Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Neurobiol Dis ; 124: 531-543, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30625365

RESUMEN

Temporal Lobe Epilepsy (TLE) is frequently associated with changes in protein composition and post-translational modifications (PTM) that exacerbate the disorder. O-linked-ß-N-acetyl glucosamine (O-GlcNAc) is a PTM occurring at serine/threonine residues that is derived from and closely associated with metabolic substrates. The enzymes O-GlcNActransferase (OGT) and O-GlcNAcase (OGA) mediate the addition and removal, respectively, of the O-GlcNAc modification. The goal of this study was to characterize OGT/OGA and protein O-GlcNAcylation in the epileptic hippocampus and to determine and whether direct manipulation of these proteins and PTM's alter epileptiform activity. We observed reduced global and protein specific O-GlcNAcylation and OGT expression in the kainate rat model of TLE and in human TLE hippocampal tissue. Inhibiting OGA with Thiamet-G elevated protein O-GlcNAcylation, and decreased both seizure duration and epileptic spike events, suggesting that OGA may be a therapeutic target for seizure control. These findings suggest that loss of O-GlcNAc homeostasis in the kainate model and in human TLE can be reversed via targeting of O-GlcNAc related pathways.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Glucosamina/metabolismo , Hipocampo/metabolismo , Homeostasis/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Animales , Histona Acetiltransferasas/metabolismo , Humanos , Masculino , N-Acetilglucosaminiltransferasas/metabolismo , Ratas , Ratas Sprague-Dawley
2.
J Neurosci ; 37(34): 8207-8215, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28760863

RESUMEN

O-GlcNAcylation is a ubiquitous and dynamic post-translational modification involving the O-linkage of ß-N-acetylglucosamine to serine/threonine residues of membrane, cytosolic, and nuclear proteins. This modification is similar to phosphorylation and regarded as a key regulator of cell survival and homeostasis. Previous studies have shown that phosphorylation of serine residues on synaptic proteins is a major regulator of synaptic strength and long-term plasticity, suggesting that O-GlcNAcylation of synaptic proteins is likely as important as phosphorylation; however, few studies have investigated its role in synaptic efficacy. We recently demonstrated that acutely increasing O-GlcNAcylation induces a novel form of LTD at CA3-CA1 synapses, O-GlcNAc LTD. Here, using hippocampal slices from young adult male rats and mice, we report that epileptiform activity at CA3-CA1 synapses, generated by GABAAR inhibition, is significantly attenuated when protein O-GlcNAcylation is pharmacologically increased. This dampening effect is lost in slices from GluA2 KO mice, indicating a requirement of GluA2-containing AMPARs, similar to expression of O-GlcNAc LTD. Furthermore, we find that increasing O-GlcNAcylation decreases spontaneous CA3 pyramidal cell activity under basal and hyperexcitable conditions. This dampening effect was also observed on cortical hyperexcitability during in vivo EEG recordings in awake mice where the effects of the proconvulsant pentylenetetrazole are attenuated by acutely increasing O-GlcNAcylation. Collectively, these data demonstrate that the post-translational modification, O-GlcNAcylation, is a novel mechanism by which neuronal and synaptic excitability can be regulated, and suggest the possibility that increasing O-GlcNAcylation could be a novel therapeutic target to treat seizure disorders and epilepsy.SIGNIFICANCE STATEMENT We recently reported that an acute pharmacological increase in protein O-GlcNAcylation induces a novel form of long-term synaptic depression at hippocampal CA3-CA1 synapses (O-GlcNAc LTD). This synaptic dampening effect on glutamatergic networks suggests that increasing O-GlcNAcylation will depress pathological hyperexcitability. Using in vitro and in vivo models of epileptiform activity, we show that acutely increasing O-GlcNAc levels can significantly attenuate ongoing epileptiform activity and prophylactically dampen subsequent seizure activity. Together, our findings support the conclusion that protein O-GlcNAcylation is a regulator of neuronal excitability, and it represents a promising target for further research on seizure disorder therapeutics.


Asunto(s)
Acetilglucosamina/metabolismo , Epilepsia/metabolismo , Epilepsia/fisiopatología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Depresión Sináptica a Largo Plazo/fisiología , Animales , Epilepsia/prevención & control , Femenino , Glicosilación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Ratas Sprague-Dawley
3.
Hum Mol Genet ; 24(23): 6667-74, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26358779

RESUMEN

CD2-associated protein (CD2AP) is a leading genetic risk factor for Alzheimer's disease, but little is known about the function of CD2AP in the brain. We studied CD2AP(-/-) mice to address this question. Because CD2AP(-/-) mice normally die by 6 weeks from nephrotic syndrome, we used mice that also express a CD2AP transgene in the kidney, but not brain, to attenuate this phenotype. CD2AP-deficient mice had no behavioral abnormalities except for mild motor and anxiety deficits in a subset of CD2AP(-/-) mice exhibiting severe nephrotic syndrome, associated with systemic illness. Pentylenetetrazol (PTZ)-induced seizures occurred with shorter latency in CD2AP(-/-) mice, but characteristics of these seizures on electroencephalography were not altered. As CD2AP is expressed in brain-adjacent endothelial cells, we hypothesized that the shorter latency to seizures without detectably different seizure characteristics may be due to increased penetration of PTZ related to compromised blood-brain barrier integrity. Using sodium fluorescein extravasation, we found that CD2AP(-/-) mice had reduced blood-brain barrier integrity. Neither seizure severity nor blood-brain barrier integrity was correlated with nephrotic syndrome, indicating that these effects are dissociable from the systemic illness associated with CD2AP deficiency. Confirming this dissociation, wild-type mice with induced nephrotic syndrome maintained an intact blood-brain barrier. Taken together, our results support a role of CD2AP in mediating blood-brain barrier integrity and suggest that cerebrovascular roles of CD2AP could contribute to its effects on Alzheimer's disease risk.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Barrera Hematoencefálica/metabolismo , Proteínas del Citoesqueleto/genética , Enfermedad de Alzheimer/genética , Animales , Ansiedad/genética , Ansiedad/metabolismo , Electroencefalografía , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Actividad Motora/genética , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo
4.
J Neurosci ; 35(15): 6221-30, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878292

RESUMEN

Neuronal hyperexcitability occurs early in the pathogenesis of Alzheimer's disease (AD) and contributes to network dysfunction in AD patients. In other disorders with neuronal hyperexcitability, dysfunction in the dendrites often contributes, but dendritic excitability has not been directly examined in AD models. We used dendritic patch-clamp recordings to measure dendritic excitability in the CA1 region of the hippocampus. We found that dendrites, more so than somata, of hippocampal neurons were hyperexcitable in mice overexpressing Aß. This dendritic hyperexcitability was associated with depletion of Kv4.2, a dendritic potassium channel important for regulating dendritic excitability and synaptic plasticity. The antiepileptic drug, levetiracetam, blocked Kv4.2 depletion. Tau was required, as crossing with tau knock-out mice also prevented both Kv4.2 depletion and dendritic hyperexcitability. Dendritic hyperexcitability induced by Kv4.2 deficiency exacerbated behavioral deficits and increased epileptiform activity in hAPP mice. We conclude that increased dendritic excitability, associated with changes in dendritic ion channels including Kv4.2, may contribute to neuronal dysfunction in early stages AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Región CA1 Hipocampal/patología , Dendritas/fisiología , Neuronas/patología , Canales de Potasio Shal/deficiencia , Proteínas tau/deficiencia , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Ondas Encefálicas/efectos de los fármacos , Ondas Encefálicas/genética , Región CA1 Hipocampal/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Técnicas In Vitro , Levetiracetam , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Nootrópicos/farmacología , Piracetam/análogos & derivados , Piracetam/farmacología , Canales de Potasio Shal/genética , Proteínas tau/genética
5.
J Neurosci ; 35(8): 3330-45, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716834

RESUMEN

Epilepsy is one of the most common chronic neurologic diseases, yet approximately one-third of affected patients do not respond to anticonvulsive drugs that target neurons or neuronal circuits. Reactive astrocytes are commonly found in putative epileptic foci and have been hypothesized to be disease contributors because they lose essential homeostatic capabilities. However, since brain pathology induces astrocytes to become reactive, it is difficult to distinguish whether astrogliosis is a cause or a consequence of epileptogenesis. We now present a mouse model of genetically induced, widespread chronic astrogliosis after conditional deletion of ß1-integrin (Itgß1). In these mice, astrogliosis occurs in the absence of other pathologies and without BBB breach or significant inflammation. Electroencephalography with simultaneous video recording revealed that these mice develop spontaneous seizures during the first six postnatal weeks of life and brain slices show neuronal hyperexcitability. This was not observed in mice with neuronal-targeted ß1-integrin deletion, supporting the hypothesis that astrogliosis is sufficient to induce epileptic seizures. Whole-cell patch-clamp recordings from astrocytes further suggest that the heightened excitability was associated with impaired astrocytic glutamate uptake. Moreover, the relative expression of the cation-chloride cotransporters (CCC) NKCC1 (Slc12a2) and KCC2 (Slc12a5), which are responsible for establishing the neuronal Cl(-) gradient that governs GABAergic inhibition were altered and the NKCC1 inhibitor bumetanide eliminated seizures in a subgroup of mice. These data suggest that a shift in the relative expression of neuronal NKCC1 and KCC2, similar to that observed in immature neurons during development, may contribute to astrogliosis-associated seizures.


Asunto(s)
Astrocitos/metabolismo , Gliosis/metabolismo , Integrina beta1/metabolismo , Convulsiones/metabolismo , Potenciales de Acción , Animales , Astrocitos/fisiología , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Gliosis/fisiopatología , Ácido Glutámico/metabolismo , Integrina beta1/genética , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Convulsiones/etiología , Convulsiones/patología , Convulsiones/fisiopatología , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Simportadores/genética , Simportadores/metabolismo , Cotransportadores de K Cl
6.
Glia ; 63(1): 23-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25066727

RESUMEN

Seizures frequently accompany gliomas and often escalate to peritumoral epilepsy. Previous work revealed the importance of tumor-derived excitatory glutamate (Glu) release mediated by the cystine-glutamate transporter (SXC) in epileptogenesis. We now show a novel contribution of GABAergic disinhibition to disease pathophysiology. In a validated mouse glioma model, we found that peritumoral parvalbumin-positive GABAergic inhibitory interneurons are significantly reduced, corresponding with deficits in spontaneous and evoked inhibitory neurotransmission. Most remaining peritumoral neurons exhibit elevated intracellular Cl(-) concentration ([Cl(-) ]i ) and consequently depolarizing, excitatory gamma-aminobutyric acid (GABA) responses. In these neurons, the plasmalemmal expression of KCC2, which establishes the low [Cl(-) ]i required for GABAA R-mediated inhibition, is significantly decreased. Interestingly, reductions in inhibition are independent of Glu release, but the presence of both decreased inhibition and decreased SXC expression is required for epileptogenesis. We suggest GABAergic disinhibition renders peritumoral neuronal networks hyper-excitable and susceptible to seizures triggered by excitatory stimuli, and propose KCC2 as a therapeutic target.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Epilepsia/metabolismo , Glioma/metabolismo , Receptores de GABA-A/metabolismo , Simportadores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Neoplasias Encefálicas/complicaciones , Epilepsia/etiología , Femenino , Glioma/complicaciones , Interneuronas/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Cotransportadores de K Cl
7.
Epilepsia ; 55(12): e139-42, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25385326

RESUMEN

Studies have demonstrated that the membrane attack complex (MAC) of complement can evoke seizures when injected directly into rodent brain. In the course of studies that examine the role of complement in the development of experimental cerebral malaria (ECM), we observed fewer seizures in mice deficient in C5, a component required for MAC formation. To determine if the MAC contributed to the tonic-clonic seizures characteristic of ECM, we performed long-term video-electroencephalography (EEG) on C5(-/-) mice with Plasmodium berghei ANKA-induced cerebral malaria and observed significantly reduced spike and seizure frequency compared to wild-type mice. Our data suggest a role for the MAC in malaria-induced seizures and that inhibition of the terminal complement pathway may reduce seizures and seizure-related neurocognitive deficits.


Asunto(s)
Complemento C5/deficiencia , Malaria Cerebral/complicaciones , Convulsiones , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Malaria Cerebral/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/patogenicidad , Convulsiones/etiología , Convulsiones/genética , Convulsiones/prevención & control
8.
bioRxiv ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38798487

RESUMEN

Tau reduction is a promising therapeutic strategy for Alzheimer's disease. In numerous models, tau reduction via genetic knockout is beneficial, at least in part due to protection against hyperexcitability and seizures, but the underlying mechanisms are unclear. Here we describe the generation and initial study of a new conditional Tau flox model to address these mechanisms. Given the protective effects of tau reduction against hyperexcitability, we compared the effects of selective tau reduction in excitatory or inhibitory neurons. Tau reduction in excitatory neurons mimicked the protective effects of global tau reduction, while tau reduction in inhibitory neurons had the opposite effect and increased seizure susceptibility. Since most prior studies used knockout mice lacking tau throughout development, we crossed Tau flox mice with inducible Cre mice and found beneficial effects of tau reduction in adulthood. Our findings support the effectiveness of tau reduction in adulthood and indicate that excitatory neurons may be a key site for its excitoprotective effects. SUMMARY: A new conditional tau knockout model was generated to study the protective effects of tau reduction against hyperexcitability. Conditional tau reduction in excitatory, but not inhibitory, neurons was excitoprotective, and induced tau reduction in adulthood was excitoprotective without adverse effects.

9.
Epilepsia ; 53(8): 1360-70, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22709330

RESUMEN

PURPOSE: Patients with gliomas frequently present with seizures, but the factors associated with seizure development are still poorly understood. In this study, we assessed peritumoral synaptic network activity in a glioma animal model and tested the contribution of aberrant glutamate release from gliomas on glioma-associated epileptic network activity. METHODS: In vitro brain slices were made from glioma-implanted mice. Using extracellular field recordings, we analyzed peritumoral epileptiform activity induced by Mg(2+)-free medium in slices from tumor-bearing animals and sham-operated controls. We assessed the effect of sulfasalazine (SAS), a blocker of system and glutamate release, on spontaneous and evoked activity in tumor-associated slices. KEY FINDINGS: Tumor-associated cortical networks were hyperexcitable. The onset latency of Mg(2+)-free-induced epileptiform activity was significantly shorter in tumor-bearing slices, and the incidence of Mg(2+)-free-induced ictal-like events was higher. Block of glutamate release from system decreased the response area of evoked activity and completely blocked Mg(2+)-free-induced ictal-like, but not interictal-like events. SIGNIFICANCE: Control of seizures in patients with gliomas is an essential component of clinical management; therefore, understanding the origin of seizures is vital. This work provides evidence that peritumoral synaptic network activity is disrupted by tumor masses resulting in network excitability. We show that blocking glutamate release via system with SAS, a drug already approved by the U.S. Food and Drug Administration (FDA), can inhibit Mg(2+)-free-induced ictal-like epileptiform events similar to other chemicals used to decrease seizure activity. We, therefore, suggest that further studies should consider SAS a promising agent to aid in the treatment of seizures associated with gliomas.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Glioma/fisiopatología , Red Nerviosa/fisiopatología , Convulsiones/fisiopatología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Neoplasias Encefálicas/complicaciones , Modelos Animales de Enfermedad , Electrofisiología , Femenino , Glioma/complicaciones , Ácido Glutámico/fisiología , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Convulsiones/etiología , Sulfasalazina/farmacología , Células Tumorales Cultivadas
10.
Ann Clin Transl Neurol ; 2(4): 401-16, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25909085

RESUMEN

OBJECTIVE: Temporal lobe epilepsy (TLE) patients exhibit signs of memory impairments even when seizures are pharmacologically controlled. Surprisingly, the underlying molecular mechanisms involved in TLE-associated memory impairments remain elusive. Memory consolidation requires epigenetic transcriptional regulation of genes in the hippocampus; therefore, we aimed to determine how epigenetic DNA methylation mechanisms affect learning-induced transcription of memory-permissive genes in the epileptic hippocampus. METHODS: Using the kainate rodent model of TLE and focusing on the brain-derived neurotrophic factor (Bdnf) gene as a candidate of DNA methylation-mediated transcription, we analyzed DNA methylation levels in epileptic rats following learning. After detection of aberrant DNA methylation at the Bdnf gene, we investigated functional effects of altered DNA methylation on hippocampus-dependent memory formation in our TLE rodent model. RESULTS: We found that behaviorally driven BdnfDNA methylation was associated with hippocampus-dependent memory deficits. Bisulfite sequencing revealed that decreased BdnfDNA methylation levels strongly correlated with abnormally high levels of BdnfmRNA in the epileptic hippocampus during memory consolidation. Methyl supplementation via methionine (Met) increased BdnfDNA methylation and reduced BdnfmRNA levels in the epileptic hippocampus during memory consolidation. Met administration reduced interictal spike activity, increased theta rhythm power, and reversed memory deficits in epileptic animals. The rescue effect of Met treatment on learning-induced BdnfDNA methylation, Bdnf gene expression, and hippocampus-dependent memory, were attenuated by DNA methyltransferase blockade. INTERPRETATION: Our findings suggest that manipulation of DNA methylation in the epileptic hippocampus should be considered as a viable treatment option to ameliorate memory impairments associated with TLE.

11.
Sci Transl Med ; 7(289): 289ra86, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019222

RESUMEN

Glioma is the most common malignant primary brain tumor. Its rapid growth is aided by tumor-mediated glutamate release, creating peritumoral excitotoxic cell death and vacating space for tumor expansion. Glioma glutamate release may also be responsible for seizures, which complicate the clinical course for many patients and are often the presenting symptom. A hypothesized glutamate release pathway is the cystine/glutamate transporter System xc (-) (SXC), responsible for the cellular synthesis of glutathione (GSH). However, the relationship of SXC-mediated glutamate release, seizures, and tumor growth remains unclear. Probing expression of SLC7A11/xCT, the catalytic subunit of SXC, in patient and mouse-propagated tissues, we found that ~50% of patient tumors have elevated SLC7A11 expression. Compared with tumors lacking this transporter, in vivo propagated and intracranially implanted SLC7A11-expressing tumors grew faster, produced pronounced peritumoral glutamate excitotoxicity, induced seizures, and shortened overall survival. In agreement with animal data, increased SLC7A11 expression predicted shorter patient survival according to genomic data in the REMBRANDT (National Institutes of Health Repository for Molecular Brain Neoplasia Data) database. In a clinical pilot study, we used magnetic resonance spectroscopy to determine SXC-mediated glutamate release by measuring acute changes in glutamate after administration of the U.S. Food and Drug Administration-approved SXC inhibitor, sulfasalazine (SAS). In nine glioma patients with biopsy-confirmed SXC expression, we found that expression positively correlates with glutamate release, which is acutely inhibited with oral SAS. These data suggest that SXC is the major pathway for glutamate release from gliomas and that SLC7A11 expression predicts accelerated growth and tumor-associated seizures.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Glioma/complicaciones , Glioma/metabolismo , Convulsiones/complicaciones , Convulsiones/metabolismo , Edema/patología , Genómica , Glioma/clasificación , Glutamatos/metabolismo , Humanos , Neuronas/efectos de los fármacos , Neuronas/patología , Neurotoxinas/toxicidad , Análisis de Supervivencia
12.
Neurochem Int ; 63(7): 696-701, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23385090

RESUMEN

Seizures are a serious and debilitating co-morbidity of primary brain tumors that affect most patients, yet their etiology is poorly understood. In many CNS pathologies, including epilepsy and brain injury, high levels of extracellular glutamate have been implicated in seizure generation. It has been shown that gliomas release neurotoxic levels of glutamate through their high expression of system xc-. More recently it was shown that the surrounding peritumoral cortex is spontaneously hyperexcitable. In this review, we discuss how gliomas induce changes in the surrounding environment that may further contribute to elevated extracellular glutamate and tumor-associated seizures. Peritumoral astrocytes become reactive and lose their ability to remove glutamate, while microglia, in response to signals from glioma cells, may release glutamate. In addition, gliomas increase blood brain barrier permeability, allowing seizure-inducing serum components, including glutamate, into the peritumoral region. These factors, working together or alone, may influence the frequency and severity of tumor-associated epilepsy.


Asunto(s)
Epilepsia/etiología , Ácido Glutámico/fisiología , Neoplasias/complicaciones , Neuroglía/patología , Barrera Hematoencefálica , Epilepsia/metabolismo , Epilepsia/patología , Homeostasis , Humanos , Potasio/metabolismo
13.
Nat Med ; 17(10): 1269-74, 2011 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-21909104

RESUMEN

Epileptic seizures are a common and poorly understood comorbidity for individuals with primary brain tumors. To investigate peritumoral seizure etiology, we implanted human-derived glioma cells into severe combined immunodeficient mice. Within 14-18 d, glioma-bearing mice developed spontaneous and recurring abnormal electroencephalogram events consistent with progressive epileptic activity. Acute brain slices from these mice showed marked glutamate release from the tumor mediated by the system x(c)(-) cystine-glutamate transporter (encoded by Slc7a11). Biophysical and optical recordings showed glutamatergic epileptiform hyperexcitability that spread into adjacent brain tissue. We inhibited glutamate release from the tumor and the ensuing hyperexcitability by sulfasalazine (SAS), a US Food and Drug Administration-approved drug that blocks system x(c)(-). We found that acute administration of SAS at concentrations equivalent to those used to treat Crohn's disease in humans reduced epileptic event frequency in tumor-bearing mice compared with untreated controls. SAS should be considered as an adjuvant treatment to ameliorate peritumoral seizures associated with glioma in humans.


Asunto(s)
Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/metabolismo , Epilepsia/etiología , Glioma/metabolismo , Ácido Glutámico/metabolismo , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/metabolismo , Análisis de Varianza , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Trasplante de Células , Electroencefalografía , Electrofisiología , Epilepsia/patología , Glioma/tratamiento farmacológico , Humanos , Ratones , Ratones SCID , Sulfasalazina/farmacología , Sulfasalazina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA