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.
J Mol Neurosci ; 73(4-5): 225-236, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36930428

RESUMO

Posttraumatic stress disorder (PTSD) is a persistent and severe psychological and mental disorder resulting from experiences of serious trauma or stress and is suffered by many individuals. Previous studies have shown that pretreatment with sevoflurane is efficient in reducing the incidence of PTSD. However, we require a more comprehensive understanding of the specific mechanisms by which sevoflurane works. Enhancer of zeste homolog 2 (EZH2) has been reported to be regulated by sevoflurane, and to improve patient cognition. In this study, we aimed to explore the mechanisms of sevoflurane and the role of EZH2 in PTSD cases. We explored the effects of sevoflurane and EPZ-6438 (inhibitor of EZH2) on rat behavior, followed by an investigation of EZH2 mRNA and protein expression. The effects of sevoflurane and EZH2 on neuronal survival were assessed by western blotting and TUNEL staining, while western blotting was used to examine the expression of PSD95 and the AKT/mTOR proteins. Sevoflurane preconditioning restored EZH2 expression and significantly inhibited apoptosis by regulating phosphorylation of the AKT/mTOR pathway. Synaptic plasticity was also significantly improved. These results suggest that pretreatment with sevoflurane could play an important role in PTSD prevention by regulating EZH2 expression.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Transtornos de Estresse Pós-Traumáticos , Ratos , Animais , Sevoflurano/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Hipocampo/metabolismo , Apoptose , Plasticidade Neuronal
3.
Immunol Invest ; 52(2): 224-240, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36562687

RESUMO

Interleukin-34 (IL-34) is a cytokine that plays important roles at steady state and in diseases. The induced or inhibited expression of IL-34 by stimuli has been deeply investigated. However, the regulation of IL-34 basal expression is largely unknown. The aim of this study is to investigate whether IL-34 expression is regulated by a general transcription factor Specificity Protein 1 (Sp1) at transcription level. By using bioinformatic software, four putative Sp1-binding sites overlapping GC boxes were found in the core promoter region of IL-34. Alignment of the core promoter sequences of mammalian IL-34 showed GC box-C (-62/-57) and D (-11/-6) were conserved in some mammals. Luciferase assay results showed that only deletion of GC box-C (-62/-57) significantly reduced luciferase activities of IL-34 core promoter in SH-SY5Y cells. By using electrophoretic mobility shift assay (EMSA), it was found that Sp1 specifically interacted with GC box-C sequence CCCGCC (-62/-57) in the core promoter of IL-34. By using chromatin immunoprecipitation (ChIP), it was discovered that Sp1 bound to the core promoter of IL-34 in living cells. In addition, silencing of Sp1 expression by its specific siRNA reduced IL-34 mRNA and protein levels significantly in SH-SY5Y cells. Likewise, IL-34 expression was inhibited in a dose-dependent manner by a Sp1 inhibitor Plicamycin. Furthermore, silencing of Sp1 also downregulated mRNA and protein expression of IL-34 in GES-1 and 293T cell lines, suggesting that IL-34 transcription regulated by Sp1 was not cell-type specific. Taken together, these results indicate that Sp1 controls the basal level of IL-34 transcription.


Assuntos
Neuroblastoma , Animais , Humanos , Neuroblastoma/genética , Regiões Promotoras Genéticas , Sítios de Ligação , Interleucinas/genética , Interleucinas/metabolismo , RNA Mensageiro/genética , Luciferases/genética , Luciferases/metabolismo , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Regulação da Expressão Gênica , Mamíferos/genética , Mamíferos/metabolismo
4.
Int J Mol Sci ; 23(17)2022 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-36077130

RESUMO

DNA Holliday junction (HJ) is a four-way stranded DNA intermediate that formed in replication fork regression, homology-dependent repair and mitosis, performing a significant role in genomic stability. Failure to remove HJ can induce an acceptable replication fork stalling and DNA damage in normal cells, leading to a serious chromosomal aberration and even cell death in HJ nuclease-deficient tumor cells. Thus, HJ is becoming an attractive target in cancer therapy. However, the development of HJ-targeting ligand faces great challenges because of flexile cavities on the center of HJs. This review introduces the discovery history of HJ, elucidates the formation and dissociation procedures of HJ in corresponding bio-events, emphasizes the importance of prompt HJ-removing in genome stability, and summarizes recent advances in HJ-based ligand discovery. Our review indicate that target HJ is a promising approach in oncotherapy.


Assuntos
DNA Cruciforme , DNA , DNA/metabolismo , Replicação do DNA , DNA Cruciforme/genética , Instabilidade Genômica , Humanos , Ligantes
5.
Glia ; 69(2): 281-295, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32652708

RESUMO

Brain edema is a grave complication of brain ischemia and is the main cause of herniation and death. Although astrocytic swelling is the main contributor to cytotoxic edema, the molecular mechanism involved in this process remains elusive. N-myc downstream-regulated gene 2 (NDRG2), a well-studied tumor suppressor gene, is mainly expressed in astrocytes in mammalian brains. Here, we found that NDRG2 deficiency leads to worsened cerebral edema, imbalanced Na+ transfer, and astrocyte swelling after ischemia. We also found that NDRG2 deletion in astrocytes dramatically changed the expression and distribution of aquaporin-4 and Na+ -K+ -ATPase ß1, which are strongly associated with cell polarity, in the ischemic brain. Brain edema and astrocyte swelling were significantly alleviated by rescuing the expression of astrocytic Na+ -K+ -ATPase ß1 in NDRG2-knockout mouse brains. In addition, the upregulation of astrocytic NDRG2 by lentiviral constructs notably attenuated brain edema, astrocytic swelling, and blood-brain barrier destruction. Our results indicate a particular role of NDRG2 in maintaining astrocytic polarization to facilitate Na+ and water transfer balance and to protect the brain from ischemic edema. These findings provide insight into NDRG2 as a therapeutic target in cerebral edema.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Astrócitos , Edema Encefálico , Acidente Vascular Cerebral , Adenosina Trifosfatases , Animais , Encéfalo , Edema Encefálico/etiologia , Camundongos
6.
Transl Stroke Res ; 11(2): 214-227, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31250377

RESUMO

Glutamate is a prominent neurotransmitter responsible for excitatory synaptic transmission and is taken up by sodium-dependent excitatory amino acid transporters (EAATs) on astrocytes to maintain synaptic homeostasis. Here, we report that N-myc downstream regulated gene 2 (NDRG2), a known tumor suppressor, is required to facilitate astroglial glutamate uptake and protect the brain from glutamate excitotoxicity after ischemia. NDRG2 knockout (Ndrg2-/-) mice exhibited an increase in cerebral interstitial glutamate and a reduction in glutamate uptake into astrocytes. The ability of NDRG2 to control EAAT-mediated glutamate uptake into astrocytes required NDRG2 to interact with and promote the function of Na+/K+-ATPase ß1, which could be disrupted by a Na+/K+-ATPase ß1 peptide. The deletion of NDRG2 or treatment with the Na+/K+-ATPase ß1 peptide significantly increased neuronal death upon a glutamate challenge and aggravated brain damage after ischemia. Our findings demonstrate that NDRG2 plays a pivotal role in promoting astroglial glutamate uptake from the interstitial space and protecting the brain from glutamate excitotoxicity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Astrócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , ATPase Trocadora de Sódio-Potássio/metabolismo
7.
J Clin Invest ; 127(12): 4270-4284, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29058689

RESUMO

Attention-deficit/hyperactivity disorder (ADHD) is a prevalent psychiatric disorder in children. Although an imbalance of excitatory and inhibitory inputs has been proposed as contributing to this disorder, the mechanisms underlying this highly heterogeneous disease remain largely unknown. Here, we show that N-myc downstream-regulated gene 2 (NDRG2) deficiency is involved in the development of ADHD in both mice and humans. Ndrg2-knockout (Ndrg2-/-) mice exhibited ADHD-like symptoms characterized by attention deficits, hyperactivity, impulsivity, and impaired memory. Furthermore, interstitial glutamate levels and excitatory transmission were markedly increased in the brains of Ndrg2-/- mice due to reduced astroglial glutamate clearance. We developed an NDRG2 peptide that rescued astroglial glutamate clearance and reduced excitatory glutamate transmission in NDRG2-deficient astrocytes. Additionally, NDRG2 peptide treatment rescued ADHD-like hyperactivity in the Ndrg2-/- mice, while routine methylphenidate treatment had no effect on hyperactivity in these animals. Finally, children who were heterozygous for rs1998848, a SNP in NDRG2, had a higher risk of ADHD than children who were homozygous for rs1998848. Our results indicate that NDRG2 deficiency leads to ADHD phenotypes and that impaired astroglial glutamate clearance, a mechanism distinct from the well-established dopamine deficit hypothesis for ADHD, underlies the resultant behavioral abnormalities.


Assuntos
Astrócitos/metabolismo , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Comportamento Animal , Proteínas/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Transtorno do Deficit de Atenção com Hiperatividade/patologia , Dopamina/genética , Dopamina/metabolismo , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Metilfenidato/farmacologia , Camundongos , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Proteínas/metabolismo , Proteínas Supressoras de Tumor/metabolismo
8.
Sci Rep ; 6: 21467, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26891996

RESUMO

The incidence of ischemic stroke is significantly increased in postmenopausal women. However, the neuroprotective effects of estrogen replacement therapy (ERT) against stroke remain controversial, and the role of astrocytes in ERT has rarely been explored. In this study, we investigated the effects of estrogen and selective estrogen receptor (ER) agonists on astrocytes activation and neuronal apoptosis in mice under conditions of cell culture oxygen and glucose deprivation and reperfusion (OGD-R), and global cerebral ischemia (GCI). We demonstrated that hippocampal astrocytes primarily express ERß. In astrocytes, 2.5-20 nM 17ß-estradiol (E2) or 10 nM DPN (ERß agonist) not 10 nM PPT (ERα agonist), significantly increased GFAP expression. And 10 nM E2, DPN or E2+MPP (ERα antagonist), but not PPT or E2+PHTPP (ERß antagonist), significantly reduced neuronal apoptosis following the subjection of astrocyte and neuronal cocultures to OGD-R. We also found that either 50 µg/kg E2 or 8 mg/kg DPN replacement (3 weeks) significantly increased GFAP expression and reduced GCI-induced neuronal apoptosis in hippocampal CA1 region of ovariectomized mice. These results indicate that estrogen-induced neuroprotection against ischemia-reperfusion injury involves activation of astrocytes via ERß. Thus, the discovery and design of astrocyte-selective ERß modulators may offer a new strategy for ERT of ischemic stroke.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Isquemia Encefálica/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Isquemia Encefálica/patologia , Caspase 3/metabolismo , Modelos Animais de Doenças , Receptor alfa de Estrogênio/metabolismo , Terapia de Reposição de Estrogênios , Estrogênios/administração & dosagem , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia
9.
Oncol Res ; 23(1-2): 43-51, 2016 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-26802650

RESUMO

Medullary thyroid carcinoma (MTC) is an uncommon and highly aggressive tumor of the neuroendocrine system, which derives from the neuroendocrine C cells of the thyroid gland. Except for surgical resection, there are not very many effective systemic treatment options for MTC. N-Myc downstream-regulated gene 2 (NDRG2) had a significantly lower expression in MTC compared with normal thyroid tissue. However, the function of NDRG2 in MTC oncogenesis is largely unknown. In this study, we found that overexpression of NDRG2 inhibited the proliferation of TT cells (human medullary thyroid carcinoma cells) in vitro and suppressed the development of MTC in a nude mouse xenograft model. Further analysis revealed that NDRG2 arrested the cell cycle G0/G1 phase progression and induced TT cell apoptosis. Moreover, NDRG2 overexpression may mediate the antiproliferative effect by reducing cyclin D1 and cyclin E protein levels. We also found aberrant NDRG2-mitigated TT cell migration and invasion in vitro. Sodium/iodide symporter (NIS) mediates active I(-) transport into the thyroid follicular cells, and radionuclide treatment is a promising therapy for MTC. Our current data revealed that NDRG2 overexpression enhanced NIS level in TT cells and increased their iodine uptake in vitro. Furthermore, (99m)TcO4(-) radionuclide imaging of the xenograft tumors indicated that NDRG2 could promote NIS-mediated radionuclide transport. In conclusion, the present study suggested that NDRG2 is a critical molecule in the regulation of MTC biological behavior and a potential promoter in radioactive iodine therapy.


Assuntos
Radioisótopos do Iodo/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Neoplasias da Glândula Tireoide/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose , Carcinogênese/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Expressão Gênica , Humanos , Radioisótopos do Iodo/metabolismo , Camundongos Nus , Transplante de Neoplasias , Cintilografia , Compostos Radiofarmacêuticos/metabolismo , Pertecnetato Tc 99m de Sódio/farmacocinética , Simportadores/metabolismo , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/patologia , Proteínas Supressoras de Tumor/genética
10.
Acta Biochim Biophys Sin (Shanghai) ; 47(10): 761-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26341979

RESUMO

Human N-myc downstream-regulated gene 2 (NDRG2) has been shown to be a multifunctional protein associated with cell proliferation, differentiation, transmembrane transport, and stress responses. In most mammalian brains, NDRG2 is principally expressed in astrocytic cells throughout different regions. NDRG2 has been increasingly implicated in the regulation of neurogenesis and in the development of nervous system diseases, including neurodegeneration, ischemia, and glioblastoma. This review summarizes the distribution and subcellular localization of NDRG2 in brain tissues, highlights the physiological actions of NDRG2 in the nervous system, and further discusses the roles of NDRG2 during the occurrence and development of several nervous system diseases.


Assuntos
Astrócitos/metabolismo , Encefalopatias/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica , Estresse Psicológico/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Humanos , Distribuição Tecidual
11.
Brain Res ; 1569: 1-8, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24796879

RESUMO

N-myc downstream-regulated gene 2 (Ndrg2) is a newly identified molecule that is mainly expressed in astrocytes within the central nervous system (CNS) and is involved in the proliferation and activation of astrocytes. 17ß-estradiol (E2) is one of the most important circulating hormones, and in the CNS, astrocytes are a target and potential mediator of the action of E2. Our most recent study found that DPN, an estrogen receptor (ER) ß-specific agonist, activated the Ndrg2 promoter and elevated endogenous NDRG2 protein expression in MCF7, HSG and T-47D cells. However, whether E2 regulates Ndrg2 expression in astrocytes remains unknown. Here, we conducted both in vivo and in vitro experiments and found that ERß co-localized with NDRG2 in astrocytes. Furthermore, in primary cultured astrocytes, we demonstrated that E2 up-regulated Ndrg2 mRNA and protein expression in a dose- and time-dependent manner and that the ERß agonist DPN but not the ERα agonist PPT up-regulated Ndrg2 expression. In vivo, we found that in the hippocampus of adult ovariectomized (OVX) female mice, Ndrg2 mRNA and protein expression were significantly decreased compared with those in normal adult female mice. After the OVX mice received continuous subcutaneous injections of 50µg/kg E2, 100µg/kg E2 or the ERß agonist DPN for 10 days, the Ndrg2 expression significantly increased compared with that of the OVX mice. Our results indicate that E2 may affect astrocytes by regulating Ndrg2 expression.


Assuntos
Astrócitos/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA