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











Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 10(4): e25916, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38390161

RESUMO

Objective: This study aimed to explore the impact of late night shift work on the functional outcomes of patients with acute ischemic stroke (AIS) treated with endovascular thrombectomy (EVT). Methods: Consecutive AIS patients who underwent EVT between June 2019 and June 2021 were enrolled and divided into non-night shift work and night shift work groups based on their occupational histories. The primary outcome was the modified Rankin Scale score defined 3-month functional outcome. The secondary outcomes were 3-month mortality, symptomatic intracerebral hemorrhage (sICH), ICH and early recanalization. Results: A total of 285 patients were enrolled, 35 patients (12.3%) were night shift workers, who were younger (P < 0.001) and had a significantly higher prevalence of smoking (P < 0.001), hyperlipidemia (P = 0.002), coronary heart disease (P = 0.031), and atrial fibrillation (P < 0.001). The 3-month favorable outcomes were achieved in 44.8% and 25.7% of patients in the non-night shift work and night shift work groups, respectively (adjusted odds ratio [OR]: 0.24, 95% CI: 0.10-0.57; adjusted P = 0.001). No difference was found in 3-month mortality (adjusted OR: 0.43; 95% CI: 0.14-1.25, adjusted P = 0.121), rates of ICH (adjusted OR: 0.73; 95% CI: 0.33-1.60; adjusted P = 0.430), sICH (adjusted OR: 0.75; 95% CI: 0.34-1.67; adjusted P = 0.487), or early successful recanalization (adjusted OR: 0.42; 95% CI: 0.12-1.56; adjusted P = 0.197). These results were consistent after PSM analysis. Conclusion: Our findings suggest that late night shift work is significantly associated with unfavorable outcomes in patients with AIS after EVT.

2.
Nat Protoc ; 17(9): 2054-2084, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35760857

RESUMO

Over decades of research into the treatment of stroke, nearly all attempts to translate experimental treatments from discovery in cells and rodents to use in humans have failed. The prevailing belief is that it might be necessary to pretest pharmacological neuroprotection in higher-order brains, especially those of nonhuman primates (NHPs). Over the past few years, chemical thrombolysis and mechanical thrombectomy have been established as the standard of care for ischemic stroke in patients. The spotlight is now shifting towards emphasizing both focal ischemia and subsequent reperfusion in developing a clinically relevant stroke model in NHPs. This protocol describes an embolic model of middle cerebral artery occlusion in adult rhesus monkeys. An autologous clot is combined with a microcatheter or microwire through endovascular procedures, and reperfusion is achieved through local intra-artery thrombolysis with tissue plasminogen activator. These NHP models formed relatively stable infarct sizes, delivered predictable reperfusion and survival outcomes, and recapitulated key characteristics of patients with ischemic stroke as observed on MRI images and behavioral assays. Importantly, treated animals could survive 30 d after the surgery for post-stroke neurologic deficit analyses. Thus far, this model has been used in several translational studies. Here we describe in detail the teamwork necessary for developing stroke models of NHPs, including the preoperation preparations, endovascular surgery, postoperation management and histopathological analysis. The model can be established by the following procedures over a 45-d period, including preparation steps (14 d), endovascular operation (1 d) and evaluation steps (30 d).


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Trombose , Animais , Isquemia Encefálica/tratamento farmacológico , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Macaca mulatta , Acidente Vascular Cerebral/terapia , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/uso terapêutico
3.
CNS Neurosci Ther ; 28(9): 1303-1314, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35702957

RESUMO

INTRODUCTION: The neuroprotection of acute ischemic stroke patients can be achieved by intra-arterial selective cooling infusion using cold saline, which can decrease brain temperature without influencing the body core temperature. This approach can lead to high burdens on the heart and decreased hematocrit in the scenario of loading a high amount of liquid for longtime usage. Therefore, autologous blood is utilized as perfusate to circumvent those side effects. METHODS: In this study, a prototype instrument with an autologous blood cooling system was developed and further evaluated by a mathematical model for brain temperature estimation. RESULTS: Hypothermia could be achieved due to the adequate cooling capacity of the prototype system, which could provide the lowest cooling temperature into the blood vessel of 10.5°C at 25 rpm (209.7 ± 0.8 ml/min). And, the core body temperature did not alter significantly (-0.7 ~ -0.2°C) after 1-h perfusion. The cooling rate and temperature distributions of the brain were analyzed, which showed a 2°C decrease within the initial 5 min infusion by 44 ml/min and 13.7°C perfusate. CONCLUSION: This prototype instrument system could safely cool simulated blood in vitro and reperfuse it to the target cerebral blood vessel. This technique could promote the clinical application of an autologous blood perfusion system for stroke therapy.


Assuntos
Hipotermia Induzida , AVC Isquêmico , Acidente Vascular Cerebral , Temperatura Corporal , Encéfalo , Temperatura Baixa , Humanos , Hipotermia Induzida/métodos , Acidente Vascular Cerebral/terapia , Temperatura
4.
Comput Biol Med ; 145: 105497, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35398811

RESUMO

Intra-arterial selective cooling infusion with the autologous blood (IA-SCAI) is a promising therapeutic hypothermia induction method for conferring neuroprotection to acute ischemic stroke (AIS) patients. The blood heat exchanger (BHE) plays a crucial role in IA-SCAI's cooling capacity. However, there are no BHEs currently available that are specifically designed for the IA-SCAI, which requires a low blood flow to be compatible with cerebral hemodynamics. In an effort to develop a BHE for AIS patients, a prototype of a commercial BHE, Medtronic MYOtherm XP®, was mathematically modeled; specifically, computational fluid dynamics (CFD) was used to analyze its hemo- and thermo-dynamic characteristics under low blood flow including temperature distribution, velocity field and shear stress. Our numerical model predicted the hemolysis index to be 0.0041%-0.0581% inside the BHE with blood flows rates of 10 ml min-1-50 ml min-1. The in vitro heat transfer experiment showed that the BHE efficiently cooled the simulated blood from the initial 37 °C-5.8 °C within 150 s by using cold water (200 ml·min-1, 0 °C). The cooled simulated blood was able to cool the simulated blood in the middle cerebral artery of an artificial circulating system from 37 °C to 16.8 °C-33.7 °C depending on the blood perfusion rate (10-50 ml/min). A biological heat transfer mathematical model showed that brain tissue could be cooled by 2 °C within the initial 1min of infusion. This study verified the feasibility of using a commercial BHE for IA-SCAI and provided insights into its cooling capacity for therapeutic hypothermia.


Assuntos
Hipotermia Induzida , AVC Isquêmico , Temperatura Corporal , Encéfalo/fisiologia , Temperatura Alta , Humanos , Hipotermia Induzida/métodos , Temperatura , Termodinâmica
5.
Neurotherapeutics ; 19(1): 386-398, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35044645

RESUMO

The translational failure of neuroprotective therapies in stroke may be influenced by the mismatch of existing comorbidities between animal models and patients. Previous studies found that single-target neuroprotective agents reduced infarction in Sprague-Dawley but not in spontaneously hypertensive rats. It is of great interest to explore whether multi-target neuroprotectants and stroke models with comorbidities should be used in further translational researches. Ischemic stroke was induced in normotensive or hypertensive rats by 90- or 120-min middle cerebral artery occlusion (MCAO) and reperfusion. Intra-Arterial Selective Cooling Infusion (IA-SCI) was started at the onset of reperfusion for 30 minutes. Acute neurological deficits, infarct volumes, gene expression and markers of A1-like and A2-like astrocytes were evaluated. In further analysis, TNFα and IL-1α were administrated intracerebroventricularly, phenotype shifting of astrocytes and infarct volumes were assessed. Normobaric oxygen treatment, as a negative control, was also assessed in hypertensive rats. IA-SCI led to similar benefits in normotensive rats with 120-min MCAO and hypertensive rats with both 90-min and 120-min MCAO, including mitigated functional deficit and reduced infarct volumes. IA-SCI shifted astrocyte phenotypes partly by downregulating A1-like astrocytes and upregulating A2-like astrocytes in both RNA and protein levels. Upregulated A1-type astrocyte markers levels, induced by intracerebroventricular injection of TNFα and IL-1α, were closely related to increased infarct volumes in hypertensive rats, despite receiving IA-SCI treatment. In addition, infarct volumes and A1/A2-like genes were not affected by normobaric oxygen treatment. IA-SCI reduced infarction in both normotensive and hypertensive rats. Our results demonstrated the neuroprotective effects of IA-SCI in hypertensive rats may be related with phenotype shifting of astrocytes.


Assuntos
Hipertensão , AVC Isquêmico , Fármacos Neuroprotetores , Acidente Vascular Cerebral , Animais , Astrócitos/metabolismo , Modelos Animais de Doenças , Humanos , Hipertensão/complicações , Hipertensão/terapia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/terapia , Fármacos Neuroprotetores/farmacologia , Oxigênio/metabolismo , Oxigênio/farmacologia , Fenótipo , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/terapia , Fator de Necrose Tumoral alfa/metabolismo
6.
Med Eng Phys ; 99: 103736, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35058029

RESUMO

Intra-arterial selective cooling infusion (IA-SCI) is a promising method for neuroprotection of patients with acute ischemic stroke. One shortcoming of IA-SCI is the elevated delivery temperature caused by the cold perfusate warming along the catheter pathway. Therefore, increasing the thermal resistance of the catheter is of significant importance. In this manuscript, an air-insulated catheter was designed and manufactured through extrusion molding technique. The computational fluid dynamics (CFD)-based thermo-/hemo-dynamics models were exploited to evaluate the thermal conductivity of the catheter. Compared with commercially available endovascular catheters, its thermal insulation property was analyzed through an in vitro experiment. The temperature of the 4°C perfusate (20 ml/min) increased to 14.2°C ± 0.2°C after being transferred to the distal tip of the air-insulated catheter, which was significantly lower than that (30°C) of commercially available alternatives. Moreover, the simulated blood (56% glycerin and 44% bi-distilled water, 37°C) in the middle cerebral artery of the artificial circulating system was cooled down to 29.7°C ± 0.1°C by this perfusate. The cooling process of the brain tissue was also estimated by a biological heat-transfer mathematical model, which showed a 2°C decrease within the initial 1 min infusion. This study demonstrated that the air-insulated catheter for IA-SCI was promising in vitro in terms of its high cooling efficiency and could be a competitive intervention catheter for therapeutic hypothermia.


Assuntos
Isquemia Encefálica , Hipotermia Induzida , AVC Isquêmico , Encéfalo/irrigação sanguínea , Catéteres , Humanos
7.
Brain Res ; 1724: 146406, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31454517

RESUMO

OBJECTIVE: The present study aimed to determine if hypothermia augments the neuroprotection conferred by MSC administration by providing a conducive micro-environment. METHODS: Sprague-Dawley rats were subjected to 1.5 h middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion for molecular analyses, as well as 1, 14 and 28 days for brain infarction or functional outcomes. Rats were treated with either MSC (1 × 105), LCI (cold saline, 0.6 ml/min, 5 min) or both. Brain damage was determined by Infarct volume and neurological deficits. Long-term functional outcomes were evaluated using foot-fault and Rota-rod testing. Human neural SHSY5Y cells were investigated in vitro using 2 h oxygen-glucose deprivation (OGD) followed by MSC with or without hypothermia (HT) (34 °C, 4 h). Mitochondrial transfer was assessed by confocal microscope, and cell damage was determined by cell viability, ATP, and ROS level. Protein levels of IL-1ß, BAX, Bcl-2, VEGF and Miro1 were measured by Western blot following 6 h and 24 h of reperfusion and reoxygenation. RESULTS: MSC, LCI, and LCI + MSC significantly reduced infarct volume and deficit scores. Combination therapy of LCI + MSC precipitated better long-term functional outcomes than monotherapy. Upregulation of Miro1 in the combination group increased mitochondrial transfer and lead to a greater increase in neuronal cell viability and ATP, as well as a decrease in ROS. Further, combination therapy significantly decreased expression of IL-1ß and BAX while increasing Bcl-2 and VEGF expression. CONCLUSION: Therapeutic hypothermia upregulated Miro1 and enhanced MSC mitochondrial transfer-mediated neuroprotection in ischemic stroke. Combination of LCI with MSC therapy may facilitate clinical translation of this approach.


Assuntos
Isquemia Encefálica/metabolismo , Hipotermia Induzida/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Lesões Encefálicas/metabolismo , Isquemia Encefálica/terapia , Modelos Animais de Doenças , Hipotermia/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Isquemia/metabolismo , Isquemia/terapia , Masculino , Células-Tronco Mesenquimais/metabolismo , Neurônios/metabolismo , Neuroproteção/fisiologia , Ratos , Ratos Sprague-Dawley , Reperfusão , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/metabolismo
9.
Sci Rep ; 9(1): 10432, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31320706

RESUMO

Retrospective studies have found that left upper lobectomy (LUL) may be a new risk factor for stroke, and the potential mechanism is pulmonary vein thrombosis, which more likely develops in the left superior pulmonary vein (LSPV) stump. The LSPV remaining after left pneumonectomy is similar to that remaining after LUL. However, the association between left pneumonectomy, LUL, and postoperative stroke remains unclear. Thus, we sought to analyze whether both LUL and left pneumonectomy are risk factors for postoperative stroke. We prospectively included consecutive patients who underwent resection between November 2016 and March 2018 at our institution with 6 months of follow-up. Baseline demographic and clinical data were taken. A logistic regression model was used to determine independent predictors of postoperative stroke. In our study, 756 patients who underwent an isolated pulmonary lobectomy procedure were screened; of these, 637 patients who completed the 6-month follow-up were included in the analysis. Multivariable logistic regression analysis adjusted for common risk factors showed that the LUL and left pneumonectomy were independent predictors of stroke (odds ratio, 18.12; 95% confidence interval, 2.12-155.24; P = 0.008). Moreover, diabetes mellitus also was a predictor of postoperative stroke. In conclusion, both LUL and left pneumonectomy are significant risk factors for postoperative stroke.


Assuntos
Pulmão/cirurgia , Pneumonectomia/efeitos adversos , Complicações Pós-Operatórias/etiologia , Acidente Vascular Cerebral/etiologia , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Veias Pulmonares/cirurgia , Fatores de Risco
10.
Neural Regen Res ; 14(10): 1780-1786, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31169196

RESUMO

Combinations of biomaterials and cells can effectively target delivery of cells or other therapeutic factors to the brain to rebuild damaged nerve pathways after brain injury. Porous collagen-chitosan scaffolds were prepared by a freeze-drying method based on brain tissue engineering. The scaffolds were impregnated with rat bone marrow mesenchymal stem cells. A traumatic brain injury rat model was established using the 300 g weight free fall impact method. Bone marrow mesenchymal stem cells/collagen-chitosan scaffolds were implanted into the injured brain. Modified neurological severity scores were used to assess the recovery of neurological function. The Morris water maze was employed to determine spatial learning and memory abilities. Hematoxylin-eosin staining was performed to measure pathological changes in brain tissue. Immunohistochemistry was performed for vascular endothelial growth factor and for 5-bromo-2-deoxyuridine (BrdU)/neuron specific enolase and BrdU/glial fibrillary acidic protein. Our results demonstrated that the transplantation of bone marrow mesenchymal stem cells and collagen-chitosan scaffolds to traumatic brain injury rats remarkably reduced modified neurological severity scores, shortened the average latency of the Morris water maze, increased the number of platform crossings, diminished the degeneration of damaged brain tissue, and increased the positive reaction of vascular endothelial growth factor in the transplantation and surrounding areas. At 14 days after transplantation, increased BrdU/glial fibrillary acidic protein expression and decreased BrdU/neuron specific enolase expression were observed in bone marrow mesenchymal stem cells in the injured area. The therapeutic effect of bone marrow mesenchymal stem cells and collagen-chitosan scaffolds was superior to stereotactic injection of bone marrow mesenchymal stem cells alone. To test the biocompatibility and immunogenicity of bone marrow mesenchymal stem cells and collagen-chitosan scaffolds, immunosuppressive cyclosporine was intravenously injected 12 hours before transplantation and 1-5 days after transplantation. The above indicators were similar to those of rats treated with bone marrow mesenchymal stem cells and collagen-chitosan scaffolds only. These findings indicate that transplantation of bone marrow mesenchymal stem cells in a collagen-chitosan scaffold can promote the recovery of neuropathological injury in rats with traumatic brain injury. This approach has the potential to be developed as a treatment for traumatic brain injury in humans. All experimental procedures were approved by the Institutional Animal Investigation Committee of Capital Medical University, China (approval No. AEEI-2015-035) in December 2015.

11.
Neurotoxicology ; 68: 212-221, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29217406

RESUMO

Numerous long non-coding RNAs (lncRNAs) have been identified as aberrantly expressed in Parkinson's disease (PD). However, limited knowledge is available concerning the roles of dysregulated lncRNAs and the underlying molecular regulatory mechanism in the pathological process of PD. In this study, we found that lncRNA small nucleolar RNA host gene 1 (SNHG1) and seven in absentia homolog 1 (SIAH1) were upregulated, but microRNA-15b-5p (miR-15b-5p) was downregulated in SH-SY5Y cells pretreated with MPP+, as well as in MPTP-induced mouse model of PD. Overexpression of SIAH1 enhanced cellular toxicity of α-synuclein in SH-SY5Y cells, as indicated by the reduction of cell viability and elevation of LDH release. The percentage of α-synuclein aggregate-positive cells and the number of α-synuclein aggregates per cell were increased in SH-SY5Y cells transfected with pcDNA-SIAH1, while decreased after transfection with short interfering RNA specific for SIAH1 (si-SIAH1). Bioinformatics and luciferase reporter assay revealed that SIAH1 was a direct target of miR-15b-5p. We also found that SNHG1 could directly bind to miR-15-5p and repress miR-15-5p expression. Upregulation of miR-15b-5p alleviated α-synuclein aggregation and apoptosis by targeting SIAH1 in SH-SY5Y cells overexpressing α-synuclein. Overexpression of SNHG1 enhanced, whereas SNHG1 knockdown inhibited α-synuclein aggregation and α-synuclein-induced apoptosis. Moreover, the neuroprotective effect of si-SNHG1 was abrogated by downregulation of miR-15b-5p. In summary, our data suggest that SNHG1, as a pathogenic factor, promotes α-synuclein aggregation and toxicity by targeting the miR-15b-5p/SIAH1 axis, contributing to a better understanding of the mechanisms of Lewy body formation and loss of dopaminergic neurons in PD.


Assuntos
MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Doença de Parkinson/metabolismo , RNA Longo não Codificante/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidade , Animais , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Transtornos Parkinsonianos/metabolismo , Agregação Patológica de Proteínas/metabolismo
12.
Behav Pharmacol ; 28(2 and 3-Spec Issue): 207-213, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27984208

RESUMO

Chronic cerebral hypoperfusion (CCH) has been recognized as an important cause of both vascular dementia and Alzheimer's disease (AD), the two most prominent neurodegenerative diseases causing memory impairment in the elderly. However, an effective therapy for CCH-induced memory impairment has not yet been established. Grape seed polyphenol extract (GSPE) has powerful antioxidant properties and protects neurons and glia during ischemic injury, but its potential use in the prevention of CCH-induced memory impairment has not yet been investigated. Here, CCH-related memory impairment was modeled in rats using permanent bilateral occlusion of the common carotid artery. A Morris water maze task was used to evaluate memory, the levels of acetylcholinesterase, choline acetyltransferase, acetylcholine were used to evaluate cholinergic function, and oxidative stress was assessed by measuring the enzyme activity of superoxide dismutase, glutathione peroxidase, malonic dialdehyde, and catalase. We found that oral administration of GSPE for 1 month can rescue memory deficits. We also found that GSPE restores cholinergic neuronal function and represses oxidative damage in the hippocampus of CCH rats. We propose that GSPE protects memory in CCH rats by reducing ischemia-induced oxidative stress and cholinergic dysfunction. These findings provide a novel application of GSPE in CCH-related memory impairments.


Assuntos
Extrato de Sementes de Uva/farmacologia , Transtornos da Memória/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Polifenóis/farmacologia , Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Administração Oral , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Isquemia Encefálica/complicações , Demência Vascular , Modelos Animais de Doenças , Extrato de Sementes de Uva/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Polifenóis/administração & dosagem , Polifenóis/isolamento & purificação , Ratos , Ratos Wistar , Vitis/química
13.
Pain Physician ; 19(5): E761-5, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27389119

RESUMO

BACKGROUND: Post-dural puncture headache (PDPH) is the most common complication of lumbar puncture. Aminophylline has been reported to be effective in the prevention of PDPH in some clinical studies, but its efficacy for the treatment of PDPH has been unproven. OBJECTIVE: To evaluate the efficacy and safety of an intravenous (IV) injection of aminophylline on PDPH. STUDY DESIGN: The study was a multicenter, open-label study to assess the effectiveness and safety of aminophylline on PDPH. SETTING: The First Affiliated Hospital of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, and Henan Province Hospital of Traditional Chinese Medicine. METHODS: Thirty-two PDPH patients received an IV injection of aminophylline. The primary and secondary endpoints were the degree of headache and the patient's overall response to the treatment, respectively. Treatment safety was evaluated based on the occurrence of adverse reactions. RESULTS: Thirty-one patients completed the study. Before the initial aminophylline administration, the visual analog scale (VAS) score was 7.72 ± 1.65. The VAS scores at 30 minutes, one hour, 8 hours, one day, and 2 days post-treatment were 4.84 ± 2.53, 3.53 ± 2.06, 2.38 ± 1.96, 1.44 ± 1.87, and 0.81 ± 1.79, respectively, and were statistically significantly different (P < 0.05) compared with those before treatment. More than 50% (17/32) of the patients reported that they were "very much improved" or "much improved" 30 minutes after the initial treatment, increasing to 93.8% (30/32) at 2 days post-treatment. One patient experienced mild allergic reaction after treatment. LIMITATIONS: Although this study had the largest sample size among current studies on treating PDPH with theophylline drugs, the sample size was still relatively small and the method employed was not compared with a placebo or other current clinical treatments for PDPH. CONCLUSION: An IV injection of aminophylline may be an effective and safe early-stage treatment for PDPH.


Assuntos
Aminofilina/farmacologia , Avaliação de Resultados em Cuidados de Saúde , Cefaleia Pós-Punção Dural/tratamento farmacológico , Antagonistas de Receptores Purinérgicos P1/farmacologia , Adulto , Idoso , Aminofilina/administração & dosagem , Feminino , Humanos , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Antagonistas de Receptores Purinérgicos P1/administração & dosagem , Resultado do Tratamento , Adulto Jovem
14.
J Chem Theory Comput ; 12(1): 281-96, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26584231

RESUMO

The parametrization and validation of the OPLS3 force field for small molecules and proteins are reported. Enhancements with respect to the previous version (OPLS2.1) include the addition of off-atom charge sites to represent halogen bonding and aryl nitrogen lone pairs as well as a complete refit of peptide dihedral parameters to better model the native structure of proteins. To adequately cover medicinal chemical space, OPLS3 employs over an order of magnitude more reference data and associated parameter types relative to other commonly used small molecule force fields (e.g., MMFF and OPLS_2005). As a consequence, OPLS3 achieves a high level of accuracy across performance benchmarks that assess small molecule conformational propensities and solvation. The newly fitted peptide dihedrals lead to significant improvements in the representation of secondary structure elements in simulated peptides and native structure stability over a number of proteins. Together, the improvements made to both the small molecule and protein force field lead to a high level of accuracy in predicting protein-ligand binding measured over a wide range of targets and ligands (less than 1 kcal/mol RMS error) representing a 30% improvement over earlier variants of the OPLS force field.


Assuntos
Algoritmos , Proteínas/química , Bibliotecas de Moléculas Pequenas/química , Quinase 2 Dependente de Ciclina/química , Quinase 2 Dependente de Ciclina/metabolismo , Ligantes , Modelos Moleculares , Peptídeos/química , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas/metabolismo , Teoria Quântica , Bibliotecas de Moléculas Pequenas/metabolismo , Termodinâmica
15.
J Neurochem ; 134(3): 551-65, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25952107

RESUMO

Hypoxia-mediated neurotoxicity contributes to various neurodegenerative disorders, including Alzheimer's disease and multiple sclerosis. Tetramethylpyrazine (TMP), a major bioactive component purified from Ligusticum wallichii Franchat, exhibited potent neuroprotective effect. However, the mechanism of TMP-exerted neuroprotective effect against hypoxia was not clear. In the study, we investigated the mechanism of the neuroprotective effect of TMP against hypoxia induced by CoCl2 in vitro and in vivo. The results showed that TMP could protect against CoCl2 -induced neurotoxicity in PC12 cells and in rats, as evidenced by enhancement of cell viability in PC12 cells and improvement of learning and memory ability in rats treated with CoCl2 . TMP could inhibit mitochondrial dysfunction, mitochondrial apoptotic molecular events, and thus apoptosis induced by CoCl2 . TMP inhibited CoCl2 -increased reactive oxygen species (ROS) level, which may contribute to hypoxia-related neurotoxicity induced by CoCl2 . The antioxidant and neuroprotective activities of TMP involved two pathways: one was the enhancement of nuclear factor erythroid 2-related factor 2 (Nrf2)/catalytic subunit of γ-glutamylcysteine ligase-mediated regulation of GSH and the other was the inhibition of hypoxia-inducible factor 1 α/NADPH oxidase 2 (NOX2)-mediated ROS generation. These two pathways contributed to improvement of oxidative stress and thus the amelioration of apoptosis under hypoxic conditions. These results have appointed a new path toward the understanding of pathogenesis and TMP-related therapy of hypoxia-related neurodegenerative diseases. We proposed two cascades for tetramethylpyrazine-exhibited protective effects against CoCl2 -induced neurotoxicity: One is enhancement of nuclear factor erythroid 2-related factor 2-catalytic subunit of γ-glutamylcysteine ligase-mediated regulation of glutathone and the other was the inhibition of hypoxia-inducible factor 1 α-NADPH oxidase-2-mediated ROS generation. We think these findings should provide a new understanding of pathogenesis and tetramethylpyrazine-related therapy of hypoxia-related neurodegenerative diseases.


Assuntos
Hipóxia Celular/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Pirazinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Imunoprecipitação da Cromatina , Cobalto/toxicidade , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Células PC12 , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA