Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Eur J Paediatr Neurol ; 51: 79-83, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38880066

ABSTRACT

OBJECTIVE: This retrospective study aimed to describe a cohort of 38 pediatric patients with MOGAD and to investigate the clinical differences between patients with CSF-negativity and CSF-positivity for MOG-abs. METHODS: The clinical and laboratory characteristics of pediatric patients with MOGAD were retrospectively studied. Demographics, clinical characteristics, CSF analysis, treatments and prognosis of patients were recorded. All patients' serums and CSF were tested for MOG-IgG by live cell-based assays (CBA). The data were statistically analysed. RESULTS: A total of 38 pediatric MOGAD patients were enrolled in the study, including 22 (57.9 %) females and 16 male (42.1 %) with a mean age of 8.4 ± 4.0 years at disease onset. Twenty-seven (71.7 %) patients were CSF-positive for MOG-abs while 11 (28.9 %) patients were CSF-negative for MOG-abs. The median follow-up was 25.5 months (IQR 5.5-73.25). Seventeen (44.7 %) patients presented a relapsing disease course, and the majority of these patients was CSF positive with a significant difference between the two groups (p = 0.038) in terms of recurrent diseases. CSF-positive patients presented more often an increased white cell count (p = 0.043), and in this cohort clinical phenotypes with spinal involvement were more frequent while encephalitis-like phenotypes were more frequent in the CSF negative cohort (p = 0.019). CONCLUSIONS: CSF-status appears to identify two subgroups in this pediatric MOGAD population; thus, CSF-status requires further studies in pediatric patients with MOGAD.


Subject(s)
Autoantibodies , Myelin-Oligodendrocyte Glycoprotein , Humans , Male , Female , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/cerebrospinal fluid , Child , Autoantibodies/cerebrospinal fluid , Autoantibodies/blood , Retrospective Studies , Child, Preschool , Adolescent , Follow-Up Studies , Clinical Relevance
2.
Nat Genet ; 9(3): 256-60, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7773288

ABSTRACT

Transplantations for neurological disorders are limited by the supply of human fetal tissue. To generate larger numbers of cells of appropriate phenotype, we investigated whether human neural progenitors expanded in vitro could be modified with recombinant adenoviruses. Strong expression of beta-galactosidase was obtained in vitro. Two or three weeks after transplantation of engineered cells to the rat brain, we observed a small percentage of surviving neuroblasts strongly expressing beta-galactosidase in four out of 13 rats. Thus human precursor cells that have been genetically modified using adenoviruses are a promising tool for ex vivo gene therapy of neurodegenerative diseases.


Subject(s)
Brain Tissue Transplantation/methods , Fetal Tissue Transplantation/methods , Adenoviridae/genetics , Animals , Cells, Cultured , Gene Expression , Gene Transfer Techniques , Genetic Engineering , Genetic Vectors , Humans , Lac Operon , Neurons/transplantation , Rats , Stem Cell Transplantation , beta-Galactosidase/genetics
3.
Neuron ; 5(4): 393-402, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2119630

ABSTRACT

Fibroblastic 3T3 and endocrine RIN cells were genetically modified by infection with a recombinant retrovirus encoding the form I of human tyrosine hydroxylase (TH) and selection in tyrosine-free medium. These cells were grafted to rats unilaterally lesioned with 6-hydroxy-dopamine. Both cell types survived implantation into the striatum, expressed TH immunoreactivity, and as assessed by microdialysis 8-9 days after implantation, secreted high amounts of DOPA and/or dopamine into the surrounding host striatum. The modified 3T3 cells secreted large amounts of DOPA that was efficiently decarboxylated to dopamine by the host striatal tissue; the newly synthesized dopamine was stored only to a limited extent in the denervated striatum. The modified RIN cells synthesized dopamine that was stored intracellularly and released in a regulated fashion. The grafted DOPA-secreting cells produced 4-5 times higher extracellular dopamine levels than the dopamine-secreting cells, and they were more efficient in reducing apomorphine-induced rotation. No effect was observed with either cell type on amphetamine-induced turning behavior.


Subject(s)
Corpus Striatum/physiology , Dihydroxyphenylalanine/metabolism , Dopamine/metabolism , Endocrine Glands/metabolism , Fibroblasts/metabolism , Genetic Engineering , Animals , Behavior, Animal , Catechols/metabolism , Denervation , Dialysis/methods , Endocrine Glands/cytology , Fibroblasts/transplantation , Humans
4.
Nat Biotechnol ; 17(4): 349-54, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10207882

ABSTRACT

Ex vivo gene transfer is emerging as a promising therapeutic approach to human neurodegenerative diseases. By combining efficient methodologies for cell amplification and gene delivery, large numbers of cells can be generated with the capacity to synthesize therapeutic molecules. These cells can then be transplanted into the degenerating central nervous system (CNS). Applying this approach to human diseases will require the development of suitable cellular vehicles, as well as safe gene delivery systems capable of tightly controlled transgene expression. For such brain repair technologies, human neural progenitors may be extremely valuable, because of their human CNS origin and developmental potential. We have used these cells to develop a system for the regulated expression of a gene of therapeutic potential. We report the construction of a single adenovirus encoding human tyrosine hydroxylase 1 (hTH-1) under the negative control of the tetracycline-based gene regulatory system. Human neural progenitors infected with this vector produced large amounts of hTH-1. Most importantly, doxycycline allowed a reversible switch of transgene transcription both in vitro and in vivo. This system may be applied to the development of therapies for human neurodegenerative diseases.


Subject(s)
Adenoviridae/genetics , Brain Tissue Transplantation , Brain/cytology , Gene Transfer Techniques , Stem Cell Transplantation , Tyrosine 3-Monooxygenase/genetics , Animals , Brain/embryology , Brain/enzymology , Cell Transplantation , Doxycycline/pharmacology , Fetal Tissue Transplantation , Gene Expression Regulation , Genetic Vectors , Humans , Rats , Stem Cells/virology , Tyrosine 3-Monooxygenase/biosynthesis
6.
Prog Neurobiol ; 55(4): 333-41, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9654383

ABSTRACT

Adenovirus is an efficient vector for neuronal gene therapy due to its ability to infect post-mitotic cells, its high efficacy of cell transduction and its low pathogenicity. Recombinant adenoviruses encoding for therapeutical agents can be delivered in vivo after direct intracerebral injection into specific brain areas. They can be transported in a retrograde manner from the injection site to the projection cell bodies offering promising applications for the specific targeting of selected neuronal populations not easily accessible by direct injection, such as the motor neurons in the spinal cord. Adenoviral vectors are also efficient tools for the ex vivo gene therapy, that is, the genetical modification of cells prior to their transplantation into the nervous system. Recently, the efficacy of the adenovirus as a gene vector system has been demonstrated in several models of neurodegenerative diseases including Parkinson's disease (PD) and motor neuron diseases. In rat models of PD, adenoviruses encoding for either tyrosine hydroxylase, superoxide dismutase or glial-derived neurotrophic factor improved the survival and the functional efficacy of dopaminergic cells. Similarly, the intramuscular injection of an adenovirus encoding for neurotrophin-3 had substantial therapeutic effects in a mutant mouse model of motor neuron degenerative disease. However, although adenoviruses are highly attractive for neuronal gene transfer, they can trigger a strong inflammatory reaction leading in particular to the destruction of infected cells. The recent development of new generations of adenoviral vectors could shed light on the nature of the immune reaction caused by adenoviral vectors in the brain. The use of these new vectors, combined with that of neurospecific and regulatable promoters, should improve adenovirus gene transfer into the central nervous system.


Subject(s)
Adenoviridae , Brain Diseases/therapy , Genetic Therapy/trends , Neurodegenerative Diseases/therapy , Animals , Humans
7.
Circulation ; 100(5): 533-40, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430768

ABSTRACT

BACKGROUND: To define the role of metalloproteinases (MMPs) in the development of lipid-rich atherosclerotic lesions in relation to the balance between proteolytic and antiproteolytic activities, we investigated the impact of adenovirus-mediated elevation in the circulating levels of human tissue inhibitor of MMP (TIMP-1) in atherosclerosis-susceptible apolipoprotein E-deficient (apoE(-/-)) mice. METHODS AND RESULTS: Infusion of apoE(-/-) mice fed a lipid-rich diet with rAd.RSV.TIMP-1 (1x10(11) viral particles) resulted in high hepatic expression of TIMP-1. At 2 weeks after injection, plasma TIMP-1 levels ranged from 7 to 24 micrograms/mL (mean 14.8+/-6.8). Marked overexpression of TIMP-1 was transient, with levels of TIMP-1 decreasing to 2.5 to 8 micrograms/mL (mean 4.3+/-2.1) at 4 weeks. Plasma lipid and lipoprotein levels in mice treated with rAd.RSV.TIMP-1 were similar to those treated with rAd.RSV.betaGal. However, rAd.RSV.TIMP-1-infused mice displayed a marked reduction (approximately 32%; P<0.05) in mean lesion area per section (512+/-121 micrometers(2)x10(3); n=12 sections from 4 animals) as compared with rAd.RSV.betaGal-infused mice (750+/-182 micrometers(2)x10(3); n=12 sections from 4 animals). Similarly, marked reduction in macrophage deposition as well as MMP-2, MMP-3, and MMP-13 antigens was observed. CONCLUSIONS: Histological and immunohistologic analyses of atherosclerotic lesions revealed increases in collagen, elastin, and smooth muscle alpha-actin content in mice treated with rAd.RSV.TIMP-1. These qualitative and quantitative features were the consequence of TIMP-1 infiltration from plasma to arterial intima, as immunohistochemical analyses revealed an abundance of TIMP-1 specifically in lesions of rAd.RSV. TIMP-1-treated mice.


Subject(s)
Adenoviridae , Apolipoproteins E/deficiency , Arteriosclerosis/pathology , Arteriosclerosis/prevention & control , Gene Transfer Techniques , Genetic Vectors , Tissue Inhibitor of Metalloproteinase-1/blood , Animals , Arteriosclerosis/etiology , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Up-Regulation
8.
Hum Gene Ther ; 10(18): 2987-97, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10609659

ABSTRACT

Huntington's disease (HD) is a genetic disorder leading to the degeneration of striatal GABA-ergic output neurons. No treatment is currently available for this devastating disorder, although several neurotrophic factors, including brain-derived neurotrophic factor (BDNF), have been shown to be beneficial for striatal neuron survival. We analyzed the effect of adenovirus-mediated transfer of the BDNF gene in a model of HD. Using a stereological procedure, three groups of rats were given an intrastriatal injection of adenovirus encoding BDNF, beta-galactosidase, or sham surgery. Two weeks after treatment, the animals were lesioned with quinolinic acid (QUIN), a toxin that induces striatal neuron death by an excitotoxic process. One month after the lesion, histological study revealed that striatal neurons were protected only in rats treated with the BDNF adenovirus. Volume measurements showed that the QUIN-induced lesions were 55% smaller in the BDNF adenovirus-treated group than in the beta-galactosidase adenovirus-treated group (p < 0.05), and the sham-treated group (p < 0.05). To determine the survival of striatal GABA-ergic output neurons after the QUIN-induced lesion, we immunostained brain sections with DARPP-32, an antibody specific for striatal output neurons. Prior treatment with the BDNF adenovirus resulted in a cell survival of 64%, whereas that after beta-galactosidase treatment was 46% (p < 0.05), showing that the BDNF adenovirus protected the striatal neurons. These results indicate that transfer of the BDNF gene is of therapeutic value for Huntington's disease.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Corpus Striatum/pathology , Gene Transfer Techniques , Huntington Disease/therapy , Adenoviridae/genetics , Animals , Base Sequence , Brain-Derived Neurotrophic Factor/genetics , DNA Primers , Disease Models, Animal , Female , Genetic Therapy , Genetic Vectors , Huntington Disease/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics
9.
Mol Neurobiol ; 15(2): 241-56, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9396012

ABSTRACT

Gene therapy is a potentially powerful approach to the treatment of neurological diseases. The discovery of neurotrophic factors inhibiting neurodegenerative processes and neurotransmitter-synthesizing enzymes provides the basis for current gene therapy strategies for Parkinson's disease. Genes can be transferred by viral or nonviral vectors. Of the various possible vectors, recombinant retroviruses are the most efficient for genetic modification of cells in vitro that can thereafter be used for transplantation (ex vivo gene therapy approach). Recently, in vivo gene transfer to the brain has been developed using adenovirus vectors. One of the advantages of recombinant adenovirus is that it can transduce both quiescent and actively dividing cells, thereby allowing both direct in vivo gene transfer and ex vivo gene transfer to neural cells. Probably because the brain is partially protected from the immune system, the expression of adenoviral vectors persists for several months with little inflammation. Novel therapeutic tools, such as vectors for gene therapy have to be evaluated in terms of efficacy and safety for future clinical trials. These vectors still need to be improved to allow long-term and possibly regulatable expression of the transgene.


Subject(s)
Genetic Therapy , Nerve Growth Factors , Nerve Tissue Proteins/metabolism , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/metabolism , Adenoviruses, Human/genetics , Animals , Astrocytes/metabolism , Astrocytes/transplantation , Cell Survival , Cell Transplantation , Cells, Cultured/transplantation , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dependovirus/genetics , Dopamine/metabolism , Fibroblasts/metabolism , Fibroblasts/transplantation , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor , Humans , Nerve Degeneration , Nerve Tissue Proteins/genetics , Neurons/pathology , Oxidopamine/toxicity , Parkinson Disease/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/therapy , Rats , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Stem Cell Transplantation , Stem Cells/metabolism , Tyrosine 3-Monooxygenase/genetics
10.
FEBS Lett ; 205(1): 6-10, 1986 Sep 01.
Article in English | MEDLINE | ID: mdl-2427363

ABSTRACT

Tyrosine hydroxylase, the rate limiting enzyme in the biosynthesis of catecholamine, is a tetramer composed of four subunits of the same molecular mass. A full length cDNA clone encoding tyrosine hydroxylase has been inserted into the SP6 expression system. Translation of the corresponding RNA in Xenopus oocyte results in enzymatic activity, demonstrating that a single gene contains all the necessary genetic information to code for a functional enzyme. The potential of this system in the analysis of posttranslational tyrosine hydroxylase modifications is discussed.


Subject(s)
Oocytes/enzymology , RNA/physiology , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Cloning, Molecular , DNA , In Vitro Techniques , Promoter Regions, Genetic , Protein Biosynthesis , Transcription, Genetic , Tyrosine 3-Monooxygenase/genetics , Xenopus laevis
11.
Brain Res Mol Brain Res ; 21(3-4): 225-34, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8170347

ABSTRACT

A mouse line transgenic for nerve growth factor (NGF) was developed using the mouse prepro-NGF cDNA inserted within a plasmid containing the proximal region (-10 to -550 bp) of the c-fos promoter and the transcription termination and polyadenylation signals of the rabbit beta-globin gene. No significant modification of gross behavior or central nervous system anatomy was detected in adult animals as assessed by immunohistochemistry and in situ hybridization for NGF and choline acetyltransferase. The expression of the transgene and the possible regulation of its expression by agents acting on the promoter were investigated in vitro. Despite the presence of an additional pool of NGF mRNA specific to the transgene, basal levels of NGF in the supernatant of transgenic astrocytes were similar to normal ones. On the other hand, transgenic neurons spontaneously synthesized and released levels of NGF two to three times higher than normal neurons, while mRNA levels were barely detectable by conventional Northern blotting. The tissue-specificity of NGF expression was respected, with higher levels in hippocampal than neocortical neurons. Increases of NGF mRNA by agents acting on the promoter could be observed in normal and transgenic astrocytes only after inhibition of the protein synthesis by cycloheximide, suggesting a similar rapid turnover of normal and transgenic transcripts. Cyclic AMP agonists specifically increased the secretion of NGF protein by transgenic astrocytes and neurons, while activators of the protein kinase C had a similar effect on transgenic and normal cells. Differences between amounts of NGF secreted by neurons and astrocytes with regards to their respective content in mRNA suggest that transgenic transcripts are subject to normal cell- and tissue-specific post-transcriptional regulations. Agents acting on the c-fos promoter through the protein kinase C or cyclic AMP routes differentially increased the secretion of NGF by transgenic astrocytes or neurons, supporting this hypothesis.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation/physiology , Genes, fos , Nerve Growth Factors/genetics , Neurons/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cloning, Molecular , Embryonic and Fetal Development/genetics , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Transcription, Genetic/drug effects
12.
J Chem Neuroanat ; 5(1): 11-8, 1992.
Article in English | MEDLINE | ID: mdl-1351396

ABSTRACT

In situ hybridization experiments, using oligodeoxyribonucleotides specific for the two major expressed human tyrosine hydroxylase mRNAs, were performed on human brain sections at the level of the mesencephalon. The specificity of the probes was ascertained by Northern blot experiments carried out with independently in vitro synthesized human tyrosine hydroxylase mRNAs. For in situ hybridization experiments, oligodeoxyribonucleotides were labelled with nucleotides tagged with digoxigenin or biotin molecules. The hybridized oligonucleotides were detected by antibodies coupled with peroxidase and alkaline phosphatase enzymes, which yield, with appropriate substrates, brown and purple products, respectively. The simultaneous detection of the two mRNAs with digoxigeninated and biotinylated probes revealed that these two mRNAs are co-expressed in single cells. The purple product obtained with alkaline phosphatase exhibits a discrete distribution within the dopaminergic cells suggesting these mRNAs are associated with sub-cellular structures. Finally, a heterogeneity in the intensity of the labelling of reactive cells with both probes was visualized as well as the expression of the two mRNA species in neurites.


Subject(s)
Mesencephalon/metabolism , Oligodeoxyribonucleotides , RNA, Messenger/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis , Aged , Alkaline Phosphatase/metabolism , Base Sequence , Biotin , Blotting, Northern , Digoxigenin , Humans , Mesencephalon/enzymology , Middle Aged , Molecular Sequence Data , Nucleic Acid Hybridization , Tyrosine 3-Monooxygenase/genetics
13.
Neurochem Int ; 22(5): 511-6, 1993 May.
Article in English | MEDLINE | ID: mdl-8485456

ABSTRACT

We have constructed a recombinant retrovirus that expresses choline acetyltransferase (ChAT) by placing the porcine enzyme cDNA under the control of the 5' long terminal repeat of the retroviral vector pMMuLV. Using retrovirus-mediated gene transfer, we have expressed ChAT in astroglial (STR-SVLT) and neuroendocrine (RIN) cell lines. Both genetically modified cell types synthesize acetylcholine (ACh). ACh is also present in the culture medium at a low concentration relative to that found in the modified cells. This result suggests that the synthesized ACh is retained within the cells and released by these two cell types. Release of ACh is not increased in the presence of the calcium ionophore A23187 or by depolarizing concentrations of potassium in either STR-SVLT or in RIN cells. The implications of these studies for understanding ACh release mechanisms are discussed.


Subject(s)
Acetylcholine/biosynthesis , Acetylcholine/metabolism , Choline O-Acetyltransferase/genetics , Models, Biological , Retroviridae/genetics , Transfection , Animals , Calcimycin/pharmacology , Calcium/pharmacology , Cell Line , Gene Expression , Genetic Vectors , Swine
14.
Neuroreport ; 7(10): 1655-9, 1996 Jul 08.
Article in English | MEDLINE | ID: mdl-8904776

ABSTRACT

Tight control of the activity of a therapeutic gene introduced in vivo is a major issue in gene therapy research. Appropriate levels of expression may be crucial for gene correction. The tetracycline-sensitive regulatory system is highly effective for transcriptional regulation of foreign genes in mammalian cells. Here we report tight tetracycline-dependent regulation of a luciferase reporter gene transferred into the rat brain in the genetically modified neural precursor cell line ST14A as early as 2 days and until at least 6 days after transplantation. This is the first demonstration of the potential of this regulatory system for the modulation of the expression of therapeutic genes introduced into the central nervous system.


Subject(s)
Brain/drug effects , Gene Expression/drug effects , Tetracycline/pharmacology , Animals , Brain Tissue Transplantation , Cell Line , Cell Transplantation , Female , Gene Transfer Techniques , Rats , Rats, Sprague-Dawley
15.
Neuroreport ; 6(1): 49-53, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7703427

ABSTRACT

Direct intracerebral gene transfer to neural cells has been demonstrated with recombinant adenovirus encoding beta-galactosidase. To explore the potential of recombinant adenovirus for the therapy of neurological disease we constructed a recombinant adenovirus encoding tyrosine hydroxylase and optimized intracerebral injection to express the gene in the striatum of unilaterally denervated rats. These animals have dopamine depletion in their lesioned striatum, causing a rotation asymmetry induced by apomorphine. One and two weeks after intracerebral injection this sensorimotor asymmetry was decreased by the adenovirus encoding tyrosine hydroxylase and not by a control adenovirus encoding beta-galactosidase. Histological analysis showed that tyrosine hydroxylase was preferentially expressed in astrocytes.


Subject(s)
Adenoviridae/physiology , Brain/physiopathology , Gene Transfer Techniques , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/genetics , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Denervation , Female , Genetic Vectors , Oxidopamine/pharmacology , Parkinson Disease/genetics , Rats , Rats, Sprague-Dawley , Recombination, Genetic , Stereotyped Behavior , beta-Galactosidase/genetics
16.
Neuroreport ; 7(2): 497-501, 1996 Jan 31.
Article in English | MEDLINE | ID: mdl-8730814

ABSTRACT

Superoxide dismutase (SOD), a key enzyme in the detoxification of free radicals, catalyses the dismutation of superoxide O2.- to oxygen and hydrogen peroxide (H2O2). It is therefore a promising candidate for gene transfer therapy of neurological diseases in which free radicals are thought to be involved. We have constructed a recombinant adenoviral vector containing the human copper-zinc SOD cDNA. Using this vector we were able to drive the production of an active human copper-zinc SOD protein (hCuZnSOD) in various cell lines and primary cultures. Infection of striatal cells with a recombinant adenovirus expressing hCuZnSOD protected these cells from glutamate-induced cell death.


Subject(s)
Adenoviridae/metabolism , Genetic Vectors/genetics , Glutamic Acid/toxicity , Neostriatum/cytology , Neurons/drug effects , Superoxide Dismutase/metabolism , Adenoviridae/genetics , Animals , Cell Survival/physiology , Cells, Cultured , Humans , Immunohistochemistry , Mice , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/genetics
17.
J Psychiatr Res ; 21(4): 559-68, 1987.
Article in English | MEDLINE | ID: mdl-2894459

ABSTRACT

Manic depressive illness has been clearly established to exhibit a strong genetic component and is therefore amenable to linkage analysis using random DNA markers. In view of the catecholamine hypothesis of this disorder, the gene encoding tyrosine hydroxylase (TH) the limiting enzyme in catecholamines is a good candidate to investigate. This gene has been localized to chromosome 11 in close linkage with Harvey-ras-1. The various transcriptional and post-transcriptional mechanisms that modulate short and long-term TH activity are discussed. Human tyrosine hydroxylase is coded by at least three distinct mRNAs derived from a single gene. This variation has clear functional consequences and could represent a novel mode of regulating catecholamines levels in normal and pathological neurons.


Subject(s)
Bipolar Disorder/genetics , Catecholamines/metabolism , Genetic Markers , Humans , Tyrosine 3-Monooxygenase/genetics
20.
Exp Neurol ; 139(1): 39-53, 1996 May.
Article in English | MEDLINE | ID: mdl-8635567

ABSTRACT

Astrocytes secreting high levels of L-3,4-dihydroxyphenylalanine (DOPA) have been generated by retrovirus-mediated transfer of the human tyrosine hydroxylase (TH) gene. Immature astrocytes obtained from prenatal rat brain were cocultured with TH virus producing psi-2 cells that had been pretreated with the mitosis inhibitor mitomycin-C. During the first week of coculture DOPA production gradually increased to reach a plateau after 7-9 days. At this time point virtually all cells were GFAP positive and over 80% of them expressed TH. DOPA production in the transduced astrocytes was largely independent of exogenous cofactor, and DOPA release into the medium was not influenced by addition of either KCl or tetrodotoxin or by removal of Ca2+ from the culture medium, indicating that the newly synthesized DOPA was constitutively released from the cells. Transplantation of the TH-transduced astrocytes to the striatum in unilaterally 6-hydroxydopamine lesioned rats reduced apomorphine-induced turning by about 50% at 2 weeks postgrafting. Microscopic analysis revealed that the transduced astrocytes survived very well after transplantation and that some of the grafted cells had migrated out, partly along blood vessels, into the surrounding striatum. TH expression was observed in cells with both the appearance of mature GFAP-positive astrocytes, as well as in more immature-looking cells. However, only a few percent of all transplanted cells maintained significant expression of the transgene, as determined by TH immuno-histochemistry. The results show that primary astrocytes may be highly useful as gene carriers for ex vivo gene therapy in the CNS. With future improvement in the gene transduction procedure for more efficient, sustained expression of the TH transgene in vivo, genetically engineered DOPA-producing astrocytes hold great promise as a tool to explore the potential of ex vivo gene therapy in Parkinson's disease.


Subject(s)
Astrocytes/metabolism , Dihydroxyphenylalanine/biosynthesis , Genetic Vectors , Parkinson Disease/therapy , Retroviridae/genetics , Tyrosine 3-Monooxygenase/genetics , Animals , Astrocytes/enzymology , Astrocytes/transplantation , Cell Division/physiology , Cell Transplantation , Cells, Cultured/cytology , Cells, Cultured/enzymology , Cells, Cultured/virology , Cytomegalovirus/genetics , Dihydroxyphenylalanine/metabolism , Disease Models, Animal , Female , Gene Expression/physiology , Humans , Neostriatum/surgery , Rats , Rats, Sprague-Dawley , Rats, Wistar , Transgenes/physiology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL