Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Neurol ; 219(1): 126-35, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19460372

ABSTRACT

Pharmacotherapy for epilepsy is limited by high incidence of pharmacoresistance and failure to prevent development and progression of epilepsy. Using the rat hippocampal kindling model, we report on the therapeutic potential of novel silk-based polymers engineered to release the anticonvulsant adenosine. Polymers were designed to release 1000 ng adenosine per day during a time span of ten days. In the first experiment rats were kindled by hippocampal electrical stimulation until all animals reacted with stage 5 seizures. Adenosine-releasing or control polymers were then implanted into the infrahippocampal fissure ipsilateral to the site of stimulation. Subsequently, only recipients of adenosine-releasing implants were completely protected from generalized seizures over a period of ten days corresponding to the duration of sustained adenosine release. To monitor seizure development in the presence of adenosine, adenosine-releasing or control polymers were implanted prior to kindling. After 30 stimulations--delivered from days 4 to 8 after implantation--control animals had developed convulsive stage 5 seizures, whereas recipients of adenosine-releasing implants were still protected from convulsive seizures. Kindling was resumed after nine days to allow expiration of adenosine release. During additional 30 stimulations, recipients of adenosine-releasing implants gradually resumed kindling development at seizure stages corresponding to those when kindling was initially suspended, while control rats resumed kindling development at convulsive seizure stages. Blockade of adenosine A1 receptors did not exacerbate seizures in protected animals. We conclude that silk-based adenosine delivery exerts potent anti-ictogenic effects, but might also have at least partial anti-epileptogenic effects. Thus, silk-based adenosine augmentation holds promise for the treatment of epilepsy.


Subject(s)
Adenosine/pharmacology , Anticonvulsants/pharmacology , Epilepsy/drug therapy , Kindling, Neurologic/drug effects , Polymers/therapeutic use , Silk/therapeutic use , Adenosine/metabolism , Adenosine/therapeutic use , Adenosine A1 Receptor Antagonists , Animals , Anticonvulsants/metabolism , Anticonvulsants/therapeutic use , Disease Models, Animal , Drug Delivery Systems/methods , Drug Implants/pharmacology , Electric Stimulation , Epilepsy/etiology , Epilepsy/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Kindling, Neurologic/physiology , Male , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/metabolism , Seizures/drug therapy , Seizures/etiology , Seizures/physiopathology , Treatment Outcome
2.
Biomaterials ; 29(26): 3609-16, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18514814

ABSTRACT

Adenosine augmentation therapies (AAT) make rational use of the brain's own adenosine-based seizure control system and hold promise for the therapy of refractory epilepsy. In an effort to develop an AAT compatible with future clinical application, we developed a novel silk protein-based release system for adenosine. Adenosine releasing brain implants with target release doses of 0, 40, 200, and 1000ng adenosine per day were prepared by embedding adenosine containing microspheres into nanofilm-coated silk fibroin scaffolds. In vitro, the respective polymers released 0, 33.4, 170.5, and 819.0ng adenosine per day over 14 days. The therapeutic potential of the implants was validated in a dose-response study in the rat model of kindling epileptogenesis. Four days prior to the onset of kindling, adenosine releasing polymers were implanted into the infrahippocampal cleft and progressive acquisition of kindled seizures was monitored over a total of 48 stimulations. We document a dose-dependent retardation of seizure acquisition. In recipients of polymers releasing 819ng adenosine per day, kindling epileptogenesis was delayed by one week corresponding to 18 kindling stimulations. Histological analysis of brain samples confirmed the correct location of implants and electrodes. We conclude that silk-based delivery of around 1000ng adenosine per day is a safe and efficient strategy to suppress seizures.


Subject(s)
Adenosine/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems , Epilepsy/drug therapy , Polymers/chemistry , Silk/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Epilepsy/physiopathology , Fibroins/chemistry , Hippocampus/surgery , Humans , Kindling, Neurologic , Male , Materials Testing , Microspheres , Prostheses and Implants , Rats , Rats, Sprague-Dawley
3.
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
4.
J Cereb Blood Flow Metab ; 28(1): 17-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17457365

ABSTRACT

The rate of ischemic brain injury varies with the brain region, requiring only hours in striatum but days in hippocampus. Such maturation implies the existence of endogenous neuroprotective mechanisms. Adenosine is an endogenous neuroprotectant regulated by adenosine kinase (ADK). To investigate, whether adenosine might play a role in protecting the hippocampus after focal ischemia, we subjected transgenic mice, which overexpress ADK in hippocampal neurons (Adk-tg mice) to transient middle cerebral artery occlusion (MCAO). Although the hippocampus of wild-type (wt) mice was consistently spared from injury after 60 mins of MCAO, hippocampal injury became evident in Adk-tg mice after only 15 mins of MCAO. To determine, whether downregulation of hippocampal ADK might qualify as candidate mechanism mediating endogenous neuroprotection, we evaluated ADK expression in wt mice after several periods of reperfusion after 15 or 60 mins of MCAO. After 60 mins of MCAO, hippocampal ADK was significantly reduced in both hemispheres after 1, 3, and 24 h of reperfusion. Reduction of ADK-immunoreactivity corresponded to a 2.2-fold increase in hippocampal adenosine at 3 h of reperfusion. Remarkably, a significant reduction of ADK immunoreactivity was also found in the ipsilateral (stroked) hippocampus after 15 mins of MCAO and 3 h of reperfusion. Thus, transient downregulation of hippocampal ADK after stroke might be a protective mechanism during maturation hippocampal cell loss.


Subject(s)
Adenosine Kinase/biosynthesis , Gene Expression Regulation, Enzymologic , Hippocampus/enzymology , Infarction, Middle Cerebral Artery/enzymology , Neuroprotective Agents/metabolism , Adenosine/genetics , Adenosine/metabolism , Adenosine Kinase/genetics , Animals , Corpus Striatum/enzymology , Corpus Striatum/pathology , Down-Regulation , Hippocampus/pathology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Transgenic , Neurons/enzymology , Neurons/pathology , Organ Specificity , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Time Factors
5.
Exp Neurol ; 208(1): 26-37, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17716659

ABSTRACT

Cell therapies based on focal delivery of the inhibitory neuromodulator adenosine were previously shown to provide potent seizure suppression in animal models of epilepsy. However, hitherto used therapeutic cells were derived from rodents and thus not suitable for clinical applications. Autologous patient-derived adenosine-releasing cell implants would constitute a major therapeutic advance to avoid both xenotransplantation and immunosuppression. Here we describe a novel approach based on lentiviral RNAi mediated downregulation of adenosine kinase (ADK), the major adenosine-removing enzyme, in human mesenchymal stem cells (hMSCs), which would be compatible with autologous cell grafting in patients. Following lentiviral transduction of hMSCs with anti-ADK miRNA expression cassettes we demonstrate up to 80% downregulation of ADK and a concentration of 8.5 ng adenosine per ml of medium after incubating 10(5) cells for 8 h. hMSCs with a knockdown of ADK or cells expressing a scrambled control sequence were transplanted into hippocampi of mice 1 week prior to the intraamygdaloid injection of kainic acid (KA). While mice with control implants expressing a scrambled miRNA sequence or sham treated control animals were characterized by KA-induced status epilepticus and subsequent CA3 neuronal cell loss, animals with therapeutic ADK knockdown implants displayed a 35% reduction in seizure duration and 65% reduction in CA3 neuronal cell loss, when analyzed 24 h after KA-injection. We conclude that lentiviral expression of anti-ADK miRNA constitutes a versatile tool to generate therapeutically effective adenosine releasing hMSCs, thus representing a model system to generate patient identical autologous adult stem cell grafts.


Subject(s)
Adenosine Kinase/metabolism , Lentivirus/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/enzymology , RNA Interference , Status Epilepticus/surgery , Adenosine/metabolism , Adenosine Kinase/genetics , Animals , Down-Regulation , Excitatory Amino Acid Agonists , Genetic Vectors , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Kainic Acid , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/physiopathology , Transduction, Genetic
6.
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
7.
J Cereb Blood Flow Metab ; 27(5): 919-27, 2007 May.
Article in English | MEDLINE | ID: mdl-17119544

ABSTRACT

Protective mechanisms of the brain may reduce the extent of injury after focal cerebral ischemia. Here, we explored in a mouse model of focal cerebral ischemia potential synergistic neuroprotective effects of two mediators of neuroprotection: (i) neuronal or glial precursor cells and (ii) the inhibitory neuromodulator adenosine. Embryonic stem (ES) cells, engineered to release adenosine by biallelic disruption of the adenosine kinase gene, and respective wild-type cells were induced to differentiate into either neural or glial precursor cells and were injected into the striatum of mice 1 week before middle cerebral artery occlusion. All stem cell-derived graft recipients were characterized by a significant reduction in infarct volume, an effect that was augmented by the release of adenosine. Neuroprotection was strongest in adenosine-releasing glial precursor cell recipients, which were characterized by an 85% reduction of the infarct area. Graft-mediated neuroprotection correlated with a significant improvement of general and focal neurologic scores. Histologic analysis before and after ischemia revealed clusters of implanted cells within the striatum of all treated mice. We conclude that ES cell derived adenosine-releasing brain implants provide neuroprotection by synergism of endogenous precursor cell-mediated effects and paracrine adenosine release.


Subject(s)
Brain Ischemia/therapy , Neurons/transplantation , Stem Cell Transplantation , Adenosine/metabolism , Adenosine Kinase/genetics , Adenosine Kinase/physiology , Alleles , Animals , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...