Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 118(2): 571-82, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18172552

ABSTRACT

Astrogliosis is a pathological hallmark of the epileptic brain. The identification of mechanisms that link astrogliosis to neuronal dysfunction in epilepsy may provide new avenues for therapeutic intervention. Here we show that astrocyte-expressed adenosine kinase (ADK), a key negative regulator of the brain inhibitory molecule adenosine, is a potential predictor and modulator of epileptogenesis. In a mouse model of focal epileptogenesis, in which astrogliosis is restricted to the CA3 region of the hippocampus, we demonstrate that upregulation of ADK and spontaneous focal electroencephalographic seizures were both restricted to the affected CA3. Furthermore, spontaneous seizures in CA3 were mimicked in transgenic mice by overexpression of ADK in this brain region, implying that overexpression of ADK without astrogliosis is sufficient to cause seizures. Conversely, after pharmacological induction of an otherwise epileptogenesis-precipitating acute brain injury, transgenic mice with reduced forebrain ADK were resistant to subsequent epileptogenesis. Likewise, ADK-deficient ES cell-derived brain implants suppressed astrogliosis, upregulation of ADK, and spontaneous seizures in WT mice when implanted after the epileptogenesis-precipitating brain injury. Our findings suggest that astrocyte-based ADK provides a critical link between astrogliosis and neuronal dysfunction in epilepsy.


Subject(s)
Adenosine Kinase/metabolism , Epilepsies, Partial/genetics , Seizures/genetics , Adenosine Kinase/analysis , Adenosine Kinase/genetics , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/pathology , Brain/drug effects , Brain/enzymology , Brain/pathology , Epilepsies, Partial/chemically induced , Epilepsies, Partial/pathology , Kainic Acid/toxicity , Male , Mice , Mice, Transgenic , Prognosis , Seizures/chemically induced , Seizures/pathology
2.
Brain ; 130(Pt 5): 1276-88, 2007 May.
Article in English | MEDLINE | ID: mdl-17472985

ABSTRACT

Epilepsy therapy is largely symptomatic and no effective therapy is available to prevent epileptogenesis. We therefore analysed the potential of stem cell-derived brain implants and of paracrine adenosine release to suppress the progressive development of seizures in the rat kindling-model. Embryonic stem (ES) cells, engineered to release the inhibitory neuromodulator adenosine by biallelic genetic disruption of the adenosine kinase gene (Adk-/-), and respective wild-type (wt) cells, were differentiated into neural precursor cells (NPs) and injected into the hippocampus of rats prior to kindling. Therapeutic effects of NP-derived brain implants were compared with those of wt baby hamster kidney cells (BHK) and adenosine releasing BHK cell implants (BHK-AK2), which were previously shown to suppress seizures by paracrine adenosine release. Wild-type NP-graft recipients were characterized by an initial delay of seizure development, while recipients of adenosine releasing NPs displayed sustained protection from developing generalized seizures. In contrast, recipients of wt BHK cells failed to display any effects on kindling development, while recipients of BHK-AK2 cells were only moderately protected from seizure development. The therapeutic effect of Adk(-/-)-NPs was due to graft-mediated adenosine release, since seizures could transiently be provoked after blocking adenosine A1 receptors. Histological analysis of NP-implants at day 26 revealed cell clusters within the infrahippocampal cleft as well as intrahippocampal location of graft-derived cells expressing mature neuronal markers. In contrast, BHK and BHK-AK2 cell implants only formed cell clusters within the infrahippocampal cleft. We conclude that ES cell-derived adenosine releasing brain implants are superior to paracrine adenosine release from BHK-AK2 cell implants in suppressing seizure progression in the rat kindling-model. These findings may indicate a potential antiepileptogenic function of stem cell-mediated adenosine delivery.


Subject(s)
Adenosine/metabolism , Brain/surgery , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Epilepsy/surgery , Kindling, Neurologic/metabolism , Adenosine Kinase/metabolism , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Epilepsy/metabolism , Epilepsy/pathology , Genetic Engineering , Kindling, Neurologic/pathology , Male , Models, Animal , Paracrine Communication , Rats , Rats, Sprague-Dawley
3.
Exp Neurol ; 200(1): 184-90, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16750195

ABSTRACT

Adenosine is an endogenous neuromodulator with anticonvulsant and neuroprotective properties presumably mediated by activation of adenosine A1 receptors (A1Rs). To study the involvement of A1Rs in neuroprotection during epileptogenesis, we induced status epilepticus by a unilateral intrahippocampal kainic acid (KA) injection (1 nmol) in wild-type C57BL/6 and homozygous adenosine A1R knock out (A1R-KO) mice of the same genetic background. Whereas the KA injection caused non-convulsive status epilepticus in wild-type mice, in A1R-KO mice KA induced status epilepticus with severe convulsions and subsequent death of the animals within 5 days. 24 h after KA injection, brains from wild-type C57BL/6 mice were characterized by slight neuronal cell loss confined to the immediate location of the KA injection. In contrast, KA-injected A1R-KO mice displayed massive neuronal cell loss in the ipsilateral hippocampus, and, importantly, the contralateral hippocampus was also affected with significant cell loss in the hilus and in the CA1 region of the pyramidal cell layer. We conclude that activation of A1 receptors by ambient adenosine is crucial in keeping epileptic foci localized. These results open up a new dimension of the A1 receptor's role in controlling excitotoxic cell death and further demonstrate its importance in preventing the progression of status epilepticus to lethal consequences.


Subject(s)
Epilepsy/metabolism , Epilepsy/pathology , Hippocampus/metabolism , Hippocampus/pathology , Receptor, Adenosine A1/physiology , Animals , Cell Death , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/genetics , Female , Hippocampus/drug effects , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics , Status Epilepticus/genetics , Status Epilepticus/metabolism , Status Epilepticus/mortality , Status Epilepticus/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...