Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging Dis ; 14(2): 450-467, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37008060

RESUMO

Ischemic stroke is an extremely common pathology with strikingly high morbidity and mortality rates. The endoplasmic reticulum (ER) is the primary organelle responsible for conducting protein synthesis and trafficking as well as preserving intracellular Ca2+ homeostasis. Mounting evidence shows that ER stress contributes to stroke pathophysiology. Moreover, insufficient circulation to the brain after stroke causes suppression of ATP production. Glucose metabolism disorder is an important pathological process after stroke. Here, we discuss the relationship between ER stress and stroke and treatment and intervention of ER stress after stroke. We also discuss the role of glucose metabolism, particularly glycolysis and gluconeogenesis, post-stroke. Based on recent studies, we speculate about the potential relationship and crosstalk between glucose metabolism and ER stress. In conclusion, we describe ER stress, glycolysis, and gluconeogenesis in the context of stroke and explore how the interplay between ER stress and glucose metabolism contributes to the pathophysiology of stroke.

2.
Mediators Inflamm ; 2022: 6886752, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35873710

RESUMO

Cerebral ischemia-reperfusion (I/R) incites neurologic damage through a myriad of complex pathophysiological mechanisms, most notably, inflammation and oxidative stress. In I/R injury, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) produces reactive oxygen species (ROS), which promote inflammatory and apoptotic pathways, augmenting ROS production and promoting cell death. Inhibiting ischemia-induced oxidative stress would be beneficial for reducing neuroinflammation and promoting neuronal cell survival. Studies have demonstrated that chlorpromazine and promethazine (C+P) induce neuroprotection. This study investigated how C+P minimizes oxidative stress triggered by ischemic injury. Adult male Sprague-Dawley rats were subject to middle cerebral artery occlusion (MCAO) and subsequent reperfusion. 8 mg/kg of C+P was injected into the rats when reperfusion was initiated. Neurologic damage was evaluated using infarct volumes, neurological deficit scoring, and TUNEL assays. NOX enzymatic activity, ROS production, protein expression of NOX subunits, manganese superoxide dismutase (MnSOD), and phosphorylation of PKC-δ were assessed. Neural SHSY5Y cells underwent oxygen-glucose deprivation (OGD) and subsequent reoxygenation and C+P treatment. We also evaluated ROS levels and NOX protein subunit expression, MnSOD, and p-PKC-δ/PKC-δ. Additionally, we measured PKC-δ membrane translocation and the level of interaction between NOX subunit (p47phox) and PKC-δ via coimmunoprecipitation. As hypothesized, treatment with C+P therapy decreased levels of neurologic damage. ROS production, NOX subunit expression, NOX activity, and p-PKC-δ/PKC-δ were all significantly decreased in subjects treated with C+P. C+P decreased membrane translocation of PKC-δ and lowered the level of interaction between p47phox and PKC-δ. This study suggests that C+P induces neuroprotective effects in ischemic stroke through inhibiting oxidative stress. Our findings also indicate that PKC-δ, NOX, and MnSOD are vital regulators of oxidative processes, suggesting that C+P may serve as an antioxidant.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , AVC Isquêmico , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Animais , Isquemia Encefálica/tratamento farmacológico , Clorpromazina/farmacologia , Clorpromazina/uso terapêutico , Masculino , NADPH Oxidases/metabolismo , Estresse Oxidativo , Prometazina/farmacologia , Prometazina/uso terapêutico , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Superóxido Dismutase/metabolismo
3.
Aging Dis ; 13(2): 521-533, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35371601

RESUMO

Stroke is one of the most prevalent causes of death around the world. When a stroke occurs, many cellular signaling cascades and regulators are activated, which results in severe cellular dysfunction and debilitating long-term disability. One crucial regulator of cell fate and function is mammalian Forkhead box protein O1 (FoxO1). Many studies have found FoxO1 to be implicated in many cellular processes, including regulating gluconeogenesis and glycogenolysis. During a stroke, modifications of FoxO1 have been linked to a variety of functions, such as inducing cell death and inflammation, inhibiting oxidative injury, affecting the blood brain barrier (BBB), and regulating hepatic gluconeogenesis. For these functions of FoxO1, different measures and treatments were applied to FoxO1 after ischemia. However, the subtle mechanisms of post-transcriptional modification and the role of FoxO1 are still elusive and even contradictory in the development of stroke. The determination of these mechanisms will lead to further enlightenment for FoxO1 signal transduction and the identification of targeted drugs. The regulation and function of FoxO1 may provide an important way for the prevention and treatment of diseases. Overall, the functions of FoxO1 are multifactorial, and this paper will summarize all of the significant pathways in which FoxO1 plays an important role during stroke damage and recovery.

4.
Neurol Res ; 44(6): 483-495, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34983317

RESUMO

OBJECTIVES:  Neuroinflammation plays a key role in cerebrovascular disease (CVD). Neuropsychiatric disorders appear to share an epidemiological association with inflammation, but the mechanisms are unclear. Forkhead box 1 (FoxO1) regulates inflammatory signaling in diabetes and cardiovascular diseases, but its role in psychological stress-induced neuroinflammation remains unknown. Therefore, we investigated the potential involvement of FoxO1 in repeated social defeat stress (RSDS)-induced neuroinflammation. METHODS:  6-week-old male C57BL/6 J mice were randomly divided into RSDS or control groups. In the RSDS group, mice (18-22 g) were individually subjected to social defeat by an 8-week-old CD-1 mouse (28-32 g) for 10 min daily for 10 consecutive days. At 24 h after this 10-day process, corticosterone (CORT), epinephrine (EPI), hydrogen peroxide, and inflammatory factors (TNF-α, IL-6, IL-1ß, and VCAM-1) from serum and brain tissues were assayed using ELISA, real-time PCR, and Western blot. Iba-1 was determined by immunofluorescence (IF), and FoxO1 siRNA was transfected into BV2 cells to further analyze the expression of inflammatory factors. RESULTS: RSDS significantly increased the levels of TNF-α, IL-6, IL-1ß, and VCAM-1 in the serum; it also increased both mRNA and protein expression of these in the brain. FoxO1 was significantly increased after stress, while its knockdown significantly suppressed stress-induced inflammation. Immunofluorescence demonstrated the activation of microglia in the setting of RSDS. CONCLUSION: RSDS induced a measurable inflammatory response in the blood and brain, and FoxO1 was demonstrated in vitro to aggravate stress-induced inflammation.


Assuntos
Interleucina-6 , Fator de Necrose Tumoral alfa , Animais , Proteína Forkhead Box O1/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias , Estresse Psicológico/complicações , Molécula 1 de Adesão de Célula Vascular/efeitos adversos
5.
Mol Neurobiol ; 58(12): 6136-6152, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34455546

RESUMO

A depressive or hibernation-like effect of chlorpromazine and promethazine (C + P) on brain activity was reported to induce neuroprotection, with or without induced-hypothermia. However, the underlying mechanisms remain unclear. The current study evaluated the pharmacological function of C + P on the inhibition of neuroinflammatory response and inflammasome activation after ischemia/reperfusion. A total of 72 adult male Sprague-Dawley rats were subjected to 2 h middle cerebral artery occlusion (MCAO) followed by 6 or 24 h reperfusion. At the onset of reperfusion, rats received C + P (8 mg/kg) with temperature control. Brain cell death was detected by measuring CD68 and myeloperoxidase (MPO) levels. Inflammasome activation was measured by mRNA levels of NLRP3, IL-1ß, and TXNIP, and protein quantities of NLRP3, IL-1ß, TXNIP, cleaved-Caspase-1, and IL-18. Activation of JAK2/STAT3 pathway was detected by the phosphorylation of STAT3 (p-STAT3) and JAK2 (p-JAK2), and the co-localization of p-STAT3 and NLRP3. Activation of the p38 pathway was assessed with the protein levels of p-p38/p38. The mRNA and protein levels of HIF-1α, FoxO1, and p-FoxO1, and the co-localization of p-STAT3 with HIF-1α or FoxO1 were quantitated. As expected, C + P significantly reduced cell death and attenuated the neuroinflammatory response as determined by reduced CD68 and MPO. C + P decreased ischemia-induced inflammasome activation, shown by reduced mRNA and protein expressions of NLRP3, IL-1ß, TXNIP, cleaved-Caspase-1, and IL-18. Phosphorylation of JAK2/STAT3 and p38 pathways and the co-localization of p-STAT3 with NLRP3 were also inhibited by C + P. Furthermore, mRNA levels of HIF-1α and FoxO1 were decreased in the C + P group. While C + P inhibited HIF-1α protein expression, it increased FoxO1 phosphorylation, which promoted the exclusion of FoxO1 from the nucleus and inhibited FoxO1 activity. At the same time, C + P reduced the co-localization of p-STAT3 with HIF-1α or FoxO1. In conclusion, C + P treatment conferred neuroprotection in stroke by suppressing neuroinflammation and NLRP3 inflammasome activation. The present study suggests that JAK2/STAT3/p38/HIF-1α/FoxO1 are vital regulators and potential targets for efficacious therapy following ischemic stroke.


Assuntos
Inflamassomos/efeitos dos fármacos , AVC Isquêmico/tratamento farmacológico , Doenças Neuroinflamatórias/tratamento farmacológico , Fenotiazinas/uso terapêutico , Animais , Morte Celular/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , AVC Isquêmico/metabolismo , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças Neuroinflamatórias/metabolismo , Fenotiazinas/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
6.
Brain Res ; 1763: 147463, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33811844

RESUMO

BACKGROUND: After ischemic stroke, the increased catabolism of glucose (hyperglycolysis) results in the production of reactive oxygen species (ROS) via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). A depressive or hibernation-like effect of C + P on brain activity was reported to induce neuroprotection. The current study assesses the effect of C + P on hyperglycolysis and NOX activation. METHODS: Adult male Sprague-Dawley rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion. At the onset of reperfusion, rats received C + P with or without temperature control, or phloretin [glucose transporter (GLUT)-1 inhibitor], or cytochalasin B (GLUT-3 inhibitor). We detected brain ROS, apoptotic cell death, and ATP levels along with HIF-1α expression. Cerebral hyperglycolysis was measured by glucose, protein expression of GLUT-1/3, and phosphofructokinase-1 (PFK-1), as well as lactate and lactate dehydrogenase (LDH) at 6 and 24 h of reperfusion. The enzymatic activity of NOX and protein expression of its subunits (gp91phox) were detected. Neural SHSY5Y cells were placed under 2 h of oxygen-glucose deprivation (OGD) followed by reoxygenation for 6 and 24 h with C + P treatment. Cell viability and protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured. A HIF-1α overexpression vector was transfected into the cells, and then protein levels of HIF-1α, GLUT-1/3, PFK-1, and LDH were quantitated. In sham-operated rats and control cells, the protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured at 6 and 24 h after C + P administration. RESULTS: C + P reduced the protein elevations after stroke in HIF-1α, glycolytic enzymes, as well as in ROS, cell death, glucose and lactate, but raised ATP levels in the brain. In ischemic rats exposed to GLUT-1/3 inhibitors, ROS, cell death, glucose, and lactate were all decreased, as well as GLUT-1, GLUT-3, LDH, and PFK-1 protein levels. C + P decreased ischemia-induced NOX activation by reducing the enzymatic activity and protein expression of the NOX subunit gp91phox, as was observed in the presence of GLUT-1/3 inhibitors. These markers were significantly decreased following C + P administration with the induced hypothermia, while C + P administration with temperature control at 37 °C induced lesser protection after ischemia stroke. In the OGD/reoxygenation model, C + P treatment increased cell viability and diminished protein levels of HIF-1α, GLUT-1, GLUT-3, PFK-1, LDH, and gp91phox. However, in OGD with HIF-1α overexpression, C + P was unable to effectively reduce the upregulated GLUT-1, GLUT-3, and LDH. In normal conditions, C + P reduced HIF-1α and the levels of key glycolytic enzymes depending on its pharmacological effect. CONCLUSION: C + P, partially depending on hypothermia, attenuates hyperglycolysis and NOX activation through HIF-1α regulation.


Assuntos
Clorpromazina/uso terapêutico , Glicólise/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , AVC Isquêmico/tratamento farmacológico , Prometazina/uso terapêutico , Animais , Clorpromazina/farmacologia , Glucose/deficiência , Transportador de Glucose Tipo 1/efeitos dos fármacos , Transportador de Glucose Tipo 3/efeitos dos fármacos , Hipóxia , Infarto da Artéria Cerebral Média/tratamento farmacológico , L-Lactato Desidrogenase/efeitos dos fármacos , Masculino , NADPH Oxidase 2/efeitos dos fármacos , Fosfofrutoquinase-1/efeitos dos fármacos , Prometazina/farmacologia , Ratos , Ratos Sprague-Dawley
7.
Curr Neurovasc Res ; 16(3): 232-240, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31232236

RESUMO

BACKGROUND: The inflammatory response to acute cerebral ischemia is a major factor in stroke pathobiology and patient outcome. In the clinical setting, no effective pharmacologic treatments are currently available. Phenothiazine drugs, such as chlorpromazine and promethazine, (C+P) have been widely studied because of their ability to induce neuroprotection through artificial hibernation after stroke. The present study determined their effect on the inflammatory response. METHODS: Sprague-Dawley rats were divided into 4 groups: (1) sham, (2) stroke, (3) stroke treated by C+P without temperature control and (4) stroke treated by C+P with temperature control (n=8 per group). To assess the neuroprotective effect of C+P, brain damage was measured using infarct volume and neurological deficits. The expression of inflammatory response molecules tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and nuclear factor kappa light chain enhancer of activated B cells (NF-κB) was determined by real-time PCR and Western blotting. RESULTS: TNF-α, IL-1ß, ICAM-1, VCAM-1, and NF-κB mRNA and protein expressions were upregulated, and brain damage and neurological deficits were increased after stroke. These markers of cerebral injury were significantly reduced following C+P administration under drug-induced hypothermia, while C+P administration under normal body temperature reduced them by a lesser degree. CONCLUSION: This study showed an inhibitory effect of C+P on brain inflammation, which may be partially dependent on drug-induced hibernation, as well as other mechanisms of action by these drugs. These findings further suggest the great potential of C+P in the clinical treatment of ischemic stroke.


Assuntos
Antipsicóticos/uso terapêutico , Encéfalo/fisiopatologia , Hibernação/fisiologia , Neuroproteção/fisiologia , Fenotiazinas/uso terapêutico , Acidente Vascular Cerebral/prevenção & controle , Animais , Antipsicóticos/farmacologia , Encéfalo/efeitos dos fármacos , Hibernação/efeitos dos fármacos , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Masculino , Neuroproteção/efeitos dos fármacos , Fenotiazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/fisiopatologia
8.
Neurol Res ; 41(8): 762-769, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31092145

RESUMO

Objectives: Cerebrovascular disease (CVD) is the leading cause of permanent disability worldwide. Inflammation has been reported to play an important role in the progression of CVD. Neuropsychiatric disorders such as depression are associated with increased incidence of CVD epidemiologically, although the mechanisms underlying this association are not clear. In this study, we assessed the effect of the acute repeated social defeat stress (RSDS) and chronic restraint stress (CRS) on neuroinflammation in mice. Methods: A total of 40 6-week-old male C57BL/6J mice were divided into RSDS, CRS, and corresponding control groups. In the RSDS group, male C57BL/6J mice were repeatedly subjected to bouts of social defeat by a larger CD-1 mouse for 10 min daily for 10 consecutive days. In the CRS group, the mice were exposed to restraint stress for 6 h per day for 28 consecutive days. Depressive behavior was evaluated by conducting sucrose preference test over 24 h. Peripheral blood serum and brain tissues were collected for measurement of corticosterone (CORT), epinephrine (EPI), and inflammatory factors (TNF-α and IL-6) using ELISA or real-time PCR 24 h after the sucrose preference test. Results: Both RSDS and CRS decreased the sucrose preference ratio. The acute stress increased serum CORT and EPI, while the chronic stress did not significantly influence them. Both stress models induced an inflammatory response in peripheral serum and the brain. Conclusions: RSDS and CRS are two effective models of depressive behavior, and both models cause neuroinflammation, which may be responsible for the increased risk of CVD seen in patients with depression.


Assuntos
Encefalite/sangue , Encefalite/etiologia , Estresse Psicológico/sangue , Estresse Psicológico/complicações , Animais , Corticosterona/sangue , Modelos Animais de Doenças , Epinefrina/sangue , Interleucina-6/sangue , Masculino , Camundongos Endogâmicos C57BL , Restrição Física , Comportamento Social , Fator de Necrose Tumoral alfa/sangue
9.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 39(4): 518-524, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-28877830

RESUMO

Objective To build an efficient random short hairpin RNA(shRNA)library. Methods shRNA expression vector was constructed with enhanced green fluorescent protein(EGFP)in the upstream of shRNA,driven by pol Ⅱ promoter(CMV).After the constructs were transfected into cells,the proteins were collected.The inhibition efficiency of shRNA was determined by Western blot and dual luciferase reporter system.After the shRNA expression vector was constructed with EGFP in the upstream of shRNA,driven by pol Ⅱ promoter(CMV),shRNA was further embedded into microRNA(miRNA)context.The constructs were transfected into cells,and then the inhibition efficiency of shRNA against target genes was evaluated by quantificational real-time polymerase chain reaction.According to the result of quantificational real-time polymerase chain reaction,a new random shRNA library was constructed based on miRNA context. Results shRNA downstream of a large transcript was transcripted efficiently by pol Ⅱ promoter(CMV).The efficiency of shRNA interference on target gene was improved when shRNA was embedded into miRNA context.Thus,we constructed a new random shRNA library sized 1.8×107 based on miRNA context.Conclusion We successfully constructed a new large random shRNA library.


Assuntos
Biblioteca Gênica , MicroRNAs/genética , RNA Interferente Pequeno/genética , Vetores Genéticos , Luciferases , Regiões Promotoras Genéticas
10.
Nucleic Acids Res ; 45(5): 2849-2864, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27789685

RESUMO

Most of our knowledge about translation regulatory mechanisms comes from studies on lower organisms. However, the translation control system of higher organisms is less understood. Here we find that in 5' untranslated region (5'UTR) of human Annexin II receptor (AXIIR) mRNA, there are two upstream open reading frames (uORFs) acting in a fail-safe manner to inhibit the translation from the main AUG. These uORFs are unfavorable for re-initiation after termination of uORF translation. Heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1), hnRNPA0 and ELAV like RNA binding protein 1 (ELAVL1) bind to the 5'UTR of AXIIR mRNA. They focus the translation of uORFs on uORF1 and attenuate leaky scanning that bypasses uORFs. The cooperation between the two uORFs and the three proteins formed a multiple fail-safe system that tightly inhibits the translation of downstream AXIIR. Such cooperation between multiple molecules and elements reflects that higher organism develops a complex translation regulatory system to achieve accurate and flexible gene expression control.

11.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(5): 470-6, 2014 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-25360642

RESUMO

OBJECTIVE: To investigate the role of TXNDC5 in serum starvation-induced proliferation inhibition of HeLa cell. METHODS: TXNDC5 was either over-expressed or knocked down by small interfering RNA (siRNA) in HeLa cells which were then cultured in conventional medium or serum starvation medium. The protein level of TXNDC5 was evaluated by Western blot analysis. The mRNA level of TXNDC5 was measured by quantitative real-time PCR. Cell growth rate was determined by cell proliferation assay kit (MTS method). Cell cycle distribution and apoptosis were detected by flow cytometry. RESULTS: Serum starvation mildly reduced the mRNA level of TXNDC5 (P<0.05), but dramatically increased the protein level of TXNDC5 in HeLa cells. The stability of TXNDC5 mRNA remained unchanged. Cycloheximide abolished the serum starvation-induced up-regulation of TXNDC5 protein. Over-expression of TXNDC5 had no effect on cell proliferation. However, suppression of TXNDC5 attenuated the proliferation inhibition of HeLa cell induced by serum starvation (P<0.05), increased the proportion of cells in S phase (P<0.05), but had no effect on cell apoptosis. CONCLUSION: TXNDC5 mediates serum starvation-induced proliferation inhibition of HeLa cell.


Assuntos
Proliferação de Células , Isomerases de Dissulfetos de Proteínas/metabolismo , Apoptose , Ciclo Celular , Meios de Cultura/química , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Isomerases de Dissulfetos de Proteínas/genética , Soro/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...