1
|
Cortes MA, Bartley AF, Li Q, Davis TR, Cunningham SE, Garner MA, Perez PJ, Harvey AC, Gross AK, Dobrunz LE. Modulation of temporoammonic-CA1 synapses by neuropeptide Y is through Y1 receptors in mice. Neuropeptides 2025; 110:102504. [PMID: 39951960 DOI: 10.1016/j.npep.2025.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Reduced levels of neuropeptide Y (NPY), an abundant neuromodulator in the brain, are linked to multiple neuropsychiatric disorders, including post-traumatic stress disorder (PTSD). The CA1 region of hippocampus is important for anxiety regulation and highly expresses NPY. Injecting NPY into CA1 is anxiolytic and alleviates behavioral symptoms in a model of traumatic stress; these anxiolytic effects are blocked by a Y1 receptor antagonist. However the location of Y1Rs that mediate NPY's anxiolytic effects in CA1 remains unclear. CA1 receives inputs from entorhinal cortex through the temporammonic pathway (TA), which is important for fear learning and sensitive to stress. Our lab previously showed that NPY reduces TA-evoked synaptic responses, however, the subtype of NPY receptor mediating this reduction is unknown. Here we demonstrate that in mice both exogenous (bath-applied) and endogenously-released NPY act through Y1 receptors in the TA pathway. This is the first demonstration of Y1 receptor-mediated effect on synaptic function in CA1. Interestingly, chronic overexpression of NPY (in NPY-expressing interneurons) impairs the sensitivity of the TA-evoked synaptic response to a Y1 receptor agonist. However, the long-known NPY Y2 receptor-mediated effect on the Schaffer collateral (SC) pathway is unaffected by NPY overexpression. Therefore, NPY can have a pathway-specific impact on synaptic transmission in CA1 based on the differential expression of NPY receptors and their response to overexpression of NPY. Our results demonstrating that NPY acts at Y1 receptors in the TA pathway are consistent with the idea that the TA pathway underlies the anxiolytic effects of NPY in CA1.
Collapse
Affiliation(s)
- Mariana A Cortes
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Aundrea F Bartley
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Qin Li
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Taylor R Davis
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Stephen E Cunningham
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Mary Anne Garner
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Patric J Perez
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Adela C Harvey
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Alecia K Gross
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States of America
| | - Lynn E Dobrunz
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States of America.
| |
Collapse
|
2
|
Chesnokova E, Bal N, Alhalabi G, Balaban P. Regulatory Elements for Gene Therapy of Epilepsy. Cells 2025; 14:236. [PMID: 39937026 PMCID: PMC11816724 DOI: 10.3390/cells14030236] [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: 12/28/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The problem of drug resistance in epilepsy means that in many cases, a surgical treatment may be advised. But this is only possible if there is an epileptic focus, and resective brain surgery may have adverse side effects. One of the promising alternatives is gene therapy, which allows the targeted expression of therapeutic genes in different brain regions, and even in specific cell types. In this review, we provide detailed explanations of some key terms related to genetic engineering, and describe various regulatory elements that have already been used in the development of different approaches to treating epilepsy using viral vectors. We compare a few universal promoters for their strength and duration of transgene expression, and in our description of cell-specific promoters, we focus on elements driving expression in glutamatergic neurons, GABAergic neurons and astrocytes. We also explore enhancers and some other cis-regulatory elements currently used in viral vectors for gene therapy, and consider future perspectives of state-of-the-art technologies for designing new, stronger and more specific regulatory elements. Gene therapy has multiple advantages and should become more common in the future, but there is still a lot to study and invent in this field.
Collapse
Affiliation(s)
- Ekaterina Chesnokova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Natalia Bal
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Ghofran Alhalabi
- Laboratory of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia;
| | - Pavel Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| |
Collapse
|
3
|
Cai AJ, Gao K, Zhang F, Jiang YW. Recent advances and current status of gene therapy for epilepsy. World J Pediatr 2024; 20:1115-1137. [PMID: 39395088 DOI: 10.1007/s12519-024-00843-w] [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: 02/14/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Epilepsy is a common neurological disorder with complex pathogenic mechanisms, and refractory epilepsy often lacks effective treatments. Gene therapy is a promising therapeutic option, with various preclinical experiments achieving positive results, some of which have progressed to clinical studies. DATA SOURCES This narrative review was conducted by searching for papers published in PubMed/MEDLINE with the following single and/or combination keywords: epilepsy, children, neurodevelopmental disorders, genetics, gene therapy, vectors, transgenes, receptors, ion channels, micro RNAs (miRNAs), clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas)9 (CRISPR/Cas9), expression regulation, optogenetics, chemical genetics, mitochondrial epilepsy, challenges, ethics, and disease models. RESULTS Currently, gene therapy research in epilepsy primarily focuses on symptoms attenuation mediated by viral vectors such as adeno-associated virus and other types. Advances in gene therapy technologies, such as CRISPR/Cas9, have provided a new direction for epilepsy treatment. However, the clinical application still faces several challenges, including issues related to vectors, models, expression controllability, and ethical considerations. CONCLUSIONS Here, we summarize the relevant research and clinical advances in gene therapy for epilepsy and outline the challenges facing its clinical application. In addition to the shortcomings inherent in gene therapy components, the reconfiguration of excitatory and inhibitory properties in epilepsy treatment is a delicate process. On-demand, cell-autonomous treatments and multidisciplinary collaborations may be crucial in addressing these issues. Understanding gene therapy for epilepsy will help clinicians gain a clearer perception of the research progress and challenges, guiding the design of future clinical protocols and research decisions.
Collapse
Affiliation(s)
- Ao-Jie Cai
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Henan Province, Zhengzhou, 450052, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Fan Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.
- Children Epilepsy Center, Peking University First Hospital, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
4
|
Barde S, Aguila J, Zhong W, Solarz A, Mei I, Prud'homme J, Palkovits M, Turecki G, Mulder J, Uhlén M, Nagy C, Mechawar N, Hedlund E, Hökfelt T. Substance P, NPY, CCK and their receptors in five brain regions in major depressive disorder with transcriptomic analysis of locus coeruleus neurons. Eur Neuropsychopharmacol 2024; 78:54-63. [PMID: 37931511 DOI: 10.1016/j.euroneuro.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 11/08/2023]
Abstract
Major depressive disorder (MDD) is a serious disease and a burden to patients, families and society. Rodent experiments and human studies suggest that several neuropeptide systems are involved in mood regulation. The aim of this study is two-fold: (i) to monitor, with qPCR, transcript levels of the substance P/tachykinin (TAC), NPY and CCK systems in bulk samples from control and suicide subjects, targeting five postmortem brain regions including locus coeruleus (LC); and (ii) to analyse expression of neuropeptide family transcripts in LC neurons of 'normal' postmortem brains by using laser capture microdissection with Smart-Seq2 RNA sequencing. qPCR revealed distinct regional expression patterns in male and female controls with higher levels for the TAC system in the dorsal raphe nucleus and LC, versus higher transcripts levels of the NPY and CCK systems in prefrontal cortex. In suicide patients, TAC, TAC receptors and a few NPY family transcript levels were increased mainly in prefrontal cortex and LC. The second study on 'normal' noradrenergic LC neurons revealed expression of transcripts for GAL, NPY, TAC1, CCK, and TACR1 and many other peptides (e.g. Cerebellin4 and CARTPT) and receptors (e.g. Adcyap1R1 and GPR173). These data and our previous results on suicide brains indicates that the tachykinin and galanin systems may be valid targets for developing antidepressant medicines. Moreover, the perturbation of neuropeptide systems in MDD patients, and the detection of further neuropeptide and receptor transcripts in LC, shed new light on signalling in noradrenergic LC neurons and on mechanisms possibly associated with mood disorders.
Collapse
Affiliation(s)
- Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Julio Aguila
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, 11428, Sweden
| | - Anna Solarz
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Irene Mei
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Josee Prud'homme
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Miklos Palkovits
- The Hungarian Academy of Sciences, Budapest, Hungary and Human Brain Tissue Bank and Laboratory, Semmelweis University, H-1085, Budapest, Hungary
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, 11428, Sweden
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Eva Hedlund
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Santos VR, Tilelli CQ, Fernandes A, de Castro OW, Del-Vecchio F, Garcia-Cairasco N. Different types of Status Epilepticus may lead to similar hippocampal epileptogenesis processes. IBRO Neurosci Rep 2023; 15:68-76. [PMID: 37457787 PMCID: PMC10338355 DOI: 10.1016/j.ibneur.2023.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
About 1-2% of people worldwide suffer from epilepsy, which is characterized by unpredictable and intermittent seizure occurrence. Despite the fact that the exact origin of temporal lobe epilepsy is frequently unknown, it is frequently linked to an early triggering insult like brain damage, tumors, or Status Epilepticus (SE). We used an experimental approach consisting of electrical stimulation of the amygdaloid complex to induce two behaviorally and structurally distinct SE states: Type I (fully convulsive), with more severe seizure behaviors and more extensive brain damage, and Type II (partial convulsive), with less severe seizure behaviors and brain damage. Our goal was to better understand how the various types of SE impact the hippocampus leading to the development of epilepsy. Despite clear variations between the two behaviors in terms of neurodegeneration, study of neurogenesis revealed a comparable rise in the number of Ki-67 + cells and an increase in Doublecortin (DCX) in both kinds of SE.
Collapse
Affiliation(s)
- Victor R. Santos
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, MG, Brazil
| | - Cristiane Q. Tilelli
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Campus Centro-Oeste Dona Lindu, Federal University of São João Del Rey, Divinópolis, MG, Brazil
| | - Artur Fernandes
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Olagide Wagner de Castro
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Pharmacology and Physiology, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Flávio Del-Vecchio
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Norberto Garcia-Cairasco
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
6
|
Melin E, Andersson M, Gøtzsche CR, Wickham J, Huang Y, Szczygiel JA, Boender A, Christiansen SH, Pinborg L, Woldbye DPD, Kokaia M. Combinatorial gene therapy for epilepsy: Gene sequence positioning and AAV serotype influence expression and inhibitory effect on seizures. Gene Ther 2023; 30:649-658. [PMID: 37029201 PMCID: PMC10457185 DOI: 10.1038/s41434-023-00399-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/24/2023] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
Gene therapy with AAV vectors carrying genes for neuropeptide Y and its receptor Y2 has been shown to inhibit seizures in multiple animal models of epilepsy. It is however unknown how the AAV serotype or the sequence order of these two transgenes in the expression cassette affects the actual parenchymal gene expression levels and the seizure-suppressant efficacy. To address these questions, we compared three viral vector serotypes (AAV1, AAV2 and AAV8) and two transgene sequence orders (NPY-IRES-Y2 and Y2-IRES-NPY) in a rat model of acutely induced seizures. Wistar male rats were injected bilaterally with viral vectors and 3 weeks later acute seizures were induced by a subcutaneous injection of kainate. The latency until 1st motor seizure, time spent in motor seizure and latency to status epilepticus were measured to evaluate the seizure-suppressing efficacy of these vectors compared to an empty cassette control vector. Based on the results, the effect of the AAV1-NPY-IRES-Y2 vector was further investigated by in vitro electrophysiology, and its ability to achieve transgene overexpression in resected human hippocampal tissue was evaluated. The AAV1-NPY-IRES-Y2 proved to be better to any other serotype or gene sequence considering both transgene expression and ability to suppress induced seizures in rats. The vector also demonstrated transgene-induced decrease of glutamate release from excitatory neuron terminals and significantly increased both NPY and Y2 expression in resected human hippocampal tissue from patients with drug-resistant temporal lobe epilepsy. These results validate the feasibility of NPY/Y2 receptor gene therapy as a therapeutic opportunity in focal epilepsies.
Collapse
Affiliation(s)
- Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, 17 Sölvegatan, 221 84, Lund, Sweden.
| | - My Andersson
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, 17 Sölvegatan, 221 84, Lund, Sweden
| | - Casper R Gøtzsche
- CombiGene AB, Medicon Village, 2 Scheelevägen, 223 81, Lund, Sweden
- Department of Neuroscience, Panum Institute, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark
| | - Jenny Wickham
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, 17 Sölvegatan, 221 84, Lund, Sweden
| | - Yuzhe Huang
- Department of Neuroscience, Panum Institute, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark
| | - Julia Alicja Szczygiel
- Department of Neuroscience, Panum Institute, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark
| | - Arnie Boender
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, 17 Sölvegatan, 221 84, Lund, Sweden
| | - Søren H Christiansen
- Department of Neuroscience, Panum Institute, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark
| | - Lars Pinborg
- Department of Neurology and Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, 9 Blegdamsvej, DK-2100, Copenhagen, Denmark
| | - David P D Woldbye
- Department of Neuroscience, Panum Institute, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark
| | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, 17 Sölvegatan, 221 84, Lund, Sweden
| |
Collapse
|
7
|
Franz J, Barheier N, Wilms H, Tulke S, Haas CA, Häussler U. Differential vulnerability of neuronal subpopulations of the subiculum in a mouse model for mesial temporal lobe epilepsy. Front Cell Neurosci 2023; 17:1142507. [PMID: 37066079 PMCID: PMC10090355 DOI: 10.3389/fncel.2023.1142507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Selective loss of inhibitory interneurons (INs) that promotes a shift toward an excitatory predominance may have a critical impact on the generation of epileptic activity. While research on mesial temporal lobe epilepsy (MTLE) has mostly focused on hippocampal changes, including IN loss, the subiculum as the major output region of the hippocampal formation has received less attention. The subiculum has been shown to occupy a key position in the epileptic network, but data on cellular alterations are controversial. Using the intrahippocampal kainate (KA) mouse model for MTLE, which recapitulates main features of human MTLE such as unilateral hippocampal sclerosis and granule cell dispersion, we identified cell loss in the subiculum and quantified changes in specific IN subpopulations along its dorso-ventral axis. We performed intrahippocampal recordings, FluoroJade C-staining for degenerating neurons shortly after status epilepticus (SE), fluorescence in situ hybridization for glutamic acid decarboxylase (Gad) 67 mRNA and immunohistochemistry for neuronal nuclei (NeuN), parvalbumin (PV), calretinin (CR) and neuropeptide Y (NPY) at 21 days after KA. We observed remarkable cell loss in the ipsilateral subiculum shortly after SE, reflected in lowered density of NeuN+ cells in the chronic stage when epileptic activity occurred in the subiculum concomitantly with the hippocampus. In addition, we show a position-dependent reduction of Gad67-expressing INs by ∼50% (along the dorso-ventral as well as transverse axis of the subiculum). This particularly affected the PV- and to a lesser extent CR-expressing INs. The density of NPY-positive neurons was increased, but the double-labeling for Gad67 mRNA expression revealed that an upregulation or de novo expression of NPY in non-GABAergic cells with a concomitant reduction of NPY-positive INs underlies this observation. Our data suggest a position- and cell type-specific vulnerability of subicular INs in MTLE, which might contribute to hyperexcitability of the subiculum, reflected in epileptic activity.
Collapse
Affiliation(s)
- Julia Franz
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Nicole Barheier
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Henrike Wilms
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Susanne Tulke
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Carola A. Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Ute Häussler
- Experimental Epilepsy Research, Department of Neurosurgery, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- *Correspondence: Ute Häussler,
| |
Collapse
|
8
|
Drexel M, Sperk G. Seizure-induced overexpression of NPY induces epileptic tolerance in a mouse model of spontaneous recurrent seizures. Front Mol Neurosci 2022; 15:974784. [PMID: 36311021 PMCID: PMC9608171 DOI: 10.3389/fnmol.2022.974784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Epileptic seizures result in pronounced over-expression of neuropeptide Y (NPY). In vivo and in vitro studies revealed that NPY exerts potent anticonvulsive actions through presynaptic Y2 receptors by suppressing glutamate release from principal neurons. We now investigated whether seizure-induced over-expression of NPY contributes to epileptic tolerance induced by preceding seizures. We used a previously established animal model based on selective inhibition of GABA release from parvalbumin (PV)-containing interneurons in the subiculum in mice. The animals present spontaneous recurrent seizures (SRS) and clusters of interictal spikes (IS). The frequency of SRS declined after five to six weeks, indicating development of seizure tolerance. In interneurons of the subiculum and sector CA1, SRS induced over-expression of NPY that persisted there for a prolonged time despite of a later decrease in SRS frequency. In contrast to NPY, somatostatin was not overexpressed in the respective axon terminals. Contrary to interneurons, NPY was only transiently expressed in mossy fibers. To demonstrate a protective function of endogenous, over-expressed NPY, we injected the selective NPY-Y2 receptor antagonist JNJ 5207787 simultaneously challenging the mice by a low dose of pentylenetetrazol (PTZ, 30 or 40 mg/kg, i.p.). In control mice, neither PTZ nor PTZ plus JNJ 5207787 induced convulsions. In mice with silenced GABA/PV neurons, PTZ alone only modestly enhanced EEG activity. When we injected JNJ 5207787 together with PTZ (either dose) the number of seizures, however, became significantly increased. In addition, in the epileptic mice CB1 receptor immunoreactivity was reduced in terminal areas of basket cells pointing to reduced presynaptic inhibition of GABA release from these neurons. Our experiments demonstrate that SRS result in overexpression of NPY in hippocampal interneurons. NPY overexpression persists for several weeks and may be related to later decreasing SRS frequency. Injection of the Y2 receptor antagonist JNJ 5207787 prevents this protective action of NPY only when release of the peptide is triggered by injection of PTZ and induces pronounced convulsions. Thus, over-expressed NPY released “on demand” by seizures may help terminating acute seizures and may prevent from recurrent epileptic activity.
Collapse
|
9
|
Li J, Ek F, Olsson R, Belting M, Bengzon J. Glioblastoma CD105 + cells define a SOX2 - cancer stem cell-like subpopulation in the pre-invasive niche. Acta Neuropathol Commun 2022; 10:126. [PMID: 36038950 PMCID: PMC9426031 DOI: 10.1186/s40478-022-01422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Glioblastoma (GBM) is the most common and most aggressive primary brain tumor in adults. Glioma stem like cells (GSC) represent the highest cellular hierarchy in GBM and have a determining role in tumor growth, recurrence and patient prognosis. However, a better definition of GSC subpopulations, especially at the surgical resection margin, is warranted for improved oncological treatment options. The present study interrogated cells expressing CD105 (CD105+) specifically within the tumor front and the pre-invasive niche as a potential GSC subpopulation. GBM primary cell lines were generated from patients (n = 18) and CD105+ cells were isolated and assessed for stem-like characteristics. In vitro, CD105+ cells proliferated and enriched in serum-containing medium but not in serum-free conditions. CD105+ cells were characterized by Nestin+, Vimentin+ and SOX2-, clearly distinguishing them from SOX2+ GCS. GBM CD105+ cells differentiated into osteocytes and adipocytes but not chondrocytes. Exome sequencing revealed that GBM CD105+ cells matched 83% of somatic mutations in the Cancer cell line encyclopedia, indicating a malignant phenotype and in vivo xenotransplantation assays verified their tumorigenic potential. Cytokine assays showed that immunosuppressive and protumorigenic cytokines such as IL6, IL8, CCL2, CXCL-1 were produced by CD105+ cells. Finally, screening for 88 clinical drugs revealed that GBM CD105+ cells are resistant to most chemotherapeutics except Doxorubicin, Idarubicin, Fludarabine and ABT-751. Our study provides a rationale for targeting tumoral CD105+ cells in order to reshape the tumor microenvironment and block GBM progression.
Collapse
Affiliation(s)
- Jiaxin Li
- Stem Cell Center, Lund University, Lund, Sweden.
- Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Roger Olsson
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Mattias Belting
- Section of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiophysics, Skane University Hospital, Lund, Sweden
- Science for Life Laboratory, Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Bengzon
- Stem Cell Center, Lund University, Lund, Sweden
- Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Neurosurgery, Skane University Hospital, Lund, Sweden
| |
Collapse
|
10
|
You J, Huang H, Chan CTY, Li L. Pathological Targets for Treating Temporal Lobe Epilepsy: Discoveries From Microscale to Macroscale. Front Neurol 2022; 12:779558. [PMID: 35069411 PMCID: PMC8777077 DOI: 10.3389/fneur.2021.779558] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common and severe types of epilepsy, characterized by intractable, recurrent, and pharmacoresistant seizures. Histopathology of TLE is mostly investigated through observing hippocampal sclerosis (HS) in adults, which provides a robust means to analyze the related histopathological lesions. However, most pathological processes underlying the formation of these lesions remain elusive, as they are difficult to detect and observe. In recent years, significant efforts have been put in elucidating the pathophysiological pathways contributing to TLE epileptogenesis. In this review, we aimed to address the new and unrecognized neuropathological discoveries within the last 5 years, focusing on gene expression (miRNA and DNA methylation), neuronal peptides (neuropeptide Y), cellular metabolism (mitochondria and ion transport), cellular structure (microtubule and extracellular matrix), and tissue-level abnormalities (enlarged amygdala). Herein, we describe a range of biochemical mechanisms and their implication for epileptogenesis. Furthermore, we discuss their potential role as a target for TLE prevention and treatment. This review article summarizes the latest neuropathological discoveries at the molecular, cellular, and tissue levels involving both animal and patient studies, aiming to explore epileptogenesis and highlight new potential targets in the diagnosis and treatment of TLE.
Collapse
Affiliation(s)
- Jing You
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | - Haiyan Huang
- Department of Nutrition and Food Science, Texas Women University, Denton, TX, United States
| | - Clement T Y Chan
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | - Lin Li
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Gupta S, Bansal RN, Sodhi SPS, Brar GK. Animal models - Mimicking the pain of trigeminal neuralgia. Indian J Pharmacol 2022; 54:138-145. [PMID: 35546466 PMCID: PMC9249160 DOI: 10.4103/ijp.ijp_296_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Trigeminal neuralgia (TN) is an episodic facial pain which feels like an electric shock of unilateral origin. This neuropathic disorder is an intensely stressful to bear for patient and impacts the quality of life. Most of the cases of TN arise when the root of fifth cranial nerve, i.e., trigeminal nerve is compressed after a few millimeters of its entry into the pons. This article describes various animal models and the role of biomarkers to study the underlying mechanisms of neuropathic pain in animal models as well as different modes of management of TN.
Collapse
Affiliation(s)
- Sonu Gupta
- Department of Clinical Research, Dasmesh Institute of Research and Dental Sciences, Faridkot Punjab, India
| | - Ravinder Nath Bansal
- Department of Hospital Administration, Guru Gobind Singh Medical Hospital, Faridkot Punjab, India,Address for correspondence: Dr. Ravinder Nath Bansal, Hospital Administration, Guru Gobind Singh Medical Hospital, Faridkot, Punjab, India. E-mail:
| | - Surender Pal Singh Sodhi
- Department of Oral & Maxillofacial Surgery, Dasmesh Institute of Research and Dental Sciences,Faridkot Punjab, India
| | - Gursimrat Kaur Brar
- Department of Oral & Maxillofacial Surgery, Dasmesh Institute of Research and Dental Sciences,Faridkot Punjab, India
| |
Collapse
|
12
|
Zhang L, Wang Y. Gene therapy in epilepsy. Biomed Pharmacother 2021; 143:112075. [PMID: 34488082 DOI: 10.1016/j.biopha.2021.112075] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 01/15/2023] Open
Abstract
Gene therapy may constitute a promising alternative to conventional pharmacological tools and surgeries for epilepsy. For primary epilepsy, a single variant leading to a significant effect is relatively rare, while other forms are considered complex in inheritances with multiple susceptible mutations and impacts from the environment. Gene therapy in preclinical models of epilepsy has attempted to perform antiepileptogenic, anticonvulsant, or disease-modifying effects during epileptogenesis or after establishing the disease. Creating gene vectors tailored for different situations is the key to expanding gene therapy, and choosing the appropriate therapeutic target remains another fundamental problem. A variety of treatment strategies, from overexpressing inhibitory neuropeptides to modulating the expression of neurotransmitters or ion channels, have been tested in animal models. Additionally, emerging new approaches of optogenetics and chemogenetics, as well as genome-editing tools will further boost the prosperity of gene therapy. This review summarizes the experience obtained to date and discusses the challenges and opportunities in clinical translations.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Neurology at Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuping Wang
- Beijing Key Laboratory of Neuromodulation, Capital Medical University, Beijing, China; Center of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
13
|
Przykaza Ł, Kozniewska E. Ligands of the Neuropeptide Y Y2 Receptors as a Potential Multitarget Therapeutic Approach for the Protection of the Neurovascular Unit Against Acute Ischemia/Reperfusion: View from the Perspective of the Laboratory Bench. Transl Stroke Res 2021; 13:12-24. [PMID: 34292517 PMCID: PMC8766383 DOI: 10.1007/s12975-021-00930-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Ischemic stroke is the third leading cause of death and disability worldwide, with no available satisfactory prevention or treatment approach. The current treatment is limited to the use of “reperfusion methods,” i.e., an intravenous or intra-arterial infusion of a fibrinolytic agent, mechanical removal of the clot by thrombectomy, or a combination of both methods. It should be stressed, however, that only approximately 5% of all acute strokes are eligible for fibrinolytic treatment and fewer than 10% for thrombectomy. Despite the tremendous progress in understanding of the pathomechanisms of cerebral ischemia, the promising results of basic research on neuroprotection are not currently transferable to human stroke. A possible explanation for this failure is that experiments on in vivo animal models involve healthy young animals, and the experimental protocols seldom consider the importance of protecting the whole neurovascular unit (NVU), which ensures intracranial homeostasis and is seriously damaged by ischemia/reperfusion. One of the endogenous protective systems activated during ischemia and in neurodegenerative diseases is represented by neuropeptide Y (NPY). It has been demonstrated that activation of NPY Y2 receptors (Y2R) by a specific ligand decreases the volume of the postischemic infarction and improves performance in functional tests of rats with arterial hypertension subjected to middle cerebral artery occlusion/reperfusion. This functional improvement suggests the protection of the NVU. In this review, we focus on NPY and discuss the potential, multidirectional protective effects of Y2R agonists against acute focal ischemia/reperfusion injury, with special reference to the NVU.
Collapse
Affiliation(s)
- Łukasz Przykaza
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland
| | - Ewa Kozniewska
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
14
|
Kalkan ÖF, Şahin Z, Öztürk H, Keser H, Aydın-Abidin S, Abidin İ. Phoenixin-14 reduces the frequency of interictal-like events in mice brain slices. Exp Brain Res 2021; 239:2841-2849. [PMID: 34283252 DOI: 10.1007/s00221-021-06179-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/13/2021] [Indexed: 01/17/2023]
Abstract
Phoenixin-14 (PNX-14) has a wide bioactivity in the central nervous system. Its role in the hypothalamus has been investigated, and it has been reported that it is involved in the regulation of excitability in hypothalamic neurons. However, its role in the regulation of excitability in entorhinal cortex and the hippocampus is unknown. In this study, we investigated whether i. PNX-14 induces any synchronous discharges or epileptiform activity and ii. PNX-14 has any effect on already initiated epileptiform discharges. We used 350 µm thick acute horizontal hippocampal-entorhinal cortex slices obtained from 30- to 35-day-old mice. Extracellular field potential recordings were evaluated in the entorhinal cortex and hippocampus CA1 region. Bath application of PNX-14 did not initiate any epileptiform activity or abnormal discharges. 4-Aminopyridine was applied to induce epileptiform activity in the slices. We found that 200 nM PNX-14 reduced the frequency of interictal-like events in both the entorhinal cortex and hippocampus CA1 region which was induced by 4-aminopyridine. Furthermore, PNX-14 led to a similar suppression in the total power of local field potentials of 1-120 Hz. The frequency or the duration of the ictal events was not affected. These results exhibited for the first time that PNX-14 has a modulatory effect on synchronized neuronal discharges which should be considered in future therapeutic approaches.
Collapse
Affiliation(s)
- Ömer Faruk Kalkan
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Zafer Şahin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Hilal Öztürk
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Hatice Keser
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Selcen Aydın-Abidin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - İsmail Abidin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| |
Collapse
|
15
|
Shih PY, Kreir M, Kumar D, Seibt F, Pestana F, Schmid B, Holst B, Clausen C, Steeg R, Fischer B, Pita-Almenar J, Ebneth A, Cabrera-Socorro A. Development of a fully human assay combining NGN2-inducible neurons co-cultured with iPSC-derived astrocytes amenable for electrophysiological studies. Stem Cell Res 2021; 54:102386. [PMID: 34229210 DOI: 10.1016/j.scr.2021.102386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/15/2021] [Accepted: 04/30/2021] [Indexed: 11/19/2022] Open
Abstract
Neurogenin 2 encodes a neural-specific transcription factor (NGN2) able to drive neuronal fate on somatic and stem cells. NGN2 is expressed in neural progenitors within the developing central and peripheral nervous systems. Overexpression of NGN2 in human induced pluripotent stem cells (hiPSCs) or human embryonic stem cells has been shown to efficiently trigger conversion to neurons. Here we describe two gene-edited hiPSC lines harbouring a doxycycline (DOX)-inducible cassette in the AAVS1 locus driving expression of NGN2 (BIONi010-C-13) or NGN2-T2A-GFP (BIONi010-C-15). By introducing NGN2-expressing cassette, we reduce variability associated with conventional over-expression methods such as viral transduction, making these lines amenable for scale-up production and screening processes. DOX-treated hiPSCs convert to neural phenotype within one week and display the expression of structural neuronal markers such as Beta-III tubulin and tau. We performed functional characterization of NGN2-neurons co-cultured with hiPSC-derived astrocytes in a "fully-humanized" set up. Passive properties of NGN2-neurons were indistinguishable from mouse primary cells while displaying variable activity in extracellular recordings performed in multi-electrode arrays (MEAs). We demonstrate that hiPSC-derived astrocytes and neurons can be co-cultured and display functional properties comparable to the gold standard used in electrophysiology. Both lines are globally available via EBiSC repository at https://cells.ebisc.org/.
Collapse
Affiliation(s)
- Pei-Yu Shih
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Mohamed Kreir
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Devesh Kumar
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Frederik Seibt
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | | | - Bjørn Holst
- Bioneer S/A, Kogle Allé 2, 2970 Hørsholm, Denmark
| | | | | | - Benjamin Fischer
- Project Centre for Stem Cell Process Engineering, Fraunhofer Institute for Biomedical Engineering IBMT, Würzburg, Germany
| | | | - Andreas Ebneth
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | |
Collapse
|
16
|
Cattaneo S, Verlengia G, Marino P, Simonato M, Bettegazzi B. NPY and Gene Therapy for Epilepsy: How, When,... and Y. Front Mol Neurosci 2021; 13:608001. [PMID: 33551745 PMCID: PMC7862707 DOI: 10.3389/fnmol.2020.608001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Neuropeptide Y (NPY) is a neuropeptide abundantly expressed in the mammalian central and peripheral nervous system. NPY is a pleiotropic molecule, which influences cell proliferation, cardiovascular and metabolic function, pain and neuronal excitability. In the central nervous system, NPY acts as a neuromodulator, affecting pathways that range from cellular (excitability, neurogenesis) to circuit level (food intake, stress response, pain perception). NPY has a broad repertoire of receptor subtypes, each activating specific signaling pathways in different tissues and cellular sub-regions. In the context of epilepsy, NPY is thought to act as an endogenous anticonvulsant that performs its action through Y2 and Y5 receptors. In fact, its overexpression in the brain with the aid of viral vectors can suppress seizures in animal models of epilepsy. Therefore, NPY-based gene therapy may represent a novel approach for the treatment of epilepsy patients, particularly for pharmaco-resistant and genetic forms of the disease. Nonetheless, considering all the aforementioned aspects of NPY signaling, the study of possible NPY applications as a therapeutic molecule is not devoid of critical aspects. The present review will summarize data related to NPY biology, focusing on its anti-epileptic effects, with a critical appraisal of key elements that could be exploited to improve the already existing NPY-based gene therapy approaches for epilepsy.
Collapse
Affiliation(s)
- Stefano Cattaneo
- Vita-Salute San Raffaele University, Milan, Italy.,San Raffaele Scientific Institute, Milan, Italy
| | - Gianluca Verlengia
- San Raffaele Scientific Institute, Milan, Italy.,Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Pietro Marino
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, Section of Pediatrics, University of Ferrara, Ferrara, Italy
| | - Michele Simonato
- Vita-Salute San Raffaele University, Milan, Italy.,San Raffaele Scientific Institute, Milan, Italy.,Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Barbara Bettegazzi
- Vita-Salute San Raffaele University, Milan, Italy.,San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
17
|
Kvist G. Derivation of Adult Human Cortical Organotypic Slice Cultures for Coculture with Reprogrammed Neuronal Cells. Methods Mol Biol 2021; 2352:253-259. [PMID: 34324192 DOI: 10.1007/978-1-0716-1601-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Adult human cortical organotypic slice culture is an attractive model system to explore mechanisms of human brain pathology as well as to test drug candidates for treatment of neurodegeneration. Acute studies in human brain slices are limited by the lifetime of the tissue and focus mainly on hippocampus slice preparation. Here we describe the derivation of human organotypic slice cultures of cortical origin, which can be kept in culture for up to 6 weeks. This method enabled us to test the system in coculture with reprogrammed neurons and show its feasibility in neuronal cell integration experiments in human-to-human grafting situation.
Collapse
Affiliation(s)
- Giedre Kvist
- Lund Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
18
|
Szczygieł JA, Danielsen KI, Melin E, Rosenkranz SH, Pankratova S, Ericsson A, Agerman K, Kokaia M, Woldbye DPD. Gene Therapy Vector Encoding Neuropeptide Y and Its Receptor Y2 for Future Treatment of Epilepsy: Preclinical Data in Rats. Front Mol Neurosci 2020; 13:232. [PMID: 33343295 PMCID: PMC7746806 DOI: 10.3389/fnmol.2020.603409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
Gene therapy to treat pharmacoresistant temporal lobe epilepsy in humans is now being developed using an AAV vector (CG01) that encodes the combination of neuropeptide Y and its antiepileptic receptor Y2. With this in mind, the present study aimed to provide important preclinical data on the effects of CG01 on the duration of transgene expression, cellular tropism, and potential side effects on body weight and cognitive function. The CG01 vector was administered unilaterally into the dorsal and ventral hippocampus of adult male rats and expression of both transgenes was found to remain elevated without a sign of decline at 6 months post-injection. CG01 appeared to mediate expression selectively in hippocampal neurons, without expression in astrocytes or oligodendrocytes. No effects were seen on body weight as well as on short- or long-term memory as revealed by testing in the Y-maze or Morris water maze tests. Thus these data show that unilateral CG01 vector treatment as future gene therapy in pharmacoresistant temporal lobe epilepsy patients should result in stable and long-term expression predominantly in neurons and be well tolerated without side effects on body weight and cognitive function.
Collapse
Affiliation(s)
| | - Kira Iben Danielsen
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Lund, Sweden
| | - Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Lund, Sweden
| | | | | | | | | | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Lund, Sweden
| | | |
Collapse
|
19
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
20
|
Grønning Hansen M, Laterza C, Palma-Tortosa S, Kvist G, Monni E, Tsupykov O, Tornero D, Uoshima N, Soriano J, Bengzon J, Martino G, Skibo G, Lindvall O, Kokaia Z. Grafted human pluripotent stem cell-derived cortical neurons integrate into adult human cortical neural circuitry. Stem Cells Transl Med 2020; 9:1365-1377. [PMID: 32602201 PMCID: PMC7581452 DOI: 10.1002/sctm.20-0134] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Several neurodegenerative diseases cause loss of cortical neurons, leading to sensory, motor, and cognitive impairments. Studies in different animal models have raised the possibility that transplantation of human cortical neuronal progenitors, generated from pluripotent stem cells, might be developed into a novel therapeutic strategy for disorders affecting cerebral cortex. For example, we have shown that human long‐term neuroepithelial‐like stem (lt‐NES) cell‐derived cortical neurons, produced from induced pluripotent stem cells and transplanted into stroke‐injured adult rat cortex, improve neurological deficits and establish both afferent and efferent morphological and functional connections with host cortical neurons. So far, all studies with human pluripotent stem cell‐derived neurons have been carried out using xenotransplantation in animal models. Whether these neurons can integrate also into adult human brain circuitry is unknown. Here, we show that cortically fated lt‐NES cells, which are able to form functional synaptic networks in cell culture, differentiate to mature, layer‐specific cortical neurons when transplanted ex vivo onto organotypic cultures of adult human cortex. The grafted neurons are functional and establish both afferent and efferent synapses with adult human cortical neurons in the slices as evidenced by immuno‐electron microscopy, rabies virus retrograde monosynaptic tracing, and whole‐cell patch‐clamp recordings. Our findings provide the first evidence that pluripotent stem cell‐derived neurons can integrate into adult host neural networks also in a human‐to‐human grafting situation, thereby supporting their potential future clinical use to promote recovery by neuronal replacement in the patient's diseased brain.
Collapse
Affiliation(s)
| | - Cecilia Laterza
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Giedre Kvist
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emanuela Monni
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Oleg Tsupykov
- Bogomoletz Institute of Physiology and State Institute of Genetic and Regenerative Medicine, Kyiv, Ukraine
| | - Daniel Tornero
- Laboratory of Stem Cells and Regenerative Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Naomi Uoshima
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Institute of Complex Systems (UBICS), Universitat de Barcelona, Barcelona, Spain
| | - Johan Bengzon
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Division of Neurosurgery, Department of Clinical Sciences Lund, University Hospital, Lund, Sweden
| | - Gianvito Martino
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Galyna Skibo
- Bogomoletz Institute of Physiology and State Institute of Genetic and Regenerative Medicine, Kyiv, Ukraine
| | - Olle Lindvall
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Pottoo FH, Tabassum N, Javed MN, Nigar S, Sharma S, Barkat MA, Harshita, Alam MS, Ansari MA, Barreto GE, Ashraf GM. Raloxifene potentiates the effect of fluoxetine against maximal electroshock induced seizures in mice. Eur J Pharm Sci 2020; 146:105261. [PMID: 32061655 DOI: 10.1016/j.ejps.2020.105261] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 01/17/2023]
Abstract
The evidence to guide clinicians regarding rationale polytherapy with current antiepileptic drugs (AEDs) is lacking, and current practice recommendations are largely empirical. The excessive drug loading with combinatorial therapies of existing AEDs are associated with escalated neurotoxicity, and that emergence of pharmacoresistant seizures couldn't be averted. In pursuit of judicious selection of novel AEDs in combinatorial therapies with mechanism based evidences, standardized dose of raloxifene, fluoxetine, bromocriptine and their low dose combinations, were experimentally tested for their impact on maximal electroshock (MES) induced tonic hind limb extension (THLE) in mice. Hippocampal neuropeptide Y (NPY) levels, oxidative stress and histopathological studies were undertaken. The results suggest the potentiating effect of 4 mg/kg raloxifene on 14 mg/kg fluoxetine against MES induced THLE, as otherwise monotherapy with 4 mg/kg raloxifene was unable to produce an effect. The results also depicted better efficacy than carbamazepine (20 mg/kg), standard AED. Most profoundly, MES-induced significant (P < 0.001) reduction in hippocampal NPY levels, that were escalated insignificantly with the duo-drug combination, suggesting some other mechanism in mitigation of electroshock induced seizures. These results were later corroborated with assays to assess oxidative stress and neuronal damage. In conclusion, the results demonstrated the propitious therapeutic benefit of duo-drug low dose combination of drugs; raloxifene and fluoxetine, with diverse mode of actions fetching greater effectiveness in the management of generalized tonic clonic seizures (GTCS).
Collapse
Affiliation(s)
- Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam, 31441 Saudi Arabia.
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech, University of Kashmir, Srinagar, India.
| | - Md Noushad Javed
- Department of Pharmaceutics, School of Pharmaceutical Sciences and Research, Jamia Hamdard University, New Delhi, India; School of Pharmaceutical Sciences, Apeejay Stya University, Gurugram, Haryana, India
| | - Shah Nigar
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech, University of Kashmir, Srinagar, India
| | - Shrestha Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, K.R.Mangalam University, Gurgaon, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al Batin 39524, Saudi Arabia
| | - Harshita
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al Batin 39524, Saudi Arabia
| | - Md Sabir Alam
- Department of Pharmacy, School of Medical and Allied Sciences, K.R.Mangalam University, Gurgaon, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam, 31441 Saudi Arabia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Ireland.
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
22
|
Inhibition of epileptiform activity by neuropeptide Y in brain tissue from drug-resistant temporal lobe epilepsy patients. Sci Rep 2019; 9:19393. [PMID: 31852985 PMCID: PMC6920462 DOI: 10.1038/s41598-019-56062-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 11/18/2019] [Indexed: 01/30/2023] Open
Abstract
In epilepsy patients, drug-resistant seizures often originate in one of the temporal lobes. In selected cases, when certain requirements are met, this area is surgically resected for therapeutic reasons. We kept the resected tissue slices alive in vitro for 48 h to create a platform for testing a novel treatment strategy based on neuropeptide Y (NPY) against drug-resistant epilepsy. We demonstrate that NPY exerts a significant inhibitory effect on epileptiform activity, recorded with whole-cell patch-clamp, in human hippocampal dentate gyrus. Application of NPY reduced overall number of paroxysmal depolarising shifts and action potentials. This effect was mediated by Y2 receptors, since application of selective Y2-receptor antagonist blocked the effect of NPY. This proof-of-concept finding is an important translational milestone for validating NPY-based gene therapy for targeting focal drug-resistant epilepsies, and increasing the prospects for positive outcome in potential clinical trials.
Collapse
|
23
|
Effects of Huazhuo Jiedu Shugan Decoction on Cognitive and Emotional Disorders in a Rat Model of Epilepsy: Possible Involvement of AC-cAMP-CREB Signaling and NPY Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4352879. [PMID: 31915447 PMCID: PMC6930777 DOI: 10.1155/2019/4352879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/29/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
Abstract
Background Huazhuo Jiedu Shugan decoction (HJSD), a traditional Chinese medicine (TCM), has been used to treat epileptic seizures for many years. Some ingredients in these herbs have been demonstrated to be effective for the treatment of brain damage caused by epilepsy. Aim of the Study The object of the study is to determine the effects of HJSD on cognitive and emotional disorders in a rat model of epilepsy. Materials and Methods After a predetermined time period, rats were intraperitoneally injected with pentylenetetrazol and observed in different phases of convulsions. The cognitive and emotional changes in the epileptic rats were assessed using behavioral and immunohistochemical tests. Results Compared with the epilepsy group, the seizure grade was reduced and seizure latency was prolonged following HJSD-H treatment (P < 0.01). Compared with the control group, the epilepsy group displayed marked worse performance on the animal behavior tests (P < 0.05) and the HJSD-H group displayed improved behavioral performance (P < 0.05). After HJSD-H treatment, the expression of adenylate cyclase (AC), cyclic adenosine monophosphate (cAMP), cAMP-response element binding protein (CREB), and neuropeptide Y (NPY) immunoreactive cells markedly increased in the hippocampus, compared with that of the epilepsy group (P < 0.05). Conclusions The current results demonstrate that HJSD treatment in epileptic rats markedly inhibits epileptic seizures and improves cognitive and emotional disorders, which may be related to the regulation of AC-cAMP-CREB signaling and NPY expression in the hippocampus. The effects of the HJSD treatment may provide a foundation for the use of HJSD as a prescription medicinal herb in the TCM for the treatment of epilepsy.
Collapse
|
24
|
Agostinho AS, Mietzsch M, Zangrandi L, Kmiec I, Mutti A, Kraus L, Fidzinski P, Schneider UC, Holtkamp M, Heilbronn R, Schwarzer C. Dynorphin-based "release on demand" gene therapy for drug-resistant temporal lobe epilepsy. EMBO Mol Med 2019; 11:e9963. [PMID: 31486590 PMCID: PMC6783645 DOI: 10.15252/emmm.201809963] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 08/01/2019] [Accepted: 08/06/2019] [Indexed: 11/13/2022] Open
Abstract
Focal epilepsy represents one of the most common chronic CNS diseases. The high incidence of drug resistance, devastating comorbidities, and insufficient responsiveness to surgery pose unmet medical challenges. In the quest of novel, disease-modifying treatment strategies of neuropeptides represent promising candidates. Here, we provide the "proof of concept" that gene therapy by adeno-associated virus (AAV) vector transduction of preprodynorphin into the epileptogenic focus of well-accepted mouse and rat models for temporal lobe epilepsy leads to suppression of seizures over months. The debilitating long-term decline of spatial learning and memory is prevented. In human hippocampal slices obtained from epilepsy surgery, dynorphins suppressed seizure-like activity, suggestive of a high potential for clinical translation. AAV-delivered preprodynorphin expression is focally and neuronally restricted and release is dependent on high-frequency stimulation, as it occurs at the onset of seizures. The novel format of "release on demand" dynorphin delivery is viewed as a key to prevent habituation and to minimize the risk of adverse effects, leading to long-term suppression of seizures and of their devastating sequel.
Collapse
Affiliation(s)
| | - Mario Mietzsch
- Institute of VirologyCampus Benjamin Franklin, Charité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Luca Zangrandi
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Iwona Kmiec
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Anna Mutti
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Larissa Kraus
- Department of NeurologyCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Epilepsy‐Center Berlin‐BrandenburgBerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
| | - Pawel Fidzinski
- Department of NeurologyCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Epilepsy‐Center Berlin‐BrandenburgBerlinGermany
| | - Ulf C Schneider
- Department of NeurosurgeryCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Martin Holtkamp
- Department of NeurologyCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Epilepsy‐Center Berlin‐BrandenburgBerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
| | - Regine Heilbronn
- Institute of VirologyCampus Benjamin Franklin, Charité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
| | | |
Collapse
|
25
|
Melin E, Nanobashvili A, Avdic U, Gøtzsche CR, Andersson M, Woldbye DPD, Kokaia M. Disease Modification by Combinatorial Single Vector Gene Therapy: A Preclinical Translational Study in Epilepsy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:179-193. [PMID: 31660420 PMCID: PMC6807261 DOI: 10.1016/j.omtm.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Gene therapy has been suggested as a plausible novel approach to achieve seizure control in patients with focal epilepsy that do not adequately respond to pharmacological treatment. We investigated the seizure-suppressant potential of combinatorial neuropeptide Y and Y2 receptor single vector gene therapy based on adeno-associated virus serotype 1 (AAV1) in rats. First, a dose-response study in the systemic kainate-induced acute seizure model was performed, whereby the 1012 genomic particles (gp)/mL titer of the vector was selected as an optimal concentration. Second, an efficacy study was performed in the intrahippocampal kainate chronic model of spontaneous recurrent seizures (SRSs), designed to reflect a likely clinical scenario, with magnetic resonance image (MRI)-guided focal unilateral administration of the vector in the hippocampus during the chronic stage of the disease. The efficacy study demonstrated a favorable outcome of the gene therapy, with a 31% responder rate (more than 50% reduction in SRS frequency) and 13% seizure-freedom rate, whereas no such effects were observed in the control animals. The inter-SRS and SRS cluster intervals were also significantly prolonged in the treated group compared to controls. In addition, the SRS duration was significantly reduced in the treated group but not in the controls. This study establishes the SRS-suppressant ability of the single vector combinatorial neuropeptide Y/Y2 receptor gene therapy in a clinically relevant chronic model of epilepsy.
Collapse
Affiliation(s)
- Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Avtandil Nanobashvili
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden.,CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden
| | - Una Avdic
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Casper R Gøtzsche
- CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden.,Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - My Andersson
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - David P D Woldbye
- Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| |
Collapse
|
26
|
Kraus L, Hetsch F, Schneider UC, Radbruch H, Holtkamp M, Meier JC, Fidzinski P. Dimethylethanolamine Decreases Epileptiform Activity in Acute Human Hippocampal Slices in vitro. Front Mol Neurosci 2019; 12:209. [PMID: 31551707 PMCID: PMC6743366 DOI: 10.3389/fnmol.2019.00209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/09/2019] [Indexed: 01/13/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common form of focal epilepsy with about 30% of patients developing pharmacoresistance. These patients continue to suffer from seizures despite polytherapy with antiepileptic drugs (AEDs) and have an increased risk for premature death, thus requiring further efforts for the development of new antiepileptic therapies. The molecule dimethylethanolamine (DMEA) has been tested as a potential treatment in various neurological diseases, albeit the functional mechanism of action was never fully understood. In this study, we investigated the effects of DMEA on neuronal activity in single-cell recordings of primary neuronal cultures. DMEA decreased the frequency of spontaneous synaptic events in a concentration-dependent manner with no apparent effect on resting membrane potential (RMP) or action potential (AP) threshold. We further tested whether DMEA can exert antiepileptic effects in human brain tissue ex vivo. We analyzed the effect of DMEA on epileptiform activity in the CA1 region of the resected hippocampus of TLE patients in vitro by recording extracellular field potentials in the pyramidal cell layer. Epileptiform burst activity in resected hippocampal tissue from TLE patients remained stable over several hours and was pharmacologically suppressed by lacosamide, demonstrating the applicability of our platform to test antiepileptic efficacy. Similar to lacosamide, DMEA also suppressed epileptiform activity in the majority of samples, albeit with variable interindividual effects. In conclusion, DMEA might present a new approach for treatment in pharmacoresistant TLE and further studies will be required to identify its exact mechanism of action and the involved molecular targets.
Collapse
Affiliation(s)
- Larissa Kraus
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Hetsch
- Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ulf C. Schneider
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurosurgery, Berlin, Germany
| | - Helena Radbruch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
| | - Martin Holtkamp
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Jochen C. Meier
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
- Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Pawel Fidzinski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
| |
Collapse
|
27
|
Ingusci S, Cattaneo S, Verlengia G, Zucchini S, Simonato M. A Matter of Genes: The Hurdles of Gene Therapy for Epilepsy. Epilepsy Curr 2019; 19:38-43. [PMID: 30838918 PMCID: PMC6610370 DOI: 10.1177/1535759718822846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Gene therapy has recently advanced to the level of standard of care for several
diseases. However, its application to neurological disorders is still in the
experimental phase. In this review, we discuss recent advancements in the field
that provide optimism on the possibility to have first-in-human studies for gene
therapy of some forms of epilepsy in the not so distant future.
Collapse
Affiliation(s)
- Selene Ingusci
- 1 Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Stefano Cattaneo
- 2 School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Gianluca Verlengia
- 1 Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,2 School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvia Zucchini
- 1 Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,3 Technopole of Ferrara, LTTA Laboratory for the Technologies for Advanced Therapies, Ferrara, Italy
| | - Michele Simonato
- 1 Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,2 School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
28
|
Soud K, Jørgensen SH, Woldbye DPD, Sørensen AT. The C-terminal flanking peptide of neuropeptide Y (NPY) is not essential for seizure-suppressant actions of prepro-NPY overexpression in male rats. J Neurosci Res 2018; 97:362-372. [PMID: 30367522 DOI: 10.1002/jnr.24350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 11/11/2022]
Abstract
The full coding sequence of neuropeptide Y (NPY), prepro-NPY, is sequentially metabolized into three peptides; an N-terminus 28-amino acid signaling peptide, the NPY peptide itself (NPY1-36), and a 30-amino acid C-terminus peptide, known as the C-terminal flanking peptide of neuropeptide-Y (CPON). While the signaling peptide directs intracellular trafficking and NPY1-36 is well characterized, the biological function of CPON is unknown. This is noteworthy because CPON is co-stored and co-released along with NPY1-36 and could thus potentially serve important functions. To assess the role of CPON, we adapted a viral genetic approach using two different vector designs encoding NPY, but where the CPON coding sequence was excluded from one of the vectors. Thus, the effect of CPON was indirectly assessed. Male rats received intrahippocampal injections of either a vector encoding NPY1-39 whose metabolism yields NPY1-36 and not CPON, or a prepro-NPY vector encoding both NPY1-36 and CPON. A third vector encoding EGFP served as control. We subsequently studied to what extent CPON might affect seizure susceptibility and memory performance, respectively, to address two important questions to evaluate the potential of NPY gene therapy in epilepsy. Both NPY vectors, as compared to EGFP control, were found to be equally effective at suppressing acute kainate-induced seizures, and both did not influence learning and memory performance in the Morris water maze. Thus CPON itself does not appear to aid actions governed by vector-mediated overexpression of NPY1-36 within the hippocampus. Whether CPON serves other important functions remains to be determined.
Collapse
Affiliation(s)
- Katia Soud
- Laboratory of Neural Plasticity, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Søren Heide Jørgensen
- Neuropharmacology and Genetics Laboratory, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - David Paul Drucker Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Toft Sørensen
- Neuropharmacology and Genetics Laboratory, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Ali I, Gandrathi A, Zheng T, Morris MJ, O'Brien TJ, French C. Neuropeptide Y affects thalamic reticular nucleus neuronal firing and network synchronization associated with suppression of spike-wave discharges. Epilepsia 2018; 59:1444-1454. [PMID: 29923603 DOI: 10.1111/epi.14451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Neuropeptide Y (NPY) potently suppresses spike-wave discharges (SWDs) in a genetic rat model of absence epilepsy (GAERS), but the underlying neurophysiologic mechanisms are not clear. We therefore sought to determine the in vivo effects of NPY on neuronal firing in the cortico-thalamo-cortical network activity, known to play a critical role in the generation of SWDs in these rats. METHODS NPY was administered intracerebroventricularly (ICV) or in separate experiments locally on the neurons of caudal thalamic reticular nucleus (NRT) by use of juxtacellular iontophoresis in triple-barrel electrodes in male GAERS aged 12-15 weeks, in vivo under neuroleptic anesthesia. Drug infusions and electroencephalography (EEG) monitoring were performed simultaneously with juxtacellular single neuronal recordings. Effect of NPY on electrically induced SWD induction threshold were also measured. RESULTS NPY administration ICV led to a decrease in the total length of SWDs in EEG recordings. Both ICV administration and iontophoresis of NPY on NRT neurons led to an increase in interictal neuronal firing of NRT neurons. During ictal periods, ICV NPY administration reduced the number of thalamic action potentials per SWDs, as well as reduced waveform correlations between field potentials within the NRT and the cortical EEG. NPY administration ICV did not significantly alter the firing patterns of relay thalamic neurons interictally and cortical neurons during ictal and interictal periods. In addition, SWD induction threshold in the S2 region of the cortex was significantly increased after NPY administration. SIGNIFICANCE Our results show alterations in cortico-thalamo-cortical local and network properties following ICV administration of NPY, suggesting mechanisms of SWD suppression in GAERS. Cellular and network alteration of NRT activity, resulting from a direct action of NPY, may be a contributor to this effect.
Collapse
Affiliation(s)
- Idrish Ali
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville, Vic., Australia.,Departments of Neuroscience and Neurology, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia
| | - Arun Gandrathi
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville, Vic., Australia
| | - Thomas Zheng
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville, Vic., Australia
| | - Margaret J Morris
- Department of Physiology and Pharmacology, The University of New South Wales, Randwick, NSW, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville, Vic., Australia.,Departments of Neuroscience and Neurology, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia
| | - Chris French
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
30
|
Ting JT, Kalmbach B, Chong P, de Frates R, Keene CD, Gwinn RP, Cobbs C, Ko AL, Ojemann JG, Ellenbogen RG, Koch C, Lein E. A robust ex vivo experimental platform for molecular-genetic dissection of adult human neocortical cell types and circuits. Sci Rep 2018; 8:8407. [PMID: 29849137 PMCID: PMC5976666 DOI: 10.1038/s41598-018-26803-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 05/16/2018] [Indexed: 12/20/2022] Open
Abstract
The powerful suite of available genetic tools is driving tremendous progress in understanding mouse brain cell types and circuits. However, the degree of conservation in human remains largely unknown in large part due to the lack of such tools and healthy tissue preparations. To close this gap, we describe a robust and stable adult human neurosurgically-derived ex vivo acute and cultured neocortical brain slice system optimized for rapid molecular-genetic manipulation. Surprisingly, acute human brain slices exhibited exceptional viability, and neuronal intrinsic membrane properties could be assayed for at least three days. Maintaining adult human slices in culture under sterile conditions further enabled the application of viral tools to drive rapid expression of exogenous transgenes. Widespread neuron-specific labeling was achieved as early as two days post infection with HSV-1 vectors, with virally-transduced neurons exhibiting membrane properties largely comparable to uninfected neurons over this short timeframe. Finally, we demonstrate the suitability of this culture paradigm for optical manipulation and monitoring of neuronal activity using genetically encoded probes, opening a path for applying modern molecular-genetic tools to study human brain circuit function.
Collapse
Affiliation(s)
| | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA.,Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Peter Chong
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Ryder P Gwinn
- Epilepsy Surgery and Functional Neurosurgery, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Charles Cobbs
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Andrew L Ko
- Regional Epilepsy Center at Harborview Medical Center, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Jeffrey G Ojemann
- Regional Epilepsy Center at Harborview Medical Center, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Richard G Ellenbogen
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
31
|
Abstract
Resected hippocampal tissue from patients with drug-resistant epilepsy presents a unique possibility to test novel treatment strategies directly in target tissue. The post-resection time for testing and analysis however is normally limited. Acute tissue slices allow for electrophysiological recordings typically up to 12 hours. To enable longer time to test novel treatment strategies such as, e.g., gene-therapy, we developed a method for keeping acute human brain slices viable over a longer period. Our protocol keeps neurons viable well up to 48 hours. Using a dual-flow chamber, which allows for microscopic visualisation of individual neurons with a submerged objective for whole-cell patch-clamp recordings, we report stable electrophysiological properties, such as action potential amplitude and threshold during this time. We also demonstrate that epileptiform activity, monitored by individual dentate granule whole-cell recordings, can be consistently induced in these slices, underlying the usefulness of this methodology for testing and/or validating novel treatment strategies for epilepsy.
Collapse
|
32
|
Agarwal AK, Tunison K, Dalal JS, Nagamma SS, Hamra FK, Sankella S, Shao X, Auchus RJ, Garg A. Metabolic, Reproductive, and Neurologic Abnormalities in Agpat1-Null Mice. Endocrinology 2017; 158:3954-3973. [PMID: 28973305 PMCID: PMC5695831 DOI: 10.1210/en.2017-00511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/20/2017] [Indexed: 12/11/2022]
Abstract
Defects in the biosynthesis of phospholipids and neutral lipids are associated with cell membrane dysfunction, disrupted energy metabolism, and diseases including lipodystrophy. In these pathways, the 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) enzymes transfer a fatty acid to the sn-2 carbon of sn-1-acylglycerol-3-phosphate (lysophosphatidic acid) to form sn-1, 2-acylglycerol-3-phosphate [phosphatidic acid (PA)]. PA is a precursor for key phospholipids and diacylglycerol. AGPAT1 and AGPAT2 are highly homologous isoenzymes that are both expressed in adipocytes. Genetic defects in AGPAT2 cause congenital generalized lipodystrophy, indicating that AGPAT1 cannot compensate for loss of AGPAT2 in adipocytes. To further explore the physiology of AGPAT1, we characterized a loss-of-function mouse model (Agpat1-/-). The majority of Agpat1-/- mice died before weaning and had low body weight and low plasma glucose levels, independent of plasma insulin and glucagon levels, with reduced percentage of body fat but not generalized lipodystrophy. These mice also had decreased hepatic messenger RNA expression of Igf-1 and Foxo1, suggesting a decrease in gluconeogenesis. In male mice, sperm development was impaired, with a late meiotic arrest near the onset of round spermatid production, and gonadotropins were elevated. Female mice showed oligoanovulation yet retained responsiveness to gonadotropins. Agpat1-/- mice also demonstrated abnormal hippocampal neuron development and developed audiogenic seizures. In summary, Agpat1-/- mice developed widespread disturbances of metabolism, sperm development, and neurologic function resulting from disrupted phospholipid homeostasis. AGPAT1 appears to serve important functions in the physiology of multiple organ systems. The Agpat1-deficient mouse provides an important model in which to study the contribution of phospholipid and triacylglycerol synthesis to physiology and diseases.
Collapse
Affiliation(s)
- Anil K. Agarwal
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Katie Tunison
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jasbir S. Dalal
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Sneha S. Nagamma
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - F. Kent Hamra
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Shireesha Sankella
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Xinli Shao
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Richard J. Auchus
- Department of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
33
|
Lepeta K, Lourenco MV, Schweitzer BC, Martino Adami PV, Banerjee P, Catuara-Solarz S, de La Fuente Revenga M, Guillem AM, Haidar M, Ijomone OM, Nadorp B, Qi L, Perera ND, Refsgaard LK, Reid KM, Sabbar M, Sahoo A, Schaefer N, Sheean RK, Suska A, Verma R, Vicidomini C, Wright D, Zhang XD, Seidenbecher C. Synaptopathies: synaptic dysfunction in neurological disorders - A review from students to students. J Neurochem 2016; 138:785-805. [PMID: 27333343 PMCID: PMC5095804 DOI: 10.1111/jnc.13713] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
Synapses are essential components of neurons and allow information to travel coordinately throughout the nervous system to adjust behavior to environmental stimuli and to control body functions, memories, and emotions. Thus, optimal synaptic communication is required for proper brain physiology, and slight perturbations of synapse function can lead to brain disorders. In fact, increasing evidence has demonstrated the relevance of synapse dysfunction as a major determinant of many neurological diseases. This notion has led to the concept of synaptopathies as brain diseases with synapse defects as shared pathogenic features. In this review, which was initiated at the 13th International Society for Neurochemistry Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental disorders (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer and Parkinson disease). We finally discuss the appropriateness and potential implications of gathering synapse diseases under a single term. Understanding common causes and intrinsic differences in disease-associated synaptic dysfunction could offer novel clues toward synapse-based therapeutic intervention for neurological and neuropsychiatric disorders. In this Review, which was initiated at the 13th International Society for Neurochemistry (ISN) Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer's and Parkinson's diseases), gathered together under the term of synaptopathies. Read the Editorial Highlight for this article on page 783.
Collapse
Affiliation(s)
- Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara C Schweitzer
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Pamela V Martino Adami
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir-IIBBA-CONICET, Buenos Aires, Argentina
| | - Priyanjalee Banerjee
- Department of Biochemistry, Institute of Post Graduate Medical Education & Research, Kolkata, West Bengal, India
| | - Silvina Catuara-Solarz
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Mario de La Fuente Revenga
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Alain Marc Guillem
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F. 07000, Mexico
| | - Mouna Haidar
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, Cross River University of Technology, Okuku Campus, Cross River, Nigeria
| | - Bettina Nadorp
- The Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Israel
| | - Lin Qi
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Nirma D Perera
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Louise K Refsgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kimberley M Reid
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Mariam Sabbar
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Arghyadip Sahoo
- Department of Biochemistry, Midnapore Medical College, West Bengal University of Health Sciences, West Bengal, India
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Rebecca K Sheean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anna Suska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Rajkumar Verma
- Department of Neurosciences Uconn Health Center, Farmington, CT, United States of America
| | | | - Dean Wright
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Xing-Ding Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Constanze Seidenbecher
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences (CBBS) Magdeburg, Magdeburg, Germany.
| |
Collapse
|
34
|
Avaliani N, Andersson M, Runegaard AH, Woldbye D, Kokaia M. DREADDs suppress seizure-like activity in a mouse model of pharmacoresistant epileptic brain tissue. Gene Ther 2016; 23:760-766. [PMID: 27416078 DOI: 10.1038/gt.2016.56] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Epilepsy is a neurological disorder with a prevalence of ≈1% of general population. Available antiepileptic drugs (AEDs) have multiple side effects and are ineffective in 30% of patients. Therefore, development of effective treatment strategies is highly needed, requiring drug-screening models that are relevant and reliable. We investigated novel chemogenetic approach, using DREADDs (designer receptors exclusively activated by designer drugs) as possible inhibitor of epileptiform activity in organotypic hippocampal slice cultures (OHSCs). The OHSCs are characterized by increased overall excitability and closely resemble features of human epileptic tissue. Studies suggest that chemically induced epileptiform activity in rat OHSCs is pharmacoresistant to most of AEDs. However, high-frequency electric stimulus train-induced bursting (STIB) in OHSCs is responsive to carbamazepine and phenytoin. We investigated whether inhibitory DREADD, hM4Di, would be effective in suppressing STIB in OHSC. hM4Di is a mutated muscarinic receptor selectively activated by otherwise inert clozapine-N-oxide, which leads to hyperpolarization in neurons. We demonstrated that this hyperpolarization effectively suppresses STIB in mouse OHSCs. As we also found that STIB in mouse OHSCs is resistant to common AED, valproic acid, collectively our findings suggest that DREADD-based strategy may be effective in suppressing epileptiform activity in a pharamcoresitant epileptic brain tissue.
Collapse
Affiliation(s)
- N Avaliani
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - M Andersson
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - A H Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - M Kokaia
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| |
Collapse
|
35
|
Optogenetic control of human neurons in organotypic brain cultures. Sci Rep 2016; 6:24818. [PMID: 27098488 PMCID: PMC4838935 DOI: 10.1038/srep24818] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/06/2016] [Indexed: 01/16/2023] Open
Abstract
Optogenetics is one of the most powerful tools in neuroscience, allowing for selective control of specific neuronal populations in the brain of experimental animals, including mammals. We report, for the first time, the application of optogenetic tools to human brain tissue providing a proof-of-concept for the use of optogenetics in neuromodulation of human cortical and hippocampal neurons as a possible tool to explore network mechanisms and develop future therapeutic strategies.
Collapse
|
36
|
Tasan RO, Verma D, Wood J, Lach G, Hörmer B, de Lima TCM, Herzog H, Sperk G. The role of Neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016; 55:111-26. [PMID: 26444585 DOI: 10.1016/j.npep.2015.09.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/10/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
While anxiety disorders are the brain disorders with the highest prevalence and constitute a major burden for society, a considerable number of affected people are still treated insufficiently. Thus, in an attempt to identify potential new anxiolytic drug targets, neuropeptides have gained considerable attention in recent years. Compared to classical neurotransmitters they often have a regionally restricted distribution and may bind to several distinct receptor subtypes. Neuropeptide Y (NPY) is a highly conserved neuropeptide that is specifically concentrated in limbic brain areas and signals via at least 5 different G-protein-coupled receptors. It is involved in a variety of physiological processes including the modulation of emotional-affective behaviors. An anxiolytic and stress-reducing property of NPY is supported by many preclinical studies. Whether NPY may also interact with processing of learned fear and fear extinction is comparatively unknown. However, this has considerable relevance since pathological, inappropriate and generalized fear expression and impaired fear extinction are hallmarks of human post-traumatic stress disorder and a major reason for its treatment-resistance. Recent evidence from different laboratories emphasizes a fear-reducing role of NPY, predominantly mediated by exogenous NPY acting on Y1 receptors. Since a reduction of fear expression was also observed in Y1 receptor knockout mice, other Y receptors may be equally important. By acting on Y2 receptors, NPY promotes fear extinction and generates a long-term suppression of fear, two important preconditions that could support cognitive behavioral therapies in human patients. A similar effect has been demonstrated for the closely related pancreatic polypeptide (PP) when acting on Y4 receptors. Preliminary evidence suggests that NPY modulates fear in particular by activation of Y1 and Y2 receptors in the basolateral and central amygdala, respectively. In the basolateral amygdala, NPY signaling activates inhibitory G protein-coupled inwardly-rectifying potassium channels or suppresses hyperpolarization-induced I(h) currents in a Y1 receptor-dependent fashion, favoring a general suppression of neuronal activity. A more complex situation has been described for the central extended amygdala, where NPY reduces the frequency of inhibitory and excitatory postsynaptic currents. In particular the inhibition of long-range central amygdala output neurons may result in a Y2 receptor-dependent suppression of fear. The role of NPY in processes of learned fear and fear extinction is, however, only beginning to emerge, and multiple questions regarding the relevance of endogenous NPY and different receptor subtypes remain elusive. Y2 receptors may be of particular interest for future studies, since they are the most prominent Y receptor subtype in the human brain and thus among the most promising therapeutic drug targets when translating preclinical evidence to potential new therapies for human anxiety disorders.
Collapse
Affiliation(s)
- R O Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - D Verma
- Institute of Physiology I, University of Münster, D-48149 Münster, Germany
| | - J Wood
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - G Lach
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria; Capes Foundation, Ministry of Education of Brazil, 70040-020 Brasília/DF, Brazil
| | - B Hörmer
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - T C M de Lima
- Department of Pharmacology, Federal University of Santa Catarina, 88049-970 Florianópolis, Brazil
| | - H Herzog
- Neuroscience Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - G Sperk
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|