Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 19(3): e3001096, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33705388

RESUMO

The regulation of protein synthesis is essential for maintaining cellular homeostasis, especially during stress responses, and its dysregulation could underlie the development of human diseases. The critical step during translation regulation is the phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α). Here we report the identification of a direct kinase of eIF2α, microtubule affinity-regulating kinase 2 (MARK2), which phosphorylates eIF2α in response to proteotoxic stress. The activity of MARK2 was confirmed in the cells lacking the 4 previously known eIF2α kinases. MARK2 itself was found to be a substrate of protein kinase C delta (PKCδ), which serves as a sensor for protein misfolding stress through a dynamic interaction with heat shock protein 90 (HSP90). Both MARK2 and PKCδ are activated via phosphorylation in proteotoxicity-associated neurodegenerative mouse models and in human patients with amyotrophic lateral sclerosis (ALS). These results reveal a PKCδ-MARK2-eIF2α cascade that may play a critical role in cellular proteotoxic stress responses and human diseases.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Fator de Iniciação 2 em Eucariotos/fisiologia , Proteínas de Choque Térmico HSP90/metabolismo , Homeostase , Humanos , Camundongos , Camundongos Knockout , Microtúbulos/metabolismo , Fosforilação , Biossíntese de Proteínas , Estresse Fisiológico/fisiologia , eIF-2 Quinase/metabolismo
2.
Br J Nutr ; 130(8): 1329-1337, 2023 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36756752

RESUMO

This study aimed to explore the mediation effects of one-carbon metabolism (OCM) related nutrients on the association between MTHFR rs1801133 polymorphism and gestational diabetes mellitus (GDM). Folate, vitamin B12 and homocysteine (Hcy) were measured in the serum of 1254 pregnant women. Linear and logistic regressions were used to estimate the associations of OCM nutrients and MTHFR rs1801133 polymorphism with blood glucose levels and GDM risk. Mediation analysis was applied to test the mediation effects of folate, vitamin B12 and Hcy on the association of MTHFR rs1801133 polymorphism with blood glucose concentrations and GDM. Pregnant women with MTHFR rs1801133 CC genotype had higher serum folate (10·75 v. 8·90 and 9·40 ng/ml) and lower serum Hcy (4·84 v. 4·93 and 5·20 µmol/l) than those with CT and TT genotypes. Folate concentrations were positively associated with fasting plasma glucose (FPG), 1-h plasma glucose (1-h PG), 2-h plasma glucose (2-h PG) and GDM risk. Vitamin B12 levels were negatively correlated with FPG and GDM. Although no direct association was found between MTHFR rs1801133 genotypes and GDM, there were significant indirect effects of MTHFR rs1801133 CC genotype on FPG (ß: 0·005; 95 % CI: 0·001, 0·013), 1-h PG (ß: 0·006; 95 % CI: 0·001, 0·014), 2-h PG (ß: 0·007; 95 % CI: 0·001, 0·015) and GDM (ß: 0·006; 95 % CI: 0·001, 0·014) via folate. In conclusion, serum folate mediates the effect of MTHFR rs1801133 on blood glucose levels and GDM. Our findings potentially provide a feasible GDM prevention strategy via individualised folate supplementation according to the MTHFR genotypes.


Assuntos
Diabetes Gestacional , Ácido Fólico , Feminino , Humanos , Gravidez , Glicemia/análise , Diabetes Gestacional/sangue , Diabetes Gestacional/genética , População do Leste Asiático , Ácido Fólico/genética , Genótipo , Homocisteína , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Vitamina B 12 , Vitaminas
3.
Environ Res ; 221: 115331, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36681142

RESUMO

BACKGROUND: Single nucleotide polymorphisms (SNPs) in N6AMT1 and AS3MT are associated with arsenic (As) metabolism, and efficient As methylation capacity has been associated with diabetes. However, little is known about the gene-As interaction on gestational diabetes mellitus (GDM). OBJECTIVE: This study aimed to explore the individual and combined effects of N6AMT1 and AS3MT SNPs with As metabolism on GDM. METHODS: A cross-sectional study was performed among 385 Chinese pregnant women (86 GDM and 299 Non-GDM). Four SNPs in N6AMT1 (rs1997605 and rs1003671) and AS3MT (rs1046778 and rs11191453) were genotyped. Urinary inorganic arsenic (iAs), monomethylarsonic acid (MMA), and dimethylarsinic acid (DMA) were determined, and the percentages of As species (iAs%, MMA%, and DMA%) were calculated to assess the efficiency of As metabolism. RESULTS: Pregnant women with N6AMT1 rs1997605 AA genotype had lower iAs% (B: 2.11; 95% CI: 4.08, -0.13) and MMA% (B: 0.21; 95% CI: 0.39, -0.04) than pregnant women with GG genotype. The AS3MT rs1046778 and rs11191453 C alleles were negatively associated with iAs% and MMA% but positively associated with DMA%. Higher urinary MMA% was significantly associated with a lower risk of GDM (OR: 0.54; 95% CI: 0.30, 0.97). The A allele in N6AMT1 rs1997605 (OR: 0.46; 95% CI: 0.26, 0.79) was associated with a decreased risk of GDM. The additive interactions between N6AMT1 rs1997605 GG genotypes and lower iAs% (AP: 0.50; 95% CI: 0.01, 0.99) or higher DMA% (AP: 0.52; 95% CI: 0.04, 0.99) were statistically significant. Similar additive interactions were also found between N6AMT1 rs1003671 GG genotypes and lower iAs% or higher DMA%. CONCLUSIONS: Genetic variants in N6AMT1 and efficient As metabolism (indicated by lower iAs% and higher DMA%) can interact to influence GDM occurrence synergistically in Chinese pregnant women.


Assuntos
Arsênio , Diabetes Gestacional , Humanos , Feminino , Gravidez , Arsênio/metabolismo , Polimorfismo de Nucleotídeo Único , Diabetes Gestacional/genética , Gestantes , Metiltransferases/genética , Metiltransferases/metabolismo , Estudos Transversais , População do Leste Asiático , Ácido Cacodílico , DNA Metiltransferases Sítio Específica (Adenina-Específica)/genética , DNA Metiltransferases Sítio Específica (Adenina-Específica)/metabolismo
4.
Nutr Neurosci ; 26(5): 445-455, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35385370

RESUMO

Brain aging is a complex biological process often associated with a decline in cognitive functions and motility. Astaxanthin (AST) is a strong antioxidant capable of crossing the blood-brain barrier. The effect of AST on brain aging and its physiological and molecular mechanism are still unclear. The study aimed to investigate whether AST from AstaReal A1010 improved brain aging by inducing autophagy in SAMP10 mice. Different concentrations of AstaReal A1010 were intragastrically administered to 6-month-old SAMP10 mice for 3 months. The results demonstrated that AST delayed age-related cognitive decline, motor ability and neurodegeneration, upregulated the expression levels of autophagy-related genes beclin-1 and LC3 in the brain. It may induce autophagy by regulating IGF-1/Akt/mTOR and IGF-1/Akt/FoxO3a signaling. Treatment with autophagy inhibitor 3-methyladenine (3MA) partly reversed the anti-aging effect of AST. In conclusion, our findings suggest that AST may induce autophagy by regulating IGF-1/Akt/mTOR and IGF-1/Akt/FoxO3a signaling, thereby delaying age-related neurodegeneration and cognitive decline in SAMP10 mice.


Assuntos
Fator de Crescimento Insulin-Like I , Proteínas Proto-Oncogênicas c-akt , Camundongos , Animais , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Envelhecimento/fisiologia , Encéfalo/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia , Autofagia
5.
Nutr Neurosci ; 26(6): 483-495, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35416761

RESUMO

BACKGROUND: Post-stroke depression (PSD), the most frequent psychiatric complication following stroke, could have a negative impact on the recuperation of stroke patients. Hyperhomocysteinemia (HHCY) has been reported to be a modifiable risk factor of stroke. OBJECTIVE: The study tries to explore the effect of HHCY on PSD and the role of N-methyl-d-aspartate receptors (NMDARs)-mediated synaptic alterations. METHODS: Forty-five adult male Sprague-Dawley rats were randomly allocated into five groups: sham operation group, middle cerebral artery occlusion group (MCAO), HCY-treated MCAO group HCY and MK-801 co-treated MCAO group and MK-801-treated MCAO group. 1.6 mg/kg/d D, L-HCY was administered by tail vein injection for 28 d prior to SHAM or MCAO operationand up to 14 d after surgery. The MK-801 (3 mg/kg) was administered by intraperitoneal injection 15 min prior to MCAO operation. RESULTS: HCY treatment aggravated depressive-like disorders of post-stroke rats by the open field test and sucrose preference test. Further, HCY significantly decreased central monoamines levels in the MCAO rats by HPLC. The transmission electron microscopy results showed that the number of synapses and the area of postsynaptic density decreased in the hippocampus of the HCY-treated MCAO rats. Additionally, HCY augmented ischemia-induced up-regulation of NMDARs, decreased the levels of synaptic structure-related marker PSD-95and the synaptic transmission-associated synaptic proteins (VGLUT1, SNAP-25 and Complexin Ι/ΙΙ). These effects of HCY were partly reversed by the NMDA antagonist MK-801. CONCLUSIONS: The current study suggested that NMDARs-mediated synaptic plasticity may be involved in the adverse effect of HCY on PSD.


Assuntos
Infarto da Artéria Cerebral Média , Acidente Vascular Cerebral , Ratos , Animais , Masculino , Ratos Sprague-Dawley , Infarto da Artéria Cerebral Média/complicações , Receptores de N-Metil-D-Aspartato , Maleato de Dizocilpina/farmacologia , Acidente Vascular Cerebral/complicações , Reperfusão , Homocisteína
6.
Int J Mol Sci ; 24(5)2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36902471

RESUMO

The pathophysiology of Alzheimer's disease is thought to be directly linked to the abnormal aggregation of ß-amyloid (Aß) in the nervous system as a common neurodegenerative disease. Consequently, researchers in many areas are actively looking for factors that affect Aß aggregation. Numerous investigations have demonstrated that, in addition to chemical induction of Aß aggregation, electromagnetic radiation may also affect Aß aggregation. Terahertz waves are an emerging form of non-ionizing radiation that has the potential to affect the secondary bonding networks of biological systems, which in turn could affect the course of biochemical reactions by altering the conformation of biological macromolecules. As the primary radiation target in this investigation, the in vitro modeled Aß42 aggregation system was examined using fluorescence spectrophotometry, supplemented by cellular simulations and transmission electron microscopy, to see how it responded to 3.1 THz radiation in various aggregation phases. The results demonstrated that in the nucleation aggregation stage, 3.1 THz electromagnetic waves promote Aß42 monomer aggregation and that this promoting effect gradually diminishes with the exacerbation of the degree of aggregation. However, by the stage of oligomer aggregation into the original fiber, 3.1 THz electromagnetic waves exhibited an inhibitory effect. This leads us to the conclusion that terahertz radiation has an impact on the stability of the Aß42 secondary structure, which in turn affects how Aß42 molecules are recognized during the aggregation process and causes a seemingly aberrant biochemical response. Molecular dynamics simulation was employed to support the theory based on the aforementioned experimental observations and inferences.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Radiação Terahertz , Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Estrutura Secundária de Proteína
7.
Eur J Neurol ; 29(10): 2913-2924, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35735052

RESUMO

BACKGROUND: The longitudinal association between serum folate concentrations and the risk of cognitive impairment remains unclear in populations with low folate levels. We examined the association between serum folate concentrations and mild cognitive impairment (MCI) in older adults in China, where mandatory fortification of foods with folic acid has not been implemented. We further explored if homocysteine (Hcy) and leukocyte telomere length (LTL) mediate the association between serum folate and MCI. METHODS: We performed a longitudinal analysis of 3974 participants aged ≥60 years from the Tianjin Elderly Nutrition and Cognition (TENC) cohort study. The associations between serum folate level and the risk of cognitive impairment overall and stratified by apolipoprotein E (APOE) ε4 genotypes were evaluated using multivariable Cox proportional hazards models. The mediating effects of Hcy and LTL on the folate-MCI association were explored via a path analysis approach. RESULTS: Within a 3-year follow-up, we documented 560 incident MCI cases. After multivariable adjustment, higher serum folate concentrations were associated with lower incidence of MCI, with hazard ratios (95% confidence interval) across quartiles of folate (from lowest to highest concentrations) of 1.00 (reference), 0.66 (0.52, 0.83), 0.57 (0.45, 0.73), 0.66 (0.52, 0.84), respectively (p for trend <0.001). In mediation analyses, the status of serum folate deficiency and MCI were correlated via two intermediary pathways, Hcy and Hcy-telomere (p < 0.05). CONCLUSIONS: Lower folate concentrations, independently of APOE genotype, were associated with increased risk of MCI among elderly Chinese people, a population with relatively low folate intake. Our data were compatible with the mediation hypothesis that the association between folate status and MCI was mediated by Hcy and LTL.


Assuntos
Disfunção Cognitiva , Ácido Fólico , Idoso , Apolipoproteína E4 , China/epidemiologia , Disfunção Cognitiva/epidemiologia , Estudos de Coortes , Homocisteína , Humanos , Estudos Prospectivos , Vitamina B 12
8.
Sensors (Basel) ; 22(17)2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36081086

RESUMO

Machine tools, as an indispensable equipment in the manufacturing industry, are widely used in industrial production. The harsh and complex working environment can easily cause the failure of machine tools during operation, and there is an urgent requirement to improve the fault diagnosis ability of machine tools. Through the identification of the operating state (OS) of the machine tools, defining the time point of machine tool failure and the working energy-consuming unit can be assessed. In this way, the fault diagnosis time of the machine tool is shortened and the fault diagnosis ability is improved. Aiming at the problems of low recognition accuracy, slow convergence speed and weak generalization ability of traditional OS recognition methods, a deep learning method based on data-driven machine tool OS recognition is proposed. Various power data (such as signals or images) of CNC machine tools can be used to recognize the OS of the machine tool, followed by an intuitive judgement regarding whether the energy-consuming units included in the OS are faulty. First, the power data are collected, and the data are preprocessed by noise reduction and cropping using the data preprocessing method of wavelet transform (WT). Then, an AlexNet Convolutional Neural Network (ACNN) is built to identify the OS of the machine tool. In addition, a parameter adaptive adjustment mechanism of the ACNN is studied to improve identification performance. Finally, a case study is presented to verify the effectiveness of the proposed approach. To illustrate the superiority of this method, the approach was compared with traditional classification methods, and the results reveal the superiority in the recognition accuracy and computing speed of this AI technology. Moreover, the technique uses power data as a dataset, and also demonstrates good progress in portability and anti-interference.


Assuntos
Aprendizado Profundo , Redes Neurais de Computação , Fenômenos Físicos , Análise de Ondaletas
9.
Int J Neurosci ; 130(11): 1142-1150, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32064985

RESUMO

Purpose: In this study, we sought to test the hypothesis that oxidative stress injury in ischemic brains and H2O2-treated mouse neuroblastoma Neuro-2a cells (N2a) was related to STAT3 activation.Materials and methods: Rat middle cerebral artery occlusion (MCAO) model and H2O2-treated mouse neuroblastoma Neuro-2a cells (N2a) were used to investigate the relationship between oxidative stress injury and STAT3 activation.Results: 8-Hydroxy-2'-deoxyguanosine (8-OHdG) content and STAT3 protein phosphorylation level were significantly increased after cerebral ischemia-reperfusion. H2O2 treatment inhibited the cell viability, induced the apoptosis, and further raised pSTAT3 protein level in N2a cells. Moreover, the addition of AG490, the protein inhibitor of JAK2, significantly alleviated cerebral ischemic damage in vivo and H2O2-induced injury in vitro, and JAK2 siRNA also alleviated H2O2-induced injury in N2a cell.Conclusions: JAK2/STAT3 pathway may play a crucial role in mediating reactive oxidative species (ROS)-induced cell injury in rat middle cerebral artery occlusion (MCAO) model and N2a cells. ROS scavenging and down-regulation of STAT3 activation might be a candidate design of therapeutic strategies against oxidative stress-related neurological diseases.


Assuntos
Apoptose , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Janus Quinase 2/metabolismo , Neuroblastoma , Oxidantes/farmacologia , Estresse Oxidativo , Traumatismo por Reperfusão/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , 8-Hidroxi-2'-Desoxiguanosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/efeitos dos fármacos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Fator de Transcrição STAT3/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
10.
J Cell Mol Med ; 23(7): 4795-4807, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087489

RESUMO

Recent studies revealed that folic acid deficiency (FD) increased the likelihood of stroke and aggravated brain injury after focal cerebral ischaemia. The microglia-mediated inflammatory response plays a crucial role in the complicated pathologies that lead to ischaemic brain injury. However, whether FD is involved in the activation of microglia and the neuroinflammation after experimental stroke and the underlying mechanism is still unclear. The aim of the present study was to assess whether FD modulates the Notch1/nuclear factor kappa B (NF-κB) pathway and enhances microglial immune response in a rat middle cerebral artery occlusion-reperfusion (MCAO) model and oxygen-glucose deprivation (OGD)-treated BV-2 cells. Our results exhibited that FD worsened neuronal cell death and exaggerated microglia activation in the hippocampal CA1, CA3 and Dentate gyrus (DG) subregions after cerebral ischaemia/reperfusion. The hippocampal CA1 region was more sensitive to ischaemic injury and FD treatment. The protein expressions of proinflammatory cytokines such as tumour necrosis factor-α, interleukin-1ß and interleukin-6 were also augmented by FD treatment in microglial cells of the post-ischaemic hippocampus and in vitro OGD-stressed microglia model. Moreover, FD not only dramatically enhanced the protein expression levels of Notch1 and NF-κB p65 but also promoted the phosphorylation of pIkBα and the nuclear translocation of NF-κB p65. Blocking of Notch1 with N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester partly attenuated the nuclear translocation of NF-κB p65 and the protein expression of neuroinflammatory cytokines in FD-treated hypoxic BV-2 microglia. These results suggested that Notch1/NF-κB p65 pathway-mediated microglial immune response may be a molecular mechanism underlying cerebral ischaemia-reperfusion injury worsened by FD treatment.


Assuntos
Lesões Encefálicas/complicações , Isquemia Encefálica/complicações , Deficiência de Ácido Fólico/imunologia , Hipocampo/metabolismo , Microglia/imunologia , Receptor Notch1/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Linhagem Celular , Citocinas/metabolismo , Dipeptídeos/farmacologia , Glucose/deficiência , Infarto da Artéria Cerebral Média/complicações , Inflamação/patologia , Masculino , Camundongos , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oxigênio , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
11.
Acta Biochim Biophys Sin (Shanghai) ; 51(5): 501-508, 2019 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-30939192

RESUMO

Elevation of angiotensin II (Ang II) in the serum of patients with diabetes is known to promote apoptosis of islet ß cells, but the underlying mechanism remains unclear. The aim of the present study was to explore the role of Nod-like receptor protein 3 (NLRP3) inflammasome in Ang II-induced apoptosis of pancreatic islet ß cells and investigate the possible underlying mechanism. The effect of Ang II on INS-1 cell (a rat insulinoma cell line) viability was detected by CCK-8 method. The cell apoptosis was detected by flow cytometry and western blot analysis. The effect of Ang II on the expressions of thioredoxin-interacting protein (TXNIP) and NLRP3 protein was detected by western blot analysis. The expression of TXNIP mRNA was detected by real-time polymerase chain reaction. The results showed that Ang II was able to reduce INS-1 cell viability and promote apoptosis and at the same time up-regulate the expressions of TXNIP and NLRP3 components. Ang II-induced apoptosis was inhibited after administration of the NLRP3 inhibitor MCC950, and TXNIP silencing could reduce the NLRP3 expression and apoptosis, while both effects of Ang II on TXNIP-NLRP3 and its apoptosis-inducing effect were inhibited by angiotensin II type I receptor (AT1R) blocker Telmisartan. Our results demonstrated that the TXNIP-NLRP3 inflammasome pathway mediated Ang II-induced INS-1 cell apoptosis and might hopefully become a novel target for the treatment of diabetes mellitus.


Assuntos
Angiotensina II/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Inflamassomos/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Apoptose/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Humanos , Inflamassomos/genética , Células Secretoras de Insulina/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Ratos , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
12.
Sheng Li Xue Bao ; 70(2): 149-157, 2018 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-29691579

RESUMO

This study investigated the effect of angiotensin II (Ang II) on apoptosis and thioredoxin-interacting protein (TXNIP) expression in INS-1 islet cells and the underlying mechanism. INS-1 cells cultured in vitro were treated with different concentration of Ang II for different time, and the viability was measured using cell counting kit-8 (CCK-8). After treatment with 1 × 10-6 mol/L Ang II for 24 h, flow cytometry and Western blot were used to measure the cell apoptosis, and Western blot was used to analyze the protein expression of TXNIP, carbohydrate response element-binding protein (ChREBP) and angiotensin II type 1 receptor (AT1R). Real-time PCR was used to detect TXNIP and ChREBP mRNA expression. IF/ICC was used to observe the TXNIP, ChREBP and AT1R expression. The results showed that Ang II reduced cell viability and induced the expression of TXNIP in a dose- and time-dependent manner (P < 0.05, n = 6) compared with the control group. Ang II induced apoptosis and up-regulated the expression of ChREBP and AT1R (P < 0.05, n = 6). AT1R inhibitor, telmisartan (TM), blocked Ang II-induced TXNIP and ChREBP overexpression (P < 0.05, n = 6) and inhibited Ang II-induced apoptosis. Taken together, Ang II increased ChREBP activation through AT1R, which subsequently increased TXNIP expression and promoted cell apoptosis. These findings suggest a therapeutic potential of targeting TXNIP in preventing Ang II-induced INS-1 cell apoptosis in diabetes.


Assuntos
Angiotensina II/farmacologia , Apoptose , Proteínas de Transporte/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Proteínas de Ciclo Celular , Linhagem Celular , Ratos , Receptor Tipo 1 de Angiotensina/fisiologia , Telmisartan/farmacologia , Regulação para Cima
13.
Sheng Li Xue Bao ; 70(2): 158-166, 2018 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-29691580

RESUMO

Diabetes can cause a significant increase in the expression of thioredoxin (Trx)-interacting protein (TXNIP), which binds to Trx and inhibits its activity. The present study was aimed to investigate the effect of TXNIP on proliferation of rat INS-1 islet ß cells and the underlying mechanism. TXNIP overexpressing adenovirus vectors (Ad-TXNIP-GFP and Ad-TXNIPc247s-GFP) were constructed and used to infect INS-1 cells. Ad-TXNIPc247s-GFP vector carries a mutant C247S TXNIP gene, and its expression product (TXNIPc247s) cannot attach and inhibit Trx activity. The expression of TXNIP was detected by real-time PCR and Western blot. EdU and Ki67 methods were used to detect cell proliferation. Protein phosphorylation levels of ERK and AKT were detected by Western blot. The results showed that both TXNIP and TXNIPc247s protein overexpressions inhibited the proliferation of INS-1 cells, and the former's inhibitory effect was greater. Moreover, both of the two kinds of overexpressions inhibited the phosphorylation of ERK and AKT. These results suggest that TXNIP overexpression may inhibit the proliferation of INS-1 cells through Trx-dependent and non-Trx-dependent pathways, and the mechanism involves the inhibition of ERK and AKT phosphorylation.


Assuntos
Proteínas de Transporte/fisiologia , Vetores Genéticos , Células Secretoras de Insulina/citologia , Adenoviridae , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proliferação de Células , Diabetes Mellitus , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Oxirredução , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos
14.
J Neuroinflammation ; 14(1): 187, 2017 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-28923114

RESUMO

BACKGROUND: Elevated plasma homocysteine (Hcy) levels have been indicated as a strong and modifiable risk factor of ischemic stroke; the previous studies have shown that exposure to Hcy activates cultured microglia. However, whether neurotoxicity of Hcy involves microglia activation following brain ischemia and the underlying mechanisms remains incompletely understood. METHODS: The cerebral damage was evaluated by staining with 2,3,5-triphenyltetrazolium chloride, hematoxylin-eosin, and Fluoro Jade B. The activation state of microglia was assessed via immunoreaction using the microglial markers Iba1 and OX-42. Then, the inflammatory factors such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and phosphorylated signal transducer and activator of transcription 3 (pSTAT3) were examined by Western blot analysis and fluorescence immunohistochemistry. RESULTS: Elevated Hcy level augmented brain damage and neural cell toxicity in the brain cortex and the dentate gyrus region of the hippocampus after cerebral ischemia/reperfusion. Meanwhile, Hcy activated microglia and induced the expression of the inflammatory factors such as TNF-α and IL-6. Moreover, Hcy caused an increase in pSTAT3 expression which occurs in microglial cells. AG490, a JAK2-STAT3 inhibitor, effectively inhibited the phosphorylation of STAT3, microglial cell activation and the secretion of IL-6, TNF-α raised by Hcy treatment. CONCLUSIONS: STAT3 signaling pathway located in microglia plays a critical role in mediating Hcy-induced activation of microglia and neuroinflammation in rat MCAO model. This suggests the feasibility of targeting the JAK2/STAT3 pathway as an effective therapeutic strategy to alleviate the progression of Hcy-associated ischemia stroke.


Assuntos
Homocisteína/metabolismo , Inflamação/metabolismo , Microglia/metabolismo , Fator de Transcrição STAT3/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Inflamação/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/patologia
15.
J Bioenerg Biomembr ; 49(2): 131-138, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27914013

RESUMO

Increased blood plasma concentration of the sulphur amino acid homocysteine (Hcy) is considered as an independent risk factor of the neurodegenerative diseases. However, the detailed molecular mechanisms by which Hcy leads to neurotoxicity have yet to be clarified. Recent research has suggested that neurotoxicity of Hcy may involve negative regulation of neural stem cell (NSC) proliferation. In the current study, primary NSCs were isolated from neonatal rat brain hippocampus and the inhibition in cell growth was observed after exposure to l50 µM and 500 µM L-Hcy. The changes in protein expression were monitored with densitometric 2D-gel electrophoresis coupled with MALDI-TOF mass spectrometry. Proteomic analysis revealed that the expression levels of two mitochondrial proteins, cytochrome bc1 complex2 (UQCRC2, the major component of electron transport chain complex III) and aconitase (an enzyme involved in the tricarboxylic acid cycle), were decreased in Hcy treatment group, compared to control group. Protein expression was further verified by Western blot, and their enzymatic activities were also down-regulated in NSCs after Hcy treatment. Restoration of aconitase and UQCRC2 protein levels using their expression vectors could partly rescue the cell viability inhibition caused by Hcy. Moreover, Hcy caused the increase in the intracellular levels of reactive oxygen species (ROS) and the decrease in ATP content, which are known to play important roles in the cellular stress response of the cell growth. Altogether, the results suggest that the decreased expression and enzymatic activities of the mitochondrial proteins may be possible causes of the overproduction of ROS and depletion of ATP. The inhibition in cell growth at the end of Hcy treatment was probably due to the changes in protein expression and mitochondrial dysfunction in vitro cultures of NSCs.


Assuntos
Proliferação de Células/efeitos dos fármacos , Homocisteína/farmacologia , Células-Tronco Neurais/citologia , Aconitato Hidratase/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Hipocampo/citologia , Homocisteína/sangue , Proteômica , Ratos , Espécies Reativas de Oxigênio/metabolismo
16.
Eur J Nutr ; 55(5): 1849-62, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26224648

RESUMO

PURPOSE: Alzheimer's disease (AD) is a highly prevalent type of dementia. The epigenetic mechanism of gene methylation provides a putative link between nutrition, one-carbon metabolism, and disease progression because folate deficiency may cause hypomethylation of promoter regions in AD-relevant genes. We hypothesized that folic acid supplementation may protect neuron cells from amyloid ß (Aß) oligomer-induced toxicity by modulating DNA methylation of APP and PS1 in AD models. METHODS: Primary hippocampal neuronal cells and hippocampal HT-22 cells were incubated for 24 h with a combination of folic acid and either Aß oligomers or vehicle and were then incubated for 72 h with various concentrations of folic acid. AD transgenic mice were fed either folate-deficient or control diets and gavaged daily with various doses of folic acid (0 or 600 µg/kg). DNA methyltransferase (DNMT) activity, cell viability, methylation potential of cells, APP and PS1 expression, and the methylation of the respective promoters were determined. RESULTS: Aß oligomers lowered DNMT activity, increased PS1 and APP expression, and decreased cell viability. Folic acid dose-dependently stimulated methylation potential and DNMT activity, altered PS1 and APP promoter methylation, decreased PS1 and APP expression, and partially preserved cell viability. Folic acid increased PS1 and APP promoter methylation in AD transgenic mice. CONCLUSION: These results suggest a mechanism by which folic acid may prevent Aß oligomer-induced neuronal toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Metilação de DNA/efeitos dos fármacos , Ácido Fólico/farmacologia , Neurônios/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ácido Fólico/sangue , Deficiência de Ácido Fólico/sangue , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Regiões Promotoras Genéticas
17.
Pediatr Crit Care Med ; 17(3): 194-202, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26808622

RESUMO

OBJECTIVES: Extracorporeal membrane oxygenation is often used in children with single-ventricle anomalies. We aimed to describe extracorporeal membrane oxygenation use in single-ventricle patients to test the hypothesis that despite increasing prevalence, mortality has not improved and overall burden measure by hospital charges and length of stay have increased. DESIGN: Retrospective analysis of the Healthcare Cost and Utilization Project Kids' Inpatient Database was performed with sample weighting to generate national estimates. PATIENTS: Pediatric patients (age ≤ 20) with a diagnosis of single ventricle heart disease requiring extracorporeal membrane oxygenation support from 2000 to 2009. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Seven hundred one children (95% CI, 559-943) with single ventricle were supported with extracorporeal membrane oxygenation in the reporting period. Mortality was 57% and did not improve over time (2000 = 52%, 2003 = 63%, 2006 = 57%, and 2009 = 55%; p = 0.66). Single-ventricle patients who required extracorporeal membrane oxygenation were more likely to have had a cardiac procedure (90% vs 46%; p < 0.001), a diagnosis of arrhythmia (22% vs 13%; p < 0.001), cerebrovascular or neurologic insult (9% vs 1%; p < 0.001), heart failure (24% vs 12%; p < 0.001), acute renal failure (28% vs 3%; p < 0.001), or sepsis (28% vs 8%; p < 0.001). By multivariable analysis, acute renal failure was a risk factor for mortality (adjusted odds ratio, 3.12; 95% CI, 1.95-4.98; p < 0.001). The length of stay for single-ventricle patients with extracorporeal membrane oxygenation increased from 25.2 days in 2000 to 55.6 days in 2009 (p < 0.001). Total inflation-adjusted charges increased from $358,021 (95% CI, $278,658-439,765) in 2000 to $732,349 (95% CI, $671,781-792,917) in 2009 (p < 0.001). CONCLUSIONS: Extracorporeal membrane oxygenation support is uncommon with single-ventricle admissions occurring in 2.3% of all hospitalizations. Among those patients, the mortality rate was 57% with no change over time. Acute renal failure was an independent risk factor for mortality during hospitalization. In addition, length of stay for these patients increased and hospital charges doubled. Further studies are needed to determine suitability and cost-effectiveness of extracorporeal membrane oxygenation in single-ventricle patients.


Assuntos
Oxigenação por Membrana Extracorpórea/mortalidade , Cardiopatias Congênitas/terapia , Preços Hospitalares/tendências , Tempo de Internação/tendências , Adolescente , Criança , Pré-Escolar , Oxigenação por Membrana Extracorpórea/economia , Feminino , Ventrículos do Coração/anormalidades , Mortalidade Hospitalar , Humanos , Lactente , Recém-Nascido , Tempo de Internação/economia , Masculino , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento , Estados Unidos/epidemiologia
18.
Int J Mol Sci ; 17(8)2016 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-27455253

RESUMO

Elevated homocysteine (Hcy) levels have been reported to be involved in neurotoxicity after ischemic stroke. However, the underlying mechanisms remain incompletely understood to date. In the current study, we hypothesized that neuronal autophagy activation may be involved in the toxic effect of Hcy on cortical neurons following cerebral ischemia. Brain cell injury was determined by hematoxylin-eosin (HE) staining and TdT-mediated dUTP Nick-End Labeling (TUNEL) staining. The level and localization of autophagy were detected by transmission electron microscopy, western blot and immunofluorescence double labeling. The oxidative DNA damage was revealed by immunofluorescence of 8-Hydroxy-2'-deoxyguanosine (8-OHdG). Hcy treatment aggravated neuronal cell death, significantly increased the formation of autophagosomes and the expression of LC3B and Beclin-1 in the brain cortex after middle cerebral artery occlusion-reperfusion (MCAO). Immunofluorescence analysis of LC3B and Beclin-1 distribution indicated that their expression occurred mainly in neurons (NeuN-positive) and hardly in astrocytes (GFAP-positive). 8-OHdG expression was also increased in the ischemic cortex of Hcy-treated animals. Conversely, LC3B and Beclin-1 overexpression and autophagosome accumulation caused by Hcy were partially blocked by the autophagy inhibitor 3-methyladenine (3-MA). Hcy administration enhanced neuronal autophagy, which contributes to cell death following cerebral ischemia. The oxidative damage-mediated autophagy may be a molecular mechanism underlying neuronal cell toxicity of elevated Hcy level.


Assuntos
Autofagia/efeitos dos fármacos , Isquemia Encefálica/fisiopatologia , Córtex Cerebral/lesões , Homocisteína/toxicidade , Neurônios/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Imunofluorescência , Infarto da Artéria Cerebral Média , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão
19.
Int J Mol Sci ; 16(10): 25002-13, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26492244

RESUMO

Alzheimer's disease (AD) is a common neurodegenerative disease resulting in progressive dementia, and is a principal cause of dementia among older adults. Folate acts through one-carbon metabolism to support the methylation of multiple substrates. We hypothesized that folic acid supplementation modulates DNA methyltransferase (DNMT) activity and may alter amyloid ß-peptide (Aß) production in AD. Mouse Neuro-2a cells expressing human APP695 were incubated with folic acid (2.8-40 µmol/L), and with or without zebularine (the DNMT inhibitor). DNMT activity, cell viability, Aß and DNMTs expression were then examined. The results showed that folic acid stimulated DNMT gene and protein expression, and DNMT activity. Furthermore, folic acid decreased Aß protein production, whereas inhibition of DNMT activity by zebularine increased Aß production. The results indicate that folic acid induces methylation potential-dependent DNMT enzymes, thereby attenuating Aß production.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Fólico/uso terapêutico , Doença de Alzheimer/enzimologia , Animais , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Metilases de Modificação do DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ácido Fólico/farmacologia , Humanos , Camundongos
20.
Clin Exp Obstet Gynecol ; 41(5): 578-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25864264

RESUMO

A middle-aged woman with rheumatic heart disease, mitral valve prolapse and incompletely closed mitral valve medium, patent foramen ovale, merge multiple uterine fibroids, and moderate blood loss anemia underwent mitral valve replacement surgery with total abdominal hysterectomy under general anesthesia and cardiopulmonary bypass condition. The surgery was successful, and postoperative bleeding, blood clots, heart failure, and other related complications did not occur. Heart valve replacement surgery with the surgical treatment of uterine fibroids effectively improves the safety of surgical treatment for patients as well as reduces the patient's medical expenses and risk of secondary surgery and trauma.


Assuntos
Implante de Prótese de Valva Cardíaca/métodos , Histerectomia/métodos , Leiomioma/cirurgia , Prolapso da Valva Mitral/cirurgia , Neoplasias Uterinas/cirurgia , Adulto , Feminino , Seguimentos , Humanos , Leiomioma/complicações , Prolapso da Valva Mitral/complicações , Fatores de Tempo , Neoplasias Uterinas/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA