Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Br J Neurosurg ; 37(6): 1580-1587, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33538212

RESUMO

BACKGROUND AND OBJECTIVE: Several studies have examined the prognostic significance of IDH1/2 mutation, 1p/19q codeletion and MGMT promoter methylation in lower-grade gliomas but most of these used the 2007 fourth edition of the WHO classification. We evaluate prognostic significance of these indicators in the 2016 WHO updated fourth edition of CNS tumor classification. METHODS: A total of 180 intracranial glioma patients diagnosed according WHO 2016 edition between December 2016 and December 2018 Jinling Hospital (Nanjing, China) were reviewed retrospectively. We performed survival analysis on 109 patients with complete molecular pathology and follow-up data. RESULTS: Histologically, 52 were diagnosed as astrocytoma (WHO grade II and III), 17 as oligodendrogliomas (WHO grade II and III) and 40 as GBMs. At last follow-up, 50.5% patients had experienced tumor progression and 34.9% had died. Among grade II and III cases 36.2% experienced tumor progression and 27.5% died. In univariate Kaplan-Meier analysis, multifocal tumor, EGFR mutation or amplification, PIK3CA mutation and IDHwt/TERTpwt group were associated with shorter PFS (p < 0.001, p = 0.003, p = 0.005, p < 0.001, respectively) and OS (p = 0.010, p = 0.020, p = 0.018, p < 0.001, respectively) as were older age (≥55 years), multifocal tumor, IDH1/2 wild type, 1p/19q non-codeletion and negative methylation in the MGMT promoter region. A Cox proportional hazards model was created demonstrating that single tumor (HR = 0.180, p = 0.04), MGMTp methylation (HR = 0.095, p = 0.003) and chemoradiotherapy (HR = 0.006, p = 0.002) were independent prognostic factors for OS. CONCLUSIONS: Beyond histological classification as well as IDH1/2 mutation, 1p/19q codeletion status, we could incorporate IDH1/2mt combined with TERTpmt, EGFR mutation or amplification and PIK3CA mutation into the diagnostic criteria for DLGGs to supplement WHO 2016 pathological criteria.


Assuntos
Neoplasias Encefálicas , Glioma , Adulto , Humanos , Prognóstico , Estudos Retrospectivos , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/diagnóstico , Glioma/genética , Glioma/patologia , Mutação , Receptores ErbB/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Isocitrato Desidrogenase/genética
2.
Acta Pharmacol Sin ; 42(2): 218-229, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32747718

RESUMO

Aconitine (ACO), a main active ingredient of Aconitum, is well-known for its cardiotoxicity. However, the mechanisms of toxic action of ACO remain unclear. In the current study, we investigated the cardiac effects of ACO and mesaconitine (MACO), a structurally related analog of ACO identified in Aconitum with undocumented cardiotoxicity in guinea pigs. We showed that intravenous administration of ACO or MACO (25 µg/kg) to guinea pigs caused various types of arrhythmias in electrocardiogram (ECG) recording, including ventricular premature beats (VPB), atrioventricular blockade (AVB), ventricular tachycardia (VT), and ventricular fibrillation (VF). MACO displayed more potent arrhythmogenic effect than ACO. We conducted whole-cell patch-clamp recording in isolated guinea pig ventricular myocytes, and observed that treatment with ACO (0.3, 3 µM) or MACO (0.1, 0.3 µM) depolarized the resting membrane potential (RMP) and reduced the action potential amplitude (APA) and durations (APDs) in a concentration-dependent manner. The ACO- and MACO-induced AP remodeling was largely abolished by an INa blocker tetrodotoxin (2 µM) and partly abolished by a specific Na+/K+ pump (NKP) blocker ouabain (0.1 µM). Furthermore, we observed that treatment with ACO or MACO attenuated NKP current (INa/K) and increased peak INa by accelerating the sodium channel activation with the EC50 of 8.36 ± 1.89 and 1.33 ± 0.16 µM, respectively. Incubation of ventricular myocytes with ACO or MACO concentration-dependently increased intracellular Na+ and Ca2+ concentrations. In conclusion, the current study demonstrates strong arrhythmogenic effects of ACO and MACO resulted from increasing the peak INa via accelerating sodium channel activation and inhibiting the INa/K. These results may help to improve our understanding of cardiotoxic mechanisms of ACO and MACO, and identify potential novel therapeutic targets for Aconitum poisoning.


Assuntos
Aconitina/análogos & derivados , Aconitina/toxicidade , Arritmias Cardíacas/induzido quimicamente , Cardiotoxicidade/etiologia , Aconitina/isolamento & purificação , Aconitum/química , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/fisiopatologia , Cardiotoxicidade/fisiopatologia , Eletrocardiografia , Cobaias , Masculino , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
3.
Int J Neurosci ; 129(8): 801-807, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30648894

RESUMO

Background: Treatment of blast-induced traumatic brain injury (bTBI) has been hindered. Previous studies have demonstrated that oxidative stress may contribute to the pathophysiological process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) signaling pathway exhibits a protective effect after traumatic brain injury (TBI). This study explored whether the Nrf2-ARE pathway was activated in a modified bTBI mouse model. Method: Mice were randomly divided into six groups: the 6 h, 1 d, 3 d, 7 d and 14 d after bTBI groups and a sham group. The protein levels of nuclear Nrf2, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO1) were detected using western blot, and HO-1 and NQO1 mRNA levels were determined by real-time quantitative polymerase chain reaction. Moreover, HO-1 and Nrf2 were localized using histological staining. Results: The protein level of the Nrf2-ARE pathway in the frontal lobe increased significantly in the 3 d after bTBI. The HO-1 and NQO1 mRNA levels also reached a peak in the frontal lobe 3 d after bTBI. The histological staining demonstrated higher expression of HO-1 in the frontal lobe and hippocampus 3 d after bTBI, when nuclear import of Nrf2 reached a peak in the frontal lobe. Conclusions: bTBI activated the Nrf2-ARE signaling pathway in the brain. The peak activation time in the frontal lobe may be 3 d after injury, and activating the Nrf2 pathway could be a new direction for treatment.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Lobo Frontal/lesões , Lobo Frontal/metabolismo , Heme Oxigenase-1/metabolismo , Proteínas de Membrana/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Animais , Modelos Animais de Doenças , Masculino , Camundongos
4.
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
5.
Cell Mol Neurobiol ; 35(5): 713-21, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25732597

RESUMO

Traumatic brain injury (TBI) is a worldwide public health and medical problem. Oxidative stress is recognized as an important contributing factor in the pathogenesis of TBI. The present study was designed to explore the anti-oxidative effect of Nuclear factor erythroid 2-related factor 2 (Nrf2) on brain damage induced by traumatic injury in a mouse model. Moderate weight-drop impact head injury was induced in adult male mice. The mice were randomly divided into four groups: Nrf2(+/+) sham-operation, Nrf2(-/-) sham-operation, Nrf2(+/+) TBI, and Nrf2(-/-) TBI group. Neurological scores were evaluated 24 h after TBI, followed by collection of the brain specimens. Brain edema was detected by the wet-dry ratio method. The expression of NOX2 protein in the brain specimen was investigated using Western Blot analysis and immunohistochemical staining. In addition, malondialdehyde (MDA) level and superoxide dismutase (SOD) activity were evaluated in the brain tissues. Twenty-four hours after TBI, our results showed Nrf2(+/+) TBI mice have more severe neurological deficits and brain edema than Nrf2(+/+) sham group. On the other hand, the Nrf2(-/-) TBI mice were found to have significantly increased neurological deficits and brain edema, compared to Nrf2(+/+) TBI mice (P < 0.05). At the same time, we found that the expression of NOX2 protein, MDA level were significantly increased in Nrf2(-/-) mice, while SOD activity was considerably decreased after TBI compared to Nrf2(+/+) mice (P < 0.05). We demonstrated that deletion of Nrf2 exacerbates brain injury after TBI in mice, suggesting that Nrf2 may play an important role in protecting brain injury after TBI, possibly by modulating oxidative stress.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Deleção de Genes , Fator 2 Relacionado a NF-E2/deficiência , Estresse Oxidativo , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Edema Encefálico/patologia , Imuno-Histoquímica , Masculino , Malondialdeído/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos ICR , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Superóxido Dismutase/metabolismo
6.
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
7.
J Neurooncol ; 116(1): 41-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24078215

RESUMO

Resistance to chemoradiotherapy is a major obstacle to successful treatment of glioblastoma. Recently, the role of NF-E2-related factor 2 (Nrf2) in enhancing chemoradiation sensitivity has been reported in several types of cancers. Here, we investigated whether temozolomide (TMZ) and irradiation (IR) combined treatment induced Nrf2 activation in human glioblastoma cells. And we further performed a preliminary study about the effect of Nrf2 on chemoradiation sensitivity. Immunohistochemical staining for Nrf2 in paired clinical specimens showed that TMZ and IR combined treatment increased the expression and nuclear localization of Nrf2 in human glioblastoma tissues. Moreover, we found nuclear Nrf2 expression in the glioblastoma tissues obtained from the patients undergoing TMZ and IR combined treatment was associated with the time to tumor recurrence. In vitro, we further verified these findings. First, we detected increased nuclear localization of Nrf2 following treatment with TMZ+IR in human glioblastoma cell lines. Second, we demonstrated TMZ+IR increased the levels of Nrf2 protein in both nuclear and cytoplasmic fractions of U251 cells and induced Nrf2 target genes expression. Finally, downregulating Nrf2 expression increased TMZ+IR-induced cell death in the U251 cells. These findings suggest TMZ+IR combined treatment induces Nrf2 activation in human glioblastoma cells. The activation of Nrf2 may be associate with enhancing chemoradiation sensitivity in human glioblastoma cell. Blocking Nrf2 activation may be a promising method enhancing chemoradiation sensitivity of glioblastoma cells.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas , Dacarbazina/análogos & derivados , Glioblastoma , Fator 2 Relacionado a NF-E2/metabolismo , Radiação , Adulto , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , RNA Mensageiro/metabolismo , Temozolomida
8.
J Surg Res ; 188(1): 206-12, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24387843

RESUMO

BACKGROUND: Traumatic brain injury (TBI) is a worldwide health problem, identified as a major cause of death and disability. Increasing evidence has shown that oxidative stress plays an important role in TBI pathogenesis. The antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), is a known mediator in protection against TBI-induced brain damage. The objective of this study was to test whether tert-butylhydroquinone (tBHQ), a novel Nrf2 activator, can protect against TBI-induced oxidative stress. METHODS: Adult male imprinting control region mice were randomly divided into three groups: (1) sham + vehicle group; (2) TBI + vehicle group; and (3) TBI + tBHQ group. Closed-head brain injury was applied using the Feeney weight-drop method. We accessed the neurologic outcome of mice at 24 h after TBI, and subsequently measured protein levels of Nrf2 and the NOX2 subunit of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), the concentration of malondialdehyde, superoxide dismutase activity, and brain edema. RESULT: The NOX2 protein level was increased fivefold in the TBI + vehicle group, whereas pretreatment with tBHQ markedly attenuated the NOX2 protein expression relative to that in the TBI + vehicle group. TBI increased Nrf2 formation by 5% compared with the sham group, whereas treatment with tBHQ further upregulated the Nrf2 protein level by 12% compared with the sham group. The level of the oxidative damage marker malondialdehyde was reduced by 29% in the TBI + tBHQ group compared with the TBI + vehicle group, Moreover, pretreatment with tBHQ significantly increased the antioxidant enzyme superoxide dismutase activity. Administration of tBHQ also significantly decreased TBI-induced brain edema and neurologic deficits. CONCLUSIONS: Pretreatment with tBHQ effectively attenuated markers of cerebral oxidative stress after TBI, thus supporting the testing of tBHQ as a potential neuroprotectant and adjunct therapy for TBI patients.


Assuntos
Antioxidantes/uso terapêutico , Edema Encefálico/prevenção & controle , Lesões Encefálicas/tratamento farmacológico , Hidroquinonas/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Animais , Dano Encefálico Crônico/etiologia , Dano Encefálico Crônico/prevenção & controle , Edema Encefálico/etiologia , Lesões Encefálicas/complicações , Avaliação Pré-Clínica de Medicamentos , Masculino , Malondialdeído/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Exame Neurológico , Distribuição Aleatória , Superóxido Dismutase/metabolismo
9.
J Surg Res ; 186(1): 328-37, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24079811

RESUMO

BACKGROUND: A wealth of evidence has suggested that oxidative stress is involved in the secondary brain injury after traumatic brain injury (TBI). Recently, numerous in vivo and in vitro studies were reported that puerarin could inhibit oxidative stress through the activation of phosphatidylinositol 3-kinase (PI3K)-Akt pathway. It is unknown, however, whether puerarin can provide neuroprotection and reduce oxidative stress after TBI. The present study investigated the effects of puerarin on the TBI-induced neurodegeneration, oxidative stress, and the possible role of PI3K-Akt pathway in the neuroprotection of puerarin, in a rat model of TBI. MATERIALS AND METHODS: Rats were randomly distributed into various subgroups undergoing the sham surgery or TBI procedures. Puerarin (200 mg/kg) was given intraperitoneally at 10 min before injury and PI3K-Akt pathway inhibitor LY294002 was also administered intracerebroventricular in one subgroup. All rats were killed at 24 h after TBI for examination. RESULTS: Our data indicated that puerarin could significantly reduce TBI-induced neuronal degeneration, accompanied by the partial restoration of the redox disturbance and enhanced expression of phospho-Akt in the pericontusional cortex after TBI. Moreover, PI3K-Akt pathway inhibitor LY294002 could partially abrogate the neuroprotection of puerarin in rats with TBI. CONCLUSIONS: These results indicate that puerarin can ameliorate oxidative neurodegeneration after TBI, at least in part, through the activation of PI3K-Akt pathway.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Isoflavonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Lesões Encefálicas/metabolismo , Membrana Celular/efeitos dos fármacos , Masculino , Degeneração Neural/induzido quimicamente , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Acta Pharmacol Sin ; 35(6): 825-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24793313

RESUMO

AIM: Trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB) is a soluble epoxide hydrolase inhibitor that suppresses glioblastoma cell growth in vitro. The aim of this study was to examine whether the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) could sensitize glioma cells to t-AUCB-induced apoptosis. METHODS: Both U251 and U87 human glioblastoma cell lines were tested. Cell growth was assessed using the cell counting kit-8. Cell apoptosis was detected with caspase-3 activity assay kits and flow cytometry. The protein levels in the p38 MAPK/MAPKAPK2/Hsp27 pathway in the cells were analyzed using Western blots. RESULTS: Pretreatment with DAPT (2 µmol/L) substantially potentiated the growth inhibition caused by t-AUCB (200 µmol/L) in U251 and U87 cells. Furthermore, pretreatment with DAPT markedly increased t-AUCB-induced apoptosis of U251 and U87 cells. T-AUCB alone did not significant affect caspase-3 activity in the cells, but t-AUCB plus DAPT pretreatment caused significant increase of caspase-3 activity. Furthermore, pretreatment with DAPT completely blocked t-AUCB-induced phosphorylation of p38 MAPK, MAPKAPK2 and Hsp27 in the cells. CONCLUSION: The γ-secretase inhibitor DAPT sensitizes t-AUCB-induced apoptosis of human glioblastoma cells in vitro via blocking the p38 MAPK/MAPKAPK2/Hsp27 pathway, suggesting that the combination of t-AUCB and DAPT may be a potentially effective strategy for the treatment of glioblastoma.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Benzoatos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Dipeptídeos/farmacologia , Glioblastoma/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Ureia/análogos & derivados , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glioblastoma/metabolismo , Proteínas de Choque Térmico HSP27/antagonistas & inibidores , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ureia/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Mar Drugs ; 12(12): 6125-41, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25528957

RESUMO

Astaxanthin (ATX) has been proven to ameliorate early brain injury (EBI) after experimental subarachnoid hemorrhage (SAH) by modulating cerebral oxidative stress. This study was performed to assess the effect of ATX on the Nrf2-ARE pathway and to explore the underlying molecular mechanisms of antioxidant properties of ATX in EBI after SAH. A total of 96 male SD rats were randomly divided into four groups. Autologous blood was injected into the prechiasmatic cistern of the rat to induce an experimental SAH model. Rats in each group were sacrificed at 24 h after SAH. Expressions of Nrf2 and heme oxygenase-1 (HO-1) were measured by Western blot and immunohistochemistry analysis. The mRNA levels of HO-1, NAD (P) H: quinone oxidoreductase 1 (NQO-1), and glutathione S-transferase-α1 (GST-α1) were determined by real-time polymerase chain reaction (PCR). It was observed that administration of ATX post-SAH could up-regulate the cortical expression of these agents, mediated in the Nrf2-ARE pathway at both pretranscriptional and posttranscriptional levels. Meanwhile, oxidative damage was reduced. Furthermore, ATX treatment significantly attenuated brain edema, blood-brain barrier (BBB) disruption, cellular apoptosis, and neurological dysfunction in SAH models. This study demonstrated that ATX treatment alleviated EBI in SAH model, possibly through activating the Nrf2-ARE pathway by inducing antioxidant and detoxifying enzymes.


Assuntos
Elementos de Resposta Antioxidante/efeitos dos fármacos , Antioxidantes/farmacologia , Lesões Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Hemorragia Subaracnóidea/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças , Glutationa Transferase/metabolismo , Heme Oxigenase-1/metabolismo , Isoenzimas/metabolismo , Masculino , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Hemorragia Subaracnóidea/metabolismo , Xantofilas/farmacologia
12.
Brain Res ; 1837: 148855, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38471644

RESUMO

Subarachnoid hemorrhage (SAH) is characterized by the extravasation of blood into the subarachnoid space, in which erythrocyte lysis is the primary contributor to cell death and brain injuries. New evidence has indicated that meningeal lymphatic vessels (mLVs) are essential in guiding fluid and macromolecular waste from cerebrospinal fluid (CSF) into deep cervical lymph nodes (dCLNs). However, the role of mLVs in clearing erythrocytes after SAH has not been completely elucidated. Hence, we conducted a cross-species study. Autologous blood was injected into the subarachnoid space of rabbits and rats to induce SAH. Erythrocytes in the CSF were measured with/without deep cervical lymph vessels (dCLVs) ligation. Additionally, prior to inducing SAH, we administered rats with vascular endothelial growth factor C (VEGF-C), which is essential for meningeal lymphangiogenesis and maintaining integrity and survival of lymphatic vessels. The results showed that the blood clearance rate was significantly lower after dCLVs ligation in both the rat and rabbit models. DCLVs ligation aggravated neuroinflammation, neuronal damage, brain edema, and behavioral impairment after SAH. Conversely, the treatment of VEGF-C enhanced meningeal lymphatic drainage of erythrocytes and improved outcomes in SAH. In summary, our research highlights the indispensable role of the meningeal lymphatic pathway in the clearance of blood and mediating consequences after SAH.


Assuntos
Vasos Linfáticos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea , Animais , Coelhos , Hemorragia Subaracnóidea/metabolismo , Ratos , Masculino , Ligadura/métodos , Eritrócitos/metabolismo , Modelos Animais de Doenças , Fator C de Crescimento do Endotélio Vascular/metabolismo , Meninges , Edema Encefálico/metabolismo
13.
Biochem Biophys Res Commun ; 430(3): 1016-21, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23261470

RESUMO

A wealth of evidence has shown that microglia-associated neuro-inflammation is involved in the secondary brain injury contributed to the poor outcome after traumatic brain injury (TBI). In vitro studies were reported that activation of metabotropic glutamate receptor 5 (mGluR5) could inhibit the microglia-associated inflammation in response to lipopolysaccharide and our previous study indicated that mGluR5 was expressed in activated microglia following TBI. However, there is little known about whether mGluR5 activation can provide neuro-protection and reduce microglia-associated neuro-inflammation in rats after TBI. The goal of the present study was to investigate the effects of mGluR5 activation with selective agonist CHPG, on cerebral edema, neuronal degeneration, microglia activation and the releasing of pro-inflammatory cytokines, in a rat model of TBI. Rats were randomly distributed into various subgroups undergoing the sham surgery or TBI procedures, and 250 nmol of CHPG or equal volume vehicle was given through intracerebroventricular injection at 30 min post-TBI. All rats were sacrificed at 24 h after TBI for the further measurements. Our data indicated that post-TBI treatment with CHPG could significantly reduce the secondary brain injury characterized by the cerebral edema and neuronal degeneration, lead to the inhibition of microglia activation and decrease the expression of pro-inflammatory cytokines in both mRNA transcription and protein synthesis. These results provide the substantial evidence that activation of mGluR5 reduces the secondary brain injury after TBI, in part, through modulating microglia-associated neuro-inflammation.


Assuntos
Edema Encefálico/metabolismo , Lesões Encefálicas/metabolismo , Degeneração Neural/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Animais , Edema Encefálico/patologia , Lesões Encefálicas/patologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Degeneração Neural/patologia , Fenilacetatos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5
14.
Neurol India ; 61(3): 265-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23860146

RESUMO

BACKGROUND: Aneurysms located at distal posterior inferior cerebellar artery (PICA) are rare. These aneurysms are difficult for surgical or endovascular treatment, especially for ruptured aneurysms. AIMS: To investigate the clinical and radiologic efficacy of parent artery occlusion (PAO) with embolic agent Onyx in the treatment of distal PICA aneurysm. MATERIALS AND METHODS: Case records of 15 consecutive patients with 15 ruptured distal PICA aneurysms treated with Onyx embolization were reviewed retrospectively. The follow-up ranged between 6 and 52 months. Cerebral angiography or cerebra computed tomography-angiogram (CTA) was performed for follow-up radiological study. Two aneurysms had origin from tonsillomedullary segment, nine from telovelotonsillar segments, and four from cortical segments. All patients were treated with Onyx to occlude aneurysm and proximal portion of vessel in front of aneurysm via endovascular approach. RESULTS: Aneurysm was occluded completely in every patient. One patient died because of intra-procedure haemorrhage. Fourteen patients had good recovery and the last follow-up Glasgow outcome scale was 5. Head CT scan was performed in every survived patient before discharge. CT in 3 patients revealed cerebellar infarctions but without any neurological deficits. None of the 14 patients had rebleeding or fresh neurologic deficits during the follow-up period. Aneurysmal recanalization had not been observed in any of the survived patients. CONCLUSIONS: Onyx occlusion of proximal parent artery and aneurysm in the treatment of distal PICA aneurysm is safe and effective according to this study. Morphology and location of aneurysm are important to decide the therapeutic strategy.


Assuntos
Aneurisma Roto/terapia , Cerebelo/irrigação sanguínea , Embolização Terapêutica/métodos , Aneurisma Intracraniano/terapia , Polivinil/uso terapêutico , Tantálio/uso terapêutico , Adulto , Combinação de Medicamentos , Feminino , Escala de Resultado de Glasgow , Humanos , Masculino , Pessoa de Meia-Idade
15.
Brain Res ; 1808: 148324, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36921750

RESUMO

BACKGROUND: Ketones are not only utilized to produce energy but also play a neuroprotective role in many neurodegenerative diseases. However, whether this process has an impact on secondary brain damage after traumatic brain injury (TBI) remains unknown. OXCT1 (3-Oxoacid CoA-Transferase 1) is the rate-limiting enzyme in the intra-neuronal utilization of ketones. In this study, we investigated whether reduced expression of OXCT1 after TBI could impact neuroprotective mechanisms and exacerbate neurological dysfunction. MATERIALS AND METHODS: Experimental TBI was induced by a modified version of the weight drop model, it is a model of severe head trauma. Expression of OXCT1 in the injured hippocampus of mice was measured at different time points using immunoblotting assays. The release of abnormal mitochondrial cytochrome c from neurons of the mouse injured lateral hippocampus was measured 1 week after TBI using immunoblotting assays. Neuronal death was assessed by Nissl staining and the level of reactive oxygen species (ROS) within the neurons of the injured lateral hippocampus was assessed by Dihydroethidium staining. RESULTS: OXCT1 was overexpressed in hippocampal neurons by injection of adeno-associated virus into the lateral ventricle. OXCT1 expression levels decreased significantly 1 week post-TBI. After comparing the data obtained from different groups of mice, OXCT1 was found to significantly increase the expression of SIRT3 and reduce the proportion of acetylated SOD2, thus decreasing the production of ROS in the injured hippocampal neurons, reducing neuronal death, and improving cognitive function. CONCLUSIONS: OXCT1 has a critical previously unappreciated protective role in neurological impairment following TBI via the SIR3-SOD2 pathway. These findings highlight the potential of OXCT1 as a simple treatment for patients with TBI.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Fármacos Neuroprotetores , Sirtuína 3 , Animais , Camundongos , Lesões Encefálicas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Cetonas , Fármacos Neuroprotetores/farmacologia , Espécies Reativas de Oxigênio/metabolismo
16.
Neurol Sci ; 33(4): 855-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22076483

RESUMO

Zinc is the second most prevalent trace element in the body and is present in particularly large concentrations in the mammalian brain. In animal experiments, several lines of studies show that zinc is involved in the pathological process of ischemia, epilepsy, and mechanical head trauma. However, little is known about the change of zinc in the human neural diseases. In the present study, using immersion autometallography, we found that pathological zinc accumulation was extensively present in the human neurons following ischemia, epilepsy, and mechanical head trauma. Under H&E, apparently damaged neurons were observed in all of the brain regions that showed zinc ions labeled neuronal somata. In conclusion, the data provide the evidence of an active involvement of zinc in neuronal damage.


Assuntos
Encéfalo/metabolismo , Histocitoquímica/métodos , Doenças do Sistema Nervoso/patologia , Zinco/metabolismo , Doença Aguda , Adulto , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Criança , Cristalização , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nanotecnologia , Adulto Jovem
17.
Brain Inj ; 26(10): 1267-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22642418

RESUMO

BACKGROUND: Peroxisome proliferator activated receptor γ co-activator-1α (PGC-1α) is a transcriptional co-activator that co-ordinately regulates genes required for mitochondrial biogenesis and is a key contributor to the up-regulation of antioxidant activities in response to oxidative stress. The expression pattern of PGC-1α after traumatic brain injury (TBI) has not been studied. MATERIALS AND METHODS: Ninety male ICR mice (28-32 g) were randomly assigned to six groups: sham, 3, 6, 12, 24 and 48 hours after TBI. PGC-1α mRNA levels in mice brain were detected by reverse-transcriptase polymerase chain reaction and its nuclear protein levels by Western blot from 3-48 hours after TBI. PGC-1α distribution in the cerebral cortex after TBI was investigated by immunohistochemistry. MAIN OUTCOMES AND RESULTS: The PGC-1α mRNA level significantly increased from 3 hours after TBI, peaked at 6 hours and gradually decreased from 12 to 48 hours. The nuclear PGC-1α protein level increased from 6 to 24 hours after TBI and decreased at 48 hours after TBI. Increased PGC-1α immunostaining was detected in the neurons of the cerebral cortex at 12 hours after TBI. CONCLUSION: PGC-1α may play an important role in the brain after TBI.


Assuntos
Lesões Encefálicas/metabolismo , RNA Mensageiro/metabolismo , Transativadores/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Estresse Oxidativo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transativadores/genética , Fatores de Transcrição , Regulação para Cima
18.
J Neurotrauma ; 39(5-6): 423-434, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34861788

RESUMO

Clinically, the renin-angiotensin-aldosterone system is activated intensely in patients with moderate to severe traumatic brain injury (TBI). Increased angiotensin II in circulatory blood after TBI can enter the brain through the disrupted blood-brain barrier. Angiotensin-converting enzyme 2 (ACE2) is an enzyme that metabolizes angiotensin II into angiotensin (1-7), which has been shown to have neuroprotective results. The expression and role of ACE2 in the brain after TBI remains elusive, however. We found that ACE2 protein abundance was downregulated around the contusional area in the brains of both humans and mice. Endogenous ACE2 was expressed in neurons, astrocytes, and microglia in the cortex of the mouse brain. Administration of recombinant human ACE2 intracerebroventricularly alleviated neurological defects after TBI in mice. Treatment of recombinant human ACE2 suppressed TBI-induced increase of angiotensin II and the decrease of angiotensin (1-7) in the brain, mitigated neural cell death, reduced the activation of NLRP3 and caspase3, decreased phosphorylation of mitogen-activated protein kinases, and nuclear factor kappa B, and reduced inflammatory cytokines tumor necrosis factor alpha and interleukin-1ß. Administration of ACE2 enzyme activator diminazene aceturate intraperitoneally rescued downregulation of ACE2 enzymatic activity and protein abundance in the brain. Diminazene aceturate treatment once per day in the acute stage after TBI alleviated long-term cognitive defects and neuronal loss in mice. Collectively, these results indicated that restoration of ACE2 alleviated neurological deficits after TBI by mitigation of pyroptosis and apoptosis.


Assuntos
Enzima de Conversão de Angiotensina 2 , Lesões Encefálicas Traumáticas , Angiotensina II/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Humanos , Camundongos , Peptidil Dipeptidase A/metabolismo , Piroptose
19.
Brain Inj ; 25(3): 300-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21280976

RESUMO

BACKGROUND: Aquaporin-4 (AQP4) is a water channel protein and it is an important determinant of outcome after brain injury. Sulphoraphane (SFN) increases AQP4 levels with reduction of brain oedema at 3 days post-traumatic brain injury. However, little is known about the effect of SFN on AQP4 expression and oedema after spinal cord injury (SCI). METHODS AND PROCEDURES: The present study used a murine SCI model induced by compression injury. AQP4 protein level and mRNA level were detected by Western blot and by RT-PCR at 48 hours after SCI, respectively. In addition, immunohistochemical study was used to show AQP4 expression in the spinal cord segments and water content of the spinal cord segments were measured by wet?:?dry weight ratio. MAIN OUTCOMES AND RESULTS: This study shows that AQP4 level was decreased in the injured spinal cord segments at 48 hours following SCI. Post-injury administration of SFN increased AQP4 levels, which was accompanied by a significant reduction in spinal cord segment oedema at 48 hours post-injury. CONCLUSION: These findings suggest that the reduction of spinal cord oedema in response to SFN administration could be due, in part, to water clearance by AQP4 from the injured spinal cord segments.


Assuntos
Aquaporina 4/metabolismo , Edema/metabolismo , Isotiocianatos/farmacologia , Doenças da Medula Espinal , Traumatismos da Medula Espinal , Animais , Western Blotting , Edema/tratamento farmacológico , Masculino , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doenças da Medula Espinal/tratamento farmacológico , Doenças da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo
20.
World J Clin Cases ; 9(19): 5054-5063, 2021 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-34307556

RESUMO

BACKGROUND: High-grade aneurysmal subarachnoid hemorrhage is a devastating disease with a low favorable outcome. Elevated intracranial pressure is a substantial feature of high-grade aneurysmal subarachnoid hemorrhage that can result to secondary brain injury. Early control of intracranial pressure including decompressive craniectomy and external ventricular drainage had been reported to be associated with improved outcomes. But in recent years, little is known whether external ventricular drainage and intracranial pressure monitoring after coiling could improve outcomes in high-grade aneurysmal subarachnoid hemorrhage. AIM: To investigate the outcomes of high-grade aneurysmal subarachnoid hemorrhage patients with coiling and ventricular intracranial pressure monitoring. METHODS: A retrospective analysis of a consecutive series of high-grade patients treated between Jan 2016 and Jun 2017 was performed. In our center, followed by continuous intracranial pressure monitoring, the use of ventricular pressure probe for endovascular coiling and invasive intracranial pressure monitoring in the acute phase is considered to be the first choice for the treatment of high-grade patients. We retrospectively analyzed patient characteristics, radiological features, intracranial pressure monitoring parameters, complications, mortality and outcome. RESULTS: A total of 36 patients were included, and 32 (88.89%) survived. The overall mortality rate was 11.11%. No patient suffered from aneurysm re-rupture. The intracranial pressure in 33 patients (91.67%) was maintained within the normal range by ventricular drainage during the treatment. A favorable outcome was achieved in 18 patients (50%) with 6 mo follow-up. Delayed cerebral ischemia and Glasgow coma scale were considered as significant predictors of outcome (2.066 and -0.296, respectively, P < 0.05). CONCLUSION: Ventricular intracranial pressure monitoring may effectively maintain the intracranial pressure within the normal range. Despite the small number of cases in the current work, high-grade patients may benefit from a combination therapy of early coiling and subsequent ventricular intracranial pressure monitoring.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA