Skip to main content

ORIGINAL RESEARCH article

Front. Neurol., 07 June 2021
Sec. Pediatric Neurology

Gene and Phenotype Expansion of Unexplained Early Infantile Epileptic Encephalopathy

\nXianyu Liu&#x;Xianyu Liu1Qiyang Shen&#x;Qiyang Shen2Guo ZhengGuo Zheng1Hu GuoHu Guo1Xiaopeng LuXiaopeng Lu1Xiaoyu WangXiaoyu Wang1Xiao YangXiao Yang1Zixuan CaoZixuan Cao1Jing Chen
Jing Chen1*
  • 1Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, China
  • 2Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China

Objective: The genetic aetiology of epileptic encephalopathy (EE) is growing rapidly based on next generation sequencing (NGS) results. In this single-centre study, we aimed to investigate a cohort of Chinese children with early infantile epileptic encephalopathy (EIEE).

Methods: NGS was performed on 50 children with unexplained EIEE. The clinical profiles of children with pathogenic variants were characterised and analysed in detail. Conservation analysis and homology modelling were performed to predict the impact of STXBP1 variant on the protein structure.

Results: Pathogenic variants were identified in 17 (34%) of 50 children. Sixteen variants including STXBP1 (n = 2), CDKL5 (n = 2), PAFAH1B1, SCN1A (n = 9), SCN2A, and KCNQ2 were de novo, and one (PIGN) was a compound heterozygous variant. The phenotypes of the identified genes were broadened. PIGN phenotypic spectrum may include EIEE. The STXBP1 variants were predicted to affect protein stability.

Significance: NGS is a useful diagnostic tool for EIEE and contributes to expanding the EIEE-associated genotypes. Early diagnosis may lead to precise therapeutic interventions and can improve the developmental outcome.

Introduction

Epileptic encephalopathy (EE) describes a group of severe intractable seizures and neurological disorders in which epileptic activity itself may contribute to severe neurologic and cognitive impairment, beyond the damage that would be expected from the underlying pathology alone, and these effects can worsen over time (1). Early infantile epileptic encephalopathies (EIEEs) occur during infancy and comprise a spectrum of syndromes such as Ohtahara syndrome (OS), West syndrome (WS), epilepsy of infancy with migrating focal seizures, and Dravet syndrome (DS) (2).

In addition to common metabolic and structural abnormalities, genetics play an important role in the aetiology of EIEEs (3). EIEEs exhibit overlap and pleiotropy, which makes it challenging to establish genotype-phenotype correlations. The recent advent and improvement of next generation sequencing (NGS) have provided a significant advancement in the diagnosis of epileptic patients (4). In this study, NGS was performed in a cohort of Chinese children with unexplained EIEEs. We analysed genotype-phenotype correlations and elucidated therapeutic effects and adjustments.

Methods

Patients

A total of 50 patients with EIEEs without specific aetiologies in the department of neurology, at the Children's Hospital of Nanjing Medical University in China were recruited between 2015 and 2018. Other aetiologies such as definite perinatal brain injury, infection of the central nervous system, typical inherited metabolic disorders and abnormal detection of chromosomal microarrays were excluded in these patients. Clinical data including age of seizure onset, type and frequency of seizures, video electroencephalography (VEEG) recording, brain magnetic resonance imaging (MRI), response to antiseizure medications (ASMs), and comorbidities were collected. Electro-clinical syndromes were defined and classified according to the International League Against Epilepsy (ILAE) (5). All patients were followed up with by telephone or outpatient visits every 3 months until October 2019. The study protocol was approved by the hospital ethics committee and all parents provided informed consent for their children's participation.

Next Generation Sequencing

Genomic DNA was extracted from peripheral leukocytes from patients and their parents. Whole-exome sequencing (WES) or epilepsy-associated gene panel analyses with the Illumina NextSeq 500 platform (Illumina, San Diego, CA, USA) were performed at accredited commercial labs. The epilepsy-associated gene panel contained more than 500 genes and continued updating over time. Copy number variations (CNVs) were analysed by copy number variation sequencing (CNV-Seq) based on the NGS platform. Sanger sequencing was used to validate variants and to analyse segregation in pedigrees. Variants were analysed and annotated according to Exome Aggregation Consortium (ExAC), 1,000 Genomes, Clinvar, dbSNP, Online Mendelian Inheritance in Man (OMIM), and the Human Gene Mutation Database (HGMD). Effects of variants on function were predicted by SIFT, PolyPhen-2, and MutationTaster. Variant pathogenicity was evaluated by the American College of Medical Genetics and Genomics (ACMG) scoring system, together with clinical presentation (6).

Homology Modelling

Multiple sequences including mutated regions from different species were aligned using the Clustal Omega website, to determine whether the variants are located in an evolutionarily conserved domain. We selected the protein crystal structures of 4JEH and 4JEU from Protein Data Bank (PDB; www.rcsb.org) as templates for STXBP1. Molecular structure modelling of mutant and wild-type proteins was achieved using the SWISS-MODEL online server (7) and PyMOL (www.pymol.org).

Results

Pathogenic variants of 17 genes were identified in 50 patients (34%) and pathogenic CNVs were not detected. Sixteen variants including STXBP1 (n = 2), CDKL5 (n = 2), PAFAH1B1, SCN1A (n = 9), SCN2A, and KCNQ2 were de novo heterozygous variants, and one (PIGN) was a compound heterozygous variant; 11 out of 17 were novel variants. Five patients underwent WES and 12 patients had epilepsy-associated gene panel testing. The male to female ratio was 9:8. The mean seizure onset was 4.5 months (range: 8 days to 11 months). All variants are predicted to be “Pathogenic” according to ACMG standards. The variants and clinical features of the patients are listed in Table 1. Multiple sequence alignments and comparative protein structures of two STXBP1 missense variants are shown in Supplementary Figures 1, 2.

TABLE 1
www.frontiersin.org

Table 1. Summary of gene variants and clinical features of patients.

STXBP1 missense variants were identified in a female and a male patient. One patient carrying c.1439C>T was diagnosed with OS and had experienced early-onset seizures since 8 days of age. The patient exhibited tonic spasms, occurring in clusters, during sleep or awakening. VEEG at seizure onset recorded burst-suppression pattern; brain MRI was unremarkable. Postnatal development was severely delayed. The patient did not respond to sodium valproate (VPA), levetiracetam (LEV), topiramate (TPM), vitamin B6 (VB6), and phenobarbital (PB). Interestingly, worsening of the seizures during the night was observed when VB6 was tapered, which improved when the initial VB6 dose was re-established. A ketogenic diet (KD) with ketone concentrations of ~3.0 mmol/L resulted in the patient being seizure-free for 10 months and was associated with cognitive improvement. After seizure relapse, lacosamide (LCM) was introduced, with good response, and the patient remained seizure-free for 6 months; developmental improvement followed. Concomitant LEV and VB6 treatment were also well-tolerated. Notably, we found that a KD was significantly associated with cognitive improvement. And the patient had no tendency to progress to WS with follow-up. The other patient, who harboured c.707G>A, was diagnosed with non-syndromic EIEE at 11 months of age, presenting with epileptic spasms, tonic seizures and focal seizures. VEEG patterns were burst suppressions, mixed with multiple focal spikes or polyspikes; brain MRI was normal. VPA and LEV were effective. After 2.5 years of treatment, the patient was free of seizures and no epileptic discharges were observed. The patient presented with moderate intellectual disability and a lisp.

CDKL5 variants were detected in two female patients: a frameshift variant and a non-sense variant. The patient with c.426_429del had a seizure onset at 2 months and developed myoclonic, tonic and absence seizures, with no response to VPA and TPM; global developmental impairment followed. She also showed some characteristic features of Rett syndrome, such as head growth deceleration, limited hand skills, and autonomic dysfunction. Unfortunately, the patient died of frequent seizures and severe pulmonary infection at 10 months of age. The other patient, carrying c.868C>T, was diagnosed with WS at 4 months of age. She presented with moderate developmental delay and VEEG features of hypsarrhythmia and multiple focal epileptic discharges. The types included epileptic spasms, absence seizures, and focal seizures. She was treated with VPA, TPM, and KD, and being seizure free was achieved for 1 year.

PIGN variants were detected in one male patient. A compound heterozygous variant was identified, whereby p.778fs was inherited from his father and p.Lys254Ile from his mother. Seizure onset was noted at 4 months of age, and the patient was diagnosed with non-syndromic EIEE. The seizure type was classical epileptic spasms. VEEG showed generalised or multifocal spike, sharp, spike-slow and polyspike-and-slow waves. Brain MRI revealed myelination delay in the cerebral white matter. The patient was treated with VPA, TPM, LEV, and clonazepam (CZP), but frequent episodes could not be controlled. Later, vigabatrin (VGB) was introduced and the seizure frequency decreased markedly. The patient exhibited severe intellectual delay; he could sit alone but could not walk by himself or speak until he was 5 years old. An additional clinical feature was anomalous ongoing hypotonia.

A PAFAH1B1 non-sense variant was detected in a male patient with intractable epilepsy and severe developmental milestone delay. The onset of seizures started at 2 months and types included epileptic spasms and tonic seizures. Brain MRI demonstrated a thickened cortex and gyral malformations. Hypsarrhythmia was identified on the VEEG record. The patient was diagnosed with WS and pachygyria, and received VPA and TPM for 4 months, but without seizure control. Then, a KD was initiated at 6 months of age, and seizure attacks in clusters decreased from more than 10 times per day to fewer than three times per day after 1 month. Nevertheless, the benefits for neurodevelopment are unclear, and frequent respiratory infections often result in second epileptic hits.

SCN1A variants were detected in three female patients and six male patients; Seven patients were diagnosed with DS and two with non-syndromic EIEEs. The mean onset age was 5 months. Various seizure types and developmental delays presented in the course of the disease, and VEEG showed multiple discharge abnormalities. VPA, TPM, and CZP were effective in controlling seizures in most of these patients, and seizure frequency was significantly reduced in three patients with KD treatment. After seizure remission, marked behavioural and cognitive improvements were observed. The SCN2A variant was detected in one female patient; the onset age was 7 months. Brain MRI revealed delayed myelin development and cerebral atrophy. High seizure frequency and seizure clustering were observed, which led to an exacerbation of early developmental delay.

A missense variant in KCNQ2 was detected in a female patient. Her seizures started within 2 months after birth; the seizure type was epileptic spasms. Brain MRI revealed delayed myelin development and VEEG hypsarrhythmia patterns. After the diagnosis of WS, she was treated with VPA, LEV, TPM, and adrenocorticotropic hormone (ACTH), but her seizures remained uncontrolled. The patient presents severe developmental delay and suffers from recurrent pulmonary infections.

Discussion

Because of the early age of onset and heterogeneity in the genotype and phenotype, it is difficult to identify several key phenotypic features of some of the genes responsible for EIEEs. We identified pathogenic variants in 34% of children with suspected genetic aetiology by NGS, which was similar to previous studies (8, 9). Despite progress in EIEE genetic diagnosis, there is still a limit for diagnosis using a gene panel. In the past, the gene panel was chosen prior to WES because of its cost-effectiveness, but targeted testing and inadequate coverage restricted the genetic diagnostic yield, which may miss disease-causing variants and lead a second cost of WES or whole-genome sequencing (WGS) and time. Routine use of WES or WGS can result in cost savings and higher diagnostic yields along with falling prices over time, which will lead to the discovery of new candidate genes and explanatory polygenic burdens (10). Moreover, detailed gene-specific phenotyping allowed us to seek diagnostic precision and to explore more specific approaches to treatment.

STXBP1 is predominantly expressed in the brain and plays an important role in synaptic vesicle docking and fusion (11). Variants in the STXBP1 gene have recently been described in OS, IS, DS, and non-syndromic EIEE, termed “STXBP1 encephalopathy” (STXBP1-E). The missense variant c.1439C>T has been reported to cause a severe phenotype, and is one of the recurrent variants (12, 13). Similarly, the patient in our study presented with developmental and epileptic encephalopathy (DEE) associated with frequent seizures and early onset. The patient with c.707G>A variant also displayed DEE but with later seizure onset. The ASMs reported to be most effective are VPA and LEV (13). Both the patients responded well to VPA and LEV, but also to VB6, LCM and KD. For early-onset seizures, especially neonatal seizures, early adoption of VB6 for management might be beneficial as a promising adjunctive therapy with an unproven night “depressant” role. Although poor long-term tolerance of KD remains a barrier, we still suggest early consideration of KD for significant seizure control and improved cognition in intractable epilepsy. In fact, a previous study reported complete seizure freedom with ketone concentrations of ~4.0 mmol/L (14); in our study, we maintained it at ~3.0 mmol/L considering potential complications and tolerability at the infant stage. Recurrent episodes and active epileptiform activity can aggravate cognitive problems, and we suggest that neurologic improvement might mainly result from amelioration of epileptic activity itself through a KD. LCM is associated with a novel mechanism of selectively enhancing slow inactivation of voltage-gated sodium channels (15). A previous study showed that LCM can exacerbate tonic seizures and electrical patterns in LGS (16), and LCM tolerance with concomitant sodium channel blocking drugs remains controversial (17). A previous study also reported that infant OS patients with STXBP1 variants may display transient responsiveness to folinic acid (18). The superiority of an ASM or a combination in STXBP1-E may require further research.

Variants of or deletions in the X-linked gene CDKL5 have been reported to cause EEs such as X-linked infantile spasms, EIEE-2, autism spectrum disorders, and Rett-like syndrome (19), which is termed CDKL5 deficiency disorder (CDD) (20). According to a previous study (21), the hallmarks of CDD usually include an early onset, a female predominance, intractability, typically polymorphic seizures, and severe developmental delay. Two children in this study had CDKL5 variants that were previously unreported. The types of CDKL5 variants may lead to differing severities of clinical phenotypes, and a deletion variant may exhibit a higher disease burden. In addition, one patient presented with a phenotype that might overlap with Rett syndrome, which was associated with MECP2 phosphorylation (22), but this feature was not detected in the WS patient. The WS patient showed a good response for a honeymoon period of 1 year without cognitive improvement. Whether the seizure-free outcome was associated with the honeymoon phase of natural disease progression in CDD or a result of ASMs is unclear (23). The severe phenotypes after an early-onset age caused by CDKL5 variant warrants more attention.

PIGN is one of more than 20 genes in the phosphatidylinositol family involved in glycosylphosphatidylinositol (GPI) biosynthesis, which is associated with neurodevelopmental disorders (NDD) (24). PIGA and PIGQ variants are associated with OS, WS, and early myoclonic encephalopathy (25). Variants in PIGN comprised a variable phenotypes including congenital anomalies, developmental delay, hypotonia, epilepsy, gastroesophageal reflux, and progressive cerebellar atrophy (26, 27). Here, we report that a male patient diagnosed with non-syndromic EIEE carried compound heterozygous PIGN variants. The clinical phenotype included early onset, drug-resistant epilepsy, hypotonia, severe developmental delay, and myelination delay. The unique manifestation was epileptic spasms, and VGB is significantly effective for treating epileptic spasms without hypsarrhythmia (ESWH). Significant dysmorphic features were not observed in this patient, and previous studies also supported that congenital anomalies associated with biallelic truncating variants are not a core feature of PIGN variants (28, 29). Biallelic PIGN variations should be considered as one of the causes of EIEE, especially when associated with seizures, hypotonia and intellectual disability.

As previously reported, some cases of isolated lissencephaly sequences are caused by variations in the PAFAH1B1 gene on chromosome 17p13.3 (30). Here, we reported a non-sense variant of PAFAH1B1, not previously reported, in a male patient. He showed an abnormal gyral pattern, drug-resistant epilepsy, and severe developmental delay. The brain MRI finding is consistent with pachygyria, a simplified convolutional pattern with few, broadened gyri and shallow sulci (31). Combined with the previous research results, non-sense variants and large deletions may be associated with more severe phenotypes than missense variants (32). We also observed the benefit of early KD implementation in the intractable condition.

SCN1A and SCN2A encode the major brain sodium channel subunits Nav1.1 and Nav1.2, respectively, which play crucial roles in the generation and propagation of neuronal firing (33). Variants in SCN1A and SCN2A have been linked to a spectrum of epilepsy phenotypes, including DS, generalised epilepsy with febrile seizures plus (GEFS+), WS and some other infantile epileptic disorders (34). The patients with SCN1A variants presented with early-onset seizures, drug-resistant epilepsy, developmental slowing and cognitive impairment, consistent with previous findings (35). Treatment strategies with some evidence of positive effects include VPA, clobazam, TPM, LEV, fenfluramine and bromides, or a KD (25, 36). In our study, the seizures were controlled by VPA, CZP, and KD, and we were delighted to observe improved cognitive development. In addition, the phenotypes of late-infantile onset, DEE and refractory epilepsy in the SCN2A variant may be associated with the loss of channel function (37). Patients with early developmental delay can benefit from genetic testing. Early and effective intervention may limit disease progression and significantly improve developmental outcomes and associated comorbidities.

KCNQ2 variants are associated with a wide phenotypic spectrum ranging from an age-dependent, self-limiting epilepsy, to a severe DEE but also an intermediate phenotype in terms of intellectual outcomes and time to reach seizure freedom (38). In the previous study (39), the recurrent KCNQ2 variant R198Q is associated with stereotypes with refractory seizures, severely abnormal VEEG and developmental delay. In our study, the KCNQ2 variant caused early-onset DEE and intractable seizures, and failed multidrug therapy led to a more severe developmental disorder.

Here, we explored the genetic aetiology of unexplained EIEEs and expanded the variant and phenotypic spectra. Furthermore, we elucidated the impact of the STXBP1 variant on the protein structure. The comparison of the structures of mutant and wild-type proteins showed that the variants affected protein stability, as described further in Supplementary Figures 1, 2. The nature and position of altered amino acids might affect the protein structure at different levels, which possibly explains the differences in clinical manifestations. The major limitations of this study are the mentioned limits of gene panels and the small sample size, which does not allow for exploring statistical correlations between genes and phenotypes.

Conclusion

NGS is a crucial tool to improve the diagnosis in EIEE and DEE and allows expanding the genotypic spectrum of this condition. Early molecular testing and detailed clinical phenotyping may lead to precise therapeutic interventions and can improve the developmental outcome. Functional studies further our understanding of novel target drugs, facilitating development in the future.

Data Availability Statement

The datasets presented in this study can be found in online repositories. The name of the repository and accession number can be found below: National Centre for Biotechnology Information (NCBI) BioProject, https://www.ncbi.nlm.nih.gov/bioproject/, PRJNA718023.

Ethics Statement

Written informed consent was obtained from the individual(s), and minor(s)' legal guardian/next of kin, for the publication of any potentially identifiable images or data included in this article.

Author Contributions

JC and XLiu: conceptualisation and writing. XLiu and QS: analysis. XW, XY, and ZC: data collection and confirmation. JC, GZ, HG, and XLu: project administration and supervision. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the Six Talent Peaks Project of China (Grant No. 2014-WSN-062).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fneur.2021.633637/full#supplementary-material

References

1. Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, van Emde BW, et al. Revised terminology and concepts for organization of seizures and epilepsies: report of the ILAE commission on classification and terminology, 2005-2009. Epilepsia. (2010) 51:676–85. doi: 10.1111/j.1528-1167.2010.02522.x

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Stafstrom CE, Kossoff EM. Epileptic encephalopathy in infants and children. Epilepsy Curr. (2016) 16:273–9. doi: 10.5698/1535-7511-16.4.273

CrossRef Full Text | Google Scholar

3. Mctague A, Howell KB, Cross JH, Kurian MA, Scheffer IE. The genetic landscape of the epileptic encephalopathies of infancy and childhood. Lancet Neurol. (2016) 15:304–16. doi: 10.1016/S1474-4422(15)00250-1

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Scala M, Bianchi A, Bisulli F, Coppola A, Elia M, Trivisano M, et al. Advances in genetic testing and optimization of clinical management in children and adults with epilepsy. Expert Rev Neurother. (2020) 20:251–69. doi: 10.1080/14737175.2020.1713101

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Scheffer IE, Berkovic S, Capovilla G, Connolly MB, French J, Guilhoto L, et al. ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology. Epilepsia. (2017) 58:512–21. doi: 10.1111/epi.13709

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology. Genet Med. (2015) 17:405–24. doi: 10.1038/gim.2015.30

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Biasini M, Bienert S, Waterhouse A, Arnold K, Studer G, Schmidt T, et al. SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Res. (2014) 42:W252–8. doi: 10.1093/nar/gku340

PubMed Abstract | CrossRef Full Text | Google Scholar

8. 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) 91:717–24. doi: 10.1111/cge.12901

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Na JH, Shin S, Yang D, Kim B, Kim HD, Kim S, et al. Targeted gene panel sequencing in early infantile onset developmental and epileptic encephalopathy. Brain Dev. (2020) 42:438–48. doi: 10.1016/j.braindev.2020.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Lionel AC, Costain G, Monfared N, Walker S, Reuter MS, Hosseini SM, et al. Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test. Genet Med. (2018) 20:435–43. doi: 10.1038/gim.2017.119

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Swanson DA, Steel JM, Valle D. Identification and characterization of the human ortholog of rat STXBP1, a protein implicated in vesicle trafficking and neurotransmitter release. Genomics. (1998) 48:373–6. doi: 10.1006/geno.1997.5202

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Di Meglio C, Lesca G, Villeneuve N, Lacoste C, Abidi A, Cacciagli P, et al. Epileptic patients with de novo STXBP1 mutations: key clinical features based on 24 cases. Epilepsia. (2015) 56:1931–40. doi: 10.1111/epi.13214

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Stamberger H, Nikanorova M, Willemsen MH, Accorsi P, Angriman M, Baier H, et al. STXBP1 encephalopathy: a neurodevelopmental disorder including epilepsy. Neurology. (2016) 86:954–62. doi: 10.1212/WNL.0000000000002457

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Li T, Cheng M, Wang J, Hong S, Li M, Liao S, et al. De novo mutations of STXBP1 in Chinese children with early onset epileptic encephalopathy. Genes Brain Behav. (2018) 17:e12492. doi: 10.1111/gbb.12492

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Beydoun A, D'Souza J, Hebert D, Doty P. Lacosamide: pharmacology, mechanisms of action and pooled efficacy and safety data in partial-onset seizures. Expert Rev Neurother. (2009) 9:33–42. doi: 10.1586/14737175.9.1.33

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Andrade-Machado R, Benjumea-Cuartas V, Jaramillo-Jimenez E. Lacosamide in lennox-Gastaut syndrome: case report. Clin Neuropharmacol. (2012) 35:148–9. doi: 10.1097/WNF.0b013e318255b649

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Ruegger AD, Freeman JL, Harvey AS. Lacosamide in children with drug-resistant epilepsy. J Paediatr Child Health. (2019) 55:194–8. doi: 10.1111/jpc.14156

CrossRef Full Text | Google Scholar

18. Tso WW, Kwong AK, Fung CW, Wong VC. Folinic acid responsive epilepsy in Ohtahara syndrome caused by STXBP1 mutation. Pediatr Neurol. (2014) 50:177–80. doi: 10.1016/j.pediatrneurol.2013.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Mei D, Marini C, Novara F, Bernardina BD, Granata T, Fontana E, et al. Xp22.3 genomic deletions involving the CDKL5 gene in girls with early onset epileptic encephalopathy. Epilepsia. (2010) 51:647–54. doi: 10.1111/j.1528-1167.2009.02308.x

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Pintaudi M, Baglietto MG, Gaggero R, Parodi E, Pessagno A, Marchi M, et al. Clinical and electroencephalographic features in patients with CDKL5 mutations: two new Italian cases and review of the literature. Epilepsy Behav. (2008) 12:326–31. doi: 10.1016/j.yebeh.2007.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Archer HL, Evans J, Edwards S, Colley J, Newbury-Ecob R, O'Callaghan F, et al. CDKL5 mutations cause infantile spasms, early onset seizures, and severe mental retardation in female patients. J Med Genet. (2006) 43:729–34. doi: 10.1136/jmg.2006.041467

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Almomen M, Rho JM, Scantlebury MH. Infantile epileptic encephalopathy with multiple genetic mutations: how important are variants of undetermined significance? Semin Pediatr Neurol. (2018) 26:33–6. doi: 10.1016/j.spen.2018.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Bahi-Buisson N, Kaminska A, Boddaert N, Rio M, Afenjar A, Gerard M, et al. The three stages of epilepsy in patients with CDKL5 mutations. Epilepsia. (2008) 49:1027–37. doi: 10.1111/j.1528-1167.2007.01520.x

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Freeze HH, Eklund EA, Ng BG, Patterson MC. Neurology of inherited glycosylation disorders. Lancet Neurol. (2012) 11:453–66. doi: 10.1016/S1474-4422(12)70040-6

CrossRef Full Text | Google Scholar

25. Balestrini S, Sisodiya SM. Treatment of epileptic encephalopathies. Curr Pharm Des. (2017) 23:5667–90. doi: 10.2174/1381612823666170809115827

CrossRef Full Text | Google Scholar

26. Couser NL, Masood MM, Strande NT, Foreman AK, Crooks K, Weck KE, et al. The phenotype of multiple congenital anomalies-hypotonia-seizures syndrome 1: report and review. Am J Med Genet A. (2015) 167A:2176–81. doi: 10.1002/ajmg.a.37129

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Ohba C, Okamoto N, Murakami Y, Suzuki Y, Tsurusaki Y, Nakashima M, et al. PIGN mutations cause congenital anomalies, developmental delay, hypotonia, epilepsy, and progressive cerebellar atrophy. Neurogenetics. (2014) 15:85–92. doi: 10.1007/s10048-013-0384-7

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Thiffault I, Zuccarelli B, Welsh H, Yuan X, Farrow E, Zellmer L, et al. Hypotonia and intellectual disability without dysmorphic features in a patient with PIGN-related disease. BMC Med Genet. (2017) 18:124. doi: 10.1186/s12881-017-0481-9

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Fleming L, Lemmon M, Beck N, Johnson M, Mu W, Murdock D, et al. Genotype-phenotype correlation of congenital anomalies in multiple congenital anomalies hypotonia seizures syndrome (MCAHS1)/PIGN-related epilepsy. Am J Med Genet A. (2016) 170A:77–86. doi: 10.1002/ajmg.a.37369

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Blazejewski SM, Bennison SA, Smith TH, Toyo-Oka K. Neurodevelopmental genetic diseases associated with microdeletions and microduplications of chromosome 17p13.3. Front Genet. (2018) 9:80. doi: 10.3389/fgene.2018.00080

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Severino M, Geraldo AF, Utz N, Tortora D, Pogledic I, Klonowski W, et al. Definitions and classification of malformations of cortical development: practical guidelines. Brain. (2020) 143:2874–94. doi: 10.1093/brain/awaa174

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Cardoso C, Leventer RJ, Dowling JJ, Ward HL, Chung J, Petras KS, et al. Clinical and molecular basis of classical lissencephaly: mutations in the LIS1 gene (PAFAH1B1). Hum Mutat. (2002) 19:4–15. doi: 10.1002/humu.10028

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Maljevic S, Reid CA, Petrou S. Models for discovery of targeted therapy in genetic epileptic encephalopathies. J Neurochem. (2017) 143:30–48. doi: 10.1111/jnc.14134

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Claes L, Del-Favero J, Ceulemans B, Lagae L, Van Broeckhoven C, De Jonghe P. De novo mutations in the sodium-channel gene SCN1A cause severe myoclonic epilepsy of infancy. Am J Hum Genet. (2001) 68:1327–32. doi: 10.1086/320609

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Cetica V, Chiari S, Mei D, Parrini E, Grisotto L, Marini C, et al. Clinical and genetic factors predicting Dravet syndrome in infants with SCN1A mutations. Neurology. (2017) 88:1037–44. doi: 10.1212/WNL.0000000000003716

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Balagura G, Cacciatore M, Grasso EA, Striano P, Verrotti A. Fenfluramine for the treatment of dravet syndrome and lennox-gastaut syndrome. CNS Drugs. (2020) 34:1001–7. doi: 10.1007/s40263-020-00755-z

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Shi X, Yasumoto S, Kurahashi H, Nakagawa E, Fukasawa T, Uchiya S, et al. Clinical spectrum of SCN2A mutations. Brain Dev. (2012) 34:541–5. doi: 10.1016/j.braindev.2011.09.016

CrossRef Full Text | Google Scholar

38. Malerba F, Alberini G, Balagura G, Marchese F, Amadori E, Riva A, et al. Genotype-phenotype correlations in patients with de novo KCNQ2 pathogenic variants. Neurol Genet. (2020) 6:e528. doi: 10.1212/NXG.0000000000000528

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Millichap JJ, Miceli F, De Maria M, Keator C, Joshi N, Tran B, et al. Infantile spasms and encephalopathy without preceding neonatal seizures caused by KCNQ2 R198Q, a gain-of-function variant. Epilepsia. (2017) 58:e10–5. doi: 10.1111/epi.13601

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: encephalopathy, epilepsy, infantile, gene, phenotype

Citation: Liu X, Shen Q, Zheng G, Guo H, Lu X, Wang X, Yang X, Cao Z and Chen J (2021) Gene and Phenotype Expansion of Unexplained Early Infantile Epileptic Encephalopathy. Front. Neurol. 12:633637. doi: 10.3389/fneur.2021.633637

Received: 21 January 2021; Accepted: 14 April 2021;
Published: 07 June 2021.

Edited by:

Pasquale Striano, University of Genoa, Italy

Reviewed by:

Andrea Accogli, Istituto Giannina Gaslini, Italy
Ganna Balagura, Department of Pediatric Neurology and Muscle Diseases, Giannina Gaslini Institute (IRCCS), Italy

Copyright © 2021 Liu, Shen, Zheng, Guo, Lu, Wang, Yang, Cao and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jing Chen, dr.chenj@njmu.edu.cn

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.