Elsevier

Epilepsy Research

Volume 85, Issues 2–3, August 2009, Pages 131-141
Epilepsy Research

Review
Adenosine augmentation therapies (AATs) for epilepsy: Prospect of cell and gene therapies

https://doi.org/10.1016/j.eplepsyres.2009.03.019Get rights and content

Summary

Deficiencies in the brain's own adenosine-based seizure control system contribute to seizure generation. Consequently, reconstitution of adenosinergic neuromodulation constitutes a rational approach for seizure control. This review will critically discuss focal adenosine augmentation strategies and their potential for antiepileptic and disease modifying therapy. Due to systemic side effects of adenosine focal adenosine augmentation – ideally targeted to an epileptic focus – becomes a therapeutic necessity. This has experimentally been achieved in kindled seizure models as well as in post-status epilepticus models of spontaneous recurrent seizures using three different therapeutic strategies that will be discussed here: (i) polymer-based brain implants that were loaded with adenosine; (ii) brain implants comprised of cells engineered to release adenosine and embedded in a cell-encapsulation device; (iii) direct transplantation of stem cells engineered to release adenosine. To meet the therapeutic goal of focal adenosine augmentation, genetic disruption of the adenosine metabolizing enzyme adenosine kinase (ADK) in rodent and human cells was used as a molecular strategy to induce adenosine release from cellular brain implants, which demonstrated antiepileptic and neuroprotective properties. New developments and therapeutic challenges in using AATs for epilepsy therapy will critically be evaluated.

Introduction

The key roles of purines in neurotransmission and neuromodulation were first recognized by Burnstock in 1972 with the identification of 5′-adenosine-triphosphate (ATP) as a novel neurotransmitter, a finding that led to the concept of purinergic neurotransmission (Burnstock, 1972). Subsequently, the release of the endogenous purine ribonucleoside adenosine, a degradation product of ATP, was shown to regulate hippocampal excitability in vitro (Dunwiddie, 1980, Dunwiddie and Hoffer, 1980). A few years later it was demonstrated that adenosine and its analogues modulated amygdala kindling in rats and adenosine was proposed to be the brain's endogenous anticonvulsant (Dragunow and Goddard, 1984, Dragunow et al., 1985, Dragunow, 1986). The crucial role of adenosinergic neuromodulation in the control of seizure activity is now well established and has recently been reviewed (Boison, 2005). In addition, adenosine is involved in one of several endogenous mechanisms of the brain that have evolved to terminate seizures (Lado and Moshe, 2008).

Adenosine exerts its neuromodulatory functions by binding to four known adenosine receptor subtypes (A1R, A2AR, A2BR, A3R) that all belong to the family of seven-membrane-spanning G-protein coupled receptors (Fredholm et al., 2001, Fredholm et al., 2005, Fredholm et al., 2007). Binding of adenosine to the high affinity A1R, which is prominently expressed at pre- and postsynaptic sites within the hippocampal formation, leads to decreased neuronal transmission and reduced excitability that are largely based on inhibition of presynaptic transmitter release and stabilization of the postsynaptic membrane potential through increased potassium efflux via G protein-coupled inwardly rectifying potassium (GIRK) channels (Sebastiao and Ribeiro, 2000). The A1R-mediated functions are largely responsible for the anticonvulsant and neuroprotective activity of adenosine. Thus, A1R knockout mice experience spontaneous hippocampal seizures (Li et al., 2007a) and are hypersensitive to status epilepticus- or trauma-induced brain injury (Fedele et al., 2006, Kochanek et al., 2006). While the A1R is thought to set a global inhibitory environment within the brain and to provide heterosynaptic depression, the stimulatory A2AR on postsynaptic locations is thought to be preferentially activated by high frequency stimulation and thus is ideally suited to potentiate selected synaptic transmission within a globally inhibited network (Cunha, 2008). In contrast to the well characterized role of the A1R in epilepsy, A2A receptor activation in epilepsy appears to have both proconvulsant as well as anticonvulsant characteristics depending on the context of activation (Boison, 2005, Boison, 2007b). Whereas A1Rs and A2ARs are primarily responsible for the central effects of adenosine (Ribeiro et al., 2003), the low affinity and low abundance A2BRs and A3Rs are currently not considered as therapeutic targets for epilepsy (Boison, 2005, Boison, 2007b). Functional receptor–receptor interactions of A1Rs and different types of metabotropic and ionotropic receptors allow a further complexity in adenosinergic neuromodulation (Sichardt and Nieber, 2007).

Synaptic levels of adenosine in adult brain are largely regulated by an astrocyte-based adenosine-cycle (Boison, 2008c), and conversely, adenosine plays important roles for astrocyte physiology (Bjorklund et al., 2008). Synaptic adenosine largely originates from extracellular breakdown of ATP (Dunwiddie et al., 1997, Ziganshin et al., 1994, Zimmermann, 2000), which in turn is derived from vesicular release from astrocytes or neurons (Fields and Burnstock, 2006, Halassa et al., 2007, Haydon and Carmignoto, 2006, Pascual et al., 2005). Alternatively, adenosine as such can directly be released from astrocytes (Frenguelli et al., 2007, Martin et al., 2007). Under physiological conditions, extra- and intracellular levels of adenosine are rapidly equilibrated via distinct families of nucleoside transporters (Baldwin et al., 2004, Gray et al., 2004). Intracellularly, adenosine is rapidly phosphorylated into 5′-adenosine-monophosphate (AMP) via adenosine kinase (ADK; EC 2.7.1.20), an evolutionary conserved member of the ribokinase family of proteins (Park and Gupta, 2008). Due to the high metabolic activity of ADK and the existence of equilibrative transport systems for adenosine, synaptic levels of adenosine are thought to be controlled by intracellular metabolism of adenosine via ADK that assumes the role of a metabolic reuptake system for adenosine; in contrast to classical neurotransmitters, which all have their specific re-uptake transporters, a comparable transporter-controlled re-uptake system for adenosine appears to be lacking (Boison, 2006). It is important to note that in adult brain ADK is almost exclusively expressed in astrocytes (Studer et al., 2006).

Section snippets

Adenosine augmentation: therapeutic rationale

Based on the failure of traditional neuron-centered pharmacotherapy in about one third of patients with epilepsy, the exploitation of non-neuronal and non-chemical synaptic signalling pathways may offer alternatives for epilepsy therapy (Szente, 2008). Several lines of evidence suggest that astrocyte dysfunction and deficiencies in endogenous adenosinergic neuromodulation contribute to seizure generation. In healthy adult brain, physiological adenosine concentrations (25–250 nM) are kept in the

Polymer-based drug delivery

The prolonged focal delivery of adenosine or other small molecule drugs can be achieved by including the drug within a biocompatible polymer. The usefulness of focal brain implants of biocompatible polymers in epilepsy therapy has been evaluated in only a few experimental paradigms. As an example, intracerebral implants of GABA-, but not of noradrenaline-releasing polymer matrices manufactured from ethylene vinyl acetate copolymers (EVAc) were shown to suppress seizures in kindled rats (Kokaia

Encapsulated cell systems

In contrast to polymer-based drug delivery, encapsulated cell biodelivery (ECB), first developed by Aebischer and colleagues in 1991 (Aebischer et al., 1991, Winn et al., 1991) and reviewed recently (Hauser et al., 2004, Tseng and Aebischer, 2000), constitutes an ex vivo gene therapy approach. In ECB approaches a cell line is first genetically modified to produce and release a therapeutic molecule of interest (e.g. adenosine). In a second step the cells are then enclosed within an ECB device,

Stem cells

Stem cells, stem cell-derived neural precursors, neuronal cell lines, and fetal hippocampal neurons have recently received much attention as direct transplantation tools for epilepsy therapy (Boison, 2007c, Loscher et al., 2008, Raedt et al., 2007, Shetty and Hattiangady, 2007, Suter and Krause, 2008). In general, two different therapeutic strategies are possible that may synergistically augment each other (Fig. 1): the first strategy is based on reconstitution of hippocampal circuitry by

Conclusions and outlook

Based on the demonstrated anticonvulsant efficacy of intrafocal polymer and/or cell mediated adenosine release and the fact that adenosine is able to suppress pharmacoresistant seizures, the foundation is laid to move this novel therapeutic approach forwards towards clinical applications. The clinical target is pharmacoresistant temporal lobe epilepsy (TLE). Prior to clinical application of adenosine-releasing devices the following requirements must be considered: (a) refined

Acknowledgments

The work of the author is supported by grants RO1NS058780-01, R21NS057475-01, and R21NS057538-01 from the National Institute of Neurological Disorders and Stroke (NINDS), the Good Samaritan Hospital Foundation, the Epilepsy Research Foundation through the generous support of Arlene & Arnold Goldstein Family Foundation, and Citizens United in Research against Epilepsy (CURE) in collaboration with the Department of Defense (DoD).

References (126)

  • G. Buzsaki et al.

    Hippocampal grafts into the intact brain induce epileptic patterns

    Brain Res.

    (1991)
  • K. Chu et al.

    Human neural stem cells can migrate, differentiate, and integrate after intravenous transplantation in adult rats with transient forebrain ischemia

    Neurosci. Lett.

    (2003)
  • K. Chu et al.

    Human neural stem cell transplantation reduces spontaneous recurrent seizures following pilocarpine-induced status epilepticus in adult rats

    Brain Res.

    (2004)
  • R.A. Cunha

    Different cellular sources and different roles of adenosine: A(1) receptor-mediated inhibition through astrocytic-driven volume transmission and synapse-restricted A(2A) receptor-mediated facilitation of plasticity

    Neurochem. Int.

    (2008)
  • N. Déglon et al.

    Continuous delivery of erythropoietin in mice using encapsulated genetically engineered cell lines

    Cell Transplant.

    (1996)
  • M. Dragunow et al.

    Adenosine modulation of amygdala kindling

    Exp. Neurol.

    (1984)
  • M. Dragunow

    Adenosine: the brain's natural anticonvulsant?

    Trends Pharmacol. Sci.

    (1986)
  • M. Fatar et al.

    Lipoaspirate-derived adult mesenchymal stem cells improve functional outcome during intracerebral hemorrhage by proliferation of endogenous progenitor cells. Stem cells in intracerebral hemorrhages

    Neurosci. Lett.

    (2008)
  • D.E. Fedele et al.

    Engineering embryonic stem cell derived glia for adenosine delivery

    Neurosci. Lett.

    (2004)
  • D.E. Fedele et al.

    Adenosine A1 receptors are crucial in keeping an epileptic focus localized

    Exp. Neurol.

    (2006)
  • K.J.L. Fernandes et al.

    Analysis of the neurogenic potential of multipotent skin-derived precursors

    Exp. Neurol.

    (2006)
  • A. Freese et al.

    Controlled release of dopamine from a polymeric brain implant: in vitro characterization

    Exp. Neurol.

    (1989)
  • M. Glass et al.

    Loss of A1 adenosine receptors in human temporal lobe epilepsy

    Brain Res.

    (1996)
  • M. Güttinger et al.

    Seizure suppression and lack of adenosine A1 receptor desensitization after focal long-term delivery of adenosine by encapsulated myoblasts

    Exp. Neurol.

    (2005)
  • M.M. Halassa et al.

    The tripartite synapse: roles for gliotransmission in health and disease

    Trends Mol. Med.

    (2007)
  • D. Hoffman et al.

    NGF released from a polymer matrix prevents loss of ChAT expression in basal forebrain neurons following a fimbria-fornix lesion

    Exp. Neurol.

    (1990)
  • S. Hofmann et al.

    Silk fibroin as an organic polymer for controlled drug delivery

    J. Control. Release

    (2006)
  • R.L. Horan et al.

    In vitro degradation of silk fibroin

    Biomaterials

    (2005)
  • M.F. Jarvis et al.

    Comparison of the ability of adenosine kinase inhibitors and adenosine receptor agonists to attenuate thermal hyperalgesia and reduce motor performance in rats

    Pharmacol. Biochem. Behav.

    (2002)
  • I. Kanter-Schlifke et al.

    GDNF released from encapsulated cells suppresses seizure activity in the epileptic hippocampus

    Exp. Neurol.

    (2009)
  • H. Kishima et al.

    Encapsulated GDNF-producing C2C12 cells for Parkinson's disease: a pre-clinical study in chronic MPTP-treated baboons

    Neurobiol. Dis.

    (2004)
  • C. Li et al.

    Electrospun silk-BMP-2 scaffolds for bone tissue engineering

    Biomaterials

    (2006)
  • Z. Li et al.

    Formulation of spray-dried phenytoin loaded poly(epsilon-caprolactone) microcarrier intended for brain delivery to treat epilepsy

    J. Pharm. Sci.

    (2007)
  • O. Lindvall et al.

    Encapsulated cell biodelivery of GDNF: a novel clinical strategy for neuroprotection and neuroregeneration in Parkinson's disease?

    Exp. Neurol.

    (2008)
  • W. Loscher et al.

    Cell and gene therapies in epilepsy—promising avenues or blind alleys?

    Trends Neurosci.

    (2008)
  • K.E. Nilsen et al.

    Focal treatment for refractory epilepsy: hope for the future?

    Brain Res. Brain Res. Rev.

    (2004)
  • S. Okabe et al.

    Development of neuronal precursor cells and functional postmitotic neurons from embryonic stem cells in vitro

    Mech. Dev.

    (1996)
  • P. Aebischer et al.

    Transplantation in humans of encapsulated xenogeneic cells without immunosuppression. A preliminary report

    Transplant

    (1994)
  • P. Aebischer et al.

    Gene therapy for amyotrophic lateral sclerosis (ALS) using a polymer encapsulated xenogenic cell line engineered to secrete hCNTF

    Hum. Gene Ther.

    (1996)
  • S.A. Baldwin et al.

    The equilibrative nucleoside transporter family, SLC29

    Pflugers Arch.

    (2004)
  • D. Boison et al.

    Seizure suppression by adenosine-releasing cells is independent of seizure frequency

    Epilepsia

    (2002)
  • D. Boison et al.

    Neonatal hepatic steatosis by disruption of the adenosine kinase gene

    Proc. Natl. Acad. Sci. U.S.A.

    (2002)
  • D. Boison

    Adenosine and epilepsy: from therapeutic rationale to new therapeutic strategies

    Neuroscientist

    (2005)
  • D. Boison

    Adenosine, astrogliosis and seizures: a new perspective of epileptogenesis

    Neuron Glia Biol.

    (2007)
  • D. Boison

    Adenosine as a modulator of brain activity

    Drug News Persp.

    (2007)
  • D. Boison

    Cell and gene therapies for refractory epilepsy

    Curr. Neuropharmacol.

    (2007)
  • D. Boison

    Astrogliosis and adenosine kinase: a glial basis of epilepsy

    Future Neurol.

    (2008)
  • G. Burnstock

    Purinergic nerves

    Pharmacol. Rev.

    (1972)
  • G. Buzsaki et al.

    Suppression and induction of epileptic activity by neuronal grafts

    Proc. Natl. Acad. Sci. U.S.A.

    (1988)
  • B. Dalle et al.

    Improvement of mouse beta-thalassemia upon erythropoietin delivery by encapsulated myoblasts

    Gene Ther.

    (1999)
  • Cited by (68)

    • Impairment of adenosinergic system in Rett syndrome: Novel therapeutic target to boost BDNF signalling

      2020, Neurobiology of Disease
      Citation Excerpt :

      Low intracellular adenosine levels may favour DNA methylation and hence epileptogenesis (Williams-Karnesky et al., 2013). Consequently, adenosine augmentation therapies constitute an effective strategy to reduce seizures, even in cases refractory to conventional antiepileptic drugs (Boison, 2013, 2009). In contrast with A1Rs, the protein levels of A2AR were decreased in cortex of Mecp2−/y mice.

    • The role of adenosine in epilepsy

      2019, Brain Research Bulletin
    View all citing articles on Scopus
    View full text