1
|
Wu C, Wang Q, Li W, Han M, Zhao H, Xu Z. Research progress on pathogenesis and treatment of febrile seizures. Life Sci 2025; 362:123360. [PMID: 39746603 DOI: 10.1016/j.lfs.2024.123360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/26/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Febrile seizures (FSs) are the most common pediatric neurological disorder, affecting approximately 5 % of children aged 6 months to 5 years. While most FSs are self-limiting and benign, about 20-30 % present as complex FSs (CFSs), which pose a risk of acute brain injury and the development of temporal lobe epilepsy. Various factors, including age, geographical distribution, and type of infection influence the occurrence of FS. Infection is the primary external trigger for FS, while the underlying intrinsic factors are linked to the immature and incomplete myelination of the brain during specific developmental stages. Although the precise pathogenesis of FS is not yet fully understood, it is likely caused by the interaction of immature brain development, fever, neuroinflammation, and genetic susceptibility. This review discussed the pathogenesis of febrile seizures, focusing on factors such as age, fever, neuroinflammation, genetics, and intestinal microbiota, and summarized existing therapeutic approaches. Our review may facilitate the identification of new targets for mechanistic studies and clinical treatment of febrile seizures.
Collapse
Affiliation(s)
- Chang Wu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Qingmei Wang
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Wenmi Li
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Mingxuan Han
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Huawei Zhao
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Zhenghao Xu
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
2
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
3
|
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
|
4
|
Egido-Betancourt HX, Strowd III RE, Raab-Graham KF. Potential roles of voltage-gated ion channel disruption in Tuberous Sclerosis Complex. Front Mol Neurosci 2024; 17:1404884. [PMID: 39253727 PMCID: PMC11381416 DOI: 10.3389/fnmol.2024.1404884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 09/11/2024] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a lynchpin disorder, as it results in overactive mammalian target of rapamycin (mTOR) signaling, which has been implicated in a multitude of disease states. TSC is an autosomal dominant disease where 90% of affected individuals develop epilepsy. Epilepsy results from aberrant neuronal excitability that leads to recurring seizures. Under neurotypical conditions, the coordinated activity of voltage-gated ion channels keep neurons operating in an optimal range, thus providing network stability. Interestingly, loss or gain of function mutations in voltage-gated potassium, sodium, or calcium channels leads to altered excitability and seizures. To date, little is known about voltage-gated ion channel expression and function in TSC. However, data is beginning to emerge on how mTOR signaling regulates voltage-gated ion channel expression in neurons. Herein, we provide a comprehensive review of the literature describing common seizure types in patients with TSC, and suggest possible parallels between acquired epilepsies with known voltage-gated ion channel dysfunction. Furthermore, we discuss possible links toward mTOR regulation of voltage-gated ion channels expression and channel kinetics and the underlying epileptic manifestations in patients with TSC.
Collapse
Affiliation(s)
- Hailey X. Egido-Betancourt
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Roy E. Strowd III
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Xu F, Cai W, Liu B, Qiu Z, Zhang X. Natural L-type calcium channels antagonists from Chinese medicine. Chin Med 2024; 19:72. [PMID: 38773596 PMCID: PMC11107034 DOI: 10.1186/s13020-024-00944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
L-type calcium channels (LTCCs), the largest subfamily of voltage-gated calcium channels (VGCCs), are the main channels for Ca2+ influx during extracellular excitation. LTCCs are widely present in excitable cells, especially cardiac and cardiovascular smooth muscle cells, and participate in various Ca2+-dependent processes. LTCCs have been considered as worthy drug target for cardiovascular, neurological and psychological diseases for decades. Natural products from Traditional Chinese medicine (TCM) have shown the potential as new drugs for the treatment of LTCCs related diseases. In this review, the basic structure, function of LTCCs, and the related human diseases caused by structural or functional abnormalities of LTCCs, and the natural LTCCs antagonist and their potential usages were summarized.
Collapse
Affiliation(s)
- Fangfang Xu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Wanna Cai
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Bo Liu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Zhenwen Qiu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| | - Xiaoqi Zhang
- Guangdong Provincial Engineering Research Center for Modernization of TCM, NMPA Key Laboratory for Quality Evaluation of TCM, Jinan University, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
6
|
Cipriano L, Piscopo R, Aiello C, Novelli A, Iolascon A, Piscopo C. Expanding the Phenotype of the CACNA1C-Associated Neurological Disorders in Children: Systematic Literature Review and Description of a Novel Mutation. CHILDREN (BASEL, SWITZERLAND) 2024; 11:541. [PMID: 38790536 PMCID: PMC11119747 DOI: 10.3390/children11050541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024]
Abstract
Background: CACNA1C gene encodes the alpha 1 subunit of the CaV1.2 L-type Ca2+ channel. Pathogenic variants in this gene have been associated with cardiac rhythm disorders such as long QT syndrome, Brugada syndrome and Timothy syndrome. Recent evidence has suggested the possible association between CACNA1C mutations and neurologically-isolated (in absence of cardiac involvement) phenotypes in children, giving birth to a wider spectrum of CACNA1C-related clinical presentations. However, to date, little is known about the variety of both neurological and non-neurological signs/symptoms in the neurologically-predominant phenotypes. Methods and Results: We conducted a systematic review of neurologically-predominant presentations without cardiac conduction defects, associated with CACNA1C mutations. We also reported a novel de novo missense pathogenic variant in the CACNA1C gene of a children patient presenting with constructional, dressing and oro-buccal apraxia associated with behavioral abnormalities, mild intellectual disability, dental anomalies, gingival hyperplasia and mild musculoskeletal defects, without cardiac conduction defects. Conclusions: The present study highlights the importance of considering the investigation of the CACNA1C gene in children's neurological isolated syndromes, and expands the phenotype of the CACNA1C related conditions. In addition, the present study highlights that, even in absence of cardiac conduction defects, nuanced clinical manifestations of the Timothy syndrome (e.g., dental and gingival defects) could be found. These findings suggest the high variable expressivity of the CACNA1C gene and remark that the absence of cardiac involvement should not mislead the diagnosis of a CACNA1C related disorder.
Collapse
Affiliation(s)
- Lorenzo Cipriano
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy; (L.C.); (A.I.)
| | - Raffaele Piscopo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University Federico II, 80131 Naples, Italy;
| | - Chiara Aiello
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy; (C.A.); (A.N.)
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy; (C.A.); (A.N.)
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy; (L.C.); (A.I.)
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. “Antonio Cardarelli”, 80131 Naples, Italy
| |
Collapse
|
7
|
Hatakama H, Asaoka N, Nagayasu K, Shirakawa H, Kaneko S. Amelioration of obsessive-compulsive disorder by intracellular acidification of cortical neurons with a proton pump inhibitor. Transl Psychiatry 2024; 14:27. [PMID: 38228604 DOI: 10.1038/s41398-024-02731-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
Obsessive-compulsive disorder (OCD) is a highly prevalent neuropsychiatric disorder poorly controlled with pharmacological treatment because of the wide variation in symptom patterns. We analysed real-world data on adverse self-reports and insurance claims to identify a novel therapeutic target for OCD. We found that dopamine D2 receptor (D2R) agonists increased the incidence of OCD-like symptoms, which were suppressed by the concomitant use of proton pump inhibitors (PPIs). Further, OCD-like repetitive and habitual behaviours were observed in mice repeatedly injected with a D2R agonist, quinpirole. However, these abnormalities were suppressed by short-term PPI treatment. In quinpirole-treated mice, PPI inhibited pyramidal neuron hyperactivity in the lateral orbitofrontal cortex, a region where the P-type proton pump gene Atp4a is abundantly expressed. In primary cultured cortical neurons, short-term PPI treatment lowered intracellular pH and decreased firing activity, which was mimicked by Atp4a knockdown. Our findings show that inhibition of P-type proton pumps may be a novel therapeutic strategy for OCD.
Collapse
Affiliation(s)
- Hikari Hatakama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nozomi Asaoka
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
8
|
Harris SA, George AG, Barrett KT, Scantlebury MH, Teskey GC. Febrile seizures lead to prolonged epileptiform activity and hyperoxia that when blocked prevents learning deficits. Epilepsia 2022; 63:2650-2663. [DOI: 10.1111/epi.17371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 07/18/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Sydney A. Harris
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
| | - Antis G. George
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
| | - Karlene T. Barrett
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Departments of Pediatrics and Clinical Neurosciences University of Calgary Calgary AB Canada
| | - Morris H. Scantlebury
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Departments of Pediatrics and Clinical Neurosciences University of Calgary Calgary AB Canada
| | - G. Campbell Teskey
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Department of Cell Biology and Anatomy University of Calgary Calgary AB Canada
| |
Collapse
|
9
|
Lanzetti S, Di Biase V. Small Molecules as Modulators of Voltage-Gated Calcium Channels in Neurological Disorders: State of the Art and Perspectives. Molecules 2022; 27:1312. [PMID: 35209100 PMCID: PMC8879281 DOI: 10.3390/molecules27041312] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.
Collapse
Affiliation(s)
| | - Valentina Di Biase
- Institute of Pharmacology, Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Peter-Mayr Strasse 1, A-6020 Innsbruck, Austria;
| |
Collapse
|
10
|
Seizure Phenotype and Underlying Cellular Defects in Drosophila Knock-In Models of DS (R1648C) and GEFS+ (R1648H) SCN1A Epilepsy. eNeuro 2021; 8:ENEURO.0002-21.2021. [PMID: 34475263 PMCID: PMC8454921 DOI: 10.1523/eneuro.0002-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 08/05/2021] [Accepted: 08/14/2021] [Indexed: 11/21/2022] Open
Abstract
Mutations in the voltage-gated sodium channel gene SCN1A are associated with human epilepsy disorders, but how most of these mutations alter channel properties and result in seizures is unknown. This study focuses on two different mutations occurring at one position within SCN1A. R1648C (R-C) is associated with the severe disorder Dravet syndrome, and R1648H (R-H), with the milder disorder GEFS+. To explore how these different mutations contribute to distinct seizure disorders, Drosophila lines with the R-C or R-H mutation, or R1648R (R-R) control substitution in the fly sodium channel gene para were generated by CRISPR-Cas9 gene editing. The R-C and R-H mutations are homozygous lethal. Animals heterozygous for R-C or R-H mutations displayed reduced life spans and spontaneous and temperature-induced seizures not observed in R-R controls. Electrophysiological recordings from adult GABAergic neurons in R-C and R-H mutants revealed the appearance of sustained neuronal depolarizations and altered firing frequency that were exacerbated at elevated temperature. The only significant change observed in underlying sodium currents in both R-C and R-H mutants was a hyperpolarized deactivation threshold at room and elevated temperature compared with R-R controls. Since this change is constitutive, it is likely to interact with heat-induced changes in other cellular properties to result in the heat-induced increase in sustained depolarizations and seizure activity. Further, the similarity of the behavioral and cellular phenotypes in the R-C and R-H fly lines, suggests that disease symptoms of different severity associated with these mutations in humans could be due in large part to differences in genetic background.
Collapse
|
11
|
Kawano S, Itoh K, Ishihara Y. Maternal intake of docosahexaenoic acid decreased febrile seizure sensitivity by increasing estrogen synthesis in offspring. Epilepsy Behav 2021; 121:108038. [PMID: 34052639 DOI: 10.1016/j.yebeh.2021.108038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
Febrile seizures, which are convulsion in children, are caused by an abrupt increase in body temperature. They are sometimes recurrent, and the more seizures are triggered, the higher the risk of epilepsy and psychiatric disorders increase after growing up. Prevention of febrile seizure is considered to be one of the effective countermeasures in protecting the future health of children; however, pharmacological prevention in the developmental stage is not realistic from the health aspects of the offspring. Docosahexaenoic acid (DHA) is an important nutrient especially during pregnancy and childhood and is reported to suppress several types of epilepsy. The purpose of this study was to examine the effect of DHA intake during pregnancy and infancy on febrile seizures in mice. We used a heat chamber for febrile seizure induction in offspring at the age of from 10 to 11 days old. Intake of DHA during pregnancy and infancy significantly increased the amount of DHA in the brain of offspring. Although DHA had no effect on seizure severity, DHA significantly prolonged the seizure latency and increased body temperature at which the first seizure occurred, indicating that maternal DHA intake decreases febrile seizure sensitivity. Brain estrogen levels significantly increased by DHA intake and administration of an inhibitor for cytochrome P450 aromatase, which is a rate-limiting enzyme for estrogen synthesis, clearly decreased seizure latency and body temperature at which the first seizure occurred. Taken together, DHA could reduce susceptibility to febrile seizures owing to increases in brain estrogen contents. DHA intake during pregnancy and infancy is of significance in protecting infant from seizures as well as conserving health after growth.
Collapse
Affiliation(s)
- Shinji Kawano
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan.
| |
Collapse
|
12
|
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
|
13
|
Mante PK, Adomako NO, Antwi P, Kusi-Boadum NK, Osafo N. Solid-lipid nanoparticle formulation improves antiseizure action of cryptolepine. Biomed Pharmacother 2021; 137:111354. [DOI: 10.1016/j.biopha.2021.111354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/25/2021] [Accepted: 01/31/2021] [Indexed: 10/22/2022] Open
|
14
|
Gulcebi MI, Bartolini E, Lee O, Lisgaras CP, Onat F, Mifsud J, Striano P, Vezzani A, Hildebrand MS, Jimenez-Jimenez D, Junck L, Lewis-Smith D, Scheffer IE, Thijs RD, Zuberi SM, Blenkinsop S, Fowler HJ, Foley A, Sisodiya SM, Berkovic S, Cavalleri G, Correa DJ, Martins Custodio H, Galovic M, Guerrini R, Henshall D, Howard O, Hughes K, Katsarou A, Koeleman BP, Krause R, Lowenstein D, Mandelenaki D, Marini C, O'Brien TJ, Pace A, De Palma L, Perucca P, Pitkänen A, Quinn F, Selmer KK, Steward CA, Swanborough N, Thijs R, Tittensor P, Trivisano M, Weckhuysen S, Zara F. Climate change and epilepsy: Insights from clinical and basic science studies. Epilepsy Behav 2021; 116:107791. [PMID: 33578223 PMCID: PMC9386889 DOI: 10.1016/j.yebeh.2021.107791] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/24/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Climate change is with us. As professionals who place value on evidence-based practice, climate change is something we cannot ignore. The current pandemic of the novel coronavirus, SARS-CoV-2, has demonstrated how global crises can arise suddenly and have a significant impact on public health. Global warming, a chronic process punctuated by acute episodes of extreme weather events, is an insidious global health crisis needing at least as much attention. Many neurological diseases are complex chronic conditions influenced at many levels by changes in the environment. This review aimed to collate and evaluate reports from clinical and basic science about the relationship between climate change and epilepsy. The keywords climate change, seasonal variation, temperature, humidity, thermoregulation, biorhythm, gene, circadian rhythm, heat, and weather were used to search the published evidence. A number of climatic variables are associated with increased seizure frequency in people with epilepsy. Climate change-induced increase in seizure precipitants such as fevers, stress, and sleep deprivation (e.g. as a result of more frequent extreme weather events) or vector-borne infections may trigger or exacerbate seizures, lead to deterioration of seizure control, and affect neurological, cerebrovascular, or cardiovascular comorbidities and risk of sudden unexpected death in epilepsy. Risks are likely to be modified by many factors, ranging from individual genetic variation and temperature-dependent channel function, to housing quality and global supply chains. According to the results of the limited number of experimental studies with animal models of seizures or epilepsy, different seizure types appear to have distinct susceptibility to seasonal influences. Increased body temperature, whether in the context of fever or not, has a critical role in seizure threshold and seizure-related brain damage. Links between climate change and epilepsy are likely to be multifactorial, complex, and often indirect, which makes predictions difficult. We need more data on possible climate-driven altered risks for seizures, epilepsy, and epileptogenesis, to identify underlying mechanisms at systems, cellular, and molecular levels for better understanding of the impact of climate change on epilepsy. Further focussed data would help us to develop evidence for mitigation methods to do more to protect people with epilepsy from the effects of climate change.
Collapse
Affiliation(s)
- Medine I. Gulcebi
- Department of Medical Pharmacology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Emanuele Bartolini
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100 Prato, Italy.
| | - Omay Lee
- Department of Neurology and Clinical Neurophysiology, St. George's University Hospitals NHS Foundation Trust, London, UK.
| | - Christos Panagiotis Lisgaras
- New York University Langone Health, 100 First Ave., New York, NY 10016, USA; The Nathan S. Kline Institute for Psychiatric Research, Center for Dementia Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA.
| | - Filiz Onat
- Department of Medical Pharmacology, Marmara University School of Medicine, Istanbul, Turkey,Department of Medical Pharmacology, Acibadem University School of Medicine, Istanbul, Turkey
| | - Janet Mifsud
- Department of Clinical Pharmacology and Therapeutics, University of Malta, Msida MSD2040, Malta.
| | - Pasquale Striano
- Paediatric Neurology and Muscular Diseases Unit, DINOGMI-Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, IRCCS “Giannina Gaslini” Institute, Genova, Italy
| | - Annamaria Vezzani
- Laboratory of Experimental Neurology, Department of Neuroscience, IRCCS 'Mario Negri' Institute for Pharmacological Research, Milan, Italy.
| | - Michael S. Hildebrand
- Department of Medicine (Austin Health), University of Melbourne, and Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Diego Jimenez-Jimenez
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK and Chalfont Centre for Epilepsy, Bucks, UK.
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| | - David Lewis-Smith
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Ingrid E. Scheffer
- University of Melbourne, Austin Health and Royal Children’s Hospital, Florey Institute and Murdoch Children’s Research Institute, Melbourne, Australia
| | - Roland D. Thijs
- Department of Neurology, Leiden University Medical Centre (LUMC), PO Box 9600, 2300 RC Leiden, the Netherlands
| | - Sameer M. Zuberi
- Paediatric Neurosciences Research Group, Royal Hospital for Children & Institute of Health & Wellbeing, University of Glasgow, Fraser of Allander Neurosciences Unit, Royal Hospital for Children, UK
| | | | - Hayley J. Fowler
- Centre for Earth Systems Engineering Research, School of Engineering, Newcastle University, UK
| | - Aideen Foley
- Department of Geography, Birkbeck College University of London, London, UK.
| | - Epilepsy Climate Change ConsortiumBalestriniSimonaaaBerkovicSamuelabCavalleriGianpieroacCorreaDaniel JoséadMartins CustodioHelenaaeGalovicMarianafGuerriniRenzoagHenshallDavidahHowardOlgaaiHughesKelvinajKatsarouAnnaakKoelemanBobby P.C.alKrauseRolandamLowensteinDanielanMandelenakiDespoinaaoMariniCarlaapO’BrienTerence J.aqPaceAdrianarDe PalmaLucaasPeruccaPieroatPitkänenAslaauQuinnFinolaavSelmerKaja KristineawStewardCharles A.axSwanboroughNicolaayThijsRolandazTittensorPhilbaTrivisanoMarinabbWeckhuysenSarahbcZaraFedericobdDepartment of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK and Chalfont Centre for Epilepsy, Bucks, UKEpilepsy Research Centre, Department of Medicine, Austin Health, University of Melbourne, Melbourne, Victoria, AustraliaDepartment of Molecular and Cellular Therapeutics, The Royal College of Surgeons in Ireland, Dublin 2, Ireland; The FutureNeuro Research Centre, Dublin 2, IrelandSaul R. Korey Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical Center, 1410 Pelham Parkway South, K-312, Bronx, NY 10461, USADepartment of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK; Chalfont Centre for Epilepsy, Bucks, UKUniversity Hospital Zurich, SwitzerlandDepartment of Child Neurology and Psychiatry, University of Pisa and IRCCS Fondazione Stella Maris, 56018 Calambrone, Pisa, ItalyFutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin D02 YN77, IrelandUCB Pharma Ltd, Slough, UKDravet Syndrome UK, UKLaboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USAUniversity Medical Center, Utrecht, The NetherlandsLuxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, LuxembourgDepartment of Neurology, University of California, San Francisco, CA, USADepartment of Pediatric Neurology, Queen Fabiola Children’s University Hospital, Brussels, Brussels Capital Region, BelgiumNeuroscience Department, Children’s Hospital A. Meyer-University of Florence, Florence, ItalyMelbourne Brain Centre, Departments of Medicine and Neurology, Royal Melbourne Hospital, University of Melbourne, VIC, Australia; Departments of Neuroscience and Neurology, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, AustraliaGozo General Hospital, MaltaNeurology Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, ItalyDepartment of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, AustraliaA.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, FinlandILAE-IBE Congress Secretariat, Dublin, IrelandNational Centre for Rare Epilepsy-related Disorders, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, NorwayCongenica Ltd, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1DR, UK; Wellcome Sanger InstituteWellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UKEpilepsy Society, Bucks, UKStichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands; NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London, UKRoyal Wolverhampton NHS Trust, Wolverhampton, UKRare and Complex Epilepsy Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children’s Hospital, IRCCS, Rome, ItalyNeurogenetics Group, Center for Molecular Neurology, VIB, University of Antwerp, Antwerp 2610, BelgiumUnit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Italy
| | - Sanjay M. Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK and Chalfont Centre for Epilepsy, Bucks, UK,Corresponding author at: Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Picollo F, Tomagra G, Bonino V, Carabelli V, Mino L, Olivero P, Pasquarelli A, Truccato M. Triggering Neurotransmitters Secretion from Single Cells by X-ray Nanobeam Irradiation. NANO LETTERS 2020; 20:3889-3894. [PMID: 32227961 PMCID: PMC7997629 DOI: 10.1021/acs.nanolett.0c01046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The employment of ionizing radiation is a powerful tool in cancer therapy, but beyond targeted effects, many studies have highlighted the relevance of its off-target consequences. An exhaustive understanding of the mechanisms underlying these effects is still missing, and no real-time data about signals released by cells during irradiation are presently available. We employed a synchrotron X-ray nanobeam to perform the first real-time simultaneous measurement of both X-ray irradiation and in vitro neurotransmitter release from individual adrenal phaeochromocytoma (PC12) cells plated over a diamond-based multielectrode array. We have demonstrated that, in specific conditions, X-rays can alter cell activity by promoting dopamine exocytosis, and such an effect is potentially very attractive for a more effective treatment of tumors.
Collapse
Affiliation(s)
- Federico Picollo
- Department
of Physics, NIS Interdepartmental Centre, University of Torino and Italian Institute of Nuclear Physics, via Giuria 1, 10125 Torino, Italy
| | - Giulia Tomagra
- Department
of Drug and Science Technology, NIS Interdepartmental Centre, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Valentina Bonino
- Department
of Physics, NIS Interdepartmental Centre, University of Torino and Italian Institute of Nuclear Physics, via Giuria 1, 10125 Torino, Italy
| | - Valentina Carabelli
- Department
of Drug and Science Technology, NIS Interdepartmental Centre, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Lorenzo Mino
- Department
of Chemistry, NIS Interdepartmental Centre, University of Torino, via Giuria 7, 10125 Torino, Italy
| | - Paolo Olivero
- Department
of Physics, NIS Interdepartmental Centre, University of Torino and Italian Institute of Nuclear Physics, via Giuria 1, 10125 Torino, Italy
| | - Alberto Pasquarelli
- Institute
of Electron Devices and Circuits, University
of Ulm, 89069 Ulm, Germany
| | - Marco Truccato
- Department
of Physics, NIS Interdepartmental Centre, University of Torino and Italian Institute of Nuclear Physics, via Giuria 1, 10125 Torino, Italy
| |
Collapse
|
16
|
Horváth ÁC, Borbély S, Boros ÖC, Komáromi L, Koppa P, Barthó P, Fekete Z. Infrared neural stimulation and inhibition using an implantable silicon photonic microdevice. MICROSYSTEMS & NANOENGINEERING 2020; 6:44. [PMID: 34567656 PMCID: PMC8433474 DOI: 10.1038/s41378-020-0153-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/19/2019] [Accepted: 02/24/2020] [Indexed: 05/07/2023]
Abstract
Brain is one of the most temperature sensitive organs. Besides the fundamental role of temperature in cellular metabolism, thermal response of neuronal populations is also significant during the evolution of various neurodegenerative diseases. For such critical environmental factor, thorough mapping of cellular response to variations in temperature is desired in the living brain. So far, limited efforts have been made to create complex devices that are able to modulate temperature, and concurrently record multiple features of the stimulated region. In our work, the in vivo application of a multimodal photonic neural probe is demonstrated. Optical, thermal, and electrophysiological functions are monolithically integrated in a single device. The system facilitates spatial and temporal control of temperature distribution at high precision in the deep brain tissue through an embedded infrared waveguide, while it provides recording of the artefact-free electrical response of individual cells at multiple locations along the probe shaft. Spatial distribution of the optically induced temperature changes is evaluated through in vitro measurements and a validated multi-physical model. The operation of the multimodal microdevice is demonstrated in the rat neocortex and in the hippocampus to increase or suppress firing rate of stimulated neurons in a reversible manner using continuous wave infrared light (λ = 1550 nm). Our approach is envisioned to be a promising candidate as an advanced experimental toolset to reveal thermally evoked responses in the deep neural tissue.
Collapse
Affiliation(s)
- Ágoston Csaba Horváth
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Microsystems Laboratory, Institute for Technical Physics & Material Science, Centre for Energy Research, Budapest, Hungary
- Óbuda University Doctoral School on Materials Sciences and Technologies, Budapest, Hungary
| | - Sándor Borbély
- MTA TTK NAP Sleep Oscillations Research Group, Budapest, Hungary
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Örs Csanád Boros
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Lili Komáromi
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Pál Koppa
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Péter Barthó
- MTA TTK NAP Sleep Oscillations Research Group, Budapest, Hungary
| | - Zoltán Fekete
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Microsystems Laboratory, Institute for Technical Physics & Material Science, Centre for Energy Research, Budapest, Hungary
| |
Collapse
|
17
|
Bongetta D, De Pirro A, Iotti GA, Assietti R. Letter to the Editor Regarding "Intraarterial Dantrolene for Refractory Cerebral Vasospasm in Patients with Aneurysmal Subarachnoid Hemorrhage": Is Malignant Hyperthermia a Model for Showing Dantrolene Efficacy in Limiting Brain Damage? World Neurosurg 2019; 133:431-432. [PMID: 31881561 DOI: 10.1016/j.wneu.2019.08.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Daniele Bongetta
- Neurosurgery Unit, ASST Fatebenefratelli Sacco, Milano, Italy; Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, The University of Pavia, Pavia, Italy.
| | - Antonella De Pirro
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, The University of Pavia, Pavia, Italy
| | - Giorgio Antonio Iotti
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, The University of Pavia, Pavia, Italy; Department of Anesthesia and Intensive Care, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
18
|
Ghotbeddin Z, Heysieattalab S, Borjkhani M, Mirnajafi-Zadeh J, Semnanian S, Hosseinmardi N, Janahmadi M. Ca 2+ Channels Involvement in Low-Frequency Stimulation-Mediated Suppression of Intrinsic Excitability of Hippocampal CA1 Pyramidal Cells in a Rat Amygdala Kindling Model. Neuroscience 2019; 406:234-248. [PMID: 30885638 DOI: 10.1016/j.neuroscience.2019.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/26/2022]
Abstract
Low-frequency stimulation has demonstrated promising seizure suppression in animal models of epilepsy, while the mechanism of the effect is still debated. Changes in intrinsic properties have been recognized as a prominent pathophysiologically relevant feature of numerous neurological disorders including epilepsy. Here, it was evaluated whether LFS can preserve the intrinsic neuronal electrophysiological properties in a rat model of epilepsy, focusing on the possible involvement of voltage-gated Ca2+ channels. The amygdala kindling model was induced by 3 s monophasic square wave pulses (50 Hz, 1 ms duration, 12times/day at 5 min intervals). Both LFS alone and kindled plus LFS (KLFS) groups received four packages of LFS (each consisting of 200 monophasic square pulses, 0.1 ms pulse duration at 1 Hz with the after discharge threshold intensity), which in KLFS rats was applied immediately after kindling induction. Whole-cell patch-clamp recordings were made in the presence of fast synaptic blockers 24 h after the last kindling stimulations or following kindling stimulations plus LFS application. In the KLFS group, both the rebound excitation and kindling-induced intrinsic hyperexcitability were decreased, associated with a regular intrinsic firing as indicated by a lower coefficient of variation. The amplitude of afterdepolarization (ADP) and its area under the curve were both decreased in the KLFS group compared to the kindled group. LFS prevented the increasing effect of kindling on Ca2+ currents in the KLFS group. Findings provided evidence for a novel form of epileptiform activity suppression by LFS in the presence of synaptic blockade possibly by decreasing Ca2+ currents.
Collapse
Affiliation(s)
- Zohreh Ghotbeddin
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Physiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | - Mehdi Borjkhani
- Department of Electrical Engineering, Urmia University of Technology, Urmia, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Hosseinmardi
- Neuroscience Research Center and Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center and Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Heine M, Heck J, Ciuraszkiewicz A, Bikbaev A. Dynamic compartmentalization of calcium channel signalling in neurons. Neuropharmacology 2019; 169:107556. [PMID: 30851307 DOI: 10.1016/j.neuropharm.2019.02.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/16/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022]
Abstract
Calcium fluxes through the neuronal membrane are strictly limited in time due to biophysical properties of voltage-gated and ligand-activated ion channels and receptors. Being embedded into the crowded dynamic environment of biological membranes, Ca2+-permeable receptors and channels undergo perpetual spatial rearrangement, which enables their temporary association and formation of transient signalling complexes. Thus, efficient calcium-mediated signal transduction requires mechanisms to support very precise spatiotemporal alignment of the calcium source and Ca2+-binding lipids and proteins in a highly dynamic environment. The mobility of calcium channels and calcium-sensing proteins themselves can be considered as a physiologically meaningful variable that affects calcium-mediated signalling in neurons. In this review, we will focus on voltage-gated calcium channels (VGCCs) and activity-induced relocation of stromal interaction molecules (STIMs) in the endoplasmic reticulum (ER) to show that particularly in time ranges between milliseconds to minutes, dynamic rearrangement of calcium conducting channels and sensor molecules is of physiological relevance. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Martin Heine
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, 39106, Germany; RG Functional Neurobiology, Institute for Development Biology and Neurobiology, Johannes Gutenberg University Mainz, Germany.
| | - Jennifer Heck
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany
| | - Anna Ciuraszkiewicz
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, 39106, Germany
| | - Arthur Bikbaev
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany
| |
Collapse
|
20
|
Latent Sex Differences in Molecular Signaling That Underlies Excitatory Synaptic Potentiation in the Hippocampus. J Neurosci 2018; 39:1552-1565. [PMID: 30578341 DOI: 10.1523/jneurosci.1897-18.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/27/2018] [Accepted: 12/13/2018] [Indexed: 01/25/2023] Open
Abstract
Excitatory synapses can be potentiated by chemical neuromodulators, including 17β-estradiol (E2), or patterns of synaptic activation, as in long-term potentiation (LTP). Here, we investigated kinases and calcium sources required for acute E2-induced synaptic potentiation in the hippocampus of each sex and tested whether sex differences in kinase signaling extend to LTP. We recorded EPSCs from CA1 pyramidal cells in hippocampal slices from adult rats and used specific inhibitors of kinases and calcium sources. This revealed that, although E2 potentiates synapses to the same degree in each sex, cAMP-activated protein kinase (PKA) is required to initiate potentiation only in females. In contrast, mitogen-activated protein kinase, Src tyrosine kinase, and rho-associated kinase are required for initiation in both sexes; similarly, Ca2+/calmodulin-activated kinase II is required for expression/maintenance of E2-induced potentiation in both sexes. Calcium source experiments showed that L-type calcium channels and calcium release from internal stores are both required for E2-induced potentiation in females, whereas in males, either L-type calcium channel activation or calcium release from stores is sufficient to permit potentiation. To investigate the generalizability of a sex difference in the requirement for PKA in synaptic potentiation, we tested how PKA inhibition affects LTP. This showed that, although the magnitude of both high-frequency stimulation-induced and pairing-induced LTP is the same between sexes, PKA is required for LTP in females but not males. These results demonstrate latent sex differences in mechanisms of synaptic potentiation in which distinct molecular signaling converges to common functional endpoints in males and females.SIGNIFICANCE STATEMENT Chemical- and activity-dependent neuromodulation alters synaptic strength in both male and female brains, yet few studies have compared mechanisms of neuromodulation between the sexes. Here, we studied molecular signaling that underlies estrogen-induced and activity-dependent potentiation of excitatory synapses in the hippocampus. We found that, despite similar magnitude increases in synaptic strength in males and females, the roles of cAMP-regulated protein kinase, internal calcium stores, and L-type calcium channels differ between the sexes. Therefore, latent sex differences in which the same outcome is achieved through distinct underlying mechanisms in males and females include kinase and calcium signaling involved in synaptic potentiation, demonstrating that sex is an important factor in identification of molecular targets for therapeutic development based on mechanisms of neuromodulation.
Collapse
|
21
|
Saboory E, Ghadimkhani M, Roshan-Milani S, Derafshpour L, Mohammadi S, Dindarian S, Mohammadi H. Effect of early-life inflammation and magnesium sulfate on hyperthermia-induced seizures in infant rats: Susceptibility to pentylenetetrazol-induced seizures later in life. Dev Psychobiol 2018; 61:96-106. [PMID: 30338516 DOI: 10.1002/dev.21781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Abstract
This study investigated the effect of inflammation and MgSO4 pretreatment on behaviors caused by hyperthermia (HT) and the effect of these interventions on PTZ-induced seizure a week later. In this experimental study, rat pups experienced inflammation on postnatal day 10 (P10). On P18-19, the pups received either saline or MgSO4 then subjected to hyperthermia. On P25-26, PTZ-induced seizure was initiated in the rats. Neonatal inflammation increased the susceptibility to HT-induced seizure. Inflammation and HT increased the susceptibility to PTZ-induced seizure. Pretreatment with MgSO4 before hyperthermia decreased the susceptibility to both HT- and PTZ-induced seizure. Furthermore, calcium and magnesium blood levels significantly decreased compared to control rats. It can be concluded that neonatal inflammation potentiates while pretreatment with MgSO4 attenuates HT-induced seizures. Also, neonatal inflammation and HT potentiate PTZ-induced seizure initiated one week later.
Collapse
Affiliation(s)
- Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Ghadimkhani
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Shiva Roshan-Milani
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sedra Mohammadi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sina Dindarian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hozan Mohammadi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
22
|
Cajigas I, Chakraborty A, Swyter KR, Luo H, Bastidas M, Nigro M, Morris ER, Chen S, VanGompel MJW, Leib D, Kohtz SJ, Martina M, Koh S, Ay F, Kohtz JD. The Evf2 Ultraconserved Enhancer lncRNA Functionally and Spatially Organizes Megabase Distant Genes in the Developing Forebrain. Mol Cell 2018; 71:956-972.e9. [PMID: 30146317 PMCID: PMC6428050 DOI: 10.1016/j.molcel.2018.07.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/30/2018] [Accepted: 07/20/2018] [Indexed: 02/06/2023]
Abstract
Gene regulation requires selective targeting of DNA regulatory enhancers over megabase distances. Here we show that Evf2, a cloud-forming Dlx5/6 ultraconserved enhancer (UCE) lncRNA, simultaneously localizes to activated (Umad1, 1.6 Mb distant) and repressed (Akr1b8, 27 Mb distant) chr6 target genes, precisely regulating UCE-gene distances and cohesin binding in mouse embryonic forebrain GABAergic interneurons (INs). Transgene expression of Evf2 activates Lsm8 (12 Mb distant) but fails to repress Akr1b8, supporting trans activation and long-range cis repression. Through both short-range (Dlx6 antisense) and long-range (Akr1b8) repression, the Evf2-5'UCE links homeodomain and mevalonate pathway-regulated enhancers to IN diversity. The Evf2-3' end is required for long-range activation but dispensable for RNA cloud localization, functionally dividing the RNA into 3'-activator and 5'UCE repressor and targeting regions. Together, these results support that Evf2 selectively regulates UCE interactions with multi-megabase distant genes through complex effects on chromosome topology, linking lncRNA-dependent topological and transcriptional control with interneuron diversity and seizure susceptibility.
Collapse
Affiliation(s)
- Ivelisse Cajigas
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Abhijit Chakraborty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Kelsey R Swyter
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Hao Luo
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Monique Bastidas
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Maximilliano Nigro
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Elizabeth R Morris
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Sean Chen
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Michael J W VanGompel
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - David Leib
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Sara J Kohtz
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA
| | - Marco Martina
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Sooky Koh
- Department of Pediatrics, Emory University, Atlanta, GA 30307, USA
| | - Ferhat Ay
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA; School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jhumku D Kohtz
- Department of Pediatrics and Developmental Biology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Box 204, 2430 N. Halsted, Chicago, IL 60614, USA.
| |
Collapse
|
23
|
Inhibition of DPP4 enhances inhibitory synaptic transmission through activating the GLP-1/GLP-1R signaling pathway in a rat model of febrile seizures. Biochem Pharmacol 2018; 156:78-85. [PMID: 30086287 DOI: 10.1016/j.bcp.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
Dipeptidyl peptidase-IV (DPP4) is a cell surface serine peptidase widely expressed in the brain. Recent studies suggest that DPP4 contributes to the development of febrile seizures (FS); however, the underlying mechanism is still unclear. Thus, we investigated the role of DPP4 in the progression of FS at the molecular and electrophysiological levels using FS models in vivo and in vitro. Herein, we found that both the mRNA and protein levels of DPP4 were upregulated in the FS model. Administration of the pharmacological DPP4 inhibitor sitagliptin suppressed the hyperthermia-induced neuronal excitability as determined via whole-cell patch-clamp recordings in vitro. Interestingly, sitagliptin administration activated the glucagon-like peptide-1 (GLP-1)/GLP-1 receptor (GLP-1R) pathway by increasing the expression of GLP-1 and GLP-1R in a rat model of FS. Moreover, administration of the GLP-1R inhibitor exendin9-39 increased seizure severity, and sitagliptin reversed the effect, as shown in the electroencephalogram (EEG) and patch-clamp results in a rat model of FS. Furthermore, the GLP-1R-mediated reduction in GABAergic transmission was enhanced by sitagliptin and DPP4 knockdown through increasing miniature inhibitory post-synaptic currents (mIPSCs) in vitro accompanied by increased synaptic release of GABA in vivo. Taken together, our results demonstrate a role of DPP4 in regulating GABAergic transmission via the GLP-1/GLP-1R pathway. These findings indicated that DPP4 may represent a novel therapeutic strategy and target for FS.
Collapse
|
24
|
Rosenberg EC, Lippman-Bell JJ, Handy M, Soldan SS, Rakhade S, Hilario-Gomez C, Folweiler K, Jacobs L, Jensen FE. Regulation of seizure-induced MeCP2 Ser421 phosphorylation in the developing brain. Neurobiol Dis 2018; 116:120-130. [PMID: 29738885 DOI: 10.1016/j.nbd.2018.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/23/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
Neonatal seizures disrupt normal synaptic maturation and often lead to later-life epilepsy and cognitive deficits. During early life, the brain exhibits heightened synaptic plasticity, in part due to a developmental overabundance of CaV1.2 L-type voltage gated calcium (Ca2+) channels (LT-VGCCs) and Ca2+-permeable AMPARs (CP-AMPARs) lacking GluA2 subunits. We hypothesized that early-life seizures overactivate these channels, in turn dysregulating Ca2+-dependent signaling pathways including that of methyl CPG binding protein 2 (MeCP2), a transcription factor implicated in the autism spectrum disorder (ASD) Rett Syndrome. Here, we show that in vivo hypoxia-induced seizures (HS) in postnatal day (P)10 rats acutely induced phosphorylation of the neuronal-specific target of activity-dependent MeCP2 phosphorylation, S421, as well as its upstream activator CaMKII T286. We next identified mechanisms by which activity-dependent Ca2+ influx induced MeCP2 phosphorylation using in vitro cortical and hippocampal neuronal cultures at embryonic day (E)18 + 10 days in vitro (DIV). In contrast to the prevalent role of NMDARs in the adult brain, we found that both CP-AMPARs and LT-VGCCs mediated MeCP2 S421 and CaMKII T286 phosphorylation induced by kainic acid (KA) or high potassium chloride (KCl) stimulation. Furthermore, in vivo post-seizure treatment with the broad-spectrum AMPAR antagonist NBQX, the CP-AMPAR blocker IEM-1460, or the LT-VGCC antagonist nimodipine blocked seizure-induced MeCP2 phosphorylation. Collectively, these results demonstrate that early-life seizures dysregulate critical activity-dependent developmental signaling pathways, in part via CP-AMPAR and LT-VGCC activation, providing novel age-specific therapeutic targets for convergent pathways underlying epilepsy and ASDs.
Collapse
Affiliation(s)
- Evan C Rosenberg
- Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States; New York University Langone Medical Center, New York, NY 10016, United States
| | - Jocelyn J Lippman-Bell
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States; Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States; Philadelphia College of Osteopathic Medicine, Department of Biomedical Sciences, Philadelphia, PA 19131, United States
| | - Marcus Handy
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Samantha S Soldan
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Sanjay Rakhade
- Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States
| | | | - Kaitlyn Folweiler
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Leah Jacobs
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Frances E Jensen
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States; Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States.
| |
Collapse
|
25
|
Abstract
Fever-associated seizures or epilepsy (FASE) is primarily characterised by the occurrence of a seizure or epilepsy usually accompanied by a fever. It is common in infants and children, and generally includes febrile seizures (FS), febrile seizures plus (FS+), Dravet syndrome (DS) and genetic epilepsy with febrile seizures plus (GEFSP). The aetiology of FASE is unclear. Genetic factors may play crucial roles in FASE. Mutations in certain genes may cause a wide spectrum of phenotypical overlap ranging from isolated FS, FS+ and GEFSP to DS. Synapse-associated proteins, postsynaptic GABAA receptor, and sodium channels play important roles in synaptic transmission. Mutations in these genes may involve in the pathogenesis of FASE. Elevated temperature promotes synaptic vesicle (SV) recycling and enlarges SV size, which may enhance synaptic transmission and contribute to FASE occurring. This review provides an overview of the loci, genes, underlying pathogenesis and the fever-inducing effect of FASE. It may provide a more comprehensive understanding of pathogenesis and contribute to the clinical diagnosis of FASE.
Collapse
|
26
|
Abdelsayed M, Ruprai M, Ruben PC. The efficacy of Ranolazine on E1784K is altered by temperature and calcium. Sci Rep 2018; 8:3643. [PMID: 29483621 PMCID: PMC5827758 DOI: 10.1038/s41598-018-22033-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/15/2018] [Indexed: 12/19/2022] Open
Abstract
E1784K is the most common mixed syndrome SCN5a mutation underpinning both Brugada syndrome type 1 (BrS1) and Long-QT syndrome type 3 (LQT3). The charge reversal mutant enhances the late sodium current (INa) passed by the cardiac voltage-gated sodium channel (NaV1.5), delaying cardiac repolarization. Exercise-induced triggers, like elevated temperature and cytosolic calcium, exacerbate E1784K late INa. In this study, we tested the effects of Ranolazine, the late INa blocker, on voltage-dependent and kinetic properties of E1784K at elevated temperature and cytosolic calcium. We used whole-cell patch clamp to measure INa from wild type and E1784K channels expressed in HEK293 cells. At elevated temperature, Ranolazine attenuated gain-of-function in E1784K by decreasing late INa, hyperpolarizing steady-state fast inactivation, and increasing use-dependent inactivation. Both elevated temperature and cytosolic calcium hampered the capacity of Ranolazine to suppress E1784K late INa. In-silico action potential (AP) simulations were done using a modified O'Hara Rudy (ORd) cardiac model. Simulations showed that Ranolazine failed to shorten AP duration, an effect augmented at febrile temperatures. The drug-channel interaction is clearly affected by external triggers, as reported previously with ischemia. Determining drug efficacy under various physiological states in SCN5a cohorts is crucial for accurate management of arrhythmias.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Manpreet Ruprai
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
27
|
An X, Yang P, Chen S, Zhang F, Yu D. An Additional Prior Retrieval Alters the Effects of a Retrieval-Extinction Procedure on Recent and Remote Fear Memory. Front Behav Neurosci 2018; 11:259. [PMID: 29358910 PMCID: PMC5766663 DOI: 10.3389/fnbeh.2017.00259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 12/19/2017] [Indexed: 11/19/2022] Open
Abstract
Several studies have shown that the isolated retrieval of a consolidated fear memory can induce a labile phase, during which extinction training can prevent the reinstatement, a form of relapse in which fear response to a fear-provoking context returns when a mild shock is presented. However, fear memory retrieval may also have another opposing result: the enhancement of fear memory. This implies that the fear memory trace can be modified by a brief retrieval. Unclear is whether the fear-impairing effect of retrieval-extinction (RE) is altered by a prior brief retrieval. The present study investigated the responses of recent and remote fear memories to the RE procedure after the presentation of an additional prior retrieval (priRet). We found that a single RE procedure effectively blocked the reinstatement of 2-day recent contextual fear memory. The memory-impairing effect of the RE procedure on recent fear was not observed when priRet was presented 6 or 24 h before the RE procedure. In contrast to the 2-day recent memory, the RE procedure failed to block the reinstatement of 36-day remote fear memory but successfully disrupted the return of remote fear memory after priRet. This memory-disruptive effect on remote memory did not occur when priRet was performed in a novel context. Nimodipine administration revealed that the blockade of priRet-induced processes recovered the effects of the RE procedure on both recent and remote fear memories. Our findings suggest that the susceptibility of recent and remote fear memories to RE procedures can be altered by an additional retrieval.
Collapse
Affiliation(s)
- Xianli An
- School of Educational Science, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, China
| | - Ping Yang
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Siguang Chen
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Fenfen Zhang
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, China.,RNA Center, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou, China
| |
Collapse
|
28
|
Schampel A, Kuerten S. Danger: High Voltage-The Role of Voltage-Gated Calcium Channels in Central Nervous System Pathology. Cells 2017; 6:E43. [PMID: 29140302 PMCID: PMC5755501 DOI: 10.3390/cells6040043] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely distributed within the central nervous system (CNS) and presumed to play an important role in the pathophysiology of a broad spectrum of CNS disorders including Alzheimer's and Parkinson's disease as well as multiple sclerosis. Several calcium channel blockers have been in clinical practice for many years so that their toxicity and side effects are well studied. However, these drugs are primarily used for the treatment of cardiovascular diseases and most if not all effects on brain functions are secondary to peripheral effects on blood pressure and circulation. While the use of calcium channel antagonists for the treatment of CNS diseases therefore still heavily depends on the development of novel strategies to specifically target different channels and channel subunits, this review is meant to provide an impulse to further emphasize the importance of future research towards this goal.
Collapse
Affiliation(s)
- Andrea Schampel
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg 97070, Germany.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91054, Germany.
| |
Collapse
|
29
|
Rungta RL, Osmanski BF, Boido D, Tanter M, Charpak S. Light controls cerebral blood flow in naive animals. Nat Commun 2017; 8:14191. [PMID: 28139643 PMCID: PMC5290324 DOI: 10.1038/ncomms14191] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 12/07/2016] [Indexed: 01/05/2023] Open
Abstract
Optogenetics is increasingly used to map brain activation using techniques that rely on functional hyperaemia, such as opto-fMRI. Here we test whether light stimulation protocols similar to those commonly used in opto-fMRI or to study neurovascular coupling modulate blood flow in mice that do not express light sensitive proteins. Combining two-photon laser scanning microscopy and ultrafast functional ultrasound imaging, we report that in the naive mouse brain, light per se causes a calcium decrease in arteriolar smooth muscle cells, leading to pronounced vasodilation, without excitation of neurons and astrocytes. This photodilation is reversible, reproducible and energy-dependent, appearing at about 0.5 mJ. These results impose careful consideration on the use of photo-activation in studies involving blood flow regulation, as well as in studies requiring prolonged and repetitive stimulations to correct cellular defects in pathological models. They also suggest that light could be used to locally increase blood flow in a controlled fashion. Combination of optogenetics and BOLD fMRI is routinely used to map neuronal activity upon photostimulation. Here the authors show that light, shone at intensities used in optogenetic studies, dilates vessels and increases blood flow independently of exogenous light-sensitive proteins in the mouse brain.
Collapse
Affiliation(s)
- Ravi L Rungta
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris 75006, France
| | - Bruno-Félix Osmanski
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris 75006, France
| | - Davide Boido
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris 75006, France
| | - Mickael Tanter
- Institut Langevin, Espci Paris, CNRS UMR 7587, INSERM U979, PSL Research University, 17 rue Moreau, Paris 75012, France
| | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris 75006, France
| |
Collapse
|
30
|
Wang Z, Zhang Y, Fang J, Yu F, Heng D, Fan Y, Xu J, Peng B, Liu W, Han S, He X. Decreased Methylation Level of H3K27me3 Increases Seizure Susceptibility. Mol Neurobiol 2016; 54:7343-7352. [PMID: 27815838 DOI: 10.1007/s12035-016-0197-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/11/2016] [Indexed: 01/01/2023]
Abstract
Epigenetic modifications including histone modifications are associated with seizure development and epileptogenesis; however, its underlying mechanism remains to be elucidated. Dipeptidyl peptidase 4 (DPP4) and IL6 are identified as febrile seizure (FS)-related genes using gene microarray analysis in hyperthermia prone (HP) rats. This purpose of the study focused on exploring whether epigenetic modifications marker histone H3 lysine 27 trimethylation (H3K27me3)-regulated DPP4 and IL6 expression further affected seizures development. Herein, we reported broad between-group differences in the global levels of H3K27me3 with increased seizure severity in vivo. Using chromatin immunoprecipitation (ChIP), we identified markedly decreased H3K27me3 enrichment at their promoters of DPP4 and IL6 in vivo. We further showed that hyperthermia significantly decreased protein levels of H3K27me3, increased mRNA levels of DPP4 and IL6 by decreasing H3K27me3 enrichment at their promoters of DPP4 and IL6 in vitro. Importantly, H3K27me3 loss via enhancer of zeste homolog 2 (EZH2) knockdown promoted expression of DPP4 and IL6 via the same mechanism in vitro. EZH2 knockdown also increased neuronal firing frequency in vitro and FS susceptibility in vivo companied with upregulation expression of DPP4 and IL6. Taken together, our study provided the first evidence that hyperthermia-induced decreased of H3K27me3 promoted seizure susceptibility via regulating the expression pattern of DPP4 and IL6. These findings suggested that the methylation level of H3K27me3 might be a key regulator of seizure susceptibility.
Collapse
Affiliation(s)
- Zhongcheng Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China
| | - Yusong Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China
| | - Jian Fang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China
| | - Fang Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China
| | - Duanhe Heng
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China
| | - Yuanteng Fan
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jian Xu
- Weifang Maternity and Child Hospital, Weifang, 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
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China.
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Donghu Road No. 185, Wuchang, Wuhan, 430071, China.
| |
Collapse
|
31
|
Radzicki D, Liu E, Deng HX, Siddique T, Martina M. Early Impairment of Synaptic and Intrinsic Excitability in Mice Expressing ALS/Dementia-Linked Mutant UBQLN2. Front Cell Neurosci 2016; 10:216. [PMID: 27703430 PMCID: PMC5028382 DOI: 10.3389/fncel.2016.00216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/30/2016] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are believed to represent the different outcomes of a common pathogenic mechanism. However, while researchers have intensely studied the involvement of motor neurons in the ALS/FTD syndrome, very little is known about the function of hippocampal neurons, although this area is critical for memory and other cognitive functions. We investigated the electrophysiological properties of CA1 pyramidal cells in slices from 1 month-old UBQLN2P497H mice, a recently generated model of ALS/FTD that shows heavy depositions of ubiquilin2-positive aggregates in this brain region. We found that, compared to wild-type mice, cells from UBQLN2P497H mice were hypo-excitable. The amplitude of the glutamatergic currents elicited by afferent fiber stimulation was reduced by ~50%, but no change was detected in paired-pulse plasticity. The maximum firing frequency in response to depolarizing current injection was reduced by ~30%; the fast afterhyperpolarization in response to a range of depolarizations was reduced by almost 10 mV; the maximum slow afterhyperpolarization (sAHP) was also significantly decreased, likely in consequence of the decreased number of spikes. Finally, the action potential (AP) upstroke was blunted and the threshold depolarized compared to controls. Thus, synaptic and intrinsic excitability are both impaired in CA1 pyramidal cells of UBQLN2P497H mice, likely constituting a cellular mechanism for the cognitive impairments. Because these alterations are detectable before the establishment of overt pathology, we hypothesize that they may affect the further course of the disease.
Collapse
Affiliation(s)
- Daniel Radzicki
- Department of Physiology, Northwestern University Feinberg School of Medicine Chicago, IL, USA
| | - Erdong Liu
- Department of Neurology, Northwestern University Feinberg School of Medicine Chicago, IL, USA
| | - Han-Xiang Deng
- Department of Neurology, Northwestern University Feinberg School of Medicine Chicago, IL, USA
| | - Teepu Siddique
- Department of Neurology, Northwestern University Feinberg School of Medicine Chicago, IL, USA
| | - Marco Martina
- Department of Physiology, Northwestern University Feinberg School of Medicine Chicago, IL, USA
| |
Collapse
|
32
|
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
|
33
|
Striessnig J, Ortner NJ, Pinggera A. Pharmacology of L-type Calcium Channels: Novel Drugs for Old Targets? Curr Mol Pharmacol 2016; 8:110-22. [PMID: 25966690 PMCID: PMC5384371 DOI: 10.2174/1874467208666150507105845] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/10/2015] [Accepted: 04/20/2015] [Indexed: 11/22/2022]
Abstract
Inhibition of voltage-gated L-type calcium channels by organic calcium channel blockers is a well-established pharmacodynamic concept for the treatment of hypertension and cardiac ischemia. Since decades these antihypertensives (such as the dihydropyridines amlodipine, felodipine or nifedipine) belong to the most widely prescribed drugs
world-wide. Their tolerability is excellent because at therapeutic doses their pharmacological effects in humans are limited to the cardiovascular system. During the last years substantial progress has been made to reveal the physiological role of different L-type calcium channel isoforms in many other tissues, including the brain, endocrine and sensory cells.
Moreover, there is accumulating evidence about their involvement in various human diseases, such as Parkinson's disease, neuropsychiatric disorders and hyperaldosteronism. In this review we discuss the pathogenetic role of L-type calcium channels, potential new indications for existing or isoform-selective compounds and strategies to minimize potential side effects.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
34
|
Ghadimkhani M, Saboory E, Roshan-Milani S, Mohammdi S, Rasmi Y. Effect of magnesium sulfate on hyperthermia and pentylen-tetrazol-induced seizure in developing rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:608-14. [PMID: 27482341 PMCID: PMC4951599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
OBJECTIVES Febrile seizures (FS) are the most common type of convulsive events among children. Its prevalence has been estimated to be 2-5% in children between 3 months and 5 years old. Also, blood and CSF magnesium levels have been demonstrated to be reduced in children with FS. This study investigates the effect of MgSo4 pretreatment on the behaviors caused by hyperthermia (HT) and effect of these two on pentylen-tetrazol (PTZ)-induced seizure later in life. MATERIALS AND METHODS Thirty two Wistar rats were assigned to 2 groups: saline-hyperthermia-pentylentetrazol (SHP) and magnesium-hyperthermia-pentylentetrazol (MHP). In both groups, HT was induced at the age of 18-19 days old. Before the HT, MHP group received MgSo4 and SHP group received normal saline intraperitoneally (IP). Behaviors of the rats were recorded during the HT. Then, in half of each group (n=8) at the age of 25-26 days old and in other half at the age of 78-79 days, seizure was induced by PTZ. RESULTS The HT successfully caused convulsive behaviors in the rats and pretreatment with MgSo4 before HT attenuated HT-induced convulsive behaviors. PTZ-induced seizures a week later was more severe than those of 2 months later. CONCLUSION It can be concluded that pretreatment with MgSO4 inhibits HT-induced seizure and, in a long run, this intervention reduced PTZ-induced seizure later in life.
Collapse
Affiliation(s)
- Maryam Ghadimkhani
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran,Corresponding author: Ehsan Saboory. Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran. Tel: +98-443-2770698; Fax: +98-443-2780801; ;
| | - Shiva Roshan-Milani
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sedra Mohammdi
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
35
|
Kim I, Mlsna LM, Yoon S, Le B, Yu S, Xu D, Koh S. A postnatal peak in microglial development in the mouse hippocampus is correlated with heightened sensitivity to seizure triggers. Brain Behav 2015; 5:e00403. [PMID: 26807334 PMCID: PMC4714636 DOI: 10.1002/brb3.403] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 07/30/2015] [Accepted: 09/02/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Explosive synaptogenesis and synaptic pruning occur in the hippocampus during the first two weeks of postnatal life, coincident with a heightened susceptibility to seizures in rodents. To determine the temporal correlation between microglial development and age-dependent susceptibility and response to seizures, we quantified developmental changes in basal microglia levels and seizure-induced microglial activation in the hippocampus of Cx3Cr1(GFP /+) transgenic mice. METHODS Basal levels of microglia were quantified in the hippocampi of Cx3Cr1(GFP /+) mice at P0, P5, P10, P15, P20, P25, P30, P40, and P60. Seizure susceptibility and seizure-induced microglial activation were assessed in response to febrile seizures (lipopolysaccharide followed by hyperthermia) and kainic acid-induced status epilepticus. RESULTS The density of microglia within the hippocampus increased rapidly after birth, reaching a peak during the second week of life - the age at which the animals became most vulnerable to seizure triggers. In addition, this peak of microglial development and seizure vulnerability during the second postnatal week represented the time of maximal seizure-induced microglia activation. CONCLUSIONS Overreactive innate immunity mediated by activated microglia may exacerbate acute injury to neuronal synapses and contribute to the long-term epileptogenic effects of early-life seizures. Anti-inflammatory therapy targeting excessive production of inflammatory mediators by activated microglia, therefore, may be an effective age-specific therapeutic strategy to minimize neuronal dysfunction and prevent increases in susceptibility to subsequent seizures in developing animals.
Collapse
Affiliation(s)
- Iris Kim
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Lauren M Mlsna
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Stella Yoon
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Brandy Le
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Songtao Yu
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Dan Xu
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| | - Sookyong Koh
- Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute Department of Pediatrics Feinberg School of Medicine Northwestern University Chicago Illinois
| |
Collapse
|
36
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
37
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 786] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
38
|
Suchomelova L, Lopez-Meraz ML, Niquet J, Kubova H, Wasterlain CG. Hyperthermia aggravates status epilepticus-induced epileptogenesis and neuronal loss in immature rats. Neuroscience 2015; 305:209-24. [PMID: 26259902 DOI: 10.1016/j.neuroscience.2015.08.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/20/2015] [Accepted: 08/03/2015] [Indexed: 01/16/2023]
Abstract
This study tightly controlled seizure duration and severity during status epilepticus (SE) in postnatal day 10 (P10) rats, in order to isolate hyperthermia as the main variable and to study its consequences. Body temperature was maintained at 39 ± 1 °C in hyperthermic SE rats (HT+SE) or at 35 ± 1 °C in normothermic SE animals (NT+SE) during 30 min of SE, which was induced by lithium-pilocarpine (3 mEq/kg, 60 mg/kg) and terminated by diazepam and cooling to NT. All video/EEG measures of SE severity were similar between HT+SE and NT+SE pups. At 24h, neuronal injury was present in the amygdala in the HT+SE group only, and was far more severe in the hippocampus in HT+SE than NT+SE pups. Separate groups of animals were monitored four months later for spontaneous recurrent seizures (SRS). Only HT+SE animals developed convulsive SRS. Both HT+SE and NT+SE animals developed electrographic SRS (83% vs. 55%), but SRS frequency and severity were higher in hyperthermic animals (12.5 ± 3.5 vs. 4.2 ± 2.0 SRS/day). The density of hilar neurons was lower, thickness of the amygdala and perirhinal cortex was reduced, and lateral ventricles were enlarged in HT+SE over NT+SE littermates and HT/NT controls. In this model, hyperthermia greatly increased the epileptogenicity of SE and its neuropathological sequelae.
Collapse
Affiliation(s)
- L Suchomelova
- Veterans Administration Greater Los Angeles Healthcare System, Epilepsy Research (151), 11 301 Wilshire Boulevard, Building 114, Room 139, Los Angeles, CA 90073, USA.
| | - M L Lopez-Meraz
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Veracruz 91190, Mexico
| | - J Niquet
- Veterans Administration Greater Los Angeles Healthcare System, Epilepsy Research (151), 11 301 Wilshire Boulevard, Building 114, Room 139, Los Angeles, CA 90073, USA
| | - H Kubova
- Department of Developmental Epileptology, Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 14220, Czech Republic
| | - C G Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, VA Medical Center (127), 11 301 Wilshire Boulevard, Los Angeles, CA 90073, USA
| |
Collapse
|
39
|
Eun BL, Abraham J, Mlsna L, Kim MJ, Koh S. Lipopolysaccharide potentiates hyperthermia-induced seizures. Brain Behav 2015; 5:e00348. [PMID: 26357586 PMCID: PMC4559014 DOI: 10.1002/brb3.348] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/25/2015] [Accepted: 04/05/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Prolonged febrile seizures (FS) have both acute and long-lasting effects on the developing brain. Because FS are often associated with peripheral infection, we aimed to develop a preclinical model of FS that simulates fever and immune activation in order to facilitate the implementation of targeted therapy after prolonged FS in young children. METHODS The innate immune activator lipopolysaccharide (LPS) was administered to postnatal day 14 rat (200 μg/kg) and mouse (100 μg/kg) pups 2-2.5 h prior to hyperthermic seizures (HT) induced by hair dryer or heat lamp. To determine whether simulation of infection enhances neuronal excitability, latency to seizure onset, threshold temperature and total number of seizures were quantified. Behavioral seizures were correlated with electroencephalographic changes in rat pups. Seizure-induced proinflammatory cytokine production was assessed in blood samples at various time points after HT. Seizure-induced microglia activation in the hippocampus was quantified using Cx3cr1(GFP/+) mice. RESULTS Lipopolysaccharide priming increased susceptibility of rats and mice to hyperthemic seizures and enhanced seizure-induced proinflammatory cytokine production and microglial activation. CONCLUSIONS Peripheral inflammation appears to work synergistically with hyperthermia to potentiate seizures and to exacerbate seizure-induced immune responses. By simulating fever, a regulated increase in body temperature from an immune challenge, we developed a more clinically relevant animal model of prolonged FS.
Collapse
Affiliation(s)
- Baik-Lin Eun
- Department of Pediatrics, Korea University College of Medicine Seoul, Korea
| | - Jayne Abraham
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute, Northwestern University Feinberg School of Medicine Chicago, Illinois
| | - Lauren Mlsna
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute, Northwestern University Feinberg School of Medicine Chicago, Illinois
| | - Min Jung Kim
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute, Northwestern University Feinberg School of Medicine Chicago, Illinois
| | - Sookyong Koh
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago Stanley Manne Children's Research Institute, Northwestern University Feinberg School of Medicine Chicago, Illinois
| |
Collapse
|
40
|
Abstract
L-type calcium channels are present in most electrically excitable cells and are needed for proper brain, muscle, endocrine and sensory function. There is accumulating evidence for their involvement in brain diseases such as Parkinson disease, febrile seizures and neuropsychiatric disorders. Pharmacological inhibition of brain L-type channel isoforms, Cav1.2 and Cav1.3, may therefore be of therapeutic value. Organic calcium channels blockers are clinically used since decades for the treatment of hypertension, cardiac ischemia, and arrhythmias with a well-known and excellent safety profile. This pharmacological benefit is mainly mediated by the inhibition of Cav1.2 channels in the cardiovascular system. Despite their different biophysical properties and physiological functions, both brain channel isoforms are similarly inhibited by existing calcium channel blockers. In this review we will discuss evidence for altered L-type channel activity in human brain pathologies, new therapeutic implications of existing blockers and the rationale and current efforts to develop Cav1.3-selective compounds.
Collapse
Affiliation(s)
- Nadine J Ortner
- a Department of Pharmacology and Toxicology ; Center for Molecular Biosciences ; University of Innsbruck ; Innsbruck , Austria
| | - Jörg Striessnig
- a Department of Pharmacology and Toxicology ; Center for Molecular Biosciences ; University of Innsbruck ; Innsbruck , Austria
| |
Collapse
|
41
|
Moreira-Filho CA, Bando SY, Bertonha FB, Iamashita P, Silva FN, Costa LDF, Silva AV, Castro LHM, Wen HT. Community structure analysis of transcriptional networks reveals distinct molecular pathways for early- and late-onset temporal lobe epilepsy with childhood febrile seizures. PLoS One 2015; 10:e0128174. [PMID: 26011637 PMCID: PMC4444281 DOI: 10.1371/journal.pone.0128174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/24/2015] [Indexed: 12/21/2022] Open
Abstract
Age at epilepsy onset has a broad impact on brain plasticity and epilepsy pathomechanisms. Prolonged febrile seizures in early childhood (FS) constitute an initial precipitating insult (IPI) commonly associated with mesial temporal lobe epilepsy (MTLE). FS-MTLE patients may have early disease onset, i.e. just after the IPI, in early childhood, or late-onset, ranging from mid-adolescence to early adult life. The mechanisms governing early (E) or late (L) disease onset are largely unknown. In order to unveil the molecular pathways underlying E and L subtypes of FS-MTLE we investigated global gene expression in hippocampal CA3 explants of FS-MTLE patients submitted to hippocampectomy. Gene coexpression networks (GCNs) were obtained for the E and L patient groups. A network-based approach for GCN analysis was employed allowing: i) the visualization and analysis of differentially expressed (DE) and complete (CO) - all valid GO annotated transcripts - GCNs for the E and L groups; ii) the study of interactions between all the system's constituents based on community detection and coarse-grained community structure methods. We found that the E-DE communities with strongest connection weights harbor highly connected genes mainly related to neural excitability and febrile seizures, whereas in L-DE communities these genes are not only involved in network excitability but also playing roles in other epilepsy-related processes. Inversely, in E-CO the strongly connected communities are related to compensatory pathways (seizure inhibition, neuronal survival and responses to stress conditions) while in L-CO these communities harbor several genes related to pro-epileptic effects, seizure-related mechanisms and vulnerability to epilepsy. These results fit the concept, based on fMRI and behavioral studies, that early onset epilepsies, although impacting more severely the hippocampus, are associated to compensatory mechanisms, while in late MTLE development the brain is less able to generate adaptive mechanisms, what has implications for epilepsy management and drug discovery.
Collapse
Affiliation(s)
| | - Silvia Yumi Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Fernanda Bernardi Bertonha
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Priscila Iamashita
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | | | | | | | - Luiz Henrique Martins Castro
- Department of Neurology, FMUSP, São Paulo, SP, Brazil
- Clinical Neurology Division, Hospital das Clínicas, FMUSP, São Paulo, SP, Brazil
| | - Hung-Tzu Wen
- Epilepsy Surgery Group, Hospital das Clínicas, FMUSP, São Paulo, SP, Brazil
| |
Collapse
|
42
|
N'Gouemo P, Akinfiresoye LR, Allard JS, Lovinger DM. Alcohol Withdrawal-Induced Seizure Susceptibility is Associated with an Upregulation of CaV1.3 Channels in the Rat Inferior Colliculus. Int J Neuropsychopharmacol 2015; 18:pyu123. [PMID: 25556199 PMCID: PMC4458366 DOI: 10.1093/ijnp/pyu123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/30/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND We previously reported increased current density through L-type voltage-gated Ca(2+) (CaV1) channels in inferior colliculus (IC) neurons during alcohol withdrawal. However, the molecular correlate of this increased CaV1 current is currently unknown. METHODS Rats received three daily doses of ethanol every 8 hours for 4 consecutive days; control rats received vehicle. The IC was dissected at various time intervals following alcohol withdrawal, and the mRNA and protein levels of the CaV1.3 and CaV1.2 α1 subunits were measured. In separate experiments, rats were tested for their susceptibility to alcohol withdrawal-induced seizures (AWS) 3, 24, and 48 hours after alcohol withdrawal. RESULTS In the alcohol-treated group, AWS were observed 24 hours after withdrawal; no seizures were observed at 3 or 48 hours. No seizures were observed at any time in the control-treated rats. Compared to control-treated rats, the mRNA level of the CaV1.3 α1 subunit was increased 1.4-fold, 1.9-fold, and 1.3-fold at 3, 24, and 48 hours, respectively. In contrast, the mRNA level of the CaV1.2 α1 subunit increased 1.5-fold and 1.4-fold at 24 and 48 hours, respectively. At 24 hours, Western blot analyses revealed that the levels of the CaV1.3 and CaV1.2 α1 subunits increased by 52% and 32%, respectively, 24 hours after alcohol withdrawal. In contrast, the CaV1.2 and CaV1.3 α1 subunits were not altered at either 3 or 48 hours during alcohol withdrawal. CONCLUSIONS Expression of the CaV1.3 α1 subunit increased in parallel with AWS development, suggesting that altered L-type CaV1.3 channel expression is an important feature of AWS pathogenesis.
Collapse
Affiliation(s)
- Prosper N'Gouemo
- Georgetown University Medical Center, Department of Pediatrics, Washington DC (Drs N'Gouemo and Akinfiesoye); Howard University College of Medicine, Department of Physiology & Biophysics, Washington DC (Dr Allard); Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD (Dr Lovinger).
| | - Luli R Akinfiresoye
- Georgetown University Medical Center, Department of Pediatrics, Washington DC (Drs N'Gouemo and Akinfiesoye); Howard University College of Medicine, Department of Physiology & Biophysics, Washington DC (Dr Allard); Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD (Dr Lovinger)
| | - Joanne S Allard
- Georgetown University Medical Center, Department of Pediatrics, Washington DC (Drs N'Gouemo and Akinfiesoye); Howard University College of Medicine, Department of Physiology & Biophysics, Washington DC (Dr Allard); Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD (Dr Lovinger)
| | - David M Lovinger
- Georgetown University Medical Center, Department of Pediatrics, Washington DC (Drs N'Gouemo and Akinfiesoye); Howard University College of Medicine, Department of Physiology & Biophysics, Washington DC (Dr Allard); Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD (Dr Lovinger)
| |
Collapse
|
43
|
Feng B, Tang YS, Chen B, Dai YJ, Xu CL, Xu ZH, Zhang XN, Zhang SH, Hu WW, Chen Z. Dysfunction of thermoregulation contributes to the generation of hyperthermia-induced seizures. Neurosci Lett 2014; 581:129-34. [PMID: 25172570 DOI: 10.1016/j.neulet.2014.08.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/31/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022]
Abstract
Febrile seizures (FS) are generally defined as seizures taking place during fever. Long-term prognosis, including development of epilepsy and malformation of cognitive function, has been demonstrated after infantile FS. However, the mechanism that triggers seizures in hyperthermic environment is still unclear. We here found that the body temperature of rat pups that experienced experimental FS was markedly decreased (∼28°C) after they were removed from the hyperthermic environment. Both the seizure generation and the temperature drop after seizure attack were abolished by either pre-treatment with chlorpromazine (CPZ), which impairs the thermoregulation, or by an electrolytic lesion of the preoptic area and anterior hypothalamus (PO/AH). However, the non-steroidal anti-inflammatory drug celecoxib did not affect the seizure incidence and the decrease in body temperature after seizure attack. In addition, pentobarbital prevented the generation of seizures, but did not reverse the decrease of body temperature after FS. Therefore, our work indicates that an over-regulation of body temperature occurs during hyperthermic environment, and that the dysfunction of thermoregulation in the PO/AH following hyperthermia contributes to the generation of FS.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang-Shun Tang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bin Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yun-Jian Dai
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ceng-Lin Xu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zheng-Hao Xu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiang-Nan Zhang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shi-Hong Zhang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei-Wei Hu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
44
|
Hasreiter J, Goldnagl L, Böhm S, Kubista H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am J Physiol Cell Physiol 2014; 306:C1200-13. [PMID: 24760982 DOI: 10.1152/ajpcell.00329.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the central nervous system, L-type voltage-gated calcium channels (LTCCs) come in two isoforms, namely Cav1.2 and Cav1.3 channels. It has been shown previously that these channels differ in biophysical properties, in subcellular localization, and in the coupling to the gene transcription machinery. In previous work on rat hippocampal neurons we have identified an excitatory cation conductance and an inhibitory potassium conductance as important LTCC coupling partners. Notably, a stimulus-dependent interplay of LTCC-mediated Ca(2+) influx and activation of these Ca(2+)-dependent conductances was found to give rise to characteristic voltage responses. However, the contribution of Cav1.2 and Cav1.3 to these voltage responses remained unknown. Hence, the relative contribution of the LTCC isoforms therein was the focus of the current study on hippocampal neurons derived from genetically modified mice, which either lack a LTCC isoform (Cav1.3 knockout mice) or express a dihydropyridine-insensitive LTCC isoform (Cav1.2DHP(-)-knockin mice). We identified common and alternate ion channel couplings of Cav1.2 and Cav1.3 channels. Whereas hyperpolarizing Ca(2+)-dependent conductances were coupled to both Cav1.2 and Cav1.3 channels, an afterdepolarizing potential was only induced by the activity of Cav1.2 channels. Unexpectedly, the activity of Cav1.2 channels was found at relatively hyperpolarized membrane voltages. Our data add important information about the differences between Cav1.2 and Cav1.3 channels that furthers our understanding of the physiological and pathophysiological neuronal roles of these calcium channels. Moreover, our findings suggest that Cav1.3 knockout mice together with Cav1.2DHP(-)-knockin mice provide valuable models for future investigation of hippocampal LTCC-dependent afterdepolarizations.
Collapse
Affiliation(s)
- Julia Hasreiter
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lena Goldnagl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Böhm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
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
|
46
|
Murphy GG. The Heat is On: L-type Calcium Channels and Febrile Seizures. Epilepsy Curr 2014; 14:93-4. [PMID: 24872789 PMCID: PMC4010887 DOI: 10.5698/1535-7597-14.2.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
|
47
|
Persistent sodium current drives conditional pacemaking in CA1 pyramidal neurons under muscarinic stimulation. J Neurosci 2013; 33:15011-21. [PMID: 24048831 DOI: 10.1523/jneurosci.0577-13.2013] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hippocampal CA1 pyramidal neurons are normally quiescent but can fire spontaneously when stimulated by muscarinic agonists. In brain slice recordings from mouse CA1 pyramidal neurons, we examined the ionic basis of this activity using interleaved current-clamp and voltage-clamp experiments. Both in control and after muscarinic stimulation, the steady-state current-voltage curve was dominated by inward TTX-sensitive persistent sodium current (I(NaP)) that activated near -75 mV and increased steeply with depolarization. In control, total membrane current was net outward (hyperpolarizing) near -70 mV so that cells had a stable resting potential. Muscarinic stimulation activated a small nonselective cation current so that total membrane current near -70 mV shifted to become barely net inward (depolarizing). The small depolarization triggers regenerative activation of I(NaP), which then depolarizes the cell from -70 mV to spike threshold. We quantified the relative contributions of I(NaP), hyperpolarization-activated cation current (I(h)), and calcium current to pacemaking by using the cell's own firing as a voltage command along with specific blockers. TTX-sensitive sodium current was substantial throughout the entire interspike interval, increasing as the membrane potential approached threshold, while both Ih and calcium current were minimal. Thus, spontaneous activity is driven primarily by activation of I(NaP) in a positive feedback loop starting near -70 mV and providing increasing inward current to threshold. These results show that the pacemaking "engine" from I(NaP) is an inherent property of CA1 pyramidal neurons that can be engaged or disengaged by small shifts in net membrane current near -70 mV, as by muscarinic stimulation.
Collapse
|