1
|
Kovács Z, Rauch E, D’Agostino DP, Ari C. Putative Role of Adenosine A1 Receptors in Exogenous Ketone Supplements-Evoked Anti-Epileptic Effect. Int J Mol Sci 2024; 25:9869. [PMID: 39337356 PMCID: PMC11432942 DOI: 10.3390/ijms25189869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Approximately 30% of patients with epilepsy are drug-refractory. There is an urgent need to elucidate the exact pathophysiology of different types of epilepsies and the mechanisms of action of both antiseizure medication and metabolic therapies to treat patients more effectively and safely. For example, it has been demonstrated that exogenous ketone supplement (EKS)-generated therapeutic ketosis, as a metabolic therapy, may decrease epileptic activity in both animal models and humans, but its exact mechanism of action is unknown. However, it was demonstrated that therapeutic ketosis, among others, can increase adenosine level, which may enhance activity of A1 adenosine receptors (A1Rs) in the brain. It has also been demonstrated previously that adenosine has anti-epileptic effect through A1Rs in different models of epilepsies. Thus, it is possible that (i) therapeutic ketosis generated by the administration of EKSs may exert its anti-epileptic effect through, among other mechanisms, increased adenosine level and A1R activity and that (ii) the enhanced activity of A1Rs may be a necessary anti-epileptic mechanism evoked by EKS administration-generated ketosis. Moreover, EKSs can evoke and maintain ketosis without severe side effects. These results also suggest that the therapeutic application of EKS-generated ketosis may be a promising opportunity to treat different types of epilepsies. In this literature review, we specifically focus on the putative role of A1Rs in the anti-epileptic effect of EKS-induced ketosis.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Biology, BDTTC, ELTE Eötvös Loránd University, Károlyi Gáspár tér 4., 9700 Szombathely, Hungary or (Z.K.); (E.R.)
| | - Enikő Rauch
- Department of Biology, BDTTC, ELTE Eötvös Loránd University, Károlyi Gáspár tér 4., 9700 Szombathely, Hungary or (Z.K.); (E.R.)
- Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary
| | - Dominic P. D’Agostino
- Ketone Technologies LLC., Tampa, FL 33612, USA;
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA
| | - Csilla Ari
- Ketone Technologies LLC., Tampa, FL 33612, USA;
- Department of Psychology, Behavioral Neuroscience Research Laboratory, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
2
|
Ho SY, Chen IC, Tsai CW, Chang KC, Lin CJ, Chern Y, Liou HH. Anticonvulsant effect of equilibrative nucleoside transporters 1 inhibitor in a mouse model of Dravet syndrome. Hippocampus 2024; 34:7-13. [PMID: 37933097 DOI: 10.1002/hipo.23584] [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: 01/31/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023]
Abstract
There are limited therapeutic options for patients with Dravet syndrome (DS). The equilibrative nucleoside transporters 1 (ENT1) mediate both the influx and efflux of adenosine across the cell membrane exerted beneficial effects in the treatment of epilepsy. This study aimed to evaluate the anticonvulsant effect of the ENT1 inhibitor in an animal model of DS (Scn1aE1099X/+ mice). J7 (5 mg/kg) treatment was efficacious in elevating seizure threshold in Scn1aE1099X/+ mice after hyperthermia exposure. Moreover, the J7 treatment significantly reduced the frequency of spontaneous excitatory post-synaptic currents (sEPSCs, ~35% reduction) without affecting the amplitude in dentate gyrus (DG) granule cells. Pretreatment with the adenosine A1 receptor (A1R) antagonist, DPCPX, abolished the J7 effects on sEPSCs. These observations suggest that the J7 shows an anticonvulsant effect in hyperthermia-induced seizures in Scn1aE1099X/+ mice. This effect possibly acts on presynaptic A1R-mediated signaling modulation in granule cells.
Collapse
Affiliation(s)
- Shih-Yin Ho
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
| | - I-Chun Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Che-Wen Tsai
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Chieh Chang
- Department of Neurology, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Horng-Huei Liou
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| |
Collapse
|
3
|
Gernert M, Feja M. Bypassing the Blood-Brain Barrier: Direct Intracranial Drug Delivery in Epilepsies. Pharmaceutics 2020; 12:pharmaceutics12121134. [PMID: 33255396 PMCID: PMC7760299 DOI: 10.3390/pharmaceutics12121134] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsies are common chronic neurological diseases characterized by recurrent unprovoked seizures of central origin. The mainstay of treatment involves symptomatic suppression of seizures with systemically applied antiseizure drugs (ASDs). Systemic pharmacotherapies for epilepsies are facing two main challenges. First, adverse effects from (often life-long) systemic drug treatment are common, and second, about one-third of patients with epilepsy have seizures refractory to systemic pharmacotherapy. Especially the drug resistance in epilepsies remains an unmet clinical need despite the recent introduction of new ASDs. Apart from other hypotheses, epilepsy-induced alterations of the blood-brain barrier (BBB) are thought to prevent ASDs from entering the brain parenchyma in necessary amounts, thereby being involved in causing drug-resistant epilepsy. Although an invasive procedure, bypassing the BBB by targeted intracranial drug delivery is an attractive approach to circumvent BBB-associated drug resistance mechanisms and to lower the risk of systemic and neurologic adverse effects. Additionally, it offers the possibility of reaching higher local drug concentrations in appropriate target regions while minimizing them in other brain or peripheral areas, as well as using otherwise toxic drugs not suitable for systemic administration. In our review, we give an overview of experimental and clinical studies conducted on direct intracranial drug delivery in epilepsies. We also discuss challenges associated with intracranial pharmacotherapy for epilepsies.
Collapse
Affiliation(s)
- Manuela Gernert
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany;
- Center for Systems Neuroscience, D-30559 Hannover, Germany
- Correspondence: ; Tel.: +49-(0)511-953-8527
| | - Malte Feja
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany;
- Center for Systems Neuroscience, D-30559 Hannover, Germany
| |
Collapse
|
4
|
Mateus JM, Ribeiro FF, Alonso-Gomes M, Rodrigues RS, Marques JM, Sebastião AM, Rodrigues RJ, Xapelli S. Neurogenesis and Gliogenesis: Relevance of Adenosine for Neuroregeneration in Brain Disorders. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Joana M. Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Filipa F. Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marta Alonso-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui S. Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana M. Marques
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ricardo J. Rodrigues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
5
|
Poppe D, Doerr J, Schneider M, Wilkens R, Steinbeck JA, Ladewig J, Tam A, Paschon DE, Gregory PD, Reik A, Müller CE, Koch P, Brüstle O. Genome Editing in Neuroepithelial Stem Cells to Generate Human Neurons with High Adenosine-Releasing Capacity. Stem Cells Transl Med 2018; 7:477-486. [PMID: 29589874 PMCID: PMC5980162 DOI: 10.1002/sctm.16-0272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
As a powerful regulator of cellular homeostasis and metabolism, adenosine is involved in diverse neurological processes including pain, cognition, and memory. Altered adenosine homeostasis has also been associated with several diseases such as depression, schizophrenia, or epilepsy. Based on its protective properties, adenosine has been considered as a potential therapeutic agent for various brain disorders. Since systemic application of adenosine is hampered by serious side effects such as vasodilatation and cardiac suppression, recent studies aim at improving local delivery by depots, pumps, or cell-based applications. Here, we report on the characterization of adenosine-releasing human embryonic stem cell-derived neuroepithelial stem cells (long-term self-renewing neuroepithelial stem [lt-NES] cells) generated by zinc finger nuclease (ZFN)-mediated knockout of the adenosine kinase (ADK) gene. ADK-deficient lt-NES cells and their differentiated neuronal and astroglial progeny exhibit substantially elevated release of adenosine compared to control cells. Importantly, extensive adenosine release could be triggered by excitation of differentiated neuronal cultures, suggesting a potential activity-dependent regulation of adenosine supply. Thus, ZFN-modified neural stem cells might serve as a useful vehicle for the activity-dependent local therapeutic delivery of adenosine into the central nervous system. Stem Cells Translational Medicine 2018;7:477-486.
Collapse
Affiliation(s)
- Daniel Poppe
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Jonas Doerr
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of BonnBonnGermany
| | - Ruven Wilkens
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Julius A. Steinbeck
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Julia Ladewig
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | | | | | | | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of BonnBonnGermany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| |
Collapse
|
6
|
Zhu QL, Luo Y, Xue QS, Zhang FJ, Yu BW. Different doses of sevoflurane facilitate and impair learning and memory function through activation of the ERK pathway and synthesis of ARC protein in the rat hippocampus. Brain Res 2017; 1678:174-179. [PMID: 29074343 DOI: 10.1016/j.brainres.2017.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 10/04/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sevoflurane has been shown to stimulate or depress memory in adult rats; however, the cellular mechanism of this bidirectional effect has not been fully investigated. METHODS We used an intra-hippocampal microinfusion of U0126 to suppress ERK activation. Male SD rats were randomly assigned to four groups: Sham, 0.11%SEV, 0.3%SEV and 0.3%+U0126. They received bilateral injections of U0126 or saline. Rats were anesthetized, and Inhibitory Avoidance (IA) training was performed immediately after anesthesia. The memory retention latency was observed 24 h later. In another experiment, the hippocampus was removed 45 min after IA training to assess ARC expression, the synapsin 1 protein levels and the phosphorylation level of ERK. RESULTS Treatment with 0.11%SEV led to rapid phosphorylation of ERK, while 0.3%SEV inhibited phosphorylation; the latter change was reversed by the microinfusion of U0126 in the hippocampus. The memory latency result had similar tendencies. The local infusion of U0126 abolished the 0.3%SEV-induced memory impairment and ERK inhibition. Selective upregulations of ARC and synapsin 1 proteins were observed in the 0.3%SEV group compared with the 0.11%SEV group. CONCLUSIONS The results indicate that different doses of sevoflurane trigger synaptic plasticity-related cytoskeleton proteins through the ERK signaling pathway. This novel modulation by inhalational agents may help to reduce their side-effects on memory function.
Collapse
Affiliation(s)
- Qian-Lin Zhu
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Sheng Xue
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Jun Zhang
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bu-Wei Yu
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Boison D. The Biochemistry and Epigenetics of Epilepsy: Focus on Adenosine and Glycine. Front Mol Neurosci 2016; 9:26. [PMID: 27147960 PMCID: PMC4829603 DOI: 10.3389/fnmol.2016.00026] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/31/2016] [Indexed: 12/14/2022] Open
Abstract
Epilepsy, one of the most prevalent neurological conditions, presents as a complex disorder of network homeostasis characterized by spontaneous non-provoked seizures and associated comorbidities. Currently used antiepileptic drugs have been designed to suppress neuronal hyperexcitability and thereby to suppress epileptic seizures. However, the current armamentarium of antiepileptic drugs is not effective in over 30% of patients, does not affect the comorbidities of epilepsy, and does not prevent the development and progression of epilepsy (epileptogenesis). Prevention of epilepsy and its progression remains the Holy Grail for epilepsy research and therapy development, requiring novel conceptual advances to find a solution to this urgent medical need. The methylation hypothesis of epileptogenesis suggests that changes in DNA methylation are implicated in the progression of the disease. In particular, global DNA hypermethylation appears to be associated with chronic epilepsy. Clinical as well as experimental evidence demonstrates that epilepsy and its progression can be prevented by biochemical manipulations and those that target previously unrecognized epigenetic functions contributing to epilepsy development and maintenance of the epileptic state. This mini-review will discuss, epigenetic mechanisms implicated in epileptogenesis and biochemical interactions between adenosine and glycine as a conceptual advance to understand the contribution of maladaptive changes in biochemistry as a major contributing factor to the development of epilepsy. New findings based on biochemical manipulation of the DNA methylome suggest that: (i) epigenetic mechanisms play a functional role in epileptogenesis; and (ii) therapeutic reconstruction of the epigenome is an effective antiepileptogenic therapy.
Collapse
Affiliation(s)
- Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute Portland, OR, USA
| |
Collapse
|
8
|
Boison D, Aronica E. Comorbidities in Neurology: Is adenosine the common link? Neuropharmacology 2015; 97:18-34. [PMID: 25979489 PMCID: PMC4537378 DOI: 10.1016/j.neuropharm.2015.04.031] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Abstract
Comorbidities in Neurology represent a major conceptual and therapeutic challenge. For example, temporal lobe epilepsy (TLE) is a syndrome comprised of epileptic seizures and comorbid symptoms including memory and psychiatric impairment, depression, and sleep dysfunction. Similarly, Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic Lateral Sclerosis (ALS) are accompanied by various degrees of memory dysfunction. Patients with AD have an increased likelihood for seizures, whereas all four conditions share certain aspects of psychosis, depression, and sleep dysfunction. This remarkable overlap suggests common pathophysiological mechanisms, which include synaptic dysfunction and synaptotoxicity, as well as glial activation and astrogliosis. Astrogliosis is linked to synapse function via the tripartite synapse, but astrocytes also control the availability of gliotransmitters and adenosine. Here we will specifically focus on the 'adenosine hypothesis of comorbidities' implying that astrocyte activation, via overexpression of adenosine kinase (ADK), induces a deficiency in the homeostatic tone of adenosine. We present evidence from patient-derived samples showing astrogliosis and overexpression of ADK as common pathological hallmark of epilepsy, AD, PD, and ALS. We discuss a transgenic 'comorbidity model', in which brain-wide overexpression of ADK and resulting adenosine deficiency produces a comorbid spectrum of seizures, altered dopaminergic function, attentional impairment, and deficits in cognitive domains and sleep regulation. We conclude that dysfunction of adenosine signaling is common in neurological conditions, that adenosine dysfunction can explain co-morbid phenotypes, and that therapeutic adenosine augmentation might be effective for the treatment of comorbid symptoms in multiple neurological conditions.
Collapse
Affiliation(s)
- Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA.
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands; Stichting Epilepsie Instellingen (SEIN) Nederland, Heemstede, The Netherlands
| |
Collapse
|
9
|
Mangubat EZ, Kellogg RG, Harris TJ, Rossi MA. On-demand pulsatile intracerebral delivery of carisbamate with closed-loop direct neurostimulation therapy in an electrically induced self-sustained focal-onset epilepsy rat model. J Neurosurg 2015; 122:1283-92. [PMID: 25723302 DOI: 10.3171/2015.1.jns14946] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECT The authors evaluated the preclinical feasibility of acutely stabilizing an active bihemispheric limbic epileptic circuit using closed-loop direct neurostimulation therapy in tandem with "on-demand'" convection-enhanced intracerebral delivery of the antiepileptic drug (AED) carisbamate. A rat model of electrically induced self-sustained focal-onset epilepsy was employed. METHODS A 16-contact depth-recording microelectrode was implanted bilaterally in the dentate gyrus (DG) of the hippocampus of Fischer 344 rats. The right microelectrode array included an integrated microcatheter for drug delivery at the distal tip. Bihemispheric spontaneous self-sustained limbic status epilepticus (SSLSE) was induced in freely moving rats using a 90-minute stimulation paradigm delivered to the right medial perforant white matter pathway. Immediately following SSLSE induction, closed-loop right PP stimulation therapy concurrent with on-demand nanoboluses of the AED [(14)C]-carisbamate (n = 4), or on-demand [(14)C]-carisbamate alone (n = 4), was introduced for a mean of 10 hours. In addition, 2 reference groups received either closed-loop stimulation therapy alone (n = 4) or stimulation therapy with saline vehicle only (n = 4). All animals were sacrificed after completing the specified therapy regimen. In situ [(14)C]-autoradiography was used to determine AED distribution. RESULTS Closed-loop direct stimulation therapy delivered unilaterally in the right PP aborted ictal runs detected in either ipsi- or contralateral hippocampi. Freely moving rats receiving closed-loop direct stimulation therapy with ondemand intracerebral carisbamate delivery experienced a significant reduction in seizure frequency (p < 0.001) and minimized seizure frequency variability during the final 50% of the therapy/recording session compared with closed-loop stimulation therapy alone. CONCLUSIONS Unilateral closed-loop direct stimulation therapy delivered to afferent hippocampal white matter pathways concurrent with on-demand ipsilateral intracerebral delivery of nano-bolused carisbamate can rapidly decrease the frequency of electrographic seizures in an active bihemispheric epileptic network. Additionally, direct pulsatile delivery of carisbamate can stabilize seizure frequency variability compared with direct stimulation therapy alone.
Collapse
Affiliation(s)
| | | | - Timothy J Harris
- 2Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Marvin A Rossi
- 2Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
10
|
Kazemzadeh-Narbat M, Annabi N, Tamayol A, Oklu R, Ghanem A, Khademhosseini A. Adenosine-associated delivery systems. J Drug Target 2015; 23:580-96. [PMID: 26453156 PMCID: PMC4863639 DOI: 10.3109/1061186x.2015.1058803] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Adenosine is a naturally occurring purine nucleoside in every cell. Many critical treatments such as modulating irregular heartbeat (arrhythmias), regulation of central nervous system (CNS) activity and inhibiting seizural episodes can be carried out using adenosine. Despite the significant potential therapeutic impact of adenosine and its derivatives, the severe side effects caused by their systemic administration have significantly limited their clinical use. In addition, due to adenosine's extremely short half-life in human blood (<10 s), there is an unmet need for sustained delivery systems to enhance efficacy and reduce side effects. In this article, various adenosine delivery techniques, including encapsulation into biodegradable polymers, cell-based delivery, implantable biomaterials and mechanical-based delivery systems, are critically reviewed and the existing challenges are highlighted.
Collapse
Affiliation(s)
- Mehdi Kazemzadeh-Narbat
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Nasim Annabi
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston 02115, MA, USA
| | - Ali Tamayol
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - Rahmi Oklu
- Massachusetts General Hospital, Harvard Medical School, Division of Interventional Radiology, Boston 02114, MA, USA
| | - Amyl Ghanem
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, MA, USA
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
11
|
Miranda MF, Hamani C, de Almeida ACG, Amorim BO, Macedo CE, Fernandes MJS, Nobrega JN, Aarão MC, Madureira AP, Rodrigues AM, Andersen ML, Tufik S, Mello LE, Covolan L. Role of adenosine in the antiepileptic effects of deep brain stimulation. Front Cell Neurosci 2014; 8:312. [PMID: 25324724 PMCID: PMC4183090 DOI: 10.3389/fncel.2014.00312] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022] Open
Abstract
Despite the effectiveness of anterior thalamic nucleus (AN) deep brain stimulation (DBS) for the treatment of epilepsy, mechanisms responsible for the antiepileptic effects of this therapy remain elusive. As adenosine modulates neuronal excitability and seizure activity in animal models, we hypothesized that this nucleoside could be one of the substrates involved in the effects of AN DBS. We applied 5 days of stimulation to rats rendered chronically epileptic by pilocarpine injections and recorded epileptiform activity in hippocampal slices. We found that slices from animals given DBS had reduced hippocampal excitability and were less susceptible to develop ictal activity. In live animals, AN DBS significantly increased adenosine levels in the hippocampus as measured by microdialysis. The reduced excitability of DBS in vitro was completely abolished in animals pre-treated with A1 receptor antagonists and was strongly potentiated by A1 receptor agonists. We conclude that some of the antiepileptic effects of DBS may be mediated by adenosine.
Collapse
Affiliation(s)
- Maisa F Miranda
- Laboratório de Neurociência Experimental e Computacional, Universidade Federal de São João del-Rei São João del-Rei, Brazil
| | - Clement Hamani
- Disciplina de Neurofisiologia, Universidade Federal de São Paulo São Paulo, Brazil ; Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health Toronto, Canada ; Division of Neurosurgery, Toronto Western Hospital, University of Toronto Toronto, Canada
| | - Antônio-Carlos G de Almeida
- Laboratório de Neurociência Experimental e Computacional, Universidade Federal de São João del-Rei São João del-Rei, Brazil
| | - Beatriz O Amorim
- Disciplina de Neurofisiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Carlos E Macedo
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Maria José S Fernandes
- Disciplina de Neurologia Experimental, Universidade Federal de São Paulo São Paulo, Brazil
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health Toronto, Canada
| | - Mayra C Aarão
- Laboratório de Neurociência Experimental e Computacional, Universidade Federal de São João del-Rei São João del-Rei, Brazil
| | - Ana Paula Madureira
- Laboratório de Neurociência Experimental e Computacional, Universidade Federal de São João del-Rei São João del-Rei, Brazil
| | - Antônio M Rodrigues
- Laboratório de Neurociência Experimental e Computacional, Universidade Federal de São João del-Rei São João del-Rei, Brazil
| | - Monica L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Luiz E Mello
- Disciplina de Neurofisiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Luciene Covolan
- Disciplina de Neurofisiologia, Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
12
|
Ramgopal S, Thome-Souza S, Jackson M, Kadish NE, Sánchez Fernández I, Klehm J, Bosl W, Reinsberger C, Schachter S, Loddenkemper T. Seizure detection, seizure prediction, and closed-loop warning systems in epilepsy. Epilepsy Behav 2014; 37:291-307. [PMID: 25174001 DOI: 10.1016/j.yebeh.2014.06.023] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/04/2014] [Accepted: 06/10/2014] [Indexed: 12/16/2022]
Abstract
Nearly one-third of patients with epilepsy continue to have seizures despite optimal medication management. Systems employed to detect seizures may have the potential to improve outcomes in these patients by allowing more tailored therapies and might, additionally, have a role in accident and SUDEP prevention. Automated seizure detection and prediction require algorithms which employ feature computation and subsequent classification. Over the last few decades, methods have been developed to detect seizures utilizing scalp and intracranial EEG, electrocardiography, accelerometry and motion sensors, electrodermal activity, and audio/video captures. To date, it is unclear which combination of detection technologies yields the best results, and approaches may ultimately need to be individualized. This review presents an overview of seizure detection and related prediction methods and discusses their potential uses in closed-loop warning systems in epilepsy.
Collapse
Affiliation(s)
- Sriram Ramgopal
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sigride Thome-Souza
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Psychiatry Department of Clinics Hospital of School of Medicine of University of Sao Paulo, Brazil
| | - Michele Jackson
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Navah Ester Kadish
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neuropediatrics and Department of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Iván Sánchez Fernández
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Jacquelyn Klehm
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - William Bosl
- Department of Health Informatics, University of San Francisco School of Nursing and Health Professions, San Francisco, CA, USA
| | - Claus Reinsberger
- Edward B. Bromfield Epilepsy Center, Dept. of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Institute of Sports Medicine, Department of Exercise and Health, Faculty of Science, University of Paderborn, Germany; Institute of Sports Medicine, Faculty of Science, University of Paderborn, Warburger Str. 100, 33098 Paderborn, Germany
| | - Steven Schachter
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Tobias Loddenkemper
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Coelho JE, Alves P, Canas PM, Valadas JS, Shmidt T, Batalha VL, Ferreira DG, Ribeiro JA, Bader M, Cunha RA, do Couto FS, Lopes LV. Overexpression of Adenosine A2A Receptors in Rats: Effects on Depression, Locomotion, and Anxiety. Front Psychiatry 2014; 5:67. [PMID: 24982640 PMCID: PMC4055866 DOI: 10.3389/fpsyt.2014.00067] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 05/24/2014] [Indexed: 11/25/2022] Open
Abstract
Adenosine A2A receptors (A2AR) are a sub-type of receptors enriched in basal ganglia, activated by the neuromodulator adenosine, which interact with dopamine D2 receptors. Although this reciprocal antagonistic interaction is well-established in motor function, the outcome in dopamine-related behaviors remains uncertain, in particular in depression and anxiety. We have demonstrated an upsurge of A2AR associated to aging and chronic stress. Furthermore, Alzheimer's disease patients present A2AR accumulation in cortical areas together with depressive signs. We now tested the impact of overexpressing A2AR in forebrain neurons on dopamine-related behavior, namely depression. Adult male rats overexpressing human A2AR under the control of CaMKII promoter [Tg(CaMKII-hA2AR)] and aged-matched wild-types (WT) of the same strain (Sprague-Dawley) were studied. The forced swimming test (FST), sucrose preference test (SPT), and the open-field test (OFT) were performed to evaluate behavioral despair, anhedonia, locomotion, and anxiety. Tg(CaMKII-hA2AR) animals spent more time floating and less time swimming in the FST and presented a decreased sucrose preference at 48 h in the SPT. They also covered higher distances in the OFT and spent more time in the central zone than the WT. The results indicate that Tg(CaMKII-hA2AR) rats exhibit depressive-like behavior, hyperlocomotion, and altered exploratory behavior. This A2AR overexpression may explain the depressive signs found in aging, chronic stress, and Alzheimer's disease.
Collapse
Affiliation(s)
- Joana E Coelho
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Pedro Alves
- Faculty of Medicine of Lisbon, Institute of Pharmacology and Neurosciences, University of Lisbon , Lisbon , Portugal
| | - Paula M Canas
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra , Coimbra , Portugal ; Faculty of Medicine, University of Coimbra , Coimbra , Portugal
| | - Jorge S Valadas
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Tatiana Shmidt
- Max-Delbrück-Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Vânia L Batalha
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Diana G Ferreira
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Joaquim A Ribeiro
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal ; Faculty of Medicine of Lisbon, Institute of Pharmacology and Neurosciences, University of Lisbon , Lisbon , Portugal
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Rodrigo A Cunha
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra , Coimbra , Portugal ; Faculty of Medicine, University of Coimbra , Coimbra , Portugal
| | - Frederico Simões do Couto
- Faculty of Medicine of Lisbon, Institute of Pharmacology and Neurosciences, University of Lisbon , Lisbon , Portugal
| | - Luísa V Lopes
- Faculty of Medicine of Lisbon, Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| |
Collapse
|
14
|
Cross JH, Kluger G, Lagae L. Advancing the management of childhood epilepsies. Eur J Paediatr Neurol 2013; 17:334-47. [PMID: 23558251 DOI: 10.1016/j.ejpn.2013.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 02/20/2013] [Accepted: 02/27/2013] [Indexed: 12/27/2022]
Abstract
Childhood epilepsies comprise a heterogeneous group of disorders and syndromes that vary in terms of severity, prognosis and treatment requirements. Effective management requires early, accurate recognition and diagnosis, and a holistic approach that addresses each individual's medical and psychosocial needs within the context of their overall health status and quality of life. With increasing understanding of underlying aetiologies, new approaches to management and treatment are emerging. For example, genetic testing is beginning to provide a tool to aid differential diagnosis and a means of predicting predisposition to particular types of epilepsy. Despite the availability of an increasing number of antiepileptic drugs (AEDs)--due not only to the development of new AEDs, but also to changes in regulatory requirements that have facilitated clinical development--seizure control and tolerability continue to be suboptimal in many patients, and there is therefore a continuing need for new treatment strategies. Surgery and other non-pharmacological treatments (e.g. vagus nerve stimulation, ketogenic diet) are already relatively well established in paediatric epilepsy. New pharmacological treatments include generational advances on existing AEDs and AEDs with novel modes of action, and non-AED pharmacological interventions, such as immunomodulation. Emerging technologies include novel approaches allowing the delivery of medicinal agents to specific areas of the brain, and 'closed-loop' experimental devices employing algorithms that allow treatment (e.g., electrical stimulation) to be targeted both spatially and temporally. Although in early stages of development, cell-based approaches (e.g., focal targeting of adenosine augmentation) and gene therapy may also provide new treatment choices in the future.
Collapse
Affiliation(s)
- J Helen Cross
- UCL-Institute of Child Health, Great Ormond Street Hospital for Children NHS Foundation Trust, London.
| | | | | |
Collapse
|
15
|
Rocha L. Interaction between electrical modulation of the brain and pharmacotherapy to control pharmacoresistant epilepsy. Pharmacol Ther 2013; 138:211-28. [DOI: 10.1016/j.pharmthera.2013.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 01/07/2013] [Indexed: 12/15/2022]
|
16
|
Abstract
Approximately one-third of patients with epilepsy continue to have seizures despite antiepileptic therapy. Many seizures occur in diurnal, sleep/wake, circadian, or even monthly patterns. The relationship between biomarkers and state changes is still being investigated, but early results suggest that some of these patterns may be related to endogenous circadian patterns whereas others may be related to wakefulness and sleep or both. Chronotherapy, the application of treatment at times of greatest seizure susceptibility, is a technique that may optimize seizure control in selected patients. It may be used in the form of differential dosing, as preparations designed to deliver sustained or pulsatile drug delivery or in the form of 'zeitgebers' that shift endogenous rhythms. Early trials in epilepsy suggest that chronopharmacology may provide improved seizure control compared with conventional treatment in some patients. The present article reviews chronopharmacology in the treatment of epilepsy as well as future treatment avenues.
Collapse
Affiliation(s)
- Sriram Ramgopal
- Division of Epilepsy and Clinical Neurophysiology, Harvard Medical School, Fegan 9, Boston, MA USA
- Department of Neurology, Children’s Hospital Boston, 300 Longwood Ave, Boston, MA 02115 USA
| | - Sigride Thome-Souza
- Division of Epilepsy and Clinical Neurophysiology, Harvard Medical School, Fegan 9, Boston, MA USA
- Department of Neurology, Children’s Hospital Boston, 300 Longwood Ave, Boston, MA 02115 USA
- Psychiatry Department of Clinics Hospital of Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Tobias Loddenkemper
- Division of Epilepsy and Clinical Neurophysiology, Harvard Medical School, Fegan 9, Boston, MA USA
- Department of Neurology, Children’s Hospital Boston, 300 Longwood Ave, Boston, MA 02115 USA
| |
Collapse
|
17
|
|
18
|
Kang H, Hu Q, Liu X, Liu Y, Xu F, Li X, Zhu S. Reconstruction of the adenosine system by bone marrow-derived mesenchymal stem cell transplantation. Neural Regen Res 2012; 7:251-5. [PMID: 25806064 PMCID: PMC4353095 DOI: 10.3969/j.issn.1673-5374.2012.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/27/2011] [Indexed: 11/18/2022] Open
Abstract
In the present study, we transplanted bone marrow-derived mesenchymal stem cells into the CA3 area of the hippocampus of chronic epilepsy rats kindled by lithium chloride-pilocarpine. Immunofluorescence and western blotting revealed an increase in adenosine A1 receptor expression and a decrease in adenosine A2a receptor expression in the brain tissues of epileptic rats 3 months after transplantation. Moreover, the imbalance in the A1 adenosine receptor/A2a adenosine receptor ratio was improved. Electroencephalograms showed that frequency and amplitude of spikes in the hippocampus and frontal lobe were reduced. These results suggested that mesenchymal stem cell transplantation can reconstruct the normal function of the adenosine system in the brain and greatly improve epileptiform discharges.
Collapse
Affiliation(s)
- Huicong Kang
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Qi Hu
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Xiaoyan Liu
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yinhe Liu
- Ministry of Water Resources of China, Beijing 100000, China
| | - Feng Xu
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Xiang Li
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Suiqiang Zhu
- Department of Neurology, Tongji Hospital of Tongji Medical University, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
19
|
Hamil NE, Cock HR, Walker MC. Acute down-regulation of adenosine A1 receptor activity in status epilepticus. Epilepsia 2011; 53:177-88. [DOI: 10.1111/j.1528-1167.2011.03340.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Updating hippocampal representations: CA2 joins the circuit. Trends Neurosci 2011; 34:526-35. [DOI: 10.1016/j.tins.2011.07.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/13/2011] [Accepted: 07/25/2011] [Indexed: 12/20/2022]
|
21
|
Siebel AM, Piato AL, Capiotti KM, Seibt KJ, Bogo MR, Bonan CD. PTZ-induced seizures inhibit adenosine deamination in adult zebrafish brain membranes. Brain Res Bull 2011; 86:385-9. [PMID: 21907764 DOI: 10.1016/j.brainresbull.2011.08.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 08/10/2011] [Accepted: 08/29/2011] [Indexed: 12/20/2022]
Abstract
Adenosine exerts neuromodulatory functions with mostly inhibitory effects, being considered an endogenous anticonvulsant. The hydrolysis of ATP by ectonucleotidases is an important source of adenosine, and adenosine deaminase (ADA) contributes to the regulation of this nucleoside concentration through its deamination. In this study, we tested the effect of pentylenetetrazole (PTZ)-induced seizures on ectonucleotidase and ADA activities in adult zebrafish brain. Our results have demonstrated that PTZ treatments did not alter ectonucleotidase and ADA activities in membranes and soluble fraction, respectively. However, ecto-ADA activity was significantly decreased in brain membranes of animals exposed to 5mM and 15 mM PTZ treatments (22.4% and 29.5%, respectively) when compared to the control group. Semiquantitative RT-PCR analysis did not show significant changes after the PTZ exposure on ADA gene expression. The decreased adenosine deamination observed in this study suggests a modulation of extracellular adenosine levels during PTZ-induced seizures in zebrafish.
Collapse
Affiliation(s)
- Anna Maria Siebel
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil
| | | | | | | | | | | |
Collapse
|
22
|
Rico EP, Rosemberg DB, Seibt KJ, Capiotti KM, Da Silva RS, Bonan CD. Zebrafish neurotransmitter systems as potential pharmacological and toxicological targets. Neurotoxicol Teratol 2011; 33:608-17. [PMID: 21907791 DOI: 10.1016/j.ntt.2011.07.007] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/20/2011] [Accepted: 07/20/2011] [Indexed: 01/23/2023]
Abstract
Recent advances in neurobiology have emphasized the study of brain structure and function and its association with numerous pathological and toxicological events. Neurotransmitters are substances that relay, amplify, and modulate electrical signals between neurons and other cells. Neurotransmitter signaling mediates rapid intercellular communication by interacting with cell surface receptors, activating second messenger systems and regulating the activity of ion channels. Changes in the functional balance of neurotransmitters have been implicated in the failure of central nervous system function. In addition, abnormalities in neurotransmitter production or functioning can be induced by several toxicological compounds, many of which are found in the environment. The zebrafish has been increasingly used as an animal model for biomedical research, primarily due to its genetic tractability and ease of maintenance. These features make this species a versatile tool for pre-clinical drug discovery and toxicological investigations. Here, we present a review regarding the role of different excitatory and inhibitory neurotransmitter systems in zebrafish, such as dopaminergic, serotoninergic, cholinergic, purinergic, histaminergic, nitrergic, glutamatergic, glycinergic, and GABAergic systems, and emphasizing their features as pharmacological and toxicological targets. The increase in the global knowledge of neurotransmitter systems in zebrafish and the elucidation of their pharmacological and toxicological aspects may lead to new strategies and appropriate research priorities to offer insights for biomedical and environmental research.
Collapse
Affiliation(s)
- E P Rico
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, Brazil
| | | | | | | | | | | |
Collapse
|
23
|
|
24
|
Van Dycke A, Raedt R, Verstraete A, Theofilas P, Wadman W, Vonck K, Boison D, Boon P. Astrocytes derived from fetal neural progenitor cells as a novel source for therapeutic adenosine delivery. Seizure 2010; 19:390-6. [PMID: 20688264 DOI: 10.1016/j.seizure.2010.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/08/2010] [Accepted: 05/21/2010] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Intracerebral delivery of anti-epileptic compounds represents a novel strategy for the treatment of refractory epilepsy. Adenosine is a possible candidate for local delivery based on its proven anti-epileptic effects. Neural stem cells constitute an ideal cell source for intracerebral transplantation and long-term drug delivery. In order to develop a cell-based system for the long-term delivery of adenosine, we isolated neural progenitor cells from adenosine kinase deficient mice (Adk(-/-)) and compared their differentiation potential and adenosine release properties with corresponding wild-type cells. METHODS Fetal neural progenitor cells were isolated from the brains of Adk(-/-) and C57BL/6 mice fetuses and expanded in vitro. Before and after neural differentiation, supernatants were collected and assayed for adenosine release using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS Adk(-/-) cells secreted significantly more adenosine compared to wild-type cells at any time point of differentiation. Undifferentiated Adk(-/-) cells secreted 137+/-5 ng adenosine per 10(5) cells during 24 h in culture, compared to 11+/-1 ng released from corresponding wild-type cells. Adenosine release was maintained after differentiation as differentiated Adk(-/-) cells continued to release significantly more adenosine per 24 h (47+/-1 ng per 10(5) cells) compared to wild-type cells (3+/-0.2 ng per 10(5) cells). CONCLUSIONS Fetal neural progenitor cells isolated from Adk(-/-) mice--but not those from C57BL/6 mice--release amounts of adenosine considered to be of therapeutic relevance.
Collapse
Affiliation(s)
- Annelies Van Dycke
- Laboratory for Clinical and Experimental Neurophysiology, Department of Neurology, Ghent University Hospital, 1K12, 185 De Pintelaan, 9000 Ghent, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|