatment of LAST is as follows: a bolus administration of 20% LE at 1.5 ml/kg over 2-3 min followed by 20% LE at 0.25 ml/kg/min. LAST most commonly occurs after intravenous administration of local anesthetics. However, non-local anesthetic drugs that cause drug toxicity are orally administered. Further studies are needed to determine the optimal dosing schedule of LE treatment for non-local anesthetic drug toxicity.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Emulsões Gordurosas Intravenosas , Humanos , Preparações Farmacêuticas , Emulsões Gordurosas Intravenosas/farmacologia , Emulsões Gordurosas Intravenosas/uso terapêutico , Anestésicos Locais/toxicidade , Bupivacaína/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/tratamento farmacológico
12.
Pediatr Emerg Care ; 39(1): 53-59, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-35981328

RESUMO

OBJECTIVE: Lipid emulsion (LE) has been used to treat children with cardiovascular collapse induced by toxic doses of nonlocal anesthetics with high lipid solubility. We aimed to analyze case reports on LE administration for resuscitation of toxicity induced by these drugs in pediatric patients. METHODS: Case reports involving pediatric patients undergoing LE treatment for toxicity caused by nonlocal anesthetic drugs until December 31, 2021, were searched through PubMed and Scopus using the following terms: "toxicity, or intoxication, or poisoning, or overdose" and "LE or intralipid." RESULTS: Twenty-eight cases on LE treatment for toxicity induced by nonlocal anesthetic drugs in pediatric patients (younger than 19 years) were retrieved. The total number of patients was 31. Lipid emulsion treatment was carried out during toxicity caused by amitriptyline, flecainide, bupropion, propranolol, and lamotrigine, which was unresponsive to supportive treatment. These drugs are highly lipid-soluble and inhibit cardiac sodium channels, which is similar to pharmacological properties of the local anesthetic bupivacaine. The most frequent method of delivery involved bolus administration followed by continuous infusion; 1.5 mL/kg LE administration followed by 0.25 mL/kg/min LE was most frequently used. Lipid emulsion improved various symptoms of drug toxicity in 29 patients (29/31, 93.54%), and symptoms were improved in 14 patients (14/31, 45.16%) within an h after LE administration. The trend in frequency of improved symptoms after LE treatment was as follows: the cardiovascular symptom alone > symptoms of the central nervous system alone > symptoms of the cardiovascular and central nervous systems. The adverse effects of LE treatment in the reported cases were hypertriglyceridemia, mild pancreatitis, and elevated levels of aspartate and alanine aminotransaminases. CONCLUSIONS: Lipid emulsion treatment may be effective in ameliorating intractable cardiovascular depression when systemic toxicity caused by drugs, including cardiac sodium channel blockers, is unresponsive to supportive treatments.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Emulsões Gordurosas Intravenosas , Humanos , Criança , Emulsões Gordurosas Intravenosas/uso terapêutico , Anestésicos Locais , Bupivacaína/toxicidade , Ressuscitação
13.
Anesthesiology ; 137(6): 687-703, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36170651

RESUMO

BACKGROUND: Long-lasting local anesthetic use for perioperative pain control is limited by possible cardiotoxicity (e.g., arrhythmias and contractile depression), potentially leading to cardiac arrest. Off-target cardiac sodium channel blockade is considered the canonical mechanism behind cardiotoxicity; however, it does not fully explain the observed toxicity variability between anesthetics. The authors hypothesize that more cardiotoxic anesthetics (e.g., bupivacaine) differentially perturb other important cardiomyocyte functions (e.g., calcium dynamics), which may be exploited to mitigate drug toxicity. METHODS: The authors investigated the effects of clinically relevant concentrations of racemic bupivacaine, levobupivacaine, or ropivacaine on human stem cell-derived cardiomyocyte tissue function. Contractility, rhythm, electromechanical coupling, field potential profile, and intracellular calcium dynamics were quantified using multielectrode arrays and optical imaging. Calcium flux differences between bupivacaine and ropivacaine were probed with pharmacologic calcium supplementation or blockade. In vitro findings were correlated in vivo using an anesthetic cardiotoxicity rat model (females; n = 5 per group). RESULTS: Bupivacaine more severely dysregulated calcium dynamics than ropivacaine in vitro (e.g., contraction calcium amplitude to 52 ± 11% and calcium-mediated repolarization duration to 122 ± 7% of ropivacaine effects, model estimate ± standard error). Calcium supplementation improved tissue contractility and restored normal beating rhythm (to 101 ± 6%, and 101 ± 26% of control, respectively) for bupivacaine-treated tissues, but not ropivacaine (e.g., contractility at 80 ± 6% of control). Similarly, calcium pretreatment mitigated anesthetic-induced arrhythmias and cardiac depression in rats, improving animal survival for bupivacaine by 8.3 ± 2.4 min, but exacerbating ropivacaine adverse effects (reduced survival by 13.8 ± 3.4 min and time to first arrhythmia by 12.0 ± 2.9 min). Calcium channel blocker nifedipine coadministration with bupivacaine, but not ropivacaine, exacerbated cardiotoxicity, supporting the role of calcium flux in differentiating toxicity. CONCLUSIONS: Our data illustrate differences in calcium dynamics between anesthetics and how calcium may mitigate bupivacaine cardiotoxicity. Moreover, our findings suggest that bupivacaine cardiotoxicity risk may be higher than for ropivacaine in a calcium deficiency context.


Assuntos
Anestésicos Locais , Cálcio , Feminino , Ratos , Humanos , Animais , Anestésicos Locais/toxicidade , Cardiotoxicidade , Miócitos Cardíacos , Amidas/farmacologia , Bupivacaína/toxicidade , Ropivacaina/toxicidade , Arritmias Cardíacas/induzido quimicamente
14.
Clin Toxicol (Phila) ; 60(8): 902-911, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35658706

RESUMO

INTRODUCTION: Ropivacaine is considered to have a wider margin of cardiovascular safety. However, several reports of ventricular arrhythmias (VA) due to ropivacaine toxicity have been documented. Intravenous lipid emulsions (ILEs) have recently been used successfully in the treatment of local anesthetic intoxication. The main objective of the present study was to evaluate the efficacy of the ILEs in the prevention of pacing-induced-VA and electrophysiological alterations in an animal model of ropivacaine toxicity. METHODS: Nineteen pigs were anesthetized and instrumentalized. A baseline programmed electrical ventricular stimulation protocol (PEVSP) to induce VA was performed. Ropivacaine (5 mg·kg-1 + 100 µg·kg-1·min-1) followed by normal saline infusion (control group n = 8) or intralipid 20% (1.5 mL·kg-1 + 0.25 mL·kg-1·min-1) for the ILE group (n = 8), were administered three minutes after the ropivacaine bolus. PEVSP was repeated 25 min after the onset of ropivacaine infusion. Pacing-induced VA and electrophysiological abnormalities were assessed in both groups. A sham-control group (n = 3) without ropivacaine infusion was included. RESULTS: Most of the electrophysiological parameters evaluated were affected by ropivacaine: PR interval by 28% (p = 0.001), AV interval by 40% (p = 0.001), sinus QRS by 101% (p = 0.001), paced QRS at a rate of 150 bpm by 258% (p = 0.001), and at 120 bpm by 241% (p = 0.001). Seven animals (87.5%) in the control group and eight animals (100%) in the ILE group developed sustained-VA (p = 0.30). Successful resuscitation occurred in 100% of animals in the ILE group vs. 57% of animals in the control group, p = 0.038. Pacing-induced-VA terminated at the first defibrillation attempt in 75% of the animals in the ILE group vs. 0% in the control group, p = 0.01. CONCLUSION: Ropivacaine strongly altered the parameters of ventricular conduction, thus facilitating the induction of VA. ILEs did not prevent pacing-induced VA. However, facilitated resuscitation and termination of VA were delivered at the first defibrillation attempt compared to the control group.


Assuntos
Anestésicos Locais , Emulsões Gordurosas Intravenosas , Anestésicos Locais/toxicidade , Animais , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/tratamento farmacológico , Bupivacaína/toxicidade , Modelos Animais de Doenças , Emulsões Gordurosas Intravenosas/farmacologia , Emulsões Gordurosas Intravenosas/uso terapêutico , Ropivacaina/toxicidade , Solução Salina , Suínos
15.
Cutan Ocul Toxicol ; 41(3): 210-214, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35607763

RESUMO

PURPOSE: To compare the histopathological effects of injecting two concentrations of Bupivacaine (5 mg/ml and 7.5 mg/ml) in the superior rectus muscle of rabbits, and to compare these to conventional extraocular muscle surgery in previous studies. METHODS: Eighteen albino rabbits' eyes were used. The superior rectus muscles were injected with Bupivacaine 5 mg/ml (Group B5, 10 eyes) or 7.5 mg/ml (Group B7, 8 eyes). The rabbits were sacrificed and eyes enucleated 6 weeks later for histopathological evaluation. Results were compared to the average of those obtained, by three previous studies, after conventional superior rectus resection in rabbits. RESULTS: Foreign body reaction was absent in all specimens. Conjunctival and scleral inflammation, perimuscular adhesions, intramuscular fibrosis, conjunctival and scleral oedema and muscle atrophy were higher in group B7, while conjunctival hyperaemia and muscle hypertrophy were higher in group B5 (p > 0.05). On comparison to conventional surgery, conjunctival inflammation and hyperaemia, foreign body reaction, and adhesions were less after bupivacaine injection (p > 0.05 for all except for intensity of conjunctival inflammation in B5 versus conventional surgery). Scleral inflammation was more frequent after bupivacaine injection (p < 0.05). Muscle fibrosis was more frequent in group B7 and conventional surgery than in group B5 (p > 0.05). CONCLUSIONS: Both Bupivacaine concentrations effectively produced the desired muscle hypertrophy and fibrosis, so the lower concentration may be used for muscle strengthening to correct strabismus. Bupivacaine injection, although produced no foreign body reaction, did not significantly lower the development of undesired postoperative adhesions and caused more scleral inflammation.


Assuntos
Hiperemia , Estrabismo , Bupivacaína/toxicidade , Fibrose , Humanos , Hiperemia/complicações , Hiperemia/patologia , Hipertrofia/complicações , Hipertrofia/patologia , Inflamação/induzido quimicamente , Inflamação/patologia , Músculos Oculomotores/patologia , Músculos Oculomotores/cirurgia , Estrabismo/induzido quimicamente , Estrabismo/complicações , Estrabismo/cirurgia , Aderências Teciduais
16.
Hum Exp Toxicol ; 41: 9603271221089001, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35410500

RESUMO

Long non-coding RNA (lncRNA) small nucleolar RNA host gene 12 (SNHG12) has been reported to participate in the regulation of various nervous system disorders. Bupivacaine (BV), a commonly used local anesthetic, could generate neurotoxicity in neurons. This work intended to investigate the role and specific mechanism of SNHG12 in BV-induced neurotoxicity. In this study, we established an in vitro cell model of BV-induced neurotoxicity by exposing human neuroblastoma cells (SH-SY5Y) to BV. It was found that SNHG12 and NLRX1 levels were gradually downregulated, while miR-497-5p enrichment was upregulated accordingly with the increase of BV concentration. As indicated by functional assays, SNHG12 overexpression promoted cell viability but inhibited cell apoptosis and oxidative stress in BV-treated SH-SY5Y cells. In addition, it was identified that SNHG12 directly targeted miR-497-5p and attenuated BV-induced neurotoxicity via interaction with miR-497-5p. Besides, it was confirmed that SNHG12 could upregulate NLRX1 expression by absorbing miR-497-5p. Moreover, miR-497-5p decreased cell viability and induced cell apoptosis and oxidative stress, which was partly reversed by NLRX1 upregulation. In conclusion, our findings indicated that SNHG12 might relieve BV-associated neurotoxicity by upregulating NLRX1 via miR-497-5p in vitro, providing novel clues and biomarkers for the treatment and prevention of BV-associated neurotoxicity.


Assuntos
MicroRNAs , Síndromes Neurotóxicas , RNA Longo não Codificante , Apoptose/genética , Bupivacaína/toxicidade , Linhagem Celular Tumoral , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Mitocondriais , Síndromes Neurotóxicas/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
17.
Bioengineered ; 13(3): 7439-7456, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35271399

RESUMO

The research aimed to explore the biological role of p53 protein and long non-coding RNA (lncRNA) taurine upregulated gene 1 (TUG1) in bupivacaine (bup)-induced neurotoxicity. Our work treated dorsal root ganglion (DRG) cells with bup, detected cell viability through CCK-8, apoptosis through TUNEL assays, DeoxyriboNucleic Acid (DNA) damage through γ-H2AX protein and comet assay, including p53 mRNA, protein and TUG1 expression through q-PCR and western blot, furthermore, cell viability and DNA damage were determined after the silencing of p53 and TUG1, biological information and TUG1 FISH combined with p53 protein immunofluorescence (IF) was performed to determine the cellular localization of these molecule. In vivo experiments, we explored the impact of intrathecal injection of bup on p53 mRNA and protein, TUG1, γ-H2AX protein expression. The results showed that bup was available to signally decreased cell viability, promoted apoptosis rate and DNA damage, additionally, bup increased p53 mRNA and protein and TUG1 expression. P53 siRNA and TUG1 siRNA significantly increased DNA damage. Furthermore, bioinformatics analysis and colocalization experiments revealed that the p53 protein is a transcription factor of TUG1, in vivo experiment, intrathecal injection of bup increased the p53 mRNA, p53 protein, TUG1 and γ-H2AX protein in the murine DRG. In this study, it was found p53 and TUG1 promote the repair of the DNA damage induced by bup in murine dorsal root ganglion cells, suggesting a new strategy for the amelioration of bup-induced neurotoxicity.


Assuntos
Bupivacaína , MicroRNAs , Animais , Apoptose/genética , Bupivacaína/toxicidade , Proliferação de Células/genética , DNA , Camundongos , RNA Mensageiro , RNA Interferente Pequeno/genética , Células Receptoras Sensoriais , Taurina , Proteína Supressora de Tumor p53/genética
18.
Int J Oral Maxillofac Surg ; 51(10): 1337-1344, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35120788

RESUMO

The aim of this study was to investigate the chondrotoxic effects of a single-dose intra-articular injection of articaine, lidocaine, and bupivacaine on the rabbit temporomandibular joint (TMJ). Twenty-four rabbits were divided into four groups: control (group 1), articaine (group 2), lidocaine (group 3), and bupivacaine (group 4). Synovial fluid samples and venous blood were taken to evaluate matrix metalloproteinase 3 (MMP-3) levels. One millilitre of local anaesthetic solution was injected in the study groups and saline solution in the control group. The rabbits were euthanized after 4 weeks and the mandibular condyles and articular discs were evaluated. On histological examination, the study group samples had irregular joint surfaces, decreased collagen, and a thinner cartilage layer. Apoptotic cells were evaluated with the TUNEL method. TUNEL-positive apoptotic cell counts were higher in all study groups compared to the control group, and the difference was significant (P < 0.001). The mean preoperative serum MMP-3 level for all groups was 5.71 ± 3.33 ng/mL, while the mean postoperative level was 22.61 ± 6.36 ng/mL; this difference was significant (P < 0.001). A single-dose intra-articular injection of local anaesthetic had apoptotic effects on chondrocytes, leading to degenerative changes in the TMJ articular structures. Articaine was found to have less harmful effects than lidocaine and bupivacaine. Intra-articular injection of local anaesthetics should be limited in the TMJ because of the potential toxic effects.


Assuntos
Anestésicos Locais , Cartilagem Articular , Anestésicos Locais/toxicidade , Animais , Bupivacaína/toxicidade , Carticaína/toxicidade , Injeções Intra-Articulares , Lidocaína/toxicidade , Metaloproteinase 3 da Matriz/farmacologia , Coelhos , Solução Salina/farmacologia , Articulação Temporomandibular
19.
Hum Exp Toxicol ; 41: 9603271211065978, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35135371

RESUMO

The aim of this study was to examine the effects of lipid emulsions on carnitine palmitoyltransferase I (CPT-I), carnitine acylcarnitine translocase (CACT), carnitine palmitoyltransferase II (CPT-II), and the mitochondrial dysfunctions induced by toxic doses of local anesthetics in H9c2 rat cardiomyoblasts. The effects of local anesthetics and lipid emulsions on the activities of CPT-I, CACT, and CPT-II, and concentrations of local anesthetics were examined. The effects of lipid emulsions, N-acetyl-L-cysteine (NAC), and mitotempo on the bupivacaine-induced changes in cell viability, reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP), and intracellular calcium levels were examined. CACT, without significantly altering CPT-I and CPT-II, was inhibited by toxic concentration of local anesthetics. The levobupivacaine- and bupivacaine-induced inhibition of CACT was attenuated by all concentrations of lipid emulsion, whereas the ropivacaine-induced inhibition of CACT was attenuated by medium and high concentrations of lipid emulsion. The concentration of levobupivacaine was slightly attenuated by lipid emulsion. The bupivacaine-induced increase of ROS and calcium and the bupivacaine-induced decrease of MMP were attenuated by ROS scavengers NAC and mitotempo, and the lipid emulsion. Collectively, these results suggested that the lipid emulsion attenuated the levobupivacaine-induced inhibition of CACT, probably through the lipid emulsion-mediated sequestration of levobupivacaine.


Assuntos
Bupivacaína/toxicidade , Carnitina Aciltransferases/efeitos dos fármacos , Carnitina Aciltransferases/metabolismo , Levobupivacaína/toxicidade , Mioblastos Cardíacos/efeitos dos fármacos , Mioblastos Cardíacos/metabolismo , Ropivacaina/toxicidade , Anestésicos Locais/administração & dosagem , Anestésicos Locais/toxicidade , Animais , Bupivacaína/administração & dosagem , Emulsões/administração & dosagem , Emulsões/toxicidade , Inibidores Enzimáticos/metabolismo , Levobupivacaína/administração & dosagem , Masculino , Ratos , Ropivacaina/administração & dosagem
20.
Neurosci Lett ; 772: 136472, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35065245

RESUMO

Bupivacaine is a common local anesthetic that causes neurotoxicity when used at clinical concentrations. Melatonin (MT), is a potent neuroprotective molecule. The study aimed to characterize the neuroprotective effects of MT on spinal neurotoxicity induced by bupivacaine in rats. It showed that bupivacaine, by intrathecal injection, induced spinal injury, and that the protein levels of Nod-like receptor protein 3 (NLRP3), cleaved caspase-1, and the N-terminal region of gasdermin D (GSDMD-N) were significantly increased. NLRP3 was expressed mainly in neurons and microglia. MT treatment ameliorated bupivacaine-induced spinal cord injury in rats by suppressing activation of neuronal NLRP3 inflammasomes.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Melatonina/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Bupivacaína/toxicidade , Masculino , Melatonina/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...