Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Cereb Blood Flow Metab ; : 271678X241264401, 2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39068534

RESUMEN

Blood-brain barrier (BBB) damage significantly affects the prognosis of ischemic stroke patients. This project employed multi-omics analysis to identify key factors regulating BBB disruption during cerebral ischemia-reperfusion. An integrated analysis of three transcriptome sequencing datasets from mouse middle cerebral artery occlusion/reperfusion (MCAO/R) models identified eight downregulated genes in endothelial cells. Additionally, transcriptome analysis of BBB (cortex) and non-BBB (lung) endothelium of E13.5 mice revealed 2,102 upregulated genes potentially associated with BBB integrity. The eight downregulated genes were intersected with the 2,102 BBB-related genes and mapped using single-cell RNA sequencing data, revealing that solute carrier family 22 member 8 (Slc22a8) is specifically expressed in endothelial cells and pericytes and significantly decreases after MCAO/R. This finding was validated in the mouse MCAO/R model at both protein and mRNA levels in this study. External overexpression of Slc22a8 using a lentivirus carrying Tie2 improved Slc22a8 and tight junction protein levels and reduced BBB leakage after MCAO/R, accompanied by Wnt/ß-catenin signaling activation. In conclusion, this study suggested that MCAO/R-induced downregulation of Slc22a8 expression may be a crucial mechanism underlying BBB disruption. Interventions that promote Slc22a8 expression or enhance its function hold promise for improving the prognosis of patients with cerebral ischemia.

3.
CNS Neurosci Ther ; 30(3): e14679, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38528842

RESUMEN

AIMS: Intracerebral hemorrhage (ICH) is a disease with high rates of disability and mortality. The role of epidermal growth factor receptor 1 (ERBB1) in ICH was elucidated in this study. METHODS: ICH model was constructed by injecting autologous arterial blood into the right basal ganglia. The protein level of ERBB1 was detected by western blot analysis. To up- and downregulation of ERBB1 in rats, intraventricular injection of a lentivirus overexpression vector of ERBB1 and AG1478 (a specific inhibitor of ERBB1) was used. The cell apoptosis, neuronal loss, and pro-inflammatory cytokines were assessed by TUNEL, Nissl staining, and ELISA. Meanwhile, behavioral cognitive impairment of ICH rats was evaluated after ERBB1-targeted interventions. RESULTS: ERBB1 increased significantly in brain tissue of ICH rats. Overexpression of ERBB1 remarkably reduced cell apoptosis and neuronal loss induced by ICH, as well as pro-inflammatory cytokines and oxidative stress. Meanwhile, the behavioral and cognitive impairment of ICH rats were alleviated after upregulation of ERBB1; however, the secondary brain injury (SBI) was aggravated by AG1478 treatment. Furthermore, the upregulation of PLC-γ and PKC in ICH rats was reversed by AG1478 treatment. CONCLUSIONS: ERBB1 can improve SBI and has a neuroprotective effect in experimental ICH rats via PLC-γ/PKC pathway.


Asunto(s)
Lesiones Encefálicas , Hemorragia Cerebral , Receptores ErbB , Quinazolinas , Animales , Ratas , Apoptosis , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Citocinas/metabolismo , Fosfolipasa C gamma/metabolismo , Ratas Sprague-Dawley , Tirfostinos , Receptores ErbB/metabolismo , Proteína Quinasa C/metabolismo
4.
Neuroscience ; 535: 108-123, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37913857

