Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Acta Neuropathol ; 142(4): 729-759, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34292399

RESUMO

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.


Assuntos
Epilepsia do Lobo Temporal/complicações , Hipocampo/metabolismo , Distúrbios do Metabolismo do Ferro/etiologia , Ferro/metabolismo , Estado Epiléptico/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Estudos de Casos e Controles , Técnicas de Cultura de Células , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/patologia , Feminino , Humanos , Distúrbios do Metabolismo do Ferro/patologia , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo/fisiologia , Ratos , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia
2.
Sci Rep ; 7(1): 8089, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808237

RESUMO

Tuberous Sclerosis Complex (TSC) is a rare genetic disorder that results from a mutation in the TSC1 or TSC2 genes leading to constitutive activation of the mechanistic target of rapamycin complex 1 (mTORC1). TSC is associated with autism, intellectual disability and severe epilepsy. Cortical tubers are believed to represent the neuropathological substrates of these disabling manifestations in TSC. In the presented study we used high-throughput RNA sequencing in combination with systems-based computational approaches to investigate the complexity of the TSC molecular network. Overall we detected 438 differentially expressed genes and 991 differentially expressed small non-coding RNAs in cortical tubers compared to autopsy control brain tissue. We observed increased expression of genes associated with inflammatory, innate and adaptive immune responses. In contrast, we observed a down-regulation of genes associated with neurogenesis and glutamate receptor signaling. MicroRNAs represented the largest class of over-expressed small non-coding RNA species in tubers. In particular, our analysis revealed that the miR-34 family (including miR-34a, miR-34b and miR-34c) was significantly over-expressed. Functional studies demonstrated the ability of miR-34b to modulate neurite outgrowth in mouse primary hippocampal neuronal cultures. This study provides new insights into the TSC transcriptomic network along with the identification of potential new treatment targets.


Assuntos
Transcrição Gênica/genética , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/genética , Esclerose Tuberosa/genética , Adolescente , Adulto , Animais , Córtex Cerebral/fisiologia , Criança , Pré-Escolar , Epilepsia/genética , Feminino , Humanos , Lactente , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Pessoa de Meia-Idade , Mutação/genética , Neurônios/fisiologia , Transdução de Sinais/genética , Adulto Jovem
3.
Epilepsia ; 57(1): 70-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26691741

RESUMO

OBJECTIVE: Blood-brain barrier (BBB) leakage may play a pro-epileptogenic role after status epilepticus. In the accompanying contrast-enhanced magnetic resonance imaging (CE-MRI) study we showed that the mammalian target of rapamycin (mTOR) inhibitor rapamycin reduced BBB leakage and seizure activity during the chronic epileptic phase. Given rapamycin's role in growth and immune response, the potential therapeutic effects of rapamycin after status epilepticus with emphasis on brain inflammation and brain vasculature were investigated. METHODS: Seven weeks after kainic acid-induced status epilepticus, rats were perfusion fixed and (immuno)histochemistry was performed using several glial and vascular markers. In addition, an in vitro model for the human BBB was used to determine the effects of rapamycin on transendothelial electrical resistance as a measure for BBB integrity. RESULTS: (Immuno)histochemistry showed that local blood vessel density, activated microglia, and astrogliosis were reduced in rapamycin-treated rats compared to vehicle-treated rats. In vitro studies showed that rapamycin could attenuate TNFα-induced endothelial barrier breakdown. SIGNIFICANCE: These data suggest that rapamycin improves BBB function during the chronic epileptic phase by a reduction of local brain inflammation and blood vessel density that can contribute to a milder form of epilepsy.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Encéfalo/metabolismo , Imunossupressores/efeitos adversos , Sirolimo/efeitos adversos , Estado Epiléptico/tratamento farmacológico , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Impedância Elétrica , Agonistas de Aminoácidos Excitatórios/toxicidade , Gliose/etiologia , Gliose/metabolismo , Gliose/patologia , Ácido Caínico/toxicidade , Lectinas/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Fibras Musgosas Hipocampais/metabolismo , Fibras Musgosas Hipocampais/patologia , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Fator de Necrose Tumoral alfa/farmacologia
4.
Epilepsy Res ; 108(3): 396-404, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24556423

RESUMO

It has been reported that glucocorticoids (GCs) can effectively control seizures in pediatric epilepsy syndromes, possibly by inhibition of inflammation. Since inflammation is supposed to be involved in epileptogenesis, we hypothesized that treatment with GCs would reduce brain inflammation and thereby modify epileptogenesis in a rat model for temporal lobe epilepsy, in which epilepsy gradually develops after electrically induced status epilepticus (SE). To prevent the severe adverse effects that are inevitable with long-term GC treatment, we used liposome nanotechnology (G-Technology(®)) to enhance the sustained delivery to the brain. Starting 4h after onset of SE, rats were treated with glutathione pegylated liposomal methylprednisolone (GSH-PEG liposomal MP) according to a treatment protocol (1× per week; 10mg/kg) that is effective in other models of neuroinflammation. Continuous electro-encephalogram (EEG) recordings revealed that SE duration and onset of spontaneous seizures were not affected by GSH-PEG liposomal MP treatment. The number and duration of spontaneous seizures were also not different between vehicle and GSH-PEG liposomal MP-treated animals. Six weeks after SE, brain inflammation, as assessed by quantification of microglia activation, was not reduced by GSH-PEG liposomal MP-treatment. Also, neuronal cell loss and mossy fiber sprouting were not affected. Our study shows that the selected GSH-PEG liposomal MP treatment regimen that was administered beyond the acute SE phase does not reduce brain inflammation and development of temporal lobe epilepsy.


Assuntos
Anti-Inflamatórios/administração & dosagem , Glutationa/administração & dosagem , Metilprednisolona/administração & dosagem , Fosfolipídeos/administração & dosagem , Estado Epiléptico/tratamento farmacológico , Albuminas , Animais , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Eletrodos Implantados/efeitos adversos , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Metilprednisolona/sangue , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/etiologia , Fatores de Tempo
5.
Epilepsia ; 54(4): 589-95, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23398413

RESUMO

PURPOSE: Brain inflammation occurs during epileptogenesis and may contribute to the development and progression of temporal lobe epilepsy. Recently, several studies have indicated that seizures may also increase specific blood plasma cytokine levels in animal models as well as in human patients with epilepsy, suggesting that peripheral inflammation may serve as a biomarker for epilepsy. Moreover, studies in epilepsy animal models have shown that peripheral inflammation may play either a pathogenic or neuroprotective role. METHODS: We evaluated the inflammatory response in blood plasma after electrically induced status epilepticus (SE) in a rat model for temporal lobe epilepsy. We measured blood plasma levels of the inflammation markers interleukin 1ß (IL-1ß), interleukin 6 (IL-6), by enzyme-linked immunosorbent assays (ELISAs) and C-reactive protein (CRP) by immunoturbidimetry, at 1 day after SE (acute period), at 1 week (during the latent period), and at 2 months after SE, which is the chronic epileptic phase when spontaneous seizures occur. Plasma levels were also measured during pilocarpine-induced SE. These were compared with plasma levels after lipopolysaccharide injection, which causes sepsis. KEY FINDINGS: Although sepsis induced a huge surge in IL-1ß and IL-6 levels, we did not detect a change in IL-1ß, IL-6, or CRP plasma levels at any time point after electrically induced SE compared to control animals. SE induced by pilocarpine produced a rise in IL-6 and CRP but not IL-1ß levels. SIGNIFICANCE: These findings suggest that plasma levels of these inflammatory proteins cannot be used as biomarkers for temporal lobe epileptogenesis.


Assuntos
Biomarcadores/sangue , Epilepsia do Lobo Temporal/sangue , Mediadores da Inflamação/sangue , Estado Epiléptico/sangue , Animais , Proteína C-Reativa/metabolismo , Eletroencefalografia , Ensaio de Imunoadsorção Enzimática , Epilepsia do Lobo Temporal/induzido quimicamente , Imunoquímica , Interleucina-1beta/sangue , Interleucina-6/sangue , Lipopolissacarídeos , Masculino , Agonistas Muscarínicos , Pilocarpina , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente
6.
J Neuroinflammation ; 9: 280, 2012 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-23270518

RESUMO

OBJECTIVE: Decreased expression of inwardly rectifying potassium (Kir) channels in astrocytes and glioma cells may contribute to impaired K⁺ buffering and increased propensity for seizures. Here, we evaluated the potential effect of inflammatory molecules, such as interleukin-1ß (IL-1ß) on Kir4.1 mRNA and protein expression. METHODS: We investigated Kir4.1 (Kcnj10) and IL-1ß mRNA expression in the temporal cortex in a rat model of temporal lobe epilepsy 24 h and 1 week after induction of status epilepticus (SE), using real-time PCR and western blot analysis. The U373 glioblastoma cell line and human fetal astrocytes were used to study the regulation of Kir4.1 expression in response to pro-inflammatory cytokines. Expression of Kir4.1 protein was also evaluated by means of immunohistochemistry in surgical specimens of patients with astrocytic tumors (n = 64), comparing the expression in tumor patients with (n = 38) and without epilepsy (n = 26). RESULTS: Twenty-four hours after onset of SE, Kir4.1 mRNA and protein were significantly down-regulated in temporal cortex of epileptic rats. This decrease in expression was followed by a return to control level at 1 week after SE. The transient downregulation of Kir4.1 corresponded to the time of prominent upregulation of IL-1ß mRNA. Expression of Kir4.1 mRNA and protein in glial cells in culture was downregulated after exposure to IL-1ß. Evaluation of Kir4.1 in tumor specimens showed a significantly lower Kir4.1 expression in the specimens of patients with epilepsy compared to patients without epilepsy. This paralleled the increased presence of activated microglial cells, as well as the increased expression of IL-1ß and the cytoplasmic translocation of high mobility group box 1 (HMGB1). CONCLUSIONS: Taken together, these findings indicate that alterations in expression of Kir4.1 occurring in epilepsy-associated lesions are possibly influenced by the local inflammatory environment and in particular by the inflammatory cytokine IL-1ß.


Assuntos
Astrócitos/metabolismo , Astrocitoma/metabolismo , Regulação para Baixo/fisiologia , Interleucina-1beta/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Astrocitoma/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Epilepsia do Lobo Temporal/patologia , Feto , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Interleucina-1beta/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/metabolismo , Ratos , Lobo Temporal/patologia
7.
PLoS One ; 7(9): e44789, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028621

RESUMO

Increasing evidence supports the involvement of microRNAs (miRNA) in the regulation of inflammation in human neurological disorders. In the present study we investigated the role of miR-146a, a key regulator of the innate immune response, in the modulation of astrocyte-mediated inflammation. Using Taqman PCR and in situ hybridization, we studied the expression of miR-146a in epilepsy-associated glioneuronal lesions which are characterized by prominent activation of the innate immune response. In addition, cultured human astrocytes were used to study the regulation of miR-146a expression in response to proinflammatory cytokines. qPCR and western blot were used to evaluate the effects of overexpression or knockdown of miR-146a on IL-1ß signaling. Downstream signaling in the IL-1ß pathway, as well as the expression of IL-6 and COX-2 were evaluated by western blot and ELISA. Release several cytokines was evaluated using a human magnetic multiplex cytokine assay on a Luminex® 100™/200™ platform. Increased expression of miR-146a was observed in glioneuronal lesions by Taqman PCR. MiR-146a expression in human glial cell cultures was strongly induced by IL-1ß and blocked by IL-1ß receptor antagonist. Modulation of miR-146a expression by transfection of astrocytes with anti-miR146a or mimic, regulated the mRNA expression levels of downstream targets of miR-146a (IRAK-1, IRAK-2 and TRAF-6) and the expression of IRAK-1 protein. In addition, the expression of IL-6 and COX-2 upon IL-1ß stimulation was suppressed by increased levels of miR-146a and increased by the reduction of miR-146a. Modulation of miR-146a expression affected also the release of several cytokines such as IL-6 and TNF-α. Our observations indicate that in response to inflammatory cues, miR-146a was induced as a negative-feedback regulator of the astrocyte-mediated inflammatory response. This supports an important role of miR-146a in human neurological disorders associated with chronic inflammation and suggests that this miR may represent a novel target for therapeutic strategies.


Assuntos
Astrócitos/metabolismo , Mediadores da Inflamação/metabolismo , MicroRNAs/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encefalopatias/genética , Encefalopatias/imunologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patologia , Feminino , Ganglioglioma/genética , Ganglioglioma/metabolismo , Ganglioglioma/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/farmacologia , Interleucina-6/metabolismo , Masculino , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/imunologia , Malformações do Desenvolvimento Cortical/metabolismo , Malformações do Desenvolvimento Cortical/patologia , Malformações do Desenvolvimento Cortical do Grupo I , MicroRNAs/genética , NF-kappa B/metabolismo , Gravidez , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo
8.
Epilepsia ; 53(7): 1119-30, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22578218

RESUMO

The mammalian target of rapamycin (mTOR) signaling pathway regulates cell growth, differentiation, proliferation, and metabolism. Loss-of-function mutations in upstream regulators of mTOR have been highly associated with dysplasias, epilepsy, and neurodevelopmental disorders. These include tuberous sclerosis, which is due to mutations in TSC1 or TSC2 genes; mutations in phosphatase and tensin homolog (PTEN) as in Cowden syndrome, polyhydramnios, megalencephaly, symptomatic epilepsy syndrome (PMSE) due to mutations in the STE20-related kinase adaptor alpha (STRADalpha); and neurofibromatosis type 1 attributed to neurofibromin 1 mutations. Inhibition of the mTOR pathway with rapamycin may prevent epilepsy and improve the underlying pathology in mouse models with disrupted mTOR signaling, due to PTEN or TSC mutations. However the timing and duration of its administration appear critical in defining the seizure and pathology-related outcomes. Rapamycin application in human cortical slices from patients with cortical dysplasias reduces the 4-aminopyridine-induced oscillations. In the multiple-hit model of infantile spasms, pulse high-dose rapamycin administration can reduce the cortical overactivation of the mTOR pathway, suppresses spasms, and has disease-modifying effects by partially improving cognitive deficits. In post-status epilepticus models of temporal lobe epilepsy, rapamycin may ameliorate the development of epilepsy-related pathology and reduce the expression of spontaneous seizures, but its effects depend on the timing and duration of administration, and possibly the model used. The observed recurrence of seizures and epilepsy-related pathology after rapamycin discontinuation suggests the need for continuous administration to maintain the benefit. However, the use of pulse administration protocols may be useful in certain age-specific epilepsy syndromes, like infantile spasms, whereas repetitive-pulse rapamycin protocols may suffice to sustain a long-term benefit in genetic disorders of the mTOR pathway. In summary, mTOR dysregulation has been implicated in several genetic and acquired forms of epileptogenesis. The use of mTOR inhibitors can reverse some of these epileptogenic processes, although their effects depend upon the timing and dose of administration as well as the model used.


Assuntos
Anticonvulsivantes/uso terapêutico , Epilepsia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Epilepsia/etiologia , Humanos , Camundongos
9.
Epilepsia ; 52(9): 1645-55, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21635241

RESUMO

PURPOSE: Adenosine kinase (ADK) represents the key metabolic enzyme for the regulation of extracellular adenosine levels in the brain. In adult brain, ADK is primarily present in astrocytes. Several lines of experimental evidence support a critical role of ADK in different types of brain injury associated with astrogliosis, which is also a prominent morphologic feature of temporal lobe epilepsy (TLE). We hypothesized that dysregulation of ADK is an ubiquitous pathologic hallmark of TLE. METHODS: Using immunocytochemistry and Western blot analysis, we investigated ADK protein expression in a rat model of TLE during epileptogenesis and the chronic epileptic phase and compared those findings with tissue resected from TLE patients with mesial temporal sclerosis (MTS). KEY FINDINGS: In rat control hippocampus and cortex, a low baseline expression of ADK was found with mainly nuclear localization. One week after the electrical induction of status epilepticus (SE), prominent up-regulation of ADK became evident in astrocytes with a characteristic cytoplasmic localization. This increase in ADK persisted at least for 3-4 months after SE in rats developing a progressive form of epilepsy. In line with the findings from the rat model, expression of astrocytic ADK was also found to be increased in the hippocampus and temporal cortex of patients with TLE. In addition, in vitro experiments in human astrocyte cultures showed that ADK expression was increased by several proinflammatory molecules (interleukin-1ß and lipopolysaccharide). SIGNIFICANCE: These results suggest that dysregulation of ADK in astrocytes is a common pathologic hallmark of TLE. Moreover, in vitro data suggest the existence of an additional layer of modulatory crosstalk between the astrocyte-based adenosine cycle and inflammation. Whether this interaction also can play a role in vivo needs to be further investigated.


Assuntos
Adenosina Quinase/metabolismo , Astrócitos/metabolismo , Epilepsia do Lobo Temporal/patologia , Regulação para Cima/fisiologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Eletroencefalografia/métodos , Epilepsia do Lobo Temporal/induzido quimicamente , Feto , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/patologia , Humanos , Interleucina-1beta/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Lobo Temporal/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Epilepsia ; 51(9): 1763-73, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20345941

RESUMO

PURPOSE: Induction of inflammatory pathways has been reported in epileptic patients with focal malformations of cortical development. In the present study we examined the innate and adaptive immune responses in focal cortical dysplasia (FCD) with different histopathologic and pathogenetic features. METHODS: The inflammatory cell components and the induction of major proinflammatory pathways and molecules [complement pathway, interleukin (IL)-1ß, and chemokine monocyte chemotactic protein-1 (MCP1)] was investigated in surgical specimens of sporadic type IA and type IIB FCD using immunocytochemical methods. RESULTS: FCD II but not FCD I cases exhibit activation of the mammalian target of rapamycin (mTOR) cascade with strong neuronal expression of the phosphorylated isoform of S6 protein. Microglia reactivity was increased in all lesions (FCD I and II) compared to control tissue; however, the number of HLA-DR-positive cells was significantly higher in FCD II than in FCD I. In FCD II specimens we also observed perivascular and parenchymal T lymphocytes (CD3(+) ), with a predominance of CD8(+) T-cytotoxic/suppressor lymphocytes, as well as a few dendritic cells. Expression of components of the complement cascade, IL-1ß, and MCP1 was prominent in FCD II cases. DISCUSSION: Our findings indicate a prominent activation of both innate and adaptive immunity, with involvement of different inflammatory pathways in FCD II cases, supporting the possible involvement of inflammation in the epileptogenesis of these lesions, as well as the notion that FCD II is pathologically distinct from FCD I.


Assuntos
Imunidade Adaptativa/fisiologia , Epilepsia/imunologia , Imunidade Inata/fisiologia , Malformações do Desenvolvimento Cortical/imunologia , Imunidade Adaptativa/imunologia , Córtex Cerebral/imunologia , Córtex Cerebral/patologia , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Epilepsia/genética , Epilepsia/patologia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Imuno-Histoquímica , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-1/genética , Interleucina-1/imunologia , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/patologia , Microglia/imunologia , Microglia/patologia , Neurônios/imunologia , Neurônios/patologia , Neurônios/fisiologia , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/imunologia
11.
Brain Pathol ; 20(4): 704-19, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19912235

RESUMO

Cortical tubers in patients with tuberous sclerosis complex are associated with disabling neurological manifestations, including intractable epilepsy. While these malformations are believed to result from the effects of TSC1 or TSC2 gene mutations, the molecular mechanisms leading to tuber formation, as well as the onset of seizures, remain largely unknown. We used the Affymetrix Gene Chip platform to provide the first genome-wide investigation of gene expression in surgically resected tubers, compared with histological normal perituberal tissue from the same patients or autopsy control tissue. We identified 2501 differentially expressed genes in cortical tubers compared with autopsy controls. Expression of genes associated with cell adhesion, for example, VCAM1, integrins and CD44, or with the inflammatory response, including complement factors, serpinA3, CCL2 and several cytokines, was increased in cortical tubers, whereas genes related to synaptic transmission, for example, the glial glutamate transporter GLT-1, and voltage-gated channel activity, exhibited lower expression. Gene expression in perituberal cortex was distinct from autopsy control cortex suggesting that even in the absence of tissue pathology the transcriptome is altered in TSC. Changes in gene expression yield insights into new candidate genes that may contribute to tuber formation or seizure onset, representing new targets for potential therapeutic development.


Assuntos
Adesão Celular/genética , Córtex Cerebral/metabolismo , Inflamação/genética , Esclerose Tuberosa/genética , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Córtex Cerebral/patologia , Criança , Pré-Escolar , Feminino , Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Imuno-Histoquímica , Lactente , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esclerose Tuberosa/metabolismo , Esclerose Tuberosa/patologia
12.
Epilepsia ; 50(6): 1409-18, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19220410

RESUMO

PURPOSE: Synaptic vesicle protein 2A (SV2A), the binding site for the antiepileptic drug (AED) levetiracetam, has been shown to be involved in the control of neuronal excitability. The aim of the study was to define the expression and cell-specific distribution of SV2A in developmental focal lesions associated with medically intractable epilepsy. METHODS: SV2A immunocytochemistry and Western blotting was performed in focal cortical dysplasia (FCD type IIB) and cortical tubers from patients with tuberous sclerosis complex (TSC). RESULTS: Autopsy and surgical control neocortical specimens were characterized by strong SV2A immunoreactivity throughout all cortical layers, with punctate labeling around the somata and dendrites of neurons. In FCD and cortical tuber specimens less intense, SV2A immunoreactivity was observed in the neuropil. The reduction in expression was confirmed by Western blot analysis. In both FCD and tuber specimens, clusters of punctate labeling were detected along cell borders and processes (perisomatic synapses) of dysplastic neuronal cells localized in both gray and white matter. The large majority of balloon cells in FCD, or giant cells in tubers, did not show punctate labeling around their somata. SV2A immunoreactivity was observed occasionally within the neuronal perikarya. CONCLUSIONS: The pattern of SV2A immunoreactivity with reduced neuropil expression and altered cellular and subcellular distribution suggests a possible contribution of SV2A to the epileptogenicity of these malformations of cortical development. Knowledge of the expression pattern of SV2A in epilepsy-associated pathologies may be valuable for the evaluation of the effectiveness of AEDs targeting this protein.


Assuntos
Malformações do Desenvolvimento Cortical/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Esclerose Tuberosa/metabolismo , Adolescente , Adulto , Autopsia/métodos , Córtex Cerebral/metabolismo , Criança , Feminino , Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Humanos , Masculino , Malformações do Desenvolvimento Cortical/patologia , Sinaptofisina/metabolismo , Esclerose Tuberosa/patologia , Adulto Jovem
13.
Acta Neuropathol ; 115(6): 683-96, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18317782

RESUMO

Members of the vascular endothelial growth factor (VEGF) family are key signaling proteins in the induction and regulation of angiogenesis, both during development and in pathological conditions. However, signaling mediated through VEGF family proteins and their receptors has recently been shown to have direct effects on neurons and glial cells. In the present study, we immunocytochemically investigated the expression and cellular distribution of VEGFA, VEGFB, and their associated receptors (VEGFR-1 and VEGFR-2) in focal cortical dysplasia (FCD) type IIB from patients with medically intractable epilepsy. Histologically normal temporal cortex and perilesional regions displayed neuronal immunoreactivity (IR) for VEGFA, VEGFB, and VEGF receptors (VEGFR-1 and VEGFR-2), mainly in pyramidal neurons. Weak IR was observed in blood vessels and there was no notable glial IR within the grey and white matter. In all FCD specimens, VEGFA, VEGFB, and both VEGF receptors were highly expressed in dysplastic neurons. IR in astroglial and balloon cells was observed for VEGFA and its receptors. VEGFR-1 displayed strong endothelial staining in FCD. Double-labeling also showed expression of VEGFA, VEGFB and VEGFR-1 in cells of the microglia/macrophage lineage. The neuronal expression of both VEGFA and VEGFB, together with their specific receptors in FCD, suggests autocrine/paracrine effects on dysplastic neurons. These autocrine/paracrine effects could play a role in the development of FCD, preventing the death of abnormal neuronal cells. In addition, the expression of VEGFA and its receptors in glial cells within the dysplastic cortex indicates that VEGF-mediated signaling could contribute to astroglial activation and associated inflammatory reactions.


Assuntos
Malformações do Desenvolvimento Cortical/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adolescente , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Criança , Epilepsia/complicações , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Malformações do Desenvolvimento Cortical/classificação , Malformações do Desenvolvimento Cortical/etiologia , Malformações do Desenvolvimento Cortical/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neurofilamentos/metabolismo
14.
Epilepsia ; 46(10): 1569-80, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16190927

RESUMO

PURPOSE: Overexpression of multidrug transporters may play a role in the development of pharmacoresistance by decreasing extracellular drug levels in the brain. However, it is not known whether overexpression is due to an initial insult or evolves more gradually because of recurrent spontaneous seizures. In the present study, we investigated the expression of different multidrug transporters during epileptogenesis in the rat. In addition, we determined whether these transporters affected phenytoin (PHT) distribution in the brain. METHODS: Expression of multidrug resistance-associated proteins MRP1 and MRP2 and breast cancer-resistance protein (BCRP) was examined after electrically induced status epilepticus (SE) by immunocytochemistry and Western blot analysis. Brain/blood PHT levels were determined by high-performance liquid chromatography (HPLC) analysis in the presence and absence of the MRP inhibitor probenecid. RESULTS: Shortly after SE, MRP1, MRP2, and BCRP were upregulated in astrocytes within several limbic structures, including hippocampus. In chronic epileptic rats, these proteins were overexpressed in the parahippocampal cortex, specifically in blood vessels and astrocytes surrounding these vessels. Overexpression was related to the occurrence of SE and was present mainly in rats with a high seizure frequency. Brain PHT levels were significantly lower in epileptic rats compared with control rats, but pharmacologic inhibition of MRPs increased the PHT levels. CONCLUSIONS: Overexpression of MRP and BCRP was induced by SE as well as recurrent seizures. Moreover, overexpression was associated with lower PHT levels in the brain, which was reversed through inhibition of MRPs. These data suggest that administration of antiepileptic drugs in combination with specific inhibitors for multidrug transporters may be a promising therapeutic strategy in pharmacoresistant patients.


Assuntos
Encéfalo/metabolismo , Resistência a Múltiplos Medicamentos , Epilepsia/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Anticonvulsivantes/metabolismo , Anticonvulsivantes/uso terapêutico , Doença Crônica , Estimulação Elétrica , Epilepsia/tratamento farmacológico , Humanos , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Fenitoína/metabolismo , Probenecid/farmacologia , Ratos , Ratos Sprague-Dawley , Recidiva , Estado Epiléptico/metabolismo , Distribuição Tecidual
15.
Epilepsia ; 46(9): 1371-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16146431

RESUMO

PURPOSE: Iron accumulation in the brain has been associated with neurodegenerative disorders, including epilepsy. In our previous SAGE study, we showed that ferritin, an iron-storage protein, was one of the genes (Ferritin-H) that showed overexpression before the chronic epileptic phase. In this study we used ferritin as indicator for disturbed iron homeostasis to acquire insight into whether this could play a role in the pathogenesis of temporal lobe epilepsy. METHODS: With immunocytochemistry, we studied the regional and cellular distribution of ferritin protein in an animal model for temporal lobe epilepsy in which spontaneous seizures develop a few weeks after electrically induced status epilepticus (SE). RESULTS: Increased ferritin expression was observed in regions known to be vulnerable to cell death, mainly in reactive microglial cells of epileptic rats. Ferritin expression after SE was initially high, especially throughout the hippocampus, but decreased over time. In the chronic epileptic phase, it was still upregulated in regions where extensive cell loss occurs during the early acute and latent period. Within the parahippocampal region, the most persistent ferritin overexpression was present in microglial cells in layer III of the medial entorhinal area. The upregulation was most extensive in rats that had developed a progressive form of epilepsy with frequent seizures (approximately five to 10 seizures per day). CONCLUSIONS: The fact that ferritin upregulation is still present in specific limbic regions in chronic epileptic rats, when neuronal loss is absent or minimal, suggests a role of iron in the pathogenesis and progression of epilepsy.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Ferritinas/metabolismo , Giro Para-Hipocampal/metabolismo , Animais , Morte Celular/genética , Modelos Animais de Doenças , Estimulação Elétrica , Epilepsia do Lobo Temporal/genética , Ferritinas/genética , Regulação da Expressão Gênica , Hipocampo/metabolismo , Imuno-Histoquímica , Microglia/metabolismo , Neurônios/metabolismo , Ratos , Estado Epiléptico/metabolismo , Distribuição Tecidual , Regulação para Cima
16.
Epilepsia ; 46(6): 849-57, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15946326

RESUMO

PURPOSE: Breast cancer resistance protein (BCRP) is a half adenosine triphosphate (ATP)-binding cassette (ABC) transporter expressed on cellular membranes and included in the group of multidrug resistant (MDR)-related proteins. Recently, upregulation of different MDR proteins has been shown in human epilepsy-associated conditions. This study investigated the expression and cellular distribution of BCRP in human control and epileptic brain, including a large number of both neoplastic and nonneoplastic specimens from patients with chronic pharmacoresistant epilepsy. METHODS: Several epileptogenic pathologies, such as hippocampal sclerosis (HS), focal cortical dysplasia (FCD), dysembryoplastic neuroepithelial tumor, oligodendroglioma astrocytoma, and glioblastoma multiforme were studied by using Western blot and immunocytochemistry. RESULTS: With Western blot, we could demonstrate the presence of BCRP in both normal and epileptic human brain tissue. In contrast to P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) 2, BCRP expression levels did not change in tissue from patients with HS, compared with control hippocampus. No BCRP immunoreactivity was observed in glial or neuronal cells, including reactive astrocytes and dysplastic neurons in FCD. BCRP expression was, however, increased in tumor brain tissue. Immunocytochemistry demonstrated that BCRP was exclusively located in blood vessels and was highly expressed at the luminal cell surface and in newly formed tumor capillaries. This localization closely resembles that of P-gp. The higher expression observed in astrocytomas by Western blot analysis was related to the higher vascular density within the tumor tissue. CONCLUSIONS: These results indicate a constitutive expression of BCRP in human endothelial cells, representing an important barrier against drug access to the brain. In particular, the strong BCRP expression in the microvasculature of epileptogenic brain tumors could critically influence the bioavailability of drugs within the tumor and contribute to pharmacoresistance.


Assuntos
Transportadores de Cassetes de Ligação de ATP/análise , Neoplasias Encefálicas/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Endotélio Vascular/química , Epilepsias Parciais/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/análise , Proteínas de Neoplasias/análise , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrocitoma/química , Western Blotting , Encéfalo/patologia , Córtex Cerebral/anormalidades , Córtex Cerebral/química , Córtex Cerebral/metabolismo , Endotélio Vascular/metabolismo , Epilepsias Parciais/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Microcirculação , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas de Neoplasias/metabolismo , Oligodendroglioma/química , Oligodendroglioma/metabolismo , Esclerose , Distribuição Tecidual
17.
Epilepsia ; 45(10): 1189-201, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15461673

RESUMO

PURPOSE: The plasma membrane Ca2+ -adenosine triphosphatase (ATPase) (PMCA) and (potassium-dependent) sodium-calcium exchange [NC(K)X] represent two main calcium-extrusion mechanisms that are important for the restoration of [Ca2+]i levels after electrical activity. We investigated whether the expression of these calcium-extrusion proteins is altered in the course of epileptogenesis. METHODS: Hippocampal-parahippocampal protein expression of NCX1, 2, and 3, PMCA1-4, and NCKX2 at an early and late stage after kainate-induced status epilepticus (SE) was compared with that in control rats by using immunocytochemistry. RESULTS: Several alterations were found in chronic epileptic rats: (a) NCX1 expression was permanently decreased in the inner molecular layer (IML) of the dentate gyrus (DG) and entorhinal cortex layer III (ECIII), related to neuronal loss in hilus and ECIII, respectively; (b) PMCA and NCKX2 expression was transiently upregulated in the IML, and decreased in several areas where cell loss had occurred, (c) NCX3 expression, which in control rats is abundant in presynaptic terminals of mossy fibers (MF), was extensively and permanently decreased in stratum lucidum and hilar region. In addition, newly formed MF sprouts that project to the DG iml did not noticeably express NCX3; (d) NCX2 and NCKX2 were (transiently) upregulated in astrocytes of epileptic rats throughout the hippocampal formation, including ECIII. CONCLUSIONS: These region-specific changes in calcium-extrusion proteins reflect a change in calcium regulation. Whether these regional-specific changes of calcium-extrusion proteins are associated with an abnormal calcium homeostasis must be determined. Because some alterations of calcium-extrusion protein expression are already present at an early stage of epileptogenesis, they could be involved in this process.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Hipocampo/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo , Animais , Astrócitos/metabolismo , Proteínas de Transporte de Cátions , Epilepsia do Lobo Temporal/metabolismo , Agonistas de Aminoácidos Excitatórios , Imuno-Histoquímica , Ácido Caínico , Masculino , Fibras Musgosas Hipocampais/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática , Ratos , Ratos Sprague-Dawley , Regulação para Cima/fisiologia
18.
Acta Neuropathol ; 108(5): 422-34, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15375667

RESUMO

An increasing number of observations suggests an important and complex role for both high- (tyrosine kinase receptor, trk) and low- (p75) affinity neurotrophin receptors (NTRs) during development in human brain. In the present study, the cell-specific distribution of NTRs was studied in different developmental lesions, including focal cortical dysplasia (FCD, n = 15), ganglioglioma (GG, n = 15) and dysembryoplastic neuroepithelial tumors, (DNT, n = 10), from patients with medically intractable epilepsy. Lesional, perilesional, as well as normal brain regions were examined for the expression of trkA, trkB, trkC and p75(NTR) by immunocytochemistry. In normal postmortem human cortex, immunoreactivity (IR) for trk and p75(NTR) was mainly observed in pyramidal neurons, whereas no notable glial IR was found within the white matter. All three trk receptors were encountered in high levels in the neuronal component of the majority of FCD, GG and DNT specimens. Strong trkA, trkB and trkC IR was found in neurons of different size, including large dysplastic neurons and balloon cells in FCD cases. In contrast, p75(NTR) IR was observed in only a small number of neuronal cells, which also contain trk receptors. Glial cells with astrocytic morphology showed predominantly IR for trkA in FCD and GG specimens, whereas oligodendroglial-like cells in DNT showed predominently IR for trkB. P75(NTR) IR was observed in a population of cells of the microglial/macrophage lineage in both FCD and glioneuronal tumors. Taken together, our findings indicate that the neuronal and the glial components of malformations of cortical development express both high- and low-affinity NTRs. Further research is necessary to investigate how activation of these specific receptors could contribute to the development and the epileptogenicity of these developmental disorders.


Assuntos
Córtex Cerebral/anormalidades , Epilepsia/fisiopatologia , Receptores de Fator de Crescimento Neural/metabolismo , Adolescente , Adulto , Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Criança , Pré-Escolar , Epilepsia/etiologia , Epilepsia/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Microglia/metabolismo , Pessoa de Meia-Idade , Malformações do Sistema Nervoso/complicações , Neurônios/metabolismo
19.
Epilepsia ; 44(9): 1166-75, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12919388

RESUMO

PURPOSE: Recent evidence has been obtained that the major vault protein (MVP) may play a role in multidrug resistance (MDR). We investigated the expression and cellular localization of MVP in gangliogliomas (GGs), which are increasingly recognized causes of chronic pharmacoresistant epilepsy. METHODS: Surgical tumor specimens (n = 30), as well as peritumoral and control brain tissues, were examined for the cellular distribution pattern of MVP with immunocytochemistry. Western blot analysis showed a consistent increase in MVP expression in GGs compared with that in control cortex. RESULTS: In normal brain, MVP expression was below detection in glial and neuronal cells, and only low immunoreactivity (IR) levels were detected in blood vessels. MVP expression was observed in the neuronal component of 30 of 30 GGs and in a population of tumor glial cells. In the majority of the tumors, strong MVP IR was found in lesional vessels. Perilesional regions did not show increased staining in vessels or in neuronal and glial cells compared with normal cortex. However, expression of MVP was detected in the hippocampus in cases with dual pathology. CONCLUSIONS: The increased expression of MVP in GGs is another example of an MDR-related protein that is upregulated in patients with refractory epilepsy. Further research is necessary to investigate whether it could play role in the mechanisms underlying drug resistance in chronic human epilepsy.


Assuntos
Neoplasias Encefálicas/metabolismo , Ganglioglioma/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/biossíntese , Adolescente , Adulto , Análise de Variância , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patologia , Feminino , Seguimentos , Ganglioglioma/genética , Ganglioglioma/patologia , Regulação Neoplásica da Expressão Gênica , Hipocampo/química , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética
20.
Eur J Neurosci ; 17(10): 2106-18, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12786977

RESUMO

We examined the regulation of glutamate transporter protein expression after stimulation with selective metabotropic glutamate receptor (mGluR) agonists in cultured human glial cells. mGluR3 and mGluR5 are expressed in human astrocytes and in human glioma cells in vivo as well as in vitro, as shown by either RT-PCR or western blot analysis. The selective group I agonist (S)-3,5-dihydroxyphenylglycine produced a significant down-regulation of both GLAST and GLT-1 protein expression in astrocytes cultured in the presence of growth factors. This condition mimics the morphology of reactive glial cells in vivo including an increased expression of mGluR5 protein (observed in pathological conditions). In contrast, (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine, a selective agonist of group II metabotropic glutamate receptors, positively modulates the expression of GLAST and GLT-1 proteins. A similar opposite effect of (S)-3,5-dihydroxyphenylglycine and (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine was observed for the expression of EAAT3 protein in U373 glioblastoma cell line. Selective group I and II antagonists prevented these effects. Pharmacological inhibition of mitogen-activated protein kinase and phosphatidylinositol-3-K pathways reduces the induction of GLT-1 observed in response to the group II metabotropic glutamate receptor agonist (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine. Thus, mGluR3 and mGluR5 can critically and differentially modulate the expression of glutamate transporters and may represent interesting pharmacological targets to regulate the extracellular levels of glutamate in pathological conditions.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/genética , Astrócitos/fisiologia , Transportador 2 de Aminoácido Excitatório/genética , Glioblastoma , Receptores de Glutamato Metabotrópico/genética , Simportadores/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Astrócitos/citologia , Transportador 1 de Aminoácido Excitatório , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Proteínas de Transporte de Glutamato da Membrana Plasmática , Substâncias de Crescimento/farmacologia , Humanos , Imunofenotipagem , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/metabolismo , Simportadores/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA