1
|
Kushner JK, Hoffman PB, Brzezinski CR, Svalina MN, O'Neill BR, Hankinson TC, Wilkinson CC, Handler MH, Baca SM, Huntsman MM, Alexander AL. Characterizing the Diversity of Layer 2/3 Human Neocortical Neurons in Pediatric Epilepsy. eNeuro 2025; 12:ENEURO.0247-24.2025. [PMID: 40246555 PMCID: PMC12061357 DOI: 10.1523/eneuro.0247-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Childhood epilepsy is a common and devastating condition, for which many children still do not have adequate treatment. Some children with drug-resistant epilepsy require surgical excision of epileptogenic brain tissue for seizure control, affording the opportunity to study this tissue ex vivo to interrogate human epileptic neurons for potentially hyperexcitable perturbations in intrinsic electrophysiological properties. In this study, we characterized the diversity of layer L2/3 (L2/3) pyramidal neurons (PNs) in ex vivo brain slices from pediatric patients with epilepsy. We found a remarkable diversity in the firing properties of epileptic L2/3 PNs: five distinct subpopulations were identified. Additionally, we investigated whether the etiology of epilepsy influenced the intrinsic neuronal properties of L2/3 PNs when comparing tissue from patients with epilepsy due to malformations of cortical development (MCDs), other forms of epilepsy (OEs), or with deep-seated tumors. When comparing epileptic with control L2/3 PNs, we observed a decrease in voltage sag and lower maximum firing rates. Moreover, we found that MCD and OE L2/3 PNs were mostly similar indicating that epilepsy etiology may not outweigh the influences of epileptiform activity on L2/3 PN physiology. Lastly, we show that the proconvulsant drug, 4-aminopyridine (4-AP), leads to increased AP half-width, reduced firing rate accommodation, and slower AHPs. These changes imply that 4-AP induces an increase in [K+]o and a resultant increase in AP duration, leading to the release of more excitatory neurotransmitters per action potential, thereby promoting network hyperexcitability.
Collapse
Affiliation(s)
- J Keenan Kushner
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Paige B Hoffman
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Christine R Brzezinski
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Matthew N Svalina
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Medical Scientist Training Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Cell and Developmental Biology, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Brent R O'Neill
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Todd C Hankinson
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Charles C Wilkinson
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Michael H Handler
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Serapio M Baca
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Molly M Huntsman
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pediatrics, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| | - Allyson L Alexander
- Neuroscience Graduate Program, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Neurosurgery, School of Medicine, University of Colorado | Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
2
|
Visvaratnam H, Korff CM, Seeck M, Fitsiori A. Pearls & Oy-sters: Increased Visibility of Focal Cortical Dysplasia in Cerebral MRI During the First Year of Life. Neurology 2025; 104:e210128. [PMID: 39729603 DOI: 10.1212/wnl.0000000000210128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/05/2024] [Indexed: 12/29/2024] Open
Abstract
Early detection of focal cortical dysplasia (FCD) using brain MRI in young children presenting with drug-resistant epilepsy may facilitate prompt surgical treatment, resulting in better control of seizures and decreased associated cognitive difficulties. Characteristics of FCD described in the literature are predominantly based on MRI findings in a fully myelinated brain; therefore, changes occurring during early brain maturation are not well known. In this case report, we describe distinct MRI features of a FCD visualized best before completion of myelination of the cortex and subcortical white matter. Specifically, this lesion exhibited increased visibility because of better delineation of the borders of the pathological area involved in the malformation compared with imaging obtained after completion of myelination. Although the pathophysiology of these imaging features necessitates further investigation, one hypothesis is that excessive neuronal input in young epileptic patients could trigger premature myelination of overstimulated fibers underlying the cortical epileptogenic focus. The aim of this case report was to raise awareness that seizures might induce local changes in brain myelination, which can be detected by MRI during the first year of life. Early identification of FCD may prompt surgical treatment in selected patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
| | - Christian M Korff
- Faculty of Medicine, University of Geneva, Switzerland
- Neuropediatrics Unit, Woman, Child and Adolescent Department, University Hospitals of Geneva, Switzerland
| | - Margitta Seeck
- Faculty of Medicine, University of Geneva, Switzerland
- EEG and Epilepsy Unit, Department of Clinical Neurosciences, University Hospitals of Geneva, Switzerland; and
| | - Aikaterini Fitsiori
- Faculty of Medicine, University of Geneva, Switzerland
- Diagnostic and Interventional Neuroradiology Unit, Diagnostic Department, University Hospitals of Geneva, Switzerland
| |
Collapse
|
3
|
van Lanen RHGJ, Uher D, Tse DHY, Steijvers E, Colon AJ, Jansen JFA, Drenthen GS, Ivanov D, Hoogland G, Rijkers K, Hoeberigs CM, Hofman PAM, Backes WH, Schijns OEMG. In vivo 9.4 Tesla MRI of a patient with drug-resistant epilepsy: Technical report. Acta Neurochir (Wien) 2025; 167:18. [PMID: 39820684 PMCID: PMC11739318 DOI: 10.1007/s00701-024-06385-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/07/2024] [Indexed: 01/19/2025]
Abstract
PURPOSE In resective epilepsy surgery for drug-resistant focal epilepsy (DRE), good seizure outcome is strongly associated with visualization of an epileptogenic lesion on MRI. Standard clinical MRI (≤ 3 Tesla (T)) may fail to detect subtle lesions. 7T MRI enhances detection and delineation, the potential benefits of increasing field strength to 9.4T are explored. METHODS A 36 years old male patient with DRE evaluated for resective surgery, in which 3T and 7T MRI failed to detect any epileptogenic lesions, was submitted to a dedicated epilepsy scan protocol using T1 and T2* weighted imaging at 9.4T. Images were evaluated independently by two neuroradiologists and one neurosurgeon. RESULTS 9.4T MRI offered increased spatial resolution and enhanced depiction of anatomical structures vital for epilepsy imaging, exemplified by regions mesio-temporal (hippocampus, amygdala), latero-temporal, insula, frontal and temporal operculum, and gray-white matter junction (precentral gyrus/frontal lobe) compared to 3T and 7T, albeit with challenges in mesial-temporal and antero-inferior temporal lobe imaging. No epileptogenic lesion was identified. CONCLUSION 9.4T demonstrates promise in the identification and delineation of anatomical structures and small epileptogenic lesions in patients with DRE eligible for resective surgery. Whether clinical 9.4T MRI in DRE has clinical advantages over 7T or leads to a more complete resection of the epileptogenic zone and improved seizure outcome after epilepsy surgery needs to be established.
Collapse
Affiliation(s)
- Rick H G J van Lanen
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands.
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands.
| | - Daniel Uher
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Desmond H Y Tse
- Scannexus, Ultra-High Field MRI Research Centre, Maastricht, Netherlands
| | - Esther Steijvers
- Scannexus, Ultra-High Field MRI Research Centre, Maastricht, Netherlands
| | - Albert J Colon
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
- Service de la recherche et traitement d'epilepsie, Centre Hospitalier Universitaire Martinique, Fort-de-France, France
| | - Jacobus F A Jansen
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Gerhard S Drenthen
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Dimo Ivanov
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
| | - Kim Rijkers
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
| | - Christianne M Hoeberigs
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
| | - Paul A M Hofman
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
| | - Walter H Backes
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
- Research Institute for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, Netherlands
| | - Olaf E M G Schijns
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre, Heeze, Kempenhaeghe, Maastricht, Netherlands
| |
Collapse
|
4
|
Arceneaux JS, Brockman AA, Khurana R, Chalkley MBL, Geben LC, Krbanjevic A, Vestal M, Zafar M, Weatherspoon S, Mobley BC, Ess KC, Ihrie RA. Multiparameter quantitative analyses of diagnostic cells in brain tissues from tuberous sclerosis complex. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2025; 108:35-54. [PMID: 38953209 PMCID: PMC11693778 DOI: 10.1002/cyto.b.22194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 07/03/2024]
Abstract
The advent of high-dimensional imaging offers new opportunities to molecularly characterize diagnostic cells in disorders that have previously relied on histopathological definitions. One example case is found in tuberous sclerosis complex (TSC), a developmental disorder characterized by systemic growth of benign tumors. Within resected brain tissues from patients with TSC, detection of abnormally enlarged balloon cells (BCs) is pathognomonic for this disorder. Though BCs can be identified by an expert neuropathologist, little is known about the specificity and broad applicability of protein markers for these cells, complicating classification of proposed BCs identified in experimental models of this disorder. Here, we report the development of a customized machine learning pipeline (BAlloon IDENtifier; BAIDEN) that was trained to prospectively identify BCs in tissue sections using a histological stain compatible with high-dimensional cytometry. This approach was coupled to a custom 36-antibody panel and imaging mass cytometry (IMC) to explore the expression of multiple previously proposed BC marker proteins and develop a descriptor of BC features conserved across multiple tissue samples from patients with TSC. Here, we present a modular workflow encompassing BAIDEN, a custom antibody panel, a control sample microarray, and analysis pipelines-both open-source and in-house-and apply this workflow to understand the abundance, structure, and signaling activity of BCs as an example case of how high-dimensional imaging can be applied within human tissues.
Collapse
Affiliation(s)
- Jerome S. Arceneaux
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, Meharry Medical College
| | - Asa A. Brockman
- Department of Cell & Developmental Biology, Vanderbilt University
| | - Rohit Khurana
- Department of Cell & Developmental Biology, Vanderbilt University
| | | | | | - Aleksandar Krbanjevic
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center
| | | | | | - Sarah Weatherspoon
- Neuroscience Institute, Le Bonheur Children’s Hospital
- University of Tennessee Health Science Center
| | - Bret C. Mobley
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center
| | - Kevin C. Ess
- Department of Cell & Developmental Biology, Vanderbilt University
- Department of Pediatrics, Vanderbilt University Medical Center
- Department of Neurology, Vanderbilt University Medical Center
- Section of Child Neurology, University of Colorado Anschutz Medical Center
| | - Rebecca A. Ihrie
- Department of Cell & Developmental Biology, Vanderbilt University
- Department of Neurological Surgery, Vanderbilt University Medical Center
| |
Collapse
|
5
|
Cao Y, Meng W, Zheng J, Zhang C, Gao D, Zhou L. Prenatal MRI for the diagnosis of foetal pial arteriovenous fistula: a case report and literature review. J OBSTET GYNAECOL 2024; 44:2299372. [PMID: 38174958 DOI: 10.1080/01443615.2023.2299372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Affiliation(s)
- Yimin Cao
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Weixin Meng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - JianXiong Zheng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Chengye Zhang
- Clinical Medicine of Chengde Medical College, Chengde City, China
| | - Duo Gao
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - LiXia Zhou
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| |
Collapse
|
6
|
Brunelli JM, Lopes TJP, Alves IS, Delgado DS, Lee HW, Martin MGM, Docema MFL, Alves SS, Pinho PC, Gonçalves VT, Oliveira LRLB, Takahashi JT, Maralani PJ, Amancio CT, Leite CC. Malformations of Cortical Development: Updated Imaging Review. Radiographics 2024; 44:e230239. [PMID: 39446612 DOI: 10.1148/rg.230239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Malformations of cortical development (MCD) are structural anomalies that disrupt the normal process of cortical development. Patients with these anomalies frequently present with seizures, developmental delay, neurologic deficits, and cognitive impairment, resulting in a wide spectrum of neurologic outcomes. The severity and type of malformation, in addition to the genetic pathways of brain development involved, contribute to the observed variability. While neuroimaging plays a central role in identifying congenital anomalies in vivo, the precise definition and classification of cortical developmental defects have undergone significant transformations in recent years due to advances in molecular and genetic knowledge. The authors provide a concise overview of embryologic brain development, recently standardized nomenclature, and the categorization system for abnormalities in cortical development, offering valuable insights into the interpretation of their neuroradiologic patterns. ©RSNA, 2024 Supplemental material is available for this article. The slide presentation from the RSNA Annual Meeting is available for this article.
Collapse
Affiliation(s)
- Julia M Brunelli
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Thiago J P Lopes
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Isabela S Alves
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Daniel S Delgado
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Hae W Lee
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Maria G M Martin
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Marcos F L Docema
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Samya S Alves
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Paula C Pinho
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Vinicius T Gonçalves
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Lucas R L B Oliveira
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Jorge T Takahashi
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Pejman J Maralani
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Camila T Amancio
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| | - Claudia C Leite
- From the Department of Radiology, Hospital Sírio-Libanês, Adma Jafet 91, Bela Vista, São Paulo, Brazil 01308-050 (J.M.B., T.J.P.L., I.S.A., D.S.D., H.W.L., M.G.M.M., M.F.L.D., S.S.A., P.C.P., V.T.G., J.T.T., C.T.A.); Departments of Radiology (M.G.M.M., P.C.P., L.R.L.B.O., C.C.L.) and Oncology (C.C.L.), University of São Paulo, São Paulo, Brazil; and Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (P.J.M.)
| |
Collapse
|
7
|
Bustros S, Kaur M, Ritchey E, Szaflarski JP, McGwin GJ, Riley KO, Bentley JN, Memon AA, Jaisani Z. Non-lesional epilepsy does not necessarily convey poor outcomes after invasive monitoring followed by resection or thermal ablation. Neurol Res 2024; 46:653-661. [PMID: 38602305 DOI: 10.1080/01616412.2024.2340879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE We aimed to compare outcomes including seizure-free status at the last follow-up in adult patients with medically refractory focal epilepsy identified as lesional vs. non-lesional based on their magnetic resonance imaging (MRI) findings who underwent invasive evaluation followed by subsequent resection or thermal ablation (LiTT). METHODS We identified 88 adult patients who underwent intracranial monitoring between 2014 and 2021. Of those, 40 received resection or LiTT, and they were dichotomized based on MRI findings, as lesional (N = 28) and non-lesional (N = 12). Patient demographics, seizure characteristics, non-invasive interventions, intracranial monitoring, and surgical variables were compared between the groups. Postsurgical seizure outcome at the last follow-up was rated according to the Engel classification, and postoperative seizure freedom was determined by Kaplan-Meyer survival analysis. Statistical analyses employed Fisher's exact test to compare categorical variables, while a t-test was used for continuous variables. RESULTS There were no differences in baseline characteristics between groups except for more often noted PET abnormality in the lesional group (p = 0.0003). 64% of the lesional group and 57% of the non-lesional group received surgical resection or LiTT (p = 0.78). At the last follow-up, 78.5% of the patients with lesional MRI findings achieved Engel I outcomes compared to 66.7% of non-lesional patients (p = 0.45). Kaplan-Meier curves did not show a significant difference in seizure-free duration between both groups after surgical intervention (p = 0.49). SIGNIFICANCE In our sample, the absence of lesion on brain MRI was not associated with worse seizure outcomes in adult patients who underwent invasive intracranial monitoring followed by resection or thermal ablation.
Collapse
Affiliation(s)
- Stephanie Bustros
- Division of Epilepsy, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Manmeet Kaur
- Division of Neurocritical Care, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Elizabeth Ritchey
- Division of Epilepsy, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Jerzy P Szaflarski
- Division of Epilepsy, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- Division of Neurocritical Care, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Gerald Jr McGwin
- Department of Epidemiology, School of Public Health, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kristen O Riley
- Department of Neurosurgery, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - J Nicole Bentley
- Department of Neurosurgery, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Adeel A Memon
- Department of Neurology, West Virginia University, Morgantown, WV, USA
| | - Zeenat Jaisani
- Division of Epilepsy, Department of Neurology, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| |
Collapse
|
8
|
Lee M, Kim EJ, Yum MS. Early developmental changes in a rat model of malformations of cortical development: Abnormal neuronal migration and altered response to NMDA-induced excitotoxic injury. Exp Neurol 2024; 376:114759. [PMID: 38519010 DOI: 10.1016/j.expneurol.2024.114759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
Malformations of cortical development (MCDs) are caused by abnormal neuronal migration processes during the fetal period and are a major cause of intractable epilepsy in infancy. However, the timing of hyperexcitability or epileptogenesis in MCDs remains unclear. To identify the early developmental changes in the brain of the MCD rat model, which exhibits increased seizure susceptibility during infancy (P12-15), we analyzed the pathological changes in the brains of MCD model rats during the neonatal period and tested NMDA-induced seizure susceptibility. Pregnant rats were injected with two doses of methylazoxymethanol acetate (MAM, 15 mg/kg, i.p.) to induce MCD, while controls were administered normal saline. The cortical development of the offspring was measured by performing magnetic resonance imaging (MRI) on postnatal days (P) 1, 5, and 8. At P8, some rats were sacrificed for immunofluorescence, Golgi staining, and Western analysis. In another set of rats, the number and latency to onset of spasms were monitored for 90 min after the NMDA (5 mg/kg i.p.) injection at P8. In MCD rats, in vivo MR imaging showed smaller brain volume and thinner cortex from day 1 after birth (p < 0.001). Golgi staining and immunofluorescence revealed abnormal neuronal migration, with a reduced number of neuronal cell populations and less dendritic arborization at P8. Furthermore, MCD rats exhibited a significant reduction in the expression of NMDA receptors and AMPAR4, along with an increase in AMPAR3 expression (p < 0.05). Although there was no difference in the latency to seizure onset between MCD rats and controls, the MCD rats survived significantly longer than the controls. These results provide insights into the early developmental changes in the cortex of a MCD rat model and suggest that delayed and abnormal neuronal development in the immature brain is associated with a blunted response to NMDA-induced excitotoxic injury. These developmental changes may be involved in the sudden onset of epilepsy in patients with MCD or prenatal brain injury.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.
| | - Eun-Jin Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Mi-Sun Yum
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Department of Pediatrics, Asan Medical Center Children's Hospital, Seoul 05505, Republic of Korea.
| |
Collapse
|
9
|
Procházková N, Nguyenová MT, Řehořová M, Kudláček J, Chvojka J, Ziak J, Balaštík M, Otáhal J, Jiruška P, Novák O. NeuroPorator: An open-source, current-limited electroporator for safe in utero gene transfer. J Neurosci Methods 2024; 406:110126. [PMID: 38554786 DOI: 10.1016/j.jneumeth.2024.110126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/13/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Electroporation is an effective technique for genetic manipulation of cells, both in vitro and in vivo. In utero electroporation (IUE) is a special case, which represents a fine application of this technique to genetically modify specific tissues of embryos during prenatal development. Commercially available electroporators are expensive and not fully customizable. We have designed and produced an inexpensive, open-design, and customizable electroporator optimized for safe IUE. We introduce NeuroPorator. METHOD We used off-the-shelf electrical parts, a single-board microcontroller, and a cheap data logger to build an open-design electroporator. We included a safety circuit to limit the applied electrical current to protect the embryos. We added full documentation, design files, and assembly instructions. RESULT NeuroPorator output is on par with commercially available devices. Furthermore, the adjustable current limiter protects both the embryos and the uterus from overcurrent damage. A built-in data acquisition module provides real-time visualization and recordings of the actual voltage/current pulses applied to each embryo. Function of NeuroPorator has been demonstrated by inducing focal cortical dysplasia in mice. SIGNIFICANCE AND CONCLUSION The simple and fully open design enables quick and cheap construction of the device and facilitates further customization. The features of NeuroPorator can accelerate the IUE technique implementation in any laboratory and speed up its learning curve.
Collapse
Affiliation(s)
- Natálie Procházková
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Minh-Thao Nguyenová
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Monika Řehořová
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Jan Kudláček
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Jan Chvojka
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Jakub Ziak
- Laboratory of Molecular Neurobiology, Institute of Physiology, The Czech Academy of Sciences, Videnska 1083, Prague 14200, Czech Republic
| | - Martin Balaštík
- Laboratory of Molecular Neurobiology, Institute of Physiology, The Czech Academy of Sciences, Videnska 1083, Prague 14200, Czech Republic
| | - Jakub Otáhal
- Department of Pathophysiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Přemysl Jiruška
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic
| | - Ondřej Novák
- Department of Physiology, Second Faculty of Medicine, Charles University, Plzenska 311, Prague 15000, Czech Republic.
| |
Collapse
|
10
|
Nemoto H, Imagawa K, Enokizono T, Masuda Y, Ide M, Deguchi T, Hara M, Morita A, Kido T, Tanaka M, Ohto T, Takada H. A Case of Anorexia Nervosa with Focal Cortical Dysplasia. Case Rep Psychiatry 2024; 2024:7478666. [PMID: 38716398 PMCID: PMC11074877 DOI: 10.1155/2024/7478666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024] Open
Abstract
Anorexia nervosa (AN) is a fatal condition associated with extreme underweight and undernutrition. It is more common in young females, with a female-to-male ratio of 10 : 1. Focal cortical dysplasia (FCD) is characterized by dysplasia of the cerebral cortex and is a common cause of pharmacoresistant epilepsy. However, FCD associated with AN has never been reported. We report the first case of AN in a 12-year-old male diagnosed with FCD-type 2 on head magnetic resonance imaging (MRI). He became concerned about lower abdominal distention and began reducing his food intake. He was admitted to our hospital after weight loss of 10 kg in a 1 year. Head MRI showed a localized high-signal area from the cortex to the white matter of the fusiform gyrus near the left hippocampus, with no associated decreased blood flow or electroencephalography (EEG) abnormalities. These findings were characteristic of FCD type II. In males with AN, the search for underlying disease is particularly important. The pathophysiology of the association between AN and FCD is unclear. However, both conditions are reportedly associated with autism spectrum disorder. Further cases are needed to clarify whether FCD is associated with eating disorders.
Collapse
Affiliation(s)
- Hiroki Nemoto
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Kazuo Imagawa
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Takashi Enokizono
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Yosuke Masuda
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Masayuki Ide
- Center for Medical Sciences, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami, Inashiki-gun, Ami-machi, Ibaraki, Japan
| | - Takuma Deguchi
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Monami Hara
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Atsushi Morita
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Takahiro Kido
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Mai Tanaka
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Tatsuyuki Ohto
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki, Japan
| |
Collapse
|
11
|
Pizzo F, Fichera V, Zanghì A, Praticò AD, Vecchio M, Falsaperla R, Lavalle S, Marino F, Palmucci S, Belfiore G, Polizzi A. Focal Cortical Dysplasia: Diagnosis, Classification, and Treatment Options. JOURNAL OF PEDIATRIC NEUROLOGY 2024; 22:166-171. [DOI: 10.1055/s-0044-1786781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
AbstractFocal cortical dysplasias (FCDs) include a spectrum of anomalies of cortical development that consist in one or more areas with altered lamination and in some cases, neurons of abnormal morphology. Clinically, these structural anomalies led to arise of epilepsy, which is more often a focal, drug-resistant type with onset in pediatric or adolescent age. Occasionally, other symptoms have been reported in patients with FCDs, such as headache, movement disorders, and cognitive impairment. According to International League against Epilepsy scheme of 2011, three main subtypes of FCD can be distinguished, based of anatomopathological feature, radiological signs, and clinical expression. Magnetic resonance imaging (MRI), fluorodeoxyglucose positron emission tomography, and neurophysiology are the cornerstones of diagnosis, although their negativity cannot exclude FCD in symptomatic patients, especially in FCD type I which often is elusive. In MRI, the main finding is the irregularity of the cortical–subcortical signal, specifically reduction of cortical thickness and absence of clear demarcation between gray and white matters, which is strongly diagnostic for FCD. Epilepsy related to FCD is difficult to manage and until now there is not a clear direction for treatment's rules. FCD shows poor response to antiepileptic drugs (AEDs), and there is no evidence of some AED that has proved more efficacy than others in patients with FCDs. Considering genetical and pathophysiological recent acquisitions, mammalian target of rapamycin inhibitors may play a fundamental role in future treatment of FCDs, but nowadays, surgery still remains the main weapon, with 50% of patients who undergo neurosurgery.
Collapse
Affiliation(s)
- Francesco Pizzo
- Pediatrics Postgraduate Residency Program, University of Catania, Catania, Italy
| | - Valeria Fichera
- Pediatrics Postgraduate Residency Program, University of Catania, Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technologies, Research Center for Surgery of Complex Malformation Syndromes of Transition and Adulthood, University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Chair of Pediatrics, Department of Medicine and Surgery, Kore University, Enna, Italy
| | - Michele Vecchio
- Rehabilitation Unit, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Neonatology and Neonatal Intensive Care Unit, University Hospital Policlinico “G. Rodolico-San Marco,” Catania, Italy
| | - Salvatore Lavalle
- Chair of Pediatrics, Department of Medicine and Surgery, Kore University, Enna, Italy
| | - Francesco Marino
- Department of Medical Surgical Sciences and Advanced Technologies, University Hospital Policlinico “G. Rodolico-San Marco,” Catania, Italy
| | - Stefano Palmucci
- IPTRA Unit, Department of Medical Surgical Sciences and Advanced Technologies, University Hospital Policlinico “G. Rodolico-San Marco,” Catania, Italy
| | - Giuseppe Belfiore
- Unit of Radiology 1, Department of Medical Surgical Sciences and Advanced Technologies, University Hospital Policlinico “G. Rodolico-San Marco,” Catania, Italy
| | - Agata Polizzi
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
| |
Collapse
|
12
|
Fu CH, You JC, Mohila C, Rissman RA, Yoshor D, Viaene AN, Chin J. Hippocampal ΔFosB expression is associated with cognitive impairment in a subgroup of patients with childhood epilepsies. Front Neurol 2024; 14:1331194. [PMID: 38274865 PMCID: PMC10808715 DOI: 10.3389/fneur.2023.1331194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Epilepsy is a chronic neurological disorder characterized by recurrent seizures, and is often comorbid with other neurological and neurodegenerative diseases, such as Alzheimer's disease (AD). Patients with recurrent seizures often present with cognitive impairment. However, it is unclear how seizures, even when infrequent, produce long-lasting deficits in cognition. One mechanism may be seizure-induced expression of ΔFosB, a long-lived transcription factor that persistently regulates expression of plasticity-related genes and drives cognitive dysfunction. We previously found that, compared with cognitively-intact subjects, the activity-dependent expression of ΔFosB in the hippocampal dentate gyrus (DG) was increased in individuals with mild cognitive impairment (MCI) and in individuals with AD. In MCI patients, higher ΔFosB expression corresponded to lower Mini-Mental State Examination scores. Surgically resected DG tissue from patients with temporal lobe epilepsy also showed robust ΔFosB expression; however, it is unclear whether ΔFosB expression also corresponds to cognitive dysfunction in non-AD-related epilepsy. To test whether DG ΔFosB expression is indicative of cognitive impairment in epilepsies with different etiologies, we assessed ΔFosB expression in surgically-resected hippocampal tissue from 33 patients with childhood epilepsies who had undergone Wechsler Intelligence Scale for Children (WISC) testing prior to surgery. We found that ΔFosB expression is inversely correlated with Full-Scale Intelligence Quotient (FSIQ) in patients with mild to severe intellectual disability (FSIQ < 85). Our data indicate that ΔFosB expression corresponds to cognitive impairment in epilepsies with different etiologies, supporting the hypothesis that ΔFosB may epigenetically regulate gene expression and impair cognition across a wide range of epilepsy syndromes.
Collapse
Affiliation(s)
- Chia-Hsuan Fu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Jason C. You
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Carrie Mohila
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, United States
| | - Robert A. Rissman
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, United States
- Veteran's Affairs (VA) San Diego Healthcare System, San Diego, CA, United States
| | - Daniel Yoshor
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - Angela N. Viaene
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
13
|
Kumari S, Brewster AL. Exploring Dendritic and Spine Structural Profiles in Epilepsy: Insights From Human Studies and Experimental Animal Models. Epilepsy Curr 2024; 24:40-46. [PMID: 38327540 PMCID: PMC10846509 DOI: 10.1177/15357597231218603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Dendrites are tree-like structures with tiny spines specialized to receive excitatory synaptic transmission. Spino-dendritic plasticity, driven by neural activity, underlies the maintenance of neuronal connections crucial for proper circuit function. Abnormalities in dendritic morphology are frequently seen in epilepsy. However, the exact etiology or functional implications are not yet known. Therefore, to better comprehend the structure-function significance of this dendritic pathology in epilepsy, it is necessary to identify the common spino-dendritic disturbances present in both human and experimental models. Here, we describe the dendritic and spine structural profiles found across human refractory epilepsy as well as in animal models of developmental, acquired, and genetic epilepsies.
Collapse
Affiliation(s)
- Shikha Kumari
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, USA
| | - Amy L. Brewster
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, USA
| |
Collapse
|
14
|
Ghatan S. Pediatric Neurostimulation and Practice Evolution. Neurosurg Clin N Am 2024; 35:1-15. [PMID: 38000833 DOI: 10.1016/j.nec.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Since the late nineteenth century, the prevailing view of epilepsy surgery has been to identify a seizure focus in a medically refractory patient and eradicate it. Sadly, only a select number of the many who suffer from uncontrolled seizures benefit from this approach. With the development of safe, efficient stereotactic methods and targeted surgical therapies that can affect deep structures and modulate broad networks in diverse disorders, epilepsy surgery in children has undergone a paradigmatic evolutionary change. With modern diagnostic techniques such as stereo electroencephalography combined with closed loop neuromodulatory systems, pediatric epilepsy surgery can reach a much broader population of underserved patients.
Collapse
Affiliation(s)
- Saadi Ghatan
- Neurological Surgery Icahn School of Medicine at Mt Sinai, New York, NY 10128, USA.
| |
Collapse
|
15
|
Rodrigo Marinowic D, Bottega Pazzin D, Prates da Cunha de Azevedo S, Pinzetta G, Victor Machado de Souza J, Tonon Schneider F, Thor Ramos Previato T, Jean Varella de Oliveira F, Costa Da Costa J. Epileptogenesis and drug-resistant in focal cortical dysplasias: Update on clinical, cellular, and molecular markers. Epilepsy Behav 2024; 150:109565. [PMID: 38070410 DOI: 10.1016/j.yebeh.2023.109565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/24/2023] [Accepted: 11/26/2023] [Indexed: 01/14/2024]
Abstract
Focal cortical dysplasia (FCD) is a cortical malformation in brain development and is considered as one of the major causes of drug-resistant epilepsiesin children and adults. The pathogenesis of FCD is yet to be fully understood. Imaging markers such as MRI are currently the surgeons major obstacle due to the difficulty in delimiting the precise dysplasic area and a mosaic brain where there is epileptogenic tissue invisible to MRI. Also increased gene expression and activity may be responsible for the alterations in cell proliferation, migration, growth, and survival. Altered expressions were found, particularly in the PI3K/AKT/mTOR pathway. Surgery is still considered the most effective treatment option, due to drug-resistance, and up to 60 % of patients experience complete seizure control, varying according to the type and location of FCD. Both genetic and epigenetic factors may be involved in the pathogenesis of FCD, and there is no conclusive evidence whether these alterations are inherited or have an environmental origin.
Collapse
Affiliation(s)
- Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Giulia Pinzetta
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Victor Machado de Souza
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Tonon Schneider
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fábio Jean Varella de Oliveira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaderson Costa Da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
16
|
Costa FV, Zabegalov KN, Kolesnikova TO, de Abreu MS, Kotova MM, Petersen EV, Kalueff AV. Experimental models of human cortical malformations: from mammals to 'acortical' zebrafish. Neurosci Biobehav Rev 2023; 155:105429. [PMID: 37863278 DOI: 10.1016/j.neubiorev.2023.105429] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Human neocortex controls and integrates cognition, emotions, perception and complex behaviors. Aberrant cortical development can be triggered by multiple genetic and environmental factors, causing cortical malformations. Animal models, especially rodents, are a valuable tool to probe molecular and physiological mechanisms of cortical malformations. Complementing rodent studies, the zebrafish (Danio rerio) is an important model organism in biomedicine. Although the zebrafish (like other fishes) lacks neocortex, here we argue that this species can still be used to model various aspects and brain phenomena related to human cortical malformations. We also discuss novel perspectives in this field, covering both advantages and limitations of using mammalian and zebrafish models in cortical malformation research. Summarizing mounting evidence, we also highlight the importance of translationally-relevant insights into the pathogenesis of cortical malformations from animal models, and discuss future strategies of research in the field.
Collapse
Affiliation(s)
- Fabiano V Costa
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Konstantin N Zabegalov
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Tatiana O Kolesnikova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Maria M Kotova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Allan V Kalueff
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; Ural Federal University, Yekaterinburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia.
| |
Collapse
|
17
|
Mohsin SN, Grezenko H, Khan S, Eshete FD, Shrestha S, Kamran M, Affaf M, Jama A, Gasim RW, Zubaer Ahmad D, Yadav I, Arif S, K C A, Khaliq AS. Bridging Development and Disruption: Comprehensive Insights into Focal Cortical Dysplasia and Epileptic Management. Cureus 2023; 15:e45996. [PMID: 37900524 PMCID: PMC10601976 DOI: 10.7759/cureus.45996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Focal cortical dysplasia (FCD) is a prominent neurological disorder characterized by disruptions in localized brain cell organization and development. This narrative review delineates the multi-faceted nature of FCD, emphasizing its correlation with drug-resistant epilepsy, predominantly in children and young adults. We explore the historical context of FCD, highlighting its indispensable role in shaping our comprehension of epilepsy and cortical anomalies. The clinical spectrum of FCD is broad, encompassing diverse seizure patterns, cognitive impairments, and associated neuropsychiatric disorders. We underscore the importance of differential diagnosis, with techniques ranging from electroencephalogram (EEG) interpretations to microscopic evaluations, and discuss advanced diagnostic modalities, such as the 3T magnetic resonance imaging (MRI) epilepsy protocols. Therapeutically, while anti-seizure medications are often first-line interventions, surgically refractory cases necessitate more invasive procedures, underscoring the importance of individualized treatment. Furthermore, the review touches upon the prognostic aspects of FCD, highlighting the importance of personalized care regimens, and provides insights into emerging therapeutic avenues, including the potential of the mammalian target of rapamycin (mTOR) pathway. Conclusively, this review accentuates the complex relationship between brain development and epileptogenicity inherent to FCD and underscores the promise of future research in enhancing patient outcomes.
Collapse
Affiliation(s)
| | - Han Grezenko
- Translational Neuroscience, Barrow Neurological Institute, Phoenix, USA
| | - Saadia Khan
- Community Medicine, Khyber Girls Medical College, Peshawar, PAK
| | | | - Shraddha Shrestha
- Internal Medicine, Nepal Korea Friendship Municipality Hospital, Bhaktapur, NPL
| | | | - Maryam Affaf
- Internal Medicine, Women's Medical and Dental College, Abbottabad, PAK
| | - Ayat Jama
- Internal Medicine, Nishtar Medical University, Multan, PAK
| | - Rayan W Gasim
- Internal Medicine, University of Khartoum, Khartoum, SDN
| | | | - Indresh Yadav
- Internal Medicine, Samar Hospital and Research Center Pvt. Ltd., Janakpur, NPL
- Internal Medicine, Community Based Medical College, Mymensingh, BGD
| | - Sidra Arif
- Urology, Jinnah Postgraduate Medical Center, Karachi, PAK
| | - Anil K C
- Medicine and Surgery, Patan Academy of Health Sciences, Kathmandu, NPL
- Internal Medicine and Neurology, California Institute of Behavioral Neurosciences & Psychology, California, USA
| | | |
Collapse
|
18
|
Guo M, Zhang J, Wang J, Wang X, Gao Q, Tang C, Deng J, Xiong Z, Kong X, Guan Y, Zhou J, Boison D, Luan G, Li T. Aberrant adenosine signaling in patients with focal cortical dysplasia. Mol Neurobiol 2023; 60:4396-4417. [PMID: 37103687 PMCID: PMC10330374 DOI: 10.1007/s12035-023-03351-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Focal cortical dysplasia (FCD), a common malformation of cortical development, is frequently associated with pharmacoresistant epilepsy in both children and adults. Adenosine is an inhibitory modulator of brain activity and a prospective anti-seizure agent with potential for clinical translation. Our previous results demonstrated that the major adenosine-metabolizing enzyme adenosine kinase (ADK) was upregulated in balloon cells (BCs) within FCD type IIB lesions, suggesting that dysfunction of the adenosine system is implicated in the pathophysiology of FCD. In our current study, we therefore performed a comprehensive analysis of adenosine signaling in surgically resected cortical specimens from patients with FCD type I and type II via immunohistochemistry and immunoblot analysis. Adenosine enzyme signaling was assessed by quantifying the levels of the key enzymes of adenosine metabolism, i.e., ADK, adenosine deaminase (ADA), and ecto-5'-nucleotidase (CD73). Adenosine receptor signaling was assessed by quantifying the levels of adenosine A2A receptor (A2AR) and putative downstream mediators of adenosine, namely, glutamate transporter-1 (GLT-1) and mammalian target of rapamycin (mTOR). Within lesions in FCD specimens, we found that the adenosine-metabolizing enzymes ADK and ADA, as well as the adenosine-producing enzyme CD73, were upregulated. We also observed an increase in A2AR density, as well as a decrease in GLT-1 levels and an increase in mTOR levels, in FCD specimens compared with control tissue. These results suggest that dysregulation of the adenosine system is a common pathologic feature of both FCD type I and type II. The adenosine system might therefore be a therapeutic target for the treatment of epilepsy associated with FCD.
Collapse
Affiliation(s)
- Mengyi Guo
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jing Zhang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jing Wang
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Xiongfei Wang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Qing Gao
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Chongyang Tang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jiahui Deng
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Zhonghua Xiong
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Xiangru Kong
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Yuguang Guan
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jian Zhou
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson & New Jersey Medical Schools, Rutgers University, Piscataway, NJ, 08854, USA
| | - Guoming Luan
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
| | - Tianfu Li
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
| |
Collapse
|
19
|
Takebayashi Y, Neshige S, Shishido T, Hayashi Y, Segawa A, Nakamori M, Nezu T, Aoki S, Yamazaki Y, Ueno H, Ohshita T, Iida K, Maruyama H. Paradoxical effects of levetiracetam in people with epilepsy with rhythmic epileptiform discharges. Epilepsy Behav 2023; 143:109225. [PMID: 37130461 DOI: 10.1016/j.yebeh.2023.109225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
OBJECTIVE To elucidate the incidence and risk factors for paradoxical effects (i.e., increased seizure frequency, increased seizure severity, or onset of new seizure types) of levetiracetam (LEV) in people with epilepsy (PWE) and identify the usefulness of electroencephalography (EEG) in predicting these effects. METHODS We examined data for consecutive PWE treated with LEV. All PWE underwent EEG and magnetic resonance imaging (MRI) before LEV administration. We also evaluated the incidence of paradoxical LEV effects and conducted multivariate logistic regression analyses to identify the associated factors. RESULTS In total, 210 (66.2%) of 317 PWEs treated in our department had a history of LEV use. The incidence of paradoxical LEV effects was 5.2% (n = 11) and was significantly associated with a high LEV dose (p = 0.029), high seizure frequency (p = 0.005), temporal lobe epilepsy (p = 0.004), focal awareness seizure (p = 0.004), focal impaired awareness seizure (p = 0.007), spike (p = 0.015), rhythmic epileptiform discharges (REDs; p = 0.003), and MRI-identified focal cortical dysplasia (FCD; p < 0.0001). Multivariate analyses revealed that REDs (odds ratio [OR] = 5.35, p = 0.048, 95% confidence interval [CI]: 1.01-28.21) were independently associated with paradoxical LEV effects. CONCLUSIONS Paradoxical LEV effects occurred in PWE, particularly in those with drug-resistant focal epilepsy. Furthermore, the occurrence of REDs in EEG was an independent factor associated with the paradoxical effects of LEV in PWE.
Collapse
Affiliation(s)
- Yoshiko Takebayashi
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Shuichiro Neshige
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan; Epilepsy Center, Hiroshima University Hospital, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan.
| | - Takeo Shishido
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Yuki Hayashi
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Akiko Segawa
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Masahiro Nakamori
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Tomohisa Nezu
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Shiro Aoki
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Yu Yamazaki
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Hiroki Ueno
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Tomohiko Ohshita
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Koji Iida
- Epilepsy Center, Hiroshima University Hospital, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan; Department of Neurosurgery, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan; Epilepsy Center, Hiroshima University Hospital, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| |
Collapse
|
20
|
Jadav D, Gupta V, Khera S, Meshram V. Focal Cortical Dysplasia with hippocampal sclerosis. AUTOPSY AND CASE REPORTS 2023; 13:e2023420. [PMID: 36741591 PMCID: PMC9886384 DOI: 10.4322/acr.2023.420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 01/02/2023] [Indexed: 02/07/2023] Open
Abstract
Focal Cortical Dysplasia (FCD) is a group of focal developmental malformations of the cerebral cortex cytoarchitecture. FCD usually manifests as medically intractable epilepsy, especially in young children. Live patients are diagnosed by radiological examination such as magnetic resonance imaging (MRI), fluorodeoxyglucose positron emission tomography (FDG PET), magnetoencephalography (MEG), diffusion-tensor imaging (DTI), and intracranial electroencephalogram (EEG). While some cases can be missed by radiological examination, they are usually diagnosed on the histopathological examination of the surgically removed specimens of medically intractable epilepsy patients. We report a case of a young girl with cerebral palsy, mental retardation, and seizure disorder who died in her sleep. The deceased was diagnosed with FCD type III with hippocampal sclerosis on histopathological examination at autopsy. H & E stain and NeuN immunohistochemistry neuronal cell marker were used to demonstrate the findings of FCD.
Collapse
Affiliation(s)
- Devendra Jadav
- All India Institute of Medical Sciences, Department of Forensic Medicine and Toxicology, Jodhpur, Rajasthan, India
| | - Vaibhav Gupta
- Vardhman Mahavir Medical College and Safdarjung Hospital, Department of Forensic Medicine and Toxicology, New Delhi, India
| | - Sudeep Khera
- All India Institute of Medical Sciences, Department of Pathology and Lab Medicine, Jodhpur, Rajasthan, India
| | - Vikas Meshram
- All India Institute of Medical Sciences, Department of Forensic Medicine and Toxicology, Jodhpur, Rajasthan, India
| |
Collapse
|
21
|
Sufianov A, Simfukwe K, Iakimov IA, Sufianov RA, Rassi MS, Mastronardi L, Borba LAB, Campero A, Rangel CC, Baldoncini M. Usefulness of Intraoperative ultrasound for cortical dysplasia type I treatment - A single-center experience. Surg Neurol Int 2023; 14:62. [PMID: 36895230 PMCID: PMC9990806 DOI: 10.25259/sni_926_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/13/2022] [Indexed: 03/11/2023] Open
Abstract
Background Focal cortical dysplasias (FCD) cause a subgroup of malformations of cortical development that has been closely linked to cause drug intractable epilepsy. Attaining adequate and safe resection of the dysplastic lesion has proved to be a viable option to archive meaningful seizure control. Of the three types of FCD (types I, II, and III), type I has the least detectable architectural and radiological abnormalities. This makes it challenging (preoperatively and intraoperatively) to achieve adequate resection. Intraoperatively, ultrasound navigation has proven an effective tool during the resection of these lesions. We evaluate our institutional experience in surgical management of FCD type I using intraoperative ultrasound (IoUS). Methods Our work is a retrospective and descriptive study, where we analyzed patients diagnosed with refractory epilepsy who underwent IoUS-guided epileptogenic tissue resection. The surgical cases analyzed were from January 2015 to June 2020 at the Federal Center of Neurosurgery, Tyumen, only patients with histological confirmation of postoperative CDF type I were included in the study. Results Of the 11 patients with histologically diagnosed FCD type I, 81.8% of the patients postoperatively had a significant reduction in seizure frequency (Engel outcome I-II). Conclusion IoUS is a critical tool for detecting and delineating FCD type I lesions, which is necessary for effective post-epilepsy surgery results.
Collapse
Affiliation(s)
- Albert Sufianov
- Department of Neurosurgery, Federal Center of Neurosurgery, Tyumen, Russian Federation
| | - Keith Simfukwe
- Department of Neurosurgery, First Moscow Medical University, Moscow, Russian Federation
| | - Iurii A Iakimov
- Department of Neurosurgery, First Moscow Medical University, Tyumen, Russian Federation
| | - Rinat A Sufianov
- Department of Neurosurgery, First Moscow Medical University, Moscow, Russian Federation
| | - Marcio S Rassi
- Department of Neurosurgery, Camargo Cancer Center, Sao Paulo, Brazil
| | | | - Luis A B Borba
- Department of Neurosurgery, Mackenzie Evangelical University Hospital, Curitiba, Brazil
| | - Alvaro Campero
- Department of Neurosurgery, Hospital Padilla de Tucuman, Tucuman, Mexico
| | - Carlos Castillo Rangel
- Department of Neurosurgery, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Mexico City, Mexico
| | - Matias Baldoncini
- Department of Neurosurgery, San Fernando Hospital, San Fernando, Buenos Aires, Argentina
| |
Collapse
|
22
|
Lee M, Kim EJ, Kim MJ, Yum MS. Rapamycin Cannot Reduce Seizure Susceptibility in Infantile Rats with Malformations of Cortical Development Lacking mTORC1 Activation. Mol Neurobiol 2022; 59:7439-7449. [PMID: 36194361 DOI: 10.1007/s12035-022-03033-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022]
Abstract
The mechanistic target of the rapamycin (mTOR) pathway is involved in cortical development. However, the efficacy of mTOR inhibitors in malformations of cortical dysplasia (MCD) outside of the tuberous sclerosis complex is unknown. We selected the MCD rat model with prenatal MAM exposure to test the efficacy of mTOR inhibitors in MCDs. We explored the early cortical changes of mTOR pathway protein expression in rats aged P15. We also monitored the early treatment effect of the mTOR inhibitor, rapamycin, on N-methyl-D-aspartate (NMDA)-induced spasms at P15 and their behavior in the juvenile stage. In vivo MR spectroscopy was performed after rapamycin treatment and compared with vehicle controls. There was no difference in mTORC1 pathway protein expression between MAM-exposed MCD rats and controls at P15, and prolonged treatment of rapamycin had no impact on NMDA-induced spasms despite poor weight gain. Prenatal MAM-exposed juvenile rats treated with rapamycin showed increased social approaching and freezing behavior during habituation. MR spectroscopy showed altered neurometabolites, including Gln, Glu+Gln, Tau, and Cr. Despite behavioral changes and in vivo neurometabolic alteration with early prolonged rapamycin treatment, rapamycin had no effect on spasms susceptibility in prenatal MAM-exposed infantile rats with MCD without mTORC1 activation. For MAM-exposed MCD rats without mTORC1 activation, treatment options outside of mTOR pathway inhibitors should be explored.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Eun-Jin Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Min-Jee Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea
| | - Mi-Sun Yum
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea. .,Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea.
| |
Collapse
|
23
|
Ellis EM, Trybula SJ, Adney SK, Lee PKJ, Bandt SK. Meningeal defects and focal cortical dysplasia: an unrecognized relationship? Illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2022; 4:CASE22112. [PMID: 36097744 PMCID: PMC9469904 DOI: 10.3171/case22112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/19/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND
Focal cortical dysplasias (FCDs) are a heterogenous cluster of histopathologic entities classically associated with medically refractory epilepsy. Because there is substantial histopathologic variation among different types of FCD, there are likely multiple pathogenic mechanisms leading to these disorders. The meninges are known to play a role in cortical development, and disruption of meningeal-derived signaling pathways has been shown to impact neurodevelopment. To our knowledge, there has not yet been an investigation into whether genetic pathways regulating meningeal development may be involved in the development of FCD.
OBSERVATIONS
The authors reported a patient with refractory epilepsy and evidence of FCD on imaging who received surgical intervention and was found to have an unusual dural anomaly overlying a region of type Ic FCD. To the authors’ knowledge, this was the first report describing a lesion of this nature in the context of FCD.
LESSONS
The dural anomaly exhibited by the patient presented what could be a potentially novel pathogenic mechanism of FCD. Resection of the cortical tissue underlying the dural anomaly resulted in improvement in seizure control. Although the pathogenesis is unclear, this case highlighted the importance of further investigation into the developmental origins of FCD, which may help elucidate whether a connection between meningeal development and FCD exists.
Collapse
Affiliation(s)
- Erin M. Ellis
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | | | - Scott K. Adney
- Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | - Paula K. J. Lee
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | |
Collapse
|
24
|
Broadband-NIRS System Identifies Epileptic Focus in a Child with Focal Cortical Dysplasia—A Case Study. Metabolites 2022; 12:metabo12030260. [PMID: 35323703 PMCID: PMC8951122 DOI: 10.3390/metabo12030260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/10/2022] Open
Abstract
Epileptic seizures are transiently occurring symptoms due to abnormal excessive or synchronous neuronal activity in the brain. Previous functional near-infrared spectroscopy (fNIRS) studies during seizures have focused in only monitoring the brain oxygenation and haemodynamic changes. However, few tools are available to measure actual cellular metabolism during seizures, especially at the bedside. Here we use an in-house developed multichannel broadband NIRS (or bNIRS) system, that, alongside the changes in oxy-, deoxy- haemoglobin concentration (HbO2, HHb), also quantifies the changes in oxidised cytochrome-c-oxidase Δ(oxCCO), a marker of cellular oxygen metabolism, simultaneously over 16 different brain locations. We used bNIRS to measure metabolic activity alongside brain tissue haemodynamics/oxygenation during 17 epileptic seizures at the bedside of a 3-year-old girl with seizures due to an extensive malformation of cortical development in the left posterior quadrant. Simultaneously Video-EEG data was recorded from 12 channels. Whilst we did observe the expected increase in brain tissue oxygenation (HbD) during seizures, it was almost diminished in the area of the focal cortical dysplasia. Furthermore, in the area of seizure origination (epileptic focus) ΔoxCCO decreased significantly at the time of seizure generalization when compared to the mean change in all other channels. We hypothesize that this indicates an incapacity to sustain and increase brain tissue metabolism during seizures in the region of the epileptic focus.
Collapse
|
25
|
Sandoval-Bonilla BA, Palmini A, Paglioli E, Monroy-Sosa A, De la Cerda-Vargas MF, Rodríguez-Hernández JJ, Chávez-Herrera VR, Perez-Reyes SP, Castro-Prado FC, Perez-Cardenas S, Sánchez-Dueñas JJ, Lagunes-Padilla LN. Extended resection for seizure control of pure motor strip focal cortical dysplasia during awake craniotomy: illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2022; 3:CASE21605. [PMID: 36130534 PMCID: PMC9379631 DOI: 10.3171/case21605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/03/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND
Focal cortical dysplasias (FCD) represent highly intrinsically epileptogenic lesions that require complete resection for seizure control. Resection of pure motor strip FCD can be challenging. Effective control of postoperative seizures is crucial and extending the boundaries of resection in an eloquent zone remains controversial.
OBSERVATIONS
The authors report a 52-year-old right-handed male with refractory epilepsy. The seizure phenotype was a focal crisis with preserved awareness and a clonic motor onset of right-hemibody. Epilepsy surgery protocol demonstrated a left pure motor strip FCD and a full-awake resective procedure with motor brain mapping was performed. Further resection of surgical boundaries monitoring function along intraoperative motor tasks with no direct electrical stimulation corroborated by intraoperative-neuromonitorization was completed as the final part of the surgery. In the follow-up period of 3-years, the patient has an Engel-IB seizure-control with mild distal lower limb palsy and no gate compromise.
LESSONS
This report represents one of the few cases with pure motor strip FCD resection. In a scenario similar to this case, the authors consider that this variation can be useful to improve seizure control and the quality of life of these patients by extending the resection of a more extensive epileptogenic zone minimizing functional damage.
Collapse
Affiliation(s)
- Bayron A. Sandoval-Bonilla
- Department of Neurosurgery, Epilepsy Surgery Program, Hospital de Especialidades, CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - André Palmini
- Department of Neurology and Neurosurgery, Epilepsy Surgery Program, Hospital São Lucas da Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Eliseu Paglioli
- Department of Neurology and Neurosurgery, Epilepsy Surgery Program, Hospital São Lucas da Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Alejandro Monroy-Sosa
- Department of Neurosurgery, Aurora Neuroscience Innovation Institute, Aurora St. Luke’s Medical Center, Milwaukee, Wisconsin
- Skull Base, Brain & Cerebrovascular Laboratory, Advocate Aurora Research Health Institute, Milwaukee, Wisconsin
| | - Maria F. De la Cerda-Vargas
- Department of Neurosurgery, Hospital de Especialidades No. 71, Instituto Mexicano del Seguro Social, Torreón Coahuila, Mexico
| | - Job J. Rodríguez-Hernández
- Department of Neurosurgery, Epilepsy Surgery Program, Hospital de Especialidades, CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Victor R. Chávez-Herrera
- Department of Neurosurgery, Epilepsy Surgery Program, Hospital de Especialidades, CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Sara P. Perez-Reyes
- Department of Neurosurgery, Hospital Regional de Alta Especialidad del Bajío, Instituto Mexicano del Seguro Social, Leon, Guanajuato, Mexico
| | - Fernando C. Castro-Prado
- Department of Neurosurgery, Epilepsy Surgery Program, Hospital de Especialidades, CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Department of Neurosurgery, High Specialties Regional Hospital Gral. I. Zaragoza, ISSSTE, Mexico City, Mexico
| | | | - Josafat J. Sánchez-Dueñas
- Department of Neurosurgery, Epilepsy Surgery Program, Hospital de Especialidades, CMN Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | |
Collapse
|
26
|
Practical Genetics for the Neuroradiologist: Adding Value in Neurogenetic Disease. Acad Radiol 2022; 29 Suppl 3:S1-S27. [PMID: 33495073 DOI: 10.1016/j.acra.2020.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/19/2020] [Accepted: 12/27/2020] [Indexed: 11/23/2022]
Abstract
Genetic discoveries have transformed our understanding of many neurologic diseases. Identification of specific causal pathogenic variants has improved understanding of pathophysiology and enabled replacement of many confusing eponyms and acronyms with more meaningful and clinically relevant genetics-based terminology. In this era of rapid scientific advancement, multidisciplinary collaboration among pediatricians, neurologists, geneticists, radiologists, and other members of the health care team is increasingly important in the care of patients with genetic neurologic diseases. Radiologists familiar with neurogenetic disease add value by (1) recognizing constellations of characteristic imaging findings that are associated with a genetic disease before one is clinically suspected; (2) predicting the most likely genotypes for a given imaging phenotype in clinically suspected genetic disease; and (3) providing detailed and accurate descriptions of the imaging phenotype in challenging cases with unknown or uncertain genotypes. This review aims to increase awareness and understanding of pathogenic variants relating to neurologic disease by (1) briefly reviewing foundational knowledge of chromosomes, inheritance patterns, and mutagenesis; (2) providing concrete examples of and detailed information about specific neurologic diseases resulting from pathogenic variants; and (3) highlighting clinical and imaging features that are of greatest relevance for the radiologist.
Collapse
|
27
|
Wang JY, Li Y, Lv YY, Jiang L. Screening and identification of novel candidate biomarkers of focal cortical dysplasia type II via bioinformatics analysis. Childs Nerv Syst 2022; 38:953-960. [PMID: 35112146 PMCID: PMC8809227 DOI: 10.1007/s00381-022-05454-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/14/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Focal cortical dysplasia (FCD) is the most common developmental malformation that causes refractory epilepsy. FCD II is a common neuropathological finding in tissues resected therapeutically from patients with drug-resistant epilepsy. However, its molecular genetic etiology remains unclear. This study aimed to identify potential molecular markers of FCD II using bioinformatics analysis. METHODS We downloaded two datasets for FCD II from the Gene Expression Omnibus data repository. Differentially expressed genes (DEGs) between FCD II and normal brain tissues were identified, and functional enrichment analysis was performed. A protein-protein interaction network was constructed, and hub genes were identified from the DEGs. The hub gene expression was validated using WB in vitro. IHC staining was performed to verify the feasibility of the target molecular markers identified in the bioinformatics analysis. RESULTS One hundred sixty-seven common DEGs were identified between the datasets. The GO and KEGG analyses showed that variations were prominently enriched in some functions associated with gene expression. Five hub genes (i.e., FANCI, FANCA, BRCA2, RAD18, and KEAP1) were identified. Western blotting confirmed that all hub gene expressions were higher in the FCD II tissue than in the normal brain tissue. IHC staining showed that the FANCI expression significantly increased in the FCD II tissue. CONCLUSION There are DEGs between FCD II and normal brain tissues, which may be considered biomarkers for FCD II, along with FANCI. The DEGs and hub genes identified in the bioinformatics analysis could serve as candidate targets for diagnosing and treating FCD II.
Collapse
Affiliation(s)
- Jiang-ya Wang
- Department of Pediatrics, Hebei Medical University, Shijiazhuang, China ,Department of Pediatrics, Hebei General Hospital, Shijiazhuang, China
| | - Yang Li
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Chang’an district, Shijiazhuang, 050000 China
| | - Yuan-yuan Lv
- Department of Pediatrics, Baoding First Central Hospital, Baoding, China
| | - Lian Jiang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Chang'an district, Shijiazhuang, 050000, China.
| |
Collapse
|
28
|
Aroor A, Brewster AL. Dendritic and Spine Loss in Epilepsy: What Seizures Got to Do With It? Epilepsy Curr 2021; 21:186-188. [PMID: 34867100 PMCID: PMC8609590 DOI: 10.1177/15357597211003096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
[Box: see text]
Collapse
|
29
|
Lee DA, Lee HJ, Kim HC, Park KM. Alterations of structural connectivity and structural co-variance network in focal cortical dysplasia. BMC Neurol 2021; 21:330. [PMID: 34452597 PMCID: PMC8394627 DOI: 10.1186/s12883-021-02358-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Background The aim of this study was to investigate alterations in structural connectivity and structural co-variance network in patients with focal cortical dysplasia (FCD). Methods We enrolled 37 patients with FCD and 35 healthy controls. All subjects underwent brain MRI with the same scanner and with the same protocol, which included diffusion tensor imaging (DTI) and T1-weighted imaging. We analyzed the structural connectivity based on DTI, and structural co-variance network based on the structural volume with T1-weighted imaging. We created a connectivity matrix and obtained network measures from the matrix using the graph theory. We tested the difference in network measure between patients with FCD and healthy controls. Results In the structural connectivity analysis, we found that the local efficiency in patients with FCD was significantly lower than in healthy controls (2.390 vs. 2.578, p = 0.031). Structural co-variance network analysis revealed that the mean clustering coefficient, global efficiency, local efficiency, and transitivity were significantly decreased in patients with FCD compared to those in healthy controls (0.527 vs. 0.635, p = 0.036; 0.545 vs. 0.648, p = 0.026; 2.699 vs. 3.801, p = 0.019; 0.791 vs. 0.954, p = 0.026, respectively). Conclusions We demonstrate that there are significant alterations in structural connectivity, based on DTI, and structural co-variance network, based on the structural volume, in patients with FCD compared to healthy controls. These findings suggest that focal lesions with FCD could affect the whole-brain network and that FCD is a network disease.
Collapse
Affiliation(s)
- Dong Ah Lee
- Neurology Department, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, 48108, Busan, Korea
| | - Ho-Joon Lee
- Radiology Department, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Hyung Chan Kim
- Neurology Department, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, 48108, Busan, Korea
| | - Kang Min Park
- Neurology Department, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, 48108, Busan, Korea.
| |
Collapse
|
30
|
Pracucci E, Pillai V, Lamers D, Parra R, Landi S. Neuroinflammation: A Signature or a Cause of Epilepsy? Int J Mol Sci 2021; 22:6981. [PMID: 34209535 PMCID: PMC8267969 DOI: 10.3390/ijms22136981] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/11/2021] [Accepted: 06/23/2021] [Indexed: 12/21/2022] Open
Abstract
Epilepsy can be both a primary pathology and a secondary effect of many neurological conditions. Many papers show that neuroinflammation is a product of epilepsy, and that in pathological conditions characterized by neuroinflammation, there is a higher probability to develop epilepsy. However, the bidirectional mechanism of the reciprocal interaction between epilepsy and neuroinflammation remains to be fully understood. Here, we attempt to explore and discuss the relationship between epilepsy and inflammation in some paradigmatic neurological and systemic disorders associated with epilepsy. In particular, we have chosen one representative form of epilepsy for each one of its actual known etiologies. A better understanding of the mechanistic link between neuroinflammation and epilepsy would be important to improve subject-based therapies, both for prophylaxis and for the treatment of epilepsy.
Collapse
Affiliation(s)
- Enrico Pracucci
- National Enterprise for Nanoscience and Nanotechnology (NEST), Istituto Nanoscienze Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy; (E.P.); (V.P.); (D.L.); (R.P.)
| | - Vinoshene Pillai
- National Enterprise for Nanoscience and Nanotechnology (NEST), Istituto Nanoscienze Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy; (E.P.); (V.P.); (D.L.); (R.P.)
| | - Didi Lamers
- National Enterprise for Nanoscience and Nanotechnology (NEST), Istituto Nanoscienze Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy; (E.P.); (V.P.); (D.L.); (R.P.)
| | - Riccardo Parra
- National Enterprise for Nanoscience and Nanotechnology (NEST), Istituto Nanoscienze Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy; (E.P.); (V.P.); (D.L.); (R.P.)
| | - Silvia Landi
- National Enterprise for Nanoscience and Nanotechnology (NEST), Istituto Nanoscienze Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy; (E.P.); (V.P.); (D.L.); (R.P.)
- Institute of Neuroscience CNR, 56127 Pisa, Italy
| |
Collapse
|
31
|
Seizure outcome after epilepsy surgery in tuberous sclerosis complex: Results and analysis of predictors from a multicenter study. J Neurol Sci 2021; 427:117506. [PMID: 34087568 DOI: 10.1016/j.jns.2021.117506] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/22/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022]
Abstract
Epilepsy surgery is recommended in selected patients with Tuberous Sclerosis Complex (TSC). However, reports on predictive factors of seizure outcome are variable. Here we report on seizure and cognitive outcome of 35 TSC patients who received surgery for refractory epilepsy in 7 Italian centers over a period of 22 years (1997-2019). The rate of seizure-free individuals at last follow-up (mean 7.5 years, range 1-21 years) was 51%. Patients with longer follow-up (≥10 years) had a lower rate of Engel I outcome (11.1%) than those who received surgery in the last 10 years (65.4%, p = 0.003). Factors associated with Engel II, III, IV outcome in our cohort included: high number of cortical tubers (≥5); presence of subependymal nodules (SENs); seizure onset before age 1 year; and multifocal interictal epileptic discharges (IEDs) on electroencephalogram (EEG). A subset of patients evaluated with Vineland Adaptive Behaviour Scales (VABS) showed developmental gains, in line with their developmental trajectories, but no improvement in standard scores after surgery was noted. Our study demonstrates that the rates of successful seizure outcome of epilepsy surgery in TSC have improved in the last 10 years. More than half of the patients achieved seizure freedom, and a high proportion of affected individuals experienced a reduction in seizure burden and in antiseizure medications. A comprehensive assessment after surgery should be performed in TSC patients to evaluate the overall neurodevelopmental outcome, as measures that are based only on seizure control do not adequately identify the benefits of surgery on global functioning in these patients.
Collapse
|
32
|
Wang Z, Huang K, Yang X, Shen K, Yang L, Ruan R, Shi X, Wang M, Zhu G, Yang M, Zhang C, Lv S, Yang H, Fan X, Liu S. Downregulated GPR30 expression in the epileptogenic foci of female patients with focal cortical dysplasia type IIb and tuberous sclerosis complex is correlated with 18 F-FDG PET-CT values. Brain Pathol 2021; 31:346-364. [PMID: 33314369 PMCID: PMC8018162 DOI: 10.1111/bpa.12925] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/20/2020] [Accepted: 12/07/2020] [Indexed: 01/19/2023] Open
Abstract
Focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC) are typical causes of developmental delay and refractory epilepsy. G‐protein‐coupled receptor 30 (GPR30) is a specific estrogen receptor that is critical in neurodevelopment, neuroinflammation, and neuronal excitability, suggesting that it plays a potential role in the epilepsy of patients with FCDIIb and TSC. Therefore, we investigated the role of GPR30 in patients with FCDIIb and TSC. We found that the expression of GPR30 and its downstream protein kinase A (PKA) pathway were decreased and negatively correlated with seizure frequency in female patients with FCDIIb and TSC, but not in male patients. GPR30 was widely distributed in neurons, astrocytes, and microglia, and its downregulation was especially notable in microglia. The GPR30 agonist G‐1 increased the expression of PKA and p‐PKA in cultured cortical neurons, and the GPR30 antagonist G‐15 exhibited the opposite effects of G‐1. The NF‐κB signaling pathway was also activated in the specimens of female patients with FCDIIb and TSC, and was regulated by G‐1 and G‐15 in cultured cortical neurons. We also found that GPR30 regulated cortical neuronal excitability by altering the frequency of spontaneous excitatory postsynaptic currents and the expression of NR2A/B. Further, the relationship between GPR30 and glycometabolism was evaluated by analyzing the correlations between GPR30 and 18F‐FDG PET‐CT values (standardized uptake values, SUVs). Positive correlations between GPR30 and SUVs were found in female patients, but not in male patients. Intriguingly, GPR30 expression and SUVs were significantly decreased in the epileptogenic tubers of female TSC patients, and ROC curves indicated that SUVs could predict the localization of epileptogenic tubers. Taken together, our results suggest a potential protective effect of GPR30 in the epileptogenesis of female patients with FCDIIb and TSC.
Collapse
Affiliation(s)
- Zhongke Wang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kaixuan Huang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaolin Yang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kaifeng Shen
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ruotong Ruan
- Department of Basic Medical College, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianjun Shi
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Miao Wang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Gang Zhu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Meihua Yang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chunqing Zhang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shengqing Lv
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hui Yang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shiyong Liu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
33
|
Hsieh LS, Wen JH, Nguyen LH, Zhang L, Getz S, Torres-Reveron J, Wang Y, Spencer DD, Bordey A. Ectopic HCN4 expression drives mTOR-dependent epilepsy in mice. Sci Transl Med 2020; 12:12/570/eabc1492. [PMID: 33208499 PMCID: PMC9888000 DOI: 10.1126/scitranslmed.abc1492] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/31/2020] [Indexed: 02/03/2023]
Abstract
The causative link between focal cortical malformations (FCMs) and epilepsy is well accepted, especially among patients with focal cortical dysplasia type II (FCDII) and tuberous sclerosis complex (TSC). However, the mechanisms underlying seizures remain unclear. Using a mouse model of TSC- and FCDII-associated FCM, we showed that FCM neurons were responsible for seizure activity via their unexpected abnormal expression of the hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4 (HCN4), which is normally not present in cortical pyramidal neurons after birth. Increasing intracellular cAMP concentrations, which preferentially affects HCN4 gating relative to the other isoforms, drove repetitive firing of FCM neurons but not control pyramidal neurons. Ectopic HCN4 expression was dependent on the mechanistic target of rapamycin (mTOR), preceded the onset of seizures, and was also found in diseased neurons in tissue resected from patients with TSC and FCDII. Last, blocking HCN4 channel activity in FCM neurons prevented epilepsy in the mouse model. These findings suggest that HCN4 play a main role in seizure and identify a cAMP-dependent seizure mechanism in TSC and FCDII. Furthermore, the unique expression of HCN4 exclusively in FCM neurons suggests that gene therapy targeting HCN4 might be effective in reducing seizures in FCDII or TSC.
Collapse
Affiliation(s)
- Lawrence S. Hsieh
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - John H. Wen
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Lena H. Nguyen
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Longbo Zhang
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Stephanie Getz
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Juan Torres-Reveron
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Ying Wang
- Emergency Department, Xiangya Hospital, Central South University, 87 Xiangya Street, Changsha, Hunan 410008, China
| | - Dennis D. Spencer
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Angélique Bordey
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA,Department of Cellular & Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA,To whom correspondence should be addressed: Angélique Bordey, Ph.D., Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, FMB 422, New Haven, CT 06520-8082, Phone: 203-737-2515, Fax: 203-737-2159,
| |
Collapse
|
34
|
Klofas LK, Short BP, Snow JP, Sinnaeve J, Rushing GV, Westlake G, Weinstein W, Ihrie RA, Ess KC, Carson RP. DEPDC5 haploinsufficiency drives increased mTORC1 signaling and abnormal morphology in human iPSC-derived cortical neurons. Neurobiol Dis 2020; 143:104975. [PMID: 32574724 PMCID: PMC7462127 DOI: 10.1016/j.nbd.2020.104975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/21/2020] [Accepted: 06/13/2020] [Indexed: 01/21/2023] Open
Abstract
Mutations in the DEPDC5 gene can cause epilepsy, including forms with and without brain malformations. The goal of this study was to investigate the contribution of DEPDC5 gene dosage to the underlying neuropathology of DEPDC5-related epilepsies. We generated induced pluripotent stem cells (iPSCs) from epilepsy patients harboring heterozygous loss of function mutations in DEPDC5. Patient iPSCs displayed increases in both phosphorylation of ribosomal protein S6 and proliferation rate, consistent with elevated mTORC1 activation. In line with these findings, we observed increased soma size in patient iPSC-derived cortical neurons that was rescued with rapamycin treatment. These data indicate that human cells heterozygous for DEPDC5 loss-of-function mutations are haploinsufficient for control of mTORC1 signaling. Our findings suggest that human pathology differs from mouse models of DEPDC5-related epilepsies, which do not show consistent phenotypic differences in heterozygous neurons, and support the need for human-based models to affirm and augment the findings from animal models of DEPDC5-related epilepsy.
Collapse
Affiliation(s)
- Lindsay K Klofas
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Brittany P Short
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John P Snow
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Justine Sinnaeve
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Grant Westlake
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Will Weinstein
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca A Ihrie
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kevin C Ess
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert P Carson
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
35
|
Chen SD, Pan HY, Huang JB, Liu XP, Li JH, Ho CJ, Tsai MH, Yang JL, Chen SF, Chen NC, Chuang YC. Circulating MicroRNAs from Serum Exosomes May Serve as a Putative Biomarker in the Diagnosis and Treatment of Patients with Focal Cortical Dysplasia. Cells 2020; 9:cells9081867. [PMID: 32785072 PMCID: PMC7465068 DOI: 10.3390/cells9081867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Focal cortical dysplasia (FCD) is a congenital malformation of cortical development where the cortical neurons located in the brain area fail to migrate in the proper formation. Epilepsy, particularly medically refractory epilepsy, is the most common clinical presentation for all types of FCD. This study aimed to explore the expression change of circulating miRNAs in patients with FCD from serum exosomes. A total of nine patients with FCD and four healthy volunteers were enrolled in this study. The serum exosomes were isolated from the peripheral blood of the subjects. Transmission electron microscopy (TEM) was used to identify the exosomes. Both exosomal markers and neuronal markers were detected by Western blotting analysis to prove that we could obtain central nervous system-derived exosomes from the circulation. The expression profiles of circulating exosomal miRNAs were assessed using next-generation sequencing analysis (NGS). We obtained a total of 107 miRNAs with dominant fold change (>2-fold) from both the annotated 5p-arm and 3p-arm of 2780 mature miRNAs. Based on the integrated platform of HMDD v3.2, miRway DB and DIANA-miRPath v3.0 online tools, and confirmed by MiRBase analysis, four potentially predicted miRNAs from serum exosomes in patients with FCD were identified, including miR194-2-5p, miR15a-5p, miR-132-3p, and miR-145-5p. All four miRNAs presented upregulated expression in patients with FCD compared with controls. Through Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and pathway category of four target miRNAs, we found eight possible signaling pathways that may be related to FCD. Among them, we suggest that the mTOR signaling pathway, PI3K-Akt signaling pathway, p53 signaling pathway, and cell cycle regulation and TGF-beta signaling pathway are high-risk pathways that play a crucial role in the pathogenesis of FCD and refractory epilepsy. Our results suggest that the circulating miRNAs from exosomes may provide a potential biomarker for diagnostic, prognostic, and therapeutic adjuncts in patients with FCD and refractory epilepsy.
Collapse
Affiliation(s)
- Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.); (J.-L.Y.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiu-Yung Pan
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (H.-Y.P.); (J.-B.H.)
| | - Jyun-Bin Huang
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (H.-Y.P.); (J.-B.H.)
| | - Xuan-Ping Liu
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.); (J.-L.Y.)
| | - Jie-Hau Li
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.); (J.-L.Y.)
| | - Chen-Jui Ho
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Jenq-Lin Yang
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.); (J.-L.Y.)
| | - Shu-Fang Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
| | - Nai-Ching Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
| | - Yao-Chung Chuang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (S.-D.C.); (C.-J.H.); (M.-H.T.); (S.-F.C.); (N.-C.C.)
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.); (J.-L.Y.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Correspondence:
| |
Collapse
|
36
|
Feliciano DM. The Neurodevelopmental Pathogenesis of Tuberous Sclerosis Complex (TSC). Front Neuroanat 2020; 14:39. [PMID: 32765227 PMCID: PMC7381175 DOI: 10.3389/fnana.2020.00039] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a model disorder for understanding brain development because the genes that cause TSC are known, many downstream molecular pathways have been identified, and the resulting perturbations of cellular events are established. TSC, therefore, provides an intellectual framework to understand the molecular and biochemical pathways that orchestrate normal brain development. The TSC1 and TSC2 genes encode Hamartin and Tuberin which form a GTPase activating protein (GAP) complex. Inactivating mutations in TSC genes (TSC1/TSC2) cause sustained Ras homologue enriched in brain (RHEB) activation of the mammalian isoform of the target of rapamycin complex 1 (mTORC1). TOR is a protein kinase that regulates cell size in many organisms throughout nature. mTORC1 inhibits catabolic processes including autophagy and activates anabolic processes including mRNA translation. mTORC1 regulation is achieved through two main upstream mechanisms. The first mechanism is regulation by growth factor signaling. The second mechanism is regulation by amino acids. Gene mutations that cause too much or too little mTORC1 activity lead to a spectrum of neuroanatomical changes ranging from altered brain size (micro and macrocephaly) to cortical malformations to Type I neoplasias. Because somatic mutations often underlie these changes, the timing, and location of mutation results in focal brain malformations. These mutations, therefore, provide gain-of-function and loss-of-function changes that are a powerful tool to assess the events that have gone awry during development and to determine their functional physiological consequences. Knowledge about the TSC-mTORC1 pathway has allowed scientists to predict which upstream and downstream mutations should cause commensurate neuroanatomical changes. Indeed, many of these predictions have now been clinically validated. A description of clinical imaging and histochemical findings is provided in relation to laboratory models of TSC that will allow the reader to appreciate how human pathology can provide an understanding of the fundamental mechanisms of development.
Collapse
Affiliation(s)
- David M Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
| |
Collapse
|
37
|
Re CJ, Batterman AI, Gerstner JR, Buono RJ, Ferraro TN. The Molecular Genetic Interaction Between Circadian Rhythms and Susceptibility to Seizures and Epilepsy. Front Neurol 2020; 11:520. [PMID: 32714261 PMCID: PMC7344275 DOI: 10.3389/fneur.2020.00520] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022] Open
Abstract
Seizure patterns observed in patients with epilepsy suggest that circadian rhythms and sleep/wake mechanisms play some role in the disease. This review addresses key topics in the relationship between circadian rhythms and seizures in epilepsy. We present basic information on circadian biology, but focus on research studying the influence of both the time of day and the sleep/wake cycle as independent but related factors on the expression of seizures in epilepsy. We review studies investigating how seizures and epilepsy disrupt expression of core clock genes, and how disruption of clock mechanisms impacts seizures and the development of epilepsy. We focus on the overlap between mechanisms of circadian-associated changes in SCN neuronal excitability and mechanisms of epileptogenesis as a means of identifying key pathways and molecules that could represent new targets or strategies for epilepsy therapy. Finally, we review the concept of chronotherapy and provide a perspective regarding its application to patients with epilepsy based on their individual characteristics (i.e., being a “morning person” or a “night owl”). We conclude that better understanding of the relationship between circadian rhythms, neuronal excitability, and seizures will allow both the identification of new therapeutic targets for treating epilepsy as well as more effective treatment regimens using currently available pharmacological and non-pharmacological strategies.
Collapse
Affiliation(s)
- Christopher J Re
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Alexander I Batterman
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Jason R Gerstner
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Russell J Buono
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
38
|
Han P, Welsh CT, Smith MT, Schmidt RE, Carroll SL. Complex Patterns of GABAergic Neuronal Deficiency and Type 2 Potassium-Chloride Cotransporter Immaturity in Human Focal Cortical Dysplasia. J Neuropathol Exp Neurol 2020; 78:365-372. [PMID: 30856249 DOI: 10.1093/jnen/nlz009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Focal cortical dysplasia (FCD) is a common histopathologic finding in cortical specimens resected for refractory epilepsy. GABAergic neuronal abnormalities and K-Cl cotransporter type 2 (KCC2) immaturity may be contributing factors for FCD-related epilepsy. We examined surgical specimens from 12 cases diagnosed with FCD, and brain tissues without developmental abnormality obtained from 6 autopsy cases. We found that GABAergic neuronal density was abnormal in FCD with 2 distinct patterns. In 7 of 12 (58%) FCD subjects, the GABAergic neuron density in dysplastic regions and in neighboring nondysplastic regions was equally reduced, hence we call this a "broad pattern." In the remaining cases, GABAergic neuron density was decreased in dysplastic regions but not in the neighboring nondysplastic regions; we designate this "restricted pattern." The different patterns are not associated with pathologic subtypes of FCD. Intracytoplasmic retention of KCC2 is evident in dysmorphic neurons in the majority of FCD type II subjects (5/7) but not in FCD type I. Our study suggests that (1) "broad" GABAergic deficiency may reflect epileptic vulnerability outside the dysplastic area; and (2) abnormal distribution of KCC2 may contribute to seizure generation in patients with FCD type II but not in type I.
Collapse
Affiliation(s)
- Pengcheng Han
- Department of Pathology and Laboratory Medicine.,Department of Pathology and Laboratory Medicine Residency Program, Medical University of South Carolina, Charleston, South Carolina
| | | | | | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | | |
Collapse
|
39
|
Han G, Zhang J, Ma Y, Gui Q, Yin S. Clinical characteristics, treatment and prognosis of angiocentric glioma. Oncol Lett 2020; 20:1641-1648. [PMID: 32724405 PMCID: PMC7377082 DOI: 10.3892/ol.2020.11723] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 05/18/2020] [Indexed: 01/21/2023] Open
Abstract
Angiocentric glioma (AG) is a rare subtype of neuroepithelial tumor in children and young adults that commonly presents with seizures. To study the clinical characteristics, treatment and prognosis of patients with AG, the features of two cases of AG were described and 108 cases reported in the literature were assessed. The cases of the present study were two males aged 8 and 16 years, who mainly presented with seizures. MRI revealed superficial, non-enhanced lesions in the left temporal and right frontal lobe, respectively. The two patients underwent gross total resection (GTR) and remained seizure-free without neurological deficits after 3.5 and 2.5 years, respectively. Histopathological examination revealed that the tumors consisted of monomorphous cells that surrounded the blood vessels and neurons in the cerebral cortex, and formed concentric sleeves or pseudorosettes. Furthermore, immunostaining indicated that the diffuse infiltrative neoplastic cells were positive for glial fibrillary acidic protein and a dot-like pattern of epithelial membrane antigen was observed. AG mostly appeared similar to low-grade gliomas on MRI. GTR of the lesions was curative and radiation or chemotherapy were not required. AG typically has a favorable prognosis, with low mortality and incidence of disability.
Collapse
Affiliation(s)
- Guoqing Han
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Junsi Zhang
- Department of Neurology, Tianjin Children's Hospital, Tianjin 300074, P.R. China
| | - Yue Ma
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Qiuping Gui
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Shi Yin
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
40
|
Jackson HN, Gadgil N, Pan IW, Clarke DF, Wagner KM, Cronkite CA, Lam S. Sociodemographic Factors in Pediatric Epilepsy Surgery. Pediatr Neurol 2020; 107:71-76. [PMID: 32284204 DOI: 10.1016/j.pediatrneurol.2019.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/24/2019] [Accepted: 09/02/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Despite documented efficacy of surgical treatment in carefully selected patients, surgery is delayed and/or underutilized in both adult and children with focal onset epilepsy. The reasons for surgical delay are often assumed or theorized, and studies have predominantly targeted the adult population. To focus on a more targeted pediatric population and to determine identifiable reasons for intervention, this study aimed to investigate time to epilepsy surgery among pediatric patients with medically intractable epilepsy associated with focal cortical dysplasia and to identify sociodemographic and clinical associations in time to epilepsy surgery. METHODS We reviewed 96 consecutive pediatric patients who underwent surgery for medically intractable epilepsy with a diagnosis of focal cortical dysplasia. Descriptive statistics, univariate and multivariate analyses were conducted to study the association of sociodemographic variables of patients with focal cortical dysplasia and time to epilepsy surgery and postoperative seizure control. RESULTS We identified that non-white patients on average had a longer duration of epilepsy before surgery and traveled shorter distances for care. Non-white patients were more likely to have government-funded insurance. Patients who traveled the shortest distance to the surgical center underwent epilepsy surgery at an older age. CONCLUSIONS Sociodemographic factors of travel distance, insurance, and race influenced time to epilepsy surgery for children with focal cortical dysplasia. Further research is warranted to target barriers in access to subspecialty care and develop ways to identify earlier the patients who may benefit from evaluation and deployment of surgical intervention.
Collapse
Affiliation(s)
- Hudin N Jackson
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Nisha Gadgil
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - I-Wen Pan
- Division of Cancer Prevention and Population Sciences, Department of Health Services Research, MD Anderson Cancer Center, Houston, Texas
| | - Dave F Clarke
- Division of Pediatric Neurology, Department of Neurology, Dell Medical School University of Texas at Austin, Austin, Texas
| | - Kathryn M Wagner
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | | | - Sandi Lam
- Division of Pediatric Neurosurgery, Ann and Robert H Lurie Children's Hospital, Chicago, Illinois; Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
41
|
|
42
|
Bonilha L, Small SS, Lin JJ. Editorial for the special issue on language and epilepsy. BRAIN AND LANGUAGE 2019; 193:1-3. [PMID: 30929763 DOI: 10.1016/j.bandl.2019.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Affiliation(s)
- Leonardo Bonilha
- Department of Neurology, Medical University of South Carolina, Charleston, SC, United States.
| | - Steven S Small
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Jack J Lin
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, United States; Department of Biomedical Engineering, The Henry Samueli School of Engineering, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
43
|
Abstract
Disorders of the developing nervous system may be of genetic origin, comprising congenital malformations of spine and brain as well as metabolic or vascular disorders that affect normal brain development. Acquired causes include congenital infections, hypoxic-ischemic or traumatic brain injury, and a number of rare neoplasms. This chapter focuses on the clinical presentation and workup of neurogenetic disorders presenting in the fetal or neonatal period. After a summary of the most frequent clinical presentations, clues from history taking and clinical examination are illustrated with short case reports. This is followed by a discussion of the different tools available for the workup of neurogenetic disorders, including the various genetic techniques with their advantages and disadvantages. The implications of a molecular genetic diagnosis for the patient and family are addressed in the section on counseling. The chapter concludes with a proposed workflow that may help the clinician when confronted with a potential neurogenetic disorder in the fetal or neonatal period.
Collapse
|
44
|
San-Juan D, Sarmiento CI, González KM, Orenday Barraza JM. Successful Treatment of a Drug-Resistant Epilepsy by Long-term Transcranial Direct Current Stimulation: A Case Report. Front Neurol 2018; 9:65. [PMID: 29479337 PMCID: PMC5811469 DOI: 10.3389/fneur.2018.00065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 01/25/2018] [Indexed: 12/03/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) is a reemerged noninvasive cerebral therapy used to treat patients with epilepsy, including focal cortical dysplasia, with controversial results. We present a case of a 28-year-old female with left frontal cortical dysplasia refractory to antiepileptic drugs, characterized by 10–15 daily right tonic hemi-body seizures. The patient received a total of seven sessions of cathodal tDCS (2 mA, 30 min). The first three sessions were applied over three consecutive days, and the remaining four sessions of tDCS were given each at 2-week intervals. At the 1-year follow-up, the patient reported to have a single seizure per month and only mild adverse events.
Collapse
Affiliation(s)
- Daniel San-Juan
- Department of Clinical Research, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Carlos Ignacio Sarmiento
- Department of Basic Sciences and Engineering, Autonomous Metropolitan University Campus Iztapalapa, Mexico City, Mexico
| | | | | |
Collapse
|
45
|
Shrot S, Hwang M, Stafstrom CE, Huisman TAGM, Soares BP. Dysplasia and overgrowth: magnetic resonance imaging of pediatric brain abnormalities secondary to alterations in the mechanistic target of rapamycin pathway. Neuroradiology 2017; 60:137-150. [PMID: 29279945 DOI: 10.1007/s00234-017-1961-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/07/2017] [Indexed: 12/22/2022]
Abstract
The current classification of malformations of cortical development is based on the type of disrupted embryological process (cell proliferation, migration, or cortical organization/post-migrational development) and the resulting morphological anomalous pattern of findings. An ideal classification would include knowledge of biological pathways. It has recently been demonstrated that alterations affecting the mechanistic target of rapamycin (mTOR) signaling pathway result in diverse abnormalities such as dysplastic megalencephaly, hemimegalencephaly, ganglioglioma, dysplastic cerebellar gangliocytoma, focal cortical dysplasia type IIb, and brain lesions associated with tuberous sclerosis. We review the neuroimaging findings in brain abnormalities related to alterations in the mTOR pathway, following the emerging trend from morphology towards genetics in the classification of malformations of cortical development. This approach improves the understanding of anomalous brain development and allows precise diagnosis and potentially targeted therapies that may regulate mTOR pathway function.
Collapse
Affiliation(s)
- Shai Shrot
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 1800 Orleans Street, Zayed 4174, Baltimore, MD, 21287, USA
- Department of Diagnostic Imaging, Sheba Medical Center, 52621, Ramat-Gan, Israel
| | - Misun Hwang
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 1800 Orleans Street, Zayed 4174, Baltimore, MD, 21287, USA
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Thierry A G M Huisman
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 1800 Orleans Street, Zayed 4174, Baltimore, MD, 21287, USA
| | - Bruno P Soares
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 1800 Orleans Street, Zayed 4174, Baltimore, MD, 21287, USA.
| |
Collapse
|
46
|
Choi SA, Kim SY, Kim H, Kim WJ, Kim H, Hwang H, Choi JE, Lim BC, Chae JH, Chong S, Lee JY, Phi JH, Kim SK, Wang KC, Kim KJ. Surgical outcome and predictive factors of epilepsy surgery in pediatric isolated focal cortical dysplasia. Epilepsy Res 2017; 139:54-59. [PMID: 29197666 DOI: 10.1016/j.eplepsyres.2017.11.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/26/2017] [Accepted: 11/24/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Focal cortical dysplasia (FCD) is a common cause of medically intractable epilepsy in children. Epilepsy surgery has been a valuable treatment option to achieve seizure freedom in these intractable epilepsy patients. We aimed to present long-term surgical outcome, in relation to pathological severity, and to assess predictive factors of epilepsy surgery in pediatric isolated FCD. METHODS We retrospectively analyzed the data of 58 children and adolescents, with FCD International League Against Epilepsy (ILAE) task force classification types I and II, who underwent resective epilepsy surgery and were followed for at least 2 years after surgery. RESULTS The mean age at epilepsy onset was 4.3 years (0-14.2 years), and mean age at epilepsy surgery was 9.4 years (0.4-17.5 years). The mean duration of postoperative follow-up was 5.1±2.6 years (2-12.4 years). Of 58 patients, 62% of patients achieved Engel class I at 2 years postoperatively, 58% at 5 years postoperatively, and 53% at the last follow up. Forty eight percent of our cohort successfully discontinued antiepileptic medication. Of 30 patients with seizure recurrence, 83% of seizures recurred within 2 years after surgery. We observed that FCD type IIb was significantly associated with a better surgical outcome. At fifth postoperative year, 88% of FCD IIb patients were seizure free compared with 21% of type I and 57% of type IIa patients (P=0.043). By multivariate analysis, lesion on MRI (P=0.02) and complete resection (P<0.01) were the most important predictive factors for a seizure-free outcome. SIGNIFICANCE Epilepsy surgery is highly effective; more than half of medically intractable epilepsy patients achieved seizure freedom after surgery. In addition, we found significant difference in surgical outcomes according to the ILAE task force classification. Lesion on MRI and complete resection were the most important predictive factors for favorable seizure outcome in isolated FCD patients.
Collapse
Affiliation(s)
- Sun Ah Choi
- Department of Pediatrics, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuna Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Joong Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hunmin Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Hee Hwang
- Department of Pediatrics, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Ji Eun Choi
- Department of Pediatrics, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Byung Chan Lim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sangjoon Chong
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea; Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Hoon Phi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Ki Joong Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Abstract
Epilepsy is one of the most common neurologic disorders, affecting about 50 million people worldwide. The disease is characterized by recurrent seizures, which are due to aberrant neuronal networks resulting in synchronous discharges. The term epilepsy encompasses a large spectrum of syndromes and diseases with different etiopathogenesis. The recent development of imaging and epilepsy surgery techniques is now enabling the identification of structural abnormalities that are part of the epileptic network, and the removal of these lesions may result in control of seizures. Access of this clinically well-characterized neurosurgical material has provided neuropathologists with the opportunity to study a variety of structural brain abnormalities associated with epilepsy, by combining traditional routine histopathologic methods with molecular genetics and functional analysis of the resected tissue. This approach has contributed greatly to a better diagnosis and classification of these structural lesions, and has provided important new insights into their pathogenesis and epileptogenesis. The present chapter provides a detailed description of the large spectrum of histopathologic findings encountered in epilepsy surgery patients, addressing in particular the nonneoplastic pathologies, including hippocampal sclerosis, malformations of cortical development, Sturge-Weber syndrome, and Rasmussen encephalitis, and reviews current knowledge regarding the underlying molecular pathomechanisms and cellular mechanisms mediating hyperexcitability.
Collapse
Affiliation(s)
- Eleonora Aronica
- Department of Neuropathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland, the Netherlands.
| | - Angelika Mühlebner
- Department of Neuropathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
48
|
Williams AJ, Zhou C, Sun QQ. Enhanced Burst-Suppression and Disruption of Local Field Potential Synchrony in a Mouse Model of Focal Cortical Dysplasia Exhibiting Spike-Wave Seizures. Front Neural Circuits 2016; 10:93. [PMID: 27891080 PMCID: PMC5102891 DOI: 10.3389/fncir.2016.00093] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/31/2016] [Indexed: 11/28/2022] Open
Abstract
Focal cortical dysplasias (FCDs) are a common cause of brain seizures and are often associated with intractable epilepsy. Here we evaluated aberrant brain neurophysiology in an in vivo mouse model of FCD induced by neonatal freeze lesions (FLs) to the right cortical hemisphere (near S1). Linear multi-electrode arrays were used to record extracellular potentials from cortical and subcortical brain regions near the FL in anesthetized mice (5–13 months old) followed by 24 h cortical electroencephalogram (EEG) recordings. Results indicated that FL animals exhibit a high prevalence of spontaneous spike-wave discharges (SWDs), predominately during sleep (EEG), and an increase in the incidence of hyper-excitable burst/suppression activity under general anesthesia (extracellular recordings, 0.5%–3.0% isoflurane). Brief periods of burst activity in the local field potential (LFP) typically presented as an arrhythmic pattern of increased theta-alpha spectral peaks (4–12 Hz) on a background of low-amplitude delta activity (1–4 Hz), were associated with an increase in spontaneous spiking of cortical neurons, and were highly synchronized in control animals across recording sites in both cortical and subcortical layers (average cross-correlation values ranging from +0.73 to +1.0) with minimal phase shift between electrodes. However, in FL animals, cortical vs. subcortical burst activity was strongly out of phase with significantly lower cross-correlation values compared to controls (average values of −0.1 to +0.5, P < 0.05 between groups). In particular, a marked reduction in the level of synchronous burst activity was observed, the closer the recording electrodes were to the malformation (Pearson’s Correlation = 0.525, P < 0.05). In a subset of FL animals (3/9), burst activity also included a spike or spike-wave pattern similar to the SWDs observed in unanesthetized animals. In summary, neonatal FLs increased the hyperexcitable pattern of burst activity induced by anesthesia and disrupted field potential synchrony between cortical and subcortical brain regions near the site of the cortical malformation. Monitoring the altered electrophysiology of burst activity under general anesthesia with multi-dimensional micro-electrode arrays may serve to define distinct neurophysiological biomarkers of epileptogenesis in human brain and improve techniques for surgical resection of epileptogenic malformed brain tissue.
Collapse
Affiliation(s)
- Anthony J Williams
- Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| | - Chen Zhou
- Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| | - Qian-Quan Sun
- Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| |
Collapse
|
49
|
Epilepsy surgery for pediatric low-grade gliomas of the cerebral hemispheres: neurosurgical considerations and outcomes. Childs Nerv Syst 2016; 32:1923-30. [PMID: 27659834 DOI: 10.1007/s00381-016-3162-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Pediatric low-grade tumors are found in roughly 1-3 % of patients with childhood epilepsy; seizures associated with these tumors are often medically refractory and often present a significant morbidity, greater than the presence of the tumor itself. DISCUSSION The unique morbidity of the seizures often requires an epilepsy surgical approach over a standard oncologic resection to achieve a reduction in morbidity for the child. Multiple quality-of-life studies have shown that unless a patient is seizure-free, they remain disabled throughout their life; the best way to achieve this in our patient population is with a multidisciplinary team approach with treatment goals focusing primarily on the epilepsy. CONCLUSION In those patients treated with gross total resection, roughly 80 % will have an Engel class I outcome and 90 % will achieve some reduction in seizure frequency with a significant improvement in quality of life.
Collapse
|
50
|
Abstract
Pediatric neurology relies on ultrasound, computed tomography (CT), and magnetic resonance (MR) imaging. CT prevails in acute neurologic presentations, including traumatic brain injury (TBI), nontraumatic coma, stroke, and status epilepticus, because of easy availability, with images of diagnostic quality, e.g., to exclude hemorrhage, usually completed quickly enough to avoid sedation. Concerns over the risks of ionizing radiation mean re-imaging and higher-dose procedures, e.g., arteriography and venography, require justification. T1/T2-weighted imaging (T1/T2-WI) MR with additional sequences (arteriography, venography, T2*, spectroscopy, diffusion tensor, perfusion, diffusion- (DWI) and susceptibility-weighted imaging (SWI)) often clarifies the diagnosis, which may alter management in acute settings, as well as chronic conditions, e.g., epilepsy. Clinical acumen remains essential to avoid imaging, e.g., in genetic epilepsies or migrainous headaches responding to treatment, or to target sequences to specific diagnosis, e.g., T1/T2-WI for shunt dysfunction (with SWI for TBI); DWI, arteriography including neck vessels, and venography for acute hemiplegia or coma; coronal temporal cuts for partial epilepsy; or muscle imaging for motor delay. The risk of general anesthesia is low; "head-only" scanners may allow rapid MRI without sedation. Timely and accurate reporting, with discrepancy discussion between expert neuroradiologists, is important for management of the child and the family's expectations.
Collapse
|