1
|
Wang Q, Pan L, Chen S, Zhang Y, Liu G, Wu Y, Qin X, Zhang P, Zhang W, Zhang J, Kong D. BBB proteomic analysis reveals that complex febrile seizures in infancy enhance susceptibility to epilepsy in adulthood through dysregulation of ECM-receptor interaction signaling pathway. Fluids Barriers CNS 2025; 22:49. [PMID: 40361173 PMCID: PMC12070522 DOI: 10.1186/s12987-025-00660-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Complex febrile seizures (CFS) have been associated with an increased risk of epilepsy in adulthood. However, the specific link between blood-brain barrier (BBB) and the predisposition to epilepsy in adults who experienced CFS during infancy remains unclear. The objective of this study was to investigate the alteration of BBB in adult mice who had experienced CFS during infancy, and to explore the mechanisms of increased susceptibility to epilepsy after CFS. METHODS The CFS pup model was induced using hot air, and the seizure susceptibility was examined using low-dose pentylenetetrazole (PTZ) after 8 W. The brain microvessels representing BBB function were isolated and their protein expression changes were analyzed using data-independent acquisition (DIA) proteomic techniques. Subsequently, the bioinformatic analyses were performed using ClusterProfiler, STRING, Gene Set Enrichment Analysis (GSEA), etc. The enriched pathways, changes in the expression of BBB-related proteins, and alterations in metabolites including certain neurotransmitters were subsequently validated by Western Blotting, quantitative real-time polymerase chain reaction (qRT-PCR), and mass spectrometric imaging (MSI). In addition, we selected the MMP inhibitor Incyclinide to verify that dysregulation of the ECM-receptor interaction signaling pathway increases epilepsy susceptibility in adult mice. RESULTS Mice that experienced CFS in infancy show increased susceptibility to epilepsy in adulthood, and BBB proteomic profile was significantly altered in the CFS mice. The network analysis suggests that dysregulation of the extracellular matrix (ECM)-receptor interaction pathway is a key mechanism. Moreover, MSI analysis uncovered notable changes in differential metabolites, including amino acids and nucleotide-derived neurotransmitters associated with the function of BBB maintaining neuronal homeostasis. Subsequent validation experiments showed that dysregulation of the ECM-receptor interaction signaling pathway exacerbated epilepsy susceptibility in adult mice. CONCLUSION Our research represents the pioneering demonstration of the modified BBB proteomics associated with epilepsy susceptibility in adult mice previously exposed to CFS in infancy. Notably, the increased susceptibility is attributed to the dysregulation of the ECM-receptor interaction pathway. These findings may help to elucidate the role of BBB alterations in the progression of epilepsy susceptibility, and provide new orientations for subsequent prevention and treatment of epilepsy.
Collapse
Affiliation(s)
- Qian Wang
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Liangyu Pan
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Siruan Chen
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Yuyu Zhang
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Guangyuan Liu
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Yiying Wu
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Xia Qin
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Panpan Zhang
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China
| | - Wei Zhang
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China.
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China.
- Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Shijiazhuang, China.
| | - Jianghua Zhang
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China.
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China.
| | - Dezhi Kong
- Institute of Integrative Medicine, College of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China.
- Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, Shijiazhuang, 050017, China.
- Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Shijiazhuang, China.
| |
Collapse
|
2
|
Griflyuk AV, Postnikova TY, Zaitsev AV. Animal Models of Febrile Seizures: Limitations and Recent Advances in the Field. Cells 2024; 13:1895. [PMID: 39594643 PMCID: PMC11592604 DOI: 10.3390/cells13221895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Febrile seizures (FSs) are defined as seizures occurring in children aged 6 months to 5 years with a background of elevated body temperature. It is one of the most common neurological disorders of childhood, emphasizing the importance of understanding the causes of FSs and their impact on the developing nervous system. However, there are significant limitations to the technologies currently available for studying the etiology and pathophysiology of seizures in humans. It is currently not possible to adequately capture the subtle molecular and structural rearrangements of the nervous system that can occur after seizures in humans. The use of animal models can be invaluable for these purposes. The most commonly used models in modern research are hyperthermic models in rats and mice aged 10-12 days. While these models can reproduce many of the characteristics of FSs, they have certain limitations. This review outlines the key considerations when working with models of FSs, provides an overview of current approaches to producing seizures in different model subjects, and presents a summary of key findings regarding morphological and functional changes in the brain and behavioral alterations that have been identified in studies using animal models of FSs.
Collapse
Affiliation(s)
| | | | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 44, Toreza Prospekt, Saint Petersburg 194223, Russia; (A.V.G.); (T.Y.P.)
| |
Collapse
|
3
|
de Labra C, Pardo-Vazquez JL, Cudeiro J, Rivadulla C. Hyperthermia-Induced Changes in EEG of Anesthetized Mice Subjected to Passive Heat Exposure. Front Syst Neurosci 2021; 15:709337. [PMID: 34566589 PMCID: PMC8458808 DOI: 10.3389/fnsys.2021.709337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/24/2021] [Indexed: 11/21/2022] Open
Abstract
Currently, the role of hypothermia in electroencephalography (EEG) is well-established. However, few studies have investigated the effect of hyperthermia on EEG, an important physiological parameter governing brain function. The aim of this work was to determine how neuronal activity in anesthetized mice is affected when the temperature rises above the physiological threshold mandatory to maintain the normal body functions. In this study, a temperature-elevation protocol, from 37 to 42°C, was applied to four female mice of 2–3 months old while EEG was recorded simultaneously. We found that hyperthermia reduces EEG amplitude by 4.36% when rising from 37 to 38 degrees and by 24.33% when it is increased to 42 degrees. Likewise, increasing the body temperature produces a very large impact on the EEG spectral parameters, reducing the frequency power at the delta, theta, alpha, and beta bands. Our results show that hyperthermia has a global effect on the EEG, being able to change the electrical activity of the brain.
Collapse
Affiliation(s)
- Carmen de Labra
- NEUROcom, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), A Coruña, Spain.,Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Jose L Pardo-Vazquez
- NEUROcom, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), A Coruña, Spain.,Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Javier Cudeiro
- NEUROcom, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), A Coruña, Spain.,Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain.,Centro de Estimulación Cerebral de Galicia, A Coruña, Spain
| | - Casto Rivadulla
- NEUROcom, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), A Coruña, Spain.,Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| |
Collapse
|
4
|
TMEM16C is involved in thermoregulation and protects rodent pups from febrile seizures. Proc Natl Acad Sci U S A 2021; 118:2023342118. [PMID: 33972431 PMCID: PMC8157992 DOI: 10.1073/pnas.2023342118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
As the most common convulsive disorder in infancy and childhood, affecting 2 to 5% of American children in their first 5 y of life, febrile seizures (FSs) are associated with genetic risk factors, including the Tmem16c (Ano3) gene. Whereas central neuronal hyperexcitability has been implicated in FSs, whether FSs may result from compromised body temperature regulation is unknown. To approach this question, we developed rodent models of FSs associated with deficient thermoregulation, including conditional knockout mice with TMEM16C eliminated from a hypothalamic neuronal population important for maintaining body temperature but not from most of the cortical and hippocampal neurons and sensory neurons. Our findings raise the possibility that impaired homeostatic thermoregulation could contribute to the risk of FSs. Febrile seizures (FSs) are the most common convulsion in infancy and childhood. Considering the limitations of current treatments, it is important to examine the mechanistic cause of FSs. Prompted by a genome-wide association study identifying TMEM16C (also known as ANO3) as a risk factor of FSs, we showed previously that loss of TMEM16C function causes hippocampal neuronal hyperexcitability [Feenstra et al., Nat. Genet. 46, 1274–1282 (2014)]. Our previous study further revealed a reduction in the number of warm-sensitive neurons that increase their action potential firing rate with rising temperature of the brain region harboring these hypothalamic neurons. Whereas central neuronal hyperexcitability has been implicated in FSs, it is unclear whether the maximal temperature reached during fever or the rate of body temperature rise affects FSs. Here we report that mutant rodent pups with TMEM16C eliminated from all or a subset of their central neurons serve as FS models with deficient thermoregulation. Tmem16c knockout (KO) rat pups at postnatal day 10 (P10) are more susceptible to hyperthermia-induced seizures. Moreover, they display a more rapid rise of body temperature upon heat exposure. In addition, conditional knockout (cKO) mouse pups (P11) with TMEM16C deletion from the brain display greater susceptibility of hyperthermia-induced seizures as well as deficiency in thermoregulation. We also found similar phenotypes in P11 cKO mouse pups with TMEM16C deletion from Ptgds-expressing cells, including temperature-sensitive neurons in the preoptic area (POA) of the anterior hypothalamus, the brain region that controls body temperature. These findings suggest that homeostatic thermoregulation plays an important role in FSs.
Collapse
|
5
|
Mishima T, Fujiwara T, Kofuji T, Saito A, Terao Y, Akagawa K. Syntaxin 1B regulates synaptic GABA release and extracellular GABA concentration, and is associated with temperature-dependent seizures. J Neurochem 2020; 156:604-613. [PMID: 32858780 DOI: 10.1111/jnc.15159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/13/2020] [Accepted: 08/11/2020] [Indexed: 11/29/2022]
Abstract
De novo heterozygous mutations in the STX1B gene, encoding syntaxin 1B, cause a familial, fever-associated epilepsy syndrome. Syntaxin 1B is an essential component of the pre-synaptic neurotransmitter release machinery as a soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein that regulates the exocytosis of synaptic vesicles. It is also involved in regulating the functions of the SLC6 family of neurotransmitter transporters that reuptake neurotransmitters, including inhibitory neurotransmitters, such as γ-aminobutyric acid (GABA) and glycine. The purpose of the present study was to elucidate the molecular mechanisms underlying the development of febrile seizures by examining the effects of syntaxin 1B haploinsufficiency on inhibitory synaptic transmission during hyperthermia in a mouse model. Stx1b gene heterozygous knockout (Stx1b+/- ) mice showed increased susceptibility to febrile seizures and drug-induced seizures. In cultured hippocampal neurons, we examined the temperature-dependent properties of neurotransmitter release and reuptake by GABA transporter-1 (GAT-1) at GABAergic neurons using whole-cell patch-clamp recordings. The rate of spontaneous quantal GABA release was reduced in Stx1b+/- mice. The hyperthermic temperature increased the tonic GABAA current in wild-type (WT) synapses, but not in Stx1b+/- synapses. In WT neurons, recurrent bursting activities were reduced in a GABA-dependent manner at hyperthermic temperature; however, this was abolished in Stx1b+/- neurons. The blockade of GAT-1 increased the tonic GABAA current and suppressed recurrent bursting activities in Stx1b+/- neurons at the hyperthermic temperature. These data suggest that functional abnormalities associated with GABA release and reuptake in the pre-synaptic terminals of GABAergic neurons may increase the excitability of the neural circuit with hyperthermia.
Collapse
Affiliation(s)
- Tatsuya Mishima
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Tomonori Fujiwara
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Faculty of Health and Medical Care, Saitama Medical University, Hidaka, Saitama, Japan
| | - Takefumi Kofuji
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Radioisotope Laboratory, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Ayako Saito
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Yasuo Terao
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Kimio Akagawa
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| |
Collapse
|
6
|
Semple BD, Dill LK, O'Brien TJ. Immune Challenges and Seizures: How Do Early Life Insults Influence Epileptogenesis? Front Pharmacol 2020; 11:2. [PMID: 32116690 PMCID: PMC7010861 DOI: 10.3389/fphar.2020.00002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/03/2020] [Indexed: 12/16/2022] Open
Abstract
The development of epilepsy, a process known as epileptogenesis, often occurs later in life following a prenatal or early postnatal insult such as cerebral ischemia, stroke, brain trauma, or infection. These insults share common pathophysiological pathways involving innate immune activation including neuroinflammation, which is proposed to play a critical role in epileptogenesis. This review provides a comprehensive overview of the latest preclinical evidence demonstrating that early life immune challenges influence neuronal hyperexcitability and predispose an individual to later life epilepsy. Here, we consider the range of brain insults that may promote the onset of chronic recurrent spontaneous seizures at adulthood, spanning intrauterine insults (e.g. maternal immune activation), perinatal injuries (e.g. hypoxic–ischemic injury, perinatal stroke), and insults sustained during early postnatal life—such as fever-induced febrile seizures, traumatic brain injuries, infections, and environmental stressors. Importantly, all of these insults represent, to some extent, an immune challenge, triggering innate immune activation and implicating both central and systemic inflammation as drivers of epileptogenesis. Increasing evidence suggests that pro-inflammatory cytokines such as interleukin-1 and subsequent signaling pathways are important mediators of seizure onset and recurrence, as well as neuronal network plasticity changes in this context. Our current understanding of how early life immune challenges prime microglia and astrocytes will be explored, as well as how developmental age is a critical determinant of seizure susceptibility. Finally, we will consider the paradoxical phenomenon of preconditioning, whereby these same insults may conversely provide neuroprotection. Together, an improved appreciation of the neuroinflammatory mechanisms underlying the long-term epilepsy risk following early life insults may provide insight into opportunities to develop novel immunological anti-epileptogenic therapeutic strategies.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Kurata H, Saito K, Kawashima F, Ikenari T, Oguri M, Saito Y, Maegaki Y, Mori T. Developing a mouse model of acute encephalopathy using low-dose lipopolysaccharide injection and hyperthermia treatment. Exp Biol Med (Maywood) 2019; 244:743-751. [PMID: 31046452 DOI: 10.1177/1535370219846497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IMPACT STATEMENT Acute encephalopathy (AE), mainly reported in East Asia, is classified into four categories based on clinical and neuropathological findings. Among them, AE caused by cytokine storm is known as the severest clinical entity that causes cerebral edema with poor prognosis. Because suitable and convenient model animal of AE had not been developed, the treatment of patients with AE is not established. In the present study, we established a simple and convenient protocol to mimic AE due to cytokine storm. Our model animal should be useful to elucidate the pathogenesis of AE.
Collapse
Affiliation(s)
- Hirofumi Kurata
- 1 Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan.,2 Division of Child Neurology, Department of Brain and Neurosciences, Tottori University, Yonago 683-8504, Japan.,3 Department of Pediatrics, National Hospital Organization, Kumamoto Saishunso National Hospital, Koshi, 861-1196, Japan
| | - Kengo Saito
- 1 Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Fumiaki Kawashima
- 1 Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Takuya Ikenari
- 1 Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masayoshi Oguri
- 4 Department of Pathobiological Science and Technology, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Yoshiaki Saito
- 2 Division of Child Neurology, Department of Brain and Neurosciences, Tottori University, Yonago 683-8504, Japan
| | - Yoshihiro Maegaki
- 2 Division of Child Neurology, Department of Brain and Neurosciences, Tottori University, Yonago 683-8504, Japan
| | - Tetsuji Mori
- 1 Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
8
|
Umeoka EHL, Robinson EJ, Turimella SL, van Campen JS, Motta-Teixeira LC, Sarabdjitsingh RA, Garcia-Cairasco N, Braun K, de Graan PN, Joëls M. Hyperthermia-induced seizures followed by repetitive stress are associated with age-dependent changes in specific aspects of the mouse stress system. J Neuroendocrinol 2019; 31:e12697. [PMID: 30773738 DOI: 10.1111/jne.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 11/28/2022]
Abstract
Stress is among the most frequently self-reported factors provoking epileptic seizures in children and adults. It is still unclear, however, why some people display stress-sensitive seizures and others do not. Recently, we showed that young epilepsy patients with stress-sensitive seizures exhibit a dysregulated hypothalamic-pituitary-adrenal (HPA)-axis. Most likely, this dysregulation gradually develops, and is triggered by stressors occurring early in life (early-life stress [ELS]). ELS may be particularly impactful when overlapping with the period of epileptogenesis. To examine this in a controlled and prospective manner, the present study investigated the effect of repetitive variable stressors or control treatment between postnatal day (PND) 12 and 24 in male mice exposed on PND10 to hyperthermia (HT)-induced prolonged seizures (control: normothermia). A number of peripheral and central indices of HPA-axis activity were evaluated at pre-adolescent and young adult age (ie, at PND25 and 90, respectively). At PND25 but not at PND90, body weight gain and absolute as well as relative (to body weight) thymus weight were reduced by ELS (vs control), whereas relative adrenal weight was enhanced, confirming the effectiveness of the stress treatment. Basal and stress-induced corticosterone levels were unaffected, though, by ELS at both ages. HT by itself did not affect any of these peripheral markers of HPA-axis activity, nor did it interact with ELS. However, centrally we did observe age-specific interaction effects of HT and ELS with regard to hippocampal glucocorticoid receptor mRNA expression, neurogenesis with the immature neurone marker doublecortin and the number of hilar (ectopic) granule cells using Prox1 staining. This lends some support to the notion that exposure to repetitive stress after HT-induced seizures may dysregulate central components of the stress system in an age-dependent manner. Such dysregulation could be one of the mechanisms conferring higher vulnerability of individuals with epilepsy to develop seizures in the face of stress.
Collapse
Affiliation(s)
- Eduardo H L Umeoka
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Edward J Robinson
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Sada Lakshmi Turimella
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jolien S van Campen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lívia C Motta-Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - R Angela Sarabdjitsingh
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Norberto Garcia-Cairasco
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kees Braun
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pierre N de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
9
|
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
|
10
|
van Campen JS, Hessel EVS, Bohmbach K, Rizzi G, Lucassen PJ, Lakshmi Turimella S, Umeoka EHL, Meerhoff GF, Braun KPJ, de Graan PNE, Joëls M. Stress and Corticosteroids Aggravate Morphological Changes in the Dentate Gyrus after Early-Life Experimental Febrile Seizures in Mice. Front Endocrinol (Lausanne) 2018; 9:3. [PMID: 29434572 PMCID: PMC5790804 DOI: 10.3389/fendo.2018.00003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/05/2018] [Indexed: 12/17/2022] Open
Abstract
Stress is the most frequently self-reported seizure precipitant in patients with epilepsy. Moreover, a relation between ear stress and epilepsy has been suggested. Although ear stress and stress hormones are known to influence seizure threshold in rodents, effects on the development of epilepsy (epileptogenesis) are still unclear. Therefore, we studied the consequences of ear corticosteroid exposure for epileptogenesis, under highly controlled conditions in an animal model. Experimental febrile seizures (eFS) were elicited in 10-day-old mice by warm-air induced hyperthermia, while a control group was exposed to a normothermic condition. In the following 2 weeks, mice received either seven corticosterone or vehicle injections or were left undisturbed. Specific measures indicative for epileptogenesis were examined at 25 days of age and compared with vehicle injected or untreated mice. We examined structural [neurogenesis, dendritic morphology, and mossy fiber sprouting (MFS)] and functional (glutamatergic postsynaptic currents and long-term potentiation) plasticity in the dentate gyrus (DG). We found that differences in DG morphology induced by eFS were aggravated by repetitive (mildly stressful) vehicle injections and corticosterone exposure. In the injected groups, eFS were associated with decreases in neurogenesis, and increases in cell proliferation, dendritic length, and spine density. No group differences were found in MFS. Despite these changes in DG morphology, no effects of eFS were found on functional plasticity. We conclude that corticosterone exposure during early epileptogenesis elicited by eFS aggravates morphological, but not functional, changes in the DG, which partly supports the hypothesis that ear stress stimulates epileptogenesis.
Collapse
Affiliation(s)
- Jolien S. van Campen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Child Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ellen V. S. Hessel
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kirsten Bohmbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Giorgio Rizzi
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Sada Lakshmi Turimella
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Eduardo H. L. Umeoka
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
- Neursocience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Gideon F. Meerhoff
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Kees P. J. Braun
- Department of Child Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pierre N. E. de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
11
|
Löscher W, Ferland RJ, Ferraro TN. The relevance of inter- and intrastrain differences in mice and rats and their implications for models of seizures and epilepsy. Epilepsy Behav 2017; 73. [PMID: 28651171 PMCID: PMC5909069 DOI: 10.1016/j.yebeh.2017.05.040] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It is becoming increasingly clear that the genetic background of mice and rats, even in inbred strains, can have a profound influence on measures of seizure susceptibility and epilepsy. These differences can be capitalized upon through genetic mapping studies to reveal genes important for seizures and epilepsy. However, strain background and particularly mixed genetic backgrounds of transgenic animals need careful consideration in both the selection of strains and in the interpretation of results and conclusions. For instance, mice with targeted deletions of genes involved in epilepsy can have profoundly disparate phenotypes depending on the background strain. In this review, we discuss findings related to how this genetic heterogeneity has and can be utilized in the epilepsy field to reveal novel insights into seizures and epilepsy. Moreover, we discuss how caution is needed in regards to rodent strain or even animal vendor choice, and how this can significantly influence seizure and epilepsy parameters in unexpected ways. This is particularly critical in decisions regarding the strain of choice used in generating mice with targeted deletions of genes. Finally, we discuss the role of environment (at vendor and/or laboratory) and epigenetic factors for inter- and intrastrain differences and how such differences can affect the expression of seizures and the animals' performance in behavioral tests that often accompany acute and chronic seizure testing.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States; Department of Neurology, Albany Medical College, Albany, NY, United States
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
12
|
Isaksen TJ, Kros L, Vedovato N, Holm TH, Vitenzon A, Gadsby DC, Khodakhah K, Lykke-Hartmann K. Hypothermia-induced dystonia and abnormal cerebellar activity in a mouse model with a single disease-mutation in the sodium-potassium pump. PLoS Genet 2017; 13:e1006763. [PMID: 28472154 PMCID: PMC5436892 DOI: 10.1371/journal.pgen.1006763] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 05/18/2017] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
Mutations in the neuron-specific α3 isoform of the Na+/K+-ATPase are found in patients suffering from Rapid onset Dystonia Parkinsonism and Alternating Hemiplegia of Childhood, two closely related movement disorders. We show that mice harboring a heterozygous hot spot disease mutation, D801Y (α3+/D801Y), suffer abrupt hypothermia-induced dystonia identified by electromyographic recordings. Single-neuron in vivo recordings in awake α3+/D801Y mice revealed irregular firing of Purkinje cells and their synaptic targets, the deep cerebellar nuclei neurons, which was further exacerbated during dystonia and evolved into abnormal high-frequency burst-like firing. Biophysically, we show that the D-to-Y mutation abolished pump-mediated Na+/K+ exchange, but allowed the pumps to bind Na+ and become phosphorylated. These findings implicate aberrant cerebellar activity in α3 isoform-related dystonia and add to the functional understanding of the scarce and severe mutations in the α3 isoform Na+/K+-ATPase. The neurological spectrum associated with mutations in the ATP1A3 gene, encoding the α3 isoform of the Na+/K+-ATPase, is complex and still poorly understood. To elucidate the disease-specific pathophysiology, we examined a mouse model harboring the mutation D801Y, which was originally found in a patient with Rapid onset Dystonia Parkinsonism, but recently, also in a patient with Alternating Hemiplegia of Childhood. We found that this model exhibited motor deficits and developed dystonia when exposed to a drop in body temperature. Cerebellar in vivo recordings in awake mice revealed irregular firing of Purkinje cells and their synaptic targets, the deep cerebellar nuclei neurons, which was further exacerbated and evolved into abnormal high-frequency burst firing during dystonia. The development of specific neurological features within the ATP1A3 mutation spectrum, such as dystonia, are thought to reflect the functional consequences of each mutation, thus to investigate the consequence of the D801Y mutations we characterized mutated D-to-Y Na+/K+-ATPases expressed in Xenopus oocytes. These in vitro studies showed that the D-to-Y mutation abolishes pump-mediated Na+/K+ exchange, but still allows the pumps to bind Na+ and become phosphorylated, trapping them in conformations that instead support proton influx.
Collapse
Affiliation(s)
- Toke Jost Isaksen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Lieke Kros
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Natascia Vedovato
- The Laboratory of Cardiac/Membrane Physiology, The Rockefeller University, New York, New York, United States of America
| | - Thomas Hellesøe Holm
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Ariel Vitenzon
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - David C. Gadsby
- The Laboratory of Cardiac/Membrane Physiology, The Rockefeller University, New York, New York, United States of America
| | - Kamran Khodakhah
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
13
|
Hessel EVS, van Lith HA, Wolterink-Donselaar IG, de Wit M, Groot Koerkamp MJA, Holstege FCP, Kas MJH, Fernandes C, de Graan PNE. Mapping of aFEB3homologous febrile seizure locus on mouse chromosome 2 containing candidate genesScn1aandScn3a. Eur J Neurosci 2016; 44:2950-2957. [DOI: 10.1111/ejn.13420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/10/2016] [Accepted: 08/09/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Ellen V. S. Hessel
- Brain Center Rudolf Magnus; Department of Translational Neuroscience; University Medical Center Utrecht; Universiteitsweg 100 3584 CG Utrecht The Netherlands
| | - Hein A. van Lith
- Division of Animal Welfare & Laboratory Animal Science; Department of Animals in Science & Society; Faculty of Veterinary Medicine and Brain Center Rudolf Magnus; Utrecht University; Utrecht The Netherlands
| | - Inge G. Wolterink-Donselaar
- Brain Center Rudolf Magnus; Department of Translational Neuroscience; University Medical Center Utrecht; Universiteitsweg 100 3584 CG Utrecht The Netherlands
| | - Marina de Wit
- Brain Center Rudolf Magnus; Department of Translational Neuroscience; University Medical Center Utrecht; Universiteitsweg 100 3584 CG Utrecht The Netherlands
| | | | - Frank C. P. Holstege
- Department of Molecular Cancer Research; University Medical Center Utrecht; Utrecht The Netherlands
| | - Martien J. H. Kas
- Brain Center Rudolf Magnus; Department of Translational Neuroscience; University Medical Center Utrecht; Universiteitsweg 100 3584 CG Utrecht The Netherlands
- Groningen Institute for Evolutionary Life Sciences; University of Groningen; Groningen The Netherlands
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre; Institute of Psychiatry; Psychology and Neuroscience; King's College London; London UK
| | - Pierre N. E. de Graan
- Brain Center Rudolf Magnus; Department of Translational Neuroscience; University Medical Center Utrecht; Universiteitsweg 100 3584 CG Utrecht The Netherlands
| |
Collapse
|
14
|
Khan D, Dupper A, Deshpande T, Graan PNED, Steinhäuser C, Bedner P. Experimental febrile seizures impair interastrocytic gap junction coupling in juvenile mice. J Neurosci Res 2016; 94:804-13. [PMID: 26931373 DOI: 10.1002/jnr.23726] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/25/2016] [Accepted: 02/04/2016] [Indexed: 02/02/2023]
Abstract
Prolonged and focal febrile seizures (FSs) have been associated with the development of temporal lobe epilepsy (TLE), although the underlying mechanism and the contribution of predisposing risk factors are still poorly understood. Using a kainate model of TLE, we previously provided strong evidence that interruption of astrocyte gap junction-mediated intercellular communication represents a crucial event in epileptogenesis. To elucidate this aspect further, we induced seizures in immature mice by hyperthermia (HT) to study the consequences of FSs on the hippocampal astrocytic network. Changes in interastrocytic coupling were assessed by tracer diffusion studies in acute slices from mice 5 days after experimental FS induction. The results reveal that HT-induced FSs cause a pronounced reduction of astrocyte gap junctional coupling in the hippocampus by more than 50%. Western blot analysis indicated that reduced connexin43 protein expression and/or changes in the phosphorylation status account for this astrocyte dysfunction. Remarkably, uncoupling occurred in the absence of neuronal death and reactive gliosis. These data provide a mechanistic link between FSs and the subsequent development of TLE and further strengthen the emerging view that astrocytes have a central role in the pathogenesis of this disorder. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dilaware Khan
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alexander Dupper
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Tushar Deshpande
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Pierre N E De Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
15
|
Feng B, Chen Z. Generation of Febrile Seizures and Subsequent Epileptogenesis. Neurosci Bull 2016; 32:481-92. [PMID: 27562688 DOI: 10.1007/s12264-016-0054-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/13/2016] [Indexed: 11/24/2022] Open
Abstract
Febrile seizures (FSs) occur commonly in children aged from 6 months to 5 years. Complex (repetitive or prolonged) FSs, but not simple FSs, can lead to permanent brain modification. Human infants and immature rodents that have experienced complex FSs have a high risk of subsequent temporal lobe epilepsy. However, the causes of FSs and the mechanisms underlying the subsequent epileptogenesis remain unknown. Here, we mainly focus on two major questions concerning FSs: how fever triggers seizures, and how epileptogenesis occurs after FSs. The risk factors responsible for the occurrence of FSs and the epileptogenesis after prolonged FSs are thoroughly summarized and discussed. An understanding of these factors can provide potential therapeutic targets for the prevention of FSs and also yield biomarkers for identifying patients at risk of epileptogenesis following FSs.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Jongbloets BC, van Gassen KLI, Kan AA, Olde Engberink AHO, de Wit M, Wolterink-Donselaar IG, Groot Koerkamp MJA, van Nieuwenhuizen O, Holstege FCP, de Graan PNE. Expression Profiling after Prolonged Experimental Febrile Seizures in Mice Suggests Structural Remodeling in the Hippocampus. PLoS One 2015; 10:e0145247. [PMID: 26684451 PMCID: PMC4684321 DOI: 10.1371/journal.pone.0145247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022] Open
Abstract
Febrile seizures are the most prevalent type of seizures among children up to 5 years of age (2–4% of Western-European children). Complex febrile seizures are associated with an increased risk to develop temporal lobe epilepsy. To investigate short- and long-term effects of experimental febrile seizures (eFS), we induced eFS in highly febrile convulsion-susceptible C57BL/6J mice at post-natal day 10 by exposure to hyperthermia (HT) and compared them to normotherm-exposed (NT) mice. We detected structural re-organization in the hippocampus 14 days after eFS. To identify molecular candidates, which entrain this structural re-organization, we investigated temporal changes in mRNA expression profiles eFS 1 hour to 56 days after eFS. We identified 931 regulated genes and profiled several candidates using in situ hybridization and histology at 3 and 14 days after eFS. This is the first study to report genome-wide transcriptome analysis after eFS in mice. We identify temporal regulation of multiple processes, such as stress-, immune- and inflammatory responses, glia activation, glutamate-glutamine cycle and myelination. Identification of the short- and long-term changes after eFS is important to elucidate the mechanisms contributing to epileptogenesis.
Collapse
Affiliation(s)
- Bart C Jongbloets
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Koen L I van Gassen
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anne A Kan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anneke H O Olde Engberink
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marina de Wit
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Inge G Wolterink-Donselaar
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marian J A Groot Koerkamp
- Molecular Cancer Research, Division Biomedical Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Onno van Nieuwenhuizen
- Brain Center Rudolf Magnus, Department of Pediatric Neurology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Frank C P Holstege
- Molecular Cancer Research, Division Biomedical Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Pierre N E de Graan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
17
|
Hamelin S, Pouyatos B, Khalaf-Nazzal R, Chabrol T, Francis F, David O, Depaulis A. Long-term modifications of epileptogenesis and hippocampal rhythms after prolonged hyperthermic seizures in the mouse. Neurobiol Dis 2014; 69:156-68. [DOI: 10.1016/j.nbd.2014.05.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/08/2014] [Accepted: 05/17/2014] [Indexed: 01/15/2023] Open
|
18
|
Wang Z, Fan Y, Xu J, Li L, Heng D, Han S, Yin J, Peng B, Liu W, He X. Transcriptome analysis of the hippocampus in novel rat model of febrile seizures. PLoS One 2014; 9:e95237. [PMID: 24736375 PMCID: PMC3988142 DOI: 10.1371/journal.pone.0095237] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/24/2014] [Indexed: 01/12/2023] Open
Abstract
Febrile seizures (FS) are the most common type of convulsive events in infants and young children, but the precise underlying genetic mechanism remains to be explored. To investigate the underlying pathogenic factors in FS and subsequent epilepsy, alterations in gene expression between the two new strains of rats (hyperthermia-prone [HP] vs hyperthermia-resistant [HR]), were investigated by using the Whole Rat Genome Oligo Microarray. This process identified 1,140 differentially expressed genes (DEGs; 602 upregulated and 538 downregulated), which were analyzed to determine significant Gene Ontology (GO) categories, signaling pathways and gene networks. Based on the GO analyses, the modified genes are closely related to various FS pathogenesis factors, including immune and inflammatory responses and ion transport. Certain DEGs identified have not been previously examined in relation to FS pathogenesis. Among these genes is dipeptidyl peptidase 4 (DPP4), a gene closely linked to interleukin 6 (IL-6), which played a key role in the gene network analysis. Furthermore, sitagliptin, a DPP4 inhibitor significantly decreased epileptic discharge in rats, observed via electroencephalogram, suggesting an important role for DPP4 in FS. The effectiveness of sitagliptin in reducing seizure activity may occur through a mechanism that stabilizes cellular Ca2+ homeostasis. In addition, DPP4 expression may be regulated by DNA methylation. The hippocampal gene expression profiles in novel rat models of FS provides a large database of candidate genes and pathways, which will be useful for researchers interested in disorders of neuronal excitability.
Collapse
Affiliation(s)
- Zhongcheng Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yuanteng Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jian Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Liang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Duanhe Heng
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Biwen Peng
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wanhong Liu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- * E-mail: (WL); (XH)
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- * E-mail: (WL); (XH)
| |
Collapse
|
19
|
Patterson KP, Baram TZ, Shinnar S. Origins of temporal lobe epilepsy: febrile seizures and febrile status epilepticus. Neurotherapeutics 2014; 11:242-50. [PMID: 24604424 PMCID: PMC3996115 DOI: 10.1007/s13311-014-0263-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Temporal lobe epilepsy (TLE) and hippocampal sclerosis (HS) commonly arise following early-life long seizures, and especially febrile status epilepticus (FSE). However, there are major gaps in our knowledge regarding the causal relationships of FSE, TLE, HS and cognitive disturbances that hamper diagnosis, biomarker development and prevention. The critical questions include: What is the true probability of developing TLE after FSE? Are there predictive markers for those at risk? A fundamental question is whether FSE is simply a marker of individuals who are destined to develop TLE, or if FSE contributes to the risk of developing TLE. If FSE does contribute to epileptogenesis, then does this happen only in the setting of a predisposed brain? These questions are addressed within this review, using information gleaned over the past two decades from clinical studies as well as animal models.
Collapse
Affiliation(s)
- Katelin P. Patterson
- />Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA USA
| | - Tallie Z. Baram
- />Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA USA
- />Department of Pediatrics, University of California-Irvine, Irvine, CA USA
- />Department of Neurology, University of California-Irvine Medical Center, Irvine, CA USA
| | - Shlomo Shinnar
- />Department of Neurology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY USA
- />Department of Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY USA
- />Department of Epidemiology and Population Health, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY USA
- />Comprehensive Epilepsy Management Center, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY USA
| |
Collapse
|
20
|
Hessel EVS, de Wit M, Wolterink-Donselaar IG, Karst H, de Graaff E, van Lith HA, de Bruijn E, de Sonnaville S, Verbeek NE, Lindhout D, de Kovel CGF, Koeleman BPC, van Kempen M, Brilstra E, Cuppen E, Loos M, Spijker SS, Kan AA, Baars SE, van Rijen PC, Gosselaar PH, Groot Koerkamp MJA, Holstege FCP, van Duijn C, Vergeer J, Moll HA, Taubøll E, Heuser K, Ramakers GMJ, Pasterkamp RJ, van Nieuwenhuizen O, Hoogenraad CC, Kas MJH, de Graan PNE. Identification of Srp9 as a febrile seizure susceptibility gene. Ann Clin Transl Neurol 2014; 1:239-50. [PMID: 25590037 PMCID: PMC4292741 DOI: 10.1002/acn3.48] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/07/2014] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Febrile seizures (FS) are the most common seizure type in young children. Complex FS are a risk factor for mesial temporal lobe epilepsy (mTLE). To identify new FS susceptibility genes we used a forward genetic strategy in mice and subsequently analyzed candidate genes in humans. METHODS We mapped a quantitative trait locus (QTL1) for hyperthermia-induced FS on mouse chromosome 1, containing the signal recognition particle 9 (Srp9) gene. Effects of differential Srp9 expression were assessed in vivo and in vitro. Hippocampal SRP9 expression and genetic association were analyzed in FS and mTLE patients. RESULTS Srp9 was differentially expressed between parental strains C57BL/6J and A/J. Chromosome substitution strain 1 (CSS1) mice exhibited lower FS susceptibility and Srp9 expression than C57BL/6J mice. In vivo knockdown of brain Srp9 reduced FS susceptibility. Mice with reduced Srp9 expression and FS susceptibility, exhibited reduced hippocampal AMPA and NMDA currents. Downregulation of neuronal Srp9 reduced surface expression of AMPA receptor subunit GluA1. mTLE patients with antecedent FS had higher SRP9 expression than patients without. SRP9 promoter SNP rs12403575(G/A) was genetically associated with FS and mTLE. INTERPRETATION Our findings identify SRP9 as a novel FS susceptibility gene and indicate that SRP9 conveys its effects through endoplasmic reticulum (ER)-dependent synthesis and trafficking of membrane proteins, such as glutamate receptors. Discovery of this new FS gene and mechanism may provide new leads for early diagnosis and treatment of children with complex FS at risk for mTLE.
Collapse
Affiliation(s)
- Ellen V S Hessel
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Inge G Wolterink-Donselaar
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Esther de Graaff
- Cell Biology, Faculty of Science, Utrecht UniversityUtrecht, The Netherlands
| | - Hein A van Lith
- Program Emotion and Cognition, Division of Animal Welfare and Laboratory Animal Science, Department of Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University and Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Ewart de Bruijn
- Hubrecht Institute-KNAW and University Medical Center UtrechtUtrecht, The Netherlands
| | - Sophietje de Sonnaville
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Nienke E Verbeek
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Dick Lindhout
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
- SEIN Epilepsy Institute in the NetherlandsHeemstede, The Netherlands
| | - Carolien G F de Kovel
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Bobby P C Koeleman
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Marjan van Kempen
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Eva Brilstra
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Edwin Cuppen
- Hubrecht Institute-KNAW and University Medical Center UtrechtUtrecht, The Netherlands
- Department of Medical Genetics, University Medical Center UtrechtUtrecht, The Netherlands
| | - Maarten Loos
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU UniversityAmsterdam, The Netherlands
| | - Sabine S Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU UniversityAmsterdam, The Netherlands
| | - Anne A Kan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Susanne E Baars
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
- Master program Neuroscience and Cognition, Utrecht UniversityUtrecht, The Netherlands
| | - Peter C van Rijen
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Peter H Gosselaar
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | | | - Frank C P Holstege
- Department of Molecular Cancer Research, University Medical Center UtrechtUtrecht, The Netherlands
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical CenterRotterdam, The Netherlands
| | - Jeanette Vergeer
- Department of Epidemiology, Erasmus University Medical CenterRotterdam, The Netherlands
| | - Henriette A Moll
- Department of Pediatrics, Erasmus Medical CenterRotterdam, The Netherlands
| | - Erik Taubøll
- Department of Neurology, Oslo University HospitalOslo, Norway
| | - Kjell Heuser
- Department of Neurology, Oslo University HospitalOslo, Norway
| | - Geert M J Ramakers
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Onno van Nieuwenhuizen
- Department of Child Neurology, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht UniversityUtrecht, The Netherlands
| | - Martien J H Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| | - Pierre N E de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, The Netherlands
| |
Collapse
|
21
|
Xu J, Fan Y, Li L, Qiu Y, Wang Z, Han S, Yin J, Liu W, Peng B, He X. Hyperthermia-induced seizures: Development of hyperthermia-prone and hyperthermia-resistant rats. Epilepsy Res 2013; 106:311-7. [DOI: 10.1016/j.eplepsyres.2013.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 07/03/2013] [Accepted: 07/06/2013] [Indexed: 11/26/2022]
|
22
|
Neuronal carbonic anhydrase VII provides GABAergic excitatory drive to exacerbate febrile seizures. EMBO J 2013; 32:2275-86. [PMID: 23881097 PMCID: PMC3746197 DOI: 10.1038/emboj.2013.160] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 06/20/2013] [Indexed: 11/08/2022] Open
Abstract
Brain carbonic anhydrases (CAs) are known to modulate neuronal signalling. Using a novel CA VII (Car7) knockout (KO) mouse as well as a CA II (Car2) KO and a CA II/VII double KO, we show that mature hippocampal pyramidal neurons are endowed with two cytosolic isoforms. CA VII is predominantly expressed by neurons starting around postnatal day 10 (P10). The ubiquitous isoform II is expressed in neurons at P20. Both isoforms enhance bicarbonate-driven GABAergic excitation during intense GABAA-receptor activation. P13-14 CA VII KO mice show behavioural manifestations atypical of experimental febrile seizures (eFS) and a complete absence of electrographic seizures. A low dose of diazepam promotes eFS in P13-P14 rat pups, whereas seizures are blocked at higher concentrations that suppress breathing. Thus, the respiratory alkalosis-dependent eFS are exacerbated by GABAergic excitation. We found that CA VII mRNA is expressed in the human cerebral cortex before the age when febrile seizures (FS) occur in children. Our data indicate that CA VII is a key molecule in age-dependent neuronal pH regulation with consequent effects on generation of FS.
Collapse
|
23
|
Hessel EVS, van Lith HA, Wolterink-Donselaar IG, de Wit M, Hendrickx DAE, Kas MJH, de Graan PNE. Mapping an X-linked locus that influences heat-induced febrile seizures in mice. Epilepsia 2012; 53:1399-410. [PMID: 22780306 DOI: 10.1111/j.1528-1167.2012.03575.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE Febrile seizures (FS) are the most common seizure type in children between the age of 6 months and 5 years. Although FS are largely benign, recurrent FS are a major risk factor for developing temporal lobe epilepsy (TLE) later in life. The mechanisms underlying FS are largely unknown; however, family and twin studies indicate that FS susceptibility is under complex genetic control. We have recently developed a phenotypic screen to study the genetics of FS susceptibility in mice. Using this screen in a phenotype-driven genetic strategy we analyzed the C57BL/6J-Chr #(A)/NaJ chromosome substitution strain (CSS) panel. In each CSS line one chromosome of the A/J strain is substituted in a genetically homogeneous C57BL/6J background. The analysis of the CSS panel revealed that A/J chromosomes 1, 2, 6, 10, 13, and X carry at least one quantitative trait locus (QTL) for heat-induced FS susceptibility. The fact that many X-linked genes are highly expressed in the brain and have been implicated in human developmental disorders often presenting with seizures (like fragile X mental retardation) prompted us to map the chromosome X QTL. METHODS C57BL/6J mice were mated with C57BL/6J-Chr X(A) /NaJ (CSSX) to generate F(2)-generations-CXBL6 and BL6CX-originating from CSSX or C57BL/6J mothers, respectively. Heat-induced FS were elicited on postnatal day 14 by exposure to a controlled warm airstream of 50°C. The latency to heat-induced FS is our phenotype. This phenotype has previously been validated by video-electroencephalography (EEG) monitoring. After phenotyping and genotyping the F(2)-population, QTL analysis was performed using R/QTL software. KEY FINDINGS QTL analysis revealed a significant peak with an LOD-score of 3.25. The 1-LOD confidence interval (149,886,866-158,836,462 bp) comprises 52 protein coding genes, of which 34 are known to be brain expressed. Two of these brain-expressed genes have previously been linked to X-linked epilepsies, namely Cdkl5 and Pdha1. SIGNIFICANCE Our results show that the mouse genetics of X-linked FS susceptibility is complex, and that our heat-induced FS-driven genetic approach is a powerful tool for use in unraveling the complexities of this trait in mice. Fine-mapping and functional studies will be required to further identify the X-linked FS susceptibility genes.
Collapse
Affiliation(s)
- Ellen V S Hessel
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
24
|
McClelland S, Flynn C, Dubé C, Richichi C, Zha Q, Ghestem A, Esclapez M, Bernard C, Baram TZ. Neuron-restrictive silencer factor-mediated hyperpolarization-activated cyclic nucleotide gated channelopathy in experimental temporal lobe epilepsy. Ann Neurol 2011; 70:454-64. [PMID: 21905079 DOI: 10.1002/ana.22479] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Enduring, abnormal expression and function of the ion channel hyperpolarization-activated cyclic adenosine monophosphate gated channel type 1 (HCN1) occurs in temporal lobe epilepsy (TLE). We examined the underlying mechanisms, and investigated whether interfering with these mechanisms could modify disease course. METHODS Experimental TLE was provoked by kainic acid-induced status epilepticus (SE). HCN1 channel repression was examined at mRNA, protein, and functional levels. Chromatin immunoprecipitation was employed to identify the transcriptional mechanism of repressed HCN1 expression, and the basis for their endurance. Physical interaction of the repressor, NRSF, was abolished using decoy oligodeoxynucleotides (ODNs). Video/electroencephalographic recordings were performed to assess the onset and initial pattern of spontaneous seizures. RESULTS Levels of NRSF and its physical binding to the Hcn1 gene were augmented after SE, resulting in repression of HCN1 expression and HCN1-mediated currents (I(h) ), and reduced I(h) -dependent resonance in hippocampal CA1 pyramidal cell dendrites. Chromatin changes typical of enduring, epigenetic gene repression were apparent at the Hcn1 gene within a week after SE. Administration of decoy ODNs comprising the NRSF DNA-binding sequence (neuron restrictive silencer element [NRSE]), in vitro and in vivo, reduced NRSF binding to Hcn1, prevented its repression, and restored I(h) function. In vivo, decoy NRSE ODN treatment restored theta rhythm and altered the initial pattern of spontaneous seizures. INTERPRETATION Acquired HCN1 channelopathy derives from NRSF-mediated transcriptional repression that endures via chromatin modification and may provide insight into the mechanisms of a number of channelopathies that coexist with, and may contribute to, the conversion of a normal brain into an epileptic one.
Collapse
Affiliation(s)
- Shawn McClelland
- Departments of Anatomy/Neurobiology, Pediatrics, and Neurology, University of California-Irvine, Irvine, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ayadi A, Ferrand G, Cruz IGD, Warot X. Mouse Breeding and Colony Management. CURRENT PROTOCOLS IN MOUSE BIOLOGY 2011; 1:239-64. [PMID: 26068995 DOI: 10.1002/9780470942390.mo100214] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The possibility to genetically modify the mouse genome has enabled the creation of numerous lines of genetically engineered mouse models (GEMMs). As a result, the demand for housing space in research facilities is increasing. Knowledge of the basis of mouse reproduction and of the methods to handle colonies of GEMMs is therefore mandatory to efficiently populate facilities. The mouse has a short generation period, produces large progenies, and can breed all year round. However, environmental parameters (bedding, diet, cage type, temperature, hygrometry, light, noise, and sanitary status) strongly influence the breeding efficiency and experimental data, and must be tightly controlled. Efficient GEMM colony management requires adequate recording of breeding and proper identification and genotyping of animals. Various mating types and breeding schemes can be used, depending on the type of studies conducted. The recent development of assisted reproduction methods helps circumvent some of the issues faced with those lines especially difficult to breed. Curr. Protoc. Mouse Biol. 1:239-264. © 2011 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Gisèle Ferrand
- Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Xavier Warot
- Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
26
|
Ferraro TN, Smith GG, Ballard D, Zhao H, Schwebel CL, Gupta A, Rappaport EF, Ruiz SE, Lohoff FW, Doyle GA, Berrettini WH, Buono RJ. Quantitative trait loci for electrical seizure threshold mapped in C57BLKS/J and C57BL/10SnJ mice. GENES BRAIN AND BEHAVIOR 2010; 10:309-15. [PMID: 21129161 DOI: 10.1111/j.1601-183x.2010.00668.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We mapped the quantitative trait loci (QTL) that contribute to the robust difference in maximal electroshock seizure threshold (MEST) between C57BLKS/J (BKS) and C57BL10S/J (B10S) mice. BKS, B10S, BKS × B10S F1 and BKS × B10S F2 intercross mice were tested for MEST at 8-9 weeks of age. Results of F2 testing showed that, in this cross, MEST is a continuously distributed trait determined by polygenic inheritance. Mice from the extremes of the trait distribution were genotyped using microarray technology. MEST correlated significantly with body weight and sex; however, because of the high correlation between these factors, the QTL mapping was conditioned on sex alone. A sequential series of statistical analyses was used to map QTLs including single-point, multipoint and multilocus methods. Two QTLs reached genome-wide levels of significance based upon an empirically determined permutation threshold: chromosome 6 (LOD = 6.0 at ∼69 cM) and chromosome 8 (LOD = 5.7 at ∼27 cM). Two additional QTLs were retained in a multilocus regression model: chromosome 3 (LOD = 2.1 at ∼68 cM) and chromosome 5 (LOD = 2.7 at ∼73 cM). Together the four QTLs explain one third of the total phenotypic variance in the mapping population. Lack of overlap between the major MEST QTLs mapped here in BKS and B10S mice and those mapped previously in C57BL/6J and DBA/2J mice (strains that are closely related to BKS and B10S) suggest that BKS and B10S represent a new polygenic mouse model for investigating susceptibility to seizures.
Collapse
Affiliation(s)
- T N Ferraro
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Koyama R, Matsuki N. Novel etiological and therapeutic strategies for neurodiseases: mechanisms and consequences of febrile seizures: lessons from animal models. J Pharmacol Sci 2010; 113:14-22. [PMID: 20424387 DOI: 10.1254/jphs.09r19fm] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Febrile seizures (FS) are the most common type of convulsive events in infancy and childhood. Genetic and environmental elements have been suggested to contribute to FS. FS can be divided into simple and complex types, the former being benign, whereas it is controversial whether complex FS have an association with the development of temporal lobe epilepsy (TLE) in later life. In the hippocampus of TLE patients, several structural and functional alterations take place that render the region an epileptic foci. Thus, it is important to clarify the cellular and molecular changes in the hippocampus after FS and to determine whether they are epileptogenic. To achieve this goal, human studies are too limited because the sample tissues are only available from adult patients in the advanced and drug-resistant stages of the disease, masking the underlying etiology. These facts have inspired researchers to take advantage of well-established animal models of FS to answer the following questions: 1) How does hyperthermia induce FS? 2) Do FS induce neuroanatomical changes? 3) Do FS induce neurophysiological changes? 4) Do FS affect the behavior in later life? Here we introduce and discuss accumulating reports to answer these questions.
Collapse
Affiliation(s)
- Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| | | |
Collapse
|
28
|
de Jong S, Fuller TF, Janson E, Strengman E, Horvath S, Kas MJH, Ophoff RA. Gene expression profiling in C57BL/6J and A/J mouse inbred strains reveals gene networks specific for brain regions independent of genetic background. BMC Genomics 2010; 11:20. [PMID: 20064228 PMCID: PMC2823687 DOI: 10.1186/1471-2164-11-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 01/11/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We performed gene expression profiling of the amygdala and hippocampus taken from inbred mouse strains C57BL/6J and A/J. The selected brain areas are implicated in neurobehavioral traits while these mouse strains are known to differ widely in behavior. Consequently, we hypothesized that comparing gene expression profiles for specific brain regions in these strains might provide insight into the molecular mechanisms of human neuropsychiatric traits. We performed a whole-genome gene expression experiment and applied a systems biology approach using weighted gene co-expression network analysis. RESULTS We were able to identify modules of co-expressed genes that distinguish a strain or brain region. Analysis of the networks that are most informative for hippocampus and amygdala revealed enrichment in neurologically, genetically and psychologically related pathways. Close examination of the strain-specific gene expression profiles, however, revealed no functional relevance but a significant enrichment of single nucleotide polymorphisms in the probe sequences used for array hybridization. This artifact was not observed for the modules of co-expressed genes that distinguish amygdala and hippocampus. CONCLUSIONS The brain-region specific modules were found to be independent of genetic background and are therefore likely to represent biologically relevant molecular networks that can be studied to complement our knowledge about pathways in neuropsychiatric disease.
Collapse
Affiliation(s)
- Simone de Jong
- Department of Medical Genetics and Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
29
|
Castelhano-Carlos MJ, Sousa N, Ohl F, Baumans V. Identification methods in newborn C57BL/6 mice: a developmental and behavioural evaluation. Lab Anim 2009; 44:88-103. [PMID: 19854756 DOI: 10.1258/la.2009.009044] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The use of group-housed rodents in many fields of biomedical research imposes a need to identify individuals in a cage. Few studies have been designed to assess possible negative effects of identification methods of newborn mice on their development and wellbeing. In the present study, three different identification methods were applied to newborn C57BL/6J mice on postnatal day (pnd) 5 (toe clipping, toe tattoo ink puncture and subcutaneous implantation of a small transponder). All identification methods used proved to be effective for long-term marking of individual animals. Newborn mice showed the least reaction to toe clipping followed by toe tattoo ink puncture and transponder implantation was the most distressful individual identification procedure in newborn mice. Importantly, clipped toe tissue proved to be enough for genotyping purposes. No overall consistent differences in somatic and neurological reflex development during the postnatal period were shown as a result of the newborn individual identification procedures used. Further, none of the methods interfered significantly with the adult animals' general normal behaviour (e.g. ability to move, grasp, climb) and sensory-motor functions as assessed with a simplified SHIRPA battery of tests, as well as Rotarod and Elevated Plus Maze tests. Postmortem thymus and adrenal gland weights gave no indication of chronic stress as a consequence of the identification method. We conclude that toe clipping might even be advisable in newborn mice at a very young age, when genotyping is needed. Toe tattoo ink puncture is also a good identification method for newborn mice and transponder implantation should only be used in older newborns or applied at weaning.
Collapse
|
30
|
Comi AM, Trescher WH, Abi-Raad R, Johnston MV, Wilson MA. Impact of age and strain on ischemic brain injury and seizures after carotid ligation in immature mice. Int J Dev Neurosci 2009; 27:271-7. [PMID: 19154784 PMCID: PMC2652514 DOI: 10.1016/j.ijdevneu.2008.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 12/02/2008] [Accepted: 12/16/2008] [Indexed: 10/21/2022] Open
Abstract
Stroke is an important cause of neurologic injury in the neonatal period and frequently results in lifelong neurologic impairments. We reported previously that unilateral carotid ligation on postnatal day (P)12 in CD1 mice causes acute behavioral seizures and unilateral brain injury and provides a model for neonatal stroke in human infants. In the present study we confirmed that behavioral seizures observed after ligation on P12 in the CD1 strain are associated with rhythmic ictal discharges that show temporal progression on electrocorticograms. We also examined the effects of carotid ligation performed at different ages in CD1 mice or performed in the C57Bl/6 strain. The right common carotid was ligated at P7, P10, P12 or P21 in CD1 mice or at P12 in C57Bl/6 mice. Littermate controls received sham surgery. Seizures were rated for 4h after surgery; brain injury was scored one week later. In a separate group of P12 CD1 mice, electrocorticographic activity was recorded continuously for 4h after carotid ligation or sham surgery. Brain injury and cumulative seizure score varied significantly with age (p<0.001) and strain (p<0.001). In CD1 mice, injury was greatest after ligation on P10 to P12 and seizure score was maximal at P12. Seizure scores were significantly correlated with injury after ligation on P10 or P12. C57Bl/6 mice, like C3Heb/FeJ mice examined previously, were much less vulnerable to seizures and injury than CD1 mice after ligation on P12. This study demonstrates that carotid ligation in the CD1 mouse on P12 causes acute electrographic rhythmic discharges that correlate with behavioral seizures. We also found that the age at which ligation is performed and genetic strain have a strong influence on the severity of injury.
Collapse
Affiliation(s)
- Anne M. Comi
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - William H. Trescher
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Division of Pediatric Neurology, M.S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Ronnie Abi-Raad
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
| | - Michael V. Johnston
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mary Ann Wilson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
31
|
Schuchmann S, Vanhatalo S, Kaila K. Neurobiological and physiological mechanisms of fever-related epileptiform syndromes. Brain Dev 2009; 31:378-82. [PMID: 19201562 DOI: 10.1016/j.braindev.2008.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 10/30/2008] [Accepted: 11/04/2008] [Indexed: 10/21/2022]
Abstract
Febrile seizures (FS) are the most common type of convulsive events in children. FS have been extensively studied using animal models, where rat and mice pups are placed in a hyperthermic environment. Such work has largely focused on the consequences rather than on the mechanisms of experimental febrile seizures (eFS). We have recently shown that eFS are preceded by a dramatic rise in the rate of respiration. The consequent respiratory alkalosis affecting the brain and increasing neuronal excitability is a direct cause of the eFS [1]. If a similar mechanism contributes to human FS and other fever-related epileptiform syndromes, a number of factors operating at the molecular, cellular and systems level that have not been previously thought to be involved in their etiology must be considered. These include physiological and pathophysiological factors affecting CO(2) chemosensitivity as well as cellular and systemic mechanisms of acid-base regulation. Furthermore, a critical role for brain pH in FS points to novel types of susceptibility genes, which include genes coding pH-sensitive target proteins (e.g. neuronal ion channels) and pH-regulatory proteins. We will discuss these novel ideas and putative therapies based on them.
Collapse
Affiliation(s)
- Sebastian Schuchmann
- Department of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
32
|
Hessel EVS, Van Gassen KLI, Wolterink-Donselaar IG, Stienen PJ, Fernandes C, Brakkee JH, Kas MJH, De Graan PNE. Phenotyping mouse chromosome substitution strains reveal multiple QTLs for febrile seizure susceptibility. GENES BRAIN AND BEHAVIOR 2009; 8:248-55. [DOI: 10.1111/j.1601-183x.2008.00466.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
van Gassen KLI, van der Hel WS, Hakvoort TBM, Lamers WH, de Graan PNE. Haploinsufficiency of glutamine synthetase increases susceptibility to experimental febrile seizures. GENES BRAIN AND BEHAVIOR 2008; 8:290-5. [PMID: 19170755 DOI: 10.1111/j.1601-183x.2008.00471.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glutamine synthetase (GS) is a pivotal glial enzyme in the glutamate-glutamine cycle. GS is important in maintaining low extracellular glutamate concentrations and is downregulated in the hippocampus of temporal lobe epilepsy patients with mesial-temporal sclerosis, an epilepsy syndrome that is frequently associated with early life febrile seizures (FS). Human congenital loss of GS activity has been shown to result in brain malformations, seizures and death within days after birth. Recently, we showed that GS knockout mice die during embryonic development and that haploinsufficient GS mice have no obvious abnormalities or behavioral seizures. In the present study, we investigated whether reduced expression/activity of GS in haploinsufficient GS mice increased the susceptibility to experimentally induced FS. FS were elicited by warm-air-induced hyperthermia in 14-day-old mice and resulted in seizures in most animals. FS susceptibility was measured as latencies to four behavioral FS characteristics. Our phenotypic data show that haploinsufficient mice are more susceptible to experimentally induced FS (P < 0.005) than littermate controls. Haploinsufficient animals did not differ from controls in hippocampal amino acid content, structure (Nissl and calbindin), glial properties (glial fibrillary acidic protein and vimentin) or expression of other components of the glutamate-glutamine cycle (excitatory amino acid transporter-2 and vesicular glutamate transporter-1). Thus, we identified GS as a FS susceptibility gene. GS activity-disrupting mutations have been described in the human population, but heterozygote mutations were not clearly associated with seizures or epilepsy. Our results indicate that individuals with reduced GS activity may have reduced FS seizure thresholds. Genetic association studies will be required to test this hypothesis.
Collapse
Affiliation(s)
- K L I van Gassen
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|