Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 503
Filter
1.
BMC Neurol ; 24(1): 169, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783211

ABSTRACT

BACKGROUND: Progressive Myoclonic Epilepsy (PME) is a group of rare diseases that are difficult to differentiate from one another based on phenotypical characteristics. CASE REPORT: We report a case of PME type 7 due to a pathogenic variant in KCNC1 with myoclonus improvement after epileptic seizures. DISCUSSION: Myoclonus improvement after seizures may be a clue to the diagnosis of Progressive Myoclonic Epilepsy type 7.


Subject(s)
Myoclonic Epilepsies, Progressive , Seizures , Humans , Myoclonic Epilepsies, Progressive/complications , Myoclonic Epilepsies, Progressive/diagnosis , Seizures/diagnosis , Seizures/complications , Seizures/etiology , Seizures/drug therapy , Myoclonus/diagnosis , Myoclonus/etiology , Myoclonus/complications , Myoclonus/drug therapy , Male , Shaw Potassium Channels/genetics , Female , Electroencephalography/methods
2.
Epilepsy Res ; 203: 107380, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38781737

ABSTRACT

OBJECTIVE: North Sea Progressive Myoclonus Epilepsy (NS-PME) is a rare genetic disorder characterized by ataxia, myoclonus and seizures with a progressive course. Although the cause of NS-PME is known, namely a homozygous mutation in the GOSR2 gene (c.430 G>T; p. Gly144Trp), sufficient treatment is lacking. Despite combinations of on average 3-5 anti-seizure medications (ASMs), debilitating myoclonus and seizures persist. Here we aimed to gain insight into the most effective anti-convulsive target in NS-PME by evaluating the individual effects of ASMs in a NS-PME Drosophila model. METHOD: A previously generated Drosophila model for NS-PME was used displaying progressive heat-sensitive seizures. We used this model to test 1. a first-generation ASM (sodium barbital), 2. common ASMs used in NS-PME (clonazepam, valproic acid, levetiracetam, ethosuximide) and 3. a novel third-generation ASM (ganaxolone) with similar mode of action to sodium barbital. Compounds were administered by adding them to the food in a range of concentrations. After 7 days of treatment, the percentage of heat-induced seizures was determined and compared to non-treated but affected controls. RESULTS: As previously reported in the NS-PME Drosophila model, sodium barbital resulted in significant seizure suppression, with increasing effect at higher dosages. Of the commonly prescribed ASMs, clonazepam and ethosuximide resulted in significant seizure suppression, whereas both valproic acid and levetiracetam did not show any changes in seizures. Interestingly, ganaxolone did result in seizure suppression as well. CONCLUSION: Of the six drugs tested, three of the four that resulted in seizure suppression (sodium barbital, clonazepam, ganaxolone) are primary known for their direct effect on GABAA receptors. This suggests that GABAA could be a potentially important target in the treatment of NS-PME. Consequently, these findings add rationale to the exploration of the clinical effect of ganaxolone in NS-PME and other progressive myoclonus epilepsies.


Subject(s)
Anticonvulsants , Disease Models, Animal , Drosophila , Myoclonic Epilepsies, Progressive , Animals , Anticonvulsants/therapeutic use , Anticonvulsants/pharmacology , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/drug therapy , Animals, Genetically Modified , Receptors, GABA-A/genetics , Receptors, GABA-A/drug effects
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 432-436, 2024 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-38565508

ABSTRACT

OBJECTIVE: To retrospectively analyze the clinical phenotype and pathogenic variants in patients with Progressive myoclonus epilepsy (PME). METHODS: Clinical data and results of genetic testing for 11 patients diagnosed with PME at the Department of Neurology, the First Affiliated Hospital of Zhejiang University School of Medicine from June 2017 to December 2022 were collected and analyzed. RESULTS: All of the patients, including 4 males and 7 females, had predominant action myoclonus. Three patients had myoclonus as the initial manifestation, whilst eight were diagnosed through genetic testing, including three cases with NEU1 gene variants, two with EPM2A gene variants (1 was novel), one with MT-TK gene variant, one with ATN1 gene variant, and one with CSTB gene variant. No pathogenic variant was identified in the remaining three cases. Among the eight patients with a genetic diagnosis, three were diagnosed with sialidosis, two with Lafora disease, one with Dentatorubral-pallidoluysian atrophy (DRPLA), one with Unverricht-Lundborg disease (ULD), and one with Myoclonic epilepsy with ragging red fibers (MERRF). CONCLUSION: Compared with pediatric patients, adult patients with PME represent a distinct subtype with slower progression and milder cognitive impairment.


Subject(s)
Epilepsies, Myoclonic , Myoclonic Epilepsies, Progressive , Unverricht-Lundborg Syndrome , Male , Adult , Female , Humans , Child , Unverricht-Lundborg Syndrome/genetics , Retrospective Studies , Myoclonic Epilepsies, Progressive/genetics , Epilepsies, Myoclonic/genetics , Genetic Testing
4.
Neurosci Res ; 204: 58-63, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38458494

ABSTRACT

Neurodegenerative diseases (ND) affect distinct populations of neurons and manifest various clinical and pathological symptoms. A subset of ND prognoses has been linked to vascular risk factors. Consequently, the current study investigated retinal vascular abnormalities in a murine model of Lafora neurodegenerative disease (LD), a fatal and genetic form of progressive myoclonus epilepsy that affects children. Here, arterial rigidity was evaluated by measuring pulse wave velocity and vasculature deformations in the retina. Our findings in the LD mouse model indicate altered pulse wave velocity, retinal vascular thinning, and convoluted retinal arteries.


Subject(s)
Disease Models, Animal , Lafora Disease , Retinal Vessels , Animals , Lafora Disease/genetics , Lafora Disease/pathology , Lafora Disease/physiopathology , Mice , Retinal Vessels/pathology , Mice, Inbred C57BL , Male , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/physiopathology , Myoclonic Epilepsies, Progressive/pathology
6.
Genes (Basel) ; 15(2)2024 01 27.
Article in English | MEDLINE | ID: mdl-38397161

ABSTRACT

The progressive myoclonus epilepsies (PME) are a diverse group of disorders that feature both myoclonus and seizures that worsen gradually over a variable timeframe. While each of the disorders is individually rare, they collectively make up a non-trivial portion of the complex epilepsy and myoclonus cases that are seen in tertiary care centers. The last decade has seen substantial progress in our understanding of the pathophysiology, diagnosis, prognosis, and, in select disorders, therapies of these diseases. In this scoping review, we examine English language publications from the past decade that address diagnostic, phenotypic, and therapeutic advances in all PMEs. We then highlight the major lessons that have been learned and point out avenues for future investigation that seem promising.


Subject(s)
Myoclonic Epilepsies, Progressive , Myoclonus , Humans , Myoclonic Epilepsies, Progressive/diagnosis , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/therapy
7.
Cell Rep Med ; 5(2): 101425, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38382469

ABSTRACT

Progressive myoclonus epilepsy type 7, a debilitating neurological disorder, is caused by a loss-of-function mutation in the KV3.1 channel. Exciting work by Feng et al.1 utilizes a new knockin mouse model to identify a potential therapeutic intervention.


Subject(s)
Myoclonic Epilepsies, Progressive , Animals , Mice , Myoclonic Epilepsies, Progressive/genetics , Mutation
8.
Cell Rep Med ; 5(2): 101389, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38266642

ABSTRACT

The recurrent variant KCNC1-p.Arg320His causes progressive myoclonus epilepsy (EPM) type 7, defined by progressive myoclonus, epilepsy, and ataxia, and is without effective treatment. KCNC1 encodes the voltage-gated potassium channel subunit Kv3.1, specifically expressed in high-frequency-firing neurons. Variant subunits act via loss of function; hence, EPM7 pathogenesis may involve impaired excitability of Kv3.1-expressing neurons, while enhancing Kv3 activity could represent a viable therapeutic strategy. We generate a mouse model, Kcnc1-p.Arg320His/+, which recapitulates the core features of EPM7, including progressive ataxia and seizure susceptibility. Kv3.1-expressing cerebellar granule cells and neocortical parvalbumin-positive GABAergic interneurons exhibit abnormalities consistent with Kv3 channel dysfunction. A Kv3-specific positive modulator (AUT00206) selectively enhances the firing frequency of Kv3.1-expressing neurons and improves motor function and seizure susceptibility in Kcnc1-Arg320His/+ mice. This work identifies a cellular and circuit basis of dysfunction in EPM7 and demonstrates that Kv3 positive modulators such as AUT00206 have therapeutic potential for the treatment of EPM7.


Subject(s)
Myoclonic Epilepsies, Progressive , Mice , Animals , Myoclonic Epilepsies, Progressive/genetics , Ataxia/genetics , Seizures/genetics , Neurons , Brain
9.
Cells ; 13(2)2024 01 16.
Article in English | MEDLINE | ID: mdl-38247861

ABSTRACT

Progressive myoclonic epilepsy type 1 (EPM1) is an autosomal recessive disorder, also known as Unverricht-Lundborg disease (ULD). EPM1 patients suffer from photo-sensitive seizures, stimulus-sensitive myoclonus, nocturnal myoclonic seizures, ataxia and dysarthria. In addition, cerebral ataxia and impaired GABAergic inhibition are typically present. EPM1 is caused by mutations in the Cystatin B gene (CSTB). The CSTB protein functions as an intracellular thiol protease inhibitor and inhibits Cathepsin function. It also plays a crucial role in brain development and regulates various functions in neurons beyond maintaining cellular proteostasis. These include controlling cell proliferation and differentiation, synaptic functions and protection against oxidative stress, likely through regulation of mitochondrial function. Depending on the differentiation stage and status of neurons, the protein localizes either to the cytoplasm, nucleus, lysosomes or mitochondria. Further, CSTB can also be secreted to the extracellular matrix for interneuron rearrangement and migration. In this review, we will review the various functions of CSTB in the brain and discuss the putative pathophysiological mechanism underlying EPM1.


Subject(s)
Cystatin B , Myoclonic Epilepsies, Progressive , Unverricht-Lundborg Syndrome , Humans , Ataxia , Brain/pathology , Cystatin B/genetics , Myoclonic Epilepsies, Progressive/genetics , Transcription Factors
10.
Epilepsia ; 65(3): 709-724, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38231304

ABSTRACT

OBJECTIVE: KCTD7-related progressive myoclonic epilepsy (PME) is a rare autosomal-recessive disorder. This study aimed to describe the clinical details and genetic variants in a large international cohort. METHODS: Families with molecularly confirmed diagnoses of KCTD7-related PME were identified through international collaboration. Furthermore, a systematic review was done to identify previously reported cases. Salient demographic, epilepsy, treatment, genetic testing, electroencephalographic (EEG), and imaging-related variables were collected and summarized. RESULTS: Forty-two patients (36 families) were included. The median age at first seizure was 14 months (interquartile range = 11.75-22.5). Myoclonic seizures were frequently the first seizure type noted (n = 18, 43.9%). EEG and brain magnetic resonance imaging findings were variable. Many patients exhibited delayed development with subsequent progressive regression (n = 16, 38.1%). Twenty-one cases with genetic testing available (55%) had previously reported variants in KCTD7, and 17 cases (45%) had novel variants in KCTD7 gene. Six patients died in the cohort (age range = 1.5-21 years). The systematic review identified 23 eligible studies and further identified 59 previously reported cases of KCTD7-related disorders from the literature. The phenotype for the majority of the reported cases was consistent with a PME (n = 52, 88%). Other reported phenotypes in the literature included opsoclonus myoclonus ataxia syndrome (n = 2), myoclonus dystonia (n = 2), and neuronal ceroid lipofuscinosis (n = 3). Eight published cases died over time (14%, age range = 3-18 years). SIGNIFICANCE: This study cohort and systematic review consolidated the phenotypic spectrum and natural history of KCTD7-related disorders. Early onset drug-resistant epilepsy, relentless neuroregression, and severe neurological sequalae were common. Better understanding of the natural history may help future clinical trials.


Subject(s)
Epilepsies, Myoclonic , Myoclonic Epilepsies, Progressive , Unverricht-Lundborg Syndrome , Adolescent , Child , Child, Preschool , Humans , Infant , Young Adult , Electroencephalography , Epilepsies, Myoclonic/genetics , Myoclonic Epilepsies, Progressive/genetics , Potassium Channels/genetics , Seizures
11.
Pract Neurol ; 24(1): 41-44, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37802651

ABSTRACT

A young man from Pakistan had his first-ever tonic-clonic seizure while playing cricket. Since age 12 years, he had reported involuntary jerks and tremulousness, sometimes with falls, particularly with bright lights. Family history included a brother who developed seizures with myoclonus in his mid-20s and parental consanguinity. Developmental history was normal. Examination identified cognitive impairment with action myoclonus. His clinical presentation raised suspicion of a progressive myoclonus epilepsy. MR scan of the brain showed white matter changes suggesting leucodystrophy with cortical atrophy. Electroencephalogram showed generalised epileptiform abnormalities with photoparoxysmal responses, including at low frequencies (1 Hz). Cortical hyperexcitability was confirmed with giant median somatosensory evoked potentials and long loop reflexes at rest. Multichannel electromyography showed action myoclonus with variable synchronous and asynchronous agonist and antagonist muscle activation with short-burst duration of 25-75 ms, and jerk-locked back-averaging showed premyoclonic potentials consistent with cortical myoclonus. Genetic sequencing identified a homozygous missense variant in the CLN6 gene (c.768C>G p.(Asp256Glu), confirming Kufs disease type A.


Subject(s)
Myoclonic Epilepsies, Progressive , Myoclonus , Neuronal Ceroid-Lipofuscinoses , Male , Adult , Humans , Child , Brain , Electroencephalography , Seizures , Electromyography , Membrane Proteins
12.
Eur J Med Genet ; 67: 104895, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38070824

ABSTRACT

INTRODUCTION: NGLY1-associated congenital disorder of deglycosylation (CDDG1: OMIM #615273) is a rare autosomal recessive disorder caused by a functional impairment of endoplasmic reticulum in degradation of glycoproteins. Neurocognitive dysfunctions have been documented in patients with CDDG1; however, deteriorating phenotypes of affected individuals remain elusive. CASE PRESENTATION: A Japanese boy with delayed psychomotor development showed ataxic movements from age 5 years and myoclonic seizures from age 12 years. Appetite loss, motor and cognitive decline became evident at age 12 years. Electrophysiological studies identified paroxysmal discharges on myoclonic seizure and a giant somatosensory evoked potential. Perampanel was effective for controlling myoclonic seizures. Exome sequencing revealed that the patient carried compound heterozygous variants in NGLY1, NM_018297.4: c.857G > A and c.-17_12del, which were inherited from mother and father, respectively. A literature review confirmed that myoclonic seizures were observed in 28.5% of patients with epilepsy. No other patients had progressive myoclonic epilepsy or cognitive decline in association with loss-of-function variations in NGLY1. CONCLUSION: Our data provides evidence that a group of patients with CDDG1 manifest slowly progressive myoclonic epilepsy and cognitive decline during the long-term clinical course.


Subject(s)
Congenital Disorders of Glycosylation , Epilepsies, Myoclonic , Myoclonic Epilepsies, Progressive , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Male , Humans , Child , Child, Preschool , Mutation , Myoclonic Epilepsies, Progressive/genetics , Phenotype , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/genetics , Seizures
13.
Gene Ther ; 31(5-6): 234-241, 2024 05.
Article in English | MEDLINE | ID: mdl-38135787

ABSTRACT

EPM1 is the most common form of Progressive Myoclonus Epilepsy characterized by late-childhood onset, ever-worsening and disabling myoclonus, seizures, ataxia, psychiatric disease, and shortened lifespan. EPM1 is caused by expansions of a dodecamer repeat sequence in the promoter of CSTB (cystatin B), which dramatically reduces, but does not eliminate, gene expression. The relatively late onset and consistent presence of a minimal amount of protein product makes EPM1 a favorable target for gene replacement therapy. If treated early, these children's normally developed brains could be rescued from the neurodegeneration that otherwise follows, and their cross-reactive immunological material (CRIM) positive status greatly reduces transgene related toxicity. We performed a proof-of-concept CSTB gene replacement study in Cstb knockout mice by introducing full-length human CSTB driven by the CBh promoter packaged in AAV9 and administered at postnatal days 21 and 60. Mice were sacrificed at 2 or 9 months of age, respectively. We observed significant improvements in expression levels of neuroinflammatory pathway genes and cerebellar granule cell layer apoptosis, as well as amelioration of motor impairment. The data suggest that gene replacement is a promising therapeutic modality for EPM1 and could spare affected children and families the ravages of this otherwise severe neurodegenerative disease.


Subject(s)
Cystatin B , Genetic Therapy , Mice, Knockout , Neuroinflammatory Diseases , Animals , Mice , Genetic Therapy/methods , Cystatin B/genetics , Neuroinflammatory Diseases/therapy , Neuroinflammatory Diseases/genetics , Humans , Ataxia/genetics , Ataxia/therapy , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/therapy , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/administration & dosage
14.
Stem Cell Res ; 73: 103248, 2023 12.
Article in English | MEDLINE | ID: mdl-37951142

ABSTRACT

Progressive myoclonic epilepsy type 1 (EPM1) is an autosomal recessive disorder caused by mutations in the cystatin B gene (CSTB). Affected individual's manifest stimulus-sensitive and action myoclonus and tonic-clonic epileptic seizures. In this study, we have generated iPSCs from an EPM1 patient's skin fibroblasts with Sendai virus mediated transgene delivery. The iPSCs retained the patient specific promoter region expansion mutation, expressed pluripotency markers, differentiated into all three germ layers, and presented a normal karyotype. The line can in future be used to develop an in-vitro model for EPM1 and may help in understanding disease mechanisms at cellular and molecular level.


Subject(s)
Cystatins , Induced Pluripotent Stem Cells , Myoclonic Epilepsies, Progressive , Unverricht-Lundborg Syndrome , Humans , Cystatin B , Cystatins/genetics , Cystatins/metabolism , Induced Pluripotent Stem Cells/metabolism , Unverricht-Lundborg Syndrome/genetics , Myoclonic Epilepsies, Progressive/genetics
15.
Genes (Basel) ; 14(10)2023 09 25.
Article in English | MEDLINE | ID: mdl-37895210

ABSTRACT

Biallelic variants in the Golgi SNAP receptor complex member 2 gene (GOSR2) have been reported in progressive myoclonus epilepsy with neurodegeneration. Typical clinical features include ataxia and areflexia during early childhood, followed by seizures, scoliosis, dysarthria, and myoclonus. Here, we report two novel patients from unrelated families with a GOSR2-related disorder and novel genetic and clinical findings. The first patient, a male compound heterozygous for the GOSR2 splice site variant c.336+1G>A and the novel c.364G>A,p.Glu122Lys missense variant showed global developmental delay and seizures at the age of 2 years, followed by myoclonus at the age of 8 years with partial response to clonazepam. The second patient, a female homozygous for the GOSR2 founder variant p.Gly144Trp, showed only mild fine motor developmental delay and generalized tonic-clonic seizures triggered by infections during adolescence, with seizure remission on levetiracetam. The associated movement disorder progressed atypically slowly during adolescence compared to its usual speed, from initial intention tremor and myoclonus to ataxia, hyporeflexia, dysmetria, and dystonia. These findings expand the genotype-phenotype spectrum of GOSR2-related disorders and suggest that GOSR2 should be included in the consideration of monogenetic causes of dystonia, global developmental delay, and seizures.


Subject(s)
Dystonia , Dystonic Disorders , Myoclonic Epilepsies, Progressive , Myoclonus , Adolescent , Child , Child, Preschool , Female , Humans , Male , Ataxia/genetics , Mutation , Myoclonic Epilepsies, Progressive/genetics , Qb-SNARE Proteins/genetics , Seizures
16.
Orphanet J Rare Dis ; 18(1): 263, 2023 09 02.
Article in English | MEDLINE | ID: mdl-37658439

ABSTRACT

BACKGROUND: Lafora disease (LD) is a fatal form of progressive myoclonic epilepsy caused by biallelic pathogenic variants in EPM2A or NHLRC1. With a few exceptions, the influence of genetic factors on disease progression has yet to be confirmed. We present a systematic review and meta-analysis of the known pathogenic variants to identify genotype-phenotype correlations. METHODS: We collected all reported cases with genetically-confirmed LD containing data on disease history. Pathogenic variants were classified into missense (MS) and protein-truncating (PT). Three genotype classes were defined according to the combination of the variants: MS/MS, MS/PT, and PT/PT. Time-to-event analysis was performed to evaluate survival and loss of autonomy. RESULTS: 250 cases described in 70 articles were included. The mutated gene was NHLRC1 in 56% and EPM2A in 44% of cases. 114 pathogenic variants (67 EPM2A; 47 NHLRC1) were identified. The NHLRC1 genotype PT/PT was associated with shorter survival [HR 2.88; 95% CI 1.23-6.78] and a trend of higher probability of loss of autonomy [HR 2.03, 95% CI 0.75-5.56] at the multivariable Cox regression analysis. The population carrying the homozygous p.Asp146Asn variant of NHLRC1 genotype was confirmed to have a more favourable prognosis in terms of disease duration. CONCLUSIONS: This study demonstrates the existence of prognostic genetic factors in LD, namely the genotype defined according to the functional impact of the pathogenic variants. Although the reasons why NHLRC1 genotype PT/PT is associated with a poorer prognosis have yet to be fully elucidated, it may be speculated that malin plays a pivotal role in LD pathogenesis.


Subject(s)
Lafora Disease , Myoclonic Epilepsies, Progressive , Humans , Lafora Disease/genetics , Prognosis , Tandem Mass Spectrometry , Disease Progression , Ubiquitin-Protein Ligases/genetics
17.
Epileptic Disord ; 25(5): 670-680, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37616028

ABSTRACT

Progressive Myoclonus Epilepsy (PME) is a rare epilepsy syndrome characterized by the development of progressively worsening myoclonus, ataxia, and seizures. A molecular diagnosis can now be established in approximately 80% of individuals with PME. Almost fifty genetic causes of PME have now been established, although some remain extremely rare. Herein, we provide a review of clinical phenotypes and genotypes of the more commonly encountered PMEs. Using an illustrative case example, we describe appropriate clinical investigation and therapeutic strategies to guide the management of this often relentlessly progressive and devastating epilepsy syndrome. This manuscript in the Genetic Literacy series maps to Learning Objective 1.2 of the ILAE Curriculum for Epileptology (Epileptic Disord. 2019;21:129).


Subject(s)
Myoclonic Epilepsies, Progressive , Myoclonus , Unverricht-Lundborg Syndrome , Humans , Literacy , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/diagnosis , Ataxia
18.
Neurobiol Dis ; 185: 106258, 2023 09.
Article in English | MEDLINE | ID: mdl-37573956

ABSTRACT

The progressive myoclonic epilepsies (PMEs) are a group of rare neurodegenerative diseases characterized by myoclonus, epileptic seizures, and progressive neurological deterioration with cerebellar involvement. They include storage diseases like Gaucher disease, Lafora disease, and forms of neuronal ceroid lipofuscinosis (NCL). To date, 13 NCLs have been reported (CLN1-CLN8, CLN10-CLN14), associated with mutations in different genes. These forms, which affect both children and adults, are characterized by seizures, cognitive and motor impairments, and in most cases visual loss. In NCLs, as in other PMEs, central nervous system (CNS) neurodegeneration is widespread and involves different subpopulations of neurons. One of the most affected regions is the cerebellar cortex, where motor and non-motor information is processed and transmitted to deep cerebellar nuclei through the axons of Purkinje cells (PCs). PCs, being GABAergic, have an inhibitory effect on their target neurons, and provide the only inhibitory output of the cerebellum. Degeneration of PCs has been linked to motor impairments and epileptic seizures. Seizures occur when some insult upsets the normal balance in the CNS between excitatory and inhibitory impulses, causing hyperexcitability. Here we review the role of PCs in epilepsy onset and progression following their PME-related loss. In particular, we focus on the involvement of PCs in seizure phenotype in NCLs, highlighting findings from case reports and studies of animal models in which epilepsy can be linked to PC loss.


Subject(s)
Epilepsy , Myoclonic Epilepsies, Progressive , Neuronal Ceroid-Lipofuscinoses , Animals , Neuronal Ceroid-Lipofuscinoses/genetics , Purkinje Cells , Myoclonic Epilepsies, Progressive/genetics , Seizures
19.
Brain Dev ; 45(10): 597-602, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37550174

ABSTRACT

BACKGROUND: MECP2 is a well-known causative gene for Rett syndrome but other phenotypes have also been reported. Here, we report a case of a female patient with adolescent-onset progressive myoclonus epilepsy (PME) carrying a novel truncating mutation in the MECP2 gene. CASE REPORT: The patient was a 29-year-old woman with infantile-onset intellectual disability of unspecified cause. She had demonstrated slow but steady development with moderate intellectual disability until the age of 16, when she started having epileptic seizures. Her epilepsy progressed intractably with multiple seizure types accompanied by myoclonus, tremor, and gradual regression. She is currently apathetic and requires extensive assistance in all aspects of life. After an extensive work-up for underlying diseases for PME turned out negative, whole-exome sequencing revealed a de novo 113-bp deletion and 3-bp insertion in MECP2, a variant of NM_004992.4:c.1099_1211delinsGGG, p.(His367Glyfs*32). CONCLUSIONS: The clinical presentation of this case was inconsistent with Rett syndrome, and the rapid regression in the patient's twenties was considered characteristic. Mutations of MECP2 may result in variable neurodevelopmental phenotypes and may also be considered a causative gene for adolescent-onset PME.


Subject(s)
Epilepsy , Intellectual Disability , Myoclonic Epilepsies, Progressive , Rett Syndrome , Humans , Adolescent , Female , Adult , Rett Syndrome/complications , Rett Syndrome/genetics , Intellectual Disability/genetics , Methyl-CpG-Binding Protein 2/genetics , Mutation/genetics , Epilepsy/genetics , Phenotype
20.
Neuroimage Clin ; 39: 103459, 2023.
Article in English | MEDLINE | ID: mdl-37541097

ABSTRACT

PURPOSE: Progressive myoclonic epilepsy, type 1A (EPM1, Unverricht-Lundborg disease), is a rare neurodegenerative autosomal recessive disorder characterized by stimulus-sensitive and action myoclonus and tonic-clonic epileptic seizures. Patients develop neurological symptoms, including ataxia, intention tremor, and dysarthria, over time, with relatively limited and nonspecific MRI atrophy findings. The effects of the disease on brain metabolism are largely unknown. METHOD: Eighteen EPM1 patients (9 M, 9F) underwent clinical evaluation and neuropsychological testing, which included the assessment of intellectual ability, verbal memory, and psychomotor and executive functions. Magnetic resonance spectroscopy (MRS) and imaging (MRI) were performed on a 1.5 T MRI system. 2D MRS chemical shift imaging (CSI) maps (TE = 270) were obtained from the following regions of the brain: basal ganglia, thalamus, insula, splenium, and occipital white and gray matter, and N-acetyl-aspartate (NAA)-, choline (Cho)-, and lactate (Lac)-to-creatine (Cr) ratios were analyzed. Ten healthy age-and sex-matched subjects (5M, 5F) were used as controls for MRS. RESULTS: We found significant brain metabolic changes involving lactate, NAA, and choline, which are widespread in the basal ganglia, thalamic nuclei, insula, and occipital areas of EPM1 patients. Changes, especially in the right insula, basal ganglia, and thalamus, were associated with intellectual abilities and impairment of the psychomotor and executive functions of EPM1 patients. CONCLUSION: Multiple brain metabolic alterations suggest the presence of neurodegeneration associated with EPM1 progression. The changes in metabolite ratios are associated with the neurocognitive dysfunction caused by the disease. However, the role of MRS findings in understanding pathophysiology of EPM1 warrants further studies.


Subject(s)
Myoclonic Epilepsies, Progressive , Unverricht-Lundborg Syndrome , Humans , Unverricht-Lundborg Syndrome/metabolism , Proton Magnetic Resonance Spectroscopy , Brain , Myoclonic Epilepsies, Progressive/metabolism , Magnetic Resonance Spectroscopy , Magnetic Resonance Imaging , Cognition , Metabolome , Choline/metabolism , Aspartic Acid , Creatine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL