Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
2.
Exp Ther Med ; 26(1): 341, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37383377

ABSTRACT

[This corrects the article DOI: 10.3892/etm.2021.10014.].

3.
Biomed Pharmacother ; 152: 113229, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35679721

ABSTRACT

Ketamine is a widely-used anesthetic in the field of pediatrics and obstetrics. Multiple studies have revealed that ketamine causes neurotoxicity in developing animals. However, further studies are needed to determine whether clinical doses of ketamine (20 mg/kg) are able to cause kidney damage in developing animals. Herein, we investigated the effects of continuous ketamine exposure on kidney injury and pyroptosis in seven-day-old rats. Serum renal function indicators, renal histopathological analysis, pyroptosis, as well as oxidative stress indicators, were tested. Additionally, the NLRP3 inhibitor MCC950 and the Caspase-1 inhibitor VX765 were used to evaluate the role of the NLRP3/Caspase-1 axis in ketamine-induced kidney injury among developing rats. Our findings indicate that ketamine exposure causes renal histopathological injury, increased the levels of blood urea nitrogen (BUN) and creatinine (Cre), and led to upregulation in the levels of pyroptosis. Furthermore, we found that ketamine induced an increase in levels of reactive oxygen species (ROS) and malonaldehyde (MDA), as well as a decrease in the content of glutathione (GSH) and catalase (CAT) in the kidneys of neonatal rats. Moreover, targeting NLRP3 and caspase-1 with MCC950 or VX765 improved pyroptosis and reduced renal damage after continuous ketamine exposure. In conclusion, this study suggested that continued exposure to ketamine caused kidney damage among neonatal rats and that the NLRP3/Caspase-1 axis-related pyroptosis may be involved in this process.


Subject(s)
Ketamine , Pyroptosis , Animals , Animals, Newborn , Caspase 1 , Child , Humans , Inflammasomes , Ketamine/toxicity , Kidney/physiology , NLR Family, Pyrin Domain-Containing 3 Protein , Rats
4.
J Neuroinflammation ; 18(1): 239, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34666787

ABSTRACT

BACKGROUND: Multiple studies have revealed that repeated or long-term exposure to ketamine causes neurodegeneration and cognitive dysfunction. Pyroptosis is an inflammatory form of programmed cell death that has been linked to various neurological diseases. However, the role of NLRP3/caspase-1 axis-related pyroptosis in ketamine-induced neurotoxicity and cognitive dysfunction remains uncertain. METHODS: To evaluate whether ketamine caused NLRP3/caspase1-dependent pyroptosis, flow cytometry analysis, western blotting, ELISA test, histopathological analysis, Morris water maze (MWM) test, cell viability assay, and lactate dehydrogenase release (LDH) assay were carried out on PC12 cells, HAPI cells, and 7-day-old rats. In addition, the NLRP3 inhibitor MCC950 or the caspase-1 inhibitor VX-765 was used to investigate the role of the NLRP3/caspase-1 axis in ketamine-induced neurotoxicity and cognitive dysfunction. RESULTS: Our findings demonstrated that ketamine exposure caused cell damage and increased the levels of pyroptosis in PC12 cells, HAPI cells, and the hippocampus of neonatal rats. After continuous exposure to ketamine, targeting NLRP3 and caspase-1 with MCC950 or VX765 improved pyroptosis, reduced neuropathological damages, and alleviated cognitive dysfunction. CONCLUSION: NLRP3/Caspase-1 axis-dependent pyroptosis is involved in ketamine-induced neuroinflammation and cognitive dysfunction, and it provides a promising strategy to treat ketamine-related neurotoxicity.


Subject(s)
Caspase 1/metabolism , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Ketamine/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/physiology , Animals , Animals, Newborn , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/prevention & control , Dipeptides/pharmacology , Dipeptides/therapeutic use , Excitatory Amino Acid Antagonists/toxicity , Female , Furans/pharmacology , Furans/therapeutic use , Hippocampus/drug effects , Indenes/pharmacology , Indenes/therapeutic use , Male , Maze Learning/drug effects , Maze Learning/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , PC12 Cells , Pyroptosis/drug effects , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , para-Aminobenzoates/pharmacology , para-Aminobenzoates/therapeutic use
5.
Front Vet Sci ; 8: 674862, 2021.
Article in English | MEDLINE | ID: mdl-34336971

ABSTRACT

The aim of the present study was to evaluate the anesthetic and cardiopulmonary effects of dexmedetomidine in combination with tiletamine (without zolazepam) as a general anesthetic. The study was divided into two phases. In Phase 1, 18 adult healthy mixed-breed dogs were randomly allocated into three groups: Group TD8 (4.5 mg kg-1 tiletamine and 8 µg kg-1 dexmedetomidine), Group TD10 (4.5 mg kg-1 tiletamine and 10 µg kg-1 dexmedetomidine), or Group TD12 (4.5 mg kg-1 tiletamine and 12 µg kg-1 dexmedetomidine). After drug administration, the heart rate (HR), respiratory rate (f R), mean arterial pressure (MAP), systolic arterial pressure (SAP), diastolic arterial pressure (DAP), peripheral hemoglobin oxygen saturation (SpO2), behavioral score, quality of induction and recovery, extent of ataxia, the time taken for induction, and the duration of anesthesia were recorded. The recovery time and quality were recorded after administration of atipamezole (50 µg kg-1) after 60 min. In phase 2, the feasibility of combining dexmedetomidine (10 µg kg-1) and tiletamine (4.5 mg kg-1) as general anesthetics for orchiectomy was evaluated in dogs (n = 6). HR, f R, MAP, SAP, DAP, temperature, SpO2, behavioral scores, and adverse reactions were recorded during each surgical procedure. In phase 1, the dogs were anesthetized for 5 min after administration of drugs and achieved a maximum behavioral score in TD10 and TD12 after 10 min. Although HR, MAP, SAP, DAP, and NIBP decreased in all three groups, they still maintained within the normal range. In phase 2, orchiectomy was completed smoothly in all dogs with little fluctuation in the physiological variables. We found that a combination of tiletamine (4.5 mg kg-1) and dexmedetomidine (10 µg kg-1) intramuscularly induced moderate anesthesia in dogs and could be utilized for short-term anesthesia and minor surgery.

6.
Exp Ther Med ; 21(6): 582, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33850554

ABSTRACT

Osteoarthritis (OA) is characterized by progressive degeneration of cartilage, formation of cartilage at the cartilage edge, and remodeling of the subchondral bone. Pro-inflammatory cytokines [e.g., interleukin (IL)-1ß] that induce inflammation and promote chondrocyte damage induce OA. Currently, the diagnosis of OA is commonly based on imaging examinations (e.g., X-ray) and evaluations of clinical symptoms; however, biomarkers that can effectively diagnose OA are currently not available. By studying the mechanism underlying OA cartilage injury and changes in the concentrations of the biomarkers procollagen type II carboxy-terminal propeptide (PIICP), collagen type-II C-telopeptide fragments (CTX-II), and type II collagen cleavage neoepitope (C2C) during pathogenesis, the present study established a theoretical basis for the evaluation and early diagnosis of OA. In an experiment, 10 ng/ml IL-1ß was used to the treat chondrocyte-induced OA models in vitro for 0, 12, 24 and 48 h. Western blotting was used to detect the expression levels of matrix metalloproteinase (MMP)-3, MMP-13, and inducible nitric oxide synthase (iNOS) protein at each time-point. The concentrations of CTX-II, C2C, and PIICP in the cell culture supernatant were detected by ELISA kit. A biochemical kit was used to detect changes of nitric oxide (NO) in the cell culture supernatant. In addition, chondrocytes were treated with 10 ng/ml IL-1ß for 0, 30, 60 and 90 min and the translocation and phosphorylation of the NF-κB pathway were investigated by western blotting. Following IL-1ß stimulation, the NF-κB pathway was activated to increase the expression levels of MMPs and iNOS synthesis downstream of the pathway, resulting in an increased degradation of type II collagen (Col II). To sum up, pro-inflammatory IL-1ß induced an OA chondrocyte model. During the development of OA, the expression of MMPs and NO increased and Col II was degraded.

7.
Mol Neurobiol ; 58(7): 3471-3483, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33733293

ABSTRACT

Ketamine is a widely used analgesic and anesthetic in obstetrics and pediatrics. Ketamine is known to promote neuronal death and cognitive dysfunction in the brains of humans and animals during development. Monosialotetrahexosyl ganglioside (GM1), a promoter of brain development, exerts neuroprotective effects in many neurological disease models. Here, we investigated the neuroprotective effect of GM1 and its potential underlying mechanism against ketamine-induced apoptosis of rats. Seven-day-old Sprague Dawley (SD) rats were randomly divided into the following four groups: (1) group C (control group: normal saline was injected intraperitoneally); (2) group K (ketamine); (3) group GM1 (GM1 was given before normal saline injection); and (4) GM1+K group (received GM1 30 min before continuous exposure to ketamine). Each group contained 15 rats, received six doses of ketamine (20 mg/kg), and was injected with saline every 90 min. The Morris water maze (MWM) test, the number of cortical and hippocampal cells, apoptosis, and AKT/GSK3ß pathway were analyzed. To determine whether GM1 exerted its effect via the PI3K/AKT/GSK3ß pathway, PC12 cells were incubated with LY294002, a PI3K inhibitor. We found that GM1 protected against ketamine-induced apoptosis in the hippocampus and cortex by reducing the expression of Bcl-2 and Caspase-3, and by increasing the expression of Bax. GM1 treatment increased the expression of p-AKT and p-GSK3ß. However, the anti-apoptotic effect of GM1 was eliminated after inhibiting the phosphorylation of AKT. We showed that GM1 lessens ketamine-induced apoptosis in the hippocampus and cortex of young rats by regulating the PI3K/AKT/GSK3ß pathway. Taken together, GM1 may be a potential preventive treatment for the neurotoxicity caused by continuous exposure to ketamine.


Subject(s)
Cerebral Cortex/metabolism , G(M1) Ganglioside/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Ketamine/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Anesthetics, Dissociative/toxicity , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Cerebral Cortex/drug effects , Hippocampus/drug effects , Neuroprotection/drug effects , Neuroprotection/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...