1
|
Shandra O, Wang Y, Coles LD, Mowrey WB, Li Q, Liu W, Moshé SL, Galanopoulou AS. Efficacy and tolerability of celastrol and edaravone in the multiple-hit rat model of infantile spasms. Epilepsy Behav 2025; 162:110159. [PMID: 39577370 DOI: 10.1016/j.yebeh.2024.110159] [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: 10/28/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE To test whether anti-inflammatory and antioxidant drugs that inhibit the nuclear factor kappa light chain enhancer of activated B cells (NF-kB), celastrol and edaravone, suppress spasms and improve developmental outcomes in the multiple-hit rat model of refractory infantile spasms (IS) due to structural lesions. METHODS Postnatal day 3 (PN3) Sprague-Dawley rats were treated according to the multiple-hit IS model protocol. Using a randomized, blinded, vehicle-controlled, dose- and time-response study design, we tested the effects of single celastrol [1, 2, or 4 mg/kg intraperitoneally (i.p.), 10-14 rats/group] or edaravone (1, 10 or 30 mg/kg i.p., 14-17 rats/group) injections vs their vehicles on behavioral and electroclinical spasms and developmental milestones. Video-EEG monitoring was done on PN6-7 (n = 11-12 rats/group). Pulse celastrol treatment effects (PN4: 4 mg/kg, PN5-6: 2 mg/kg/day i.p.) were determined on spasms, developmental milestones and Barnes maze. Celastrol and edaravone pharmacokinetics in plasma and neocortex were assessed. Linear mixed model analysis of raw or normalized log-transformed spasm frequencies, considering repeated observations was used. RESULTS Single (2-4 mg/kg i.p) or pulse celastrol, but not edaravone, reduced behavioral and electroclinical spasms frequencies within 5hrs. Pulse celastrol did not affect spasm-freedom, survival, developmental milestones or Barnes maze performance. Celastrol had erratic i.p. absorption with maximum concentrations observed between 2-4 h, when effects on spasms were seen. Edaravone had low blood-to-brain permeability. CONCLUSIONS Celastrol's efficacy on spasms is partially explained by its better brain penetration than edaravone's. NFkB inhibitors may be useful in treating drug-resistant IS but delivery methods with improved bioavailability and brain permeability are needed.
Collapse
Affiliation(s)
- Oleksii Shandra
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Yongjun Wang
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Lisa D Coles
- University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Wenzhu B Mowrey
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qianyun Li
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Wei Liu
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA
| | - Solomon L Moshé
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA; Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aristea S Galanopoulou
- Albert Einstein College of Medicine, Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Bronx, NY, USA; Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
2
|
Snyder HE, Jain P, RamachandranNair R, Jones KC, Whitney R. Genetic Advancements in Infantile Epileptic Spasms Syndrome and Opportunities for Precision Medicine. Genes (Basel) 2024; 15:266. [PMID: 38540325 PMCID: PMC10970414 DOI: 10.3390/genes15030266] [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: 01/19/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 06/15/2024] Open
Abstract
Infantile epileptic spasms syndrome (IESS) is a devastating developmental epileptic encephalopathy (DEE) consisting of epileptic spasms, as well as one or both of developmental regression or stagnation and hypsarrhythmia on EEG. A myriad of aetiologies are associated with the development of IESS; broadly, 60% of cases are thought to be structural, metabolic or infectious in nature, with the remainder genetic or of unknown cause. Epilepsy genetics is a growing field, and over 28 copy number variants and 70 single gene pathogenic variants related to IESS have been discovered to date. While not exhaustive, some of the most commonly reported genetic aetiologies include trisomy 21 and pathogenic variants in genes such as TSC1, TSC2, CDKL5, ARX, KCNQ2, STXBP1 and SCN2A. Understanding the genetic mechanisms of IESS may provide the opportunity to better discern IESS pathophysiology and improve treatments for this condition. This narrative review presents an overview of our current understanding of IESS genetics, with an emphasis on animal models of IESS pathogenesis, the spectrum of genetic aetiologies of IESS (i.e., chromosomal disorders, single-gene disorders, trinucleotide repeat disorders and mitochondrial disorders), as well as available genetic testing methods and their respective diagnostic yields. Future opportunities as they relate to precision medicine and epilepsy genetics in the treatment of IESS are also explored.
Collapse
Affiliation(s)
- Hannah E. Snyder
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Puneet Jain
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Rajesh RamachandranNair
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Kevin C. Jones
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| |
Collapse
|
3
|
Koul R. Hypsarrhythmia and triphasic waves seem to be akin(similar) age-dependent responses of the brain to different insults. J Neurosci Rural Pract 2024; 15:156-158. [PMID: 38476411 PMCID: PMC10927049 DOI: 10.25259/jnrp_319_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/30/2023] [Indexed: 03/14/2024] Open
|
4
|
Ren LH, Zhang J, Li SX, Liu P, Chen H, Hu W. Infantile epileptic spasms syndrome: a cohort study of 88 children. Ital J Pediatr 2023; 49:159. [PMID: 38041198 PMCID: PMC10693141 DOI: 10.1186/s13052-023-01563-z] [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: 04/20/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND This study aimed to investigate and analyze the risk factors for non-etiology-specific infantile spasms (IS) and unrelieved clinical symptoms after treatment. METHODS Eighty-eight children with IS who were treated at our hospital from March 2018 to December 2021 were included in the study. The children were divided into etiology-specific (n = 46) and nonetiology-specific (n = 42) groups, based on the diagnostic results, and remission (n = 45) and nonremission (n = 43) groups, based on clinical outcomes after treatment. The clinical data from patients in the etiology-specific and nonetiology-specific groups and the remission and nonremission groups were compared. Risk factors for non-etiology-specific IS were identified using logistic regression analysis. RESULTS Gender, family history, birth status, and metabolic abnormalities were significantly different between the etiology-specific and non-etiology-specific groups. Gender and metabolic abnormalities were risk factors for nonetiology-specific IS. Family history, birth status, metabolic abnormalities, and brain magnetic resonance imaging were significantly different between the remission and nonremission groups, and different etiologies were risk factors for unrelieved symptoms after treatment. CONCLUSION The occurrence of nonetiology-specific IS is associated with gender and metabolic abnormalities in children. After medication, unrelieved IS symptoms are associated with etiologies.
Collapse
Affiliation(s)
- Li-Hong Ren
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China
| | - Jing Zhang
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China
| | - Si-Xiu Li
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China
| | - Ping Liu
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China
| | - Hui Chen
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China
| | - Wenguang Hu
- Department of Pediatric Neurology, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617, Riyue Aveneue, Chengdu, 611731, China.
| |
Collapse
|
5
|
Soylu S, Cherkezzade M, Akbayır E, Yüceer Korkmaz H, Koral G, Şanlı E, Topaloğlu P, Yılmaz V, Tüzün E, Küçükali Cİ. Distribution of peripheral blood mononuclear cell subtypes in patients with West syndrome: Impact of synacthen treatment. Immunol Lett 2023; 261:17-24. [PMID: 37459957 DOI: 10.1016/j.imlet.2023.07.007] [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: 08/22/2022] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND West Syndrome (WS) is an epileptic encephalopathy that typically occurs in infants and is characterized by hypsarrhythmia, infantile spasms, and neurodevelopmental impairment. Demonstration of autoantibodies and cytokines in some WS patients and favorable response to immunotherapy have implicated inflammation as a putative trigger of epileptiform activity in WS. Our aim was to provide additional support for altered inflammatory responses in WS through peripheral blood immunophenotype analysis. METHODS Eight WS cases treated with synacthen and 11 age- and sex-matched healthy volunteers were included. Peripheral blood mononuclear cells (PBMC) were isolated and immunophenotyping was performed in pre-treatment baseline (8 patients) and 3 months post-treatment (6 patients) samples. The analysis included PBMC expressing NFκB transcription and NLRP3 inflammasome factors. RESULTS In pre-treatment baseline samples, switched memory B cells (CD19+IgD-CD27+) were significantly reduced, whereas plasma cells (CD19+CD38+CD138+) and cytotoxic T cells (CD3+CD8+) were significantly increased. Regulatory T and B cell ratios were not significantly altered. Synacthen treatment only marginally reduced helper T cell ratios and did not significantly change other T, B, NK and NKT cell and monocyte ratios. CONCLUSIONS Our findings lend further support for the involvement of inflammation-related mechanisms in WS. New-onset WS patients are inclined to display increased plasma cells in the peripheral blood. Synacthen treatment does not show a beneficial effect on most effector acquired and innate immunity subsets.
Collapse
Affiliation(s)
- Selen Soylu
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey; Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Minara Cherkezzade
- Istanbul University, Istanbul Medical Faculty, Department of Neurology, Istanbul, Turkey
| | - Ece Akbayır
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey; Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Hande Yüceer Korkmaz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey; Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Gizem Koral
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey; Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Elif Şanlı
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey; Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Pınar Topaloğlu
- Istanbul University, Istanbul Medical Faculty, Department of Neurology, Istanbul, Turkey
| | - Vuslat Yılmaz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey
| | - Erdem Tüzün
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey
| | - Cem İsmail Küçükali
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Neuroscience, Istanbul, Turkey.
| |
Collapse
|
6
|
Riikonen R. Biochemical mechanisms in pathogenesis of infantile epileptic spasm syndrome. Seizure 2023; 105:1-9. [PMID: 36634586 DOI: 10.1016/j.seizure.2023.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanisms leading to infantile epileptic spasm syndrome (IESS) remain obscure. The only common factor seems to be that the spasms are restricted to a limited period of infancy, during a certain maturational state. Here the current literature regarding the biochemical mechanisms of brain maturation in IESS is reviewed, and various hypotheses of the pathophysiology are put together. They include: (1) imbalance of inhibitory (NGF, IGF-1, ACTH, GABA) and excitatory factors (glutamate, nitrites) which distinguishes the different etiological subgroups, (2) abnormality of the hypothalamic pituitary adrenal (HPA) axis linking insults and early life stress, (3) inflammation (4) yet poorly known genetic and epigenetic factors, and (5) glucocorticoid and vigabatrin action on brain development, pinpointing at molecular targets of the pathophysiology from another angle. An altered maturational process may explain why so many, seemingly independent etiological factors lead to the same clinical syndrome and frequently to developmental delay. Understanding these factors can provide ideas for novel therapies.
Collapse
Affiliation(s)
- Raili Riikonen
- Children's Hospital, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
7
|
Del Pozo A, Barker-Haliski M. Cannabidiol reveals a disruptive strategy for 21st century epilepsy drug discovery. Exp Neurol 2023; 360:114288. [PMID: 36471511 PMCID: PMC9789191 DOI: 10.1016/j.expneurol.2022.114288] [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: 09/27/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 11/27/2022]
Abstract
Over 30 antiseizure medicines (ASMs) have been uncovered in a diversity of preclinical seizure and epilepsy models, with several critical inflection points in the 20th century fundamentally transforming ASM discovery. This commentary aims to review the historical relevance of cannabidiol's (CBD; Epidiolex) approval for epilepsy in the context of other ASMs brought to market. Further, we highlight how CBD's approval may represent an inflection point for 21st century ASM discovery. CBD is one of the main phytocannabinoids of Cannabis sativa. Unlike its related phytocannabinoid, Δ9-tetrahydrocannabinol, CBD does not exert any euphorigenic, tolerance, or withdrawal effects at anticonvulsant doses. CBD also possess marked anti-inflammatory effects, offering the tantalizing potential of a new pharmacological approach in epilepsy. For decades, hints of the anticonvulsant profile of CBD had been suggested with a small handful of studies in rodent seizure models, yet difficulties in formulation, compounded by the social and regulatory pressures related to medical use of cannabis plant-derived agents constrained any clinical implementation. Nonetheless, CBD possesses a broad antiseizure profile in preclinical seizure and epilepsy models, but the transformative impact of CBD'-s approval came because of studies in a rodent model of the orphan disease Dravet syndrome (DS). DS is a pediatric developmental epileptic encephalopathy with high mortality, frequent spontaneous recurrent seizures, and marked resistance to conventional ASMs, such as phenytoin and carbamazepine. CBD was approved for DS by the US Food and Drug Administration in 2018 after convincing efficacy was established in randomized, placebo-controlled trials in children. Because of the clinical approval of CBD as a novel, cannabis plantderived ASM for DS, CBD has revealed a new strategy in ASM discovery to reignite 21st century therapeutic development for epilepsy. In this commentary, we review the major preclinical and clinical milestones of the late 20th century that made CBD, a compound historically subjected to regulatory restrictions, a key driver of a new discovery strategy for epilepsy in the 21st century.
Collapse
Affiliation(s)
- Aaron Del Pozo
- Department of Pharmacy, University of Washington, Seattle, WA 98195, United States of America
| | - Melissa Barker-Haliski
- Department of Pharmacy, University of Washington, Seattle, WA 98195, United States of America.
| |
Collapse
|
8
|
Duc NM, Thu NTM, Bui CB, Hoa G, Le Trung Hieu N. Genotype and phenotype characteristics of West syndrome in 20 Vietnamese children: Two novel variants detected by next-generation sequencing. Epilepsy Res 2023; 190:107094. [PMID: 36689859 DOI: 10.1016/j.eplepsyres.2023.107094] [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: 11/17/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND In children with West syndrome (WS), whose treatment is challenging due to drug resistance and poor prognosis, investigation of genetic etiology and genotype-phenotype characteristics might assist in treatment optimization and genetic counseling. OBJECTIVE In this study, we aimed to present the results of genetic analysis and the corresponding phenotypes in a cohort of twenty children with WS in Vietnam. METHODS Our study was designed as a single-institution retrospective case series, in which consecutive sampling was used to select WS children having undergone genetic testing. Identified variants were investigated individually or as a variant combination by bioinformatics platforms. Clinical data were used to establish the genotype-phenotype correlation and compare clinical characteristics between groups of genetic causes and unknown causes. RESULTS Genetic testing identified at least one variant in 17/20 children. According to ACMG 2015, of all variants, one variant (3.9%) was classified as a benign variant, 16 variants (61.5%) were variants of uncertain significance, 4 (15.4%) were likely pathogenic variants, and 5 (19.2%) were pathogenic variants. These 26 variants belonged to 21 genes, of which eight candidate genes were CREBBP, MED25, HDAC8, SCN3A, ABCD1, TSC2, COL4A1, and NDUFA10. Two novel variants of SCN3A and TSC2 were found. Predicted pathogenic variant combinations were identified in two cases. Compared to three children of unknown etiology, five children with genetic causes had a higher rate of abnormal brain structures, developmental delay, and treatment resistance. CONCLUSIONS WS has a genetically heterogeneous etiology, and some cases might be polygenically susceptible. Our findings expand the disease's genotype-phenotype spectrum and support previous literature results that genetic etiology poses an unfavorable outcome in WS.
Collapse
Affiliation(s)
- Nguyen Minh Duc
- Neurology Department, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam.
| | - Nguyen Thuy Minh Thu
- Neurology Department, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam; Neurology Department, Children Hospital 2, Ho Chi Minh City 70000, Vietnam.
| | - Chi-Bao Bui
- School of Medicine, Vietnam National University, Ho Chi Minh City 70701, Vietnam.
| | - Giang Hoa
- Medical Genetics Institute, Ho Chi Minh City 70000, Vietnam.
| | - Nguyen Le Trung Hieu
- Neurology Department, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam; Neurology Department, Children Hospital 2, Ho Chi Minh City 70000, Vietnam.
| |
Collapse
|
9
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Pestana Knight EM. Hypsarrhythmia and Epileptic Spasms: A Look at One Old Epilepsy in the Modern Era. J Clin Neurophysiol 2022; 39:519-520. [PMID: 35323130 DOI: 10.1097/wnp.0000000000000909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
11
|
Biagioni F, Celli R, Giorgi FS, Nicoletti F, Fornai F. Perspective on mTOR-dependent Protection in Status Epilepticus. Curr Neuropharmacol 2022; 20:1006-1010. [PMID: 34636300 PMCID: PMC9886823 DOI: 10.2174/1570159x19666211005152618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The piriform cortex, known as area tempestas, has a high propensity to trigger limbic epileptic seizures. Recent studies on human patients indicate that a resection containing the piriform cortex produces a marked improvement in patients suffering from intractable limbic seizures. This calls for looking back at the pharmacological and anatomical data on area tempestas. Within the piriform cortex, status epilepticus can be induced by impairing the desensitization of AMPA receptors. The mechanistic target of rapamycin complex1 (mTORC1) is a promising candidate. OBJECTIVE The present perspective aims to link the novel role of the piriform cortex with recent evidence on the modulation of AMPA receptors under the influence of mTORC1. This is based on recent evidence and preliminary data, leading to the formulation of interaction between mTORC1 and AMPA receptors to mitigate the onset of long-lasting, self-sustaining, neurotoxic status epilepticus. METHODS The perspective grounds its method on recent literature along with the actual experimental procedure to elicit status epilepticus from the piriform cortex and the method to administer the mTORC1 inhibitor rapamycin to mitigate seizure expression and brain damage. RESULTS The available and present perspectives converge to show that rapamycin may disrupt the seizure circuitry initiated in the piriform cortex to mitigate seizure duration, severity, and brain damage. CONCLUSION The perspective provides a novel scenario to understand refractory epilepsy and selfsustaining status epilepticus. It is expected to provide a beneficial outcome in patients suffering from temporal lobe epilepsy.
Collapse
Affiliation(s)
| | - Roberta Celli
- I.R.C.C.S. Neuromed, Pozzilli, Italy;,Co-First Authors
| | - Filippo Sean Giorgi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, Pozzilli, Italy;,Departments of Physiology and Pharmacology, University of Sapienza, Rome, Italy
| | - Francesco Fornai
- I.R.C.C.S. Neuromed, Pozzilli, Italy;,Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy;,Address correspondence to this author at I.R.C.C.S. Neuromed, via dell’elettronica, 86077 Pozzilli, Italy and Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, The University of Pisa, via Roma 55, 56126 Pisa, Italy; Tel: +39 0502218667; E-mails: ;
| |
Collapse
|
12
|
Menon R, Chandrasekharan S, Nanda S, Nair J, Radhakrishnan A, Cherian A, Thomas S. Does Etiology and Hypsarrhythmia Subtype Influence Outcome in West Syndrome? Challenges Encountered from a Referral Center Perspective. Neurol India 2022; 70:188-196. [DOI: 10.4103/0028-3886.336325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
13
|
Hassan W, Noreen H, Rehman S, Kamal MA, Teixeira da Rocha JB. Association of Oxidative Stress with Neurological Disorders. Curr Neuropharmacol 2022; 20:1046-1072. [PMID: 34781871 PMCID: PMC9886831 DOI: 10.2174/1570159x19666211111141246] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/05/2021] [Accepted: 10/06/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGORUND Oxidative stress is one of the main contributing factors involved in cerebral biochemical impairment. The higher susceptibility of the central nervous system to reactive oxygen species mediated damage could be attributed to several factors. For example, neurons use a greater quantity of oxygen, many parts of the brain have higher concentraton of iron, and neuronal mitochondria produce huge content of hydrogen peroxide. In addition, neuronal membranes have polyunsaturated fatty acids, which are predominantly vulnerable to oxidative stress (OS). OS is the imbalance between reactive oxygen species generation and cellular antioxidant potential. This may lead to various pathological conditions and diseases, especially neurodegenerative diseases such as, Parkinson's, Alzheimer's, and Huntington's diseases. OBJECTIVES In this study, we explored the involvement of OS in neurodegenerative diseases. METHODS We used different search terms like "oxidative stress and neurological disorders" "free radicals and neurodegenerative disorders" "oxidative stress, free radicals, and neurological disorders" and "association of oxidative stress with the name of disorders taken from the list of neurological disorders. We tried to summarize the source, biological effects, and physiologic functions of ROS. RESULTS Finally, it was noted that more than 190 neurological disorders are associated with oxidative stress. CONCLUSION More elaborated studies in the future will certainly help in understanding the exact mechanism involved in neurological diseases and provide insight into revelation of therapeutic targets.
Collapse
Affiliation(s)
- Waseem Hassan
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Hamsa Noreen
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Shakila Rehman
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia
| | - Joao Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Programa de Pós-Graduação em Bioquímica, Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
14
|
Spoto G, Saia MC, Amore G, Gitto E, Loddo G, Mainieri G, Nicotera AG, Di Rosa G. Neonatal Seizures: An Overview of Genetic Causes and Treatment Options. Brain Sci 2021; 11:brainsci11101295. [PMID: 34679360 PMCID: PMC8534058 DOI: 10.3390/brainsci11101295] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/16/2021] [Accepted: 09/26/2021] [Indexed: 01/04/2023] Open
Abstract
Seizures are the most frequent neurological clinical symptoms of the central nervous system (CNS) during the neonatal period. Neonatal seizures may be ascribed to an acute event or symptomatic conditions determined by genetic, metabolic or structural causes, outlining the so-called 'Neonatal Epilepsies'. To date, three main groups of neonatal epilepsies are recognised during the neonatal period: benign familial neonatal epilepsy (BFNE), early myoclonic encephalopathy (EME) and 'Ohtahara syndrome' (OS). Recent advances showed the role of several genes in the pathogenesis of these conditions, such as KCNQ2, KCNQ3, ARX, STXBP1, SLC25A22, CDKL5, KCNT1, SCN2A and SCN8A. Herein, we reviewed the current knowledge regarding the pathogenic variants most frequently associated with neonatal seizures, which should be considered when approaching newborns affected by these disorders. In addition, we considered the new possible therapeutic strategies reported in these conditions.
Collapse
Affiliation(s)
- Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.S.); (M.C.S.); (G.A.); (G.D.R.)
| | - Maria Concetta Saia
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.S.); (M.C.S.); (G.A.); (G.D.R.)
| | - Greta Amore
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.S.); (M.C.S.); (G.A.); (G.D.R.)
| | - Eloisa Gitto
- Unit of Neonatal Intensive Care, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | | | - Greta Mainieri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.S.); (M.C.S.); (G.A.); (G.D.R.)
- Correspondence: ; Tel.: +39-090-221-2911
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.S.); (M.C.S.); (G.A.); (G.D.R.)
| |
Collapse
|
15
|
Galanopoulou AS, Löscher W, Lubbers L, O’Brien TJ, Staley K, Vezzani A, D’Ambrosio R, White HS, Sontheimer H, Wolf JA, Twyman R, Whittemore V, Wilcox KS, Klein B. Antiepileptogenesis and disease modification: Progress, challenges, and the path forward-Report of the Preclinical Working Group of the 2018 NINDS-sponsored antiepileptogenesis and disease modification workshop. Epilepsia Open 2021; 6:276-296. [PMID: 34033232 PMCID: PMC8166793 DOI: 10.1002/epi4.12490] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/04/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Epilepsy is one of the most common chronic brain diseases and is often associated with cognitive, behavioral, or other medical conditions. The need for therapies that would prevent, ameliorate, or cure epilepsy and the attendant comorbidities is a priority for both epilepsy research and public health. In 2018, the National Institute of Neurological Disease and Stroke (NINDS) convened a workshop titled "Accelerating the Development of Therapies for Antiepileptogenesis and Disease Modification" that brought together preclinical and clinical investigators and industry and regulatory bodies' representatives to discuss and propose a roadmap to accelerate the development of antiepileptogenic (AEG) and disease-modifying (DM) new therapies. This report provides a summary of the discussions and proposals of the Preclinical Science working group. Highlights of the progress of collaborative preclinical research projects on AEG/DM of ongoing research initiatives aiming to improve infrastructure and translation to clinical trials are presented. Opportunities and challenges of preclinical epilepsy research, vis-à-vis clinical research, were extensively discussed, as they pertain to modeling of specific epilepsy types across etiologies and ages, the utilization of preclinical models in AG/DM studies, and the strategies and study designs, as well as on matters pertaining to transparency, data sharing, and reporting research findings. A set of suggestions on research initiatives, infrastructure, workshops, advocacy, and opportunities for expanding the borders of epilepsy research were discussed and proposed as useful initiatives that could help create a roadmap to accelerate and optimize preclinical translational AEG/DM epilepsy research.
Collapse
Affiliation(s)
- Aristea S. Galanopoulou
- Saul R. Korey Department of NeurologyDominick P. Purpura Department of NeuroscienceIsabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNYUSA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and PharmacyUniversity of Veterinary Medicine HannoverHannoverGermany
| | | | - Terence J. O’Brien
- Department of NeuroscienceCentral Clinical SchoolAlfred HealthMonash UniversityMelbourneVic.Australia
| | - Kevin Staley
- Department of NeurologyMassachusetts General HospitalBostonMAUSA
| | - Annamaria Vezzani
- Department of NeuroscienceIRCCS‐Mario Negri Institute for Pharmacological ResearchMilanoItaly
| | | | - H. Steve White
- Department of PharmacySchool of PharmacyUniversity of WashingtonSeattleWAUSA
| | | | - John A. Wolf
- Center for Brain Injury and RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
- Corporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPAUSA
| | | | - Vicky Whittemore
- National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Karen S. Wilcox
- Department of Pharmacology & ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Brian Klein
- National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
16
|
Scalia B, Venti V, Ciccia LM, Criscione R, Lo Bianco M, Sciuto L, Falsaperla R, Zanghì A, Praticò AD. Aristaless-Related Homeobox (ARX): Epilepsy Phenotypes beyond Lissencephaly and Brain Malformations. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1727140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractThe Aristaless-related homeobox (ARX) transcription factor is involved in the development of GABAergic and cholinergic neurons in the forebrain. ARX mutations have been associated with a wide spectrum of neurodevelopmental disorders in humans and are responsible for both malformation (in particular lissencephaly) and nonmalformation complex phenotypes. The epilepsy phenotypes related to ARX mutations are West syndrome and X-linked infantile spasms, X-linked myoclonic epilepsy with spasticity and intellectual development and Ohtahara and early infantile epileptic encephalopathy syndrome, which are related in most of the cases to intellectual disability and are often drug resistant. In this article, we shortly reviewed current knowledge of the function of ARX with a particular attention on its consequences in the development of epilepsy during early childhood.
Collapse
Affiliation(s)
- Bruna Scalia
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Valeria Venti
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Lina M. Ciccia
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberta Criscione
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Manuela Lo Bianco
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura Sciuto
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Neonatal Intensive Care unit and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| |
Collapse
|
17
|
Lubbers L, Iyengar SS. A team science approach to discover novel targets for infantile spasms (IS). Epilepsia Open 2021; 6:49-61. [PMID: 33681648 PMCID: PMC7918303 DOI: 10.1002/epi4.12441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/28/2020] [Accepted: 10/24/2020] [Indexed: 12/20/2022] Open
Abstract
Infantile spasms (IS) is a devastating epilepsy syndrome that typically begins in the first year of life. Symptoms consist of stereotypical spasms, developmental delay, and electroencephalogram (EEG) that may demonstrate Hypsarhythmia. Current therapeutic approaches are not always effective, and there is no reliable way to predict which patient will respond to therapy. Given this disorder's complexity and the potential impact of a disease-modifying approach, Citizens United for Research in Epilepsy (CURE) employed a "team science" approach to advance the understanding of IS pathology and explore therapeutic modalities that might lead to the development of new ways to potentially prevent spasms and Hypsarhythmia. This approach was a first-of-its-kind collaborative initiative in epilepsy. The IS initiative funded 8 investigative teams over the course of 1-3 years. Projects included the following: discovery on the basic biology of IS, discovery of novel therapeutic targets, cross-validation of targets, discovery of biomarkers, and prognosis and treatment of IS. The combined efforts of a strong investigative team led to numerous advances in understanding the neural pathways underlying IS, testing of small molecules in preclinical models of IS and generated preliminary data on potential biomarkers. Thus far, the initiative has resulted in over 19 publications and subsequent funding for several investigators. Investigators reported that the IS initiative generally affected their research positively due to its collaborative and iterative nature. It also provided a unique opportunity to mentor junior investigators with an interest in translational research. Learnings included the need for a dedicated project manager and more transparent and real-time communication with investigators. The CURE IS initiative represents a unique approach to fund scientific discoveries on epilepsy. It brought together an interdisciplinary group of investigators-who otherwise would not have collaborated-to find transformative therapies for IS. Learnings from this initiative are being utilized for subsequent initiatives at CURE.
Collapse
Affiliation(s)
- Laura Lubbers
- Citizens United for Research in Epilepsy (CURE)ChicagoILUSA
| | | |
Collapse
|
18
|
Löscher W, Potschka H, Sisodiya SM, Vezzani A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Pharmacol Rev 2020; 72:606-638. [PMID: 32540959 PMCID: PMC7300324 DOI: 10.1124/pr.120.019539] [Citation(s) in RCA: 386] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epilepsy is a chronic neurologic disorder that affects over 70 million people worldwide. Despite the availability of over 20 antiseizure drugs (ASDs) for symptomatic treatment of epileptic seizures, about one-third of patients with epilepsy have seizures refractory to pharmacotherapy. Patients with such drug-resistant epilepsy (DRE) have increased risks of premature death, injuries, psychosocial dysfunction, and a reduced quality of life, so development of more effective therapies is an urgent clinical need. However, the various types of epilepsy and seizures and the complex temporal patterns of refractoriness complicate the issue. Furthermore, the underlying mechanisms of DRE are not fully understood, though recent work has begun to shape our understanding more clearly. Experimental models of DRE offer opportunities to discover, characterize, and challenge putative mechanisms of drug resistance. Furthermore, such preclinical models are important in developing therapies that may overcome drug resistance. Here, we will review the current understanding of the molecular, genetic, and structural mechanisms of ASD resistance and discuss how to overcome this problem. Encouragingly, better elucidation of the pathophysiological mechanisms underpinning epilepsies and drug resistance by concerted preclinical and clinical efforts have recently enabled a revised approach to the development of more promising therapies, including numerous potential etiology-specific drugs (“precision medicine”) for severe pediatric (monogenetic) epilepsies and novel multitargeted ASDs for acquired partial epilepsies, suggesting that the long hoped-for breakthrough in therapy for as-yet ASD-resistant patients is a feasible goal.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Heidrun Potschka
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Sanjay M Sisodiya
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Annamaria Vezzani
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| |
Collapse
|
19
|
Kats DJ, Roche KJ, Skotko BG. Epileptic spasms in individuals with Down syndrome: A review of the current literature. Epilepsia Open 2020; 5:344-353. [PMID: 32913943 PMCID: PMC7469826 DOI: 10.1002/epi4.12412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/27/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy can occur in individuals with Down syndrome (DS), with epileptic spasms representing the most frequent seizure type in this population. Epileptic spasms can have devastating consequences on the development of individuals with the condition. This review sought to explore the lifetime prevalence and underlying mechanism of epileptic spasms in this population. We also aimed to review the response rate to various treatments, the relapse rate, and the development of subsequent epilepsy or autism in this population. A comprehensive literature search was conducted for articles discussing the lifetime prevalence, diagnosis, treatment, outcomes, or underlying etiology of epileptic spasms in animal models or individuals with DS. According to available literature, the global clinic-based lifetime prevalence of epilepsy in individuals with DS ranged from 1.6% to 23.1%, with epileptic spasms representing 6.7%-66.7% of these cases. Response rate to treatment with adrenocorticotropic hormone/corticosteroids was highest (81%) and has the most literature supporting its use, with other regimens, including vigabatrin and other antiepileptic drugs, having lower response rates. Epileptic spasms occur more frequently in children with DS than in the general population, though more studies are needed to determine the true lifetime prevalence of epileptic spasms in this population. Generally, children with DS and epileptic spasms tend to be more responsive to treatment and have better outcomes than children with epileptic spasms of unknown etiology (ie, without DS), in terms of response and relapse rates as well as the development of intractable epilepsy (eg, Lennox-Gastaut syndrome).
Collapse
Affiliation(s)
- Daniel J. Kats
- Case Western Reserve University School of MedicineClevelandOHUSA
- Down Syndrome ProgramDivision of Medical Genetics and MetabolismDepartment of PediatricsMassachusetts General HospitalBostonMAUSA
| | - Katherine J. Roche
- Department of PediatricsHarvard Medical SchoolBostonMAUSA
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMAUSA
| | - Brian G. Skotko
- Down Syndrome ProgramDivision of Medical Genetics and MetabolismDepartment of PediatricsMassachusetts General HospitalBostonMAUSA
- Department of PediatricsHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
20
|
Infantile Spasms: An Update on Pre-Clinical Models and EEG Mechanisms. CHILDREN-BASEL 2020; 7:children7010005. [PMID: 31935804 PMCID: PMC7023485 DOI: 10.3390/children7010005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/06/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022]
Abstract
Infantile spasms (IS) is an epileptic encephalopathy with unique clinical and electrographic features, which affects children in the middle of the first year of life. The pathophysiology of IS remains incompletely understood, despite the heterogeneity of IS etiologies, more than 200 of which are known. In particular, the neurobiological basis of why multiple etiologies converge to a relatively similar clinical presentation has defied explanation. Treatment options for this form of epilepsy, which has been described as “catastrophic” because of the poor cognitive, developmental, and epileptic prognosis, are limited and not fully effective. Until the pathophysiology of IS is better clarified, novel treatments will not be forthcoming, and preclinical (animal) models are essential for advancing this knowledge. Here, we review preclinical IS models, update information regarding already existing models, describe some novel models, and discuss exciting new data that promises to advance understanding of the cellular mechanisms underlying the specific EEG changes seen in IS—interictal hypsarrhythmia and ictal electrodecrement.
Collapse
|
21
|
Liao J, Huang T, Srour M, Xiao Y, Chen Y, Lin S, Chen L, Hu Y, Men L, Wen J, Li B, Wen F, Xiong L. Status Epilepticus Manifested as Continuous Epileptic Spasms. Front Neurol 2020; 11:65. [PMID: 32117026 PMCID: PMC7034528 DOI: 10.3389/fneur.2020.00065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/17/2020] [Indexed: 02/05/2023] Open
Abstract
Objective: The etiology and outcome of status epilepticus with continuous epileptic spasms have not been fully understood; and only rare cases have been reported in the literature. Here, we described 11 children, who manifested continuous epileptic spasms with various etiologies and different outcomes. Methods: This is a case series study designed to systematically review the charts, video-electroencephalography (video-EEG), magnetic resonance images, and longitudinal follow-up of patients who presented continuous epileptic spasms lasting more than 30 min. Results: Median age at onset was 2 years old, ranging from 2 months to 5.6 years. The etiology of continuous epileptic spasms for these 11 cases consisted of not only some known electro-clinical epilepsy syndromes like West Syndrome and Ohtahara Syndrome, but also secondary symptomatic continuous epileptic spasms, caused by acute encephalitis or encephalopathy, which extends the etiological spectrum of continuous epileptic spasms. The most characteristic feature of these 11 cases was prolonged epileptic spasms, lasting for a median of 13.00 days (95% CI: 7.26-128.22 days). The interictal EEG findings typically manifested as hypsarrhythmia or its variants, including burst suppression. Hospital stays were much longer in acute symptomatic cases than in primary epileptic syndromic cases (59.67 ± 50.82 vs. 15.00 ± 1.41 days). However, the long-term outcomes were extremely poor in the patients with defined electro-clinical epilepsy syndromes, including severe motor and intellectual developmental deficits (follow-up of 4.94 ± 1.56 years), despite early diagnosis and treatment. Continuous epileptic spasms were refractory to corticosteroids, immuno-modulation or immunosuppressive therapies, and ketogenic diet. Conclusion: Continuous epileptic spasms were associated with severe brain impairments in patients with electro-clinical syndromes; and required long hospital stays in patients with acute symptomatic causes. We suggest to include continuous epileptic spasms in the international classification of status epilepticus, as a special form. Further investigations are required to better recognize this condition, better understand the etiology, as well as to explore more effective treatments to improve outcomes.
Collapse
Affiliation(s)
- Jianxiang Liao
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Tieshuan Huang
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Myriam Srour
- Montreal Children's Hospital, McGill University, Montreal, QC, Canada
| | - Yuhan Xiao
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Yan Chen
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Sufang Lin
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Li Chen
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Yan Hu
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Lina Men
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Jialun Wen
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Bing Li
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Feiqiu Wen
- Shenzhen Children's Hospital Affiliated With China Medical University, Shenzhen, China
| | - Lan Xiong
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- *Correspondence: Lan Xiong
| |
Collapse
|
22
|
Higgins GA, Fletcher PJ, Shanahan WR. Lorcaserin: A review of its preclinical and clinical pharmacology and therapeutic potential. Pharmacol Ther 2020; 205:107417. [DOI: 10.1016/j.pharmthera.2019.107417] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022]
|
23
|
Yang Q, Huang Z, Luo Y, Zheng F, Hu Y, Liu H, Zhu S, He M, Xu D, Li Y, Yang M, Yang Y, Wei X, Gao X, Wang W, Ma J, Ma Y, Wang X, Wang Q. Inhibition of Nwd1 activity attenuates neuronal hyperexcitability and GluN2B phosphorylation in the hippocampus. EBioMedicine 2019; 47:470-483. [PMID: 31474551 PMCID: PMC6796588 DOI: 10.1016/j.ebiom.2019.08.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/04/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND NACHT and WD repeat domain-containing protein 1 (Nwd1) is a member of the innate immune protein subfamily. Nwd1 contributes to the androgen receptor signaling pathway and is involved in axonal growth. However, the mechanisms that underlie pathophysiological dysfunction in seizures remain unclear. METHODS Biochemical methods were used to assess Nwd1 expression and localization in a mouse model of kainic acid (KA)-induced acute seizures and temporal lobe epilepsy (TLE) patients. Electrophysiological recordings were used to measure the role of Nwd1 in regulating synaptic transmission and neuronal hyperexcitability in a model of magnesium-free-induced seizure in vitro. Behavioral experiments were performed, and seizure-induced pathological changes were evaluated in a KA-induced seizure model in vivo. GluN2B expression was measured and its correlation with Tyr1472-GluN2B phosphorylation was analyzed in primary hippocampal neurons. FINDINGS We demonstrated high protein levels of Nwd1 in brain tissues obtained from mice with acute seizures and TLE patients. Silencing Nwd1 in mice using an adeno-associated virus (AAV) profoundly suppressed neuronal hyperexcitability and the occurrence of acute seizures, which may have been caused by reducing GluN2B-containing NMDA receptor-dependent glutamatergic synaptic transmission. Moreover, the decreased activation of Nwd1 reduced GluN2B expression and the phosphorylation of the GluN2B subunit at Tyr1472. INTERPRETATION Here, we report a previously unrecognized but important role of Nwd1 in seizure models in vitro and in vivo, i.e., modulating the phosphorylation of the GluN2B subunit at Tyr1472 and regulating neuronal hyperexcitability. Meanwhile, our findings may provide a therapeutic strategy for the treatment of epilepsy or other hyperexcitability-related neurological disorders. FUND: The funders have not participated in the study design, data collection, data analysis, interpretation, or writing of the report.
Collapse
Affiliation(s)
- Qin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China; Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Yangfu Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Fangshuo Zheng
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yida Hu
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Hui Liu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Miaoqing He
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Demei Xu
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yun Li
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Min Yang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yi Yang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Wei Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Junhong Ma
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Yuanlin Ma
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Xuefeng Wang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China; Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100101, PR China.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China.
| |
Collapse
|
24
|
Silenieks LB, Carroll NK, Van Niekerk A, Van Niekerk E, Taylor C, Upton N, Higgins GA. Evaluation of Selective 5-HT 2C Agonists in Acute Seizure Models. ACS Chem Neurosci 2019; 10:3284-3295. [PMID: 31082204 DOI: 10.1021/acschemneuro.8b00739] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The 5-HT releaser/reuptake inhibitor fenfluramine has been recently reported to provide benefit as an adjunctive treatment for Dravet and Lennox-Gastaut syndromes, two types of severe childhood epilepsy. Despite its enhancement of 5-HT function, many effects of fenfluramine have been demonstrated to be dependent on 5-HT2C receptor activation, suggesting that 5-HT2C receptor activation may have an anticonvulsant property. The present study was designed to evaluate fenfluramine and 5-HT agonists of varying 5-HT2C agonist selectivity, the relatively nonselective mCPP and Ro 60-0175, and the selective 5-HT2C agonists lorcaserin and CP-809101 across a variety of acute seizure tests conducted in adult rats and mice, which have been instrumental in identifying the majority of clinically efficacious antiepileptic drugs. Tests included the maximal electroshock seizure (MES), MES threshold, and 6 Hz electrical convulsive seizure models and the chemoconvulsant pentylenetetrazole test. The effect of mCPP, lorcaserin, and CP-809101 against electrically evoked seizures in amygdala kindled rats was also investigated. Overall, at doses known to interact with 5-HT2CR, there was no clear class-related effect of these agonists in any test. The only notable antiseizure effect of fenfluramine was inhibition of MES-induced tonic seizures in the rat. The current preclinical studies using the classical acute seizure tests and an amygdala kindling model do not identify a reliable antiseizure effect of fenfluramine, an agent now used in the treatment of human epilepsies, including Dravet syndrome and Lennox-Gastaut syndrome. Given the nature of these epilepsies, early life and/or genetic models may have better construct validity and be more appropriate for further study.
Collapse
Affiliation(s)
- Leonardo B. Silenieks
- Intervivo Solutions Inc, Toronto, ON M5A 4K2, Canada
- Vivocore, Toronto, ON N1M 2W4, Canada
| | | | | | | | | | - Neil Upton
- Transpharmation Ltd, London NW10NH, United Kingdom
| | - Guy A. Higgins
- Intervivo Solutions Inc, Toronto, ON M5A 4K2, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON ON M5S, Canada
| |
Collapse
|
25
|
Katsarou AM, Li Q, Liu W, Moshé SL, Galanopoulou AS. Acquired parvalbumin-selective interneuronopathy in the multiple-hit model of infantile spasms: A putative basis for the partial responsiveness to vigabatrin analogs? Epilepsia Open 2018; 3:155-164. [PMID: 30564774 PMCID: PMC6293059 DOI: 10.1002/epi4.12280] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2018] [Indexed: 12/13/2022] Open
Abstract
West syndrome, an age-specific epileptic encephalopathy, manifests with infantile spasms (IS) and impaired neurodevelopmental outcomes and epilepsy. The multiple-hit rat model of IS is a chronic model of IS due to structural etiology, in which spasms respond partially to vigabatrin analogs. Using this model, we investigated whether IS due to structural etiology may have deficits in parvalbumin (PRV) and somatostatin (SST) immunoreactive (-ir) interneurons, and calretinin-ir (CR-ir) neurons of the primary somatosensory cortex of postnatal day (PN) 20-24 rats, using specific immunohistochemical assays. PN3 Sprague-Dawley male rats underwent the multiple-hit induction protocol, were monitored until PN20-24, and were transcardially perfused to collect brains for histology. Age-matched sham and naive control male rats were also used. Coronal brain cryosections were stained with anti-PRV, anti-CR, and anti-SST antibodies, and regions of interest (ROIs) from the primary somatosensory cortices were selected to determine PRV-, CR-, and SST-ir cell counts and cortical ROI volumes, with blinding to experimental group. Statistical analyses were done using a linear mixed model accounting for repeated measures. We found PRV-ir interneuronal selective reduction, sparing of the CR-ir and SST-ir neurons, and bilateral cortical atrophy. Our findings provide evidence for acquired PRV-selective interneuronopathy, possibly underlying the pathogenesis of IS, neurodevelopmental deficits, and epilepsy, and potentially contributing to the partial response to vigabatrin analogs in this model.
Collapse
Affiliation(s)
- Anna-Maria Katsarou
- Laboratory of Developmental Epilepsy Saul R. Korey Department of Neurology Albert Einstein College of Medicine Bronx New York U.S.A
| | - Qianyun Li
- Laboratory of Developmental Epilepsy Saul R. Korey Department of Neurology Albert Einstein College of Medicine Bronx New York U.S.A
| | - Wei Liu
- Laboratory of Developmental Epilepsy Saul R. Korey Department of Neurology Albert Einstein College of Medicine Bronx New York U.S.A
| | - Solomon L Moshé
- Laboratory of Developmental Epilepsy Saul R. Korey Department of Neurology Albert Einstein College of Medicine Bronx New York U.S.A.,Laboratory of Developmental Epilepsy Isabelle Rapin Division of Child Neurology Dominick P. Purpura Department of Neuroscience Albert Einstein College of Medicine Einstein/Montefiore Epilepsy Center Montefiore Medical Center Bronx New York U.S.A.,Department of Pediatrics Albert Einstein College of Medicine Einstein/Montefiore Epilepsy Center Montefiore Medical Center Bronx New York U.S.A
| | - Aristea S Galanopoulou
- Laboratory of Developmental Epilepsy Saul R. Korey Department of Neurology Albert Einstein College of Medicine Bronx New York U.S.A.,Laboratory of Developmental Epilepsy Isabelle Rapin Division of Child Neurology Dominick P. Purpura Department of Neuroscience Albert Einstein College of Medicine Einstein/Montefiore Epilepsy Center Montefiore Medical Center Bronx New York U.S.A
| |
Collapse
|
26
|
Moshé SL. The 2017 Sachs Lecture: Kindling Knowledge in Epilepsy. Pediatr Neurol 2018; 85:5-12. [PMID: 29958806 DOI: 10.1016/j.pediatrneurol.2018.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York; Dominick P. Purpura Department of Neuroscience, Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
27
|
Abstract
Epilepsy is a chronic neurological disorder affecting 65 million people worldwide. The etiologies of seizures can often be identified as genetic, metabolic, structural, immunologic or infectious, but in many cases the cause is unknown with the current diagnostic tools. Epileptogenesis is a process during which genetic or other acquired etiologies/insults lead to functional, structural, or network reorganization changes in the brain that may lead to the development of, or progression of, spontaneous seizures. During development, there are continuous changes in the structure, function, and network operation that also show sex specificity, which may alter the mechanisms underlying the generation of seizures (ictogenesis) and epileptogenesis. Understanding the mechanisms of early life epileptogenesis will enable the development of rationally designed age- and sex-appropriate therapies that would improve the overall quality of patients' lives. Here, we discuss some of these processes that may affect seizure generation and epileptogenesis in the neonatal brain.
Collapse
Affiliation(s)
- Anna-Maria Katsarou
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA,Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA,Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York, USA,Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, Bronx, New York, USA,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA,Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York, USA,Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, Bronx, New York, USA,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA,Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA,Corresponding author. Address: Department of Neurology, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Rose F. Kennedy Center, Rm 316, Bronx, NY 10461, USA. Tel.: +1 718-430-2447; fax: +1 718-430-8899. (S.L. Moshé)
| |
Collapse
|
28
|
Abstract
West syndrome (WS) is an early life epileptic encephalopathy associated with infantile spasms, interictal electroencephalography (EEG) abnormalities including high amplitude, disorganized background with multifocal epileptic spikes (hypsarrhythmia), and often neurodevelopmental impairments. Approximately 64% of the patients have structural, metabolic, genetic, or infectious etiologies and, in the rest, the etiology is unknown. Here we review the contribution of etiologies due to various metabolic disorders in the pathology of WS. These may include metabolic errors in organic molecules involved in amino acid and glucose metabolism, fatty acid oxidation, metal metabolism, pyridoxine deficiency or dependency, or acidurias in organelles such as mitochondria and lysosomes. We discuss the biochemical, clinical, and EEG features of these disorders as well as the evidence of how they may be implicated in the pathogenesis and treatment of WS. The early recognition of these etiologies in some cases may permit early interventions that may improve the course of the disease.
Collapse
Affiliation(s)
- Seda Salar
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Solomon L. Moshé
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Department of PediatricsMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Aristea S. Galanopoulou
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| |
Collapse
|
29
|
Katsarou A, Moshé SL, Galanopoulou AS. INTERNEURONOPATHIES AND THEIR ROLE IN EARLY LIFE EPILEPSIES AND NEURODEVELOPMENTAL DISORDERS. Epilepsia Open 2017; 2:284-306. [PMID: 29062978 PMCID: PMC5650248 DOI: 10.1002/epi4.12062] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2017] [Indexed: 12/22/2022] Open
Abstract
GABAergic interneurons control the neural circuitry and network activity in the brain. The advances in genetics have identified genes that control the development, maturation and integration of GABAergic interneurons and implicated them in the pathogenesis of epileptic encephalopathies or neurodevelopmental disorders. For example, mutations of the Aristaless-Related homeobox X-linked gene (ARX) may result in defective GABAergic interneuronal migration in infants with epileptic encephalopathies like West syndrome (WS), Ohtahara syndrome or X-linked lissencephaly with abnormal genitalia (XLAG). The concept of "interneuronopathy", i.e. impaired development, migration or function of interneurons, has emerged as a possible etiopathogenic mechanism for epileptic encephalopathies. Treatments that enhance GABA levels, may help seizure control but do not necessarily show disease modifying effect. On the other hand, interneuronopathies can be seen in other conditions in which epilepsy may not be the primary manifestation, such as autism. In this review, we plan to outline briefly the current state of knowledge on the origin, development, and migration and integration of GABAergic interneurons, present neurodevelopmental conditions, with or without epilepsy, that have been associated with interneuronopathies and discuss the evidence linking certain types of interneuronal dysfunction with epilepsy and/or cognitive or behavioral deficits.
Collapse
Affiliation(s)
- Anna‐Maria Katsarou
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Solomon L. Moshé
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Aristea S. Galanopoulou
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| |
Collapse
|
30
|
Barker-Haliski ML, Löscher W, White HS, Galanopoulou AS. Neuroinflammation in epileptogenesis: Insights and translational perspectives from new models of epilepsy. Epilepsia 2017; 58 Suppl 3:39-47. [PMID: 28675559 PMCID: PMC5604891 DOI: 10.1111/epi.13785] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2017] [Indexed: 12/25/2022]
Abstract
Animal models have provided a wealth of information on mechanisms of epileptogenesis and comorbidogenesis, and have significantly advanced our ability to investigate the potential of new therapies. Processes implicating brain inflammation have been increasingly observed in epilepsy research. Herein we discuss the progress on animal models of epilepsy and comorbidities that inform us on the potential role of inflammation in epileptogenesis and comorbidity pathogenesis in rodent models of West syndrome and the Theiler's murine encephalomyelitis virus (TMEV) mouse model of viral encephalitis-induced epilepsy. Rat models of infantile spasms were generated in rat pups after right intracerebral injections of proinflammatory compounds (lipopolysaccharides with or without doxorubicin, or cytokines) and were longitudinally monitored for epileptic spasms and neurodevelopmental and cognitive deficits. Anti-inflammatory treatments were tested after the onset of spasms. The TMEV mouse model was induced with intracerebral administration of TMEV and prospective monitoring for handling-induced seizures or seizure susceptibility, as well as long-term evaluations of behavioral comorbidities of epilepsy. Inflammatory processes are evident in both models and are implicated in the pathogenesis of the observed seizures and comorbidities. A common feature of these models, based on the data so far available, is their pharmacoresistant profile. The presented data support the role of inflammatory pathways in epileptogenesis and comorbidities in two distinct epilepsy models. Pharmacoresistance is a common feature of both inflammation-based models. Utilization of these models may facilitate the identification of age-specific, syndrome- or etiology-specific therapies for the epilepsies and attendant comorbidities, including the drug-resistant forms.
Collapse
Affiliation(s)
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - H. Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Bronx NY USA
| |
Collapse
|
31
|
Kim EH, Yum MS, Lee M, Kim EJ, Shim WH, Ko TS. A New Rat Model of Epileptic Spasms Based on Methylazoxymethanol-Induced Malformations of Cortical Development. Front Neurol 2017; 8:271. [PMID: 28659857 PMCID: PMC5466970 DOI: 10.3389/fneur.2017.00271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/26/2017] [Indexed: 02/03/2023] Open
Abstract
Malformations of cortical development (MCDs) can cause medically intractable epilepsies and cognitive disabilities in children. We developed a new model of MCD-associated epileptic spasms by treating rats prenatally with methylazoxymethanol acetate (MAM) to induce cortical malformations and postnatally with N-methyl-d-aspartate (NMDA) to induce spasms. To produce cortical malformations to infant rats, two dosages of MAM (15 mg/kg, intraperitoneally) were injected to pregnant rats at gestational day 15. In prenatally MAM-exposed rats and the controls, spasms were triggered by single (6 mg/kg on postnatal day 12 (P12) or 10 mg/kg on P13 or 15 mg/kg on P15) or multiple doses (P12, P13, and P15) of NMDA. In prenatally MAM-exposed rats with single NMDA-provoked spasms at P15, we obtain the intracranial electroencephalography and examine the pretreatment response to adrenocorticotropic hormone (ACTH) or vigabatrin. Rat pups prenatally exposed to MAM exhibited a significantly greater number of spasms in response to single and multiple postnatal NMDA doses than vehicle-exposed controls. Vigabatrin treatment prior to a single NMDA dose on P15 significantly suppressed spasms in MAM group rats (p < 0.05), while ACTH did not. The MAM group also showed significantly higher fast oscillation (25–100 Hz) power during NMDA-induced spasms than controls (p = 0.047). This new model of MCD-based epileptic spasms with corresponding features of human spasms will be valuable for future research of the developmental epilepsy.
Collapse
Affiliation(s)
- Eun-Hee Kim
- Department of Pediatrics, CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Mi-Sun Yum
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, South Korea
| | - Minyoung Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Jin Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, South Korea
| | - Woo-Hyun Shim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Tae-Sung Ko
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Galanopoulou AS, Mowrey WB, Liu W, Li Q, Shandra O, Moshé SL. Preclinical Screening for Treatments for Infantile Spasms in the Multiple Hit Rat Model of Infantile Spasms: An Update. Neurochem Res 2017; 42:1949-1961. [PMID: 28462453 DOI: 10.1007/s11064-017-2282-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 04/20/2017] [Accepted: 04/22/2017] [Indexed: 12/16/2022]
Abstract
Infantile spasms are the typical seizures of West syndrome, an infantile epileptic encephalopathy with poor outcomes. There is an increasing need to identify more effective and better tolerated treatments for infantile spasms. We have optimized the rat model of infantile spasms due to structural etiology, the multiple-hit rat model, for therapy discovery. Here, we test three compounds administered after spasms induction in the multiple hit model for efficacy and tolerability. Specifically, postnatal day 3 (PN3) male Sprague-Dawley rats were induced by right intracerebral injections of doxorubicin and lipopolysaccharide. On PN5 p-chlorophenylalanine was given intraperitoneally (i.p.). Daily monitoring of weights and developmental milestones was done and rats were intermittently video monitored. A blinded, randomized, vehicle-controlled study design was followed. The caspase 1 inhibitor VX-765 (50-200 mg/kg i.p.) and the GABAB receptor inhibitor CGP35348 (12.5-100 mg/kg i.p.) each was administered in different cohorts as single intraperitoneal injections on PN4, using a dose- and time-response design with intermittent monitoring till PN5. 17β-estradiol (40 ng/g/day subcutaneously) was given daily between PN3-10 and intermittent monitoring was done till PN12. None of the treatments demonstrated acute or delayed effects on spasms, yet all were well tolerated. We discuss the implications for therapy discovery and challenges of replication trials.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA.
- Dominick P. Purpura Department of Neuroscience, Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 306, Bronx, NY, 10461, USA.
| | - Wenzhu B Mowrey
- Division of Biostatistics, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Oleksii Shandra
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 306, Bronx, NY, 10461, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
33
|
Animal Models of Seizures and Epilepsy: Past, Present, and Future Role for the Discovery of Antiseizure Drugs. Neurochem Res 2017; 42:1873-1888. [PMID: 28290134 DOI: 10.1007/s11064-017-2222-z] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
The identification of potential therapeutic agents for the treatment of epilepsy requires the use of seizure models. Except for some early treatments, including bromides and phenobarbital, the antiseizure activity of all clinically used drugs was, for the most part, defined by acute seizure models in rodents using the maximal electroshock and subcutaneous pentylenetetrazole seizure tests and the electrically kindled rat. Unfortunately, the clinical evidence to date would suggest that none of these models, albeit useful, are likely to identify those therapeutics that will effectively manage patients with drug resistant seizures. Over the last 30 years, a number of animal models have been developed that display varying degrees of pharmacoresistance, such as the phenytoin- or lamotrigine-resistant kindled rat, the 6-Hz mouse model of partial seizures, the intrahippocampal kainate model in mice, or rats in which spontaneous recurrent seizures develops after inducing status epilepticus by chemical or electrical stimulation. As such, these models can be used to study mechanisms of drug resistance and may provide a unique opportunity for identifying a truly novel antiseizure drug (ASD), but thus far clinical evidence for this hope is lacking. Although animal models of drug resistant seizures are now included in ASD discovery approaches such as the ETSP (epilepsy therapy screening program), it is important to note that no single model has been validated for use to identify potential compounds for as yet drug resistant seizures, but rather a battery of such models should be employed, thus enhancing the sensitivity to discover novel, highly effective ASDs. The present review describes the previous and current approaches used in the search for new ASDs and offers some insight into future directions incorporating new and emerging animal models of therapy resistance.
Collapse
|
34
|
Shandra O, Moshé SL, Galanopoulou AS. Inflammation in Epileptic Encephalopathies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 108:59-84. [PMID: 28427564 DOI: 10.1016/bs.apcsb.2017.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
West syndrome (WS) is an infantile epileptic encephalopathy that manifests with infantile spasms (IS), hypsarrhythmia (in ~60% of infants), and poor neurodevelopmental outcomes. The etiologies of WS can be structural-metabolic pathologies (~60%), genetic (12%-15%), or of unknown origin. The current treatment options include hormonal treatment (adrenocorticotropic hormone and high-dose steroids) and the GABA aminotransferase inhibitor vigabatrin, while ketogenic diet can be given as add-on treatment in refractory IS. There is a need to identify new therapeutic targets and more effective treatments for WS. Theories about the role of inflammatory pathways in the pathogenesis and treatment of WS have emerged, being supported by both clinical and preclinical data from animal models of WS. Ongoing advances in genetics have revealed numerous genes involved in the pathogenesis of WS, including genes directly or indirectly involved in inflammation. Inflammatory pathways also interact with other signaling pathways implicated in WS, such as the neuroendocrine pathway. Furthermore, seizures may also activate proinflammatory pathways raising the possibility that inflammation can be a consequence of seizures and epileptogenic processes. With this targeted review, we plan to discuss the evidence pro and against the following key questions. Does activation of inflammatory pathways in the brain cause epilepsy in WS and does it contribute to the associated comorbidities and progression? Can activation of certain inflammatory pathways be a compensatory or protective event? Are there interactions between inflammation and the neuroendocrine system that contribute to the pathogenesis of WS? Does activation of brain inflammatory signaling pathways contribute to the transition of WS to Lennox-Gastaut syndrome? Are there any lead candidates or unexplored targets for future therapy development for WS targeting inflammation?
Collapse
Affiliation(s)
- Oleksii Shandra
- Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Solomon L Moshé
- Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, United States; Montefiore/Einstein Epilepsy Center, Montefiore Medical Center, Bronx, NY, United States
| | - Aristea S Galanopoulou
- Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, NY, United States; Montefiore/Einstein Epilepsy Center, Montefiore Medical Center, Bronx, NY, United States.
| |
Collapse
|
35
|
Ismail FY, Fatemi A, Johnston MV. Cerebral plasticity: Windows of opportunity in the developing brain. Eur J Paediatr Neurol 2017; 21:23-48. [PMID: 27567276 DOI: 10.1016/j.ejpn.2016.07.007] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/06/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Neuroplasticity refers to the inherently dynamic biological capacity of the central nervous system (CNS) to undergo maturation, change structurally and functionally in response to experience and to adapt following injury. This malleability is achieved by modulating subsets of genetic, molecular and cellular mechanisms that influence the dynamics of synaptic connections and neural circuitry formation culminating in gain or loss of behavior or function. Neuroplasticity in the healthy developing brain exhibits a heterochronus cortex-specific developmental profile and is heightened during "critical and sensitive periods" of pre and postnatal brain development that enable the construction and consolidation of experience-dependent structural and functional brain connections. PURPOSE In this review, our primary goal is to highlight the essential role of neuroplasticity in brain development, and to draw attention to the complex relationship between different levels of the developing nervous system that are subjected to plasticity in health and disease. Another goal of this review is to explore the relationship between plasticity responses of the developing brain and how they are influenced by critical and sensitive periods of brain development. Finally, we aim to motivate researchers in the pediatric neuromodulation field to build on the current knowledge of normal and abnormal neuroplasticity, especially synaptic plasticity, and their dependence on "critical or sensitive periods" of neural development to inform the design, timing and sequencing of neuromodulatory interventions in order to enhance and optimize their translational applications in childhood disorders of the brain. METHODS literature review. RESULTS We discuss in details five patterns of neuroplasticity expressed by the developing brain: 1) developmental plasticity which is further classified into normal and impaired developmental plasticity as seen in syndromic autism spectrum disorders, 2) adaptive (experience-dependent) plasticity following intense motor skill training, 3) reactive plasticity to pre and post natal CNS injury or sensory deprivation, 4) excessive plasticity (loss of homeostatic regulation) as seen in dystonia and refractory epilepsy, 6) and finally, plasticity as the brain's "Achilles tendon" which induces brain vulnerability under certain conditions such as hypoxic ischemic encephalopathy and epileptic encephalopathy syndromes. We then explore the unique feature of "time-sensitive heightened plasticity responses" in the developing brain in the in the context of neuromodulation. CONCLUSION The different patterns of neuroplasticity and the unique feature of heightened plasticity during critical and sensitive periods are important concepts for researchers and clinicians in the field of pediatric neurology and neurodevelopmental disabilities. These concepts need to be examined systematically in the context of pediatric neuromodulation. We propose that critical and sensitive periods of brain development in health and disease can create "windows of opportunity" for neuromodulatory interventions that are not commonly seen in adult brain and probably augment plasticity responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Fatima Yousif Ismail
- Department of neurology and developmental medicine, The Kennedy Krieger Institute, Johns Hopkins Medical Institutions, MD, USA; Department of pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al- Ain, UAE.
| | - Ali Fatemi
- Departments of Neurology and Pediatrics, The Kennedy Krieger Institute, and Johns Hopkins University School of Medicine, MD, USA
| | - Michael V Johnston
- Departments of Neurology and Pediatrics, The Kennedy Krieger Institute, and Johns Hopkins University School of Medicine, MD, USA
| |
Collapse
|
36
|
Galanopoulou AS, Mowrey WB. Not all that glitters is gold: A guide to critical appraisal of animal drug trials in epilepsy. Epilepsia Open 2016; 1:86-101. [PMID: 28497130 PMCID: PMC5421644 DOI: 10.1002/epi4.12021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Preclinical studies have produced numerous drugs with antiseizure properties that currently are the standard of care. One third of the human population with epilepsy still continues to have seizures despite the ongoing discoveries. The recognized clinical gaps of care that need to be addressed are the identification of antiepileptogenic and disease‐modifying treatments, and treatments for refractory seizures or for seizures and epilepsies with limited or unsatisfactory treatments, such as early life epileptic encephalopathies. In this invited review, we provide a historical summary of the international efforts to reevaluate the strategies adopted in preclinical epilepsy therapy discovery studies. We discuss issues that may affect the quality, interpretation, and validation of preclinical studies and their translation to successful therapies for humans affected with epilepsy. These include the selection of animal models and the study design; research practices that affect rigor (such as appropriate use of statistics and reporting of study methods and results, their validation across models, labs, and preclinical‐clinical studies); the need to harmonize research methods and outcome assessment; and the importance of improving translation to clinically appropriate situations. The epilepsy research community is incrementally adopting collaborative research, including consortia or multicenter studies to meet these needs. Improving the infrastructure that can support these efforts will be instrumental in future success.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Montefiore / Einstein Epilepsy Center, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx NY USA
| | - Wenzhu B Mowrey
- Division of Biostatistics, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx NY USA
| |
Collapse
|
37
|
Joshi K, Shen L, Michaeli A, Salter M, Thibault-Messier G, Hashmi S, Eubanks JH, Cortez MA, Snead OC. Infantile spasms in down syndrome: Rescue by knockdown of the GIRK2 channel. Ann Neurol 2016; 80:511-21. [PMID: 27462820 DOI: 10.1002/ana.24749] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The Ts65Dn (Ts) mouse model of Down syndrome (DS) is exquisitely sensitive to an infantile spasms phenotype induced by γ-aminobutyric acidB receptor (GABAB R) agonists. The Ts mouse contains the core genomic triplication of the DS critical region, which includes 3 copies of the Kcnj6 gene that encodes the GABAB R-coupled G protein-coupled inward rectifying potassium channel subunit 2 (GIRK2) channel. We test the hypothesis that GIRK2 is necessary for the GABAB R agonist-induced infantile spasms phenotype in Ts. METHODS We assessed the result of either genetic or pharmacological knockdown of the GIRK2 channel in Ts brain upon the GABAB R agonist-induced infantile spasms phenotype in the Ts mouse model of DS. As well, we examined GABAB R currents in hippocampal neurons prepared from GIRK2-trisomic Ts control mice and GIRK2-disomic Ts mice in which Kcnj6 had been genetically knocked down from 3 to 2 copies. RESULTS The reduction of the copy number of Kcnj6 in Ts mice rescued the GABAB R agonist-induced infantile spasms phenotype. There was an increase in GABAB R-mediated GIRK2 currents in GIRK2-trisomic Ts mouse hippocampal neurons, which were normalized in the GIRK2-disomic Ts mice. Similarly, pharmacological knockdown of the GIRK2 channel in Ts brain using the GIRK antagonist tertiapin-Q also rescued the GABAB R agonist-induced infantile spasms phenotype in Ts mutants. INTERPRETATION The GABAB R-coupled GIRK2 channel is necessary for the GABAB R agonist-induced infantile spasms phenotype in the Ts mouse and may represent a novel therapeutic target for the treatment of infantile spasms in DS. Ann Neurol 2016;80:511-521.
Collapse
Affiliation(s)
- Krutika Joshi
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada. .,Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Lily Shen
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Avner Michaeli
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Salter
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Sumaiya Hashmi
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - James H Eubanks
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,University Health Network, Toronto Western Research Institute, Toronto, Ontario, Canada
| | - Miguel A Cortez
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - O Carter Snead
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Fit for purpose application of currently existing animal models in the discovery of novel epilepsy therapies. Epilepsy Res 2016; 126:157-84. [PMID: 27505294 DOI: 10.1016/j.eplepsyres.2016.05.016] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/06/2016] [Accepted: 05/30/2016] [Indexed: 01/10/2023]
Abstract
Animal seizure and epilepsy models continue to play an important role in the early discovery of new therapies for the symptomatic treatment of epilepsy. Since 1937, with the discovery of phenytoin, almost all anti-seizure drugs (ASDs) have been identified by their effects in animal models, and millions of patients world-wide have benefited from the successful translation of animal data into the clinic. However, several unmet clinical needs remain, including resistance to ASDs in about 30% of patients with epilepsy, adverse effects of ASDs that can reduce quality of life, and the lack of treatments that can prevent development of epilepsy in patients at risk following brain injury. The aim of this review is to critically discuss the translational value of currently used animal models of seizures and epilepsy, particularly what animal models can tell us about epilepsy therapies in patients and which limitations exist. Principles of translational medicine will be used for this discussion. An essential requirement for translational medicine to improve success in drug development is the availability of animal models with high predictive validity for a therapeutic drug response. For this requirement, the model, by definition, does not need to be a perfect replication of the clinical condition, but it is important that the validation provided for a given model is fit for purpose. The present review should guide researchers in both academia and industry what can and cannot be expected from animal models in preclinical development of epilepsy therapies, which models are best suited for which purpose, and for which aspects suitable models are as yet not available. Overall further development is needed to improve and validate animal models for the diverse areas in epilepsy research where suitable fit for purpose models are urgently needed in the search for more effective treatments.
Collapse
|
39
|
Current understanding and neurobiology of epileptic encephalopathies. Neurobiol Dis 2016; 92:72-89. [DOI: 10.1016/j.nbd.2016.03.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 03/01/2016] [Accepted: 03/09/2016] [Indexed: 12/25/2022] Open
|
40
|
Affiliation(s)
- Carl E. Stafstrom
- Division of Pediatric Neurology, John M. Freeman Pediatric Epilepsy Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eric H. Kossoff
- Division of Pediatric Neurology, John M. Freeman Pediatric Epilepsy Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
41
|
|
42
|
Dhindsa RS, Goldstein DB. Genetic Discoveries Drive Molecular Analyses and Targeted Therapeutic Options in the Epilepsies. Curr Neurol Neurosci Rep 2016; 15:70. [PMID: 26319171 DOI: 10.1007/s11910-015-0587-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Epilepsy is a serious neurological disease with substantial genetic contribution. We have recently made major advances in understanding the genetics and etiology of the epilepsies. However, current antiepileptic drugs are ineffective in nearly one third of patients. Most of these drugs were developed without knowledge of the underlying causes of the epilepsy to be treated; thus, it seems reasonable to assume that further improvements require a deeper understanding of epilepsy pathophysiology. Although once the rate-limiting step, gene discovery is now occurring at an unprecedented rapid rate, especially in the epileptic encephalopathies. However, to place these genetic findings in a biological context and discover treatment options for patients, we must focus on developing an efficient framework for functional evaluation of the mutations that cause epilepsy. In this review, we discuss guidelines for gene discovery, emerging functional assays and models, and novel therapeutics to highlight the developing framework of precision medicine in the epilepsies.
Collapse
Affiliation(s)
- Ryan S Dhindsa
- Institute for Genomic Medicine, Columbia University, Hammer Building, 701 West 168th Street, Box 149, New York, NY, 10032, USA,
| | | |
Collapse
|
43
|
Shao LR, Stafstrom CE. Pediatric Epileptic Encephalopathies: Pathophysiology and Animal Models. Semin Pediatr Neurol 2016; 23:98-107. [PMID: 27544466 DOI: 10.1016/j.spen.2016.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Epileptic encephalopathies are syndromes in which seizures or interictal epileptiform activity contribute to or exacerbate brain function, beyond that caused by the underlying pathology. These severe epilepsies begin early in life, are associated with poor lifelong outcome, and are resistant to most treatments. Therefore, they represent an immense challenge for families and the medical care system. Furthermore, the pathogenic mechanisms underlying the epileptic encephalopathies are poorly understood, hampering attempts to devise novel treatments. This article reviews animal models of the three classic epileptic encephalopathies-West syndrome (infantile spasms), Lennox-Gastaut syndrome, and continuous spike waves during sleep or Landau-Kleffner syndrome-with discussion of how animal models are revealing underlying pathophysiological mechanisms that might be amenable to targeted therapy.
Collapse
Affiliation(s)
- Li-Rong Shao
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
44
|
Franco V, French JA, Perucca E. Challenges in the clinical development of new antiepileptic drugs. Pharmacol Res 2016; 103:95-104. [DOI: 10.1016/j.phrs.2015.11.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 12/26/2022]
|
45
|
Galanopoulou AS, Moshé SL. Neonatal and Infantile Epilepsy: Acquired and Genetic Models. Cold Spring Harb Perspect Med 2015; 6:a022707. [PMID: 26637437 DOI: 10.1101/cshperspect.a022707] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The incidence of seizures and epilepsies is particularly high during the neonatal and infantile periods. We will review selected animal models of early-life epileptic encephalopathies that have addressed the dyscognitive features of frequent interictal spikes, the pathogenesis and treatments of infantile spasms (IS) or Dravet syndrome, disorders with mammalian target of rapamycin (mTOR) dysregulation, and selected early-life epilepsies with genetic defects. Potentially pathogenic mechanisms in these conditions include interneuronopathies in IS or Dravet syndrome and mTOR dysregulation in brain malformations, tuberous sclerosis, and related genetic disorders, or IS of acquired etiology. These models start to generate the first therapeutic drugs, which have been specifically developed in immature animals. However, there are challenges in translating preclinical discoveries into clinically relevant findings. The advances made so far hold promise that the new insights may potentially have curative or disease-modifying potential for many of these devastating conditions.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York 10461 Department of Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
46
|
Sánchez Fernández I, Loddenkemper T, Galanopoulou AS, Moshé SL. Should epileptiform discharges be treated? Epilepsia 2015; 56:1492-504. [PMID: 26293670 DOI: 10.1111/epi.13108] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2015] [Indexed: 01/09/2023]
Abstract
To evaluate the impact of epileptiform discharges (EDs) that do not occur within seizure patterns--such as spikes, sharp waves or spike waves--on cognitive function and to discuss the circumstances under which treatment of EDs might be considered. Methods used in this article is "Review of the literature". EDs may disrupt short-term cognition in humans. Frequent EDs for a prolonged period can potentially impair long-term cognitive function in humans. However, there is conflicting evidence on the impact of EDs on long-term cognitive outcome because this relationship may be confounded by multiple factors such as underlying etiology, seizures, and medication effects. Limitations of existing studies include the lack of standardized ED quantification methods and of widely accepted automated spike quantification methods. Although there is no solid evidence for or against treatment of EDs, a non-evidence-based practical approach is suggested. EDs in otherwise asymptomatic individuals should not be treated because the risks of treatment probably outweigh its dubious benefits. A treatment trial for EDs may be considered when there is cognitive dysfunction or regression or neurologic symptoms that are unexplained by the underlying etiology, comorbid conditions, or seizure severity. In patients with cognitive or neurologic dysfunction with epilepsy or EDs, treatment may be warranted to control the underlying epileptic syndrome. EDs may cause cognitive or neurologic dysfunction in humans in the short term. There is conflicting evidence on the impact of EDs on long-term cognitive outcome. There is no evidence for or against treatment of asymptomatic ED.
Collapse
Affiliation(s)
- Iván Sánchez Fernández
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, U.S.A.,Department of Child Neurology, Hospital Sant Joan de Déu, University of Barcelona, Spain
| | - Tobias Loddenkemper
- Department of Child Neurology, Hospital Sant Joan de Déu, University of Barcelona, Spain
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Medicine, Montefiore/Einstein Epilepsy Management Center, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, U.S.A
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Medicine, Montefiore/Einstein Epilepsy Management Center, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, U.S.A.,Department of Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, U.S.A
| |
Collapse
|