Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Epilepsia ; 46(8): 1162-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16060924

ABSTRACT

PURPOSE: Stem cells and their derivatives have emerged as a promising tool for cell-based drug delivery because of (a) their unique ability to differentiate into various somatic cell types, (b) the virtually unlimited donor source for transplantation, and (c) the advantage of being amenable to a wide spectrum of genetic manipulations. Previously, adenosine-releasing embryonic stem (ES) cells have been generated by disruption of both alleles of adenosine kinase (Adk-/-). Lack of ADK did not compromise the cells' differentiation potential into embryoid bodies or glial precursor cells. The aim of the present study was to investigate the potential of differentiated Adk-/- ES cell progeny for seizure suppression by paracrine adenosine release. METHODS: To isolate paracrine effects of stem cell-derived implants from effects caused by network integration, ES cell-derived embryoid bodies and glial precursor cells were encapsulated into semipermeable polymer membranes and grafted into the lateral brain ventricles of kindled rats. RESULTS: While seizure activity in kindled rats with wild-type Adk+/+ implants remained unaltered, rats with adenosine-releasing Adk-/- ES cell-derived implants displayed transient protection from convulsive seizures and a profound reduction of afterdischarge activity in EEG recordings. Long-term seizure suppression was precluded by limited viability of the encapsulated cells. CONCLUSIONS: We thereby provide a proof-of-principle that Adk-/- ES cell-derived brain implants can suppress seizure activity by a paracrine mode of action. Adk-deficient stem cells therefore represent a potential tool for the treatment of epileptic disorders.


Subject(s)
Adenosine/administration & dosage , Adenosine/metabolism , Brain/surgery , Kindling, Neurologic , Seizures/prevention & control , Stem Cell Transplantation/methods , Stem Cells/physiology , Adenosine/biosynthesis , Adenosine Kinase/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Drug Delivery Systems , Electroencephalography , Paracrine Communication/physiology , Rats , Seizures/drug therapy , Stem Cells/metabolism
2.
Exp Neurol ; 193(1): 53-64, 2005 May.
Article in English | MEDLINE | ID: mdl-15817264

ABSTRACT

Adenosine is an important inhibitory modulator of brain activity. In a previous ex vivo gene therapy approach, local release of adenosine by encapsulated fibroblasts implanted into the vicinity of an epileptic focus, was sufficient to provide transient protection from seizures (Huber, A., Padrun, V., Deglon, N., Aebischer, P., Mohler, H., Boison, D., 2001. Grafts of adenosine-releasing cells suppress seizures in kindling epilepsy. Proc. Natl. Acad. Sci. U. S. A. 98, 7611-7616). Long-term seizure suppression beyond 2 weeks was precluded by limited life expectancy of the encapsulated fibroblasts. To study the feasibility for long-term seizure suppression by adenosine releasing brain implants, in the present contribution, mouse C2C12 myoblasts were engineered to release adenosine by genetic inactivation of adenosine kinase. After encapsulation, the myoblasts were grafted into the lateral brain ventricles of epileptic rats kindled in the hippocampus. While seizure activity in animals with wild-type implants remained unaltered, 1 week after grafting all rats with adenosine-releasing implants (n = 25) displayed complete protection from convulsive seizures and a corresponding reduction of afterdischarges in EEG-recordings. The duration of seizure suppression was maintained for a period of 3 weeks in 50% of the animals ranging to a maximum of 8 weeks in one animal. During the course of these experiments, adenosine A1 receptors remained responsive to selective agonists and antagonists indicating a lack of desensitization of A1 receptors after local long-term exposure to adenosine. Furthermore, local release of adenosine did not affect locomotor activity, whereas systemic application of the A1 agonist 2-chloro-N6-cyclopentyladenosine caused strong sedation. Thus, the local release of adenosine by cellular implants provides a feasible option for a potential side-effect free approach for the long-term treatment of focal epilepsies.


Subject(s)
Adenosine/administration & dosage , Adenosine/metabolism , Drug Delivery Systems/methods , Myoblasts/transplantation , Receptor, Adenosine A1/metabolism , Seizures/therapy , Adenosine A1 Receptor Agonists , Animals , Capsules , Cell Survival/physiology , Delayed-Action Preparations/administration & dosage , Male , Mice , Mice, Inbred C3H , Myoblasts/cytology , Myoblasts/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/physiology , Seizures/metabolism , Seizures/prevention & control , Time Factors
3.
Neurobiol Dis ; 16(2): 428-39, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15193299

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF), a potent neurotrophic factor with restorative effects in a variety of rodent and primate models of Parkinson's disease (PD), could be of therapeutic value to PD. In this study, we show that intraventricular chronic infusion of low doses of GDNF using encapsulated genetically engineered C2C12 cells can exert: (1) transient recovery of motor deficits (hypokinesia); (2) significant protection of intrinsic striatal dopaminergic function in the immediate vicinity of the site of implantation of the capsule in the caudate nucleus, and (3) significant-long-lasting-neurotrophic properties at the nigral level with an increase volume of the cell bodies. These observations confirm the potent neurorestorative potential of GDNF in PD and the safety/efficacy of the encapsulation technology as a means to deliver in situ this neurotrophic cytokine even using an intraventricular approach.


Subject(s)
Dihydroxyphenylalanine/analogs & derivatives , Genetic Therapy/methods , Nerve Growth Factors/genetics , Neuroglia/transplantation , Parkinsonian Disorders/therapy , Animals , Capsules , Fluorine Radioisotopes , Glial Cell Line-Derived Neurotrophic Factor , Lateral Ventricles , Magnetic Resonance Imaging , Male , Motor Activity/drug effects , Neurons/enzymology , Neurons/pathology , Papio , Parkinsonian Disorders/diagnostic imaging , Parkinsonian Disorders/pathology , Recovery of Function/drug effects , Substantia Nigra/pathology , Tomography, Emission-Computed , Tyrosine 3-Monooxygenase/metabolism
4.
Mol Ther ; 7(4): 506-14, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12727114

ABSTRACT

The present study investigates the respective roles of both the host immune response and the metabolic requirements in determining the long-term survival of erythropoietin-secreting myoblasts within encapsulating polymer membranes. Hollow-fiber capsules loaded with a high density of erythropoietin-secreting C(2)C(12) myoblasts survived poorly in the subcutaneous tissue of syngeneic mice, inducing variable hematocrit responses. To determine the role and the nature of the host response, recipients were treated with anti-inflammatory (diclofenac) and immunosuppressive (dexamethasone, FK506) agents. Only immunosuppressive drugs led to improved graft survival after 5 weeks of implantation, indicating an immune process as the cause of cell death. Furthermore, transient blocking of this process allowed long-term preservation of the implanted cells (> 100 days). The formation of necrotic cell cores inside densely packed devices elicited a local chronic inflammatory reaction. Hence, implants were designed to limit early cell death by inserting a supporting matrix and decreasing the number of loaded cells. The most efficient erythropoietin delivery was obtained with matrix-containing capsules that had received the lowest myoblast density. These results highlight the critical role of initial engraftment in the long-term acceptance of encapsulated myoblasts and the need to limit early cell death in the device to prevent subsequent host immuno-inflammatory responses.


Subject(s)
Cell Transplantation/methods , Erythropoietin/genetics , Graft Survival/immunology , Inflammation/prevention & control , Myoblasts/transplantation , Animals , Capsules , Cells, Cultured , Dexamethasone/pharmacology , Diclofenac/pharmacology , Erythropoietin/metabolism , Female , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Hematocrit , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C3H , Myoblasts/cytology , Myoblasts/metabolism , Plasmids/genetics , Tacrolimus/pharmacology , Transplantation Tolerance
5.
Hum Gene Ther ; 13(18): 2197-201, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12542850

ABSTRACT

RNA interference (RNAi) is a form of posttranscriptional gene silencing mediated by short double-stranded RNA, known as small interfering RNA (siRNA). These siRNAs are capable of binding to a specific mRNA sequence and causing its degradation. The recent demonstration of a plasmid vector that directs siRNA synthesis in mammalian cells prompted us to examine the ability of lentiviral vectors to encode siRNA as a means of providing long-term gene silencing in mammalian cells. The RNA-polymerase III dependent promoter (H1-RNA promoter) was inserted in the lentiviral genome to drive the expression of a small hairpin RNA (shRNA) against enhanced green fluorescent protein (EGFP). This construct successfully silenced EGFP expression in two stable cell lines expressing this protein, as analyzed by fluorescence microscopy, flow cytometry, and Western blotting. The silencing, which is dose dependent, occurs as early as 72 hr postinfection and persists for at least 25 days postinfection. The ability of lentiviruses encoding siRNA to silence genes specifically makes it possible to take full advantage of the possibilities offered by the lentiviral vector and provides a powerful tool for gene therapy and gene function studies.


Subject(s)
Genetic Vectors , Lentivirus , RNA Interference , RNA/genetics , Cell Line , Gene Silencing , Humans , In Vitro Techniques , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...