1
|
Pastor-Alonso O, Durá I, Bernardo-Castro S, Varea E, Muro-García T, Martín-Suárez S, Encinas-Pérez JM, Pineda JR. HB-EGF activates EGFR to induce reactive neural stem cells in the mouse hippocampus after seizures. Life Sci Alliance 2024; 7:e202201840. [PMID: 38977310 PMCID: PMC11231495 DOI: 10.26508/lsa.202201840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Hippocampal seizures mimicking mesial temporal lobe epilepsy cause a profound disruption of the adult neurogenic niche in mice. Seizures provoke neural stem cells to switch to a reactive phenotype (reactive neural stem cells, React-NSCs) characterized by multibranched hypertrophic morphology, massive activation to enter mitosis, symmetric division, and final differentiation into reactive astrocytes. As a result, neurogenesis is chronically impaired. Here, using a mouse model of mesial temporal lobe epilepsy, we show that the epidermal growth factor receptor (EGFR) signaling pathway is key for the induction of React-NSCs and that its inhibition exerts a beneficial effect on the neurogenic niche. We show that during the initial days after the induction of seizures by a single intrahippocampal injection of kainic acid, a strong release of zinc and heparin-binding epidermal growth factor, both activators of the EGFR signaling pathway in neural stem cells, is produced. Administration of the EGFR inhibitor gefitinib, a chemotherapeutic in clinical phase IV, prevents the induction of React-NSCs and preserves neurogenesis.
Collapse
Affiliation(s)
- Oier Pastor-Alonso
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Irene Durá
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Sara Bernardo-Castro
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Emilio Varea
- Faculty of Biology, University of Valencia, Valencia, Spain
| | - Teresa Muro-García
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Soraya Martín-Suárez
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Juan Manuel Encinas-Pérez
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
- Ikerbasque, The Basque Foundation for Science, Bizkaia, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Bizkaia, Spain
| | - Jose Ramon Pineda
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
- Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bizkaia, Spain
| |
Collapse
|
2
|
Hong R, Zheng T, Marra V, Yang D, Liu JK. Multi-scale modelling of the epileptic brain: advantages of computational therapy exploration. J Neural Eng 2024; 21:021002. [PMID: 38621378 DOI: 10.1088/1741-2552/ad3eb4] [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: 08/29/2023] [Accepted: 04/15/2024] [Indexed: 04/17/2024]
Abstract
Objective: Epilepsy is a complex disease spanning across multiple scales, from ion channels in neurons to neuronal circuits across the entire brain. Over the past decades, computational models have been used to describe the pathophysiological activity of the epileptic brain from different aspects. Traditionally, each computational model can aid in optimizing therapeutic interventions, therefore, providing a particular view to design strategies for treating epilepsy. As a result, most studies are concerned with generating specific models of the epileptic brain that can help us understand the certain machinery of the pathological state. Those specific models vary in complexity and biological accuracy, with system-level models often lacking biological details.Approach: Here, we review various types of computational model of epilepsy and discuss their potential for different therapeutic approaches and scenarios, including drug discovery, surgical strategies, brain stimulation, and seizure prediction. We propose that we need to consider an integrated approach with a unified modelling framework across multiple scales to understand the epileptic brain. Our proposal is based on the recent increase in computational power, which has opened up the possibility of unifying those specific epileptic models into simulations with an unprecedented level of detail.Main results: A multi-scale epilepsy model can bridge the gap between biologically detailed models, used to address molecular and cellular questions, and brain-wide models based on abstract models which can account for complex neurological and behavioural observations.Significance: With these efforts, we move toward the next generation of epileptic brain models capable of connecting cellular features, such as ion channel properties, with standard clinical measures such as seizure severity.
Collapse
Affiliation(s)
- Rongqi Hong
- School of Computer Science, Centre for Human Brain Health, University of Birmingham, Birmingham, United Kingdom
| | - Tingting Zheng
- School of Computer Science, Centre for Human Brain Health, University of Birmingham, Birmingham, United Kingdom
| | | | - Dongping Yang
- Research Centre for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| | - Jian K Liu
- School of Computer Science, Centre for Human Brain Health, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Vasilieva AA, Timechko EE, Lysova KD, Paramonova AI, Yakimov AM, Kantimirova EA, Dmitrenko DV. MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review. Int J Mol Sci 2023; 24:15366. [PMID: 37895044 PMCID: PMC10607802 DOI: 10.3390/ijms242015366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Structural or post-traumatic epilepsy often develops after brain tissue damage caused by traumatic brain injury, stroke, infectious diseases of the brain, etc. Most often, between the initiating event and epilepsy, there is a period without seizures-a latent period. At this time, the process of restructuring of neural networks begins, leading to the formation of epileptiform activity, called epileptogenesis. The prediction of the development of the epileptogenic process is currently an urgent and difficult task. MicroRNAs are inexpensive and minimally invasive biomarkers of biological and pathological processes. The aim of this study is to evaluate the predictive ability of microRNAs to detect the risk of epileptogenesis. In this study, we conducted a systematic search on the MDPI, PubMed, ScienceDirect, and Web of Science platforms. We analyzed publications that studied the aberrant expression of circulating microRNAs in epilepsy, traumatic brain injury, and ischemic stroke in order to search for microRNAs-potential biomarkers for predicting epileptogenesis. Thus, 31 manuscripts examining biomarkers of epilepsy, 19 manuscripts examining biomarkers of traumatic brain injury, and 48 manuscripts examining biomarkers of ischemic stroke based on circulating miRNAs were analyzed. Three miRNAs were studied: miR-21, miR-181a, and miR-155. The findings showed that miR-21 and miR-155 are associated with cell proliferation and apoptosis, and miR-181a is associated with protein modifications. These miRNAs are not strictly specific, but they are involved in processes that may be indirectly associated with epileptogenesis. Also, these microRNAs may be of interest when they are studied in a cohort with each other and with other microRNAs. To further study the microRNA-based biomarkers of epileptogenesis, many factors must be taken into account: the time of sampling, the type of biological fluid, and other nuances. Currently, there is a need for more in-depth and prolonged studies of epileptogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Diana V. Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia; (A.A.V.); (E.E.T.); (K.D.L.); (A.I.P.)
| |
Collapse
|
4
|
Călin A, Waseem T, Raimondo JV, Newey SE, Akerman CJ. A genetically targeted ion sensor reveals distinct seizure-related chloride and pH dynamics in GABAergic interneuron populations. iScience 2023; 26:106363. [PMID: 37034992 PMCID: PMC10074576 DOI: 10.1016/j.isci.2023.106363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/03/2023] [Accepted: 03/05/2023] [Indexed: 03/12/2023] Open
Abstract
Intracellular chloride and pH play fundamental roles in determining a neuron's synaptic inhibition and excitability. Yet it has been difficult to measure changes in these ions during periods of heightened network activity, such as occur in epilepsy. Here we develop a version of the fluorescent reporter, ClopHensorN, to enable simultaneous quantification of chloride and pH in genetically defined neurons during epileptiform activity. We compare pyramidal neurons to the major GABAergic interneuron subtypes in the mouse hippocampus, which express parvalbumin (PV), somatostatin (SST), or vasoactive intestinal polypeptide (VIP). Interneuron populations exhibit higher baseline chloride, with PV interneurons exhibiting the highest levels. During an epileptiform discharge, however, all subtypes converge upon a common elevated chloride level. Concurrent with these dynamics, epileptiform activity leads to different degrees of intracellular acidification, which reflect baseline pH. Thus, a new optical tool for dissociating chloride and pH reveals neuron-specific ion dynamics during heightened network activity.
Collapse
Affiliation(s)
- Alexandru Călin
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Tatiana Waseem
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Joseph V. Raimondo
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Sarah E. Newey
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Colin J. Akerman
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
5
|
İMDAT NN, ÇİLİNGİR-KAYA ÖT, TURGAN ÂŞIK ZN, KARAMAHMUTOĞLU T, GÜLÇEBİ İDRİZ OĞLU M, AKAKIN D, ONAT F, ŞİRVANCI S. Electron microscopic GABA evaluation in hippocampal mossy terminals of genetic absence epilepsy rats receiving kindling stimulations. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2022. [DOI: 10.33808/clinexphealthsci.1030132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Objective: The hypotheses related to the fact of epileptic mechanisms are mainly based on excitation-inhibition imbalance in central nervous system. GAERS (Genetic Absence Epilepsy Rats from Strasbourg) is a well-known animal model of absence epilepsy, and frequently used in experimental studies. In the present study, we aimed to examine possible morphological and gamma-aminobutyric acid (GABA) density changes in GAERS hippocampus after electrical kindling stimulations.
Methods: All control and test group rats received 6 kindling stimulations. Rats were decapitated 1 h after the last stimulation. Ultrastructural GABA immunocytochemistry was used to evaluate GABA density quantitatively in mossy terminals of hippocampal CA3 region.
Results: GABA levels were less in kindling groups compared to their controls, and in GAERS groups compared to Wistar groups; mitochondrial and dendritic spine area ratios were greater in GAERS groups compared to Wistar groups, although all these evaluations were statistically nonsignificant. Depletion of synaptic vesicles was evident in the mossy terminals of kindling groups.
Conclusion: The reason of decreased levels of GABA found in the present study might be that GABA has been released from the synaptic pool rapidly at an early time period after the last stimulation, for compansation mechanisms. Depletion of synaptic vesicles observed in kindling groups shows that even 6 kindling stimulations have an impact of changing hippocampal morphology in trisynaptic cycle. The increased mitochondrial area in GAERS might be related to the increased mitochondrial activity. The increased dendritic spine area might be related to the increased performance of learning in GAERS. Our findings indicating that absence epilepsy and temporal lobe epilepsy have different mechanisms of epileptogenesis might be a basis for further experimental studies
Collapse
|
6
|
DeFelipe J, DeFelipe-Oroquieta J, Furcila D, Muñoz-Alegre M, Maestú F, Sola RG, Blázquez-Llorca L, Armañanzas R, Kastanaskaute A, Alonso-Nanclares L, Rockland KS, Arellano JI. Neuroanatomical and psychological considerations in temporal lobe epilepsy. Front Neuroanat 2022; 16:995286. [PMID: 36590377 PMCID: PMC9794593 DOI: 10.3389/fnana.2022.995286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/11/2022] [Indexed: 01/03/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common form of focal epilepsy and is associated with a variety of structural and psychological alterations. Recently, there has been renewed interest in using brain tissue resected during epilepsy surgery, in particular 'non-epileptic' brain samples with normal histology that can be found alongside epileptic tissue in the same epileptic patients - with the aim being to study the normal human brain organization using a variety of methods. An important limitation is that different medical characteristics of the patients may modify the brain tissue. Thus, to better determine how 'normal' the resected tissue is, it is fundamental to know certain clinical, anatomical and psychological characteristics of the patients. Unfortunately, this information is frequently not fully available for the patient from which the resected tissue has been obtained - or is not fully appreciated by the neuroscientists analyzing the brain samples, who are not necessarily experts in epilepsy. In order to present the full picture of TLE in a way that would be accessible to multiple communities (e.g., basic researchers in neuroscience, neurologists, neurosurgeons and psychologists), we have reviewed 34 TLE patients, who were selected due to the availability of detailed clinical, anatomical, and psychological information for each of the patients. Our aim was to convey the full complexity of the disorder, its putative anatomical substrates, and the wide range of individual variability, with a view toward: (1) emphasizing the importance of considering critical patient information when using brain samples for basic research and (2) gaining a better understanding of normal and abnormal brain functioning. In agreement with a large number of previous reports, this study (1) reinforces the notion of substantial individual variability among epileptic patients, and (2) highlights the common but overlooked psychopathological alterations that occur even in patients who become "seizure-free" after surgery. The first point is based on pre- and post-surgical comparisons of patients with hippocampal sclerosis and patients with normal-looking hippocampus in neuropsychological evaluations. The second emerges from our extensive battery of personality and projective tests, in a two-way comparison of these two types of patients with regard to pre- and post-surgical performance.
Collapse
Affiliation(s)
- Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain,Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain,*Correspondence: Javier DeFelipe,
| | - Jesús DeFelipe-Oroquieta
- Gerencia Asistencial de Atención Primaria, Servicio Madrileño de Salud, Madrid, Spain,Facultad de Educación, Universidad Camilo José Cela, Madrid, Spain
| | - Diana Furcila
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Mar Muñoz-Alegre
- Facultad de Educación y Psicología, Universidad Francisco de Vitoria, Madrid, Spain
| | - Fernando Maestú
- Department of Experimental Psychology, Complutense University of Madrid, Madrid, Spain,Center for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid, Spain
| | - Rafael G. Sola
- Cátedra UAM de “Innovación en Neurocirugía”, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lidia Blázquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain,Sección Departamental de Anatomía y Embriología, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Armañanzas
- Institute of Data Science and Artificial Intelligence, Universidad de Navarra, Pamplona, Spain,Tecnun School of Engineering, Universidad de Navarra, Donostia-San Sebastian, Spain
| | - Asta Kastanaskaute
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain,Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Lidia Alonso-Nanclares
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Madrid, Spain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain,Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Kathleen S. Rockland
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jon I. Arellano
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
7
|
Erisken S, Nune G, Chung H, Kang JW, Koh S. Time and age dependent regulation of neuroinflammation in a rat model of mesial temporal lobe epilepsy: Correlation with human data. Front Cell Dev Biol 2022; 10:969364. [PMID: 36172274 PMCID: PMC9512631 DOI: 10.3389/fcell.2022.969364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Acute brain insults trigger diverse cellular and signaling responses and often precipitate epilepsy. The cellular, molecular and signaling events relevant to the emergence of the epileptic brain, however, remain poorly understood. These multiplex structural and functional alterations tend also to be opposing - some homeostatic and reparative while others disruptive; some associated with growth and proliferation while others, with cell death. To differentiate pathological from protective consequences, we compared seizure-induced changes in gene expression hours and days following kainic acid (KA)-induced status epilepticus (SE) in postnatal day (P) 30 and P15 rats by capitalizing on age-dependent differential physiologic responses to KA-SE; only mature rats, not immature rats, have been shown to develop spontaneous recurrent seizures after KA-SE. To correlate gene expression profiles in epileptic rats with epilepsy patients and demonstrate the clinical relevance of our findings, we performed gene analysis on four patient samples obtained from temporal lobectomy and compared to four control brains from NICHD Brain Bank. Pro-inflammatory gene expressions were at higher magnitudes and more sustained in P30. The inflammatory response was driven by the cytokines IL-1β, IL-6, and IL-18 in the acute period up to 72 h and by IL-18 in the subacute period through the 10-day time point. In addition, a panoply of other immune system genes was upregulated, including chemokines, glia markers and adhesion molecules. Genes associated with the mitogen activated protein kinase (MAPK) pathways comprised the largest functional group identified. Through the integration of multiple ontological databases, we analyzed genes belonging to 13 separate pathways linked to Classical MAPK ERK, as well as stress activated protein kinases (SAPKs) p38 and JNK. Interestingly, genes belonging to the Classical MAPK pathways were mostly transiently activated within the first 24 h, while genes in the SAPK pathways had divergent time courses of expression, showing sustained activation only in P30. Genes in P30 also had different regulatory functions than in P15: P30 animals showed marked increases in positive regulators of transcription, of signaling pathways as well as of MAPKKK cascades. Many of the same inflammation-related genes as in epileptic rats were significantly upregulated in human hippocampus, higher than in lateral temporal neocortex. They included glia-associated genes, cytokines, chemokines and adhesion molecules and MAPK pathway genes. Uniquely expressed in human hippocampus were adaptive immune system genes including immune receptors CDs and MHC II HLAs. In the brain, many immune molecules have additional roles in synaptic plasticity and the promotion of neurite outgrowth. We propose that persistent changes in inflammatory gene expression after SE leads not only to structural damage but also to aberrant synaptogenesis that may lead to epileptogenesis. Furthermore, the sustained pattern of inflammatory genes upregulated in the epileptic mature brain was distinct from that of the immature brain that show transient changes and are resistant to cell death and neuropathologic changes. Our data suggest that the epileptogenic process may be a result of failed cellular signaling mechanisms, where insults overwhelm the system beyond a homeostatic threshold.
Collapse
Affiliation(s)
- Sinem Erisken
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, United States
| | - George Nune
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Neurology, University of Southern California, Los Angeles, CA, United States
| | - Hyokwon Chung
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
| | - Joon Won Kang
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
- Department of Pediatrics & Medical Science, Brain Research Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Sookyong Koh
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
- *Correspondence: Sookyong Koh,
| |
Collapse
|
8
|
Adenosine A 2A receptors control synaptic remodeling in the adult brain. Sci Rep 2022; 12:14690. [PMID: 36038626 PMCID: PMC9424208 DOI: 10.1038/s41598-022-18884-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/22/2022] [Indexed: 01/04/2023] Open
Abstract
The molecular mechanisms underlying circuit re-wiring in the mature brain remains ill-defined. An eloquent example of adult circuit remodelling is the hippocampal mossy fiber (MF) sprouting found in diseases such as temporal lobe epilepsy. The molecular determinants underlying this retrograde re-wiring remain unclear. This may involve signaling system(s) controlling axon specification/growth during neurodevelopment reactivated during epileptogenesis. Since adenosine A2A receptors (A2AR) control axon formation/outgrowth and synapse stabilization during development, we now examined the contribution of A2AR to MF sprouting. A2AR blockade significantly attenuated status epilepticus(SE)-induced MF sprouting in a rat pilocarpine model. This involves A2AR located in dentate granule cells since their knockdown selectively in dentate granule cells reduced MF sprouting, most likely through the ability of A2AR to induce the formation/outgrowth of abnormal secondary axons found in rat hippocampal neurons. These A2AR should be activated by extracellular ATP-derived adenosine since a similar prevention/attenuation of SE-induced hippocampal MF sprouting was observed in CD73 knockout mice. These findings demonstrate that A2AR contribute to epilepsy-related MF sprouting, most likely through the reactivation of the ability of A2AR to control axon formation/outgrowth observed during neurodevelopment. These results frame the CD73-A2AR axis as a regulator of circuit remodeling in the mature brain.
Collapse
|
9
|
Molecular Mechanism and Regulation of Autophagy and Its Potential Role in Epilepsy. Cells 2022; 11:cells11172621. [PMID: 36078029 PMCID: PMC9455075 DOI: 10.3390/cells11172621] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an evolutionally conserved degradation mechanism for maintaining cell homeostasis whereby cytoplasmic components are wrapped in autophagosomes and subsequently delivered to lysosomes for degradation. This process requires the concerted actions of multiple autophagy-related proteins and accessory regulators. In neurons, autophagy is dynamically regulated in different compartments including soma, axons, and dendrites. It determines the turnover of selected materials in a spatiotemporal control manner, which facilitates the formation of specialized neuronal functions. It is not surprising, therefore, that dysfunctional autophagy occurs in epilepsy, mainly caused by an imbalance between excitation and inhibition in the brain. In recent years, much attention has been focused on how autophagy may cause the development of epilepsy. In this article, we overview the historical landmarks and distinct types of autophagy, recent progress in the core machinery and regulation of autophagy, and biological roles of autophagy in homeostatic maintenance of neuronal structures and functions, with a particular focus on synaptic plasticity. We also discuss the relevance of autophagy mechanisms to the pathophysiology of epileptogenesis.
Collapse
|
10
|
Benassi SK, Alves JGSM, Guidoreni CG, Massant CG, Queiroz CM, Garrido-Sanabria E, Loduca RDDS, Susemihl MA, Paiva WS, de Andrade AF, Teixeira MJ, Andrade JQ, Garzon E, Foresti ML, Mello LE. Two decades of research towards a potential first anti-epileptic drug. Seizure 2021; 90:99-109. [DOI: 10.1016/j.seizure.2021.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/20/2021] [Accepted: 02/26/2021] [Indexed: 11/27/2022] Open
|
11
|
do Canto AM, Donatti A, Geraldis JC, Godoi AB, da Rosa DC, Lopes-Cendes I. Neuroproteomics in Epilepsy: What Do We Know so Far? Front Mol Neurosci 2021; 13:604158. [PMID: 33488359 PMCID: PMC7817846 DOI: 10.3389/fnmol.2020.604158] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Epilepsies are chronic neurological diseases that affect approximately 2% of the world population. In addition to being one of the most frequent neurological disorders, treatment for patients with epilepsy remains a challenge, because a proportion of patients do not respond to the antiseizure medications that are currently available. This results in a severe economic and social burden for patients, families, and the healthcare system. A characteristic common to all forms of epilepsy is the occurrence of epileptic seizures that are caused by abnormal neuronal discharges, leading to a clinical manifestation that is dependent on the affected brain region. It is generally accepted that an imbalance between neuronal excitation and inhibition generates the synchronic electrical activity leading to seizures. However, it is still unclear how a normal neural circuit becomes susceptible to the generation of seizures or how epileptogenesis is induced. Herein, we review the results of recent proteomic studies applied to investigate the underlying mechanisms leading to epilepsies and how these findings may impact research and treatment for these disorders.
Collapse
Affiliation(s)
- Amanda M. do Canto
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Amanda Donatti
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Jaqueline C. Geraldis
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Alexandre B. Godoi
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Douglas C. da Rosa
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| |
Collapse
|
12
|
Carstens KE, Gloss BR, Alexander GM, Dudek SM. Modified adeno-associated virus targets the bacterial enzyme chondroitinase ABC to select mouse neuronal populations in vivo using the Cre-LoxP system. Eur J Neurosci 2020; 53:4005-4015. [PMID: 33220084 PMCID: PMC8137719 DOI: 10.1111/ejn.15050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/01/2023]
Abstract
Current methods of experimentally degrading the specialized extracellular matrix (ECM), perineuronal nets (PNNs) have several limitations. Genetic knockout of ECM components typically has only partial effects on PNNs, and knockout of the major ECM component aggrecan is lethal in mice. Direct injection of the chondroitinase ABC (ChABC) enzyme into the mammalian brain is effective at degrading PNNs in vivo but this method typically lacks consistent, localized spatial targeting of PNN degradation. PNNs also regenerate within weeks after a ChABC injection, thus limiting the ability to perform long‐term studies. Previous work has demonstrated that viral delivery of ChABC in mammalian neurons can successfully degrade PNNs for much longer periods, but the effects are similarly diffuse beyond the injection site. In an effort to gain cell‐specific targeting of ChABC, we designed an adeno‐associated virus encoding ChABC under the control of the Cre‐LoxP system. We show that this virus is effective at targeting the synthesis of ChABC to Cre‐expressing mouse neurons in vivo. Although ChABC expression is localized to the Cre‐expressing neurons, we also note that ChABC is apparently trafficked and secreted at projection sites, as was previously reported for the non‐Cre dependent construct. Overall, this method allows for cell‐specific targeting of ChABC and long‐term degradation of PNNs, which will ultimately serve as an effective tool to study the function of cell‐autonomous regulation of PNNs in vivo. This novel approach may also aid in determining whether specific, long‐term PNN loss is an appropriate strategy for treatment of neurodevelopmental disorders associated with PNN pathology.
Collapse
Affiliation(s)
- Kelly E Carstens
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Bernd R Gloss
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| |
Collapse
|
13
|
Cannabidiol anticonvulsant effect is mediated by the PI3Kγ pathway. Neuropharmacology 2020; 176:108156. [DOI: 10.1016/j.neuropharm.2020.108156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
|
14
|
Abstract
We aimed to explore the link between NREM sleep and epilepsy. Based on human and experimental data we propose that a sleep-related epileptic transformation of normal neurological networks underlies epileptogenesis. Major childhood epilepsies as medial temporal lobe epilepsy (MTLE), absence epilepsy (AE) and human perisylvian network (PN) epilepsies - made us good models to study. These conditions come from an epileptic transformation of the affected functional systems. This approach allows a system-based taxonomy instead of the outworn generalized-focal classification. MTLE links to the memory-system, where epileptic transformation results in a switch of normal sharp wave-ripples to epileptic spikes and pathological high frequency oscillations, compromising sleep-related memory consolidation. Absence epilepsy (AE) and juvenile myoclonic epilepsy (JME) belong to the corticothalamic system. The burst-firing mode of NREM sleep normally producing sleep-spindles turns to an epileptic working mode ejecting bilateral synchronous spike-waves. There seems to be a progressive transition from AE to JME. Shared absences and similar bilateral synchronous discharges show the belonging of the two conditions, while the continuous age windows - AE affecting schoolchildren, JME the adolescents - and the increased excitability in JME compared to AE supports the notion of progression. In perisylvian network epilepsies - idiopathic focal childhood epilepsies and electrical status epilepticus in sleep including Landau-Kleffner syndrome - centrotemporal spikes turn epileptic, with the potential to cause cognitive impairment. Postinjury epilepsies modeled by the isolated cortex model highlight the shared way of epileptogenesis suggesting the derailment of NREM sleep-related homeostatic plasticity as a common step. NREM sleep provides templates for plasticity derailing to epileptic variants under proper conditions. This sleep-origin explains epileptiform discharges' link and similarity with NREM sleep slow oscillations, spindles and ripples. Normal synaptic plasticity erroneously overgrowing homeostatic processes may derail toward an epileptic working-mode manifesting the involved system's features. The impact of NREM sleep is unclear in epileptogenesis occurring in adolescence and adulthood, when plasticity is lower. The epileptic process interferes with homeostatic synaptic plasticity and may cause cognitive impairment. Its type and degree depends on the affected network's function. We hypothesize a vicious circle between sleep end epilepsy. The epileptic derailment of normal plasticity interferes with sleep cognitive functions. Sleep and epilepsy interconnect by the pathology of plasticity.
Collapse
Affiliation(s)
- Péter Halász
- Szentágothai János School of Ph.D Studies, Clinical Neurosciences, Semmelweis University, Budapest, Hungary
| | - Anna Szűcs
- Institute of Behavioral Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Ahmed Juvale II, Che Has AT. The evolution of the pilocarpine animal model of status epilepticus. Heliyon 2020; 6:e04557. [PMID: 32775726 PMCID: PMC7393986 DOI: 10.1016/j.heliyon.2020.e04557] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/05/2020] [Accepted: 07/22/2020] [Indexed: 02/02/2023] Open
Abstract
The pilocarpine animal model of status epilepticus is a well-established, clinically translatable model that satisfies all of the criteria essential for an animal model of status epilepticus: a latency period followed by spontaneous recurrent seizures, replication of behavioural, electrographic, metabolic, and neuropathological changes, as well as, pharmacoresistance to anti-epileptic drugs similar to that observed in human status epilepticus. However, this model is also characterized by high mortality rates and studies in recent years have also seen difficulties in seizure induction due to pilocarpine resistant animals. This can be attributed to differences in rodent strains, species, gender, and the presence of the multi-transporter, P-glycoprotein at the blood brain barrier. The current paper highlights the various alterations made to the original pilocarpine model over the years to combat both the high mortality and low induction rates. These range from the initial lithium-pilocarpine model to the more recent Reduced Intensity Status Epilepticus (RISE) model, which finally brought the mortality rates down to 1%. These modifications are essential to improve animal welfare and future experimental outcomes.
Collapse
Affiliation(s)
- Iman Imtiyaz Ahmed Juvale
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Tarmizi Che Has
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Dombroski TCD, Peixoto-Santos JE, Maciel K, Baqui MMA, Velasco TR, Sakamoto AC, Assirati JA, Carlotti CG, Machado HR, Sousa GKD, Hanamura K, Leite JP, Costa da Costa J, Palmini AL, Paglioli E, Neder L, Spreafico R, Shirao T, Garbelli R, Martins AR. Drebrin expression patterns in patients with refractory temporal lobe epilepsy and hippocampal sclerosis. Epilepsia 2020; 61:1581-1594. [PMID: 32662890 DOI: 10.1111/epi.16595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Drebrins are crucial for synaptic function and dendritic spine development, remodeling, and maintenance. In temporal lobe epilepsy (TLE) patients, a significant hippocampal synaptic reorganization occurs, and synaptic reorganization has been associated with hippocampal hyperexcitability. This study aimed to evaluate, in TLE patients, the hippocampal expression of drebrin using immunohistochemistry with DAS2 or M2F6 antibodies that recognize adult (drebrin A) or adult and embryonic (pan-drebrin) isoforms, respectively. METHODS Hippocampal sections from drug-resistant TLE patients with hippocampal sclerosis (HS; TLE, n = 33), of whom 31 presented with type 1 HS and two with type 2 HS, and autopsy control cases (n = 20) were assayed by immunohistochemistry and evaluated for neuron density, and drebrin A and pan-drebrin expression. Double-labeling immunofluorescences were performed to localize drebrin A-positive spines in dendrites (MAP2), and to evaluate whether drebrin colocalizes with inhibitory (GAD65) and excitatory (VGlut1) presynaptic markers. RESULTS Compared to controls, TLE patients had increased pan-drebrin in all hippocampal subfields and increased drebrin A-immunopositive area in all hippocampal subfields but CA1. Drebrin-positive spine density followed the same pattern as total drebrin quantification. Confocal microscopy indicated juxtaposition of drebrin-positive spines with VGlut1-positive puncta, but not with GAD65-positive puncta. Drebrin expression in the dentate gyrus of TLE cases was associated negatively with seizure frequency and positively with verbal memory. TLE patients with lower drebrin-immunopositive area in inner molecular layer (IML) than in outer molecular layer (OML) had a lower seizure frequency than those with higher or comparable drebrin-immunopositive area in IML compared with OML. SIGNIFICANCE Our results suggest that changes in drebrin-positive spines and drebrin expression in the dentate gyrus of TLE patients are associated with lower seizure frequency, more preserved verbal memory, and a better postsurgical outcome.
Collapse
Affiliation(s)
| | - Jose Eduardo Peixoto-Santos
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Paulista Medical School, UNIFESP, São Paulo, Brazil
| | - Karina Maciel
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Munira Muhammad Abdel Baqui
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tonicarlo Rodrigues Velasco
- Ribeirao Preto Epilepsy Surgery Center, Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Americo Ceiki Sakamoto
- Ribeirao Preto Epilepsy Surgery Center, Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Alberto Assirati
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos Gilberto Carlotti
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Hélio Rubens Machado
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gleice Kelly de Sousa
- Graduate Program of Health Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - João Pereira Leite
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jaderson Costa da Costa
- Department of Internal Medicine, School of Medicine, Epilepsy Surgery Program and Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - André Luiz Palmini
- Department of Internal Medicine, School of Medicine, Epilepsy Surgery Program and Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Eliseu Paglioli
- Department of Surgery, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Roberto Spreafico
- Clinical Epileptology and Experimental Neurophysiology Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Rita Garbelli
- Clinical Epileptology and Experimental Neurophysiology Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Antonio Roberto Martins
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Institute for Neuroscience and Behavior, Ribeirão Preto, Brazil
| |
Collapse
|
17
|
Genetically Modified Mesenchymal Stem Cells: The Next Generation of Stem Cell-Based Therapy for TBI. Int J Mol Sci 2020; 21:ijms21114051. [PMID: 32516998 PMCID: PMC7312789 DOI: 10.3390/ijms21114051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are emerging as an attractive approach for restorative medicine in central nervous system (CNS) diseases and injuries, such as traumatic brain injury (TBI), due to their relatively easy derivation and therapeutic effect following transplantation. However, the long-term survival of the grafted cells and therapeutic efficacy need improvement. Here, we review the recent application of MSCs in TBI treatment in preclinical models. We discuss the genetic modification approaches designed to enhance the therapeutic potency of MSCs for TBI treatment by improving their survival after transplantation, enhancing their homing abilities and overexpressing neuroprotective and neuroregenerative factors. We highlight the latest preclinical studies that have used genetically modified MSCs for TBI treatment. The recent developments in MSCs’ biology and potential TBI therapeutic targets may sufficiently improve the genetic modification strategies for MSCs, potentially bringing effective MSC-based therapies for TBI treatment in humans.
Collapse
|
18
|
Chen Y, Feng Z, Shen M, Lin W, Wang Y, Wang S, Li C, Wang S, Chen M, Shan W, Xie XQ. Insight into Ginkgo biloba L. Extract on the Improved Spatial Learning and Memory by Chemogenomics Knowledgebase, Molecular Docking, Molecular Dynamics Simulation, and Bioassay Validations. ACS OMEGA 2020; 5:2428-2439. [PMID: 32064403 PMCID: PMC7017398 DOI: 10.1021/acsomega.9b03960] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/16/2020] [Indexed: 05/08/2023]
Abstract
Epilepsy is a common cause of serious cognitive disorders and is known to have impact on patients' memory and executive functions. Therefore, the development of antiepileptic drugs for the improvement of spatial learning and memory in patients with epileptic cognitive dysfunction is important. In the present work, we systematically predicted and analyzed the potential effects of Ginkgo terpene trilactones (GTTL) on cognition and pathologic changes utilizing in silico and in vivo approaches. Based on our established chemogenomics knowledgebase, we first conducted the network systems pharmacology analysis to predict that ginkgolide A/B/C may target 5-HT 1A, 5-HT 1B, and 5-HT 2B. The detailed interactions were then further validated by molecular docking and molecular dynamics (MD) simulations. In addition, status epilepticus (SE) was induced by lithium-pilocarpine injection in adult Wistar male rats, and the results of enzyme-linked immunosorbent assay (ELISA) demonstrated that administration with GTTL can increase the expression of brain-derived neurotrophic factor (BDNF) when compared to the model group. Interestingly, recent studies suggest that the occurrence of a reciprocal involvement of 5-HT receptor activation along with the hippocampal BDNF-increased expression can significantly ameliorate neurologic changes and reverse behavioral deficits in status epilepticus rats while improving cognitive function and alleviating neuronal injury. Therefore, we evaluated the effects of GTTL (bilobalide, ginkgolide A, ginkgolide B, and ginkgolide C) on synergistic antiepileptic effect. Our experimental data showed that the spatial learning and memory abilities (e.g., electroencephalography analysis and Morris water maze test for behavioral assessment) of rats administrated with GTTL were significantly improved under the middle dose (80 mg/kg, GTTL) and high dose (160 mg/kg, GTTL). Moreover, the number of neurons in the hippocampus of the GTTL group increased when compared to the model group. Our studies showed that GTTL not only protected rat cerebral hippocampal neurons against epilepsy but also improved the learning and memory ability. Therefore, GTTL may be a potential drug candidate for the prevention and/or treatment of epilepsy.
Collapse
Affiliation(s)
- Yan Chen
- College
of Pharmacology Sciences, Zhejiang University
of Technology, Hangzhou 310014, P. R. China
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Mingzhe Shen
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Weiwei Lin
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Yuanqiang Wang
- School of
Pharmacy and Bioengineering, Chongqing University
of Technology, Chongqing 400054, P. R. China
| | - Siyi Wang
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Caifeng Li
- College
of Pharmacology Sciences, Zhejiang University
of Technology, Hangzhou 310014, P. R. China
| | - Shengfeng Wang
- College
of Pharmacology Sciences, Zhejiang University
of Technology, Hangzhou 310014, P. R. China
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Weiguang Shan
- College
of Pharmacology Sciences, Zhejiang University
of Technology, Hangzhou 310014, P. R. China
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational
Chemical
Genomics Screening Center, School of Pharmacy, National Center of Excellence for
Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology
and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
19
|
Royero PX, Higa GSV, Kostecki DS, Dos Santos BA, Almeida C, Andrade KA, Kinjo ER, Kihara AH. Ryanodine receptors drive neuronal loss and regulate synaptic proteins during epileptogenesis. Exp Neurol 2020; 327:113213. [PMID: 31987836 DOI: 10.1016/j.expneurol.2020.113213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
Status epilepticus (SE) is a clinical emergency that can lead to the development of temporal lobe epilepsy (TLE). The development and maintenance of spontaneous seizures in TLE are linked to calcium (Ca+2)-dependent processes such as neuronal cell loss and pathological synaptic plasticity. It has been shown that SE produces an increase in ryanodine receptor-dependent intracellular Ca+2 levels in hippocampal neurons, which remain elevated during the progression of the disease. However, the participation of ryanodine receptors (RyRs) in the neuronal loss and circuitry rewiring that take place in the hippocampus after SE remains unknown. In this context, we first investigated the functional role of RyRs on the expression of synaptic and plasticity-related proteins during epileptogenesis induced by pilocarpine in Wistar rats. Intrahippocampal injection of dantrolene, a selective pharmacological blocker of RyRs, caused the increase of the presynaptic protein synapsin I (SYN) and synaptophysin (SYP) 48 h after SE induction. Specifically, we observed that SYN and SYP were regulated in hippocampal regions known to receive synaptic inputs, revealing that RyRs could be involved in network changes and/or neuronal protection after SE induction. In order to investigate whether the changes in SYN and SYP were related to neuroplastic changes that could contribute to pathological processes that occur after SE, we evaluated the levels of activity-regulated cytoskeleton-associated protein (ARC) and mossy fiber sprouting in the dentate gyrus (DG). Interestingly, we observed that although SE induced the appearance of intense ARC-positive cells, dantrolene treatment did not change the levels of ARC in both western blot and immunofluorescence analyses. Accordingly, in the same experimental conditions, we were not able to detect changes in the levels of both pre- and post-synaptic plasticity-related proteins, growth associated protein-43 (GAP-43) and postsynaptic density protein-95 (PSD-95), respectively. Additionally, the density of mossy fiber sprouting in the DG was not increased by dantrolene treatment. We next examined the effects of intrahippocampal injection of dantrolene on neurodegeneration. Notably, dantrolene promoted neuroprotective effects by decreasing neuronal cell loss in CA1 and CA3, which explains the increased levels of synaptic proteins, and the apparent lack of positive effect on pathological plasticity. Taken together, our results revealed that RyRs may have a major role in the hippocampal neurodegeneration associated to the development of acquired epilepsy.
Collapse
Affiliation(s)
- Pedro Xavier Royero
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil; Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Daiane Soares Kostecki
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Bianca Araújo Dos Santos
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Cayo Almeida
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Kézia Accioly Andrade
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Erika Reime Kinjo
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil; Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
20
|
Pottoo FH, Javed MN, Barkat MA, Alam MS, Nowshehri JA, Alshayban DM, Ansari MA. Estrogen and Serotonin: Complexity of Interactions and Implications for Epileptic Seizures and Epileptogenesis. Curr Neuropharmacol 2019; 17:214-231. [PMID: 29956631 PMCID: PMC6425080 DOI: 10.2174/1570159x16666180628164432] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/01/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
A burgeoning literature documents the confluence of ovarian steroids and central serotonergic systems in the injunction of epileptic seizures and epileptogenesis. Estrogen administration in animals reduces neuronal death from seizures by up-regulation of the prosurvival molecule i.e. Bcl-2, anti-oxidant potential and protection of NPY interneurons. Serotonin modulates epileptiform activity in either direction i.e administration of 5-HT agonists or reuptake inhibitors leads to the activation of 5-HT3 and 5-HT1A receptors tending to impede focal and generalized seizures, while depletion of brain 5-HT along with the destruction of serotonergic terminals leads to expanded neuronal excitability hence abatement of seizure threshold in experimental animal models. Serotonergic neurotransmission is influenced by the organizational activity of steroid hormones in the growing brain and the actuation effects of steroids which come in adulthood. It is further established that ovarian steroids bring induction of dendritic spine proliferation on serotonin neurons thus thawing a profound effect on serotonergic transmission. This review features 5-HT1A and 5-HT3 receptors as potential targets for ameliorating seizure-induced neurodegeneration and recurrent hypersynchronous neuronal activity. Indeed 5-HT3 receptors mediate cross-talk between estrogenic and serotonergic pathways, and could be well exploited for combinatorial drug therapy against epileptogenesis.
Collapse
Affiliation(s)
- Faheem Hyder Pottoo
- Address correspondence to these authors at the Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University (Formerly University of Dammam), Dammam 31441, Saudi Arabia; E-mail: and Department of Pharmacology, College of Clinical Pharmacy, 31441 Imam Abdulrahman Bin Faisal University, (Formerly University of Dammam), Dammam, Saudi Arabia; E-mail:
| | | | | | | | | | | | - Mohammad Azam Ansari
- Address correspondence to these authors at the Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University (Formerly University of Dammam), Dammam 31441, Saudi Arabia; E-mail: and Department of Pharmacology, College of Clinical Pharmacy, 31441 Imam Abdulrahman Bin Faisal University, (Formerly University of Dammam), Dammam, Saudi Arabia; E-mail:
| |
Collapse
|
21
|
Pansani AP, Cysneiros RM, Colugnati DB, Janjoppi L, Ferrari D, de Lima E, Ghazale PP, Sinigaglia-Coimbra R, Scorza FA. Long-term monotherapy treatment with vitamin E reduces oxidative stress, but not seizure frequency in rats submitted to the pilocarpine model of epilepsy. Epilepsy Behav 2018; 88:301-307. [PMID: 30342389 DOI: 10.1016/j.yebeh.2018.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 09/20/2018] [Indexed: 01/29/2023]
Abstract
The imbalance between antioxidant system and reactive oxygen species (ROS) generation is related to epileptogenesis, neuronal death, and seizure frequency. Treatment with vitamin E has been associated with neuroprotection and control of seizures. In most experimental studies, vitamin E treatment has short duration. Therefore, the aim of this study was to verify the role of long-term treatment with vitamin E in rats submitted to the pilocarpine model of epilepsy. Rats were divided into two main groups: control (Ctr) and pilocarpine (Pilo). Each one was subdivided according to treatment: vehicle (Ctr V and Pilo V) or vitamin E at dosages of 6 IU/kg/day (Ctr E6 and Pilo E6) or 60 IU/kg/day (Ctr E60 and Pilo E60). Treatment lasted 120 days from status epilepticus (SE). There were no statistical differences concerning treatment in the Ctr group for all variables, so the data were grouped. Carbonyl content in the hippocampus of Pilo V and Pilo E6 was higher compared with that of the Ctr group (8 ± 1.5, 7.1 ± 1, and 3.1 ± 0.3 nmol carbonyl/mg protein, respectively for Pilo V, Pilo E6, and Ctr; p < 0.05). Carbonyl content was restored to control values in Pilo E60 rats (4.2 ± 1.1 and 3.1 ± 0.3 nmol carbonyl/mg protein, respectively for Pilo E60 and Ctr; p > 0.05). The volume of the hippocampal formation (6.5 ± 0.3, 6.6 ± 0.4, 6.3 ± 0.3, and 7.4 ± 0.2, respectively for Pilo V, Pilo E6, Pilo E60, and Ctr) and subfields CA1 (1.6 ± 0.1, 1.4 ± 0.2, 1.5 ± 0.1, and 2 ± 0.05, respectively for Pilo V, Pilo E6, Pilo E60, and Ctr) and CA3 (1.7 ± 0.1, 1.5 ± 0.2, 1.4 ± 0.1, and 2 ± 0.1, respectively for Pilo V, Pilo E6, Pilo E60, and Ctr) was reduced in the Pilo group regardless of treatment. Parvalbumin immunostaining was increased in the hilus of the Pilo E60 group compared with that in the Ctr group (26 ± 2 and 39.6 ± 8.3 neurons, respectively for Ctr and Pilo E60). No difference was found in seizure frequency and Neo-Timm staining. Therefore, long-term treatment with 60 IU/kg/day of vitamin E prevented oxidative damage in the hippocampus and increased hilar parvalbumin expression in rats with epilepsy without a reduction in seizure frequency.
Collapse
Affiliation(s)
- Aline Priscila Pansani
- Laboratório Integrado de Fisiopatologia Cardiovascular e Neurológica, Departamento de Ciências Fisiológicas, Universidade Federal de Goiás, Brazil.
| | - Roberta Monterazzo Cysneiros
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento do Centro de Ciências Biológicas e da Saúde - Universidade Presbiteriana Mackenzie, São Paulo, Brazil
| | - Diego Basile Colugnati
- Laboratório Integrado de Fisiopatologia Cardiovascular e Neurológica, Departamento de Ciências Fisiológicas, Universidade Federal de Goiás, Brazil
| | - Luciana Janjoppi
- Laboratório de Neurologia Experimental - Universidade Federal de São Paulo (UNIFESP), Brazil
| | - Danuza Ferrari
- Laboratório de Neurologia Experimental - Universidade Federal de São Paulo (UNIFESP), Brazil
| | - Eliângela de Lima
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde - Universidade Federal de Mato Grosso (UFMT), Brazil
| | - Poliana Peres Ghazale
- Laboratório de Neurologia Experimental - Universidade Federal de São Paulo (UNIFESP), Brazil
| | | | - Fulvio Alexandre Scorza
- Laboratório de Neurologia Experimental - Universidade Federal de São Paulo (UNIFESP), Brazil
| |
Collapse
|
22
|
Miller ER, Kharlamov EA, Hu Z, Klein EC, Shiau DS, Kelly KM. Transient and permanent arterial occlusions modeling poststroke epilepsy in aging rats. Epilepsy Res 2018; 148:69-77. [PMID: 30391633 DOI: 10.1016/j.eplepsyres.2018.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/31/2018] [Accepted: 10/23/2018] [Indexed: 11/16/2022]
Abstract
The pathophysiological mechanisms of epileptogenesis following ischemic stroke in the aged brain are not well understood, largely due to limited developments in animal modeling of poststroke epilepsy (PSE). A recent study in our laboratory (Kelly et al., 2018) using transient (3 h) unilateral middle cerebral artery (MCA) and common carotid artery (CCA) occlusion (MCA/CCAo) in 4- and 20-month-old Fischer (F344) rats resulted in epileptic seizures in both age groups; age and infarction factors independently had effects on seizure frequency. We hypothesized that permanent unilateral MCA/CCAo, a simpler model, was capable of producing results comparable to those of transient MCA/CCAo. In this study, we performed permanent MCA/CCAo and compared it to transient MCA/CCAo in 76 4-, 12-, and 20-month-old F344 rats; 41 (54%) animals experienced early, unexpected mortality. The remaining 35 (46%) animals had depth electrodes implanted. Prior to implantation of depth electrodes, 9 (26%) of these 35 animals (26%) were monitored periodically by video alone before video-EEG monitoring (17,837 h total) to assess the potential development of PSE. No EEG recordings were obtained from 12- or 20-month-old transient occlusion or 20-month-old permanent occlusion animals due to premature deaths. Five animals (14%) demonstrated epileptic seizure activity after MCA/CCAo: one 4-month-old transient occlusion animal, one 4-month-old permanent occlusion animal, and three 12-month-old permanent occlusion animals. Of these 5 animals, all but the 4-month-old permanent animal demonstrated 1-4 Hz spike-wave discharges variably associated with inactivity or frank motor arrest, and 2 animals (4- and 12-month-old permanent) demonstrated generalized ictal EEG discharges associated with grade 5 convulsive activity. All animals monitored with video-EEG demonstrated generalized 7-9 Hz spike-wave discharges, innate in F344 animals and distinct from lesion-induced epileptic seizures. Gross inspection of brains revealed variability in lesion presence and size among age groups and occlusion types. Comparison of infarct volumes of permanent MCA/CCAo animals (2.9 ± 1.29 mm3, n = 6) with those of transient MCA/CCAo animals (1.7 ± 0.31 mm3, n = 3) was not significant (p = 0.44) due to the small sample size. Timm staining revealed no evidence of mossy fiber sprouting in 7 animals tested, only one of which was known to be epileptic (4-month-old transient). These results provide evidence of focal nonconvulsive electrographic ictal discharges and behavioral seizures in both permanent and transient MCA/CCAo animals lesioned at 4- or 12-months-of-age and support the use of arterial ligation as a viable method for modeling PSE.
Collapse
Affiliation(s)
- Eric R Miller
- Neurology and Neuroscience Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA, 15212, United States
| | - Elena A Kharlamov
- Neurology and Neuroscience Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA, 15212, United States; Neurology, Drexel University College of Medicine, Philadelphia, PA, 19102, United States
| | - Zeyu Hu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, United States
| | - Edwin C Klein
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA, 15260, United States
| | | | - Kevin M Kelly
- Neurology and Neuroscience Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA, 15212, United States; Neurology, Drexel University College of Medicine, Philadelphia, PA, 19102, United States; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, United States; Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, United States.
| |
Collapse
|
23
|
Abstract
Evidence from both preclinical and clinical studies suggest the importance of zinc homeostasis in seizures/epilepsy. Undoubtedly, zinc, via modulation of a variety of targets, is necessary for maintaining the balance between neuronal excitation and inhibition, while an imbalance between excitation and inhibition underlies seizures. However, the relationship between zinc signaling and seizures/epilepsy is complex as both extracellular and intracellular zinc may produce either protective or detrimental effects. This review provides an overview of preclinical/behavioral, functional and molecular studies, as well as clinical data on the involvement of zinc in the pathophysiology and treatment of seizures/epilepsy. Furthermore, the potential of targeting elements associated with zinc signaling or homeostasis and zinc levels as a therapeutic strategy for epilepsy is discussed.
Collapse
Affiliation(s)
- Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Katarzyna Młyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Aleksandra Wlaź
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland; Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
24
|
Takahashi DK, Jin S, Prince DA. Gabapentin Prevents Progressive Increases in Excitatory Connectivity and Epileptogenesis Following Neocortical Trauma. Cereb Cortex 2018; 28:2725-2740. [PMID: 28981586 PMCID: PMC6041890 DOI: 10.1093/cercor/bhx152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/30/2017] [Accepted: 06/01/2017] [Indexed: 11/12/2022] Open
Abstract
Neocortical injury initiates a cascade of events, some of which result in maladaptive epileptogenic reorganization of surviving neural circuits. Research focused on molecular and organizational changes that occur following trauma may reveal processes that underlie human post-traumatic epilepsy (PTE), a common and unfortunate consequence of traumatic brain injury. The latency between injury and development of PTE provides an opportunity for prophylactic intervention, once the key underlying mechanisms are understood. In rodent neocortex, injury to pyramidal neurons promotes axonal sprouting, resulting in increased excitatory circuitry that is one important factor promoting epileptogenesis. We used laser-scanning photostimulation of caged glutamate and whole-cell recordings in in vitro slices from injured neocortex to assess formation of new excitatory synapses, a process known to rely on astrocyte-secreted thrombospondins (TSPs), and to map the distribution of maladaptive circuit reorganization. We show that this reorganization is centered principally in layer V and associated with development of epileptiform activity. Short-term blockade of the synaptogenic effects of astrocyte-secreted TSPs with gabapentin (GBP) after injury suppresses the new excitatory connectivity and epileptogenesis for at least 2 weeks. Results reveal that aberrant circuit rewiring is progressive in vivo and provide further rationale for prophylactic anti-epileptogenic use of gabapentinoids following cortical trauma.
Collapse
Affiliation(s)
- D K Takahashi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Sha Jin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - D A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
25
|
Koyama R, Ikegaya Y. The Molecular and Cellular Mechanisms of Axon Guidance in Mossy Fiber Sprouting. Front Neurol 2018; 9:382. [PMID: 29896153 PMCID: PMC5986954 DOI: 10.3389/fneur.2018.00382] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/11/2018] [Indexed: 01/25/2023] Open
Abstract
The question of whether mossy fiber sprouting is epileptogenic has not been resolved; both sprouting-induced recurrent excitatory and inhibitory circuit hypotheses have been experimentally (but not fully) supported. Therefore, whether mossy fiber sprouting is a potential therapeutic target for epilepsy remains under debate. Moreover, the axon guidance mechanisms of mossy fiber sprouting have attracted the interest of neuroscientists. Sprouting of mossy fibers exhibits several uncommon axonal growth features in the basically non-plastic adult brain. For example, robust branching of axonal collaterals arises from pre-existing primary mossy fiber axons. Understanding the branching mechanisms in adulthood may contribute to axonal regeneration therapies in neuroregenerative medicine in which robust axonal re-growth is essential. Additionally, because granule cells are produced throughout life in the neurogenic dentate gyrus, it is interesting to examine whether the mossy fibers of newly generated granule cells follow the pre-existing trajectories of sprouted mossy fibers in the epileptic brain. Understanding these axon guidance mechanisms may contribute to neuron transplantation therapies, for which the incorporation of transplanted neurons into pre-existing neural circuits is essential. Thus, clarifying the axon guidance mechanisms of mossy fiber sprouting could lead to an understanding of central nervous system (CNS) network reorganization and plasticity. Here, we review the molecular and cellular mechanisms of axon guidance in mossy fiber sprouting by discussing mainly in vitro studies.
Collapse
Affiliation(s)
- Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Demars F, Clark K, Wyeth MS, Abrams E, Buckmaster PS. A single subconvulsant dose of domoic acid at mid-gestation does not cause temporal lobe epilepsy in mice. Neurotoxicology 2018; 66:128-137. [PMID: 29625197 PMCID: PMC5940543 DOI: 10.1016/j.neuro.2018.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 11/19/2022]
Abstract
Harmful blooms of domoic acid (DA)-producing algae are a problem in oceans worldwide. DA is a potent glutamate receptor agonist that can cause status epilepticus and in survivors, temporal lobe epilepsy. In mice, one-time low-dose in utero exposure to DA was reported to cause hippocampal damage and epileptiform activity, leading to the hypothesis that unrecognized exposure to DA from contaminated seafood in pregnant women can damage the fetal hippocampus and initiate temporal lobe epileptogenesis. However, development of epilepsy (i.e., spontaneous recurrent seizures) has not been tested. In the present study, long-term seizure monitoring and histology was used to test for temporal lobe epilepsy following prenatal exposure to DA. In Experiment One, the previous study's in utero DA treatment protocol was replicated, including use of the CD-1 mouse strain. Afterward, mice were video-monitored for convulsive seizures from 2 to 6 months old. None of the CD-1 mice treated in utero with vehicle or DA was observed to experience spontaneous convulsive seizures. After seizure monitoring, mice were evaluated for pathological evidence of temporal lobe epilepsy. None of the mice treated in utero with DA displayed the hilar neuron loss that occurs in patients with temporal lobe epilepsy and in the mouse pilocarpine model of temporal lobe epilepsy. In Experiment Two, a higher dose of DA was administered to pregnant FVB mice. FVB mice were tested as a potentially more sensitive strain, because they have a lower seizure threshold, and some females spontaneously develop epilepsy. Female offspring were monitored with continuous video and telemetric bilateral hippocampal local field potential recording at 1-11 months old. A similar proportion of vehicle- and DA-treated female FVB mice spontaneously developed epilepsy, beginning in the fourth month of life. Average seizure frequency and duration were similar in both groups. Seizure frequency was lower than that of positive-control pilocarpine-treated mice, but seizure duration was similar. None of the mice treated in utero with vehicle or DA displayed hilar neuron loss or intense mossy fiber sprouting, a form of aberrant synaptic reorganization that develops in patients with temporal lobe epilepsy and in pilocarpine-treated mice. FVB mice that developed epilepsy (vehicle- and DA-treated) displayed mild mossy fiber sprouting. Results of this study suggest that a single subconvulsive dose of DA at mid-gestation does not cause temporal lobe epilepsy in mice.
Collapse
Affiliation(s)
- Fanny Demars
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA, United States; VetAgro Sup, School of Veterinary Medicine, Lyon, France; Paris Descartes University, Paris, France
| | - Kristen Clark
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA, United States; College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States
| | - Megan S Wyeth
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA, United States
| | - Emily Abrams
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA, United States
| | - Paul S Buckmaster
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA, United States; Department of Neurology & Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
27
|
Wong JC, Makinson CD, Lamar T, Cheng Q, Wingard JC, Terwilliger EF, Escayg A. Selective targeting of Scn8a prevents seizure development in a mouse model of mesial temporal lobe epilepsy. Sci Rep 2018; 8:126. [PMID: 29317669 PMCID: PMC5760706 DOI: 10.1038/s41598-017-17786-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/30/2017] [Indexed: 11/08/2022] Open
Abstract
We previously found that genetic mutants with reduced expression or activity of Scn8a are resistant to induced seizures and that co-segregation of a mutant Scn8a allele can increase survival and seizure resistance of Scn1a mutant mice. In contrast, Scn8a expression is increased in the hippocampus following status epilepticus and amygdala kindling. These findings point to Scn8a as a promising therapeutic target for epilepsy and raise the possibility that aberrant overexpression of Scn8a in limbic structures may contribute to some epilepsies, including temporal lobe epilepsy. Using a small-hairpin-interfering RNA directed against the Scn8a gene, we selectively reduced Scn8a expression in the hippocampus of the intrahippocampal kainic acid (KA) mouse model of mesial temporal lobe epilepsy. We found that Scn8a knockdown prevented the development of spontaneous seizures in 9/10 mice, ameliorated KA-induced hyperactivity, and reduced reactive gliosis. These results support the potential of selectively targeting Scn8a for the treatment of refractory epilepsy.
Collapse
Affiliation(s)
- Jennifer C Wong
- Department of Human Genetics, Emory University, Atlanta, Georgia, 30322, USA
| | | | - Tyra Lamar
- Department of Human Genetics, Emory University, Atlanta, Georgia, 30322, USA
| | - Qi Cheng
- Department of Human Genetics, Emory University, Atlanta, Georgia, 30322, USA
| | - Jeffrey C Wingard
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ernest F Terwilliger
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, Georgia, 30322, USA.
| |
Collapse
|
28
|
Dunn R, Queenan BN, Pak DTS, Forcelli PA. Divergent effects of levetiracetam and tiagabine against spontaneous seizures in adult rats following neonatal hypoxia. Epilepsy Res 2017; 140:1-7. [PMID: 29227795 DOI: 10.1016/j.eplepsyres.2017.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/09/2017] [Accepted: 12/02/2017] [Indexed: 01/03/2023]
Abstract
Animal models are valuable tools for screening novel therapies for patients who suffer from epilepsy. However, a wide array of models are necessary to cover the diversity of human epilepsies. In humans, neonatal hypoxia (or hypoxia-ischemia) is one of the most common causes of epilepsy early in life. Hypoxia-induced seizures (HS) during the neonatal period can also lead to spontaneous seizures in adulthood. This phenomenon, i.e., early-life hypoxia leading to adult epilepsy - is also seen in experimental models, including rats. However, it is not known which anti-seizure medications are most effective at managing adult epilepsy resulting from neonatal HS. Here, we examined the efficacy of three anti-seizure medications against spontaneous seizures in adult rats with a history of neonatal HS: (1) phenobarbital (PHB), the oldest epilepsy medicine still in use today; (2) levetiracetam (LEV); and (3) tiagabine (TGB). Both LEV and TGB are relatively new anticonvulsant drugs that are ineffective in traditional seizure models, but strikingly effective in other models. We found that PHB and LEV decreased seizures in adult rats with a history of HS, whereas TGB exacerbated seizures. These divergent drug effects indicate that the HS model may be useful for differentiating the clinical efficacy of putative epilepsy therapies.
Collapse
Affiliation(s)
- Raymond Dunn
- Department of Pharmacology and Physiology, United States
| | - Bridget N Queenan
- Department of Pharmacology and Physiology, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States; Neuroscience Research Institute, Department of Mechanical Engineering; Department of Physics, University sof California, Santa Barbara, Santa Barbara, CA, United States
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States.
| | - Patrick A Forcelli
- Department of Pharmacology and Physiology, United States; Department of Neuroscience, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States.
| |
Collapse
|
29
|
Amorim RP, Araújo MGL, Valero J, Lopes-Cendes I, Pascoal VDB, Malva JO, da Silva Fernandes MJ. Silencing of P2X7R by RNA interference in the hippocampus can attenuate morphological and behavioral impact of pilocarpine-induced epilepsy. Purinergic Signal 2017; 13:467-478. [PMID: 28707031 PMCID: PMC5714836 DOI: 10.1007/s11302-017-9573-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 06/28/2017] [Indexed: 12/01/2022] Open
Abstract
Cell signaling mediated by P2X7 receptors (P2X7R) has been suggested to be involved in epileptogenesis, via modulation of intracellular calcium levels, excitotoxicity, activation of inflammatory cascades, and cell death, among other mechanisms. These processes have been described to be involved in pilocarpine-induced status epilepticus (SE) and contribute to hyperexcitability, resulting in spontaneous and recurrent seizures. Here, we aimed to investigate the role of P2X7R in epileptogenesis in vivo using RNA interference (RNAi) to inhibit the expression of this receptor. Small interfering RNA (siRNA) targeting P2X7R mRNA was injected into the lateral ventricles (icv) 6 h after SE. Four groups were studied: Saline-Vehicle, Saline-siRNA, Pilo-Vehicle, and Pilo-siRNA. P2X7R was quantified by western blotting and neuronal death assessed by Fluoro-Jade B histochemistry. The hippocampal volume (edema) was determined 48 h following RNAi. Behavioral parameters as latency to the appearance of spontaneous seizures and the number of seizures were determined until 60 days after the SE onset. The Saline-siRNA and Pilo-siRNA groups showed a 43 and 37% reduction, respectively, in P2X7R protein levels compared to respective vehicle groups. Neuroprotection was observed in CA1 and CA3 of the Pilo-siRNA group compared to Pilo-Vehicle. P2X7R silencing in pilocarpine group reversed the increase in the edema detected in the hilus, suprapyramidal dentate gyrus, CA1, and CA3; reduced mortality rate following SE; increased the time to onset of spontaneous seizure; and reduced the number of seizures, when compared to the Pilo-Vehicle group. Therefore, our data highlights the potential of P2X7R as a therapeutic target for the adjunct treatment of epilepsy.
Collapse
Affiliation(s)
- Rebeca Padrão Amorim
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil
| | - Michelle Gasparetti Leão Araújo
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil
| | - Jorge Valero
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
- Achucarro Basque Center for Neuroscience, Zamudio, Bizkaia, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Iscia Lopes-Cendes
- Departamento de Genética Médica, Faculdade de Medicina da Unicamp, Campinas, SP, Brazil
| | | | - João Oliveira Malva
- Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria José da Silva Fernandes
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil.
| |
Collapse
|
30
|
Ferreira ES, Vieira LG, Moraes DM, Amorim BO, Malheiros JM, Hamani C, Covolan L. Long-Term Effects of Anterior Thalamic Nucleus Deep Brain Stimulation on Spatial Learning in the Pilocarpine Model of Temporal Lobe Epilepsy. Neuromodulation 2017; 21:160-167. [PMID: 28960670 DOI: 10.1111/ner.12688] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/29/2017] [Accepted: 07/25/2017] [Indexed: 01/20/2023]
Abstract
INTRODUCTION AND OBJECTIVES Cognitive impairment is a significant comorbidity of temporal lobe epilepsy that is associated with extensive hippocampal cell loss. Deep brain stimulation (DBS) of the anterior thalamic nucleus (ANT) has been used for the treatment of refractory partial seizures. In the pilocarpine model of epilepsy, ANT DBS applied during status epilepticus (SE) reduces hippocampal inflammation and apoptosis. When given to chronic epileptic animals it reduces hippocampal excitability and seizure frequency. Here, we tested whether ANT DBS delivered during SE and the silent phase of the pilocarpine model would reduce cognitive impairment when animals became chronically epileptic. MATERIALS AND METHODS SE was induced by a systemic pilocarpine injection (320 mg/kg). Immediately after SE onset, rats were assigned to receive DBS during the first six hours of SE (n = 8; DBSa group) or during SE + the silent period (i.e., 6 h/day until the animals developed the first spontaneous recurrent seizure; n = 10; DBSs group). Four months following SE, animals underwent water maze testing and histological evaluation. Nonstimulated chronic epileptic animals (n = 13; PCTL group) and age-matched naïve rats (n = 11, CTL group) were used as controls. Results were analyzed by repeated-measures analyses of variance (RM_ANOVA) and one-way ANOVAs, followed by Newman-Keuls post hoc tests. RESULTS Although all groups learned the spatial task, epileptic animals with or without DBS spent significantly less time in the platform quadrant, denoting a spatial memory deficit (p < 0.02). Despite these negative behavioral results, we found that animals given DBS had a significantly higher number of cells in the CA1 region and dentate gyrus. Mossy fiber sprouting was similar among all epileptic groups. CONCLUSIONS Despite lesser hippocampal neuronal loss, ANT DBS delivered either during SE or during SE and the silent phase of the pilocarpine model did not mitigate memory deficits in chronic epileptic rats.
Collapse
Affiliation(s)
- Elenn Soares Ferreira
- Department of de Physiology, Universidade Federal de São Paulo, São Paulo, Sao Paulo, Brazil
| | - Laís Gabrielle Vieira
- Department of de Physiology, Universidade Federal de São Paulo, São Paulo, Sao Paulo, Brazil
| | - Daniela Macedo Moraes
- Department of de Physiology, Universidade Federal de São Paulo, São Paulo, Sao Paulo, Brazil
| | - Beatriz O Amorim
- Department of de Physiology, Universidade Federal de São Paulo, São Paulo, Sao Paulo, Brazil
| | | | - Clement Hamani
- Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health, Toronto, Canada.,Division of Neurosurgery, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Luciene Covolan
- Department of de Physiology, Universidade Federal de São Paulo, São Paulo, Sao Paulo, Brazil
| |
Collapse
|
31
|
Buckmaster PS, Abrams E, Wen X. Seizure frequency correlates with loss of dentate gyrus GABAergic neurons in a mouse model of temporal lobe epilepsy. J Comp Neurol 2017; 525:2592-2610. [PMID: 28425097 PMCID: PMC5963263 DOI: 10.1002/cne.24226] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/19/2023]
Abstract
Epilepsy occurs in one of 26 people. Temporal lobe epilepsy is common and can be difficult to treat effectively. It can develop after brain injuries that damage the hippocampus. Multiple pathophysiological mechanisms involving the hippocampal dentate gyrus have been proposed. This study evaluated a mouse model of temporal lobe epilepsy to test which pathological changes in the dentate gyrus correlate with seizure frequency and help prioritize potential mechanisms for further study. FVB mice (n = 127) that had experienced status epilepticus after systemic treatment with pilocarpine 31-61 days earlier were video-monitored for spontaneous, convulsive seizures 9 hr/day every day for 24-36 days. Over 4,060 seizures were observed. Seizure frequency ranged from an average of one every 3.6 days to one every 2.1 hr. Hippocampal sections were processed for Nissl stain, Prox1-immunocytochemistry, GluR2-immunocytochemistry, Timm stain, glial fibrillary acidic protein-immunocytochemistry, glutamic acid decarboxylase in situ hybridization, and parvalbumin-immunocytochemistry. Stereological methods were used to measure hilar ectopic granule cells, mossy cells, mossy fiber sprouting, astrogliosis, and GABAergic interneurons. Seizure frequency was not significantly correlated with the generation of hilar ectopic granule cells, the number of mossy cells, the extent of mossy fiber sprouting, the extent of astrogliosis, or the number of GABAergic interneurons in the molecular layer or hilus. Seizure frequency significantly correlated with the loss of GABAergic interneurons in or adjacent to the granule cell layer, but not with the loss of parvalbumin-positive interneurons. These findings prioritize the loss of granule cell layer interneurons for further testing as a potential cause of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Paul S. Buckmaster
- Department of Comparative Medicine, Stanford University, Stanford, California
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, California
| | - Emily Abrams
- Department of Comparative Medicine, Stanford University, Stanford, California
| | - Xiling Wen
- Department of Comparative Medicine, Stanford University, Stanford, California
| |
Collapse
|
32
|
Kinjo ER, Rodríguez PXR, Dos Santos BA, Higa GSV, Ferraz MSA, Schmeltzer C, Rüdiger S, Kihara AH. New Insights on Temporal Lobe Epilepsy Based on Plasticity-Related Network Changes and High-Order Statistics. Mol Neurobiol 2017; 55:3990-3998. [PMID: 28555345 DOI: 10.1007/s12035-017-0623-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/16/2017] [Indexed: 12/21/2022]
Abstract
Epilepsy is a disorder of the brain characterized by the predisposition to generate recurrent unprovoked seizures, which involves reshaping of neuronal circuitries based on intense neuronal activity. In this review, we first detailed the regulation of plasticity-associated genes, such as ARC, GAP-43, PSD-95, synapsin, and synaptophysin. Indeed, reshaping of neuronal connectivity after the primary, acute epileptogenesis event increases the excitability of the temporal lobe. Herein, we also discussed the heterogeneity of neuronal populations regarding the number of synaptic connections, which in the theoretical field is commonly referred as degree. Employing integrate-and-fire neuronal model, we determined that in addition to increased synaptic strength, degree correlations might play essential and unsuspected roles in the control of network activity. Indeed, assortativity, which can be described as a condition where high-degree correlations are observed, increases the excitability of neural networks. In this review, we summarized recent topics in the field, and data were discussed according to newly developed or unusual tools, as provided by mathematical graph analysis and high-order statistics. With this, we were able to present new foundations for the pathological activity observed in temporal lobe epilepsy.
Collapse
Affiliation(s)
- Erika Reime Kinjo
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Pedro Xavier Royero Rodríguez
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Bianca Araújo Dos Santos
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Mariana Sacrini Ayres Ferraz
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Christian Schmeltzer
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
- Institute of Physics, Humboldt University at Berlin, Berlin, Germany
| | - Sten Rüdiger
- Institute of Physics, Humboldt University at Berlin, Berlin, Germany
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil.
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
33
|
Do Val-da Silva RA, Peixoto-Santos JE, Kandratavicius L, De Ross JB, Esteves I, De Martinis BS, Alves MNR, Scandiuzzi RC, Hallak JEC, Zuardi AW, Crippa JA, Leite JP. Protective Effects of Cannabidiol against Seizures and Neuronal Death in a Rat Model of Mesial Temporal Lobe Epilepsy. Front Pharmacol 2017; 8:131. [PMID: 28367124 PMCID: PMC5355474 DOI: 10.3389/fphar.2017.00131] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/02/2017] [Indexed: 11/30/2022] Open
Abstract
The present study reports the behavioral, electrophysiological, and neuropathological effects of cannabidiol (CBD), a major non-psychotropic constituent of Cannabis sativa, in the intrahippocampal pilocarpine-induced status epilepticus (SE) rat model. CBD was administered before pilocarpine-induced SE (group SE+CBDp) or before and after SE (group SE+CBDt), and compared to rats submitted only to SE (SE group), CBD, or vehicle (VH group). Groups were evaluated during SE (behavioral and electrophysiological analysis), as well as at days one and three post-SE (exploratory activity, electrophysiological analysis, neuron density, and neuron degeneration). Compared to SE group, SE+CBD groups (SE+CBDp and SE+CBDt) had increased SE latency, diminished SE severity, increased contralateral afterdischarge latency and decreased relative powers in delta (0.5–4 Hz) and theta (4–10 Hz) bands. Only SE+CBDp had increased vertical exploratory activity 1-day post SE and decreased contralateral relative power in delta 3 days after SE, when compared to SE group. SE+CBD groups also showed decreased neurodegeneration in the hilus and CA3, and higher neuron density in granule cell layer, hilus, CA3, and CA1, when compared to SE group. Our findings demonstrate anticonvulsant and neuroprotective effects of CBD preventive treatment in the intrahippocampal pilocarpine epilepsy model, either as single or multiple administrations, reinforcing the potential role of CBD in the treatment of epileptic disorders.
Collapse
Affiliation(s)
- Raquel A Do Val-da Silva
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo São Paulo, Brazil
| | - Jose E Peixoto-Santos
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo São Paulo, Brazil
| | - Ludmyla Kandratavicius
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil; National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil
| | - Jana B De Ross
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo São Paulo, Brazil
| | - Ingrid Esteves
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo São Paulo, Brazil
| | - Bruno S De Martinis
- National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil; Department of Chemistry, Faculty of Philosophy, Science and Languages of Ribeirao Preto, University of São PauloSão Paulo, Brazil
| | - Marcela N R Alves
- Department of Chemistry, Faculty of Philosophy, Science and Languages of Ribeirao Preto, University of São Paulo São Paulo, Brazil
| | - Renata C Scandiuzzi
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil; National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil
| | - Antonio W Zuardi
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil; National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil
| | - Jose A Crippa
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil; National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil
| | - Joao P Leite
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil; National Institute of Science and Technology for Translational Medicine, Conselho Nacional de Desenvolvimento Cientifico e TecnologicoBrasília, Brazil
| |
Collapse
|
34
|
Inhibition of Calcineurin A by FK506 Suppresses Seizures and Reduces the Expression of GluN2B in Membrane Fraction. Neurochem Res 2017; 42:2154-2166. [PMID: 28299629 DOI: 10.1007/s11064-017-2221-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 02/08/2023]
Abstract
FK506, a calcineurin inhibitor, shows neuroprotective effects and has been associated with neurodegenerative diseases. Calcineurin A (CaNA), a catalytic subunit of calcineurin, mediates the dephosphorylation of various proteins. N-methyl-D-aspartate receptor (GluN) is closely related to epileptogenesis, and various phosphorylation sites of GluN2B, a regulatory subunit of the GluN complex, have different functions. Thus, we hypothesized that one of the potential anti-epileptic mechanisms of FK506 is mediated by its ability to promote the phosphorylation of GluN2B and reduce the expression of GluN2B in membrane fraction by down-regulating CaNA. CaNA expression was increased in the cortex of patients with temporal lobe epilepsy and pentylenetetrazol (PTZ)-induced epileptic models. CaNA was shown to be expressed in neurons using immunofluorescence staining. According to our behavioral observations, epileptic rats exhibited less severe seizures and were less sensitive to PTZ after a systemic injection of FK506. The levels of phosphorylated GluN2B were decreased in epileptic rats but increased after the FK506 treatment. Moreover, there was no difference in the total GluN2B levels before and after FK506 treatment. However, the expression of GluN2B in membrane fraction was suppressed after FK506 treatment. Based on these results, FK506 may reduce the severity and frequency of seizures by reducing the expression of GluN2B in membrane fraction.
Collapse
|
35
|
Vannucci Campos D, Lopim GM, da Silva DA, de Almeida AA, Amado D, Arida RM. Epilepsy and exercise: An experimental study in female rats. Physiol Behav 2017; 171:120-126. [DOI: 10.1016/j.physbeh.2016.12.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/25/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
|
36
|
Extent of mossy fiber sprouting in patients with mesiotemporal lobe epilepsy correlates with neuronal cell loss and granule cell dispersion. Epilepsy Res 2017; 129:51-58. [DOI: 10.1016/j.eplepsyres.2016.11.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/20/2016] [Indexed: 11/21/2022]
|
37
|
Opačić M, Ristić AJ, Savić D, Šelih VS, Živin M, Sokić D, Raičević S, Baščarević V, Spasojević I. Metal maps of sclerotic hippocampi of patients with mesial temporal lobe epilepsy. Metallomics 2017; 9:141-148. [DOI: 10.1039/c6mt00293e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Petralia RS, Wang YX, Mattson MP, Yao PJ. The Diversity of Spine Synapses in Animals. Neuromolecular Med 2016; 18:497-539. [PMID: 27230661 PMCID: PMC5158183 DOI: 10.1007/s12017-016-8405-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/11/2016] [Indexed: 12/23/2022]
Abstract
Here we examine the structure of the various types of spine synapses throughout the animal kingdom. Based on available evidence, we suggest that there are two major categories of spine synapses: invaginating and non-invaginating, with distributions that vary among different groups of animals. In the simplest living animals with definitive nerve cells and synapses, the cnidarians and ctenophores, most chemical synapses do not form spine synapses. But some cnidarians have invaginating spine synapses, especially in photoreceptor terminals of motile cnidarians with highly complex visual organs, and also in some mainly sessile cnidarians with rapid prey capture reflexes. This association of invaginating spine synapses with complex sensory inputs is retained in the evolution of higher animals in photoreceptor terminals and some mechanoreceptor synapses. In contrast to invaginating spine synapse, non-invaginating spine synapses have been described only in animals with bilateral symmetry, heads and brains, associated with greater complexity in neural connections. This is apparent already in the simplest bilaterians, the flatworms, which can have well-developed non-invaginating spine synapses in some cases. Non-invaginating spine synapses diversify in higher animal groups. We also discuss the functional advantages of having synapses on spines and more specifically, on invaginating spines. And finally we discuss pathologies associated with spine synapses, concentrating on those systems and diseases where invaginating spine synapses are involved.
Collapse
Affiliation(s)
- Ronald S Petralia
- Advanced Imaging Core, NIDCD/NIH, 35A Center Drive, Room 1E614, Bethesda, MD, 20892-3729, USA.
| | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD/NIH, 35A Center Drive, Room 1E614, Bethesda, MD, 20892-3729, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Pamela J Yao
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| |
Collapse
|
39
|
Decreased neuron loss and memory dysfunction in pilocarpine-treated rats pre-exposed to hypoxia. Neuroscience 2016; 332:88-100. [DOI: 10.1016/j.neuroscience.2016.06.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/04/2016] [Accepted: 06/24/2016] [Indexed: 01/03/2023]
|
40
|
Sharma AK, Reams RY, Jordan WH, Miller MA, Thacker HL, Snyder PW. Mesial Temporal Lobe Epilepsy: Pathogenesis, Induced Rodent Models and Lesions. Toxicol Pathol 2016; 35:984-99. [PMID: 18098044 DOI: 10.1080/01926230701748305] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Mesial temporal lobe epilepsy (MTLE), the most common epilepsy in adults, is generally intractable and is suspected to be the result of recurrent excitation or inhibition circuitry. Recurrent excitation and the development of seizures have been associated with aberrant mossy fiber sprouting in the hippocampus. Of the animal models developed to investigate the pathogenesis of MTLE, post-status epilepticus models have received the greatest acceptance because they are characterized by a latency period, the development of spontaneous motor seizures, and a spectrum of lesions like those of MTLE. Among post-status epilepticus models, induction of systemic kainic acid or pilocarpine-induced epilepsy is less labor-intensive than electrical-stimulation models and these models mirror the clinicopathologic features of MTLE more closely than do kindling, tetanus toxin, hyperthermia, post-traumatic, and perinatal hypoxia/ischemia models. Unfortunately, spontaneous motor seizures do not develop in kindling or adult hyperthermia models and are not a consistent finding in tetanus toxin-induced or perinatal hypoxia/ischemia models. This review presents the mechanistic hypotheses for seizure induction, means of model induction, and associated pathology, especially as compared to MTLE patients. Animal models are valuable tools not only to study the pathogenesis of MTLE, but also to evaluate potential antiepileptogenic drugs.
Collapse
Affiliation(s)
- Alok K. Sharma
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
- Department of Pathology, Covance Laboratories Inc., Madison, WI, 53704, USA
| | - Rachel Y. Reams
- Department of Pathology, Lilly Research Laboratories, Division of Eli Lilly and Co., Greenfield, IN, 46140, USA
| | - William H. Jordan
- Department of Pathology, Lilly Research Laboratories, Division of Eli Lilly and Co., Greenfield, IN, 46140, USA
| | - Margaret A. Miller
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - H. Leon Thacker
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Paul W. Snyder
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
41
|
|
42
|
Sun H, Juul HM, Jensen FE. Models of hypoxia and ischemia-induced seizures. J Neurosci Methods 2015; 260:252-60. [PMID: 26434705 DOI: 10.1016/j.jneumeth.2015.09.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 01/19/2023]
Abstract
Despite greater understanding and improved management, seizures continue to be a major problem in childhood. Neonatal seizures are often refractory to conventional antiepileptic drugs, and can result in later life epilepsy and cognitive deficits, conditions for which there are no specific treatments. Hypoxic and/or ischemic encephalopathy (HIE) is the most common cause for neonatal seizures, and accounts for more than two-thirds of neonatal seizure cases. A better understanding of the cellular and molecular mechanisms is essential for identifying new therapeutic strategies that control the neonatal seizures and its cognitive consequences. This heavily relies on animal models that play a critical role in discovering novel mechanisms underlying both epileptogenesis and associated cognitive impairments. To date, a number of animal models have provided a tremendous amount of information regarding the pathophysiology of HIE-induced neonatal seizures. This review provides an overview on the most important features of the main animal models of HIE-induced seizures. In particular, we focus on the methodology of seizure induction and the characterizations of post-HIE injury consequences. These aspects of HIE-induced seizure models are discussed in the light of the suitability of these models in studying human HIE-induced seizures.
Collapse
Affiliation(s)
- Hongyu Sun
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Halvor M Juul
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
43
|
Van Nieuwenhuyse B, Raedt R, Sprengers M, Dauwe I, Gadeyne S, Carrette E, Delbeke J, Wadman WJ, Boon P, Vonck K. The systemic kainic acid rat model of temporal lobe epilepsy: Long-term EEG monitoring. Brain Res 2015; 1627:1-11. [PMID: 26381287 DOI: 10.1016/j.brainres.2015.08.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 02/03/2023]
Abstract
Animal models reproducing the characteristics of human epilepsy are essential for the elucidation of the pathophysiological mechanisms. In epilepsy research there is ongoing debate on whether the epileptogenic process is a continuous process rather than a step function. The aim of this study was to assess progression of epileptogenesis over the long term and to evaluate possible correlations between SE duration and severity with the disease progression in the kainic acid model. Rats received repeated KA injections (5mg/kg) until a self-sustained SE was elicited. Continuous depth EEG recording started before KA injection and continued for 30 weeks. Mean seizure rate progression could be expressed as a sigmoid function and increased from 1 ± 0.2 seizures per day during the second week after SE to 24.4 ± 6.4 seizures per day during week 30. Seizure rate progressed to a plateau phase 122 ± 9 days after SE. However, the individual seizure rate during this plateau phase varied between 14.5 seizures and 48.6 seizures per day. A circadian rhythm in seizure occurrence was observed in all rats. Histological characterization of damage to the dentate gyrus in the KA treated rats confirmed the presence of astrogliosis and aberrant mossy fiber sprouting in the dentate gyrus. This long-term EEG monitoring study confirms that epileptogenesis is a continuous process rather than a step function.
Collapse
Affiliation(s)
- B Van Nieuwenhuyse
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - R Raedt
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - M Sprengers
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - I Dauwe
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - S Gadeyne
- Swammerdam Institute of Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands.
| | - E Carrette
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - J Delbeke
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - W J Wadman
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium; Swammerdam Institute of Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands.
| | - P Boon
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - K Vonck
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology, and Neuropsychology, Department of Neurology, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
44
|
Posttraumatic seizures and epilepsy in adult rats after controlled cortical impact. Epilepsy Res 2015; 117:104-16. [PMID: 26432760 DOI: 10.1016/j.eplepsyres.2015.09.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 08/26/2015] [Accepted: 09/10/2015] [Indexed: 12/27/2022]
Abstract
Posttraumatic epilepsy (PTE) has been modeled with different techniques of experimental traumatic brain injury (TBI) using mice and rats at various ages. We hypothesized that the technique of controlled cortical impact (CCI) could be used to establish a model of PTE in young adult rats. A total of 156 male Sprague-Dawley rats of 2-3 months of age (128 CCI-injured and 28 controls) was used for monitoring and/or anatomical studies. Provoked class 3-5 seizures were recorded by video monitoring in 7/57 (12.3%) animals in the week immediately following CCI of the right parietal cortex; none of the 7 animals demonstrated subsequent spontaneous convulsive seizures. Monitoring with video and/or video-EEG was performed on 128 animals at various time points 8-619 days beyond one week following CCI during which 26 (20.3%) demonstrated nonconvulsive or convulsive epileptic seizures. Nonconvulsive epileptic seizures of >10s were demonstrated in 7/40 (17.5%) animals implanted with 2 or 3 depth electrodes and usually characterized by an initial change in behavior (head raising or animal alerting) followed by motor arrest during an ictal discharge that consisted of high-amplitude spikes or spike-waves with frequencies ranging between 1 and 2Hz class 3-5 epileptic seizures were recorded by video monitoring in 17/88 (19%) and by video-EEG in 2/40 (5%) CCI-injured animals. Ninety of 156 (58%) animals (79 CCI-injured, 13 controls) underwent transcardial perfusion for gross and microscopic studies. CCI caused severe brain tissue loss and cavitation of the ipsilateral cerebral hemisphere associated with cell loss in the hippocampal CA1 and CA3 regions, hilus, and dentate granule cells, and thalamus. All Timm-stained CCI-injured brains demonstrated ipsilateral hippocampal mossy fiber sprouting in the inner molecular layer. These results indicate that the CCI model of TBI in adult rats can be used to study the structure-function relationships that underlie epileptogenesis and PTE.
Collapse
|
45
|
Bernhardt BC, Bonilha L, Gross DW. Network analysis for a network disorder: The emerging role of graph theory in the study of epilepsy. Epilepsy Behav 2015; 50:162-70. [PMID: 26159729 DOI: 10.1016/j.yebeh.2015.06.005] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 01/01/2023]
Abstract
Recent years have witnessed a paradigm shift in the study and conceptualization of epilepsy, which is increasingly understood as a network-level disorder. An emblematic case is temporal lobe epilepsy (TLE), the most common drug-resistant epilepsy that is electroclinically defined as a focal epilepsy and pathologically associated with hippocampal sclerosis. In this review, we will summarize histopathological, electrophysiological, and neuroimaging evidence supporting the concept that the substrate of TLE is not limited to the hippocampus alone, but rather is broadly distributed across multiple brain regions and interconnecting white matter pathways. We will introduce basic concepts of graph theory, a formalism to quantify topological properties of complex systems that has recently been widely applied to study networks derived from brain imaging and electrophysiology. We will discuss converging graph theoretical evidence indicating that networks in TLE show marked shifts in their overall topology, providing insight into the neurobiology of TLE as a network-level disorder. Our review will conclude by discussing methodological challenges and future clinical applications of this powerful analytical approach.
Collapse
Affiliation(s)
- Boris C Bernhardt
- Neuroimaging of Epilepsy Laboratory, Brain Imaging Center, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada; Department of Social Neuroscience, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Leonardo Bonilha
- Department of Neurology, Medical University of South Carolina, SC, USA
| | - Donald W Gross
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
46
|
Peixoto-Santos JE, Velasco TR, Galvis-Alonso OY, Araujo D, Kandratavicius L, Assirati JA, Carlotti CG, Scandiuzzi RC, Santos ACD, Leite JP. Temporal lobe epilepsy patients with severe hippocampal neuron loss but normal hippocampal volume: Extracellular matrix molecules are important for the maintenance of hippocampal volume. Epilepsia 2015. [PMID: 26218733 DOI: 10.1111/epi.13082] [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] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Hippocampal sclerosis is a common finding in patients with temporal lobe epilepsy (TLE), and magnetic resonance imaging (MRI) studies associate the reduction of hippocampal volume with the neuron loss seen on histologic evaluation. Astrogliosis and increased levels of chondroitin sulfate, a major component of brain extracellular matrix, are also seen in hippocampal sclerosis. Our aim was to evaluate the association between hippocampal volume and chondroitin sulfate, as well as neuronal and astroglial populations in the hippocampus of patients with TLE. METHODS Patients with drug-resistant TLE were subdivided, according to hippocampal volume measured by MRI, into two groups: hippocampal atrophy (HA) or normal volume (NV) cases. Hippocampi from TLE patients and age-matched controls were submitted to immunohistochemistry to evaluate neuronal population, astroglial population, and chondroitin sulfate expression with antibodies against neuron nuclei protein (NeuN), glial fibrillary acidic protein (GFAP), and chondroitin sulfate (CS-56) antigens, respectively. RESULTS Both TLE groups were clinically similar. NV cases had higher hippocampal volume, both ipsilateral and contralateral, when compared to HA. Compared to controls, NV and HA patients had reduced neuron density, and increased GFAP and CS-56 immunopositive area. There was no statistical difference between NV and HA groups in neuron density or immunopositive areas for GFAP and CS-56. Hippocampal volume correlated positively with neuron density in CA1 and prosubiculum, and with immunopositive areas for CS-56 in CA1, and negatively with immunopositive area for GFAP in CA1. Multiple linear regression analysis indicated that both neuron density and CS-56 immunopositive area in CA1 were statistically significant predictors of hippocampal volume. SIGNIFICANCE Our findings indicate that neuron density and chondroitin sulfate immunopositive area in the CA1 subfield are crucial for the hippocampal volume, and that chondroitin sulfate is important for the maintenance of a normal hippocampal volume in some cases with severe neuron loss.
Collapse
Affiliation(s)
- Jose Eduardo Peixoto-Santos
- Department of Neurosciences and Behavior, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Tonicarlo Rodrigues Velasco
- Department of Neurosciences and Behavior, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Orfa Yineth Galvis-Alonso
- Department of Molecular Biology, São José do Rio Preto Medical School, Sao Jose do Rio Preto, Sao Paulo, Brazil
| | - David Araujo
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Ludmyla Kandratavicius
- Department of Neurosciences and Behavior, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Joao Alberto Assirati
- Department of Surgery, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Brazil
| | - Carlos Gilberto Carlotti
- Department of Surgery, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Brazil
| | - Renata Caldo Scandiuzzi
- Department of Neurosciences and Behavior, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Antonio Carlos dos Santos
- Department of Internal Medicine, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Brazil
| | - Joao Pereira Leite
- Department of Neurosciences and Behavior, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
47
|
Weissberg I, Wood L, Kamintsky L, Vazquez O, Milikovsky DZ, Alexander A, Oppenheim H, Ardizzone C, Becker A, Frigerio F, Vezzani A, Buckwalter MS, Huguenard JR, Friedman A, Kaufer D. Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol Dis 2015; 78:115-25. [PMID: 25836421 DOI: 10.1016/j.nbd.2015.02.029] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/07/2015] [Accepted: 02/19/2015] [Indexed: 01/26/2023] Open
Abstract
Post-injury epilepsy (PIE) is a common complication following brain insults, including ischemic, and traumatic brain injuries. At present, there are no means to identify the patients at risk to develop PIE or to prevent its development. Seizures can occur months or years after the insult, do not respond to anti-seizure medications in over third of the patients, and are often associated with significant neuropsychiatric morbidities. We have previously established the critical role of blood-brain barrier dysfunction in PIE, demonstrating that exposure of brain tissue to extravasated serum albumin induces activation of inflammatory transforming growth factor beta (TGF-β) signaling in astrocytes and eventually seizures. However, the link between the acute astrocytic inflammatory responses and reorganization of neural networks that underlie recurrent spontaneous seizures remains unknown. Here we demonstrate in vitro and in vivo that activation of the astrocytic ALK5/TGF-β-pathway induces excitatory, but not inhibitory, synaptogenesis that precedes the appearance of seizures. Moreover, we show that treatment with SJN2511, a specific ALK5/TGF-β inhibitor, prevents synaptogenesis and epilepsy. Our findings point to astrocyte-mediated synaptogenesis as a key epileptogenic process and highlight the manipulation of the TGF-β-pathway as a potential strategy for the prevention of PIE.
Collapse
Affiliation(s)
- Itai Weissberg
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Lydia Wood
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Lyn Kamintsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Oscar Vazquez
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Allyson Alexander
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Hannah Oppenheim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Carolyn Ardizzone
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Albert Becker
- Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Federica Frigerio
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3140, USA; Canadian Institute for Advanced Research (CIFAR) Program in Child and Brain Development Toronto, ON M5G 1Z8, Canada.
| |
Collapse
|
48
|
Kandratavicius L, Peixoto-Santos JE, Monteiro MR, Scandiuzzi RC, Carlotti CG, Assirati JA, Hallak JE, Leite JP. Mesial temporal lobe epilepsy with psychiatric comorbidities: a place for differential neuroinflammatory interplay. J Neuroinflammation 2015; 12:38. [PMID: 25889039 PMCID: PMC4347571 DOI: 10.1186/s12974-015-0266-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 02/10/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Despite the strong association between epilepsy and psychiatric comorbidities, few biological substrates are currently described. We have previously reported neuropathological alterations in mesial temporal lobe epilepsy (MTLE) patients with major depression and psychosis that suggest a morphological and neurochemical basis for psychopathological symptoms. Neuroinflammatory-related structures and molecules might be part of the altered neurochemical milieu underlying the association between epilepsy and psychiatric comorbidities, and such features have not been previously investigated in humans. METHODS MTLE hippocampi of subjects without psychiatric history (MTLEW), MTLE + major depression (MTLE + D), and MTLE + interictal psychosis (MTLE + P) derived from epilepsy surgery and control necropsies were investigated for reactive astrocytes (glial fibrillary acidic protein (GFAP)), activated microglia (human leukocyte antigen, MHC class II (HLA-DR)), glial metallothionein-I/II (MT-I/II), and aquaporin 4 (AQP4) immunohistochemistry. RESULTS We found an increased GFAP immunoreactive area in the molecular layers, granule cell layer, and cornus ammonis region 2 (CA2) and cornus ammonis region 1 (CA1) of MTLEW and MTLE + P, respectively, compared to MTLE + D. HLA-DR immunoreactive area was higher in cornus ammonis region 3 (CA3) of MTLE + P, compared to MTLE + D and MTLEW, and in the hilus, when compared to MTLEW. MTLEW cases showed increased MT-I/II area in the granule cell layer and CA1, compared to MTLE + P, and in the parasubiculum, when compared to MTLE + D and MTLE + P. Differences between MTLE and control, such as astrogliosis, microgliosis, increased MT-I/II, and decreased perivascular AQP4 in the epileptogenic hippocampus, were in agreement to what is currently described in the literature. CONCLUSIONS Neuroinflammatory-related molecules in MTLE hippocampus show a distinct pattern of expression when patients present with a comorbid psychiatric diagnosis, similar to what is found in the pure forms of schizophrenia and major depression. Future studies focusing on inflammatory characteristics of MTLE with psychiatric comorbidities might help in the design of better therapeutic strategies.
Collapse
Affiliation(s)
- Ludmyla Kandratavicius
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| | - Jose Eduardo Peixoto-Santos
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | - Mariana Raquel Monteiro
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | - Renata Caldo Scandiuzzi
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | | | | | - Jaime Eduardo Hallak
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil. .,National Institute of Science and Technology in Translational Medicine (INCT-TM - CNPq), Ribeirao Preto, Brazil.
| | - Joao Pereira Leite
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| |
Collapse
|
49
|
Bittencourt S, Covolan L, Hamani C, Longo BM, Faria FP, Freymuller E, Ottersen OP, Mello LE. Replacement of Asymmetric Synaptic Profiles in the Molecular Layer of Dentate Gyrus Following Cycloheximide in the Pilocarpine Model in Rats. Front Psychiatry 2015; 6:157. [PMID: 26635635 PMCID: PMC4647073 DOI: 10.3389/fpsyt.2015.00157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/23/2015] [Indexed: 11/13/2022] Open
Abstract
Mossy fiber sprouting is among the best-studied forms of post-lesional synaptic plasticity and is regarded by many as contributory to seizures in both humans and animal models of epilepsy. It is not known whether mossy fiber sprouting increases the number of synapses in the molecular layer or merely replaces lost contacts. Using the pilocarpine (Pilo) model of status epilepticus to induce mossy fiber sprouting, and cycloheximide (CHX) to block this sprouting, we evaluated at the ultrastructural level the number and type of asymmetric synaptic contacts in the molecular layer of the dentate gyrus. As expected, whereas Pilo-treated rats had dense silver grain deposits in the inner molecular layer (IML) (reflecting mossy fiber sprouting), pilocarpine + cycloheximide (CHX + Pilo)-treated animals did not differ from controls. Both groups of treated rats (Pilo group and CHX + Pilo group) had reduced density of asymmetric synaptic profiles (putative excitatory synaptic contacts), which was greater for CHX-treated animals. For both treated groups, the loss of excitatory synaptic contacts was even greater in the outer molecular layer than in the best-studied IML (in which mossy fiber sprouting occurs). These results indicate that mossy fiber sprouting tends to replace lost synaptic contacts rather than increase the absolute number of contacts. We speculate that the overall result is more consistent with restored rather than with increased excitability.
Collapse
Affiliation(s)
- Simone Bittencourt
- Department of Physiology, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Luciene Covolan
- Department of Physiology, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Clement Hamani
- Division of Neurosurgery, Toronto Western Hospital , Toronto, ON , Canada
| | - Beatriz M Longo
- Department of Physiology, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Flávio P Faria
- Electron Microscopy Center, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Edna Freymuller
- Electron Microscopy Center, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Ole P Ottersen
- Department of Anatomy, Centre for Molecular Biology and Neuroscience, University of Oslo , Oslo , Norway
| | - Luiz E Mello
- Department of Physiology, Universidade Federal de São Paulo , São Paulo , Brazil
| |
Collapse
|
50
|
Bramlett HM, Dietrich WD. Long-Term Consequences of Traumatic Brain Injury: Current Status of Potential Mechanisms of Injury and Neurological Outcomes. J Neurotrauma 2014; 32:1834-48. [PMID: 25158206 DOI: 10.1089/neu.2014.3352] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a significant clinical problem with few therapeutic interventions successfully translated to the clinic. Increased importance on the progressive, long-term consequences of TBI have been emphasized, both in the experimental and clinical literature. Thus, there is a need for a better understanding of the chronic consequences of TBI, with the ultimate goal of developing novel therapeutic interventions to treat the devastating consequences of brain injury. In models of mild, moderate, and severe TBI, histopathological and behavioral studies have emphasized the progressive nature of the initial traumatic insult and the involvement of multiple pathophysiological mechanisms, including sustained injury cascades leading to prolonged motor and cognitive deficits. Recently, the increased incidence in age-dependent neurodegenerative diseases in this patient population has also been emphasized. Pathomechanisms felt to be active in the acute and long-term consequences of TBI include excitotoxicity, apoptosis, inflammatory events, seizures, demyelination, white matter pathology, as well as decreased neurogenesis. The current article will review many of these pathophysiological mechanisms that may be important targets for limiting the chronic consequences of TBI.
Collapse
Affiliation(s)
- Helen M Bramlett
- The Miami Project to Cure Paralysis/Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis/Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|