1
|
García-García L, Gómez-Oliver F, Fernández de la Rosa R, Pozo MÁ. Dantrolene paradoxically exacerbates short-term brain glucose hypometabolism, hippocampal damage and neuroinflammation induced by status epilepticus in the rat lithium-pilocarpine model. Eur J Pharmacol 2024; 985:177073. [PMID: 39481630 DOI: 10.1016/j.ejphar.2024.177073] [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: 08/08/2024] [Revised: 09/23/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Status epilepticus (SE) is a neurologic emergency characterized by prolonged or rapidly recurring seizures. Increased intracellular calcium concentration ([Ca2+]i) occurring after SE is a key mediator of excitotoxicity that contributes to the brain damage associated with the development of epilepsy. Accumulated evidence indicates that dantrolene, a ryanodine receptor (RyR) blocker may have protective effects against the SE-induced damage. We evaluated whether dantrolene (10 mg/kg, i.p.) administered twice, 5 min and 24 h after the lithium-pilocarpine-induced SE in rats, had neuroprotective effects. Dantrolene by itself had no effects on control rats. However, it exacerbated the signs of damage in rats that underwent SE, increasing brain glucose hypometabolism as measured by PET neuroimaging 3 days after SE. Likewise, the neurohistochemical studies revealed that dantrolene aggravated signs of hippocampal neurodegeneration, neuronal death and microglia-induced neuroinflammation. Besides, the damaging effects were reflected by severe body weight loss. Overall, our results point towards a deleterious effect of dantrolene in the lithium-pilocarpine-induced SE model. Nonetheless, our results are in opposition to the reported neuroprotective effects of dantrolene. Whether the mechanisms underlying [Ca2+]i increase might significantly differ depending on the particularities of the model of epilepsy used and general experimental conditions need further studies. Besides, it is yet to be determined which isoform of RyRs significantly contributes to Ca2+-induced excitotoxicity in the lithium-pilocarpine SE rat model.
Collapse
Affiliation(s)
- Luis García-García
- Department of Pharmacology, Pharmacognosy and Botany. Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Brain Mapping Unit, Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain; Health Research Institute, Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Francisca Gómez-Oliver
- Department of Pharmacology, Pharmacognosy and Botany. Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Brain Mapping Unit, Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain; Health Research Institute, Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Rubén Fernández de la Rosa
- Brain Mapping Unit, Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain; ICTS Bioimagen Complutense (BIOIMAC), Complutense University of Madrid, Madrid, Spain
| | - Miguel Ángel Pozo
- Brain Mapping Unit, Instituto Pluridisciplinar, Complutense University of Madrid, Madrid, Spain; Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain; Health Research Institute, Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
2
|
Maciel CB, Ahmad B, Jose Bruzzone Giraldez M, Eisenschenk S, Ramsay E, Maranchick NF, Peloquin CA, Hirsch L, Busl KM. Early vigabatrin to augment GABAergic pathways in post-anoxic status epilepticus. Epilepsy Behav 2024; 160:110082. [PMID: 39393141 DOI: 10.1016/j.yebeh.2024.110082] [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: 09/09/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
The outcomes of patients who experience status epilepticus during the post-cardiac arrest period, or post-anoxic status epilepticus (PASE), remain dismal despite advances in resuscitation. The combination of therapeutic nihilism and the refractoriness of seizures in a setting where pessimistic prognostic impressions prevail is likely the main driver of such poor outcomes. The resulting pervasive vicious cycle perpetuates this knowledge gap, whereby hypoxic-ischemic insults as the etiology for seizures remain a ubiquitous exclusion criterion for clinal trials in status epilepticus. Effective therapies targeting hyperexcitability resulting from hypoxic-ischemic brain injury are urgently needed. Early inhibition of gamma-aminobutyric acid (GABA) transaminase with vigabatrin holds potential as an effective adjunctive therapy for PASE. This scientific premise is based on the resulting halted GABA catabolism thereby promoting synergistic augmentation of GABAergic pathway when used in combination with positive GABAergic allosteric modulators. This paper is based on a lecture presented at the 9th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures, in London 8-10 April 2024.
Collapse
Affiliation(s)
- Carolina B Maciel
- Department of Neurology, Division of Neurocritical Care, University of Florida College of Medicine, Gainesville, FL 32611, USA; Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA.
| | - Bakhtawar Ahmad
- Department of Neurology, Division of Neurocritical Care, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Maria Jose Bruzzone Giraldez
- Department of Neurology, Division of Epilepsy, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Stephan Eisenschenk
- Department of Neurology, Division of Epilepsy, University of Florida College of Medicine, Gainesville, FL 32611, USA; North Florida/South Georgia Department of Veterans Affairs Medical Center, Gainesville, FL 32608, USA; Veterans Affairs Medical Center National TeleEEG Program, Gainesville, FL 32608, USA
| | - Eugene Ramsay
- Department of Neurology, University of South Alabama, Mobile, AL 36688, USA
| | - Nicole F Maranchick
- Department of Pharmacotherapy & Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32611, USA
| | - Charles A Peloquin
- Department of Pharmacotherapy & Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32611, USA
| | - Lawrence Hirsch
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Katharina M Busl
- Department of Neurology, Division of Neurocritical Care, University of Florida College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
3
|
Kyllo T, Allocco D, Hei LV, Wulff H, Erickson JD. Riluzole attenuates acute neural injury and reactive gliosis, hippocampal-dependent cognitive impairments and spontaneous recurrent generalized seizures in a rat model of temporal lobe epilepsy. Front Pharmacol 2024; 15:1466953. [PMID: 39539628 PMCID: PMC11558044 DOI: 10.3389/fphar.2024.1466953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Background Riluzole exhibits neuroprotective and therapeutic effects in several neurological disease models associated with excessive synaptic glutamate (Glu) release. We recently showed riluzole prevents acute excitotoxic hippocampal neural injury at 3 days in the kainic acid (KA) model of temporal lobe epilepsy (TLE). Currently, it is unknown if preventing acute neural injury and the neuroinflammatory response is sufficient to suppress epileptogenesis. Methods The KA rat model of TLE was used to determine if riluzole attenuates acute hippocampal neural injury and reactive gliosis. KA was administered to adult male Sprague-Dawley (250 g) rats at 5 mg/kg/hr until status epilepticus (SE) was observed, and riluzole was administered at 10 mg/kg 1 h and 4 h after SE and once per day for the next 2 days. Immunostaining was used to assess neural injury (FJC and NeuN), microglial activation (Iba1 and ED-1/CD68) and astrogliosis (GFAP and vimentin) at day 7 and day 14 after KA-induced SE. Learning and memory tests (Y-maze, Novel object recognition test, Barnes maze), behavioral hyperexcitability tests, and spontaneous generalized recurrent seizure (SRS) activity (24-hour video monitoring) were assessed at 11-15 weeks. Results Here we show that KA-induced hippocampal neural injury precedes the neuroimmune response and that riluzole attenuates acute neural injury, microglial activation, and astrogliosis at 7 and 14 days. We find that reducing acute hippocampal injury and the associated neuroimmune response following KA-induced SE by riluzole attenuates hippocampal-dependent cognitive impairment, behavioral hyperexcitability, and tonic/clonic generalized SRS activity after 3 months. We also show that riluzole attenuates SE-associated body weight loss during the first week after KA-induced SE. Discussion Riluzole acts on multiple targets that are involved to prevent excessive synaptic Glu transmission and excitotoxic neuronal injury. Attenuating KA-induced neural injury and subsequent microglia/astrocyte activation in the hippocampus and extralimbic regions with riluzole reduces TLE-associated cognitive deficits and generalized SRS and suggests that riluzole could be a potential antiepileptogenic drug.
Collapse
Affiliation(s)
- Thomas Kyllo
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, United States
| | - Dominic Allocco
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, United States
| | - Laine Vande Hei
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, CA, United States
| | - Jeffrey D. Erickson
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, United States
| |
Collapse
|
4
|
Löscher W. Mammalian models of status epilepticus - Their value and limitations. Epilepsy Behav 2024; 158:109923. [PMID: 38944026 DOI: 10.1016/j.yebeh.2024.109923] [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: 05/24/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Status epilepticus (SE) is a medical and neurologic emergency that may lead to permanent brain damage, morbidity, or death. Animal models of SE are particularly important to study the pathophysiology of SE and mechanisms of SE resistance to antiseizure medications with the aim to develop new, more effective treatments. In addition to rodents (rats or mice), larger mammalian species such as dogs, pigs, and nonhuman primates are used. This short review describes and discusses the value and limitations of the most frequently used mammalian models of SE. Issues that are discussed include (1) differences between chemical and electrical SE models; (2) the role of genetic background and environment on SE in rodents; (3) the use of rodent models (a) to study the pathophysiology of SE and mechanisms of SE resistance; (b) to study developmental aspects of SE; (c) to study the efficacy of new treatments, including drug combinations, for refractory SE; (d) to study the long-term consequences of SE and identify biomarkers; (e) to develop treatments that prevent or modify epilepsy; (e) to study the pharmacology of spontaneous seizures; (4) the limitations of animal models of induced SE; and (5) the advantages (and limitations) of naturally (spontaneously) occurring SE in epileptic dogs and nonhuman primates. Overall, mammalian models of SE have significantly increased our understanding of the pathophysiology and drug resistance of SE and identified potential targets for new, more effective treatments. This paper was presented at the 9th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures held in April 2024.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
5
|
Xiao C, Gu X, Feng Y, Shen J. Two-sample Mendelian randomization analysis of 91 circulating inflammatory protein levels and amyotrophic lateral sclerosis. Front Aging Neurosci 2024; 16:1367106. [PMID: 38601850 PMCID: PMC11004327 DOI: 10.3389/fnagi.2024.1367106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease with poorly understood pathophysiology. Recent studies have highlighted systemic inflammation, especially the role of circulating inflammatory proteins, in ALS. Methods This study investigates the potential causal link between these proteins and ALS. We employed a two-sample Mendelian Randomization(MR) approach, analyzing data from large-scale genome-wide association studies to explore the relationship between 91 circulating inflammatory proteins and ALS. This included various MR methods like MR Egger, weighted median, and inverse-variance weighted, complemented by sensitivity analyses for robust results. Results Significant associations were observed between levels of inflammatory proteins, including Adenosine Deaminase, Interleukin-17C, Oncostatin-M, Leukemia Inhibitory Factor Receptor, and Osteoprotegerin, and ALS risk. Consistencies were noted across different P-value thresholds. Bidirectional MR suggested that ALS risk might influence levels of certain inflammatory proteins. Discussion Our findings, via MR analysis, indicate a potential causal relationship between circulating inflammatory proteins and ALS. This sheds new light on ALS pathophysiology and suggests possible therapeutic targets. Further research is required to confirm these results and understand the specific roles of these proteins in ALS.
Collapse
Affiliation(s)
- Chenxu Xiao
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Xiaochu Gu
- Medical Laboratory, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Yu Feng
- The University of New South Wales, Kensington, NSW, Australia
- The University of Melbourne, Parkville, VIC, Australia
| | - Jing Shen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
6
|
Zierath DK, Davidson S, Manoukian J, White HS, Meeker S, Ericsson A, Barker-Haliski M. Diet composition and sterilization modifies intestinal microbiome diversity and burden of Theiler's virus infection-induced acute seizures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562694. [PMID: 37905123 PMCID: PMC10614857 DOI: 10.1101/2023.10.17.562694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Objective Central nervous system infection with Theiler's murine encephalomyelitis virus (TMEV) in C57BL/6J mice can model acquired epileptogenesis. Diet alters the acute seizure incidence in TMEV-infected mice; yet it is unclear whether intestinal dysbiosis may also impact acute or chronic behavioral comorbidities. This study thus assessed the impact of diet sterilization in a specific pathogen-free vivarium on acute seizure presentation, the composition of the gut microbiome, and chronic behavioral comorbidities of epilepsy. Methods Baseline fecal samples were collected from male C57BL/6J mice (4-5 weeks-old; Jackson Labs) upon arrival. Mice were randomized to either autoclaved (AC) or irradiated (IR) diet (Prolab RMH 3000 - UU diets) or IR (Picolab 5053 - UW IR diet). Mice then underwent intracerebral TMEV or PBS injection three days later. Fecal samples were collected from a subset of mice at infection (Day 0) and Day 7 post-infection. Epilepsy-related working memory deficits and seizure threshold were assessed 6 weeks post-infection. Gut microbiome diversity was determined by 16S rRNA amplicon sequencing of fecal samples. Results TMEV-infected mice displayed acute handling-induced seizures, regardless of diet: 28/57 UW IR (49.1%), 30/41 UU IR (73.2%), and 47/77 UU AC (61%) mice displayed seizures. The number of observed seizures significantly differed: UW IR mice had 2.2±2.8 seizures (mean±standard deviation), UU IR mice had 3.5±2.9 seizures, and UU AC mice had 4.4±3.8 seizures during the 7-day monitoring period. The composition of the gut microbiome significantly differed in TMEV-infected mice fed the UU AC diet, with most measured differences occurring in Gram-positive bacteria. TMEV-infected mice fed the UU AC diet displayed worsened chronic working memory. Significance Intestinal dysbiosis evokes stark differences in acute seizure presentation in the TMEV model and vastly influences the trajectory of post-TMEV infection-induced behavioral comorbidities of epilepsy. Our study reveals a novel disease-modifying contribution of intestinal bacterial species after TMEV-induced acute seizures.
Collapse
Affiliation(s)
- Dannielle K. Zierath
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Stephanie Davidson
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Jonathan Manoukian
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - H. Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA
| | - Stacey Meeker
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | - Aaron Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | | |
Collapse
|
7
|
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
|
8
|
Guarino A, Bettegazzi B, Aziz N, Barbieri M, Bochicchio D, Crippa L, Marino P, Sguizzato M, Soukupova M, Zucchini S, Simonato M. Low-dose 7,8-Dihydroxyflavone Administration After Status Epilepticus Prevents Epilepsy Development. Neurotherapeutics 2022; 19:1951-1965. [PMID: 36180719 PMCID: PMC9723075 DOI: 10.1007/s13311-022-01299-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2022] [Indexed: 12/14/2022] Open
Abstract
Temporal lobe epilepsy often manifests months or even years after an initial epileptogenic insult (e.g., stroke, trauma, status epilepticus) and, therefore, may be preventable. However, no such preventive treatment is currently available. Aim of this study was to test an antioxidant agent, 7,8-dihydroxyflavone (7,8-DHF), that is well tolerated and effective in preclinical models of many neurological disorders, as an anti-epileptogenic drug. However, 7,8-DHF also acts as a TrkB receptor agonist and, based on the literature, this effect may imply an anti- or a pro-epileptogenic effect. We found that low- (5 mg/kg), but not high-dose 7,8-DHF (10 mg/kg) can exert strong anti-epileptogenic effects in the lithium-pilocarpine model (i.e., highly significant reduction in the frequency of spontaneous seizures and in the time to first seizure after status epilepticus). The mechanism of these different dose-related effects remains to be elucidated. Nonetheless, considering its excellent safety profile and antioxidant properties, as well as its putative effects on TrkB receptors, 7,8-DHF represents an interesting template for the development of effective and well-tolerated anti-epileptogenic drugs.
Collapse
Affiliation(s)
- Annunziata Guarino
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Barbara Bettegazzi
- University Vita-Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy
| | - Nimra Aziz
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Mario Barbieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Daniela Bochicchio
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Lucia Crippa
- University Vita-Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy
| | - Pietro Marino
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Maddalena Sguizzato
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Marie Soukupova
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy.
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy.
| | - Michele Simonato
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
- Division of Neuroscience, IRCCS San Raffaele Hospital, via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
9
|
Mallmann MP, Mello FK, Neuberger B, Sobral KG, Fighera MR, Royes LFF, Furian AF, Oliveira MS. Beta-caryophyllene attenuates short-term recurrent seizure activity and blood-brain-barrier breakdown after pilocarpine-induced status epilepticus in rats. Brain Res 2022; 1784:147883. [PMID: 35300975 DOI: 10.1016/j.brainres.2022.147883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/18/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Status epilepticus (SE) is a neurological life-threatening condition, resulting from the failure of the mechanisms responsible for seizure termination. SE is often pharmacoresistant and associated with significant morbidity and mortality. Hence, ceasing or attenuating SE and its consequences is of fundamental importance. Beta-caryophyllene is a functional CB2 receptor agonist and exhibit a good safety profile. Besides, it displays beneficial effects in several experimental conditions, including neuroprotective activity. In the present study we aimed to investigate the effects of beta-caryophyllene on pilocarpine-induced SE. METHODS Wistar rats were submitted to pilocarpine-induced SE and monitored for 24 hours by video and EEG for short-term recurrence of seizure activity (i.e. seizures occurring within 24 hours after termination of SE). Rats received beta-caryophyllene (100 mg/kg, ip) at 1, 8- and 16-hours after SE. Twenty-four hours after SE we evaluated sensorimotor response, neuronal damage (fluoro jade C staining) and serum albumin infiltration into brain parenchyma. RESULTS Beta-caryophyllene-treated animals presented fewer short-term recurrent seizures than vehicle-treated counterparts, suggesting an anticonvulsant effect after SE. Behavioral recovery from SE and the number of fluoro jade C positive cells in the hippocampus and thalamus were not modified by beta-caryophyllene. Treatment with beta-caryophyllene attenuated the SE-induced increase of albumin immunoreactivity in the hippocampus, indicating a protective effect against blood-brain-barrier breakdown. CONCLUSIONS Given the inherent difficulties in the treatment of SE and its consequences, present results suggest that beta-caryophyllene deserve further investigation as an adjuvant therapeutic strategy for SE.
Collapse
Affiliation(s)
| | | | - Bruna Neuberger
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.
| | - Karine Gabriela Sobral
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.
| | - Michele Rechia Fighera
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.
| | - Luiz Fernando Freire Royes
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil.
| | - Ana Flávia Furian
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil; Graduate Program in Food Science and Technology, Federal University of Santa Maria, Santa Maria, Brazil.
| | | |
Collapse
|
10
|
Eid T. Progressive Neuronal Loss in Epilepsy - A Long-Standing Conundrum Finally Resolved? Epilepsy Curr 2021; 21:366-368. [PMID: 34924838 PMCID: PMC8655263 DOI: 10.1177/15357597211030385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
11
|
Jahreis I, Bascuñana P, Ross TL, Bankstahl JP, Bankstahl M. Choice of anesthesia and data analysis method strongly increases sensitivity of 18F-FDG PET imaging during experimental epileptogenesis. PLoS One 2021; 16:e0260482. [PMID: 34818362 PMCID: PMC8612569 DOI: 10.1371/journal.pone.0260482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose Alterations in brain glucose metabolism detected by 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) may serve as an early predictive biomarker and treatment target for epileptogenesis. Here, we aimed to investigate changes in cerebral glucose metabolism before induction of epileptogenesis, during epileptogenesis as well as during chronic epilepsy. As anesthesia is usually unavoidable for preclinical PET imaging and influences the distribution of the radiotracer, four different protocols were compared. Procedures We investigated 18F-FDG uptake phase in conscious rats followed by a static scan as well as dynamic scans under continuous isoflurane, medetomidine-midazolam-fentanyl (MMF), or propofol anesthesia. Furthermore, we applied different analysis approaches: atlas-based regional analysis, statistical parametric mapping, and kinetic analysis. Results At baseline and compared to uptake in conscious rats, isoflurane and propofol anesthesia resulted in decreased cortical 18F-FDG uptake while MMF anesthesia led to a globally decreased tracer uptake. During epileptogenesis, MMF anesthesia was clearly best distinctive for visualization of prominently increased glucometabolism in epilepsy-related brain areas. Kinetic modeling further increased sensitivity, particularly for continuous isoflurane anesthesia. During chronic epilepsy, hypometabolism affecting more or less the whole brain was detectable with all protocols. Conclusion This study reveals evaluation of anesthesia protocols for preclinical 18F-FDG PET imaging as a critical step in the study design. Together with an appropriate data analysis workflow, the chosen anesthesia protocol may uncover otherwise concealed disease-associated regional glucometabolic changes.
Collapse
Affiliation(s)
- Ina Jahreis
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias L. Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Jens P. Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- * E-mail:
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
12
|
Akyuz E, Koklu B, Uner A, Angelopoulou E, Paudel YN. Envisioning the role of inwardly rectifying potassium (Kir) channel in epilepsy. J Neurosci Res 2021; 100:413-443. [PMID: 34713909 DOI: 10.1002/jnr.24985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 01/29/2023]
Abstract
Epilepsy is a devastating neurological disorder characterized by recurrent seizures attributed to the disruption of the dynamic excitatory and inhibitory balance in the brain. Epilepsy has emerged as a global health concern affecting about 70 million people worldwide. Despite recent advances in pre-clinical and clinical research, its etiopathogenesis remains obscure, and there are still no treatment strategies modifying disease progression. Although the precise molecular mechanisms underlying epileptogenesis have not been clarified yet, the role of ion channels as regulators of cellular excitability has increasingly gained attention. In this regard, emerging evidence highlights the potential implication of inwardly rectifying potassium (Kir) channels in epileptogenesis. Kir channels consist of seven different subfamilies (Kir1-Kir7), and they are highly expressed in both neuronal and glial cells in the central nervous system. These channels control the cell volume and excitability. In this review, we discuss preclinical and clinical evidence on the role of the several subfamilies of Kir channels in epileptogenesis, aiming to shed more light on the pathogenesis of this disorder and pave the way for future novel therapeutic approaches.
Collapse
Affiliation(s)
- Enes Akyuz
- Faculty of International Medicine, Department of Biophysics, University of Health Sciences, Istanbul, Turkey
| | - Betul Koklu
- Faculty of Medicine, Namık Kemal University, Tekirdağ, Turkey
| | - Arda Uner
- Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
13
|
Jiang G, Wang S, Chen M, Ding X, He W, Wang L, Wang S, Yu J, Wang X. Linsitinib (OSI-906) modulates brain energy metabolism and seizure activity in the lithium-pilocarpine rat model. ACTA EPILEPTOLOGICA 2021. [DOI: 10.1186/s42494-021-00054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Epileptic seizure is a process of energy accumulation, bursting, and depletion accompanied by the production, spread, and termination of epileptic discharges. The energy required for a seizure is mainly provided through mitochondrial production of ATP. Mitochondrial diseases often lead to epileptic seizures, and energy depletion caused by seizures can lead to mitochondrial dysfunction. The energy metabolism has become a key target for treatment of epileptic diseases.
Method
The effect of OSI-906, an insulin receptor (IR)/ insulin-like growth factor 1 receptor (IGF-1R) inhibitor, on behaviors and electroencephalographic activity in the lithium-pilocarpine rats were tested. 18F-FDG positron emission tomography (PET)/ computed tomography (CT) was performed to detect the relative whole-brain glucose uptake values. Electron microscopy was performed to observe the ultrastructure of neuronal and mitochondrial damage. The changes in blood glucose at different time points before and after the intervention were tested and the effects of OSI-906 on IR/IGF-1R and downstream Akt signaling in the context of seizures were evaluated.
Results
The OSI-906 treatment applied 3 days before the pilocarpine-induced seizures significantly reduced the seizure severity, prolonged the seizure latency and decreased the EEG energy density. MicroPET/CT revealed that 50 mg/kg of OSI-906 inhibited the 18F-FDG glucose uptake after epileptic seizures, suggesting that OSI-906, through inhibiting IR/IGF-1R and the downstream AKT signaling, may regulate the excessive energy consumption of the epileptic brain. The OSI-906 treatment also reduced the mitochondrial damage caused by epileptic seizures.
Conclusion
The IR/IGF-1R inhibitor OSI-906 can significantly reduce the sensitivity and severity of pilocarpine-induced seizures by inhibiting the IR/IGF-1R and the downstream Akt signaling pathway.
Collapse
|
14
|
Pototskiy E, Dellinger JR, Bumgarner S, Patel J, Sherrerd-Smith W, Musto AE. Brain injuries can set up an epileptogenic neuronal network. Neurosci Biobehav Rev 2021; 129:351-366. [PMID: 34384843 DOI: 10.1016/j.neubiorev.2021.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/01/2021] [Indexed: 10/20/2022]
Abstract
Development of epilepsy or epileptogenesis promotes recurrent seizures. As of today, there are no effective prophylactic therapies to prevent the onset of epilepsy. Contributing to this deficiency of preventive therapy is the lack of clarity in fundamental neurobiological mechanisms underlying epileptogenesis and lack of reliable biomarkers to identify patients at risk for developing epilepsy. This limits the development of prophylactic therapies in epilepsy. Here, neural network dysfunctions reflected by oscillopathies and microepileptiform activities, including neuronal hyperexcitability and hypersynchrony, drawn from both clinical and experimental epilepsy models, have been reviewed. This review suggests that epileptogenesis reflects a progressive and dynamic dysfunction of specific neuronal networks which recruit further interconnected groups of neurons, with this resultant pathological network mediating seizure occurrence, recurrence, and progression. In the future, combining spatial and temporal resolution of neuronal non-invasive recordings from patients at risk of developing epilepsy, together with analytics and computational tools, may contribute to determining whether the brain is undergoing epileptogenesis in asymptomatic patients following brain injury.
Collapse
Affiliation(s)
- Esther Pototskiy
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA; College of Sciences, Old Dominion University, Norfolk, Virginia
| | - Joshua Ryan Dellinger
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA
| | - Stuart Bumgarner
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA
| | - Jay Patel
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA
| | - William Sherrerd-Smith
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA
| | - Alberto E Musto
- Department of Anatomy & Pathology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA; Department of Neurology, Eastern Virginia Medical School, Department of Pathology, Norfolk, Virginia, USA.
| |
Collapse
|
15
|
Scopolamine prevents aberrant mossy fiber sprouting and facilitates remission of epilepsy after brain injury. Neurobiol Dis 2021; 158:105446. [PMID: 34280524 DOI: 10.1016/j.nbd.2021.105446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/01/2021] [Accepted: 07/13/2021] [Indexed: 11/21/2022] Open
Abstract
Prevention or modification of acquired epilepsy in patients at risk is an urgent, yet unmet, clinical need. Following acute brain insults, there is an increased risk of mesial temporal lobe epilepsy (mTLE), which is often associated with debilitating comorbidities and reduced life expectancy. The latent period between brain injury and the onset of epilepsy may offer a therapeutic window for interfering with epileptogenesis. The pilocarpine model of mTLE is widely used in the search for novel antiepileptogenic treatments. Recent biochemical studies indicated that cholinergic mechanisms play a role in the epileptogenic alterations induced by status epilepticus (SE) in this and other models of mTLE, which prompted us to evaluate whether treatment with the muscarinic antagonist scopolamine during the latent period after SE is capable of preventing or modifying epilepsy and associated behavioral and cognitive alterations in female Sprague-Dawley rats. First, in silico pharmacokinetic modeling was used to select a dosing protocol by which M-receptor inhibitory brain levels of scopolamine are maintained during prolonged treatment. This protocol was verified by drug analysis in vivo. Rats were then treated twice daily with scopolamine over 17 days after SE, followed by drug wash-out and behavioral and video/EEG monitoring up to ~6 months after SE. Compared to vehicle controls, rats that were treated with scopolamine during the latent period exhibited a significantly lower incidence of spontaneous recurrent seizures during periods of intermittent recording in the chronic phase of epilepsy, less behavioral excitability, less cognitive impairment, and significantly reduced aberrant mossy fiber sprouting in the hippocampus. The present data may indicate that scopolamine exerts antiepileptogenic/disease-modifying activity in the lithium-pilocarpine rat model, possibly involving increased remission of epilepsy as a new mechanism of disease-modification. For evaluating the rigor of the present data, we envision a study that more thoroughly addresses the gender bias and video-EEG recording limitations of the present study.
Collapse
|
16
|
Löscher W, Klein P. New approaches for developing multi-targeted drug combinations for disease modification of complex brain disorders. Does epilepsy prevention become a realistic goal? Pharmacol Ther 2021; 229:107934. [PMID: 34216705 DOI: 10.1016/j.pharmthera.2021.107934] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
Over decades, the prevailing standard in drug discovery was the concept of designing highly selective compounds that act on individual drug targets. However, more recently, multi-target and combinatorial drug therapies have become an important treatment modality in complex diseases, including neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The development of such network-based approaches is facilitated by the significant advance in our understanding of the pathophysiological processes in these and other complex brain diseases and the adoption of modern computational approaches in drug discovery and repurposing. However, although drug combination therapy has become an effective means for the symptomatic treatment of many complex diseases, the holy grail of identifying clinically effective disease-modifying treatments for neurodegenerative and other brain diseases remains elusive. Thus, despite extensive research, there remains an urgent need for novel treatments that will modify the progression of the disease or prevent its development in patients at risk. Here we discuss recent approaches with a focus on multi-targeted drug combinations for prevention or modification of epilepsy. Over the last ~10 years, several novel promising multi-targeted therapeutic approaches have been identified in animal models. We envision that synergistic combinations of repurposed drugs as presented in this review will be demonstrated to prevent epilepsy in patients at risk within the next 5-10 years.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| |
Collapse
|
17
|
The Kainic Acid Models of Temporal Lobe Epilepsy. eNeuro 2021; 8:ENEURO.0337-20.2021. [PMID: 33658312 PMCID: PMC8174050 DOI: 10.1523/eneuro.0337-20.2021] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/14/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Experimental models of epilepsy are useful to identify potential mechanisms of epileptogenesis, seizure genesis, comorbidities, and treatment efficacy. The kainic acid (KA) model is one of the most commonly used. Several modes of administration of KA exist, each producing different effects in a strain-, species-, gender-, and age-dependent manner. In this review, we discuss the advantages and limitations of the various forms of KA administration (systemic, intrahippocampal, and intranasal), as well as the histologic, electrophysiological, and behavioral outcomes in different strains and species. We attempt a personal perspective and discuss areas where work is needed. The diversity of KA models and their outcomes offers researchers a rich palette of phenotypes, which may be relevant to specific traits found in patients with temporal lobe epilepsy.
Collapse
|
18
|
Chen X, Liu K, Lin Z, Huang K, Pan S. Knockout of Transient Receptor Potential Melastatin 4 Channel Mitigates Cerebral Edema and Neuronal Injury After Status Epilepticus in Mice. J Neuropathol Exp Neurol 2021; 79:1354-1364. [PMID: 33186453 DOI: 10.1093/jnen/nlaa134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study aimed to evaluate whether the knockout of transient receptor potential melastatin 4 (TRPM4) could reduce cerebral edema and improve neurologic outcome in a mouse model of status epilepticus (SE). Wild-type (WT) (n = 61) and Trpm4-/- mice (n = 61) with behavioral seizures induced by lithium (10 mEq/kg) and pilocarpine (30-40 mg/kg) were terminated 2.5 hours after the onset of SE. After SE, 28 WT-SE and 27 Trpm4-/--SE mice were observed for 28 days and assessed for survival and cognitive function; the others were killed after 24 hours, 72 hours, or 7 days, and evaluated for cerebral edema and histological injury. In comparison to WT-SE mice, the mortality and cognitive deficit for Trpm4-/--SE mice following SE after 28 days were significantly ameliorated. Trpm4-/--SE mice also showed less water content and cerebral edema assessed by magnetic resonance imaging, and decreased blood-brain barrier breakdown after SE. Moreover, Trpm4 deficiency significantly mitigated neuronal loss, cellular necrosis and apoptosis in the hippocampus and piriform cortex and mitigated astrocytosis and microgliosis. In conclusion, this study suggests that Trmp4 may represent a new target for improving outcomes after SE.
Collapse
Affiliation(s)
- Xing Chen
- From the Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kewei Liu
- From the Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenzhou Lin
- From the Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaibin Huang
- From the Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- From the Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Rojas A, McCarren HS, Wang J, Wang W, Abreu-Melon J, Wang S, McDonough JH, Dingledine R. Comparison of neuropathology in rats following status epilepticus induced by diisopropylfluorophosphate and soman. Neurotoxicology 2021; 83:14-27. [PMID: 33352274 PMCID: PMC7987879 DOI: 10.1016/j.neuro.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022]
Abstract
The increasing number of cases involving the use of nerve agents as deadly weapons has spurred investigation into the molecular mechanisms underlying nerve agent-induced pathology. The highly toxic nature of nerve agents restrict their use in academic research laboratories. Less toxic organophosphorus (OP) based agents including diisopropylfluorophosphate (DFP) are used as surrogates in academic research laboratories to mimic nerve agent poisoning. However, neuropathology resulting from DFP-induced status epilepticus (SE) has not been compared directly to neuropathology observed following nerve agent poisoning in the same study. Here, the hypothesis that neuropathology measured four days after SE is the same for rats exposed to DFP and soman was tested. Adult Sprague-Dawley rats were injected with soman or DFP to induce SE. Cortical electroencephalography (EEG) was recorded prior to and during soman-induced SE. EEG power analysis of rats administered soman revealed prolonged electrographic SE similar to that of rats that endure uninterrupted SE following injection of DFP. Rats that experienced soman-induced SE displayed less hippocampal neuroinflammation and gliosis compared to rats administered DFP. Seizure-induced weight change, blood-brain barrier (BBB) leakiness and neurodegeneration in most seizure sensitive limbic brain regions were similar for rats that endured SE following soman or DFP. The amalgamated pathology score calculated by combining pathological measures (weight loss, hippocampal neuroinflammation, gliosis, BBB integrity and neurodegeneration) was similar in rats administered the OP agents. These findings support use of the rat DFP model of SE as a suitable surrogate for investigating some, but not all delayed consequences produced by nerve agents.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States.
| | - Hilary S McCarren
- Neuroscience Department, Medical Toxicology Research Division, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, United States
| | - Jennifer Wang
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States
| | - JuanMartin Abreu-Melon
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States
| | - Sarah Wang
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States
| | - John H McDonough
- Neuroscience Department, Medical Toxicology Research Division, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, United States
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, 30322, United States
| |
Collapse
|
20
|
Pawlik MJ, Obara-Michlewska M, Popek MP, Czarnecka AM, Czuczwar SJ, Łuszczki J, Kołodziej M, Acewicz A, Wierzba-Bobrowicz T, Albrecht J. Pretreatment with a glutamine synthetase inhibitor MSO delays the onset of initial seizures induced by pilocarpine in juvenile rats. Brain Res 2021; 1753:147253. [PMID: 33422530 DOI: 10.1016/j.brainres.2020.147253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/26/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
The contribution of glutamatergic transmission to generation of initial convulsive seizures (CS) is debated. We tested whether pretreatment with a glutamine synthetase (GS) inhibitor, methionine sulfoximine (MSO), affects the onset and progression of initial CS by cholinergic stimulus in juvenile rats. Male rats (24 days old, Sprague Dawley) sequentially received i.p. injections of lithium-carbonate, MSO, methyl-scopolamine, and pilocarpine (Pilo). Pilo was given 150 min after MSO. Animals were continuously monitored using the Racine scale, EEG/EMG and intrahippocampal glutamate (Glu) biosensors. GS activity as measured in hippocampal homogenates, was not altered by MSO at 150 min, showed initial, varied inhibition at 165 (15 min post-Pilo), and dropped down to 11% of control at 60 min post-Pilo, whereas GS protein expression remained unaltered throughout. Pilo did neither modulate the effect of MSO on GS activity nor affect GS activity itself, at any time point. MSO reduced from 32% to 4% the number of animals showing CS during the first 12 min post-Pilo, delayed by ~6 min the appearance of electrographic seizures, and tended to decrease EMG power during ~15 min post-Pilo. The results indicate that MSO impairs an aspect of glutamatergic transmission involved in the transition from the first cholinergic stimulus to the onset of seizures. A continuous rise of extracellular Glu lasting 60 min was insignificantly affected by MSO, leaving the nature of the Glu pool(s) involved in altered glutamatergic transmission undefined.
Collapse
Affiliation(s)
- Marek J Pawlik
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Marta Obara-Michlewska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Mariusz P Popek
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Anna Maria Czarnecka
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Jarogniew Łuszczki
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Marcin Kołodziej
- Institute of Theory of Electrical Engineering, Measurement and Information Systems, Warsaw University of Technology, Koszykowa 75, 00-662 Warsaw, Poland.
| | - Albert Acewicz
- Department of Neuropathology, Institute of Psychiatry and Neurology, Jana III Sobieskiego 9, 02-957 Warsaw, Poland.
| | - Teresa Wierzba-Bobrowicz
- Department of Neuropathology, Institute of Psychiatry and Neurology, Jana III Sobieskiego 9, 02-957 Warsaw, Poland.
| | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| |
Collapse
|
21
|
Rojas A, Wang J, Glover A, Dingledine R. Urethane attenuates early neuropathology of diisopropylfluorophosphate-induced status epilepticus in rats. Neurobiol Dis 2020; 140:104863. [PMID: 32283202 PMCID: PMC7266093 DOI: 10.1016/j.nbd.2020.104863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/17/2020] [Accepted: 04/02/2020] [Indexed: 12/27/2022] Open
Abstract
Seizures can be evident within minutes of exposure to an organophosphorus (OP) agent and often progress to status epilepticus (SE) resulting in a high mortality if left untreated. Effective medical countermeasures are necessary to sustain patients suffering from OP poisoning and to mitigate the ensuing brain injury. Here, the hypothesis was tested that a single subanesthetic dose of urethane prevents neuropathology measured 24 h following diisopropylfluorophosphate (DFP)-induced SE. Adult Sprague-Dawley rats were injected with DFP to induce SE. During SE rats displayed increased neuronal activity in the hippocampus and an upregulation of immediate early genes as well as pro-inflammatory mediators. In additional experiments rats were administered diazepam (10 mg/kg, ip) or urethane (0.8 g/kg, sc) 1 h after DFP-induced SE and compared to rats that experienced uninterrupted SE. Cortical electroencephalography (EEG) and power analysis strengthen the conclusion that urethane effectively terminates SE and prevents the overnight return of seizure activity. Neurodegeneration in limbic brain regions and the seizure-induced upregulation of key inflammatory mediators present 24 h after DFP-induced SE were strongly attenuated by administration of urethane. A trivial explanation for these beneficial effects, that urethane simply reactivates acetylcholinesterase, has been ruled out. These findings indicate that, by contrast to rats administered diazepam or rats that experience uninterrupted SE, the early neuropathology after SE is prevented by subanesthetic urethane, which terminates rather than interrupts, SE.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, United States of America.
| | - Jennifer Wang
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, United States of America
| | - Avery Glover
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, United States of America
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, United States of America
| |
Collapse
|
22
|
Fan J, Shan W, Yang H, Zhu F, Liu X, Wang Q. Neural Activities in Multiple Rat Brain Regions in Lithium-Pilocarpine-Induced Status Epilepticus Model. Front Mol Neurosci 2020; 12:323. [PMID: 32009899 PMCID: PMC6974466 DOI: 10.3389/fnmol.2019.00323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/17/2019] [Indexed: 11/13/2022] Open
Abstract
To clarify the different regional brain electroencephalogram (EEG) activities and biochemical responses in seizure and epilepsy models, we assessed the EEG and c-Fos immunolabeling characteristics in a lithium-pilocarpine-induced status epilepticus (SE) model and pentylenetetrazol (PTZ)-induced seizure model. The regional brain activities were evaluated by EEG and c-Fos immunolabeling. ZnT3 immunostaining was performed to observe hippocampal mossy fiber sprouting (MFS) within 7 days after the induction of SE in the lithium-pilocarpine model. The EEG recordings showed distinctive features of activation in different brain areas. With the aggravation of the behavioral manifestations of the seizures, the frequency and amplitude of the discharges on EEG gradually increased. SE was eventually induced and sustained. The labeling of c-Fos was enhanced in the cortex and hippocampal CA1, CA3, and dentate gyrus (DG); however, compared to the PTZ-induced seizure model, c-Fos staining could only be observed in the striatum and thalamus in the lithium-pilocarpine-induced epilepsy model. In each brain region, prominent c-Fos labeling was observed 2 h and 4 h after the induction of SE or seizures and diminished at 24 h. During the lithium-pilocarpine-induced chronic epilepsy phase after SE induction, MFS was observed 7 days after SE and was accompanied by the dynamic evolution of epileptic EEG activities. These findings validated the lithium-pilocarpine-induced SE model as an epilepsy model with a specific spatial-temporal profile of neural activation. The EEG characteristics and c-Fos expression patterns differ from those presented in a previous study using a PTZ-induced seizure model. Hippocampal mossy fiber spouting might be associated with spontaneous seizures during the chronic phase and can be detected at least within 1 week by ZnT3 staining after stimulation.
Collapse
Affiliation(s)
- Jingjing Fan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Wei Shan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Huajun Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Fei Zhu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Xiao Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Center for Clinical Medicine of Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
23
|
Bascuñana P, Brackhan M, Leiter I, Keller H, Jahreis I, Ross TL, Bengel FM, Bankstahl M, Bankstahl JP. Divergent metabolic substrate utilization in brain during epileptogenesis precedes chronic hypometabolism. J Cereb Blood Flow Metab 2020; 40:204-213. [PMID: 30375913 PMCID: PMC6928550 DOI: 10.1177/0271678x18809886] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/17/2018] [Accepted: 10/05/2018] [Indexed: 01/08/2023]
Abstract
Alterations in metabolism during epileptogenesis may be a therapy target. Recently, an increase in amino acid transport into the brain was proposed to play a role in epileptogenesis. We aimed to characterize alterations of substrate utilization during epileptogenesis and in chronic epilepsy. The lithium-pilocarpine post status epilepticus (SE) rat model was used. We performed longitudinal O-(2-[(18)F]fluoroethyl)-l-tyrosine (18F-FET) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) and calculated 18F-FET volume of distribution (Vt) and 18F-FDG uptake. Correlation analyses were performed with translocator protein-PET defined neuroinflammation from previously acquired data. We found reduced 18F-FET Vt at 48 h after SE (amygdala: -30.2%, p = 0.014), whereas 18F-FDG showed increased glucose uptake 4 and 24 h after SE (hippocampus: + 43.6% and +42.5%, respectively; p < 0.001) returning to baseline levels thereafter. In chronic epileptic animals, we found a reduction in 18F-FET and 18F-FDG in the hippocampus. No correlation was found for 18F-FET or 18F-FDG to microglial activation at seven days post SE. Whereas metabolic alterations do not reflect higher metabolism associated to activated microglia, they might be partially driven by chronic neuronal loss. However, both metabolisms diverge during early epileptogenesis, pointing to amino acid turnover as a possible biomarker and/or therapeutic target for epileptogenesis.
Collapse
Affiliation(s)
- Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Mirjam Brackhan
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Ina Leiter
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Heike Keller
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Ina Jahreis
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
24
|
Bascuñana P, Gendron T, Sander K, Jahreis I, Polyak A, Ross TL, Bankstahl M, Arstad E, Bankstahl JP. Ex vivo characterization of neuroinflammatory and neuroreceptor changes during epileptogenesis using candidate positron emission tomography biomarkers. Epilepsia 2019; 60:2325-2333. [PMID: 31571210 DOI: 10.1111/epi.16353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/04/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Identification of patients at risk of developing epilepsy before the first spontaneous seizure may promote the development of preventive treatment providing opportunity to stop or slow down the disease. METHODS As development of novel radiotracers and on-site setup of existing radiotracers is highly time-consuming and expensive, we used dual-centre in vitro autoradiography as an approach to characterize the potential of innovative radiotracers in the context of epilepsy development. Using brain slices from the same group of rats, we aimed to characterise the evolution of neuroinflammation and expression of inhibitory and excitatory neuroreceptors during epileptogenesis using translational positron emission tomography (PET) tracers; 18 F-flumazenil (18 F-FMZ; GABAA receptor), 18 F-FPEB (metabotropic glutamate receptor 5; mGluR5), 18 F-flutriciclamide (translocator protein; TSPO, microglia activation) and 18 F-deprenyl (monoamine oxidase B, astroglia activation). Autoradiography images from selected time points after pilocarpine-induced status epilepticus (SE; baseline, 24 and 48 hours, 5, 10 and 15 days and 6 and 12-14 weeks after SE) were normalized to a calibration curve, co-registered to an MRI-based 2D region-of-interest atlas, and activity concentration (Bq/mm2 ) was calculated. RESULTS In epileptogenesis-associated brain regions, 18 F-FMZ and 18 F-FPEB showed an early decrease after SE. 18 F-FMZ decrease was maintained in the latent phase and further reduced in the chronic epileptic animals, while 18 F-FPEB signal recovered from day 10, reaching baseline levels in chronic epilepsy. 18 F-flutriciclamide showed an increase of activated microglia at 24 hours after SE, peaking at 5-15 days and decreasing during the chronic phase. On the other hand, 18 F-deprenyl autoradiography showed late astrogliosis, peaking in the chronic phase. SIGNIFICANCE Autoradiography revealed different evolution of the selected targets during epileptogenesis. Our results suggest an advantage of combined imaging of inter-related targets like glutamate and GABAA receptors, or microglia and astrocyte activation, in order to identify important interactions, especially when using PET imaging for the evaluation of novel treatments.
Collapse
Affiliation(s)
- Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Thibault Gendron
- Institute of Nuclear Medicine, University College London, London, UK.,Department of Chemistry, University College London, London, UK
| | - Kerstin Sander
- Institute of Nuclear Medicine, University College London, London, UK.,Department of Chemistry, University College London, London, UK
| | - Ina Jahreis
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany.,Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Erik Arstad
- Institute of Nuclear Medicine, University College London, London, UK.,Department of Chemistry, University College London, London, UK
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
25
|
Jain S, LaFrancois JJ, Botterill JJ, Alcantara-Gonzalez D, Scharfman HE. Adult neurogenesis in the mouse dentate gyrus protects the hippocampus from neuronal injury following severe seizures. Hippocampus 2019; 29:683-709. [PMID: 30672046 PMCID: PMC6640126 DOI: 10.1002/hipo.23062] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/29/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023]
Abstract
Previous studies suggest that reducing the numbers of adult-born neurons in the dentate gyrus (DG) of the mouse increases susceptibility to severe continuous seizures (status epilepticus; SE) evoked by systemic injection of the convulsant kainic acid (KA). However, it was not clear if the results would be the same for other ways to induce seizures, or if SE-induced damage would be affected. Therefore, we used pilocarpine, which induces seizures by a different mechanism than KA. Also, we quantified hippocampal damage after SE. In addition, we used both loss-of-function and gain-of-function methods in adult mice. We hypothesized that after loss-of-function, mice would be more susceptible to pilocarpine-induced SE and SE-associated hippocampal damage, and after gain-of-function, mice would be more protected from SE and hippocampal damage after SE. For loss-of-function, adult neurogenesis was suppressed by pharmacogenetic deletion of dividing radial glial precursors. For gain-of-function, adult neurogenesis was increased by conditional deletion of pro-apoptotic gene Bax in Nestin-expressing progenitors. Fluoro-Jade C (FJ-C) was used to quantify neuronal injury and video-electroencephalography (video-EEG) was used to quantify SE. Pilocarpine-induced SE was longer in mice with reduced adult neurogenesis, SE had more power and neuronal damage was greater. Conversely, mice with increased adult-born neurons had shorter SE, SE had less power, and there was less neuronal damage. The results suggest that adult-born neurons exert protective effects against SE and SE-induced neuronal injury.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Justin J. Botterill
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
26
|
Meller S, Brandt C, Theilmann W, Klein J, Löscher W. Commonalities and differences in extracellular levels of hippocampal acetylcholine and amino acid neurotransmitters during status epilepticus and subsequent epileptogenesis in two rat models of temporal lobe epilepsy. Brain Res 2019; 1712:109-123. [DOI: 10.1016/j.brainres.2019.01.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/09/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023]
|
27
|
Pinellia Total Alkaloids Modulate the GABAergic System in Hippocampal Formation on Pilocarpine-Induced Epileptic Rats. Chin J Integr Med 2019; 26:138-145. [DOI: 10.1007/s11655-019-2944-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 01/13/2023]
|
28
|
Löscher W. The holy grail of epilepsy prevention: Preclinical approaches to antiepileptogenic treatments. Neuropharmacology 2019; 167:107605. [PMID: 30980836 DOI: 10.1016/j.neuropharm.2019.04.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
A variety of acute brain insults can induce epileptogenesis, a complex process that results in acquired epilepsy. Despite advances in understanding mechanisms of epileptogenesis, there is currently no approved treatment that prevents the development or progression of epilepsy in patients at risk. The current concept of epileptogenesis assumes a window of opportunity following acute brain insults that allows intervention with preventive treatment. Recent results suggest that injury-induced epileptogenesis can be a much more rapid process than previously thought, suggesting that the 'therapeutic window' may only be open for a brief period, as in stroke therapy. However, experimental data also suggest a second, possibly delayed process ("secondary epileptogenesis") that influences the progression and refractoriness of the epileptic state over time, allowing interfering with this process even after onset of epilepsy. In this review, both methodological issues in preclinical drug development and novel targets for antiepileptogenesis will be discussed. Several promising drugs that either prevent epilepsy (antiepileptogenesis) or slow epilepsy progression and alleviate cognitive or behavioral comorbidities of epilepsy (disease modification) have been described in recent years, using diverse animal models of acquired epilepsy. Promising agents include TrkB inhibitors, losartan, statins, isoflurane, anti-inflammatory and anti-oxidative drugs, the SV2A modulator levetiracetam, and epigenetic interventions. Research on translational target validity and on prognostic biomarkers that can be used to stratify patients (or experimental animals) at high risk of developing epilepsy will hopefully soon lead to proof-of-concept clinical trials with the most promising drugs, which will be essential to make prevention of epilepsy a reality. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
29
|
Welzel L, Twele F, Schidlitzki A, Töllner K, Klein P, Löscher W. Network pharmacology for antiepileptogenesis: Tolerability and neuroprotective effects of novel multitargeted combination treatments in nonepileptic vs. post-status epilepticus mice. Epilepsy Res 2019; 151:48-66. [PMID: 30831337 DOI: 10.1016/j.eplepsyres.2019.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/06/2019] [Accepted: 02/23/2019] [Indexed: 01/08/2023]
Abstract
Network-based approaches in drug discovery comprise both development of novel drugs interacting with multiple targets and repositioning of drugs with known targets to form novel drug combinations that interact with cellular or molecular networks whose function is disturbed in a disease. Epilepsy is a complex network phenomenon that, as yet, cannot be prevented or cured. We recently proposed multitargeted, network-based approaches to prevent epileptogenesis by combinations of clinically available drugs chosen to impact diverse epileptogenic processes. In order to test this strategy preclinically, we developed a multiphase sequential study design for evaluating such drug combinations in rodents, derived from human clinical drug development phases. Because pharmacokinetics of such drugs are known, only the tolerability of novel drug combinations needs to be evaluated in Phase I in öhealthy" controls. In Phase IIa, tolerability is assessed following an epileptogenic brain insult, followed by antiepileptogenic efficacy testing in Phase IIb. Here, we report Phase I and Phase IIa evaluation of 7 new drug combinations in mice, using 10 drugs (levetiracetam, topiramate, gabapentin, deferoxamine, fingolimod, ceftriaxone, α-tocopherol, melatonin, celecoxib, atorvastatin) with diverse mechanisms thought to be important in epileptogenesis. Six of the 7 drug combinations were well tolerated in mice during prolonged treatment at the selected doses in both controls and during the latent phase following status epilepticus induced by intrahippocampal kainate. However, none of the combinations prevented hippocampal damage in response to kainate, most likely because treatment started only 16-18 h after kainate. This suggests that antiepileptogenic or disease-modifying treatment may need to start earlier after the brain insult. The present data provide a rich collection of tolerable, network-based combinatorial therapies as a basis for antiepileptogenic or disease-modifying efficacy testing.
Collapse
Affiliation(s)
- Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD 20817, USA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
30
|
Barker-Haliski M, Harte-Hargrove LC, Ravizza T, Smolders I, Xiao B, Brandt C, Löscher W. A companion to the preclinical common data elements for pharmacologic studies in animal models of seizures and epilepsy. A Report of the TASK3 Pharmacology Working Group of the ILAE/AES Joint Translational Task Force. Epilepsia Open 2018; 3:53-68. [PMID: 30450485 PMCID: PMC6210039 DOI: 10.1002/epi4.12254] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Preclinical pharmacology studies in animal models of seizures and epilepsy have provided a platform to identify more than 20 antiseizure drugs in recent decades. To minimize variability in lab‐to‐lab studies and to harmonize approaches to data collection and reporting methodology in pharmacologic evaluations of the next generation of therapies, we present common data elements (CDEs), case report forms (CRFs), and this companion manuscript to help with the implementation of methods for studies in established preclinical seizure and epilepsy models in adult rodents. The development of and advocacy for CDEs in preclinical research has been encouraged previously by both clinical and preclinical groups. It is anticipated that adoption and implementation of these CDEs in preclinical studies may help standardize approaches to minimize variability and increase the reproducibility of preclinical studies. Moreover, they may provide a methodologic framework for pharmacology studies in atypical animal models or models in development, which may ultimately promote novel therapy development. In the present document, we refer selectively to animal models that have a long history of preclinical use, and in some cases, are clinically validated.
Collapse
Affiliation(s)
- Melissa Barker-Haliski
- Department of Pharmacy School of Pharmacy University of Washington Seattle Washington U.S.A
| | - Lauren C Harte-Hargrove
- ILAE/AES Joint Translational Task Force International League Against Epilepsy West Hartford Connecticut U.S.A
| | - Teresa Ravizza
- Department of Neuroscience IRCCS-Institute for Pharmacological Research Mario Negri Milan Italy
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry Drug Analysis and Drug Information Center for Neurosciences Vrije Universiteit Brussel Brussels Belgium
| | - Bo Xiao
- Department of Neurology Xiangya Hospital Central South University Changsha China
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy University of Veterinary Medicine Hannover Hannover Germany.,Center for Systems Neuroscience Hannover Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy University of Veterinary Medicine Hannover Hannover Germany.,Center for Systems Neuroscience Hannover Germany
| |
Collapse
|
31
|
Koepp MJ. The help of biomarkers in the prevention of epilepsy. Lancet Neurol 2018; 15:782-784. [PMID: 27302346 DOI: 10.1016/s1474-4422(16)30081-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London WC1N 3BG, UK.
| |
Collapse
|
32
|
Blood-Brain Barrier Leakage during Early Epileptogenesis Is Associated with Rapid Remodeling of the Neurovascular Unit. eNeuro 2018; 5:eN-NWR-0123-18. [PMID: 29854942 PMCID: PMC5975718 DOI: 10.1523/eneuro.0123-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/01/2018] [Indexed: 01/26/2023] Open
Abstract
Increased permeability of the blood-brain barrier (BBB) following cerebral injury results in regional extravasation of plasma proteins and can critically contribute to the pathogenesis of epilepsy. Here, we comprehensively explore the spatiotemporal evolution of a main extravasation component, albumin, and illuminate associated responses of the neurovascular unit (NVU) contributing to early epileptogenic neuropathology. We applied translational in vivo MR imaging and complementary immunohistochemical analyses in the widely used rat pilocarpine post-status epilepticus (SE) model. The observed rapid BBB leakage affected major epileptogenesis-associated brain regions, peaked between 1 and 2 d post-SE, and rapidly declined thereafter, accompanied by cerebral edema generally following the same time course. At peak of BBB leakage, serum albumin colocalized with NVU constituents, such as vascular components, neurons, and brain immune cells. Surprisingly, astroglial markers did not colocalize with albumin, and aquaporin-4 (AQP4) was clearly reduced in areas of leaky BBB, indicating a severe disturbance of astrocyte-mediated endothelial-neuronal coupling. In addition, a distinct adaptive reorganization process of the NVU vasculature apparently takes place at sites of albumin presence, substantiated by reduced immunoreactivity of endothelial and changes in vascular basement membrane markers. Taken together, degenerative events at the level of the NVU, affecting vessels, astrocytes, and neurons, seem to outweigh reconstructive processes. Considering the rapidly occurring BBB leakage and subsequent impairment of the NVU, our data support the necessity of a prompt BBB-restoring treatment as one component of rational therapeutic intervention to prevent epileptogenesis and the development of other detrimental sequelae of SE.
Collapse
|
33
|
Rojas A, Wang W, Glover A, Manji Z, Fu Y, Dingledine R. Beneficial Outcome of Urethane Treatment Following Status Epilepticus in a Rat Organophosphorus Toxicity Model. eNeuro 2018; 5:ENEURO.0070-18.2018. [PMID: 29766039 PMCID: PMC5952304 DOI: 10.1523/eneuro.0070-18.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/07/2023] Open
Abstract
The efficacy of benzodiazepines to terminate electrographic status epilepticus (SE) declines the longer a patient is in SE. Therefore, alternative methods for ensuring complete block of SE and refractory SE are necessary. We compared the ability of diazepam and a subanesthetic dose of urethane to terminate prolonged SE and mitigate subsequent pathologies. Adult Sprague Dawley rats were injected with diisopropylfluorophosphate (DFP) to induce SE. Rats were administered diazepam (10 mg/kg, ip) or urethane (0.8 g/kg, s.c.) 1 h after DFP-induced SE and compared to rats that experienced uninterrupted SE. Large-amplitude and high-frequency spikes induced by DFP administration were quenched for at least 46 h in rats administered urethane 1 h after SE onset as demonstrated by cortical electroencephalography (EEG). By contrast, diazepam interrupted SE but seizures with high power in the 20- to 70-Hz band returned 6-10 h later. Urethane was more effective than diazepam at reducing hippocampal neurodegeneration, brain inflammation, gliosis and weight loss as measured on day 4 after SE. Furthermore, rats administered urethane displayed a 73% reduction in the incidence of spontaneous recurrent seizures after four to eight weeks and a 90% reduction in frequency of seizures in epileptic rats. By contrast, behavioral changes in the light/dark box, open field and a novel object recognition task were not improved by urethane. These findings indicate that in typical rodent SE models, it is the return of SE overnight, and not the initially intense 1-2 h of SE experience, that is largely responsible for neurodegeneration, accompanying inflammation, and the subsequent development of epilepsy.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Wenyi Wang
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Avery Glover
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Zahra Manji
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Yujiao Fu
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | | |
Collapse
|
34
|
Smith ZZ, Benison AM, Bercum FM, Dudek FE, Barth DS. Progression of convulsive and nonconvulsive seizures during epileptogenesis after pilocarpine-induced status epilepticus. J Neurophysiol 2018; 119:1818-1835. [PMID: 29442558 DOI: 10.1152/jn.00721.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although convulsive seizures occurring during pilocarpine-induced epileptogenesis have received considerable attention, nonconvulsive seizures have not been closely examined, even though they may reflect the earliest signs of epileptogenesis and potentially guide research on antiepileptogenic interventions. The definition of nonconvulsive seizures based on brain electrical activity alone has been controversial. Here we define and quantify electrographic properties of convulsive and nonconvulsive seizures in the context of the acquired epileptogenesis that occurs after pilocarpine-induced status epilepticus (SE). Lithium-pilocarpine was used to induce the prolonged repetitive seizures characteristic of SE; when SE was terminated with paraldehyde, seizures returned during the 2-day period after pilocarpine treatment. A distinct latent period ranging from several days to >2 wk was then measured with continuous, long-term video-EEG. Nonconvulsive seizures dominated the onset of epileptogenesis and consistently preceded the first convulsive seizures but were still present later. Convulsive and nonconvulsive seizures had similar durations. Postictal depression (background suppression of the EEG) lasted for >100 s after both convulsive and nonconvulsive seizures. Principal component analysis was used to quantify the spectral evolution of electrical activity that characterized both types of spontaneous recurrent seizures. These studies demonstrate that spontaneous nonconvulsive seizures have electrographic properties similar to convulsive seizures and confirm that nonconvulsive seizures link the latent period and the onset of convulsive seizures during post-SE epileptogenesis in an animal model. Nonconvulsive seizures may also reflect the earliest signs of epileptogenesis in human acquired epilepsy, when intervention could be most effective. NEW & NOTEWORTHY Nonconvulsive seizures usually represent the first bona fide seizure following a latent period, dominate the early stages of epileptogenesis, and change in severity in a manner consistent with the progressive nature of epileptogenesis. This analysis demonstrates that nonconvulsive and convulsive seizures have different behavioral outcomes but similar electrographic signatures. Alternatively, epileptiform spike-wave discharges fail to recapitulate several key seizure features and represent a category of electrical activity separate from nonconvulsive seizures in this model.
Collapse
Affiliation(s)
- Zachariah Z Smith
- Department of Psychology and Neuroscience, University of Colorado , Boulder, Colorado
| | - Alexander M Benison
- Department of Psychology and Neuroscience, University of Colorado , Boulder, Colorado
| | - Florencia M Bercum
- Department of Psychology and Neuroscience, University of Colorado , Boulder, Colorado
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine , Salt Lake City, Utah
| | - Daniel S Barth
- Department of Psychology and Neuroscience, University of Colorado , Boulder, Colorado
| |
Collapse
|
35
|
McCarren HS, Arbutus JA, Ardinger C, Dunn EN, Jackson CE, McDonough JH. Dexmedetomidine stops benzodiazepine-refractory nerve agent-induced status epilepticus. Epilepsy Res 2018; 141:1-12. [PMID: 29414381 DOI: 10.1016/j.eplepsyres.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/21/2017] [Accepted: 01/07/2018] [Indexed: 12/15/2022]
Abstract
Nerve agents are highly toxic chemicals that pose an imminent threat to soldiers and civilians alike. Nerve agent exposure leads to an increase in acetylcholine within the central nervous system, resulting in development of protracted seizures known as status epilepticus (SE). Currently, benzodiazepines are the standard of care for nerve agent-induced SE, but their efficacy quickly wanes as the time to treatment increases. Here, we examine the role of the α2-adrenoceptor in termination of nerve agent-induced SE using the highly specific agonist dexmedetomidine (DEX). Adult male rats were exposed to soman and entered SE as confirmed by electroencephalograph (EEG). We observed that administration of DEX in combination with the benzodiazepine midazolam (MDZ) 20 or 40 min after the onset of SE stopped seizures and returned processed EEG measurements to baseline levels. The protective effect of DEX was blocked by the α2-adrenoceptor antagonist atipamezole (ATI), but ATI failed to restore seizure activity after it was already halted by DEX in most cases, suggesting that α2-adrenoceptors may be involved in initiating SE cessation rather than merely suppressing seizure activity. Histologically, treatment with DEX + MDZ significantly reduced the number of dying neurons as measured by FluoroJade B in the amygdala, thalamus, and piriform cortex, but did not protect the hippocampus or parietal cortex even when SE was successfully halted. We conclude that DEX serves not just as a valuable potential addition to the anticonvulsant regimen for nerve agent exposure, but also as a tool for dissecting the neural circuitry that drives SE.
Collapse
Affiliation(s)
- Hilary S McCarren
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States.
| | - Julia A Arbutus
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cherish Ardinger
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Emily N Dunn
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cecelia E Jackson
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - John H McDonough
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| |
Collapse
|
36
|
Terrone G, Pauletti A, Salamone A, Rizzi M, Villa BR, Porcu L, Sheehan MJ, Guilmette E, Butler CR, Piro JR, Samad TA, Vezzani A. Inhibition of monoacylglycerol lipase terminates diazepam-resistant status epilepticus in mice and its effects are potentiated by a ketogenic diet. Epilepsia 2017; 59:79-91. [DOI: 10.1111/epi.13950] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Gaetano Terrone
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Alberto Pauletti
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Alessia Salamone
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Massimo Rizzi
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Bianca R. Villa
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Luca Porcu
- Department of Oncology; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| | - Mark J. Sheehan
- Internal Medicine Research Unit; Pfizer Worldwide Research and Development; Cambridge MA USA
| | - Edward Guilmette
- Internal Medicine Research Unit; Pfizer Worldwide Research and Development; Cambridge MA USA
| | | | - Justin R. Piro
- Internal Medicine Research Unit; Pfizer Worldwide Research and Development; Cambridge MA USA
| | - Tarek A. Samad
- Internal Medicine Research Unit; Pfizer Worldwide Research and Development; Cambridge MA USA
| | - Annamaria Vezzani
- Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Milano Italy
| |
Collapse
|
37
|
Zhang T, Todorovic MS, Williamson J, Kapur J. Flupirtine and diazepam combination terminates established status epilepticus: results in three rodent models. Ann Clin Transl Neurol 2017; 4:888-896. [PMID: 29296617 PMCID: PMC5740237 DOI: 10.1002/acn3.497] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/30/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022] Open
Abstract
Objective Status epilepticus (SE) is a neurological emergency requiring rapid termination of seizures. New treatment choices are needed for benzodiazepine-refractory SE or established SE (ESE). Previous studies have demonstrated that the potassium-channel opener flupirtine terminates seizures in neonatal animals. However, its effectiveness in adult ESE has not been tested. We tested whether flupirtine alone or in combination with the benzodiazepine diazepam would terminate ESE in three animal models. Methods SE was induced by administration of lithium followed by pilocarpine, by electrical stimulation of the hippocampus or by diisopropylfluorophosphate (DFP) administration. Seizures were assessed by EEG recorded from the hippocampus and cortex. Results Flupirtine alone did not terminate ESE within 60 min of administration in any of the three models of ESE. A combination of flupirtine and diazepam terminated ESE within 60 min in all the three models. The drug combination shortened the duration of ESE in all three models. Drug responsiveness was distinct between each model. Conclusion A combination of the potassium channel opener flupirtine and diazepam is a potential therapy for ESE.
Collapse
Affiliation(s)
- Terry Zhang
- Department of Neurology University of Virginia Health Sciences Center Charlottesville Virginia 22908
| | - Marko S Todorovic
- Department of Neurology University of Virginia Health Sciences Center Charlottesville Virginia 22908
| | - John Williamson
- Department of Neurology University of Virginia Health Sciences Center Charlottesville Virginia 22908
| | - Jaideep Kapur
- Department of Neurology University of Virginia Health Sciences Center Charlottesville Virginia 22908.,Department of Neuroscience University of Virginia Health Sciences Center Charlottesville Virginia 22908
| |
Collapse
|
38
|
Epileptogenesis meets Occam's Razor. Curr Opin Pharmacol 2017; 35:105-110. [PMID: 28781107 DOI: 10.1016/j.coph.2017.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/23/2017] [Accepted: 07/23/2017] [Indexed: 01/01/2023]
Abstract
Pharmacological treatment to prevent brain injury-induced temporal lobe epileptogenesis has been generally unsuccessful, raising the issues of exactly when the conversion process to an epileptic brain state occurs and reaches completion, and which cellular or network processes might be the most promising therapeutic targets. The time course of epileptogenesis is a central issue, with recent results suggesting that injury-induced epileptogenesis can be a much more rapid process than previously thought, and may be inconsistent with a delayed epileptogenic mechanism. Simplification of the seemingly complex issues involved in the use of epilepsy animal models might lead to a better understanding of the nature of injury-induced epileptogenesis, the significance of the 'latent' period, and whether current strategies should focus on preventing or modifying epilepsy.
Collapse
|
39
|
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
|
40
|
Wu T, Ido K, Osada Y, Kotani S, Tamaoka A, Hanada T. The neuroprotective effect of perampanel in lithium-pilocarpine rat seizure model. Epilepsy Res 2017. [PMID: 28624183 DOI: 10.1016/j.eplepsyres.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Status epilepticus (SE) causes irreversible neurodegeneration if not terminated quickly. Perampanel (PER), a potent AMPA receptor antagonist, has previously been shown to terminate seizures in the lithium-pilocarpine SE model. In the present study, we assessed whether PER would also prevent neuronal damage in this model. METHODS SE was induced in rats using lithium chloride and pilocarpine. Initiation of SE was defined as continuous seizures that exhibited as rearing accompanied by bilateral forelimb clonus (Racine score 4). Either PER (0.6, 2, or 6mg/kg) or diazepam (DZP, 10mg/kg) was administered intravenously 30min after SE initiation. Histopathological samples from treated and seizure-naive rats were taken one week after treatment and then stained with an anti-neuronal nuclei (NeuN) antibody. The sections were analyzed by using a pixel-counting algorithm to quantify the amount of staining in the CA1 subregion of the hippocampus, piriform cortex (Pir), and mediodorsal thalamic nucleus (MD). RESULTS DZP administration did not suppress seizures or the degeneration of neurons in the examined areas. Seizures were terminated in 100% of rats treated with 6mg/kg PER (n=8) and in 47% (7/15) of rats treated with 2mg/kg PER, and neurons in the analyzed areas of these animals were preserved to the level seen in naive rats. In the eight animals in which 2mg/kg PER did not terminate the seizures, neuronal loss was partially attenuated in CA1 and Pir, and neurons were fully preserved in MD. Treatment with 0.6mg/kg PER did not terminate the seizures or significantly preserve neurons. The anti-seizure effect of PER correlated well with the degree of neuroprotection in each analyzed area. CONCLUSIONS PER exhibited a strong neuroprotective effect in a drug-refractory SE model, and this effect was correlated with its attenuation of seizure.
Collapse
Affiliation(s)
- Ting Wu
- Neurology, Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd., Japan; Clinical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Japan.
| | - Katsutoshi Ido
- Neurology, Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd., Japan
| | - Yoshihide Osada
- Neurology, Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd., Japan
| | - Sadaharu Kotani
- Neurology, Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd., Japan
| | - Akira Tamaoka
- Institute of Clinical Medicine, Department of Neurology, University of Tsukuba, Japan
| | - Takahisa Hanada
- Neurology, Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd., Japan
| |
Collapse
|
41
|
Pitkänen A, Löscher W, Vezzani A, Becker AJ, Simonato M, Lukasiuk K, Gröhn O, Bankstahl JP, Friedman A, Aronica E, Gorter JA, Ravizza T, Sisodiya SM, Kokaia M, Beck H. Advances in the development of biomarkers for epilepsy. Lancet Neurol 2017; 15:843-856. [PMID: 27302363 DOI: 10.1016/s1474-4422(16)00112-5] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 12/13/2022]
Abstract
Over 50 million people worldwide have epilepsy. In nearly 30% of these cases, epilepsy remains unsatisfactorily controlled despite the availability of over 20 antiepileptic drugs. Moreover, no treatments exist to prevent the development of epilepsy in those at risk, despite an increasing understanding of the underlying molecular and cellular pathways. One of the major factors that have impeded rapid progress in these areas is the complex and multifactorial nature of epilepsy, and its heterogeneity. Therefore, the vision of developing targeted treatments for epilepsy relies upon the development of biomarkers that allow individually tailored treatment. Biomarkers for epilepsy typically fall into two broad categories: diagnostic biomarkers, which provide information on the clinical status of, and potentially the sensitivity to, specific treatments, and prognostic biomarkers, which allow prediction of future clinical features, such as the speed of progression, severity of epilepsy, development of comorbidities, or prediction of remission or cure. Prognostic biomarkers are of particular importance because they could be used to identify which patients will develop epilepsy and which might benefit from preventive treatments. Biomarker research faces several challenges; however, biomarkers could substantially improve the management of people with epilepsy and could lead to prevention in the right person at the right time, rather than just symptomatic treatment.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, A I Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Annamaria Vezzani
- Department of Neuroscience, Experimental Neurology, IRCCS-Istituto di Recerche Farmacologiche "Mario Negri", Milan, Italy
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, University of Bonn, Bonn, Germany
| | - Michele Simonato
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy; Unit of Gene Therapy of Neurodegenerative Diseases, Division of Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Katarzyna Lukasiuk
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Olli Gröhn
- Department of Neurobiology, A I Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jens P Bankstahl
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Alon Friedman
- Department of Brain and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Israel; Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Teresa Ravizza
- Department of Neuroscience, Experimental Neurology, IRCCS-Istituto di Recerche Farmacologiche "Mario Negri", Milan, Italy
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK; Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Merab Kokaia
- Epilepsy Center, Experimental Epilepsy Group, Division of Neurology, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Heinz Beck
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
42
|
Oxidative Stress Contributes to Status Epilepticus Associated Mortality. Neurochem Res 2017; 42:2024-2032. [PMID: 28462450 DOI: 10.1007/s11064-017-2273-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 10/19/2022]
Abstract
Status epilepticus is a common manifestation of nerve agent toxicity and represents a serious medical emergency with high rates of mortality and neurologic injury in those that survive. The aim of the current study was to determine if targeting oxidative stress with the catalytic antioxidant, AEOL10150, would reduce pilocarpine-induced mortality and attenuate neuronal death and neuroinflammation. We found that treatment with AEOL10150 in conjunction with scopolamine and diazepam following pilocarpine-induced SE was able to significantly reduce mortality compared to treatment with just scopolamine and diazepam. Mortality was further reduced when AEOL10150 was used in conjunction with atropine and diazepam which is considered the standard of care for nerve agent exposures. Both treatment paradigms offered significant protection against SE-induced oxidative stress. Additionally, treatment with scopolamine, AEOL10150 and diazepam attenuated SE-induced neuronal loss and neuroinflammation. Taken together, the data suggest that pharmacological targeting of oxidative stress can improve survival and attenuate secondary neurological damage following SE induced by the nerve agent surrogate pilocarpine.
Collapse
|
43
|
Niquet J, Baldwin R, Norman K, Suchomelova L, Lumley L, Wasterlain CG. Simultaneous triple therapy for the treatment of status epilepticus. Neurobiol Dis 2017; 104:41-49. [PMID: 28461248 DOI: 10.1016/j.nbd.2017.04.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/27/2017] [Accepted: 04/27/2017] [Indexed: 02/08/2023] Open
Abstract
Early maladaptive internalization of synaptic GABAA receptors (GABAAR) and externalization of NMDA receptors (NMDAR) may explain the time-dependent loss of potency of standard anti-epileptic drugs (AED) in refractory status epilepticus (SE). We hypothesized that correcting the effects of changes in GABAAR and NMDAR would terminate SE, even when treatment is delayed 40 minutes. SE was induced in adult Sprague-Dawley rats with a high dose of lithium and pilocarpine. The GABAAR agonist midazolam, the NMDAR antagonist ketamine and the AED valproate were injected 40 min after SE onset in combination or as monotherapy. The midazolam-ketamine-valproate combination was more efficient than triple-dose midazolam, ketamine or valproate monotherapy or higher-dose dual therapy in reducing several parameters of SE severity. Triple therapy also reduced SE-induced acute neuronal injury and spatial memory deficits. In addition, simultaneous triple therapy was more efficient than sequential triple therapy: giving the three drugs simultaneously was more efficient at stopping seizures than the standard practice of giving them sequentially. Furthermore, midazolam-ketamine-valproate therapy suppressed seizures far better than the midazolam-fosphenytoin-valproate therapy, which follows evidence-based AES guidelines. These results show that a treatment aimed at correcting maladaptive GABAAR and NMDAR trafficking can reduce the severity of SE and its long-term consequences.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Roger Baldwin
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Keith Norman
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lucie Suchomelova
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lucille Lumley
- US Army Medical Research Institute of Chemical Defense (USAMRICD), 2900 Ricketts Point Rd., Aberdeen Proving Ground, MD 21010, USA
| | - Claude G Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
44
|
Lin Z, Huang H, Gu Y, Huang K, Hu Y, Ji Z, Wu Y, Wang S, Yang T, Pan S. Glibenclamide ameliorates cerebral edema and improves outcomes in a rat model of status epilepticus. Neuropharmacology 2017; 121:1-11. [PMID: 28412320 DOI: 10.1016/j.neuropharm.2017.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/06/2017] [Accepted: 04/09/2017] [Indexed: 12/30/2022]
Abstract
Glibenclamide (GBC), a sulfonylurea receptor 1 blocker, emerges recently as a promising neuron protectant in various neurological disorders. This study aimed to determine whether GBC improves survival and neurological outcome of status epilepticus (SE). Male Sprague-Dawley rats successfully undergoing SE for 2.5 h (n = 134) were randomly assigned to GBC or vehicle group. Rats in the GBC group received a loading dose of 10 μg/kg of GBC, followed by 1.2 μg/6 h for 3 days, while same dose of vehicle was used as control. The 28-day survival rate in the GBC group (11/23) was significantly higher than that in the vehicle group (8/36). In addition, the frequency and duration of spontaneous recurrent seizures in SE rats were profoundly reduced by GBC but not by vehicle treatment. Moreover, cognitive impairment was observed in the SE rats at day 28, which was reversed by GBC treatment. Meanwhile, cerebral edema, as well as neuronal loss, was decreased in several brain areas in the GBC group. Additionally, on the molecular basis, the subunits of sulfonylurea receptor 1/transient receptor potential M4 (SUR1-TRPM4) heterodimer were both strongly upregulated after SE but partly suppressed by GBC treatment. Furthermore, gene knockdown of Trpm4 in SE rats reduced BBB disruption and neuronal loss, similar to the inhibitory effects with GBC treatment. Taken together, GBC treatment markedly improved survival and neurologic outcomes after SE. The salutary effects of GBC were correlated to the alleviation of cerebral edema and reduction in neurological injury via down-regulation of SUR1-TRPM4 channel.
Collapse
Affiliation(s)
- Zhenzhou Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Gu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong Ji
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongming Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengnan Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Yang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
45
|
Niquet J, Baldwin R, Suchomelova L, Lumley L, Eavey R, Wasterlain CG. Treatment of experimental status epilepticus with synergistic drug combinations. Epilepsia 2017; 58:e49-e53. [PMID: 28225161 DOI: 10.1111/epi.13695] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
During status epilepticus (SE), synaptic γ-aminobutyric acid A receptors (GABAA Rs) become internalized and inactive, whereas spare N-methyl-d-aspartate receptors (NMDARs) assemble, move to the membrane, and become synaptically active. When treatment of SE is delayed, the number of synaptic GABAA Rs is drastically reduced, and a GABAA agonist cannot fully restore inhibition. We used a combination of low-dose diazepam (to stimulate the remaining GABAA Rs), ketamine (to mitigate the effect of the NMDAR increase), and valproate (to enhance inhibition at a nonbenzodiazepine site) to treat seizures in a model of severe cholinergic SE. High doses of diazepam failed to stop electrographic SE, showing that benzodiazepine pharmacoresistance had developed. The diazepam-ketamine-valproate combination was far more effective in stopping SE than triple-dose monotherapy using the same individual drugs. Isobolograms showed that this drug combination's therapeutic actions were synergistic, with positive cooperativity between drugs, whereas drug toxicity was simply additive, without positive or negative cooperativity. As a result, the therapeutic index was improved by this drug combination compared to monotherapy. These results suggest that synergistic drug combinations that target receptor changes can control benzodiazepine-refractory SE.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Roger Baldwin
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucie Suchomelova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucille Lumley
- US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland, U.S.A
| | - Roland Eavey
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Claude G Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A.,Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A
| |
Collapse
|
46
|
Barker-Haliski ML, Heck TD, Dahle EJ, Vanegas F, Pruess TH, Wilcox KS, White HS. Acute treatment with minocycline, but not valproic acid, improves long-term behavioral outcomes in the Theiler's virus model of temporal lobe epilepsy. Epilepsia 2016; 57:1958-1967. [PMID: 27739576 PMCID: PMC5154893 DOI: 10.1111/epi.13577] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Infection with Theiler's murine encephalomyelitis virus (TMEV) in C57Bl/6J mice induces acute seizures and development of spontaneous recurrent seizures and behavioral comorbidities weeks later. The present studies sought to determine whether acute therapeutic intervention with an anti-inflammatory-based approach could prevent or modify development of TMEV-induced long-term behavioral comorbidities. Valproic acid (VPA), in addition to its prototypical anticonvulsant properties, inhibits histone deacetylase (HDAC) activity, which may alter expression of the inflammasome. Minocycline (MIN) has previously demonstrated an antiseizure effect in the TMEV model via direct anti-inflammatory mechanisms, but the long-term effect of MIN treatment on the development of chronic behavioral comorbidities is unknown. METHODS Mice infected with TMEV were acutely administered MIN (50 mg/kg, b.i.d. and q.d.) or VPA (100 mg/kg, q.d.) during the 7-day viral infection period. Animals were evaluated for acute seizure severity and subsequent development of chronic behavioral comorbidities and seizure threshold. RESULTS Administration of VPA reduced the proportion of mice with seizures, delayed onset of symptomatic seizures, and reduced seizure burden during the acute infection. This was in contrast to the effects of administration of once-daily MIN, which did not affect the proportion of mice with seizures or delay onset of acute symptomatic seizures. However, VPA-treated mice were no different from vehicle (VEH)-treated mice in long-term behavioral outcomes, including open field activity and seizure threshold. Once-daily MIN treatment, despite no effect on the maximum observed Racine stage seizure severity, was associated with improved long-term behavioral outcomes and normalized seizure threshold. SIGNIFICANCE Acute seizure control alone is insufficient to modify chronic disease comorbidities in the TMEV model. This work further supports the role of an inflammatory response in the development of chronic behavioral comorbidities and further highlights the utility of this platform for the development of mechanistically novel pharmacotherapies for epilepsy.
Collapse
Affiliation(s)
- Melissa L. Barker-Haliski
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
- Department of Pharmacy, School of Pharmacy, University of Washington, Box 357630, H375 Health Sciences Building, Seattle, WA 98195-7630
| | - Taylor D. Heck
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
| | - E. Jill Dahle
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
| | - Fabiola Vanegas
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
| | - Timothy H. Pruess
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
| | - Karen S. Wilcox
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
| | - H. Steve White
- Anticonvulsant Drug Development Program, Dept. of Pharmacology & Toxicology, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, 801-581-6733
- Department of Pharmacy, School of Pharmacy, University of Washington, Box 357630, H375 Health Sciences Building, Seattle, WA 98195-7630
| |
Collapse
|
47
|
The cannabinoid receptor agonist WIN55.212 reduces consequences of status epilepticus in rats. Neuroscience 2016; 334:191-200. [PMID: 27520083 DOI: 10.1016/j.neuroscience.2016.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/03/2016] [Accepted: 08/03/2016] [Indexed: 12/29/2022]
Abstract
An acute brain insult can cause a spectrum of primary and secondary pathologies including increased risk for epilepsy, mortality and neurodegeneration. The endocannabinoid system, involved in protecting the brain against network hyperexcitability and excitotoxicity, is profoundly dysregulated by acute brain insults. We hypothesize that post-insult dysregulation of the endocannabinoid signaling may contribute to deleterious effects of an acute brain injury and potentiation of endocannabinoid transmission soon after an insult may reduce its pathological outcomes. Effects of an acute post-insult administration of the endocannabinoid receptor agonist WIN55,212-2 on early seizure occurrence, animal mortality and hippocampal cell loss were studied in the lithium-pilocarpine status model. A single dose of WIN55,212-2 (5mg/kg) administered four hours after the end of status epilepticus (SE) reduced the incidence of early seizures during the first two post-SE days though did not change their duration and latency. Brief 4-6-Hz spike-wave discharges appeared de novo in the latent post-SE period and the acute administration of WIN55,212-2 also reduced the incidence of the epileptiform events. A single dose of WIN55,212-2 administered soon after SE improved survival of animals and reduced cell loss in the dentate hilus but did not prevent appearance of spontaneous recurrent seizures in the chronic period. Thus, a brief pharmacological stimulation of the endocannabinoid system soon after a brain insult exerts beneficial effects on its pathological outcome though does not prevent epileptogenesis.
Collapse
|
48
|
Niquet J, Baldwin R, Norman K, Suchomelova L, Lumley L, Wasterlain CG. Midazolam-ketamine dual therapy stops cholinergic status epilepticus and reduces Morris water maze deficits. Epilepsia 2016; 57:1406-15. [PMID: 27500978 DOI: 10.1111/epi.13480] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pharmacoresistance remains an unsolved therapeutic challenge in status epilepticus (SE) and in cholinergic SE induced by nerve agent intoxication. SE triggers a rapid internalization of synaptic γ-aminobutyric acid A (GABAA ) receptors and externalization of N-methyl-d-aspartate (NMDA) receptors that may explain the loss of potency of standard antiepileptic drugs (AEDs). We hypothesized that a drug combination aimed at correcting the consequences of receptor trafficking would reduce SE severity and its long-term consequences. METHODS A severe model of SE was induced in adult Sprague-Dawley rats with a high dose of lithium and pilocarpine. The GABAA receptor agonist midazolam, the NMDA receptor antagonist ketamine, and/or the AED valproate were injected 40 min after SE onset in combination or as monotherapy. Measures of SE severity were the primary outcome. Secondary outcomes were acute neuronal injury, spontaneous recurrent seizures (SRS), and Morris water maze (MWM) deficits. RESULTS Midazolam-ketamine dual therapy was more efficient than double-dose midazolam or ketamine monotherapy or than valproate-midazolam or valproate-ketamine dual therapy in reducing several parameters of SE severity, suggesting a synergistic mechanism. In addition, midazolam-ketamine dual therapy reduced SE-induced acute neuronal injury, epileptogenesis, and MWM deficits. SIGNIFICANCE This study showed that a treatment aimed at correcting maladaptive GABAA receptor and NMDA receptor trafficking can stop SE and reduce its long-term consequences. Early midazolam-ketamine dual therapy may be superior to monotherapy in the treatment of benzodiazepine-refractory SE.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Roger Baldwin
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Keith Norman
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucie Suchomelova
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucille Lumley
- US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland, U.S.A
| | - Claude G Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A.,Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A
| |
Collapse
|
49
|
Terrone G, Pauletti A, Pascente R, Vezzani A. Preventing epileptogenesis: A realistic goal? Pharmacol Res 2016; 110:96-100. [DOI: 10.1016/j.phrs.2016.05.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 12/16/2022]
|
50
|
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
|