Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Gene Ther ; 22(2): 181-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25474439

ABSTRACT

Sandhoff disease (SD) is caused by deficiency of N-acetyl-ß-hexosaminidase (Hex) resulting in pathological accumulation of GM2 ganglioside in lysosomes of the central nervous system (CNS) and progressive neurodegeneration. Currently, there is no treatment for SD, which often results in death by the age of five years. Adeno-associated virus (AAV) gene therapy achieved global CNS Hex restoration and widespread normalization of storage in the SD mouse model. Using a similar treatment approach, we sought to translate the outcome in mice to the feline SD model as an important step toward human clinical trials. Sixteen weeks after four intracranial injections of AAVrh8 vectors, Hex activity was restored to above normal levels throughout the entire CNS and in cerebrospinal fluid, despite a humoral immune response to the vector. In accordance with significant normalization of a secondary lysosomal biomarker, ganglioside storage was substantially improved, but not completely cleared. At the study endpoint, 5-month-old AAV-treated SD cats had preserved neurological function and gait compared with untreated animals (humane endpoint, 4.4±0.6 months) demonstrating clinical benefit from AAV treatment. Translation of widespread biochemical disease correction from the mouse to the feline SD model provides optimism for treatment of the larger human CNS with minimal modification of approach.


Subject(s)
Genetic Therapy , Sandhoff Disease/therapy , Animals , Cats , Dependovirus/genetics , Dependovirus/immunology , Disease Progression , Gangliosides/metabolism , Genetic Vectors , Humans , Immunity, Humoral , Injections, Intraventricular , Sandhoff Disease/pathology , Transduction, Genetic , Treatment Outcome , beta-N-Acetylhexosaminidases/biosynthesis , beta-N-Acetylhexosaminidases/genetics
2.
Gene Ther ; 21(12): 1041-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25273354

ABSTRACT

Familial amyloidotic polyneuropathy (FAP) is a neurodegenerative disorder characterized by extracellular deposition of amyloid fibrils composed by mutated transthyretin (TTR) mainly in the peripheral nervous system. At present, liver transplantation is still the standard treatment to halt the progression of clinical symptoms in FAP, but new therapeutic strategies are emerging, including the use of TTR stabilizers. Here we propose to establish a new gene therapy approach using adeno-associated virus (AAV) vectors to deliver the trans-suppressor TTR T119M variant to the liver of transgenic TTR V30M mice at different ages. This TTR variant is known for its ability to stabilize the tetrameric protein. Analysis of the gastrointestinal tract of AAV-treated animals revealed a significant reduction in deposition of TTR non-fibrillar aggregates in as much as 34% in stomach and 30% in colon, as well as decreased levels of biomarkers associated with TTR deposition, namely the endoplasmic reticulum stress marker BiP and the extracellular matrix protein MMP-9. Moreover, we showed with different studies that our approach leads to an increase in tetrameric and more stable forms of TTR, in favor of destabilized monomers. Altogether our data suggest the possibility to use this gene therapy approach in a prophylactic manner to prevent FAP pathology.


Subject(s)
Amyloid Neuropathies, Familial/therapy , Genetic Therapy/methods , Prealbumin/genetics , Amyloid Neuropathies, Familial/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Stress/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Genetic Markers , Genetic Vectors , Liver/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Peripheral Nervous System/metabolism , Prealbumin/metabolism , Proteomics
3.
Cancer Gene Ther ; 20(9): 514-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23949282

ABSTRACT

The antifolate methotrexate (MTX) is an important chemotherapeutic agent for treatment of osteosarcoma. This drug is converted intracellularly into polyglutamate derivates by the enzyme folylpolyglutamate synthase (FPGS). MTX polyglutamates show an enhanced and prolonged cytotoxicity in comparison to the monoglutamate. In the present study, we proved the hypothesis that transfer of the human fpgs gene into osteosarcoma cells may augment their MTX sensitivity. For this purpose, we employed the human osteocalcin (OC) promoter, which had shown marked osteosarcoma specificity in promoter studies using different luciferase assays in osteosarcoma and non-osteosarcoma cell lines. A recombinant lentiviral vector was generated with the OC promoter driving the expression of fpgs and the gene for enhanced green fluorescent protein (egfp), which was linked to fpgs by an internal ribosomal entry site (IRES). As the vector backbone contained only a self-inactivating viral LTR promoter, any interference of the OC promoter by unspecific promoter elements was excluded. We tested the expression of FPGS and enhanced green fluorescent protein (EGFP) after lentiviral transduction in various osteosarcoma cell lines (human MG-63 cells and TM 791 cells; rat osteosarcoma (ROS) 17/2.8 cells) and non-osteogenic tumor cell lines (293T human embryonic kidney cells, HeLa human cervix carcinoma cells). EGFP expression and MTX sensitivity were assessed in comparison with non-transduced controls. Whereas the OC promoter failed to enhance MTX sensitivity via FPGS expression in non-osteogenic tumor cell lines, the OC promoter mediated a markedly increased MTX cytotoxicity in all osteosarcoma cell lines after lentiviral transduction. The present chemotherapy-enhancing gene therapy system may have great potential to overcome in future MTX resistance in human osteosarcomas.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Bone Neoplasms/genetics , Gene Expression/drug effects , Methotrexate/pharmacology , Osteosarcoma/genetics , Peptide Synthases/genetics , Cell Line, Tumor , Cloning, Molecular , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Organ Specificity/genetics , Osteocalcin/genetics , Osteocalcin/metabolism , Promoter Regions, Genetic , Transduction, Genetic , Transfection , Tumor Cells, Cultured
4.
J Huntingtons Dis ; 2(1): 41-5, 2013.
Article in English | MEDLINE | ID: mdl-25063428

ABSTRACT

Our goal is delivery of a long-term treatment for Huntington's disease. We administer intracerebrally in sheep adeno-associated virus (AAV) to establish optimal safety, spread and neuronal uptake of AAV based therapeutics. Sheep have large gyrencephalic brains and offer the opportunity to study a transgenic Huntington's disease model. However, lack of a relevant brain stereotactic atlas and the difficulty of skull fixation make conventional stereotaxy unreliable. We describe a multi-modal image-guidance technique to achieve accurate placement of therapeutics into the sheep striatum.


Subject(s)
Corpus Striatum/surgery , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Huntington Disease , Animals , Animals, Genetically Modified , Convection , Corpus Striatum/anatomy & histology , Dependovirus , Huntington Disease/therapy , Sheep, Domestic , Stereotaxic Techniques
5.
Gene Ther ; 18(12): 1173-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21900963

ABSTRACT

Adeno-associated virus (AAV)-mediated gene replacement for lysosomal disorders have been spurred by the ability of some serotypes to efficiently transduce neurons in the brain and by the ability of lysosomal enzymes to cross-correct among cells. Here, we explored enzyme replacement therapy in a knock-out mouse model of congenital neuronal ceroid lipofuscinosis (NCL), the most severe of the NCLs in humans. The missing protease in this disorder, cathepsin D (CathD) has high levels in the central nervous system. This enzyme has the potential advantage for assessing experimental therapy in that it can be imaged using a near-infrared fluorescence (NIRF) probe activated by CathD. Injections of an AAV2/rh8 vector-encoding mouse CathD (mCathD) into both cerebral ventricles and peritoneum of newborn knock-out mice resulted in a significant increase in lifespan. Successful delivery of active CathD by the AAV2/rh8-mCathD vector was verified by NIRF imaging of mouse embryonic fibroblasts from knock-out mice in culture, as well as by ex vivo NIRF imaging of the brain and liver after gene transfer. These studies support the potential effectiveness and imaging evaluation of enzyme replacement therapy to the brain and other organs in CathD null mice via AAV-mediated gene delivery in neonatal animals.


Subject(s)
Cathepsin D/genetics , Fluorescent Dyes , Gene Transfer Techniques , Genetic Therapy/methods , Infrared Rays , Neuronal Ceroid-Lipofuscinoses/therapy , Animals , Animals, Newborn , Brain Chemistry , Dependovirus/genetics , Disease Models, Animal , Enzyme Replacement Therapy/methods , Genetic Vectors , Liver/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/genetics
6.
Cancer Gene Ther ; 17(9): 655-63, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20508670

ABSTRACT

Oncolytic herpes simplex virus (HSV) vectors have been used in early phase human clinical trials as a therapy for recurrent malignant glioblastoma. This treatment proved safe but limited improvements in patient survival were observed. The potency of these vectors might be enhanced by targeting vector infectivity to tumor cells. Glioma tumors often express a mutant form (vIII) of the epidermal growth factor receptor (EGFR) resulting in the presence of a novel epitope on the cell surface. This epitope is specifically recognized by a single-chain antibody designated MR1-1. HSV-1 infection involves initial binding to heparan sulfate (HS) on the cell surface mediated primarily by the viral envelope, glycoprotein C (gC). Here we joined the MR1-1 single-chain antibody (scFv) to the gC sequence deleted for the HS-binding domain as a means of targeting viral attachment to EGFRvIII on glial tumor cells. Virions bearing MR1-1-modified gC had fivefold increased infectivity for EGFRvIII-bearing human glioma U87 cells compared to mutant receptor-deficient cells. Further, MR1-1/EGFRvIII-mediated infection was more efficient for EGFRvIII-positive cells than was wild-type virus for either positive or negative cells. Sustained infection of EGFRvIII+ glioma cells by MR1-1-modified gC-bearing oncolytic virus, as compared to wild-type gC oncolytic virus, was also shown in subcutaneous tumors in vivo using firefly luciferase as a reporter of infection. These data show that HSV tropism can be manipulated so that virions recognize a cell-specific binding site with increased infectivity for the target cell. The retargeting of HSV infection to tumor cells should enhance vector specificity, tumor cell killing and vector safety.


Subject(s)
ErbB Receptors/metabolism , Glioma/virology , Heparitin Sulfate/metabolism , Herpesvirus 1, Human/physiology , Viral Envelope Proteins/metabolism , Virion/physiology , Animals , Cell Membrane/metabolism , Chlorocebus aethiops , Female , Genetic Vectors , Glioma/metabolism , Helper Viruses/genetics , Humans , Mice , Mice, Nude , Vero Cells
7.
Cancer Gene Ther ; 17(4): 266-74, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19834516

ABSTRACT

Schwannomas are benign tumors forming along peripheral nerves that can cause deafness, pain and paralysis. Current treatment involves surgical resection, which can damage associated nerves. To achieve tumor regression without damage to nerve fibers, we generated an HSV amplicon vector in which the apoptosis-inducing enzyme, caspase-1 (ICE), was placed under the Schwann cell-specific P0 promoter. Infection of schwannoma, neuroblastoma and fibroblastic cells in culture with ICE under the P0 promoter showed selective toxicity to schwannoma cells, while ICE under a constitutive promoter was toxic to all cell types. After direct intratumoral injection of the P0-ICE amplicon vector, we achieved marked regression of schwannoma tumors in an experimental xenograft mouse model. Injection of this amplicon vector into the sciatic nerve produced no apparent injury to the associated dorsal root ganglia neurons or myelinated nerve fibers. The P0-ICE amplicon vector provides a potential means of 'knifeless resection' of schwannoma tumors by injection of the vector into the tumor with low risk of damage to associated nerve fibers.


Subject(s)
Caspase 1/genetics , Diagnostic Imaging , Neurilemmoma/pathology , Neurilemmoma/therapy , Oncolytic Virotherapy , Promoter Regions, Genetic/genetics , Simplexvirus/genetics , Animals , Fluorescence , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/therapeutic use , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpes Simplex/metabolism , Herpes Simplex/pathology , Herpes Simplex/therapy , Humans , Luminescence , Mice , Mice, Nude , Neurilemmoma/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Schwann Cells/virology , Transduction, Genetic
8.
Cancer Gene Ther ; 16(8): 664-71, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19197327

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive type of all primary brain tumors, with an overall median survival <1 year after diagnosis. Despite introduction of multimodal treatment approaches, the prognosis has not improved significantly over the past 50 years. In this study we investigated the effect of intracerebroventricular (ICV) injection of an adeno-associated virus (AAV) vector encoding human interferon-beta (AAV-hIFN-beta) on glioblastoma growth. Recently, we found that peritumoral parenchymal transduction with an AAV-hIFN-beta was exceptionally efficient in eradicating GBM brain tumors. However, the extensive infiltration and migration displayed by glioblastoma cells in patients may leave a significant number of tumor cells outside a local therapeutic zone created by intraparenchymal delivery of AAV vectors. Here we show that pretreatment of mice by ICV infusion of an AAV-IFN-beta completely prevents tumor growth in an orthotopic model of GBM. Furthermore, ICV infusion of AAV-IFN-beta into mice bearing preestablished U87 intracranial tumors improved their survival compared to mice infused through the same route with a control AAV vector. These data suggest that ICV injection of AAV vectors encoding antitumor proteins is a promising approach deserving further consideration for the treatment of GBM.


Subject(s)
Brain Neoplasms/therapy , Dependovirus/genetics , Genetic Vectors/administration & dosage , Glioblastoma/therapy , Interferon-beta/genetics , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Genetic Therapy/methods , Genetic Vectors/metabolism , Genetic Vectors/pharmacokinetics , Glioblastoma/metabolism , Humans , Injections, Intraventricular , Interferon-beta/administration & dosage , Interferon-beta/pharmacokinetics , Male , Mice , Mice, Nude , Neoplasm Transplantation
9.
Gene Ther ; 16(2): 303-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18818671

ABSTRACT

GM1-gangliosidosis is a lysosomal storage disease (LSD) caused by an autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal). This leads to accumulation of GM1-ganglioside and its asialo derivative GA1 in the central nervous system (CNS), and progressive neurodegeneration. Therapeutic AAV-mediated gene delivery to the brain for LSDs has proven very successful in several animal models. GM1-gangliosidosis is also a prime candidate for AAV-mediated gene therapy in the CNS. As global neuropathology characterizes the most severe forms of this disease, therapeutic interventions need to achieve distribution of betagal throughout the entire CNS. Therefore, careful consideration of routes of administration and target structures from where metabolically active enzyme can be produced, released and distributed throughout the CNS, is necessary. The goal of this study was to investigate the pattern and mechanism of distribution of betagal in the adult GM1-gangliosidosis mouse brain upon hippocampal injection of an AAV vector-encoding betagal. We found evidence that three different mechanisms contribute to its distribution in the brain: (1) diffusion; (2) axonal transport within neurons from the site of production; (3) CSF flow in the perivascular space of Virchow-Robin. In addition, we found evidence of axonal transport of vector-encoded mRNA.


Subject(s)
Brain/enzymology , Gangliosidosis, GM1/enzymology , Genetic Therapy/methods , beta-Galactosidase/genetics , Animals , Axonal Transport , Dependovirus/genetics , Disease Models, Animal , Gangliosidosis, GM1/therapy , Genetic Vectors/pharmacokinetics , Hippocampus/enzymology , Mice , Mice, Knockout , Neurons/physiology , RNA, Messenger/genetics , Tissue Distribution , beta-Galactosidase/biosynthesis , beta-Galactosidase/deficiency , beta-Galactosidase/pharmacokinetics
10.
Proc Natl Acad Sci U S A ; 104(43): 17204-9, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17940007

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a CAG repeat in the huntingtin (Htt) gene. HD is autosomal dominant and, in theory, amenable to therapeutic RNA silencing. We introduced cholesterol-conjugated small interfering RNA duplexes (cc-siRNA) targeting human Htt mRNA (siRNA-Htt) into mouse striata that also received adeno-associated virus containing either expanded (100 CAG) or wild-type (18 CAG) Htt cDNA encoding huntingtin (Htt) 1-400. Adeno-associated virus delivery to striatum and overlying cortex of the mutant Htt gene, but not the wild type, produced neuropathology and motor deficits. Treatment with cc-siRNA-Htt in mice with mutant Htt prolonged survival of striatal neurons, reduced neuropil aggregates, diminished inclusion size, and lowered the frequency of clasping and footslips on balance beam. cc-siRNA-Htt was designed to target human wild-type and mutant Htt and decreased levels of both in the striatum. Our findings indicate that a single administration into the adult striatum of an siRNA targeting Htt can silence mutant Htt, attenuate neuronal pathology, and delay the abnormal behavioral phenotype observed in a rapid-onset, viral transgenic mouse model of HD.


Subject(s)
Cerebral Cortex/pathology , Gene Silencing , Genetic Therapy , Mutant Proteins/antagonists & inhibitors , Neostriatum/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cholesterol/metabolism , Dependovirus , Disease Models, Animal , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/therapy , Injections , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/pathology , Intranuclear Inclusion Bodies/ultrastructure , Mice , Motor Neuron Disease/pathology , Neostriatum/drug effects , Nerve Tissue Proteins/immunology , Neurons/pathology , Neurons/ultrastructure , Neuropil Threads/drug effects , Neuropil Threads/ultrastructure , Nuclear Proteins/immunology
11.
Gene Ther ; 14(14): 1078-91, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17460718

ABSTRACT

Vectors based on herpes simplex virus type-1 (HSV-1) permit delivery of transgenes of up to 150 kb, while the inverted terminal repeats and Rep of the adeno-associated virus (AAV) can confer site-specific integration into the AAVS1 site, which allows sustained expression of a transgene. In this study, combination of the viral elements in HSV/AAV hybrid vectors has been applied for the infectious transfer of the human lysosomal beta-galactosidase (BGAL) gene of 100 kb. Temporary expression and functional activity of beta-galactosidase (beta-gal) could be detected in human beta-gal-deficient patient and glioblastoma (Gli36) cells upon infection with the basic BGAL amplicon vector. Sustained expression of beta-gal was achieved in Gli36 cells infected with rep-plus, but not rep-minus, HSV/AAV hybrid vectors. None of five clones isolated after rep-minus hybrid vector infection showed elevated beta-gal activity or site-specific integration. In contrast, 80% of the rep-plus clones possessed beta-gal activity at least twofold greater than normal levels for up to 4 months of continuous growth, and 33% of the clones exhibited AAVS1-specific integration of the ITR-flanked transgene. One of the rep-plus clones displayed integration of the ITR cassette only at the AAVS1 site, with no sequences outside the cassette detectable and beta-gal activity fourfold above normal levels. These data demonstrate AAVS1-specific integration of an entire genomic locus and expression of the transgene from the endogenous promoter mediated by an HSV/AAV hybrid vector.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , beta-Galactosidase/genetics , Animals , Blotting, Southern/methods , Cell Line, Transformed , Chlorocebus aethiops , Fibroblasts/virology , Gangliosidosis, GM1/therapy , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Genome , Green Fluorescent Proteins/genetics , Humans , Time Factors , Transduction, Genetic/methods , Transgenes , Virus Integration , beta-Galactosidase/analysis , beta-Galactosidase/metabolism
12.
Mol Ther ; 15(1): 30-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164772

ABSTRACT

GM1-gangliosidosis is a glycosphingolipid (GSL) lysosomal storage disease caused by autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal), and characterized by accumulation of GM1-ganglioside and GA1 in the brain. Here we examined the effect of neonatal intracerebroventricular (i.c.v.) injection of an adeno-associated virus (AAV) vector encoding mouse betagal on enzyme activity and brain GSL content in GM1-gangliosidosis (betagal(-/-)) mice. Histological analysis of betagal distribution in 3-month-old AAV-treated betagal(-/-) mice showed that enzyme was present at high levels throughout the brain. Biochemical quantification showed that betagal activity in AAV-treated brains was 7- to 65-fold higher than in wild-type controls and that brain GSL levels were normalized. Cerebrosides and sulfatides, which were reduced in untreated betagal(-/-) mice, were restored to normal levels by AAV treatment. In untreated betagal(-/-) brains, cholesterol was present at normal levels but showed abnormal cellular distribution consistent with endosomal/lysosomal localization. This feature was also corrected in AAV-treated mice. The biochemical and histological parameters analyzed in this study showed that normal brain neurochemistry was achieved in AAV-treated betagal(-/-) mice. Therefore we show for the first time that neonatal AAV-mediated gene delivery of lysosomal betagal to the brain may be an effective approach for treatment of GM1-gangliosidosis.


Subject(s)
Dependovirus/genetics , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/therapy , Genetic Therapy , Lysosomes/enzymology , beta-Galactosidase/deficiency , beta-Galactosidase/metabolism , Animals , Animals, Newborn , Chromatography, High Pressure Liquid , Gangliosidosis, GM1/enzymology , Gangliosidosis, GM1/pathology , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , beta-Galactosidase/genetics
13.
Neuroscience ; 138(2): 501-10, 2006.
Article in English | MEDLINE | ID: mdl-16414198

ABSTRACT

Adeno-associated virus (AAV) vectors have gained a preeminent position in the field of gene delivery to the normal brain through their ability to achieve extensive transduction of neurons and to mediate long-term gene expression with no apparent toxicity. In adult animals direct infusion of AAV vectors into the brain parenchyma results in highly efficient transduction of target structures. However AAV-mediated global delivery to the adult brain has been an elusive goal. In contrast, widespread global gene delivery has been obtained by i.c.v. injection of AAV1 or AAV2 in neonates. Among the novel AAV serotypes cloned and engineered for production of recombinant vectors, AAV8 has shown a tremendous potential for in vivo gene delivery with nearly complete transduction of many tissues in rodents after intravascular infusion. Here we compare the efficiency of an AAV8 serotyped vector with that of AAV1 and AAV2 serotyped vectors for the extent of gene delivery to the brain after neonatal injection into the lateral ventricles. The vectors all encoded green fluorescent protein (GFP) under control of a hybrid CMV enhancer/chicken beta-actin promoter with AAV2 inverted terminal repeats, but differed from each other with respect to the capsid type. A total of 6.8 x 10(10) genome copies were injected into the lateral ventricles of postnatal day 0 mice. Mice were killed at postnatal day 30 and brains analyzed for distribution of GFP-positive cells. AAV8 proved to be more efficient than AAV1 or AAV2 vectors for gene delivery to all of the structures analyzed, including the cerebral cortex, hippocampus, olfactory bulb, and cerebellum. Moreover the intensity of gene expression, assessed using a microarray reader, was considerably higher for AAV8 in all structures analyzed. In conclusion, the enhanced transduction achieved by AAV8 compared with AAV1 and AAV2 indicates that AAV8 is the superior serotype for gene delivery to the CNS.


Subject(s)
Brain/physiology , Dependovirus/genetics , Transfection/methods , Animals , Animals, Newborn , Base Sequence , Brain/growth & development , Capsid , Cell Line , Cerebellum/physiology , Cerebral Cortex/physiology , DNA Primers , Dependovirus/classification , Genetic Vectors , Hippocampus/physiology , Humans , Kidney , Mice , Mice, Inbred C57BL , Olfactory Bulb , Serotyping
14.
Neuroscience ; 125(3): 651-61, 2004.
Article in English | MEDLINE | ID: mdl-15099679

ABSTRACT

TorsinA is a novel protein identified in the search for mutations underlying the human neurologic movement disorder, early onset torsion dystonia. Relatively little is understood about the normal function of torsinA or the physiological effects of the codon deletion associated with most cases of disease. Overexpression of wild-type torsinA in cultured cells by DNA transfection results in a reticular distribution of immunoreactive protein that co-localizes with endoplasmic reticulum resident chaperones, while the dystonia-related mutant form accumulates within concentric membrane whorls and nuclear-associated membrane stacks. In this study we examined the biogenesis of mutant torsinA-positive membrane inclusions using tetracycline-regulated herpes simplex virus amplicon vectors. At low expression levels, mutant torsinA was localized predominantly around the nucleus, while at high levels it was also concentrated within cytosolic spheroid inclusions. In contrast, the distribution of wild-type torsinA did not vary, appearing diffuse and reticular at all expression levels. These observations are consistent with descriptions of inducible membrane synthesis in other systems in which cytosolic membrane whorls are derived from multilayered membrane stacks that first form around the nuclear envelope. These results also suggest that formation of mutant torsinA-positive inclusions occurs at high expression levels in culture, whereas the perinuclear accumulation of the mutant protein is present even at low expression levels that are more likely to resemble those of the endogenous protein. These nuclear-associated membrane structures enriched in mutant torsinA may therefore be of greater relevance to understanding how the dystonia-related mutation compromises cellular physiology.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , Inclusion Bodies/metabolism , Intracellular Membranes/metabolism , Molecular Chaperones/metabolism , Organelles/metabolism , Animals , Biomarkers , Carrier Proteins/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/pathology , Cytosol/metabolism , Cytosol/pathology , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/metabolism , Dystonia Musculorum Deformans/physiopathology , Genes, Reporter/genetics , Genetic Vectors/genetics , Herpes Simplex/genetics , Humans , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Intracellular Membranes/pathology , Molecular Chaperones/genetics , Mutation/genetics , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Organelles/genetics , Organelles/pathology , Tetracycline/pharmacology , Transgenes/genetics
15.
Hum Gene Ther ; 12(14): 1771-83, 2001 Sep 20.
Article in English | MEDLINE | ID: mdl-11560770

ABSTRACT

Mutations in the alpha-chain of lysosomal hexosaminidase (EC 3.2.1.52) underlie two distinct biochemical phenotypes known as variant B and variant B1 of G(M2) gangliosidosis. This paper shows that the transduction of human B1-type fibroblasts (producing catalytically inactive alpha-chains) with a retroviral vector encoding the human hexosaminidase alpha-chain leads to a complete correction of HexA (alpha beta dimer) activity with both synthetic and natural substrates. The alpha-subunit overexpression leads to a partial HexB (beta beta dimer) depletion corresponding to about 10% of control HexB activity. The newly synthesized enzyme is correctly processed and targeted to the lysosomes in transduced cells. The high levels of recombinant enzyme correctly produced the metabolic defect, enabling the cells efficiently to degrade the accumulated storage product present in lysosomes. The transduced fibroblasts are also able to secrete HexA efficiently into the culture medium. Moreover, transfer of the human transgene product to B1-type deficient fibroblasts lead to an increase of activity against 4MUGS, the alpha-chain specific synthetic substrate, up to 30% of the control mean activity level. This level of activity might be sufficient to restore the normal ganglioside G(M2) metabolism in recipient cells. The data obtained demonstrate that B1-type phenotype can be efficiently corrected by retrovirus-mediated gene transfer.


Subject(s)
DNA, Complementary/metabolism , Fibroblasts/metabolism , G(M2) Ganglioside/genetics , Gangliosidoses, GM2/genetics , Gene Transfer Techniques , Retroviridae/genetics , beta-N-Acetylhexosaminidases/genetics , 3T3 Cells , Animals , Cell Line , Dimerization , Electrophoresis, Polyacrylamide Gel , G(M2) Ganglioside/metabolism , Gangliosidoses, GM2/metabolism , Genetic Vectors , Hexosaminidase A , Hexosaminidase B , Humans , Immunoglobulin M/metabolism , Lysosomes/metabolism , Mice , Microscopy, Fluorescence , Mutation , Phenotype , Precipitin Tests , Recombinant Proteins/metabolism , Temperature , Time Factors , Transduction, Genetic , Transgenes , beta-N-Acetylhexosaminidases/chemistry
16.
Mol Ther ; 1(4): 347-57, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10933953

ABSTRACT

Cellular delivery of a replication-conditional herpes simplex virus type 1 (HSV-1) vector provides a means for gene therapy of invasive tumor cells. LacZ-bearing neural precursor cells, which can migrate and differentiate in the brain, were infected with a ribonucleotide reductase-deficient HSV-1 mutant virus (rRp450) that replicates only in dividing cells. Replication of rRp450 in neural precursor cells was blocked prior to implantation into the tumor by growth arrest in late G1 phase through treatment with mimosine. Viral titers in the medium of mimosine-treated, rRp450-infected neural precursor cells were below detection levels 3 days after infection. In culture, after removal of mimosine and passaging, cells resumed growth and replication of rRp450 so that, 7 days later, virus was present in the medium and cell death was evident. Mimosine-treated neural precursor cells injected into established intracerebral CNS-1 gliomas in nude mice migrated extensively throughout the tumor and into the surrounding parenchyma beyond the tumor over 3 days. Mimosine-treated neural precursor cells, infected with rRp450 and injected into intracerebral CNS-1 tumors, also migrated within the tumor with the appearance of foci of HSV-thymidine kinase-positive (TK+) cells, presumably including tumor cells, distributed throughout the tumor and in the surrounding parenchyma over a similar period. This migratory cell delivery method has the potential to expand the range of delivery of HSV-1 vectors to tumor cells in the brain.


Subject(s)
Brain Neoplasms/therapy , Genetic Vectors , Glioma/therapy , Herpesvirus 1, Human/genetics , Neurons/virology , Stem Cells/virology , Animals , Brain Neoplasms/pathology , Cell Movement , Ganciclovir/pharmacology , Genes, Viral , Genetic Therapy/methods , Glioma/pathology , Herpes Simplex Virus Protein Vmw65/genetics , Herpesvirus 1, Human/physiology , Mice , Mice, Nude , Mimosine/pharmacology , Mutation , Neurons/cytology , Neurons/drug effects , Ribonucleotide Reductases/genetics , Stem Cells/cytology , Stem Cells/drug effects , Thymidine Kinase/genetics , Virus Replication/drug effects
18.
J Neuropathol Exp Neurol ; 59(7): 607-13, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10901232

ABSTRACT

A large animal tumor model for anaplastic glioma has been recently developed using immunotolerant allogeneic Beagle dogs and an established canine glioma cell line, J3T. This model offers advantages in terms of tumor morphology and similarity to human anaplastic glioma. The present study was aimed at evaluating the biological characteristics of the J3T canine glioma cell line as related to experimental gene therapy studies. Furthermore, development and morphology of canine brain tumors in a xenogeneic immunodeficient SCID mouse model was investigated. It was demonstrated that cultured J3T cells can be efficiently infected by adenovirus (AV), herpes-simplex type I (HSV), or retrovirus (RV) vectors, as well as by non-virus vectors such as cationic liposome/DNA complexes. Thus, in terms of infectability and transfectability, J3T cells seem to be closer to human glioma than the 9L rodent gliosarcoma. Cytotoxicity of selection antibiotics such as G418, puromycin, and hygromycin on J3T cells essentially resemble cytotoxicity seen with other established glioma lines, for example, 9L, U87, or U343. RV-mediated HSV-TK/GCV gene therapy demonstrated comparable LD50 for TK-expressing and control (non-expressing) J3T and 9L cells treated with Ganciclovir. Further, it was proven that J3T cells are tumorigenic and may grow heterotopically and orthotopically in a xenogeneic immunodeficient host, the SCID mouse, although morphology and growth pattern of these xenogeneic tumors differ from the demonstrated invasive phenotype in the Beagle dog.


Subject(s)
Brain Neoplasms , Cell Culture Techniques/methods , Cinnamates , Glioblastoma , Neoplasms, Experimental , 3T3 Cells , Adenoviridae/genetics , Animals , Anti-Bacterial Agents/pharmacology , Antiviral Agents/pharmacology , Cell Division/drug effects , Cell Division/genetics , Chlorocebus aethiops , Dogs , Drug Resistance, Microbial , Ganciclovir/pharmacology , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Gene Transfer Techniques , Genetic Therapy , Gliosarcoma , Herpesvirus 1, Human/genetics , Humans , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , Kidney/cytology , Male , Mice , Mice, SCID , Neoplasm Transplantation , Rats , Thymidine Kinase/genetics , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/transplantation , Vero Cells
19.
Hum Gene Ther ; 11(10): 1429-38, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910140

ABSTRACT

Subcutaneous vaccination therapy with glioma cells, which are retrovirally transduced to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF), has previously proven effective in C57BL/6 mice harboring intracerebral GL261 gliomas. However, clinical ex vivo gene therapy for human gliomas would be difficult, as transgene delivery via retroviral vectors occurs only in dividing cells and ex vivo glioma cells have a low growth fraction. To circumvent this problem, a helper virus-free herpes simplex virus type 1 (HSV-1) amplicon vector was used. When primary cultures of human glioblastoma cells were infected with HSV-1 amplicon vectors at an MOI of 1, more than 90% of both dividing and nondividing cells were transduced. When cells were infected with an amplicon vector, HSVGM, bearing the GM-CSF cDNA in the presence of Polybrene, GM-CSF secretion into the medium during the first 24 hr after infection was 1026 ng/10(6) cells, whereas mock-infected cells did not secrete detectable GM-CSF. Subcutaneous vaccination of C57BL/6 mice with 5 x 10(5) irradiated HSVGM-transduced GL261 cells 7 days prior to intracerebral implantation of 10(6) wild-type GL261 cells yielded 60% long-term survivors (>80 days), similar to the 50% long-term survivors obtained by vaccination with retrovirally GM-CSF-transduced GL261 cells. In contrast, animals vaccinated with the same number of nontranduced GL261 cells or with GL261 cells infected with helper virus-free packaged HSV-1 amplicon vectors carrying no transgene showed only 10% long-term survivors. In conclusion, helper virus-free HSV-1 amplicon vectors appear to be effective for cytokine-enhanced vaccination therapy of glioma, with the advantages that both dividing and nondividing tumor cells can be infected, no viral proteins are expressed, and these vectors are safe and compatible with clinical use.


Subject(s)
Cancer Vaccines , Glioma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Helper Viruses/genetics , Herpesvirus 1, Human/genetics , Neoplasms, Experimental/therapy , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Hexadimethrine Bromide/pharmacology , Humans , Lac Operon , Mice , Mice, Inbred C57BL , Time Factors , Transduction, Genetic , Transgenes/genetics , Tumor Cells, Cultured , Vero Cells
20.
Hum Gene Ther ; 11(5): 715-27, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10757351

ABSTRACT

Mutations in the lysosomal acid beta-galactosidase (EC 3.2.1.23) underlie two different disorders: GM1 gangliosidosis, which involves the nervous system and visceral organs to varying extents, and Morquio's syndrome type B (Morquio B disease), which is a skeletal-connective tissue disease without any CNS symptoms. This article shows that transduction of human GM1 gangliosidosis fibroblasts with retrovirus vectors encoding the human acid beta-galactosidase cDNA leads to complete correction of the enzymatic deficiency. The newly synthesized enzyme is correctly processed and targeted to the lysosomes in transduced cells. Cross-correction experiments using retrovirus-modified cells as enzyme donors showed, however, that the human enzyme is transferred at low efficiencies. Experiments using a different retrovirus vector carrying the human cDNA confirmed this observation. Transduction of human GM1 fibroblasts and mouse NIH 3T3 cells with a retrovirus vector encoding the mouse beta-galactosidase cDNA resulted in high levels of enzymatic activity. Furthermore, the mouse enzyme was found to be transferred to human cells at high efficiency. Enzyme activity measurements in medium conditioned by genetically modified cells suggest that the human beta-galactosidase enzyme is less efficiently released to the extracellular space than its mouse counterpart. This study suggests that lysosomal enzymes, contrary to the generalized perception in the field of gene therapy, may differ significantly in their properties and provides insights for design of future gene therapy interventions in acid beta-galactosidase deficiency.


Subject(s)
Gangliosidosis, GM1/enzymology , Gene Transfer Techniques , Retroviridae/genetics , beta-Galactosidase/deficiency , 3T3 Cells/enzymology , 3T3 Cells/virology , Animals , Culture Media, Conditioned , Fibroblasts/enzymology , Fibroblasts/virology , Gangliosidosis, GM1/genetics , Genetic Vectors , Humans , Lysosomes/metabolism , Mice , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...