1
|
Abend NS, Wusthoff CJ, Jensen FE, Inder TE, Volpe JJ. Neonatal Seizures. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:381-448.e17. [DOI: 10.1016/b978-0-443-10513-5.00015-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
2
|
Schmidt R, Welzel B, Merten A, Naundorf H, Löscher W. Temporal development of seizure threshold and spontaneous seizures after neonatal asphyxia and the effect of prophylactic treatment with midazolam in rats. Exp Neurol 2024; 383:115042. [PMID: 39505250 DOI: 10.1016/j.expneurol.2024.115042] [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: 08/19/2024] [Revised: 10/15/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Birth asphyxia (BA) and subsequent hypoxic-ischemic encephalopathy (HIE) is one of the most serious birth complications affecting full-term infants and can result in severe disabilities including mental retardation, cerebral palsy, and epilepsy. Animal models of BA and HIE are important to characterize the functional and behavioral correlates of injury, explore cellular and molecular mechanisms, and assess the potential of novel therapeutic strategies. Here we used a non-invasive, physiologically validated rat model of BA and acute neonatal seizures that mimics many features of BA and HIE in human infants to study (i) the temporal development of epilepsy with spontaneous recurrent seizures (SRS) in the weeks and months after the initial brain injury, (ii) alterations in seizure threshold and hippocampal EEG that may precede the onset of SRS, and (iii) the effect of prophylactic treatment with midazolam. For this purpose, a total of 89 rat pups underwent asphyxia or sham asphyxia at postnatal day 11 and were examined over 8-10.5 months. In vehicle-treated animals, the incidence of electroclinical SRS progressively increased from 0 % at 2.5 months to 50 % at 6.5 months, 75 % at 8.5 months, and > 80 % at 10.5 months after asphyxia. Unexpectedly, post-asphyxial rats did not differ from sham-exposed rats in seizure threshold or interictal epileptiform discharges in the EEG. Treatment with midazolam (1 mg/kg i.p.) after asphyxia, which suppressed acute symptomatic neonatal seizures in about 60 % of the rat pups, significantly reduced the incidence of SRS regardless of its effect on neonatal seizures. This antiepileptogenic effect of midazolam adds to the recently reported prophylactic effects of this drug on BA-induced neuroinflammation, brain damage, behavioral alterations, and cognitive impairment in the rat asphyxia model of HIE.
Collapse
Affiliation(s)
- Ricardo Schmidt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany
| | - Björn Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Annika Merten
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Hannah Naundorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
3
|
Quinlan S, Witherspoon E, Forcelli PA. Padsevonil suppresses seizures without inducing cell death in neonatal rats. Pharmacol Rep 2024; 76:1055-1066. [PMID: 39028384 PMCID: PMC11584979 DOI: 10.1007/s43440-024-00628-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Padsevonil (PSL) is a rationally designed anti-seizure medication (ASM) which has overlapping mechanisms of action with the two most common ASMs used for neonatal seizures, phenobarbital (PB) and levetiracetam (LEV). Here we evaluated the anti-seizure properties of PSL across the neonatal and adolescent period in rats in the pentlyenetetrazole (PTZ) induced seizures model. METHODS Postnatal day (P)7, P14 and P21 Sprague-Dawley rat pups were pre-treated with PSL (1-30 mg/kg), and assessed for seizure latency and severity 30 min later following injection of PTZ. A separate cohort of P7 pups were treated with neonatal ASMs and euthanized 24 h later (on P8) to assess induction of cell death, a feature common to many ASMs when given to P7 rodents. This effect has been extensively reported with PB, but not with LEV. Cell death was assessed by PathoGreen staining. RESULTS PSL suppressed PTZ-evoked seizures across multiple age groups, particularly at higher doses, without producing increased cell death compared to vehicle. The effects of PSL were particularly notable at suppressing tonic-clonic seizure manifestations (82% of P7 and 100% of P14 and P21 animals were protected from tonic-clonic seizures with the 30 mg/kg dose). CONCLUSIONS PSL displayed dose-dependent anti-seizure effects in immature rodents in the PTZ model of seizures in immature rats. While many ASMs, including PB, induce cell death in neonatal rats, PSL does not. This suggests that PSL may offer therapeutic benefit and a favorable safety profile for the treatment of neonatal seizures.
Collapse
Affiliation(s)
- Sean Quinlan
- Department of Pharmacology & Physiology, Georgetown University, New Research Bldg., W209B, 3970 Reservoir Road NW, Washington, DC, 20007, USA
| | - Eric Witherspoon
- Department of Pharmacology & Physiology, Georgetown University, New Research Bldg., W209B, 3970 Reservoir Road NW, Washington, DC, 20007, USA
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University, New Research Bldg., W209B, 3970 Reservoir Road NW, Washington, DC, 20007, USA.
- Department of Neuroscience, Georgetown University, Washington, DC, USA.
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
| |
Collapse
|
4
|
Lee KKY, Chattopadhyaya B, do Nascimento ASF, Moquin L, Rosa-Neto P, Amilhon B, Di Cristo G. Neonatal hypoxia impairs serotonin release and cognitive functions in adult mice. Neurobiol Dis 2024; 193:106465. [PMID: 38460800 DOI: 10.1016/j.nbd.2024.106465] [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: 08/04/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
Children who experienced moderate perinatal asphyxia (MPA) are at risk of developing long lasting subtle cognitive and behavioral deficits, including learning disabilities and emotional problems. The prefrontal cortex (PFC) regulates cognitive flexibility and emotional behavior. Neurons that release serotonin (5-HT) project to the PFC, and compounds modulating 5-HT activity influence emotion and cognition. Whether 5-HT dysregulations contribute to MPA-induced cognitive problems is unknown. We established a MPA mouse model, which displays recognition and spatial memory impairments and dysfunctional cognitive flexibility. We found that 5-HT expression levels, quantified by immunohistochemistry, and 5-HT release, quantified by in vivo microdialysis in awake mice, are reduced in PFC of adult MPA mice. MPA mice also show impaired body temperature regulation following injection of the 5-HT1A receptor agonist 8-OH-DPAT, suggesting the presence of deficits in 5-HT auto-receptor function on raphe neurons. Finally, chronic treatment of adult MPA mice with fluoxetine, an inhibitor of 5-HT reuptake transporter, or the 5-HT1A receptor agonist tandospirone rescues cognitive flexibility and memory impairments. All together, these data demonstrate that the development of 5-HT system function is vulnerable to moderate perinatal asphyxia. 5-HT hypofunction might in turn contribute to long-term cognitive impairment in adulthood, indicating a potential target for pharmacological therapies.
Collapse
Affiliation(s)
- Karen Ka Yan Lee
- Neurosciences Department, Université de Montréal, Montréal, Canada; CHU Sainte-Justine Azrieli Research Center, Montréal, Canada
| | | | | | - Luc Moquin
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Canada
| | - Pedro Rosa-Neto
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Canada
| | - Bénédicte Amilhon
- Neurosciences Department, Université de Montréal, Montréal, Canada; CHU Sainte-Justine Azrieli Research Center, Montréal, Canada.
| | - Graziella Di Cristo
- Neurosciences Department, Université de Montréal, Montréal, Canada; CHU Sainte-Justine Azrieli Research Center, Montréal, Canada.
| |
Collapse
|
5
|
Abbasi H, Davidson JO, Dhillon SK, Zhou KQ, Wassink G, Gunn AJ, Bennet L. Deep Learning for Generalized EEG Seizure Detection after Hypoxia-Ischemia-Preclinical Validation. Bioengineering (Basel) 2024; 11:217. [PMID: 38534490 DOI: 10.3390/bioengineering11030217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
Brain maturity and many clinical treatments such as therapeutic hypothermia (TH) can significantly influence the morphology of neonatal EEG seizures after hypoxia-ischemia (HI), and so there is a need for generalized automatic seizure identification. This study validates efficacy of advanced deep-learning pattern classifiers based on a convolutional neural network (CNN) for seizure detection after HI in fetal sheep and determines the effects of maturation and brain cooling on their accuracy. The cohorts included HI-normothermia term (n = 7), HI-hypothermia term (n = 14), sham-normothermia term (n = 5), and HI-normothermia preterm (n = 14) groups, with a total of >17,300 h of recordings. Algorithms were trained and tested using leave-one-out cross-validation and k-fold cross-validation approaches. The accuracy of the term-trained seizure detectors was consistently excellent for HI-normothermia preterm data (accuracy = 99.5%, area under curve (AUC) = 99.2%). Conversely, when the HI-normothermia preterm data were used in training, the performance on HI-normothermia term and HI-hypothermia term data fell (accuracy = 98.6%, AUC = 96.5% and accuracy = 96.9%, AUC = 89.6%, respectively). Findings suggest that HI-normothermia preterm seizures do not contain all the spectral features seen at term. Nevertheless, an average 5-fold cross-validated accuracy of 99.7% (AUC = 99.4%) was achieved from all seizure detectors. This significant advancement highlights the reliability of the proposed deep-learning algorithms in identifying clinically translatable post-HI stereotypic seizures in 256Hz recordings, regardless of maturity and with minimal impact from hypothermia.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Auckland Bioengineering Institute (ABI), University of Auckland, Auckland 1010, New Zealand
| | - Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Simerdeep K Dhillon
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Kelly Q Zhou
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Guido Wassink
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Alistair J Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
6
|
Sanchez-Brualla I, Ghosh A, Gibatova VA, Quinlan S, Witherspoon E, Vicini S, Forcelli PA. Phenobarbital does not worsen outcomes of neonatal hypoxia on hippocampal LTP on rats. Front Neurol 2023; 14:1295934. [PMID: 38073649 PMCID: PMC10703306 DOI: 10.3389/fneur.2023.1295934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/06/2023] [Indexed: 10/28/2024] Open
Abstract
Introduction Neonatal hypoxia is a common cause of early-life seizures. Both hypoxia-induced seizures (HS), and the drugs used to treat them (e.g., phenobarbital, PB), have been reported to have long-lasting impacts on brain development. For example, in neonatal rodents, HS reduces hippocampal long-term potentiation (LTP), while PB exposure disrupts GABAergic synaptic maturation in the hippocampus. Prior studies have examined the impact of HS and drug treatment separately, but in the clinic, PB is unlikely to be given to neonates without seizures, and neonates with seizures are very likely to receive PB. To address this gap, we assessed the combined and separate impacts of neonatal HS and PB treatment on the development of hippocampal LTP. Methods Male and female postnatal day (P)7 rat pups were subjected to graded global hypoxia (or normoxia as a control) and treated with either PB (or vehicle as a control). On P13-14 (P13+) or P29-37 (P29+), we recorded LTP of the Schaffer collaterals into CA1 pyramidal layer in acute hippocampal slices. We compared responses to theta burst stimulation (TBS) and tetanization induction protocols. Results Under the TBS induction protocol, female rats showed an LTP impairment caused by HS, which appeared only at P29+. This impairment was delayed compared to male rats. While LTP in HS males was impaired at P13+, it normalized by P29+. Under the tetanization protocol, hypoxia produced larger LTP in males compared to female rats. PB injection, under TBS, did not exacerbate the effects of hypoxia. However, with the tetanization protocol, PB - on the background of HS - compensated for these effects, returning LTP to control levels. Discussion These results point to different susceptibility to hypoxia as a function of sex and age, and a non-detrimental effect of PB when administered after hypoxic seizures.
Collapse
Affiliation(s)
- Irene Sanchez-Brualla
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Anjik Ghosh
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Viktoriya A. Gibatova
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Sean Quinlan
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Eric Witherspoon
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- Department of Neuroscience, Georgetown University, Washington, DC, United States
| | - Patrick A. Forcelli
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- Department of Neuroscience, Georgetown University, Washington, DC, United States
| |
Collapse
|
7
|
Primiani CT, Lee JK, O’Brien CE, Chen MW, Perin J, Kulikowicz E, Santos P, Adams S, Lester B, Rivera-Diaz N, Olberding V, Niedzwiecki MV, Ritzl EK, Habela CW, Liu X, Yang ZJ, Koehler RC, Martin LJ. Hypothermic Protection in Neocortex Is Topographic and Laminar, Seizure Unmitigating, and Partially Rescues Neurons Depleted of RNA Splicing Protein Rbfox3/NeuN in Neonatal Hypoxic-Ischemic Male Piglets. Cells 2023; 12:2454. [PMID: 37887298 PMCID: PMC10605428 DOI: 10.3390/cells12202454] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
The effects of hypothermia on neonatal encephalopathy may vary topographically and cytopathologically in the neocortex with manifestations potentially influenced by seizures that alter the severity, distribution, and type of neuropathology. We developed a neonatal piglet survival model of hypoxic-ischemic (HI) encephalopathy and hypothermia (HT) with continuous electroencephalography (cEEG) for seizures. Neonatal male piglets received HI-normothermia (NT), HI-HT, sham-NT, or sham-HT treatments. Randomized unmedicated sham and HI piglets underwent cEEG during recovery. Survival was 2-7 days. Normal and pathological neurons were counted in different neocortical areas, identified by cytoarchitecture and connectomics, using hematoxylin and eosin staining and immunohistochemistry for RNA-binding FOX-1 homolog 3 (Rbfox3/NeuN). Seizure burden was determined. HI-NT piglets had a reduced normal/total neuron ratio and increased ischemic-necrotic/total neuron ratio relative to sham-NT and sham-HT piglets with differing severities in the anterior and posterior motor, somatosensory, and frontal cortices. Neocortical neuropathology was attenuated by HT. HT protection was prominent in layer III of the inferior parietal cortex. Rbfox3 immunoreactivity distinguished cortical neurons as: Rbfox3-positive/normal, Rbfox3-positive/ischemic-necrotic, and Rbfox3-depleted. HI piglets had an increased Rbfox3-depleted/total neuron ratio in layers II and III compared to sham-NT piglets. Neuronal Rbfox3 depletion was partly rescued by HT. Seizure burdens in HI-NT and HI-HT piglets were similar. We conclude that the neonatal HI piglet neocortex has: (1) suprasylvian vulnerability to HI and seizures; (2) a limited neuronal cytopathological repertoire in functionally different regions that engages protective mechanisms with HT; (3) higher seizure burden, insensitive to HT, that is correlated with more panlaminar ischemic-necrotic neurons in the somatosensory cortex; and (4) pathological RNA splicing protein nuclear depletion that is sensitive to HT. This work demonstrates that HT protection of the neocortex in neonatal HI is topographic and laminar, seizure unmitigating, and restores neuronal depletion of RNA splicing factor.
Collapse
Affiliation(s)
- Christopher T. Primiani
- Department of Neurology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Jennifer K. Lee
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Caitlin E. O’Brien
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - May W. Chen
- Department Pediatrics, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Jamie Perin
- Department of Biostatistics and Epidemiology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Ewa Kulikowicz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Polan Santos
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Shawn Adams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Bailey Lester
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Natalia Rivera-Diaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Valerie Olberding
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Mark V. Niedzwiecki
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Eva K. Ritzl
- Department of Neurology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Christa W. Habela
- Department of Neurology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Xiuyun Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Zeng-Jin Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Raymond C. Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
| | - Lee J. Martin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA; (J.K.L.); (E.K.); (V.O.); (M.V.N.)
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
- The Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| |
Collapse
|
8
|
Roberts NS, Handy MJ, Ito Y, Hashimoto K, Jensen FE, Talos DM. Anti-seizure efficacy of perampanel in two established rodent models of early-life epilepsy. Epilepsy Behav 2023; 143:109194. [PMID: 37119576 DOI: 10.1016/j.yebeh.2023.109194] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/10/2023] [Accepted: 03/23/2023] [Indexed: 05/01/2023]
Abstract
Early-life seizures can be refractory to conventional antiseizure medications (ASMs) and can also result in chronic epilepsy and long-term behavioral and cognitive deficits. Treatments targeting age-specific mechanisms contributing to epilepsy would be of clinical benefit. One such target is the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subtype of excitatory glutamate receptor, which is upregulated in the developing brain. Perampanel is a non-competitive, selective AMPAR antagonist that is FDA-approved for focal onset seizures (FOS) or primary generalized tonic-clonic seizures (PGTC) in children and adults. However, the efficacy of perampanel treatment in epilepsy patients younger than 4 years has been less documented. We thus tested the efficacy of perampanel in two early-life seizure models: (1) a rat model of hypoxia-induced neonatal seizures and (2) a mouse model of Dravet syndrome with hyperthermia-induced seizures. Pretreatment with perampanel conferred dose-dependent protection against early-life seizures in both experimental models. These findings suggest that AMPAR-mediated hyperexcitability could be involved in the pathophysiology of early-life seizures, which may be amenable to treatment with perampanel.
Collapse
Affiliation(s)
- Nicholas S Roberts
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcus J Handy
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yoshimasa Ito
- Formerly: Neurology Business Group, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Keisuke Hashimoto
- Deep Human Biology Learning, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Delia M Talos
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Guez-Barber D, Eisch AJ, Cristancho AG. Developmental Brain Injury and Social Determinants of Health: Opportunities to Combine Preclinical Models for Mechanistic Insights into Recovery. Dev Neurosci 2023; 45:255-267. [PMID: 37080174 PMCID: PMC10614252 DOI: 10.1159/000530745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Epidemiological studies show that social determinants of health are among the strongest factors associated with developmental outcomes after prenatal and perinatal brain injuries, even when controlling for the severity of the initial injury. Elevated socioeconomic status and a higher level of parental education correlate with improved neurologic function after premature birth. Conversely, children experiencing early life adversity have worse outcomes after developmental brain injuries. Animal models have provided vital insight into mechanisms perturbed by developmental brain injuries, which have indicated directions for novel therapeutics or interventions. Animal models have also been used to learn how social environments affect brain maturation through enriched environments and early adverse conditions. We recognize animal models cannot fully recapitulate human social circumstances. However, we posit that mechanistic studies combining models of developmental brain injuries and early life social environments will provide insight into pathways important for recovery. Some studies combining enriched environments with neonatal hypoxic injury models have shown improvements in developmental outcomes, but further studies are needed to understand the mechanisms underlying these improvements. By contrast, there have been more limited studies of the effects of adverse conditions on developmental brain injury extent and recovery. Uncovering the biological underpinnings for early life social experiences has translational relevance, enabling the development of novel strategies to improve outcomes through lifelong treatment. With the emergence of new technologies to analyze subtle molecular and behavioral phenotypes, here we discuss the opportunities for combining animal models of developmental brain injury with social construct models to deconvolute the complex interactions between injury, recovery, and social inequity.
Collapse
Affiliation(s)
- Danielle Guez-Barber
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana G. Cristancho
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Salyha N, Oliynyk I. Hypoxia modeling techniques: A review. Heliyon 2023; 9:e13238. [PMID: 36718422 PMCID: PMC9877323 DOI: 10.1016/j.heliyon.2023.e13238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Hypoxia is the main cause and effect of a large number of diseases, including the most recent one facing the world, the coronavirus disease (COVID-19). Hypoxia is divided into short-term, long-term, and periodic, it can be the result of diseases, climate change, or living and traveling in the high mountain regions of the world. Since each type of hypoxia can be a cause and a consequence of various physiological changes, the methods for modeling these hypoxias are also different. There are many techniques for modeling hypoxia under experimental conditions. The most common animal for modeling hypoxia is a rat. Hypoxia models (hypoxia simulations) in rats are a tool to study the effect of various conditions on the oxygen supply of the body. These models can provide a necessary information to understand hypoxia and also provide effective treatment, highlighting the importance of various reactions of the body to hypoxia. The main parameters when choosing a model should be reproducibility and the goal that the scientist wants to achieve. Hypoxia in rats can be reproduced both ways exogenously and endogenously. The reason for writing this review was the aim to systematize the models of rats available in the literature in order to facilitate their selection by scientists. The relative strengths and limitations of each model need to be identified and understood in order to evaluate the information obtained from these models and extrapolate these results to humans to develop the necessary generalizations. Despite these problems, animal models have been and remain vital to understanding the mechanisms involved in the development and progression of hypoxia. The eligibility criteria for the selected studies was a comprehensive review of the methods and results obtained from the studies. This made it possible to make generalizations and give recommendations on the application of these methods. The review will assist scientists in choosing an appropriate hypoxia simulation method, as well as assist in interpreting the results obtained with these methods.
Collapse
Affiliation(s)
- Nataliya Salyha
- Institute of Animal Biology NAAS, Lviv, Ukraine,Corresponding author
| | | |
Collapse
|
11
|
Welzel B, Schmidt R, Johne M, Löscher W. Midazolam Prevents the Adverse Outcome of Neonatal Asphyxia. Ann Neurol 2023; 93:226-243. [PMID: 36054632 DOI: 10.1002/ana.26498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/09/2022] [Accepted: 08/29/2022] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Birth asphyxia (BA) is the most frequent cause of neonatal death as well as central nervous system (CNS) injury. BA is often associated with neonatal seizures, which only poorly respond to anti-seizure medications and may contribute to the adverse neurodevelopmental outcome. Using a non-invasive rat model of BA, we have recently reported that the potent benzodiazepine, midazolam, prevents neonatal seizures in ~50% of rat pups. In addition to its anti-seizure effect, midazolam exerts anti-inflammatory actions, which is highly relevant for therapeutic intervention following BA. The 2 major aims of the present study were to examine (1) whether midazolam reduces the adverse outcome of BA, and (2) whether this effect is different in rats that did or did not exhibit neonatal seizures after drug treatment. METHODS Behavioral and cognitive tests were performed over 14 months after asphyxia, followed by immunohistochemical analyses. RESULTS All vehicle-treated rats had seizures after asphyxia and developed behavioral and cognitive abnormalities, neuroinflammation in gray and white matter, neurodegeneration in the hippocampus and thalamus, and hippocampal mossy fiber sprouting in subsequent months. Administration of midazolam (1 mg/kg i.p.) directly after asphyxia prevented post-asphyctic seizures in ~50% of the rats and resulted in the prevention or decrease of neuroinflammation and the behavioral, cognitive, and neurodegenerative consequences of asphyxia. Except for neurodegeneration in the thalamus, seizures did not seem to contribute to the adverse outcome of asphyxia. INTERPRETATION The disease-modifying effect of midazolam identified here strongly suggests that this drug provides a valuable option for improving the treatment and outcome of BA. ANN NEUROL 2023;93:226-243.
Collapse
Affiliation(s)
- Björn Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Ricardo Schmidt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
12
|
Arida RM. Physical Exercise as a Strategy to Reduce Seizure Susceptibility. PHARMACORESISTANCE IN EPILEPSY 2023:453-477. [DOI: 10.1007/978-3-031-36526-3_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
13
|
Johnson KJ, Moy B, Rensing N, Robinson A, Ly M, Chengalvala R, Wong M, Galindo R. Functional neuropathology of neonatal hypoxia-ischemia by single-mouse longitudinal electroencephalography. Epilepsia 2022; 63:3037-3050. [PMID: 36054439 PMCID: PMC10176800 DOI: 10.1111/epi.17403] [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: 04/08/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Neonatal cerebral hypoxia-ischemia (HI) results in symptomatic seizures and long-term neurodevelopmental disability. The Rice-Vannucci model of rodent neonatal HI has been used extensively to examine and translate the functional consequences of acute and chronic HI-induced encephalopathy. Yet, longitudinal electrophysiological characterization of this brain injury model has been limited by the size of the neonatal mouse's head and postnatal maternal dependency. We overcome this challenge by employing a novel method of longitudinal single-mouse electroencephalography (EEG) using chronically implanted subcranial electrodes in the term-equivalent mouse pup. We characterize the neurophysiological disturbances occurring during awake and sleep states in the acute and chronic phases following newborn brain injury. METHODS C57BL/6 mice underwent long-term bilateral subcranial EEG and electromyographic electrode placement at postnatal day 9 followed by unilateral carotid cauterization and exposure to 40 minutes of hypoxia the following day. EEG recordings were obtained prior, during, and intermittently after the HI procedure from postnatal day 10 to weaning age. Quantitative EEG and fast Fourier transform analysis were used to evaluate seizures, cortical cerebral dysfunction, and disturbances in vigilance states. RESULTS We observed neonatal HI-provoked electrographic focal and bilateral seizures during or immediately following global hypoxia and most commonly contralateral to the ischemic injury. Spontaneous chronic seizures were not seen. Injured mice developed long-term asymmetric EEG background attenuation in all frequencies and most prominently during non-rapid eye movement (NREM) sleep. HI mice also showed transient impairments in vigilance state duration and transitions during the first 2 days following injury. SIGNIFICANCE The functional burden of mouse neonatal HI recorded by EEG resembles closely that of the injured human newborn. The use of single-mouse longitudinal EEG in this immature model can advance our understanding of the developmental and pathophysiological mechanisms of neonatal cerebral injury and help translate novel therapeutic strategies against this devastating condition.
Collapse
Affiliation(s)
- Kevin J Johnson
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brianna Moy
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas Rensing
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexia Robinson
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Ly
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ramya Chengalvala
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Wong
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rafael Galindo
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
14
|
Zayachkivsky A, Lehmkuhle MJ, Ekstrand JJ, Dudek FE. Background suppression of electrical activity is a potential biomarker of subsequent brain injury in a rat model of neonatal hypoxia-ischemia. J Neurophysiol 2022; 128:118-130. [PMID: 35675445 DOI: 10.1152/jn.00024.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Electrographic seizures and abnormal background activity in the neonatal electroencephalogram (EEG) may differentiate between harmful versus benign brain insults. Using two animal models of neonatal seizures, electrical activity was recorded in freely behaving rats and examined quantitatively during successive time periods with field-potential recordings obtained shortly after the brain insult (i.e., 0-4 days). Single-channel, differential recordings with miniature wireless telemetry were used to analyze spontaneous electrographic seizures and background suppression of electrical activity after 1) hypoxia-ischemia (HI), which is a model of neonatal encephalopathy that causes acute seizures and a large brain lesion with possible development of epilepsy, 2) hypoxia alone (Ha), which causes severe acute seizures without an obvious lesion or subsequent epilepsy, and 3) sham control rats. Background EEG exhibited increases in power as a function of age in control animals. Although background electrical activity was depressed in all frequency bands immediately after HI, suppression in the β and γ bands was greatest and lasted longest. Spontaneous electrographic seizures were recorded, but only in a few HI-treated animals. Ha-treated rat pups were similar to sham controls, they had no subsequent spontaneous electrographic seizures after the treatment and background suppression was only briefly observed in one frequency band. Thus, the normal age-dependent maturation of electrical activity patterns in control animals was significantly disrupted after HI. Suppression of the background EEG observed here after HI-induced acute seizures and subsequent brain injury may be a noninvasive biomarker for detecting severe brain injuries and may help predict subsequent epilepsy.NEW & NOTEWORTHY Biomarkers of neonatal brain injury are needed. Hypoxia-ischemia (HI) in immature rat pups caused severe brain injury, which was associated with strongly suppressed background EEG. The suppression was most robust in the β and γ bands; it started immediately after the HI injury and persisted for days. Thus, background suppression may be a noninvasive biomarker for detecting severe brain injuries and may help predict subsequent epilepsy.
Collapse
Affiliation(s)
- A Zayachkivsky
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - M J Lehmkuhle
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - J J Ekstrand
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - F E Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
15
|
Nehlig A, Sperling MR. The debate on the treatment of neonatal seizures introductory remarks. Epilepsia 2022; 63:1862. [PMID: 35524449 DOI: 10.1111/epi.17275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
|
16
|
Kaila K, Löscher W. Bumetanide for neonatal seizures: no light in the pharmacokinetic/dynamic tunnel. Epilepsia 2022; 63:1868-1873. [PMID: 35524446 PMCID: PMC9545618 DOI: 10.1111/epi.17279] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
In his editorial, Kevin Staley criticizes our recent work demonstrating the lack of effect of bumetanide in a novel model of neonatal seizures. The main points in our response are that (1) our work is on an asphyxia model, not one on "hypercarbia only"; (2) clinically relevant parenteral doses of bumetanide applied in vivo lead to concentrations in the brain parenchyma that are at least an order of magnitude lower than what would be sufficient to exert any direct effect—even a transient one—on neuronal functions, including neonatal seizures; and (3) moreover, bumetanide's molecular target in the brain is the Na‐K‐2Cl cotransporter NKCC1, which has vital functions in neurons, astrocytes, and oligodendrocytes as well as microglia. This would make it impossible even for highly brain‐permeant NKCC1 blockers to specifically target depolarizing and excitatory actions of γ‐aminobutyric acid in principal neurons of the brain, which is postulated as the rationale of clinical trials on neonatal seizures.
Collapse
Affiliation(s)
- Kai Kaila
- Molecular and Integrative Biosciences (MIBS) and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
17
|
Cristancho AG, Gadra EC, Samba IM, Zhao C, Ouyang M, Magnitsky S, Huang H, Viaene AN, Anderson SA, Marsh ED. Deficits in Seizure Threshold and Other Behaviors in Adult Mice without Gross Neuroanatomic Injury after Late Gestation Transient Prenatal Hypoxia. Dev Neurosci 2022; 44:246-265. [PMID: 35279653 PMCID: PMC9464267 DOI: 10.1159/000524045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/07/2022] [Indexed: 11/19/2022] Open
Abstract
Intrauterine hypoxia is a common cause of brain injury in children resulting in a broad spectrum of long-term neurodevelopmental sequela, including life-long disabilities that can occur even in the absence of severe neuroanatomic damage. Postnatal hypoxia-ischemia rodent models are commonly used to understand the effects of ischemia and transient hypoxia on the developing brain. Postnatal models, however, have some limitations. First, they do not test the impact of placental pathologies on outcomes from hypoxia. Second, they primarily recapitulate severe injury because they provoke substantial cell death, which is not seen in children with mild hypoxic injury. Lastly, they do not model preterm hypoxic injury. Prenatal models of hypoxia in mice may allow us to address some of these limitations to expand our understanding of developmental brain injury. The published rodent models of prenatal hypoxia employ multiple days of hypoxic exposure or complicated surgical procedures, making these models challenging to perform consistently in mice. Furthermore, large animal models suggest that transient prenatal hypoxia without ischemia is sufficient to lead to significant functional impairment to the developing brain. However, these large animal studies are resource-intensive and not readily amenable to mechanistic molecular studies. Therefore, here we characterized the effect of late gestation (embryonic day 17.5) transient prenatal hypoxia (5% inspired oxygen) on long-term anatomical and neurodevelopmental outcomes in mice. Late gestation transient prenatal hypoxia increased hypoxia-inducible factor 1 alpha protein levels (a marker of hypoxic exposure) in the fetal brain. Hypoxia exposure predisposed animals to decreased weight at postnatal day 2, which normalized by day 8. However, hypoxia did not affect gestational age at birth, litter size at birth, or pup survival. No differences in fetal brain cell death or long-term gray or white matter changes resulted from hypoxia. Animals exposed to prenatal hypoxia did have several long-term functional consequences, including sex-dichotomous changes. Hypoxia exposure was associated with a decreased seizure threshold and abnormalities in hindlimb strength and repetitive behaviors in males and females. Males exposed to hypoxia had increased anxiety-related deficits, whereas females had deficits in social interaction. Neither sex developed any motor or visual learning deficits. This study demonstrates that late gestation transient prenatal hypoxia in mice is a simple, clinically relevant paradigm for studying putative environmental and genetic modulators of the long-term effects of hypoxia on the developing brain.
Collapse
Affiliation(s)
- Ana G. Cristancho
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Elyse C. Gadra
- Department of Child and Adolescent Psychiatry and Behavioral Services, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Ima M. Samba
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Chenying Zhao
- Radiology Research, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, U.S.A
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Minhui Ouyang
- Radiology Research, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, U.S.A
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, PA, U.S.A
| | - Sergey Magnitsky
- Radiology Research, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, U.S.A
| | - Hao Huang
- Radiology Research, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, U.S.A
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, PA, U.S.A
| | - Angela N. Viaene
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A. and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry and Behavioral Services, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Eric D. Marsh
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| |
Collapse
|
18
|
Wang Y, Wei P, Yan F, Luo Y, Zhao G. Animal Models of Epilepsy: A Phenotype-oriented Review. Aging Dis 2022; 13:215-231. [PMID: 35111370 PMCID: PMC8782545 DOI: 10.14336/ad.2021.0723] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022] Open
Abstract
Epilepsy is a serious neurological disorder characterized by abnormal, recurrent, and synchronous discharges in the brain. Long-term recurrent seizure attacks can cause serious damage to brain function, which is usually observed in patients with temporal lobe epilepsy. Controlling seizure attacks is vital for the treatment and prognosis of epilepsy. Animal models, such as the kindling model, which was the most widely used model in the past, allow the understanding of the potential epileptogenic mechanisms and selection of antiepileptic drugs. In recent years, various animal models of epilepsy have been established to mimic different seizure types, without clear merits and demerits. Accordingly, this review provides a summary of the views mentioned above, aiming to provide a reference for animal model selection.
Collapse
Affiliation(s)
- Yilin Wang
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Penghu Wei
- 1Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| | - Feng Yan
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yumin Luo
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| | - Guoguang Zhao
- 1Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.,3Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Zen R, Terashima T, Tsuji S, Katagi M, Ohashi N, Nobuta Y, Higuchi A, Kanai H, Murakami T, Kojima H. Ambient Temperature Is Correlated With the Severity of Neonatal Hypoxic-Ischemic Brain Injury via Microglial Accumulation in Mice. Front Pediatr 2022; 10:883556. [PMID: 35601427 PMCID: PMC9120824 DOI: 10.3389/fped.2022.883556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The pathophysiology of neonatal hypoxic-ischemic encephalopathy (HIE) has been studied in several rodent models to develop novel treatments. Although it is well known that high ambient temperature results in severe HIE, the effect of subtle changes in ambient temperature during a hypoxic-ischemic (HI) insult has not been studied. Therefore, in order to clarify the difference of pathophysiological change among the HIE models due to the influence of small changes in chamber temperature, three-step gradual change of 0.5°C each were prepared in ambient temperature during hypoxic exposure. METHODS Blood flow in the left common carotid artery (CCA) of neonatal mice was interrupted using bipolar electronic forceps under general and local anesthesia. The mice were subsequently subjected to 10% hypoxic exposure for 50 min at 36.0, 36.5, or 37.0°C. A control group was also included in the study. The size of the striatum and hippocampus and the volume reduction rate of the hemisphere in the section containing them on the ischemic side were evaluated using microtubule associated protein 2 (MAP2) immunostaining. The accumulation of Iba1-positive cells was investigated to assess inflammation. Additionally, rotarod and open-field tests were performed 2 weeks after HI insult to assess its effect on physiological conditions. RESULTS MAP2 staining revealed that the higher the temperature during hypoxia, the more severe the volume reduction rate in the hemisphere, striatum, and hippocampus. The number of Iba1-positive cells in the ipsilateral lesion gradually increased with increasing temperature, and there was a significant difference in motor function in the 36.5 and 37.0°C groups compared with the sham group. In the open-field tests, there was a significant decrease in performance in the 37.0°C groups compared with the 36.0°C and sham groups. CONCLUSIONS Even a small gradual change of 0.5°C produced a significant difference in pathological and behavioral changes and contributed to the accumulation of Iba1-positive cells. The arrangement of ambient temperature is useful for creating a rodent model with the appropriate severity of the targeted neuropsychological symptoms to establish a novel therapy for HIE.
Collapse
Affiliation(s)
- Rika Zen
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan.,Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yuri Nobuta
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Asuka Higuchi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hirohiko Kanai
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
20
|
Löscher W, Kaila K. CNS pharmacology of NKCC1 inhibitors. Neuropharmacology 2021; 205:108910. [PMID: 34883135 DOI: 10.1016/j.neuropharm.2021.108910] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022]
Abstract
The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used off-label in attempts to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been initiated over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs lead to significantly higher brain levels than the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. In parallel, a major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drugs within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., the blood-brain barrier, the choroid plexus, and the endocrine system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| |
Collapse
|
21
|
Chen L, Ren SY, Li RX, Liu K, Chen JF, Yang YJ, Deng YB, Wang HZ, Xiao L, Mei F, Wang F. Chronic Exposure to Hypoxia Inhibits Myelinogenesis and Causes Motor Coordination Deficits in Adult Mice. Neurosci Bull 2021; 37:1397-1411. [PMID: 34292513 PMCID: PMC8490606 DOI: 10.1007/s12264-021-00745-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/06/2021] [Indexed: 12/18/2022] Open
Abstract
Exposure to chronic hypoxia is considered to be a risk factor for deficits in brain function in adults, but the underlying mechanisms remain largely unknown. Since active myelinogenesis persists in the adult central nervous system, here we aimed to investigate the impact of chronic hypoxia on myelination and the related functional consequences in adult mice. Using a transgenic approach to label newly-generated myelin sheaths (NG2-CreERTM; Tau-mGFP), we found that myelinogenesis was highly active in most brain regions, such as the motor cortex and corpus callosum. After exposure to hypoxia (10% oxygen) 12 h per day for 4 weeks, myelinogenesis was largely inhibited in the 4-month old brain and the mice displayed motor coordination deficits revealed by the beam-walking test. To determine the relationship between the inhibited myelination and functional impairment, we induced oligodendroglia-specific deletion of the transcription factor Olig2 by tamoxifen (NG2-CreERTM; Tau-mGFP; Olig2 fl/fl) in adult mice to mimic the decreased myelinogenesis caused by hypoxia. The deletion of Olig2 inhibited myelinogenesis and consequently impaired motor coordination, suggesting that myelinogenesis is required for motor function in adult mice. To understand whether enhancing myelination could protect brain functions against hypoxia, we treated hypoxic mice with the myelination-enhancing drug-clemastine, which resulted in enhanced myelogenesis and improved motor coordination. Taken together, our data indicate that chronic hypoxia inhibits myelinogenesis and causes functional deficits in the brain and that enhancing myelinogenesis protects brain functions against hypoxia-related deficits.
Collapse
Affiliation(s)
- Lin Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Rui-Xue Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Kun Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Yu-Jian Yang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Yong-Bin Deng
- Department of Neurosurgery, Chongqing Emergency Medical Center, Chongqing University, Chongqing, 400014, China
| | - Han-Zhi Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China.
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
22
|
Merelli A, Repetto M, Lazarowski A, Auzmendi J. Hypoxia, Oxidative Stress, and Inflammation: Three Faces of Neurodegenerative Diseases. J Alzheimers Dis 2021; 82:S109-S126. [PMID: 33325385 DOI: 10.3233/jad-201074] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cerebral hypoxia-ischemia can induce a wide spectrum of biologic responses that include depolarization, excitotoxicity, oxidative stress, inflammation, and apoptosis, and result in neurodegeneration. Several adaptive and survival endogenous mechanisms can also be activated giving an opportunity for the affected cells to remain alive, waiting for helper signals that avoid apoptosis. These signals appear to help cells, depending on intensity, chronicity, and proximity to the central hypoxic area of the affected tissue. These mechanisms are present not only in a large list of brain pathologies affecting commonly older individuals, but also in other pathologies such as refractory epilepsies, encephalopathies, or brain trauma, where neurodegenerative features such as cognitive and/or motor deficits sequelae can be developed. The hypoxia inducible factor 1α (HIF-1α) is a master transcription factor driving a wide spectrum cellular response. HIF-1α may induce erythropoietin (EPO) receptor overexpression, which provides the therapeutic opportunity to administer pharmacological doses of EPO to rescue and/or repair affected brain tissue. Intranasal administration of EPO combined with other antioxidant and anti-inflammatory compounds could become an effective therapeutic alternative, to avoid and/or slow down neurodegenerative deterioration without producing adverse peripheral effects.
Collapse
Affiliation(s)
- Amalia Merelli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Marisa Repetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (IBIMOL, UBA-CONICET), Argentina
| | - Alberto Lazarowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Jerónimo Auzmendi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
23
|
Gailus B, Naundorf H, Welzel L, Johne M, Römermann K, Kaila K, Löscher W. Long-term outcome in a noninvasive rat model of birth asphyxia with neonatal seizures: Cognitive impairment, anxiety, epilepsy, and structural brain alterations. Epilepsia 2021; 62:2826-2844. [PMID: 34458992 DOI: 10.1111/epi.17050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/30/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Birth asphyxia is a major cause of hypoxic-ischemic encephalopathy (HIE) in neonates and often associated with mortality, neonatal seizures, brain damage, and later life motor, cognitive, and behavioral impairments and epilepsy. Preclinical studies on rodent models are needed to develop more effective therapies for preventing HIE and its consequences. Thus far, the most popular rodent models have used either exposure of intact animals to hypoxia-only, or a combination of hypoxia and carotid occlusion, for the induction of neonatal seizures and adverse outcomes. However, such models lack systemic hypercapnia, which is a fundamental constituent of birth asphyxia with major effects on neuronal excitability. Here, we use a recently developed noninvasive rat model of birth asphyxia with subsequent neonatal seizures to study later life adverse outcome. METHODS Intermittent asphyxia was induced for 30 min by exposing male and female postnatal day 11 rat pups to three 7 + 3-min cycles of 9% and 5% O2 at constant 20% CO2 . All pups exhibited convulsive seizures after asphyxia. A set of behavioral tests were performed systematically over 14 months following asphyxia, that is, a large part of the rat's life span. Video-electroencephalographic (EEG) monitoring was used to determine whether asphyxia led to the development of epilepsy. Finally, structural brain alterations were examined. RESULTS The animals showed impaired spatial learning and memory and increased anxiety when tested at an age of 3-14 months. Video-EEG at ~10 months showed an abundance of spontaneous seizures, which was paralleled by neurodegeneration in the hippocampus and thalamus, and by aberrant mossy fiber sprouting. SIGNIFICANCE The present model of birth asphyxia recapitulates several of the later life consequences associated with human HIE. This model thus allows evaluation of the efficacy of novel therapies designed to prevent HIE and seizures following asphyxia, and of how such therapies might alleviate long-term adverse consequences.
Collapse
Affiliation(s)
- Björn Gailus
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Hannah Naundorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, Helsinki, Finland.,Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
24
|
Volpe JJ. Commentary - Early discontinuation of antiseizure medication in neonatal seizures - Proceed with caution. J Neonatal Perinatal Med 2021; 15:203-207. [PMID: 34459421 PMCID: PMC9108580 DOI: 10.3233/npm-210849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- J J Volpe
- Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Newborn Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Ziobro JM, Eschbach K, Shellhaas RA. Novel Therapeutics for Neonatal Seizures. Neurotherapeutics 2021; 18:1564-1581. [PMID: 34386906 PMCID: PMC8608938 DOI: 10.1007/s13311-021-01085-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/04/2023] Open
Abstract
Neonatal seizures are a common neurologic emergency for which therapies have not significantly changed in decades. Improvements in diagnosis and pathophysiologic understanding of the distinct features of acute symptomatic seizures and neonatal-onset epilepsies present exceptional opportunities for development of precision therapies with potential to improve outcomes. Herein, we discuss the pathophysiology of neonatal seizures and review the evidence for currently available treatment. We present emerging therapies in clinical and preclinical development for the treatment of acute symptomatic neonatal seizures. Lastly, we discuss the role of precision therapies for genetic neonatal-onset epilepsies and address barriers and goals for developing new therapies for clinical care.
Collapse
Affiliation(s)
- Julie M Ziobro
- Department of Pediatrics, Michigan Medicine, C.S. Mott Children's Hospital, University of Michigan, 1540 E. Hospital Dr, Ann Arbor, MI, USA.
| | - Krista Eschbach
- Department of Pediatrics, Section of Neurology, Denver Anschutz School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, CO, 80045, USA
| | - Renée A Shellhaas
- Department of Pediatrics, Michigan Medicine, C.S. Mott Children's Hospital, University of Michigan, 1540 E. Hospital Dr, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Pan W, Song X, Hu Q, Zhang Y. miR-485 inhibits histone deacetylase HDAC5, HIF1α and PFKFB3 expression to alleviate epilepsy in cellular and rodent models. Aging (Albany NY) 2021; 13:14416-14432. [PMID: 34021541 PMCID: PMC8202868 DOI: 10.18632/aging.203058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/05/2020] [Indexed: 01/15/2023]
Abstract
We investigated the role of microRNA (miR)-485 and its downstream signaling molecules on mediating epilepsy in cellular and rat models. We established a cellular epilepsy model by exposing hippocampal neurons to magnesium and a rat model by treating ICR mice with lithium chloride (127 mg/kg) and pilocarpine (30 mg/kg). We confirmed that miR-485 could bind and inhibit histone deacetylase 5 (HDAC5) and then measured expression of miR-485 and in mice and cells. Cells were transfected with overexpression or knockdown of miR-485, HDAC5, hypoxia-inducible factor-1alpha (HIF1α), or 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 enzyme (PFKFB3) to verify their roles in apoptosis, oxidative stress, and inflammation in epileptic hippocampal neurons. Binding relationship between miR-485, HDAC5, HIF1α, and PFKFB3 was verified. Oxidative stress and inflammation marker levels in epilepsy model mice were assessed. miR-485 was downregulated and HDAC5 was upregulated in cell and animal model of epilepsy. Seizure, neuronal apoptosis, oxidative stress (increased SOD and GSH-Px expression and decreased MDA and 8-OHdG expression) and inflammation (reduced IL-1β, TNF-α, and IL-6 expression) were reduced by miR-485 in epileptic cells. HIF1α and PFKFB3 expression was reduced by HDAC5 knockdown in cells, which was recapitulated in vivo. Thus, miR-485 alleviates neuronal damage and epilepsy by inhibiting HDAC5, HIF1α, and PFKFB3.
Collapse
Affiliation(s)
- Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Xingyu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qibo Hu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yunfeng Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
27
|
Johne M, Käufer C, Römermann K, Gailus B, Gericke B, Löscher W. A combination of phenobarbital and the bumetanide derivative bumepamine prevents neonatal seizures and subsequent hippocampal neurodegeneration in a rat model of birth asphyxia. Epilepsia 2021; 62:1460-1471. [PMID: 33955541 DOI: 10.1111/epi.16912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Bumetanide was suggested as an adjunct to phenobarbital for suppression of neonatal seizures. This suggestion was based on the idea that bumetanide, by reducing intraneuronal chloride accumulation through inhibition of the Na-K-2Cl cotransporter NKCC1, may attenuate or abolish depolarizing γ-aminobutyric acid (GABA) responses caused by birth asphyxia. However, a first proof-of-concept clinical trial failed. This could have had several reasons, including bumetanide's poor brain penetration, the wide cellular NKCC1 expression pattern in the brain, and problems with the general concept of NKCC1's role in neonatal seizures. We recently replicated the clinical failure of bumetanide to potentiate phenobarbital's effect in a novel rat model of birth asphyxia. In this study, a clinically relevant dose (0.3 mg/kg) of bumetanide was used that does not lead to NKCC1-inhibitory brain levels. The aim of the present experiments was to examine whether a much higher dose (10 mg/kg) of bumetanide is capable of potentiating phenobarbital in this rat model. Furthermore, the effects of the two lipophilic bumetanide derivatives, the ester prodrug N,N-dimethylaminoethylester of bumetanide (DIMAEB) and the benzylamine derivative bumepamine, were examined at equimolar doses. METHODS Intermittent asphyxia was induced for 30 min by exposing male and female P11 rat pups to three 7 + 3 min cycles of 9% and 5% O2 at constant 20% CO2 . All control pups exhibited neonatal seizures after the asphyxia. RESULTS Even at 10 mg/kg, bumetanide did not potentiate the effect of a submaximal dose (15 mg/kg) of phenobarbital on seizure incidence, whereas a significant suppression of neonatal seizures was determined for combinations of phenobarbital with DIMAEB or, more effectively, bumepamine, which, however, does not inhibit NKCC1. Of interest, the bumepamine/phenobarbital combination prevented the neurodegenerative consequences of asphyxia and seizures in the hippocampus. SIGNIFICANCE Both bumepamine and DIMAEB are promising tools that may help to develop more effective lead compounds for clinical trials.
Collapse
Affiliation(s)
- Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Björn Gailus
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
28
|
Ala‐Kurikka T, Pospelov A, Summanen M, Alafuzoff A, Kurki S, Voipio J, Kaila K. A physiologically validated rat model of term birth asphyxia with seizure generation after, not during, brain hypoxia. Epilepsia 2021; 62:908-919. [PMID: 33338272 PMCID: PMC8246723 DOI: 10.1111/epi.16790] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Birth asphyxia (BA) is often associated with seizures that may exacerbate the ensuing hypoxic-ischemic encephalopathy. In rodent models of BA, exposure to hypoxia is used to evoke seizures, that commence already during the insult. This is in stark contrast to clinical BA, in which seizures are typically seen upon recovery. Here, we introduce a term-equivalent rat model of BA, in which seizures are triggered after exposure to asphyxia. METHODS Postnatal day 11-12 male rat pups were exposed to steady asphyxia (15 min; air containing 5% O2 + 20% CO2 ) or to intermittent asphyxia (30 min; three 5 + 5-min cycles of 9% and 5% O2 at 20% CO2 ). Cortical activity and electrographic seizures were recorded in freely behaving animals. Simultaneous electrode measurements of intracortical pH, Po2 , and local field potentials (LFPs) were made under urethane anesthesia. RESULTS Both protocols decreased blood pH to <7.0 and brain pH from 7.3 to 6.7 and led to a fall in base excess by 20 mmol·L-1 . Electrographic seizures with convulsions spanning the entire Racine scale were triggered after intermittent but not steady asphyxia. In the presence of 20% CO2 , brain Po2 was only transiently affected by 9% ambient O2 but fell below detection level during the steps to 5% O2 , and LFP activity was nearly abolished. Post-asphyxia seizures were strongly suppressed when brain pH recovery was slowed down by 5% CO2 . SIGNIFICANCE The rate of brain pH recovery has a strong influence on post-asphyxia seizure propensity. The recurring hypoxic episodes during intermittent asphyxia promote neuronal excitability, which leads to seizures only after the suppressing effect of the hypercapnic acidosis is relieved. The present rodent model of BA is to our best knowledge the first one in which, consistent with clinical BA, behavioral and electrographic seizures are triggered after and not during the BA-mimicking insult.
Collapse
Affiliation(s)
- Tommi Ala‐Kurikka
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Alexey Pospelov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Milla Summanen
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Aleksander Alafuzoff
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Samu Kurki
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Juha Voipio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
| | - Kai Kaila
- Faculty of Biological and Environmental Sciences, Molecular and Integrative BiosciencesUniversity of HelsinkiHelsinkiFinland
- Neuroscience Center (HiLIFE)University of HelsinkiHelsinkiFinland
| |
Collapse
|
29
|
Johne M, Römermann K, Hampel P, Gailus B, Theilmann W, Ala-Kurikka T, Kaila K, Löscher W. Phenobarbital and midazolam suppress neonatal seizures in a noninvasive rat model of birth asphyxia, whereas bumetanide is ineffective. Epilepsia 2020; 62:920-934. [PMID: 33258158 DOI: 10.1111/epi.16778] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Neonatal seizures are the most frequent type of neurological emergency in newborn infants, often being a consequence of prolonged perinatal asphyxia. Phenobarbital is currently the most widely used antiseizure drug for treatment of neonatal seizures, but fails to stop them in ~50% of cases. In a neonatal hypoxia-only model based on 11-day-old (P11) rats, the NKCC1 inhibitor bumetanide was reported to potentiate the antiseizure activity of phenobarbital, whereas it was ineffective in a human trial in neonates. The aim of this study was to evaluate the effect of clinically relevant doses of bumetanide as add-on to phenobarbital on neonatal seizures in a noninvasive model of birth asphyxia in P11 rats, designed for better translation to the human term neonate. METHODS Intermittent asphyxia was induced for 30 minutes by exposing the rat pups to three 7 + 3-minute cycles of 9% and 5% O2 at constant 20% CO2 . Drug treatments were administered intraperitoneally either before or immediately after asphyxia. RESULTS All untreated rat pups had seizures within 10 minutes after termination of asphyxia. Phenobarbital significantly blocked seizures when applied before asphyxia at 30 mg/kg but not 15 mg/kg. Administration of phenobarbital after asphyxia was ineffective, whereas midazolam (0.3 or 1 mg/kg) exerted significant antiseizure effects when administered before or after asphyxia. In general, focal seizures were more resistant to treatment than generalized convulsive seizures. Bumetanide (0.3 mg/kg) alone or in combination with phenobarbital (15 or 30 mg/kg) exerted no significant effect on seizure occurrence. SIGNIFICANCE The data demonstrate that bumetanide does not increase the efficacy of phenobarbital in a model of birth asphyxia, which is consistent with the negative data of the recent human trial. The translational data obtained with the novel rat model of birth asphyxia indicate that it is a useful tool to evaluate novel treatments for neonatal seizures.
Collapse
Affiliation(s)
- Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Philip Hampel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Björn Gailus
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Wiebke Theilmann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Tommi Ala-Kurikka
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
30
|
Arida RM. Physical exercise and seizure activity. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165979. [PMID: 32980461 DOI: 10.1016/j.bbadis.2020.165979] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 09/05/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Neuroprotective and antiepileptogenic therapies have been extensively investigated for epilepsy prevention and treatment. This review gives an overview of the promising contribution of the ketogenic diet, a complementary treatment, on the intestinal microbiota to reduce seizure susceptibility. Next, the relevance of physical exercise is extensively addressed as a complementary therapy to reduce seizure susceptibility, and thereby impact beneficially on the epilepsy condition. In this context, particular attention is given to the potential risks and benefits of physical exercise, possible precipitant factors related to exercise and proposed mechanisms by which exercise can reduce seizures, and its antiepileptogenic effects. Finally, this review points to emerging evidence of exercise reducing comorbidities from epilepsy and improving the quality of life of people with epilepsy. Based on evidence from current literature, physical or sport activities represent a potential non-pharmacological intervention that can be integrated with conventional therapy for epilepsy.
Collapse
Affiliation(s)
- Ricardo Mario Arida
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
31
|
Pospelov AS, Puskarjov M, Kaila K, Voipio J. Endogenous brain-sparing responses in brain pH and PO 2 in a rodent model of birth asphyxia. Acta Physiol (Oxf) 2020; 229:e13467. [PMID: 32174009 DOI: 10.1111/apha.13467] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
AIM To study brain-sparing physiological responses in a rodent model of birth asphyxia which reproduces the asphyxia-defining systemic hypoxia and hypercapnia. METHODS Steady or intermittent asphyxia was induced for 15-45 minutes in anaesthetized 6- and 11-days old rats and neonatal guinea pigs using gases containing 5% or 9% O2 plus 20% CO2 (in N2 ). Hypoxia and hypercapnia were induced with low O2 and high CO2 respectively. Oxygen partial pressure (PO2 ) and pH were measured with microsensors within the brain and subcutaneous ("body") tissue. Blood lactate was measured after asphyxia. RESULTS Brain and body PO2 fell to apparent zero with little recovery during 5% O2 asphyxia and 5% or 9% O2 hypoxia, and increased more than twofold during 20% CO2 hypercapnia. Unlike body PO2 , brain PO2 recovered rapidly to control after a transient fall (rat), or was slightly higher than control (guinea pig) during 9% O2 asphyxia. Asphyxia (5% O2 ) induced a respiratory acidosis paralleled by a progressive metabolic (lact)acidosis that was much smaller within than outside the brain. Hypoxia (5% O2 ) produced a brain-confined alkalosis. Hypercapnia outlasting asphyxia suppressed pH recovery and prolonged the post-asphyxia PO2 overshoot. All pH changes were accompanied by consistent shifts in the blood-brain barrier potential. CONCLUSION Regardless of brain maturation stage, hypercapnia can restore brain PO2 and protect the brain against metabolic acidosis despite compromised oxygen availability during asphyxia. This effect extends to the recovery phase if normocapnia is restored slowly, and it is absent during hypoxia, demonstrating that exposure to hypoxia does not mimic asphyxia.
Collapse
Affiliation(s)
- Alexey S. Pospelov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Martin Puskarjov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Kai Kaila
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
- Neuroscience Center (HiLIFE) University of Helsinki Helsinki Finland
| | - Juha Voipio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| |
Collapse
|
32
|
Seizures severity during rewarming can predict seizure outcomes of infants with neonatal hypoxic-ischemic encephalopathy following therapeutic hypothermia. Biomed J 2020; 43:285-292. [PMID: 32684487 PMCID: PMC7424086 DOI: 10.1016/j.bj.2020.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background The aim of this study was to examine the predictive value of amplitude-integrated electroencephalography (aEEG) on 12-month seizure outcomes of infants with neonatal hypoxic-ischemic encephalopathy (HIE) treated with therapeutic hypothermia. Methods We conducted this retrospective cohort study in a tertiary neonatal intensive care unit between May 2012 and September 2017. Neonates with HIE who received both therapeutic hypothermia (TH) and aEEG were enrolled. Results A total of 23 infants (14 boys, nine girls) with a mean gestational age of 38.9 weeks were enrolled. Fifteen (65%) infants had moderate HIE and eight (35%) had severe HIE according to modified Sarnat staging. The mean aEEG recording time was 107.5 h. Twenty (86.9%) infants had seizure activity during the first 24 h after cooling and 14 (60.8%) had seizure activity during the first 24 h after rewarming. At 12 months, five (21.7%) infants had poor seizure outcomes. Repetitive seizures or status epilepticus pattern during the first 24 h after rewarming, but not the first 24 h after cooling, were associated with the presence of epilepsy at 12 months (p = 0.037). Conclusions We identified a high incidence of electrographic seizures in infants with neonatal HIE treated with therapeutic hypothermia, and post-neonatal epilepsy in the children who survived after HIE. Repetitive seizures or status epilepticus pattern during the first 24 h after rewarming, but not in the first 24 h after cooling, were associated with the presence of epilepsy at 12 months.
Collapse
|
33
|
Abbasi H, Unsworth CP. Electroencephalogram studies of hypoxic ischemia in fetal and neonatal animal models. Neural Regen Res 2020; 15:828-837. [PMID: 31719243 PMCID: PMC6990791 DOI: 10.4103/1673-5374.268892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alongside clinical achievements, experiments conducted on animal models (including primate or non-primate) have been effective in the understanding of various pathophysiological aspects of perinatal hypoxic/ischemic encephalopathy (HIE). Due to the reasonably fair degree of flexibility with experiments, most of the research around HIE in the literature has been largely concerned with the neurodevelopmental outcome or how the frequency and duration of HI seizures could relate to the severity of perinatal brain injury, following HI insult. This survey concentrates on how EEG experimental studies using asphyxiated animal models (in rodents, piglets, sheep and non-human primate monkeys) provide a unique opportunity to examine from the exact time of HI event to help gain insights into HIE where human studies become difficult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Aravamuthan BR, Gandham S, Young AB, Rutkove SB. Sex may influence motor phenotype in a novel rodent model of cerebral palsy. Neurobiol Dis 2019; 134:104711. [PMID: 31841677 PMCID: PMC9128630 DOI: 10.1016/j.nbd.2019.104711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/27/2019] [Accepted: 12/09/2019] [Indexed: 12/30/2022] Open
Abstract
Cerebral palsy (CP) is the most common cause of childhood motor disability, manifesting most often as spasticity and/or dystonia. Spasticity and dystonia are often co-morbid clinically following severe injury at term gestation. Currently available animal CP models have not demonstrated or differentiated between these two motor phenotypes, limiting their clinical relevance. We sought to develop an animal CP model displaying objectively identifiable spasticity and dystonia. We exposed rat pups at post-natal day 7–8 (equivalent to human 37 postconceptional weeks) to global hypoxia. Since spasticity and dystonia can be difficult to differentiate from each other in CP, objective electrophysiologic markers of motor phenotypes were assessed. Spasticity was inferred using an electrophysiologic measure of hyperreflexia: soleus Hoffman reflex suppression with 2 Hz tibial nerve stimulation. Dystonia was assessed during voluntary isometric hindlimb withdrawal at different levels of arousal by calculating tibialis anterior and triceps surae electromyographic co-activation as a surrogate of overflow muscle activity. Hypoxia affected spasticity and dystonia measures in a sex-dependent manner. Males had attenuated Hoffman reflex suppression suggestive of spasticity but no change in antagonist muscle co-activation. In contrast, females demonstrated increased co-activation suggestive of dystonia but no change in Hoffman reflex suppression. Therefore, there was an unexpected segregation of electrophysiologically-defined motor phenotypes based on sex with males predominantly demonstrating spasticity and females predominantly demonstrating dystonia. These results require human clinical confirmation but suggest that sex could play a critical role in the motor manifestations of neonatal brain injury.
Collapse
Affiliation(s)
- Bhooma R Aravamuthan
- Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Sushma Gandham
- Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Anne B Young
- Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA
| | - Seward B Rutkove
- Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
35
|
Hagag AA, El Frargy MS, Abd El-Latif AE. Vitamin D as an Adjuvant Therapy in Neonatal Hypoxia: Is it Beneficial? Endocr Metab Immune Disord Drug Targets 2019; 19:341-348. [PMID: 30514196 PMCID: PMC7040512 DOI: 10.2174/1871530319666181204151044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/19/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022]
Abstract
Background Neonatal hypoxic ischemic encephalopathy (HIE) is a potentially devastating disorder associated with significant mortality and long-term morbidity. Objective The aim of this study was to study the role of vitamin D as an adjuvant therapy for management of neonatal HIE. Patients and Methods This study was carried out on 60 neonates with HIE grade II who were diagnosed according to modified Sarnat staging and were divided in to 2 groups: Group I: Included 30 neonates with Sarnat grade II HIE who received single daily oral dose of vitamin D3 (1000 IU) for 2 weeks in addition to daily subcutaneous (SC) human recombinant erythropoietin (2500 IU/kg) for 5 days and IM or IV magnesium sulphate 250 mg/kg within half an hour of birth, and subsequently 125 mg/kg at 24 and 48 hours of life. Group II: Included 30 neonates with HIE grade II who received erythropoietin and magnesium sulphate as group I but without vitamin D. Two blood samples were taken from all neonates included in both groups; the 1st at diagnosis and the 2nd after 2 weeks of therapy. This study included also 30 healthy neonates as a control group. All neonates included in this study were subjected to: complete clinical examination with assessment of Apgar score at 5 and 10 minutes, measurement of arterial blood gases and serum 25 (OH) vitamin D, calcium, phosphorus, S100-B and IL-17 levels. Results Before therapy, there were no significant differences between group I and II in PH, PO2 and PCO2 (p= 0.294, 0.462, 0.758 respectively), but after 2 weeks of therapy, there were significantly higher PH levels in group I compared with group II (p <0.001) while there were no significant differences between group I and II regarding PO2 and PCO2. Before therapy, there were no significant differences in serum 25(OH) vitamin D levels between group I and II while there were significantly lower serum 25(OH) vitamin D levels in group I and II compared with controls (P1; comparison between group I and II = 0.742, P2; comparison between group I and controls = 0.001 and P3; comparison between group II and controls = 0. 001). There were no significant differences between group I and II and between group I and II and control as regard serum calcium (P1= 0.943, P2= 0.875 and P3= 0.764) and phosphorus (P1= 0.862, P2= 0.921, P3= 0.786). There were no significant differences between group I and II regarding serum IL-17 levels while there were significantly lower serum IL-17 levels in group I and II compared with controls (P1 = 0.457, P2 = 0.043 and P3 = 0.023). Before therapy, there were no significant differences in serum S100-B levels between group I and II while there were significantly higher serum S100-B levels in group I and II compared with control (P1 = 0.381, P2 = 0.001 and P3= 0.001) but after therapy, there were significantly higher S100-B levels in group II compared with group I and significantly higher S100-B levels in group I and II compared with control (P1= 0.001, P2= 0.043, P3 = 0.001). There were significant negative correlations in group I between serum S100-B and PH and between S100-B and serum vitamin D before and after therapy. Conclusion Vitamin D was found to improve the cases of group I as demonstrated by the reduction of serum S100-B levels after vitamin D therapy. Recommendations Extensive multicenter studies are required on a large number of patients with Sarnat grade II HIE with longer duration of follow up to give valid recommendations about the use of vitamin D as an adjuvant therapy in Sarnat grade II HIE.
Collapse
Affiliation(s)
- Adel A Hagag
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta, Gharbia, Egypt
| | - Mohamed S El Frargy
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta, Gharbia, Egypt
| | - Amal E Abd El-Latif
- Department of Clinical Pathology, Faculty of Medicine, Tanta University, Tanta, Gharbia, Egypt
| |
Collapse
|
36
|
Li X, Giri V, Cui Y, Yin M, Xian Z, Li J. LncRNA FTX inhibits hippocampal neuron apoptosis by regulating miR-21-5p/SOX7 axis in a rat model of temporal lobe epilepsy. Biochem Biophys Res Commun 2019; 512:79-86. [DOI: 10.1016/j.bbrc.2019.03.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/03/2019] [Indexed: 12/14/2022]
|
37
|
Abbasi H, Bennet L, Gunn AJ, Unsworth CP. Latent Phase Detection of Hypoxic-Ischemic Spike Transients in the EEG of Preterm Fetal Sheep Using Reverse Biorthogonal Wavelets & Fuzzy Classifier. Int J Neural Syst 2019; 29:1950013. [PMID: 31184228 DOI: 10.1142/s0129065719500138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic (HI) studies in preterms lack reliable prognostic biomarkers for diagnostic tests of HI encephalopathy (HIE). Our group's observations from in utero fetal sheep models suggest that potential biomarkers of HIE in the form of developing HI micro-scale epileptiform transients emerge along suppressed EEG/ECoG background during a latent phase of 6-7h post-insult. However, having to observe for the whole of the latent phase disqualifies any chance of clinical intervention. A precise automatic identification of these transients can help for a well-timed diagnosis of the HIE and to stop the spread of the injury before it becomes irreversible. This paper reports fusion of Reverse-Biorthogonal Wavelets with Type-1 Fuzzy classifiers, for the accurate real-time automatic identification and quantification of high-frequency HI spike transients in the latent phase, tested over seven in utero preterm sheep. Considerable high performance of 99.78 ± 0.10% was obtained from the Rbio-Wavelet Type-1 Fuzzy classifier for automatic identification of HI spikes tested over 42h of high-resolution recordings (sampling-freq:1024Hz). Data from post-insult automatic time-localization of high-frequency HI spikes reveals a promising trend in the average rate of the HI spikes, even in the animals with shorter occlusion periods, which highlights considerable higher number of transients within the first 2h post-insult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
38
|
Salas J, Reddy N, Orru E, Carson KA, Chavez-Valdez R, Burton VJ, Stafstrom CE, Northington FJ, Huisman TAGM. The Role of Diffusion Tensor Imaging in Detecting Hippocampal Injury Following Neonatal Hypoxic-Ischemic Encephalopathy. J Neuroimaging 2018; 29:252-259. [PMID: 30325083 DOI: 10.1111/jon.12572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/12/2018] [Accepted: 10/03/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Neonatal hypoxic-ischemic injury of the brain and resultant encephalopathy (HIE) leads to major developmental impairments by school age. Conventional/anatomical MRI often fails to detect hippocampal injury in mild cases. We hypothesize that diffusion tensor imaging (DTI) has greater sensitivity for identifying subtle hippocampal injury. METHODS We retrospectively analyzed DTI data collected from a cohort of neonates with HIE and controls. Conventional MRI sequences were classified qualitatively according to severity using a modified Barkovich scale. Using multivariate linear regression, we compared hippocampal DTI scalars of HIE patients and controls. Spearman correlation was used to test the association of DTI scalars in the hippocampal and thalamic regions. A multiple regression analysis tested the association of the DTI scalars with short-term outcomes. RESULTS Fifty-five neonates with HIE (42% males) and 13 controls (54% males) were included. Hippocampal DTI scalars were similar between HIE and control groups, even when restricting the HIE group to those with moderate-to-severe injury (8 subjects). DTI scalars of the thalamus were significantly lower in the moderate-to-severely affected patients compared to controls (right fractional anisotropy [FA] .148 vs. .182, P = .01; left FA .147 vs. .181, P = .03). Hippocampal and thalamic DTI scalars were correlated (P < .001). Hippocampal DTI scalars were not associated with short-term outcomes. CONCLUSIONS Quantitative DTI analysis of the hippocampus in neonates following HIE is a feasible technique to examine neuronal injury. Although DTI scalars were useful in identifying thalamic injury in our cohort, hippocampal DTI analysis did not provide additional information regarding hippocampal injury following HIE.
Collapse
Affiliation(s)
- Jacqueline Salas
- Division of Neonatology, Department of Pediatrics, The Johns Hopkins University School of Medicine, The Charlotte R. Bloomberg Children's Center, Baltimore, MD.,Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Nihaal Reddy
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Emanuele Orru
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kathryn A Carson
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, The Johns Hopkins University School of Medicine, The Charlotte R. Bloomberg Children's Center, Baltimore, MD.,Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Vera Joanna Burton
- Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD.,Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD
| | - Carl E Stafstrom
- Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD.,Division of Pediatric Neurology, Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Frances J Northington
- Division of Neonatology, Department of Pediatrics, The Johns Hopkins University School of Medicine, The Charlotte R. Bloomberg Children's Center, Baltimore, MD.,Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Thierry A G M Huisman
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD.,Neuro-Intensive Care Nursery Group, The Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
39
|
Blauwblomme T, Dzhala V, Staley K. Transient ischemia facilitates neuronal chloride accumulation and severity of seizures. Ann Clin Transl Neurol 2018; 5:1048-1061. [PMID: 30250862 PMCID: PMC6144438 DOI: 10.1002/acn3.617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Objective Preceding oxygen glucose deprivation (OGD) and ongoing seizures have both been reported to increase neuronal chloride concentration ([Cl−]i), which may contribute to anticonvulsant failure by reversing the direction of chloride currents at inhibitory GABAA synapses. Methods The effects of OGD on [Cl−]i, seizure activity, and anticonvulsant efficacy were studied in a chronically epileptic in vitro preparation. Results Seizures initially increased during OGD, followed by suppression. On reperfusion, seizure frequency and [Cl−]i progressively increased, and phenobarbital efficacy was reduced. Bumetanide (10 μmol/L) and furosemide (1 mmol/L) prevented or reduced the OGD induced [Cl−]i increase. Phenobarbital efficacy was enhanced by bumetanide (10 μmol/L). Furosemide (1 mmol/L) suppressed recurrent seizures. Interpretation [Cl−]i increases after OGD and is associated with worsened seizure activity, reduced efficacy of GABAergic anticonvulsants, and amelioration by antagonists of secondary chloride transport.
Collapse
Affiliation(s)
- Thomas Blauwblomme
- Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts 02114.,Universite Paris Descartes Sorbonne Paris Cite Paris France
| | - Volodymyr Dzhala
- Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts 02114
| | - Kevin Staley
- Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts 02114
| |
Collapse
|
40
|
Quinlan SMM, Rodriguez-Alvarez N, Molloy EJ, Madden SF, Boylan GB, Henshall DC, Jimenez-Mateos EM. Complex spectrum of phenobarbital effects in a mouse model of neonatal hypoxia-induced seizures. Sci Rep 2018; 8:9986. [PMID: 29968748 PMCID: PMC6030182 DOI: 10.1038/s41598-018-28044-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/11/2018] [Indexed: 11/09/2022] Open
Abstract
Seizures in neonates, mainly caused by hypoxic-ischemic encephalopathy, are thought to be harmful to the brain. Phenobarbital remains the first line drug therapy for the treatment of suspected neonatal seizures but concerns remain with efficacy and safety. Here we explored the short- and long-term outcomes of phenobarbital treatment in a mouse model of hypoxia-induced neonatal seizures. Seizures were induced in P7 mice by exposure to 5% O2 for 15 minutes. Immediately after hypoxia, pups received a single dose of phenobarbital (25 mg.kg-1) or saline. We observed that after administration of phenobarbital seizure burden and number of seizures were reduced compared to the hypoxic period; however, PhB did not suppress acute histopathology. Behavioural analysis of mice at 5 weeks of age previously subjected to hypoxia-seizures revealed an increase in anxiety-like behaviour and impaired memory function compared to control littermates, and these effects were not normalized by phenobarbital. In a seizure susceptibility test, pups previously exposed to hypoxia, with or without phenobarbital, developed longer and more severe seizures in response to kainic acid injection compared to control mice. Unexpectedly, mice treated with phenobarbital developed less hippocampal damage after kainic acid than untreated counterparts. The present study suggests phenobarbital treatment in immature mice does not improve the long lasting functional deficits induces by hypoxia-induced seizures but, unexpectedly, may reduce neuronal death caused by exposure to a second seizure event in later life.
Collapse
Affiliation(s)
- Sean M M Quinlan
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123, St Stephen Green, Dublin, 2, Dublin, Ireland
| | - Natalia Rodriguez-Alvarez
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123, St Stephen Green, Dublin, 2, Dublin, Ireland
| | - Eleanor J Molloy
- Paediatrics, Academic Centre, Tallaght Hospital, Trinity College, The University of Dublin, Dublin, Ireland.,Neonatology, Coombe Women and Infants' University Hospital, Dublin, Ireland.,Neonatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Beaux Lane House, Royal College of Surgeons in Ireland, Dublin, 2, Ireland
| | - Geraldine B Boylan
- Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123, St Stephen Green, Dublin, 2, Dublin, Ireland.,Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123, St Stephen Green, Dublin, 2, Dublin, Ireland.
| |
Collapse
|
41
|
Kasahara Y, Ikegaya Y, Koyama R. Neonatal Seizure Models to Study Epileptogenesis. Front Pharmacol 2018; 9:385. [PMID: 29720941 PMCID: PMC5915831 DOI: 10.3389/fphar.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Current therapeutic strategies for epilepsy include anti-epileptic drugs and surgical treatments that are mainly focused on the suppression of existing seizures rather than the occurrence of the first spontaneous seizure. These symptomatic treatments help a certain proportion of patients, but these strategies are not intended to clarify the cellular and molecular mechanisms underlying the primary process of epilepsy development, i.e., epileptogenesis. Epileptogenic changes include reorganization of neural and glial circuits, resulting in the formation of an epileptogenic focus. To achieve the goal of developing “anti-epileptogenic” drugs, we need to clarify the step-by-step mechanisms underlying epileptogenesis for patients whose seizures are not controllable with existing “anti-epileptic” drugs. Epileptogenesis has been studied using animal models of neonatal seizures because such models are useful for studying the latent period before the occurrence of spontaneous seizures and the lowering of the seizure threshold. Further, neonatal seizure models are generally easy to handle and can be applied for in vitro studies because cells in the neonatal brain are suitable for culture. Here, we review two animal models of neonatal seizures for studying epileptogenesis and discuss their features, specifically focusing on hypoxia-ischemia (HI)-induced seizures and febrile seizures (FSs). Studying these models will contribute to identifying the potential therapeutic targets and biomarkers of epileptogenesis.
Collapse
Affiliation(s)
- Yuka Kasahara
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
42
|
Auzmendi J, Buchholz B, Salguero J, Cañellas C, Kelly J, Men P, Zubillaga M, Rossi A, Merelli A, Gelpi RJ, Ramos AJ, Lazarowski A. Pilocarpine-Induced Status Epilepticus Is Associated with P-Glycoprotein Induction in Cardiomyocytes, Electrocardiographic Changes, and Sudden Death. Pharmaceuticals (Basel) 2018; 11:ph11010021. [PMID: 29462915 PMCID: PMC5874717 DOI: 10.3390/ph11010021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the major cause of death in those patients suffering from refractory epilepsy (RE), with a 24-fold higher risk relative to the normal population. SUDEP risk increases with seizure frequency and/or seizure-duration as in RE and Status Epilepticus (SE). P-glycoprotein (P-gp), the product of the multidrug resistant ABCB1-MDR-1 gene, is a detoxifying pump that extrudes drugs out of the cells and can confer pharmacoresistance to the expressing cells. Neurons and cardiomyocytes normally do not express P-gp, however, it is overexpressed in the brain of patients or in experimental models of RE and SE. P-gp was also detected after brain or cardiac hypoxia. We have previously demonstrated that repetitive pentylenetetrazole (PTZ)-induced seizures increase P-gp expression in the brain, which is associated with membrane depolarization in the hippocampus, and in the heart, which is associated with fatal SE. SE can produce hypoxic-ischemic altered cardiac rhythm (HIACR) and severe arrhythmias, and both are related with SUDEP. Here, we investigate whether SE induces the expression of hypoxia-inducible transcription factor (HIF)-1α and P-gp in cardiomyocytes, which is associated with altered heart rhythm, and if these changes are related with the spontaneous death rate. SE was induced in Wistar rats once a week for 3 weeks, by lithium-pilocarpine-paradigm. Electrocardiograms, HIF-1α, and P-gp expression in cardiomyocytes, were evaluated in basal conditions and 72 h after SE. All spontaneous deaths occurred 48 h after each SE was registered. We observed that repeated SE induced HIF-1α and P-gp expression in cardiomyocytes, electrocardiographic (ECG) changes, and a high rate of spontaneous death. Our results suggest that the highly accumulated burden of convulsive stress results in a hypoxic heart insult, where P-gp expression may play a depolarizing role in cardiomyocyte membranes and in the development of the ECG changes, such as QT interval prolongation, that could be related with SUDEP. We postulate that this mechanism could explain, in part, the higher SUDEP risk in patients with RE or SE.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Bruno Buchholz
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Jimena Salguero
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Carlos Cañellas
- Laboratorio Tecnonuclear SA, Arias 4176, Buenos Aires C1430CRP, Argentina.
| | - Jazmín Kelly
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Paula Men
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Marcela Zubillaga
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Alicia Rossi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Amalia Merelli
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Ricardo J Gelpi
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Alberto J Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Alberto Lazarowski
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| |
Collapse
|
43
|
Abend NS, Jensen FE, Inder TE, Volpe JJ. Neonatal Seizures. VOLPE'S NEUROLOGY OF THE NEWBORN 2018:275-321.e14. [DOI: 10.1016/b978-0-323-42876-7.00012-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Rudy M, Mayer-Proschel M. Iron Deficiency Affects Seizure Susceptibility in a Time- and Sex-Specific Manner. ASN Neuro 2017; 9:1759091417746521. [PMID: 29243938 PMCID: PMC5734468 DOI: 10.1177/1759091417746521] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Iron deficiency (ID) affects more than three billion people worldwide making it the most common micronutrient deficiency. ID is most prevalent during gestation and early life, which is of particular concern since its impact on the developing central nervous system is associated with an increased risk of a wide range of different psychiatric disorders later in life. The cause for this association is not known, but many of these same disorders are also associated with an imbalance between excitation and inhibition (E/I) within the brain. Based on this shared impairment, we asked whether ID could contribute to such an imbalance. Disruptions in the E/I balance can be uncovered by the brain’s response to seizure inducing insults. We therefore tested the seizure threshold under different nutritional models of ID. We found that mice which were postnatally exposed to ID (and were acutely ID) had a decreased seizure threshold and increased susceptibility to certain seizure types. In contrast, mice that were exposed to ID only during gestation had an increased seizure threshold and low seizure incidence. We suggest that exposure to ID during gestation might alter the cellular components that contribute to the establishment of a proper E/I balance later in life. In addition, our data highlight the importance of considering the window of vulnerability since gestational ID and postnatal ID have significantly different consequences on seizure probability.
Collapse
Affiliation(s)
- Michael Rudy
- 1 Department of Environmental Medicine, University of Rochester, NY, USA.,2 Department of Biomedical Genetics, University of Rochester, NY, USA
| | - Margot Mayer-Proschel
- 2 Department of Biomedical Genetics, University of Rochester, NY, USA.,3 Department of Neuroscience, University of Rochester, NY, USA
| |
Collapse
|
45
|
Dunn R, Queenan BN, Pak DTS, Forcelli PA. Divergent effects of levetiracetam and tiagabine against spontaneous seizures in adult rats following neonatal hypoxia. Epilepsy Res 2017; 140:1-7. [PMID: 29227795 DOI: 10.1016/j.eplepsyres.2017.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/09/2017] [Accepted: 12/02/2017] [Indexed: 01/03/2023]
Abstract
Animal models are valuable tools for screening novel therapies for patients who suffer from epilepsy. However, a wide array of models are necessary to cover the diversity of human epilepsies. In humans, neonatal hypoxia (or hypoxia-ischemia) is one of the most common causes of epilepsy early in life. Hypoxia-induced seizures (HS) during the neonatal period can also lead to spontaneous seizures in adulthood. This phenomenon, i.e., early-life hypoxia leading to adult epilepsy - is also seen in experimental models, including rats. However, it is not known which anti-seizure medications are most effective at managing adult epilepsy resulting from neonatal HS. Here, we examined the efficacy of three anti-seizure medications against spontaneous seizures in adult rats with a history of neonatal HS: (1) phenobarbital (PHB), the oldest epilepsy medicine still in use today; (2) levetiracetam (LEV); and (3) tiagabine (TGB). Both LEV and TGB are relatively new anticonvulsant drugs that are ineffective in traditional seizure models, but strikingly effective in other models. We found that PHB and LEV decreased seizures in adult rats with a history of HS, whereas TGB exacerbated seizures. These divergent drug effects indicate that the HS model may be useful for differentiating the clinical efficacy of putative epilepsy therapies.
Collapse
Affiliation(s)
- Raymond Dunn
- Department of Pharmacology and Physiology, United States
| | - Bridget N Queenan
- Department of Pharmacology and Physiology, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States; Neuroscience Research Institute, Department of Mechanical Engineering; Department of Physics, University sof California, Santa Barbara, Santa Barbara, CA, United States
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States.
| | - Patrick A Forcelli
- Department of Pharmacology and Physiology, United States; Department of Neuroscience, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States.
| |
Collapse
|
46
|
Harutyunyan G, Hauer L, Dünser MW, Moser T, Pikija S, Leitinger M, Novak HF, Aichhorn W, Trinka E, Sellner J. Risk Factors for Intensive Care Unit Admission in Patients with Autoimmune Encephalitis. Front Immunol 2017; 8:835. [PMID: 28804482 PMCID: PMC5532517 DOI: 10.3389/fimmu.2017.00835] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/03/2017] [Indexed: 01/15/2023] Open
Abstract
Background Prevention and early recognition of critical illness in patients with autoimmune encephalitis (AE) is essential to achieve better outcome. Aim of the study To evaluate risk factors for intensive care unit (ICU) admission and its prognostic impact in patients with AE. Patients and methods A reclassification of patients hospitalized between 2011 and 2016 revealed 17 “definite” and 15 “probable” AE cases. Thirteen patients (41%) developed critical illness and required ICU admission. The underlying conditions were intractable seizures or status epilepticus (54%), altered mental state (39%), and respiratory failure (8%). Results ICU admission was associated with longer time from first symptoms to hospitalization (p = 0.046). Regression analysis revealed that anemia on hospital admission and definite diagnosis of AE was associated with a higher risk of acquiring critical illness. At last follow-up after a median of 31 months (range 2.5–52.4), seven patients had died (23%) and 63% had a good outcome [modified Rankin Scale (mRS) 0–3]. Anemia was associated with poor prognosis (p = 0.021), whereas development of critical illness did not impact mortality and functional outcome. Conclusion We confirmed the need for ICU care in a subgroup of patients and the prevailing objective is improved seizure control, and definite diagnosis of AE and anemia were identified as risk factors for development of critical illness. However, prognosis was not affected by ICU admission.
Collapse
Affiliation(s)
- Gayane Harutyunyan
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Larissa Hauer
- Department of Psychiatry and Psychotherapy, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Martin W Dünser
- Department of Critical Care, University College of London Hospital, London, United Kingdom
| | - Tobias Moser
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Slaven Pikija
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Markus Leitinger
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Helmut F Novak
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Wolfgang Aichhorn
- Department of Psychiatry and Psychotherapy, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria.,Department of Neurology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| |
Collapse
|
47
|
Huizenga MN, Wicker E, Beck VC, Forcelli PA. Anticonvulsant effect of cannabinoid receptor agonists in models of seizures in developing rats. Epilepsia 2017; 58:1593-1602. [PMID: 28691158 DOI: 10.1111/epi.13842] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Although drugs targeting the cannabinoid system (e.g., CB1 receptor agonists) display anticonvulsant efficacy in adult animal models of seizures/epilepsy, they remain unexplored in developing animal models. However, cannabinoid system functions emerge early in development, providing a rationale for targeting this system in neonates. We examined the therapeutic potential of drugs targeting the cannabinoid system in three seizure models in developing rats. METHODS Postnatal day (P) 10, Sprague-Dawley rat pups were challenged with the chemoconvulsant methyl-6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate (DMCM) or pentylenetetrazole (PTZ), after treatment with either CB1/2 mixed agonist (WIN 55,212-2), CB1 agonist (arachidonyl-2'-chloroethylamide [ACEA]), CB2 agonist (HU-308), CB1 antagonist (AM-251), CB2 antagonist (AM-630), fatty acid amide hydrolase inhibitor (URB-597), or G protein-coupled receptor 55 agonist (O-1602). P20 Sprague-Dawley pups were challenged with DMCM after treatment with WIN, ACEA, or URB. Finally, after pretreatment with WIN, P10 Sprague-Dawley rats were challenged against acute hypoxia-induced seizures. RESULTS The mixed CB1/2 agonist and the CB1-specific agonist, but no other drugs, displayed anticonvulsant effects against clonic seizures in the DMCM model. By contrast, both CB1 and CB2 antagonism increased seizure severity. Similarly, we found that the CB1/2 agonist displayed antiseizure efficacy against acute hypoxia-induced seizures (automatisms, clonic and tonic-clonic seizures) and tonic-clonic seizures evoked by PTZ. Anticonvulsant effects were seen in P10 animals but not P20 animals. SIGNIFICANCE Early life seizures represent a significant cause of morbidity, with 30-40% of infants and children with epilepsy failing to achieve seizure remission with current pharmacotherapy. Identification of new therapies for neonatal/infantile epilepsy syndromes is thus of high priority. These data indicate that the anticonvulsant action of the CB system is specific to CB1 receptor activation during early development and provide justification for further examination of CB1 receptor agonists as novel antiepileptic drugs targeting epilepsy in infants and children.
Collapse
Affiliation(s)
- Megan N Huizenga
- Pharmacology & Physiology, Georgetown University, Washington, District of Columbia, U.S.A
| | - Evan Wicker
- Pharmacology & Physiology, Georgetown University, Washington, District of Columbia, U.S.A
| | - Veronica C Beck
- Pharmacology & Physiology, Georgetown University, Washington, District of Columbia, U.S.A
| | - Patrick A Forcelli
- Pharmacology & Physiology, Georgetown University, Washington, District of Columbia, U.S.A.,Neuroscience, Georgetown University, Washington, District of Columbia, U.S.A.,Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, U.S.A
| |
Collapse
|
48
|
Farrell JS, Colangeli R, Wolff MD, Wall AK, Phillips TJ, George A, Federico P, Teskey GC. Postictal hypoperfusion/hypoxia provides the foundation for a unified theory of seizure-induced brain abnormalities and behavioral dysfunction. Epilepsia 2017. [DOI: 10.1111/epi.13827] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jordan S. Farrell
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Roberto Colangeli
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Marshal D. Wolff
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Alexandra K. Wall
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Thomas J. Phillips
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Antis George
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Paolo Federico
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - G. Campbell Teskey
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| |
Collapse
|
49
|
RILJAK V, KRAF J, DARYANANI A, JIRUŠKA P, OTÁHAL J. Pathophysiology of Perinatal Hypoxic-Ischemic Encephalopathy – Biomarkers, Animal Models and Treatment Perspectives. Physiol Res 2016; 65:S533-S545. [DOI: 10.33549/physiolres.933541] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the leading pediatric neurological conditions causing long-term disabilities and socio-economical burdens. Nearly 20-50 % of asphyxiated newborns with HIE die within the newborn period and another third will develop severe health consequences and permanent handicaps. HIE is the result of severe systemic oxygen deprivation and reduced cerebral blood flow, commonly occurring in full-term infants. Hypoxic-ischemic changes trigger several molecular and cellular processes leading to cell death and inflammation. Generated reactive oxygen species attack surrounding cellular components resulting in functional deficits and mitochondrial dysfunction. The aim of the present paper is to review present knowledge about the pathophysiology of perinatal hypoxic-ischemic encephalopathy, especially with respect to novel treatment strategies and biomarkers that might enhance early detection of this disorder and thus improve the general outcome of patients.
Collapse
Affiliation(s)
| | | | | | | | - J. OTÁHAL
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
50
|
Abbasi H, Bennet L, Gunn AJ, Unsworth CP. Robust Wavelet Stabilized 'Footprints of Uncertainty' for Fuzzy System Classifiers to Automatically Detect Sharp Waves in the EEG after Hypoxia Ischemia. Int J Neural Syst 2016; 27:1650051. [PMID: 27760476 DOI: 10.1142/s0129065716500519] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Currently, there are no developed methods to detect sharp wave transients that exist in the latent phase after hypoxia-ischemia (HI) in the electroencephalogram (EEG) in order to determine if these micro-scale transients are potential biomarkers of HI. A major issue with sharp waves in the HI-EEG is that they possess a large variability in their sharp wave profile making it difficult to build a compact 'footprint of uncertainty' (FOU) required for ideal performance of a Type-2 fuzzy logic system (FLS) classifier. In this paper, we develop a novel computational EEG analysis method to robustly detect sharp waves using over 30[Formula: see text]h of post occlusion HI-EEG from an equivalent, in utero, preterm fetal sheep model cohort. We demonstrate that initial wavelet transform (WT) of the sharp waves stabilizes the variation in their profile and thus permits a highly compact FOU to be built, hence, optimizing the performance of a Type-2 FLS. We demonstrate that this method leads to higher overall performance of [Formula: see text] for the clinical [Formula: see text] sampled EEG and [Formula: see text] for the high resolution [Formula: see text] sampled EEG that is improved upon over conventional standard wavelet [Formula: see text] and [Formula: see text], respectively, and fuzzy approaches [Formula: see text] and [Formula: see text], respectively, when performed in isolation.
Collapse
Affiliation(s)
- Hamid Abbasi
- 1 Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- 2 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- 2 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- 1 Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|