Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 545: 158-170, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38513765

RESUMO

Thioredoxin-reductase 2 (Txnrd2) belongs to the thioredoxin-reductase family of selenoproteins and is a key antioxidant enzyme in mammalian cells to regulate redox homeostasis. Here, we reported that Txnrd2 exerted a major influence in brain damage caused by Intracerebral hemorrhage (ICH) by suppressing endoplasmic reticulum (ER) stress oxidative stress and via Trx2/Prx3 pathway. Furthermore, we demonstrated that pharmacological selenium (Se) rescued the brain damage after ICH by enhancing Txnrd2 expression. Primarily, expression and localization of Txnrd2, Trx2 and Prx3 were determined in collagenase IV-induced ICH model. Txnrd2 was then knocked down using siRNA interference in rats which were found to develop more severe encephaledema and neurological deficits. Mechanistically, we observed that loss of Txnrd2 leads to increased lipid peroxidation levels and ER stress protein expression in neurons and astrocytes. Additionally, it was revealed that Se effectively restored the expression of Txnrd2 in brain and inhibited both the activity of ER stress protein activity and the generation of reactive oxygen species (ROS) by promoting Trx2/Prx3 kilter when administrating sodium selenite in lateral ventricle. This study shed light on the effect of Txnrd2 in regulating oxidative stress and ER stress via Trx2/Prx3 pathway upon ICH and its promising potential as an ICH therapeutic target.


Assuntos
Hemorragia Cerebral , Estresse do Retículo Endoplasmático , Estresse Oxidativo , Ratos Sprague-Dawley , Tiorredoxina Redutase 2 , Tiorredoxinas , Animais , Masculino , Ratos , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/fisiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Peroxirredoxina III/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Selênio/farmacologia , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tiorredoxina Redutase 2/metabolismo , Tiorredoxinas/metabolismo
2.
Patterns (N Y) ; 4(3): 100702, 2023 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-36960450

RESUMO

The accurate identification of anticancer peptides (ACPs) and antimicrobial peptides (AMPs) remains a computational challenge. We propose a tri-fusion neural network termed TriNet for the accurate prediction of both ACPs and AMPs. The framework first defines three kinds of features to capture the peptide information contained in serial fingerprints, sequence evolutions, and physicochemical properties, which are then fed into three parallel modules: a convolutional neural network module enhanced by channel attention, a bidirectional long short-term memory module, and an encoder module for training and final classification. To achieve a better training effect, TriNet is trained via a training approach using iterative interactions between the samples in the training and validation datasets. TriNet is tested on multiple challenging ACP and AMP datasets and exhibits significant improvements over various state-of-the-art methods. The web server and source code of TriNet are respectively available at http://liulab.top/TriNet/server and https://github.com/wanyunzh/TriNet.

3.
Free Radic Biol Med ; 199: 67-80, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36805044

RESUMO

Intracerebral hemorrhage (ICH) induces high mortality and disability. Neuronal death is the principal factor to unfavourable prognosis in ICH. However, the mechanisms underlying this association remain unclear. In this study, we investigated the molecular mechanisms by which neuronal ferroptosis occurs after ICH and whether the use of corresponding modulators can inhibit neuronal death and improve early outcomes in a rat ICH model. Our findings indicated that Nox4 and TF/TfR were upregulated in the perihematomal tissues of ICH rats. Oxidative stress and iron overload induced by Nox4 and TF/TfR promoted neuronal ferroptosis post-ICH. In contrast, application of Nox4-siRNA and the deferoxamine (DFO) attenuated peroxidation and iron deposition in the hemorrhagic brain, alleviated neuronal ferroptosis, and improved sensorimotor function in ICH rats. Additionally, our findings indicated that the post-ICH neuronal reduced glutathione (GSH) depletion were not related to dysfunctional glutamine delivery in astrocytes but rather to downregulation of EAAT3 due to lipid peroxidation-induced dysfunction in the neuronal membrane. These findings indicate that ferroptosis is involved in neuronal death in model rats with collagenase-induced ICH. Oxidative stress and iron overload induced by Nox4 and TF/TfR exacerbate ferroptosis after ICH, while Nox4 downregulation and iron chelation exert neuroprotective effects. The present results highlight the cysteine importer EAAT3 as a potential biomarker of ferroptosis and provide insight into the neuronal death process that occurs following ICH, which may aid in the development of translational treatment strategies for ICH.


Assuntos
Ferroptose , Sobrecarga de Ferro , Animais , Ratos , Morte Celular , Hemorragia Cerebral/genética , Ferroptose/genética , Sobrecarga de Ferro/genética , NADPH Oxidase 4/genética , Estresse Oxidativo/fisiologia
4.
Neural Regen Res ; 18(8): 1734-1742, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36751799

RESUMO

Intracerebral hemorrhage is often accompanied by oxidative stress induced by reactive oxygen species, which causes abnormal mitochondrial function and secondary reactive oxygen species generation. This creates a vicious cycle leading to reactive oxygen species accumulation, resulting in progression of the pathological process. Therefore, breaking the cycle to inhibit reactive oxygen species accumulation is critical for reducing neuronal death after intracerebral hemorrhage. Our previous study found that increased expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NADPH oxidase 4, NOX4) led to neuronal apoptosis and damage to the blood-brain barrier after intracerebral hemorrhage. The purpose of this study was to investigate the role of NOX4 in the circle involving the neuronal tolerance to oxidative stress, mitochondrial reactive oxygen species and modes of neuronal death other than apoptosis after intracerebral hemorrhage. We found that NOX4 knockdown by adeno-associated virus (AAV-NOX4) in rats enhanced neuronal tolerance to oxidative stress, enabling them to better resist the oxidative stress caused by intracerebral hemorrhage. Knockdown of NOX4 also reduced the production of reactive oxygen species in the mitochondria, relieved mitochondrial damage, prevented secondary reactive oxygen species accumulation, reduced neuronal pyroptosis and contributed to relieving secondary brain injury after intracerebral hemorrhage in rats. Finally, we used a mitochondria-targeted superoxide dismutase mimetic to explore the relationship between reactive oxygen species and NOX4. The mitochondria-targeted superoxide dismutase mimetic inhibited the expression of NOX4 and neuronal pyroptosis, which is similar to the effect of AAV-NOX4. This indicates that NOX4 is likely to be an important target for inhibiting mitochondrial reactive oxygen species production, and NOX4 inhibitors can be used to alleviate oxidative stress response induced by intracerebral hemorrhage.

5.
Neuroscience ; 492: 1-17, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35405301

RESUMO

Toll-like receptor-4 (TLR4), a member of the TLR family, plays a key role in inflammation-related diseases of the nervous system. TLR4 knockout mice are widely used in various neurological disease studies, and there is a clear correlation between inflammation and behavior. Therefore, elucidating the effect of TLR4 on neurobehavioral function is essential, and the related mechanisms need to be explored. Male TLR4 knockout (TLR4-/-) and wild-type (TLR4+/+) mice of different ages (4, 8, and 16 months) were used for behavioral experiments. Synaptic spine, blood-brain barrier (BBB) integrity, memory regulatory proteins, cortical blood flow, and inflammatory factor examinations were also conducted to explore the possible mechanism by which TLR4 works. Here, we found that compared with 16-m-old TLR4+/+ mice, age-matched TLR4-/- mice had better learning and memory abilities, increased expression of neuronal synaptic spines, and increased memory-related regulatory proteins in the hippocampus. TLR4 knockout significantly attenuated the fear response in 16-m-old mice. The TLR4-/- mice also had better blood-brain barrier integrity, increased expression of tight junction-associated proteins, increased cerebral cortical blood flow and reduced proinflammatory cytokine expression in the cortex and cerebrospinal fluid. Our results suggest that TLR4 deletion ameliorates significant neurobehavioral dysfunction during the aging stage, as well as multiple abnormalities in brain function and structure due to alterations in tight junction-associated proteins and inflammatory factors.


Assuntos
Encéfalo , Receptor 4 Toll-Like , Animais , Encéfalo/metabolismo , Cognição , Deleção de Genes , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Junções Íntimas/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
6.
Front Oncol ; 11: 644492, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34221965

RESUMO

BACKGROUND: P7C3 is a neurogenic compound that exhibits neuroprotective properties in neural cells. However, its target proteins and effects in glioma are unknown. METHODS: The candidate P7C3 target proteins were analyzed using a human protein microarray containing 23136 human proteins. A streptavidin agarose affinity assay was used to verify the direct interaction between P7C3 and phosphoglycerate kinase 1 (PGK1). Mass spectrometry was used to identify the binding sites of PGK1 for P7C3 binding. Seahorse XF96 extracellular flux analyzer was used to measure the cell oxygen consumption rate and extracellular acidification rate. Glycolytic metabolites were measured using the related kits. Protein level was detected by western blotting and immunohistochemical staining. Autophagy was analyzed using a transmission electron microscope and western blotting. The malignancy of tumor progression in vitro and in vivo was analyzed based on cell viability, apoptosis and proliferation, migration and invasion, and xenograft model. Glial cells were marked by antibodies via immunohistochemical staining. RESULTS: The human protein microarray identified 577 candidate P7C3 target proteins. The global profile of P7C3 target proteins indicated that P7C3 regulates glycolysis. Metabolic experiments confirmed that P7C3 regulates aerobic glycolysis in glioma cells. The underlying mechanism of P7C3 was found to be direct targeting PGK1 at lysine residues and asparagine residues, and the specific P7C3-PGK1 interaction led to decreased protein level and total intracellular kinase activity of PGK1. The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases indicated that the mRNA level of PGK1 is significantly increased in high-grade glioma, and the abnormally high mRNA level of PGK1 is associated with a poor prognosis in patients with glioma, suggesting that PGK1 is a promising target for glioma therapy. The inhibition of PGK1 and the subsequent suppression of aerobic glycolysis caused by P7C3 inhibited the malignant growth of glioma in vitro and in vivo. Furthermore, P7C3 did not damage normal glial cells under concentration, which exhibit an inhibitory effect on gliomas. CONCLUSIONS: This study revealed that P7C3 suppresses glioma by regulating aerobic glycolysis via directly targeting PGK1. Furthermore, we identified the P7C3 target proteins for the first time which is expected to provide scientific clues for future studies.

7.
Am J Cancer Res ; 11(4): 1369-1390, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33948363

RESUMO

Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a new-found tumor suppressor in a variety of tumors. While, it is still unknown about its role in glioma. In this study, we found that LHPP is abnormally decreasing or absent in glioblastoma, and the low expression of LHPP is associated with poor median survival in glioma patients. Functional assay revealed that LHPP-overexpression significantly inhibited U87MG and U118MG growth in vitro and in vivo. As to the mechanism, mass-spectrometric analysis indicated that the LHPP interacting proteins were mainly enriched in regulation of energy metabolism, including Carbon metabolism, Oxidative phosphorylation, and Glycolysis. Seahorse assay and metabolites detection confirmed that LHPP-overexpression obviously impeded glycolysis and respiration in U87MG and U118MG cells. For the further study, western blot assay showed that the protein level of PKM2 at dimeric, tetrameric, and total protein, were all decreased significantly, and its enzymatic activity was decreased as well. ChIP and RNAseq integrated analysis indicated that the decreased protein level of PKM2 was independent of PKM2 transcription, and LHPP did not reprogram transcription level of metabolic genome. Co-IP and immunofluorescence assay manifested that LHPP interacted with PKM2, and this interaction interfered the protein stability, then induced ubiquitin-mediated degradation of PKM2. Rescue assay confirmed that restoring the expression of PKM2 effectively reversed the restrained energy metabolism and the inhibited cancer cell growth caused by LHPP-overexpression in U87MG and U118MG cells. Taking together, we demonstrated that LHPP impedes the glycolysis and respiration during energy metabolic process via inducing ubiquitin-mediated degradation of PKM2, thus inhibits the growth of glioblastoma.

8.
Stem Cell Res Ther ; 12(1): 128, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579360

RESUMO

BACKGROUND: Recently, growing evidence has indicated an important role of the complement system, a crucial component of immunity, in mediating neuroinflammation and promoting neuronal apoptosis following closed head injury (CHI). We previously reported that transplanted induced neural stem cells (iNSCs) pre-treated with CHI mouse serum could enhance complement receptor type 1-related protein y (Crry) expression and ameliorate complement-mediated damage in mouse CHI models. However, the mechanism underlying the elevated levels of Crry expression remains elusive. METHODS: CHI models were established using a standardized weight-drop device. We collected CHI mouse serum at 12 h post-trauma. RT-QPCR assay, western blot analysis, complement deposition assay, Akt inhibition assay, flow cytometry, cell transplantation, and functional assay were utilized to clarify the mechanism of Crry expression in iNSCs receiving CHI mouse serum treatment. RESULTS: We observed dramatic increases in the levels of Crry expression and Akt activation in iNSCs receiving CHI mouse serum treatment. Remarkably, Akt inhibition led to the reduction of Crry expression in iNSCs. Intriguingly, the treatment of iNSC-derived neurons with recombinant complement receptor 2-conjugated Crry (CR2-Crry), which inhibits all complement pathways, substantially enhanced Crry expression and Akt activation in neurons after CHI mouse serum treatment. In subsequent in vitro experiments of pre-treatment of iNSCs with CR2-Crry, we observed significant increases in the levels of Crry expression and Akt activation in iNSCs and iNSC-derived astrocytes and neurons post-treatment with CHI mouse serum. Additionally, an in vivo study showed that intracerebral-transplanted iNSCs pre-treated with CR2-Crry markedly enhanced Crry expression in neurons and protected neurons from complement-dependent damage in the brains of CHI mice. CONCLUSION: INSCs receiving CR2-Crry pre-treatment increased the levels of Crry expression in iNSCs and iNSC-derived astrocytes and neurons and attenuated complement-mediated injury following CHI.


Assuntos
Traumatismos Cranianos Fechados , Células-Tronco Neurais , Animais , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/transplante , Neuroproteção , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Complemento 3b , Proteínas Recombinantes de Fusão
9.
J Neuroinflammation ; 16(1): 234, 2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31771613

RESUMO

BACKGROUND: Inflammation and apoptosis caused by intracerebral hemorrhage (ICH) are two important factors that affect patient prognosis and survival. Toll-like receptor 4 (TLR4) triggers activation of the inflammatory pathway, causing synthesis and release of inflammatory factors. The inflammatory environment also causes neuronal apoptosis. However, no studies have reported the role of TLR4 in inflammation and apoptosis. METHODS: We performed survival curve analysis and behavioral scores on TLR4 knockout mice and wild-type mice after inducing ICH. We used TLR4 knockout mice and wild-type mice to make ICH models with type VII collagenase and explored the link between TLR4 in inflammation and apoptosis. We used Western blot to detect the expression of apoptosis-related proteins, inflammatory factors, and their receptors at different time points after ICH induction. The effects of TLR4 on apoptosis were observed by TUNEL, Hoechst, and HE staining techniques. The association with TLR4 in inflammation and apoptosis was explored using IL-1ß and TNF-α antagonists. Data conforming to a normal distribution are expressed as mean ± standard deviation. Grade and quantitative data were compared with rank sum test and t test between two groups. P < 0.05 was considered statistically significant. RESULTS: TLR4 knockout significantly increased the survival rate of ICH mice. The scores of TLR4 knockout mice were significantly lower than those of wild-type mice. We found that TLR4 knockout mice significantly inhibited apoptosis and the expression of inflammatory factors after the induction of ICH. The apoptosis of ICH-induced mice was significantly improved after injecting IL-1ß and TNF-α antagonists. Moreover, the anti-apoptotic effect of the antagonist in wild-type mice is more pronounced. A single injection of the antagonist failed to improve apoptosis in TLR4 knockout mice. CONCLUSIONS: We conclude that TLR4-induced inflammation after ICH promotes neuronal apoptosis. IL-1ß and TNF-α antagonists attenuate this apoptotic effect. Therefore, targeting TLR4 in patients with clinical ICH may attenuate inflammatory response, thereby attenuating apoptosis and improving prognosis.


Assuntos
Apoptose/fisiologia , Encéfalo/metabolismo , Hemorragia Cerebral/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Hemorragia Cerebral/genética , Colagenases/metabolismo , Interleucina-1beta/antagonistas & inibidores , Camundongos , Camundongos Knockout , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores
10.
Int Immunopharmacol ; 76: 105837, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31476693

RESUMO

BACKGROUND: Intracranial hemorrhage (ICH) is one of the most common brain traumas, and inflammation caused by ICH seriously affects the quality of life and prognosis of patients. Eupatilin has been shown to have anti-inflammatory effects in various diseases. However, only one paper has reported that Eupatilin has a therapeutic effect on the inflammatory response caused by ICH and the underlying mechanism needs to be studied. METHODS: We used erythrocyte lysis stimulation (ELS) to induce mouse microglia BV2 as the inflammation model. CCK-8 and Transwell assays were used to detect cell viability and migration. RT-PCR, western blotting, and ELISA were used to detect the secretion of inflammatory factors and the expression of related mechanism proteins. HE staining was used to detect cell edema and death. RESULT: We found that ELS significantly increased protein and mRNA levels and secretion of inflammatory factors IL-1ß and TNF-α, which Eupatilin attenuated through the Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) pathway. The anti-inflammatory effect of Eupatilin was significantly attenuated after siRNA was used to reduce TLR4 expression. The experimental results and mechanism were also verified in TLR4 knockout mice in vivo. CONCLUSION: Eupatilin has a therapeutic effect on inflammation caused by ICH. The underlying mechanism may be related to TLR4/MyD88, which brings new hope for clinical patients to improve symptoms and prognosis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Hemorragia Cerebral/tratamento farmacológico , Flavonoides/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Hemorragia Cerebral/genética , Hemorragia Cerebral/imunologia , Flavonoides/farmacologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/fisiologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
11.
Gene ; 717: 143998, 2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31381951

RESUMO

Eid1 is a member of the EID protein family, which regulates differentiation, transcription and acetyltransferase activity. Accumulating evidence suggests that Eid1 is relevant to neurological disorder, but the main function of Eid1 is still unclear, especially in the brain. To better understand this issue, we generated Eid1-knockout (Eid1-KO) mice and profiled its gene expression changes in the brain by RNA sequencing. This study identified 2531 genes differentially expressed in Eid1-KO mice compared with the wild-type, then qRT-PCR verification demonstrated that the transcriptomic data are reliable. By protein-protein interaction cluster analysis, 'regulation of cell proliferation' were unexpectedly discovered as important Eid1 functions. We then isolated neural progenitor cells (NPCs) and showed that the number of neurospheres and the proliferation rate of Eid1-KO NPCs were obviously lower than that in the control group, furthermore, CCK-8 and immunofluorescence assay clearly demonstrated that the Eid1-KO NPCs showed significantly less cell proliferation than the control group. To the best of our knowledge, this is the first comprehensive report of the Eid1-KO transcriptome of mice brain. Our analysis and experimental data provide a foundation for further studies on understanding function of Eid1 in the brain.


Assuntos
Encéfalo/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/fisiologia , Proliferação de Células/genética , Feminino , Perfilação da Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Gravidez , Mapas de Interação de Proteínas , Análise de Sequência de RNA
12.
Cancer Manag Res ; 11: 4781-4796, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31213900

RESUMO

Purpose: Eupatilin is a pharmacologically active flavonoid extracted from Asteraceae argyi that has been identified as having antitumor effects. Gliomas are the most common intracranial malignant tumors and are associated with high mortality and a poor postoperative prognosis. There are few studies on the therapeutic effects of eupatilin on glioma. Therefore, we explored the efficacy and the underlying molecular mechanism of eupatilin on glioma. Methods: The effect of eupatilin on cell proliferation and viability was detected using Cell Counting Kit-8 assays. Cell migration was analyzed with a scratch wound healing assay and invasion was analyzed using transwell assays. Results: We found that eupatilin significantly inhibits the viability and proliferation of glioma cells by arresting the cell cycle at the G1/S phase. In addition, eupatilin disrupts the structure of the cytoskeleton and affects F-actin depolymerization via the "P-LIMK"/cofilin pathway, thereby inhibiting the migration of glioma. We also found that eupatilin inhibits the invasion of gliomas. The underlying mechanism may be related to the destruction of epithelial-mesenchymal transition, with eupatilin also affecting the RECK/matrix metalloproteinase pathway. However, we did not observe the proapoptotic effect of eupatilin on glioma, which is inconsistent with other studies. Finally, we observed a significant inhibitory effect of eupatilin on U87MG glioma in xenograft nude mice. Conclusion: Eupatilin inhibits the viability and proliferation of glioma cells, attenuates the migration and invasion, and inhibits tumor growth in vivo, but does not promote apoptosis. Therefore, due to the poor clinical efficacy of drug treatment of glioma and high drug resistance, the emergence of eupatilin brings a new dawn for glioma patients.

13.
Biochem Biophys Res Commun ; 512(4): 763-769, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-30926163

RESUMO

EP300-interacting inhibitor of differentiation 1 (Eid1) regulates differentiation, transcription and acetyltransferase activity. But the main function of Eid1 in the brain is still unclear. To better understand this issue, we generated Eid1-knockout (Eid1-KO) mice. We found poorer learning and memory ability, and smaller volume of neonatal telencephalon in Eid1-KO group than wild-type (WT). Bioinformatics implied that Eid1 may directly regulate cell proliferation. We then isolated neural stem cells (NSCs) and discovered a slower proliferation rate in Eid1-KO NSCs. Moreover, based on bioinformatics results, we investigated the expression of phosphatidylinositol 3-kinase (PI3K)/AKT/GSK3ß pathway by Western blotting assay, which showed attenuated in Eid1-KO group. Our data proved the first comprehensive report of Eid1 regulating NSCs proliferation via PI3K/AKT/GSK3ß pathway, and provide a foundation for the role of EID1 in the brain.


Assuntos
Proliferação de Células , Células-Tronco Neurais/citologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA