1
|
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
|
2
|
Zhou F, Hu R, Wang Y, Wu X, Chen X, Xi Z, Zeng K. Calsyntenin-1 expression and function in brain tissue of lithium-pilocarpine rat seizure models. Synapse 2024; 78:e22307. [PMID: 39171546 DOI: 10.1002/syn.22307] [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: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
To present the expression of calsyntenin-1 (Clstn1) in the brain and investigate the potential mechanism of Clstn1 in lithium-pilocarpine rat seizure models. Thirty-five male SD adult rats were induced to have seizures by intraperitoneal injection of lithium chloride pilocarpine. Rats exhibiting spontaneous seizures were divided into the epilepsy (EP) group (n = 15), whereas those without seizures were divided into the control group (n = 14). Evaluate the expression of Clstn1 in the temporal lobe of two groups using Western blotting, immunohistochemistry, and immunofluorescence. Additionally, 55 male SD rats were subjected to status epilepticus (SE) using the same induction method. Rats experiencing seizures exceeding Racine's level 4 (n = 48) were randomly divided into three groups: SE, SE + control lentivirus (lentiviral vector expressing green fluorescent protein [LV-GFP]), and SE + Clstn1-targeted RNA interference lentivirus (LV-Clstn1-RNAi). The LV-GFP group served as a control for the lentiviral vector, whereas the LV-Clstn1-RNAi group received a lentivirus designed to silence Clstn1 expression. These lentiviral treatments were administered via hippocampal stereotactic injection 2 days after SE induction. Seven days after SE, Western blot analysis was performed to evaluate the expression of Clstn1 in the hippocampus and temporal lobe. Meanwhile, we observed the latency of spontaneous seizures and the frequency of spontaneous seizures within 8 weeks among the three groups. The expression of Clstn1 in the cortex and hippocampus of the EP group was significantly increased compared to the control group (p < .05). Immunohistochemistry and immunofluorescence showed that Clstn1 was widely distributed in the cerebral cortex and hippocampus of rats, and colocalization analysis revealed that it was mainly co expressed with neurons in the cytoplasm. Compared with the SE group (11.80 ± 2.17 days) and the SE + GFP group (12.40 ± 1.67 days), there was a statistically significant difference (p < .05) in the latency period of spontaneous seizures (15.14 ± 2.41 days) in the SE + Clstn1 + RNAi group rats. Compared with the SE group (4.60 ± 1.67 times) and the SE + GFP group (4.80 ± 2.05 times), the SE + Clstn1 + RNAi group (2.0 ± .89 times) showed a significant reduction in the frequency of spontaneous seizures within 2 weeks of chronic phase in rats (p < .05). Elevated Clstn1 expression in EP group suggests its role in EP onset. Targeting Clstn1 may be a potential therapeutic approach for EP management.
Collapse
Affiliation(s)
- Fu Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rong Hu
- Department of Neurology, Pizhou People's Hospital, Jiangsu, China
| | - Yuzhu Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohui Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiqin Xi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kebin Zeng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Hung SC, Dahmoush H, Lee HJ, Chen HC, Guimaraes CV. Prenatal Imaging of Supratentorial Fetal Brain Malformation. Magn Reson Imaging Clin N Am 2024; 32:395-412. [PMID: 38944430 DOI: 10.1016/j.mric.2024.03.002] [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: 07/01/2024]
Abstract
This review article provides a comprehensive overview of fetal MR imaging in supratentorial cerebral malformations. It emphasizes the importance of fetal MR imaging as an adjunct diagnostic tool used alongside ultrasound, improving the detection and characterization of prenatal brain abnormalities. This article reviews a spectrum of cerebral malformations, their MR imaging features, and the clinical implications of these findings. Additionally, it outlines the growing importance of fetal MR imaging in the context of perinatal care.
Collapse
Affiliation(s)
- Sheng-Che Hung
- Division of Neuroradiology, Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, NC, USA; Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill
| | - Hisham Dahmoush
- Division of Pediatric Neuroradiology, Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | - Han-Jui Lee
- Division of Neuroradiology, Department of Radiology, Taipei Veterans General Hospital, Taiwan; National Yang Ming Chiao Tung University, Taiwan
| | - Hung-Chieh Chen
- National Yang Ming Chiao Tung University, Taiwan; Division of Neuroradiology, Department of Radiology, Taichung Veterans General Hospital, Taiwan
| | - Carolina V Guimaraes
- Division of Pediatric Radiology, Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Flanders TM, Schreiber JE, Punchak MA, Land SD, Reynolds TA, Soni S, Adzick NS, Heuer GG. Periventricular nodular heterotopia in patients with a prenatal diagnosis of myelomeningocele/myeloschisis: associations with seizures and neurodevelopmental outcomes during early childhood. Childs Nerv Syst 2024; 40:2521-2526. [PMID: 38676719 DOI: 10.1007/s00381-024-06424-6] [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: 03/14/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
PURPOSE Historically, the presence of gray matter heterotopia was a concern for adverse postnatal neurocognitive status in patients undergoing fetal closure of open spinal dysraphism. The purpose of this study was to evaluate neurodevelopmental outcomes and the onset of seizures during early childhood in patients with a prenatal diagnosis of myelomeningocele/myeloschisis (MMC) and periventricular nodular heterotopia (PVNH). METHODS All patients evaluated at the Center for Fetal Diagnosis and Treatment with a diagnosis of MMC between June 2016 to March 2023 were identified. PVNH was determined from prenatal and/or postnatal MRI. The Bayley Scales of Infant and Toddler Development (edition III or IV) were used for neurodevelopmental assessments. Patients were screened for seizures/epilepsy. RESULTS Of 497 patients evaluated with a prenatal diagnosis of MMC, 99 were found to have PVNH on prenatal MRI, of which 35 had confirmed PVNH on postnatal imaging. From the 497 patients, 398 initially did not exhibit heterotopia on prenatal MRI, but 47 of these then had confirmed postnatal PVNH. The presence of PVNH was not a significant risk factor for postnatal seizures in early childhood. The average neurodevelopmental scores were not significantly different among heterotopia groups for cognitive, language, and motor domains. CONCLUSION The presence of PVNH in patients with a prenatal diagnosis of MMC does not indicate an increased risk for neurodevelopmental delay at 1 year of age. We did not demonstrate an association with seizures/epilepsy. These findings can aid clinicians in prenatal consultation regarding fetal repair of open spinal dysraphism. Long-term follow-up is required to discern the true association between PVNH seen on prenatal imaging and postnatal seizures/epilepsy and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Tracy M Flanders
- Division of Neurosurgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Jane E Schreiber
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maria A Punchak
- Division of Neurosurgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sierra D Land
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tom A Reynolds
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shelly Soni
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - N Scott Adzick
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gregory G Heuer
- Division of Neurosurgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Richard D. Wood Jr. Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
5
|
Wendling F, Koksal-Ersoz E, Al-Harrach M, Yochum M, Merlet I, Ruffini G, Bartolomei F, Benquet P. Multiscale neuro-inspired models for interpretation of EEG signals in patients with epilepsy. Clin Neurophysiol 2024; 161:198-210. [PMID: 38520800 DOI: 10.1016/j.clinph.2024.03.006] [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] [Received: 09/15/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVE The aim is to gain insight into the pathophysiological mechanisms underlying interictal epileptiform discharges observed in electroencephalographic (EEG) and stereo-EEG (SEEG, depth electrodes) recordings performed during pre-surgical evaluation of patients with drug-resistant epilepsy. METHODS We developed novel neuro-inspired computational models of the human cerebral cortex at three different levels of description: i) microscale (detailed neuron models), ii) mesoscale (neuronal mass models) and iii) macroscale (whole brain models). Although conceptually different, micro- and mesoscale models share some similar features, such as the typology of neurons (pyramidal cells and three types of interneurons), their spatial arrangement in cortical layers, and their synaptic connectivity (excitatory and inhibitory). The whole brain model consists of a large-scale network of interconnected neuronal masses, with connectivity based on the human connectome. RESULTS For these three levels of description, the fine-tuning of free parameters and the quantitative comparison with real data allowed us to reproduce interictal epileptiform discharges with a high degree of fidelity and to formulate hypotheses about the cell- and network-related mechanisms underlying the generation of fast ripples and SEEG-recorded epileptic spikes and spike-waves. CONCLUSIONS The proposed models provide valuable insights into the pathophysiological mechanisms underlying the generation of epileptic events. The knowledge gained from these models effectively complements the clinical analysis of SEEG data collected during the evaluation of patients with epilepsy. SIGNIFICANCE These models are likely to play a key role in the mechanistic interpretation of epileptiform activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fabrice Bartolomei
- APHM, Timone Hospital, Epileptology and Cerebral Rhythmology Department, Marseille, France; Univ Aix Marseille, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | | |
Collapse
|
6
|
Cheng Y, Zhai Y, Yuan Y, Wang Q, Li S, Sun H. The Contributions of Thrombospondin-1 to Epilepsy Formation. Neurosci Bull 2024; 40:658-672. [PMID: 38528256 PMCID: PMC11127911 DOI: 10.1007/s12264-024-01194-2] [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: 06/27/2023] [Accepted: 01/27/2024] [Indexed: 03/27/2024] Open
Abstract
Epilepsy is a neural network disorder caused by uncontrolled neuronal hyperexcitability induced by an imbalance between excitatory and inhibitory networks. Abnormal synaptogenesis plays a vital role in the formation of overexcited networks. Recent evidence has confirmed that thrombospondin-1 (TSP-1), mainly secreted by astrocytes, is a critical cytokine that regulates synaptogenesis during epileptogenesis. Furthermore, numerous studies have reported that TSP-1 is also involved in other processes, such as angiogenesis, neuroinflammation, and regulation of Ca2+ homeostasis, which are closely associated with the occurrence and development of epilepsy. In this review, we summarize the potential contributions of TSP-1 to epilepsy development.
Collapse
Affiliation(s)
- Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yujie Zhai
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Shucui Li
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
7
|
Su Y, Cao N, Zhang D, Wang M. The effect of ferroptosis-related mitochondrial dysfunction in the development of temporal lobe epilepsy. Ageing Res Rev 2024; 96:102248. [PMID: 38408490 DOI: 10.1016/j.arr.2024.102248] [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: 10/14/2023] [Revised: 01/27/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Temporal lobe epilepsy (TLE) is the most common form of epileptic syndrome. It has been established that due to its complex pathogenesis, a considerable proportion of TLE patients often progress to drug-resistant epilepsy. Ferroptosis has emerged as an important neuronal death mechanism in TLE, which is primarily influenced by lipid accumulation and oxidative stress. In previous studies of ferroptosis, more attention has been focused on the impact of changes in the levels of proteins related to the redox equilibrium and signaling pathways on epileptic seizures. However, it is worth noting that the oxidative-reduction changes in different organelles may have different pathophysiological significance in the process of ferroptosis-related diseases. Mitochondria, as a key organelle involved in ferroptosis, its structural damage and functional impairment can lead to energy metabolism disorders and disruption of the excitatory inhibitory balance, significantly increasing the susceptibility to epileptic seizures. Therefore, secondary mitochondrial dysfunction in the process of ferroptosis could play a crucial role in TLE pathogenesis. This review focuses on ferroptosis and mitochondria, discussing the pathogenic role of ferroptosis-related mitochondrial dysfunction in TLE, thus aiming to provide novel insights and potential implications of ferroptosis-related secondary mitochondrial dysfunction in epileptic seizures and to offer new insights for the precise exploration of ferroptosis-related therapeutic targets for TLE patients.
Collapse
Affiliation(s)
- Yang Su
- Department of Laboratory Medicine, West China Hospital of Sichuan University, China
| | - Ningrui Cao
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Dingkun Zhang
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Minjin Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, China; Department of Neurology, West China Hospital of Sichuan University, China.
| |
Collapse
|
8
|
Doğanyiğit Z, Okan A, Yılmaz S, Uğuz AC, Akyüz E. Gender-related variation expressions of neuroplastin TRAF6, GluA1, GABA(A) receptor, and PMCA in cortex, hippocampus, and brainstem in an experimental epilepsy model. Synapse 2024; 78:e22289. [PMID: 38436644 DOI: 10.1002/syn.22289] [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: 07/21/2023] [Revised: 01/19/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
Epileptic seizures are seen as a result of changing excitability balance depending on the deterioration in synaptic plasticity in the brain. Neuroplastin, and its related molecules which are known to play a role in synaptic plasticity, neurotransmitter activities that provide balance of excitability and, different neurological diseases, have not been studied before in epilepsy. In this study, a total of 34 Sprague-Dawley male and female rats, 2 months old, weighing 250-300 g were used. The epilepsy model in rats was made via pentylenetetrazole (PTZ). After the completion of the experimental procedure, the brain tissue of the rats were taken and the histopathological changes in the hippocampus and cortex parts and the brain stem were investigated, as well as the immunoreactivity of the proteins related to the immunohistochemical methods. As a result of the histopathological evaluation, it was determined that neuron degeneration and the number of dilated blood vessels in the hippocampus, frontal cortex, and brain stem were higher in the PTZ status epilepticus (SE) groups than in the control groups. It was observed that neuroplastin and related proteins TNF receptor-associated factor 6 (TRAF6), Gamma amino butyric acid type A receptors [(GABA(A)], and plasma membrane Ca2+ ATPase (PMCA) protein immunoreactivity levels increased especially in the male hippocampus, and only AMPA receptor subunit type 1 (GluA1) immunoreactivity decreased, unlike other proteins. We believe this may be caused by a problem in the mechanisms regulating the interaction of neuroplastin and GluA1 and may cause problems in synaptic plasticity in the experimental epilepsy model. It may be useful to elucidate this mechanism and target GluA1 when determining treatment strategies.
Collapse
Affiliation(s)
- Züleyha Doğanyiğit
- Faculty of Medicine, Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| | - Aslı Okan
- Faculty of Medicine, Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| | - Seher Yılmaz
- Faculty of Medicine, Department of Anatomy, Yozgat Bozok University, Yozgat, Turkey
| | - A Cihangir Uğuz
- Faculty of Medicine, Department of Biophysics, Karamanoglu Mehmetbey University, Karaman, Turkey
| | - Enes Akyüz
- Faculty of International Medicine, Department of Biophysics, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
9
|
Doğanyiğit Z, Okan A, Akyüz E, Yılmaz S, Ateş Ş, Taheri S, Yılmaz Z, Shaikh MF. Can endoplasmic reticulum stress observed in the PTZ-kindling model seizures be prevented with TUDCA and 4-PBA? Eur J Pharmacol 2023; 960:176072. [PMID: 37852571 DOI: 10.1016/j.ejphar.2023.176072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
Epilepsy is a chronic neurological disease with recurrent seizures. Increasing evidence suggests that endoplasmic reticulum (ER) stress may play a role in the pathogenesis of epilepsy. We aimed to investigate the effects of Tauroursodeoxycholic acid (TUDCA) and 4-phenyl-butyric acid (4-PBA), which are known to suppress ER stress, on developed seizures in terms of markers of ER stress, oxidative stress, and apoptosis. The pentylenetetrazole (PTZ) kindling model was induced in Wistar albino rats (n = 48) by administering 35 mg/kg PTZ intraperitoneally (I.P.) every other day for 1 month. TUDCA and 4-PBA were administered via I.P. at a dose of 500 mg/kg dose. ER stress, apoptosis, and oxidative stress were determined in the hippocampus tissues of animals in all groups. Immunohistochemistry, qRT-PCR, ELISA, and Western Blot analyzes were performed to determine the efficacy of treatments. Expressions of ATF4, ATF6, p-JNK1/2, Cleaved-Kaspase3, and Caspase12 significantly increased in PTZ-kindled seizures compared to the control group. Increased NOX2 and MDA activity in the seizures were measured. In addition, stereology analyzes showed an increased neuronal loss in the PTZ-kindled group. qRT-PCR examination showed relative mRNA levels of CHOP. Accordingly, TUDCA and 4-PBA treatment suppressed the expressions of ATF4, ATF6, Cleaved-Caspase3, Kaspase12, NOX2, MDA, and CHOP in TUDCA + PTZ and 4-PBA + PTZ groups. ER stress-induced oxidative stress and apoptosis by reducing neuronal loss and degeneration were also preserved in these groups. Our data show molecularly that TUDCA and 4-PBA treatment can suppress the ER stress process in epileptic seizures.
Collapse
Affiliation(s)
- Züleyha Doğanyiğit
- Department of Histology and Embryology, Faculty of Medicine, Yozgat Bozok University, Yozgat, 66100, Turkey.
| | - Aslı Okan
- Department of Histology and Embryology, Faculty of Medicine, Yozgat Bozok University, Yozgat, 66100, Turkey
| | - Enes Akyüz
- Department of Biophysics, Faculty of International Medicine, University of Health Sciences, Istanbul, 34468, Turkey
| | - Seher Yılmaz
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, 66100, Turkey
| | - Şükrü Ateş
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, 66100, Turkey
| | - Serpil Taheri
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38030, Turkey
| | - Zeynep Yılmaz
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38030, Turkey
| | - Mohd Farooq Shaikh
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, NSW, Australia
| |
Collapse
|
10
|
Wu J, Cao M, Peng Y, Dong B, Jiang Y, Hu C, Zhu P, Xing W, Yu L, Xu R, Chen Z. Research progress on the treatment of epilepsy with traditional Chinese medicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155022. [PMID: 37647670 DOI: 10.1016/j.phymed.2023.155022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/18/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Traditional Chinese Medicine (TCM) system is a medical system that has been expanding for thousands of years that was formed by the extensive clinical practice experience of many physicians and the accumulation of personal medication habits in China. In TCM, there is a history of long-term medication for epilepsy, the main treatment for epilepsy is TCM drugs and its prescription, supplemented by TCM modalities such as acupuncture therapy, moxibustion therapy, tuina, emotion adjustment therapy, etc. PURPOSE: With the modernization of TCM, the active ingredients and molecular mechanisms of TCM for epilepsy treatment have been gradually revealed. This review aimed to comprehensively summarize the TCM treatment of epilepsy, focusing on the current TCM drugs and some TCM formulae for the treatment of epilepsy, and to discuss the research progress of TCM for the treatment of epilepsy, and to provide a reference to develop future related studies in this field. MATERIALS AND METHODS The mechanism of action of antiepileptic drugs (AEDs) was interpreted from different perspectives by searching online databases and querying various materials identify drugs used in both modern medicine and TCM systems for the treatment of epilepsy. We collected all relevant TCM for epilepsy literature published in the last 30 years up to December 2022 from electronic databases such as PubMed, CNKI and Web of Science, and statistically analyzed the literature for the following keyword information. The search terms comprise the keywords "TCM", "phytochemistry", "pharmacological activity", "epilepsy" and "traditional application" as a combination. Scientific plant names were provided by "The Plant List" (www.theplantlist.org). RESULTS Epilepsy is a complex and serious disease of the brain and nervous system. At present, the treatment of epilepsy in modern medicine is mainly surgery and chemotherapy, but there are many serious side effects. By summarizing the treatment of epilepsy in TCM, it is found that there are various methods to treat epilepsy in TCM, mainly TCM drugs and its formulae. Many TCM drugs have antiepileptic effects. Now found that the main effective TCM drugs for the treatment of epilepsy are Curcumae Longae Rhizoma, Scorpio, Acori Tatarinowii Rhizoma, Uncariae Ramulus Cum Uncis and Ganoderma, etc. And the main compounds that play a role in the treatment of epilepsy are curcumin, gastrodin, ligustrazine, baicalin and rhynchophylline, etc. These TCM drugs have played an important role in the treatment of epilepsy in TCM clinic. However, the chemically active components of these TCM drugs are diverse and their mechanisms of action are complex, which are not fully understood and need to be further explored. CONCLUSIONS TCM treats epilepsy in a variety of ways, and with the discovery of a variety of potential bioactive substances for treatment of epilepsy. With the new progress in the research of other TCM treatment methods for epilepsy, TCM will have greater potential in the clinical application of epilepsy.
Collapse
Affiliation(s)
- Jie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Mayijie Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Ying Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Baohua Dong
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Yunxiu Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Changjiang Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Pengjin Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Weidei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China
| | - Lingying Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China.
| | - Runchun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China.
| | - Zhimin Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, school of pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Fan J, Dong X, Tang Y, Wang X, Lin D, Gong L, Chen C, Jiang J, Shen W, Xu A, Zhang X, Xie Y, Huang X, Zeng L. Preferential pruning of inhibitory synapses by microglia contributes to alteration of the balance between excitatory and inhibitory synapses in the hippocampus in temporal lobe epilepsy. CNS Neurosci Ther 2023; 29:2884-2900. [PMID: 37072932 PMCID: PMC10493672 DOI: 10.1111/cns.14224] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND A consensus has formed that neural circuits in the brain underlie the pathogenesis of temporal lobe epilepsy (TLE). In particular, the synaptic excitation/inhibition balance (E/I balance) has been implicated in shifting towards elevated excitation during the development of TLE. METHODS Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to generate a model of TLE. Next, electroencephalography (EEG) recording was applied to verify the stability and detectability of spontaneous recurrent seizures (SRS) in rats. Moreover, hippocampal slices from rats and patients with mesial temporal lobe epilepsy (mTLE) were assessed using immunofluorescence to determine the alterations of excitatory and inhibitory synapses and microglial phagocytosis. RESULTS We found that KA induced stable SRSs 14 days after status epilepticus (SE) onset. Furthermore, we discovered a continuous increase in excitatory synapses during epileptogenesis, where the total area of vesicular glutamate transporter 1 (vGluT1) rose considerably in the stratum radiatum (SR) of cornu ammonis 1 (CA1), the stratum lucidum (SL) of CA3, and the polymorphic layer (PML) of the dentate gyrus (DG). In contrast, inhibitory synapses decreased significantly, with the total area of glutamate decarboxylase 65 (GAD65) in the SL and PML diminishing enormously. Moreover, microglia conducted active synaptic phagocytosis after the formation of SRSs, especially in the SL and PML. Finally, microglia preferentially pruned inhibitory synapses during recurrent seizures in both rat and human hippocampal slices, which contributed to the synaptic alteration in hippocampal subregions. CONCLUSIONS Our findings elaborately characterize the alteration of neural circuits and demonstrate the selectivity of synaptic phagocytosis mediated by microglia in TLE, which could strengthen the comprehension of the pathogenesis of TLE and inspire potential therapeutic targets for epilepsy treatment.
Collapse
Affiliation(s)
- Jianchen Fan
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xinyan Dong
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Yejiao Tang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xuehui Wang
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Donghui Lin
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Lifen Gong
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Chen Chen
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Jie Jiang
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Weida Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Anyu Xu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xiangnan Zhang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
| | - Yicheng Xie
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Xin Huang
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Linghui Zeng
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| |
Collapse
|
12
|
Hanin A, Cespedes J, Huttner A, Strelnikov D, Gopaul M, DiStasio M, Vezzani A, Hirsch LJ, Aronica E. Neuropathology of New-Onset Refractory Status Epilepticus (NORSE). J Neurol 2023:10.1007/s00415-023-11726-x. [PMID: 37079033 DOI: 10.1007/s00415-023-11726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
New-Onset Refractory Status Epilepticus (NORSE), including its subtype with a preceding febrile illness known as FIRES (Febrile Infection-Related Epilepsy Syndrome), is one of the most severe forms of status epilepticus. Despite an extensive workup (clinical evaluation, EEG, imaging, biological tests), the majority of NORSE cases remain unexplained (i.e., "cryptogenic NORSE"). Understanding the pathophysiological mechanisms underlying cryptogenic NORSE and the related long-term consequences is crucial to improve patient management and preventing secondary neuronal injury and drug-resistant post-NORSE epilepsy. Previously, neuropathological evaluations conducted on biopsies or autopsies have been found helpful for identifying the etiologies of some cases that were previously of unknown cause. Here, we summarize the findings of studies reporting neuropathology findings in patients with NORSE, including FIRES. We identified 64 cryptogenic cases and 66 neuropathology tissue samples, including 37 biopsies, 18 autopsies, and seven epilepsy surgeries (the type of tissue sample was not detailed for 4 cases). We describe the main neuropathology findings and place a particular emphasis on cases for which neuropathology findings helped establish a diagnosis or elucidate the pathophysiology of cryptogenic NORSE, or on described cases in which neuropathology findings supported the selection of specific treatments for patients with NORSE.
Collapse
Affiliation(s)
- Aurélie Hanin
- Department of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, AP-HP, Hôpital de La Pitié-Salpêtrière, Sorbonne Université, DMU Neurosciences 6, Paris, France.
- Epilepsy Unit and Department of Clinical Neurophysiology, AP-HP, Hôpital de La Pitié-Salpêtrière, DMU Neurosciences 6, Paris, France.
| | - Jorge Cespedes
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- School of Medicine, Universidad Autonoma de Centro America, San Jose, Costa Rica
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David Strelnikov
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Margaret Gopaul
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Marcello DiStasio
- Department of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Annamaria Vezzani
- Department of Acute Brain Injury, Istituto di Recerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lawrence J Hirsch
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
13
|
Li J, Bai YC, Wu LH, Zhang P, Wei XC, Ma CH, Yan MN, Wang YT, Chen B. Synthetic relaxometry combined with MUSE DWI and 3D-pCASL improves detection of hippocampal sclerosis. Eur J Radiol 2022; 157:110571. [DOI: 10.1016/j.ejrad.2022.110571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/14/2022] [Accepted: 10/23/2022] [Indexed: 11/03/2022]
|
14
|
From spreading depolarization to epilepsy with neuroinflammation: The role of CGRP in cortex. Exp Neurol 2022; 356:114152. [PMID: 35760098 DOI: 10.1016/j.expneurol.2022.114152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/03/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022]
Abstract
CGRP release plays a major role in migraine pain by activating the trigeminal pain pathways. Here we explored putative additional effects of CGRP on cortical circuits and investigated whether CGRP affects cortical excitability, cortical spreading depolarization (CSD), a phenomenon associated with migraine aura, blood-brain-barrier (BBB) and microglial morphology. We used immunohistochemistry to localize CGRP and the CGRP receptor (CGRP-R) in native cortex and evaluated morphology of microglia and integrity of the BBB after exposure to CGRP. In anesthetized rats we applied CGRP and the CGRP-R antagonist BIBN4096BS locally to the exposed cortex and monitored the spontaneous electrocorticogram and CSDs evoked by remote KCl pressure microinjection. In mouse brain slices CGRP effects on neuronal activity were explored by multielectrode array. CGRP immunoreactivity was detectable in intracortical vessels, and all cortical neurons showed CGRP-R immunoreactivity. In rat cortex in vivo, topical CGRP induced periods of epileptiform discharges, however, also dose-dependently reduced CSD amplitudes and propagation velocity. BIBN4096BS prevented these effects. CGRP evoked synchronized bursting activity in mouse cortical but not in cerebellar slices. Topical application of CGRP to rat cortex induced plasma extravasation and this was associated with reduced ramification of microglial cells. From these findings we conclude that CGRP induces a pathophysiological state in the cortex, consisting in neuronal hyperexcitability and neuroinflammation Thus, CGRP may have a pronounced impact on brain functions during migraine episodes supporting the benefit of CGRP antagonists for clinical use. However, increased cortical CGRP may end the CSD-induced aura phase of migraine.
Collapse
|
15
|
Genetic pathogenesis of the epileptogenic lesions in Tuberous Sclerosis Complex: Therapeutic targeting of the mTOR pathway. Epilepsy Behav 2022; 131:107713. [PMID: 33431351 DOI: 10.1016/j.yebeh.2020.107713] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
Abstract
Tuberous sclerosis complex (TSC) is a genetic multisystem disease due to the mutation in one of the two genes TSC1 and TSC2, affecting several organs and systems and carrying a significant risk of early onset and refractory seizures. The pathogenesis of this complex disorder is now well known, with most of TSC-related manifestations being a consequence of the overactivation of the mammalian Target of Rapamycin (mTOR) complex. The discovery of this underlying mechanism paved the way for the use of a class of drugs called mTOR inhibitors including rapamycin and everolimus and specifically targeting this pathway. Rapamycin has been widely used in different animal models of TSC-related epilepsy and proved to be able not only to suppress seizures but also to prevent the development of epilepsy, thus demonstrating an antiepileptogenic potential. In some models, it also showed some benefit on neuropsychiatric manifestations associated with TSC. Everolimus has recently been approved by the US Food and Drug Administration and the European Medical Agency for the treatment of refractory seizures associated with TSC starting from the age of 2 years. It demonstrated a clear benefit when compared to placebo on reducing the frequency of different seizure types and exerting a higher effect in younger children. In conclusion, mTOR cascade can be a potentially major cause of TSC-associated epilepsy and neurodevelopmental disability, and additional research should investigate if early suppression of abnormal mTOR signal with mTOR inhibitors before seizure onset can be a more efficient approach and an effective antiepileptogenic and disease-modifying strategy in infants with TSC.
Collapse
|
16
|
Shawahna R, Zaid L. Caring for women with epilepsy in Palestine: A qualitative study of the current status. Epilepsy Behav 2022; 130:108689. [PMID: 35398722 DOI: 10.1016/j.yebeh.2022.108689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/26/2022] [Accepted: 03/26/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE This qualitative study was conducted to explore the current status of caring for women with epilepsy (WWE) in the Palestinian healthcare system. METHODS This study used an explorative qualitative design. A purposive sampling technique was used to recruit the participants. Semi-structured in-depth interviews were conducted with neurologists (n = 6), gynecologists (n = 5), psychiatrists (n = 3), an internist (n = 1), and clinical pharmacists (n = 5). The interpretive description methodology was used to thematically analyze the qualitative data. RESULTS A total of 745 min (12.4 h) of interview time were analyzed. The qualitative data collected in this study were categorized under 3 major themes and multiple subthemes. The 3 major themes were: (1) diagnosis and care for patients with epilepsy, (2) general issues in caring for patients with epilepsy, and (3) consideration of women's issues in the pharmacotherapy of epilepsy. Formally adopted protocols/criteria for the diagnosis and care for WWE were lacking. CONCLUSION Findings of this qualitative study showed a need to formally adopt uniform guidelines that can guide the diagnosis and care of WWE in the Palestinian healthcare system. The findings of this study might be informative to healthcare providers, decision-makers in healthcare authorities, WWE, and patient advocacy groups who could be interested in improving and benchmarking healthcare services provided to WWE. Future studies are still needed to quantitatively measure adherence to the international guidelines in caring for WWE.
Collapse
Affiliation(s)
- Ramzi Shawahna
- Department of Physiology, Pharmacology and Toxicology, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine; An-Najah BioSciences Unit, Centre for Poisons Control, Chemical and Biological Analyses, An-Najah National University, Nablus, Palestine.
| | - Lina Zaid
- Master of Pharmacology Program, Faculty of Graduate Studies, An-Najah National University, Nablus, Palestine
| |
Collapse
|
17
|
Aguilar-Castillo MJ, Cabezudo-García P, Ciano-Petersen NL, García-Martin G, Marín-Gracia M, Estivill-Torrús G, Serrano-Castro PJ. Immune Mechanism of Epileptogenesis and Related Therapeutic Strategies. Biomedicines 2022; 10:716. [PMID: 35327518 PMCID: PMC8945207 DOI: 10.3390/biomedicines10030716] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
Immunologic and neuroinflammatory pathways have been found to play a major role in the pathogenesis of many neurological disorders such as epilepsy, proposing the use of novel therapeutic strategies. In the era of personalized medicine and in the face of the exhaustion of anti-seizure therapeutic resources, it is worth looking at the current or future possibilities that neuroimmunomodulator or anti-inflammatory therapy can offer us in the management of patients with epilepsy. For this reason, we performed a narrative review on the recent advances on the basic epileptogenic mechanisms related to the activation of immunity or neuroinflammation with special attention to current and future opportunities for novel treatments in epilepsy. Neuroinflammation can be considered a universal phenomenon and occurs in structural, infectious, post-traumatic, autoimmune, or even genetically based epilepsies. The emerging research developed in recent years has allowed us to identify the main molecular pathways involved in these processes. These molecular pathways could constitute future therapeutic targets for epilepsy. Different drugs current or in development have demonstrated their capacity to inhibit or modulate molecular pathways involved in the immunologic or neuroinflammatory mechanisms described in epilepsy. Some of them should be tested in the future as possible antiepileptic drugs.
Collapse
Affiliation(s)
- María José Aguilar-Castillo
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Biotechnology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
| | - Pablo Cabezudo-García
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Nicolas Lundahl Ciano-Petersen
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Guillermina García-Martin
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Marta Marín-Gracia
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Guillermo Estivill-Torrús
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Pedro Jesús Serrano-Castro
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
- Department of Medicine, University of Málaga, 29071 Málaga, Spain
| |
Collapse
|
18
|
Shawahna R, Zaid L. Concentrations of antiseizure medications in breast milk of lactating women with epilepsy: A systematic review with qualitative synthesis. Seizure 2022; 98:57-70. [PMID: 35427849 DOI: 10.1016/j.seizure.2022.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Recent position papers and guidelines encourage women with epilepsy (WWE) to exclusively breastfeed their infants because the benefits to their infants outweigh the potential adverse effects caused by exposure to antiseizure medications (ASMs). OBJECTIVE The objectives of this review were: to evaluate concentrations of ASMs in breastmilk of lactating WWE, qualitatively synthesize evidence that can be used to estimate theoretical doses as estimated daily intake (EDI) and relative infant dose (RID) of ASMs, and to evaluate potential risks to infants as a result of exposure to ASMs from breastmilk. METHODS This systematic review was registered in the International Prospective Register of Systematic Reviews (PROSPERO) as CRD42020223645. The databases: MEDLINE/PubMed, EMBASE, CINAHL/EBSCO, COCHRANE, SpringerLink, ScienceDirect, Summon, WHO International Clinical Trials Registry Platform, and SCOPUS were systematically searched. A qualitative synthesis was adopted in this study. RESULTS A total of 15 records were included in this systematic review. The included studies reported levels of 8 ASMs in the breastmilk of WWE. The highest RIDs of carbamazepine, lamotrigine, primidone, phenobarbital, gabapentin, valproic acid, ethosuximide, levetiracetam, and topiramate were 3.70%, 36.33%, 4.96%, 3.15%, 4.37%, 1.90%, 31.49%, 12.50%, and 12.18%, respectively. Breastfeeding might be limited or even discontinued when signs of excessive sedation/drowsiness and/or poor weight gain are evident on infants exposed to primidone and phenobarbital, ethosuximide/primidone, or ethosuximide/phenobarbital. CONCLUSIONS Concentrations of ASMs can be detected in breastmilk of WWE and plasma/serum of infants exposed via breastmilk. Healthcare providers and WWE might use the findings of this study to make informed decisions on the safety of breastfeeding while taking ASMs.
Collapse
|
19
|
Gray matter heterotopia: clinical and neuroimaging report on 22 children. Acta Neurol Belg 2022; 122:153-162. [PMID: 34471972 PMCID: PMC8894204 DOI: 10.1007/s13760-021-01774-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/03/2021] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To investigate the clinical characteristics and neuroimaging features of childhood presenting with gray matter heterotopia observed in a single tertiary Pediatric Department in Catania and compare the data with those reported in the literature. METHODS A retrospectively review of the history, clinical findings, electrophysiological features and magnetic resonance images of 22 children presenting with gray matter heterotopia observed from January 2010 to January 2020. RESULTS Among the 22 children included in the study, 17 presented with periventricular heterotopia (PVNH), two with Subcortical Band Heterotopia (SBH), and three with other subcortical heterotopia (SUBH). In the affected children, the ages at first diagnosis ranged from 3 months to 16 years with a mean age of 8.2 years (± 5.4); twelve (54.5%) suffered by developmental delay and intellectual deficit; eleven children (50%) complained of epileptic seizures, mostly focal to bilateral tonic-clonic seizure. In addition, in the periventricular heterotopia group (PVNH), cerebral and systemic malformations were reported in twelve (70%) and in ten (58%) children, respectively, out of seventeen. In the SBH plus SUBH group, epileptic seizures were recorded in 3 (60%) out of 5 children, cerebral malformations in one child and systemic malformations in two children. CONCLUSIONS Heterotopic gray matter malformations include a group of disorders that manifest with a variety of neurological implications, such as cognitive impairment and epilepsy, and often related with epilepsy, other cerebral malformations and systemic anomalies.
Collapse
|
20
|
Beaty S, Rosenthal NA, Gayle J, Dongre P, Ricchetti-Masterson K. Clinical and Economic Outcomes of Intravenous Brivaracetam Compared With Levetiracetam for the Treatment of Seizures in United States Hospitals. Front Neurol 2021; 12:760855. [PMID: 34912285 PMCID: PMC8667030 DOI: 10.3389/fneur.2021.760855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Seizures are common among hospitalized patients. Levetiracetam (LEV), a synaptic vesicle protein 2A (SV2A) ligand, is a common intravenous (IV) anti-seizure medication option in hospitals. Brivaracetam (BRV), a selective SV2A ligand for treatment of focal seizures in patients ≥16 years, has greater binding affinity, higher lipophilicity, and faster brain entry than IV LEV. Differences in clinical outcomes and associated costs between IV BRV and IV LEV in treating hospitalized patients with seizure remain unknown. Objectives: To compare the clinical outcomes, costs, and healthcare resource utilization between patients with seizure treated with IV BRV and those with IV LEV within hospital setting. Design/Methods: A retrospective cohort analysis was performed using chargemaster data from 210 United States hospitals in Premier Healthcare Database. Adult patients (age ≥18 years) treated intravenously with LEV or BRV (with or without BZD) and a seizure discharge diagnosis between July 1, 2016 and December 31, 2019 were included. The cohorts were propensity score-matched 4:1 on baseline characteristics. Outcomes included intubation rates, intensive care unit (ICU) admission, length of stay (LOS), all-cause and seizure-related readmission, total hospitalization cost, and in-hospital mortality. A multivariable regression analysis was performed to determine the association between treatment and main outcomes adjusting for unbalanced confounders. Results: A total of 450 patients were analyzed (IV LEV, n = 360 vs. IV BRV, n = 90). Patients treated with IV BRV had lower crude prevalence of ICU admission (14.4 vs. 24.2%, P < 0.05), 30-day all-cause readmission (1.1 vs. 6.4%, P = 0.06), seizure-related 30-day readmission (0 vs. 4.2%, P < 0.05), similar mean total hospitalization costs ($13,715 vs. $13,419, P = 0.91), intubation (0 vs. 1.1%, P = 0.59), and in-hospital mortality (4.4 vs. 3.9%, P = 0.77). The adjusted odds for ICU admission (adjusted odds ratio [aOR] = 0.6; 95% confidence interval [CI]:0.31, 1.16; P = 0.13), 30-day all-cause readmission (aOR = 0.17; 95% CI:0.02, 1.24; P = 0.08), and in-hospital mortality (aOR = 1.15; 95% CI:0.37, 3.58, P = 0.81) were statistically similar between comparison groups. Conclusion: The use of IV BRV may provide an alternative to IV LEV for management of seizures in hospital setting due to lower or comparable prevalence of ICU admission, intubation, and 30-day seizure-related readmission. Additional studies with greater statistical power are needed to confirm these findings.
Collapse
Affiliation(s)
| | - Ning A Rosenthal
- Premier Applied Sciences, Premier Inc., Charlotte, NC, United States
| | - Julie Gayle
- Premier Applied Sciences, Premier Inc., Charlotte, NC, United States
| | | | | |
Collapse
|
21
|
The Endocannabinoid System in Glial Cells and Their Profitable Interactions to Treat Epilepsy: Evidence from Animal Models. Int J Mol Sci 2021; 22:ijms222413231. [PMID: 34948035 PMCID: PMC8709154 DOI: 10.3390/ijms222413231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is one of the most common neurological conditions. Yearly, five million people are diagnosed with epileptic-related disorders. The neuroprotective and therapeutic effect of (endo)cannabinoid compounds has been extensively investigated in several models of epilepsy. Therefore, the study of specific cell-type-dependent mechanisms underlying cannabinoid effects is crucial to understanding epileptic disorders. It is estimated that about 100 billion neurons and a roughly equal number of glial cells co-exist in the human brain. The glial population is in charge of neuronal viability, and therefore, their participation in brain pathophysiology is crucial. Furthermore, glial malfunctioning occurs in a wide range of neurological disorders. However, little is known about the impact of the endocannabinoid system (ECS) regulation over glial cells, even less in pathological conditions such as epilepsy. In this review, we aim to compile the existing knowledge on the role of the ECS in different cell types, with a particular emphasis on glial cells and their impact on epilepsy. Thus, we propose that glial cells could be a novel target for cannabinoid agents for treating the etiology of epilepsy and managing seizure-like disorders.
Collapse
|
22
|
Vergara López S, Ramos-Jiménez C, Adrián de la Cruz Reyes L, Kevin Galindo Ruiz A, Armando Baigts Arriola L, Manuel Moncayo Olivares J, Gabriela Aguirre Galindo E, Fabricio Pérez Pedroza I, San-Juan D. Epilepsy diagnosis based on one unprovoked seizure and ≥60% risk. A systematic review of the etiologies. Epilepsy Behav 2021; 125:108392. [PMID: 34740089 DOI: 10.1016/j.yebeh.2021.108392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/06/2021] [Accepted: 10/16/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND According to the International League Against Epilepsy (ILAE) criteria, epilepsy can be diagnosed after one unprovoked (or reflex) seizure when there is a ≥60% of seizure recurrence in the next decade. The application of this diagnostic criterion, however, is challenging because the risk of recurrence based on different etiologies is not easily retrievable from the literature. OBJECTIVE To assess etiologies that permit a diagnosis of epilepsy after a single unprovoked seizure. METHODS We conducted a systematic review of the literature search using PubMed, Scopus, and Cochrane library from January 1950 to December 2020 with the keywords: recurrence, risk of recurrence, absolute risk, risk ratio, risk, seizures, epilepsy, structural, infectious, metabolic, immune, and genetic. We included articles that reported estimates of risks of a subsequent unprovoked seizure. Etiologies were categorized according to the ILAE epilepsy classification. The quality of the evidence was evaluated with PRISMA. Descriptive statistics were used. RESULTS A total of 25,044 articles resulted from searching three databases. After authors removed duplicates, 18,911 articles remained. We screened by title and abstract, 40 articles were reviewed and finally, two articles were included. The mean follow-up was 8 years and the mean for a risk to present a subsequent unprovoked seizure was 66.6% and included structural etiologies as stroke, traumatic brain injury, cavernous malformation, arteriovenous malformation, and neuroinfections (unspecified agents). Study quality characteristics are classified with low strength of evidence and moderate-quality cohort. CONCLUSIONS We found that stroke, traumatic brain injury, cavernous or arteriovenous malformations, and unspecified CNS infections can meet the epilepsy diagnosis after one unprovoked seizure based on low strength of evidence and moderate quality of cohorts.
Collapse
Affiliation(s)
- Salvador Vergara López
- Dresden International University, Division of Health Care Sciences, Center for Clinical Research and Management Education, Dresden, Germany
| | - Christian Ramos-Jiménez
- Epilepsy Service, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City, Mexico
| | | | - Axel Kevin Galindo Ruiz
- Epilepsy Service, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City, Mexico
| | - Luis Armando Baigts Arriola
- Epilepsy Service, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City, Mexico
| | | | | | | | - Daniel San-Juan
- Epilepsy Service, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City, Mexico.
| |
Collapse
|
23
|
Akyuz E, Koklu B, Uner A, Angelopoulou E, Paudel YN. Envisioning the role of inwardly rectifying potassium (Kir) channel in epilepsy. J Neurosci Res 2021; 100:413-443. [PMID: 34713909 DOI: 10.1002/jnr.24985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 01/29/2023]
Abstract
Epilepsy is a devastating neurological disorder characterized by recurrent seizures attributed to the disruption of the dynamic excitatory and inhibitory balance in the brain. Epilepsy has emerged as a global health concern affecting about 70 million people worldwide. Despite recent advances in pre-clinical and clinical research, its etiopathogenesis remains obscure, and there are still no treatment strategies modifying disease progression. Although the precise molecular mechanisms underlying epileptogenesis have not been clarified yet, the role of ion channels as regulators of cellular excitability has increasingly gained attention. In this regard, emerging evidence highlights the potential implication of inwardly rectifying potassium (Kir) channels in epileptogenesis. Kir channels consist of seven different subfamilies (Kir1-Kir7), and they are highly expressed in both neuronal and glial cells in the central nervous system. These channels control the cell volume and excitability. In this review, we discuss preclinical and clinical evidence on the role of the several subfamilies of Kir channels in epileptogenesis, aiming to shed more light on the pathogenesis of this disorder and pave the way for future novel therapeutic approaches.
Collapse
Affiliation(s)
- Enes Akyuz
- Faculty of International Medicine, Department of Biophysics, University of Health Sciences, Istanbul, Turkey
| | - Betul Koklu
- Faculty of Medicine, Namık Kemal University, Tekirdağ, Turkey
| | - Arda Uner
- Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
24
|
Green JL, Dos Santos WF, Fontana ACK. Role of glutamate excitotoxicity and glutamate transporter EAAT2 in epilepsy: Opportunities for novel therapeutics development. Biochem Pharmacol 2021; 193:114786. [PMID: 34571003 DOI: 10.1016/j.bcp.2021.114786] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022]
Abstract
Epilepsy is a complex neurological syndrome characterized by seizures resulting from neuronal hyperexcitability and sudden and synchronized bursts of electrical discharges. Impaired astrocyte function that results in glutamate excitotoxicity has been recognized to play a key role in the pathogenesis of epilepsy. While there are 26 drugs marketed as anti-epileptic drugs no current treatments are disease modifying as they only suppress seizures rather than the development and progression of epilepsy. Excitatory amino acid transporters (EAATs) are critical for maintaining low extracellular glutamate concentrations and preventing excitotoxicity. When extracellular glutamate concentrations rise to abnormal levels, glutamate receptor overactivation and the subsequent excessive influx of calcium into the post-synaptic neuron can trigger cell death pathways. In this review we discuss targeting EAAT2, the predominant glutamate transporter in the CNS, as a promising approach for developing therapies for epilepsy. EAAT2 upregulation via transcriptional and translational regulation has proven successful in vivo in reducing spontaneous recurrent seizures and offering neuroprotective effects. Another approach to regulate EAAT2 activity is through positive allosteric modulation (PAM). Novel PAMs of EAAT2 have recently been identified and are under development, representing a promising approach for the advance of novel therapeutics for epilepsy.
Collapse
Affiliation(s)
- Jennifer Leigh Green
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, United States
| | | | | |
Collapse
|
25
|
Leitner DF, Faustin A, Verducci C, Friedman D, William C, Devore S, Wisniewski T, Devinsky O. Neuropathology in the North American sudden unexpected death in epilepsy registry. Brain Commun 2021; 3:fcab192. [PMID: 34514397 PMCID: PMC8417454 DOI: 10.1093/braincomms/fcab192] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 11/12/2022] Open
Abstract
Sudden unexpected death in epilepsy is the leading category of epilepsy-related death and the underlying mechanisms are incompletely understood. Risk factors can include a recent history and high frequency of generalized tonic-clonic seizures, which can depress brain activity postictally, impairing respiration, arousal and protective reflexes. Neuropathological findings in sudden unexpected death in epilepsy cases parallel those in other epilepsy patients, with no implication of novel structures or mechanisms in seizure-related deaths. Few large studies have comprehensively reviewed whole brain examination of such patients. We evaluated 92 North American Sudden unexpected death in epilepsy Registry cases with whole brain neuropathological examination by board-certified neuropathologists blinded to the adjudicated cause of death, with an average of 16 brain regions examined per case. The 92 cases included 61 sudden unexpected death in epilepsy (40 definite, 9 definite plus, 6 probable, 6 possible) and 31 people with epilepsy controls who died from other causes. The mean age at death was 34.4 years and 65.2% (60/92) were male. The average age of death was younger for sudden unexpected death in epilepsy cases than for epilepsy controls (30.0 versus 39.6 years; P = 0.006), and there was no difference in sex distribution respectively (67.3% male versus 64.5%, P = 0.8). Among sudden unexpected death in epilepsy cases, earlier age of epilepsy onset positively correlated with a younger age at death (P = 0.0005) and negatively correlated with epilepsy duration (P = 0.001). Neuropathological findings were identified in 83.7% of the cases in our cohort. The most common findings were dentate gyrus dysgenesis (sudden unexpected death in epilepsy 50.9%, epilepsy controls 54.8%) and focal cortical dysplasia (FCD) (sudden unexpected death in epilepsy 41.8%, epilepsy controls 29.0%). The neuropathological findings in sudden unexpected death in epilepsy paralleled those in epilepsy controls, including the frequency of total neuropathological findings as well as the specific findings in the dentate gyrus, findings pertaining to neurodevelopment (e.g. FCD, heterotopias) and findings in the brainstem (e.g. medullary arcuate or olivary dysgenesis). Thus, like prior studies, we found no neuropathological findings that were more common in sudden unexpected death in epilepsy cases. Future neuropathological studies evaluating larger sudden unexpected death in epilepsy and control cohorts would benefit from inclusion of different epilepsy syndromes with detailed phenotypic information, consensus among pathologists particularly for more subjective findings where observations can be inconsistent, and molecular approaches to identify markers of sudden unexpected death in epilepsy risk or pathogenesis.
Collapse
Affiliation(s)
- Dominique F Leitner
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Arline Faustin
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Chloe Verducci
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Daniel Friedman
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Christopher William
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
- Department of Pathology, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Sasha Devore
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
- Department of Pathology, NYU Langone Health and School of Medicine, New York, NY, USA
- Department of Psychiatry, NYU Langone Health and School of Medicine, New York, NY, USA
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Langone Health and School of Medicine, New York, NY, USA
| |
Collapse
|
26
|
Cao Q, Yang F, Wang H. CB2R induces a protective response against epileptic seizures through ERK and p38 signaling pathways. Int J Neurosci 2021; 131:735-744. [PMID: 32715907 DOI: 10.1080/00207454.2020.1796661] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 05/09/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Epilepsy is a pivotal neurological disorder characterized by the synchronous discharging of neurons to induce momentary brain dysfunction. Temporal lobe epilepsy is the most common type of epilepsy, with seizures originating from the mesial temporal lobe. The hippocampus forms part of the mesial temporal lobe and plays a significant role in epileptogenesis; it also has a vital influence on the mental development of children. In this study, we aimed to explore the effects of CB2 receptor (CB2R) activation on ERK and p38 signaling in nerve cells of a rat epilepsy model. MATERIALS AND METHODS We treated Sprague-Dawley rats with pilocarpine to induce an epilepsy model and treated such animals with a CB2R agonist (JWH133) alone or with a CB2R antagonist (AM630). Nissl's stain showed the neuron conditon in different groups. Western blot analyzed the level of p-ERK and p-p38. RESULTS JWH133 can increase the latent period of first seizure attack and decrease the Grades IV-V magnitude ratio after the termination of SE. Nissl's stain showed JWH133 protected neurons in the hippocampus while AM630 inhibited the functioning of CB2R in neurons. Western blot analysis showed that JWH133 decreased levels of p-ERK and p-p38, which is found at increased levels in the hippocampus of our epilepsy model. In contrast, AM630 inhibited the protective function of JWH133 and also enhanced levels of p-ERK and p-p38. CONCLUSIONS CB2R activation can induce neurons proliferation and survival through activation of ERK and p38 signaling pathways.
Collapse
Affiliation(s)
- Qingjun Cao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fenghua Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Tilelli CQ, Flôres LR, Cota VR, Castro OWD, Garcia-Cairasco N. Amygdaloid complex anatomopathological findings in animal models of status epilepticus. Epilepsy Behav 2021; 121:106831. [PMID: 31864944 DOI: 10.1016/j.yebeh.2019.106831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
Temporal lobe epileptic seizures are one of the most common and well-characterized types of epilepsies. The current knowledge on the pathology of temporal lobe epilepsy relies strongly on studies of epileptogenesis caused by experimentally induced status epilepticus (SE). Although several temporal lobe structures have been implicated in the epileptogenic process, the hippocampal formation is the temporal lobe structure studied in the greatest amount and detail. However, studies in human patients and animal models of temporal lobe epilepsy indicate that the amygdaloid complex can be also an important seizure generator, and several pathological processes have been shown in the amygdala during epileptogenesis. Therefore, in the present review, we systematically selected, organized, described, and analyzed the current knowledge on anatomopathological data associated with the amygdaloid complex during SE-induced epileptogenesis. Amygdaloid complex participation in the epileptogenic process is evidenced, among others, by alterations in energy metabolism, circulatory, and fluid regulation, neurotransmission, immediate early genes expression, tissue damage, cell suffering, inflammation, and neuroprotection. We conclude that major efforts should be made in order to include the amygdaloid complex as an important target area for evaluation in future research on SE-induced epileptogenesis. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Cristiane Queixa Tilelli
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil.
| | - Larissa Ribeiro Flôres
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil
| | - Vinicius Rosa Cota
- Laboratory of Neuroengineering and Neuroscience (LINNce), Department of Electrical Engineering, Campus Santo Antônio, Universidade Federal de São João del-Rei, Praça Frei Orlando, 170, Centro, São João Del Rei, MG 36307-352, Brazil
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Campus A. C. Simões, Universidade Federal de Alagoas, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, Maceió, AL 57072-970, Brazil
| | - Norberto Garcia-Cairasco
- Neurophysiology and Experimental Neuroethology Laboratory (LNNE), Department of Physiology, School of Medicine, Universidade de São Paulo, Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP 14049-900, Brazil.
| |
Collapse
|
28
|
Nonperiodic stimulation for the treatment of refractory epilepsy: Applications, mechanisms, and novel insights. Epilepsy Behav 2021; 121:106609. [PMID: 31704250 DOI: 10.1016/j.yebeh.2019.106609] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 11/21/2022]
Abstract
Electrical stimulation of the central nervous system is a promising alternative for the treatment of pharmacoresistant epilepsy. Successful clinical and experimental stimulation is most usually carried out as continuous trains of current or voltage pulses fired at rates of 100 Hz or above, since lower frequencies yield controversial results. On the other hand, stimulation frequency should be as low as possible, in order to maximize implant safety and battery efficiency. Moreover, the development of stimulation approaches has been largely empirical in general, while they should be engineered with the neurobiology of epilepsy in mind if a more robust, efficient, efficacious, and safe application is intended. In an attempt to reconcile evidence of therapeutic effect with the understanding of the underpinnings of epilepsy, our group has developed a nonstandard form of low-frequency stimulation with randomized interpulse intervals termed nonperiodic stimulation (NPS). The rationale was that an irregular temporal pattern would impair neural hypersynchronization, which is a hallmark of epilepsy. In this review, we start by briefly revisiting the literature on the molecular, cellular, and network level mechanisms of epileptic phenomena in order to highlight this often-overlooked emergent property of cardinal importance in the pathophysiology of the disease. We then review our own studies on the efficacy of NPS against acute and chronic experimental seizures and also on the anatomical and physiological mechanism of the method, paying special attention to the hypothesis that the lack of temporal regularity induces desynchronization. We also put forward a novel insight regarding the temporal structure of NPS that may better encompass the set of findings published by the group: the fact that intervals between stimulation pulses have a distribution that follows a power law and thus may induce natural-like activity that would compete with epileptiform discharge for the recruitment of networks. We end our discussion by mentioning ongoing research and future projects of our lab.
Collapse
|
29
|
Kerr WT, Lee JK, Karimi AH, Tatekawa H, Hickman LB, Connerney M, Sreenivasan SS, Dubey I, Allas CH, Smith JM, Savic I, Silverman DHS, Hadjiiski LM, Beimer NJ, Stacey WC, Cohen MS, Engel J, Feusner JD, Salamon N, Stern JM. A minority of patients with functional seizures have abnormalities on neuroimaging. J Neurol Sci 2021; 427:117548. [PMID: 34216975 DOI: 10.1016/j.jns.2021.117548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Functional seizures often are managed incorrectly as a diagnosis of exclusion. However, a significant minority of patients with functional seizures may have abnormalities on neuroimaging that typically are associated with epilepsy, leading to diagnostic confusion. We evaluated the rate of epilepsy-associated findings on MRI, FDG-PET, and CT in patients with functional seizures. METHODS We studied radiologists' reports from neuroimages at our comprehensive epilepsy center from a consecutive series of patients diagnosed with functional seizures without comorbid epilepsy from 2006 to 2019. We summarized the MRI, FDG-PET, and CT results as follows: within normal limits, incidental findings, unrelated findings, non-specific abnormalities, post-operative study, epilepsy risk factors (ERF), borderline epilepsy-associated findings (EAF), and definitive EAF. RESULTS Of the 256 MRIs, 23% demonstrated ERF (5%), borderline EAF (8%), or definitive EAF (10%). The most common EAF was hippocampal sclerosis, with the majority of borderline EAF comprising hippocampal atrophy without T2 hyperintensity or vice versa. Of the 87 FDG-PETs, 26% demonstrated borderline EAF (17%) or definitive EAF (8%). Epilepsy-associated findings primarily included focal hypometabolism, especially of the temporal lobes, with borderline findings including subtle or questionable hypometabolism. Of the 51 CTs, only 2% had definitive EAF. SIGNIFICANCE This large case series provides further evidence that, while uncommon, EAF are seen in patients with functional seizures. A significant portion of these abnormal findings are borderline. The moderately high rate of these abnormalities may represent framing bias from the indication of the study being "seizures," the relative subtlety of EAF, or effects of antiseizure medications.
Collapse
Affiliation(s)
- Wesley T Kerr
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA.
| | - John K Lee
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amir H Karimi
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hiroyuki Tatekawa
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - L Brian Hickman
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Internal Medicine, University of California at Irvine, Irvine, CA, USA
| | - Michael Connerney
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Ishita Dubey
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Corinne H Allas
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jena M Smith
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ivanka Savic
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Women's and Children's Health, Karolinska Institute and Neurology Clinic, Karolinksa University Hospital, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Daniel H S Silverman
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lubomir M Hadjiiski
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas J Beimer
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - William C Stacey
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mark S Cohen
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA; Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Departments of Bioengineering, Psychology and Biomedical Physics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jerome Engel
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Jamie D Feusner
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA; Department of Women's and Children's Health, Karolinska Institute and Neurology Clinic, Karolinksa University Hospital, Karolinska Universitetssjukhuset, Stockholm, Sweden; Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Noriko Salamon
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John M Stern
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
30
|
The Role of KRAS Mutations in Cortical Malformation and Epilepsy Surgery: A Novel Report of Nevus Sebaceous Syndrome and Review of the Literature. Brain Sci 2021; 11:brainsci11060793. [PMID: 34208656 PMCID: PMC8234150 DOI: 10.3390/brainsci11060793] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
The rare nevus sebaceous (NS) syndrome (NSS) includes cortical malformations and drug-resistant epilepsy. Somatic RAS-pathway genetic variants are pathogenetic in NS, but not yet described within the brain of patients with NSS. We report on a 5-year-old boy with mild psychomotor delay. A brown-yellow linear skin lesion suggestive of NS in the left temporo-occipital area was evident at birth. Epileptic spasms presented at aged six months. EEG showed continuous left temporo-occipital epileptiform abnormalities. Brain MRI revealed a similarly located diffuse cortical malformation with temporal pole volume reduction and a small hippocampus. We performed a left temporo-occipital resection with histopathological diagnosis of focal cortical dysplasia type Ia in the occipital region and hippocampal sclerosis type 1. Three years after surgery, he is seizure-and drug-free (Engel class Ia) and showed cognitive improvement. Genetic examination of brain and skin specimens revealed the c.35G > T (p.Gly12Val) KRAS somatic missense mutation. Literature review suggests epilepsy surgery in patients with NSS is highly efficacious, with 73% probability of seizure freedom. The few histological analyses reported evidenced disorganized cortex, occasionally with cytomegalic neurons. This is the first reported association of a KRAS genetic variant with cortical malformations associated with epilepsy, and suggests a possible genetic substrate for hippocampal sclerosis.
Collapse
|
31
|
Leitner DF, Mills JD, Pires G, Faustin A, Drummond E, Kanshin E, Nayak S, Askenazi M, Verducci C, Chen BJ, Janitz M, Anink JJ, Baayen JC, Idema S, van Vliet EA, Devore S, Friedman D, Diehl B, Scott C, Thijs R, Wisniewski T, Ueberheide B, Thom M, Aronica E, Devinsky O. Proteomics and Transcriptomics of the Hippocampus and Cortex in SUDEP and High-Risk SUDEP Patients. Neurology 2021; 96:e2639-e2652. [PMID: 33910938 PMCID: PMC8205452 DOI: 10.1212/wnl.0000000000011999] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To identify the molecular signaling pathways underlying sudden unexpected death in epilepsy (SUDEP) and high-risk SUDEP compared to control patients with epilepsy. METHODS For proteomics analyses, we evaluated the hippocampus and frontal cortex from microdissected postmortem brain tissue of 12 patients with SUDEP and 14 with non-SUDEP epilepsy. For transcriptomics analyses, we evaluated hippocampus and temporal cortex surgical brain tissue from patients with mesial temporal lobe epilepsy: 6 low-risk and 8 high-risk SUDEP as determined by a short (<50 seconds) or prolonged (≥50 seconds) postictal generalized EEG suppression (PGES) that may indicate severely depressed brain activity impairing respiration, arousal, and protective reflexes. RESULTS In autopsy hippocampus and cortex, we observed no proteomic differences between patients with SUDEP and those with non-SUDEP epilepsy, contrasting with our previously reported robust differences between epilepsy and controls without epilepsy. Transcriptomics in hippocampus and cortex from patients with surgical epilepsy segregated by PGES identified 55 differentially expressed genes (37 protein-coding, 15 long noncoding RNAs, 3 pending) in hippocampus. CONCLUSION The SUDEP proteome and high-risk SUDEP transcriptome were similar to those in other patients with epilepsy in hippocampus and cortex, consistent with diverse epilepsy syndromes and comorbid conditions associated with SUDEP. Studies with larger cohorts and different epilepsy syndromes, as well as additional anatomic regions, may identify molecular mechanisms of SUDEP.
Collapse
Affiliation(s)
- Dominique F Leitner
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - James D Mills
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Geoffrey Pires
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Arline Faustin
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Eleanor Drummond
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Evgeny Kanshin
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Shruti Nayak
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Manor Askenazi
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Chloe Verducci
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Bei Jun Chen
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Michael Janitz
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Jasper J Anink
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Johannes C Baayen
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Sander Idema
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Erwin A van Vliet
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Sasha Devore
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Daniel Friedman
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Beate Diehl
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Catherine Scott
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Roland Thijs
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Thomas Wisniewski
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Beatrix Ueberheide
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Maria Thom
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Eleonora Aronica
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| | - Orrin Devinsky
- From the Comprehensive Epilepsy Center (D.F.L., C.V., S.D., D.F., O.D.), Proteomics Laboratory (E.K., S.N., B.U.), Division of Advanced Research Technologies, and Department of Biochemistry and Molecular Pharmacology (B.U.), NYU School of Medicine; Department of Neurology (D.F.L., G.P., A.F., E.D., S.D., D.F., T.W., B.U., O.D.), Center for Cognitive Neurology (G.P., A.F., E.D., T.W.), Department of Pathology (T.W.), and Department of Psychiatry (T.W.), NYU Langone Health and School of Medicine, New York; Department of (Neuro)Pathology (J.D.M., J.J.A., E.A.v.V., E.A.), Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, the Netherlands; Alzheimer's and Prion Diseases Team (G.P.), Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France; Brain & Mind Centre and School of Medical Sciences (E.D.), Faculty of Medicine and Health, University of Sydney, Australia; Biomedical Hosting LLC (M.A.), Arlington, MA; School of Biotechnology and Biomolecular Sciences (B.J.C., M.J.), University of New South Wales, Sydney, Australia; Amsterdam UMC (J.C.B., S.I.), Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117; Swammerdam Institute for Life Sciences (E.A.v.V.), Center for Neuroscience, University of Amsterdam, the Netherlands; Department of Clinical and Experimental Epilepsy (B.D., C.S., M.T.), University College London Institute of Neurology, UK; and Stichting Epilepsie Instellingen Nederland (R.T., E.A.), Heemstede, the Netherlands
| |
Collapse
|
32
|
Delayed brain development of Rolandic epilepsy profiled by deep learning-based neuroanatomic imaging. Eur Radiol 2021; 31:9628-9637. [PMID: 34018056 DOI: 10.1007/s00330-021-08048-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Although Rolandic epilepsy (RE) has been regarded as a brain developmental disorder, neuroimaging studies have not yet ascertained whether RE has brain developmental delay. This study employed deep learning-based neuroanatomic biomarker to measure the changed feature of "brain age" in RE. METHODS The study constructed a 3D-CNN brain age prediction model through 1155 cases of typically developing children's morphometric brain MRI from open-source datasets and further applied to a local dataset of 167 RE patients and 107 typically developing children. The brain-predicted age difference was measured to quantitatively estimate brain age changes in RE and further investigated the relevancies with cognitive and clinical variables. RESULTS The brain age estimation network model presented a good performance for brain age prediction in typically developing children. The children with RE showed a 0.45-year delay of brain age by contrast with typically developing children. Delayed brain age was associated with neuroanatomic changes in the Rolandic regions and also associated with cognitive dysfunction of attention. CONCLUSION This study provided neuroimaging evidence to support the notion that RE has delayed brain development. KEY POINTS • The children with Rolandic epilepsy showed imaging phenotypes of delayed brain development with increased GM volume and decreased WM volume in the Rolandic regions. • The children with Rolandic epilepsy had a 0.45-year delay of brain-predicted age by comparing with typically developing children, using 3D-CNN-based brain age prediction model. • The delayed brain age was associated with morphometric changes in the Rolandic regions and attentional deficit in Rolandic epilepsy.
Collapse
|
33
|
Protective effects of lamotrigine and vitamin B12 on pentylenetetrazole-induced epileptogenesis in rats. Epilepsy Behav 2021; 118:107915. [PMID: 33743341 DOI: 10.1016/j.yebeh.2021.107915] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 12/26/2022]
Abstract
Epileptogenesis is a process that includes molecular and cellular events that foster the establishment of hyperexcitable neuronal networks in the brain. Pentylenetetrazole (PTZ)-induced kindling model in rodents has added new information to the knowledge about the pathogenesis of epilepsy and potential targets of novel antiepileptic agents. Evidence from animal and human studies suggests that oxidative and inflammatory events may play important roles in the initiation and maintaining seizure activities. Vitamin B12 has beneficial effects on the nervous system and presents pleiotropic effects with antioxidant and anti-inflammatory aspects. In the present study, we aimed to test the hypothesis that vitamin B12 and their combination with lamotrigine prevents behavioral deficits, hippocampal damage, oxidation, and proinflammatory state during epileptogenesis. Male rats were subjected to PTZ-induced epileptogenesis and pretreated with vitamin B12 (50 µg/kg) or Lamotrigine (LTG) (25 mg/kg) or B12 (50 µg/kg) + LTG (25 mg/kg). Vitamin B12 and its combination with LTG suppressed epileptogenesis and improved the performance of rats in the passive avoidance test. In addition, Vitamin B12 and its combination with LTG decreased levels of total oxidative status (TOS), oxidative stress index (OSI), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and increased total antioxidant status (TAS) levels in the hippocampus and cerebral cortex. Furthermore, it reduced hippocampal neuronal damage. Current findings support the beneficial actions of vitamin B12 due to its antioxidative and anti-inflammatory properties during the course of disease.
Collapse
|
34
|
Huang XY, Hu QP, Shi HY, Zheng YY, Hu RR, Guo Q. Everolimus inhibits PI3K/Akt/mTOR and NF-kB/IL-6 signaling and protects seizure-induced brain injury in rats. J Chem Neuroanat 2021; 114:101960. [PMID: 33915267 DOI: 10.1016/j.jchemneu.2021.101960] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Epilepsy is a common chronic neurological disease caused by the over-synchronization of neurons leading to brain dysfunction. Recurrent seizures can lead to cognitive and behavioral deficits, and irreversible brain damage. While the PI3K/Akt/mTOR pathway regulates various physiological processes of neurons and glia, it may also lead to abnormal neuronal signal transduction under pathological conditions, including that of epilepsy. Everolimus (Eve), an mTOR inhibitor, may modulate neuronal excitability and therefore exert protection against epilepsy. Therefore, this study aimed to investigate the neuroprotective effect of Everolimus on seizure-induced brain injury and its regulation of the PI3K/Akt/mTOR and NF-kB/IL-6 signaling pathway. Kainic acid (KA) 15 mg/kg was used to induce seizures and Everolimus (1, 2, 5 mg/kg) was administered as a pretreatment. Hippocampal tissue was extracted 24 h post-seizure. RESULTS The protein and mRNA expression levels of PI3K、p-AKt、p-mTOR、NF-kB and IL-6 as well as neuronal apoptosis and microglia activation, significantly increased after KA-induced seizures, however, these effects were inhibited by Everolimus treatment. Furthermore, pretreatment with Everolimus decreased seizure scores and increased seizure latency. CONCLUSIONS Everolimus can decrease the PI3K/Akt/mTOR and NF-kB/IL-6 signaling pathway, reduce neuronal apoptosis and microglia activation, and attenuate seizure susceptibility and intensity, thus having a protective effect on seizure-induced brain damage.
Collapse
Affiliation(s)
- Xiang-Yi Huang
- Department of Function Examination, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China.
| | - Qing-Peng Hu
- Department of Pediatrics, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China.
| | - Hong-Yun Shi
- Department of Pediatrics, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Ya-Yu Zheng
- Department of Pediatrics, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Rong-Rong Hu
- Department of Pediatrics, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Qian Guo
- Department of Pediatrics, The Second Hospital, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
35
|
Specchio N, Pepi C, De Palma L, Trivisano M, Vigevano F, Curatolo P. Neuroimaging and genetic characteristics of malformation of cortical development due to mTOR pathway dysregulation: clues for the epileptogenic lesions and indications for epilepsy surgery. Expert Rev Neurother 2021; 21:1333-1345. [PMID: 33754929 DOI: 10.1080/14737175.2021.1906651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Malformation of cortical development (MCD) is strongly associated with drug-resistant epilepsies for which surgery to remove epileptogenic lesions is common. Two notable technological advances in this field are identification of the underlying genetic cause and techniques in neuroimaging. These now question how presurgical evaluation ought to be approached for 'mTORpathies.'Area covered: From review of published primary and secondary articles, the authors summarize evidence to consider focal cortical dysplasia (FCD), tuber sclerosis complex (TSC), and hemimegalencephaly (HME) collectively as MCD mTORpathies. The authors also consider the unique features of these related conditions with particular focus on the practicalities of using neuroimaging techniques currently available to define surgical targets and predict post-surgical outcome. Ultimately, the authors consider the surgical dilemmas faced for each condition.Expert opinion: Considering FCD, TSC, and HME collectively as mTORpathies has some merit; however, a unified approach to presurgical evaluation would seem unachievable. Nevertheless, the authors believe combining genetic-centered classification and morphologic findings using advanced imaging techniques will eventually form the basis of a paradigm when considering candidacy for early surgery.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Chiara Pepi
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Luca De Palma
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Federico Vigevano
- Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| |
Collapse
|
36
|
Pires G, Leitner D, Drummond E, Kanshin E, Nayak S, Askenazi M, Faustin A, Friedman D, Debure L, Ueberheide B, Wisniewski T, Devinsky O. Proteomic differences in the hippocampus and cortex of epilepsy brain tissue. Brain Commun 2021; 3:fcab021. [PMID: 34159317 DOI: 10.1093/braincomms/fcab021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022] Open
Abstract
Epilepsy is a common neurological disorder affecting over 70 million people worldwide, with a high rate of pharmaco-resistance, diverse comorbidities including progressive cognitive and behavioural disorders, and increased mortality from direct (e.g. sudden unexpected death in epilepsy, accidents, drowning) or indirect effects of seizures and therapies. Extensive research with animal models and human studies provides limited insights into the mechanisms underlying seizures and epileptogenesis, and these have not translated into significant reductions in pharmaco-resistance, morbidities or mortality. To help define changes in molecular signalling networks associated with seizures in epilepsy with a broad range of aetiologies, we examined the proteome of brain samples from epilepsy and control cases. Label-free quantitative mass spectrometry was performed on the hippocampal cornu ammonis 1-3 region (CA1-3), frontal cortex and dentate gyrus microdissected from epilepsy and control cases (n = 14/group). Epilepsy cases had significant differences in the expression of 777 proteins in the hippocampal CA1 - 3 region, 296 proteins in the frontal cortex and 49 proteins in the dentate gyrus in comparison to control cases. Network analysis showed that proteins involved in protein synthesis, mitochondrial function, G-protein signalling and synaptic plasticity were particularly altered in epilepsy. While protein differences were most pronounced in the hippocampus, similar changes were observed in other brain regions indicating broad proteomic abnormalities in epilepsy. Among the most significantly altered proteins, G-protein subunit beta 1 (GNB1) was one of the most significantly decreased proteins in epilepsy in all regions studied, highlighting the importance of G-protein subunit signalling and G-protein-coupled receptors in epilepsy. Our results provide insights into common molecular mechanisms underlying epilepsy across various aetiologies, which may allow for novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Geoffrey Pires
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA.,Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
| | - Dominique Leitner
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Eleanor Drummond
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Faculty of Medicine and Health, Brain and Mind Centre and School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Shruti Nayak
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Manor Askenazi
- Biomedical Hosting LLC, USA.,Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Arline Faustin
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ludovic Debure
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Beatrix Ueberheide
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA.,Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.,Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
37
|
Markus F, Kannengießer A, Näder P, Atigbire P, Scholten A, Vössing C, Bültmann E, Korenke GC, Owczarek-Lipska M, Neidhardt J. A novel missense variant in the EML1 gene associated with bilateral ribbon-like subcortical heterotopia leads to ciliary defects. J Hum Genet 2021; 66:1159-1167. [PMID: 34211111 PMCID: PMC8612930 DOI: 10.1038/s10038-021-00947-5] [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] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Heterotopia is a brain malformation caused by a failed migration of cortical neurons during development. Clinical symptoms of heterotopia vary in severity of intellectual disability and may be associated with epileptic disorders. Abnormal neuronal migration is known to be associated with mutations in the doublecortin gene (DCX), the platelet-activating factor acetylhydrolase gene (PAFAH1B1), or tubulin alpha-1A gene (TUBA1A). Recently, a new gene encoding echinoderm microtubule-associated protein-like 1 (EML1) was reported to cause a particular form of subcortical heterotopia, the ribbon-like subcortical heterotopia (RSH). EML1 mutations are inherited in an autosomal recessive manner. Only six unrelated EML1-associated heterotopia-affected families were reported so far. The EML1 protein is a member of the microtubule-associated proteins family, playing an important role in microtubule assembly and stabilization as well as in mitotic spindle formation in interphase. Herein, we present a novel homozygous missense variant in EML1 (NM_004434.2: c.692G>A, NP_004425.2: p.Gly231Asp) identified in a male RSH-affected patient. Our clinical and molecular findings confirm the genotype-phenotype associations of EML1 mutations and RSH. Analyses of patient-derived fibroblasts showed the significantly reduced length of primary cilia. In addition, our results presented, that the mutated EML1 protein did not change binding capacities with tubulin. The data described herein will expand the mutation spectrum of the EML1 gene and provide further insight into molecular and cellular bases of the pathogenic mechanisms underlying RSH.
Collapse
Affiliation(s)
- Fenja Markus
- grid.5560.60000 0001 1009 3608Junior Research Group, Genetics of Childhood Brain Malformations, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany ,grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Annika Kannengießer
- grid.5560.60000 0001 1009 3608Junior Research Group, Genetics of Childhood Brain Malformations, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany ,grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Patricia Näder
- grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Paul Atigbire
- grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Alexander Scholten
- grid.5560.60000 0001 1009 3608Division of Biochemistry, Biochemistry of signal transduction/neurosensory processes, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Christine Vössing
- grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Eva Bültmann
- grid.10423.340000 0000 9529 9877Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - G. Christoph Korenke
- grid.419838.f0000 0000 9806 6518Department of Neuropediatrics, University Children’s Hospital, Klinikum Oldenburg, Oldenburg, Germany
| | - Marta Owczarek-Lipska
- grid.5560.60000 0001 1009 3608Junior Research Group, Genetics of Childhood Brain Malformations, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany ,grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany ,grid.5560.60000 0001 1009 3608Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - John Neidhardt
- grid.5560.60000 0001 1009 3608Human Genetics, Faculty VI-School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany ,grid.5560.60000 0001 1009 3608Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
38
|
Gernert M, Feja M. Bypassing the Blood-Brain Barrier: Direct Intracranial Drug Delivery in Epilepsies. Pharmaceutics 2020; 12:pharmaceutics12121134. [PMID: 33255396 PMCID: PMC7760299 DOI: 10.3390/pharmaceutics12121134] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsies are common chronic neurological diseases characterized by recurrent unprovoked seizures of central origin. The mainstay of treatment involves symptomatic suppression of seizures with systemically applied antiseizure drugs (ASDs). Systemic pharmacotherapies for epilepsies are facing two main challenges. First, adverse effects from (often life-long) systemic drug treatment are common, and second, about one-third of patients with epilepsy have seizures refractory to systemic pharmacotherapy. Especially the drug resistance in epilepsies remains an unmet clinical need despite the recent introduction of new ASDs. Apart from other hypotheses, epilepsy-induced alterations of the blood-brain barrier (BBB) are thought to prevent ASDs from entering the brain parenchyma in necessary amounts, thereby being involved in causing drug-resistant epilepsy. Although an invasive procedure, bypassing the BBB by targeted intracranial drug delivery is an attractive approach to circumvent BBB-associated drug resistance mechanisms and to lower the risk of systemic and neurologic adverse effects. Additionally, it offers the possibility of reaching higher local drug concentrations in appropriate target regions while minimizing them in other brain or peripheral areas, as well as using otherwise toxic drugs not suitable for systemic administration. In our review, we give an overview of experimental and clinical studies conducted on direct intracranial drug delivery in epilepsies. We also discuss challenges associated with intracranial pharmacotherapy for epilepsies.
Collapse
Affiliation(s)
- Manuela Gernert
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany;
- Center for Systems Neuroscience, D-30559 Hannover, Germany
- Correspondence: ; Tel.: +49-(0)511-953-8527
| | - Malte Feja
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany;
- Center for Systems Neuroscience, D-30559 Hannover, Germany
| |
Collapse
|
39
|
Severino M, Geraldo AF, Utz N, Tortora D, Pogledic I, Klonowski W, Triulzi F, Arrigoni F, Mankad K, Leventer RJ, Mancini GMS, Barkovich JA, Lequin MH, Rossi A. Definitions and classification of malformations of cortical development: practical guidelines. Brain 2020; 143:2874-2894. [PMID: 32779696 PMCID: PMC7586092 DOI: 10.1093/brain/awaa174] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/14/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022] Open
Abstract
Malformations of cortical development are a group of rare disorders commonly manifesting with developmental delay, cerebral palsy or seizures. The neurological outcome is extremely variable depending on the type, extent and severity of the malformation and the involved genetic pathways of brain development. Neuroimaging plays an essential role in the diagnosis of these malformations, but several issues regarding malformations of cortical development definitions and classification remain unclear. The purpose of this consensus statement is to provide standardized malformations of cortical development terminology and classification for neuroradiological pattern interpretation. A committee of international experts in paediatric neuroradiology prepared systematic literature reviews and formulated neuroimaging recommendations in collaboration with geneticists, paediatric neurologists and pathologists during consensus meetings in the context of the European Network Neuro-MIG initiative on Brain Malformations (https://www.neuro-mig.org/). Malformations of cortical development neuroimaging features and practical recommendations are provided to aid both expert and non-expert radiologists and neurologists who may encounter patients with malformations of cortical development in their practice, with the aim of improving malformations of cortical development diagnosis and imaging interpretation worldwide.
Collapse
Affiliation(s)
| | - Ana Filipa Geraldo
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Neuroradiology Unit, Imaging Department, Centro Hospitalar Vila Nova de Gaia/Espinho (CHVNG/E), Vila Nova de Gaia, Portugal
| | - Norbert Utz
- Department of Pediatric Radiology, HELIOS Klinikum Krefeld, Germany
| | - Domenico Tortora
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Ivana Pogledic
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wlodzimierz Klonowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, Poland
| | - Fabio Triulzi
- Neuroradiology Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, Università degli Studi Milano, Italy
| | - Filippo Arrigoni
- Department of Neuroimaging Lab, Scientific Institute, IRCCS E. Medea, Bosisio Parini, Italy
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, UK
| | - Richard J Leventer
- Department of Neurology Royal Children’s Hospital, Murdoch Children’s Research Institute and University of Melbourne Department of Pediatrics, Melbourne, Australia
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - James A Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Maarten H Lequin
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
40
|
Palombo M, Ianus A, Guerreri M, Nunes D, Alexander DC, Shemesh N, Zhang H. SANDI: A compartment-based model for non-invasive apparent soma and neurite imaging by diffusion MRI. Neuroimage 2020; 215:116835. [PMID: 32289460 PMCID: PMC8543044 DOI: 10.1016/j.neuroimage.2020.116835] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/26/2020] [Accepted: 04/06/2020] [Indexed: 11/29/2022] Open
Abstract
This work introduces a compartment-based model for apparent cell body (namely soma) and neurite density imaging (SANDI) using non-invasive diffusion-weighted MRI (DW-MRI). The existing conjecture in brain microstructure imaging through DW-MRI presents water diffusion in white (WM) and gray (GM) matter as restricted diffusion in neurites, modelled by infinite cylinders of null radius embedded in the hindered extra-neurite water. The extra-neurite pool in WM corresponds to water in the extra-axonal space, but in GM it combines water in the extra-cellular space with water in soma. While several studies showed that this microstructure model successfully describe DW-MRI data in WM and GM at b ≤ 3,000 s/mm2 (or 3 ms/μm2), it has been also shown to fail in GM at high b values (b≫3,000 s/mm2 or 3 ms/μm2). Here we hypothesise that the unmodelled soma compartment (i.e. cell body of any brain cell type: from neuroglia to neurons) may be responsible for this failure and propose SANDI as a new model of brain microstructure where soma of any brain cell type is explicitly included. We assess the effects of size and density of soma on the direction-averaged DW-MRI signal at high b values and the regime of validity of the model using numerical simulations and comparison with experimental data from mouse (bmax = 40,000 s/mm2, or 40 ms/μm2) and human (bmax = 10,000 s/mm2, or 10 ms/μm2) brain. We show that SANDI defines new contrasts representing complementary information on the brain cyto- and myelo-architecture. Indeed, we show maps from 25 healthy human subjects of MR soma and neurite signal fractions, that remarkably mirror contrasts of histological images of brain cyto- and myelo-architecture. Although still under validation, SANDI might provide new insight into tissue architecture by introducing a new set of biomarkers of potential great value for biomedical applications and pure neuroscience.
Collapse
Affiliation(s)
- Marco Palombo
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK.
| | - Andrada Ianus
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK; Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Michele Guerreri
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| | - Daniel Nunes
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Daniel C Alexander
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Hui Zhang
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| |
Collapse
|
41
|
Da Ré C, Souza JM, Fróes F, Taday J, dos Santos JP, Rodrigues L, Sesterheim P, Gonçalves CA, Leite MC. Neuroinflammation induced by lipopolysaccharide leads to memory impairment and alterations in hippocampal leptin signaling. Behav Brain Res 2020; 379:112360. [DOI: 10.1016/j.bbr.2019.112360] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/21/2019] [Accepted: 11/13/2019] [Indexed: 12/29/2022]
|
42
|
Chen Y, Huang T, Sun Y, Liao J, Cao D, Li L, Xiang K, Lin C, Li C, Chen Q. Surface-Based Registration of MR Scan versus Refined Anatomy-Based Registration of CT Scan: Effect on the Accuracy of SEEG Electrodes Implantation Performed in Prone Position under Frameless Neuronavigation. Stereotact Funct Neurosurg 2020; 98:73-79. [PMID: 32036377 DOI: 10.1159/000505713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/31/2019] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Stereoelectroencephalography (SEEG) refers to a commonly used diagnostic procedure to localise and define the epileptogenic zone of refractory focal epilepsies, by means of minimally invasive operation techniques without large craniotomies. OBJECTIVE This study aimed to investigate the influence of different registration methods on the accuracy of SEEG electrode implantation under neuronavigation for paediatric patients with refractory epilepsy. METHODS The clinical data of 18 paediatric patients with refractory epilepsy were retrospectively analysed. The SEEG electrodes were implanted under optical neuronavigation while the patients were in the prone position. Patients were divided into two groups on the basis of the surface-based registration of MR scan method and refined anatomy-based registration of CT scan. Registration time, accuracy, and the differences between electrode placement and preoperative planned position were analysed. RESULTS Thirty-six electrodes in 7 patients were placed under surface-based registration of MR scan, and 45 electrodes in 11 patients were placed under refined anatomy-based registration of CT scan. The registration time of surface-based registration of MR scan and refined anatomy-based registration of CT scan was 45 ± 12 min and 10 ± 4 min. In addition, the mean registration error, the error of insertion point, and target error were 3.6 ± 0.7 mm, 2.7 ± 0.7 mm, and 3.1 ± 0.5 mm in the surface-based registration of MR scan group, and 1.1 ± 0.3 mm, 1.5 ± 0.5 mm, and 2.2 ± 0.6 mm in the refined anatomy-based registration of CT scan group. The differences between the two registration methods were statistically significant. CONCLUSIONS The refined anatomy-based registration of CT scan method can improve the registration efficiency and electrode placement accuracy, and thereby can be considered as the preferred registration method in the application of SEEG electrode implantation under neuronavigation for treatment of paediatric intractable epilepsy.
Collapse
Affiliation(s)
- Yan Chen
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Tieshuan Huang
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Yang Sun
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Jianxiang Liao
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Dezhi Cao
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Lin Li
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Kui Xiang
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Chun Lin
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Cong Li
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Qian Chen
- Neurosurgery Department, Shenzhen Children's Hospital, Shenzhen, China,
| |
Collapse
|
43
|
Synchrotron Radiation-Based Three-Dimensional Visualization of Angioarchitectural Remodeling in Hippocampus of Epileptic Rats. Neurosci Bull 2019; 36:333-345. [PMID: 31823302 DOI: 10.1007/s12264-019-00450-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022] Open
Abstract
Characterizing the three-dimensional (3D) morphological alterations of microvessels under both normal and seizure conditions is crucial for a better understanding of epilepsy. However, conventional imaging techniques cannot detect microvessels on micron/sub-micron scales without angiography. In this study, synchrotron radiation (SR)-based X-ray in-line phase-contrast imaging (ILPCI) and quantitative 3D characterization were used to acquire high-resolution, high-contrast images of rat brain tissue under both normal and seizure conditions. The number of blood microvessels was markedly increased on days 1 and 14, but decreased on day 60 after seizures. The surface area, diameter distribution, mean tortuosity, and number of bifurcations and network segments also showed similar trends. These pathological changes were confirmed by histological tests. Thus, SR-based ILPCI provides systematic and detailed views of cerebrovascular anatomy at the micron level without using contrast-enhancing agents. This holds considerable promise for better diagnosis and understanding of the pathogenesis and development of epilepsy.
Collapse
|
44
|
Mendes NF, Pansani AP, Carmanhães ERF, Tange P, Meireles JV, Ochikubo M, Chagas JR, da Silva AV, Monteiro de Castro G, Le Sueur-Maluf L. The Blood-Brain Barrier Breakdown During Acute Phase of the Pilocarpine Model of Epilepsy Is Dynamic and Time-Dependent. Front Neurol 2019; 10:382. [PMID: 31040818 PMCID: PMC6477033 DOI: 10.3389/fneur.2019.00382] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/28/2019] [Indexed: 11/29/2022] Open
Abstract
The maintenance of blood-brain barrier (BBB) integrity is essential for providing a suitable environment for nervous tissue function. BBB disruption is involved in many central nervous system diseases, including epilepsy. Evidence demonstrates that BBB breakdown may induce epileptic seizures, and conversely, seizure-induced BBB disruption may cause further epileptic episodes. This study was conducted based on the premise that the impairment of brain tissue during the triggering event may determine the organization and functioning of the brain during epileptogenesis, and that BBB may have a key role in this process. Our purpose was to investigate in rats the relationship between pilocarpine-induced status epilepticus (SE), and BBB integrity by determining the time course of the BBB opening and its subsequent recovery during the acute phase of the pilocarpine model. BBB integrity was assessed by quantitative and morphological methods, using sodium fluorescein and Evans blue (EB) dyes as markers of the increased permeability to micromolecules and macromolecules, respectively. Different time-points of the pilocarpine model were analyzed: 30 min after pilocarpine injection and then 1, 5, and 24 h after the SE onset. Our results show that BBB breakdown is a dynamic phenomenon and time-dependent, i.e., it happens at specific time-points of the acute phase of pilocarpine model of epilepsy, recovering in part its integrity afterwards. Pilocarpine-induced changes on brain tissue initially increases the BBB permeability to micromolecules, and subsequently, around 5 h after SE, the BBB breakdown to macromolecules occurs. After BBB breakdown, EB dye is captured by damaged cells, especially neurons, astrocytes, and oligodendrocytes. Although the BBB permeability to macromolecules is restored 24 h after the start of SE, the leakage of micromolecules persists and the consequences of BBB degradation are widely disseminated in the brain. Our findings reveal the existence of a temporal window of BBB dysfunction in the acute phase of the pilocarpine model that is important for the development of therapeutic strategies that could prevent the epileptogenesis.
Collapse
Affiliation(s)
| | - Aline Priscila Pansani
- Departmento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Poliana Tange
- Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | | | - Mayara Ochikubo
- Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | - Jair Ribeiro Chagas
- Departmento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
45
|
Mühlebner A, Bongaarts A, Sarnat HB, Scholl T, Aronica E. New insights into a spectrum of developmental malformations related to mTOR dysregulations: challenges and perspectives. J Anat 2019; 235:521-542. [PMID: 30901081 DOI: 10.1111/joa.12956] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years the role of the mammalian target of rapamycin (mTOR) pathway has emerged as crucial for normal cortical development. Therefore, it is not surprising that aberrant activation of mTOR is associated with developmental malformations and epileptogenesis. A broad spectrum of malformations of cortical development, such as focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC), have been linked to either germline or somatic mutations in mTOR pathway-related genes, commonly summarised under the umbrella term 'mTORopathies'. However, there are still a number of unanswered questions regarding the involvement of mTOR in the pathophysiology of these abnormalities. Therefore, a monogenetic disease, such as TSC, can be more easily applied as a model to study the mechanisms of epileptogenesis and identify potential new targets of therapy. Developmental neuropathology and genetics demonstrate that FCD IIb and hemimegalencephaly are the same diseases. Constitutive activation of mTOR signalling represents a shared pathogenic mechanism in a group of developmental malformations that have histopathological and clinical features in common, such as epilepsy, autism and other comorbidities. We seek to understand the effect of mTOR dysregulation in a developing cortex with the propensity to generate seizures as well as the aftermath of the surrounding environment, including the white matter.
Collapse
Affiliation(s)
- A Mühlebner
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Bongaarts
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, AB, Canada
| | - T Scholl
- Department of Paediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - E Aronica
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Amsterdam, The Netherlands
| |
Collapse
|
46
|
Zhang Z, Qiu W, Gong H, Li G, Jiang Q, Liang P, Zheng H, Zhang P. Low-intensity ultrasound suppresses low-Mg2+-induced epileptiform discharges in juvenile mouse hippocampal slices. J Neural Eng 2019; 16:036006. [DOI: 10.1088/1741-2552/ab0b9a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
47
|
Sousa GK, Capitelli CS, Dombroski TCD, Zanella CAB, Terra VC, Velasco TR, Machado HR, Assirati JA, Carlotti CG, Alves VM, DaCosta JC, Palmini AL, Paglioli E, Sakamoto AC, Spreafico R, Garbelli R, Neder L, Martins AR. Identification and immunophenotype of abnormal cells present in focal cortical dysplasia type IIb. SURGICAL AND EXPERIMENTAL PATHOLOGY 2018. [DOI: 10.1186/s42047-018-0024-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Focal cortical dysplasias (FCDs) are malformations of cortical development that present cortical dyslamination and abnormal cell morphology and are frequently associated with refractory epilepsy. FCD type IIb presents dysmorphic neurons (DNs) and balloon cells (BCs), which are the hallmarks of this dysplasia. Moreover, hypertrophic neurons (HyNs) may be present in FCD types I, II and III. The objective of this study was to perform a detailed morphology and immunophenotype study of BCs, DNs, and HyNs in a cohort of FCD IIb patients.
Methods
Cortices resected as a treatment for refractory epilepsy from 18 cases of FCD type IIb were analysed using Bielschowsky method and haematoxylin and eosin as routine stains. Immunophenotype was performed using specific antibodies to detect epitopes differentially expressed by abnormal cells.
Results
All cases showed cortical dyslamination, BCs, DNs, and HyNs. No cell layer or column could be identified, except for cortical layer I. Lesions predominated in the frontal cortex (11 cases). DNs were large neurons and presented a clumped and or displaced Nissl substance towards the cell membrane, and a cytoplasm accumulation of neurofilament that displaced the nucleus to the cell periphery, as shown by Bielschowsky staining and immunohistochemistry. HyNs were as large as DNs, but without alterations of Nissl substance or dense neurofilament accumulation, with a central nucleus. BCs were identified as large, oval-shaped and pale eosinophilic cells, which lacked the Nissl substance, and presented an eccentric nucleus. BCs and DNs expressed epitopes of both undifferentiated and mature cells, detected using antibodies against nestin, vimentin, class III β-tubulin, pan-neuronal filaments, neurofilament proteins, β-tubulin and NeuN. Only BCs expressed GFAP.
Conclusion
FCDs present with disorganization of the cerebral cortex architecture, abnormal cell morphology, are frequently associated with refractory epilepsy, and their post-surgical prognosis depends on the type of FCD. The diagnosis of focal cortical dysplasia in a surgical specimen relies on the identification of the abnormal cells present in a dysplastic cortex specimen. The current report contributes to the identification of balloon cells, dysmorphic and hypertrophic neurons in the context of focal cortical dysplasia type IIb.
Collapse
|
48
|
Arena A, Zimmer TS, van Scheppingen J, Korotkov A, Anink JJ, Mühlebner A, Jansen FE, van Hecke W, Spliet WG, van Rijen PC, Vezzani A, Baayen JC, Idema S, Iyer AM, Perluigi M, Mills JD, van Vliet EA, Aronica E. Oxidative stress and inflammation in a spectrum of epileptogenic cortical malformations: molecular insights into their interdependence. Brain Pathol 2018; 29:351-365. [PMID: 30303592 DOI: 10.1111/bpa.12661] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/14/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress (OS) occurs in brains of patients with epilepsy and coincides with brain inflammation, and both phenomena contribute to seizure generation in animal models. We investigated whether expression of OS and brain inflammation markers co-occurred also in resected brain tissue of patients with epileptogenic cortical malformations: hemimegalencephaly (HME), focal cortical dysplasia (FCD) and cortical tubers in tuberous sclerosis complex (TSC). Moreover, we studied molecular mechanisms linking OS and inflammation in an in vitro model of neuronal function. Untangling interdependency and underlying molecular mechanisms might pose new therapeutic strategies for treating patients with drug-resistant epilepsy of different etiologies. Immunohistochemistry was performed for specific OS markers xCT and iNOS and brain inflammation markers TLR4, COX-2 and NF-κB in cortical tissue derived from patients with HME, FCD IIa, IIb and TSC. Additionally, we studied gene expression of these markers using the human neuronal cell line SH-SY5Y in which OS was induced using H2 O2 . OS markers were higher in dysmorphic neurons and balloon/giant cells in cortex of patients with FCD IIb or TSC. Expression of OS markers was positively correlated to expression of brain inflammation markers. In vitro, 100 µM, but not 50 µM, of H2 O2 increased expression of TLR4, IL-1β and COX-2. We found that NF-κB signaling was activated only upon stimulation with 100 µM H2 O2 leading to upregulation of TLR4 signaling and IL-1β. The NF-κB inhibitor TPCA-1 completely reversed this effect. Our results show that OS positively correlates with neuroinflammation and is particularly evident in brain tissue of patients with FCD IIb and TSC. In vitro, NF-κB is involved in the switch to an inflammatory state after OS. We propose that the extent of OS can predict the neuroinflammatory state of the brain. Additionally, antioxidant treatments may prevent the switch to inflammation in neurons thus targeting multiple epileptogenic processes at once.
Collapse
Affiliation(s)
- Andrea Arena
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.,Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Till S Zimmer
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Anatoly Korotkov
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jasper J Anink
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Angelika Mühlebner
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wim van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wim G Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Peter C van Rijen
- Department of Neurosurgery, Rudolf Magnus Institute for Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Johannes C Baayen
- Department of Neurosurgery, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sander Idema
- Department of Neurosurgery, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anand M Iyer
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - James D Mills
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Erwin A van Vliet
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro-)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), the Netherlands
| |
Collapse
|
49
|
Wen Y, Wu Q, Shi Q, Xie Y, Dan W, Chen Y, Ma L. UCH-L1 inhibition aggravates mossy fiber sprouting in the pentylenetetrazole kindling model. Biochem Biophys Res Commun 2018; 503:2312-2318. [PMID: 29964011 DOI: 10.1016/j.bbrc.2018.06.154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
Mossy fiber sprouting (MFS) is a pathological phenomenon that is commonly observed in epilepsy, and plentiful data reveal that abnormal phosphorylated modification of tau protein plays a critical role in MSF by the regulation of microtubule dynamics and axonal transport. Ubiquitin C-terminal hydrolase L1 (UCH-L1), a proteasomal deubiquitinating enzyme, has been proved to be associated with tau aggregation through mediating degradation of ubiquitinated and hyperphosphorylated tau. Thus, this study aimed to determine the expression of UCH-L1 in the rat hippocampus during the pentylenetetrazole (PTZ)-induced process and to demonstrate the possible correlation with MFS in epileptogenesis. Seizures were established by intraperitoneal injection of PTZ and LDN-57444 was used to inhibit the hydrolase activity of UCH-L1. We used western blot, immunofluorescence, immunoprecipitation, and timm staining to detect phosphorylated modification of tau and MSF. The results presented that LDN-57444 induced the deteriorated severity of seizures, increased phosphorylation of tau and increased distribution of Timm granules in both the supragranular region of the dentate gyrus (DG) and the stratum pyramidale of CA3 subfield. Our results suggest that UCH-L1 may be associated with hippocampal MSF followed the epileptogenesis through mediating phosphorylation of tau. UCH-L1 may be a potential and novel therapeutic target to limit epileptogenesis.
Collapse
Affiliation(s)
- Yuetao Wen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Number 1, Youyi Road. Yuzhong District, 400042, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing Three Gorges Central Hospital, Number 165, Xincheng Road, Wanzhou District, 404000, Chongqing, China
| | - Quanhong Shi
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Number 1, Youyi Road. Yuzhong District, 400042, Chongqing, China
| | - Yanfeng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Number 1, Youyi Road. Yuzhong District, 400042, Chongqing, China
| | - Wei Dan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Number 1, Youyi Road. Yuzhong District, 400042, Chongqing, China
| | - YangMei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Number 76, Linjiang Road, Yuzhong District, 400010, Chongqing, China
| | - Limin Ma
- Department of Neurology, Chongqing Three Gorges Central Hospital, Number 165, Xincheng Road, Wanzhou District, 404000, Chongqing, China.
| |
Collapse
|
50
|
Abstract
This study firstly investigated the feasibility effect and safety of Ganoderma Lucidum Spore Powder (GLSP) for treating patients with epilepsy.Eighteen eligible patients with epilepsy were included. They all received GLSP treatment for a total of 8 weeks. The primary outcome included weekly seizure frequency. The secondary outcomes consisted of each seizure episode, and quality of life, measured by the Quality of Life in Epilepsy Inventory-31 (QOLIE-31), as well as the adverse events (AEs).After treatment, GLSP can significant reduce the weekly seizure frequency, compared with it before the treatment (P = .04). However, GLSP did not exert promising effect in each seizure episode (P = .13), and quality of life, measured by the QOLIE-31 scale (P = .11). Additionally, only minor AEs occurred during the treatment period.The results of this study showed that GLSP may be effective for reducing the weekly seizure frequency. Further studies are still needed to warrant this result.
Collapse
Affiliation(s)
- Guo-hui Wang
- First Ward of Neurology Department, First Affiliated Hospital of Jiamusi University, Jiamusi
| | - Xin Li
- First Ward of Neurology Department, First Affiliated Hospital of Jiamusi University, Jiamusi
| | - Wen-hui Cao
- Second Ward of Neurology Department, Hongqi Affiliated Hospital of Mudanjiang Medical University, Mudanjiang
| | - Jing Li
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, China
| | - Li-hua Wang
- First Ward of Neurology Department, First Affiliated Hospital of Jiamusi University, Jiamusi
| |
Collapse
|