27
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Similar early characteristics but variable neurological outcome of patients with a de novo mutation of KCNQ2

      research-article
      1 , 2 , , 3 , 1 , 2 , 4 , 5 , 6 , 7 , 8 , 2 , 9 , 10 , 6 , 9 , 11 , 12 , 13 , 13 , 14 , 4 , 5 , 7 , 5 , 6 , 6 , 6 , 5 , 6 , 15 , 4 , 4 , 16 , 17 , 18 , 1 , 2 , 1 , 8 , 19 , 3 , 1 , 19
      Orphanet Journal of Rare Diseases
      BioMed Central
      Epilepsy, Genetics, KCNQ2, Encephalopathy

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Early onset epileptic encephalopathies (EOEEs) are dramatic heterogeneous conditions in which aetiology, seizures and/or interictal EEG have a negative impact on neurological development. Several genes have been associated with EOEE and a molecular diagnosis workup is challenging since similar phenotypes are associated with mutations in different genes and since mutations in one given gene can be associated with very different phenotypes. Recently, de novo mutations in KCNQ2, have been found in about 10% of EOEE patients. Our objective was to confirm that KCNQ2 was an important gene to include in the diagnosis workup of EOEEs and to fully describe the clinical and EEG features of mutated patients.

          Methods

          We have screened KCNQ2 in a cohort of 71 patients with an EOEE, without any brain structural abnormality. To be included in the cohort, patient’s epilepsy should begin before three months of age and be associated with abnormal interictal EEG and neurological impairment. Brain MRI should not show any structural abnormality that could account for the epilepsy.

          Results

          Out of those 71 patients, 16 had a de novo mutation in KCNQ2 (23%). Interestingly, in the majority of the cases, the initial epileptic features of these patients were comparable to those previously described in the case of benign familial neonatal epilepsy (BFNE) also caused by KCNQ2 mutations. However, in contrast to BFNE, the interictal background EEG was altered and displayed multifocal spikes or a suppression-burst pattern. The ongoing epilepsy and development were highly variable but overall severe: 15/16 had obvious cognitive impairment, half of the patients became seizure-free, 5/16 could walk before the age of 3 and only 2/16 patient acquired the ability to speak.

          Conclusion

          This study confirms that KCNQ2 is frequently mutated de novo in neonatal onset epileptic encephalopathy. We show here that despite a relatively stereotyped beginning of the condition, the neurological and epileptic evolution is variable.

          Related collections

          Most cited references6

          • Record: found
          • Abstract: found
          • Article: not found

          A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns.

          Idiopathic generalized epilepsies account for about 40% of epilepsy up to age 40 and commonly have a genetic basis. One type is benign familial neonatal convulsions (BFNC), a dominantly inherited disorder of newborns. We have identified a sub-microscopic deletion of chromosome 20q13.3 that co-segregates with seizures in a BFNC family. Characterization of cDNAs spanning the deleted region identified one encoding a novel voltage-gated potassium channel, KCNQ2, which belongs to a new KQT-like class of potassium channels. Five other BFNC probands were shown to have KCNQ2 mutations, including two transmembrane missense mutations, two frameshifts and one splice-site mutation. This finding in BFNC provides additional evidence that defects in potassium channels are involved in the mammalian epilepsy phenotype.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family.

            Epileptic disorders affect about 20-40 million people worldwide, and 40% of these are idiopathic generalized epilepsies (IGEs; ref. 1). Most of the IGEs that are inherited are complex, multigenic diseases. To address basic mechanisms for epilepsies, we have focused on one well-defined class of IGEs with an autosomal-dominant mode of inheritance: the benign familial neonatal convulsions (BFNC; refs 2,3). Genetic heterogeneity of BFNC has been observed. Two loci, EBN1 and EBN2, have been mapped by linkage analysis to chromosome 20q13 (refs 5,6) and chromosome 8q24 (refs 7,8), respectively. By positional cloning, we recently identified the gene for EBN1 as KCNQ2 (ref. 9). This gene, a voltage-gated potassium channel, based on homology, is a member of the KQT-like family. Here we describe an additional member, KCNQ3. We mapped this new gene to chromosome 8, between markers D8S256 and D8S284 on a radiation hybrid map. We screened KCNQ3 for mutations in the large BFNC family previously linked to chromosome 8q24 in the same marker interval. We found a missense mutation in the critical pore region in perfect co-segregation with the BFNC phenotype. The same conserved amino acid is also mutated in KVLQT1 (KCNQ1) in an LQT patient. KCNQ2, KCNQ3 and undiscovered genes of the same family of K+ channels are strong candidates for other IGEs.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genotype-phenotype correlations in neonatal epilepsies caused by mutations in the voltage sensor of K(v)7.2 potassium channel subunits.

              Mutations in the K(V)7.2 gene encoding for voltage-dependent K(+) channel subunits cause neonatal epilepsies with wide phenotypic heterogeneity. Two mutations affecting the same positively charged residue in the S4 domain of K(V)7.2 have been found in children affected with benign familial neonatal seizures (R213W mutation) or with neonatal epileptic encephalopathy with severe pharmacoresistant seizures and neurocognitive delay, suppression-burst pattern at EEG, and distinct neuroradiological features (R213Q mutation). To examine the molecular basis for this strikingly different phenotype, we studied the functional characteristics of mutant channels by using electrophysiological techniques, computational modeling, and homology modeling. Functional studies revealed that, in homomeric or heteromeric configuration with K(V)7.2 and/or K(V)7.3 subunits, both mutations markedly destabilized the open state, causing a dramatic decrease in channel voltage sensitivity. These functional changes were (i) more pronounced for channels incorporating R213Q- than R213W-carrying K(V)7.2 subunits; (ii) proportional to the number of mutant subunits incorporated; and (iii) fully restored by the neuronal K(v)7 activator retigabine. Homology modeling confirmed a critical role for the R213 residue in stabilizing the activated voltage sensor configuration. Modeling experiments in CA1 hippocampal pyramidal cells revealed that both mutations increased cell firing frequency, with the R213Q mutation prompting more dramatic functional changes compared with the R213W mutation. These results suggest that the clinical disease severity may be related to the extent of the mutation-induced functional K(+) channel impairment, and set the preclinical basis for the potential use of K(v)7 openers as a targeted anticonvulsant therapy to improve developmental outcome in neonates with K(V)7.2 encephalopathy.
                Bookmark

                Author and article information

                Journal
                Orphanet J Rare Dis
                Orphanet J Rare Dis
                Orphanet Journal of Rare Diseases
                BioMed Central
                1750-1172
                2013
                22 May 2013
                : 8
                : 80
                Affiliations
                [1 ]INSERM, UMR_S 910 Faculté de médecine, Boulevard jean MOULIN F13005, Marseille, France
                [2 ]APHM, Service de neurologie pédiatrique, CHU Timone, Marseille, France
                [3 ]Hospices civils de Lyon. Laboratoire de génétique, Hôpital Edouard Herriot. Bron, Lyon, France
                [4 ]APHP, Unité Fonctionnelle de Génétique Médicale, Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
                [5 ]Centre de Référence des Déficiences Intellectuelles de Causes Rares, Paris, France
                [6 ]APHP. Service de Neuropédiatrie, Hôpital Armand Trousseau, Paris, France
                [7 ]APHP. Service de neuropédiatrie, Hopital Robert Debré, Paris, France
                [8 ]APHM. Département de Génétique Médicale et Biologie Cellulaire CHU Timone, Marseille, France
                [9 ]CHU Montpellier. Service de neuropédiatrie, Montpellier, France
                [10 ]INSERM U1051, INM Montpellier, Montpellier, France
                [11 ]APHP. Service de neurophysiologie clinique Hôpital Necker, Paris, France
                [12 ]CHU Besancon. Service de neuropédiatrie, Besancon, France
                [13 ]Hospices civils de Lyon, Service de neuropédiatrie. HFME. Bron, Lyon, France
                [14 ]CHU de Tours. Service de neuropédiatrie, Beranger, France
                [15 ]APHP. Groupe hospitalier Pitié Salpétrière. Service de neurologie, Paris, France
                [16 ]CHU de Nantes. Service de pédiatrie, Nantes, France
                [17 ]CHU de Nantes. Service de génétique médicale, Nantes, France
                [18 ]CHU de Grenoble. Service d’électrophysiologie clinique, Grenoble, France
                [19 ]Aix Marseille Université, Faculté de Médecine, Marseille, France
                Article
                1750-1172-8-80
                10.1186/1750-1172-8-80
                3670812
                23692823
                5c474992-894d-4095-bfca-8d091f95e1c9
                Copyright ©2013 Milh et al.; licensee BioMed Central Ltd.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 8 March 2013
                : 15 May 2013
                Categories
                Research

                Infectious disease & Microbiology
                epilepsy,genetics,kcnq2,encephalopathy
                Infectious disease & Microbiology
                epilepsy, genetics, kcnq2, encephalopathy

                Comments

                Comment on this article