Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Brain ; 146(4): 1342-1356, 2023 04 19.
Article in English | MEDLINE | ID: mdl-36226386

ABSTRACT

Understanding the exact molecular mechanisms involved in the aetiology of epileptogenic pathologies with or without tumour activity is essential for improving treatment of drug-resistant focal epilepsy. Here, we characterize the landscape of somatic genetic variants in resected brain specimens from 474 individuals with drug-resistant focal epilepsy using deep whole-exome sequencing (>350×) and whole-genome genotyping. Across the exome, we observe a greater number of somatic single-nucleotide variants in low-grade epilepsy-associated tumours (7.92 ± 5.65 single-nucleotide variants) than in brain tissue from malformations of cortical development (6.11 ± 4 single-nucleotide variants) or hippocampal sclerosis (5.1 ± 3.04 single-nucleotide variants). Tumour tissues also had the largest number of likely pathogenic variant carrying cells. low-grade epilepsy-associated tumours had the highest proportion of samples with one or more somatic copy-number variants (24.7%), followed by malformations of cortical development (5.4%) and hippocampal sclerosis (4.1%). Recurring somatic whole chromosome duplications affecting Chromosome 7 (16.8%), chromosome 5 (10.9%), and chromosome 20 (9.9%) were observed among low-grade epilepsy-associated tumours. For germline variant-associated malformations of cortical development genes such as TSC2, DEPDC5 and PTEN, germline single-nucleotide variants were frequently identified within large loss of heterozygosity regions, supporting the recently proposed 'second hit' disease mechanism in these genes. We detect somatic variants in 12 established lesional epilepsy genes and demonstrate exome-wide statistical support for three of these in the aetiology of low-grade epilepsy-associated tumours (e.g. BRAF) and malformations of cortical development (e.g. SLC35A2 and MTOR). We also identify novel significant associations for PTPN11 with low-grade epilepsy-associated tumours and NRAS Q61 mutated protein with a complex malformation of cortical development characterized by polymicrogyria and nodular heterotopia. The variants identified in NRAS are known from cancer studies to lead to hyperactivation of NRAS, which can be targeted pharmacologically. We identify large recurrent 1q21-q44 duplication including AKT3 in association with focal cortical dysplasia type 2a with hyaline astrocytic inclusions, another rare and possibly under-recognized brain lesion. The clinical-genetic analyses showed that the numbers of somatic single-nucleotide variant across the exome and the fraction of affected cells were positively correlated with the age at seizure onset and surgery in individuals with low-grade epilepsy-associated tumours. In summary, our comprehensive genetic screen sheds light on the genome-scale landscape of genetic variants in epileptic brain lesions, informs the design of gene panels for clinical diagnostic screening and guides future directions for clinical implementation of epilepsy surgery genetics.


Subject(s)
Drug Resistant Epilepsy , Epilepsies, Partial , Epilepsy , Malformations of Cortical Development , Humans , Epilepsy/pathology , Brain/pathology , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/surgery , Drug Resistant Epilepsy/metabolism , Genomics , Malformations of Cortical Development/complications , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Epilepsies, Partial/metabolism , Nucleotides/metabolism
2.
Acta Neuropathol ; 145(6): 815-827, 2023 06.
Article in English | MEDLINE | ID: mdl-36973520

ABSTRACT

Exome-wide sequencing studies recently described PTPN11 as a novel brain somatic epilepsy gene. In contrast, germline mutations of PTPN11 are known to cause Noonan syndrome, a multisystem disorder characterized by abnormal facial features, developmental delay, and sporadically, also brain tumors. Herein, we performed a deep phenotype-genotype analysis of a comprehensive series of ganglioglioma (GG) with brain somatic alterations of the PTPN11/KRAS/NF1 genes compared to GG with common MAP-Kinase signaling pathway alterations, i.e., BRAFV600E. Seventy-two GG were submitted to whole exome sequencing and genotyping and 84 low grade epilepsy associated tumors (LEAT) to DNA-methylation analysis. In 28 tumours, both analyses were available from the same sample. Clinical data were retrieved from hospital files including disease onset, age at surgery, brain localization, and seizure outcome. A comprehensive histopathology staining panel was available in all cases. We identified eight GG with PTPN11 alterations, copy number variant (CNV) gains of chromosome 12, and the commonality of additional CNV gains in NF1, KRAS, FGFR4 and RHEB, as well as BRAFV600E alterations. Histopathology revealed an atypical glio-neuronal phenotype with subarachnoidal tumor spread and large, pleomorphic, and multinuclear cellular features. Only three out of eight patients with GG and PTPN11/KRAS/NF1 alterations were free of disabling-seizures 2 years after surgery (38% had Engel I). This was remarkably different from our series of GG with only BRAFV600E mutations (85% had Engel I). Unsupervised cluster analysis of DNA methylation arrays separated these tumours from well-established LEAT categories. Our data point to a subgroup of GG with cellular atypia in glial and neuronal cell components, adverse postsurgical outcome, and genetically characterized by complex alterations in PTPN11 and other RAS-/MAP-Kinase and/or mTOR signaling pathways. These findings need prospective validation in clinical practice as they argue for an adaptation of the WHO grading system in developmental, glio-neuronal tumors associated with early onset focal epilepsy.


Subject(s)
Epilepsy , Ganglioglioma , Humans , Epilepsy/pathology , Ganglioglioma/genetics , Ganglioglioma/pathology , Mutation/genetics , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Genes, ras , MAP Kinase Signaling System
3.
Acta Neuropathol ; 143(1): 93-104, 2022 01.
Article in English | MEDLINE | ID: mdl-34797422

ABSTRACT

Malformations of cortical development (MCD) comprise a broad spectrum of structural brain lesions frequently associated with epilepsy. Disease definition and diagnosis remain challenging and are often prone to arbitrary judgment. Molecular classification of histopathological entities may help rationalize the diagnostic process. We present a retrospective, multi-center analysis of genome-wide DNA methylation from human brain specimens obtained from epilepsy surgery using EPIC 850 K BeadChip arrays. A total of 308 samples were included in the study. In the reference cohort, 239 formalin-fixed and paraffin-embedded (FFPE) tissue samples were histopathologically classified as MCD, including 12 major subtype pathologies. They were compared to 15 FFPE samples from surgical non-MCD cortices and 11 FFPE samples from post-mortem non-epilepsy controls. We applied three different statistical approaches to decipher the DNA methylation pattern of histopathological MCD entities, i.e., pairwise comparison, machine learning, and deep learning algorithms. Our deep learning model, which represented a shallow neuronal network, achieved the highest level of accuracy. A test cohort of 43 independent surgical samples from different epilepsy centers was used to test the precision of our DNA methylation-based MCD classifier. All samples from the test cohort were accurately assigned to their disease classes by the algorithm. These data demonstrate DNA methylation-based MCD classification suitability across major histopathological entities amenable to epilepsy surgery and age groups and will help establish an integrated diagnostic classification scheme for epilepsy-associated MCD.


Subject(s)
DNA Methylation , Deep Learning , Malformations of Cortical Development/classification , Malformations of Cortical Development/diagnosis , Adolescent , Adult , Child , Child, Preschool , Epilepsy/etiology , Female , Humans , Infant , Male , Malformations of Cortical Development/genetics , Middle Aged , Retrospective Studies , Young Adult
4.
Epilepsia ; 63(1): 42-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34741301

ABSTRACT

OBJECTIVE: Focal cortical dysplasia (FCD) Type 1 and its three subtypes have yet not been fully characterized at the clinical, anatomopathological, and molecular level (International League Against Epilepsy [ILAE] FCD classification from 2011). We aimed to describe the clinical phenotype of patients with histopathologically confirmed FCD1A obtained from a single epilepsy center between 2002 and 2016. METHODS: Medical records were retrieved from the hospital's archive. Results from electroencephalography (EEG) video recordings, neuroimaging, and histopathology were reevaluated. Magnetic resonance imaging (MRI) post-processing was retrospectively performed in nine patients. DNA methylation studies were carried out from archival surgical brain tissue in 11 patients. RESULTS: Nineteen children with a histopathological diagnosis of FCD1A were included. The average onset of epilepsy was 0.9 years (range 0.2-10 years). All children had severe cognitive impairment and one third had mild motor deficits, yet fine finger movements were preserved in all patients. All patients had daily seizures, being drug resistant from disease onset. Interictal electroencephalography revealed bilateral multi-regional epileptiform discharges. Interictal status epilepticus was observed in 8 and countless subclinical seizures in 11 patients. Regional continuous irregular slow waves were of higher lateralizing and localizing yield than spikes. Posterior background rhythms were normal in 16 of 19 children. Neuroimaging showed unilateral multilobar hypoplasia and increased T2-FLAIR signals of the white matter in 18 of 19 patients. All children underwent tailored multilobar resections, with seizure freedom achieved in 47% (Engel class I). There was no case with frontal involvement without involvement of the posterior quadrant by MRI and histopathology. DNA methylation profiling distinguished FCD1A samples from all other epilepsy specimens and controls. SIGNIFICANCE: We identified a cohort of young children with drug resistance from seizure onset, bad EEG with posterior emphasis, lack of any focal neurological deficits but severe cognitive impairment, subtle hypoplasia of the epileptogenic area on MRI, and histopathologically defined and molecularly confirmed by DNA methylation analysis as FCD ILAE Type 1A.


Subject(s)
Epilepsy , Malformations of Cortical Development , Child, Preschool , Electroencephalography , Epilepsy/surgery , Humans , Magnetic Resonance Imaging , Malformations of Cortical Development/complications , Malformations of Cortical Development/diagnostic imaging , Malformations of Cortical Development/genetics , Retrospective Studies , Seizures/surgery , Treatment Outcome
5.
Childs Nerv Syst ; 38(10): 1965-1975, 2022 10.
Article in English | MEDLINE | ID: mdl-35680686

ABSTRACT

PURPOSE: Hypothalamic hamartomas (HH) are malformations responsible for drug-resistant epilepsy. HH are usually isolated or part of a genetic syndrome, such as Pallister-Hall. Exceptionally they can be associated with other brain malformations such as polymicrogyria (PMG) and periventricular nodular heterotopia (PNH). We discuss the origin of the seizures associated with this combination of malformations, through electrophysiological studies, and review the literature on this rarely reported syndrome. METHODS: We retrospectively reviewed the patients with HH who had surgery between 1998 and 2020 and selected those with associated focal PMG and PNH, detected on MRIs. All patients had comprehensive clinical evaluation and surface video-EEG and one underwent stereoelectroencephalography (SEEG). RESULTS: Three male patients out of 182 were identified with a mean age at surgery of 7.5 years. MRI showed unilateral focal PMG (fronto-insulo-parietal, fronto-insulo-parieto-opercular, and fronto-insular, respectively) and multiple PNH homolateral to the main HH implantation side. In two patients, there were strong clinical and scalp EEG arguments for seizure onset within the HH. In the third, due to abnormalities on scalp video-EEG in the same area as PMG and the lack of gelastic seizures, SEEG was indicated and demonstrated seizure onset within the hamartoma. With a mean follow-up of 6 years, two patients were seizure-free. CONCLUSION: Our results show that HH is the trigger of epilepsy, which confirms the high epileptogenic potential of this malformation. In patients such as ours, as in those with isolated HH, we recommend to begin by operating the HH independently of seizure semiology or electrophysiological abnormalities.


Subject(s)
Epilepsy , Hamartoma , Hypothalamic Diseases , Periventricular Nodular Heterotopia , Polymicrogyria , Child , Electroencephalography/methods , Epilepsy/complications , Hamartoma/complications , Hamartoma/diagnostic imaging , Hamartoma/surgery , Humans , Hypothalamic Diseases/complications , Hypothalamic Diseases/diagnostic imaging , Hypothalamic Diseases/surgery , Magnetic Resonance Imaging , Male , Periventricular Nodular Heterotopia/complications , Periventricular Nodular Heterotopia/diagnostic imaging , Periventricular Nodular Heterotopia/surgery , Polymicrogyria/complications , Polymicrogyria/diagnostic imaging , Polymicrogyria/surgery , Retrospective Studies , Seizures/complications , Seizures/surgery
6.
N Engl J Med ; 377(17): 1648-1656, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29069555

ABSTRACT

BACKGROUND: Detailed neuropathological information on the structural brain lesions underlying seizures is valuable for understanding drug-resistant focal epilepsy. METHODS: We report the diagnoses made on the basis of resected brain specimens from 9523 patients who underwent epilepsy surgery for drug-resistant seizures in 36 centers from 12 European countries over 25 years. Histopathological diagnoses were determined through examination of the specimens in local hospitals (41%) or at the German Neuropathology Reference Center for Epilepsy Surgery (59%). RESULTS: The onset of seizures occurred before 18 years of age in 75.9% of patients overall, and 72.5% of the patients underwent surgery as adults. The mean duration of epilepsy before surgical resection was 20.1 years among adults and 5.3 years among children. The temporal lobe was involved in 71.9% of operations. There were 36 histopathological diagnoses in seven major disease categories. The most common categories were hippocampal sclerosis, found in 36.4% of the patients (88.7% of cases were in adults), tumors (mainly ganglioglioma) in 23.6%, and malformations of cortical development in 19.8% (focal cortical dysplasia was the most common type, 52.7% of cases of which were in children). No histopathological diagnosis could be established for 7.7% of the patients. CONCLUSIONS: In patients with drug-resistant focal epilepsy requiring surgery, hippocampal sclerosis was the most common histopathological diagnosis among adults, and focal cortical dysplasia was the most common diagnosis among children. Tumors were the second most common lesion in both groups. (Funded by the European Union and others.).


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Epilepsy/pathology , Hippocampus/pathology , Malformations of Cortical Development/pathology , Adult , Age Factors , Age of Onset , Brain Neoplasms/complications , Child , Databases as Topic , Epilepsy/etiology , Epilepsy/surgery , Europe , Female , Humans , Male , Malformations of Cortical Development/complications , Temporal Lobe/pathology
7.
Acta Neuropathol ; 140(6): 881-891, 2020 12.
Article in English | MEDLINE | ID: mdl-32979071

ABSTRACT

Polymicrogyria (PMG) is a developmental cortical malformation characterized by an excess of small and frustrane gyration and abnormal cortical lamination. PMG frequently associates with seizures. The molecular pathomechanisms underlying PMG development are not yet understood. About 40 genes have been associated with PMG, and small copy number variations have also been described in selected patients. We recently provided evidence that epilepsy-associated structural brain lesions can be classified based on genomic DNA methylation patterns. Here, we analyzed 26 PMG patients employing array-based DNA methylation profiling on formalin-fixed paraffin-embedded material. A series of 62 well-characterized non-PMG cortical malformations (focal cortical dysplasia type 2a/b and hemimegalencephaly), temporal lobe epilepsy, and non-epilepsy autopsy controls was used as reference cohort. Unsupervised dimensionality reduction and hierarchical cluster analysis of DNA methylation profiles showed that PMG formed a distinct DNA methylation class. Copy number profiling from DNA methylation data identified a uniform duplication spanning the entire long arm of chromosome 1 in 7 out of 26 PMG patients, which was verified by additional fluorescence in situ hybridization analysis. In respective cases, about 50% of nuclei in the center of the PMG lesion were 1q triploid. No chromosomal imbalance was seen in adjacent, architecturally normal-appearing tissue indicating mosaicism. Clinically, PMG 1q patients presented with a unilateral frontal or hemispheric PMG without hemimegalencephaly, a severe form of intractable epilepsy with seizure onset in the first months of life, and severe developmental delay. Our results show that PMG can be classified among other structural brain lesions according to their DNA methylation profile. One subset of PMG with distinct clinical features exhibits a duplication of chromosomal arm 1q.


Subject(s)
Brain/pathology , Chromosomes/metabolism , Drug Resistant Epilepsy/pathology , Malformations of Cortical Development/pathology , Polymicrogyria/pathology , DNA Copy Number Variations/physiology , Drug Resistant Epilepsy/complications , Drug Resistant Epilepsy/genetics , Female , Humans , Male , Polymicrogyria/complications , Polymicrogyria/genetics , Seizures/pathology
8.
Epilepsia ; 60(6): 1091-1103, 2019 06.
Article in English | MEDLINE | ID: mdl-31074842

ABSTRACT

OBJECTIVES: Focal cortical dysplasia (FCD) is a major cause of drug-resistant focal epilepsy in children, and the clinicopathological classification remains a challenging issue in daily practice. With the recent progress in DNA methylation-based classification of human brain tumors we examined whether genomic DNA methylation and gene expression analysis can be used to also distinguish human FCD subtypes. METHODS: DNA methylomes and transcriptomes were generated from massive parallel sequencing in 15 surgical FCD specimens, matched with 5 epilepsy and 6 nonepilepsy controls. RESULTS: Differential hierarchical cluster analysis of DNA methylation distinguished major FCD subtypes (ie, Ia, IIa, and IIb) from patients with temporal lobe epilepsy patients and nonepileptic controls. Targeted panel sequencing identified a novel likely pathogenic variant in DEPDC5 in a patient with FCD type IIa. However, no enrichment of differential DNA methylation or gene expression was observed in mechanistic target of rapamycin (mTOR) pathway-related genes. SIGNIFICANCE: Our studies extend the evidence for disease-specific methylation signatures toward focal epilepsies in favor of an integrated clinicopathologic and molecular classification system of FCD subtypes incorporating genomic methylation.


Subject(s)
DNA Methylation/genetics , Malformations of Cortical Development/genetics , Adolescent , Adult , Child , Child, Preschool , Cluster Analysis , DNA/genetics , Epilepsies, Partial/classification , Epilepsies, Partial/genetics , Female , Gene Expression Profiling , Genome, Human , Humans , Infant , Male , Malformations of Cortical Development/classification , Malformations of Cortical Development/diagnostic imaging , Middle Aged , RNA, Messenger/genetics , TOR Serine-Threonine Kinases/genetics , Tissue Banks , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Transcriptome , Young Adult
9.
Epilepsy Behav ; 91: 68-74, 2019 02.
Article in English | MEDLINE | ID: mdl-30061008

ABSTRACT

BACKGROUND: Mild malformation of cortical development with oligodendroglial hyperplasia and epilepsy (MOGHE) is a newly described, rare histopathologic entity detected in resected brain tissue of patients with refractory epilepsies. It shows a predominantly frontal localization causing a difficult-to-treat epilepsy with onset usually in early childhood. Histologically, MOGHE is characterized by blurred gray-white-matter boundaries with increased numbers of heterotopic neurons in the subcortical white matter and increased density of oligodendroglia. Little is known, to date, about radiologic features of MOGHE. Here, we report typical and age-related magnetic resonance (MR) characteristics of MOGHE. PATIENTS AND METHODS: Retrospective analysis of 40 preoperative MR images of 25 pediatric patients with MOGHE (m/f: 13/12) who underwent epilepsy surgery at a median age of 9.3 years at our center between 2003 and 2018. Median age at magnetic resonance imaging (MRI) was 5.2 years (1.5-20.7 years). RESULTS: Two MR subtypes were found: subtype I with an increased laminar T2 and fluid attenuated inversion recovery (FLAIR) signal at the corticomedullary junction and subtype II with reduced corticomedullary differentiation because of increased signal of the adjacent white matter. Distribution of subtypes was age-related, with subtype I occurring between 1.5 and 5.1 years (median 2.6 years) and subtype II between 3.4 and 20.7 years (median 14.1 years). In one patient, MRI at the age of 2.7 years showed subtype I but had converted to subtype II by the age of 16 years. Histology revealed that in addition to the above mentioned typical findings of MOGHE, patchy areas of reduced density of myelin in 6 of 7 patients presenting subtype I out of 14 patients in which retrospective analysis regarding myelination was accessible. CONCLUSION: Magnetic resonance characteristics in patients with MOGHE are age-related and seem to change from subtype I to subtype II probably because of maturational processes between 3 and 6 years. Patchy areas of hypomyelination in histology seem to disappear during brain maturation and may therefore represent the histologic correlate of laminar T2 and FLAIR hyperintensities in subtype I. This article is part of the Special Issue "Individualized Epilepsy Management: Medicines, Surgery and Beyond".


Subject(s)
Brain/diagnostic imaging , Drug Resistant Epilepsy/diagnostic imaging , Magnetic Resonance Imaging/methods , Malformations of Cortical Development/diagnostic imaging , Oligodendroglia/pathology , Adolescent , Age Factors , Brain/surgery , Child , Child, Preschool , Drug Resistant Epilepsy/surgery , Female , Gray Matter/diagnostic imaging , Gray Matter/surgery , Humans , Hyperplasia/diagnostic imaging , Hyperplasia/surgery , Infant , Magnetic Resonance Spectroscopy/methods , Male , Malformations of Cortical Development/surgery , Pilot Projects , Retrospective Studies , White Matter/diagnostic imaging , White Matter/surgery , Young Adult
11.
Brain ; 139(Pt 9): 2456-68, 2016 09.
Article in English | MEDLINE | ID: mdl-27383529

ABSTRACT

Hemidisconnections (i.e. hemispherectomies or hemispherotomies) invariably lead to contralateral hemiparesis. Many patients with a pre-existing hemiparesis, however, experience no deterioration in motor functions, and some can still grasp with their paretic hand after hemidisconnection. The scope of our study was to predict this phenomenon. Hypothesizing that preserved contralateral grasping ability after hemidisconnection can only occur in patients controlling their paretic hands via ipsilateral corticospinal projections already in the preoperative situation, we analysed the asymmetries of the brainstem (by manual magnetic resonance imaging volumetry) and of the structural connectivity of the corticospinal tracts within the brainstem (by magnetic resonance imaging diffusion tractography), assuming that marked hypoplasia or Wallerian degeneration on the lesioned side in patients who can grasp with their paretic hands indicate ipsilateral control. One hundred and two patients who underwent hemidisconnections between 0.8 and 36 years of age were included. Before the operation, contralateral hand function was normal in 3/102 patients, 47/102 patients showed hemiparetic grasping ability and 52/102 patients could not grasp with their paretic hands. After hemidisconnection, 20/102 patients showed a preserved grasping ability, and 5/102 patients began to grasp with their paretic hands only after the operation. All these 25 patients suffered from pre- or perinatal brain lesions. Thirty of 102 patients lost their grasping ability. This group included all seven patients with a post-neonatally acquired or progressive brain lesion who could grasp before the operation, and also all three patients with a preoperatively normal hand function. The remaining 52/102 patients were unable to grasp pre- and postoperatively. On magnetic resonance imaging, the patients with preserved grasping showed significantly more asymmetric brainstem volumes than the patients who lost their grasping ability. Similarly, these patients showed striking asymmetries in the structural connectivity of the corticospinal tracts. In summary, normal preoperative hand function and a post-neonatally acquired or progressive lesion predict a loss of grasping ability after hemidisconnection. A postoperatively preserved grasping ability is possible in hemiparetic patients with pre- or perinatal lesions, and this is highly likely when the brainstem is asymmetric and especially when the structural connectivity of the corticospinal tracts within the brainstem is asymmetric.


Subject(s)
Brain Stem/diagnostic imaging , Hand/physiopathology , Hemispherectomy/adverse effects , Magnetic Resonance Imaging/methods , Motor Activity/physiology , Outcome Assessment, Health Care , Paresis/physiopathology , Postoperative Complications , Pyramidal Tracts/diagnostic imaging , Adolescent , Adult , Child , Child, Preschool , Diffusion Tensor Imaging/methods , Female , Humans , Infant , Male , Paresis/congenital , Postoperative Complications/diagnostic imaging , Postoperative Complications/physiopathology , Prognosis , Young Adult
12.
J Pharmacol Exp Ther ; 352(2): 368-78, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503388

ABSTRACT

As a member of the multidrug-resistance associated protein (MRP) family, MRP2 affects the brain entry of different endogenous and exogenous compounds. Considering the role of this transporter at the blood-brain barrier, the regulation is of particular interest. However, there is limited knowledge regarding the factors that regulate MRP2 in neurologic disease states. Thus, we addressed the hypothesis that MRP2 might be affected by a glutamate-induced signaling pathway that we previously identified as one key mechanism in the regulation of P-glycoprotein. Studies in isolated porcine brain capillaries confirmed that glutamate and N-methyl-d-aspartic acid (NMDA) exposure upregulates expression and function of MPR2. The involvement of the NMDA receptor was further suggested by the fact that the NMDA receptor antagonist MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine], as well as the NMDA receptor glycine binding site antagonist L-701,324 [7-chloro-4-hydroxy-3-(3-phenoxy)phenyl-2(1H)-quinolinone], prevented the impact of glutamate. A role of cyclooxygenase-2 was indicated by coincubation with the cyclooxygenase-2 inhibitor celecoxib and the cyclooxygenase-1/-2 inhibitor indomethacin, which both efficaciously abolished a glutamate-induced upregulation of MRP2. Translational studies in human capillaries from surgical specimen demonstrated a relevant MRP2 efflux function and indicated an effect of glutamate exposure as well as its prevention by cyclooxygenase-2 inhibition. Taken together the findings provide first evidence for a role of a glutamate-induced NMDA receptor/cyclooxygenase-2 signaling pathway in the regulation of MRP2 expression and function. The response to excessive glutamate concentrations might contribute to overexpression of MRP2, which has been reported in neurologic diseases including epilepsy. The overexpression might have implications for brain access of various compounds including therapeutic drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/biosynthesis , Brain/blood supply , Capillaries/metabolism , Glutamic Acid/pharmacology , Adolescent , Animals , Biological Transport , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Capillaries/drug effects , Child , Child, Preschool , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Epilepsy/metabolism , Female , Humans , In Vitro Techniques , Male , Receptors, N-Methyl-D-Aspartate/metabolism , Swine , Up-Regulation , ATP-Binding Cassette Sub-Family B Member 4
13.
Mol Pharm ; 12(6): 2049-60, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25898179

ABSTRACT

Breast cancer resistance protein (BCRP) functions as a major molecular gatekeeper at the blood-brain barrier. Considering its impact on access to the brain by therapeutic drugs and harmful xenobiotics, it is of particular interest to elucidate the mechanisms of its regulation. Excessive glutamate concentrations have been reported during epileptic seizures or as a consequence of different brain insults including brain ischemia. Previously, we have demonstrated that glutamate can trigger an induction of the transporter P-glycoprotein. These findings raised the question whether other efflux transporters are affected in a comparable manner. Glutamate exposure proved to down-regulate BCRP transport function and expression in isolated porcine capillaries. The reduction was efficaciously prevented by coincubation with N-methyl-d-aspartate (NMDA) receptor antagonist MK-801. The involvement of the NMDA receptor in the down-regulation of BCRP was further confirmed by experiments showing an effect of NMDA exposure on brain capillary BCRP transport function and expression. Pharmacological targeting of cyclooxygenase-1 and -2 (COX-1 and -2) using the nonselective inhibitor indomethacin, COX-1 inhibitor SC-560, and COX-2 inhibitor celecoxib revealed a contribution of COX-2 activity to the NMDA receptor's downstream signaling events affecting BCRP. Translational studies were performed using human capillaries isolated from surgical specimens of epilepsy patients. The findings confirmed a glutamate-induced down-regulation of BCRP transport activity in human capillaries, which argued against major species differences. In conclusion, our data reveal a novel mechanism of BCRP down-regulation in porcine and human brain capillaries. Moreover, together with previous data sets for P-glycoprotein, the findings point to a contrasting impact of the signaling pathway on the regulation of BCRP and P-glycoprotein. The effect of glutamate and arachidonic acid signaling on BCRP function might have implications for brain drug delivery and for radiotracer brain access in epilepsy patients and patients with other brain insults.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Brain/metabolism , Capillaries/metabolism , Glucose Transporter Type 1/metabolism , Glutamic Acid/metabolism , Animals , Female , Humans , In Vitro Techniques , Male , Swine
14.
Epilepsia ; 55(7): 1009-19, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24902755

ABSTRACT

OBJECTIVE: To report on six patients with SCN1A mutations and malformations of cortical development (MCDs) and describe their clinical course, genetic findings, and electrographic, imaging, and neuropathologic features. METHODS: Through our database of epileptic encephalopathies, we identified 120 patients with SCN1A mutations, of which 4 had magnetic resonance imaging (MRI) evidence of MCDs. We collected two further similar observations through the European Task-force for Epilepsy Surgery in Children. RESULTS: The study group consisted of five males and one female (mean age 7.4 ± 5.3 years). All patients exhibited electroclinical features consistent with the Dravet syndrome spectrum, cognitive impairment, and autistic features. Sequencing analysis of the SCN1A gene detected two missense, two truncating, and two splice-site mutations. Brain MRI revealed bilateral periventricular nodular heterotopia (PNH) in two patients and focal cortical dysplasia (FCD) in three, and disclosed no macroscopic abnormality in one. In the MRI-negative patient, neuropathologic study of the whole brain performed after sudden unexpected death in epilepsy (SUDEP), revealed multifocal micronodular dysplasia in the left temporal lobe. Two patients with FCD underwent epilepsy surgery. Neuropathology revealed FCD type IA and type IIA. Their seizure outcome was unfavorable. All four patients with FCD exhibited multiple seizure types, which always included complex partial seizures, the area of onset of which co-localized with the region of structural abnormality. SIGNIFICANCE: MCDs and SCN1A gene mutations can co-occur. Although epidemiology does not support a causative role for SCN1A mutations, loss or impaired protein function combined with the effect of susceptibility factors and genetic modifiers of the phenotypic expression of SCN1A mutations might play a role. MCDs, particularly FCD, can influence the electroclinical phenotype in patients with SCN1A-related epilepsy. In patients with MCDs and a history of polymorphic seizures precipitated by fever, SCN1A gene testing should be performed before discussing any epilepsy surgery option, due to the possible implications for outcome.


Subject(s)
Cerebral Cortex/abnormalities , Cerebral Cortex/growth & development , Epilepsy/diagnosis , Epilepsy/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Adolescent , Cerebral Cortex/pathology , Child , Child, Preschool , Female , Humans , Male , Mutation, Missense/genetics , RNA Splice Sites/genetics
15.
Childs Nerv Syst ; 30(12): 2015-26, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25255773

ABSTRACT

The prevalence of focal cortical dysplasia (FCD) in pediatric patients with focal epilepsy is not exactly known because authors of publications in which the etiologies of epilepsies are listed, but which are not dealing specifically with epilepsy surgery issues, tend to lump together the many kinds of malformations of cortical development (MCD), of which FCDs, because of their relative frequency, are the most relevant subtypes. Out of 561 patients with MCD (children and adults) operated at centers in Europe who do feed data into the "European Epilepsy Brain Bank," 426 (76 %) had FCD.


Subject(s)
Electroencephalography , Epilepsies, Partial/physiopathology , Epilepsies, Partial/surgery , Malformations of Cortical Development/physiopathology , Malformations of Cortical Development/surgery , Signal Processing, Computer-Assisted , Adolescent , Adult , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Child , Child, Preschool , Cross-Sectional Studies , Diagnosis, Differential , Epilepsies, Partial/epidemiology , Epilepsies, Partial/pathology , Female , Humans , Image Enhancement , Imaging, Three-Dimensional , Infant , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/epidemiology , Malformations of Cortical Development/pathology , Neurologic Examination
16.
Clin Neuroradiol ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918242

ABSTRACT

PURPOSE: After vertical parasagittal hemispherotomy a restricted diffusion is often seen ipsilaterally and even distant from the adjacent resection margin. This retrospective cohort study analyses the anatomic site and the time course of the diffusion restriction after vertical parasagittal hemispherotomy. METHODS: Fifty-nine patients were included into this study, all of them having had one pre-operative and at least one post-operative MRI, including diffusion imaging at b­values of 0 and 1000 s/mm2 with a calculated ADC. RESULTS: Diffusion restriction occurred exclusively on the operated site in all patients. In the basal ganglia, diffusion restriction was present in 37 of 38 patients at the first postoperative day with a duration of 38 days. In the midbrain, the posterior limb of the internal capsule and the thalamus, a restricted diffusion became postoperatively prominent at day 9 in all three localizations, with a duration of 36, 34 and 36 days, respectively. The incidence of thalamic lesions was lower if a preoperative damage had occurred. CONCLUSION: The restricted diffusion in the basal ganglia resembles direct effects of the operation at its edges, whereas the later appearing diffusion restriction in the midbrain and the posterior limb of the internal capsule rather belong to a degeneration of the descending fibers being transected by the hemispherotomy in the sense of a Wallerian degeneration. The presence of preoperative hemispheric lesions influences the development of diffusion restriction at subacute fiber degeneration.

17.
Epilepsia Open ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38845524

ABSTRACT

OBJECTIVE: Perineuronal nets (PNN) are specialized extracellular matrix (ECM) components of the central nervous system, frequently accumulating at the surface of inhibitory GABAergic interneurons. While an altered distribution of PNN has been observed in neurological disorders including Alzheimer's disease, schizophrenia and epilepsy, their anatomical distribution also changes during physiological brain maturation and aging. Such an age-dependent shift was experimentally associated also with hippocampal engram formation during brain maturation. Our aim was to histopathologically assess PNN in the hippocampus of adult and pediatric patients with temporal lobe epilepsy (TLE) compared to age-matched post-mortem control subjects and to compare PNN-related changes with memory impairment observed in our patient cohort. METHODS: Sixty-six formalin-fixed and paraffin-embedded tissue specimens of the human hippocampus were retrieved from the European Epilepsy Brain Bank. Twenty-nine patients had histopathologically confirmed hippocampal sclerosis (HS), and eleven patients suffered from TLE without HS. PNN were immunohistochemically visualized using an antibody directed against aggrecan and manually counted from hippocampus subfields and the subiculum. RESULTS: PNN density increased with age in both human controls and TLE patients. However, their density was significantly higher in all HS patients compared to age-matched controls. Intriguingly, TLE patients presented presurgically with better memory when their hippocampal PNN density was higher (p < 0.05). SIGNIFICANCE: Our results were compatible with age-dependent ECM specialization in the human hippocampus and its precocious aging in the epileptic condition. These observations confirm recent experimental animal models and also support the notion that PNN play a role in memory formation in the human brain. PLAIN LANGUAGE SUMMARY: "Perineuronal nets" (PNN) are a specialized compartment of the extracellular matrix (ECM), especially surrounding highly active neurons of the mammalian brain. There is evidence that PNN play a role in memory formation, brain maturation, and in some pathologies like Alzheimer's disease, schizophrenia or epilepsy. In this study, we investigated the role of PNN in patients suffering from drug-resistant focal epilepsy compared to controls. We found that with increasing age, more neurons are surrounded by PNN. Similarly, all epilepsy patients but especially patients with better memory performance also had more PNN. This study raises further interest in studying ECM molecules in the human brain under physiological and pathophysiological conditions.

18.
Neurology ; 102(4): e208007, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38290094

ABSTRACT

BACKGROUND AND OBJECTIVE: Patients with presumed nonlesional focal epilepsy-based on either MRI or histopathologic findings-have a lower success rate of epilepsy surgery compared with lesional patients. In this study, we aimed to characterize a large group of patients with focal epilepsy who underwent epilepsy surgery despite a normal MRI and had no lesion on histopathology. Determinants of their postoperative seizure outcomes were further studied. METHODS: We designed an observational multicenter cohort study of MRI-negative and histopathology-negative patients who were derived from the European Epilepsy Brain Bank and underwent epilepsy surgery between 2000 and 2012 in 34 epilepsy surgery centers within Europe. We collected data on clinical characteristics, presurgical assessment, including genetic testing, surgery characteristics, postoperative outcome, and treatment regimen. RESULTS: Of the 217 included patients, 40% were seizure-free (Engel I) 2 years after surgery and one-third of patients remained seizure-free after 5 years. Temporal lobe surgery (adjusted odds ratio [AOR]: 2.62; 95% CI 1.19-5.76), shorter epilepsy duration (AOR for duration: 0.94; 95% CI 0.89-0.99), and completely normal histopathologic findings-versus nonspecific reactive gliosis-(AOR: 4.69; 95% CI 1.79-11.27) were significantly associated with favorable seizure outcome at 2 years after surgery. Of patients who underwent invasive monitoring, only 35% reached seizure freedom at 2 years. Patients with parietal lobe resections had lowest seizure freedom rates (12.5%). Among temporal lobe surgery patients, there was a trend toward favorable outcome if hippocampectomy was part of the resection strategy (OR: 2.94; 95% CI 0.98-8.80). Genetic testing was only sporadically performed. DISCUSSION: This study shows that seizure freedom can be reached in 40% of nonlesional patients with both normal MRI and histopathology findings. In particular, nonlesional temporal lobe epilepsy should be regarded as a relatively favorable group, with almost half of patients achieving seizure freedom at 2 years after surgery-even more if the hippocampus is resected-compared with only 1 in 5 nonlesional patients who underwent extratemporal surgery. Patients with an electroclinically identified focus, who are nonlesional, will be a promising group for advanced molecular-genetic analysis of brain tissue specimens to identify new brain somatic epilepsy genes or epilepsy-associated molecular pathways.


Subject(s)
Epilepsies, Partial , Epilepsy, Temporal Lobe , Epilepsy , Humans , Cohort Studies , Electroencephalography , Epilepsies, Partial/diagnostic imaging , Epilepsies, Partial/surgery , Epilepsy/diagnostic imaging , Epilepsy/surgery , Epilepsy, Temporal Lobe/surgery , Magnetic Resonance Imaging , Retrospective Studies , Seizures , Treatment Outcome
19.
Neuropediatrics ; 44(4): 225-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23483447

ABSTRACT

BACKGROUND: There are only few reports on patients with duplications of the short arm of chromosome 5, with little information about concomitant epilepsy. PATIENTS: We report on two patients with generalized epilepsies since age 2.5 years, in whom array comparative genomic hybridization revealed microduplications of different sizes in the short arm of chromosome 5. Both patients showed developmental delay, and magnetic resonance images were normal. The larger duplication in patient 1, who additionally exhibited dysmorphic features and photosensivity on electroencephalogram, occurred de novo, whereas the smaller duplication in patient 2 was paternally inherited. DISCUSSION: The overlapping duplicated region in band 5p13.1 comprises four genes (RICTOR, FYB, C9, and DAB2). Further investigation on patients with duplication 5p and epilepsy are needed to possibly identify a potential susceptibility locus for epilepsy at chromosome 5p13.1.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 5/genetics , Developmental Disabilities/genetics , Epilepsy, Generalized/genetics , Gene Duplication/genetics , Adolescent , Humans , Male
20.
Methods Mol Biol ; 2702: 315-325, 2023.
Article in English | MEDLINE | ID: mdl-37679627

ABSTRACT

Antibody phage display selection on cells is a powerful tool to generate highly specific antibodies recognizing a target in its cell bound conformation. Unlike phage display selections on immobilized proteins, it is not hampered by difficulties caused by recombinant protein expression of target proteins like altered folding or loss of epitopes. It also allows the generation of antibodies against proteins that are commercially unavailable, due to high production costs or lack of production. It is also a promising approach for single and especially multi-pass membrane proteins for which the complex secondary and tertiary structures can often not be retained upon recombinant protein expression. The selected antibodies are not only tools for in vivo studies but also used for the development of diagnostic assays and for therapeutical applications. Here, we describe a straightforward protocol for generation and screening of scFv binders by phage display selections on cells.


Subject(s)
Cell Surface Display Techniques , Antibodies , Biological Assay , Membrane Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL