Journal of Pediatric Neurology 2023; 21(03): 203-211
DOI: 10.1055/s-0041-1727099
Review Article

The Spectrum of KCNQ2- and KCNQ3-Related Epilepsy

Anna Portale*
1   Unit of Pediatrics, Avola Hospital, Siracusa, Italy
,
Mattia Comella*
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Giulia Salomone
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Alessandra Di Nora
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Lidia Marino
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Roberta Leonardi
3   Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
,
Andrea D. Praticò
3   Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
,
Raffaele Falsaperla
4   Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
5   Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
› Author Affiliations

Abstract

KCNQ genes encode for a family of six transmembrane domains, single pore-loop, and K+ channel α-subunits that have a wide range of physiological correlates. In the brain, KCNQ2 and KCNQ3 heteromultimers are thought to underlie the M-current which is essential in raising the threshold for firing an action potential; mutations in these genes may cause several types of infantile epilepsies. KCNQ2-related disorders represent a continuum of overlapping neonatal epileptic phenotypes that range from KCNQ2 benign familial neonatal epilepsy (BFNE), a seizure disorder that occur in children who typically have a normal psychomotor development and are inherited as an autosomal dominant trait, to KCNQ2 early-onset epileptic encephalopathy (EOEE) as the result of a de novo pathogenic variant. KCNQ3-related disorders are rarer and include BFNE, benign familial infantile epilepsy and KCNQ3-related epileptic encephalopathy with intellectual disability with or without seizures and/or cortical visual impairment. For both KCNQ2- and KCNQ3-related disorders, it is possible to use several drugs for different classes of mutations (i.e., gain of function vs. loss of function), and usually their effects vary in relation to the clinical presentation and the phenotype of the patient. However, KCNQ2-EOEE patients have a worse response to treatment than KCNQ2-BFNE patients and usually become drug resistant with multiple daily seizures.

* Both the authors have equally contributed to the present paper.




Publication History

Received: 13 September 2020

Accepted: 12 January 2021

Article published online:
13 April 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Miceli F, Soldovieri MV, Ambrosino P. et al. Early-onset epileptic encephalopathy caused by gain-of-function mutations in the voltage sensor of Kv7.2 and Kv7.3 potassium channel subunits. J Neurosci 2015; 35 (09) 3782-3793
  • 2 Brown DA, Adams PR. Muscarinic suppression of a novel voltage-sensitive K+ current in a vertebrate neurone. Nature 1980; 283 (5748): 673-676
  • 3 Coppola G, Castaldo P, Miraglia del Giudice E. et al. A novel KCNQ2 K+ channel mutation in benign neonatal convulsions and centrotemporal spikes. Neurology 2003; 61 (01) 131-134
  • 4 Kothur K, Holman K, Farnsworth E. et al. Diagnostic yield of targeted massively parallel sequencing in children with epileptic encephalopathy. Seizure 2018; 59: 132-140
  • 5 Ambrosino P, Freri E, Castellotti B. et al. Kv7.3 compound heterozygous variants in early onset encephalopathy reveal additive contribution of C-terminal residues to PIP2-dependent K+ channel gating. Mol Neurobiol 2018; 55 (08) 7009-7024
  • 6 Maljevic S, Vejzovic S, Bernhard MK. et al. Novel KCNQ3 mutation in a large family with benign familial neonatal epilepsy: a rare cause of neonatal seizures. Mol Syndromol 2016; 7 (04) 189-196
  • 7 Pavone P, Falsaperla R, Ruggieri M. et al. Clinical course of N-methyl-D-aspartate receptor encephalitis and the effectiveness of cyclophosphamide treatment. J Pediatr Neurol 2017; 15: 84-49
  • 8 Rett A, Teubel R. Neugeborenen Krampfe im Rahmen einer epileptisch belasten Familie. Wien Klin Wochenschr 1964; 76: 609-613
  • 9 Praticò AD, Falsaperla R, Ruggieri M, Corsello G, Pavone P. Prognostic challenges of SCN1A genetic mutations: report on two children with mild features. J Pediatr Neurol 2016; 14: 82-88
  • 10 Falsaperla R, Perciavalle V, Pavone P. et al. Unilateral eye blinking arising from the ictal ipsilateral occipital area. Clin EEG Neurosci 2016; 47 (03) 243-246
  • 11 Incorpora G, Pavone P, Castellano-Chiodo D, Praticò AD, Ruggieri M, Pavone L. Gelastic seizures due to hypothalamic hamartoma: rapid resolution after endoscopic tumor disconnection. Neurocase 2013; 19 (05) 458-461
  • 12 Pavone P, Falsaperla R, Ruggieri M, Praticò AD, Pavone L. West syndrome treatment: new roads for an old syndrome. Front Neurol 2013; 4: 113
  • 13 Pavone P, Praticò AD, Falsaperla R. et al. Congenital generalized hypertrichosis: the skin as a clue to complex malformation syndromes. Ital J Pediatr 2015; 41: 55
  • 14 Al Yazidi G, Shevell MI, Srour M. Two novel KCNQ2 mutations in 2 families with benign familial neonatal convulsions. Child Neurol Open 2017; 4: 2329048  × 17691396
  • 15 Lee IC, Yang JJ, Li SYA. A KCNQ2 E515D mutation associated with benign familial neonatal seizures and continuous spike and waves during slow-wave sleep syndrome in Taiwan. J Formos Med Assoc 2017; 116 (09) 711-719
  • 16 Vilan A, Mendes Ribeiro J, Striano P. et al. A distinctive ictal amplitude-integrated electroencephalography pattern in newborns with neonatal epilepsy associated with KCNQ2 mutations. Neonatology 2017; 112 (04) 387-393
  • 17 Salafia S, Praticò AD, Pizzo E, Greco F, Di Bella D. Hemiconvulsion-hemiplegia-epilepsy syndrome. Magnetic resonance findings in a 3-year-old boy. Neurol Neurochir Pol 2013; 47 (06) 584-589
  • 18 Barbagallo M, Ruggieri M, Incorpora G. et al. Infantile spasms in the setting of Sturge-Weber syndrome. Childs Nerv Syst 2009; 25 (01) 111-118
  • 19 Ronen GM, Rosales TO, Connolly M, Anderson VE, Leppert M. Seizure characteristics in chromosome 20 benign familial neonatal convulsions. Neurology 1993; 43 (07) 1355-1360
  • 20 Ishii A, Miyajima T, Kurahashi H. et al. KCNQ2 abnormality in BECTS: benign childhood epilepsy with centrotemporal spikes following benign neonatal seizures resulting from a mutation of KCNQ2. Epilepsy Res 2012; 102 (1-2): 122-125
  • 21 Caltabiano R, Magro G, Polizzi A. et al. A mosaic pattern of INI1/SMARCB1 protein expression distinguishes Schwannomatosis and NF2-associated peripheral schwannomas from solitary peripheral schwannomas and NF2-associated vestibular schwannomas. Childs Nerv Syst 2017; 33 (06) 933-940
  • 22 Pavone P, Briuglia S, Falsaperla R. et al. Wide spectrum of congenital anomalies including choanal atresia, malformed extremities, and brain and spinal malformations in a girl with a de novo 5.6-Mb deletion of 13q12.11-13q12.13. Am J Med Genet A 2014; 164A (07) 1734-1743
  • 23 Pavone V, Signorelli SS, Praticò AD. et al. Total hemi-overgrowth in pigmentary mosaicism of the (hypomelanosis of) ITO type: eight case reports. Medicine (Baltimore) 2016; 95 (10) e2705
  • 24 Pavone P, Praticò AD, Pavone V. et al. Ataxia in children: early recognition and clinical evaluation. Ital J Pediatr 2017; 43 (01) 6
  • 25 Praticò AD, Pavone P, Scuderi MG. et al. Symptomatic hypocalcemia in an epileptic child treated with valproic acid plus lamotrigine: a case report. Cases J 2009; 2: 7394
  • 26 Pavone P, Rizzo R, Conti I. et al. Primary headaches in children: clinical findings on the association with other conditions. Int J Immunopathol Pharmacol 2012; 25 (04) 1083-1091
  • 27 Gomis-Pérez C, Urrutia J, Marcé-Grau A. et al. Homomeric Kv7.2 current suppression is a common feature in KCNQ2 epileptic encephalopathy. Epilepsia 2019; 60 (01) 139-148
  • 28 Pavone P, Praticò AD, Gentile G. et al. A neurocutaneous phenotype with paired hypo- and hyperpigmented macules, microcephaly and stunted growth as prominent features. Eur J Med Genet 2016; 59 (05) 283-289
  • 29 Ruggieri M, Praticò AD, Caltabiano R, Polizzi A. Early history of the different forms of neurofibromatosis from ancient Egypt to the British Empire and beyond: First descriptions, medical curiosities, misconceptions, landmarks, and the persons behind the syndromes. Am J Med Genet A 2018; 176 (03) 515-550
  • 30 Falsaperla R, Praticò AD, Ruggieri M. et al. Congenital muscular dystrophy: from muscle to brain. Ital J Pediatr 2016; 42 (01) 78
  • 31 Ranieri C, Di Tommaso S, Loconte DC. et al. In vitro efficacy of ARQ 092, an allosteric AKT inhibitor, on primary fibroblast cells derived from patients with PIK3CA-related overgrowth spectrum (PROS). Neurogenetics 2018; 19 (02) 77-91
  • 32 Ruggieri M, Praticò AD, Caltabiano R, Polizzi A. Rediagnosing one of Smith's patients (John McCann) with “neuromas tumours” (1849). Neurol Sci 2017; 38 (03) 493-499
  • 33 Kasinathan A, Sankhyan N, Singhi P. KCNQ2 epileptic encephalopathy in early infancy. Indian J Pediatr 2017; 84 (11) 877-878
  • 34 Reid ES, Williams H, Stabej PleQ. et al. Seizures due to a KCNQ2 mutation: treatment with vitamin B6. JIMD Rep 2016; 27: 79-84
  • 35 Reif PS, Tsai MH, Helbig I, Rosenow F, Klein KM. Precision medicine in genetic epilepsies: break of dawn?. Expert Rev Neurother 2017; 17 (04) 381-392
  • 36 Cornet MC, Sands TT, Cilio MR. Neonatal epilepsies: clinical management. Semin Fetal Neonatal Med 2018; 23 (03) 204-212
  • 37 Mulkey SB, Ben-Zeev B, Nicolai J. et al. Neonatal nonepileptic myoclonus is a prominent clinical feature of KCNQ2 gain-of-function variants R201C and R201H. Epilepsia 2017; 58 (03) 436-445
  • 38 Millichap JJ, Miceli F, De Maria M. et al. Infantile spasms and encephalopathy without preceding neonatal seizures caused by KCNQ2 R198Q, a gain-of-function variant. Epilepsia 2017; 58 (01) e10-e15
  • 39 Kojima K, Shirai K, Kobayashi M. et al. A patient with early myoclonic encephalopathy (EME) with a de novo KCNQ2 mutation. Brain Dev 2018; 40 (01) 69-73
  • 40 Devaux J, Abidi A, Roubertie A. et al. A Kv7.2 mutation associated with early onset epileptic encephalopathy with suppression-burst enhances Kv7/M channel activity. Epilepsia 2016; 57 (05) e87-e93
  • 41 Schubert-Bast S, Hofstetter P, Fischer D, Schloesser R, Ramantani G, Kieslich M. Sodium channel blockers in KCNQ2-encephalopathy: lacosamide as a new treatment option. Seizure 2017; 51: 171-173
  • 42 Duan H, Peng J, Kessi M, Yin F. De novo KCNQ2 mutation in one case of epilepsy of infancy with migrating focal seizures that evolved to infantile spasms. Child Neurol Open 2018; 5: 2329048  × 18767738
  • 43 Olson HE, Kelly M, LaCoursiere CM. et al. Genetics and genotype-phenotype correlations in early onset epileptic encephalopathy with burst suppression. Ann Neurol 2017; 81 (03) 419-429
  • 44 Ruggieri M, Praticò AD, Scuderi A, Sorge G, Polizzi A. The multiple faces of artwork diagnoses. Lancet Neurol 2017; 16 (06) 417-418
  • 45 Ruggieri M, Praticò AD, Serra A. et al. Early history of neurofibromatosis type 2 and related forms: earliest descriptions of acoustic neuromas, medical curiosities, misconceptions, landmarks and the pioneers behind the eponyms. Childs Nerv Syst 2017; 33 (04) 549-560
  • 46 Spagnoli C, Salerno GG, Iodice A, Frattini D, Pisani F, Fusco C. KCNQ2 encephalopathy: a case due to a de novo deletion. Brain Dev 2018; 40 (01) 65-68
  • 47 Hewson S, Puka K, Mercimek-Mahmutoglu S. Variable expressivity of a likely pathogenic variant in KCNQ2 in a three-generation pedigree presenting with intellectual disability with childhood onset seizures. Am J Med Genet A 2017; 173 (08) 2226-2230
  • 48 Knuutinen O, Kousi M, Suo-Palosaari M. et al. Neonatal alexander disease: novel GFAP mutation and comparison to previously published cases. Neuropediatrics 2018; 49 (04) 256-261
  • 49 Zhou P, He N, Zhang JW. et al. Novel mutations and phenotypes of epilepsy-associated genes in epileptic encephalopathies. Genes Brain Behav 2018; 17 (08) e12456
  • 50 Allen NM, Mannion M, Conroy J. et al. The variable phenotypes of KCNQ-related epilepsy. Epilepsia 2014; 55 (09) e99-e105
  • 51 Miao P, Feng J, Guo Y. et al. Genotype and phenotype analysis using an epilepsy-associated gene panel in Chinese pediatric epilepsy patients. Clin Genet 2018; 94 (06) 512-520
  • 52 Orhan G, Bock M, Schepers D. et al. Dominant-negative effects of KCNQ2 mutations are associated with epileptic encephalopathy. Ann Neurol 2014; 75 (03) 382-394
  • 53 Miceli F, Soldovieri MV, Joshi N, Weckhuysen S, Cooper EC, Taglialatela M. KCNQ3-related disorders. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A. eds. GeneReviews. Seattle WA: University of Washington, Seattle; 1993-2021
  • 54 Milh M, Boutry-Kryza N, Sutera-Sardo J. et al. Similar early characteristics but variable neurological outcome of patients with a de novo mutation of KCNQ2 . Orphanet J Rare Dis 2013; 8: 80
  • 55 Borgatti R, Zucca C, Cavallini A. et al. A novel mutation in KCNQ2 associated with BFNC, drug resistant epilepsy, and mental retardation. Neurology 2004; 63 (01) 57-65
  • 56 Grinton BE, Heron SE, Pelekanos JT. et al. Familial neonatal seizures in 36 families: Clinical and genetic features correlate with outcome. Epilepsia 2015; 56 (07) 1071-1080
  • 57 Soldovieri MV, Boutry-Kryza N, Milh M. et al. Novel KCNQ2 and KCNQ3 mutations in a large cohort of families with benign neonatal epilepsy: first evidence for an altered channel regulation by syntaxin-1A. Hum Mutat 2014; 35 (03) 356-367
  • 58 Zeng Q, Yang X, Zhang J. et al. Genetic analysis of benign familial epilepsies in the first year of life in a Chinese cohort. J Hum Genet 2018; 63 (01) 9-18
  • 59 Zhang Q, Li J, Zhao Y, Bao X, Wei L, Wang J. Gene mutation analysis of 175 Chinese patients with early-onset epileptic encephalopathy. Clin Genet 2017; 91 (05) 717-724
  • 60 Ortega-Moreno L, Giráldez BG, Soto-Insuga V. et al; Grupo Español de Genética de las Epilepsias de la Infancia (GEGEI). Molecular diagnosis of patients with epilepsy and developmental delay using a customized panel of epilepsy genes. PLoS One 2017; 12 (11) e0188978
  • 61 Arafat A, Jing P, Ma Y. et al. Unexplained early infantile epileptic encephalopathy in han chinese children: next-generation sequencing and phenotype enriching. Sci Rep 2017; 7: 46227
  • 62 Liu J, Tong L, Song S. et al. Novel and de novo mutations in pediatric refractory epilepsy. Mol Brain 2018; 11 (01) 48
  • 63 Ruggieri M, Milone P, Pavone P. et al. Nevus vascularis mixtus (cutaneous vascular twin nevi) associated with intracranial vascular malformation of the Dyke-Davidoff-Masson type in two patients. Am J Med Genet A 2012; 158A (11) 2870-2880
  • 64 Ruggieri M, Iannetti P, Pavone L. Delineation of a newly recognized neurocutaneous malformation syndrome with “cutis tricolor”. Am J Med Genet A 2003; 120A (01) 110-116
  • 65 Pavone P, Praticò AD, Vitaliti G. et al. Hydranencephaly: cerebral spinal fluid instead of cerebral mantles. Ital J Pediatr 2014; 40: 79
  • 66 Ruggieri M, Praticò AD, Serra A. et al. Childhood neurofibromatosis type 2 (NF2) and related disorders: from bench to bedside and biologically targeted therapies. Acta Otorhinolaryngol Ital 2016; 36 (05) 345-367
  • 67 Polizzi A, Pavone P, Ciancio E, La Rosa C, Sorge G, Ruggieri M. Hypertrichosis cubiti (hairy elbow syndrome): a clue to a malformation syndrome. J Pediatr Endocrinol Metab 2005; 18 (10) 1019-1025
  • 68 Polizzi A, Pavone P, Parano E, Incorpora G, Ruggieri M. Lack of progression of brain atrophy in Aicardi-Goutières syndrome. Pediatr Neurol 2001; 24 (04) 300-302
  • 69 Polizzi A, Coghill S, McShane MA, Squier W. Acute ataxia complicating Langherans cell histiocytosis. Arch Dis Child 2002; 86 (02) 130-131
  • 70 Lauritano A, Moutton S, Longobardi E. et al. A novel homozygous KCNQ3 loss-of-function variant causes non-syndromic intellectual disability and neonatal-onset pharmacodependent epilepsy. Epilepsia Open 2019; 4 (03) 464-475
  • 71 Sands TT, Miceli F, Lesca G. et al. Autism and developmental disability caused by KCNQ3 gain-of-function variants. Ann Neurol 2019; 86 (02) 181-192
  • 72 Kuersten M, Tacke M, Gerstl L, Hoelz H, Stülpnagel CV, Borggraefe I. Antiepileptic therapy approaches in KCNQ2 related epilepsy: a systematic review. Eur J Med Genet 2020; 63 (01) 103628
  • 73 Tulloch JK, Carr RR, Ensom MH. A systematic review of the pharmacokinetics of antiepileptic drugs in neonates with refractory seizures. J Pediatr Pharmacol Ther 2012; 17 (01) 31-44
  • 74 Sands TT, Balestri M, Bellini G. et al. Rapid and safe response to low-dose carbamazepine in neonatal epilepsy. Epilepsia 2016; 57 (12) 2019-2030
  • 75 Kato M, Yamagata T, Kubota M. et al. Clinical spectrum of early onset epileptic encephalopathies caused by KCNQ2 mutation. Epilepsia 2013; 54 (07) 1282-1287
  • 76 Brodie MJ. Sodium channel blockers in the treatment of epilepsy. CNS Drugs 2017; 31 (07) 527-534
  • 77 Ko A, Youn SE, Kim SH. et al. Targeted gene panel and genotype-phenotype correlation in children with developmental and epileptic encephalopathy. Epilepsy Res 2018; 141: 48-55
  • 78 Falsaperla R, D'Angelo G, Praticò AD. et al. Ketogenic diet for infants with epilepsy: a literature review. Epilepsy Behav 2020; 112: 107361
  • 79 Serino D, Specchio N, Pontrelli G, Vigevano F, Fusco L. Video/EEG findings in a KCNQ2 epileptic encephalopathy: a case report and revision of literature data. Epileptic Disord 2013; 15 (02) 158-165
  • 80 Pratico AD, Longo L, Mansueto S. et al. Off-label use of drugs and adverse drug reactions in pediatric units: a prospective, multicenter study. Curr Drug Saf 2018; 13 (03) 200-207
  • 81 Pavone P, Praticò AD, Ruggieri M. et al. Acquired peripheral neuropathy: a report on 20 children. Int J Immunopathol Pharmacol 2012; 25 (02) 513-517
  • 82 Pratico AD, Ruggieri M, Falsaperla R, Pavone P. A probable topiramate-induced limbs paraesthesia and rigid fingers flexion. Curr Drug Saf 2018; 13 (02) 131-136
  • 83 Ruggieri M, Polizzi A, Marceca GP, Catanzaro S, Praticò AD, Di Rocco C. Introduction to phacomatoses (neurocutaneous disorders) in childhood. Childs Nerv Syst 2020; 36 (10) 2229-2268
  • 84 Porter RJ, Burdette DE, Gil-Nagel A. et al. Retigabine as adjunctive therapy in adults with partial-onset seizures: integrated analysis of three pivotal controlled trials. Epilepsy Res 2012; 101 (1-2): 103-112
  • 85 Weckhuysen S, Ivanovic V, Hendrickx R. et al; KCNQ2 Study Group. Extending the KCNQ2 encephalopathy spectrum: clinical and neuroimaging findings in 17 patients. Neurology 2013; 81 (19) 1697-1703
  • 86 Singh NA, Westenskow P, Charlier C. et al; BFNC Physician Consortium. KCNQ2 and KCNQ3 potassium channel genes in benign familial neonatal convulsions: expansion of the functional and mutation spectrum. Brain 2003; 126 (Pt 12): 2726-2737