Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
In Vivo ; 38(1): 184-189, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38148065

RESUMO

BACKGROUND/AIM: Neurogenesis is an important process in the recovery from neurological damage caused by ischemic lesions. Endogenous neurogenesis is insufficient to restore neuronal damage following cerebral ischemia. Dexmedetomidine (DEX) exerts neuroprotective effects against cerebral ischemia and ischemia/reperfusion injury. DEX promotes neurogenesis, including neuronal proliferation and maturation in the hippocampus. In a previous study, we showed that early neurogenesis increased 3 days after bilateral common carotid artery occlusion (BCCAO). In this study, we investigated the effect of DEX on neurogenesis 3 days after BCCAO. MATERIALS AND METHODS: Male Sprague-Dawley (SD) rats (7-8 weeks old) were used as a BCCAO model. Right and left common carotid arteries of the rats were occluded using 4-0 silk sutures. Two hours after surgery, an intracranial DEX injection was administered to rats that underwent surgery using a stereotaxic injector. Brains were obtained from control and BCCAO rats 3 days after surgery. Immunohistochemistry was performed on the cortex and dentate gyrus of the hippocampus using a NeuN antibody. Western blot was performed with HIF1α and brain-derived neurotrophic factor (BDNF) antibodies. RESULTS: The number of mature neurons decreased 3 days after BCCAO, but DEX treatment alleviated neural loss in the parietal cortex and hippocampus. Up-regulation of BDNF was also observed after dexmedetomidine treatment. CONCLUSION: Stereotaxic injection of dexmedetomidine alleviates neural loss following BCCAO by up-regulating BDNF expression.


Assuntos
Isquemia Encefálica , Dexmedetomidina , Ratos , Masculino , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Ratos Sprague-Dawley , Regulação para Cima , Dexmedetomidina/farmacologia , Dexmedetomidina/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Artéria Carótida Primitiva/metabolismo
2.
J Neurophysiol ; 129(5): 1145-1156, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37042554

RESUMO

Postoperative neurocognitive dysfunction (PND) is a common postoperative complication. Autophagy is correlated with the pathogenesis of PND. This study investigated the potential role of autophagy in the neuroprotection of dexmedetomidine (Dex) pretreatment in PND. The PND rat model was established by abdominal surgery. The cognitive function of rats was evaluated by Y-maze 3 days after surgery. Nissl staining assessed postoperative hippocampal damage. Immunofluorescence detected the expression of microglial activation (Iba-1) and autophagy-related protein (LC3B) in hippocampal tissues. Western blot detected the autophagy-related protein expression (Beclin 1, LC3B, and p62), proinflammatory cytokines, and the protein activation of the autophagy-related LKB1/AMPK/ULK-1 signaling pathway. RT-PCR quantified the expression of IL-1ß, TNF-α, and IL6. In this study, we found that Dex pretreatment improved spatial memory function impairment and reduced abdominal surgery-induced hippocampal tissue damage. Dex pretreatment significantly increased the expression of Beclin 1 and LC3 II/I and decreased the expression of p62 in the hippocampus after surgery. Furthermore, Dex effectively inhibited microglial activation and proinflammatory cytokines by enhancing autophagy in the hippocampus. Pretreatment with 3-MA, an autophagy inhibitor, significantly weakened the inhibitory effect of Dex on postoperative neuroinflammation. We further demonstrated that Dex suppressed surgery-induced neuroinflammation by activating the LKB1/AMPK/ULK-1 signaling pathway. In conclusion, our study indicated that Dex inhibited hippocampal neuroinflammation and ameliorated PND by enhancing autophagy after surgery in rats, which was related to the LKB1/AMPK/ULK-1 signaling pathway. These findings provide a potential therapeutic prospect for PND.NEW & NOTEWORTHY Dex inhibits hippocampal neuroinflammation and attenuates early cognitive impairment by enhancing autophagy following surgery in rats. Dex may protect postoperative cognitive function by activating the LKB1/AMPK/ULK-1 signaling pathway.


Assuntos
Disfunção Cognitiva , Dexmedetomidina , Complicações Cognitivas Pós-Operatórias , Ratos , Animais , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Ratos Sprague-Dawley , Doenças Neuroinflamatórias , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/farmacologia , Proteína Beclina-1/metabolismo , Proteína Beclina-1/farmacologia , Complicações Cognitivas Pós-Operatórias/tratamento farmacológico , Citocinas , Hipocampo/metabolismo , Autofagia
3.
Eur J Pharm Biopharm ; 183: 47-60, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36565969

RESUMO

The purpose of this study was to prepare a dexmedetomidine (Dex) 72 h long-acting patch by the combined use of ion-pair strategy and chemical enhancers (CEs), and to investigate molecular mechanisms of drug-loading enhancement and controlled release. The formulation of patch was optimized by single-factor investigation and Box-Behnken design. The pharmacokinetics, analgesic pharmacodynamics and irritation of the formulation were evaluated, respectively. Moreover, the effects of ion-pairs and CEs on the patch were characterized by DSC, rheology study, FTIR, and molecular docking, and the effects on the skin were evaluated by Attenuated Total Reflection Fourier Transform Infrared Spectroscopy (ATR-FTIR), Raman study, and molecular dynamics, respectively. The optimized formulation was 17.00 % (w/w) Dex-NA (Naphthoic acid), 7.20 % Polyglyceryl-3 dioleate (POCC), 25-AAOH as pressure sensitive adhesives (PSA) and 66.50 µm in thickness. Compared with the control group (Cmax = 62.02 ± 16.55 ng/mL, MRT0-t = 26.74 ± 1.27 h), the pharmacokinetics behavior of the optimization group was more stable and durable (Cmax = 31.22 ± 13.26 ng/mL, MRT0-t = 33.62 ± 1.62 h). Besides, it also showed good analgesic effect and no obvious irritation. The results indicated that Dex-NA both increased the drug-PSA interactions and inhibited the penetration of the drug into the skin. POCC increased the molecular mobility of the PSA and disrupted skin lipids thereby improving the drug penetration rate. In summary, the Dex long-acting patch was developed, which provided a reference for the combined application of ion-pair strategy and CEs in other long-acting transdermal delivery.


Assuntos
Dexmedetomidina , Absorção Cutânea , Dexmedetomidina/farmacologia , Dexmedetomidina/metabolismo , Simulação de Acoplamento Molecular , Preparações de Ação Retardada/farmacologia , Adesivo Transdérmico , Pele/metabolismo , Administração Cutânea , Adesivos/química , Espectroscopia de Infravermelho com Transformada de Fourier
4.
Neurocrit Care ; 38(3): 688-697, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36418766

RESUMO

BACKGROUND: Microglia are a primary mediator of the neuroinflammatory response to neurologic injury, such as that in traumatic brain injury. Their response includes changes to their cytokine expression, metabolic profile, and immunophenotype. Dexmedetomidine (DEX) is an α2 adrenergic agonist used as a sedative in critically ill patients, such as those with traumatic brain injury. Given its pharmacologic properties, DEX may alter the phenotype of inflammatory microglia. METHODS: Primary microglia were isolated from Sprague-Dawley rats and cultured. Microglia were activated using multiple mediators: lipopolysaccharide (LPS), polyinosinic-polycytidylic acid (Poly I:C), and traumatic brain injury damage-associated molecular patterns (DAMP) from a rat that sustained a prior controlled cortical impact injury. After activation, cultures were treated with DEX. At the 24-h interval, the cell supernatant and cells were collected for the following studies: cytokine expression (tumor necrosis factor-α [TNFα], interleukin-10 [IL-10]) via enzyme-linked immunosorbent assay, 6-phosphofructokinase enzyme activity assay, and immunophenotype profiling with flow cytometry. Cytokine expression and metabolic enzyme activity data were analyzed using two-way analysis of variance. Cell surface marker expression was analyzed using FlowJo software. RESULTS: In LPS-treated cultures, DEX treatment decreased the expression of TNFα from microglia (mean difference = 121.5 ± 15.96 pg/mL; p < 0.0001). Overall, DEX-treated cultures had a lower expression of IL-10 than nontreated cultures (mean difference = 39.33 ± 14.50 pg/mL, p < 0.0001). DEX decreased IL-10 expression in LPS-stimulated microglia (mean difference = 74.93 ± 12.50 pg/mL, p = 0.0039) and Poly I:C-stimulated microglia (mean difference = 23.27 ± 6.405 pg/mL, p = 0.0221). In DAMP-stimulated microglia, DEX decreased the activity of 6-phosphofructokinase (mean difference = 18.79 ± 6.508 units/mL; p = 0.0421). The microglial immunophenotype was altered to varying degrees with different inflammatory stimuli and DEX treatment. CONCLUSIONS: DEX may alter the neuroinflammatory response of microglia. By altering the microglial profile, DEX may affect the progression of neurologic injury.


Assuntos
Lesões Encefálicas Traumáticas , Dexmedetomidina , Ratos , Animais , Dexmedetomidina/farmacologia , Dexmedetomidina/metabolismo , Dexmedetomidina/uso terapêutico , Interleucina-10/metabolismo , Interleucina-10/uso terapêutico , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ratos Sprague-Dawley , Lipopolissacarídeos/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Citocinas/metabolismo , Inflamação/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Poli I/metabolismo , Poli I/uso terapêutico
5.
FASEB J ; 36(11): e22608, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36250975

RESUMO

Renal microvascular endothelial cells (RMECs), which are closely related to regulation of vascular reactivity and modulation of inflammation, play a crucial role in the process of renal ischemia and reperfusion (I/R) injury. Previous studies have reported the protective effects of dexmedetomidine (DEX) against renal I/R injury, but little is known about the role of DEX on RMECs. This study aimed to investigate whether DEX alleviated renal I/R injury via acting on the RMECs. Mice underwent bilateral renal artery clamping for 45 min followed by reperfusion for 48 h, and the cultured neonatal mice RMECs were subjected to hypoxia for 1 h followed by reoxygenation (H/R) for 24 h. The results suggest that DEX alleviated renal I/R injury in vivo and improved cell viability of RMECs during H/R injury in vitro. Gene sequencing revealed that the PI3K/Akt was the top enriched signaling pathway and the endothelial cells were widely involved in renal I/R injury. DEX activated phosphorylation of PI3K and Akt, increased eNOS expression, and attenuated inflammatory responses. In addition, the results confirmed the distribution of α2 adrenoreceptor (α2 -AR) in RMECs. Furthermore, the protective effects of DEX against renal I/R injury were abolished by α2 -AR antagonist (atipamezole), which was partly reversed by the PI3K agonist (740 Y-P). These findings indicated that DEX protects against renal I/R injury by activating the PI3K/Akt-eNOS pathway and inhibiting inflammation responses via α2 -AR in RMECs.


Assuntos
Dexmedetomidina , Traumatismo por Reperfusão , Animais , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Células Endoteliais/metabolismo , Inflamação/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais
6.
Comput Math Methods Med ; 2022: 8714221, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213579

RESUMO

Objective: To study the effect of dexmedetomidine on cognitive function in rats with cognitive impairment after partial hepatectomy and its mechanism. Methods: 60 SD rats were randomly divided into 4 groups (n = 15): blank control group (CG group), sham operation group (Sham group), cognitive impairment model group (POCD group), and dexmedetomidine + cognitive impairment model group (DEX group). Rats in the POCD group underwent left lobe hepatectomy and intraperitoneal injection of the same amount of normal saline after resuscitation. Rats in the DEX group underwent left lobe hepatectomy and intraperitoneal injection of dexmedetomidine 50 µg/kg. Group CG was not operated on and the same amount of normal saline was injected intraperitoneally. In the Sham group, liver resection was not allowed after the abdominal incision, and normal saline was injected intraperitoneally. Rats were injected every 24 hours for 5 consecutive days. Morris water maze (MWM) were used to evaluate the effects of dexmedetomidine on learning and memory ability of POCD rats. TUNEL method was used to detect apoptotic neurons in the hippocampus. INOS, Arg-1, IL-6, and TNF-αexpression levels were detected. Western blot detects the expression level of TNF-α, Bcl-2, and NF-κB protein. Result: Compared with the CG group, the escape latency of the other three groups was prolonged on the 5th day after the operation, and the number of crossing the platform was reduced. Compared with the Sham group, the escape latency of the POCD group and DEX group was significantly prolonged, and the number of crossing the platform was significantly reduced on day 5 (P < 0.05). Compared with the POCD group, the DEX group shortened the escape latency and increased the number of crossing the platform on the 5th day (P < 0.05). It shows that the spatial learning and memory function of rats has been restored to a certain extent.The number of iNOS and Arg-1 positive cells in the POCD group and DEX group was higher than that in the control group, and the number of Arg-1 positive cells in the DEX group was higher than that in the POCD group (P < 0.05). Western blot results the expression of Bcl-2 and NF-κB protein in POCD group, and DEX group was higher than that of the sham group (P < 0.05). The expression of Bcl-2 and NF-κB protein was the most in POCD group. The expression of Bcl-2 and NF-κB protein in DEX group was lower than that in POCD group (P < 0.05). Conclusion: Behavioral results showed that the learning and cognitive ability of POCD model rats after hepatectomy was impaired, and inflammatory factors and activated microglia were found in the hippocampus of POCD rats. Dexmedetomidine may improve the brain function of POCD rats by inhibiting neuronal apoptosis,partly through NF-κB apoptosis pathway.


Assuntos
Disfunção Cognitiva , Dexmedetomidina , Animais , Ratos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Hepatectomia/efeitos adversos , Hipocampo , Interleucina-6 , Inflamação Neurogênica/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
7.
Clin Lab ; 68(8)2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35975484

RESUMO

BACKGROUND: The purpose of this study was to investigate the protective effects of dexmedetomidine (DEX) on total body radiation-induced acute liver injury in mice and to explore the possible mechanisms. METHODS: A total of 40 mice were randomly divided into the Control group (Group C), Dexmedetomidine group (Group Dex), Radiation group (Group R), and Group R+Dex. Mice in Group Dex and Group R+Dex were intraperitoneally injected with 10 µg/mL Dex at 50 mg/kg. Both Group C and Group R received normal saline instead of Dex. Mice were treated via continuous administration for 10 days and injection once a day (pre-administration for 3 days and 7 days after radiation). One hour after administration on the third day, the mice in Group R and R+Dex received total body radiation with a total dose of 6 Gy at a rate of 2 Gy/min. Group C received sham radiation. Levels of aspartate aminotransferase (AST), serum alanine aminotransferase (ALT), and liver levels of tumor necrosis factor (TNF-α), interleukin-1ß (IL-1ß), reactive oxygen species (ROS), superoxide dismutase (SOD), malondialdehyde (MDA) were measured. HE staining was employed to evaluate the pathological changes in liver tissues, and the expressions of Nrf2 and HO-1 proteins in the liver were measured by western blot. RESULTS: Compared with group C, serum levels of AST and ALT, liver TNF-α, IL-1ß, MDA, and ROS levels increased, and SOD decreased in Group R. Group R mice had higher liver injury scores, and the protein expressions of Nrf2 and HO-1 proteins were lower (p < 0.05). Compared with Group R, the levels of AST, ALT, TNF-α, IL-1ß, MDA, and ROS decreased, SOD increased, liver injury scores were lower, and the expressions of Nrf2 and HO-1 proteins were higher in the Group R+Dex group (all p < 0.05). CONCLUSIONS: Dex exhibits a protective effect on reducing acute radiation-induced liver injury and oxidative stress, and the mechanism may be associated with the activation of Nrf2/HO-1 pathways.


Assuntos
Dexmedetomidina , Fator 2 Relacionado a NF-E2 , Animais , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Fígado/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase , Fator de Necrose Tumoral alfa
8.
Bioengineered ; 13(5): 13300-13311, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35635079

RESUMO

Dexmedetomidine (Dex) protects the heart from ischemia/reperfusion (I/R) injury. The differential expression of long non-coding RNAs (lncRNAs) is associated with myocardial injury, but whether the lncRNA colon cancer-associated transcript 1 (CCAT1) is associated with Dex-mediated myocardial protection remains unclear. In this study, a hypoxia/reoxygenation (H/R) H9C2 model was established to simulate the in vitro characteristics of I/R. CCAT1 and microRNA (miR)-8063 expression levels in H/R H9C2 cells pretreated with Dex were determined via quantitative reverse transcription-polymerase chain reaction. The survival and apoptotic rates of H9C2 cells were determined via cell counting kit-8 and flow cytometry assays. Wnt3a, Wnt5a, and ß-catenin protein levels were measured via western blotting. Luciferase and RNA immunoprecipitation assays were used to explore the binding relationship between miR-8063 and CCAT1. Dex pretreatment increased H/R H9C2 cell viability and CCAT1 expression, while decreasing the cell apoptosis and Wnt3a, Wnt5a, and ß-catenin protein levels. Knockdown of CCAT1 abolished the protective effects of Dex on H/R H9C2 cells, and the downregulation of miR-8063 expression eliminated the effect of CCAT1 knockdown. These results revealed that CCAT1, a sponge for miR-8063, is involved in Dex-mediated H9C2 cell H/R injury by negatively targeting miR-8063 and inactivating the Wnt/ß-catenin pathway. Dex protects H9C2 cells from H/R impairment by regulating the lncRNA CCAT1/miR-8063/Wnt/ß-catenin axis.


Assuntos
Neoplasias do Colo , Dexmedetomidina , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Animais , Neoplasias do Colo/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Hipóxia/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , RNA Longo não Codificante/metabolismo , Ratos , Traumatismo por Reperfusão/metabolismo , beta Catenina/metabolismo
9.
Neural Plast ; 2022: 1353778, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35494481

RESUMO

Surgery-induced microglial activation is critical in mediating postoperative cognitive dysfunction (POCD) in elderly patients, where the important protective effect of dexmedetomidine has been indicated. However, the mechanisms of action of dexmedetomidine during the neuroinflammatory response that underlies POCD remain largely unknown. We found that lipopolysaccharide (LPS) induced substantial inflammatory responses in primary and BV2 microglial cells. The screening of differentially expressed miRNAs revealed that miR-103a-3p was downregulated in these cell culture models. Overexpression of miR-103a-3p mimics and inhibitors suppressed and enhanced the release of inflammatory factors, respectively. VAMP1 expression was upregulated in LPS-treated primary and BV-2 microglial cells, and it was validated as a downstream target of miR-103-3p. VAMP1-knockdown significantly inhibited the LPS-induced inflammatory response. Dexmedetomidine treatment markedly inhibited LPS-induced inflammation and the expression of VAMP1, and miR-103a-3p expression reversed this inhibition. Moreover, dexmedetomidine mitigated microglial activation and the associated inflammatory response in a rat model of surgical trauma that mimicked POCD. In this model, dexmedetomidine reversed miR-103a-3p and VAMP1 expression; this effect was abolished by miR-103a-3p overexpression. Taken together, the data show that miR-103a-3p/VAMP1 is critical for surgery-induced microglial activation of POCD.


Assuntos
Dexmedetomidina , MicroRNAs , Complicações Cognitivas Pós-Operatórias , Idoso , Animais , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Humanos , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Microglia/metabolismo , Ratos , Proteína 1 Associada à Membrana da Vesícula/metabolismo
10.
Psychopharmacology (Berl) ; 239(7): 2171-2186, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35412062

RESUMO

RATIONALE: Due to its anti-inflammatory effect, dexmedetomidine (DEX) can confer neuroprotection in postoperative neurocognitive disorders (NCD). Here, the mechanism responsible for this effect of DEX is rarely ascertained. OBJECTIVES: Our research was implemented to figure out mechanism governing the protection of DEX against hippocampal neuroinflammation in postoperative NCD. METHODS: Exploratory laparotomy was applied for generating a postoperative NCD mouse model before bilateral hippocampal injection with microRNA (miR)-329-3p-agomir and intraperitoneal injection with DEX. Cognitive function of mice was evaluated by water maze test and fear conditioning test. Immunofluorescence was performed to assess microglial activation in hippocampus. After cell transfection and DEX treatment, mouse microglial cells (BV-2) were stimulated by lipopolysaccharide (LPS). IL-1ß, IL-6, and TNF-α levels and the number of phagocytes were assessed by ELISA and flow cytometry. Dual-luciferase reporter assay was adopted to assess the relationship between miR-329-3p and CREB1. RESULTS: miR-329-3p expression was reduced in the postoperative NCD mice after DEX treatment. DEX treatment or miR-329-3p downregulation caused attenuated cognitive dysfunction and microglia activation as well as reduced IL-1ß, IL-6, and TNF-α levels in the hippocampus of the postoperative NCD mice. Mechanistically, miR-329-3p inversely targeted CREB1 that activated IL1RA in LPS-induced BV-2 cells. DEX treatment, miR-329-3p inhibition, or CREB1 or IL1RA upregulation curtailed the release of proinflammatory proteins and the number of phagocytes in LPS-induced BV-2 cells. CONCLUSIONS: Collectively, our data provided the novel insight of the neuroprotective mechanism of DEX in postoperative NCD pertaining to the miR-329-3p/CREB1/IL1RA axis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Dexmedetomidina , Proteína Antagonista do Receptor de Interleucina 1 , MicroRNAs , Doenças Neuroinflamatórias , Complicações Cognitivas Pós-Operatórias , Animais , Dexmedetomidina/metabolismo , Dexmedetomidina/uso terapêutico , Hipocampo , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos , Camundongos , MicroRNAs/genética , Doenças Neuroinflamatórias/tratamento farmacológico , Complicações Cognitivas Pós-Operatórias/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo
11.
Bioengineered ; 13(4): 9369-9386, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35387565

RESUMO

Myocardial hypoxia/reoxygenation (H/R) injury is a common pathological change in patients with acute myocardial infarction undergoing reperfusion therapy. Dexmedetomidine (DEX) has been found to substantially improve ischemia-mediated cell damage. Here, we focus on probing the role and mechanism of DEX in ameliorating myocardial H/R injury. Oxygen-glucose deprivation and reoxygenation (OGD/R) were applied to construct the H/R injury model in human myocardial cell lines. After different concentrations of DEX's treatment, cell counting kit-8 (CCK-8) assay and BrdU assay were employed to test cell viability. The profiles of apoptosis-related proteins Bcl2, Bax, Bad and Caspase3, 8, 9 were determined by Western blot (WB). The expression of inflammatory factors interleukin 1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) was checked by reverse transcription-polymerase chain reaction (RT-PCR). By conducting WB, we examined the expression of NF-κB, Sirt1, Tet methylcytosine dioxygenase 1 (TET1) and DNA methylation-related proteins (DNA methyltransferase 1, DNMT1; DNA methyltransferase 3 alpha, DNMT3A; and DNA methyltransferase 3 beta, DNMT3B). Our data showed that OGD/R stimulation distinctly hampered the viability and elevated apoptosis and inflammatory factor expression in cardiomyocytes. DEX treatment notably impeded myocardial apoptosis and inflammation and enhanced cardiomyocyte viability. OGD/R enhanced total DNA methylation levels in cardiomyocytes, while DEX curbed DNA methylation. In terms of mechanism, inhibiting TET1 or Sirtuin1 (Sirt1) curbed the DEX-mediated myocardial protection. TET1 strengthened demethylation of the Sirt1 promoter and up-regulated Sirt1. DEX up-regulates Sirt1 by accelerating TET1 and mediating demethylation of the Sirt1 promoter and improves H/R-mediated myocardial injury.


Assuntos
Dexmedetomidina , Dioxigenases , Apoptose/genética , DNA/metabolismo , Desmetilação do DNA , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Dioxigenases/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Metiltransferases/metabolismo , Oxigenases de Função Mista/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo
12.
Transpl Immunol ; 72: 101577, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35272011

RESUMO

BACKGROUND: This study investigates the effect of dexmedetomidine (DEX), a highly selective agonist of alpha 2-adrenergic receptors (α2-ARs), on the regulation of hepatic macrophage activation in liver regeneration. METHODS: A two-thirds partial hepatectomy (PHx) mouse model was performed. DEX (25 µg/kg) or a vehicle control (saline) was injected i.p. at 30 min before and every 12 h after PHx. The expression of α2B-ARs in the liver was detected using immunofluorescence staining. The effects of DEX on liver regeneration were assessed by Ki67 staining. The gene expression of inflammatory cytokines in isolated hepatic macrophages was quantified 36 h after the PHx. RESULTS: α2B-ARs colocalized with hepatic macrophages after the PHx. The number of Ki67-positive hepatocytes in the mice treated with DEX was markedly increased (p < 0.05). The increases in Ki67-positive hepatocytes after treatment with DEX were inhibited in the macrophage-depleted mice. DEX treatment inhibited the expression of major pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, and tumor necrosis factor and elevated the expression of anti-inflammatory cytokines IL-4, IL-10, and transforming growth factor-ß1 in hepatic macrophages 36 h after the PHx (p < 0.05). CONCLUSIONS: The α2B-AR subtype is expressed in hepatic macrophages after a PHx. DEX modulates hepatic macrophage activation toward an anti-inflammatory phenotype via α2B-AR, which promotes the process of liver regeneration.


Assuntos
Dexmedetomidina , Regeneração Hepática , Animais , Anti-Inflamatórios , Citocinas/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Hepatectomia , Antígeno Ki-67/metabolismo , Fígado/metabolismo , Regeneração Hepática/fisiologia , Ativação de Macrófagos , Camundongos
13.
Eur J Pharmacol ; 920: 174806, 2022 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-35150656

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive degeneration in nigrostriatal dopamine pathway that is essential to control motor functions. Dexmedetomidine (DEX), a sedative and analgesic drug, is often used in patients with PD undergoing surgery. Although DEX seems to have promising future applications in neuroprotection, whether and how DEX alter the function of nigrostriatal circuit and its roles on motor deficits in PD remain unclear. Here we report that DEX attenuated motor deficits in a dose-dependent manner and protected the degeneration of dopaminergic neurons in MPTP-induced PD model mice. The DEX acted on the neurons in the nigrostriatal circuits, including activation of dopaminergic neurons and the reduction of the excitabilities of striatal neurons via dopamine D2 receptors. We further found that DEX prevented the increase in glutamatergic transmission of cholinergic interneurons (CINs) to alleviate motor dysfunction. It also decreased the intrinsic excitability and glutamatergic transmission of striatal D2 medium spiny neurons (D2-MSNs). Finally, D2 receptor antagonists prevented the restoration of DEX on motor deficits. These results demonstrate that DEX, a neuroprotective drug, restores the function of nigrostriatal neurons and improves the motor deficits, providing a potential neural mechanism of the effects of anesthetic drugs on PD progression.


Assuntos
Dexmedetomidina , Doença de Parkinson , Animais , Corpo Estriado/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/metabolismo
14.
J Clin Lab Anal ; 36(7): e24119, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34882841

RESUMO

BACKGROUND: Myocardial infarction (MI) is a serious cardiovascular disease associated with myocardial ischemia/reperfusion (I/R) injury. Dexmedetomidine (Dex), an α2-adrenoceptor agonist, has been reported to protect against I/R injury. We examined the cardioprotective effects of Dex on cardiomyocytes under hypoxia/reoxygenation (H/R) conditions and explored the underlying mechanisms. MATERIALS AND METHODS: A H/R model was established to mimic the MI injury. The CCK-8 assay was performed to measure cell viability. Cellular apoptosis was measured using the Annexin V fluorescein isothiocyanate (FITC)-propidium iodide (PI) staining. The levels of interleukin (IL)-1α and tumor necrosis factor (TNF)-α, and the activity of lactate dehydrogenase (LDH) were measured using a commercial enzyme-linked immunosorbent assay (ELISA) kit. Reactive oxygen species (ROS) were measured using the 2'-7' dichlorofluorescein diacetate (DCFH-DA) staining assay. In addition, the levels of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD), catalase (CAT), and caspase-3 were measured using a commercial kit. siRNA was used to silence Bcl-2, catalase, or STAT3. Western blotting was used to measure the change in the levels of proteins. RESULTS: Dex improved the cell viability and inhibited the inflammatory response in H9c2 cells exposed to H/R treatment. In addition, Dex inhibited apoptosis and alleviated the endoplasmic reticulum (ER) stress and oxidative stress in H9c2 cells under the H/R treatment. Mechanism investigation showed that Dex inhibited the intrinsic pathway of apoptosis. Moreover, Dex enhanced the activation of the JAK2/STAT3 signaling pathway in H/R-treated H9c2 cells. CONCLUSION: Altogether, our findings suggested Dex as a promising therapeutic agent for myocardial I/R.


Assuntos
Dexmedetomidina , Traumatismo por Reperfusão Miocárdica , Apoptose , Catalase , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacologia , Humanos , Hipóxia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/metabolismo , Estresse Oxidativo
15.
Eur J Pharmacol ; 892: 173733, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33220278

RESUMO

Over the past decade, dexmedetomidine (DEX) has been found to possess an anti-inflammatory effect. However, the local anti-inflammatory mechanism of DEX has not been fully clarified. Some intracellular inflammatory pathways lead to negative feedback during the inflammatory process. The cyclooxygenase (COX) cascade synthesizes prostaglandins (PGs) and plays a key role in inflammation, but is known to also have anti-inflammatory properties through an alternative route of a PGD2 metabolite, 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2), and its receptor, peroxisome proliferator-activated receptor gamma (PPARγ). Therefore, we hypothesized that DEX inhibits LPS-induced inflammatory responses through 15d-PGJ2 and/or PPARγ activation, and evaluated the effects of DEX on these responses. The RAW264.7 mouse macrophage-like cells were pre-incubated with DEX, followed by the addition of LPS to induce inflammatory responses. Concentrations of TNFα, IL-6, PGE2, and 15d-PGJ2 in the supernatants of the cells were measured, and gene expressions of PPARγ and COX-2 were evaluated in the cells. Furthermore, we evaluated whether a selective α2 adrenoceptor antagonist, yohimbine or a selective PPARγ antagonist, T0070907, reversed the effects of DEX on the LPS-induced inflammatory responses. DEX inhibited LPS-induced TNFα, IL-6, and PGE2 productions and COX-2 mRNA expression, and the effects of DEX were reversed by yohimbine. On the other hand, DEX significantly increased 15d-PGJ2 production and PPARγ mRNA expression, and yohimbine reversed these DEX's effects. Furthermore, T0070907 reversed the anti-inflammatory effects of DEX on TNFα and IL-6 productions in the cells. These results suggest that DEX inhibits LPS-induced inflammatory responses through PPARγ activation following binding to α2 adrenoceptors.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Anti-Inflamatórios/farmacologia , Dexmedetomidina/farmacologia , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , PPAR gama/agonistas , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dexmedetomidina/metabolismo , Dinoprostona/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Ligação Proteica , Células RAW 264.7 , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
17.
Mol Med Rep ; 22(6): 5378-5384, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33173983

RESUMO

Mechanical ventilation (MV) can contribute to ventilator­induced lung injury (VILI); dexmedetomidine (Dex) treatment attenuates MV­related pulmonary inflammation, but the mechanisms remain unclear. Therefore, the present study aimed to explore the protective effect and the possible molecular mechanisms of Dex in a VILI rodent model. Adult male Sprague­Dawley rats were randomly assigned to one of seven groups (n=24 rats/group). Rats were euthanized after 4 h of continuous MV, and pathological changes, lung wet/dry (W/D) weight ratio, the levels of inflammatory cytokines (IL­1ß, TNF­α and IL­6) in the bronchoalveolar lavage fluid (BALF), and the expression levels of Bcl­2 homologous antagonist/killer (Bak), Bcl­2, pro­caspase­3, cleaved caspase­3 and the phosphorylation of ERK1/2 in the lung tissues were measured. Propidium iodide uptake and TUNEL staining were used to detect epithelial cell death. The Dex pretreatment group exhibited fewer pathological changes, lower W/D ratios and lower expression levels of inflammatory cytokines in BALF compared with the VILI group. Dex significantly attenuated the ratio of Bak/Bcl­2, cleaved caspase­3 expression levels and epithelial cell death, and increased the expression of phosphorylated ERK1/2. The protective effects of Dex could be partially reversed by PD98059, which is a mitogen­activated protein kinase (upstream of ERK1/2) inhibitor. Overall, dexmedetomidine was found to reduce the inflammatory response and epithelial cell death caused by VILI, via the activation of the ERK1/2 signaling pathway.


Assuntos
Dexmedetomidina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/metabolismo , Dexmedetomidina/metabolismo , Modelos Animais de Doenças , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Pneumonia/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
18.
Life Sci ; 259: 118249, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32798558

RESUMO

AIMS: We aimed to evaluate the effect of Dexmedetomidine (Dex) on immunology function of macrophages and inflammatory reactions in non-ventilated lung tissues from both humans and rats. MAIN METHODS: Patients scheduled for lung lobectomy were randomly assigned to traditional anesthesia group or Dex anesthesia group, 15 subjects in each group. CD68, CD86 and CD206 were used to mark activate and polarized macrophages using immunofluorescence staining in human lung tissues. Sprague-Dawley rats were used to set lung injury model and randomly divided into Control group, one-lung ventilation group (CLI group) and CLI + Dex group. Lung tissues and bronchoalveolar lavage fluid (BALF) from non-ventilated lungs were collected. The acquired lung tissues were subjected to hematoxylin-eosin (H&E) staining and the inflammatory cells in BALF were calculated. Levels of cytokines and chemokines were detected by enzyme-linked immunosorbent assays (ELISA). KEY FINDINGS: Results from humans showed that anesthesia with Dex decreased the number of both CD68 positive cells and CD86 positive cells and down-regulated level of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and monocyte chemotactic protein 1 (MCP-1) in human lung. Results from rats demonstrated that treatment with Dex reversed the increased inflammatory cells in lung and the increased levels of TNF-α, interleukin-1ß (IL-ß), MCP-1 and chemokine (C-X-C motif) ligand 1 (CXCL1) resulted from non-ventilation; Dex increased the anti-inflammatory cytokine interleukin-10 (IL-10) in BALF from non-ventilated lung. SIGNIFICANCE: This study showed that Dex modulated the activation and immunological function of macrophages in non-ventilated lung and revealed a protective role in collapsed lung injury.


Assuntos
Dexmedetomidina/farmacologia , Lesão Pulmonar/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Idoso , Animais , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Dexmedetomidina/metabolismo , Feminino , Humanos , Pulmão/citologia , Pulmão/metabolismo , Lesão Pulmonar/patologia , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Life Sci ; 257: 118004, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32621918

RESUMO

BACKGROUND: Patients undergoing cardiopulmonary bypass (CPB) often develop acute kidney injury (AKI) caused by myocardial ischemia reperfusion (MI/R), and this renal injury can be resolved notably by dexmedetomidine. Endoplasmic reticulum (ER) stress was reported to get involved in organ injury including AKI. OBJECTIVES: The current study aimed to address the correlation between MI/R induced AKI with ER stress and to assess the effects of dexmedetomidine pretreatment on AKI protection. METHOD: Patients selected for heart valve replacement surgery were randomly assigned to NS group (pre-anesthesia with 0.9% NaCl) and DEX group (pre-anesthesia with dexmedetomidine). Rat MI/R model was induced by occluding coronary artery for 30 min followed by 48-hour reperfusion. Rats were randomized into Sham (0.9% NaCl), I/R (MI/R + 0.9% NaCl) and I/R + DEX (MI/R + dexmedetomidine). Organ function and ER stress condition were evaluated by blood chemistry, pathology, and molecular test. RESULTS: Clinical data indicated dexmedetomidine pretreatment attenuated AKI and oxidative stress as well as postischemic myocardial injury in patients. Accordingly animal results suggested dexmedetomidine reduced cellular injury and improved postischemic myocardial and renal function. Dexmedetomidine also reduced myocardial and renal cells apoptosis and down-regulated ER stress. CONCLUSIONS: These results suggested that dexmedetomidine pretreatment attenuates MI/R injury-induced AKI by relieving the ER stress.


Assuntos
Dexmedetomidina/farmacologia , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Idoso , Animais , Apoptose/efeitos dos fármacos , China , Dexmedetomidina/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/fisiologia , Feminino , Humanos , Isquemia/metabolismo , Rim/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/metabolismo , Reperfusão Miocárdica/métodos , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estudos Prospectivos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Med Sci Monit ; 26: e922139, 2020 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-32419697

RESUMO

BACKGROUND Glutamate (GLU) is the most excitatory amino acid in the central nervous system and plays an important role in maintaining the normal function of the nervous system. During cerebral ischemia, massive release of GLU leads to neuronal necrosis and apoptosis. It has been reported that dexmedetomidine (DEX) possesses anti-oxidant and anti-apoptotic properties. The objective of this study was to investigate the effects of DEX on GLU-induced neurotoxicity in PC12 cells. MATERIAL AND METHODS PC12 cells were treated with 20 mM GLU to establish an ischemia-induced injury model. Cell viability was accessed by MTT assay. MDA content and SOD activity were analyzed by assay kits. Apoptosis rate, ROS production, intracellular Ca²âº concentration, and MMP were evaluated by flow cytometry. Western blot analysis was performed to analyze expressions of caspase-3, caspase-9, cyt-c, bax, and bcl-2. RESULTS PC12 cells treated with GLU exhibited reduced cell viability and increased apoptosis rates, which were ameliorated by pretreatment with DEX. DEX significantly increased SOD activity, reduced content of MDA, and decreased production of ROS in PC12 cells. In addition, DEX clearly reduced the level of intracellular Ca²âº and attenuated the decline of MMP. Moreover, DEX notably reduced expressions of caspase-3, caspase-9, cyt-c, and bax and increased expression of bcl-2. CONCLUSIONS Our findings suggest that DEX can protect PC12 cells against GLU-induced cytotoxicity, which may be attributed to its anti-oxidative property and reduction of intracellular calcium overload, as well as its ability to inhibit the mitochondria-mediated apoptotic pathway.


Assuntos
Apoptose/efeitos dos fármacos , Dexmedetomidina/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Animais , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Dexmedetomidina/metabolismo , Glucose/metabolismo , Ácido Glutâmico/efeitos adversos , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Substâncias Protetoras/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA