1
|
Gelinas JN, Khodagholy D. Interictal network dysfunction and cognitive impairment in epilepsy. Nat Rev Neurosci 2025:10.1038/s41583-025-00924-3. [PMID: 40295879 DOI: 10.1038/s41583-025-00924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
Epilepsy is diagnosed when neural networks become capable of generating excessive or hypersynchronous activity patterns that result in observable seizures. In many cases, epilepsy is associated with cognitive comorbidities that persist between seizures and negatively impact quality of life. Dysregulation of the coordinated physiological network interactions that are required for cognitive function has been implicated in mediating these enduring symptoms, but the causal mechanisms are often elusive. Here, we provide an overview of neural network abnormalities with the potential to contribute to cognitive dysfunction in epilepsy. We examine these pathological interactions across spatial and temporal scales, additionally highlighting the dynamics that arise in response to the brain's intrinsic capacity for plasticity. Understanding these processes will facilitate development of network-level interventions to address cognitive comorbidities that remain undertreated by currently available epilepsy therapeutics.
Collapse
Affiliation(s)
- Jennifer N Gelinas
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Paediatrics, University of California, Irvine, CA, USA.
- Children's Hospital of Orange County, Orange, CA, USA.
| | - Dion Khodagholy
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Electrical Engineering, University of California, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, USA.
- Department of Materials Science and Engineering, University of California, Irvine, CA, USA.
| |
Collapse
|
2
|
Yazi S, Sehirli US, Gulhan R, Onat F, Kirazli O. Evaluation of dendrite morphology in Wistar and genetic absence epileptic rats. Brain Struct Funct 2024; 230:5. [PMID: 39681662 DOI: 10.1007/s00429-024-02868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/06/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVE Genetic Absence Epilepsy Rat from Strasbourg (GAERS), a rodent model genetically predisposed to absence epilepsy, serves as an experimental tool to elucidate the neuronal mechanisms underlying human absence epilepsy. This study aimed to investigate the morphological features of dendrites and dendritic spines of pyramidal neurons in somatosensory cortex and hippocampus of Wistar and GAERS rats. MATERIAL AND METHOD Adult male GAERS (n = 5) and control Wistar (n = 5) rats were sacrificed by transcardial perfusion and brains were removed. Brain tissues were processed by Golgi impregnation method using FD Rapid GolgiStain Kit. Coronal sections were obtained with a cryostat. Pyramidal neurons in layers V-VI of the somatosensory cortex and the CA1 region of the hippocampus were examined using a light microscope and Neurolucida 360 software. Dendrite nodes, dendrite segments (dendritic branching), dendrite terminations, total dendrite length, dendritic spine density, and dendritic spine types were analyzed. RESULTS Compared to Wistar, GAERS exhibited significantly higher numbers of nodes (p = 0.0053, p = 0.0047), segments (p = 0.0036, p = 0.0036), and terminations (p = 0.0033, p = 0.0029) in the dendrites of the somatosensory cortex and the hippocampus, respectively. Furthermore, the total dendrite length (µm) (p = 0.0002, p = 0.0007) and the density of dendritic spines (1/µm) (p = 0.0168, p = 0.0120) were significantly high in GAERS compared to Wistar. When dendritic spine types were evaluated separately, stubby-type dendritic spines in the hippocampus were higher in GAERS compared to Wistar (p = 0.0045). CONCLUSION Intense synaptic connections in the somatosensory cortex and the hippocampus of genetic absence epileptic rats led to morphological alterations in the dendrites and the dendritic spines of pyramidal neurons in these regions, potentially contributing to the pathophysiology of absence seizures.
Collapse
Affiliation(s)
- Sevdenur Yazi
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey
| | - Umit S Sehirli
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey
| | - Rezzan Gulhan
- Department of Medical Pharmacology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Filiz Onat
- Department of Medical Pharmacology, School of Medicine, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Ozlem Kirazli
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey.
| |
Collapse
|
3
|
El-Shafei SMA, El-Rahman AAA, Abuelsaad ASA, Al-Khalaf AA, Shehab GMG, Abdel-Aziz AM. Assessment of the potential protective effects of culture filtrate of Trichoderma harzianum to ameliorate the damaged histoarchitecture of brain in epileptic rats. Metab Brain Dis 2024; 39:1363-1385. [PMID: 39115642 DOI: 10.1007/s11011-024-01391-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/14/2024] [Indexed: 10/29/2024]
Abstract
The simultaneous hyperexcitability of the neural network is the most well-known manifestation of epilepsy that causes recurrent seizures. The current study was aimed to examine any potential safety benefits of the culture filtrate of Trichoderma harzianum (ThCF) to ameliorate damaged histoarchitecture of the brain in epileptic rats by assessing seizure intensity scale and behavioral impairments and follow up the spontaneous motor seizures during status epilepticus phases in rats. Twenty-four rats were divided into four groups; control (C), epileptic (EP) valproic acid-treated epileptic (EP-VPA), and epileptic treated with T. harzianum cultured filtrate (ThCF). In addition to a seizure intensity score and behavioral tests, routine H&E and Golgi-Copsch histopathology, were used to examine the cell somas, dendrites, axons, and neural spines. ThCF treatment increased activity and recorded movements during grooming, rearing, and ambulation frequency. Brain tissues of epileptic rats exhibited detached meninges, hypercellularity, mild edema in the cortex and markedly degenerated neurons, degenerated glial cells, and microcyst formation in the hippocampus. Moreover, brains of EP-ThCF were noticed with average blood vessels, and increased dendritogenesis. The current data revealed some of negative effects of epileptogenesis brought on by seizure intensity score and retarded histopathological alterations in the hippocampus. Therefore, the study is forecasting to identify novel active components from the metabolites of T. harzianum with a crucial therapeutic role in various disorders.
Collapse
Affiliation(s)
- Sally M A El-Shafei
- Department of Agricultural Chemistry, Faculty of Agriculture, Minia University, 61517, El-Minya, Egypt
| | - Atef A Abd El-Rahman
- Department of Agricultural Chemistry, Faculty of Agriculture, Minia University, 61517, El-Minya, Egypt
| | - Abdelaziz S A Abuelsaad
- Immunology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62521, Egypt
| | - Areej A Al-Khalaf
- Plant Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Gaber M G Shehab
- Department of Biochemistry, College of Medicine, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ayman M Abdel-Aziz
- Cell Biology, Histology and Genetics Division, Zoology Department, Faculty of Science, Fayoum University, Fayoum, 63514, Egypt.
| |
Collapse
|
4
|
Rodemer W, Ra I, Jia E, Gujral J, Zhang B, Hoxha K, Xing B, Mehta S, Farag M, Porta S, Jensen FE, Talos DM, Lee VMY. Hyperexcitability precedes CA3 hippocampal neurodegeneration in a dox-regulatable TDP-43 mouse model of ALS-FTD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.612703. [PMID: 39386447 PMCID: PMC11463581 DOI: 10.1101/2024.09.24.612703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Neuronal hyperexcitability is a hallmark of amyotrophic lateral sclerosis (ALS) but its relationship with the TDP-43 aggregates that comprise the predominant pathology in over 90% of ALS cases remains unclear. Emerging evidence in tissue and slice culture models indicate that TDP-43 pathology induces neuronal hyperexcitability suggesting it may be responsible for the excitotoxicity long believed to be a major driver of ALS neuron death. Here, we characterized hyperexcitability and neurodegeneration in the hippocampus of doxycycline-regulatable rNLS8 mice (NEFH-tTA x tetO-hTDP-43ΔNLS), followed by treatment with AAV encoded DREADDs and anti-seizure medications to measure the effect on behavioral function and neurodegeneration. We found that approximately half of the CA3 neurons in the dorsal hippocampus are lost between 4 and 6 weeks after TDP-43ΔNLS induction. Neurodegeneration was preceded by selective hyperexcitability in the mossy fiber - CA3 circuit, leading us to hypothesize that glutamate excitotoxicity may be a significant contributor to neurodegeneration in this model. Interestingly, hippocampal injection of AAV encoded inhibitory DREADDs (hM4Di) and daily activation with CNO ligand rescued anxiety deficits on elevated zero maze (EZM) but did not reduce neurodegeneration. Therapeutic doses of the anti-seizure medications, valproic acid and levetiracetam, did not improve behavior or prevent neurodegeneration. These results highlight the complexity of TDP-43 - induced alterations to neuronal excitability and suggest that whereas targeting hyperexcitability can meliorate some behavioral deficits, it may not be sufficient to halt or slow neurodegeneration in TDP-43-related proteinopathies. Significance Statement Cytoplasmic aggregates of TAR DNA Binding Protein 43 (TDP-43) are the predominant pathology in over 90% of Amyotrophic lateral sclerosis (ALS) and the majority of frontotemporal lobar degeneration (FTLD-TDP) cases. Understanding how TDP-43 pathology promotes neurodegeneration may lead to therapeutic strategies to slow disease progression in humans. Recent reports in mouse and cell culture models suggest loss-of-normal TDP-43 function may drive neuronal hyperexcitability, a key physiological hallmark of ALS and possible contributor to neurodegeneration. In this study, we identified region-specific hyperexcitability that precedes neurodegeneration in the inducible rNLS8 TDP-43 mouse model. Suppressing hyperexcitability with chemogenetics improved behavioral function but did not reduce hippocampal neuron loss. Anti-seizure medications had no beneficial effects suggesting directly targeting hyperexcitability may not be therapeutically effective.
Collapse
|
5
|
Huang L, Wang Y, He Y, Huang D, Wen T, Han Z. Association Between COVID-19 and Neurological Diseases: Evidence from Large-Scale Mendelian Randomization Analysis and Single-Cell RNA Sequencing Analysis. Mol Neurobiol 2024; 61:6354-6365. [PMID: 38300446 PMCID: PMC11339101 DOI: 10.1007/s12035-024-03975-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/18/2024] [Indexed: 02/02/2024]
Abstract
Observational studies have suggested that SARS-CoV-2 infection increases the risk of neurological diseases, but it remains unclear whether the association is causal. The present study aims to evaluate the causal relationships between SARS-CoV-2 infections and neurological diseases and analyzes the potential routes of SARS-CoV-2 entry at the cellular level. We performed Mendelian randomization (MR) analysis with CAUSE method to investigate causal relationship of SARS-CoV-2 infections with neurological diseases. Then, we conducted single-cell RNA sequencing (scRNA-seq) analysis to obtain evidence of potential neuroinvasion routes by measuring SARS-CoV-2 receptor expression in specific cell subtypes. Fast gene set enrichment analysis (fGSEA) was further performed to assess the pathogenesis of related diseases. The results showed that the COVID-19 is causally associated with manic (delta_elpd, - 0.1300, Z-score: - 2.4; P = 0.0082) and epilepsy (delta_elpd: - 2.20, Z-score: - 1.80; P = 0.038). However, no significant effects were observed for COVID-19 on other traits. Moreover, there are 23 cell subtypes identified through the scRNA-seq transcriptomics data of epilepsy, and SARS-CoV-2 receptor TTYH2 was found to be specifically expressed in oligodendrocyte and astrocyte cell subtypes. Furthermore, fGSEA analysis showed that the cell subtypes with receptor-specific expression was related to methylation of lysine 27 on histone H3 (H3K27ME3), neuronal system, aging brain, neurogenesis, and neuron projection. In summary, this study shows causal links between SARS-CoV-2 infections and neurological disorders such as epilepsy and manic, supported by MR and scRNA-seq analysis. These results should be considered in further studies and public health measures on COVID-19 and neurological diseases.
Collapse
Affiliation(s)
- Lin Huang
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yongheng Wang
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
- International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yijie He
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Dongyu Huang
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Tong Wen
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Zhijie Han
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Postnikova TY, Diespirov GP, Malkin SL, Chernyshev AS, Vylekzhanina EN, Zaitsev AV. Morphological and Functional Alterations in the CA1 Pyramidal Neurons of the Rat Hippocampus in the Chronic Phase of the Lithium-Pilocarpine Model of Epilepsy. Int J Mol Sci 2024; 25:7568. [PMID: 39062811 PMCID: PMC11276980 DOI: 10.3390/ijms25147568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Epilepsy is known to cause alterations in neural networks. However, many details of these changes remain poorly understood. The objective of this study was to investigate changes in the properties of hippocampal CA1 pyramidal neurons and their synaptic inputs in a rat lithium-pilocarpine model of epilepsy. In the chronic phase of the model, we found a marked loss of pyramidal neurons in the CA1 area. However, the membrane properties of the neurons remained essentially unaltered. The results of the electrophysiological and morphological studies indicate that the direct pathway from the entorhinal cortex to CA1 neurons is reinforced in epileptic animals, whereas the inputs to them from CA3 are either unaltered or even diminished. In particular, the dendritic spine density in the str. lacunosum moleculare, where the direct pathway from the entorhinal cortex terminates, was found to be 2.5 times higher in epileptic rats than in control rats. Furthermore, the summation of responses upon stimulation of the temporoammonic pathway was enhanced by approximately twofold in epileptic rats. This enhancement is believed to be a significant contributing factor to the heightened epileptic activity observed in the entorhinal cortex of epileptic rats using an ex vivo 4-aminopyridine model.
Collapse
Affiliation(s)
- Tatyana Y. Postnikova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Saint Petersburg 194223, Russia; (T.Y.P.); (G.P.D.); (S.L.M.); (E.N.V.)
| | - Georgy P. Diespirov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Saint Petersburg 194223, Russia; (T.Y.P.); (G.P.D.); (S.L.M.); (E.N.V.)
| | - Sergey L. Malkin
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Saint Petersburg 194223, Russia; (T.Y.P.); (G.P.D.); (S.L.M.); (E.N.V.)
| | | | - Elizaveta N. Vylekzhanina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Saint Petersburg 194223, Russia; (T.Y.P.); (G.P.D.); (S.L.M.); (E.N.V.)
| | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Saint Petersburg 194223, Russia; (T.Y.P.); (G.P.D.); (S.L.M.); (E.N.V.)
| |
Collapse
|
7
|
Caznok Silveira AC, Antunes ASLM, Athié MCP, da Silva BF, Ribeiro dos Santos JV, Canateli C, Fontoura MA, Pinto A, Pimentel-Silva LR, Avansini SH, de Carvalho M. Between neurons and networks: investigating mesoscale brain connectivity in neurological and psychiatric disorders. Front Neurosci 2024; 18:1340345. [PMID: 38445254 PMCID: PMC10912403 DOI: 10.3389/fnins.2024.1340345] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
The study of brain connectivity has been a cornerstone in understanding the complexities of neurological and psychiatric disorders. It has provided invaluable insights into the functional architecture of the brain and how it is perturbed in disorders. However, a persistent challenge has been achieving the proper spatial resolution, and developing computational algorithms to address biological questions at the multi-cellular level, a scale often referred to as the mesoscale. Historically, neuroimaging studies of brain connectivity have predominantly focused on the macroscale, providing insights into inter-regional brain connections but often falling short of resolving the intricacies of neural circuitry at the cellular or mesoscale level. This limitation has hindered our ability to fully comprehend the underlying mechanisms of neurological and psychiatric disorders and to develop targeted interventions. In light of this issue, our review manuscript seeks to bridge this critical gap by delving into the domain of mesoscale neuroimaging. We aim to provide a comprehensive overview of conditions affected by aberrant neural connections, image acquisition techniques, feature extraction, and data analysis methods that are specifically tailored to the mesoscale. We further delineate the potential of brain connectivity research to elucidate complex biological questions, with a particular focus on schizophrenia and epilepsy. This review encompasses topics such as dendritic spine quantification, single neuron morphology, and brain region connectivity. We aim to showcase the applicability and significance of mesoscale neuroimaging techniques in the field of neuroscience, highlighting their potential for gaining insights into the complexities of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ana Clara Caznok Silveira
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
- School of Electrical and Computer Engineering, University of Campinas, Campinas, Brazil
| | | | - Maria Carolina Pedro Athié
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Bárbara Filomena da Silva
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | | | - Camila Canateli
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Marina Alves Fontoura
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Allan Pinto
- Brazilian Synchrotron Light Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | | | - Simoni Helena Avansini
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Murilo de Carvalho
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
- Brazilian Synchrotron Light Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| |
Collapse
|
8
|
Liebowitz M, Kramer KP, Rogers EE. All Care is Brain Care: Neuro-Focused Quality Improvement in the Neonatal Intensive Care Unit. Clin Perinatol 2023; 50:399-420. [PMID: 37201988 DOI: 10.1016/j.clp.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Neonates requiring intensive care are in a critical period of brain development that coincides with the neonatal intensive care unit (NICU) hospitalization, placing these infants at high risk of brain injury and long-term neurodevelopmental impairment. Care in the NICU has the potential to be both harmful and protective to the developing brain. Neuro-focused quality improvement efforts address 3 main pillars of neuroprotective care: prevention of acquired injury, protection of normal maturation, and promotion of a positive environment. Despite challenges in measurement, many centers have shown success with consistent implementation of best and potentially better practices that may improve markers of brain health and neurodevelopment.
Collapse
Affiliation(s)
- Melissa Liebowitz
- Envision Physician Services, St. Francis Hospital, 6001 East Woodmen Road, Colorado Springs, CO 80923, USA
| | - Katelin P Kramer
- Department of Pediatrics, University of California, 550 16th Avenue, 5th Floor, San Francisco, CA 94143, USA; University of California, Benioff Children's Hospital, 550 16th Avenue, 5th Floor, San Francisco, CA 94143, USA.
| | - Elizabeth E Rogers
- Department of Pediatrics, University of California, 550 16th Avenue, 5th Floor, San Francisco, CA 94143, USA; University of California, Benioff Children's Hospital, 550 16th Avenue, 5th Floor, San Francisco, CA 94143, USA. https://twitter.com/eerogersmd
| |
Collapse
|
9
|
Kramer K, Bekmezian A, Nash K, Papp E, Glass HC. Expediting Treatment of Seizures in the Intensive Care Nursery. Pediatrics 2021; 148:peds.2020-013730. [PMID: 34380776 DOI: 10.1542/peds.2020-013730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Prolonged neonatal seizures are associated with poor neurodevelopmental outcomes. The aim of this quality improvement project was to decrease the time to medical treatment of seizures by 45% within 15 months for neonates admitted to the intensive care nursery (ICN) in an academic children's hospital. METHODS A multidisciplinary team developed key drivers for timely treatment of seizures. Targeted interventions included optimizing a seizure rescue process with a mechanism that brings a pharmacist to the bedside for expedited medication delivery, in addition to interactive educational sessions. The outcome measure was time from the decision to treat seizures to medication administration. The process measure was use of the seizure rescue process with a balancing measure of unnecessary activations of this process. Data were collected from monthly chart review and displayed on statistical process control charts for analysis. The intervention period was from January 2019 to March 2020. RESULTS Between January 2016 and March 2020, there were 203 seizure treatment events (160 preintervention and 43 postintervention) in the ICN. Time to treatment of neonatal seizures decreased by 48%, from a baseline of 27 minutes (January 2016 to December 2018) to 14 minutes by March 2020, which reflected significant and sustained improvement. This was associated with improvement in the process metric during the same time periods. Unnecessary seizure rescue process activations were stable postintervention. CONCLUSIONS Implementation of an innovative seizure rescue process, in conjunction with staff and provider education, expedited antiseizure therapy in the ICN without requiring code resources.
Collapse
Affiliation(s)
- Katelin Kramer
- Departments of Pediatrics .,UCSF Benioff Children's Hospital, San Francisco, California
| | - Arpi Bekmezian
- Departments of Pediatrics.,UCSF Benioff Children's Hospital, San Francisco, California
| | - Kendall Nash
- Departments of Pediatrics.,Neurology, Weil Institute for Neurosciences.,UCSF Benioff Children's Hospital, San Francisco, California
| | - Elizabeth Papp
- UCSF Benioff Children's Hospital, San Francisco, California
| | - Hannah C Glass
- Departments of Pediatrics.,Neurology, Weil Institute for Neurosciences.,Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California.,UCSF Benioff Children's Hospital, San Francisco, California
| |
Collapse
|
10
|
Lippman-Bell JJ, Handy M, Nieder CG, Getzfread M, Jensen FE. Altered hippocampal dendritic spine maturation after hypoxia-induced seizures in neonatal rats. Mol Cell Neurosci 2021; 113:103629. [PMID: 34015497 DOI: 10.1016/j.mcn.2021.103629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/01/2022] Open
Abstract
Cognitive comorbidities often follow early-life seizures (ELS), especially in the setting of autism and other neurodevelopmental syndromes. However, there is an incomplete understanding of whether neuronal and synaptic development are concomitantly dysregulated. We have previously shown that hypoxia-induced seizures (HS) in postnatal day (P)10 rats increase acute and later-life hippocampal glutamatergic neurotransmission and spontaneous recurrent seizures, and impair cognition and behavior. As dendritic spines critically regulate synaptic function, we hypothesized that ELS can induce developmentally specific changes in dendritic spine maturation. At intervals during one month following HS in P10 rats, we assessed dendritic spine development on pyramidal neurons in the stratum radiatum of hippocampal area CA1. Compared to control rats in which spine density significantly decreased from P10 to early adulthood (P38), post-seizure rats failed to show a developmental decrease in spine density, and spines from P38 post-seizure rats appeared more immature-shaped (long, thin). In addition, compared to P38 control rats, post-seizure P38 rats expressed significantly more synaptic PSD-95, a marker of mature synapses. These changes were preceded by a transient increase in hippocampal expression of cofilin phosphorylated at Ser3, representing a decrease in cofilin activity. These results suggest that early-life seizures may impair normal dendritic spine maturation and pruning in CA1 during development, resulting in an excess of less efficient synapses, via activity-dependent modification of actin-regulating proteins such as cofilin. Given that multiple neurodevelopmental disorders show similar failures in developmental spine pruning, the current findings may represent a deficit in structural plasticity that could be a component of a mechanism leading to later-life cognitive consequences associated with early-life seizures.
Collapse
Affiliation(s)
- Jocelyn J Lippman-Bell
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| | - Marcus Handy
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cassidy G Nieder
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America
| | - Mollie Getzfread
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
11
|
Eyo UB, Haruwaka K, Mo M, Campos-Salazar AB, Wang L, Speros XS, Sabu S, Xu P, Wu LJ. Microglia provide structural resolution to injured dendrites after severe seizures. Cell Rep 2021; 35:109080. [PMID: 33951432 PMCID: PMC8164475 DOI: 10.1016/j.celrep.2021.109080] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/01/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
Although an imbalance between neuronal excitation and inhibition underlies seizures, clinical approaches that target these mechanisms are insufficient in containing seizures in patients with epilepsy, raising the need for alternative approaches. Brain-resident microglia contribute to the development and stability of neuronal structure and functional networks that are perturbed during seizures. However, the extent of microglial contributions in response to seizures in vivo remain to be elucidated. Using two-photon in vivo imaging to visualize microglial dynamics, we show that severe seizures induce formation of microglial process pouches that target but rarely engulf beaded neuronal dendrites. Microglial process pouches are stable for hours, although they often shrink in size. We further find that microglial process pouches are associated with a better structural resolution of beaded dendrites. These findings provide evidence for the structural resolution of injured dendrites by microglia as a form of neuroprotection.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Brain Immunology and Glia Center, Department of Cell Biology and Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | | | - Mingshu Mo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China
| | - Antony Brayan Campos-Salazar
- Brain Immunology and Glia Center, Department of Cell Biology and Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xenophon S Speros
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Sruchika Sabu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Pingyi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
12
|
Klemz A, Kreis P, Eickholt BJ, Gerevich Z. The actin binding protein drebrin helps to protect against the development of seizure-like events in the entorhinal cortex. Sci Rep 2021; 11:8662. [PMID: 33883605 PMCID: PMC8060314 DOI: 10.1038/s41598-021-87967-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/31/2021] [Indexed: 11/09/2022] Open
Abstract
The actin binding protein drebrin plays a key role in dendritic spine formation and synaptic plasticity. Decreased drebrin protein levels have been observed in temporal lobe epilepsy, suggesting the involvement of drebrin in the disease. Here we investigated the effect of drebrin knockout on physiological and pathophysiological neuronal network activities in mice by inducing gamma oscillations, involved in higher cognitive functions, and by analyzing pathophysiological epileptiform activity. We found that loss of drebrin increased the emergence of spontaneous gamma oscillations suggesting an increase in neuronal excitability when drebrin is absent. Further analysis showed that although the kainate-induced hippocampal gamma oscillations were unchanged in drebrin deficient mice, seizure like events measured in the entorhinal cortex appeared earlier and more frequently. The results suggest that while drebrin is not essential for normal physiological network activity, it helps to protect against the formation of seizure like activities during pathological conditions. The data indicate that targeting drebrin function could potentially be a preventive or therapeutic strategy for epilepsy treatment.
Collapse
Affiliation(s)
- Alexander Klemz
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Patricia Kreis
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| | - Britta J Eickholt
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Zoltan Gerevich
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
13
|
Rasia-Filho AA, Guerra KTK, Vásquez CE, Dall’Oglio A, Reberger R, Jung CR, Calcagnotto ME. The Subcortical-Allocortical- Neocortical continuum for the Emergence and Morphological Heterogeneity of Pyramidal Neurons in the Human Brain. Front Synaptic Neurosci 2021; 13:616607. [PMID: 33776739 PMCID: PMC7991104 DOI: 10.3389/fnsyn.2021.616607] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
Human cortical and subcortical areas integrate emotion, memory, and cognition when interpreting various environmental stimuli for the elaboration of complex, evolved social behaviors. Pyramidal neurons occur in developed phylogenetic areas advancing along with the allocortex to represent 70-85% of the neocortical gray matter. Here, we illustrate and discuss morphological features of heterogeneous spiny pyramidal neurons emerging from specific amygdaloid nuclei, in CA3 and CA1 hippocampal regions, and in neocortical layers II/III and V of the anterolateral temporal lobe in humans. Three-dimensional images of Golgi-impregnated neurons were obtained using an algorithm for the visualization of the cell body, dendritic length, branching pattern, and pleomorphic dendritic spines, which are specialized plastic postsynaptic units for most excitatory inputs. We demonstrate the emergence and development of human pyramidal neurons in the cortical and basomedial (but not the medial, MeA) nuclei of the amygdala with cells showing a triangular cell body shape, basal branched dendrites, and a short apical shaft with proximal ramifications as "pyramidal-like" neurons. Basomedial neurons also have a long and distally ramified apical dendrite not oriented to the pial surface. These neurons are at the beginning of the allocortex and the limbic lobe. "Pyramidal-like" to "classic" pyramidal neurons with laminar organization advance from the CA3 to the CA1 hippocampal regions. These cells have basal and apical dendrites with specific receptive synaptic domains and several spines. Neocortical pyramidal neurons in layers II/III and V display heterogeneous dendritic branching patterns adapted to the space available and the afferent inputs of each brain area. Dendritic spines vary in their distribution, density, shapes, and sizes (classified as stubby/wide, thin, mushroom-like, ramified, transitional forms, "atypical" or complex forms, such as thorny excrescences in the MeA and CA3 hippocampal region). Spines were found isolated or intermingled, with evident particularities (e.g., an extraordinary density in long, deep CA1 pyramidal neurons), and some showing a spinule. We describe spiny pyramidal neurons considerably improving the connectional and processing complexity of the brain circuits. On the other hand, these cells have some vulnerabilities, as found in neurodegenerative Alzheimer's disease and in temporal lobe epilepsy.
Collapse
Affiliation(s)
- Alberto A. Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Kétlyn T. Knak Guerra
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos Escobar Vásquez
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Aline Dall’Oglio
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Roman Reberger
- Medical Engineering Program, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cláudio R. Jung
- Institute of Informatics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry and Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
14
|
Sharma S, Tiarks G, Haight J, Bassuk AG. Neuropathophysiological Mechanisms and Treatment Strategies for Post-traumatic Epilepsy. Front Mol Neurosci 2021; 14:612073. [PMID: 33708071 PMCID: PMC7940684 DOI: 10.3389/fnmol.2021.612073] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death in young adults and a risk factor for acquired epilepsy. Severe TBI, after a period of time, causes numerous neuropsychiatric and neurodegenerative problems with varying comorbidities; and brain homeostasis may never be restored. As a consequence of disrupted equilibrium, neuropathological changes such as circuit remodeling, reorganization of neural networks, changes in structural and functional plasticity, predisposition to synchronized activity, and post-translational modification of synaptic proteins may begin to dominate the brain. These pathological changes, over the course of time, contribute to conditions like Alzheimer disease, dementia, anxiety disorders, and post-traumatic epilepsy (PTE). PTE is one of the most common, devastating complications of TBI; and of those affected by a severe TBI, more than 50% develop PTE. The etiopathology and mechanisms of PTE are either unknown or poorly understood, which makes treatment challenging. Although anti-epileptic drugs (AEDs) are used as preventive strategies to manage TBI, control acute seizures and prevent development of PTE, their efficacy in PTE remains controversial. In this review, we discuss novel mechanisms and risk factors underlying PTE. We also discuss dysfunctions of neurovascular unit, cell-specific neuroinflammatory mediators and immune response factors that are vital for epileptogenesis after TBI. Finally, we describe current and novel treatments and management strategies for preventing PTE.
Collapse
Affiliation(s)
- Shaunik Sharma
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Grant Tiarks
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Joseph Haight
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
15
|
Lee TH, Christie BR, van Praag H, Lin K, Siu PMF, Xu A, So KF, Yau SY. AdipoRon Treatment Induces a Dose-Dependent Response in Adult Hippocampal Neurogenesis. Int J Mol Sci 2021; 22:2068. [PMID: 33669795 PMCID: PMC7922380 DOI: 10.3390/ijms22042068] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022] Open
Abstract
AdipoRon, an adiponectin receptor agonist, elicits similar antidiabetic, anti-atherogenic, and anti-inflammatory effects on mouse models as adiponectin does. Since AdipoRon can cross the blood-brain barrier, its chronic effects on regulating hippocampal function are yet to be examined. This study investigated whether AdipoRon treatment promotes hippocampal neurogenesis and spatial recognition memory in a dose-dependent manner. Adolescent male C57BL/6J mice received continuous treatment of either 20 mg/kg (low dose) or 50 mg/kg (high dose) AdipoRon or vehicle intraperitoneally for 14 days, followed by the open field test to examine anxiety and locomotor activity, and the Y maze test to examine hippocampal-dependent spatial recognition memory. Immunopositive cell markers of neural progenitor cells, immature neurons, and newborn cells in the hippocampal dentate gyrus were quantified. Immunosorbent assays were used to measure the serum levels of factors that can regulate hippocampal neurogenesis, including adiponectin, brain-derived neurotrophic factor (BDNF), and corticosterone. Our results showed that 20 mg/kg AdipoRon treatment significantly promoted hippocampal cell proliferation and increased serum levels of adiponectin and BDNF, though there were no effects on spatial recognition memory and locomotor activity. On the contrary, 50 mg/kg AdipoRon treatment impaired spatial recognition memory, suppressed cell proliferation, neuronal differentiation, and cell survival associated with reduced serum levels of BDNF and adiponectin. The results suggest that a low-dose AdipoRon treatment promotes hippocampal cell proliferation, while a high-dose AdipoRon treatment is detrimental to the hippocampus function.
Collapse
Affiliation(s)
- Thomas H. Lee
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong;
| | - Brian R. Christie
- Division of Biomedical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Henriette van Praag
- FAU Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33431, USA;
| | - Kangguang Lin
- Department of Affective Disorder, Guangzhou Brain Hospital, The Brain Affiliated Hospital of Guangzhou Medical University, Guangzhou 510370, China;
| | - Parco Ming-Fai Siu
- Division of Kinesiology, School of Public Health, The University of Hong Kong, Hong Kong;
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong;
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
- The State Key Laboratory of Pharmacology, The University of Hong Kong, Hong Kong
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China;
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
- Department of Ophthalmology, The University of Hong Kong, Hong Kong
| | - Suk-yu Yau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong;
| |
Collapse
|
16
|
Guo Y, Chen Y, Yang M, Xu X, Lin Z, Ma J, Chen H, Hu Y, Ma Y, Wang X, Tian X. A Rare KIF1A Missense Mutation Enhances Synaptic Function and Increases Seizure Activity. Front Genet 2020; 11:61. [PMID: 32174959 PMCID: PMC7056823 DOI: 10.3389/fgene.2020.00061] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
Although genetic factors are considered a main etiology of epilepsy, the causes of genetic epilepsy in the majority of epilepsy patients remain unknown. Kinesin family member 1A (KIF1A), a neuron-specific motor protein that moves along with microtubules, is responsible for the transport of membranous organelles and synaptic vesicles. Variants of KIF1A have recently been associated with hereditary spastic paraplegia (HSP), hereditary sensory and autonomic neuropathy type 2 (HSANII), and intellectual disability. However, mutations in KIF1A have not been detected in patients with epilepsy. In our study, we conducted customized sequencing of epilepsy-related genes of a family with six patients with generalized epilepsy over three generations and identified a rare heterozygous mutation (c.1190C > A, p. Ala397Asp) in KIF1A. Whole-cell recordings from primary cultured neurons revealed that the mutant KIF1A increases the excitatory synaptic transmission but not the intrinsic excitability of neurons, and phenotype testing in zebrafish showed that this rare mutation results in epileptic seizure-like activity. These results provide new evidence demonstrating that KIF1A dysfunction is involved in epileptogenesis.
Collapse
Affiliation(s)
- Yi Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuanyuan Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Min Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xin Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Zijun Lin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Junhong Ma
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Hongnian Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yida Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
17
|
Mohamed HK, Eltony SA. Effect of acute pentylenetetrazol injection induced epileptic seizures on rat dentate gyrus at different postnatal ages. Anat Cell Biol 2020; 53:84-94. [PMID: 32274253 PMCID: PMC7118254 DOI: 10.5115/acb.19.083] [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: 04/10/2019] [Revised: 06/30/2019] [Accepted: 09/20/2019] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is one of the most common neurological disorders, its prevalence approximately from 0.5% to 2% of the general population. Generalized seizures could lead to several morphological changes in the brain. This study aimed to investigate the morphological effects of a single convulsive dose of pentylenetetrazol (PTZ) on rat dentate gyrus at different postnatal ages. Thirty-six male Wistar rats were used at the following postnatal ages: P10, P21, and P90 (12 rats per each age). The animals in each age were equally divided into two groups: group I, control and group II, treated with a single intraperitoneal injection of PTZ (55 mg/kg). After confirmation of generalized tonic-clonic seizures, specimens from the right dentate gyrus were processed for light and electron microscopy. In PTZ-treated groups, the number of granule cells significantly decreased. Dark granule cells appeared in the deep layers of the granule cells in P10 and with the progress of age, they significantly increased in number and extended into the superficial layers of the granule cells. The dendritic spines diminished. Glial fibrillary acidic protein and caspase-3 expression increased. Ultrastructurally, granule cells showed irregular shaped nucleus, dilated rough endoplasmic reticulum (RER) cisternae, mitochondria with damaged cristae, large vacuoles, lysosomes, and lipofuscin granules. Dark granule cells characterized by electron-dense nucleus and cytoplasm containing disorganized Golgi bodies, swollen mitochondria with damaged cristae, numerous free ribosomes and few long strands of RER. Astrocytes had hypertrophied cell body. Acute treatment with PTZ-induced epileptic seizures caused toxic effect on the structure of rat dentate gyrus at different postnatal ages.
Collapse
Affiliation(s)
- Heba K Mohamed
- Department of Anatomy, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sohair A Eltony
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
18
|
Expression Analysis of lncRNAs in Refractory and Non-Refractory Epileptic Patients. J Mol Neurosci 2020; 70:689-698. [PMID: 31900886 DOI: 10.1007/s12031-019-01477-8] [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: 11/22/2019] [Accepted: 12/26/2019] [Indexed: 01/03/2023]
Abstract
Long non-coding RNAs (lncRNAs) have been demonstrated to be involved in the pathogenesis of neuropsychiatric disorders such as epilepsy. In the current study, we evaluated expression of eight lncRNAs in 80 epileptic patients (40 refractory and 40 non-refractory ones) and 40 normal individual using quantitative real-time PCR. Bayesian regression model showed significant higher expression of UCA1 in both refractory and non-refractory groups compared with controls (posterior beta of relative expression (RE) = 2.03, P value = 0.003, and posterior beta of RE = 4.05, P value < 0.0001, respectively). Besides, expression of UCA1 was higher in non-refractory patients compared with refractory ones (posterior beta of RE = 2.008, P value = 0.019). When repeating statistical analyses in a gender-based manner, differences in expression of UCA1 were significant in all subgroup analyses except for male non-refractory vs. refractory subgroups analysis. Expression levels of NKILA and ANRIL were higher in both refractory and non-refractory groups compared with controls (posterior beta of RE = 1.565, P value = 0.018, and posterior beta of RE = 1.902, P value = 0.006 for NKILA; posterior beta of RE = 1.304, P value < 0.0001, and posterior beta of RE = 1.603, P value = 0.019 for ANRIL, respectively). However, expression levels of these two lncRNAs were not different between refractory and non-refractory groups. Gender-based analysis for these two lncRNAs revealed similar results except for lack of difference in ANRIL expression between male refractory group and controls. Expression of THRIL was significantly lower in both refractory and non-refractory groups compared with controls (posterior beta of RE = - 0.842, P value = 0.044 and posterior beta of RE = - 1.969, P value < 0.0001, respectively). Furthermore, expression of this lncRNA was lower in non-refractory patients compared with refractory ones (posterior beta of RE = - 1.129, P value = 0.002). However, no significant difference was detected between non-refractory and refractory patients either in males or females. The interactions between gender and relative expressions of PACER, DILC, and MALAT1 were significant, so the results were assessed in gender-based manner. In females, expression of DILC was higher in non-refractory patients compared with refractory ones (posterior beta of RE = 0.959, P value = 0.044). Expression of MALAT1 was lower in female non-refractory patients compared with controls and in female non-refractory patients compared with refractory ones (posterior beta of RE = - 1.35, P value = 0.002, and posterior beta of RE = - 0.942, P value = 0.045, respectively). Finally, expression of PACER was higher in refractory patients vs. controls and non-refractory patients vs. controls in both male and female subgroups. However, comparison between non-refractory and refractory patients revealed significant results only among females. Expression of none of the assessed lncRNAs was correlated with age of study participants. There were robust correlations between expression levels of lncRNAs. The most robust correlations were detected between UCA1 and PACER (r = 0.84, P < 0.0001) and between UCA1 and ANRIL (r = 0.75, P < 0.0001). Taken together, our study demonstrated dysregulation of lncRNAs in peripheral blood of epileptic patients and potentiated them as biomarkers for this neurologic condition.
Collapse
|
19
|
Kang SK, Ammanuel S, Adler DA, Kadam SD. Rescue of PB-resistant neonatal seizures with single-dose of small-molecule TrkB antagonist show long-term benefits. Epilepsy Res 2020; 159:106249. [PMID: 31864171 PMCID: PMC6953748 DOI: 10.1016/j.eplepsyres.2019.106249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022]
Abstract
A recently characterized CD-1 mouse model of phenobarbital (PB)-resistant neonatal ischemic-seizures (i.e.; unilateral carotid ligation) was shown to be associated with age-dependent (P7 vs. P10) acute seizure severity and PB-efficacy (i.e.; PB-resistant vs. PB-responsive). ANA12, a novel small-molecule TrkB antagonist, rescued the PB-resistance at P7 in a dose-dependent manner and prevented the post-ischemic downregulation of KCC2, the chief Cl- extruder in neurons. The long-term consequences of this novel rescue-intervention with ANA12 + PB in P7 and P10 ligated pups was investigated and compared to the standard first-line protocol of PB-alone loading dose. The mice underwent neurobehavioral testing, 24 h video-EEG-EMG monitoring, and immunohistochemistry in ipsi- and contralateral cortices as adults following the neonatal interventions. ANA12 + PB rescued the emergence of hyperactivity in post-ischemic P7, but not in P10 pups as adults. ANA12 + PB administration at neither P7 nor P10 significantly altered 24 h macro-sleep architecture in adults when compared to PB-alone. Behavioral state-dependent gamma (35-50 Hz) power homeostasis showed the most significant between-group differences that were age-dependent. ANA12 + PB treatment, but not PB-alone, rescued the loss of gamma power homeostasis present in P7 ligate-control but absent in P10 ligate group, highlighting the age-dependence. In contrast, PB-alone treatment, but not ANA12+PB, significantly reduced the elevated delta-AUC observed in P10 ligate-controls, when PB is efficacious by itself. These results indicate that the rescue of acute PB-resistant neonatal seizures using a novel intervention positively modulates the long-term outcomes at P7 when the seizures are refractory.
Collapse
Affiliation(s)
- S K Kang
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
| | - S Ammanuel
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - D A Adler
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - S D Kadam
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA; Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
20
|
Abstract
The structure of neuronal circuits that subserve cognitive functions in the brain is shaped and refined throughout development and into adulthood. Evidence from human and animal studies suggests that the cellular and synaptic substrates of these circuits are atypical in neuropsychiatric disorders, indicating that altered structural plasticity may be an important part of the disease biology. Advances in genetics have redefined our understanding of neuropsychiatric disorders and have revealed a spectrum of risk factors that impact pathways known to influence structural plasticity. In this Review, we discuss the importance of recent genetic findings on the different mechanisms of structural plasticity and propose that these converge on shared pathways that can be targeted with novel therapeutics.
Collapse
|
21
|
Shah BS, Shimell JJ, Bamji SX. Regulation of dendrite morphology and excitatory synapse formation by zDHHC15. J Cell Sci 2019; 132:jcs.230052. [PMID: 31189538 PMCID: PMC6633394 DOI: 10.1242/jcs.230052] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
Protein palmitoylation is the most common post-translational lipid modification in the brain and is mediated by a family of 24 zDHHC enzymes. There has been growing interest in zDHHCs due to mounting evidence that these enzymes play key roles in the development and function of neuronal connections, and the fact that a number of zDHHCs have been associated with neurodevelopmental and neurodegenerative diseases. Loss-of-function variants in several zDHHCs, including zDHHC15, have been identified in patients with intellectual disabilities; however, the function of zDHHC15 in the brain has not been well studied. Here, we demonstrate that knocking down zDHHC15 in primary rat hippocampal cultures reduces dendritic outgrowth and arborization, as well as spine maturation. Moreover, knockdown of zDHHC15 reduces palmitoylation of PSD-95 and its trafficking into dendrites, resulting in an overall decrease in the density of excitatory synapses being formed onto mutant cells. Summary: Disruption of zDHHC15 function in primary hippocampal cultures reduces dendritic outgrowth, spine maturation, excitatory synapse density and trafficking of PSD-95 into dendrites.
Collapse
Affiliation(s)
- Bhavin S Shah
- Department of Cellular & Physiological Sciences & the Brain Research Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada
| | - Jordan J Shimell
- Department of Cellular & Physiological Sciences & the Brain Research Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada
| | - Shernaz X Bamji
- Department of Cellular & Physiological Sciences & the Brain Research Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada
| |
Collapse
|
22
|
Comorbidities of early-onset temporal epilepsy: Cognitive, social, emotional, and morphologic dimensions. Exp Neurol 2019; 320:113005. [PMID: 31278943 DOI: 10.1016/j.expneurol.2019.113005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/16/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023]
Abstract
Epilepsy, the most common neurologic disorder in childhood, is associated with a subset of psychiatric dysfunctions, including cognitive deficits, and alterations in emotionality (e.g., anxiety and depression) and social functioning. In the present study, we evaluated an integrative set of behavioral responses, including cognitive/socio-cognitive and emotional dimensions, using a number of behavioral paradigms in the LiCl/pilocarpine model of status epilepticus (SE) in rats. The aims of the study were to examine whether SE affects: 1) non-associative learning (habituation of exploratory behavior); 2) investigatory response to an indifferent stimulus object; 3) sociability/social novelty preference; 4) social recognition or discrimination; and 4) short- and long-term memory in the Morris water maze (MWM). Finally, we investigated the morphology of key brain structures involved in the examined behavioral dysfunctions. SE did not affect habituation to an open-field arena in juvenile (P25), adolescent (P32), or adult (P80) rats. SE rats spent less time in the central part of the arena. SE adolescent rats (P32) displayed a higher number of rearings with a shorter duration. SE rats displayed a markedly attenuated investigatory response to an indifferent stimulus object. SE rats in all age groups demonstrated pronounced deficits in sociability and the preference for social novelty. In addition, SE rats spent a reduced amount of time investigating a juvenile rat upon first exposure. After 30 min re-exposure together with an additional, novel juvenile, the SE rats spent equal time investigating both juveniles. In the MWM task, acquisition was unimpaired but there was a deficit in delayed memory retention after 10 days. SE did not affect cognitive flexibility expressed by reversal learning. Together, these findings suggest that early-life SE leads to alterations in emotional/anxiety-related behavior and affects sociability/preference for social novelty and social discrimination. Early-life SE did not alter acquisition of spatial learning, but it impaired delayed retention. Using Fluoro Jade B staining performed 24 h after SE revealed apparent neurodegeneration in the dorsal hippocampus, mediodorsal thalamic nucleus and medial amygdala, brain areas that are critically involved in network underlying emotional behavior and cognitive functions.
Collapse
|
23
|
Yu X, Yang L, Li J, Li W, Li D, Wang R, Wu K, Chen W, Zhang Y, Qiu Z, Zhou W. De Novo and Inherited SETD1A Variants in Early-onset Epilepsy. Neurosci Bull 2019; 35:1045-1057. [PMID: 31197650 DOI: 10.1007/s12264-019-00400-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022] Open
Abstract
Early-onset epilepsy is a neurological abnormality in childhood, and it is especially common in the first 2 years after birth. Seizures in early life mostly result from structural or metabolic disorders in the brain, and the genetic causes of idiopathic seizures have been extensively investigated. In this study, we identified four missense mutations in the SETD1A gene (SET domain-containing 1A, histone lysine methyltransferase): three de novo mutations in three individuals and one inherited mutation in a four-generation family. Whole-exome sequencing indicated that all four of these mutations were responsible for the seizures. Mutations of SETD1A have been implicated in schizophrenia and developmental disorders, so we examined the role of the four mutations (R913C, Q269R, G1369R, and R1392H) in neural development. We found that their expression in mouse primary cortical neurons affected excitatory synapse development. Moreover, expression of the R913C mutation also affected the migration of cortical neurons in the mouse brain. We further identified two common genes (Neurl4 and Usp39) affected by mutations of SETD1A. These results suggested that the mutations of SETD1A play a fundamental role in abnormal synaptic function and the development of neurons, so they may be pathogenic factors for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiuya Yu
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Lin Yang
- Clinical Genetic Center, Children's Hospital of Fudan University, Shanghai, 201102, China.,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jin Li
- Institute of Biomedicine Sciences, Fudan University, Shanghai, 200032, China
| | - Wanxing Li
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Dongzhi Li
- Department of Prenatal Diagnosis, The Women and Children's Medical Center, Guangzhou, 510623, China
| | - Ran Wang
- Euler Genomics, Beijing, 102206, China
| | - Kai Wu
- Euler Genomics, Beijing, 102206, China
| | | | - Yi Zhang
- Euler Genomics, Beijing, 102206, China. .,School of Life Sciences, Peking University, Beijing, 100191, China.
| | - Zilong Qiu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Wenhao Zhou
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
24
|
Glass HC, Grinspan ZM, Shellhaas RA. Outcomes after acute symptomatic seizures in neonates. Semin Fetal Neonatal Med 2018; 23:218-222. [PMID: 29454756 DOI: 10.1016/j.siny.2018.02.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acute symptomatic seizures are a common sign of neurological dysfunction and brain injury in neonates and occur in approximately one to three per 1000 live births. Seizures in neonates are usually a sign of underlying brain injury and, as such, are commonly associated with adverse outcomes. Neurological morbidities in survivors often co-occur; epilepsy, cerebral palsy, and intellectual disability often occur together in the most severely affected children. Risk factors for adverse outcome include prematurity, low Apgar scores, low pH on the first day of life, seizure onset <24 or >72 h after birth, abnormal neonatal neurological examination, abnormal neonatal electroencephalographic background, status epilepticus, and presence and pattern of brain injury (particularly deep gray or brainstem injury). Despite this list of potential indicators, accurate prediction of outcome in a given child remains challenging. There is great need for long-term, multicenter studies to examine risk factors for, and pathogenesis of, adverse outcomes following acute symptomatic seizures in neonates.
Collapse
Affiliation(s)
- Hannah C Glass
- Department of Neurology, Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| | - Zachary M Grinspan
- Department of Healthcare Policy, Department of Research and Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Renée A Shellhaas
- Department of Pediatrics, Department of Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
25
|
Silva-Gómez AB, Bravo-Duran DA, Eguibar JR, Cortes C. Juvenile Taiep rats have shorter dendritic trees in the dorsal field of the hippocampus without spatial learning disabilities. Synapse 2018; 72:e22024. [PMID: 29323756 DOI: 10.1002/syn.22024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Myelin mutant taiep rats show a progressive demyelination in the central nervous system due to an abnormal accumulation of microtubules in the cytoplasm and the processes on their oligodendrocytes. Demyelination is associated with electrophysiological alterations and the mutant had a progressive astrocytosis. The illness is associated with change in cytokine levels and in the expression of different nitric oxide synthase and concomitantly lipoperoxidation in several areas of the brain. However, until now there has been no detailed anatomical analysis of neurons in this mutant. The aim of this study was to analyze the dendritic morphology in the hippocampus using Golgi-Cox staining and spatial memory through Morris water maze test in young adult (3 months old) taiep rats and compare them with normal Sprague-Dawley. Our results showed that taiep rats have altered dendritic tree morphology in pyramidal neurons in the CA1 field of the hippocampus, but not in the CA3 region. These morphological changes did not produce a concomitant deficit in spatial memory acquisition or recall at this early stage of the disease. Our results suggest that impairment of dendritic morphology in the CA1 field of the hippocampus is a landmark of the pathology of this progressive multiple sclerosis model.
Collapse
Affiliation(s)
| | | | - Jose R Eguibar
- Institute of Physiology, Benemérita Universidad Autónoma de Puebla, México.,Research Office, Vice-rectory of Research and Postgraduate Studies, Benemérita Universidad Autónoma de Puebla, México
| | - Carmen Cortes
- Institute of Physiology, Benemérita Universidad Autónoma de Puebla, México
| |
Collapse
|
26
|
Rubio-Casillas A, Fernández-Guasti A. The dose makes the poison: from glutamate-mediated neurogenesis to neuronal atrophy and depression. Rev Neurosci 2018; 27:599-622. [PMID: 27096778 DOI: 10.1515/revneuro-2015-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022]
Abstract
Experimental evidence has demonstrated that glutamate is an essential factor for neurogenesis, whereas another line of research postulates that excessive glutamatergic neurotransmission is associated with the pathogenesis of depression. The present review shows that such paradox can be explained within the framework of hormesis, defined as biphasic dose responses. Low glutamate levels activate adaptive stress responses that include proteins that protect neurons against more severe stress. Conversely, abnormally high levels of glutamate, resulting from increased release and/or decreased removal, cause neuronal atrophy and depression. The dysregulation of the glutamatergic transmission in depression could be underlined by several factors including a decreased inhibition (γ-aminobutyric acid or serotonin) or an increased excitation (primarily within the glutamatergic system). Experimental evidence shows that the activation of N-methyl-D-aspartate receptor (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPAR) can exert two opposite effects on neurogenesis and neuron survival depending on the synaptic or extrasynaptic concentration. Chronic stress, which usually underlies experimental and clinical depression, enhances glutamate release. This overactivates NMDA receptors (NMDAR) and consequently impairs AMPAR activity. Various studies show that treatment with antidepressants decreases plasma glutamate levels in depressed individuals and regulates glutamate receptors by reducing NMDAR function by decreasing the expression of its subunits and by potentiating AMPAR-mediated transmission. Additionally, it has been shown that chronic treatment with antidepressants having divergent mechanisms of action (including tricyclics, selective serotonin reuptake inhibitors, and ketamine) markedly reduced depolarization-evoked glutamate release in the hippocampus. These data, taken together, suggest that the glutamatergic system could be a final common pathway for antidepressant treatments.
Collapse
|
27
|
Sebastián-Serrano Á, Engel T, de Diego-García L, Olivos-Oré LA, Arribas-Blázquez M, Martínez-Frailes C, Pérez-Díaz C, Millán JL, Artalejo AR, Miras-Portugal MT, Henshall DC, Díaz-Hernández M. Neurodevelopmental alterations and seizures developed by mouse model of infantile hypophosphatasia are associated with purinergic signalling deregulation. Hum Mol Genet 2016; 25:4143-4156. [PMID: 27466191 PMCID: PMC5291194 DOI: 10.1093/hmg/ddw248] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/14/2016] [Accepted: 07/17/2016] [Indexed: 01/14/2023] Open
Abstract
Hypomorphic mutations in the gene encoding the tissue-nonspecific alkaline phosphatase (TNAP) enzyme, ALPL in human or Akp2 in mice, cause hypophosphatasia (HPP), an inherited metabolic bone disease also characterized by spontaneous seizures. Initially, these seizures were attributed to the impairment of GABAergic neurotransmission caused by altered vitamin B6 (vit-B6) metabolism. However, clinical cases in human newborns and adults whose convulsions are refractory to pro-GABAergic drugs but controlled by the vit-B6 administration, suggest that other factors are involved. Here, to evaluate whether neurodevelopmental alterations are underlying the seizures associated to HPP, we performed morphological and functional characterization of postnatal homozygous TNAP null mice, a model of HPP. These analyses revealed that TNAP deficient mice present an increased proliferation of neural precursors, an altered neuronal morphology, and an augmented neuronal activity. We found that these alterations were associated with a partial downregulation of the purinergic P2X7 receptor (P2X7R). Even though deficient P2X7R mice present similar neurodevelopmental alterations, they do not develop neonatal seizures. Accordingly, we found that the additional blockage of P2X7R prevent convulsions and extend the lifespan of mice lacking TNAP. In agreement with these findings, we also found that exogenous administration of ATP or TNAP antagonists induced seizures in adult wild-type mice by activating P2X7R. Finally, our results also indicate that the anticonvulsive effects attributed to vit-B6 may be due to its capacity to block P2X7R. Altogether, these findings suggest that the purinergic signalling regulates the neurodevelopmental alteration and the neonatal seizures associated to HPP.
Collapse
Affiliation(s)
- Álvaro Sebastián-Serrano
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Laura de Diego-García
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Luis A Olivos-Oré
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain and
| | - Marina Arribas-Blázquez
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain and
| | - Carlos Martínez-Frailes
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Carmen Pérez-Díaz
- Department of Medicine and Animal Surgery, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Antonio R Artalejo
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain and
| | - María Teresa Miras-Portugal
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork, Ireland
| | - Miguel Díaz-Hernández
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, Spain .,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| |
Collapse
|
28
|
Awad PN, Sanon NT, Chattopadhyaya B, Carriço JN, Ouardouz M, Gagné J, Duss S, Wolf D, Desgent S, Cancedda L, Carmant L, Di Cristo G. Reducing premature KCC2 expression rescues seizure susceptibility and spine morphology in atypical febrile seizures. Neurobiol Dis 2016; 91:10-20. [PMID: 26875662 DOI: 10.1016/j.nbd.2016.02.014] [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: 12/15/2015] [Revised: 01/26/2016] [Accepted: 02/09/2016] [Indexed: 11/23/2022] Open
Abstract
Atypical febrile seizures are considered a risk factor for epilepsy onset and cognitive impairments later in life. Patients with temporal lobe epilepsy and a history of atypical febrile seizures often carry a cortical malformation. This association has led to the hypothesis that the presence of a cortical dysplasia exacerbates febrile seizures in infancy, in turn increasing the risk for neurological sequelae. The mechanisms linking these events are currently poorly understood. Potassium-chloride cotransporter KCC2 affects several aspects of neuronal circuit development and function, by modulating GABAergic transmission and excitatory synapse formation. Recent data suggest that KCC2 downregulation contributes to seizure generation in the epileptic adult brain, but its role in the developing brain is still controversial. In a rodent model of atypical febrile seizures, combining a cortical dysplasia and hyperthermia-induced seizures (LHS rats), we found a premature and sustained increase in KCC2 protein levels, accompanied by a negative shift of the reversal potential of GABA. In parallel, we observed a significant reduction in dendritic spine size and mEPSC amplitude in CA1 pyramidal neurons, accompanied by spatial memory deficits. To investigate whether KCC2 premature overexpression plays a role in seizure susceptibility and synaptic alterations, we reduced KCC2 expression selectively in hippocampal pyramidal neurons by in utero electroporation of shRNA. Remarkably, KCC2 shRNA-electroporated LHS rats show reduced hyperthermia-induced seizure susceptibility, while dendritic spine size deficits were rescued. Our findings demonstrate that KCC2 overexpression in a compromised developing brain increases febrile seizure susceptibility and contribute to dendritic spine alterations.
Collapse
Affiliation(s)
- Patricia N Awad
- Neurosciences Department, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec H3T 1N8, Canada; CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Nathalie T Sanon
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Bidisha Chattopadhyaya
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Josianne Nunes Carriço
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Mohamed Ouardouz
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Jonathan Gagné
- Neurosciences Department, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec H3T 1N8, Canada; CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Sandra Duss
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Daniele Wolf
- Neurosciences Department, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec H3T 1N8, Canada; CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Sébastien Desgent
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada
| | - Laura Cancedda
- Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Lionel Carmant
- Neurosciences Department, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec H3T 1N8, Canada; CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada.
| | - Graziella Di Cristo
- Neurosciences Department, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec H3T 1N8, Canada; CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Sainte-Catherine, Montréal, Québec H3T 1C5, Canada.
| |
Collapse
|
29
|
Fauth M, Tetzlaff C. Opposing Effects of Neuronal Activity on Structural Plasticity. Front Neuroanat 2016; 10:75. [PMID: 27445713 PMCID: PMC4923203 DOI: 10.3389/fnana.2016.00075] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 06/16/2016] [Indexed: 12/21/2022] Open
Abstract
The connectivity of the brain is continuously adjusted to new environmental influences by several activity-dependent adaptive processes. The most investigated adaptive mechanism is activity-dependent functional or synaptic plasticity regulating the transmission efficacy of existing synapses. Another important but less prominently discussed adaptive process is structural plasticity, which changes the connectivity by the formation and deletion of synapses. In this review, we show, based on experimental evidence, that structural plasticity can be classified similar to synaptic plasticity into two categories: (i) Hebbian structural plasticity, which leads to an increase (decrease) of the number of synapses during phases of high (low) neuronal activity and (ii) homeostatic structural plasticity, which balances these changes by removing and adding synapses. Furthermore, based on experimental and theoretical insights, we argue that each type of structural plasticity fulfills a different function. While Hebbian structural changes enhance memory lifetime, storage capacity, and memory robustness, homeostatic structural plasticity self-organizes the connectivity of the neural network to assure stability. However, the link between functional synaptic and structural plasticity as well as the detailed interactions between Hebbian and homeostatic structural plasticity are more complex. This implies even richer dynamics requiring further experimental and theoretical investigations.
Collapse
Affiliation(s)
- Michael Fauth
- Department of Computational Neuroscience, Third Institute of Physics - Biophysics, Georg-August UniversityGöttingen, Germany; Bernstein Center for Computational NeuroscienceGöttingen, Germany
| | - Christian Tetzlaff
- Bernstein Center for Computational NeuroscienceGöttingen, Germany; Max Planck Institute for Dynamics and Self-OrganizationGöttingen, Germany
| |
Collapse
|
30
|
Neonatal seizures induced by pentylenetetrazol or kainic acid disrupt primary cilia growth on developing mouse cortical neurons. Exp Neurol 2016; 282:119-27. [PMID: 27181411 DOI: 10.1016/j.expneurol.2016.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 04/19/2016] [Accepted: 05/11/2016] [Indexed: 11/23/2022]
Abstract
Neonatal or early-life seizures (ELS) are often associated with life-long neurophysiological, cognitive and behavioral deficits, but the underlying mechanisms contributing to these deficits remain poorly understood. Newborn, post-migratory cortical neurons sprout ciliary buds (procilia) that mature into primary cilia. Disruption of the growth or signaling capabilities of these cilia has been linked to atypical neurite outgrowth from neurons and abnormalities in neuronal circuitry. Here, we tested the hypothesis that generalized seizures induced by pentylenetetrazol (PTZ) or kainic acid (KA) during early postnatal development impair neuronal and/or glial ciliogenesis. Mice received PTZ (50 or 100mg/kg), KA (2mg/kg), or saline either once at birth (P0), or once daily from P0 to P4. Using immunohistochemistry and electron microscopy, the cilia of neurons and glia were examined at P7, P14, and P42. A total of 83 regions were analyzed, representing 13 unique neocortical and hippocampal regions. Neuronal cilia were identified by co-expression of NeuN and type 3 adenylyl cyclase (ACIII) or somatostatin receptor 3 (SSTR3), while glial cilia were identified by co-expression of GFAP, Arl13b, and gamma-tubulin. We found that PTZ exposure at either P0 or from P0 to P4 induced convulsive behavior, followed by acute and lasting effects on neuronal cilia lengths that varied depending on the cortical region, PTZ dose, injection frequency, and time post-PTZ. Both increases and decreases in neuronal cilia length were observed. No changes in the length of glial cilia were observed under any of the test conditions. Lastly, we found that a single KA seizure at P0 led to similar abnormalities in neuronal cilia lengths. Our results suggest that seizure(s) occurring during early stages of cortical development induce persistent and widespread changes in neuronal cilia length. Given the impact neuronal cilia have on neuronal differentiation, ELS-induced changes in ciliogenesis may contribute to long-term pathology and abnormal cortical function.
Collapse
|
31
|
Pascente R, Frigerio F, Rizzi M, Porcu L, Boido M, Davids J, Zaben M, Tolomeo D, Filibian M, Gray WP, Vezzani A, Ravizza T. Cognitive deficits and brain myo-Inositol are early biomarkers of epileptogenesis in a rat model of epilepsy. Neurobiol Dis 2016; 93:146-55. [PMID: 27173096 DOI: 10.1016/j.nbd.2016.05.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/26/2016] [Accepted: 05/05/2016] [Indexed: 10/21/2022] Open
Abstract
One major unmet clinical need in epilepsy is the identification of therapies to prevent or arrest epilepsy development in patients exposed to a potential epileptogenic insult. The development of such treatments has been hampered by the lack of non-invasive biomarkers that could be used to identify the patients at-risk, thereby allowing to design affordable clinical studies. Our goal was to test the predictive value of cognitive deficits and brain astrocyte activation for the development of epilepsy following a potential epileptogenic injury. We used a model of epilepsy induced by pilocarpine-evoked status epilepticus (SE) in 21-day old rats where 60-70% of animals develop spontaneous seizures after around 70days, although SE is similar in all rats. Learning was evaluated in the Morris water-maze at days 15 and 65 post-SE, each time followed by proton magnetic resonance spectroscopy for measuring hippocampal myo-Inositol levels, a marker of astrocyte activation. Rats were video-EEG monitored for two weeks at seven months post-SE to detect spontaneous seizures, then brain histology was done. Behavioral and imaging data were retrospectively analysed in epileptic rats and compared with non-epileptic and control animals. Rats displayed spatial learning deficits within three weeks from SE. However, only epilepsy-prone rats showed accelerated forgetting and reduced learning rate compared to both rats not developing epilepsy and controls. These deficits were associated with reduced hippocampal neurogenesis. myo-Inositol levels increased transiently in the hippocampus of SE-rats not developing epilepsy while this increase persisted until spontaneous seizures onset in epilepsy-prone rats, being associated with a local increase in S100β-positive astrocytes. Neuronal cell loss was similar in all SE-rats. Our data show that behavioral deficits, together with a non-invasive marker of astrocyte activation, predict which rats develop epilepsy after an acute injury. These measures have potential clinical relevance for identifying individuals at-risk for developing epilepsy following exposure to epileptogenic insults, and consequently, for designing adequately powered antiepileptogenesis trials.
Collapse
Affiliation(s)
- Rosaria Pascente
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Federica Frigerio
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Massimo Rizzi
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Luca Porcu
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Marina Boido
- Neuroscience Institute "Cavalieri Ottolenghi", Department of Neuroscience, University of Torino, Torino, Italy
| | - Joe Davids
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Malik Zaben
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Daniele Tolomeo
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Marta Filibian
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - William P Gray
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Teresa Ravizza
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy.
| |
Collapse
|
32
|
Holley AJ, Lugo JN. Effects of an acute seizure on associative learning and memory. Epilepsy Behav 2016; 54:51-7. [PMID: 26655449 PMCID: PMC4724501 DOI: 10.1016/j.yebeh.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 01/31/2023]
Abstract
Past studies have demonstrated that inducing several seizures or continuous seizures in neonatal or adult rats results in impairments in learning and memory. The impact of a single acute seizure on learning and memory has not been investigated in mice. In this study, we exposed adult 129SvEvTac mice to the inhalant flurothyl until a behavioral seizure was induced. Our study consisted of 4 experiments where we examined the effect of one seizure before or after delay fear conditioning. We also included a separate cohort of animals that was tested in the open field after a seizure to rule out changes in locomotor activity influencing the results of memory tests. Mice that had experienced a single seizure 1h, but not 6h, prior to training showed a significant impairment in associative conditioning to the conditioned stimulus when compared with controls 24h later. There were no differences in freezing one day later for animals that experienced a single seizure 1h after associative learning. We also found that an acute seizure reduced activity levels in an open-field test 2h but not 24h later. These findings suggest that an acute seizure occurring immediately before learning can have an effect on the recall of events occurring shortly after that seizure. In contrast, an acute seizure occurring shortly after learning appears to have little or no effect on long-term memory. These findings have implications for understanding the acute effects of seizures on the acquisition of new knowledge.
Collapse
Affiliation(s)
- Andrew J. Holley
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA
| | - Joaquin N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA,Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
| |
Collapse
|
33
|
Gangarossa G, Sakkaki S, Lory P, Valjent E. Mouse hippocampal phosphorylation footprint induced by generalized seizures: Focus on ERK, mTORC1 and Akt/GSK-3 pathways. Neuroscience 2015; 311:474-83. [PMID: 26545981 DOI: 10.1016/j.neuroscience.2015.10.051] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/27/2015] [Indexed: 02/03/2023]
Abstract
Exacerbated hippocampal activity has been associated to critical modifications of the intracellular signaling pathways. We have investigated rapid hippocampal adaptive responses induced by maximal electroshock seizure (MES). Here, we demonstrate that abnormal and exacerbated hippocampal activity induced by MES triggers specific and temporally distinct patterns of phosphorylation of extracellular signal-related kinase (ERK), mammalian target of rapamycin complex (mTORC) and Akt/glycogen synthase kinase-3 (Akt/GSK-3) pathways in the mouse hippocampus. While the ERK pathway is transiently activated, the mTORC1 cascade follows a rapid inhibition followed by a transient activation. This rebound of mTORC1 activity leads to the selective phosphorylation of p70S6K, which is accompanied by an enhanced phosphorylation of the ribosomal subunit S6. In contrast, the Akt/GSK-3 pathway is weakly altered. Finally, MES triggers a rapid upregulation of several plasticity-associated genes as a consequence exacerbated hippocampal activity. The results reported in the present study are reminiscent of the one observed in other models of generalized seizures, thus defining a common molecular footprint induced by intense and aberrant hippocampal activities.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier F-34094, France; Inserm U1191, Montpellier F-34094, France; Université de Montpellier, Montpellier F-34094, France.
| | - Sophie Sakkaki
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier F-34094, France; Inserm U1191, Montpellier F-34094, France; Université de Montpellier, Montpellier F-34094, France
| | - Philippe Lory
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier F-34094, France; Inserm U1191, Montpellier F-34094, France; Université de Montpellier, Montpellier F-34094, France; LabEx 'Ion Channel Science and Therapeutics', Montpellier F-34094, France
| | - Emmanuel Valjent
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier F-34094, France; Inserm U1191, Montpellier F-34094, France; Université de Montpellier, Montpellier F-34094, France.
| |
Collapse
|
34
|
Travaglione S, Ballan G, Fortuna A, Ferri A, Guidotti M, Campana G, Fiorentini C, Loizzo S. CNF1 Enhances Brain Energy Content and Counteracts Spontaneous Epileptiform Phenomena in Aged DBA/2J Mice. PLoS One 2015; 10:e0140495. [PMID: 26457896 PMCID: PMC4601759 DOI: 10.1371/journal.pone.0140495] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/25/2015] [Indexed: 11/19/2022] Open
Abstract
Epilepsy, one of the most common conditions affecting the brain, is characterized by neuroplasticity and brain cell energy defects. In this work, we demonstrate the ability of the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1) to counteract epileptiform phenomena in inbred DBA/2J mice, an animal model displaying genetic background with an high susceptibility to induced- and spontaneous seizures. Via modulation of the Rho GTPases, CNF1 regulates actin dynamics with a consequent increase in spine density and length in pyramidal neurons of rat visual cortex, and influences the mitochondrial homeostasis with remarkable changes in the mitochondrial network architecture. In addition, CNF1 improves cognitive performances and increases ATP brain content in mouse models of Rett syndrome and Alzheimer's disease. The results herein reported show that a single dose of CNF1 induces a remarkable amelioration of the seizure phenotype, with a significant augmentation in neuroplasticity markers and in cortex mitochondrial ATP content. This latter effect is accompanied by a decrease in the expression of mitochondrial fission proteins, suggesting a role of mitochondrial dynamics in the CNF1-induced beneficial effects on this epileptiform phenotype. Our results strongly support the crucial role of brain energy homeostasis in the pathogenesis of certain neurological diseases, and suggest that CNF1 could represent a putative new therapeutic tool for epilepsy.
Collapse
Affiliation(s)
- Sara Travaglione
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Giulia Ballan
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Andrea Fortuna
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Alberto Ferri
- Institute of Cellular Biology and Neurobiology, CNR, Via del Fosso di Fiorano 64/65, 00143, Roma, Italy
| | - Marco Guidotti
- Department of Veterinary Public Health and Food Safety, Viale Regina Elena 299, 00161, Roma, Italy
| | - Gabriele Campana
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Carla Fiorentini
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Stefano Loizzo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
- * E-mail:
| |
Collapse
|
35
|
Sayin U, Hutchinson E, Meyerand ME, Sutula T. Age-dependent long-term structural and functional effects of early-life seizures: evidence for a hippocampal critical period influencing plasticity in adulthood. Neuroscience 2014; 288:120-134. [PMID: 25555928 DOI: 10.1016/j.neuroscience.2014.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 11/15/2014] [Accepted: 12/14/2014] [Indexed: 12/19/2022]
Abstract
Neural activity promotes circuit formation in developing systems and during critical periods permanently modifies circuit organization and functional properties. These observations suggest that excessive neural activity, as occurs during seizures, might influence developing neural circuitry with long-term outcomes that depend on age at the time of seizures. We systematically examined long-term structural and functional consequences of seizures induced in rats by kainic acid, pentylenetetrazol, and hyperthermia across postnatal ages from birth through postnatal day 90 in adulthood (P90). Magnetic resonance imaging (MRI), diffusion tensor imaging (DTI), and electrophysiological methods at ⩾P95 following seizures induced from P1 to P90 demonstrated consistent patterns of gross atrophy, microstructural abnormalities in the corpus callosum (CC) and hippocampus, and functional alterations in hippocampal circuitry at ⩾P95 that were independent of the method of seizure induction and varied systematically as a function of age at the time of seizures. Three distinct epochs were observed in which seizures resulted in distinct long-term structural and functional outcomes at ⩾P95. Seizures prior to P20 resulted in DTI abnormalities in CC and hippocampus in the absence of gross cerebral atrophy, and increased paired-pulse inhibition (PPI) in the dentate gyrus (DG) at ⩾P95. Seizures after P30 induced a different pattern of DTI abnormalities in the fimbria and hippocampus accompanied by gross cerebral atrophy with increases in lateral ventricular volume, as well as increased PPI in the DG at ⩾P95. In contrast, seizures between P20 and P30 did not result in cerebral atrophy or significant imaging abnormalities in the hippocampus or white matter, but irreversibly decreased PPI in the DG compared to normal adult controls. These age-specific long-term structural and functional outcomes identify P20-30 as a potential critical period in hippocampal development defined by distinctive long-term structural and functional properties in adult hippocampal circuitry, including loss of capacity for seizure-induced plasticity in adulthood that could influence epileptogenesis and other hippocampal-dependent behaviors and functional properties.
Collapse
Affiliation(s)
- U Sayin
- Department of Neurology, University of Wisconsin UW Medical Foundation Centennial Building 1685 Highland Ave Madison, WI 53705, USA
| | - E Hutchinson
- Department of Neurology, University of Wisconsin UW Medical Foundation Centennial Building 1685 Highland Ave Madison, WI 53705, USA.,Department of Medical Physics, University of Wisconsin Wisconsin Institutes Medical Research 1111 Highland Avenue Madison, WI 53705, USA
| | - M E Meyerand
- Department of Medical Physics, University of Wisconsin Wisconsin Institutes Medical Research 1111 Highland Avenue Madison, WI 53705, USA.,Department of Biomedical Engineering, University of Wisconsin Room 2130 Engineering Centers Building 1550 Engineering Drive Madison, WI 53706-1609, USA
| | - T Sutula
- Department of Neurology, University of Wisconsin UW Medical Foundation Centennial Building 1685 Highland Ave Madison, WI 53705, USA
| |
Collapse
|
36
|
Hester MS, Danzer SC. Hippocampal granule cell pathology in epilepsy - a possible structural basis for comorbidities of epilepsy? Epilepsy Behav 2014; 38:105-16. [PMID: 24468242 PMCID: PMC4110172 DOI: 10.1016/j.yebeh.2013.12.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/17/2013] [Accepted: 12/21/2013] [Indexed: 01/31/2023]
Abstract
Temporal lobe epilepsy in both animals and humans is characterized by abnormally integrated hippocampal dentate granule cells. Among other abnormalities, these cells make axonal connections with inappropriate targets, grow dendrites in the wrong direction, and migrate to ectopic locations. These changes promote the formation of recurrent excitatory circuits, leading to the appealing hypothesis that these abnormal cells may by epileptogenic. While this hypothesis has been the subject of intense study, less attention has been paid to the possibility that abnormal granule cells in the epileptic brain may also contribute to comorbidities associated with the disease. Epilepsy is associated with a variety of general findings, such as memory disturbances and cognitive dysfunction, and is often comorbid with a number of other conditions, including schizophrenia and autism. Interestingly, recent studies implicate disruption of common genes and gene pathways in all three diseases. Moreover, while neuropsychiatric conditions are associated with changes in a variety of brain regions, granule cell abnormalities in temporal lobe epilepsy appear to be phenocopies of granule cell deficits produced by genetic mouse models of autism and schizophrenia, suggesting that granule cell dysmorphogenesis may be a common factor uniting these seemingly diverse diseases. Disruption of common signaling pathways regulating granule cell neurogenesis may begin to provide mechanistic insight into the cooccurrence of temporal lobe epilepsy and cognitive and behavioral disorders.
Collapse
Affiliation(s)
- Michael S Hester
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesia, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
37
|
Stawarski M, Stefaniuk M, Wlodarczyk J. Matrix metalloproteinase-9 involvement in the structural plasticity of dendritic spines. Front Neuroanat 2014; 8:68. [PMID: 25071472 PMCID: PMC4091410 DOI: 10.3389/fnana.2014.00068] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/25/2014] [Indexed: 01/01/2023] Open
Abstract
Dendritic spines are the locus for excitatory synaptic transmission in the brain and thus play a major role in neuronal plasticity. The ability to alter synaptic connections includes volumetric changes in dendritic spines that are driven by scaffolds created by the extracellular matrix (ECM). Here, we review the effects of the proteolytic activity of ECM proteases in physiological and pathological structural plasticity. We use matrix metalloproteinase-9 (MMP-9) as an example of an ECM modifier that has recently emerged as a key molecule in regulating the morphology and dysmorphology of dendritic spines that underlie synaptic plasticity and neurological disorders, respectively. We summarize the influence of MMP-9 on the dynamic remodeling of the ECM via the cleavage of extracellular substrates. We discuss its role in the formation, modification, and maintenance of dendritic spines in learning and memory. Finally, we review research that implicates MMP-9 in aberrant synaptic plasticity and spine dysmorphology in neurological disorders, with a focus on morphological abnormalities of dendritic protrusions that are associated with epilepsy.
Collapse
Affiliation(s)
- Michal Stawarski
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mzowieckie, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| |
Collapse
|
38
|
Dendritic integration in pyramidal neurons during network activity and disease. Brain Res Bull 2014; 103:2-10. [DOI: 10.1016/j.brainresbull.2013.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/18/2013] [Accepted: 09/19/2013] [Indexed: 11/18/2022]
|
39
|
Abstract
Seizures occur in approximately 1 to 5 per 1000 live births and are among the most common neurologic conditions managed by a neonatal neurocritical care service. There are several, age-specific factors that are particular to the developing brain, which influence excitability and seizure generation, response to medications, and impact of seizures on brain structure and function. Neonatal seizures are often associated with serious underlying brain injury such as hypoxia-ischemia, stroke, or hemorrhage. Conventional, prolonged, continuous video electroencephalogram is the gold standard for detecting seizures, whereas amplitude-integrated EEG is a convenient and useful bedside tool.
Collapse
Affiliation(s)
- Hannah C. Glass
- Departments of Neurology and Pediatrics University of California, San Francisco, United States of America
| |
Collapse
|
40
|
How is homeostatic plasticity important in epilepsy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:123-31. [PMID: 25012372 DOI: 10.1007/978-94-017-8914-1_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Maintaining physiological variables within narrow operating limits by homeostatic mechanisms is a fundamental property of most if not all living cells and organisms. In recent years, research from many laboratories has shown that the activity of neurons and neural circuits are also homeostatically regulated. Here, we attempt to apply concepts of homeostasis in general, and more specifically synaptic homeostatic plasticity, to the study of epilepsy. We hypothesize that homeostatic mechanisms are actively engaged in the epileptic brain. These processes attempt to re-establish normal neuronal and network activity, but are opposed by the concurrent mechanisms underlying epileptogenesis. In forms of intractable epilepsy, seizures are so frequent and intense that homeostatic mechanisms are unable to restore normal levels of neuronal activity. In such cases, we contend that homeostatic plasticity mechanisms nevertheless remain active. However, their continuing attempts to reset neuronal activity become maladaptive and results in dyshomeostasis with neurobehavioral consequences. Using the developing hippocampus as a model system, we briefly review experimental results and present a series of arguments to propose that the cognitive neurobehavioral comorbidities of childhood epilepsy result, at least in part, from unchecked homeostatic mechanisms.
Collapse
|
41
|
Galanopoulou AS, Moshé SL. Does epilepsy cause a reversion to immature function? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:195-209. [PMID: 25012378 DOI: 10.1007/978-94-017-8914-1_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Seizures have variable effects on brain. Numerous studies have examined the consequences of seizures, in light of the way that these may alter the susceptibility of the brain to seizures, promote epileptogenesis, or functionally alter brain leading to seizure-related comorbidities. In many -but not all- situations, seizures shift brain function towards a more immature state, promoting the birth of newborn neurons, altering the dendritic structure and neuronal connectivity, or changing neurotransmitter signaling towards more immature patterns. These effects depend upon many factors, including the seizure type, age of seizure occurrence, sex, and brain region studied. Here we discuss some of these findings proposing that these seizure-induced immature features do not simply represent rejuvenation of the brain but rather a de-synchronization of the homeostatic mechanisms that were in place to maintain normal physiology, which may contribute to epileptogenesis or the cognitive comorbidities.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, The Laboratory of Developmental Epilepsy, Comprehensive Einstein/Montefiore Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 306, Bronx, NY, 10461, USA,
| | | |
Collapse
|
42
|
Lakhina V, Subramanian L, Huilgol D, Shetty AS, Vaidya VA, Tole S. Seizure evoked regulation of LIM-HD genes and co-factors in the postnatal and adult hippocampus. F1000Res 2013; 2:205. [PMID: 25110573 PMCID: PMC4111125 DOI: 10.12688/f1000research.2-205.v1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2013] [Indexed: 12/03/2022] Open
Abstract
The LIM-homeodomain (LIM-HD) family of transcription factors is well known for its functions during several developmental processes including cell fate specification, cell migration and axon guidance, and its members play fundamental roles in hippocampal development. The hippocampus is a structure that displays striking activity dependent plasticity. We examined whether LIM-HD genes and their co-factors are regulated during kainic acid induced seizure in the adult rat hippocampus as well as in early postnatal rats, when the hippocampal circuitry is not fully developed. We report a distinct and field-specific regulation of LIM-HD genes
Lhx1,Lhx2, and
Lhx9, LIM-only gene
Lmo4, and cofactor
Clim1a in the adult hippocampus after seizure induction. In contrast none of these genes displayed altered levels upon induction of seizure in postnatal animals. Our results provide evidence of temporal and spatial seizure mediated regulation of LIM-HD family members and suggest that LIM-HD gene function may be involved in activity dependent plasticity in the adult hippocampus
Collapse
Affiliation(s)
- Vanisha Lakhina
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Lewis Sigler Institute for Integrative Genomics, Princeton University, NJ, USA
| | - Lakshmi Subramanian
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Department of Neurology, University of California, San Francisco, CA, USA
| | - Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Cold Spring Harbor Laboratory, NY, USA
| | - Ashwin S Shetty
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
43
|
Casanova JR, Nishimura M, Le J, Lam TT, Swann JW. Rapid hippocampal network adaptation to recurring synchronous activity--a role for calcineurin. Eur J Neurosci 2013; 38:3115-27. [PMID: 23879713 DOI: 10.1111/ejn.12315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 06/10/2013] [Accepted: 06/19/2013] [Indexed: 11/29/2022]
Abstract
Neuronal networks are thought to gradually adapt to altered neuronal activity over many hours and days. For instance, when activity is increased by suppressing synaptic inhibition, excitatory synaptic transmission is reduced. The underlying compensatory cellular and molecular mechanisms are thought to contribute in important ways to maintaining normal network operations. Seizures, due to their massive and highly synchronised discharging, probably challenge the adaptive properties of neurons, especially when seizures are frequent and intense - a condition common in early childhood. In the experiments reported here, we used rat and mice hippocampal slice cultures to explore the effects that recurring seizure-like activity has on the developing hippocampus. We found that developing networks adapted rapidly to recurring synchronised activity in that the duration of seizure-like events was reduced by 42% after 4 h of activity. At the same time, the frequency of spontaneous excitatory postsynaptic currents in pyramidal cells, the expression of biochemical biomarkers for glutamatergic synapses and the branching of pyramidal cell dendrites were all dramatically reduced. Experiments also showed that the reduction in N-methyl-D-aspartate receptor subunits and postsynaptic density protein 95 expression were N-methyl-D-aspartate receptor-dependent. To explore calcium signaling mechanisms in network adaptation, we tested inhibitors of calcineurin, a protein phosphatase known to play roles in synaptic plasticity and activity-dependent dendrite remodeling. We found that FK506 was able to prevent all of the electrophysiological, biochemical, and anatomical changes produced by synchronised network activity. Our results show that hippocampal pyramidal cells and their networks adapt rapidly to intense synchronised activity and that calcineurin play an important role in the underlying processes.
Collapse
Affiliation(s)
- J R Casanova
- The Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; The Cain Foundation Laboratories, The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Suite 1225, Houston, TX, 77030, USA
| | | | | | | | | |
Collapse
|
44
|
Ma Y, Ramachandran A, Ford N, Parada I, Prince DA. Remodeling of dendrites and spines in the C1q knockout model of genetic epilepsy. Epilepsia 2013; 54:1232-9. [PMID: 23621154 DOI: 10.1111/epi.12195] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine whether developmental synaptic pruning defects in epileptic C1q-knockout (KO) mice are accompanied by postsynaptic abnormalities in dendrites and/or spines. METHODS Immunofluorescence staining was performed on biocytin-filled layer Vb pyramidal neurons in sensorimotor cortex. Basal dendritic arbors and their spines were reconstructed with NEUROLUCIDA software, and their morphologic characteristics were quantitated in Neuroexplorer. KEY FINDINGS Seven to nine completely filled pyramidal neurons were analyzed from the wild-type (WT) and C1q KO groups. Compared to WT controls, KO mice showed significant structural modifications in their basal dendrites including (1) higher density of dendritic spines (0.60 ± 0.03/μm vs. 0.49 ± 0.03/μm dendritic length in WT, p < 0.05); (2) remarkably increased occurrence of thin spines (0.26 ± 0.02/μm vs. 0.14 ± 0.02/μm dendritic length in control, p < 0.01); (3) longer dendritic length (2,680 ± 159 μm vs. 2,119 ± 108 μm in control); and (4) increased branching (22.6 ± 1.9 vs. 16.2 ± 1.3 in WT at 80 μm from soma center, p < 0.05; 12.4 ± 1.4 vs. 8.2 ± 0.6 in WT at 120 μm from soma center, respectively, p < 0.05). Dual immunolabeling demonstrated the expression of putative glutamate receptor 2 (GluR2) on some thin spines. These dendritic alterations are likely postsynaptic structural consequences of failure of synaptic pruning in the C1q KO mice. SIGNIFICANCE Failure to prune excessive excitatory synapses in C1q KO mice is a likely mechanism underlying abnormalities in postsynaptic dendrites, including increased branching and alterations in spine type and density. It is also possible that seizure activity contributes to these abnormalities. These structural abnormalities, together with increased numbers of excitatory synapses, likely contribute to epileptogenesis in C1q KO mice.
Collapse
Affiliation(s)
- Yunyong Ma
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5122, USA
| | | | | | | | | |
Collapse
|
45
|
Jeon BT, Jeong EA, Park SY, Son H, Shin HJ, Lee DH, Kim HJ, Kang SS, Cho GJ, Choi WS, Roh GS. The Rho-Kinase (ROCK) Inhibitor Y-27632 Protects Against Excitotoxicity-Induced Neuronal Death In Vivo and In Vitro. Neurotox Res 2012; 23:238-48. [DOI: 10.1007/s12640-012-9339-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 12/22/2022]
|
46
|
Casanova JR, Nishimura M, Owens JW, Swann JW. Impact of seizures on developing dendrites: Implications for intellectual developmental disabilities. Epilepsia 2012; 53 Suppl 1:116-24. [DOI: 10.1111/j.1528-1167.2012.03482.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
47
|
Dendritic spine pathology in epilepsy: cause or consequence? Neuroscience 2012; 251:141-50. [PMID: 22522469 DOI: 10.1016/j.neuroscience.2012.03.048] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 01/26/2012] [Accepted: 03/04/2012] [Indexed: 01/23/2023]
Abstract
Abnormalities in dendritic spines have commonly been observed in brain specimens from epilepsy patients and animal models of epilepsy. However, the functional implications and clinical consequences of this dendritic pathology for epilepsy are uncertain. Dendritic spine abnormalities may promote hyperexcitable circuits and seizures in some types of epilepsy, especially in specific genetic syndromes with documented dendritic pathology, but in these cases it is difficult to differentiate their effects on seizures versus other comorbidities, such as cognitive deficits and autism. In other situations, seizures themselves may cause damage to dendrites and dendritic spines and this seizure-induced brain injury may then contribute to progressive epileptogenesis, memory problems and other neurological deficits in epilepsy patients. The mechanistic basis of dendritic spine abnormalities in epilepsy has begun to be elucidated and suggests novel therapeutic strategies for treating epilepsy and its complications.
Collapse
|
48
|
Holmes GL, Milh MM, Dulac O. Maturation of the human brain and epilepsy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 107:135-43. [DOI: 10.1016/b978-0-444-52898-8.00007-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
49
|
Nishimura M, Gu X, Swann JW. Seizures in early life suppress hippocampal dendrite growth while impairing spatial learning. Neurobiol Dis 2011; 44:205-14. [PMID: 21777677 PMCID: PMC3167037 DOI: 10.1016/j.nbd.2011.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 06/27/2011] [Accepted: 07/01/2011] [Indexed: 11/21/2022] Open
Abstract
Impaired learning and memory are common in epilepsy syndromes of childhood. Clinical investigations suggest that the developing brain may be particularly vulnerable to the effects of intractable seizure disorders. Magnetic resonance imaging (MRI) studies have demonstrated reduced volumes in brain regions involved in learning and memory. The earlier the onset of an epilepsy the larger the effects seem to be on both brain anatomy and cognition. Thus, childhood epilepsy has been proposed to interfere in some unknown way with brain development. Experiments reported here explore these ideas by examining the effects of seizures in infant mice on learning and memory and on the growth of CA1 hippocampal pyramidal cell dendrites. Fifteen brief seizures were induced by flurothyl between postnatal days 7 and 11 in mice that express green fluorescent protein (GFP) in hippocampal pyramidal cells. One to 44days later, dendritic arbors were reconstructed to measure growth. Spatial learning and memory were also assessed in a water maze. Our results show that recurrent seizures produced marked deficits in learning and memory. Seizures also dramatically slowed the growth of basilar dendrites while neurons in littermate control mice continued to add new dendritic branches and lengthen existing branches. When experiments were performed in older mice, seizures had no measureable effects on either dendrite arbor complexity or spatial learning and memory. Our results suggest that the recurring seizures of intractable childhood epilepsy contribute to associated learning and memory deficits by suppressing dendrite growth.
Collapse
Affiliation(s)
- Masataka Nishimura
- The Cain Foundation Laboratories The Jan and Dan Duncan Neurological Research Institute Departments of Pediatrics Baylor College of Medicine Houston TX 77030
| | - Xue Gu
- The Cain Foundation Laboratories The Jan and Dan Duncan Neurological Research Institute Departments of Pediatrics Baylor College of Medicine Houston TX 77030
| | - John W. Swann
- The Cain Foundation Laboratories The Jan and Dan Duncan Neurological Research Institute Departments of Pediatrics Baylor College of Medicine Houston TX 77030
- Neuroscience Baylor College of Medicine Houston TX 77030
| |
Collapse
|
50
|
Bourne JN, Harris KM. Coordination of size and number of excitatory and inhibitory synapses results in a balanced structural plasticity along mature hippocampal CA1 dendrites during LTP. Hippocampus 2011; 21:354-73. [PMID: 20101601 DOI: 10.1002/hipo.20768] [Citation(s) in RCA: 228] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Enlargement of dendritic spines and synapses correlates with enhanced synaptic strength during long-term potentiation (LTP), especially in immature hippocampal neurons. Less clear is the nature of this structural synaptic plasticity on mature hippocampal neurons, and nothing is known about the structural plasticity of inhibitory synapses during LTP. Here the timing and extent of structural synaptic plasticity and changes in local protein synthesis evidenced by polyribosomes were systematically evaluated at both excitatory and inhibitory synapses on CA1 dendrites from mature rats following induction of LTP with theta-burst stimulation (TBS). Recent work suggests dendritic segments can act as functional units of plasticity. To test whether structural synaptic plasticity is similarly coordinated, we reconstructed from serial section transmission electron microscopy all of the spines and synapses along representative dendritic segments receiving control stimulation or TBS-LTP. At 5 min after TBS, polyribosomes were elevated in large spines suggesting an initial burst of local protein synthesis, and by 2 h only those spines with further enlarged synapses contained polyribosomes. Rapid induction of synaptogenesis was evidenced by an elevation in asymmetric shaft synapses and stubby spines at 5 min and more nonsynaptic filopodia at 30 min. By 2 h, the smallest synaptic spines were markedly reduced in number. This synapse loss was perfectly counterbalanced by enlargement of the remaining excitatory synapses such that the summed synaptic surface area per length of dendritic segment was constant across time and conditions. Remarkably, the inhibitory synapses showed a parallel synaptic plasticity, also demonstrating a decrease in number perfectly counterbalanced by an increase in synaptic surface area. Thus, TBS-LTP triggered spinogenesis followed by loss of small excitatory and inhibitory synapses and a subsequent enlargement of the remaining synapses by 2 h. These data suggest that dendritic segments coordinate structural plasticity across multiple synapses and maintain a homeostatic balance of excitatory and inhibitory inputs through local protein-synthesis and selective capture or redistribution of dendritic resources.
Collapse
Affiliation(s)
- Jennifer N Bourne
- Center for Learning and Memory, Section of Neurobiology, Institute for Neuroscience, University of Texas, Austin, Texas 78712, USA
| | | |
Collapse
|