1
|
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: 2.2] [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
|
2
|
Kostoula C, Pascente R, Ravizza T, McCown T, Schoch S, Vezzani A, Becker AJ, van Loo KMJ. Development of In Vivo Imaging Tools for Investigating Astrocyte Activation in Epileptogenesis. Mol Neurobiol 2017; 55:4463-4472. [DOI: 10.1007/s12035-017-0660-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/14/2017] [Indexed: 12/21/2022]
|
3
|
Differential Effect of Neuropeptides on Excitatory Synaptic Transmission in Human Epileptic Hippocampus. J Neurosci 2015; 35:9622-31. [PMID: 26134645 DOI: 10.1523/jneurosci.3973-14.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Development of novel disease-modifying treatment strategies for neurological disorders, which at present have no cure, represents a major challenge for today's neurology. Translation of findings from animal models to humans represents an unresolved gap in most of the preclinical studies. Gene therapy is an evolving innovative approach that may prove useful for clinical applications. In animal models of temporal lobe epilepsy (TLE), gene therapy treatments based on viral vectors encoding NPY or galanin have been shown to effectively suppress seizures. However, how this translates to human TLE remains unknown. A unique possibility to validate these animal studies is provided by a surgical therapeutic approach, whereby resected epileptic tissue from temporal lobes of pharmacoresistant patients are available for neurophysiological studies in vitro. To test whether NPY and galanin have antiepileptic actions in human epileptic tissue as well, we applied these neuropeptides directly to human hippocampal slices in vitro. NPY strongly decreased stimulation-induced EPSPs in dentate gyrus and CA1 (up to 30 and 55%, respectively) via Y2 receptors, while galanin had no significant effect. Receptor autoradiographic binding revealed the presence of both NPY and galanin receptors, while functional receptor binding was only detected for NPY, suggesting that galanin receptor signaling may be impaired. These results underline the importance of validating findings from animal studies in human brain tissue, and advocate for NPY as a more appropriate candidate than galanin for future gene therapy trials in pharmacoresistant TLE patients.
Collapse
|
4
|
Gene-directed enzyme prodrug therapy for localized chemotherapeutics in allograft and xenograft tumor models. Cancer Gene Ther 2014; 21:434-40. [PMID: 25236494 DOI: 10.1038/cgt.2014.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 02/05/2023]
Abstract
Most chemotherapy regimens rely on systemic administration of drugs leading to a wide array of toxicities. Using viral-vector-mediated gene modification of muscle tissues, we have developed a method for gene-directed enzyme prodrug therapy that allows for localized drug administration. An inactive prodrug of geldanamycin was activated locally for inhibition of tumor growth without systemic toxicities. A recombinant adeno-associated virus (rAAV) was used to deliver β-galactosidase (LacZ) to the treatment group and green fluorescent protein to the control group. After 1 week, both groups received adenocarcinoma cells in the same location as the previous rAAV injection. The geldanamycin prodrug was administered 1 h later via intraperitoneal injection. Tumor growth was significantly suppressed in animals whose muscles were gene modified to express β-galactosidase compared with the control. Serum assay to access hepatotoxicity resulted in no significant differences between the animals treated with the inactive or activated form of geldanamycin, indicating minimal damage to non-target organs. Using gene-directed enzyme prodrug therapy, in combination with novel recombinant AAV vectors, we have developed a method for localized activation of chemotherapeutic agents that limits the toxicities seen with traditional systemic administration of these potent drugs.
Collapse
|
5
|
Abstract
Gene therapy may represent an effective alternative to standard pharmacological approaches for certain forms of epilepsy. Currently, the best candidates for this therapeutic approach appear to be epilepsies characterized by a focal lesion. Gene therapy has been attempted to produce antiepileptogenic (prevention of development of epilepsy in subject at risk after having received an epileptogenic insult), antiseizure (reduction of frequency and/or severity of seizures), and disease-modifying (alteration of the natural history of the disease) effects. An example of gene therapy aimed at producing antiepileptogenic effects is a combination therapy based on the supplementation of the neurotrophic factors brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF-2). Antiseizure effects have been obtained by increasing the strength of inhibitory signals (by supplementing specific GABAA receptor subunits or inhibitory neuropeptides like galanin or neuropeptide Y) or by reducing the strength of excitatory signals (by knocking down NMDA receptor subunits). This review summarizes the results obtained to date using gene therapy in epilepsy models and discusses the challenges and the opportunities that this approach can offer for the treatment of human epilepsies.
Collapse
Affiliation(s)
- Michele Simonato
- Department of Medical Sciences, Section of Pharmacology and Neuroscience Center, University of Ferrara, Italy; National Institute of Neuroscience, University of Ferrara, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Italy.
| |
Collapse
|
6
|
Clynen E, Swijsen A, Raijmakers M, Hoogland G, Rigo JM. Neuropeptides as targets for the development of anticonvulsant drugs. Mol Neurobiol 2014; 50:626-46. [PMID: 24705860 PMCID: PMC4182642 DOI: 10.1007/s12035-014-8669-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/27/2014] [Indexed: 11/04/2022]
Abstract
Epilepsy is a common neurological disorder characterized by recurrent seizures. These seizures are due to abnormal excessive and synchronous neuronal activity in the brain caused by a disruption of the delicate balance between excitation and inhibition. Neuropeptides can contribute to such misbalance by modulating the effect of classical excitatory and inhibitory neurotransmitters. In this review, we discuss 21 different neuropeptides that have been linked to seizure disorders. These neuropeptides show an aberrant expression and/or release in animal seizure models and/or epilepsy patients. Many of these endogenous peptides, like adrenocorticotropic hormone, angiotensin, cholecystokinin, cortistatin, dynorphin, galanin, ghrelin, neuropeptide Y, neurotensin, somatostatin, and thyrotropin-releasing hormone, are able to suppress seizures in the brain. Other neuropeptides, such as arginine-vasopressine peptide, corticotropin-releasing hormone, enkephalin, β-endorphin, pituitary adenylate cyclase-activating polypeptide, and tachykinins have proconvulsive properties. For oxytocin and melanin-concentrating hormone both pro- and anticonvulsive effects have been reported, and this seems to be dose or time dependent. All these neuropeptides and their receptors are interesting targets for the development of new antiepileptic drugs. Other neuropeptides such as nesfatin-1 and vasoactive intestinal peptide have been less studied in this field; however, as nesfatin-1 levels change over the course of epilepsy, this can be considered as an interesting marker to diagnose patients who have suffered a recent epileptic seizure.
Collapse
Affiliation(s)
- Elke Clynen
- Biomedical Research Institute BIOMED, Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium,
| | | | | | | | | |
Collapse
|
7
|
Xu X, Guo F, He Q, Cai X, Min D, Wang Q, Wang S, Tian L, Cai J, Zhao Y. Altered expression of neuropeptide Y, Y1 and Y2 receptors, but not Y5 receptor, within hippocampus and temporal lobe cortex of tremor rats. Neuropeptides 2014; 48:97-105. [PMID: 24444822 DOI: 10.1016/j.npep.2013.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 12/18/2013] [Accepted: 12/22/2013] [Indexed: 01/24/2023]
Abstract
As an endogenous inhibitor of glutamate-mediated synaptic transmission in mammalian central nervous system, neuropeptide Y (NPY) plays a crucial role in regulating homeostasis of neuron excitability. Loss of balance between excitatory and inhibitory neurotransmission is thought to be a chief mechanism of epileptogenesis. The abnormal expression of NPY and its receptors observed following seizures have been demonstrated to be related to the production of epilepsy. The tremor rat (TRM) is a hereditary epileptic animal model. So far, there is no report concerning whether NPY and its receptors may be involved in TRM pathogenesis. In this study, we focused on the expression of NPY and its three receptor subtypes: Y1R, Y2R and Y5R in the TRM brain. We first found the expression of NPY in TRM hippocampus and temporal lobe cortex was increased compared with control (Wistar) rats. The mRNA and protein expression of Y1R was down-regulated in hippocampus but up-regulated in temporal lobe cortex, whereas Y2R expression was significantly increased in both areas. There was no significant change of Y5R expression in either area. The immunohistochemistry data showed that Y1R, Y2R, Y5R were present throughout CA1, CA3, dentate gyrus (DG) and the entorhinal cortex which is included in the temporal lobe cortex of TRM. In conclusion, our results showed the altered expression of NPY, Y1R and Y2R but not Y5R in hippocampus and temporal lobe cortex of TRM brain. This abnormal expression may be associated with the generation of epileptiform activity and provide a candidate target for treatment of genetic epilepsy.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China; Department of Neurology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Qun He
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Xinze Cai
- Central Lab, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dongyu Min
- Experiment Center of Traditional Chinese Medicine, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| | - Qianhui Wang
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Shaocheng Wang
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Liu Tian
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Jiqun Cai
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Yujie Zhao
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China.
| |
Collapse
|
8
|
Fat grafting as a vehicle for the delivery of recombinant adenoassociated viral vectors to achieve gene modification of muscle flaps. Ann Plast Surg 2014; 70:726-31. [PMID: 23403543 DOI: 10.1097/sap.0b013e3182414add] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The combination of gene therapy and plastic surgery may have the potential to improve the specificity that is needed to achieve clinically applicable treatment regimens. Our goal was to develop a method for gene modification that would yield sustainable production of gene products but would be less time consuming than existing protocols. METHODS An adenoassociated virus was used to deliver gene products to pectoralis muscle flaps. Gene modification was accomplished via either direct injection or novel fat grafting techniques. RESULTS The production of gene product was observable by both in vivo imaging and immunohistochemical staining. Gene products were not detected in tissues that were not in contact with the fat grafts that were incubated with the viral vector, indicating that the transduction stayed local to the flap. CONCLUSIONS Using novel recombinant adenoassociated virus vectors, we have developed a method for gene delivery that is highly efficient and applicable to muscle flaps.
Collapse
|
9
|
Hui Yin Y, Ahmad N, Makmor-Bakry M. Pathogenesis of epilepsy: challenges in animal models. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2013; 16:1119-32. [PMID: 24494063 PMCID: PMC3909622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/02/2013] [Indexed: 11/13/2022]
Abstract
Epilepsy is one of the most common chronic disorders affecting individuals of all ages. A greater understanding of pathogenesis in epilepsy will likely provide the basis fundamental for development of new antiepileptic therapies that aim to prevent the epileptogenesis process or modify the progression of epilepsy in addition to treatment of epilepsy symptomatically. Therefore, several investigations have embarked on advancing knowledge of the mechanism underlying epileptogenesis, understanding in mechanism of pharmacoresistance and discovering antiepileptogenic or disease-modifying therapy. Animal models play a crucial and significant role in providing additional insight into mechanism of epileptogenesis. With the help of these models, epileptogenesis process has been demonstrated to be involved in various molecular and biological pathways or processes. Hence, this article will discuss the known and postulated mechanisms of epileptogenesis and challenges in using the animal models.
Collapse
Affiliation(s)
- Yow Hui Yin
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurulumi Ahmad
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Makmor-Bakry
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia,Corresponding author: Mohd Makmor-Bakry. Faculty of Pharmacy, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia. Tel: +60392897244; Fax: +60326983271;
| |
Collapse
|
10
|
Association of age at onset in Huntington disease with functional promoter variations in NPY and NPY2R. J Mol Med (Berl) 2013; 92:177-84. [DOI: 10.1007/s00109-013-1092-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/09/2013] [Accepted: 10/02/2013] [Indexed: 12/23/2022]
|
11
|
Casillas-Espinosa PM, Powell KL, O'Brien TJ. Regulators of synaptic transmission: roles in the pathogenesis and treatment of epilepsy. Epilepsia 2013; 53 Suppl 9:41-58. [PMID: 23216578 DOI: 10.1111/epi.12034] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Synaptic transmission is the communication between a presynaptic and a postsynaptic neuron, and the subsequent processing of the signal. These processes are complex and highly regulated, reflecting their importance in normal brain functioning and homeostasis. Sustaining synaptic transmission depends on the continuing cycle of synaptic vesicle formation, release, and endocytosis, which requires proteins such as dynamin, syndapin, synapsin, and synaptic vesicle protein 2A. Synaptic transmission is regulated by diverse mechanisms, including presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors and signaling, and modulators of neurotransmission. Neurotransmitters released presynaptically can bind to their postsynaptic receptors, the inhibitory γ-aminobutyric acid (GABA)ergic receptors or the excitatory glutamate receptors. Once released, glutamate activates a variety of postsynaptic receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA), kainate, and metabotropic receptors. The activation of the receptors triggers downstream signaling cascades generating a vast array of effects, which can be modulated by a numerous auxiliary regulatory subunits. Moreover, different neuropeptides such as neuropeptide Y, brain-derived neurotrophic factor (BDNF), somatostatin, ghrelin, and galanin, act as regulators of diverse synaptic functions and along with the classic neurotransmitters. Abnormalities in the regulation of synaptic transmission play a critical role in the pathogenesis of numerous brain diseases, including epilepsy. This review focuses on the different mechanisms involved in the regulation of synaptic transmission, which may play a role in the pathogenesis of epilepsy: the presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors, and modulators of neurotransmission, including the mechanism by which drugs can modulate the frequency and severity of epileptic seizures.
Collapse
Affiliation(s)
- Pablo M Casillas-Espinosa
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
12
|
Chachua T, Poon KL, Yum MS, Nesheiwat L, DeSantis K, Velíšková J, Velíšek L. Rapamycin has age-, treatment paradigm-, and model-specific anticonvulsant effects and modulates neuropeptide Y expression in rats. Epilepsia 2012; 53:2015-25. [PMID: 23016669 PMCID: PMC3496841 DOI: 10.1111/j.1528-1167.2012.03674.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Rapamycin (RAP) has certain antiepileptogenic features. However, it is unclear whether these effects can be explained by the anticonvulsant action of RAP, which has not been studied. To address this question, we tested potential anticonvulsant effects of RAP in immature and adult rats using different seizure models and treatment paradigms. In addition, we studied changes in the expression of neuropeptide Y (NPY) induced by RAP, which may serve as an indirect target of the RAP action. METHODS A complex approach was adopted to evaluate the anticonvulsant potential of RAP: We used flurothyl-, pentylenetetrazole (PTZ)-, N-methyl-D-aspartate (NMDA)-, and kainic acid (KA)-induced seizures to test the effects of RAP using different pretreatment protocols in immature and adult rats. We also evaluated expression of NPY within the primary motor cortex, hippocampal CA1, and dentate gyrus (DG) after different pretreatments with RAP in immature rats. KEY FINDINGS We found the following: (1) RAP administered with short-term pretreatment paradigms has a weak anticonvulsant potential in the seizure models with compromised inhibition. (2) Lack of RAP efficacy correlates with decreased NPY expression in the cortex, CA1, and DG. Specifically in immature rats, a single dose of RAP (3 mg/kg) 4 or 24 h before seizure testing had anticonvulsant effects against PTZ-induced seizures. In the flurothyl seizure model only the 4-h pretreatment with RAP was anticonvulsant in the both age groups. Short-term pretreatments with RAP had no effects against NMDA- and KA-induced seizures tested in immature rats. Long-term pretreatments with RAP over 8 days did not show beneficial effect in all tested seizure models in developing rats. Moreover, the long-term pretreatment with RAP had a slight proconvulsant effect on KA-induced seizures. In immature rats, any lack of anticonvulsant effect (including proconvulsant effect of multiple doses of RAP) was associated with downregulation of NPY expression in the cortex and DG. In immature animals, after a single dose of RAP with 24 h delay, we found a decrease of NPY expression in DG, and CA1 as well. SIGNIFICANCE Our data show weak age-, treatment paradigm-, and model-specific anticonvulsant effects of RAP as well as loss of those effects after long-term RAP pretreatment associated with downregulation of NPY expression. These findings suggest that RAP is a poor anticonvulsant and may have beneficial effects only against epileptogenesis. In addition, our data present new insights into mechanisms of RAP action on seizures indicating a possible connection between mammalian target of rapamycin (mTOR) signaling and NPY system.
Collapse
Affiliation(s)
- Tamar Chachua
- Department of Cell Biology & Anatomy, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, U.S.A.
| | | | | | | | | | | | | |
Collapse
|
13
|
van Raay L, Jovanovska V, Morris MJ, O'Brien TJ. Focal administration of neuropeptide Y into the S2 somatosensory cortex maximally suppresses absence seizures in a genetic rat model. Epilepsia 2012; 53:477-84. [PMID: 22220638 DOI: 10.1111/j.1528-1167.2011.03370.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE Neuropeptide Y (NPY) is an inhibitory neurotransmitter that suppresses focal and generalized seizures in animal models. In this study, we investigated the sites within the thalamocortical circuit that NPY acts to suppress seizures in genetic absence epilepsy rats from Strasbourg (GAERS). METHODS In conscious freely moving GAERS, NPY was administered via intracerebral microcannulae implanted bilaterally into one of the following regions: primary somatosensory cortex (S1), secondary somatosensory cortex (S2), the primary motor cortex (M1), caudal nucleus reticular thalamus (nRT), or ventrobasal thalamus (VB). Animals received vehicle and up to three doses of NPY, in a randomized order. Electroencephalography (EEG) recordings were carried out for 30 min prior to injection and 90 min after the injection of NPY or vehicle. KEY FINDINGS Focal microinjections of NPY into the S2 cortex suppressed seizures in a dose-dependent manner, with the response being significantly different at the highest dose (1.5 mm) compared to vehicle for total time in seizures postinjection (7.2 ± 3.0% of saline, p < 0.01) and average number of seizures (9.4 ± 4.9% of saline, p < 0.05). In contrast NPY microinjections into the VB resulted in an aggravation of seizures. SIGNIFICANCE NPY produces contrasting effects on absence-like seizures in GAERS depending on the site of injection within the thalamocortical circuit. The S2 is the site at which NPY most potently acts to suppress absence-like seizures in GAERS, whereas seizure-aggravating effects are seen in the VB. These results provide further evidence to support the proposition that these electroclinically "generalized" seizures are being driven by a topographically restricted region within the somatosensory cortex.
Collapse
Affiliation(s)
- Leena van Raay
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
14
|
Olesen MV, Christiansen SH, Gøtzsche CR, Nikitidou L, Kokaia M, Woldbye DPD. Neuropeptide Y Y1 receptor hippocampal overexpression via viral vectors is associated with modest anxiolytic-like and proconvulsant effects in mice. J Neurosci Res 2011; 90:498-507. [PMID: 21971867 DOI: 10.1002/jnr.22770] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/15/2011] [Accepted: 07/24/2011] [Indexed: 02/06/2023]
Abstract
Neuropeptide Y (NPY) exerts anxiolytic- and antidepressant-like effects in rodents that appear to be mediated via Y1 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY in the hippocampus or amygdala has previously been shown to confer anxiolytic-like effect in rodents. The present study explored an alternative and more specific approach: overexpression of Y1 receptors. Using a recombinant adeno-associated viral vector (rAAV) encoding the Y1 gene (rAAV-Y1), we, for the first time, induced overexpression of functional transgene Y1 receptors in the hippocampus of adult mice and tested the animals in anxiety- and depression-like behavior. Hippocampal Y1 receptors have been suggested to mediate seizure-promoting effect, so the effects of rAAV-induced Y1 receptor overexpression were also tested in kainate-induced seizures. Y1 receptor transgene overexpression was found to be associated with modest anxiolytic-like effect in the open field and elevated plus maze tests, but no effect was seen on depression-like behavior using the tail suspension and forced swim tests. However, the rAAV-Y1 vector modestly aggravated kainate-induced seizures. These data indicate that rAAV-induced overexpression of Y1 receptors in the hippocampus could confer anxiolytic-like effect accompanied by a moderate proconvulsant adverse effect. Further studies are clearly needed to determine whether Y1 gene therapy might have a future role in the treatment of anxiety disorders.
Collapse
Affiliation(s)
- M V Olesen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
15
|
Seyhan AA. RNAi: a potential new class of therapeutic for human genetic disease. Hum Genet 2011; 130:583-605. [PMID: 21537948 DOI: 10.1007/s00439-011-0995-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/17/2011] [Indexed: 12/19/2022]
Abstract
Dominant negative genetic disorders, in which a mutant allele of a gene causes disease in the presence of a second, normal copy, have been challenging since there is no cure and treatments are only to alleviate the symptoms. Current therapies involving pharmacological and biological drugs are not suitable to target mutant genes selectively due to structural indifference of the normal variant of their targets from the disease-causing mutant ones. In instances when the target contains single nucleotide polymorphism (SNP), whether it is an enzyme or structural or receptor protein are not ideal for treatment using conventional drugs due to their lack of selectivity. Therefore, there is a need to develop new approaches to accelerate targeting these previously inaccessible targets by classical therapeutics. Although there is a cooling trend by the pharmaceutical industry for the potential of RNA interference (RNAi), RNAi and other RNA targeting drugs (antisense, ribozyme, etc.) still hold their promise as the only drugs that provide an opportunity to target genes with SNP mutations found in dominant negative disorders, genes specific to pathogenic tumor cells, and genes that are critical for mediating the pathology of various other diseases. Because of its exquisite specificity and potency, RNAi has attracted a considerable interest as a new class of therapeutic for genetic diseases including amyotrophic lateral sclerosis, Huntington's disease (HD), Alzheimer's disease (AD), Parkinson's disease (PD), spinocerebellar ataxia, dominant muscular dystrophies, and cancer. In this review, progress and challenges in developing RNAi therapeutics for genetic diseases will be discussed.
Collapse
Affiliation(s)
- Attila A Seyhan
- Pfizer Inc., Translational Immunology, Inflammation and Immunology, 200 Cambridgepark Drive, Cambridge, MA 02140, USA.
| |
Collapse
|
16
|
Abstract
Neuropeptide Y (NPY) is widely distributed in the human body and contributes to a vast number of physiological processes. Since its discovery, NPY has been implicated in metabolic regulation and, although interest in its role in central mechanisms related to food intake and obesity has somewhat diminished, the topic remains a strong focus of research concerning NPY signalling. In addition, a number of other uses for modulators of NPY receptors have been implied in a range of diseases, although the development of NPY receptor ligands has been slow, with no clinically approved receptor therapeutics currently available. Nevertheless, several interesting small molecule compounds, notably Y2 receptor antagonists, have been published recently, fueling optimism in the field. Herein we review the role of NPY in the pathophysiology of a number of diseases and highlight instances where NPY receptor signalling systems are attractive therapeutic targets.
Collapse
Affiliation(s)
- Shaun P Brothers
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | |
Collapse
|
17
|
Theofilas P, Brar S, Stewart KA, Shen HY, Sandau US, Poulsen D, Boison D. Adenosine kinase as a target for therapeutic antisense strategies in epilepsy. Epilepsia 2011; 52:589-601. [PMID: 21275977 DOI: 10.1111/j.1528-1167.2010.02947.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Given the high incidence of refractory epilepsy, novel therapeutic approaches and concepts are urgently needed. To date, viral-mediated delivery and endogenous expression of antisense sequences as a strategy to prevent seizures have received little attention in epilepsy therapy development efforts. Here we validate adenosine kinase (ADK), the astrocyte-based key negative regulator of the brain's endogenous anticonvulsant adenosine, as a potential therapeutic target for antisense-mediated seizure suppression. METHODS We developed adenoassociated virus 8 (AAV8)-based gene therapy vectors to selectively modulate ADK expression in astrocytes. Cell type selectivity was achieved by expressing an Adk-cDNA in sense or antisense orientation under the control of an astrocyte-specific gfaABC1D promoter. Viral vectors where injected into the CA3 of wild-type mice or spontaneously epileptic Adk-tg transgenic mice that overexpress ADK in brain. After virus injection, ADK expression was assessed histologically and biochemically. In addition, intracranial electroencephalography (EEG) recordings were obtained. KEY FINDINGS We demonstrate in wild-type mice that viral overexpression of ADK within astrocytes is sufficient to trigger spontaneous recurrent seizures in the absence of any other epileptogenic event, whereas ADK downregulation via AAV8-mediated RNA interference almost completely abolished spontaneous recurrent seizures in Adk-tg mice. SIGNIFICANCE Our data demonstrate that modulation of astrocytic ADK expression can trigger or prevent seizures, respectively. This is the first study to use an antisense approach to validate ADK as a rational therapeutic target for the treatment of epilepsy and suggests that gene therapies based on the knock down of ADK might be a feasible approach to control seizures in refractory epilepsy.
Collapse
Affiliation(s)
- Panos Theofilas
- RS Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon 97232, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Functional, metabolic, and synaptic changes after seizures as potential targets for antiepileptic therapy. Epilepsy Behav 2010; 19:105-13. [PMID: 20705520 DOI: 10.1016/j.yebeh.2010.06.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 06/17/2010] [Indexed: 01/11/2023]
Abstract
Little is known about how the brain limits seizure duration and terminates seizures. Depending on severity and duration, a single seizure is followed by various functional, metabolic, and synaptic changes that may form targets for novel therapeutic strategies. It is long known that most seizures are followed by a period of postictal refractoriness during which the threshold for induction of additional seizures is increased. The endogenous anticonvulsant mechanisms involved in this phenomenon may be relevant for both spontaneous seizure arrest and increase of seizure threshold after seizure arrest. Postictal refractoriness has been extensively studied in various seizure and epilepsy models, including electrically and chemically induced seizures, kindling, and genetic animal models of epilepsy. During kindling development, two antagonistic processes occur simultaneously, one responsible for kindling-like events and the other for terminating ictus and postictal refractoriness. Frequently occurring seizures may lead to an accumulation of postictal refractoriness that may last weeks. The mechanisms involved in seizure termination and postictal refractoriness include changes in ionic microenvironment, in pH, and in various endogenous neuromodulators such as adenosine and neuropeptides. In animal models, the anticonvulsant efficacy of several antiepileptic drugs (AEDs) is increased during postictal refractoriness, which is a logical consequence of the interaction between endogenous anticonvulsant processes and the mechanism of AEDs. As discussed in this review, enhanced understanding of these endogenous processes may lead to novel targets for AED development.
Collapse
|
19
|
Abstract
Since its discovery a decade ago, RNA interference (RNAi) has been developed not only into powerful experimental tools but also into promising novel therapeutics. In contrast to conventional antiepileptic drugs (AEDs) that target specific proteins such as ion channels or receptors, RNAi-based therapeutics exploit an endogenous regulatory mechanism of gene expression and thereby are poised to prevent or reverse pathogenetic mechanisms involved in seizure development. Therapeutic RNAi has been widely explored for dominant targets involved in neurodegenerative diseases; however, their use for epilepsy therapy has received less attention. This review discusses potential RNAi-based targets that are of interest for epilepsy therapy, including adenosine kinase (ADK), the key negative regulator of the brain's endogenous anticonvulsant adenosine. Overexpression of ADK, and the resulting adenosine deficiency, are pathologic hallmarks of the sclerotic epileptic brain, and have been implicated in seizure generation. Therefore, RNAi-strategies aimed at reducing ADK (and increasing adenosine) are based on a direct neurochemical rationale that has recently been explored experimentally using ex vivo and in vivo gene therapy approaches. Technical issues and challenges remain before those promising tools can be developed into future therapeutics for epilepsy.
Collapse
Affiliation(s)
- Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon 97232, USA.
| |
Collapse
|
20
|
Mitra R, Sapolsky RM. Gene therapy in rodent amygdala against fear disorders. Expert Opin Biol Ther 2010; 10:1289-303. [DOI: 10.1517/14712598.2010.509341] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Green BR, White KL, McDougle DR, Zhang L, Klein B, Scholl EA, Pruess TH, White HS, Bulaj G. Introduction of lipidization-cationization motifs affords systemically bioavailable neuropeptide Y and neurotensin analogs with anticonvulsant activities. J Pept Sci 2010; 16:486-95. [DOI: 10.1002/psc.1266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Giné E, Morales-Garcia JA, Perez-Castillo A, Santos A. Developmental hypothyroidism increases the expression of kainate receptors in the hippocampus and the sensitivity to kainic acid-induced seizures in the rat. Endocrinology 2010; 151:3267-76. [PMID: 20410204 DOI: 10.1210/en.2010-0070] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormones are essential for normal brain development, and multiple alterations at behavioral, cognitive, cellular, and molecular levels have been described in animals made hypothyroid during development. Here we analyzed the effect of developmental hypothyroidism in the rat on the sensitivity to kainic acid-induced limbic seizures and the expression of kainate receptors in the hippocampus. Our results show that hypothyroid rats are extremely sensitive to the proconvulsant and neurotoxic effects of kainic acid (KA). Hypothyroid rats entered in status epilepticus at a dose of KA three times lower than that required to reach status epilepticus in control animals. In accordance with this, high levels of glial activation and neuronal loss after low KA dose injections were observed only in the hippocampus of hypothyroid rats. These effects correlated with an increased expression of kainate receptor subunits, excluding GluR5, in the hippocampus of hypothyroid animals. The concentrations of GluR6, GluR7, KAR1, and KAR2 (ionotropic glutamate receptor subunits of the kainic acid subtype) mRNAs were increased between 50 and 250% in hypothyroid animals relative to the values in controls. In agreement with these results, Western blot and immunohistochemical analysis showed a clear increase in the hippocampal content of GluR6/7 proteins in hypothyroid animals.
Collapse
Affiliation(s)
- Elena Giné
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
23
|
Differential regulation of vasoactive intestinal peptide (VIP) in the dentate gyrus and hippocampus via the NO-cGMP pathway following kainic acid-induced seizure in the rat. J Mol Neurosci 2010; 42:359-69. [PMID: 20369387 DOI: 10.1007/s12031-010-9353-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 03/12/2010] [Indexed: 12/14/2022]
Abstract
We have previously shown that kainic acid (KA) increases nitric oxide (NO) synthase (NOS) production in the rat dentate gyrus (DG) and hippocampus (CA3), and NOS inhibition [(by N(G)-nitro-L-arginine methylester (L-NAME)] modulates the vasoactive intestinal peptide (VIP)-responsive gene, activity-dependent neuroprotective protein, and alters neuro- and astrogliogenesis (Cosgrave et al. in Neurobiol Dis 30(3):281-292 2008, J Mol Neurosci 39(1-2):9-21, 2009, 2010). In the present study, using the same model we demonstrate that VIP synthesis is differentially regulated by the NO-cyclic guanosine monophosphate (cGMP) pathway in the DG and CA3 at 3 h and 3 days post-KA. At 3 h post-KA: In L-NAME+KA/7-nitroindazole (7-NI)+KA, stratum granulosum (SG) and subgranular zone (SGZ) cells were intensely stained for VIP when compared with L-NAME/7-NI/KA alone. Soluble guanylyl cyclase inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, blocks cGMP production), suppressed astrocytic activation (glial fibrillary acidic protein) but other cell types were VIP(+); however, ODQ+KA suppressed overall VIP synthesis in the DG. At 3 days post-KA: In L-NAME+KA/7-NI+KA, SGZ and SG cells continued to express VIP, while in the KA alone, only SGZ cells were VIP(+). ODQ increased VIP(+) cells in the SG, and in contrast to 3 h, VIP-containing nNOS(+) cells increased in ODQ+KA when compared to vehicle+KA. In the hippocampus, 7-NI/ODQ had no effect on VIP at 3 h/3 days, while L-NAME+KA at 3 days increased VIP(+) cells, but reduced VIP-like immunoreactivity in astrocytes. These results suggest that the NO-cGMP pathway differentially regulates VIP in the DG and hippocampus during seizure.
Collapse
|
24
|
Boison D, Stewart KA. Therapeutic epilepsy research: from pharmacological rationale to focal adenosine augmentation. Biochem Pharmacol 2009; 78:1428-37. [PMID: 19682439 DOI: 10.1016/j.bcp.2009.08.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 08/05/2009] [Accepted: 08/05/2009] [Indexed: 01/16/2023]
Abstract
Epilepsy is a common seizure disorder affecting approximately 70 million people worldwide. Current pharmacotherapy is neuron-centered, frequently accompanied by intolerable side effects, and fails to be effective in about one third of patients. Therefore, new therapeutic concepts are needed. Recent research suggests an astrocytic basis of epilepsy, presenting the possibility of novel therapeutic targets. In particular, dysfunction of the astrocyte-controlled, endogenous, adenosine-based seizure control system of the brain is implicated in seizure generation. Thus, astrogliosis - a pathological hallmark of the epileptic brain - is associated with upregulation of the adenosine-removing enzyme adenosine kinase (ADK), resulting in focal adenosine deficiency. Both astrogliotic upregulation of ADK in epilepsy and transgenic overexpression of ADK are associated with seizures, and inhibition of ADK prevents seizures in a mouse model of pharmacoresistant epilepsy. These findings link adenosine deficiency with seizures and predict that adenosine augmentation therapies (AATs) will likely be effective in preventing seizures. Given the wide-spread systemic and central side effects of systemically administered AATs, focal AATs (i.e., limited to the astrogliotic lesion) are a necessity. This Commentary will discuss the pharmacological rationale for the development of focal AATs. Additionally, several AAT strategies will be discussed: (1) adenosine released from silk-based brain implants; (2) adenosine released from locally implanted encapsulated cells; (3) adenosine released from stem cell-derived brain implants; and (4) adenosine augmenting gene therapies. Finally, new developments and therapeutic challenges in using focal AATs for epilepsy therapy will critically be evaluated.
Collapse
Affiliation(s)
- Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research, Portland, OR 97232, USA.
| | | |
Collapse
|
25
|
Rogawski MA. Convection-enhanced delivery in the treatment of epilepsy. Neurotherapeutics 2009; 6:344-51. [PMID: 19332329 PMCID: PMC2753495 DOI: 10.1016/j.nurt.2009.01.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Revised: 01/23/2009] [Accepted: 01/23/2009] [Indexed: 11/23/2022] Open
Abstract
Convection-enhanced delivery (CED) is a novel drug-delivery technique that uses positive hydrostatic pressure to deliver a fluid containing a therapeutic substance by bulk flow directly into the interstitial space within a localized region of the brain parenchyma. CED circumvents the blood-brain barrier and provides a wider, more homogenous distribution than bolus deposition (focal injection) or other diffusion-based delivery approaches. A potential use of CED is for the local delivery of antiseizure agents, which would provide an epilepsy treatment approach that avoids the systemic toxicities of orally administered antiepileptic drugs and bystander effects on nonepileptic brain regions. Recent studies have demonstrated that brief CED infusions of nondiffusible peptides that inhibit the release of excitatory neurotransmitters, including omega-conotoxins and botulinum neurotoxins, can produce long-lasting (weeks to months) seizure protection in the rat amygdala-kindling model. Seizure protection is obtainable without detectable neurological or behavioral side effects. Although conventional diffusible antiepileptic drugs do confer seizure protection when administered locally by CED, the effect is transitory. CED is a potential approach for seizure protection that could represent an alternative to resective surgery in the treatment of focal epilepsies that are resistant to orally-administered antiepileptic drugs. The prolonged duration of action of nondiffusible toxins would allow seizure protection to be maintained chronically with infrequent reinfusions.
Collapse
Affiliation(s)
- Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, California 95817, USA.
| |
Collapse
|