Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 536
Filter
Add more filters

Publication year range
1.
J Med Genet ; 58(7): 495-504, 2021 07.
Article in English | MEDLINE | ID: mdl-32719099

ABSTRACT

BACKGROUND: Adenosine-to-inosine RNA editing is a co-transcriptional/post-transcriptional modification of double-stranded RNA, catalysed by one of two active adenosine deaminases acting on RNA (ADARs), ADAR1 and ADAR2. ADARB1 encodes the enzyme ADAR2 that is highly expressed in the brain and essential to modulate the function of glutamate and serotonin receptors. Impaired ADAR2 editing causes early onset progressive epilepsy and premature death in mice. In humans, ADAR2 dysfunction has been very recently linked to a neurodevelopmental disorder with microcephaly and epilepsy in four unrelated subjects. METHODS: We studied three children from two consanguineous families with severe developmental and epileptic encephalopathy (DEE) through detailed physical and instrumental examinations. Exome sequencing (ES) was used to identify ADARB1 mutations as the underlying genetic cause and in vitro assays with transiently transfected cells were performed to ascertain the impact on ADAR2 enzymatic activity and splicing. RESULTS: All patients showed global developmental delay, intractable early infantile-onset seizures, microcephaly, severe-to-profound intellectual disability, axial hypotonia and progressive appendicular spasticity. ES revealed the novel missense c.1889G>A, p.(Arg630Gln) and deletion c.1245_1247+1 del, p.(Leu415PhefsTer14) variants in ADARB1 (NM_015833.4). The p.(Leu415PhefsTer14) variant leads to incorrect splicing resulting in frameshift with a premature stop codon and loss of enzyme function. In vitro RNA editing assays showed that the p.(Arg630Gln) variant resulted in a severe impairment of ADAR2 enzymatic activity. CONCLUSION: In conclusion, these data support the pathogenic role of biallelic ADARB1 variants as the cause of a distinctive form of DEE, reinforcing the importance of RNA editing in brain function and development.


Subject(s)
Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Brain Diseases/genetics , Epilepsy/genetics , Neurodevelopmental Disorders/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , Alleles , Brain Diseases/enzymology , Brain Diseases/metabolism , Child , Child, Preschool , Consanguinity , Epilepsy/enzymology , Female , HEK293 Cells , Humans , Mutation , Neurodevelopmental Disorders/enzymology , Pedigree , RNA Editing , RNA-Binding Proteins/metabolism
2.
Int J Mol Sci ; 23(10)2022 May 11.
Article in English | MEDLINE | ID: mdl-35628171

ABSTRACT

Gangliosides (glycosphingolipids containing one or more sialic acids) are highly expressed in neural tissues in vertebrates, and four species (GM1a, GD1a, GD1b, GT1b) are predominant in mammalian brains. GM3 is the precursor of each of these four species and is the major ganglioside in many nonneural tissues. GM3 synthase (GM3S), encoded by ST3GAL5 gene in humans, is a sialyltransferase responsible for synthesis of GM3 from its precursor, lactosylceramide. ST3GAL5 mutations cause an autosomal recessive form of severe infantile-onset neurological disease characterized by progressive microcephaly, intellectual disability, dyskinetic movements, blindness, deafness, intractable seizures, and pigment changes. Some of these clinical features are consistently present in patients with ST3GAL5 mutations, whereas others have variable expression. GM3S knockout (KO) mice have deafness and enhanced insulin sensitivity, but otherwise do not display the above-described neurological defects reported in ST3GAL5 patients. The authors present an overview of physiological functions and pathological aspects of gangliosides based on findings from studies of GM3S KO mice and discuss differential phenotypes of GM3S KO mice versus human GM3S-deficiency patients.


Subject(s)
Deafness , Epilepsy , Sialyltransferases , Animals , Deafness/enzymology , Disease Models, Animal , Epilepsy/enzymology , Humans , Mice , Mice, Knockout , Sialyltransferases/deficiency , Sialyltransferases/metabolism
3.
Am J Hum Genet ; 102(2): 296-308, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395075

ABSTRACT

15q13.3 microdeletion syndrome is characterized by a wide spectrum of neurodevelopmental disorders, including developmental delay, intellectual disability, epilepsy, language impairment, abnormal behaviors, neuropsychiatric disorders, and hypotonia. This syndrome is caused by a deletion on chromosome 15q, which typically encompasses six genes. Here, through studies on OTU deubiquitinase 7A (Otud7a) knockout mice, we identify OTUD7A as a critical gene responsible for many of the cardinal phenotypes associated with 15q13.3 microdeletion syndrome. Otud7a-null mice show reduced body weight, developmental delay, abnormal electroencephalography patterns and seizures, reduced ultrasonic vocalizations, decreased grip strength, impaired motor learning/motor coordination, and reduced acoustic startle. We show that OTUD7A localizes to dendritic spines and that Otud7a-null mice have decreased dendritic spine density compared to their wild-type littermates. Furthermore, frequency of miniature excitatory postsynaptic currents (mEPSCs) is reduced in the frontal cortex of Otud7a-null mice, suggesting a role of Otud7a in regulation of dendritic spine density and glutamatergic synaptic transmission. Taken together, our results suggest decreased OTUD7A dosage as a major contributor to the neurodevelopmental phenotypes associated with 15q13.3 microdeletion syndrome, through the misregulation of dendritic spine density and activity.


Subject(s)
Chromosome Disorders/enzymology , Chromosome Disorders/genetics , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Seizures/enzymology , Seizures/genetics , Action Potentials , Animals , Base Sequence , Behavior, Animal , Chromosome Deletion , Chromosomes, Human, Pair 15/enzymology , Chromosomes, Human, Pair 15/genetics , Dendritic Spines/metabolism , Disease Models, Animal , Electroencephalography , Endopeptidases/deficiency , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/physiopathology , Female , Homozygote , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Synapses/metabolism
4.
Horm Metab Res ; 53(7): 425-434, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34282594

ABSTRACT

We conducted this meta-analysis to assess the effects of sodium valproate (VPA) monotherapy on blood liver enzymes in patients with epilepsy. PubMed, Web of Science, EBSCO, Cochrane Library, Wanfang, China national knowledge infrastructure databases were searched. Nine studies were included. Results showed: (1) The overall SMD for blood AST, ALT, and GGT levels of VPA monotherapy group versus control group were 0.70 (95% CI=0.31 to 1.09, Z=3.52, p=0.0004), 0.47 (95% CI=- 0.01 to 0.95, Z=1.91, p=0.06), 0.44 (95% CI=0.29 to 0.60, Z=5.55, p<0.00001), respectively. (2) In subgroup meta-analysis, increased blood AST and GGT levels were observed in epileptic minors (AST: total SMD=0.85, 95% CI=0.40 to 1.30, Z=3.69, p=0.0002; GGT: total SMD=0.46, 95% CI=0.29 to 0.63, Z=5.25, p<0.00001). Elevated blood ALT level was observed in Asian patients receiving VPA monotherapy (total SMD=0.70, 95% CI=0.51 to 0.90, Z=7.01, p<0.00001), and the early stage of VPA monotherapy (total SMD=0.93, 95% CI=0.57 to 1.29, Z=5.09, p<0.00001). Overall, our results indicated that blood AST and GGT were significantly increased in epileptic minors receiving VPA monotherapy. The elevation of blood ALT was observed in Asian patients and the early stage of VPA monotherapy. However, due to the small number of included studies, our results should be considered with caution.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Liver/enzymology , Valproic Acid/therapeutic use , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Epilepsy/enzymology , Epilepsy/pathology , Humans , Liver/drug effects , gamma-Glutamyltransferase/blood
5.
J Hum Genet ; 65(5): 481-485, 2020 May.
Article in English | MEDLINE | ID: mdl-32005903

ABSTRACT

p21-activated kinases (PAKs) are protein serine/threonine kinases stimulated by Rho-family p21 GTPases such as CDC42 and RAC. PAKs have been implicated in several human disorders, with pathogenic variants in PAK3 associated with intellectual disability and several PAK members, especially PAK1 and PAK4, overexpressed in human cancer. Recently, de novo PAK1 variants were reported to be causative of neurodevelopmental disorder (ND) with secondary macrocephaly in three patients. We herein report a fourth patient with ND, epilepsy, and macrocephaly caused by a de novo PAK1 missense variant. Two previously reported missense PAK1 variants functioned as activating alleles by reducing PAK1 homodimerization. To examine the pathogenicity of the identified novel p.Ser110Thr variant, we carried out in silico structural analysis. Our findings suggest that this variant also prevents PAK1 homodimerization, leading to constitutive PAK1 activation.


Subject(s)
Epilepsy , Megalencephaly , Mutation, Missense , Neurodevelopmental Disorders , Protein Multimerization , p21-Activated Kinases , Amino Acid Substitution , Child , Enzyme Activation/genetics , Epilepsy/enzymology , Epilepsy/genetics , Humans , Male , Megalencephaly/enzymology , Megalencephaly/genetics , Neurodevelopmental Disorders/enzymology , Neurodevelopmental Disorders/genetics , Protein Domains , p21-Activated Kinases/chemistry , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
6.
J Hum Genet ; 65(9): 751-757, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32405030

ABSTRACT

Inborn errors of metabolism can cause epileptic encephalopathies. Biallelic loss-of-function variants in the ITPA gene, encoding inosine triphosphate pyrophosphatase (ITPase), have been reported in epileptic encephalopathies with lack of myelination of the posterior limb of the internal capsule, brainstem tracts, and tracts to the primary visual and motor cortices (MIM:616647). ITPase plays an important role in purine metabolism. In this study, we identified two novel homozygous ITPA variants, c.264-1 G > A and c.489-1 G > A, in two unrelated consanguineous families. The probands had epilepsy, microcephaly with characteristic magnetic resonance imaging findings (T2 hyperintensity signals in the pyramidal tracts of the internal capsule, delayed myelination, and thin corpus callosum), hypotonia, and developmental delay; both died in early infancy. Our report expands the knowledge of clinical consequences of biallelic ITPA variants.


Subject(s)
Brain Diseases/genetics , Developmental Disabilities/genetics , Epilepsy/genetics , Genetic Predisposition to Disease , Multiple Organ Failure/genetics , Muscle Hypotonia/genetics , Pyrophosphatases/genetics , Brain Diseases/complications , Brain Diseases/enzymology , Brain Diseases/mortality , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Developmental Disabilities/complications , Developmental Disabilities/enzymology , Developmental Disabilities/mortality , Epilepsy/complications , Epilepsy/enzymology , Epilepsy/mortality , Female , Genotype , Homozygote , Humans , Infant , Magnetic Resonance Imaging , Male , Multiple Organ Failure/complications , Multiple Organ Failure/enzymology , Multiple Organ Failure/mortality , Muscle Hypotonia/complications , Muscle Hypotonia/enzymology , Muscle Hypotonia/mortality , Mutation , Pedigree , Pyramidal Tracts/diagnostic imaging , Pyramidal Tracts/pathology , Exome Sequencing
7.
FASEB J ; 33(12): 14281-14295, 2019 12.
Article in English | MEDLINE | ID: mdl-31661303

ABSTRACT

Blood-brain barrier dysfunction in epilepsy contributes to seizures and resistance to antiseizure drugs. Reports show that seizures increase brain glutamate levels, leading to barrier dysfunction. One component of barrier dysfunction is overexpression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP). Based on our previous studies, we hypothesized that glutamate released during seizures activates cytosolic phospholipase A2 (cPLA2), resulting in P-gp and BCRP overexpression. We exposed isolated rat brain capillaries to glutamate ex vivo and used an in vivo-ex vivo approach of isolating brain capillaries from rats after status epilepticus (SE) and in chronic epileptic (CE) rats. Glutamate increased cPLA2, P-gp, and BCRP protein and activity levels in isolated brain capillaries. We confirmed the role of cPLA2 in the signaling pathway in brain capillaries from male and female mice lacking cPLA2. We also demonstrated, in vivo, that cPLA2 inhibition prevents overexpression of P-gp and BCRP at the blood-brain barrier in rats after status epilepticus and in CE rats. Our data support the hypothesis that glutamate signals cPLA2 activation, resulting in overexpression of blood-brain barrier P-gp and BCRP.-Hartz, A. M. S., Rempe, R. G., Soldner, E. L. B., Pekcec, A., Schlichtiger, J., Kryscio, R., Bauer, B. Cytosolic phospholipase A2 is a key regulator of blood-brain barrier function in epilepsy.


Subject(s)
Blood-Brain Barrier/enzymology , Epilepsy/enzymology , Group IV Phospholipases A2/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/enzymology , Capillaries/enzymology , Epilepsy/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Genotype , Glutamic Acid/pharmacology , Group IV Phospholipases A2/genetics , Male , Mice , Rats , Rats, Sprague-Dawley
8.
Am J Hum Genet ; 99(4): 912-916, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27616480

ABSTRACT

The risk of epilepsy among individuals with intellectual disability (ID) is approximately ten times that of the general population. From a cohort of >5,000 families affected by neurodevelopmental disorders, we identified six consanguineous families harboring homozygous inactivating variants in MBOAT7, encoding lysophosphatidylinositol acyltransferase (LPIAT1). Subjects presented with ID frequently accompanied by epilepsy and autistic features. LPIAT1 is a membrane-bound phospholipid-remodeling enzyme that transfers arachidonic acid (AA) to lysophosphatidylinositol to produce AA-containing phosphatidylinositol. This study suggests a role for AA-containing phosphatidylinositols in the development of ID accompanied by epilepsy and autistic features.


Subject(s)
Acyltransferases/genetics , Autistic Disorder/genetics , Epilepsy/genetics , Intellectual Disability/genetics , Membrane Proteins/genetics , Mutation , Acyltransferases/metabolism , Arachidonic Acid/metabolism , Autistic Disorder/complications , Autistic Disorder/enzymology , Autistic Disorder/metabolism , Child , Child, Preschool , Consanguinity , Epilepsy/complications , Epilepsy/enzymology , Epilepsy/metabolism , Female , Homozygote , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/enzymology , Intellectual Disability/metabolism , Lysophospholipids/metabolism , Male , Membrane Proteins/metabolism , Pedigree , Phosphatidylinositols/metabolism
9.
J Neuroinflammation ; 16(1): 197, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31666079

ABSTRACT

Epilepsy, a common multifactorial neurological disease, affects about 69 million people worldwide constituting nearly 1% of the world population. Despite decades of extensive research on understanding its underlying mechanism and developing the pharmacological treatment, very little is known about the biological alterations leading to epileptogenesis. Due to this gap, the currently available antiepileptic drug therapy is symptomatic in nature and is ineffective in 30% of the cases. Mounting evidences revealed the pathophysiological role of neuroinflammation in epilepsy which has shifted the focus of epilepsy researchers towards the development of neuroinflammation-targeted therapeutics for epilepsy management. Markedly increased expression of key inflammatory mediators in the brain and blood-brain barrier may affect neuronal function and excitability and thus may increase seizure susceptibility in preclinical and clinical settings. Cyclooxygenase-2 (COX-2), an enzyme synthesizing the proinflammatory mediators, prostaglandins, has widely been reported to be induced during seizures and is considered to be a potential neurotherapeutic target for epilepsy management. However, the efficacy of such therapy involving COX-2 inhibition depends on various factors viz., therapeutic dose, time of administration, treatment duration, and selectivity of COX-2 inhibitors. This article reviews the preclinical and clinical evidences supporting the role of COX-2 in seizure-associated neuroinflammation in epilepsy and the potential clinical use of COX-2 inhibitors as a future strategy for epilepsy treatment.


Subject(s)
Anticonvulsants/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Management , Epilepsy/diagnosis , Epilepsy/drug therapy , Animals , Clinical Trials as Topic/methods , Epilepsy/enzymology , Forecasting , Humans
10.
Cell Mol Neurobiol ; 39(1): 149-160, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30539418

ABSTRACT

Epilepsy is a common disease presenting with recurrent seizures. Hippocampal sclerosis (HS) is the commonest histopathological alteration in patients with temporal lobe epilepsy (TLE) undergoing surgery. HS physiopathogenesis is debatable. We have recently studied, by using mass spectrometry-based proteomics, an experimental model of TLE induced by electrical stimulation. Specifically, protein expressions of both the beta subunit of mitochondrial ATP synthase (ATP5B) and of membrane ATPases were found to be reduced. Here, we investigated tissue distribution of ATP5B and sodium/potassium-transporting ATPase subunit alpha-3 (NKAα3), a protein associated with neuromuscular excitability disorders, in human hippocampi resected "en bloc" for HS treatment (n = 15). We used immunohistochemistry and the stained area was digitally evaluated (increase in binary contrast of microscopic fields) in the hippocampal sectors (CA1-CA4) and dentate gyrus. All HS samples were classified as Type 1, according to the International League Against Epilepsy (ILAE) 2013 Classification (predominant cell loss in CA1 and CA4). ATP5B was significantly decreased in all sectors and dentate gyrus of HS patients compared with individuals submitted to necropsy and without history of neurological alterations (n = 10). NKAα3 expression showed no difference. Moreover, we identified a negative correlation between frequency of pre-operative seizures and number of neurons in CA1. In conclusion, our data showed similarity between changes in protein expression in a model of TLE and individuals with HS. ATP5B reduction would be at least in part due to neuronal loss. Future investigations on ATP5B activity could provide insights into the process of such cell loss.


Subject(s)
Epilepsy/enzymology , Hippocampus/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Sclerosis/enzymology , Adolescent , Adult , Cell Count , Dentate Gyrus/pathology , Epilepsy/pathology , Female , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/metabolism , Neurons/pathology , Sclerosis/pathology , Sodium-Potassium-Exchanging ATPase , Young Adult
11.
J Recept Signal Transduct Res ; 38(5-6): 492-498, 2018.
Article in English | MEDLINE | ID: mdl-31038026

ABSTRACT

Epilepsy affects approximately 50-70 million people worldwide and 30-40% of patients do not benefit from medication. Therefore, it is necessary to identify novel targets for epileptic treatments. c-Jun N-terminal kinase (JNK) is a member of the mitogen-activated protein kinase (MAPK) family that activates diverse substrates, such as transcriptional factors, adaptor proteins, and signaling proteins, and has a wide variety of functions in both physiological and pathological conditions. The excessive activation of JNK is found not only in the acute phase of epilepsy, but also in the chronic phase, which potentiates it as a promising target in epilepsy control. In this review, we discuss the activation of the JNK pathway in epilepsy and its role in neuronal death, astrocyte activation, and mossy fiber sprouting (MFS) based on recent updates. Finally, we briefly introduce the current agents that target JNK signaling to control epilepsy.


Subject(s)
Epilepsy/genetics , Epilepsy/therapy , JNK Mitogen-Activated Protein Kinases/genetics , Epilepsy/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/therapeutic use , MAP Kinase Signaling System/genetics , Molecular Targeted Therapy , Neurons/pathology , Signal Transduction/genetics
12.
Neuroradiology ; 60(3): 239-245, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29353399

ABSTRACT

PURPOSE: Glutamic acid decarboxylase (GAD65) has been implicated in a number of autoimmune-associated neurologic syndromes, including autoimmune epilepsy. This study categorizes the spectrum of MRI findings in patients with a clinical diagnosis of autoimmune epilepsy and elevated serum GAD65 autoantibodies. METHODS: An institutional database search identified patients with elevated serum GAD65 antibodies and a clinical diagnosis of autoimmune epilepsy who had undergone brain MRI. Imaging studies were reviewed by three board-certified neuroradiologists and one neuroradiology fellow. Studies were evaluated for cortical/subcortical and hippocampal signal abnormality, cerebellar and cerebral volume loss, mesial temporal sclerosis, and parenchymal/leptomeningeal enhancement. The electronic medical record was reviewed for relevant clinical information and laboratory markers. RESULTS: A study cohort of 19 patients was identified. The majority of patients were female (84%), with a mean age of onset of 27Ā years. Serum GAD65 titers ranged from 33 to 4415Ā nmol/L (normal < 0.02Ā nmol/L). The most common presentation was medically intractable, complex partial seizures with temporal lobe onset. Parenchymal atrophy was the most common imaging finding (47%), with a subset of patients demonstrating cortical/subcortical parenchymal T2 hyperintensity (37%) or abnormal hippocampal signal (26%). No patients demonstrated abnormal parenchymal/leptomeningeal enhancement. CONCLUSION: The most common MRI finding in GAD65-associated autoimmune epilepsy is disproportionate parenchymal atrophy for age, often associated with abnormal cortical/subcortical T2 hyperintensities. Hippocampal abnormalities are seen in a minority of patients. This constellation of findings in a patient with medically intractable epilepsy should raise the possibility of GAD65 autoimmunity.


Subject(s)
Autoimmune Diseases/diagnostic imaging , Autoimmune Diseases/enzymology , Epilepsy/diagnostic imaging , Epilepsy/enzymology , Glutamate Decarboxylase/metabolism , Magnetic Resonance Imaging/methods , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Retrospective Studies
13.
Cereb Cortex ; 27(3): 2226-2248, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27005990

ABSTRACT

Alterations in the balance of inhibitory and excitatory synaptic transmission have been implicated in the pathogenesis of neurological disorders such as epilepsy. Eukaryotic elongation factor 2 kinase (eEF2K) is a highly regulated, ubiquitous kinase involved in the control of protein translation. Here, we show that eEF2K activity negatively regulates GABAergic synaptic transmission. Indeed, loss of eEF2K increases GABAergic synaptic transmission by upregulating the presynaptic protein Synapsin 2b and α5-containing GABAA receptors and thus interferes with the excitation/inhibition balance. This cellular phenotype is accompanied by an increased resistance to epilepsy and an impairment of only a specific hippocampal-dependent fear conditioning. From a clinical perspective, our results identify eEF2K as a potential novel target for antiepileptic drugs, since pharmacological and genetic inhibition of eEF2K can revert the epileptic phenotype in a mouse model of human epilepsy.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Epilepsy/enzymology , Neurons/enzymology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Conditioning, Psychological/physiology , Disease Models, Animal , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Epilepsy/pathology , Fear/physiology , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neurons/pathology , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
14.
Int J Clin Pharmacol Ther ; 56(7): 337-346, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29628024

ABSTRACT

OBJECTIVE: Therapeutic response to phenytoin (PHT), a first-line antiepileptic drug (AED), is highly variable, in part likely due to genetic factors. Genetic polymorphisms in cytochrome P450 (CYP) 2C9 and CYP2C19 are expected to affect the metabolism of PHT and consequently affect its maintenance doses. We aimed to clarify the effects of genetic polymorphisms in both enzymes on daily PHT maintenance dosage in Asian epileptic patients by meta-analysis. MATERIALS AND METHODS: A systematic literature search was conducted in PubMed and EMBASE for relevant studies published prior to April 14, 2017. RevMan 5.2.3 software was used to analyze the relationship between CYP2C9/2C19 polymorphisms and PHT maintenance doses. RESULTS: A total of 6 studies with 993 patients fulfilling the inclusion criteria were included in our meta-analysis. The homozygous and heterozygous CYP2C19 mutation group (i.e., CYP2C19*2/*2, CYP2C19*3/*3, or CYP2C19*2/*3 group) required significant decrease of PHT maintenance dose. The starting maintenance dose suggested in this group is 4.38Ā mg/kg/day. Patients with heterozygous CYP2C9 or both heterozygous CYP2C9 and CYP2C19 showed a trend but not a statistically-significant decrease of PHT dose, but dosage adjustment was recommended. CONCLUSION: The meta-analysis indicates that CYP2C9 and CYP2C19 polymorphisms are associated with lower PHT maintenance dosage in Asian epileptic patients. Ethnic differences can influence PHT maintenance dose.Ć¢Ā€Ā©.


Subject(s)
Anticonvulsants/administration & dosage , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C9/genetics , Epilepsy/drug therapy , Pharmacogenomic Variants , Phenytoin/administration & dosage , Polymorphism, Single Nucleotide , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Asian People/genetics , Chi-Square Distribution , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C9/metabolism , Drug Monitoring , Epilepsy/enzymology , Epilepsy/ethnology , Epilepsy/genetics , Heterozygote , Homozygote , Humans , Pharmacogenetics , Phenytoin/adverse effects , Phenytoin/pharmacokinetics , Risk Factors , Treatment Outcome
15.
Postepy Biochem ; 64(3): 222-230, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30656907

ABSTRACT

Epileptogenesis is the process responsible for converting normal brain into an epileptic. It may be triggered by an event such as brain injury or status epilepticus (SE). The main mechanisms responsible include neuroinflammation and blood-brain barrier (BBB) disruption, pathologic neuronal networks' reorganisation and aberrant synaptic plasticity. Accumulating amount of evidence from animal models and epileptic patients strongly suggest that matrix metalloproteinase 9 (MMP-9) is potentially one of the key executors of the processes of epileptogenesis. MMP-9 by affecting synaptic plasticity is suggested to enable epileptic remodelling of the brain circuitry. MMP-9's dependent cleavage of BBB followed by inflammatory cell infiltration into the brain contributes to the neuroinflamation component of epileptogenesis. The goal of this review was to analyse all possible ways MMP-9 may be involved in epileptogenesis and consider MMP-9 inhibition as potential therapeutic strategy.


Subject(s)
Epilepsy/enzymology , Epilepsy/prevention & control , Matrix Metalloproteinase 9/metabolism , Animals , Blood-Brain Barrier , Brain/enzymology , Brain/pathology , Disease Progression , Epilepsy/pathology , Humans , Neuronal Plasticity
16.
Pharmacogenomics J ; 17(3): 242-251, 2017 06.
Article in English | MEDLINE | ID: mdl-26951882

ABSTRACT

CYP1A1 gene is involved in estrogen metabolism, and previously, we have reported association of variant rs2606345 with altered anti-epileptic drugs (AED) response in North Indian women with epilepsy (WWE). The present study aims to replicate the pharmacogenetic association, perform functional characterization and study its distribution within ethnically diverse Indian population. The variant was genotyped in 351 patients to assess the pharmacogenetic association and 552 healthy individuals belonging to 24 different ethnic groups to examine the distribution in Indian population. We observed significant overrepresentation of 'A' allele and 'AA' genotype in poor responders in WWE at Bonferroni-corrected significance levels. The recessive allele was found to lower the promoter activity by ~70-80% which was further substantiated by thermally less stable hairpin formed by it (ΔTm=7 Ā°C). Among all ethnic groups, west Indo-European (IE-W-LP2) subpopulation showed highest genotypic frequency of the variant making women from this community more prone to poor AED response. Our results indicate that rs2606345 influences drug response in WWE by lowering CYP1A1 expression.


Subject(s)
Anticonvulsants/therapeutic use , Cytochrome P-450 CYP1A1/genetics , Epilepsy/drug therapy , Pharmacogenomic Variants , Adolescent , Adult , Anticonvulsants/adverse effects , Case-Control Studies , Cytochrome P-450 CYP1A1/metabolism , Epilepsy/enzymology , Epilepsy/ethnology , Epilepsy/genetics , Female , Gene Frequency , HEK293 Cells , Heterozygote , Homozygote , Humans , India/epidemiology , MCF-7 Cells , Male , Pharmacogenetics , Pharmacogenomic Testing , Phenotype , Promoter Regions, Genetic , Racial Groups/genetics , Recurrence , Transfection , Treatment Outcome , Young Adult
17.
Exp Mol Pathol ; 102(1): 115-122, 2017 02.
Article in English | MEDLINE | ID: mdl-28087392

ABSTRACT

Gliomas, especially low-grade gliomas, are highly epileptogenic brain tumors. Histopathological information is valuable in evaluating the diagnosis and/or biologic behavior of various gliomas. Here we explored the clinical data and histopathological predictors of the occurrence of epilepsy in patients with gliomas. A retrospective study examined 310 consecutive patients who had undergone surgical treatment for gliomas in our institution from January 2013 to January 2015. Clinical data and pathological examination results were analyzed. Literatures regarding the predictors and etiology of glioma associated epileptic seizures in the period of 1995-2015 were also reviewed. A total of 234 (75.5%) astrocytic tumors and 76 (24.5%) oligodendrial tumors were included. At diagnosis, 33.6% of patients had epileptic seizures. Multivariate analysis revealed cortex involvement (OR=7.991, 95%CI=1.599-39.926), lower World Health Organization grade (OR=3.584, 95%CI=1.032-12.346) and topoisomerase II (TopoII) positivity (OR=0.943, 95%CI=0.903-0.982) were strong predictors for preoperative epileptic seizures. Gender, disease course, tumor classification, location or volume did not significantly affect epileptic seizure occurrence. Forty-three publications involved glioma-associated epilepsy were found in PubMed online database and key data were extracted and summarized. The present studies on glioma-related epilepsy are relatively limited and inconsistent. Low-grade gliomas, cortex involvement and TopoII positivity were independent predictors of a history of epileptic seizures at diagnosis. Further studies to examine the underlying mechanism of topoisomerase II as well as other molecules in epilepsy occurrence in brain gliomas are needed in the future.


Subject(s)
Brain Neoplasms/epidemiology , DNA Topoisomerases, Type II/metabolism , Epilepsy/epidemiology , Glioma/epidemiology , Adolescent , Adult , Aged , Brain/metabolism , Brain/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/surgery , Child , China/epidemiology , Comorbidity , Epilepsy/enzymology , Female , Glioma/enzymology , Glioma/surgery , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Preoperative Period , Prognosis , Retrospective Studies , Young Adult
18.
Dev Med Child Neurol ; 59(10): 1063-1070, 2017 10.
Article in English | MEDLINE | ID: mdl-28543354

ABSTRACT

AIM: Mucopolysaccharidosis type II (MPS II) is caused by variants in the iduronate-2-sulphatase gene (IDS). Patients can be either neuronopathic with intellectual disability, or non-neuronopathic. Few studies have reported on the IDS genotype-phenotype relationship and on the molecular effects involved. We addressed this in a cohort study of Dutch patients with MPS II. METHOD: Intellectual performance was assessed for school performance, behaviour, and intelligence. Urinary glycosaminoglycans were quantified by mass spectrometry. IDS variants were analysed in expression studies for enzymatic activity and processing by immunoblotting. RESULTS: Six patients had a non-neuronopathic phenotype and 11 a neuronopathic phenotype, three of whom had epilepsy. Total deletion of IDS invariably resulted in the neuronopathic phenotype. Phenotypes of seven known IDS variants were consistent with the literature. Expression studies of nine variants were novel and showed impaired IDS enzymatic activity, aberrant intracellular processing, and elevated urinary excretion of heparan sulphate and dermatan sulphate irrespective of the MPS II phenotype. INTERPRETATION: We speculate that very low or cell-type-specific IDS residual activity is sufficient to prevent the neuronal phenotype of MPS II. Whereas the molecular effects of IDS variants do not distinguish between MPS II phenotypes, the IDS genotype is a strong predictor.


Subject(s)
Genetic Variation , Glycoproteins/genetics , Glycoproteins/metabolism , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/psychology , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Educational Status , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/psychology , Genetic Association Studies , Glycosaminoglycans/urine , Humans , Immunoblotting , Intelligence , Mass Spectrometry , Middle Aged , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/metabolism , Netherlands , Phenotype , Young Adult
19.
Med Arch ; 71(4): 239-242, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28974841

ABSTRACT

INTRODUCTION: Epilepsy is one of the most common neurological diseases in childhood and adolescence. Carbamazepine (CBZ) and valproate (VPA) have been widely used as the first generation of antiepileptic drugs (AED). AIM: The aim of the study has been to evaluate and compare the effect of CBZ and VPA monotherapy on aspartate aminotransferase (AST), alanine aminotransferase (ALT) and gamma-glutamyltransferase (GGT) serum levels in children. MATERIAL AND METHODS: The study has included 100 patients (boys 57/girls 43, age range 1 to 18 years), who have been treated with CBZ or VPA, as initial monotherapy, for at least 12 months. Patients with liver lesions or patients who have been treated with other drugs have been excluded from the study. The initial serum enzyme levels (AST, ALT and GGT) and after 12 months of treatment have been compared. RESULTS: 53/100 (53%) patients have been treated with CBZ and 47/100 (47%) patients have been treated with VPA.The initial level of enzymes were within the referece range. After one year-long treatment AST was elevated at 4/53 (7.5%) CBZ patients and 9/47 (19.15%) VPA patients (x2 test =3.965, p<0.05). ALT was elevated at 5/53 (9.4%) CBZ patients and 9/47 (19.15%) VPA patients (x2 test =6.953, p<0.05). GGT was elevated at 18/53 (34%) CBZ patients and 7/47 (14.9%) VPA patients (x2 test =4.831, p<0.05). CONCLUSION: The levels of enzymes AST and ALT have been elevated statistically significant in VPA group and GGT in CBZ group.


Subject(s)
Alanine Transaminase/blood , Anticonvulsants/pharmacology , Aspartate Aminotransferases/blood , Carbamazepine/pharmacology , Epilepsy/drug therapy , Epilepsy/enzymology , Valproic Acid/pharmacology , gamma-Glutamyltransferase/blood , Adolescent , Anticonvulsants/therapeutic use , Carbamazepine/therapeutic use , Child , Child, Preschool , Cross-Sectional Studies , Epilepsy/blood , Female , Humans , Infant , Liver/drug effects , Liver/enzymology , Male , Treatment Outcome , Valproic Acid/therapeutic use
20.
Radiology ; 278(3): 822-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26397127

ABSTRACT

PURPOSE: To determine if myeloperoxidase (MPO) is involved in epileptogenesis and if molecular nuclear imaging can be used to noninvasively map inflammatory changes in epileptogenesis. MATERIALS AND METHODS: The animal and human studies were approved by the institutional review boards. Pilocarpine-induced epileptic mice were treated with 4-aminobenzoic acid hydrazide (n = 46), a specific irreversible MPO inhibitor, or saline (n = 42). Indium-111-bis-5-hydroxytryptamide-diethylenetriaminepentaacetate was used to image brain MPO activity (n = 6 in the 4-aminobenzoic acid hydrazide and saline groups; n = 5 in the sham group) by using single photon emission computed tomography/computed tomography. The role of MPO in the development of spontaneous recurrent seizures was assessed by means of clinical symptoms and biochemical and histopathologic data. Human brain specimens from a patient with epilepsy and a patient without epilepsy were stained for MPO. The Student t test, one-way analysis of variance, and Mann-Whitney and Kruskal-Wallis tests were used. Differences were regarded as significant if P was less than .05. RESULTS: MPO and leukocytes increased in the brain during epileptogenesis (P < .05). Blocking MPO delayed spontaneous recurrent seizures (99.6 vs 142 hours, P = .016), ameliorated the severity of spontaneous recurrent seizures (P < .05), and inhibited mossy fiber sprouting (Timm index, 0.31 vs 0.03; P = .003). Matrix metalloproteinase activity was upregulated during epileptogenesis in an MPO-dependent manner (1.44 vs 0.94 U/mg, P = .049), suggesting that MPO acts upstream of matrix metalloproteinases. MPO activity was mapped during epileptogenesis in vivo in the hippocampal regions. Resected temporal lobe tissue from a human patient with refractory epilepsy but not the temporal lobe tissue from a patient without seizures demonstrated positive MPO immunostaining, suggesting high translational potential for this imaging technology. CONCLUSION: The findings of this study highlight an important role for MPO in epileptogenesis and show MPO to be a potential therapeutic target and imaging biomarker for epilepsy.


Subject(s)
Epilepsy/diagnostic imaging , Epilepsy/enzymology , Multimodal Imaging , Peroxidase/metabolism , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , 4-Aminobenzoic Acid , Animals , Blotting, Western , Disease Models, Animal , Flow Cytometry , Mice , Pilocarpine
SELECTION OF CITATIONS
SEARCH DETAIL