Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
World Neurosurg ; 165: e159-e168, 2022 09.
Article in English | MEDLINE | ID: mdl-35659589

ABSTRACT

BACKGROUND: The gamma-aminobutyric acid A (GABAA) receptor is an important mediator of cellular signaling in the globus pallidus and might be implicated in the pathophysiology of Parkinson disease (PD). The goal of the present study was to characterize GABAA receptor subunit expression in the normal and parkinsonian human globus pallidus. METHODS: Postmortem brain specimens were obtained from 8 patients with pathological evidence of PD at autopsy and from 4 control patients without such evidence. These tissues were exposed to primary antibodies directed against the α1 and α3 subunits of the GABAA receptor and were visualized and quantified using fluorescence microscopy. RESULTS: No differences were found in the pallidal neuronal density in the control versus PD tissues. Projection neurons strongly expressed the α1, α3, and ß2 GABAA receptor subunits. After normalizing the immunofluorescence intensities in the globus pallidus to those in the adjacent structures, no significant differences were found in GABAA receptor subunit expression in the globus pallidus between the PD specimens and the control specimens. CONCLUSIONS: Compensatory changes in GABAA receptor α1 and α3 subunit expression in response to PD-related signaling abnormalities in the globus pallidus did not occur in our PD cohort.


Subject(s)
Globus Pallidus , Receptors, GABA-A , Humans , Neurons/metabolism , Receptors, GABA , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism
2.
J Clin Invest ; 131(15)2021 08 02.
Article in English | MEDLINE | ID: mdl-34138754

ABSTRACT

BackgroundPyridoxine-dependent epilepsy (PDE-ALDH7A1) is an inborn error of lysine catabolism that presents with refractory epilepsy in newborns. Biallelic ALDH7A1 variants lead to deficiency of α-aminoadipic semialdehyde dehydrogenase/antiquitin, resulting in accumulation of piperideine-6-carboxylate (P6C), and secondary deficiency of the important cofactor pyridoxal-5'-phosphate (PLP, active vitamin B6) through its complexation with P6C. Vitamin B6 supplementation resolves epilepsy in patients, but intellectual disability may still develop. Early diagnosis and treatment, preferably based on newborn screening, could optimize long-term clinical outcome. However, no suitable PDE-ALDH7A1 newborn screening biomarkers are currently available.MethodsWe combined the innovative analytical methods untargeted metabolomics and infrared ion spectroscopy to discover and identify biomarkers in plasma that would allow for PDE-ALDH7A1 diagnosis in newborn screening.ResultsWe identified 2S,6S-/2S,6R-oxopropylpiperidine-2-carboxylic acid (2-OPP) as a PDE-ALDH7A1 biomarker, and confirmed 6-oxopiperidine-2-carboxylic acid (6-oxoPIP) as a biomarker. The suitability of 2-OPP as a potential PDE-ALDH7A1 newborn screening biomarker in dried bloodspots was shown. Additionally, we found that 2-OPP accumulates in brain tissue of patients and Aldh7a1-knockout mice, and induced epilepsy-like behavior in a zebrafish model system.ConclusionThis study has opened the way to newborn screening for PDE-ALDH7A1. We speculate that 2-OPP may contribute to ongoing neurotoxicity, also in treated PDE-ALDH7A1 patients. As 2-OPP formation appears to increase upon ketosis, we emphasize the importance of avoiding catabolism in PDE-ALDH7A1 patients.FundingSociety for Inborn Errors of Metabolism for Netherlands and Belgium (ESN), United for Metabolic Diseases (UMD), Stofwisselkracht, Radboud University, Canadian Institutes of Health Research, Dutch Research Council (NWO), and the European Research Council (ERC).


Subject(s)
Epilepsy/metabolism , Metabolomics , Pipecolic Acids/metabolism , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/metabolism , Animals , Biomarkers/metabolism , Child , Epilepsy/genetics , Female , Humans , Mice , Mice, Knockout , Spectrophotometry, Infrared , Zebrafish/genetics , Zebrafish/metabolism
3.
Ann Clin Transl Neurol ; 7(4): 527-542, 2020 04.
Article in English | MEDLINE | ID: mdl-32243088

ABSTRACT

OBJECTIVE: Neurosteroids regulate neuronal excitability by potentiating γ-aminobutyric acid type-A receptors (GABARs). In animal models of temporal lobe epilepsy, the neurosteroid sensitivity of GABARs is diminished and GABAR subunit composition is altered. We tested whether similar changes occur in patients with epilepsy and if depolarization-induced increases in neuronal activity can replicate this effect. METHODS: We determined GABAR α4 subunit expression in cortical tissue resected from pediatric epilepsy patients. Modulation of human GABARs by allopregnanolone and Ro15-4513 was measured in Xenopus oocytes using whole-cell patch clamp. To extend the findings obtained using tissue from epilepsy patients, we evaluated GABAR expression and modulation by allopregnanolone and Ro15-4513 in cultured rat hippocampal neurons exposed to high extracellular potassium (HK) to increase neuronal activity. RESULTS: Expression of α4 subunits was increased in pediatric cortical epilepsy specimens encompassing multiple pathologies. The potentiation of GABA-evoked currents by the neurosteroid allopregnanolone was decreased in Xenopus oocytes expressing GABARs isolated from epilepsy patients. Furthermore, receptors isolated from epilepsy but not control tissue were sensitive to potentiation by Ro15-4513, indicating higher expression of α4 ßx γ2 subunit-containing receptors. Correspondingly, increasing the activity of cultured rat hippocampal neurons reduced allopregnanolone potentiation of miniature inhibitory postsynaptic currents (mIPSCs), increased modulation of tonic GABAR current by Ro15-4513, upregulated the surface expression of α4 and γ2 subunits, and increased the colocalization of α4 and γ2 subunit immunoreactivity. INTERPRETATION: These findings suggest that seizure activity-induced upregulation of α4 ßx γ2 subunit-containing GABARs could affect the anticonvulsant actions of neurosteroids.


Subject(s)
Cerebral Cortex/metabolism , Drug Resistant Epilepsy/metabolism , Electrophysiological Phenomena/physiology , GABA-A Receptor Agonists/pharmacology , Neurons/metabolism , Neurosteroids/metabolism , Pregnanolone/pharmacology , Receptors, GABA-A/metabolism , Adolescent , Adult , Animals , Azides/pharmacology , Benzodiazepines/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Child , Child, Preschool , Drug Resistant Epilepsy/surgery , Electrophysiological Phenomena/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Infant , Male , Neurons/drug effects , Oocytes , Patch-Clamp Techniques , Rats , Receptors, GABA-A/drug effects , Xenopus , Young Adult
4.
Epilepsy Curr ; 20(2): 116-117, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32100554

ABSTRACT

[Box: see text].

5.
Neuron ; 106(2): 237-245.e8, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32097630

ABSTRACT

Lissencephaly (LIS), denoting a "smooth brain," is characterized by the absence of normal cerebral convolutions with abnormalities of cortical thickness. Pathogenic variants in over 20 genes are associated with LIS. The majority of posterior predominant LIS is caused by pathogenic variants in LIS1 (also known as PAFAH1B1), although a significant fraction remains without a known genetic etiology. We now implicate CEP85L as an important cause of posterior predominant LIS, identifying 13 individuals with rare, heterozygous CEP85L variants, including 2 families with autosomal dominant inheritance. We show that CEP85L is a centrosome protein localizing to the pericentriolar material, and knockdown of Cep85l causes a neuronal migration defect in mice. LIS1 also localizes to the centrosome, suggesting that this organelle is key to the mechanism of posterior predominant LIS.


Subject(s)
Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Cytoskeletal Proteins/genetics , Oncogene Proteins, Fusion/genetics , Adolescent , Adult , Age of Onset , Animals , Centrosome/pathology , Child , Child, Preschool , Chromosome Aberrations , Classical Lissencephalies and Subcortical Band Heterotopias/diagnostic imaging , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , Female , Gene Knockdown Techniques , Genetic Variation , Heterozygote , Humans , Infant , Magnetic Resonance Imaging , Male , Mice , Mutation/genetics , Pedigree , Seizures/etiology , Young Adult
6.
Epilepsy Curr ; 19(1): 54-56, 2019 01.
Article in English | MEDLINE | ID: mdl-30838928

ABSTRACT

Somatic Mutations Activating the mTOR Pathway in Dorsal Telencephalic Progenitors Cause a Continuum of Cortical Dysplasias D'Gama AM, Woodworth MB, Hossain AA, Bizzotto S, Hatem NE, LaCoursiere CM, Najm I, Ying Z, Yang E, Barkovich AJ, Kwiatkowski DJ, Vinters HV, Madsen JR, Mathern GW, Blümcke I, Poduri A, Walsh CA. Cell Rep. 2017;21:3754-3766. Focal cortical dysplasia (FCD) and hemimegalencephaly (HME) are epileptogenic neurodevelopmental malformations caused by mutations in mTOR pathway genes. Deep sequencing of these genes in FCD/HME brain tissue identified an etiology in 27 (41%) of 66 cases. Radiographically indistinguishable lesions are caused by somatic activating mutations in AKT3, MTOR, and PIK3CA and germline loss-of-function mutations in DEPDC5, NPRL2, and TSC1/2, including TSC2 mutations in isolated HME demonstrating a "two-hit" model. Mutations in the same gene cause a disease continuum from FCD to HME to bilateral brain overgrowth, reflecting the progenitor cell and developmental time when the mutation occurred. Single-cell sequencing demonstrated mTOR activation in neurons in all lesions. Conditional Pik3ca activation in the mouse cortex showed that mTOR activation in excitatory neurons and glia, but not interneurons, is sufficient for abnormal cortical overgrowth. These data suggest that mTOR activation in dorsal telencephalic progenitors, in some cases specifically the excitatory neuron lineage, causes cortical dysplasia.

7.
Epilepsy Curr ; 18(3): 197-199, 2018.
Article in English | MEDLINE | ID: mdl-29950950
8.
Epilepsy Curr ; 18(1): 51-52, 2018.
Article in English | MEDLINE | ID: mdl-29844767
9.
Epilepsy Curr ; 17(6): 377-378, 2017.
Article in English | MEDLINE | ID: mdl-29217986
10.
JAMA Neurol ; 73(7): 836-845, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27159400

ABSTRACT

IMPORTANCE: Focal cortical dysplasia (FCD), hemimegalencephaly, and megalencephaly constitute a spectrum of malformations of cortical development with shared neuropathologic features. These disorders are associated with significant childhood morbidity and mortality. OBJECTIVE: To identify the underlying molecular cause of FCD, hemimegalencephaly, and diffuse megalencephaly. DESIGN, SETTING, AND PARTICIPANTS: Patients with FCD, hemimegalencephaly, or megalencephaly (mean age, 11.7 years; range, 2-32 years) were recruited from Pediatric Hospital A. Meyer, the University of Hong Kong, and Seattle Children's Research Institute from June 2012 to June 2014. Whole-exome sequencing (WES) was performed on 8 children with FCD or hemimegalencephaly using standard-depth (50-60X) sequencing in peripheral samples (blood, saliva, or skin) from the affected child and their parents and deep (150-180X) sequencing in affected brain tissue. Targeted sequencing and WES were used to screen 93 children with molecularly unexplained diffuse or focal brain overgrowth. Histopathologic and functional assays of phosphatidylinositol 3-kinase-AKT (serine/threonine kinase)-mammalian target of rapamycin (mTOR) pathway activity in resected brain tissue and cultured neurons were performed to validate mutations. MAIN OUTCOMES AND MEASURES: Whole-exome sequencing and targeted sequencing identified variants associated with this spectrum of developmental brain disorders. RESULTS: Low-level mosaic mutations of MTOR were identified in brain tissue in 4 children with FCD type 2a with alternative allele fractions ranging from 0.012 to 0.086. Intermediate-level mosaic mutation of MTOR (p.Thr1977Ile) was also identified in 3 unrelated children with diffuse megalencephaly and pigmentary mosaicism in skin. Finally, a constitutional de novo mutation of MTOR (p.Glu1799Lys) was identified in 3 unrelated children with diffuse megalencephaly and intellectual disability. Molecular and functional analysis in 2 children with FCD2a from whom multiple affected brain tissue samples were available revealed a mutation gradient with an epicenter in the most epileptogenic area. When expressed in cultured neurons, all MTOR mutations identified here drive constitutive activation of mTOR complex 1 and enlarged neuronal size. CONCLUSIONS AND RELEVANCE: In this study, mutations of MTOR were associated with a spectrum of brain overgrowth phenotypes extending from FCD type 2a to diffuse megalencephaly, distinguished by different mutations and levels of mosaicism. These mutations may be sufficient to cause cellular hypertrophy in cultured neurons and may provide a demonstration of the pattern of mosaicism in brain and substantiate the link between mosaic mutations of MTOR and pigmentary mosaicism in skin.


Subject(s)
Malformations of Cortical Development/genetics , Megalencephaly/genetics , Mosaicism , Mutation/genetics , TOR Serine-Threonine Kinases/genetics , Adolescent , Adult , Amino Acids/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Association Studies , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Male , Malformations of Cortical Development/diagnostic imaging , Mechanistic Target of Rapamycin Complex 1 , Megalencephaly/diagnostic imaging , Multiprotein Complexes/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Rats , Retrospective Studies , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Young Adult
11.
J Genet Couns ; 25(2): 213-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26536886

ABSTRACT

Next generation sequencing panels have revolutionized the diagnostic approach to patients with epilepsy. There are several commercial epilepsy panels available. We assessed the list of genes tested and consent forms for epilepsy panels available at seven laboratories. The panels varied in the number of genes included (70-465 genes). In some panels, genes not currently associated with epilepsy were included (up to 4 % of panel content). The panels also included genes for lysosomal storage disorders (6-12 %), congenital disorders of glycosylation (0-8.5 %), metabolic disorders (3.5-34 %), neurological syndromes (18-43 %) and multisystemic genetic syndromes (6.4-21 %). Informed consents differed significantly between laboratories ranging from basic information about genetic testing and possible results to information about insurance, genetic counseling and familial testing, and incidental findings.Our findings suggest that it is important to consider the range of genes offered on epilepsy panels and their predicted phenotypes in an effort toward improving the informed consent process.


Subject(s)
Epilepsy/genetics , Genetic Counseling , Genetic Testing , High-Throughput Nucleotide Sequencing , Informed Consent , Epilepsy/diagnosis , Humans , Phenotype
12.
Am J Hum Genet ; 96(4): 682-90, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25839329

ABSTRACT

Developmental epilepsies are age-dependent seizure disorders for which genetic causes have been increasingly identified. Here we report six unrelated individuals with mutations in salt-inducible kinase 1 (SIK1) in a series of 101 persons with early myoclonic encephalopathy, Ohtahara syndrome, and infantile spasms. Individuals with SIK1 mutations had short survival in cases with neonatal epilepsy onset, and an autism plus developmental syndrome after infantile spasms in others. All six mutations occurred outside the kinase domain of SIK1 and each of the mutants displayed autophosphorylation and kinase activity toward HDAC5. Three mutations generated truncated forms of SIK1 that were resistant to degradation and also showed changes in sub-cellular localization compared to wild-type SIK1. We also report the human neuropathologic examination of SIK1-related developmental epilepsy, with normal neuronal morphology and lamination but abnormal SIK1 protein cellular localization. Therefore, these results expand the genetic etiologies of developmental epilepsies by demonstrating SIK1 mutations as a cause of severe developmental epilepsy.


Subject(s)
Autistic Disorder/genetics , Protein Serine-Threonine Kinases/genetics , Spasms, Infantile/genetics , Age Factors , Autistic Disorder/pathology , Base Sequence , Child , DNA Primers/genetics , Electroencephalography , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Immunohistochemistry , Infant , Infant, Newborn , Magnetic Resonance Imaging , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Polymerase Chain Reaction , Spasms, Infantile/pathology
13.
Epilepsia ; 56(3): 422-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25656163

ABSTRACT

OBJECTIVE: Mutations of ATP1A3 have been associated with rapid onset dystonia-parkinsonism and more recently with alternating hemiplegia of childhood. Here we report one child with catastrophic early life epilepsy and shortened survival, and another with epilepsy, episodic prolonged apnea, postnatal microcephaly, and severe developmental disability. Novel heterozygous mutations (p.Gly358Val and p.Ile363Asn) were identified in ATP1A3 in these children. METHODS: Subjects underwent next-generation sequencing under a research protocol. Clinical data were collected retrospectively. The biochemical effects of the mutations on ATP1A3 protein function were investigated. Postmortem neuropathologic specimens from control and affected subjects were studied. RESULTS: The mutations localized to the P domain of the Na,K-ATPase α3 protein, and resulted in significant reduction of Na,K-ATPase activity in vitro. We demonstrate in both control human brain tissue and that from the subject with the p.Gly358Val mutation that ATP1A3 immunofluorescence is prominently associated with interneurons in the cortex, which may provide some insight into the pathogenesis of the disease. SIGNIFICANCE: The findings indicate these mutations cause severe phenotypes of ATP1A3-related disorder spectrum that include catastrophic early life epilepsy, episodic apnea, and postnatal microcephaly.


Subject(s)
Catastrophic Illness , Epilepsy/genetics , Epilepsy/psychology , Mutation/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Brain/metabolism , Brain/pathology , Child, Preschool , DNA Mutational Analysis , Electroencephalography , Enzyme Inhibitors/pharmacology , Epilepsy/complications , Epilepsy/pathology , Female , Glutamate Decarboxylase/metabolism , HEK293 Cells , Humans , Infant , Male , Models, Molecular , Nervous System Diseases/etiology , Ouabain/pharmacology , Transfection
14.
Brain ; 138(Pt 6): 1613-28, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25722288

ABSTRACT

Malformations of cortical development containing dysplastic neuronal and glial elements, including hemimegalencephaly and focal cortical dysplasia, are common causes of intractable paediatric epilepsy. In this study we performed multiplex targeted sequencing of 10 genes in the PI3K/AKT pathway on brain tissue from 33 children who underwent surgical resection of dysplastic cortex for the treatment of intractable epilepsy. Sequencing results were correlated with clinical, imaging, pathological and immunohistological phenotypes. We identified mosaic activating mutations in PIK3CA and AKT3 in this cohort, including cancer-associated hotspot PIK3CA mutations in dysplastic megalencephaly, hemimegalencephaly, and focal cortical dysplasia type IIa. In addition, a germline PTEN mutation was identified in a male with hemimegalencephaly but no peripheral manifestations of the PTEN hamartoma tumour syndrome. A spectrum of clinical, imaging and pathological abnormalities was found in this cohort. While patients with more severe brain imaging abnormalities and systemic manifestations were more likely to have detected mutations, routine histopathological studies did not predict mutation status. In addition, elevated levels of phosphorylated S6 ribosomal protein were identified in both neurons and astrocytes of all hemimegalencephaly and focal cortical dysplasia type II specimens, regardless of the presence or absence of detected PI3K/AKT pathway mutations. In contrast, expression patterns of the T308 and S473 phosphorylated forms of AKT and in vitro AKT kinase activities discriminated between mutation-positive dysplasia cortex, mutation-negative dysplasia cortex, and non-dysplasia epilepsy cortex. Our findings identify PI3K/AKT pathway mutations as an important cause of epileptogenic brain malformations and establish megalencephaly, hemimegalencephaly, and focal cortical dysplasia as part of a single pathogenic spectrum.


Subject(s)
Brain/abnormalities , Hemimegalencephaly/genetics , Malformations of Cortical Development/genetics , Megalencephaly/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Adolescent , Brain/metabolism , Child , Child, Preschool , Class I Phosphatidylinositol 3-Kinases , Female , Genetic Predisposition to Disease/genetics , Hemimegalencephaly/metabolism , Hemimegalencephaly/pathology , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Megalencephaly/metabolism , Megalencephaly/pathology , Mutation , Neuroimaging , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism
15.
Ann Neurol ; 75(1): 22-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24122892

ABSTRACT

OBJECTIVE: A high incidence of structural brain abnormalities has been reported in individuals with pyridoxine-dependent epilepsy (PDE). PDE is caused by mutations in ALDH7A1, also known as antiquitin. How antiquitin dysfunction leads to cerebral dysgenesis is unknown. In this study, we analyzed tissue from a child with PDE as well as control human and murine brain to determine the normal distribution of antiquitin, its distribution in PDE, and associated brain malformations. METHODS: Formalin-fixed human brain sections were subjected to histopathology and fluorescence immunohistochemistry studies. Frozen brain tissue was utilized for measurement of PDE-associated metabolites and Western blot analysis. Comparative studies of antiquitin distribution were performed in developing mouse brain sections. RESULTS: Histologic analysis of PDE cortex revealed areas of abnormal radial neuronal organization consistent with type Ia focal cortical dysplasia. Heterotopic neurons were identified in subcortical white matter, as was cortical astrogliosis, hippocampal sclerosis, and status marmoratus of the basal ganglia. Highly elevated levels of lysine metabolites were present in postmortem PDE cortex. In control human and developing mouse brain, antiquitin immunofluorescence was identified in radial glia, mature astrocytes, ependyma, and choroid plexus epithelium, but not in neurons. In PDE cortex, antiquitin immunofluorescence was greatly attenuated with evidence of perinuclear accumulation in astrocytes. INTERPRETATION: Antiquitin is expressed within glial cells in the brain, and its dysfunction in PDE is associated with neuronal migration abnormalities and other structural brain defects. These malformations persist despite postnatal pyridoxine supplementation and likely contribute to neurodevelopmental impairments.


Subject(s)
Aldehyde Dehydrogenase/biosynthesis , Cerebral Cortex/metabolism , Epilepsy/diagnosis , Epilepsy/metabolism , Neuroglia/metabolism , Adolescent , Animals , Animals, Newborn , Cell Movement/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Mice , Neuroglia/chemistry , Neuroglia/pathology , Pregnancy
16.
Channels (Austin) ; 7(6): 468-72, 2013.
Article in English | MEDLINE | ID: mdl-23965409

ABSTRACT

Dravet Syndrome (DS) is an intractable genetic epilepsy caused by loss-of-function mutations in SCN1A, the gene encoding brain sodium channel Nav 1.1. DS is associated with increased frequency of sudden unexpected death in humans and in a mouse genetic model of this disease. Here we correlate the time course of declining expression of the murine embryonic sodium channel Nav 1.3 and the rise in expression of the adult sodium channel Nav 1.1 with susceptibility to epileptic seizures and increased incidence of sudden death in DS mice. Parallel studies with unaffected human brain tissue demonstrate similar decline in Nav 1.3 and increase in Nav 1.1 with age. In light of these results, we introduce the hypothesis that the natural loss Nav 1.3 channel expression in brain development, coupled with the failure of increase in functional Nav 1.1 channels in DS, defines a tipping point that leads to disinhibition of neural circuits, intractable seizures, co-morbidities, and premature death in this disease.


Subject(s)
Death, Sudden , Epilepsies, Myoclonic/metabolism , Gene Expression Regulation , Sodium Channels/metabolism , Animals , Brain/growth & development , Brain/metabolism , Humans , Mice , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Time Factors
17.
Neurosurg Focus ; 34(6): E8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23724842

ABSTRACT

OBJECT: Imaging-guided surgery (IGS) systems are widely used in neurosurgical practice. During epilepsy surgery, the authors routinely use IGS landmarks to localize intracranial electrodes and/or specific brain regions. The authors have developed a technique to coregister these landmarks with pre- and postoperative scans and the Montreal Neurological Institute (MNI) standard space brain MRI to allow 1) localization and identification of tissue anatomy; and 2) identification of Brodmann areas (BAs) of the tissue resected during epilepsy surgery. Tracking tissue in this fashion allows for better correlation of patient outcome to clinical factors, functional neuroimaging findings, and pathological characteristics and molecular studies of resected tissue. METHODS: Tissue samples were collected in 21 patients. Coordinates from intraoperative tissue localization were downloaded from the IGS system and transformed into patient space, as defined by preoperative high-resolution T1-weighted MRI volume. Tissue landmarks in patient space were then transformed into MNI standard space for identification of the BAs of the tissue samples. RESULTS: Anatomical locations of resected tissue were identified from the intraoperative resection landmarks. The BAs were identified for 17 of the 21 patients. The remaining patients had abnormal brain anatomy that could not be meaningfully coregistered with the MNI standard brain without causing extensive distortion. CONCLUSIONS: This coregistration and landmark tracking technique allows localization of tissue that is resected from patients with epilepsy and identification of the BAs for each resected region. The ability to perform tissue localization allows investigators to relate preoperative, intraoperative, and postoperative functional and anatomical brain imaging to better understand patient outcomes, improve patient safety, and aid in research.


Subject(s)
Epilepsy/pathology , Epilepsy/surgery , Neurosurgical Procedures/methods , Adolescent , Child , Child, Preschool , Electroencephalography , Female , Humans , Infant , Male , Neuroimaging , Tomography, X-Ray Computed
19.
Neurosci Lett ; 483(3): 167-72, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20692323

ABSTRACT

The neurodevelopmental disorder Angelman syndrome is most frequently caused by deletion of the maternally derived chromosome 15q11-q13 region, which includes not only the causative UBE3A gene, but also the beta(3)-alpha(5)-gamma(3) GABA(A) receptor subunit gene cluster. GABAergic dysfunction has been hypothesized to contribute to the occurrence of epilepsy and cognitive and behavioral impairments in this condition. In the present study, analysis of GABA(A) receptor subunit expression and pharmacology was performed in cerebral cortex from four subjects with Angelman syndrome and compared to that from control tissue. The membrane fraction of frozen postmortem neocortical tissue was isolated and subjected to quantitative Western blot analysis. The ratios of beta(3)/beta(2) and alpha(5)/alpha(1) subunit protein expression in Angelman syndrome cortex were significantly decreased when compared with controls. An additional membrane fraction was injected into Xenopus oocytes, resulting in incorporation of the brain membrane vesicles with their associated receptors into the oocyte cellular membrane. Two-electrode voltage-clamp analysis of GABA(A) receptor currents was then performed. Studies of GABA(A) receptor pharmacology in Angelman syndrome cortex revealed increased current enhancement by the alpha(1)-selective benzodiazepine-site agonist zolpidem and by the barbiturate phenobarbital, while sensitivity to current inhibition by zinc was decreased. GABA(A) receptor affinity and modulation by neurosteroids were unchanged. This shift in GABA(A) receptor subunit expression and pharmacology in Angelman syndrome is consistent with impaired extrasynaptic but intact to augmented synaptic cortical GABAergic inhibition, which could contribute to the epileptic, behavioral, and cognitive phenotypes of the disorder.


Subject(s)
Angelman Syndrome/pathology , Cerebral Cortex/metabolism , Gene Expression Regulation/physiology , Protein Subunits/metabolism , Receptors, GABA/metabolism , Adolescent , Adult , Animals , Case-Control Studies , Child, Preschool , Dose-Response Relationship, Drug , Female , GABA Modulators/pharmacology , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Oocytes , Patch-Clamp Techniques/methods , Phenobarbital/pharmacology , Protein Subunits/genetics , Receptors, GABA/genetics , Xenopus , gamma-Aminobutyric Acid/pharmacology
20.
Am J Med Genet A ; 152A(10): 2529-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20799318

ABSTRACT

The primary mechanism of copper transport to the brain is unknown, although this process is drastically impaired in Menkes disease, an X-linked neurodevelopmental disorder caused by mutations in an evolutionarily conserved copper transporter, ATP7A. Potential central nervous system entry routes for copper include brain capillary endothelial cells that originate from mesodermal angioblasts and form the blood-brain barrier, and the choroid plexuses, which derive from embryonic ectoderm, and form the blood-cerebrospinal fluid barrier. We exploited a rare (and first reported) example of somatic mosaicism for an ATP7A mutation to shed light on questions about copper transport into the developing brain. In a 20-month-old Menkes disease patient evaluated before copper treatment, blood copper, and catecholamine concentrations were normal, whereas levels in cerebrospinal fluid were abnormal and consistent with his neurologically severe phenotype. We documented disparate levels of mosaicism for an ATP7A missense mutation, P1001L, in tissues derived from different embryonic origins; allele quantitation showed P1001L in approximately 27% of DNA samples from blood cells (mesoderm-derived) and 88% from cultured fibroblasts (ectoderm-derived). These findings imply that the P1001L mutation in the patient preceded formation of the three primary embryonic lineages at gastrulation, with the ectoderm layer ultimately harboring a higher percentage of mutation-bearing cells than mesoderm or endoderm. Since choroid plexus epithelia are derived from neuroectoderm, and brain capillary endothelial cells from mesodermal angioblasts, the clinical and biochemical findings in this infant support a critical role for the blood-CSF barrier (choroid plexus epithelia) in copper entry to the developing brain.


Subject(s)
Adenosine Triphosphatases/genetics , Brain/metabolism , Cation Transport Proteins/genetics , Copper/metabolism , Menkes Kinky Hair Syndrome/genetics , Amino Acid Substitution , Biological Transport/genetics , Biopsy , Blood-Brain Barrier , Brain/growth & development , Copper/cerebrospinal fluid , Copper/urine , Copper-Transporting ATPases , DNA/blood , DNA/genetics , Dihydroxyphenylalanine/metabolism , Ectoderm/pathology , Humans , Infant , Male , Menkes Kinky Hair Syndrome/pathology , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/metabolism , Mosaicism , Mutation , Mutation, Missense , Reference Values , Skin/pathology , beta 2-Microglobulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...