Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36769143

RESUMEN

Traumatic brain injury (TBI) causes 10-20% of structural epilepsies and 5% of all epilepsies. The lack of prognostic biomarkers for post-traumatic epilepsy (PTE) is a major obstacle to the development of anti-epileptogenic treatments. Previous studies revealed TBI-induced alterations in blood microRNA (miRNA) levels, and patients with epilepsy exhibit dysregulation of blood miRNAs. We hypothesized that acutely altered plasma miRNAs could serve as prognostic biomarkers for brain damage severity and the development of PTE. To investigate this, epileptogenesis was induced in adult male Sprague Dawley rats by lateral fluid-percussion-induced TBI. Epilepsy was defined as the occurrence of at least one unprovoked seizure during continuous 1-month video-electroencephalography monitoring in the sixth post-TBI month. Cortical pathology was analyzed by magnetic resonance imaging on day 2 (D2), D7, and D21, and by histology 6 months post-TBI. Small RNA sequencing was performed from tail-vein plasma samples on D2 and D9 after TBI (n = 16, 7 with and 9 without epilepsy) or sham operation (n = 4). The most promising miRNA biomarker candidates were validated by droplet digital polymerase chain reaction in a validation cohort of 115 rats (8 naïve, 17 sham, and 90 TBI rats [21 with epilepsy]). These included 7 brain-enriched plasma miRNAs (miR-434-3p, miR-9a-3p, miR-136-3p, miR-323-3p, miR-124-3p, miR-212-3p, and miR-132-3p) that were upregulated on D2 post-TBI (p < 0.001 for all compared with naïve rats). The acute post-TBI plasma miRNA profile did not predict the subsequent development of PTE or PTE severity. Plasma miRNA levels, however, predicted the cortical pathology severity on D2 (Spearman ρ = 0.345-0.582, p < 0.001), D9 (ρ = 0.287-0.522, p < 0.001-0.01), D21 (ρ = 0.269-0.581, p < 0.001-0.05) and at 6 months post-TBI (ρ = 0.230-0.433, p < 0.001-0.05). We found that the levels of 6 of 7 miRNAs also reflected mild brain injury caused by the craniotomy during sham operation (ROC AUC 0.76-0.96, p < 0.001-0.05). In conclusion, our findings revealed that increased levels of neuronally enriched miRNAs in the blood circulation after TBI reflect the extent of cortical injury in the brain but do not predict PTE development.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , MicroARN Circulante , Epilepsia , MicroARNs , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Lesiones Traumáticas del Encéfalo/patología , Lesiones Encefálicas/complicaciones , MicroARNs/genética , Epilepsia/genética , Biomarcadores , Modelos Animales de Enfermedad
2.
Epilepsia ; 63(6): 1297-1313, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35218208

RESUMEN

The term neurovascular unit (NVU) describes the structural and functional liaison between specialized brain endothelium, glial and mural cells, and neurons. Within the NVU, the blood-brain barrier (BBB) is the microvascular structure regulating neuronal physiology and immune cross-talk, and its properties adapt to brain aging. Here, we analyze a research framework where NVU dysfunction, caused by acute insults or disease progression in the aging brain, represents a converging mechanism underlying late-onset seizures or epilepsy and neurological or neurodegenerative sequelae. Furthermore, seizure activity may accelerate brain aging by sustaining regional NVU dysfunction, and a cerebrovascular pathology may link seizures to comorbidities. Next, we focus on NVU diagnostic approaches that could be tailored to seizure conditions in the elderly. We also examine the impending disease-modifying strategies based on the restoration of the NVU and, more in general, the homeostatic control of anti- and pro-inflammatory players. We conclude with an outlook on current pre-clinical knowledge gaps and clinical challenges pertinent to seizure onset and conditions in an aging population.


Asunto(s)
Barrera Hematoencefálica , Epilepsia , Anciano , Envejecimiento , Encéfalo , Humanos , Convulsiones
3.
Neuropathol Appl Neurobiol ; 47(5): 679-693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33421166

RESUMEN

AIMS: Focal non-convulsive status epilepticus (FncSE) is a common emergency condition that may present as the first epileptic manifestation. In recent years, it has become increasingly clear that de novo FncSE should be promptly treated to improve post-status outcome. Whether seizure activity occurring during the course of the FncSE contributes to ensuing brain damage has not been demonstrated unequivocally and is here addressed. METHODS: We used continuous video-EEG monitoring to characterise an acute experimental FncSE model induced by unilateral intrahippocampal injection of kainic acid (KA) in guinea pigs. Immunohistochemistry and mRNA expression analysis were utilised to detect and quantify brain injury, 3-days and 1-month after FncSE. RESULTS: Seizure activity occurring during the course of FncSE involved both hippocampi equally. Neuronal loss, blood-brain barrier permeability changes, gliosis and up-regulation of inflammation, activity-induced and astrocyte-specific genes were observed in the KA-injected hippocampus. Diazepam treatment reduced FncSE duration and KA-induced neuropathological damage. In the contralateral hippocampus, transient and possibly reversible gliosis with increase of aquaporin-4 and Kir4.1 genes were observed 3 days post-KA. No tissue injury and gene expression changes were found 1-month after FncSE. CONCLUSIONS: In our model, focal seizures occurring during FncSE worsen ipsilateral KA-induced tissue damage. FncSE only transiently activated glia in regions remote from KA-injection, suggesting that seizure activity during FncSE without local pathogenic co-factors does not promote long-lasting detrimental changes in the brain. These findings demonstrate that in our experimental model, brain damage remains circumscribed to the area where the primary cause (KA) of the FncSE acts. Our study emphasises the need to use antiepileptic drugs to contain local damage induced by focal seizures that occur during FncSE.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Estado Epiléptico/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Epilepsia/patología , Cobayas , Ácido Kaínico/farmacología , Convulsiones/patología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico
4.
Neuropathol Appl Neurobiol ; 47(6): 826-839, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34003514

RESUMEN

AIMS: Focal cortical dysplasia (FCD) type 2 is an epileptogenic malformation of the neocortex associated with somatic mutations in the mammalian target of rapamycin (mTOR) pathway. Histopathologically, FCD 2 is subdivided into FCD 2a and FCD 2b, the only discriminator being the presence of balloon cells (BCs) in FCD 2b. While pro-epileptogenic immune system activation and inflammatory responses are commonly detected in both subtypes, it is unknown what contextual role BCs play. METHODS: The present study employed RNA sequencing of surgically resected brain tissue from FCD 2a (n = 11) and FCD 2b (n = 20) patients compared to autopsy control (n = 9) focusing on three immune system processes: adaptive immunity, innate immunity and cytokine production. This analysis was followed by immunohistochemistry on a clinically well-characterised FCD 2 cohort. RESULTS: Differential expression analysis revealed stronger expression of components of innate immunity, adaptive immunity and cytokine production in FCD 2b than in FCD 2a, particularly complement activation and antigen presentation. Immunohistochemical analysis confirmed these findings, with strong expression of leukocyte antigen I and II in FCD 2b as compared to FCD 2a. Moreover, T-lymphocyte tissue infiltration was elevated in FCD 2b. Expression of markers of immune system activation in FCD 2b was concentrated in subcortical white matter. Lastly, antigen presentation was strongly correlated with BC load in FCD 2b lesions. CONCLUSION: We conclude that, next to mutation-driven mTOR activation and seizure activity, BCs are crucial drivers of inflammation in FCD 2b. Our findings indicate that therapies targeting inflammation may be beneficial in FCD 2b.


Asunto(s)
Epilepsia/patología , Sistema Inmunológico/metabolismo , Malformaciones del Desarrollo Cortical de Grupo I/patología , Malformaciones del Desarrollo Cortical/patología , Serina-Treonina Quinasas TOR/metabolismo , Adolescente , Niño , Epilepsia/genética , Epilepsia/inmunología , Humanos , Masculino , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/inmunología , Malformaciones del Desarrollo Cortical de Grupo I/genética , Malformaciones del Desarrollo Cortical de Grupo I/inmunología , Persona de Mediana Edad , Mutación/genética , Neocórtex/patología , Neuronas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Sustancia Blanca/metabolismo
5.
Neuropathol Appl Neurobiol ; 47(6): 796-811, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33942341

RESUMEN

AIMS: Tuberous sclerosis complex (TSC) is a genetic disorder associated with dysregulation of the mechanistic target of rapamycin complex 1 (mTORC1) signalling pathway. Neurodevelopmental disorders, frequently present in TSC, are linked to cortical tubers in the brain. We previously reported microRNA-34a (miR-34a) among the most upregulated miRs in tubers. Here, we characterised miR-34a expression in tubers with the focus on the early brain development and assessed the regulation of mTORC1 pathway and corticogenesis by miR-34a. METHODS: We analysed the expression of miR-34a in resected cortical tubers (n = 37) compared with autopsy-derived control tissue (n = 27). The effect of miR-34a overexpression on corticogenesis was assessed in mice at E18. The regulation of the mTORC1 pathway and the expression of the bioinformatically predicted target genes were assessed in primary astrocyte cultures from three patients with TSC and in SH-SY5Y cells following miR-34a transfection. RESULTS: The peak of miR-34a overexpression in tubers was observed during infancy, concomitant with the presence of pathological markers, particularly in giant cells and dysmorphic neurons. miR-34a was also strongly expressed in foetal TSC cortex. Overexpression of miR-34a in mouse embryos decreased the percentage of cells migrated to the cortical plate. The transfection of miR-34a mimic in TSC astrocytes negatively regulated mTORC1 and decreased the expression of the target genes RAS related (RRAS) and NOTCH1. CONCLUSIONS: MicroRNA-34a is most highly overexpressed in tubers during foetal and early postnatal brain development. miR-34a can negatively regulate mTORC1; however, it may also contribute to abnormal corticogenesis in TSC.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/crecimiento & desarrollo , MicroARNs/genética , Esclerosis Tuberosa/genética , Adolescente , Adulto , Animales , Encéfalo/patología , Corteza Cerebral/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Neuronas/patología , Transducción de Señal/genética , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/patología , Adulto Joven
6.
Acta Neuropathol ; 142(4): 729-759, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34292399

RESUMEN

Neuronal dysfunction due to iron accumulation in conjunction with reactive oxygen species (ROS) could represent an important, yet underappreciated, component of the epileptogenic process. However, to date, alterations in iron metabolism in the epileptogenic brain have not been addressed in detail. Iron-related neuropathology and antioxidant metabolic processes were investigated in resected brain tissue from patients with temporal lobe epilepsy and hippocampal sclerosis (TLE-HS), post-mortem brain tissue from patients who died after status epilepticus (SE) as well as brain tissue from the electrically induced SE rat model of TLE. Magnetic susceptibility of the presumed seizure-onset zone from three patients with focal epilepsy was compared during and after seizure activity. Finally, the cellular effects of iron overload were studied in vitro using an acute mouse hippocampal slice preparation and cultured human fetal astrocytes. While iron-accumulating neurons had a pyknotic morphology, astrocytes appeared to acquire iron-sequestrating capacity as indicated by prominent ferritin expression and iron retention in the hippocampus of patients with SE or TLE. Interictal to postictal comparison revealed increased magnetic susceptibility in the seizure-onset zone of epilepsy patients. Post-SE rats had consistently higher hippocampal iron levels during the acute and chronic phase (when spontaneous recurrent seizures are evident). In vitro, in acute slices that were exposed to iron, neurons readily took up iron, which was exacerbated by induced epileptiform activity. Human astrocyte cultures challenged with iron and ROS increased their antioxidant and iron-binding capacity, but simultaneously developed a pro-inflammatory phenotype upon chronic exposure. These data suggest that seizure-mediated, chronic neuronal iron uptake might play a role in neuronal dysfunction/loss in TLE-HS. On the other hand, astrocytes sequester iron, specifically in chronic epilepsy. This function might transform astrocytes into a highly resistant, pro-inflammatory phenotype potentially contributing to pro-epileptogenic inflammatory processes.


Asunto(s)
Epilepsia del Lóbulo Temporal/complicaciones , Hipocampo/metabolismo , Trastornos del Metabolismo del Hierro/etiología , Hierro/metabolismo , Estado Epiléptico/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/metabolismo , Astrocitos/patología , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/patología , Femenino , Humanos , Trastornos del Metabolismo del Hierro/patología , Masculino , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Ratas , Estado Epiléptico/metabolismo , Estado Epiléptico/patología
7.
Glia ; 68(1): 60-75, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31408236

RESUMEN

Temporal lobe epilepsy (TLE) is a chronic neurological disease in humans, which is refractory to pharmacological treatment in about 30% of the patients. Reactive glial cells are thought to play a major role during the development of epilepsy (epileptogenesis) via regulation of brain inflammation and remodeling of the extracellular matrix (ECM). These processes can be regulated by microRNAs (miRs), a class of small non-coding RNAs, which can control entire gene networks at a post-transcriptional level. The expression of miRs is known to change dynamically during epileptogenesis. miR-132 is one of the most commonly upregulated miRs in animal TLE models with important roles shown in neurons. However, the possible role of miR-132 in glia remains largely unknown. The aim of this study was to characterize the cell-type specific expression of miR-132 in the hippocampus of patients with TLE and during epileptogenesis in a rat TLE model. Furthermore, the potential role of miR-132 was investigated by transfection of human primary cultured astrocytes that were stimulated with the cytokines IL-1ß or TGF-ß1. We showed an increased expression of miR-132 in the human and rat epileptogenic hippocampus, particularly in glial cells. Transfection of miR-132 in human primary astrocytes reduced the expression of pro-epileptogenic COX-2, IL-1ß, TGF-ß2, CCL2, and MMP3. This suggests that miR-132, particularly in astrocytes, represents a potential therapeutic target that warrants further in vivo investigation.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , MicroARNs/biosíntesis , Neuroglía/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/patología , Células Cultivadas , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/patología , Femenino , Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Neuroglía/patología , Ratas , Ratas Sprague-Dawley , Adulto Joven
8.
Neurobiol Dis ; 145: 105080, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32919030

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) causes 10-20% of acquired epilepsy, which typically develops within 2 years post-injury with poorly understood mechanisms. We investigated the location, severity, evolution and persistence of blood-brain barrier (BBB) dysfunction and associated neuroinflammation after TBI, and their contribution to post-traumatic seizure susceptibility. METHODS: TBI was induced with lateral fluid-percussion in adult male Sprague-Dawley rats (6 sham, 12 TBI). Permeability of the BBB was assessed using T1-weighted magnetic resonance imaging (MRI) with gadobutrol (Gd) contrast enhancement at 4 days, 2 weeks, 2 months, and 10 months post-injury and with intravenously administered fluorescein at 11 months post-TBI. Continuous (24/7) video-EEG monitoring was performed for 3 weeks at 11 months post-injury followed by the pentylenetetrazol (PTZ) seizure-susceptibility test. In the end, rats were perfused for histology to assess albumin extravasation, iron deposits, calcifications, reactive astrocytes, microglia and monocytes. To investigate the translational value of the data obtained, BBB dysfunction and neuroinflammation were investigated immunohistochemically in autopsy brain tissue from patients with TBI and PTE. RESULTS: MRI indicated persistent Gd leakage in the impacted cortex and thalamus of variable severity in all rats with TBI which correlated with fluorescein extravasation. In the impacted cortex BBB dysfunction was evident from 4 days post-injury onwards to the end of the 10-months follow-up. In the ipsilateral thalamus, leakage was evident at 2 and 10 months post-injury. The greater the BBB leakage in the perilesional cortex at 10 months after the injury, the greater the expression of the endothelial cell antigen RECA-1 (r = 0.734, p < 0.01) and the activated macrophages/monocytes/microglia marker CD68 (r = 0.699, p < 0.05) at 11 months post-injury. Seven of the 12 rats with TBI showed increased seizure susceptibility in the PTZ-test. Unlike expected, we did not find any association between increased Gd-leakage or neuroinflammation with seizure susceptibility at 11 months post-TBI. Analysis of human autopsy tissue indicated that similar to the animal model, chronic BBB dysfunction was also evident in the perilesional cortex and thalamus of patients with PTE, characterized by presence of albumin, iron deposits and calcifications as well as markers of neuroinflammation, including reactive astrocytes, microglia and monocytes. CONCLUSIONS: Rats and humans with TBI have long-lasting cortical BBB dysfunction and neuroinflammation. Focal Gd-enhancement matched with loci of neuroinflammation, particularly in the thalamus. Although BBB leakage did not associate with increased seizure susceptibility after TBI, our data suggest that for treatments aimed to mitigate BBB damage and its secondary pathologies like chronic neuroinflammation, there is a region-specific, long-lasting therapeutic time window.


Asunto(s)
Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Convulsiones/etiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Permeabilidad Capilar , Femenino , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Convulsiones/patología
9.
Neurobiol Dis ; 134: 104612, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31533065

RESUMEN

Our understanding of mesial temporal lobe epilepsy (MTLE), one of the most common form of drug-resistant epilepsy in humans, is derived mainly from clinical, imaging, and physiological data from humans and animal models. High-throughput gene expression studies of human MTLE have the potential to uncover molecular changes underlying disease pathogenesis along with novel therapeutic targets. Using RNA- and small RNA-sequencing in parrallel, we explored differentially expressed genes in the hippocampus and cortex of MTLE patients who had undergone surgical resection and non-epileptic controls. We identified differentially expressed genes in the hippocampus of MTLE patients and differentially expressed small RNAs across both the cortex and hippocampus. We found significant enrichment for astrocytic and microglial genes among up-regulated genes, and down regulation of neuron specific genes in the hippocampus of MTLE patients. The transcriptome profile of the small RNAs reflected disease state more robustly than mRNAs, even across brain regions which show very little pathology. While mRNAs segregated predominately by brain region for MTLE and controls, small RNAs segregated by disease state. In particular, our data suggest that specific miRNAs (e.g., let-7b-3p and let-7c-3p) may be key regulators of multiple pathways related to MTLE pathology. Further, we report a strong association of other small RNA species with MTLE pathology. As such we have uncovered novel elements that may contribute to the establishment and progression of MTLE pathogenesis and that could be leveraged as therapeutic targets.


Asunto(s)
Epilepsia del Lóbulo Temporal/genética , ARN Pequeño no Traducido/genética , Adulto , Anciano , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Transcriptoma , Adulto Joven
10.
Epilepsia ; 61(8): 1595-1605, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32652588

RESUMEN

OBJECTIVE: Depressive disorders are common among about 50% of the patients with drug-resistant temporal lobe epilepsy (TLE). The underlying etiology remains elusive, but hypothalamus-pituitary-adrenal (HPA) axis activation due to changes in glucocorticoid receptor (GR) protein expression could play an important role. Therefore, we set out to investigate expression of the GR in the hippocampus, an important brain region for HPA axis feedback, of patients with drug-resistant TLE, with and without comorbid depression. METHODS: GR expression was studied using immunohistochemistry on hippocampal sections from well-characterized TLE patients with depression (TLE + D, n = 14) and without depression (TLE - D, n = 12) who underwent surgery for drug-resistant epilepsy, as well as on hippocampal sections from autopsy control cases (n = 9). Video-electroencephalography (EEG), magnetic resonance imaging (MRI), and psychiatric and memory assessments were performed prior to surgery. RESULTS: Abundant GR immunoreactivity was present in dentate gyrus granule cells and CA1 pyramidal cells of controls. In contrast, neuronal GR expression was lower in patients with TLE, particularly in the TLE + D group. Quantitative analysis showed a smaller GR+ area in TLE + D as compared to TLE - D patients and controls. Furthermore, the ratio between the number of GR+/NeuN+ cells was lower in patients with TLE + D as compared to TLE - D and correlated negatively with the depression severity based on psychiatric history. The expression of the GR was also lower in glial cells of TLE + D compared to TLE - D patients and correlated negatively to the severity of depression. SIGNIFICANCE: Reduced hippocampal GR expression may be involved in the etiology of depression in patients with TLE and could constitute a biological marker of depression in these patients.


Asunto(s)
Trastorno Depresivo/metabolismo , Epilepsia Refractaria/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Receptores de Glucocorticoides/metabolismo , Adulto , Anciano , Región CA1 Hipocampal/metabolismo , Estudios de Casos y Controles , Giro Dentado/metabolismo , Trastorno Depresivo/complicaciones , Epilepsia Refractaria/complicaciones , Epilepsia Refractaria/cirugía , Epilepsia del Lóbulo Temporal/complicaciones , Epilepsia del Lóbulo Temporal/cirugía , Femenino , Humanos , Sistema Hipotálamo-Hipofisario , Masculino , Persona de Mediana Edad , Procedimientos Neuroquirúrgicos , Sistema Hipófiso-Suprarrenal , Células Piramidales/metabolismo , Adulto Joven
11.
Brain ; 142(7): e39, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31145451

RESUMEN

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Asunto(s)
Acetilcisteína/farmacología , Epilepsia/prevención & control , Glutatión/metabolismo , Isotiocianatos/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Astrocitos/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Recuento de Células , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/prevención & control , Modelos Animales de Enfermedad , Estimulación Eléctrica , Epilepsia/complicaciones , Proteína HMGB1/sangre , Hipocampo/metabolismo , Humanos , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Estado Epiléptico/complicaciones , Estado Epiléptico/metabolismo , Estado Epiléptico/prevención & control , Sulfóxidos
12.
Epilepsia ; 60(4): 605-614, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30747999

RESUMEN

OBJECTIVE: Inhibition of the mammalian target of rapamycin (mTOR) pathway could be antiepileptogenic in temporal lobe epilepsy (TLE), possibly via anti-inflammatory actions. We studied effects of the mTOR inhibitor rapamycin and the anti-inflammatory compound curcumin-also reported to inhibit the mTOR pathway-on epileptogenesis and inflammation in an in vitro organotypic hippocampal-entorhinal cortex slice culture model. METHODS: Brain slices containing hippocampus and entorhinal cortex were obtained from 6-day-old rat pups and maintained in culture for up to 3 weeks. Rapamycin or curcumin was added to the culture medium from day 2 in vitro onward. Electrophysiological recordings revealed epileptiformlike activity that developed over 3 weeks. RESULTS: In week 3, spontaneous seizurelike events (SLEs) could be detected using whole cell recordings from CA1 principal neurons. The percentage of recorded CA1 neurons displaying SLEs was lower in curcumin-treated slice cultures compared to vehicle-treated slices (25.8% vs 72.5%), whereas rapamycin did not reduce SLE occurrence significantly (52%). Western blot for phosphorylated-S6 (pS6) and phosphorylated S6K confirmed that rapamycin inhibited the mTOR pathway, whereas curcumin only lowered pS6 expression at one phosphorylation site. Real-time quantitative polymerase chain reaction results indicated a trend toward lower expression of inflammatory markers IL-1ß and IL-6 and transforming growth factor ß after 3 weeks of treatment with rapamycin and curcumin compared to vehicle. SIGNIFICANCE: Our results show that curcumin suppresses SLEs in the combined hippocampal-entorhinal cortex slice culture model and suggest that its antiepileptogenic effects should be further investigated in experimental models of TLE.


Asunto(s)
Antioxidantes/farmacología , Curcumina/farmacología , Corteza Entorrinal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Convulsiones/metabolismo , Animales , Corteza Entorrinal/metabolismo , Hipocampo/metabolismo , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
13.
Brain ; 141(11): 3130-3143, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30307467

RESUMEN

Epilepsy therapy is based on drugs that treat the symptoms rather than the underlying mechanisms of the disease (epileptogenesis). There are no treatments for preventing seizures or improving disease prognosis, including neurological comorbidities. The search of pathogenic mechanisms of epileptogenesis highlighted that neuroinflammatory cytokines [i.e. interleukin-1ß (IL-1ß), tumour necrosis factor-α (Tnf-α)] are induced in human and experimental epilepsies, and contribute to seizure generation in animal models. A major role in controlling the inflammatory response is played by specialized pro-resolving lipid mediators acting on specific G-protein coupled receptors. Of note, the role that these pathways have in epileptogenic tissue remains largely unexplored. Using a murine model of epilepsy, we show that specialized pro-resolving mechanisms are activated by status epilepticus before the onset of spontaneous seizures, but with a marked delay as compared to the neuroinflammatory response. This was assessed by measuring the time course of mRNA levels of 5-lipoxygenase (Alox5) and 15-lipoxygenase (Alox15), the key biosynthetic enzymes of pro-resolving lipid mediators, versus Il1b and Tnfa transcripts and proteins. In the same hippocampal tissue, we found a similar delayed expression of two main pro-resolving receptors, the lipoxin A4 receptor/formyl peptide receptor 2 and the chemerin receptor. These receptors were also induced in the human hippocampus after status epilepticus and in patients with temporal lobe epilepsy. This evidence supports the hypothesis that the neuroinflammatory response is sustained by a failure to engage pro-resolving mechanisms during epileptogenesis. Lipidomic LC-MS/MS analysis showed that lipid mediator levels apt to resolve the neuroinflammatory response were also significantly altered in the hippocampus during epileptogenesis with a shift in the biosynthesis of several pro-resolving mediator families including the n-3 docosapentaenoic acid (DPA)-derived protectin D1. Of note, intracerebroventricular injection of this mediator during epileptogenesis in mice dose-dependently reduced the hippocampal expression of both Il1b and Tnfa mRNAs. This effect was associated with marked improvement in mouse weight recovery and rescue of cognitive deficit in the novel object recognition test. Notably, the frequency of spontaneous seizures was drastically reduced by 2-fold on average and the average seizure duration was shortened by 40% after treatment discontinuation. As a result, the total time spent in seizures was reduced by 3-fold in mice treated with n-3 DPA-derived protectin D1. Taken together, the present findings demonstrate that epilepsy is characterized by an inadequate engagement of resolution pathways. Boosting endogenous resolution responses significantly improved disease outcomes, providing novel treatment avenues.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Ácidos Docosahexaenoicos/uso terapéutico , Encefalitis/tratamiento farmacológico , Epilepsia/tratamiento farmacológico , Animales , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Antígeno CD11b/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Encefalitis/inducido químicamente , Epilepsia/inducido químicamente , Epilepsia/complicaciones , Epilepsia/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/patología , Ácido Kaínico/toxicidad , Leucotrieno B4/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Lipoxinas/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
Cell Mol Life Sci ; 75(24): 4557-4581, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30155647

RESUMEN

Traumatic brain injury (TBI) initiates molecular and cellular pathologies that underlie post-injury morbidities, including hippocampus-related memory decline and epileptogenesis. Non-coding small RNAs are master regulators of gene expression with the potential to affect multiple molecular pathways. To evaluate whether hippocampal gene expression networks are chronically regulated by microRNAs after TBI, we sampled the dentate gyrus of rats with severe TBI induced by lateral fluid-percussion injury 3 months earlier. Ingenuity pathway analysis revealed 30 upregulated miR-124-3p targets, suggesting that miR-124-3p is downregulated post-TBI (z-score = - 5.146, p < 0.05). Droplet digital polymerase chain reaction (ddPCR) and in situ hybridization confirmed the chronic downregulation of miR-124-3p (p < 0.05). Quantitative PCR analysis of two targets, Plp2 and Stat3, indicated that their upregulation correlated with the miR-124-3p downregulation (r = - 0.647, p < 0.05; r = - 0.629, p < 0.05, respectively). Immunohistochemical staining of STAT3 confirmed the increased protein expression. STRING analysis showed that 9 of the 30 miR-124-3p targets belonged to a STAT3 network. Reactome analysis and data mining connected the targets especially to inflammation and signal transduction. L1000CDS2 software revealed drugs (e.g., importazole, trichostatin A, and IKK-16) that could reverse the observed molecular changes. The translational value of our data was emphasized by in situ hybridization showing chronic post-traumatic downregulation of miR-124-3p in the dentate gyrus of TBI patients. Analysis of another brain injury model, status epilepticus, highlighted the fact that chronic downregulation of miR-124 is a common phenomenon after brain injury. Together, our findings indicate that miR-124-3p is a chronic modulator of molecular networks relevant to post-injury hippocampal pathologies in experimental models and in humans.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , Regulación de la Expresión Génica , MicroARNs/genética , Anciano , Animales , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Regulación hacia Abajo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Persona de Mediana Edad , Ratas Sprague-Dawley
15.
Glia ; 66(5): 1082-1097, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29384235

RESUMEN

Astrocytes are important mediators of inflammatory processes in the brain and seem to play an important role in several neurological disorders, including epilepsy. Recent studies show that astrocytes produce several microRNAs, which may function as crucial regulators of inflammatory pathways and could be used as therapeutic target. We aim to study which miRNAs are produced by astrocytes during IL-1ß mediated inflammatory conditions in vitro, as well as their functional role and to validate these findings in human epileptogenic brain tissue. Sequencing was used to assess miRNA and mRNA expression in IL-1ß-stimulated human fetal astrocyte cultures. miRNAs were overexpressed in cell cultures using miRNA mimics. Expression of miRNAs in resected brain tissue from patients with tuberous sclerosis complex or temporal lobe epilepsy with hippocampal sclerosis was examined using in situ hybridization. Two differentially expressed miRNAs were found: miR146a and miR147b, which were associated with increased expression of genes related to the immune/inflammatory response. As previously reported for miR146a, overexpression of miR147b reduced the expression of the pro-inflammatory mediators IL-6 and COX-2 after IL-1ß stimulation in both astrocyte and tuberous sclerosis complex cell cultures. miR146a and miR147b overexpression decreased proliferation of astrocytes and promoted neuronal differentiation of human neural stem cells. Similarly to previous evidence for miR146a, miR147b was increased expressed in astrocytes in epileptogenic brain. Due to their anti-inflammatory effects, ability to restore aberrant astrocytic proliferation and promote neuronal differentiation, miR146a and miR147b deserve further investigation as potential therapeutic targets in neurological disorders associated with inflammation, such as epilepsy.


Asunto(s)
Astrocitos/inmunología , Inflamación/metabolismo , MicroARNs/metabolismo , Astrocitos/patología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/cirugía , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/cirugía , Humanos , Inflamación/patología , Interleucina-1beta , Interleucina-6/metabolismo , Células-Madre Neurales/metabolismo , ARN Mensajero/metabolismo , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/cirugía
16.
J Neuroinflammation ; 15(1): 211, 2018 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-30031401

RESUMEN

BACKGROUND: Temporal lobe epilepsy (TLE) is a chronic neurological disease, in which about 30% of patients cannot be treated adequately with anti-epileptic drugs. Brain inflammation and remodeling of the extracellular matrix (ECM) seem to play a major role in TLE. Matrix metalloproteinases (MMPs) are proteolytic enzymes largely responsible for the remodeling of the ECM. The inhibition of MMPs has been suggested as a novel therapy for epilepsy; however, available MMP inhibitors lack specificity and cause serious side effects. We studied whether MMPs could be modulated via microRNAs (miRNAs). Several miRNAs mediate inflammatory responses in the brain, which are known to control MMP expression. The aim of this study was to investigate whether an increased expression of MMPs after interleukin-1ß (IL-1ß) stimulation can be attenuated by inhibition of the inflammation-associated miR-155. METHODS: We investigated the expression of MMP2, MMP3, MMP9, and MMP14 in cultured human fetal astrocytes after stimulation with the pro-inflammatory cytokine IL-1ß. The cells were transfected with miR-155 antagomiR, and the effect on MMP3 expression was investigated using real-time quantitative PCR and Western blotting. Furthermore, we characterized MMP3 and miR-155 expression in brain tissue of TLE patients with hippocampal sclerosis (TLE-HS) and during epileptogenesis in a rat TLE model. RESULTS: Inhibition of miR-155 by the antagomiR attenuated MMP3 overexpression after IL-1ß stimulation in astrocytes. Increased expression of MMP3 and miR-155 was also evident in the hippocampus of TLE-HS patients and throughout epileptogenesis in the rat TLE model. CONCLUSIONS: Our experiments showed that MMP3 is dynamically regulated by seizures as shown by increased expression in TLE tissue and during different phases of epileptogenesis in the rat TLE model. MMP3 can be induced by the pro-inflammatory cytokine IL-1ß and is regulated by miR-155, suggesting a possible strategy to prevent epilepsy via reduction of inflammation.


Asunto(s)
Astrocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Metaloproteinasa 3 de la Matriz/metabolismo , MicroARNs/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Unión al Calcio , Citocinas/genética , Citocinas/metabolismo , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , Femenino , Feto , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 3 de la Matriz/genética , MicroARNs/genética , Proteínas de Microfilamentos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Ratas , Estado Epiléptico/metabolismo , Estado Epiléptico/patología
17.
Epilepsia ; 59(11): 2106-2117, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30306542

RESUMEN

OBJECTIVE: Dravet syndrome is a rare neurodevelopmental disease, characterized by general cognitive impairment and severe refractory seizures. The majority of patients carry the gene mutation SCN1A, leading to a defective sodium channel that contributes to pathogenic brain excitability. A γ-aminobutyric acid (GABAergic) impairment, as in other neurodevelopmental diseases, has been proposed as an additional mechanism, suggesting that seizures could be alleviated by GABAergic therapies. However, up to now the physiological mechanisms underlying the GABAergic dysfunction in Dravet syndrome are still unknown due to the scarce availability of this brain tissue. Here we studied, for the first time, human GABAA -evoked currents using cortical brain tissue from Dravet syndrome patients. METHODS: We transplanted in Xenopus oocytes cell membranes obtained from brain tissues of autopsies of Dravet syndrome patients, tuberous sclerosis complex patients as a pathological comparison, and age-matched controls. Additionally, experiments were performed on oocytes expressing human α1ß2γ2 and α1ß2 GABAA receptors. GABAA currents were recorded using the two-microelectrodes voltage-clamp technique. Quantitative real-time polymerase chain reaction, immunohistochemistry, and double-labeling techniques were carried out on the same tissue samples. RESULTS: We found (1) a decrease in GABA sensitivity in Dravet syndrome compared to controls, which was related to an increase in α4- relative to α1-containing GABAA receptors; (2) a shift of the GABA reversal potential toward more depolarizing values in Dravet syndrome, and a parallel increase of the chloride transporters NKCC1/KCC2 expression ratio; (3) an increase of GABAA currents induced by low doses of cannabidiol both in Dravet syndrome and tuberous sclerosis complex comparable to that induced by a classical benzodiazepine, flunitrazepam, that still persists in γ-less GABAA receptors. SIGNIFICANCE: Our study indicates that a dysfunction of the GABAergic system, considered as a feature of brain immaturity, together with defective sodium channels, can contribute to a general reduction of inhibitory efficacy in Dravet brain, suggesting that GABAA receptors could be a target for new therapies.


Asunto(s)
Corteza Cerebral/patología , Epilepsias Mioclónicas/patología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Adulto , Animales , Anticonvulsivantes/farmacología , Cannabidiol/farmacología , Membrana Celular/trasplante , Corteza Cerebral/ultraestructura , Niño , Preescolar , Estimulación Eléctrica , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Femenino , Humanos , Larva , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , ARN Mensajero/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Xenopus , Ácido gamma-Aminobutírico/farmacología
18.
Epilepsia ; 59(10): 1931-1944, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30194729

RESUMEN

OBJECTIVE: Because brain inflammation may contribute to the pathophysiology of temporal lobe epilepsy (TLE), we investigated the expression of various inflammatory markers of the innate and adaptive immune system in the epileptogenic human and rat hippocampus in relation to seizure activity and blood-brain barrier (BBB) dysfunction. METHODS: Immunohistochemistry was performed using various immune cell markers (for microglia, monocytes, macrophages, T lymphocytes, and dendritic cells) on hippocampal sections of drug-resistant TLE patients and patients who died after status epilepticus. The expression of these markers was also studied in the electrical post-status epilepticus rat model for TLE, during the acute, latent, and chronic epileptic phase. BBB dysfunction was assessed using albumin immunohistochemistry and the BBB tracer fluorescein. RESULTS: Monocyte infiltration, microglia, and perivascular macrophage activation were persistently increased in both epileptogenic human and rat hippocampus, whereas T lymphocytes and dendritic cells were not or were scarcely detected. In addition to this, increased expression of C-C motif ligand 2 (CCL2) and osteopontin was observed. In humans, the expression of CD68 and CCL2 was related to the duration of epilepsy and type of pathology. In rats, the expression of CD68, CCL2, and the perivascular macrophage marker CD163 was related to the duration of the initial insult and to the number of spontaneous seizures. Interestingly, the number of CD163-positive perivascular macrophages was also positively correlated to BBB dysfunction in chronic epileptic rats. SIGNIFICANCE: These data suggest a proepileptogenic role for monocytes/macrophages and other cells of the innate immune response, possibly via increased BBB leakage, and indicate that T cells and dendritic cells, which are closely associated with the adaptive immune response, are only sparsely infiltrated during epileptogenesis in the electrical post-status epilepticus rat model. Future studies should reveal the relative importance of these immune cells and whether specific manipulation can modify or prevent epileptogenesis.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Epilepsia del Lóbulo Temporal , Sistema Inmunológico/fisiopatología , Estado Epiléptico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Electroencefalografía , Epilepsia del Lóbulo Temporal/complicaciones , Epilepsia del Lóbulo Temporal/inmunología , Epilepsia del Lóbulo Temporal/patología , Femenino , Fluoresceína/metabolismo , Regulación de la Expresión Génica/inmunología , Humanos , Masculino , Osteopontina/metabolismo , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/complicaciones , Estado Epiléptico/inmunología , Estado Epiléptico/patología
19.
Brain ; 140(7): 1885-1899, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575153

RESUMEN

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented disulfide HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Asunto(s)
Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Epilepsia/tratamiento farmacológico , Dominios HMG-Box/efectos de los fármacos , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Isotiocianatos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Animales , Astrocitos/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad , Quimioterapia Combinada , Epilepsia/metabolismo , Proteína HMGB1/biosíntesis , Hipocampo/metabolismo , Isotiocianatos/farmacología , Masculino , Degeneración Nerviosa/dietoterapia , Neuronas/metabolismo , Ratas , Sulfóxidos
20.
Epilepsia ; 58(3): 315-330, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27883181

RESUMEN

Neuroimaging offers a wide range of opportunities to obtain information about neuronal activity, brain inflammation, blood-brain barrier alterations, and various molecular alterations during epileptogenesis or for the prediction of pharmacoresponsiveness as well as postoperative outcome. Imaging biomarkers were examined during the XIII Workshop on Neurobiology of Epilepsy (XIII WONOEP) organized in 2015 by the Neurobiology Commission of the International League Against Epilepsy (ILAE). Here we present an extended summary of the discussed issues and provide an overview of the current state of knowledge regarding the biomarker potential of different neuroimaging approaches for epilepsy.


Asunto(s)
Biomarcadores , Epilepsia , Neuroimagen , Biomarcadores/metabolismo , Barrera Hematoencefálica/fisiopatología , Educación , Epilepsia/diagnóstico , Epilepsia/metabolismo , Epilepsia/terapia , Humanos , Neurobiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA