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.
Mol Biomed ; 4(1): 42, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37975957

RESUMO

Glioblastoma (GBM) is an aggressive intracranial tumour, and current chemotherapy regimens have limited efficacy. Aloperine (ALO), a natural alkaline compound, has shown potential as an antitumor agent. However, the effect of ALO against GBM remains unclear. This study aimed to investigate the function of ALO in treating GBM. U87, A172, and GL261 cell lines were used for in vitro experiments, and GL261 was also used to establish in vivo models. The results showed that ALO inhibited the proliferation of GBM cells by cell cycle arrest and apoptosis. Furthermore, autophagy was found to play a critical role, suggested by observation of autophagosomes under the transmission electron microscopy. It was discovered for the first time that ALO targeted lysosomes directly in glioma cells, tested by fluo-rescence-labelled ALO and organelle-localizing probes. In addition, ALO inhibited late autophagy and induced paraptosis in GBM, verified by classical gene expression changes in qPCR and western blotting. Also, ALO inhibited tumour growth and acted synergistically with temozolomide in intracranial glioma mice models in vivo. Our findings suggest that ALO targets lysosomes to inhibit late autophagy in GBM, inducing cell cycle arrest, paraptosis, and apoptosis. ALO may therefore be a promising therapeutic agent for the treatment of GBM.

2.
Pathol Oncol Res ; 29: 1611391, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37822669

RESUMO

Purpose: The driver mutations of gliomas have been identified in cerebrospinal fluid (CSF). Here we compared the concordance between CSF and tumor tissue for integrated diagnosis in gliomas using next-generation sequencing (NGS) to evaluate the feasibility of CSF detection in gliomas. Patients and methods: 27 paired CSF/tumor tissues of glioma patients were sequenced by a customized gene panel based on NGS. All CSF samples were collected through lumbar puncture before surgery. Integrated diagnosis was made by analysis of histology and tumor DNA molecular pathology according to the 2021 WHO classification of the central nervous system tumors. Results: A total of 24 patients had detectable circulating tumor DNA (ctDNA) and 22 had at least one somatic mutation or chromosome alteration in CSF. The ctDNA levels varied significantly across different ages, Ki-67 index, magnetic resonance imaging signal and glioma subtypes (p < 0.05). The concordance between integrated ctDNA diagnosis and the final diagnosis came up to 91.6% (Kappa, 0.800). We reclassified the clinical diagnosis of 3 patients based on the results of CSF ctDNA sequencing, and 4 patients were reassessed depending on tumor DNA. Interestingly, a rare IDH1 R132C was identified in CSF ctDNA, but not in the corresponding tumor sample. Conclusion: This study demonstrates a high concordance between integrated ctDNA diagnosis and the final diagnosis of gliomas, highlighting the practicability of NGS based detection of mutations of CSF in assisting integrated diagnosis of gliomas, especially glioblastoma.


Assuntos
DNA Tumoral Circulante , Glioma , Humanos , Biomarcadores Tumorais/genética , Glioma/diagnóstico , Glioma/genética , DNA Tumoral Circulante/genética , DNA de Neoplasias , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação
3.
Oncol Lett ; 20(3): 2442-2446, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32782561

RESUMO

Expression level of long non-coding RNA (lncRNA) RHPN1-AS1 in glioma tissues was detected to determine potential risk factors influencing prognosis of glioma. This study aimed to clarify the molecular mechanisms underlying tumorigenesis of glioma and thus to improve therapeutic efficacy of glioma. RHPN1-AS1 levels in glioma tissues (n=105) and normal brain tissues (n=105) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between RHPN1-AS1 level and pathological indicators of glioma patients was analyzed. Glioma patients were followed up for 5 years. Overall survival (OS) and relapse-free survival (RFS) in glioma patients were tested by Kaplan-Meier and log-rank method. Potential factors influencing prognosis of glioma were analyzed by Cox regression model. RHPN1-AS1 was upregulated in glioma tissues. Its level was correlated to histological grade, Karnofsky (KPS) score and postoperative recurrence of glioma patients, rather than sex, age, pathological and tumor size. Glioma patients expressing high level of RHPN1-AS1 suffered worse OS and RFS than those with low level. Advanced histological grade, KPS score <80 and high level of RHPN1-AS1 were considered to be risk factors influencing postoperative prognosis of glioma. High level of RHPN1-AS1 is an independent risk factor for poor prognosis of glioma, which may be utilized as a prognostic hallmark.

4.
J Mater Chem B ; 6(33): 5343-5352, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-32254500

RESUMO

Due to inhomogeneous molecular structure and inherent flexibility of organic piezoelectric materials, the improvement in piezoelectric performances is extremely challenging. Herein, a novel sheath-gas-assisted electrospinning method was designed to induce rapid recrystallization and a stretching effect on the PVDF molecular chains, which led to significant promotion in the formation of ß and γ crystal phases in PVDF nanofibers and the highest piezoelectric properties reported to date for pure organic piezoelectric materials. By using the PVDF nanofibers for energy harvesting in vivo and in vitro, the motion sensor and implantable nanogenerators displayed excellent sensitivity under an extremely low working frequency of ∼0.1 Hz and considerable output voltage of 0.4-0.6 V cm-2 driven by the femoral artery and carotid artery for the first time. Furthermore, we demonstrated that the nanofibers have outstanding biocompatibility and can provide a spontaneous microelectronic stimulation effect to improve the growth of nerve cells. Thus, we envision that the high-performance piezoelectric nanofibers may serve as a powerful tool for developing future flexible force-sensitive microelectric devices and generators for applications in bioelectronics and biomedicine.

5.
Neurochem Int ; 111: 32-44, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28465088

RESUMO

The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway exhibits protective effects in a variety of neurological diseases. However, the role of this pathway in traumatic brain injury (TBI) is not fully understood. This study investigates whether the Nrf2-ARE pathway provides neuroprotection following TBI via regulation of the ubiquitin proteasome system (UPS), and examines the involvement of this pathway in redox homeostasis. We found that activation the Nrf2-ARE pathway can mitigate secondary brain injury induced by TBI. Furthermore, we found that inhibiting the Nrf2-ARE pathway weakened the UPS following TBI. Treatment of TBI with the proteasome inhibitor, MG132, increased neuronal apoptosis, and evidence of brain water content was found. These data suggest that the Nrf2-ARE pathway provides neuroprotection following TBI via modulation of the UPS. In addition, the results indicated that the content of glutathione (GSH) was significantly increased after activation of Nrf2, and the level of ROS decreased; however, this effect contradictory in the Nrf2 knockout mice. Further studies found that treatment with the ROS agonist, ferric ammonium citrate (FAC), resulted in additional damage exerted by the ubiquitin proteasome pathways, and a significant increase in the amount of ubiquitinated proteins. In contrast, the activity of the ubiquitin proteasome pathways was vastly enhanced, and the level of ubiquitination proteins was significantly decreased following treatment with the inhibitor, N-acetylcysteine (NAC). The above mentioned results were also verified in in vitro experiments. In conclusion, the activation the Nrf2-ARE pathway improves neurological impairment caused by TBI via modulation of the UPS, and the redox homeostasis is one of the vital regulatory mechanisms.


Assuntos
Elementos de Resposta Antioxidante/fisiologia , Lesões Encefálicas Traumáticas/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Ubiquitina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Masculino , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Neuroproteção/fisiologia , Estresse Oxidativo/efeitos dos fármacos
6.
Neurochem Res ; 42(2): 337-346, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27734181

RESUMO

Previous studies have indicated oxidative stress and inflammatory injury as significant contributors to the secondary damage associated with traumatic brain injury (TBI). Ursolic acid (UA) has been demonstrated to exert anti-oxidative and anti-inflammatory effects on cerebral ischemia by activating the nuclear factor-erythroid 2-related factor 2 (Nrf2) pathway. However, the effects of UA on TBI remain unclear. The aim of this study is to evaluate the potential roles of UA in the activation of the Nrf2 pathway using an experimental TBI model and the underlying mechanism. Wild-type (WT) and Nrf2(-/-) mice were divided into eight groups: (1) sham; (2) TBI; (3) TBI + vehicle; (4) TBI + 50 mg/kg UA; (5) TBI + 100 mg/kg UA; (6) TBI + 150 mg/kg UA; (7) TBI + Nrf2(-/-) + vehicle; (8) TBI + Nrf2(-/-) + UA. All mice underwent the TBI with the exception of the sham group. The neurologic outcomes of the mice were evaluated at 24 h after TBI, as well as the expression of Nrf2, NQO1, HO1,SOD, GPx, and MDA. Treatment of UA significantly ameliorated brain edema and the neurological insufficiencies after TBI. In addition, UA treatment markedly strengthened the nuclear translocation of Nrf2 protein and increased the expression of NQO1 and HO1. Moreover, UA significantly increased the expression of AKT, an Nrf2 upstream factor, suggesting that UA play a neuroprotective role through the activation of the Nrf2-ARE signal pathway. On the contrary, UA showed no neuroprotective effect on the Nrf2(-/-) mice. These data indicated that UA increases the activity of antioxidant enzymes and attenuated brain injury via Nrf2 factor.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Triterpenos/uso terapêutico , Animais , Lesões Encefálicas Traumáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Triterpenos/farmacologia , Ácido Ursólico
7.
Neurochem Res ; 40(9): 1829-38, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26260377

RESUMO

Evidence has shown that the activation of the autophagy pathway after experimental subarachnoid hemorrhage (SAH) protects against neuronal damage. Tert-butylhydroquinone (tBHQ), a commonly used nuclear factor erythroid 2-related factor 2 (Nrf2) activator, was found to significantly enhance autophagy activation. The aim of this study was to explore the effect of tBHQ treatment on early stage brain injury at 24 h after SAH. The results showed that tBHQ treatment failed to stimulate an effective anti-oxidative effect at 24 h after the SAH operation, but succeeded in ameliorating early brain injury, including alleviated brain edema, BBB disruption, neuronal degeneration and neurological deficits. Further exploration found that tBHQ treatment significantly increased the expression of Beclin-1 and the ratio of microtubule-associated protein 1 light chain 3 (LC3)-II to LC3-I, suggesting that autophagy was enhanced after tBHQ treatment. Moreover, tBHQ treatment restored Bcl-2 and Bax expression and reduced caspase-3 cleavage, suggesting the protective effect of tBHQ treatment in ameliorating brain injury after SAH. Furthermore, tBHQ enhanced autophagy activation, decreased neuronal degeneration and improved the neurological score after SAH in Nrf2-deficient mice. Taken together, these findings suggest that tBHQ treatment exerts neuro-protective effects against EBI following SAH by enhancing Nrf2-independent autophagy. Therefore, tBHQ is a promising therapeutic agent against EBI following SAH.


Assuntos
Autofagia/efeitos dos fármacos , Lesões Encefálicas/prevenção & controle , Modelos Animais de Doenças , Hidroquinonas/farmacologia , Fator 2 Relacionado a NF-E2/fisiologia , Hemorragia Subaracnóidea/complicações , Animais , Barreira Hematoencefálica , Lesões Encefálicas/etiologia , Camundongos
8.
Neurochem Int ; 87: 85-91, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26055972

RESUMO

Alpha lipoic acid (ALA) is a powerful antioxidant that has proven protective effects against brain damage following a traumatic brain injury (TBI) in rats. However, the molecular mechanisms underlying these effects are not well understood. This study investigated the effect of ALA on neural apoptosis and the potential mechanism of these effects in the weight-drop model of TBI in male Sprague-Dawley rats that were treated with ALA (20 or 100 mg/kg) or vehicle via intragastric administration 30 min after TBI. Brain samples were collected 48 h later for analysis. ALA treatment resulted in a downregulation of caspase-3 expression, reduced the number of positive cells in the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay and improved neuronal survival. Furthermore, the level of malondialdehyde and glutathione peroxidase activity were restored, while Bcl-2-associated X protein translocation to mitochondria and cytochrome c release into the cytosol were reduced by ALA treatment. These results demonstrate that ALA improves neurological outcome in rats by protecting neural cell against apoptosis via a mechanism that involves the mitochondria following TBI.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas/patologia , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ácido Tióctico/farmacologia , Animais , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Lesões Encefálicas/complicações , Masculino , Ratos , Ratos Sprague-Dawley
9.
Tumour Biol ; 36(6): 4723-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25758051

RESUMO

Glioblastoma multiforme (GBM) is the most malignant primary brain tumor and more resistant to radiotherapy. However, hetero-radiosensitivity occurs in different patients. MicroRNAs (miRNAs) play important roles in the initiation and progression of a multitude of tumors. The study aims to examine the different microRNAs expression profiles of postoperative radiotherapy sensitive and resistant patients with GBM, to make an inquiry about their potential role and discover a certain set of radio-sensitivity markers. Three paired samples from six GBM patients who had only been treated with postoperative radiotherapy were selected, and then, they were divided into radiotherapy sensitive group and resistant group according to their overall survivals, local recurrence rates, and Karnofsky Performance Scale scores. Expression profiles of miRNAs in these two groups were determined by the method of microarray assay. Comparing with resistant patients, 13 miRNAs were significantly upregulated and 10 miRNAs were greatly downregulated in sensitive group. Among them, four miRNAs were validated by quantitative RT-PCR. The differentially expressed miRNAs and their putative target genes were revealed by bioformatic analysis to play a role in cell signaling, proliferation, aging, and death. High-enrichment pathway analysis identified that some classical pathways participated in numerous metabolic processes, especially in cell cycle regulation, such as mTOR, MAPK, TGF-beta, and PI3K-Akt signaling pathways. Our research will contribute to identifying clinical diagnostic markers and therapeutic targets in the treatment of GBM by postoperative radiotherapy.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , MicroRNAs/biossíntese , Tolerância a Radiação/genética , Adulto , Idoso , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Proliferação de Células/genética , Glioblastoma/radioterapia , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Período Pós-Operatório
10.
Cell Mol Neurobiol ; 35(4): 493-501, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25417195

RESUMO

Subarachnoid hemorrhage (SAH) is a pervasive and devastating condition in which inflammatory and apoptotic pathways contribute to poor outcome. Interleukin-33 (IL-33) plays a crucial role in the inflammatory and apoptotic pathways through binding of the transmembrane ST2 receptor. This study investigated the expression and cellular localization of IL-33 in the cerebral cortex after SAH in order to clarify the role of IL-33 after SAH. Sprague-Dawley rats were randomly divided into sham and SAH groups and evaluated 2, 6, and 12 h and 1, 2, 3, and 5 days after the surgery, with SAH animals subjected to prechiasmatic cistern SAH. IL-33 expression was measured by western blot analysis, real-time PCR, immunohistochemistry, and immunofluorescence. The mRNA levels of tumor necrosis factor (TNF)-α and IL-1ß were also assessed. The expression of IL-33, IL-1ß, and TNF-α was markedly elevated in the SAH as compared to the sham group; IL-33 was mainly localized in neurons and astrocytes and not microglia after SAH. Moreover, a significant positive association was observed between IL-33 and IL-1ß expression. These findings indicate that IL-33 might play an important role in the inflammatory response following SAH.


Assuntos
Córtex Cerebral/metabolismo , Interleucina-33/metabolismo , Hemorragia Subaracnóidea/metabolismo , Animais , Astrócitos/metabolismo , Núcleo Celular/metabolismo , Córtex Cerebral/patologia , Imunofluorescência , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-33/genética , Masculino , Neurônios/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Hemorragia Subaracnóidea/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
11.
Brain Res ; 1582: 237-46, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25093609

RESUMO

Secondary brain insult induced by traumatic brain injury (TBI), including excitotoxicity, oxidative stress, inflammatory response, and neuronal degeneration, is sensitive to therapeutic interventions; therefore, searching for neuroprotective agents represents a promising therapeutic strategy for TBI treatment. Luteolin, a member of the flavonoid family, has recently been proven to modulate autophagy. However, whether it activates autophagy after TBI thereby alleviating the secondary insult is not yet understood. Here, we aimed to evaluate the neuroprotection of luteolin against TBI and the potential role of autophagy where it is involved. For this purpose, mice were randomly divided into four groups and then subjected to TBI. The treatment mice received luteolin at a dose of 30mg/kg 30min post-TBI based on our previous study. We employed western blot, immunofluorescence and quantitative real-time PCR to determine autophagy process and inflammatory response among different groups. Autophagy was found to be enhanced after luteolin treatment according to the expressions of autophagic markers. Furthermore, luteolin decreased nuclear accumulation of p65 induced by TBI, indicating attenuation of inflammation. In line with these observations, luteolin decreased mRNA and protein expressions of pro-inflammatory factors IL-1b and TNF-a. At last, luteolin reduced neuronal degeneration, and alleviated brain edema and blood-brain barrier (BBB) disruption. In conclusion, these results implied that luteolin protected mice brain from traumatic brain injury by inhibiting inflammatory response, and luteolin-induced autophagy might play a pivotal role in its neuroprotection.


Assuntos
Autofagia/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Luteolina/farmacologia , Neuroimunomodulação/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Autofagia/fisiologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Western Blotting , Encéfalo/imunologia , Encéfalo/patologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Modelos Animais de Doenças , Imunofluorescência , Interleucina-1beta/metabolismo , Masculino , Camundongos , Neuroimunomodulação/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/metabolismo
12.
Brain Res ; 1558: 90-9, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24576487

RESUMO

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key endogenous protective regulator in the body. This study aimed to explore the role of Nrf2 in subarachnoid hemorrhage (SAH)-induced secondary complications. Wild type (WT) and Nrf2 knockout (KO) mice were subjected to experimental SAH by injecting fresh autologous blood into pre-chiasmatic cistern. The absence of Nrf2 function in mice resulted in exacerbated brain injury with increased brain edema, blood-brain barrier (BBB) disruption, neural apoptosis, and severe neurological deficits at 24h after SAH. Moreover, cerebral vasospasm was severe at 24h after SAH, but not significantly different between WT and Nrf2 KO mice after SAH. Meanwhile, Molondialdehyde (MDA) was increased and GSH/GSSG ratio was decreased in Nrf2 KO mice after SAH. Furthermore, higher expression of TNF-α and IL-1ß was also found after SAH in Nrf2 KO mice. In conclusion, our results revealed that Nrf2 plays an important role in attenuating SAH-induced secondary complications by regulating excessive oxidative stress and inflammatory response.


Assuntos
Fator 2 Relacionado a NF-E2/deficiência , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/etiologia , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Edema Encefálico/etiologia , Modelos Animais de Doenças , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Marcação In Situ das Extremidades Cortadas , Interleucina-1beta/metabolismo , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Necrose Tumoral alfa/metabolismo , Vasoespasmo Intracraniano/diagnóstico
13.
Free Radic Biol Med ; 71: 186-195, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24642087

RESUMO

Luteolin has recently been proven to exert neuroprotection in a variety of neurological diseases; however, its roles and the underlying mechanisms in traumatic brain injury are not fully understood. The present study was aimed to investigate the neuroprotective effects of luteolin in models of traumatic brain injury (TBI) and the possible role of the Nrf2-ARE pathway in the putative neuroprotection. A modified Marmarou׳s weight-drop model in mice and the scratch model in mice primary cultured neurons were used to induce TBI. We determined that luteolin significantly ameliorated secondary brain injury induced by TBI, including neurological deficits, brain water content, and neuronal apoptosis. Furthermore, the level of malondialdehyde (MDA) and the activity of glutathione peroxidase (GPx) were restored in the group with luteolin treatment. in vitro studies showed that luteolin administration lowered the intracellular reactive oxygen species (ROS) level and increased the neuron survival. Moreover, luteolin enhanced the translocation of Nrf2 to the nucleus both in vivo and in vitro, which was proved by the results of Western blot, immunohistochemistry, and electrophoretic mobility shift assay (EMSA). Subsequently upregulation of the expression of the downstream factors such as heme oxygenase 1 (HO1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) was also examined. However, luteolin treatment failed to provide neuroprotection after TBI in Nrf2(-/-) mice. Taken together, these in vivo and in vitro data demonstrated that luteolin provided neuroprotective effects in the models of TBI, possibly through the activation of the Nrf2-ARE pathway.


Assuntos
Elementos de Resposta Antioxidante , Lesões Encefálicas/tratamento farmacológico , Córtex Cerebral/efeitos dos fármacos , Luteolina/farmacologia , Fator 2 Relacionado a NF-E2/genética , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Regulação da Expressão Gênica , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Cultura Primária de Células , Transporte Proteico , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Água/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...