Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
J Biol Chem ; 298(3): 101721, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35151685

RESUMO

Hypoxia-inducible factor prolyl 4-hydroxylases (HIF-P4Hs) regulate the hypoxic induction of >300 genes required for survival and adaptation under oxygen deprivation. Inhibition of HIF-P4H-2 has been shown to be protective in focal cerebral ischemia rodent models, while that of HIF-P4H-1 has no effects and inactivation of HIF-P4H-3 has adverse effects. A transmembrane prolyl 4-hydroxylase (P4H-TM) is highly expressed in the brain and contributes to the regulation of HIF, but the outcome of its inhibition on stroke is yet unknown. To study this, we subjected WT and P4htm-/- mice to permanent middle cerebral artery occlusion (pMCAO). Lack of P4H-TM had no effect on lesion size following pMCAO, but increased inflammatory microgliosis and neutrophil infiltration was observed in the P4htm-/- cortex. Furthermore, both the permeability of blood brain barrier and ultrastructure of cerebral tight junctions were compromised in P4htm-/- mice. At the molecular level, P4H-TM deficiency led to increased expression of proinflammatory genes and robust activation of protein kinases in the cortex, while expression of tight junction proteins and the neuroprotective growth factors erythropoietin and vascular endothelial growth factor was reduced. Our data provide the first evidence that P4H-TM inactivation has no protective effect on infarct size and increases inflammatory microgliosis and neutrophil infiltration in the cortex at early stage after pMCAO. When considering HIF-P4H inhibitors as potential therapeutics in stroke, the current data support that isoenzyme-selective inhibitors that do not target P4H-TM or HIF-P4H-3 would be preferred.


Assuntos
Barreira Hematoencefálica , Infarto da Artéria Cerebral Média , Doenças Neuroinflamatórias , Prolil Hidroxilases , Acidente Vascular Cerebral , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/metabolismo , Permeabilidade da Membrana Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/metabolismo , Camundongos , Doenças Neuroinflamatórias/enzimologia , Doenças Neuroinflamatórias/metabolismo , Permeabilidade , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
J Stroke Cerebrovasc Dis ; 30(3): 105165, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33360522

RESUMO

BACKGROUND: Cerebral infarction is one of the most common causes of disability and death worldwide. It is reported that electric acupuncture was able to improve the prognosis of cerebral infarction by promoting angiogenesis. However, the corresponding signal pathways of angiogenesis promotes by electric acupuncture treatment needs to be further explored. METHODS: MCAO rat was employed as the animal model, and clopidogrel hydrogen sulfate treatment was set as the positive control. Behaviors of rats, H&E staining, and TTC-staining was used to evaluate the recovery of infarcted brain tissue and nervous function. After that, immunocytochemical and immunofluorescence staining was used to quantify the angiogenesis and compensatory circulation, which including the analysis of microvessel density, field/ microvessel area ratio, and microvessel diameter. Western blot and RT-PCR for the detection of the related signal molecule, PI3K, Src, and EphB4/ephrinB2. RESULTS: The neurologic impairment scores were decreased, and the brain tissue damage that showed with H&E and TTC-staining was relieved by the treatment of electric acupuncture in MCAO rat. The quantification of microvessel density and field/ microvessel area ratio was improved obviously, and the microvessel diameter was decreased which represent the angiogenesis of capillary in day 3 and 7 by the electric acupuncture treatment. We also found that the level of Src and PI3K was increased markedly followed by the up-regulation of EphB4 and EphrinB2 mRNA during the electric acupuncture treatment, and the pre-treatment of Src and/or PI3K inhibitor was able to disturb the angiogenesis of capillary. CONCLUSIONS: We proved that electric acupuncture was able to accelerate the recovery of infarcted brain tissue and nervous function in MCAO rat by the promotion of angiogenesis, which was regulated by EphB4/EphrinB2 mediated Src/PI3K signal pathway. Our study provides a potential therapy and theoretical basis for the clinical treatment of cerebral infarction by the use of electric acupuncture.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/enzimologia , Eletroacupuntura , Efrina-B2/metabolismo , Infarto da Artéria Cerebral Média/terapia , Neovascularização Fisiológica , Fosfatidilinositol 3-Quinase/metabolismo , Receptor EphB4/metabolismo , Quinases da Família src/metabolismo , Animais , Modelos Animais de Doenças , Efrina-B2/genética , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/fisiopatologia , Densidade Microvascular , Ratos Sprague-Dawley , Receptor EphB4/genética , Recuperação de Função Fisiológica , Transdução de Sinais , Fatores de Tempo
3.
Transl Stroke Res ; 12(1): 147-163, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32221863

RESUMO

Some researchers have previously shown that RNAi knockdown of arginyl-tRNA synthetase (ArgRS) before or after a hypoxic injury can rescue animals from death, based on the model organism, C. elegans. However, there has been no study on the application of arginyl-tRNA synthetase knockdown in treating mammalian ischemic stroke, and its potential mechanism and effect on ischemic brain damage are still unknown. Here, we focused on the Rars gene, which encodes an arginyl-tRNA synthetase, and examined the effects of Rars knockdown in a permanent middle cerebral artery occlusion model in rats. To achieve this aim, adult male Sprague-Dawley (SD) rats were given right cerebral cortex injections of short hairpin RNA (shRNA) adenovirus (AV) particles to knock down arginyl-tRNA synthetase, and a non-targeting control (NTC) vector or phosphate-buffered solution served as the controls. After 4 days, the rats were exposed to permanent middle cerebral artery occlusion (pMCAO). Then, the right cerebral cortex level of arginyl-tRNA synthetase was examined, and the effects of the Rars knockdown were evaluated by differences in infarction volume, oxidative stress, blood-brain barrier, mitochondrial function, and glucose metabolism at 1 day and 3 days after MCAO. The injection of shRNA adenovirus particles successfully suppressed the expression of arginyl-tRNA synthetase in the cerebral cortex. We observed an improvement in oxidative stress, mitochondrial function, and glucose utilization and a reduction in brain edema compared with the non-targeting control rats with suppressed expression of arginyl-tRNA synthetase mRNA in the ipsilateral ischemic cortex of the brain. Our findings indicate that knockdown of arginyl-tRNA synthetase in the cerebral cortex exerted neuroprotective effects, which were achieved not only by the improvement of oxidative stress and glucose utilization but also by the maintenance of mitochondrial morphological integrity and the preservation of mitochondrial function. Knockdown of ArgRS administration could be a promising approach to protect ischemic stroke.


Assuntos
Arginina-tRNA Ligase/antagonistas & inibidores , Isquemia Encefálica/enzimologia , Córtex Cerebral/enzimologia , Técnicas de Silenciamento de Genes/métodos , Infarto da Artéria Cerebral Média/enzimologia , Animais , Arginina-tRNA Ligase/genética , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/genética , Córtex Cerebral/diagnóstico por imagem , Infarto da Artéria Cerebral Média/diagnóstico por imagem , Infarto da Artéria Cerebral Média/genética , Injeções Intraventriculares , Masculino , Ratos , Ratos Sprague-Dawley
4.
Biomed Pharmacother ; 133: 111021, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33227709

RESUMO

OBJECTIVE: To observe the brain protective effect of Leonuri Herba Total Alkali (LHA) on cerebral ischemia reperfusion injury in rats, so as to provide basis for clinical research. METHODS: Adult male SD rats were randomly assigned into sham group, middle cerebral artery occlusion/reperfusion (MCAO/R) group, and LHA + MCAO/R group (25 mg/kg, 50 mg/kg, and 100 mg/kg). Fourteen days before MCAO/R surgery, the rats in treatment groups were orally administered with LHA in ultrapure water once daily for 14 days, while rats in the sham and MCAO groups were given the same amount of saline in advance. After 1 h of administration on the 14th day, MCAO surgery was subjected. The neurological deficits, brain infarct volume, histopathology, immunofluorescence, inflammation indicators and the gene/protein expressions of BDNF-TrKB-PI3K/Akt signaling pathway in the rat brain tissue were evaluated 24 h after the MCAO/R-injury. RESULTS: It was found that rats in LHA pre-administration group showed significantly reduced neurological deficit scores, infarction volume, the serum levels of NSE and S100ß. Meanwhile, the content of Evans Blue (EB) in brain tissue from LHA group was decreased, as well as the levels of inflammatory cytokines and their gene levels. Moreover, LHA pre-administration inhibited the expression of CD44, GFAP, FOXO1 and promoted the expression of BDNF and NeuN. In addition, LHA pre-administration could up-regulate the protein expression of TrkB, p-PI3K, p-Akt, Bcl-2, and down-regulate the protein expression of Bax, and increase the level of Bcl-2/Bax. CONCLUSIONS: The study demonstrated that LHA pre-administration could regulate the PI3K/Akt pathway by increasing BDNF levels, and play a neuroprotective role in cerebral ischemia-reperfusion injury.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Leonurus/química , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor trkB/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fosforilação , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais
5.
J Stroke Cerebrovasc Dis ; 29(10): 105029, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32912542

RESUMO

BACKGROUND: We investigated whether exogenous lysophosphatidic acid (LPA), a phospholipid extracellular signaling molecule, would increase infarct size and blood-brain barrier (BBB) disruption during the early stage of cerebral ischemia-reperfusion, and whether it works through Akt-mTOR-S6K1 intracellular signaling. MATERIAL AND METHODS: Rats were given either vehicle or LPA 1 mg/kg iv three times during reperfusion after one hour of middle cerebral artery (MCA) occlusion. In another group, prior to administration of LPA, 30 mg/kg of PF-4708671, an S6K1 inhibitor, was injected. After one hour of MCA occlusion and two hours of reperfusion the transfer coefficient (Ki) of 14C-α-aminoisobutyric acid and the volume of 3H-dextran distribution were determined to measure the degree of BBB disruption. At the same time, the size of infarct was determined and western blot analysis was performed to determine the levels of phosphorylated Akt (p-Akt) and phosphorylated S6 (pS6). RESULTS: LPA increased the Ki in the ischemic-reperfused cortex (+43%) when compared with Control rats and PF-4708671 pretreatment prevented the increase of Ki by LPA. LPA increased the percentage of cortical infarct out of total cortical area (+36%) and PF-4708671 pretreatment prevented the increase of the infarct size. Exogenous LPA did not significantly change the levels of p-Akt as well as pS6 in the ischemic-reperfused cortex. CONCLUSION: Our data demonstrate that the increase in BBB disruption could be one of the reasons of the increased infarct size by LPA. S6K1 may not be the major target of LPA. A decrease of LPA during early cerebral ischemia-reperfusion might be beneficial for neuronal survival.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Infarto da Artéria Cerebral Média/terapia , Lisofosfolipídeos/toxicidade , Traumatismo por Reperfusão/induzido quimicamente , Reperfusão , Animais , Barreira Hematoencefálica/fisiopatologia , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Endogâmicos F344 , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Proteínas Quinases S6 Ribossômicas/metabolismo
6.
Eur J Pharmacol ; 883: 173294, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32681941

RESUMO

Neuroinflammation is critical for the pathogenesis of ischemia brain damage. Over-activated microglia-mediated inflammation plays a very important role in ischemia cerebral injuries. 6-Gingerol, obtained from edible ginger (Zingiber Officinale) exhibits protective effects against inflammation. In this study, we found that 6-Gingerol could reduce the size of infarction (P = 0.0184) and improve neurological functions (P = 0.04) at the third day after ischemic brain injury in vivo. Since 6-Gingerol has the anti-inflammatory effects, we further investigated its impacts on neuroinflammation mediated by microglia both in vivo and in vitro. We found that the levels of pro-inflammatory cytokines Interleukin-1 beta (IL-1ß, P = 0.0213), Interleukin-6 (IL-6, P = 0.0316), and inducible NO synthase (iNOS, P = 0.0229) in the infarct penumbra were lower in 6-Gingerol treated groups. Furthermore, microglia induced pro-inflammatory cytokines, such as IL-6, IL-1ß, incremental intercellular nitric oxide (NO), as well as iNOS were blocked by the treatment of 6-Gingerol in lipopolysaccharide (LPS) stimulated microglia. In terms of mechanism, 6-Gingerol potently suppressed phosphorylation of serine-threonine protein kinase (Akt) - mammalian target of rapamycin (mTOR) - signal transducer and activator of transcription 3 (STAT3) in LPS-treated microglia. Taken together, the present study suggested that 6-Gingerol improved cerebral ischemia injury by suppressing microglia-mediated neuroinflammation by down-regulating Akt-mTOR-STAT3 pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Catecóis/farmacologia , Álcoois Graxos/farmacologia , Infarto da Artéria Cerebral Média/prevenção & controle , AVC Isquêmico/prevenção & controle , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Mediadores da Inflamação/metabolismo , AVC Isquêmico/enzimologia , AVC Isquêmico/patologia , AVC Isquêmico/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/enzimologia , Microglia/patologia , Fosforilação , Transdução de Sinais
7.
Indian J Pharmacol ; 52(2): 108-116, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32565598

RESUMO

We investigate the protective effect of Carthamus tinctorius L. (CTL, also known as Honghua in China or Safflower) on cerebral ischemia-reperfusion and explored the possible mechanisms on regulating apoptosis and matrix metalloproteinases (MMPs). High-performance liquid chromatography method with diode array detection analysis was established to analyze the components of CTL. Middle cerebral artery occlusion rats model was established to evaluate Neurological Function Score and hematoxylin-eosin staining, as well as triphenyltetrazolium was used to examine the infarction area ratio. Transferase-mediated dUTP nick-end labeling was performed for the apoptosis. Apoptosis-related factors, including B-cell lymphoma-2 (Bcl-2), Bax and Caspase3, and MMPs-related MMP2, MMP9, tissue inhibitor of metalloproteinases 1 (TIMP1) in ischemic brain, were assayed by Western blot, reverse transcription polymerase chain reaction, and immunohistochemistry. The data showed that CTL (2, 4 g crude drug/kg/d) treatment could significantly reduce the ischemic damage in brain tissue and improve a significant neurological function score. In addition, CTL could also attenuate apoptosis degree of brain tissues and regulate Bcl-2, Bax, and Caspase 3 and also have a significant decrease on MMP-9 expression, followed by a significant increase of TIMP1 protein expression. These findings indicated that regulation of CTL on apoptosis and MMPs contributed to its protective effect on ischemia/reperfusion injury.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Carthamus tinctorius , Infarto da Artéria Cerebral Média/tratamento farmacológico , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/enzimologia , Encéfalo/patologia , Carthamus tinctorius/química , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Fármacos Neuroprotetores/isolamento & purificação , Extratos Vegetais/isolamento & purificação , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Inibidor Tecidual de Metaloproteinase-1/metabolismo
8.
Biomed Pharmacother ; 128: 110045, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32460187

RESUMO

Ischemic stroke is a major cause of death and disability worldwide. Necroptosis is known as a form of cell death, playing an essential role in regulating ischemia-induced brain injury. Triad3A is a ubiquitin ligase of the RING-in-between-RING family, and regulates necroptotic cell death under different pathological conditions, including neurodegenerative disorders. In the present study, the effects of Triad3A on experimental stroke were explored on a mouse model with middle cerebral artery occlusion (MCAO). The results indicated that Triad3A expression was markedly induced in the ischemic brain after MCAO operation. The neurons and microglia cells were the major cellular sources for Triad3A induction. Triad3A knockdown enhanced the infarction area, cell death, microglia activity, and the expression levels of pro-inflammatory markers including tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, inducible nitric oxide synthase (iNOS), CD32 and CD68 in MCAO mice. Triad3A and necroptosis were triggered in mouse microglia cells treated with oxygen and glucose deprivation (OGD), and in TNFα-incubated mouse hippocampal neuronal cells treated with Z-VAD-fmk, known as a pan-caspase inhibitor. Moreover, Triad3A knockdown accelerated cell death in microglial cells and neurons under these stresses. Furthermore, pre-treatment with necroptosis inhibitor markedly inhibited the cell death promoted by Triad3A silence in brain of mice with MCAO operation, demonstrating that Triad3A could regulate necroptosis to meditate the progression of cerebral I/R injury. Collectively, these finding illustrated that Triad3A could be served as a potential target for stroke therapy.


Assuntos
Encéfalo/enzimologia , Infarto da Artéria Cerebral Média/enzimologia , Microglia/enzimologia , Necroptose , Neurônios/enzimologia , Traumatismo por Reperfusão/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Encéfalo/patologia , Linhagem Celular , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/patologia , Neurônios/patologia , Proteínas Quinases/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética
9.
J Cardiovasc Pharmacol ; 76(2): 227-236, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32453073

RESUMO

Cerebral ischemia-reperfusion (I/R) is a kind of neurovascular disease that causes serious cerebral damage. MicroRNAs (miRNAs) have been widely reported to participate in multiple diseases, including cerebral I/R injury. However, the exact mechanisms of miR-7-5p in cerebral I/R injury was not fully elucidated. In this study, we explored the biological role and regulatory mechanism of miR-7-5p in cerebral I/R injury. We established an in vivo model of cerebral I/R by middle cerebral artery occlusion and an in vitro cellular model of cerebral I/R injury through treating neurons (SH-SY5Y cells) with oxygen-glucose deprivation (OGD). In addition, miR-7-5p expression was confirmed to be upregulated in the cerebral I/R rat model and OGD/R-treated SH-SY5Y cells. Moreover, miR-7-5p inhibition overtly suppressed cerebral injury, cerebral inflammation, and SH-SY5Y cells apoptosis. Sirtuin 1 (sirt1) is previously reported to alleviate I/R, and in this study, it was identified to be a target of miR-7-5p based on luciferase reporter assay. Reverse transcription-quantitative polymerase chain reaction revealed sirt1 expression was downregulated in the cerebral I/R rat model and OGD/R-treated SH-SY5Y cells. Besides, miR-7-5p negatively regulated sirt1. Finally, rescue assays delineated sirt1 overexpression recovered the miR-7-5p upregulation-induced promotion on cerebral I/R injury. In conclusion, miR-7-5p enhanced cerebral I/R injury by degrading sirt1, providing a new paradigm to investigate cerebral I/R injury.


Assuntos
Encéfalo/enzimologia , Infarto da Artéria Cerebral Média/enzimologia , MicroRNAs/metabolismo , Neurônios/enzimologia , Estabilidade de RNA , RNA Mensageiro/metabolismo , Traumatismo por Reperfusão/enzimologia , Sirtuína 1/metabolismo , Animais , Apoptose , Encéfalo/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Glucose/deficiência , Humanos , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , MicroRNAs/genética , NF-kappa B/metabolismo , Neurônios/patologia , RNA Mensageiro/genética , Ratos Wistar , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Sirtuína 1/genética
10.
J Stroke Cerebrovasc Dis ; 29(6): 104801, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32249206

RESUMO

BACKGROUND: Ischemic stroke is the leading cause of disability and death globally. Micro-RNAs (miRNAs) have been reported to play important roles in the development and pathogenesis of the nervous system. However, the exact function and mechanism of miRNAs have not been fully elucidated about brain damage caused by cerebral ischemia/reperfusion (I/R). METHODS: In this study, we explored the neuroprotective effects of miR-219a-5p on brain using an in vitro ischemia model (mouse neuroblastoma N2a cells treated with oxyglucose deprivation and reperfusion), and in vivo cerebral I/R model in mice. Western blot assay and Reverse Transcription-Polymerase Chain Reaction were used to check the expression of molecules involved. Flow cytometry and cholecystokinin were used to examine cell apoptosis, respectively. RESULTS: Our research shows that miR-219a-5p gradually decreases in cerebral I/R models in vivo and in vitro. In vitro I/R, we find that miR-219a-5p mimics provided evidently protection for cerebral I/R damage, as shown by increased cell viability and decreased the release of LDH and cell apoptosis. Mechanically, our findings indicate that miR-219a-5p binds to cAMP specific 3', 5'-cyclic phosphodiesterase 4D (PDE4D) mRNA in the 3'-UTR region, which subsequently leads to a decrease in Pde4d expression in I/R N2a cells. CONCLUSIONS: Our results provide new ideas for the study of the mechanism of cerebral ischemia/reperfusion injury, and lay the foundation for further research on the treatment of brain I/R injury. Upregulation of miR-219a-5p decreases cerebral ischemia/reperfusion injury by targeting Pde4d in vitro.


Assuntos
Apoptose , Encéfalo/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Infarto da Artéria Cerebral Média/enzimologia , MicroRNAs/metabolismo , Neurônios/enzimologia , Traumatismo por Reperfusão/enzimologia , Regiões 3' não Traduzidas , Animais , Sítios de Ligação , Encéfalo/patologia , Linhagem Celular Tumoral , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Neurônios/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais
11.
J Stroke Cerebrovasc Dis ; 29(5): 104743, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32127256

RESUMO

BACKGROUND: Bone marrow stromal cell (BMSC) transplantation is a promising therapeutic approach for cerebral ischemia, as it elicits multiple neuroprotective effects. However, it remains unclear how BMSC transplantation modulates the ubiquitin-proteasome system (UPS) and autophagy under cerebral ischemia. METHODS: In the present study, an intermediate level of cerebral ischemia (30 minutes) was chosen to examine the effect of BMSC transplantation on the molecular switch regulating UPS and autophagy. BMSC or vehicle was stereotactically injected into the penumbra 15 minutes after sham operation or transient middle cerebral artery occlusion (tMCAO). RESULTS: Thirty minutes of tMCAO artery occlusion significantly increased TUNEL-, ubiquitin-, and p62-positive cells (which peaked at 72 hours, 2 hours, and 2 hours after reperfusion, respectively) and ratios of both BAG3/BAG1 and LC3-II/LC3-I at 24 hours after reperfusion. However, intracerebral injection of BMSCs significantly reduced infarct volume and numbers of TUNEL- and p62-positive cells, and improved BAG3/BAG1 and LC3-II/LC3-I ratios. In addition, observed increases in ubiquitin-positive cells 2 hours after reperfusion were slightly suppressed by BMSC transplantation. CONCLUSIONS: These data suggest a protective role of BMSC transplantation, which drove the molecular switch from autophagy to UPS in a murine model of ischemic stroke.


Assuntos
Autofagia , Encéfalo/enzimologia , Infarto da Artéria Cerebral Média/cirurgia , Transplante de Células-Tronco Mesenquimais , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/patologia , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Sequestossoma-1/metabolismo , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/metabolismo , Ubiquitinação
12.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1753-1760, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31900521

RESUMO

Isorhapontigenin (ISO) is one of the main bioactive components of Gnetum cleistostachyum and was shown to possess antioxidant and antitumor functions. Herein, we hope to examine the neuroprotection impacts of ISO in rats subjected to transient middle cerebral artery occlusion/reperfusion (MCAO/R, 2/24 h) injuries. ISO was injected intraperitoneally into the rats immediately after cerebral ischemia. After 24 h of the reperfusion, infarct volume, brain water contents, neurological deficit, and cerebral blood flow were assessed. Hippocampus histopathology change was detected by H&E and TUNEL staining. The expressions of cleaved caspase-3, Bax and Bcl-2, and phospho-Akt (p-Akt) were investigated by real-time RT-PCR or western blot analysis. We found that ISO significantly suppressed the infarct volumes, brain water contents, and neurological deficit, increased CBF, and relieved histopathologic change in a dose-dependent manner. Reduced malondialdehyde (MDA) and elevated activities of superoxide dismutase (SOD) and GSH and glutathione peroxidase (GSH-PX) were observed in ISO group. ISO remarkably decreased caspase-3 and Bax and increased levels of Bcl-2. Additionally, ISO upregulated p-Akt expression. Blocking of PI3K activities by wortmannin can abolish the ISO-caused decrease in infarct volumes and neurologic deficit scores and abrogate the promotion of p-Akt. The data indicated that ISO played neuroprotective impacts against focal I/R injuries, possibly related to the activating of PI3K/Akt signaling.


Assuntos
Hipocampo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Estilbenos/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Modelos Animais de Doenças , Hipocampo/enzimologia , Hipocampo/patologia , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais
13.
J Stroke Cerebrovasc Dis ; 28(7): 1832-1840, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31078389

RESUMO

GOAL: The present study aimed to examine whether Am80 (tamibarotene) protects the hippocampus against cerebral ischemia-reperfusion (I/R) injury and whether phosphoinositide-3-kinase/Akt (PI3K/Akt) pathway mediates this effect. MATERIALS AND METHODS: Rats were subjected to 90 minutes of middle cerebral artery occlusion followed by 24 hours of reperfusion. The animals were randomly divided into 7 groups: sham-operated group; I/R group; groups pretreated with 2 mg/kg, 6 mg/kg, and 10 mg/kg of Am80; Am80 (6 mg/kg) combined with the selective PI3K inhibitor wortmannin (0.6 mg/kg), and wortmannin (0.6 mg/kg) only group. After 24 hours of reperfusion, neurological deficits and infarct volume were measured. Pathological changes in hippocampal neurons were analyzed by transmission electron microscopy. Neuronal survival was examined by TUNEL staining. The expression of Bcl-2, Bax, and Akt, and Akt phosphorylation (p-Akt) were measured by Western blotting and quantitative real-time polymerase chain reaction. FINDINGS: The pretreatment with Am80 improved the neurologic deficit score, reduced infarct volume, and decreased the number of TUNEL-positive cells in the hippocampus. Moreover, Am80 pretreatment downregulated the expression of Bax, upregulated the expression of Bcl-2, and increased the level of p-Akt. Wortmannin abolished in part the increase in p-Act and the neuroprotective effect exerted on the ischemic by Am80 pretreatment. CONCLUSIONS: Our results documented that Am80 pretreatment protects ischemic hippocampus after cerebral I/R by regulating the expression of apoptosis-related proteins through the activation of the PI3K/Akt signaling pathway.


Assuntos
Benzoatos/farmacologia , Hipocampo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/prevenção & controle , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Tetra-Hidronaftalenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/enzimologia , Hipocampo/ultraestrutura , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Neurônios/enzimologia , Neurônios/ultraestrutura , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
14.
J Cell Physiol ; 234(5): 7090-7103, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30552827

RESUMO

Ischemic stroke is a refractory disease caused by cerebral ischemic injury, which results in brain dysfunction. This study intends to investigate the effects of microRNA-212 (miR-212) on the recovery function and vascular regeneration of endothelial progenitor cells (EPCs) by inactivation of the Notch signaling pathway by binding to matrix metallopeptidase 9 (MMP9) in mice with ischemic stroke. According to the results of database retrieval systems and data analysis, MMP9 was predicted as a gene related to ischemic stroke and miR-212 is a potential regulating mRNA of MMP9. All 72 healthy adult C57BL6 mice were selected for middle cerebral artery occlusion (MCAO) establishment. Cerebral infarction was observed under triphenyltetrazolium chloride staining. A series of inhibitors, activators, and siRNAs were introduced to the verified regulatory functions for miR-212 governing MMP9 in ischemic stroke. Cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and tube-forming ability by tubule formation test. Reverse transcription quantitative polymerase chain reaction and Western blot analysis were used to detect the expressions of miR-212, MMP9, Hes-1, and Notch-1. The corresponding results demonstrated that the area of cerebral infarction and the number of neuronal necrosis increased in the MCAO group in contrast to the sham group. Meanwhile, upregulation of miR-212 or downregulation of MMP9 decreases the expressions of MMP9, Hes-1 Notch-1, increases cell proliferation and tube-forming ability and improves the pathological conditions of EPCs. Our study suggests that miR-212 promotes recovery function and vascular regeneration of EPCs through negative regulation of the Notch signaling pathway via downregulating expression of MMP9, thus provides a clinical theoretical basis for ischemic stroke therapy.


Assuntos
Encéfalo/irrigação sanguínea , Proliferação de Células , Células Progenitoras Endoteliais/enzimologia , Infarto da Artéria Cerebral Média/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/metabolismo , Neovascularização Fisiológica , Receptor Notch1/metabolismo , Animais , Estudos de Casos e Controles , Células Cultivadas , Bases de Dados Genéticas , Modelos Animais de Doenças , Células Progenitoras Endoteliais/patologia , Humanos , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Receptor Notch1/genética , Transdução de Sinais , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
15.
J Stroke Cerebrovasc Dis ; 28(3): 792-799, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30552029

RESUMO

OBJECTIVE: To determine the mechanism(s) involved in electroacupuncture (EA)-mediated improvements in synaptic plasticity in a rat model of middle cerebral artery occlusion and reperfusion (MCAO/R)-induced cognitive deficits. METHODS: Focal cerebral ischemic stroke was induced by (MCAO/R) surgery. Rats were randomly split into 4 groups: control group (sham operation control), MCAO group, Baihui (GV 20) and Shenting (GV 24) acupoint EA group (verum acupuncture, MCAO + VA), and nonacupoint EA group (control acupuncture, MCAO + CA). EA treatment was administered for 14 consecutive days in MCAO + VA and MCAO + CA groups. Neurological assessment, behavioral performance testing, and molecular biology assays were used to evaluate the MCAO/R model, EA therapeutic effect and potential therapeutic mechanism(s) of EA. RESULTS: Significant amelioration of neurological deficits was found in MCAO + VA rats compared with MCAO rats (P < .01). Moreover, learning and memory significantly improved in EA-treated rats compared with MCAO or MCAO + CA rats (P < .05) together with an increase in the number of PSD-95+ and SYN+ cells and synapses in the hippocampal CA1 region (P < .05). MCAO + VA rats also showed amelioration of pathological synaptic ultrastructural changes compared with MCAO or MCAO + CA groups (P < .001). In contrast, EA decreased the levels and phosphorylation of JAK2 (Janus-activated kinase 2) and STAT3 (signal transducer and activator of transcription 3) in the hippocampal CA1 region compared with MCAO or MCAO + CA group (P < .01). CONCLUSION: EA at GV 20 and GV 24 acupoints improved cognitive deficits in cerebral ischemic rats via the JAK2/STAT3 signaling pathway and mediated synaptic plasticity in the peri-infarct hippocampal CA1 region of rats following ischemic stroke.


Assuntos
Região CA1 Hipocampal/enzimologia , Eletroacupuntura/métodos , Infarto da Artéria Cerebral Média/terapia , Janus Quinase 2/metabolismo , Plasticidade Neuronal , Fator de Transcrição STAT3/metabolismo , Pontos de Acupuntura , Animais , Comportamento Animal , Região CA1 Hipocampal/fisiopatologia , Cognição , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/psicologia , Masculino , Memória , Fosforilação , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Transdução de Sinais
16.
J Stroke Cerebrovasc Dis ; 27(11): 3053-3065, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30093209

RESUMO

BACKGROUND: The present study aimed to examine the protective role of fullerenol nanoparticles against blood-brain barrier (BBB) interruption and brain edema during cerebral ischemia-reperfusion injury probably by reduction of interleukin-6 (IL-6) and matrix metalloproteinase-9 (MMP-9) transcription. METHODS: The male Wistar rats (weighting 280-320 g) were randomly assigned into four groups as follows: sham, control ischemic, pretreated ischemic, and posttreated ischemic groups. Cerebral ischemia-reperfusion (IR) injury was performed by occlusion of middle cerebral artery (MCA) for 90 minutes followed by twenty-four hours reperfusion. Rats were administered fullerenol 5mg/kg, intraperitoneally, 30 minutes before induction of IR in pretreated ischemic group and immediately after termination of MCA occlusion in posttreated ischemic group. After twenty-four hours reperfusion, the method of Evans blue dye extravasation (EBE) and RT-PCR were used for determination of BBB permeability and mRNA expression levels of MMP-9 and IL-6, respectively. Neuronal deficit score (NDS) and edema of the ischemic hemispheres were also evaluated. RESULTS: MCA occlusion increased NDS in control ischemic rats (3.16 ± 0.16) with concomitant increase in EBE (15.30 ± 3.98µg/g) and edema (3.53 ± 0.50%). Fullerenol in both pretreated and posttreated ischemic groups reduced NDS (36% and 68%, respectively), EBE (89% and 91%, respectively) and edema (53% and 81%, respectively). Although MCA occlusion increased the mRNA expression levels of MMP-9 and IL-6 in ischemic hemispheres, fullerenol in both treatment groups noticeably decreased the mRNA expression levels of these genes. CONCLUSION: In conclusion, fullerenol nanoparticles can protect BBB integrity and attenuate brain edema after cerebral ischemia-reperfusion injury possibly by reduction of IL-6 and MMP-9 transcription.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Edema Encefálico/prevenção & controle , Fulerenos/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Interleucina-6/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Nanopartículas , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Transcrição Gênica/efeitos dos fármacos , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Edema Encefálico/enzimologia , Edema Encefálico/genética , Edema Encefálico/patologia , Permeabilidade Capilar/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Interleucina-6/genética , Masculino , Metaloproteinase 9 da Matriz/genética , Ratos Wistar , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
17.
Biomed Pharmacother ; 106: 805-812, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29990874

RESUMO

(3ß,5α,16α,20S)-4,4,14-trimethyl-3,20-bis(methylamino)-9,19-cyclopregnan-16-ol-dihydrochloride (JLX001), a derivative of cyclovirobuxine D (CVB-D), is a novel compound from synthesis. This study aims to confirm the therapeutic effect of JLX001 on cerebral ischemia and researchits antiplatelet and antithrombosis activities via thromboxane (TXA2)/phospholipase C-ß-3(PLCß3)/protein kinase C (PKC) pathway suppression. The therapeutic effects of JLX001 was evaluated by infarct sizes, brain edema and neurological scores in Sprague-Dawley (SD) rats with middle cerebral artery occlusion (MCAO). Brain TXA2 and prostacyclin (PGI2) were measured by enzyme-linked immunosorbentassay (ELISA). P-PLCß3and activated PKC were detected by immunohistochemical method. Adenosine diphosphate (ADP) or 9, 11-dieoxy-11α, 9α-epoxymethanoeprostaglandin F2α (U46619) was used as platelet agonist in the in vivo and in vitro platelet aggregation experiments. Clotting time and bleeding time were determined. Besides, two whole-animal experiments including arteriovenous shunt thrombosis and pulmonary thromboembolism model were conducted. Results showed that JLX001 treatment markedly alleviated cerebral infarcts, edema, and neurological scores in permanent middle cerebral artery occlusion (pMCAO) rats. Brain TXA2 level, p-PLCß3and activated PKC were decreased, while PGI2level had no significant change. Besides, JLX001 inhibited platelet aggregation induced by ADP or U46619 and exhibited anti-coagulation effects with a minor bleeding risk. In the two whole-animal experiments, JLX001 inhibited thrombus formation. In summary, JLX001 attenuates cerebral ischemia injury and the underlying mechanisms relate to inhibiting platelet activation and thrombus formation via TXA2/PLCß3/PKC pathway suppression.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/prevenção & controle , Trombose Intracraniana/prevenção & controle , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Triterpenos/farmacologia , Animais , Aspirina/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Edema Encefálico/sangue , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/farmacologia , Epoprostenol/metabolismo , Feminino , Infarto da Artéria Cerebral Média/sangue , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Trombose Intracraniana/sangue , Trombose Intracraniana/enzimologia , Trombose Intracraniana/patologia , Masculino , Camundongos Endogâmicos ICR , Fosfolipase C beta/metabolismo , Inibidores da Agregação Plaquetária/uso terapêutico , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tromboxano A2/metabolismo , Triterpenos/uso terapêutico
18.
Cardiovasc Diabetol ; 17(1): 60, 2018 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-29776406

RESUMO

BACKGROUND: Dipeptidyl peptidase-4 (DPP-4) inhibitors (gliptins) are approved drugs for the treatment of hyperglycemia in patients with type 2 diabetes. These effects are mainly mediated by inhibiting endogenous glucagon-like peptide-1 (GLP-1) cleavage. Interestingly, gliptins can also improve stroke outcome in rodents independently from GLP1. However, the underlying mechanisms are unknown. Stromal cell-derived factor-1α (SDF-1α) is a DPP-4 substrate and CXCR4 agonist promoting beneficial effects in injured brains. However, SDF-1α involvement in gliptin-mediated neuroprotection after ischemic injury is unproven. We aimed to determine whether the gliptin linagliptin improves stroke outcome via the SDF-1α/CXCR4 pathway, and identify additional effectors behind the efficacy. METHODS: Mice were subjected to stroke by transient middle cerebral artery occlusion (MCAO). linagliptin was administered for 3 days or 3 weeks from stroke onset. The CXCR4-antagonist AMD3100 was administered 1 day before MCAO until 3 days thereafter. Stroke outcome was assessed by measuring upper-limb function, infarct volume and neuronal survival. The plasma and brain levels of active GLP-1, GIP and SDF-1α were quantified by ELISA. To identify additional gliptin-mediated molecular effectors, brain samples were analyzed by mass spectrometry. RESULTS: Linagliptin specifically increased active SDF-1α but not glucose-dependent insulinotropic peptide (GIP) or GLP-1 brain levels. Blocking of SDF-1α/CXCR4 pathway abolished the positive effects of linagliptin on upper-limb function and histological outcome after stroke. Moreover, linagliptin treatment after stroke decreased the presence of peptides derived from neurogranin and from an isoform of the myelin basic protein. CONCLUSIONS: We showed that linagliptin improves functional stroke outcome in a SDF-1α/CXCR4-dependent manner. Considering that Calpain activity and intracellular Ca2+ regulate neurogranin and myelin basic protein detection, our data suggest a gliptin-mediated neuroprotective mechanism via the SDF-1α/CXCR4 pathway that could involve the regulation of Ca2+ homeostasis and the reduction of Calpain activity. These results provide new insights into restorative gliptin-mediated effects against stroke.


Assuntos
Encéfalo/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Linagliptina/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores CXCR4/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Proteína Básica da Mielina/metabolismo , Recuperação de Função Fisiológica , Proteínas Repressoras/metabolismo
19.
Sci Rep ; 8(1): 2459, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29410497

RESUMO

Thalidomide was originally used as a sedative and found to be a teratogen, but now thalidomide and its derivatives are widely used to treat haematologic malignancies. Accumulated evidence suggests that thalidomide suppresses nerve cell death in neurologic model mice. However, detailed molecular mechanisms are unknown. Here we examined the molecular mechanism of thalidomide's neuroprotective effects, focusing on its target protein, cereblon (CRBN), and its binding protein, AMP-activated protein kinase (AMPK), which plays an important role in maintaining intracellular energy homeostasis in the brain. We used a cerebral ischemia rat model of middle cerebral artery occlusion/reperfusion (MCAO/R). Thalidomide treatment significantly decreased the infarct volume and neurological deficits of MCAO/R rats. AMPK was the key signalling protein in this mechanism. Furthermore, we considered that the AMPK-CRBN interaction was altered when neuroprotective action by thalidomide occurred in cells under ischemic conditions. Binding was strong between AMPK and CRBN in normal SH-SY5Y cells, but was weakened by the addition of H2O2. However, when thalidomide was administered at the same time as H2O2, the binding of AMPK and CRBN was partly restored. These results suggest that thalidomide inhibits the activity of AMPK via CRBN under oxidative stress and suppresses nerve cell death.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Proteases Dependentes de ATP/genética , Isquemia Encefálica/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Talidomida/farmacologia , Complexos Ubiquitina-Proteína Ligase/genética , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Proteases Dependentes de ATP/antagonistas & inibidores , Proteases Dependentes de ATP/metabolismo , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Regulação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/farmacologia , Imunossupressores/farmacologia , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Complexos Ubiquitina-Proteína Ligase/metabolismo
20.
J Neurosci ; 37(50): 12123-12140, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29114077

RESUMO

Stroke is one of the leading causes of morbidity and mortality worldwide. Inflammation, oxidative stress, apoptosis, and excitotoxicity contribute to neuronal death during ischemic stroke; however, the mechanisms underlying these complicated pathophysiological processes remain to be fully elucidated. Here, we found that the expression of tumor necrosis factor receptor-associated factor 6 (TRAF6) was markedly increased after cerebral ischemia/reperfusion (I/R) in mice. TRAF6 ablation in male mice decreased the infarct volume and neurological deficit scores and decreased proinflammatory signaling, oxidative stress, and neuronal death after cerebral I/R, whereas transgenic overexpression of TRAF6 in male mice exhibited the opposite effects. Mechanistically, we demonstrated that TRAF6 induced Rac1 activation and consequently promoted I/R injury by directly binding and ubiquitinating Rac1. Either functionally mutating the TRAF6 ubiquitination site on Rac1 or inactivating Rac1 with a specific inhibitor reversed the deleterious effects of TRAF6 overexpression during I/R injury. In conclusion, our study demonstrated that TRAF6 is a key promoter of ischemic signaling cascades and neuronal death after cerebral I/R injury. Therefore, the TRAF6/Rac1 pathway might be a promising target to attenuate cerebral I/R injury.SIGNIFICANCE STATEMENT Stroke is one of the most severe and devastating neurological diseases globally. The complicated pathophysiological processes restrict the translation of potential therapeutic targets into medicine. Further elucidating the molecular mechanisms underlying cerebral ischemia/reperfusion injury may open a new window for pharmacological interventions to promote recovery from stroke. Our study revealed that ischemia-induced tumor necrosis factor receptor-associated factor 6 (TRAF6) upregulation binds and ubiquitinates Rac1 directly, which promotes neuron death through neuroinflammation and neuro-oxidative signals. Therefore, precisely targeting the TRAF6-Rac1 axis may provide a novel therapeutic strategy for stroke recovery.


Assuntos
Infarto da Artéria Cerebral Média/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Traumatismo por Reperfusão/enzimologia , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Estresse Oxidativo , RNA Interferente Pequeno/farmacologia , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/genética , Transfecção , Ubiquitinação , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA