1
|
Gonzalez-Ramos A, Berglind F, Kudláček J, Rocha ER, Melin E, Sebastião AM, Valente CA, Ledri M, Andersson M, Kokaia M. Chemogenetics with PSAM 4-GlyR decreases excitability and epileptiform activity in epileptic hippocampus. Gene Ther 2025; 32:106-120. [PMID: 39455855 PMCID: PMC11946892 DOI: 10.1038/s41434-024-00493-7] [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: 05/17/2024] [Revised: 09/28/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Despite the availability of new drugs on the clinics in recent years, drug-resistant epilepsy remains an unresolved challenge for healthcare, and one-third of epilepsy patients remain refractory to anti-seizure medications. Gene therapy in experimental models has emerged as effective treatment targeting specific neuronal populations in the epileptogenic focus. When combined with an external chemical activator using chemogenetics, it also becomes an "on-demand" treatment. Here, we evaluate a targeted and specific chemogenetic therapy, the PSAM/PSEM system, which holds promise as a potential candidate for clinical application in treating drug-resistant epilepsy. We show that the inert ligand uPSEM817, which selectively activates the chloride-permeable channel PSAM4-GlyR, effectively reduces the number of depolarization-induced action potentials in vitro. This effect is likely due to the shunting of depolarizing currents, as evidenced by decreased membrane resistance in these cells. In organotypic slices, uPSEM817 decreased the number of bursts and peak amplitude of events of spontaneous epileptiform activity. Although administration of uPSEM817 in vivo did not significantly alter electrographic seizures in a male mouse model of temporal lobe epilepsy, it did demonstrate a strong trend toward reducing the frequency of interictal epileptiform discharges. These findings indicate that PSAM4-GlyR-based chemogenetics holds potential as an anti-seizure strategy, although further refinement is necessary to enhance its efficacy.
Collapse
Affiliation(s)
- Ana Gonzalez-Ramos
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Fredrik Berglind
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Jan Kudláček
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Elza R Rocha
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Esbjörn Melin
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Marco Ledri
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - My Andersson
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Merab Kokaia
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
| |
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
|
Anand A, Shrivastava A, Singh K, Barik R, Gayakwad D, Jailani S, Shamim, Dwivedi S. Neuroprotective Efficacy and Complementary Treatment with Medicinal Herbs: A Comprehensive Review of Recent Therapeutic Approaches in Epilepsy Management. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:60-73. [PMID: 39069797 DOI: 10.2174/0118715273332140240724093837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Central Nervous System (CNS) disorders affect millions of people worldwide, with a significant proportion experiencing drug-resistant forms where conventional medications fail to provide adequate seizure control. This abstract delves into recent advancements and innovative therapies aimed at addressing the complex challenge of CNS-related drug-resistant epilepsy (DRE) management. The idea of precision medicine has opened up new avenues for epilepsy treatment. Herbs such as curcumin, ginkgo biloba, panax ginseng, bacopa monnieri, ashwagandha, and rhodiola rosea influence the BDNF pathway through various mechanisms. These include the activation of CREB, inhibition of NF-κB, modulation of neurotransmitters, reduction of oxidative stress, and anti- inflammatory effects. By promoting BDNF expression and activity, these herbs support neuroplasticity, cognitive function, and overall neuronal health. Novel antiepileptic drugs (AEDs) with distinct mechanisms of action demonstrate efficacy in refractory cases where traditional medications falter. Additionally, repurposing existing drugs for antiepileptic purposes presents a cost-effective strategy to broaden therapeutic choices. Cannabidiol (CBD), derived from cannabis herbs, has garnered attention for its anticonvulsant properties, offering a potential adjunctive therapy for refractory seizures. In conclusion, recent advances and innovative therapies represent a multifaceted approach to managing drug-resistant epilepsy. Leveraging precision medicine, neurostimulation technologies, novel pharmaceuticals, and complementary therapies, clinicians can optimize treatment outcomes and improve the life expectancy of patients living with refractory seizures. Genetic testing and biomarker identification now allow for personalized therapeutic approaches tailored to individual patient profiles. Utilizing next-generation sequencing techniques, researchers have elucidated genetic mutations.
Collapse
Affiliation(s)
- Amit Anand
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Aman Shrivastava
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Rakesh Barik
- GITAM School of Pharmacy, GITAM University, Hyderabad, Telangana, India
| | - Devshree Gayakwad
- Acropolis Institute of Pharmaceutical Education and Research, Indore, Madhya Pradesh, India
| | - S Jailani
- Formulation R&D Department, Alpha Pharma, KAEC, Rabigh, Kingdom of Saudi Arabia
| | - Shamim
- IIMT College of Medical Sciences, IIMT University, Ganga Nagar, Meerut, Uttar Pradesh, India
| | - Sumeet Dwivedi
- Acropolis Institute of Pharmaceutical Education and Research, Indore, Madhya Pradesh, India
| |
Collapse
|
4
|
Nelson TS, Allen HN, Khanna R. Neuropeptide Y and Pain: Insights from Brain Research. ACS Pharmacol Transl Sci 2024; 7:3718-3728. [PMID: 39698268 PMCID: PMC11651174 DOI: 10.1021/acsptsci.4c00333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024]
Abstract
Neuropeptide Y (NPY) is a highly conserved neuropeptide with widespread distribution in the central nervous system and diverse physiological functions. While extensively studied for its inhibitory effects on pain at the spinal cord level, its role in pain modulation within the brain remains less clear. This review aims to summarize the complex landscape of supraspinal NPY signaling in pain processing. We discuss the expression and function of NPY receptors in key pain-related brain regions, including the parabrachial nucleus, periaqueductal gray, amygdala, and nucleus accumbens. Additionally, we highlight the potent efficacy of NPY in attenuating pain sensitivity and nociceptive processing throughout the central nervous system. NPY-based therapeutic interventions targeting the central nervous system represent a promising avenue for novel analgesic strategies and pain-associated comorbidities.
Collapse
Affiliation(s)
- Tyler S. Nelson
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Heather N. Allen
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Rajesh Khanna
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
5
|
Mullagulova AI, Timechko EE, Solovyeva VV, Yakimov AM, Ibrahim A, Dmitrenko DD, Sufianov AA, Sufianova GZ, Rizvanov AA. Adeno-Associated Viral Vectors in the Treatment of Epilepsy. Int J Mol Sci 2024; 25:12081. [PMID: 39596149 PMCID: PMC11593886 DOI: 10.3390/ijms252212081] [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: 09/25/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Epilepsy is a brain disorder characterized by a persistent predisposition to epileptic seizures. With various etiologies of epilepsy, a significant proportion of patients develop pharmacoresistance to antiepileptic drugs, which necessitates the search for new therapeutic methods, in particular, using gene therapy. This review discusses the use of adeno-associated viral (AAV) vectors in gene therapy for epilepsy, emphasizing their advantages, such as high efficiency of neuronal tissue transduction and low immunogenicity/cytotoxicity. AAV vectors provide the possibility of personalized therapy due to the diversity of serotypes and genomic constructs, which allows for increasing the specificity and effectiveness of treatment. Promising orientations include the modulation of the expression of neuropeptides, ion channels, transcription, and neurotrophic factors, as well as the use of antisense oligonucleotides to regulate seizure activity, which can reduce the severity of epileptic disorders. This review summarizes the current advances in the use of AAV vectors for the treatment of epilepsy of various etiologies, demonstrating the significant potential of AAV vectors for the development of personalized and more effective approaches to reducing seizure activity and improving patient prognosis.
Collapse
Affiliation(s)
- Aysilu I. Mullagulova
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Elena E. Timechko
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Valeriya V. Solovyeva
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Alexey M. Yakimov
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Ahmad Ibrahim
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Diana D. Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Albert A. Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119991, Russia;
- The Research and Educational Institute of Neurosurgery, Peoples’ Friendship University of Russia, Moscow 117198, Russia
| | - Galina Z. Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia;
| | - Albert A. Rizvanov
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
- Division of Medical and Biological Sciences, Academy of Sciences of the Republic of Tatarstan, Kazan 420111, Russia
| |
Collapse
|
6
|
Cattaneo S, Bettegazzi B, Crippa L, Asth L, Regoni M, Soukupova M, Zucchini S, Cantore A, Codazzi F, Valtorta F, Simonato M. Gene therapy for epilepsy targeting neuropeptide Y and its Y2 receptor to dentate gyrus granule cells. EMBO Rep 2024; 25:4387-4409. [PMID: 39251828 PMCID: PMC11467199 DOI: 10.1038/s44319-024-00244-0] [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: 12/20/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Gene therapy is emerging as an alternative option for individuals with drug-resistant focal epilepsy. Here, we explore the potential of a novel gene therapy based on Neuropeptide Y (NPY), a well-known endogenous anticonvulsant. We develop a lentiviral vector co-expressing NPY with its inhibitory receptor Y2 in which, for the first time, both transgenes are placed under the control of the minimal CamKIIa(0.4) promoter, biasing expression toward excitatory neurons and allowing autoregulation of neuronal excitability by Y2 receptor-mediated inhibition. Vector-induced NPY and Y2 expression and safety are first assessed in cultures of hippocampal neurons. In vivo experiments demonstrate efficient and nearly selective overexpression of both genes in granule cell mossy fiber terminals following vector administration in the dentate gyrus. Telemetry video-EEG monitoring reveals a reduction in the frequency and duration of seizures in the synapsin triple KO model. This study shows that targeting a small subset of neurons (hippocampal granule cells) with a combined overexpression of NPY and Y2 receptor is sufficient to reduce the occurrence of spontaneous seizures.
Collapse
Affiliation(s)
- Stefano Cattaneo
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Barbara Bettegazzi
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Lucia Crippa
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Laila Asth
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Maria Regoni
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marie Soukupova
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Alessio Cantore
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20123, Milan, Italy
| | - Franca Codazzi
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Flavia Valtorta
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Michele Simonato
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
| |
Collapse
|
7
|
Kumagai S, Nakajima T, Muramatsu SI. Intraparenchymal delivery of adeno-associated virus vectors for the gene therapy of neurological diseases. Expert Opin Biol Ther 2024; 24:773-785. [PMID: 39066718 DOI: 10.1080/14712598.2024.2386339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/07/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION In gene therapy with adeno-associated virus (AAV) vectors for diseases of the central nervous system, the vectors can be administered into blood vessels, cerebrospinal fluid space, or the brain parenchyma. When gene transfer to a large area of the brain is required, the first two methods are used, but for diseases in which local gene transfer is expected to be effective, vectors are administered directly into the brain parenchyma. AREAS COVERED Strategies for intraparenchymal vector delivery in gene therapy for Parkinson's disease, aromatic l-amino acid decarboxylase (AADC) deficiency, and epilepsy are reviewed. EXPERT OPINION Stereotactic intraparenchymal injection of AAV vectors allows precise gene delivery to the target site. Although more surgically invasive than intravascular or intrathecal administration, intraparenchymal vector delivery has the advantage of a lower vector dose, and preexisting neutralizing antibodies have little effect on the transduction efficacy. This approach improves motor function in AADC deficiency and led to regulatory approval of an AAV vector for the disease in the EU. Although further validation through clinical studies is needed, direct infusion of viral vectors into the brain parenchyma is expected to be a novel treatment for Parkinson's disease and drug-resistant epilepsy.
Collapse
Affiliation(s)
- Shinichi Kumagai
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Takeshi Nakajima
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
8
|
Chen X, Luo J, Song M, Pan L, Qu Z, Huang B, Yu S, Shu H. Challenges and prospects in geriatric epilepsy treatment: the role of the blood-brain barrier in pharmacotherapy and drug delivery. Front Aging Neurosci 2024; 16:1342366. [PMID: 38389560 PMCID: PMC10882099 DOI: 10.3389/fnagi.2024.1342366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
The blood-brain barrier (BBB) is pivotal in maintaining neuronal physiology within the brain. This review delves into the alterations of the BBB specifically in the context of geriatric epilepsy. We examine how age-related changes in the BBB contribute to the pathogenesis of epilepsy in the elderly and present significant challenges in pharmacotherapy. Subsequently, we evaluate recent advancements in drug delivery methods targeting the BBB, as well as alternative approaches that could bypass the BBB's restrictive nature. We particularly highlight the use of neurotropic viruses and various synthetic nanoparticles that have been investigated for delivering a range of antiepileptic drugs. Additionally, the advantage and limitation of these diverse delivery methods are discussed. Finally, we analyze the potential efficacy of different drug delivery approaches in the treatment of geriatric epilepsy, aiming to provide insights into more effective management of this condition in the elderly population.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Juan Luo
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Min Song
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Liang Pan
- Department of Pediatrics, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Bo Huang
- Department of Burn and Plastic, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Sixun Yu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Haifeng Shu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
9
|
Metto AC, Telgkamp P, McLane-Svoboda AK, Gilad AA, Pelled G. Closed-loop neurostimulation via expression of magnetogenetics-sensitive protein in inhibitory neurons leads to reduction of seizure activity in a rat model of epilepsy. Brain Res 2023; 1820:148591. [PMID: 37748572 DOI: 10.1016/j.brainres.2023.148591] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
On-demand neurostimulation has shown success in epilepsy patients with pharmacoresistant seizures. Seizures produce magnetic fields that can be recorded using magnetoencephalography. We developed a new closed-loop approach to control seizure activity based on magnetogenetics using the electromagnetic perceptive gene (EPG) that encodes a protein that responds to magnetic fields. The EPG transgene was expressed in inhibitory interneurons under the hDlx promoter and kainic acid was used to induce acute seizures. In vivo electrophysiological signals were recorded. We found that hDlx EPG rats exhibited a significant delay in the onset of first seizure (1142.72 ± 186.35 s) compared to controls (644.03 ± 15.06 s) and significantly less seizures (4.11 ± 1.03) compared to controls (8.33 ± 1.58). These preliminary findings suggest that on-demand activation of EPG expressed in inhibitory interneurons suppresses seizure activity, and magnetogenetics via EPG may be an effective strategy to alleviate seizure severity in a closed-loop, and cell-specific fashion.
Collapse
Affiliation(s)
- Abigael C Metto
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Petra Telgkamp
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | - Autumn K McLane-Svoboda
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Assaf A Gilad
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, United States; Department of Radiology, Michigan State University, East Lansing, MI, United States; Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States; Department of Radiology, Michigan State University, East Lansing, MI, United States; Neuroscience Program, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Abstract
In recent years, there has been a significant increase in preclinical studies to test genetic therapies for epilepsy. Some of these therapies have advanced to clinical trials and are being tested in patients with monogenetic or focal refractory epilepsy. This article provides an overview of the current state of preclinical studies that show potential for clinical translation. Specifically, we focus on genetic therapies that have demonstrated a clear effect on seizures in animal models and have the potential to be translated to clinical settings. Both therapies targeting the cause of the disease and those that treat symptoms are discussed. We believe that the next few years will be crucial in determining the potential of genetic therapies for treating patients with epilepsy.
Collapse
Affiliation(s)
- James S. Street
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
13
|
Khatri DK, Preeti K, Tonape S, Bhattacharjee S, Patel M, Shah S, Singh PK, Srivastava S, Gugulothu D, Vora L, Singh SB. Nanotechnological Advances for Nose to Brain Delivery of Therapeutics to Improve the Parkinson Therapy. Curr Neuropharmacol 2023; 21:493-516. [PMID: 35524671 PMCID: PMC10207920 DOI: 10.2174/1570159x20666220507022701] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022] Open
Abstract
Blood-Brain Barrier (BBB) acts as a highly impermeable barrier, presenting an impediment to the crossing of most classical drugs targeted for neurodegenerative diseases including Parkinson's disease (PD). About the nature of drugs and other potential molecules, they impose unavoidable doserestricted limitations eventually leading to the failure of therapy. However, many advancements in formulation technology and modification of delivery approaches have been successful in delivering the drug to the brain in the therapeutic window. The nose to the brain (N2B) drug delivery employing the nanoformulation, is one such emerging delivery approach, overcoming both classical drug formulation and delivery-associated limitations. This latter approach offers increased bioavailability, greater patient acceptance, lesser metabolic degradation of drugs, circumvention of BBB, ample drug loading along with the controlled release of the drugs. In N2B delivery, the intranasal (IN) route carries therapeutics firstly into the nasal cavity followed by the brain through olfactory and trigeminal nerve connections linked with nasal mucosa. The N2B delivery approach is being explored for delivering other biologicals like neuropeptides and mitochondria. Meanwhile, this N2B delivery system is associated with critical challenges consisting of mucociliary clearance, degradation by enzymes, and drug translocations by efflux mechanisms. These challenges finally culminated in the development of suitable surfacemodified nano-carriers and Focused- Ultrasound-Assisted IN as FUS-IN technique which has expanded the horizons of N2B drug delivery. Hence, nanotechnology, in collaboration with advances in the IN route of drug administration, has a diversified approach for treating PD. The present review discusses the physiology and limitation of IN delivery along with current advances in nanocarrier and technical development assisting N2B drug delivery.
Collapse
Affiliation(s)
- Dharmendra K. Khatri
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Kumari Preeti
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Shivraj Tonape
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Sheoshree Bhattacharjee
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Monica Patel
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Pankaj K. Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| | - Dalapathi Gugulothu
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi-110017, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast-BT9 7BL, UK
| | - Shashi B. Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana State, India
| |
Collapse
|
14
|
Proskurina EY, Zaitsev AV. Regulation of Potassium and Chloride Concentrations in Nervous Tissue as a Method of Anticonvulsant Therapy. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract
Under some pathological conditions, such as pharmacoresistant
epilepsy, status epilepticus or certain forms of genetic abnormalities,
spiking activity of GABAergic interneurons may enhance excitation
processes in neuronal circuits and provoke the generation of ictal
discharges. As a result, anticonvulsants acting on the GABAergic
system may be ineffective or even increase seizure activity. This
paradoxical effect of the inhibitory system is due to ionic imbalances
in nervous tissue. This review addresses the mechanisms of ictal
discharge initiation in neuronal networks due to the imbalance of
chloride and potassium ions, as well as possible ways to regulate
ionic concentrations. Both the enhancement (or attenuation) of the
activity of certain neuronal ion transporters and ion pumps and
their additional expression via gene therapy can be effective in
suppressing seizure activity caused by ionic imbalances. The Na+–K+-pump,
NKCC1 and KCC2 cotransporters are important for maintaining proper
K+ and Cl– concentrations
in nervous tissue, having been repeatedly considered as pharmacological
targets for antiepileptic exposures. Further progress in this direction
is hampered by the lack of sufficiently selective pharmacological
tools and methods for providing effective drug delivery to the epileptic
focus. The use of the gene therapy techniques, such as overexpressing
of the KCC2 transporter in the epileptic focus, seems to be a more promising
approach. Another possible direction could be the use of optogenetic
tools, namely specially designed light-activated ion pumps or ion
channels. In this case, photon energy can be used to create the
required gradients of chloride and potassium ions, although these
methods also have significant limitations which complicate their
rapid introduction into medicine.
Collapse
|
15
|
Nikitin ES, Balaban PM, Zaitsev AV. Prospects for Gene Therapy of Epilepsy Using Calcium-Acivated Potassium Channel Vectors. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Shaimardanova AA, Chulpanova DS, Mullagulova AI, Afawi Z, Gamirova RG, Solovyeva VV, Rizvanov AA. Gene and Cell Therapy for Epilepsy: A Mini Review. Front Mol Neurosci 2022; 15:868531. [PMID: 35645733 PMCID: PMC9132249 DOI: 10.3389/fnmol.2022.868531] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic non-infectious disease of the brain, characterized primarily by recurrent unprovoked seizures, defined as an episode of disturbance of motor, sensory, autonomic, or mental functions resulting from excessive neuronal discharge. Despite the advances in the treatment achieved with the use of antiepileptic drugs and other non-pharmacological therapies, about 30% of patients suffer from uncontrolled seizures. This review summarizes the currently available methods of gene and cell therapy for epilepsy and discusses the development of these approaches. Currently, gene therapy for epilepsy is predominantly adeno-associated virus (AAV)-mediated delivery of genes encoding neuro-modulatory peptides, neurotrophic factors, enzymes, and potassium channels. Cell therapy for epilepsy is represented by the transplantation of several types of cells such as mesenchymal stem cells (MSCs), bone marrow mononuclear cells, neural stem cells, and MSC-derived exosomes. Another approach is encapsulated cell biodelivery, which is the transplantation of genetically modified cells placed in capsules and secreting various therapeutic agents. The use of gene and cell therapy approaches can significantly improve the condition of patient with epilepsy. Therefore, preclinical, and clinical studies have been actively conducted in recent years to prove the benefits and safety of these strategies.
Collapse
Affiliation(s)
| | - Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Aysilu I. Mullagulova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Zaid Afawi
- Center for Neuroscience, Ben Gurion University of the Negev, Be’er Sheva, Israel
| | - Rimma G. Gamirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- *Correspondence: Albert A. Rizvanov,
| |
Collapse
|
17
|
Parambi DGT, Alharbi KS, Kumar R, Harilal S, Batiha GES, Cruz-Martins N, Magdy O, Musa A, Panda DS, Mathew B. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:191-233. [PMID: 34655056 PMCID: PMC8518903 DOI: 10.1007/s12035-021-02555-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022]
Abstract
The etiology of many neurological diseases affecting the central nervous system (CNS) is unknown and still needs more effective and specific therapeutic approaches. Gene therapy has a promising future in treating neurodegenerative disorders by correcting the genetic defects or by therapeutic protein delivery and is now an attraction for neurologists to treat brain disorders, like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, spinocerebellar ataxia, epilepsy, Huntington's disease, stroke, and spinal cord injury. Gene therapy allows the transgene induction, with a unique expression in cells' substrate. This article mainly focuses on the delivering modes of genetic materials in the CNS, which includes viral and non-viral vectors and their application in gene therapy. Despite the many clinical trials conducted so far, data have shown disappointing outcomes. The efforts done to improve outcomes, efficacy, and safety in the identification of targets in various neurological disorders are also discussed here. Adapting gene therapy as a new therapeutic approach for treating neurological disorders seems to be promising, with early detection and delivery of therapy before the neuron is lost, helping a lot the development of new therapeutic options to translate to the clinic.
Collapse
Affiliation(s)
- Della Grace Thomas Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Khalid Saad Alharbi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Omnia Magdy
- Department of Clinical Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014 Kingdom of Saudi Arabia
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
| | - Arafa Musa
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371 Egypt
| | - Dibya Sundar Panda
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Al Jouf, Sakaka, 72341 Kingdom of Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
18
|
The Endocannabinoid System in Glial Cells and Their Profitable Interactions to Treat Epilepsy: Evidence from Animal Models. Int J Mol Sci 2021; 22:ijms222413231. [PMID: 34948035 PMCID: PMC8709154 DOI: 10.3390/ijms222413231] [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: 11/22/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is one of the most common neurological conditions. Yearly, five million people are diagnosed with epileptic-related disorders. The neuroprotective and therapeutic effect of (endo)cannabinoid compounds has been extensively investigated in several models of epilepsy. Therefore, the study of specific cell-type-dependent mechanisms underlying cannabinoid effects is crucial to understanding epileptic disorders. It is estimated that about 100 billion neurons and a roughly equal number of glial cells co-exist in the human brain. The glial population is in charge of neuronal viability, and therefore, their participation in brain pathophysiology is crucial. Furthermore, glial malfunctioning occurs in a wide range of neurological disorders. However, little is known about the impact of the endocannabinoid system (ECS) regulation over glial cells, even less in pathological conditions such as epilepsy. In this review, we aim to compile the existing knowledge on the role of the ECS in different cell types, with a particular emphasis on glial cells and their impact on epilepsy. Thus, we propose that glial cells could be a novel target for cannabinoid agents for treating the etiology of epilepsy and managing seizure-like disorders.
Collapse
|
19
|
Comeras LB, Hörmer N, Mohan Bethuraj P, Tasan RO. NPY Released From GABA Neurons of the Dentate Gyrus Specially Reduces Contextual Fear Without Affecting Cued or Trace Fear. Front Synaptic Neurosci 2021; 13:635726. [PMID: 34122036 PMCID: PMC8187774 DOI: 10.3389/fnsyn.2021.635726] [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: 11/30/2020] [Accepted: 04/20/2021] [Indexed: 11/15/2022] Open
Abstract
Disproportionate, maladapted, and generalized fear are essential hallmarks of posttraumatic stress disorder (PTSD), which develops upon severe trauma in a subset of exposed individuals. Among the brain areas that are processing fear memories, the hippocampal formation exerts a central role linking emotional-affective with cognitive aspects. In the hippocampus, neuronal excitability is constrained by multiple GABAergic interneurons with highly specialized functions and an extensive repertoire of co-released neuromodulators. Neuropeptide Y (NPY) is one of these co-transmitters that significantly affects hippocampal signaling, with ample evidence supporting its fundamental role in emotional, cognitive, and metabolic circuitries. Here we investigated the role of NPY in relation to GABA, both released from the same interneurons of the dorsal dentate gyrus (DG), in different aspects of fear conditioning. We demonstrated that activation of dentate GABA neurons specifically during fear recall reduced cue-related as well as trace-related freezing behavior, whereas inhibition of the same neurons had no significant effects. Interestingly, concomitant overexpression of NPY in these neurons did not further modify fear recall, neither under baseline conditions nor upon chemogenetic stimulation. However, potentially increased co-release of NPY substantially reduced contextual fear, promoted extinction learning, and long-term suppression of fear in a foreground context–conditioning paradigm. Importantly, NPY in the dorsal DG was not only expressed in somatostatin neurons, but also in parvalbumin-positive basket cells and axoaxonic cells, indicating intense feedback and feedforward modulation of hippocampal signaling and precise curtailing of neuronal engrams. Thus, these findings suggest that co-release of NPY from specific interneuron populations of the dorsal DG modifies dedicated aspects of hippocampal processing by sharpening the activation of neural engrams and the consecutive fear response. Since inappropriate and generalized fear is the major impediment in the treatment of PTSD patients, the dentate NPY system may be a suitable access point to ameliorate PTSD symptoms and improve the inherent disease course.
Collapse
Affiliation(s)
- Lucas B Comeras
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Noa Hörmer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
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
|
21
|
Christian-Hinman CA. Is On-Demand Dynorphin Destined to Be in Demand to Decrease Seizures? Epilepsy Curr 2021; 21:48-50. [PMID: 34025273 PMCID: PMC7863305 DOI: 10.1177/1535759720951791] [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] [Indexed: 11/16/2022] Open
Abstract
Dynorphin-Based “Release on Demand” Gene Therapy for Drug-Resistant Temporal Lobe Epilepsy Agostinho AS, Mietzsch M, Zangrandi L, et al. EMBO Mol Med. 2019;11(10):e9963. doi:10.15252/emmm.201809963 Focal epilepsy represents one of the most common chronic central nervous system 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 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. Adeno-associated virus 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
|
22
|
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
|
23
|
Serum Neuropeptide Y Level is Associated with Post-Ischemic Stroke Epilepsy. J Stroke Cerebrovasc Dis 2020; 30:105475. [PMID: 33242785 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105475] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/01/2020] [Accepted: 11/10/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Post-ischemic stroke epilepsy (PISE) is one of the common complications of stroke. MATERIALS AND METHODS Methods To determine the risk factors of PISE, in this study, 78 patients with PISE and 86 patients without PISE were recruited. Clinical data and serum neuropeptide Y (NPY) levels were collected and the relative factors including clinical data and serum were analyzed. RESULTS Logistic regression showed that low serum NPY was significantly associated with PISE. Every 5 ng/ml increment of serum NPY was associated with 62% risk decrease in patients with PISE. The area under curve of serum NPY was 0.910 with a sensitivity of 84.62% and a specificity of 86.05%. The cut-off value of serum NPY was 90 ng/ml. According to cut-off value of serum NPY, the percentage of patients with PISE decreased from 84.6% in low serum NPY group to 14.0% in high serum NPY group. Furthermore, patients were divided into different tertiles according to serum NPY. The percentage of patients with PISE reduced from 90.0% in the lowest tertile (NPY < 85 ng/ml) to 3.5% in the highest tertile (NPY ≥ 105 ng/ml). Compared with patients with normal video-electroencephalogram (VEEG), serum NPY levels significantly decreased in patients with abnormal VEEG; however, serum NPY levels were not associaated with epileptic seizure subtypes. CONCLUSIONS Serum NPY was an independent risk factor for PISE. Targeting serum NPY may be used to the prevention and treatment of PISE.
Collapse
|
24
|
Impact of predictive, preventive and precision medicine strategies in epilepsy. Nat Rev Neurol 2020; 16:674-688. [PMID: 33077944 DOI: 10.1038/s41582-020-0409-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/15/2022]
Abstract
Over the last decade, advances in genetics, neuroimaging and EEG have enabled the aetiology of epilepsy to be identified earlier in the disease course than ever before. At the same time, progress in the study of experimental models of epilepsy has provided a better understanding of the mechanisms underlying the condition and has enabled the identification of therapies that target specific aetiologies. We are now witnessing the impact of these advances in our daily clinical practice. Thus, now is the time for a paradigm shift in epilepsy treatment from a reactive attitude, treating patients after the onset of epilepsy and the initiation of seizures, to a proactive attitude that is more broadly integrated into a 'P4 medicine' approach. This P4 approach, which is personalized, predictive, preventive and participatory, puts patients at the centre of their own care and, ultimately, aims to prevent the onset of epilepsy. This aim will be achieved by adapting epilepsy treatments not only to a given syndrome but also to a given patient and moving from the usual anti-seizure treatments to personalized treatments designed to target specific aetiologies. In this Review, we present the current state of this ongoing revolution, emphasizing the impact on clinical practice.
Collapse
|
25
|
Iqubal A, Iqubal MK, Khan A, Ali J, Baboota S, Haque SE. Gene Therapy, A Novel Therapeutic Tool for Neurological Disorders: Current Progress, Challenges and Future Prospective. Curr Gene Ther 2020; 20:184-194. [DOI: 10.2174/1566523220999200716111502] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/02/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
:
Neurological disorders are one of the major threat for health care system as they put enormous
socioeconomic burden. All aged populations are susceptible to one or other neurological problems
with symptoms of neuroinflammation, neurodegeneration and cognitive dysfunction. At present,
available pharmacotherapeutics are insufficient to treat these diseased conditions and in most cases,
they provide only palliative effect. It was also found that the molecular etiology of neurological disorders
is directly linked with the alteration in genetic makeup, which can be inherited or triggered by the
injury, environmental toxins and by some existing disease. Therefore, to take care of this situation,
gene therapy has emerged as an advanced modality that claims to permanently cure the disease by deletion,
silencing or edition of faulty genes and by insertion of healthier genes. In this modality, vectors
(viral and non-viral) are used to deliver targeted gene into a specific region of the brain via various
routes. At present, gene therapy has shown positive outcomes in complex neurological disorders, such
as Parkinson's disease, Alzheimer's disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral
sclerosis and in lysosomal storage disease. However, there are some limitations such as immunogenic
reactions non-specificity of viral vectors and a lack of effective biomarkers to understand the efficacy
of therapy. Considerable progress has been made to improve vector design, gene selection and
targeted delivery. This review article deals with the current status of gene therapy in neurological disorders
along with its clinical relevance, challenges and future prospective.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Aamir Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| |
Collapse
|
26
|
Colasante G, Qiu Y, Massimino L, Di Berardino C, Cornford JH, Snowball A, Weston M, Jones SP, Giannelli S, Lieb A, Schorge S, Kullmann DM, Broccoli V, Lignani G. In vivo CRISPRa decreases seizures and rescues cognitive deficits in a rodent model of epilepsy. Brain 2020; 143:891-905. [PMID: 32129831 PMCID: PMC7089667 DOI: 10.1093/brain/awaa045] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/31/2019] [Accepted: 01/14/2020] [Indexed: 01/20/2023] Open
Abstract
Epilepsy is a major health burden, calling for new mechanistic insights and therapies. CRISPR-mediated gene editing shows promise to cure genetic pathologies, although hitherto it has mostly been applied ex vivo. Its translational potential for treating non-genetic pathologies is still unexplored. Furthermore, neurological diseases represent an important challenge for the application of CRISPR, because of the need in many cases to manipulate gene function of neurons in situ. A variant of CRISPR, CRISPRa, offers the possibility to modulate the expression of endogenous genes by directly targeting their promoters. We asked if this strategy can effectively treat acquired focal epilepsy, focusing on ion channels because their manipulation is known be effective in changing network hyperactivity and hypersynchronziation. We applied a doxycycline-inducible CRISPRa technology to increase the expression of the potassium channel gene Kcna1 (encoding Kv1.1) in mouse hippocampal excitatory neurons. CRISPRa-mediated Kv1.1 upregulation led to a substantial decrease in neuronal excitability. Continuous video-EEG telemetry showed that AAV9-mediated delivery of CRISPRa, upon doxycycline administration, decreased spontaneous generalized tonic-clonic seizures in a model of temporal lobe epilepsy, and rescued cognitive impairment and transcriptomic alterations associated with chronic epilepsy. The focal treatment minimizes concerns about off-target effects in other organs and brain areas. This study provides the proof-of-principle for a translational CRISPR-based approach to treat neurological diseases characterized by abnormal circuit excitability.
Collapse
Affiliation(s)
- Gaia Colasante
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Luca Massimino
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | | | - Jonathan H Cornford
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Mikail Weston
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Steffan P Jones
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Serena Giannelli
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Stephanie Schorge
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Vania Broccoli
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.,CNR Institute of Neuroscience, Via Vanvitelli 32, 20129, Milan, Italy
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
27
|
Lignani G, Baldelli P, Marra V. Homeostatic Plasticity in Epilepsy. Front Cell Neurosci 2020; 14:197. [PMID: 32676011 PMCID: PMC7333442 DOI: 10.3389/fncel.2020.00197] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/05/2020] [Indexed: 11/26/2022] Open
Abstract
In the healthy brain, neuronal excitability and synaptic strength are homeostatically regulated to keep neuronal network activity within physiological boundaries. Epilepsy is characterized by episodes of highly synchronized firing across in widespread neuronal populations, due to a failure in regulation of network activity. Here we consider epilepsy as a failure of homeostatic plasticity or as a maladaptive response to perturbations in the activity. How homeostatic compensation is involved in epileptogenic processes or in the chronic phase of epilepsy, is still debated. Although several theories have been proposed, there is relatively little experimental evidence to evaluate them. In this perspective, we will discuss recent results that shed light on the potential role of homeostatic plasticity in epilepsy. First, we will present some recent insights on how homeostatic compensations are probably active before and during epileptogenesis and how their actions are temporally regulated and closely dependent on the progression of pathology. Then, we will consider the dual role of transcriptional regulation during epileptogenesis, and finally, we will underline the importance of homeostatic plasticity in the context of therapeutic interventions for epilepsy. While classic pharmacological interventions may be counteracted by the epileptic brain to maintain its potentially dysfunctional set point, novel therapeutic approaches may provide the neuronal network with the tools necessary to restore its physiological balance.
Collapse
Affiliation(s)
- Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Marra
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
28
|
Functional Access to Neuron Subclasses in Rodent and Primate Forebrain. Cell Rep 2020; 26:2818-2832.e8. [PMID: 30840900 PMCID: PMC6509701 DOI: 10.1016/j.celrep.2019.02.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 01/08/2019] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
Viral vectors enable foreign proteins to be expressed in brains of non-genetic species, including non-human primates. However, viruses targeting specific neuron classes have proved elusive. Here we describe viral promoters and strategies for accessing GABAergic interneurons and their molecularly defined subsets in the rodent and primate. Using a set intersection approach, which relies on two co-active promoters, we can restrict heterologous protein expression to cortical and hippocampal somatostatin-positive and parvalbumin-positive interneurons. With an orthogonal set difference method, we can enrich for subclasses of neuropeptide-Y-positive GABAergic interneurons by effectively subtracting the expression pattern of one promoter from that of another. These methods harness the complexity of gene expression patterns in the brain and significantly expand the number of genetically tractable neuron classes across mammals.
Collapse
|
29
|
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
|
30
|
ROCK/PKA Inhibition Rescues Hippocampal Hyperexcitability and GABAergic Neuron Alterations in a Oligophrenin-1 Knock-Out Mouse Model of X-Linked Intellectual Disability. J Neurosci 2020; 40:2776-2788. [PMID: 32098904 DOI: 10.1523/jneurosci.0462-19.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 01/19/2023] Open
Abstract
Oligophrenin-1 (Ophn1) encodes a Rho GTPase activating protein whose mutations cause X-linked intellectual disability (XLID) in humans. Loss of function of Ophn1 leads to impairments in the maturation and function of excitatory and inhibitory synapses, causing deficits in synaptic structure, function and plasticity. Epilepsy is a frequent comorbidity in patients with Ophn1-dependent XLID, but the cellular bases of hyperexcitability are poorly understood. Here we report that male mice knock-out (KO) for Ophn1 display hippocampal epileptiform alterations, which are associated with changes in parvalbumin-, somatostatin- and neuropeptide Y-positive interneurons. Because loss of function of Ophn1 is related to enhanced activity of Rho-associated protein kinase (ROCK) and protein kinase A (PKA), we attempted to rescue Ophn1-dependent pathological phenotypes by treatment with the ROCK/PKA inhibitor fasudil. While acute administration of fasudil had no impact on seizure activity, seven weeks of treatment in adulthood were able to correct electrographic, neuroanatomical and synaptic alterations of Ophn1 deficient mice. These data demonstrate that hyperexcitability and the associated changes in GABAergic markers can be rescued at the adult stage in Ophn1-dependent XLID through ROCK/PKA inhibition.SIGNIFICANCE STATEMENT In this study we demonstrate enhanced seizure propensity and impairments in hippocampal GABAergic circuitry in Ophn1 mouse model of X-linked intellectual disability (XLID). Importantly, the enhanced susceptibility to seizures, accompanied by an alteration of GABAergic markers were rescued by Rho-associated protein kinase (ROCK)/protein kinase A (PKA) inhibitor fasudil, a drug already tested on humans. Because seizures can significantly impact the quality of life of XLID patients, the present data suggest a potential therapeutic pathway to correct alterations in GABAergic networks and dampen pathological hyperexcitability in adults with XLID.
Collapse
|
31
|
Binder DK, Boison D, Eid T, Frankel WN, Mingorance A, Smith BN, Dacks PA, Whittemore V, Poduri A. Epilepsy Benchmarks Area II: Prevent Epilepsy and Its Progression. Epilepsy Curr 2020; 20:14S-22S. [PMID: 31937124 PMCID: PMC7031802 DOI: 10.1177/1535759719895274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Area II of the 2014 Epilepsy Research Benchmarks aims to establish goals for preventing the development and progression of epilepsy. In this review, we will highlight key advances in Area II since the last summary of research progress and opportunities was published in 2016. We also highlight areas of investigation that began to develop before 2016 and in which additional progress has been made more recently.
Collapse
Affiliation(s)
- Devin K Binder
- Division of Biomedical Sciences, School of Medicine, Center for Glial-Neuronal Interactions, University of California, Riverside, CA, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson and New Jersey Medical Schools, Rutgers University, Piscataway, NJ, USA
| | - Tore Eid
- Department of Laboratory Medicine, Neurosurgery and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Wayne N Frankel
- Department of Genetics & Development, Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Bret N Smith
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Vicky Whittemore
- Division of Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Annapurna Poduri
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Haggerty DL, Grecco GG, Reeves KC, Atwood B. Adeno-Associated Viral Vectors in Neuroscience Research. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:69-82. [PMID: 31890742 PMCID: PMC6931098 DOI: 10.1016/j.omtm.2019.11.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adeno-associated viral vectors (AAVs) are increasingly useful preclinical tools in neuroscience research studies for interrogating cellular and neurocircuit functions and mapping brain connectivity. Clinically, AAVs are showing increasing promise as viable candidates for treating multiple neurological diseases. Here, we briefly review the utility of AAVs in mapping neurocircuits, manipulating neuronal function and gene expression, and activity labeling in preclinical research studies as well as AAV-based gene therapies for diseases of the nervous system. This review highlights the vast potential that AAVs have for transformative research and therapeutics in the neurosciences.
Collapse
Affiliation(s)
- David L. Haggerty
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory G. Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kaitlin C. Reeves
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brady Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
- Corresponding author: Brady Atwood, PhD, Department of Pharmacology & Toxicology, Indiana University School of Medicine, 320 West 15th Street, NB-400C, Indianapolis, IN 46202, USA.
| |
Collapse
|
35
|
Kundap UP, Choo BKM, Kumari Y, Ahmed N, Othman IB, Shaikh MF. Embelin Protects Against Acute Pentylenetetrazole-Induced Seizures and Positively Modulates Cognitive Function in Adult Zebrafish. Front Pharmacol 2019; 10:1249. [PMID: 31708779 PMCID: PMC6823247 DOI: 10.3389/fphar.2019.01249] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/27/2019] [Indexed: 01/29/2023] Open
Abstract
Purpose of the research: Epilepsy is a continuous process of neurodegeneration categorized by an enduring tendency to generate uncontrolled electrical firing known as seizures causing involuntary movement all over the body. Cognitive impairment and behavioral disturbances are among the more alarming co-morbidities of epilepsy. Anti-epileptic drugs (AEDs) were found to be successful in controlling epilepsy but are reported to worsen cognitive status in patients. Embelin (EMB) is a benzoquinone derived from the plant Embelia ribes and is reported to have central nervous system (CNS) activity. This study aims to evaluate the effectiveness of EMB against pentylenetetrazole (PTZ) induced acute seizures and its associated cognitive dysfunction. This was done via docking studies as well as evaluating neurotransmitter and gene expression in the zebrafish brain. The principal results: Behavioral observations showed that EMB reduced epileptic seizures and the T-maze study revealed that EMB improved the cognitive function of the fish. The docking study of EMB showed a higher affinity toward gamma-aminobutyric acid (GABAA) receptor as compared to the standard diazepam, raising the possibility of EMB working via the alpha subunit of the GABA receptor. EMB was found to modulate several genes, neurotransmitters, and also neuronal growth, all of which play an important role in improving cognitive status after epileptic seizures. Healthy zebrafish treated with EMB alone were found to have no behavioral and biochemical interference or side effects. The immunohistochemistry data suggested that EMB also promotes neuronal protection and neuronal migration in zebrafish brains. Major Conclusions: It was perceived that EMB suppresses seizure-like behavior via GABAA receptor pathway and has a positive impact on cognitive functions. The observed effect was supported by docking study, T-maze behavior, neurotransmitter and gene expression levels, and immunohistology study. The apparatus such as the T-maze and seizure scoring behavior tank were found to be a straightforward technique to score seizure and test learning ability after acute epileptic seizures. These research findings suggest that EMB could be a promising molecule for epilepsy induced learning and memory dysfunction.
Collapse
Affiliation(s)
- Uday Praful Kundap
- Department of Neurosciences, University of Montreal Hospital Centre (CRCHUM), Montreal, Canada.,Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Kuala Lumpur, Malaysia
| | - Brandon Kar Meng Choo
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Kuala Lumpur, Malaysia
| | - Yatinesh Kumari
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Kuala Lumpur, Malaysia
| | - Nafees Ahmed
- School of Pharmacy, Monash University Malaysia, Kuala Lumpur, Malaysia
| | - Iekhsan Bin Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Kuala Lumpur, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
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
|
37
|
Walker MC, Kullmann DM. Optogenetic and chemogenetic therapies for epilepsy. Neuropharmacology 2019; 168:107751. [PMID: 31494141 DOI: 10.1016/j.neuropharm.2019.107751] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022]
Abstract
Drug-resistant epilepsy remains a significant health-care burden. The most effective treatment is surgery, but this is suitable for very few patients because of the unacceptable consequences of removing brain tissue. In contrast, gene therapy can regulate neuronal excitability in the epileptic focus whilst preserving function. Optogenetics and chemogenetics have the advantage that they are titratable therapies. Optogenetics uses light to control the excitability of specific neuronal populations. Optogenetics can be used in a closed-loop paradigm in which the light source is activated only when seizures are detected. However, expression of foreign proteins raises concerns about immunogenicity. Chemogenetics relies on the modification of an endogenous receptor or the production of a modified chimeric receptor that responds to an exogenous ligand. The main chemogenetic approach applied to epilepsy is to use designer receptors exclusively activated by designer drugs (DREADDs), which have been mainly modified muscarinic receptors or kappa-opioid receptors. Genetically modified human muscarinic receptor DREADDs are activated not by acetylcholine but by specific drugs such as clozapine-n-oxide or olanzepine. The dose of the drugs can be titrated in order to suppress seizures without adverse effects. Lastly, there is a chemogenetic approach that is activated by an endogenous ligand, glutamate. This takes advantage of invertebrate glutamate receptors that are chloride permeable. These bind glutamate released during seizure activity, and the resultant chloride current inhibits neuronal activity. The exogenous ligand, ivermectin, can also be given to reduce neuronal activity either chronically or as a rescue medication. The translation of this technology is hampered by the expression of a foreign protein. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| |
Collapse
|
38
|
Yang Q, Huang Z, Luo Y, Zheng F, Hu Y, Liu H, Zhu S, He M, Xu D, Li Y, Yang M, Yang Y, Wei X, Gao X, Wang W, Ma J, Ma Y, Wang X, Wang Q. Inhibition of Nwd1 activity attenuates neuronal hyperexcitability and GluN2B phosphorylation in the hippocampus. EBioMedicine 2019; 47:470-483. [PMID: 31474551 PMCID: PMC6796588 DOI: 10.1016/j.ebiom.2019.08.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/04/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND NACHT and WD repeat domain-containing protein 1 (Nwd1) is a member of the innate immune protein subfamily. Nwd1 contributes to the androgen receptor signaling pathway and is involved in axonal growth. However, the mechanisms that underlie pathophysiological dysfunction in seizures remain unclear. METHODS Biochemical methods were used to assess Nwd1 expression and localization in a mouse model of kainic acid (KA)-induced acute seizures and temporal lobe epilepsy (TLE) patients. Electrophysiological recordings were used to measure the role of Nwd1 in regulating synaptic transmission and neuronal hyperexcitability in a model of magnesium-free-induced seizure in vitro. Behavioral experiments were performed, and seizure-induced pathological changes were evaluated in a KA-induced seizure model in vivo. GluN2B expression was measured and its correlation with Tyr1472-GluN2B phosphorylation was analyzed in primary hippocampal neurons. FINDINGS We demonstrated high protein levels of Nwd1 in brain tissues obtained from mice with acute seizures and TLE patients. Silencing Nwd1 in mice using an adeno-associated virus (AAV) profoundly suppressed neuronal hyperexcitability and the occurrence of acute seizures, which may have been caused by reducing GluN2B-containing NMDA receptor-dependent glutamatergic synaptic transmission. Moreover, the decreased activation of Nwd1 reduced GluN2B expression and the phosphorylation of the GluN2B subunit at Tyr1472. INTERPRETATION Here, we report a previously unrecognized but important role of Nwd1 in seizure models in vitro and in vivo, i.e., modulating the phosphorylation of the GluN2B subunit at Tyr1472 and regulating neuronal hyperexcitability. Meanwhile, our findings may provide a therapeutic strategy for the treatment of epilepsy or other hyperexcitability-related neurological disorders. FUND: The funders have not participated in the study design, data collection, data analysis, interpretation, or writing of the report.
Collapse
Affiliation(s)
- Qin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China; Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Yangfu Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Fangshuo Zheng
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yida Hu
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Hui Liu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Miaoqing He
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Demei Xu
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yun Li
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Min Yang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Yi Yang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Wei Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Junhong Ma
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Yuanlin Ma
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China
| | - Xuefeng Wang
- Department of Neurology, The first Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, PR China; Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100101, PR China.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong Province 510282, PR China.
| |
Collapse
|
39
|
Carron SF, Sun M, Shultz SR, Rajan R. Inhibitory neuronal changes following a mixed diffuse‐focal model of traumatic brain injury. J Comp Neurol 2019; 528:175-198. [DOI: 10.1002/cne.24746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Simone F. Carron
- Neuroscience Discovery Program, Biomedicine Discovery Institute, Department of Physiology Monash University Melbourne Victoria Australia
| | - Mujun Sun
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Sandy R. Shultz
- Department of Medicine and Neuroscience Monash University Melbourne Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Ramesh Rajan
- Neuroscience Discovery Program, Biomedicine Discovery Institute, Department of Physiology Monash University Melbourne Victoria Australia
| |
Collapse
|
40
|
Pauletti A, Terrone G, Shekh-Ahmad T, Salamone A, Ravizza T, Rizzi M, Pastore A, Pascente R, Liang LP, Villa BR, Balosso S, Abramov AY, van Vliet EA, Del Giudice E, Aronica E, Patel M, Walker MC, Vezzani A. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain 2019; 142:e39. [PMID: 31145451 PMCID: PMC6598637 DOI: 10.1093/brain/awz130] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/17/2017] [Accepted: 03/26/2017] [Indexed: 01/07/2023] Open
Abstract
Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.
Collapse
Affiliation(s)
- Alberto Pauletti
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Gaetano Terrone
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Tawfeeq Shekh-Ahmad
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Alessia Salamone
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Teresa Ravizza
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Massimo Rizzi
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Anna Pastore
- 3 Metabolomics and Proteomics Unit, ‘Bambino Gesù’ Children’s Hospital,
IRCCS, Rome, Italy
| | - Rosaria Pascente
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Li-Ping Liang
- 4 Department of Pharmaceutical Sciences, University of Colorado Denver,
Aurora, Colorado, USA
| | - Bianca R Villa
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Silvia Balosso
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Andrey Y Abramov
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Erwin A van Vliet
- 5 Department of (Neuro)Pathology, Academic Medical Center, University of
Amsterdam, The Netherlands
| | - Ennio Del Giudice
- 6 Department of Translational Medical Sciences, Section of Pediatrics,
Federico II University, Naples, Italy
| | - Eleonora Aronica
- 5 Department of (Neuro)Pathology, Academic Medical Center, University of
Amsterdam, The Netherlands
- 7 Stichting Epilepsie Instellingen Nederland, Amsterdam, The
Netherlands
| | - Manisha Patel
- 4 Department of Pharmaceutical Sciences, University of Colorado Denver,
Aurora, Colorado, USA
| | - Matthew C Walker
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Annamaria Vezzani
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
- Correpondence to: Annamaria Vezzani, PhD Department of Neuroscience
IRCCS-Istituto di Ricerche Farmacologiche Mario Negri Via G. La Masa 19, 20156 Milano,
Italy E-mail:
| |
Collapse
|
41
|
Mehdizadeh A, Barzegar M, Negargar S, Yahyavi A, Raeisi S. The current and emerging therapeutic approaches in drug-resistant epilepsy management. Acta Neurol Belg 2019; 119:155-162. [PMID: 30868468 DOI: 10.1007/s13760-019-01120-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/06/2019] [Indexed: 01/08/2023]
Abstract
Epilepsy is a neurologic disorder consisting of recurrent spontaneous seizures. Antiepileptic drugs administration is the most commonly used therapeutic strategy in the management of epilepsy. However, 20-30% of epilepsy patients have seizure episodes that are not controlled by these medicines (drug-resistant epilepsy). The management of drug-resistant epilepsy, especially in the children, is challenging and can cause economic and social problems, and lower the patients' quality of life, cognition, and mood. Several therapeutic approaches for drug-resistant epilepsy are available including surgical methods, neurostimulation treatments, and diet therapies which lead to diminishing the epileptic seizures. An increasing number of novel and potential therapeutic approaches such as gene therapy, gene editing, cell therapy, exosome therapy, and molecular network targeting have also been explored. The present study is aimed to review these current and emerging therapeutic approaches for drug-resistant epilepsy.
Collapse
Affiliation(s)
- Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Barzegar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sohrab Negargar
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azita Yahyavi
- Department of Anesthesia, Shohada Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Raeisi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
42
|
Lieb A, Weston M, Kullmann DM. Designer receptor technology for the treatment of epilepsy. EBioMedicine 2019; 43:641-649. [PMID: 31078519 PMCID: PMC6558262 DOI: 10.1016/j.ebiom.2019.04.059] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Epilepsy remains refractory to medical treatment in ~30% of patients despite decades of new drug development. Neurosurgery to remove or disconnect the seizure focus is often curative but frequently contraindicated by risks of irreversible impairment to brain function. Novel therapies are therefore required that better balance seizure suppression against the risks of side effects. Among experimental gene therapies, chemogenetics has the major advantage that the action on the epileptogenic zone can be modulated on demand. Two broad approaches are to use a designer G-protein-coupled receptor or a modified ligand gated ion channel, targeted to specific neurons in the epileptogenic zone using viral vectors and cell-type selective promoters. The receptor can be activated on demand by either an exogenous compound or by pathological levels of extracellular glutamate that occur in epileptogenic tissue. We review the principal designer receptor technologies and their modes of action. We compare the drawbacks and benefits of each designer receptor with particular focus on the drug activators and the potential for clinical translation in epilepsy.
Collapse
Affiliation(s)
- Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| | - Mikail Weston
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK.
| |
Collapse
|
43
|
Snowball A, Chabrol E, Wykes RC, Shekh-Ahmad T, Cornford JH, Lieb A, Hughes MP, Massaro G, Rahim AA, Hashemi KS, Kullmann DM, Walker MC, Schorge S. Epilepsy Gene Therapy Using an Engineered Potassium Channel. J Neurosci 2019; 39:3159-3169. [PMID: 30755487 PMCID: PMC6468110 DOI: 10.1523/jneurosci.1143-18.2019] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 12/24/2018] [Accepted: 01/21/2019] [Indexed: 12/21/2022] Open
Abstract
Refractory focal epilepsy is a devastating disease for which there is frequently no effective treatment. Gene therapy represents a promising alternative, but treating epilepsy in this way involves irreversible changes to brain tissue, so vector design must be carefully optimized to guarantee safety without compromising efficacy. We set out to develop an epilepsy gene therapy vector optimized for clinical translation. The gene encoding the voltage-gated potassium channel Kv1.1, KCNA1, was codon optimized for human expression and mutated to accelerate the recovery of the channels from inactivation. For improved safety, this engineered potassium channel (EKC) gene was packaged into a nonintegrating lentiviral vector under the control of a cell type-specific CAMK2A promoter. In a blinded, randomized, placebo-controlled preclinical trial, the EKC lentivector robustly reduced seizure frequency in a male rat model of focal neocortical epilepsy characterized by discrete spontaneous seizures. When packaged into an adeno-associated viral vector (AAV2/9), the EKC gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. This demonstration of efficacy in a clinically relevant setting, combined with the improved safety conferred by cell type-specific expression and integration-deficient delivery, identify EKC gene therapy as being ready for clinical translation in the treatment of refractory focal epilepsy.SIGNIFICANCE STATEMENT Pharmacoresistant epilepsy affects up to 0.3% of the population. Although epilepsy surgery can be effective, it is limited by risks to normal brain function. We have developed a gene therapy that builds on a mechanistic understanding of altered neuronal and circuit excitability in cortical epilepsy. The potassium channel gene KCNA1 was mutated to bypass post-transcriptional editing and was packaged in a nonintegrating lentivector to reduce the risk of insertional mutagenesis. A randomized, blinded preclinical study demonstrated therapeutic effectiveness in a rodent model of focal neocortical epilepsy. Adeno-associated viral delivery of the channel to both hippocampi was also effective in a model of temporal lobe epilepsy. These results support clinical translation to address a major unmet need.
Collapse
Affiliation(s)
- Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Elodie Chabrol
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Robert C Wykes
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Tawfeeq Shekh-Ahmad
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Jonathan H Cornford
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Michael P Hughes
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Kevan S Hashemi
- Open Source Instruments Inc., Watertown, Massachusetts 02472
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom,
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom,
| | - Stephanie Schorge
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| |
Collapse
|
44
|
Weston M, Kaserer T, Wu A, Mouravlev A, Carpenter JC, Snowball A, Knauss S, von Schimmelmann M, During MJ, Lignani G, Schorge S, Young D, Kullmann DM, Lieb A. Olanzapine: A potent agonist at the hM4D(Gi) DREADD amenable to clinical translation of chemogenetics. SCIENCE ADVANCES 2019; 5:eaaw1567. [PMID: 31001591 PMCID: PMC6469940 DOI: 10.1126/sciadv.aaw1567] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) derived from muscarinic receptors not only are a powerful tool to test causality in basic neuroscience but also are potentially amenable to clinical translation. A major obstacle, however, is that the widely used agonist clozapine N-oxide undergoes conversion to clozapine, which penetrates the blood-brain barrier but has an unfavorable side effect profile. Perlapine has been reported to activate DREADDs at nanomolar concentrations but is not approved for use in humans by the Food and Drug Administration or the European Medicines Agency, limiting its translational potential. Here, we report that the atypical antipsychotic drug olanzapine, widely available in various formulations, is a potent agonist of the human M4 muscarinic receptor-based DREADD, facilitating clinical translation of chemogenetics to treat central nervous system diseases.
Collapse
Affiliation(s)
- Mikail Weston
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Teresa Kaserer
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Angela Wu
- Department of Pharmacology & Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Alexandre Mouravlev
- Department of Pharmacology & Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Jenna C. Carpenter
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Samuel Knauss
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | | | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Stephanie Schorge
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Deborah Young
- Department of Pharmacology & Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Dimitri M. Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
45
|
Shende P, Desai D. Physiological and Therapeutic Roles of Neuropeptide Y on Biological Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1237:37-47. [PMID: 31468359 DOI: 10.1007/5584_2019_427] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuropeptide Y (NPY), an amino acid, used for various physiological processes for management and treatment of various ailments related to central nervous system, cardiovascular system, respiratory system, gastro-intestinal system and endocrinal system. In nasal mucosa, high concentrations of NPY are stored with noradrenaline in sympathetic nerve fibers. NPY Y1 receptor mediates nitric oxide levels and reduction in blood flow in nasal mucosa of the human. NPY plays a role in dietary consumption via various factors like signaling the CNS for a prerequisite of energy in hypothalamus by mediating appetite and shows orexigenic effect. NPY emerges as a natural ligand of G-protein coupled receptors which activates the Y-receptors (Y1-Y6). But applications of NPY are limited due to shows the cost inefficiency and stability issues in the formulation design and development. In this review, authors present the findings on various therapeutic applications of NPY on different organ systems. Moreover, its role in food intake, sexual behavior, blood pressure, etc. by inhibiting calcium and activating potassium channels. The combination therapies of drugs with neuropeptide Y and its receptors will show new targets for treating diseases. Further evaluation and detection of NPY needs to be investigated for animal models of various diseases like retinal degeneration and immune mechanisms.
Collapse
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, India.
| | - Drashti Desai
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, India
| |
Collapse
|
46
|
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
|
47
|
ZDHHC8 critically regulates seizure susceptibility in epilepsy. Cell Death Dis 2018; 9:795. [PMID: 30038264 PMCID: PMC6056564 DOI: 10.1038/s41419-018-0842-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/07/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Epilepsy is one of the most prevalent and drug-refractory neurological disorders. Zinc finger DHHC-type containing 8 (ZDHHC8) is a putative palmitoyltransferase that is highly expressed in the brain. However, the impact of ZDHHC8 on seizures remains unclear. We aimed to explore the association of ZDHHC8 with epilepsy and investigate its in epileptogenesis in in vivo and in vitro models through behavioral, electrophysiological, and pathological studies. We used kainic acid- and pilocarpine-induced C57BL/6 mice and magnesium-free-induced pyramidal neurons as experimental epileptic models in this study. We first found increased ZDHHC8 expression in the brains of temporal lobe epilepsy (TLE) patients, similar to that observed in chronic epileptic mice, strongly suggesting that ZDHHC8 is correlated with human epilepsy. In the in vitro seizure models, knocking down ZDHHC8 using recombinant adeno-associated virus (rAAV) delayed seizure precipitation and decreased chronic spontaneous recurrent seizures (SRSs) and epileptiform-like discharges, while ZDHHC8 overexpression had the opposite effect. ZDHHC8 levels were consistent with seizure susceptibility in induced mice with SRSs. In an in vitro magnesium-free model, neuronal hyperexcitability and hypersynchrony were reduced in ZDHHC8-knockdown neurons but were increased in ZDHHC8-overexpressing neurons. To further explore the potential mechanisms, we observed that ZDHHC8 had a significant modulatory effect on 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid (AMPA) receptor-related excitatory, but not inhibitory, glutamatergic synaptic neurotransmission, further affecting the inward rectification of AMPA currents in acute hippocampal slices in whole-cell recordings. ZDHHC8 facilitated GluA1 trafficking to the neuronal surface in the hippocampus, as shown by immunoprecipitation and Western blotting. These results suggest that ZDHHC8 may promote the generation and propagation of seizures in humans and that knocking down ZDHHC8 might produce anti-epileptogenic effects in drug-resistant epilepsy. Our study provides evidence that may facilitate the development of an alternative approach for the treatment of epilepsy by modulating AMPA/GluA1-mediated neurotransmission.
Collapse
|
48
|
Gene therapy mediated seizure suppression in Genetic Generalised Epilepsy: Neuropeptide Y overexpression in a rat model. Neurobiol Dis 2018; 113:23-32. [DOI: 10.1016/j.nbd.2018.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/07/2018] [Accepted: 01/22/2018] [Indexed: 02/01/2023] Open
|
49
|
Patrício MI, Barnard AR, Green AL, During MJ, Sen A, MacLaren RE. A clinical-grade gene therapy vector for pharmacoresistant epilepsy successfully overexpresses NPY in a human neuronal cell line. Seizure 2018; 55:25-29. [PMID: 29367144 DOI: 10.1016/j.seizure.2017.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 11/16/2017] [Accepted: 12/14/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Epilepsy is a common neurological condition characterised by recurrent unprovoked seizures and often treatable with appropriate medication. However, almost 30% of cases are pharmacoresistant and while a proportion of these may be amenable to resective surgery, a gene therapy approach could be an attractive alternative option. Neuropeptide Y (NPY) has anticonvulsant and anti-epileptogenic properties in animal models of temporal lobe epilepsy when delivered by an adeno-associated viral (AAV) vector. Here we sought to demonstrate successful secretion of NPY from AAV-transduced human neuronal cells, which would be essential in planning any clinical trial. METHODS A human neuroblastoma cell line (SH-SY5Y) was used to assess in vitro whether an AAV vector manufactured to clinical-grade protocols would be effective at transducing these cells to express NPY. Optimal transduction efficiency was first achieved with retinoic acid and tetradecanoylphorpol-13-acetate (TPA) treatment, prior to expose to AAV1-green fluorescent protein (GFP) reporter vector, AAV1-NPY therapeutic vector or sham treated with no vector. Levels of NPY in cell supernatants were determined using two antibody-based methods RESULTS: We found that the levels of NPY released into the cell culture media supernatant, and protein extracts of the cell pellet, were significantly higher following exposure to AAV1-NPY than when compared to either a control GFP reporter vector (AAV1-GFP) or sham treated controls. CONCLUSION This first demonstration that an AAV-NPY construct can successfully transduce human neuronal cells supports the pre-clinical development of a clinical trial using AAV-based NPY for pharmacoresistant epilepsy.
Collapse
Affiliation(s)
- Maria I Patrício
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford & NIHR Oxford Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX3 9DU, UK; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Headley Way, Oxford OX3 9DU, UK
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford & NIHR Oxford Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX3 9DU, UK; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Headley Way, Oxford OX3 9DU, UK
| | - Alexander L Green
- Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Headley Way, Oxford OX3 9DU, UK; Nuffield Department of Surgery, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Matthew J During
- Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Headley Way, Oxford OX3 9DU, UK; Nuffield Department of Surgery, University of Oxford, Headley Way, Oxford OX3 9DU, UK.
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford & NIHR Oxford Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX3 9DU, UK; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Headley Way, Oxford OX3 9DU, UK.
| |
Collapse
|
50
|
Sheng J, Liu S, Qin H, Li B, Zhang X. Drug-Resistant Epilepsy and Surgery. Curr Neuropharmacol 2018; 16:17-28. [PMID: 28474565 PMCID: PMC5771378 DOI: 10.2174/1570159x15666170504123316] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/23/2017] [Accepted: 04/25/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Epilepsy is a chronic brain disease that is caused by various factors and characterized by recurrent, episodic and temporary central nervous system dysfunction which results due to excessive discharge of brain neurons. In the past decades, despite the continuous development of antiepileptic drugs, there are still many patients with epilepsy progressing to drugresistant epilepsy. Currently, surgical treatment is one of important way to cure drug-resistant epilepsy. METHODS Data were collected from Web of Science, Medline, Pubmed, through searching of these keywords: "surgery" and "drug-resistant epilepsy". RESULTS An increasing number of studies have shown that surgery plays an important role in the treatment of drug-resistant epilepsy. Moreover, the comprehensive treatment mainly based on surgery can achieve the remission and even cure of drug-resistant epilepsy. CONCLUSION In this review, we discuss the pathogenesis of drug-resistant epilepsy and the comprehensive treatment mainly based on surgery; this review may provide a reference for the clinical treatment of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Jiyao Sheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Shui Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Hanjiao Qin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Xuewen Zhang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| |
Collapse
|