Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Neurophysiol ; 129(5): 1145-1156, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37042554

RESUMEN

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.


Asunto(s)
Disfunción Cognitiva , Dexmedetomidina , Complicaciones Cognitivas Postoperatorias , Ratas , Animales , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Ratas Sprague-Dawley , Enfermedades Neuroinflamatorias , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Beclina-1/metabolismo , Beclina-1/farmacología , Complicaciones Cognitivas Postoperatorias/tratamiento farmacológico , Citocinas , Hipocampo/metabolismo , Autofagia
2.
FASEB J ; 36(11): e22608, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36250975

RESUMEN

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.


Asunto(s)
Dexmedetomidina , Daño por Reperfusión , Animales , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Células Endoteliales/metabolismo , Inflamación/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal
3.
Neurocrit Care ; 38(3): 688-697, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36418766

RESUMEN

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.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Dexmedetomidina , Ratas , Animales , Dexmedetomidina/farmacología , Dexmedetomidina/metabolismo , Dexmedetomidina/uso terapéutico , Interleucina-10/metabolismo , Interleucina-10/uso terapéutico , Microglía/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ratas Sprague-Dawley , Lipopolisacáridos/farmacología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Citocinas/metabolismo , Inflamación/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Poli I/metabolismo , Poli I/uso terapéutico
4.
Nat Chem Biol ; 16(5): 507-512, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32152538

RESUMEN

The α2 adrenergic receptors (α2ARs) are G protein-coupled receptors (GPCRs) that respond to adrenaline and noradrenaline and couple to the Gi/o family of G proteins. α2ARs play important roles in regulating the sympathetic nervous system. Dexmedetomidine is a highly selective α2AR agonist used in post-operative patients as an anxiety-reducing, sedative medicine that decreases the requirement for opioids. As is typical for selective αAR agonists, dexmedetomidine consists of an imidazole ring and a substituted benzene moiety lacking polar groups, which is in contrast to ßAR-selective agonists, which share an ethanolamine group and an aromatic system with polar, hydrogen-bonding substituents. To better understand the structural basis for the selectivity and efficacy of adrenergic agonists, we determined the structure of the α2BAR in complex with dexmedetomidine and Go at a resolution of 2.9 Å by single-particle cryo-EM. The structure reveals the mechanism of α2AR-selective activation and provides insights into Gi/o coupling specificity.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/química , Dexmedetomidina/química , Receptores Adrenérgicos alfa 2/química , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Sitios de Unión , Microscopía por Crioelectrón , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Insectos/citología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Complejos Multiproteicos/química , Receptores Adrenérgicos alfa 2/genética , Simpaticolíticos/química , Simpaticolíticos/farmacología
5.
Pulm Pharmacol Ther ; 75: 102147, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35863724

RESUMEN

BACKGROUND: Airway hyperresponsiveness (AHR) is a clinical manifestation of airflow limitation due to abnormal tracheal and bronchial sensitivity and is the main basis for the diagnosis of asthma. Patients with AHR are at high risk of perioperative tracheal and bronchospasm, which can lead to hypoxaemia and haemodynamic instability and, in severe cases, to a life-threatening 'silent lung'. It is therefore important to reduce the incidence or intensity of AHR episodes in the perioperative period. The inflammatory response is key to the development and progression of AHR. HYPOTHESIS/PURPOSE: Based on the modulatory role of dexmedetomidine (DEX) in the inflammatory response, we hypothesised that dexmedetomidine (DEX) attenuates inflammatory properties by inhibiting the toll-like receptor 4 (TLR4)/nuclear factor (NF-κB) signalling pathway and can reduce the respiratory parameters of mechanical ventilation in ovalbumin-induced allergic airway hyperresponsiveness. STUDY DESIGN: BABL/C mice were divided into control and OVA groups (ovalbumin-induced allergy. Ten mice in all OVA models were randomly selected for in vivo invasive lung function monitoring to analyse airway resistance parameters and demonstrate successful model establishment. The remaining OVA mice were treated with dexmedetomidine 25 µg/kg for 5 days (OVA + DEX group) or dexmedetomidine 25 µg/kg + yohimbine 1 mg/kg for 5 days (OVA + DEX + yohimbine). After treatment, bronchoalveolar lavage fluid (BAL) and peripheral blood (ELISA) and lung tissue (H&E and PAS) were collected for analysis of inflammatory factors, and lung tissue was verified by PCR for genes and proteins that do correlate with inflammatory mediators. RESULTS: All airway resistance parameters were increased in OVA mice by invasive lung function monitoring. Proximal airway resistance (parameter Rn) and total respiratory resistance (parameter Rrs) were attenuated after dexmedetomidine intervention treatment. Dexmedetomidine reduced total inflammatory cell count and inflammatory infiltration of lung tissue in BALF and down-regulated IL-4 and IgE levels in BALF and peripheral blood, as shown by Giemsa, H&E, PAS staining and ELISA; this mechanism of action was found to be related to the TLR4/NFκB pathway, but not to TLR4/NFκB, as measured by PCR. CONCLUSION: Dexmedetomidine reduces hyperresponsiveness and airway inflammatory responses. This mechanism of action may be related to the TLR4/NFκB signalling pathway. Overall conclusions are presented in.


Asunto(s)
Dexmedetomidina , FN-kappa B , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Regulación hacia Abajo , Inmunoglobulina E , Interleucina-4/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Ovalbúmina , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Yohimbina/metabolismo
6.
Cell Biol Int ; 46(7): 1036-1046, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35312207

RESUMEN

Dexmedetomidine (Dex), an α2-adrenergic receptor (α2-AR) agonist, possesses cardioprotection against ischemic/hypoxic injury, but the exact mechanism is not fully elucidated. Since telomere/telomerase dysfunction is involved in myocardial ischemic damage, the present study aimed to investigate whether Dex ameliorates cobalt chloride (CoCl2; a hypoxia mimic agent in vitro)-induced the damage of H9c2 cardiomyocytes by improving telomere/telomerase dysfunction and further explored the underlying mechanism focusing on extracellular signal-regulated kinase (ERK1/2)-NF-E2-related factor 2 (Nrf2) signaling pathway. The result showed that Dex increased cell viability, decreased apoptosis, and reduced cardiomyocyte hypertrophy as illustrated by the decreases in cell surface area and the biomarker levels for cardiac hypertrophy including atrial natriuretic peptide, brain natriuretic peptide, and myosin heavy chain ß messenger RNA (mRNA) and protein in CoCl2 -exposed H9c2 cells. Intriguingly, Dex increased the telomere length and telomerase activity as well as telomere reverse transcriptase protein and mRNA levels in H9c2 cells exposed to CoCl2 , indicating that Dex promotes telomere/telomerase function under hypoxia. In addition, Dex remarkably diminished the reactive oxygen species generation, reduced malondialdehyde content, and increased antioxidative signaling as evidenced by the increases in superoxide dismutase and plasma glutathione peroxidase activities. Furthermore, Dex increased the ratio of P-ERK1/2/T-ERK1/2 and P-Nrf2/T-Nrf2 and enhanced Nrf2 nuclear translocation in CoCl2 -subjected H9c2 cells, suggesting that Dex promotes the activation of the ERK1/2-Nrf2 signaling pathway. These novel findings indicated that Dex attenuates myocardial ischemic damage and reduces myocardial hypertrophy by promoting telomere/telomerase function, which may be associated with the activation of the ERK1/2-Nrf2 signaling pathway in vitro.


Asunto(s)
Dexmedetomidina , Telomerasa , Apoptosis , Cardiomegalia/metabolismo , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Humanos , Hipoxia/metabolismo , Sistema de Señalización de MAP Quinasas , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , ARN Mensajero/metabolismo , Transducción de Señal , Telomerasa/genética , Telómero/metabolismo
7.
Clin Lab ; 68(8)2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35975484

RESUMEN

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.


Asunto(s)
Dexmedetomidina , Factor 2 Relacionado con NF-E2 , Animales , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Hígado/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/farmacología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa , Factor de Necrosis Tumoral alfa
8.
J Clin Lab Anal ; 36(7): e24119, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34882841

RESUMEN

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.


Asunto(s)
Dexmedetomidina , Daño por Reperfusión Miocárdica , Apoptosis , Catalasa , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Humanos , Hipoxia , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Estrés Oxidativo
9.
Neural Plast ; 2022: 1353778, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35494481

RESUMEN

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.


Asunto(s)
Dexmedetomidina , MicroARNs , Complicaciones Cognitivas Postoperatorias , Anciano , Animales , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Humanos , Inflamación/metabolismo , Lipopolisacáridos/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Microglía/metabolismo , Ratas , Proteína 1 de Membrana Asociada a Vesículas/metabolismo
10.
Allergol Immunopathol (Madr) ; 50(2): 16-22, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35257541

RESUMEN

BACKGROUND: Airway remodeling is implicated in the pathogenesis of asthma, and abnormal proliferation of airway smooth muscle cells (ASMCs) contribute to airway remodeling. Inflammatory mediator, transforming growth factor-ß1 (TGF-ß1), stimulates the proliferation of ASMCs, and is associated with airway remodeling in asthma. Dexmedetomidine (DEX) has been widely used in the adjuvant therapy of acute asthma. OBJECTIVE: The potential effects of DEX on extracellular matrix (ECM) production and proliferation of ASMCs were investigated in this study. MATERIAL AND METHODS: Human ASMCs were incubated with TGF-ß1 for 48 hours, and then treated with different concentrations of DEX for another 24 hours. Cell proliferation was detected by MTT and BrdU (5'-bromo-2'-deoxyuridine) staining. Flow cytometry was used to assess cell apoptosis, and western blot was applied to identify the underlying mechanism. RESULTS: TGF-ß1 induced increase in cell viability and bromodeoxyuridine (BrdU) positive cells in ASMCs while repressed cell apoptosis. Second, TGF-ß1-induced ASMCs were then treated with different concentrations of DEX. Cell viability of TGF-ß1-induced ASMCs was decreased by incubation of DEX. The number of BrdU positive cells in TGF-ß1-induced ASMCs was reduced by incubation of DEX. Moreover, incubation of DEX promoted cell apoptosis of TGF-ß1-induced ASMCs. Third, incubation of DEX attenuated TGF-ß1-induced increase in fibronectin, collagen I, MMP9, and versican in ASMCs. Lastly, the up-regulation of phosphorylated extracellular receptor kinase (p-ERK), phosphorylated Jun N-terminal Kinase (p-JNK), and p-p38 in TGF-ß1-induced ASMCs was reversed by incubation of DEX. CONCLUSION: DEX suppressed TGF-ß1-induced ECM production and proliferation of ASMCs through inactivation of p38 mitogen-activated protein kinase (MAPK) pathway, providing a potential strategy for prevention of asthma.


Asunto(s)
Dexmedetomidina , Remodelación de las Vías Aéreas (Respiratorias) , Proliferación Celular , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Matriz Extracelular/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/farmacología , Miocitos del Músculo Liso , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
11.
Med Sci Monit ; 27: e929857, 2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33730014

RESUMEN

BACKGROUND Multimodal analgesic regimens are well known as the best option for total knee arthroplasty. They include the adductor canal block, combined with local infiltration analgesia and a block of the interspace between the popliteal artery and the capsule of the posterior knee. However, these analgesic techniques all require a large amount of local anesthetics. In this study, we explored whether the quantity of local anesthetics could be decreased by using dexmedetomidine for the adductor canal block. MATERIAL AND METHODS Fifty-four patients scheduled for unilateral, primary total knee arthroplasty were allocated into 2 groups: the ropivacaine group (group R) and the dexmedetomidine group (group RD). Ropivacaine 0.5% was chosen as the initial concentration, and the concentration was decreased or increased according to the response of the previous participant. Based on Dixon's up-and-down method, the median effective concentration was calculated. RESULTS The quadriceps strength was similar between the 2 groups, both at 30 min after adductor canal block and during recovery from general anesthesia in the Postanesthesia Care Unit. None of the patients in this study exhibited bradycardia or hypotension. The median effective concentration of ropivacaine for adductor canal block was 0.29% (95% confidence interval [CI], 0.28-0.31%) in group RD, which was lower than that in group R (0.38% [95% CI, 0.36-0.41%]). CONCLUSIONS This study found perineural dexmedetomidine 1 µg/kg could reduce the median effective concentration of ropivacaine for the adductor canal block.


Asunto(s)
Artroplastia de Reemplazo de Rodilla/métodos , Dexmedetomidina/uso terapéutico , Bloqueo Nervioso/métodos , Anciano , Analgesia , Analgésicos Opioides , Anestésicos Locales/farmacología , China , Dexmedetomidina/metabolismo , Femenino , Nervio Femoral/efectos de los fármacos , Humanos , Articulación de la Rodilla/cirugía , Masculino , Persona de Mediana Edad , Manejo del Dolor/métodos , Dimensión del Dolor , Músculo Cuádriceps/cirugía , Ropivacaína/uso terapéutico , Muslo/cirugía
12.
Mol Biol Rep ; 47(5): 3629-3639, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32342432

RESUMEN

Oxidative stress induced necroptosis is important in myocardial ischemia/reperfusion injury. Dexmedetomidine (Dex), an α2-adrenoceptor (α2-AR) agonist, has protective effect on oxidative stress induced cell apoptosis, but effects of Dex and Dex-mediated α2-AR activation on oxidant induced necroptosis was unclear. H9C2 cardiomyocytes were pre-treated with or without Dex and α2-AR antagonist yohimbine hydrochloride (YOH) before being exposed to H2O2 to induce oxidative cellular damage. Cell viability and lactate dehydrogenase (LDH) were detected by ELISA kits, protein expressions of Heme Oxygenase 1(HO-1), receptor interacting protein kinase 1 (RIPK1) and receptor interacting protein kinase 3 (RIPK3) were observed by WB, and TUNEL was used to detected cell apoptosis. H2O2 significantly decreased cell viability and increased LDH release and necroptotic and apoptotic cell deaths (all p < 0.05, H2O2 vs. Control). Dex preconditioning alleviated these injuries induced by H2O2. Dex preconditioning significantly increased expression of protein HO-1 and decreased expressions of proteins RIPK1 and RIPK3 induced by H2O2, while all these protective effects of Dex were reversed by YOH (all p < 0.05, Dex + H2O2 vs. H2O2; and YOH + Dex + H2O2 vs. Dex + H2O2). However, YOH did not prevent this protective effect of Dex against H2O2 induced apoptosis (YOH + Dex + H2O2 vs. Dex + H2O2, p > 0.05). These findings indicated that Dex attenuates H2O2 induced cardiomyocyte necroptotic and apoptotic cell death respectively dependently and independently of α2-AR activation.


Asunto(s)
Dexmedetomidina/farmacología , Estrés Oxidativo/efectos de los fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dexmedetomidina/metabolismo , Hemo-Oxigenasa 1/metabolismo , Peróxido de Hidrógeno/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Necroptosis/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
13.
Med Sci Monit ; 26: e922139, 2020 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-32419697

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexmedetomidina/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Animales , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Dexmedetomidina/metabolismo , Glucosa/metabolismo , Ácido Glutámico/efectos adversos , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Sustancias Protectoras/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Med Sci Monit ; 25: 1848-1856, 2019 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-30856162

RESUMEN

BACKGROUND Sepsis is among the major antecedents of lung injury characterized by mitochondrial dysfunction. The functional integrity of the cell is influenced by mitochondrial dynamics. The present investigation evaluated the protective effects of dexmedetomidine against lung injury and speculates on the possible mechanism underlying its effects on mitochondrial function. MATERIAL AND METHODS Lung injury was induced by cecal ligation and puncture (CLP) in mice treated with 0.1, 0.3, or 0.5 mg/kg intravenous dexmedetomidine after a 30-minute surgery. The effects of dexmedetomidine were determined by the oxygenation index and the wet/dry weight ratio of the lung. The expression of mitochondrial protein was assessed by western blot analyses and real-time polymerase chain reaction, to determine the effects of dexmedetomidine on mitochondrial dynamics. The histopathology of the lung tissue was determined by hematoxylin and eosin staining, and TUNEL-positive cells were counted in TUNEL assays. Activity of caspase-3, caspase-8, and caspase-9 enzymes were determined by colorimetric assay. RESULTS Treatment with dexmedetomidine significantly attenuated changes in the oxygenation index and the wet/dry weight ratio in mice with CLP-induced lung injury. There was a significant decrease in pro-inflammatory mediators and markers of oxidative stress in the lung tissue of the dexmedetomidine-treated group compared to the negative control group. Moreover, treatment with dexmedetomidine attenuated the altered gene expression caused by mitochondrial fusion and fission in the lung tissue of mice with CLP-induced lung injury. The number of TUNEL-positive cells was significantly reduced in the dexmedetomidine-treated group compared to the negative control group. Moreover, dexmedetomidine ameliorated the altered activity of caspase-3, caspase-8, and caspase-9 enzyme in the lung tissues of CLP-induced lung injure mice. CONCLUSIONS Dexmedetomidine protected mice against CLP-induced lung injury by attenuating changes in mitochondrial fusion and fission.


Asunto(s)
Dexmedetomidina/farmacología , Dinámicas Mitocondriales/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Apoptosis , Dexmedetomidina/metabolismo , Pulmón , Masculino , Ratones , Mitocondrias , Proteínas Mitocondriales , Estrés Oxidativo , Pruebas de Función Respiratoria , Sepsis
15.
Med Sci Monit ; 25: 9073-9084, 2019 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-31782408

RESUMEN

BACKGROUND Tourniquet-related complications are a common clinical problem. In the present study, we compared the effects of dexmedetomidine vs. oxycodone in patients undergoing limb ischemia-reperfusion. MATERIAL AND METHODS Fifty-four patients undergoing unilateral lower-extremity surgery under combined spinal and epidural anesthesia were randomly assigned to a control (ischemia-reperfusion, I/R) group, a dexmedetomidine (Dex) group, and an oxycodone (Oxy) group. Tourniquet-induced hemodynamic parameters changes among groups were compared. The serum concentration of malondialdehyde (MDA), superoxide dismutase (SOD), tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), fatty acid binding protein 3 (FABP3), endothelin-1 (ET-1), and brain-derived neurotrophic factor (BDNF) were measured using ELISA before anesthesia and at 30 min and at 6 h after tourniquet release. RESULTS In the control group, tourniquet use caused significant increases in systolic arterial pressure (SAP), mean arterial pressure (MAP), diastolic arterial pressure (DAP), and rate-pressure product. Compared with Oxy, Dex significantly decreased heart rate (HR). Both Dex and Oxy lowered SAP compared with the control group. No significant difference was observed in DAP between Dex and Oxy. The levels of MDA, TNF-alpha, IL-6, FABP3, and ET-1 were significantly higher, while the SOD and BDNF were significantly lower compared to baseline in the I/R group, but the variation range of those agents was significantly smaller in the Dex and Oxy groups, and the measured values were comparable between the 2 groups. CONCLUSIONS Compared with Dex, Oxy was not inferior in mitigating tourniquet-induced hyperdynamic response, ameliorating the inflammatory reaction, and protecting remote multiple organs in lower-extremity surgery patients.


Asunto(s)
Dexmedetomidina/farmacología , Oxicodona/farmacología , Daño por Reperfusión/tratamiento farmacológico , Adulto , Anciano , Factor Neurotrófico Derivado del Encéfalo/análisis , Factor Neurotrófico Derivado del Encéfalo/sangre , China , Dexmedetomidina/metabolismo , Endotelina-1/análisis , Endotelina-1/sangre , Proteína 3 de Unión a Ácidos Grasos/análisis , Proteína 3 de Unión a Ácidos Grasos/sangre , Femenino , Hemodinámica/efectos de los fármacos , Humanos , Interleucina-6/análisis , Interleucina-6/sangre , Isquemia/complicaciones , Extremidad Inferior/cirugía , Masculino , Malondialdehído/análisis , Malondialdehído/sangre , Persona de Mediana Edad , Oxicodona/metabolismo , Enfermedades Vasculares Periféricas , Estudios Prospectivos , Distribución Aleatoria , Superóxido Dismutasa/análisis , Superóxido Dismutasa/sangre , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/sangre
16.
Cell Biol Int ; 42(9): 1250-1258, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29851220

RESUMEN

Dexmedetomidine (Dex) protects different cell types during hypoxia or ischemia-reperfusion injury by inhibiting cell apoptosis. However, the underlying mechanism and its impact on hepatic ischemia reperfusion injury are still not known. In this study, we established a model of oxygen-glucose deprivation/reperfusion (OGD/R) injury in hepatocyte HL7702 cells, and studied the impact of Dex on cell proliferation, apoptosis, and cell cycle during OGD/R. In addition, we explored the role of CCAT1 in this process. We found that Dex increased cell proliferation and inhibited cell apoptosis during OGD/R, in a concentration-dependent manner. Dex partially reversed the OGD-inhibited expression of lncRNA CCAT1. Knockdown of CCAT1 by siRNA inhibited Dex-mediated protection against OGD/R-induced injury and promoted cell apoptosis, caspase-3 expression and cell cycle arrest in the G0/G1 phase, and inhibited cell proliferation and cyclin D1 expression. In contrast, overexpression of CCAT1 by pcDNA3.0-CCAT1 enhanced Dex-mediated protection against OGD/R-induced cell injury. Thus, Dex protects hepatocytes against OGD/R injury by upregulating lncRNA CCAT1. This study suggests a novel role of CCAT1 in ischemia reperfusion injury, and lays the framework for future studies.


Asunto(s)
Dexmedetomidina/farmacología , Hepatocitos/metabolismo , ARN Largo no Codificante/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dexmedetomidina/metabolismo , Glucosa/metabolismo , Humanos , Hipoxia , Hígado/metabolismo , Oxígeno/metabolismo , Sustancias Protectoras/farmacología , ARN Largo no Codificante/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
17.
Med Sci Monit ; 24: 8925-8932, 2018 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-30531676

RESUMEN

BACKGROUND This study was designed to evaluate the effects of combined usage of dexmedetomidine (DEX) and sufentanil on young patients with postoperative delirium (POD) after general anesthesia. MATERIAL AND METHODS We randomized 100 young patients with POD into 4 groups: Group D, Group S, Group DS1, and Group DS2, with loading and maintenance doses of DEX and/or sufentanil administered according to the experimental protocol. Hemodynamic variables, standard visual analogue scale (VAS) scores, sedation agitation scale (SAS) scores, stress hormones, and inflammatory biomarkers were assessed at 5 time-points: baseline (T1); 1 h (T2), 2 h (T3), 4 h (T4), and 8 h (T5) after completion of the loading dose. RESULTS At T3-T5, hemodynamic indicators in group D were obviously higher than in the other groups (P<0.05). At T2-T5, the VAS and SAS scores were noticeably lower than those at T1 in each group (P<0.05). The VAS and SAS scores were remarkably higher in group D than those in the other groups (P<0.05). Compared with DS1, the incidence of respiratory distress decreased and the incidence of POD increased in group DS2. Compared to T1, plasma concentrations of epinephrine, norepinephrine, IL-6, and TNF-a all decreased at T2 and T5 (P<0.05). CONCLUSIONS DEX and sufentanil decrease the incidence of POD, ameliorate the abnormities of hemodynamic indicators, and decrease VAS scores, SAS scores, stress hormones, and inflammatory biomarkers, but increase the incidence of respiratory distress. DEX combined with sufentanil may play a synergistic reaction in causing respiratory distress, but remarkably decreases the incidence of POD.


Asunto(s)
Delirio/tratamiento farmacológico , Dexmedetomidina/farmacología , Sufentanilo/farmacología , Adulto , Anestesia General/efectos adversos , China , Dexmedetomidina/metabolismo , Quimioterapia Combinada/métodos , Femenino , Hemodinámica/efectos de los fármacos , Humanos , Interleucina-6/análisis , Interleucina-6/sangre , Masculino , Complicaciones Posoperatorias/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
18.
Med Sci Monit ; 24: 4961-4967, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30013022

RESUMEN

BACKGROUND Traumatic brain injury (TBI) leads to acute lung injury (ALI), in which the inflammatory response plays an important role in its pathophysiology. Recent studies suggest that dexmedetomidine (Dex) plays a protective role in acute inflammatory diseases. However, whether Dex has a protective effect on TBI-induced ALI is not clear. The aim of this study was to investigate the effect of Dex on TBI-induced ALI in mice. MATERIAL AND METHODS Mice were randomly divided into 5 groups: 1) sham group; 2) TBI group; 3) TBI+Dex group; 4) TBI+atipamezole (Atip) group; and 5) TBI+Dex+Atip group. Dex (50 µg/kg) was intraperitoneal injected immediately after TBI. The α2 adrenergic antagonist Atip (250 µg/kg) was intraperitoneal injected 15 minutes prior to Dex treatment. Then 24 hours later, the protein concentration in the bronchoalveolar lavage fluid (BALF), lung wet to dry weight ratio, hematoxylin and eosin (H&E) staining of lungs, the level of high-mobility group box protein 1(HMGB1) in serum, and the receptor for advanced glycation end products (RAGE) expression in lung were detected. RESULTS Dex ameliorated the score of lung histological examination, as well as the severity of pulmonary edema and permeability. Moreover, Dex was observed to significantly suppress the expression of HMGBI and RAGE. However, the protective effects of Dex were partially reversed by the administration of Atip. CONCLUSIONS Dex may protect against TBI-induced ALI via the HMGB1-RAGE signal pathway, and this protective effect is partly dependent on its α2 adrenoceptor agonist action.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Dexmedetomidina/farmacología , Lesión Pulmonar Aguda/prevención & control , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Líquido del Lavado Bronquioalveolar , Dexmedetomidina/metabolismo , Proteína HMGB1/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Sustancias Protectoras/farmacología , Edema Pulmonar/tratamiento farmacológico , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Electrophoresis ; 36(11-12): 1349-57, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25382218

RESUMEN

Pharmacokinetic and pharmacodynamic properties of a chiral drug can significantly differ between application of the racemate and single enantiomers. During drug development, the characteristics of candidate compounds have to be assessed prior to clinical testing. Since biotransformation significantly influences drug actions in an organism, metabolism studies represent a crucial part of such tests. Hence, an optimized and economical capillary electrophoretic method for on-line studies of the enantioselective drug metabolism mediated by cytochrome P450 enzymes was developed. It comprises a diffusion-based procedure, which enables mixing of the enzyme with virtually any compound inside the nanoliter-scale capillary reactor and without the need of additional optimization of mixing conditions. For CYP3A4, ketamine as probe substrate and highly sulfated γ-cyclodextrin as chiral selector, improved separation conditions for ketamine and norketamine enantiomers compared to a previously published electrophoretically mediated microanalysis method were elucidated. The new approach was thoroughly validated for the CYP3A4-mediated N-demethylation pathway of ketamine and applied to the determination of its kinetic parameters and the inhibition characteristics in presence of ketoconazole and dexmedetomidine. The determined parameters were found to be comparable to literature data obtained with different techniques. The presented method constitutes a miniaturized and cost-effective tool, which should be suitable for the assessment of the stereoselective aspects of kinetic and inhibition studies of cytochrome P450-mediated metabolic steps within early stages of the development of a new drug.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Electroforesis Capilar/métodos , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacocinética , Ketamina/metabolismo , Ketamina/farmacocinética , Cetoconazol/metabolismo , Cetoconazol/farmacocinética , Estereoisomerismo , gamma-Ciclodextrinas/metabolismo , gamma-Ciclodextrinas/farmacocinética
20.
In Vivo ; 38(2): 587-597, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38418149

RESUMEN

BACKGROUND/AIM: Since the use of anaesthetics has the drawback of altering radiotracer distribution, preclinical positron emission tomography (PET) imaging findings of anaesthetised animals must be carefully handled. This study aimed at assessing the cerebral [18F]F-FDG uptake pattern in healthy Wistar rats under four different anaesthesia protocols using microPET/magnetic resonance imaging (MRI) examinations. MATERIALS AND METHODS: Post-injection of 15±1.2 MBq of [18F]F-FDG, either while awake or during the isoflurane-induced incubation phase was applied. Prior to microPET/MRI imaging, one group of the rats was subjected to forane-only anaesthesia while the other group was anaesthetised with the co-administration of forane and dexmedetomidine/Dexdor® Results: While as for the whole brain it was the addition of dexmedetomidine/Dexdor® to the anaesthesia protocol that generated the differences between the radiotracer concentrations of the investigated groups, regarding the cortex, the [18F]F-FDG accumulation was rather affected by the way of incubation. To ensure the most consistent and highest uptake, forane-induced anaesthesia coupled with an awake uptake condition seemed to be most suitable method of anaesthetisation for cerebral metabolic assessment. Diminished whole brain and cortical tracer accumulation detected upon dexmedetomidine/Dexdor® administration highlights the significance of the mechanism of action of different anaesthetics on radiotracer pharmacokinetics. CONCLUSION: Overall, the standardization of PET protocols is of utmost importance to avoid the confounding factors derived from anaesthesia.


Asunto(s)
Anestesia , Anestésicos , Dexmedetomidina , Isoflurano , Ratas , Animales , Fluorodesoxiglucosa F18/metabolismo , Dexmedetomidina/farmacología , Dexmedetomidina/metabolismo , Ratas Wistar , Encéfalo , Tomografía de Emisión de Positrones/métodos , Anestésicos/farmacología , Anestésicos/metabolismo , Isoflurano/farmacología , Isoflurano/metabolismo , Radiofármacos/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda