Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Neurochem Res ; 46(5): 1119-1128, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33559830

RESUMEN

This study aimed to explore the effects and function of microRNA-101a-3p (miR-101a-3p) in epilepsy. Rat model of pilocarpine-induced epilepsy was established and the seizure frequency was recorded. Expression of miR-101a-3p and c-Fos in hippocampus tissues of Rat models were detected by qRT-PCR and western blot. Besides, we established a hippocampal neuronal culture model of acquired epilepsy using Mg2+ free medium to evaluate the effects of miR-101a-3p and c-Fos in vitro. Cells were transfected with miR-101a-3p mimic, si-c-FOS, miR-101a-3p + c-FOS and its corresponding controls. MTT assay was used to detect cell viability upon transfection. Flow cytometry was performed to determine the apoptosis rate. Western blot was performed to measure the protein expression of apoptosis-related proteins (Bcl-2, Bax, and cleaved caspase 3), autophagy-related proteins (LC3 and Beclin1) and c-FOS. The targeting relationship between miR-101a-3p and c-FOS was predicted and verified by TargetScan software and dual-luciferase reporter assay. The role of miR-101a-3p was validated using epilepsy rat models in vivo. Another Rat models of pilocarpine-induced epilepsy with miR-NC or miR-101a-3p injection were established to evaluate the effect of miR-101a-3p overexpression on epilepsy in vivo. MiR-101a-3p was downregulated while c-FOS was increased in hippocampus tissues of Rat model of pilocarpine-induced epilepsy. Overexpression of miR-101a-3p or c-FOS depletion promoted cell viability, inhibited cell apoptosis and autophagy. C-FOS was a target of miR-101a-3p and miR-101a-3p negatively regulated c-FOS expression to function in epilepsy. Overexpression of miR-101a-3p attenuated pilocarpine-induced epilepsy in Rats in vivo. This study indicated that miR-101a-3p could attenuate pilocarpine-induced epilepsy by repressing c-Fos expression.


Asunto(s)
Epilepsia/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Apoptosis/fisiología , Autofagia/fisiología , Supervivencia Celular/fisiología , Regulación hacia Abajo/fisiología , Epilepsia/inducido químicamente , Hipocampo/citología , Hipocampo/metabolismo , Neuronas/metabolismo , Pilocarpina , Ratas Sprague-Dawley , Regulación hacia Arriba/fisiología
2.
Stem Cell Rev Rep ; 18(3): 1127-1142, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35257299

RESUMEN

Exosomes derived from mesenchymal stem cells (MSCs) have been considered as an alternative for cell therapy of acute spinal cord injury (ASCI). However, the underlying mechanism remains unclear. Here, ASCI mouse model and hypoxic cell model were established to evaluate the effects of MSCs and MSCs-derived exosomes (MSCs-exo). The results showed that both MSCs and MSCs-exo inhibited the production of ROS and ferrous iron, upregulated the expression of ferroptosis suppressor FSP1, and enhanced repair of neurological function in the ASCI mice. Besides, MSCs and MSCs-exo attenuated hypoxia-induced neuronal cell ferroptosis and increased cell proliferation. Further study demonstrated that lncGm36569 was enriched in the MSCs-exo. Through bioinformatics analysis and luciferase assay, we confirmed that lncGm36569 acted as a competitive RNA of miR-5627-5p to induce FSP1 upregulation. Furthermore, overexpression of miR-5627-5p reversed the therapeutic effects of lncGm36569 on neuronal cell ferroptosis. In conclusion, MSCs-exosomes lncGm36569 inhibited neuronal cell ferroptosis through miR-5627-5p/FSP1 axis, thereby attenuating neuronal dysfunction.


Asunto(s)
Exosomas , Ferroptosis , Células Madre Mesenquimatosas , MicroARNs , Traumatismos de la Médula Espinal , Animales , Exosomas/metabolismo , Ferroptosis/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/terapia
3.
Clin Transl Immunology ; 11(5): e1393, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35582627

RESUMEN

Objectives: Temozolomide (TMZ) resistance is a key factor that restricts the therapeutic effect of glioblastoma (GBM). YTH-domain family member 2 (YTHDF2) is highly expressed in GBM tissues, while the mechanism of YTHDF2 in TMZ resistance in GBM remains not fully elucidated. Methods: The YTHDF2 expression in TMZ-resistant tissues and cells was detected. Kaplan-Meier analysis was employed to evaluate the prognostic value of YTHDF2 in GBM. Effect of YTHDF2 in TMZ resistance in GBM was explored via corresponding experiments. RNA sequence, FISH in conjugation with fluorescent immunostaining, RNA immunoprecipitation, dual-luciferase reporter gene and immunofluorescence were applied to investigate the mechanism of YTHDF2 that boosted TMZ resistance in GBM. Results: YTHDF2 was up-regulated in TMZ-resistant tissues and cells, and patients with high expression of YTHDF2 showed lower survival rate than the patients with low expression of YTHDF2. The elevated YTHDF2 expression boosted TMZ resistance in GBM cells, and the decreased YTHDF2 expression enhanced TMZ sensitivity in TMZ-resistant GBM cells. Mechanically, YTHDF2 bound to the N6-methyladenosine (m6A) sites in the 3'UTR of EPHB3 and TNFAIP3 to decrease the mRNA stability. YTHDF2 activated the PI3K/Akt and NF-κB signals through inhibiting expression of EPHB3 and TNFAIP3, and the inhibition of the two pathways attenuated YTHDF2-mediated TMZ resistance. Conclusion: YTHDF2 enhanced TMZ resistance in GBM by activation of the PI3K/Akt and NF-κB signalling pathways via inhibition of EPHB3 and TNFAIP3.

4.
Cureus ; 13(4): e14373, 2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33976994

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are rare soft tissue malignancies that can occur in any part of the body. The most common sites are the proximal limbs and trunk. Intracranial MPNSTs are rare; most originate from the auditory, trigeminal, and other cranial nerves, and occurrence within the brain parenchyma is rarer. Here, we describe a malignant peripheral schwannoma in the cerebellar hemisphere of the brain parenchyma. To our knowledge, this is the first case of brain parenchymal metastasis of an MPNST. We observed no effects on the tumor after the application of multiple chemotherapy drugs; thereafter, we explored the literature surrounding the condition.

5.
Front Oncol ; 11: 756828, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35127472

RESUMEN

BACKGROUND: Isocitrate dehydrogenase (IDH) mutation and 1p19q codeletion status have been identified as significant markers for therapy and prognosis in lower-grade glioma (LGG). The current study aimed to construct a combined machine learning-based model for predicting the molecular subtypes of LGG, including (1) IDH wild-type astrocytoma (IDHwt), (2) IDH mutant and 1p19q non-codeleted astrocytoma (IDHmut-noncodel), and (3) IDH-mutant and 1p19q codeleted oligodendroglioma (IDHmut-codel), based on multiparametric magnetic resonance imaging (MRI) radiomics, qualitative features, and clinical factors. METHODS: A total of 335 patients with LGG (WHO grade II/III) were retrospectively enrolled. The sum of 5,929 radiomics features were extracted from multiparametric MRI. Selected robust, non-redundant, and relevant features were used to construct a random forest model based on a training cohort (n = 269) and evaluated on a testing cohort (n = 66). Meanwhile, preoperative MRIs of all patients were scored in accordance with Visually Accessible Rembrandt Images (VASARI) annotations and T2-fluid attenuated inversion recovery (T2-FLAIR) mismatch sign. By combining radiomics features, qualitative features (VASARI annotations and T2-FLAIR mismatch signs), and clinical factors, a combined prediction model for the molecular subtypes of LGG was built. RESULTS: The 17-feature radiomics model achieved area under the curve (AUC) values of 0.6557, 0.6830, and 0.7579 for IDHwt, IDHmut-noncodel, and IDHmut-codel, respectively, in the testing cohort. Incorporating qualitative features and clinical factors into the radiomics model resulted in improved AUCs of 0.8623, 0.8056, and 0.8036 for IDHwt, IDHmut-noncodel, and IDHmut-codel, with balanced accuracies of 0.8924, 0.8066, and 0.8095, respectively. CONCLUSION: The combined machine learning algorithm can provide a method to non-invasively predict the molecular subtypes of LGG preoperatively with excellent predictive performance.

6.
EBioMedicine ; 72: 103583, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34563923

RESUMEN

BACKGROUND: To develop and validate a deep learning signature (DLS) from diffusion tensor imaging (DTI) for predicting overall survival in patients with infiltrative gliomas, and to investigate the biological pathways underlying the developed DLS. METHODS: The DLS was developed based on a deep learning cohort (n = 688). The key pathways underlying the DLS were identified on a radiogenomics cohort with paired DTI and RNA-seq data (n=78), where the prognostic value of the pathway genes was validated in public databases (TCGA, n = 663; CGGA, n = 657). FINDINGS: The DLS was associated with survival (log-rank P < 0.001) and was an independent predictor (P < 0.001). Incorporating the DLS into existing risk system resulted in a deep learning nomogram predicting survival better than either the DLS or the clinicomolecular nomogram alone, with a better calibration and classification accuracy (net reclassification improvement 0.646, P < 0.001). Five kinds of pathways (synaptic transmission, calcium signaling, glutamate secretion, axon guidance, and glioma pathways) were significantly correlated with the DLS. Average expression value of pathway genes showed prognostic significance in our radiogenomics cohort and TCGA/CGGA cohorts (log-rank P < 0.05). INTERPRETATION: DTI-derived DLS can improve glioma stratification by identifying risk groups with dysregulated biological pathways that contributed to survival outcomes. Therapies inhibiting neuron-to-brain tumor synaptic communication may be more effective in high-risk glioma defined by DTI-derived DLS. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Transducción de Señal/genética , Adolescente , Adulto , Anciano , Estudios de Cohortes , Aprendizaje Profundo , Imagen de Difusión Tensora/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Adulto Joven
7.
Front Oncol ; 10: 558162, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117690

RESUMEN

The 2016 WHO classification of central nervous system tumors has included four molecular subgroups under medulloblastoma (MB) as sonic hedgehog (SHH), wingless (WNT), Grade 3, and Group 4. We aimed to develop machine learning models for predicting MB molecular subgroups based on multi-parameter magnetic resonance imaging (MRI) radiomics, tumor locations, and clinical factors. A total of 122 MB patients were enrolled retrospectively. After selecting robust, non-redundant, and relevant features from 5,529 extracted radiomics features, a random forest model was constructed based on a training cohort (n = 92) and evaluated on a testing cohort (n = 30). By combining radiographic features and clinical parameters, two combined prediction models were also built. The subgroup can be classified using an 11-feature radiomics model with a high area under the curve (AUC) of 0.8264 for WNT and modest AUCs of 0.6683, 0.6004, and 0.6979 for SHH, Group 3, and Group 4 in the testing cohort, respectively. Incorporating location and hydrocephalus into the radiomics model resulted in improved AUCs of 0.8403 and 0.8317 for WNT and SHH, respectively. After adding gender and age, the AUCs for WNT and SHH were further improved to 0.9097 and 0.8654, while the accuracies were 70 and 86.67% for Group 3 and Group 4, respectively. Prediction performance was excellent for WNT and SHH, while that for Group 3 and Group 4 needs further improvements. Machine learning algorithms offer potentials to non-invasively predict the molecular subgroups of MB.

8.
EBioMedicine ; 61: 103093, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33096488

RESUMEN

BACKGROUND: To develop a radiomics signature for predicting overall survival (OS)/progression-free survival (PFS) in patients with medulloblastoma (MB), and to investigate the incremental prognostic value and biological pathways of the radiomics patterns. METHODS: A radiomics signature was constructed based on magnetic resonance imaging (MRI) from a training cohort (n = 83), and evaluated on a testing cohort (n = 83). Key pathways associated with the signature were identified by RNA-seq (GSE151519). Prognostic value of pathway genes was assessed in a public GSE85218 cohort. FINDINGS: The radiomics-clinicomolecular signature predicted OS (C-index 0.762) and PFS (C-index 0.697) better than either the radiomics signature (C-index: OS: 0.649; PFS: 0.593) or the clinicomolecular signature (C-index: OS: 0.725; PFS: 0.691) alone, with a better calibration and classification accuracy (net reclassification improvement: OS: 0.298, P = 0.022; PFS: 0.252, P = 0.026). Nine pathways were significantly correlated with the radiomics signature. Average expression value of pathway genes achieved significant risk stratification in GSE85218 cohort (log-rank P = 0.016). INTERPRETATION: This study demonstrated radiomics signature, which associated with dysregulated pathways, was an independent parameter conferring incremental value over clinicomolecular factors in survival predictions for MB patients. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Asunto(s)
Biomarcadores , Imagen por Resonancia Magnética , Meduloblastoma/diagnóstico por imagen , Meduloblastoma/metabolismo , Transducción de Señal , Toma de Decisiones Clínicas , Biología Computacional/métodos , Manejo de la Enfermedad , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Estimación de Kaplan-Meier , Imagen por Resonancia Magnética/métodos , Meduloblastoma/mortalidad , Pronóstico , Reproducibilidad de los Resultados
9.
Cancer Med ; 8(8): 3811-3821, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31140757

RESUMEN

CD204 is a specific marker of tumor-associated macrophages (TAMs) in glioma. However, the expression levels of CD204 and its involvement in glioma are not fully understood. In this large-scale study, we assessed the expression and function of CD204 in whole-grade glioma molecularly and clinically. In total, 1323 glioma samples, including 301 microarray data and 325 RNA-seq data from the Chinese Glioma Genome Atlas (CGGA) dataset and 697 RNA-seq data from The Cancer Genome Atlas (TCGA) dataset, were utilized. The statistical analysis and graphical work were mainly performed using the R software. Univariate and multivariate Cox analysis demonstrated that CD204 was an independent prognosticator in glioma patients. CD204 expression was positively correlated with the grade of malignancy. CD204 was consistently upregulated in wild-type isocitrate dehydrogenase glioma and highly expressed in mesenchymal glioblastoma. Gene ontology of CD204-related genes showed that CD204 was most enriched in inflammatory response and immune response. It was associated with the stromal and immune populations, especially the monocytic lineage, fibroblasts, and T cells. Circos plots revealed that CD204 was closely associated with many immune checkpoint regulators, especially TIM-3. CD204 expression is consistent with the malignant phenotype of glioma and independently predicts poor outcomes in glioma patients. Additionally, CD204+ TAMs, collaborating with other checkpoint members, may contribute to the dysfunction of T cells. These findings suggest that CD204 may be a promising target for glioma immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Perfilación de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/genética , Receptores Depuradores de Clase A/antagonistas & inhibidores , Transcriptoma , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Biomarcadores de Tumor , Biología Computacional/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/mortalidad , Glioma/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Terapia Molecular Dirigida , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC
10.
J Clin Neurosci ; 67: 234-238, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31221576

RESUMEN

During neurological surgery, neurosurgeons have to transform the two-dimensional (2D) sectional images into three-dimensional (3D) structures at the cognitive level. The complexity of the intracranial structures increases the difficulty and risk of neurosurgery. Mixed reality (MR) applications reduce the obstacles in the transformation from 2D images to 3D visualization of anatomical structures of central nervous system. In this study, the holographic image was established by MR using computed tomography (CT), computed tomography angiography (CTA) and magnetic resonance imaging (MRI) data of patients. The surgeon's field of vision was superimposed with the 3D model of the patient's intracranial structure displayed on the mixed reality head-mounted display (MR-HMD). The neurosurgeons practiced and evaluated the feasibility of this technique in neurosurgical cases. We developed the segmentation image masks and texture mapping including brain tissue, intracranial vessels, nerves, tumors, and their relative positions by MR technologies. The results showed that the three-dimensional imaging is in a stable state in the operating room with no significant flutter and blur. And the neurosurgeon's feedback on the comfort of the equipment and the practicality of the technology was satisfactory. In conclusion, MR technology can holographically construct a 3D digital model of patient's lesions and improve the anatomical perception of neurosurgeons during craniotomy. The feasibility of the MR-HMD application in neurosurgery is confirmed.


Asunto(s)
Craneotomía/métodos , Holografía/métodos , Cirugía Asistida por Computador/métodos , Anciano , Neoplasias Encefálicas/cirugía , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Tomografía Computarizada por Rayos X/métodos
11.
Sci Rep ; 9(1): 13239, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519974

RESUMEN

Inflammation and immunoreaction markers were correlated with the survival of patients in many tumors. However, there were no reports investigating the relationships between preoperative hematological markers and the prognosis of medulloblastoma (MB) patients based on the molecular subgroups (WNT, SHH, Group 3, and Group 4). A total 144 MB patients were enrolled in the study. The differences of preoperative hematological markers among molecular subgroups of MB were compared by One-way ANOVA method. Kaplan-Meier method was used to calculate the curves of progression free survival (PFS) and overall survival (OS). The comparison of survival rates in different groups were conducted by the Log-rank test. Multivariate analysis was used to evaluate independent prognostic factors. Increased preoperative NLR (neutrophil-to-lymphocyte ratio, PFS, P = 0.004, OS, P < 0.001) and PLR (platelet-to-lymphocyte ratio, PFS, P = 0.028, OS, P = 0.003) predicted poor prognosis in patients with MB, while preoperative MLR (monocyte-to-lymphocyte ratio), MPV (mean platelet volume), PDW (platelet distribution width), and AGR (albumin-to-globulin ratio) were revealed no predictive value on the prognosis of patients with MB. Furthermore, high preoperative NLR and PLR predicted unfavorable prognosis in childhood MB patients. However, preoperative NLR and PLR were not associated with the prognosis in adult MB patients. Multivariate analysis demonstrated preoperative NLR (PFS, P = 0.029, OS, P = 0.005) and PLR (PFS, P = 0.023, OS, P = 0.005) were the independent prognostic factors in MB patients. Emphatically, the levels of preoperative NLR and PLR in Group 3 MB were significantly higher than those in WNT MB. High preoperative NLR was associated with unfavorable OS in Group 3 (P = 0.032) and Group 4 (P = 0.027) tumors. Similarly, increased preoperative PLR predicted poor PFS (P = 0.012) and OS (P = 0.009) in Group 4 tumors. Preoperative NLR and PLR were the potential prognostic markers for MB patients. Preoperative NLR and PLR were significantly associated with the survival of Group 3 and Group 4 tumors.


Asunto(s)
Biomarcadores de Tumor/análisis , Plaquetas/patología , Neoplasias Cerebelosas/patología , Linfocitos/patología , Meduloblastoma/patología , Cuidados Preoperatorios , Adolescente , Adulto , Neoplasias Cerebelosas/cirugía , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Meduloblastoma/cirugía , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
12.
Aging (Albany NY) ; 11(16): 6252-6272, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444316

RESUMEN

The prediction of clinical outcome for patients with infiltrative gliomas is challenging. Although preoperative hematological markers have been proposed as predictors of survival in glioma and other cancers, systematic investigations that combine these data with other relevant clinical variables are needed to improve prognostic accuracy and patient outcomes. We investigated the prognostic value of preoperative hematological markers, alone and in combination with molecular pathology, for the survival of 592 patients with Grade II-IV diffuse gliomas. On univariate analysis, increased neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and monocyte-to-lymphocyte ratio (MLR), and decreased albumin-to-globulin ratio (AGR), all predicted poor prognosis in Grade II/III gliomas. Multivariate analysis incorporating tumor status based on the presence of IDH mutations, TERT promoter mutations, and 1p/19q codeletion showed that in lower-grade gliomas, high NLR predicted poorer survival for the triple-negative, IDH mutation only, TERT mutation only, and IDH and TERT mutation groups. NLR was an independent prognostic factor in Grade IV glioma. We therefore propose a prognostic model for diffuse gliomas based on the presence of IDH and TERT promoter mutations, 1p/19q codeletion, and NLR. This model classifies lower-grade gliomas into nine subgroups that can be combined into four main risk groups based on survival projections.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/patología , Glioma/sangre , Glioma/patología , Patología Molecular , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Factores de Riesgo
13.
Int J Biochem Cell Biol ; 99: 133-139, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29608952

RESUMEN

BACKGROUND: Both LncRNA UCA1 and miR-495 are crucial gene regulators in various disorders. This study aims to investigate their role in epilepsy and seizure-induced brain injury. METHODS: In this research, rat model of epilepsy was established by pilocarpine induction. The RNA and protein expression in hippocampal tissues and neurons were determined by qRT-PCR and western blot, respectively. The hippocampal neurons were isolated from hippocampal tissues, and treated with magnesium-free (MGF) physiological solution for epileptiform activity induction. The endogenous expression of related genes was modulated by recombinant plasmids and cell transfection. Flow cytometry was used to analyze the cell apoptosis. Dual luciferase reporter assay was performed to determine the interaction between miR-495 and Nrf2 in HEK-293 cells. RESULTS: The lncRNA UCA1 and Nrf2 were down-regulated in epileptiform hippocampal tissues and neurons, while the miR-495 was up-regulated. Over-expression of UCA1 inhibited apoptosis of hippocampal neurons by suppressing miR-495. MiR-495 negatively regulated Nrf2. UCA1 inhibited apoptosis of hippocampal neurons through miR-495/Nrf2-ARE pathway. UCA1 suppressed pilocarpine-induced epilepsy in rat. CONCLUSION: LncRNA UCA1 suppressed pilocarpine-induced epilepsy by inhibiting apoptosis of hippocampal neurons through miR-495/Nrf2-ARE pathway, and thereby inhibiting brain injury induced by seizure.


Asunto(s)
Elementos de Respuesta Antioxidante , Lesiones Encefálicas/prevención & control , Epilepsia/complicaciones , MicroARNs/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , ARN Largo no Codificante/genética , Convulsiones/complicaciones , Animales , Apoptosis , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Células Cultivadas , Epilepsia/inducido químicamente , Epilepsia/patología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , MicroARNs/genética , Agonistas Muscarínicos/toxicidad , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Neuronas/patología , Pilocarpina/toxicidad , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA