Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(6): 448, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918408

RESUMO

Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.


Assuntos
Aquaporina 4 , Dexmedetomidina , Sistema Glinfático , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Sevoflurano , Transdução de Sinais , Animais , Dexmedetomidina/farmacologia , Sevoflurano/farmacologia , Sevoflurano/efeitos adversos , Sistema Glinfático/efeitos dos fármacos , Sistema Glinfático/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Aquaporina 4/metabolismo , Aquaporina 4/genética , Transdução de Sinais/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Masculino
2.
Biomed Pharmacother ; 175: 116739, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38759288

RESUMO

BACKGROUND: Ketamine, as a non-competitive antagonist of N-methyl-D-aspartate (NMDA) receptors, was originally used in general anesthesia. Epidemiological data show that ketamine has become one of the most commonly abused drugs in China. Ketamine administration might cause cognitive impairment; however, its molecular mechanism remains unclear. The glymphatic system is a lymphoid system that plays a key role in metabolic waste removal and cognitive regulation in the central nervous system. METHODS: Focusing on the glymphatic system, this study evaluated the behavioral performance and circulatory function of the glymphatic system by building a short-term ketamine administration model in mice, and detected the expression levels of the 5-HT2c receptor, ΔFosb, Pten, Akt, and Aqp4 in the hippocampus. Primary astrocytes were cultured to verify the regulatory relationships among related indexes using a 5-HT2c receptor antagonist, a 5-HT2c receptor short interfering RNA (siRNA), and a ΔFosb siRNA. RESULTS: Ketamine administration induced ΔFosb accumulation by increasing 5-HT2c receptor expression in mouse hippocampal astrocytes and primary astrocytes. ΔFosb acted as a transcription factor to recognize the AATGATTAAT bases in the 5' regulatory region of the Aqp4 gene (-1096 bp to -1087 bp), which inhibited Aqp4 expression, thus causing the circulatory dysfunction of the glymphatic system, leading to cognitive impairment. CONCLUSIONS: Although this regulatory mechanism does not involve the Pten/Akt pathway, this study revealed a new mechanism of ketamine-induced cognitive impairment in non-neuronal systems, and provided a theoretical basis for the safety of clinical treatment and the effectiveness of withdrawal.


Assuntos
Astrócitos , Disfunção Cognitiva , Sistema Glinfático , Hipocampo , Ketamina , Animais , Ketamina/farmacologia , Ketamina/toxicidade , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Camundongos , Masculino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sistema Glinfático/efeitos dos fármacos , Sistema Glinfático/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Aquaporina 4/metabolismo , Aquaporina 4/genética , Receptor 5-HT2C de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/genética , Camundongos Endogâmicos C57BL , Células Cultivadas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética
3.
Mol Neurobiol ; 61(4): 2049-2062, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37840071

RESUMO

Ketamine as a glutamate receptor antagonist has a rapid, potent, and long-lasting antidepressant effect, but its specific mechanism is still not fully understood. Depression is associated with elevated levels of glutamate and astrocyte loss in the brain; the exploration of the relationships between ketamine's antidepressant effect and astrocytes has drawn great attention. Astrocytes and aquaporin 4 (AQP4) are essential components of the glymphatic system, which is a brain-wide perivascular pathway to help transport nutrients to the parenchyma and remove metabolic wastes. In this study, we investigated pyroptosis-associated protein Nlrp3/Caspase-1/Gsdmd-N expression in the hippocampus of mice and the toxic effect of high levels of glutamate on primary astrocytes. On this basis, the protective mechanism of ketamine is explored. A single administration of ketamine (10 mg/kg) remarkably relieved anxious and depressive behaviors in the sucrose preference test, elevated plus maze test, and forced swim test. Meanwhile, ketamine reduced the level of hippocampus Nlrp3 and the expression of its downstream molecules in chronic unpredictable mild stress (CUMS) mice model by western blot and reduced the colocalization of Gfap and Gsdmd by nearly 25% via immunofluorescent staining. Ketamine also increased the Gfap-positive cells and AQP4 expression in the hippocampus of the CUMS mice. More important, ketamine increased the distribution of the fluorescent tracer of CUMS mice. Treatment with 128 mM glutamate in cortical and hippocampus astrocytes increased the level of Nlrp3, and Gsdmd-N, and ketamine alleviated high glutamate-induced pyroptosis-associated proteins. In summary, these results suggest that high glutamate-induced astrocyte pyroptosis through the Nlrp3/Caspase-1/Gsdmd-N pathway which was inhibited by ketamine and ketamine can improve the damaged glymphatic function of the CUMS mice. The present study indicates that inhibiting astrocyte pyroptosis and promoting the glymphatic circulation function are a new mechanism of ketamine's antidepressant effect, and astrocyte pyroptosis may be a new target for other antidepressant medicines.


Assuntos
Sistema Glinfático , Ketamina , Ketamina/farmacologia , Sistema Glinfático/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Astrócitos/metabolismo , Piroptose , Antidepressivos/farmacologia , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Caspases/metabolismo , Depressão/metabolismo , Estresse Psicológico/metabolismo
5.
Anesth Analg ; 136(4): 779-788, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35880774

RESUMO

BACKGROUND: Ketamine is an intravenous anesthetic. However, whether ketamine can induce neurotoxicity and neurobehavioral deficits remains largely unknown. Delirium is a syndrome of acute brain dysfunction associated with anesthesia and surgery in patients, and tau protein may contribute to postoperative delirium. Finally, ketamine may affect the function of the endosome, the key organelle for tau release from neurons. Therefore, we set out to determine the effects of ketamine on delirium-like behavior in mice and on tau trafficking in cultured cells. METHODS: We used the buried-food test, open-field test, and Y-maze test in adult mice to assess the presence of delirium-like behavior in mice. We quantified tau amounts in the serum of mice. We used cell fraction methods to determine the effects of ketamine on tau intracellular trafficking, extracellular release, and endosome trafficking in cultured cells. RESULTS: Ketamine induced delirium-like behavior in mice and increased tau amounts in serum of mice. The ketamine treatments also led to increased accumulation of endosomes, as evidenced by increased endosomal markers Rab5 and Rab7. Moreover, ketamine inhibited endosome maturation, demonstrated by decreased membrane-bound but increased cytoplasm amounts of Rab5 and Rab7. Consequently, ketamine increased tau in the endosomes of cultured cells and the cell culture medium. CONCLUSIONS: These data suggest that ketamine may interfere with intracellular tau trafficking and induce delirium-like behavior, promoting future research regarding the potential neurotoxicity of anesthetics.


Assuntos
Delírio , Ketamina , Camundongos , Animais , Ketamina/toxicidade , Proteínas tau/metabolismo , Endossomos/metabolismo , Neurônios/metabolismo , Delírio/induzido quimicamente
6.
Front Immunol ; 13: 955581, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36003380

RESUMO

Surgical pain is associated with delirium in patients, and acupuncture can treat pain. However, whether electroacupuncture can attenuate the surgical pain-associated delirium via the gut-brain axis remains unknown. Leveraging a mouse model of foot incision-induced surgical pain and delirium-like behavior, we found that electroacupuncture stimulation at specific acupoints (e.g., DU20+KI1) attenuated both surgical pain and delirium-like behavior in mice. Mechanistically, mice with incision-induced surgical pain and delirium-like behavior showed gut microbiota imbalance, microglia activation in the spinal cord, somatosensory cortex, and hippocampus, as well as an enhanced dendritic spine elimination in cortex revealed by two-photon imaging. The electroacupuncture regimen that alleviated surgical pain and delirium-like behavior in mice also effectively restored the gut microbiota balance, prevented the microglia activation, and reversed the dendritic spine elimination. These data demonstrated a potentially important gut-brain interactive mechanism underlying the surgical pain-induced delirium in mice. Pending further studies, these findings revealed a possible therapeutic approach in preventing and/or treating postoperative delirium by using perioperative electroacupuncture stimulation in patients.


Assuntos
Delírio , Eletroacupuntura , Microbioma Gastrointestinal , Animais , Espinhas Dendríticas , Eletroacupuntura/métodos , Camundongos , Dor
7.
Toxicology ; 471: 153163, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35378374

RESUMO

Ketamine is an anesthetic and addictive drug that can cause cognitive dysfunction and neuroinflammation. Studies have shown that carboxy-terminal fragment derived from ß-secretase (CTF-ß) and amyloid beta (Aß), the amyloidogenic products of amyloid precursor protein (APP), can also induce neuroinflammation and impair cognitive function. However, it remains unclear whether ketamine regulates the amyloidogenic pathway. In the endosome, APP is cleaved by beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), whose activity is influenced by pH. Endosomal acidification is mainly regulated by sodium hydrogen exchanger 6 (NHE6), which leaks protons out of endosomes, and vacuolar proton translocating ATPases (V-ATPase), which pump protons into endosomes. Therefore, we hypothesized that ketamine lowers the endosomal pH by reducing the endosomal NHE6 protein level, and this hyperacidification promotes the amyloidogenic pathway. We set up C57BL/6 J mouse models using 10, 20, 40, 80, and 100 mg/kg ketamine administration and SH-SY5Y cell models using 1, 10, 100, and 1000 µM ketamine administration to investigate its effects on the amyloidogenic pathway at different doses. Western blotting results showed that 100 mg/kg ketamine treatment in vivo and 1000 µM ketamine treatment in vitro increased endosomal BACE1 and CTF-ß protein levels and reduced endosomal NHE6 and APP protein levels. The endosomal accumulation of BACE1 caused by ketamine administration was also observed using confocal imaging. Moreover, flow cytometry indicated that ketamine treatment lowered the endosomal pH value of SH-SY5Y cells. Later, cells were pretreated with monensin to restore the endosomal pH. Monensin did not affect amyloidogenic-related proteins or NHE6 directly; therefore, ketamine-promoted endosomal amyloidogenic processing and BACE1 accumulation were depleted by restoring endosomal acidity through monensin pretreatment. Finally, knockdown of NHE6 promoted the amyloidogenic pathway similarly and prevented further enhancement by ketamine. These results indicated that the effects of ketamine on the amyloidogenic pathway were dependent on the reduction of NHE6 and endosomal pH.

8.
Environ Toxicol ; 36(12): 2521-2529, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34487425

RESUMO

Long-term ketamine abuse can cause significant lower urinary tract symptoms in humans, termed ketamine-associated cystitis (KC). Here, we established a model of long-term (6 months) ketamine administration in wild-type (C57BL/6) mice. We elucidated the pathological effects of ketamine in the bladder and investigated changes in autophagy-associated protein expression (i.e., LC3, Beclin-1, and P62) and inflammatory cytokines (i.e., IL-6 and IL-1ß) in the bladder smooth muscle tissue. Long-term ketamine administration reduced the number of layers in the bladder mucosal epithelial cells (4-5 layers in the saline group vs. 2-3 layers in the ketamine groups), but increased the number of mast cells and collagen fibers. LC3-II/LC3-I, Beclin-1, IL-6, and IL-1ß protein expression in the bladder smooth muscle tissues of ketamine-treated mice was significantly increased. The mRNA and protein levels of P62 in the Ket-60 mg/kg group were also significantly increased, but not the Ket-30 mg/kg group. Our results reveal that long-term ketamine administration can cause cystitis-like pathological changes in mice, and the disordered autophagy in the bladder tissue may be involved in the persistent bladder damage following long-term administration of ketamine at 60 mg/kg.


Assuntos
Ketamina , Bexiga Urinária , Animais , Autofagia , Ketamina/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso
9.
J Mol Neurosci ; 70(12): 2068-2076, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32705526

RESUMO

Ketamine is a recreational drug that causes emotional and cognitive impairments, but its specific mechanisms of action are still unclear. Recent evidence suggests that Tau protein phosphorylation and targeted delivery to the postsynaptic area are closely related to its neurotoxicity, and our recent studies have shown that long-term ketamine administration causes excessive Tau protein phosphorylation. However, the regulatory mechanism of Tau protein phosphorylation induced by ketamine has not been clarified. In the present study, we administered a single ketamine injection and long-term (6 months) ketamine injections in C57BL/6 mice, to investigate the effects of different doses of ketamine on the expression levels of Tau protein and its phosphorylation, the expression levels and activities of the related protein phosphokinases GSK-3ß and CDK5, and the expression levels and activities of the related protein phosphatases PP2A and PP2B in the mouse hippocampus. Our results showed that both single-dose and long-term ketamine administration induced excessive phosphorylation of the Tau protein at ser202/thr205 and ser396. A single ketamine administration caused an increase in the activity of GSK-3ß (at high doses) and a decrease in the activity of PP2A. On the other hand, long-term ketamine administration resulted in an increase in the activities of GSK-3ß (at high doses) and CDK5, and a decrease in the activity of PP2A. Our results indicate that GSK-3ß, CDK5, and PP2A may be involved in ketamine-induced Tau protein phosphorylation.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/efeitos dos fármacos , Ketamina/farmacologia , Proteína Fosfatase 2/metabolismo , Proteínas tau/metabolismo , Animais , Células Cultivadas , Quinase 5 Dependente de Ciclina/genética , Esquema de Medicação , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Glicogênio Sintase Quinase 3 beta/genética , Hipocampo/metabolismo , Ketamina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
10.
Front Behav Neurosci ; 14: 60, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32362817

RESUMO

The monocarboxylate transporters (MCTs) MCT1, MCT2, and MCT4 are essential components of the astrocyte-neuron lactate shuttle (ANLS), which is a fundamental element of brain energetics. Decreased expression of MCTs can induce cognitive dysfunction of the brain. In the present study, we established a mouse model of long-term ketamine administration by subjecting mice to a 6-month course of a daily intraperitoneal injection of ketamine. These mice demonstrated learning and memory deficits and a significant decline in MCT1 and MCT4 proteins in the hippocampal membrane fraction, while cytoplasmic MCT1 and MCT4 protein levels were significantly increased. In contrast, the levels of global MCT2 protein were significantly increased. Analysis of mRNA levels found no changes in MCT1/4 transcripts, although the expression of MCT2 mRNA was significantly increased. We suggest that redistribution of hippocampal MCT1 and MCT4, but not MCT2 up-regulation, may be related to learning and memory deficits induced by long-term ketamine administration.

11.
Diabetes Res Clin Pract ; 162: 108044, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32017960

RESUMO

AIMS: The objective of the present analysis was to evaluate and quantify the risk for gestational diabetes mellitus (GDM) according to maternal age. METHODS: Three electronic databases were searched for publications from inception to July 2018. Odds ratio (OR) and 95% confidence interval (95% CI) were calculated. A dose-response analsis was performed using generalised least squares regression. Subgroup and meta-regression analyses were conducted to explore the source of identified heterogeneity among studies. RESULTS: Twenty-four studies were included in the present meta-analysis. The ORs and 95% CIs for women aged <20 years vs 25-29 years, 30-34 years, 35-39 years and ≥40 years were 0.60 (95% CI = 0.50-0.72), 1.69 (95% CI = 1.49-1.93), 2.73 (95% CI = 2.28-3.27), 3.54 (95% CI = 2.88-4.34) and 4.86 (95% CI = 3.78-6.24), respectively. Dose-response analysis showed that GDM risk exhibited a linear relationship with maternal age (Ptrend < 0.001). For each one-year increase in maternal age from 18 years, GDM risk for the overall population, Asian, and Europid increased by 7.90%, 12.74%, and 6.52%, respectively. Subgroup analyses indicated that from the age of 25, Asian women had a significantly higher risk of developing GDM than Europid women (all Pinteractions < 0.001). CONCLUSIONS: This meta-analysis demonstrates that the risk of GDM increases linearly with successive age-groups.


Assuntos
Diabetes Gestacional/epidemiologia , Idade Materna , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Gravidez , Fatores de Risco , Adulto Jovem
12.
Toxicol Lett ; 315: 107-115, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31470060

RESUMO

As a recreational drug of abuse and an injectable anesthetic, ketamine has been shown to cause cognitive dysfunction and induce psychotic states. Although the specific mechanism is still unclear, it may be linked to synaptic receptors, including the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor. Recent evidence suggests that Tau protein phosphorylation and targeted delivery to the postsynaptic area is involved in maintaining neuronal plasticity, indicating that the neurotoxicity induced by ketamine may be related to the transfer of Tau protein after phosphorylation. In this study, we established a model of long-term (6 months) ketamine administration in wild-type (C57BL/6) and Tau knockout mice to investigate the effects of different doses of ketamine administration on Tau protein expression and phosphorylation in the mouse hippocampus. We also investigated changes in AMPA receptor expression in the synaptic membrane of wild-type and Tau knockout mice. Our results showed that long-term ketamine administration led to excessive Tau protein phosphorylation at Ser202/Thr205 and Ser396, but not at Ser199, Ser262 and Ser404. Most importantly, long-term ketamine administration decreased AMPA receptor levels in the hippocampal cell membrane in a Tau protein-dependent manner. Our results reveal the role of Tau protein phosphorylation in the mechanism of ketamine neurotoxicity, suggesting that the changes of membrane AMPA receptor and synaptic function induced by ketamine are mediated by abnormal phosphorylation of Tau protein at specific sites.


Assuntos
Hipocampo/efeitos dos fármacos , Ketamina/toxicidade , Fosforilação/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Proteínas tau/toxicidade , Animais , Modelos Animais de Doenças , Ketamina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Front Psychiatry ; 10: 287, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31114516

RESUMO

Tau protein is known to play an important role in maintaining microtubule assembly and stabilization, and maintaining the normal morphology of neurons, but several studies have found that chronic stress leads to Tau hyperphosphorylation. A large number of clinical trials have found that ketamine, which is an N-methyl-D-aspartate receptor antagonist, produces a rapid, long-lasting, and potent antidepressant effect in patients suffering from major depression. This rapid antidepressant effect of ketamine, which involves many mechanisms, has attracted wide attention. However, the relationship between ketamine's antidepressant effects and Tau protein has rarely been examined. We used C57BL/6 and Tau KO mice exposed to 42 days of chronic unpredictable mild stress (the CUMS model) to investigate the effect of ketamine on behavioral changes and synaptic functioning of the hippocampus. The results showed that a single treatment of ketamine rapidly relieved the CUMS-induced anhedonia, depression-like, and anxious behaviors of the C57BL/6 mice. The abnormal behaviors were accompanied by increased levels of specific alterations of hyperphosphorylated Tau protein in cytoplasm and synapse in the hippocampus of the C57BL/6 mice, but ketamine reduced the aggregation of hyperphosphorylated Tau protein only in the synapse. We also found that CUMS exposure reduced the levels of GluA1 and PSD95 in the hippocampus of the C57BL/6 mice and that these deficits were reversed by ketamine. However, the Tau KO mice did not develop any stress-induced depressive behaviors or deficits of hippocampal function. The antidepressant effect of ketamine may decrease the levels of hyperphosphorylated Tau protein in synapse of C57BL/6 mice.

14.
Med Sci Law ; 57(3): 130-133, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28587535

RESUMO

Death from hypothermia usually *We presented a fatal case of hypothermia after being bitten by dog in this article. occurs among people exposed to cold and humid environmental conditions when they are homeless, aged, suffering from natural or psychiatric diseases and drug or alcohol intoxication. A normal healthy person dying from hypothermia due to dog bites is unusual and rare. Here, we present a fatal case of hypothermia following dog bites causing blood loss and multiple wounds on the body. A 56-year-old man was found dead in a remote roadside puddle of a small village, early in the morning. He was naked, and his body trunk and limbs had multiple irregular wounds. Gray animal hairs could be seen in parts of the wound cavities and surrounding areas. In addition, there was a kennel near the scene. Family members argued that the deceased was bitten to death by a dog. However, autopsy revealed several findings which were strongly supportive of fatal hypothermia. Moreover, we saw no obvious changes caused by blood loss, either on the body surface or internal organs. Accordingly, we concluded the true cause of his death was fatal hypothermia; bites from a dog was a necessary causative factor.


Assuntos
Mordeduras e Picadas/complicações , Hipotermia/etiologia , Animais , Causas de Morte , China , Cães , Evolução Fatal , Patologia Legal , Humanos , Masculino , Pessoa de Meia-Idade
15.
Front Pharmacol ; 8: 139, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28373844

RESUMO

Ketamine is an injectable anesthetic and recreational drug of abuse commonly used worldwide. Many experimental studies have shown that ketamine can impair cognitive function and induce psychotic states. Neuroinflammation has been suggested to play an important role in neurodegeneration. Meanwhile, ketamine has been shown to modulate the levels of inflammatory cytokines. We hypothesized that the effects of ketamine on the central nervous system are associated with inflammatory cytokines. Therefore, we set out to establish acute and chronic ketamine administration models in C57BL/6 mice, to evaluate spatial recognition memory and emotional response, to analyze the changes in the levels of the inflammatory cytokines interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α) in the mouse hippocampus, employing behavioral tests, Western blot, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and immunohistochemistry. Our results showed that ketamine at the dose of 60 mg/kg induced spatial recognition memory deficit and reduced anxiety-like behaviors in mice after chronic administration. Moreover, we found that ketamine increased the hippocampal levels of IL-6 and IL-1ß after single, multiple and long-term administration in a dose-dependent manner. However, the expression level of TNF-α differed in the mouse hippocampus under different conditions. Single administration of ketamine increased the level of TNF-α, whereas multiple and long-term administration decreased it significantly. We considered that TNF-α expression could be controlled by a bi-directional regulatory pathway, which was associated with the dose and duration of ketamine administration. Our results suggest that the alterations in the levels of inflammatory cytokines IL-6, IL-1ß, and TNF-α may be involved in the neurotoxicity of ketamine.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...