RESUMEN

Subarachnoid Hemorrhage (SAH) is a cerebrovascular disorder that has been found to have severe consequences, including a high mortality and disability rate. Research has indicated that neuronal death, particularly apoptosis, plays a major role in the neurological impairment that follows SAH. RNA-binding protein Pum2 can interfere with translation or other biological functions by connecting to the UGUAHAUA sequence on RNA. Noncoding RNA activated by DNA damage (Norad) contains some Pum2 recognition sequences, which may bind to Pum2 protein and affect its capacity to attach to target mRNA. The time course expression of Norad and Pum2 after SAH is analyzed by establishing a mouse SAH model. Subsequently, the purpose of this study is to investigate the potential role and mechanism of the Norad-Pum2 axis after SAH using lentivirus overexpression of Pum2 and knockdown of Norad. Analysis of Pum2 and Norad levels reveal that the former is significantly reduce and the latter is significantly increased in the SAH group compared to the sham group. Subsequent overexpression of Pum2 and Norad knockdown is found to reduce SAH-induced oxidative stress, neuronal apoptosis, and ultimately improve behavioral and cognitive changes in SAH mice. Our study indicates that Norad-Pum2 acts as a neuromodulator in SAH, and that by increasing Pum2 and decreasing Norad levels, SAH-induced neuronal apoptosis can be reduced and neurological deficits alleviated. Consequently, Norad-Pum2 may be a promising therapeutic target for SAH.


Asunto(s)
Hemorragia Subaracnoidea , Ratones , Animales , Hemorragia Subaracnoidea/metabolismo , Neuroprotección , Modelos Animales de Enfermedad , Apoptosis/fisiología , ARN no Traducido , Proteínas de Unión al ARN/genética
5.
Exp Neurol ; 368: 114508, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37598879

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is one of the stroke subtypes with the highest mortality. Secondary brain injury is associated with neurological dysfunction and poor prognosis after ICH. Caveolin-1 (CAV1) is the key protein of Caveolae. Previous studies have shown that CAV1 plays an important role in central nervous system diseases, and pointed out that in a collagenase-induced ICH model in vivo, CAV1 is associated with neuroinflammatory activation and poor neurological prognosis. In this study, we explore the role and the molecular mechanism of CAV1 in brain injury via a rat autologous whole blood injection model and an in vitro model of ICH. METHODS: Adult male Sprague-Dawley rats ICH model was induced through autologous whole blood injecting into the right basal ganglia. The changes in protein levels of CAV1 in brain tissues of ICH rats were detected by western blot analysis. The immunofluorescent staining was used to explore the changes of CAV1 in microglia/macrophages (Iba1+ cells). Lentivirus vectors were administered by intracerebroventricular injection to induce CAV1 overexpression and knockdown respectively. The western blot analysis, immunofluorescence staining, enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining were performed to explore the role of CAV1 in secondary brain injury after ICH. Meanwhile, the rotarod test, foot fault test, adhesive-removal test, and Modified Garcia Test, as well as Morris Water Maze test, were performed to evaluate the behavioral cognitive impairment of ICH rats after genetic intervention. Additionally, BV-2 cells treated with oxygen hemoglobin for 24 h, were used as an in vitro model of ICH in this study to explore the molecular mechanism of CAV1 in brain injury; we performed western blot analysis after precise regulation of CAV1 in BV2 cells to observe changes in protein levels and phosphorylated levels of C-Src, IKK-ß, and NF-κB. RESULTS: The expression of CAV1 in microglia/macrophages (Iba1+ cells) was elevated and reached the peak at 24 h after ICH. CAV1 knockdown ameliorated ICH-induced neurological deficits, while CAV1 overexpression significantly worsened neurological dysfunction of ICH rats. CAV1 knockdown attenuated cellular apoptosis and promoted neuronal survival in brain tissues of ICH rats, while the ICH rats with CAV1 overexpression presented more cellular apoptosis and neuronal loss. Meanwhile, CAV1 knockdown inhibited the microglia activation and neuroinflammatory response, while CAV1 overexpression abolished these effects and aggravated neuroinflammation in brain tissues of ICH rats. Additionally, by inducing to CAV1 knockdown in BV2 cells in an in vitro model of ICH, the levels of p-C-Src, CAV-1, p-CAV-1, and p-IKK-ß in cytoplasm and the level of NF-κB p65 in nucleus of BV2 cells were significantly decreased, while they were increased by inducing to CAV1 overexpression. CONCLUSIONS: Our research revealed CAV1 aggravated neurological dysfunction in a rat ICH model. CAV1 knockdown exerted neuroprotective effect by suppressing microglia activation and neuroinflammation after ICH might via the C-Src/CAV1/IKK-ß/NF-κB signaling pathway.


Asunto(s)
Lesiones Encefálicas , Neoplasias Encefálicas , Animales , Masculino , Ratas , Caveolina 1 , Hemorragia Cerebral/complicaciones , Enfermedades Neuroinflamatorias , FN-kappa B , Ratas Sprague-Dawley
6.
Medicine (Baltimore) ; 102(16): e32591, 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37083791

RESUMEN

BACKGROUND: The efficacy of tivantinib for MET-high hepatocellular carcinoma remains controversial. We conduct this meta-analysis to explore the efficacy of tivantinib versus placebo for MET-high hepatocellular carcinoma. METHODS: We have searched PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through November 2022 and included randomized controlled trials (RCTs) assessing the efficacy and safety of tivantinib versus placebo for MET-high hepatocellular carcinoma. RESULTS: Three RCTs were included in the meta-analysis. Overall, compared with control group for MET-high hepatocellular carcinoma, tivantinib showed no obvious impact on overall survival (hazard ratio [HR] = 0.77; 95% confidence interval [CI] = 0.52-1.13; P = .18) or progression-free survival (HR = 0.78; 95% CI = 0.56-1.08; P = .14). In addition, tivantinib was associated with the increase in grade ≥3 neutropenia (odd ratio [OR] = 11.76; 95% CI = 2.77-49.89; P = .0008) and leukopenia (OR = 14; 95% CI = 1.68-116.82; P = .01), but demonstrated no impact on the incidence of grade ≥ 3 anemia (OR = 2.74; 95% CI = 0.14-53.43; P = .51). CONCLUSIONS: Tivantinib may not benefit to the treatment of MET-high hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Quinolinas , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Metaanálisis como Asunto , Pirrolidinonas/uso terapéutico , Quinolinas/uso terapéutico
7.
Front Neurosci ; 14: 245, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265644

RESUMEN

Nix is located in the outer membrane of mitochondria, mediates mitochondrial fission and implicated in many neurological diseases. However, the association between Nix and subarachnoid hemorrhage (SAH) has not previously been reported. Therefore, the present study was designed to evaluate the expression of Nix and its role in early brain injury (EBI) after SAH. Adult male Sprague-Dawley (SD) rats were randomly assigned to various time points for investigation after SAH. A rat model of SAH was induced by injecting 0.3 ml of autologous non-heparinized arterial blood into the prechiasmatic cistern. The expression of Nix was investigated by Western blot and immunohistochemistry. Next, Nix-specific overexpression plasmids and small interfering RNAs (siRNAs) were separately administered. Western blot, neurological scoring, Morris water maze, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining and fluoro-jade B (FJB) staining were performed to evaluate the role of Nix in EBI following SAH. We found that Nix was expressed in neurons and its expression level in the SAH groups was higher than that in the Sham group, which peaked at 24 h after SAH. Overexpression of Nix following SAH significantly decreased the expression of translocase of outer mitochondrial membrane 20 (TOMM20, a marker of mitochondria), ameliorated neurological/cognitive deficits induced by SAH, and reduced the total number of apoptotic/neurodegenerative cells, whereas siRNA knockdown of Nix yielded opposite effects. Taken together, our findings demonstrated that the expression of Nix is increased in neurons after experimental SAH in rats, and may play a neuroprotective role in EBI following SAH.

8.
Acta Neurochir Suppl ; 127: 105-119, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31407071

RESUMEN

The protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathway, which is a branch of the unfolded protein response, participates in a range of pathophysiological processes of neurological diseases. However, few studies have investigated the role of the PERK in intracerebral hemorrhage (ICH). The present study evaluated the role of the PERK pathway during the early phase of ICH-induced secondary brain injury (SBI) and its potential mechanisms. An autologous whole blood ICH model was established in rats, and cultured primary cortical neurons were treated with oxyhemoglobin to mimic ICH in vitro. We found that levels of phosphorylated alpha subunit of eukaryotic translation initiation factor 2 (p-eIF2α) and activating transcription factor 4 (ATF4) increased significantly and peaked at 12 h during the early phase of the ICH. To further elucidate the role of the PERK pathway, we assessed the effects of the PERK inhibitor, GSK2606414, and the eIF2α dephosphorylation antagonist, salubrinal, at 12 h after ICH both in vivo and in vitro. Inhibition of PERK with GSK2606414 suppressed the protein levels of p-eIF2α and ATF4, resulting in increase of transcriptional activator CCAAT/enhancer-binding protein homologous protein (CHOP) and caspase-12, which promoted apoptosis and reduced neuronal survival. Treatment with salubrinal yielded opposite results, which suggested that activation of the PERK pathway could promote neuronal survival and reduce apoptosis. In conclusion, the present study has demonstrated the neuroprotective effects of the PERK pathway during the early phase of ICH-induced SBI. These findings highlight the potential value of PERK pathway as a therapeutic target for ICH.


Asunto(s)
Lesiones Encefálicas , Hemorragia Cerebral , ARN , eIF-2 Quinasa , Animales , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Factor 2 Eucariótico de Iniciación , Ratas , eIF-2 Quinasa/metabolismo
9.
Neurobiol Dis ; 129: 144-158, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31082470

RESUMEN

Necroptosis is a regulated form of necrosis that is mediated by a variety of proteins including tumor necrosis factor-α (TNF-α) and receptor-interacting proteins (RIPs). TNF-α, a critical inflammatory molecule, is one of the initiating signals in the necroptosis pathway, and RIP3 acts as a switch that commits the cell to necroptosis. Subarachnoid hemorrhage (SAH) is a common type of hemorrhagic stroke with high mortality and disability rates. RIP3 has been studied in many central nervous system (CNS) diseases, but its role in SAH has not been investigated in depth. Here, we used an autologous-blood injection model to study the role of RIP3 in brain injury induced by SAH in rats. Several indexes such as brain edema, loss of blood-brain barrier (BBB) integrity, and behavioral tests of neurological function were used to evaluate brain damage in SAH-injured rats. We found that the expression of RIP3 was increased in the rat brain after SAH, reaching the highest point 24 h post-injury. We also showed that genetic or pharmacological inhibition of RIP3 or TNF-α reduced the brain damage induced by SAH, whereas overexpression of RIP3 aggravated brain injury and neurological damage. Additionally, we verified the presence of RIP3-mediated necroptosis in an in vitro SAH model of primary cultured neurons treated with conditioned medium from primary microglia activated by oxygen hemoglobin (OxyHb). Collectively, our findings indicated that RIP3 contributed to brain damage after SAH by inducing necroptosis.


Asunto(s)
Necroptosis/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/patología
10.
Transl Stroke Res ; 10(6): 729-743, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30941717

RESUMEN

Intracerebral hemorrhage (ICH) is a serious public health problem which causes high rates of disability and mortality in adults. Cell apoptosis is a sign of secondary brain injury (SBI) following ICH. Mammalian sterile 20-like kinase-1 (MST1), an apoptosis-promoting kinase, is a part of the Hippo signaling pathway and involved in cell death, oxidative stress, and inflammation. However, the role and underlying mechanism of MST1 in SBI induced by ICH have not yet been fully explained. The main purpose of present research was to explore the role of MST1 and its potential mechanism in SBI after ICH. An ICH model was established by injecting autologous blood into the right basal ganglia in male SD rats. We found that MST1 phosphorylation was significantly increased in brain tissues of rats after ICH. Additionally, inhibition of MST1 phosphorylation by a chemical inhibitor (Xmu-mp-1) and genetic knockdown could effectively reduce the activation of P-LATS1 and P-YAP which are downstream proteins of MST1 and decrease neuronal cell death and inflammation reaction in ICH rats. Furthermore, the decreased of MST1 phosphorylation reduced brain edema, blood-brain barrier (BBB) damage, and neurobehavioral impairment during ICH. Over-expression of MST1 resulted in opposite effects. Finally, deletion of MST1 significantly reduced neuronal apoptosis in vitro. In summary, our study revealed that MST1 played an important role in the SBI following ICH, and inhibition of MST1 could alleviate ICH-induced SBI. Therefore, MST1 may be considered as a potential therapeutic target for SBI following ICH.


Asunto(s)
Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apoptosis , Barrera Hematoencefálica/metabolismo , Edema Encefálico/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Masculino , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
11.
Cell Transplant ; 28(8): 985-1001, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30838887

RESUMEN

Sodium/hydrogen exchanger 1 (NHE1) plays an essential role in maintaining intracellular pH (pHi) homeostasis in the central nervous system (CNS) under physiological conditions, and it is also associated with neuronal death and intracellular Na+ and Ca2+ overload induced by cerebral ischemia. However, its roles and underlying mechanisms in early brain injury (EBI) induced by subarachnoid hemorrhage (SAH) have not been fully explored. In this research, a SAH model in adult male rat was established through injecting autologous arterial blood into prechiasmatic cistern. Meanwhile, primary cultured cortical neurons of rat treated with 5 µM oxygen hemoglobin (OxyHb) for 24 h were applied to mimic SAH in vitro. We find that the protein levels of NHE1 are significantly increased in brain tissues of rats after SAH. Downregulation of NHE1 by HOE642 (a specific chemical inhibitor of NHE1) and genetic-knockdown can effectively alleviate behavioral and cognitive dysfunction, brain edema, blood-brain barrier (BBB) injury, inflammatory reactions, oxidative stress, neurondegeneration, and neuronal apoptosis, all of which are involved in EBI following SAH. However, upregulation of NHE1 by genetic-overexpression can produce opposite effects. Additionally, inhibiting NHE1 significantly attenuates OxyHb-induced neuronal apoptosis in vitro and reduces interaction of NHE1 and CHP1 both in vivo and in vitro. Collectively, we can conclude that NHE1 participates in EBI induced by SAH through mediating inflammation, oxidative stress, behavioral and cognitive dysfunction, BBB injury, brain edema, and promoting neuronal degeneration and apoptosis.


Asunto(s)
Apoptosis , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Neuronas/patología , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Hemorragia Subaracnoidea/complicaciones , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Lipoproteínas/metabolismo , Masculino , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/patología , Neuronas/metabolismo , Oxihemoglobinas/metabolismo , Ratas Sprague-Dawley
12.
CNS Neurosci Ther ; 25(6): 674-684, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30677253

RESUMEN

AIMS: To investigate the critical role of Tim-3 in the polarization of microglia in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI). METHODS: An in vivo ICH model was established by autologous whole blood injection into the right basal ganglia in rats. The primary cultured microglia were treated with oxygen-hemoglobin (OxyHb) to mimic ICH in vitro. In this experiment, specific siRNA for Tim-3 and recombinant human TIM-3 were exploited both in vivo and in vitro. RESULTS: Tim-3 was increased in the brain after ICH, which mainly distributed in microglia, but not neurons and astrocytes. However, the blockade of Tim-3 by siRNA markedly reduced secretion of inflammatory factors, neuronal degeneration, neuronal cell death, and brain edema. Meanwhile, downregulation of Tim-3 promoted the transformation of microglia phenotype from M1 to M2 after ICH. Furthermore, upregulation of Tim-3 can increase the interaction between Tim-3 and Galectin-9 (Gal-9) and activate Toll-like receptor 4 (TLR-4) pathway after ICH. Increasing the expression of Tim-3 may be related to the activation of HIF-1α. CONCLUSION: Tim-3 may be an important link between neuroinflammation and microglia polarization through Tim-3/Gal-9 and TLR-4 signaling pathways which induced SBI after ICH.


Asunto(s)
Hemorragia Cerebral/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Edema Encefálico/metabolismo , Muerte Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Galectinas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuronas/metabolismo , Ratas Sprague-Dawley , Receptores de Superficie Celular/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 4/metabolismo
13.
Surg Laparosc Endosc Percutan Tech ; 29(1): 1-6, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30531447

RESUMEN

INTRODUCTION: The efficacy of melatonin to improve sleep quality after laparoscopic cholecystectomy remains controversial. We conduct a systematic review and meta-analysis to explore the influence of melatonin intervention versus placebo on sleep quality after laparoscopic cholecystectomy. METHODS: We searched PubMed, EMbase, Web of Science, EBSCO, and Cochrane library databases through July 2018 for randomized controlled trials assessing the effect of melatonin intervention versus placebo on sleep quality after laparoscopic cholecystectomy. This meta-analysis is performed using the random-effect model. RESULTS: Five randomized controlled trials involving 250 patients are included in the meta-analysis. Overall, compared with control group for laparoscopic cholecystectomy, melatonin intervention shows no substantial impact on well-being [standard mean difference (std MD)=0.05; 95% confidence interval (CI)=-0.26 to 0.36; P=0.76], sleepiness (std MD=-0.10; 95% CI=-0.44 to 0.23; P=0.54), sleep quality (std MD=0.10; 95% CI=-0.21 to 0.41; P=0.53), pain scores after 1 hour (std MD=-0.26; 95% CI=-1.08 to 0.56; P=0.53) and 3 hours (std MD=-0.86; 95% CI=-2.69 to 0.97; P=0.36), headache [risk ratio (RR)=1.25; 95% CI=0.42-3.71; P=0.68], depression (RR=1.03; 95% CI=0.15-7.21; P=0.97), dizziness (RR=1.09; 95% CI=0.14-9.40; P=0.94). CONCLUSIONS: Melatonin intervention has no significant influence on well-being, sleepiness, sleep quality, pain intensity after 1 and 3 hours, headache, depression, and dizziness for laparoscopic cholecystectomy.


Asunto(s)
Depresores del Sistema Nervioso Central/uso terapéutico , Colecistectomía Laparoscópica/efectos adversos , Melatonina/uso terapéutico , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Depresión/etiología , Mareo/etiología , Trastornos de Cefalalgia/etiología , Humanos , Dolor Postoperatorio/etiología , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/etiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Trastornos del Sueño-Vigilia/etiología
14.
Medicine (Baltimore) ; 97(45): e12747, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30407279

RESUMEN

BACKGROUND: The impact of magnesium sulfate on hemodynamic responses during laparoscopic cholecystectomy remains controversial. We conduct a systematic review and meta-analysis to explore the influence of magnesium sulfate on hemodynamic responses for laparoscopic cholecystectomy. METHODS: We search PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through June 2018 for randomized controlled trials (RCTs) assessing the effect of magnesium sulfate on hemodynamic responses for laparoscopic cholecystectomy. Meta-analysis is performed using the random-effect model. RESULTS: Four RCTs involving 208 patients are included in the meta-analysis. Overall, compared with control group in laparoscopic cholecystectomy, intravenous magnesium sulfate is associated with systolic blood pressure at 30 minutes [Std. MD = -1.34; 95% confidence interval (95% CI) = -1.86 to -0.82; P < .00001], diastolic blood pressure at 30 minutes (Std. MD = -1.40; 95% CI = -1.86 to -0.94; P < .00001), mean arterial pressure at 30 minutes (Std. MD = -1.19; 95% CI = -1.91 to -0.46; P = .001), systolic blood pressure at 10 minutes (Std. MD = -1.61; 95% CI = -2.08 to -1.13; P < .00001), diastolic blood pressure at 10 minutes (Std. MD = -1.54; 95% CI = -2.68 to -0.40; P = .008), heart rate at 30 minutes (Std. MD = -2.09; 95% CI = -2.87 to -1.32; P < .00001), but results in prolonged extubation time (Std. MD = 0.96; 95% CI = 0.18-1.74; P = .02). CONCLUSION: Magnesium sulfate can reduce blood pressure, but with the increase in extubation time for laparoscopic cholecystectomy.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Colecistectomía Laparoscópica/métodos , Hemostasis Quirúrgica/métodos , Sulfato de Magnesio/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Vasodilatadores/farmacología , Adulto , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
15.
Brain Res ; 1701: 112-125, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30205109

RESUMEN

Oxidative stress plays an important role in secondary brain injury (SBI) after intracerebral hemorrhage (ICH), but the underling mechanism has not been fully elucidated. Recently, the antioxidant enzyme glutathione peroxidase 4 (GPX4), has attracted increasing attention due to its ability to degrade reactive oxygen species (ROS) which are the major indicator of oxidative stress; However, the role of GPX4 in ICH has not been reported. This study was designed to investigate the changes in protein levels, as well as potential role and mechanism of GPX4 in SBI following ICH using a Sprague-Dawley (SD) rat model of ICH induced by autologous blood injection into the right basal ganglia. Firstly, GPX4 protein levels in the brain were reduced gradually and bottomed out at 24 h after ICH, compared with the Sham group. Secondly, genetic-overexpression of GPX4 effectively increased level of GPX4 in the brain, and clearly relieved neuronal dysfunction, brain edema, blood brain barrier (BBB) injury, oxidative stress and inflammation after ICH. In contrast, inhibiting GPX4 with a specific pharmacological inhibitor or genetic knockdown exacerbated SBI after ICH. Finally, Ferrostatin-1, a chemical inhibitor of ferroptosis, was used to explore the role of ferroptosis in brain injury after ICH. The results suggest that inhibiting ferroptosis can significantly alleviate SBI after ICH. In summary, our work indicated that GPX4 contributes to SBI following ICH by mediating ferroptosis. Therefore, inhibiting ferroptosis with specific inhibitors or upregulation of GPX4 may be a potential strategy to ameliorate brain injury induced by ICH.


Asunto(s)
Lesiones Encefálicas/enzimología , Hemorragia Cerebral/enzimología , Glutatión Peroxidasa/metabolismo , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Muerte Celular/fisiología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclohexilaminas/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fenilendiaminas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
16.
Neurobiol Dis ; 119: 53-64, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30048803

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is the genetic cause of both familial and idiopathic Parkinson's disease (PD), and it is associated with neuronal death, vesicle trafficking, mitochondrial dysfunction, and inflammation. However, its role in secondary brain injury (SBI) induced by intracerebral hemorrhage (ICH) has not been evaluated. In this study, an ICH model was induced by injecting autologous whole blood into the right basal ganglia of adult rats. Meanwhile, primary rat cortical neurons treated with Oxyhemoglobin (OxyHb) were used as an in vitro ICH model. Protein levels of LRRK2 increased significantly in brain tissues after ICH. Upregulation of LRRK2 by genetic overexpression augmented inflammatory responses, behavioral and cognitive dysfunction, brain edema, blood-brain barrier (BBB) injury, and cell death involved in SBI following ICH. Downregulation of LRRK2 by GNE7915 (a specific chemical inhibitor of LRRK2) and genetic knockdown yielded opposite effects. Additionally, inhibiting LRRK2 by GNE7915 obviously reduced OxyHb-induced neuronal apoptosis in vitro and attenuated phosphorylation of p38 MAPK and Drosha both in vivo and in vitro. Therefore, we concluded that LRRK2 participated in ICH-induced SBI by mediating inflammatory responses, behavioral and cognitive dysfunction, brain edema, and BBB injury and by modulating neuronal death and dysfunction and regulating the p38 MAPK/Drosha pathway.


Asunto(s)
Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/biosíntesis , Ribonucleasa III/biosíntesis , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/patología , Hemorragia Cerebral/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Masculino , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ribonucleasa III/antagonistas & inhibidores , Ribonucleasa III/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
Front Neurosci ; 12: 111, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29541018

RESUMEN

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) signaling pathway was reported to exert an important role in neuronal apoptosis. The present study was designed to investigate the roles of the PERK signaling pathway in the secondary brain injury (SBI) induced by intracerebral hemorrhage (ICH) and its potential mechanisms. Sprague-Dawley rats were used to establish ICH models by injecting autologous blood (100 µl), and cultured primary rat cortical neurons were exposed to oxyhemoglobin (10 µM) to mimic ICH in vitro. The PERK antagonist, GSK2606414, and inhibitor of eukaryotic translation initiation factor 2 subunit α (eIF2α) dephosphorylation, salubrinal, were used to study the roles of PERK signaling pathway in ICH-induced SBI. Our results showed that the protein levels of p-eIF2α and ATF4 were upregulated following ICH, peaking at 48 h. Application of GSK2606414 reversed this increase in vivo and in vitro, thereby preventing ICH-induced neuronal apoptosis. On the contrary, salubrinal inhibited the dephosphorylation of eIF2α, resulting in the elevation of p-eIF2α, which could activate downstream of PERK signaling and induce neuronal apoptosis and necrosis following ICH in vitro and in vivo. Thus, PERK signaling pathway plays an important role in ICH-induced apoptosis and blocking its activation has neuroprotective effects that alleviates SBI, suggesting that targeting this pathway could be a promising therapeutic strategy for improving patient outcome after ICH.

18.
Med Gas Res ; 7(2): 113-119, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744364

RESUMEN

Hydrogen sulfide (H2S) has been recognized and studied for nearly 300 years, but past researches mainly focus on its toxicity effect. During the past two decades, the majority of researches have reported that H2S is a novel endogenous gaseous signal molecule in organisms, and play an important role in various systems and diseases. H2S is mainly produced by three enzymes, including cystathionine ß-synthase, cystathionine γ-lyase and 3-mercaptopyruvate sulfurtransferase along with cysteine aminotransferase. H2S had been firstly reported as a neuromodulator in the brain, because of its essential role in the facilitating hippocampal long-term potentiation at physiological concentration. It is subsequently reported that H2S may have relevance to neurologic disorders through antioxidative, anti-inflammatory, anti-apoptotic and additional effects. Recent basic medical studies and preclinical studies on neurologic diseases have demonstrated that the administration of H2S at physiological or pharmacological levels attenuates brain injury. However, the neuroprotective effect of H2S is concentration-dependent, only a comparatively low dose of H2S can provide beneficial effect. Herein, we review the neuroprotevtive role of H2S therapy in brain diseases from its mechanism to clinical application in animal and human subjects, and therefore provide the potential strategies for further clinical treatment.

19.
Med Gas Res ; 7(2): 124-132, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744366

RESUMEN

Helium has been classified as a kind of inert gas that is not effortless to spark chemical reactions with other substances in the past decades. Nevertheless, the cognition of scientists has gradually changed accompanied with a variety of studies revealing the potential molecular mechanism underlying organ-protection induced by helium. Especially, as a non-anesthetic gas which is deficient of relevant cardiopulmonary side effects, helium conditioning is recognized as an emerging and promising approach to exert favorable effects by mimicking the cardioprotection of anesthetic gases or xenon. In this review we will summarize advances in the underlying biological mechanisms and clinical applicability with regards to the cardioprotective effects of helium.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA