Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Gene Ther ; 21(12): 1029-40, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231174

ABSTRACT

Astrocytes are an attractive cell target for gene therapy, but the validation of new therapeutic candidates is needed. We determined whether adeno-associated viral (AAV) vector-mediated overexpression of glutamine synthetase (GS) or excitatory amino-acid transporter 2 (EAAT2), or expression of microRNA targeting adenosine kinase (miR-ADK) in hippocampal astrocytes in the rat brain could modulate susceptibility to kainate-induced seizures and neuronal cell loss. Transgene expression was found predominantly in astrocytes following direct injection of glial-targeting AAV9 vectors by 3 weeks postinjection. ADK expression in miR-ADK vector-injected rats was reduced by 94-96% and was associated with an ~50% reduction in the duration of kainate-induced seizures and greater protection of dentate hilar neurons but not CA3 neurons compared with miR-control vector-injected rats. In contrast, infusion of AAV-GS and EAAT2 vectors did not afford any protection against seizures or neuronal damage as the level of transcriptional activity of the glial fibrillary acidic promoter was too low to drive any significant increase in transgenic GS or EAAT2 relative to the high endogenous levels of these proteins. Our findings support ADK as a prime therapeutic target for gene therapy of temporal lobe epilepsy and suggest that alternative approaches including the use of stronger glial promoters are needed to increase transgenic GS and EAAT2 expression to levels that may be required to affect seizure induction and propagation.


Subject(s)
Adenosine Kinase/genetics , Epilepsy, Temporal Lobe/therapy , Excitatory Amino Acid Transporter 2/genetics , Gene Targeting , Genetic Therapy/methods , Glutamate-Ammonia Ligase/genetics , Adenosine Kinase/metabolism , Animals , Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression Regulation , Genetic Vectors , Glutamate-Ammonia Ligase/metabolism , Hippocampus/metabolism , Kainic Acid/adverse effects , Male , Neuroglia/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Transgenes/genetics
2.
Cancer Gene Ther ; 21(10): 434-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25236494

ABSTRACT

Most chemotherapy regimens rely on systemic administration of drugs leading to a wide array of toxicities. Using viral-vector-mediated gene modification of muscle tissues, we have developed a method for gene-directed enzyme prodrug therapy that allows for localized drug administration. An inactive prodrug of geldanamycin was activated locally for inhibition of tumor growth without systemic toxicities. A recombinant adeno-associated virus (rAAV) was used to deliver ß-galactosidase (LacZ) to the treatment group and green fluorescent protein to the control group. After 1 week, both groups received adenocarcinoma cells in the same location as the previous rAAV injection. The geldanamycin prodrug was administered 1 h later via intraperitoneal injection. Tumor growth was significantly suppressed in animals whose muscles were gene modified to express ß-galactosidase compared with the control. Serum assay to access hepatotoxicity resulted in no significant differences between the animals treated with the inactive or activated form of geldanamycin, indicating minimal damage to non-target organs. Using gene-directed enzyme prodrug therapy, in combination with novel recombinant AAV vectors, we have developed a method for localized activation of chemotherapeutic agents that limits the toxicities seen with traditional systemic administration of these potent drugs.


Subject(s)
Enzymes/genetics , Enzymes/metabolism , Genes, Transgenic, Suicide , Neoplasms/genetics , Neoplasms/pathology , Prodrugs/metabolism , Prodrugs/pharmacology , Allografts , Animals , Antibiotics, Antineoplastic/pharmacology , Benzoquinones/pharmacology , Cell Proliferation/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/drug effects , Humans , Lactams, Macrocyclic/pharmacology , Liver Function Tests , Mice , Neoplasms/therapy , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
3.
J Bone Miner Res ; 29(10): 2238-49, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24535841

ABSTRACT

Chronic stress and depression have adverse consequences on many organ systems, including the skeleton, but the mechanisms underlying stress-induced bone loss remain unclear. Here we demonstrate that neuropeptide Y (NPY), centrally and peripherally, plays a critical role in protecting against stress-induced bone loss. Mice lacking the anxiolytic factor NPY exhibit more anxious behavior and elevated corticosterone levels. Additionally, following a 6-week restraint, or cold-stress protocol, Npy-null mice exhibit three-fold greater bone loss compared to wild-type mice, owing to suppression of osteoblast activity. This stress-protective NPY pathway acts specifically through Y2 receptors. Centrally, Y2 receptors suppress corticotropin-releasing factor expression and inhibit activation of noradrenergic neurons in the paraventricular nucleus. In the periphery, they act to control noradrenaline release from sympathetic neurons. Specific deletion of arcuate Y2 receptors recapitulates the Npy-null stress response, coincident with elevated serum noradrenaline. Importantly, specific reintroduction of NPY solely in noradrenergic neurons of otherwise Npy-null mice blocks the increase in circulating noradrenaline and the stress-induced bone loss. Thus, NPY protects against excessive stress-induced bone loss, through Y2 receptor-mediated modulation of central and peripheral noradrenergic neurons.


Subject(s)
Bone Resorption/etiology , Neuropeptide Y/metabolism , Norepinephrine/metabolism , Stress, Psychological/complications , Animals , Anxiety/complications , Arcuate Nucleus of Hypothalamus/metabolism , Behavior, Animal , Bone Resorption/blood , Mice , Models, Biological , Neurons/metabolism , Neuropeptide Y/blood , Organ Specificity , Protective Agents/metabolism , Receptors, Neuropeptide Y/metabolism , Signal Transduction , Stress, Psychological/blood
4.
Gene Ther ; 17(5): 643-52, 2010 May.
Article in English | MEDLINE | ID: mdl-20220782

ABSTRACT

Neuropeptide Y (NPY) is an endogenous peptide with powerful anticonvulsant properties. Its overexpression in the rat hippocampus, mediated by the local application of recombinant adeno-associated viral (rAAV) vectors carrying the human NPY gene, results in significant reduction of seizures in acute and chronic seizure models. In this study, we characterized a more efficient rAAV-NPY vector to improve cell transfection in the injected area. The changes included pseudotyping with the AAV vector serotype 1 (rAAV1), and using the strong constitutive hybrid CBA promoter, which contains a cytomegalovirus enhancer and chicken beta-actin promoter sequences. We compared NPY expression and the associated anticonvulsant effects of this new vector, with those mediated by the former rAAV vector with chimeric serotype 1/2 (rAAV1/2). In addition, we investigated whether rAAV serotype 1 vector-mediated chronic NPY overexpression causes behavioural deficits that may detract from the clinical utility of this therapeutic approach. We report that rAAV-NPY serotype 1 vector has significantly improved anticonvulsant activity when compared with serotype 1/2 vector, as assessed by measuring EEG seizure activity in kainic acid treated rats. rAAV1-mediated NPY overexpression in naive rats did not result in alterations of physiological functions such as learning and memory, anxiety and locomotor activity. In addition, we did not observe glia activation, or humoral immune responses against serotype 1 vector, which could inactivate gene expression. Our findings show that rAAV1-NPY vector with the CBA promoter mediates powerful anticonvulsant effects and seems to be safe in rodents, thus it may be considered a vector of choice for possible clinical applications.


Subject(s)
Epilepsy, Temporal Lobe/therapy , Genetic Therapy/methods , Hippocampus/metabolism , Neuropeptide Y/genetics , Seizures/therapy , Transduction, Genetic/methods , Actins/genetics , Animals , Dependovirus , Epilepsy, Temporal Lobe/physiopathology , Genetic Vectors , Immunity, Humoral , Kainic Acid/adverse effects , Learning , Male , Memory , Motor Activity , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Seizures/physiopathology
5.
Neuroscience ; 151(2): 352-60, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18082971

ABSTRACT

Neuronal activity regulated pentraxin (Narp) is a secreted, synaptic protein that has been implicated in modulating synaptic transmission. However, it is unclear how Narp secretion is regulated. Since we noted prominent Narp immunostaining in vasopressin neurons of the hypothalamus and in the posterior pituitary, we assessed whether it, like vasopressin, is released into the systemic circulation in an activity-dependent fashion. Consistent with this hypothesis, electron microscopic studies of the posterior pituitary demonstrated that Narp is located in secretory vesicles containing vasopressin. Using affinity chromatography, we detected Narp in plasma and found that these levels are markedly decreased by hypophysectomy. In addition, we confirmed that injection of a viral Narp construct into the hypothalamus restores plasma Narp levels in Narp knockout mice. In checking for activity-dependent secretion of Narp from the posterior pituitary, we found that several stimuli known to trigger vasopressin release, i.e. hypovolemia, dehydration and endotoxin, elevate plasma Narp levels. Taken together, these findings provide compelling evidence that Narp is secreted from vasopressin neurons in an activity-dependent fashion.


Subject(s)
C-Reactive Protein/metabolism , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/physiology , Vasopressins/physiology , Adenoviridae/genetics , Animals , Chromatography, Affinity , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dehydration/physiopathology , Genetic Vectors , Humans , Hypovolemia/physiopathology , Immunohistochemistry , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Microscopy, Electron , Microscopy, Immunoelectron , Nerve Tissue Proteins/blood , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
6.
Gene Ther ; 11(9): 797-804, 2004 May.
Article in English | MEDLINE | ID: mdl-14961066

ABSTRACT

In vivo gene transfer of glutamate decarboxylase (GAD) has been explored as a means of inducing or increasing the production of the inhibitory amino-acid neurotransmitter, GABA. This strategy has been applied to neuroprotection, seizure prevention, and neuromodulation. In the present experiment, AAV2 was used to transfer the genes for green fluorescence protein (GFP) and GAD65 into the lateral nucleus of the rat hypothalamus. Microinjection of 500 nl of AAV2 resulted in transduction of a 0.25+/-0.04 mm(3) with targeting errors of X=0.48 mm, Y=0.18 mm, Z=0.37 mm using standard stereotactic technique. Pre- and postinjection food and water consumption, urine and feces production, and weight were recorded. In comparison with rAAVCAGGFP- and PBS-injected animals, rats treated with rAAVCAGGAD65 demonstrated reduced weight gain (P<0.014) and transiently reduced daily food consumption (P<0.007) during the postoperative period. No changes in water consumption or waste production were recorded. Effective GAD65 gene transfer was confirmed with in situ hybridization using a probe to the woodchuck post-transcriptional regulatory element sequence included in the vector. These findings suggest that increased GABA production in lateral nucleus of the hypothalamus induced by GAD65 gene transfer may reduce weight gain through reduced feeding.


Subject(s)
Feeding Behavior/physiology , Gene Transfer Techniques , Glutamate Decarboxylase/metabolism , Hypothalamic Area, Lateral/enzymology , Adenoviridae/genetics , Animals , Eating/genetics , Gene Targeting/methods , Glutamate Decarboxylase/genetics , Hypothalamic Area, Lateral/physiology , Microinjections/methods , Rats , Rats, Wistar , Stereotaxic Techniques , Weight Gain/genetics , Weight Gain/physiology , gamma-Aminobutyric Acid/biosynthesis
7.
Neuroscience ; 122(3): 773-84, 2003.
Article in English | MEDLINE | ID: mdl-14622920

ABSTRACT

Gephyrin is a postsynaptic clustering molecule that forms a protein scaffold to anchor inhibitory neurotransmitter receptors at the postsynaptic membrane of neurons. Gephyrin was first identified as a protein component of the glycine receptor complex and is also colocalized with several GABAA receptor subunits in rodent brain. We have studied the distribution of gephyrin and glycine receptor subunits in the human brainstem and spinal cord using immunohistochemistry at light and confocal laser scanning microscopy levels. This study demonstrates the novel localization of gephyrin with glycine receptors in the human brainstem and spinal cord. Colocalization of immunoreactivities for gephyrin and glycine receptor subunits was detected in the dorsal and ventral horns of the spinal cord, the hypoglossal nucleus and the medial vestibular nucleus of the medulla. The results clearly establish that gephyrin is ubiquitously distributed and is colocalized, with a large proportion of glycine receptor subunits in the human brainstem and spinal cord. We therefore suggest that gephyrin functions as a clustering molecule for major subtypes of glycine receptors in the human CNS.


Subject(s)
Brain Stem/metabolism , Carrier Proteins/metabolism , Immunohistochemistry/methods , Membrane Proteins/metabolism , Receptors, Glycine/metabolism , Spinal Cord/metabolism , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
8.
Neuroscience ; 116(1): 145-56, 2003.
Article in English | MEDLINE | ID: mdl-12535948

ABSTRACT

Gephyrin is an ubiquitously expressed protein that, in the central nervous system, generates a protein scaffold to anchor inhibitory neurotransmitter receptors in the postsynaptic membrane. It was first identified as a protein component of the glycine receptor complex. Recent studies have demonstrated that gephyrin is colocalized with several subtypes of GABA(A) receptors and is part of postsynaptic GABA(A) receptor clusters. Here, we describe a study of the regional and cellular distribution of gephyrin in the human brain, determined by immunohistochemical localisation at the light and confocal laser scanning microscopic levels. At the regional level, gephyrin immunoreactivity was observed in most of the major brain regions examined. The most intense staining was in the cerebral cortex, hippocampus and caudate-putamen, in various brainstem nuclei with more moderate levels in the thalamus and cerebellum. At the cellular level gephyrin immunoreactivity was present on the plasma membranes of the soma and dendrites of pyramidal neurons throughout the various cortical regions examined. In the hippocampus, intense staining was observed on the granule cells of the dentate gyrus, and neurons of the CA1 and CA3 regions showed intense punctate gephyrin staining on their apical dendrites and cell bodies. Gephyrin immunoreactivity was also observed on neurons in the thalamus, globus pallidus and substantia nigra. In the putamen intense labelling of the striosomes was observed; most of the medium-sized neurons in the caudate-putamen were weakly labelled and many large neurons of the striatum were conspicuously stained. Many of the brainstem nuclei, notably the dorsal motor nucleus of the vagus, hypoglossal nucleus, trigeminal nucleus and inferior olive were all labelled with gephyrin. The spinal cord also showed high levels of gephyrin immunoreactivity. Our results demonstrate that the anchoring protein gephyrin is ubiquitously present in the human brain. We therefore suggest that gephyrin may have a central organizer role in assembling and stabilizing inhibitory postsynaptic membranes in human brain and is similar in function to those observed in the rodent brain. These findings contribute towards elucidating the role of gephyrin in the human brain.


Subject(s)
Brain Chemistry , Carrier Proteins/analysis , Membrane Proteins/analysis , Neurons/chemistry , Synapses/chemistry , Aged , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neural Inhibition
9.
Genomics ; 78(1-2): 3-6, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11707065

ABSTRACT

Over the past decade, viral vectors have slowly gained mainstream acceptance in the neuroscience and genetics communities for the in vivo study of gene function [1]. Using stereotactic techniques, it is possible to characterize neuroanatomical relationships through the delivery of neurotropic viral vectors to specific brain regions. More sophisticated studies combine viral vectors with other methods of genetic manipulation such as germline transgenic mice. As more is learned about the properties of different viral vectors, it has become possible to use viral vectors to test hypotheses about the function of genes, through targeted in vivo delivery to the central nervous system (CNS). The effects of gene expression in the brain can be measured on the molecular, biochemical, electrophysiological, morphological, and behavioral levels. We propose that viral vectors should be considered as part of an integrated functional genomics platform in the CNS.


Subject(s)
Genetic Vectors/genetics , Genomics/methods , Viruses/genetics , Animals , Animals, Genetically Modified , Humans , Research Design
10.
Trends Neurosci ; 24(12): 706-12, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11718875

ABSTRACT

A fundamental problem in neuroscience has been the creation of suitable in vivo model systems to study basic neurological phenomena and pathology of the central nervous system (CNS). Somatic cell genetic engineering with viral vectors provides a versatile tool to model normal brain physiology and a variety of neurological diseases.


Subject(s)
Brain/physiology , Gene Transfer Techniques , Viruses/genetics , Animals , Brain Diseases/physiopathology , Genetic Vectors , Mammals
11.
Gene Ther ; 8(17): 1323-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11571569

ABSTRACT

This study compared a range of mammalian CNS expression cassettes in recombinant adeno-associated virus (AAV-2) vectors using strong endogenous promoter sequences, with or without a strong post-regulatory element and polyadenylation signal. Changes in these elements led to transgene expression varying by over three orders of magnitude. In experiments conducted in primary cell culture and in >100 stereotactically injected rats, we observed highly efficient and stable (>15 months) gene expression in neurons and limited expression in glia; the highest expression occurred with endogenous, nonviral promoters such as neuron-specific enolase and beta-actin. The packaging size of AAV-2 was maximized at 5.7 kb without impairing gene expression, as judged by direct comparison with a number of smaller AAV-2 constructs. The genomic insert size and titer were confirmed by Southern blot and quantitative PCR, and infectivity was tested by particle titer using ELISA with a conformation-dependent epitope that requires the full intact capsid. A packaging and purification protocol we describe allows for high-titer, high-capacity AAV-2 vectors that can transduce over 2 x 10(5) neurons in vivo per microliter of vector, using the strongest expression cassette.


Subject(s)
Brain/enzymology , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Luciferases/genetics , Animals , Blotting, Southern/methods , Blotting, Western/methods , Cells, Cultured , Gene Expression , Genetic Vectors/administration & dosage , Injections , Rats , Rats, Wistar
12.
Hum Gene Ther ; 12(12): 1589-91, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11529246

ABSTRACT

This gene transfer experiment is the first Parkinson's Disease (PD) protocol to be submitted to the Recombinant DNA Advisory Committee. The principal investigators have uniquely focused their careers on both pre-clinical work on gene transfer in the brain and clinical expertise in management and surgical treatment of patients with PD. They have extensively used rodent models of PD for proof-of-principle experiments on the utility of different vector systems. PD is an excellent target for gene therapy, because it is a complex acquired disease of unknown etiology (apart from some rare familial cases) yet it is characterized by a specific neuroanatomical pathology, the degeneration of dopamine neurons of the substantia nigra (SN) with loss of dopamine input to the striatum. This pathology results in focal changes in the function of several deep brain nuclei, which have been well-characterized in humans and animal models and which account for many of the motor symptoms of PD. Our original approaches, largely to validate in vivo gene transfer in the brain, were designed to facilitate dopamine transmission in the striatum using an AAV vector expressing dopamine-synthetic enzymes. Although these confirmed the safety and potential efficacy of AAV, complex patient responses to dopamine augmenting medication as well as poor results and complications of human transplant studies suggested that this would be a difficult and potentially dangerous clinical strategy using current approaches. Subsequently, we and others investigated the use of growth factors, including GDNF. These showed some encouraging effects on dopamine neuron survival and regeneration in both rodent and primate models; however, uncertain consequences of long-term growth factor expression and question regarding timing of therapy in the disease course must be resolved before any clinical study can be contemplated. We now propose to infuse into the subthalamic nucleus (STN) recombinant AAV vectors expressing the two isoforms of the enzyme glutamic acid decarboxylase (GAD-65 and GAD-67), which synthesizes the major inhibitory neurotransmitter in the brain, GABA. The STN is a very small nucleus (140 cubic mm or 0.02% of the total brain volume, consisting of approximately 300,000 neurons) which is disinhibited in PD, leading to pathological excitation of its targets, the internal segment of the globus pallidus (GPi) and substantia nigra pars reticulata (SNpr). Increased GPi/SNpr outflow is believed responsible for many of the cardinal symptoms of PD, i.e., tremor, rigidity, bradykinesia, and gait disturbance. A large amount of data based on lesioning, electrical stimulation, and local drug infusion studies with GABA-agonists in human PD patients have reinforced this circuit model of PD and the central role of the STN. Moreover, the closest conventional surgical intervention to our proposal, deep brain stimulation (DBS) of the STN, has shown remarkable efficacy in even late stage PD, unlike the early failures associated with recombinant GDNF infusion or cell transplantation approaches in PD. We believe that our gene transfer strategy will not only palliate symptoms by inhibiting STN activity, as with DBS, but we also have evidence that the vector converts excitatory STN projections to inhibitory projections. This additional dampening of outflow GPi/SNpr outflow may provide an additional advantage over DBS. Moreover, of perhaps the greatest interest, our preclinical data suggests that this strategy may also be neuroprotective, so this therapy may slow the degeneration of dopaminergic neurons. We will use both GAD isoforms since both are typically expressed in inhibitory neurons in the brain, and our data suggest that the combination of both isoforms is likely to be most beneficial. Our preclinical data includes three model systems: (1) old, chronically lesioned parkinsonian rats in which intraSTN GAD gene transfer results not only in improvement in both drug-induced asymmetrical behavior (apomorphine symmetrical rotations), but also in spontaneous behaviors. In our second model, GAD gene transfer precedes the generation of a dopamine lesion. Here GAD gene transfer showed remarkable neuroprotection. Finally, we carried out a study where GAD-65 and GAD-67 were used separately in monkeys that were resistant to MPTP lesioning and hence showed minimal symptomatology. Nevertheless GAD gene transfer showed no adverse effects and small improvements in both Parkinson rating scales and activity measures were obtained. In the proposed clinical trial, all patients will have met criteria for and will have given consent for STN DBS elective surgery. Twenty patients will all receive DBS electrodes, but in addition they will be randomized into two groups, to receive either a solution containing rAAV-GAD, or a solution which consists just of the vector vehicle, physiological saline. Patients, care providers, and physicians will be blind as to which solution any one patient receives. All patients, regardless of group, will agree to not have the DBS activated until the completion and unblinding of the study. Patients will be assessed with a core clinical assessment program modeled on the CAPSIT, and in addition will also undergo a preop and several postop PET scans. At the conclusion of the study, if any patient with sufficient symptomatic improvement will be offered DBS removal if they so desire. Any patients with no benefit will simply have their stimulators activated, which would normally be appropriate therapy for them and which requires no additional operations. If any unforeseen symptoms occur from STN production of GABA, this might be controlled by blocking STN GABA release with DBS, or STN lesioning could be performed using the DBS electrode. Again, this treatment would not subject the patient to additional invasive brain surgery. The trial described here reflects an evolution in our thinking about the best strategy to make a positive impact in Parkinson Disease by minimizing risk and maximizing potential benefit. To our knowledge, this proposal represents the first truly blinded, completely controlled gene or cell therapy study in the brain, which still provides the patient with the same surgical procedure which they would normally receive and should not subject the patient to additional surgical procedures regardless of the success or failure of the study. This study first and foremost aims to maximally serve the safety interests of the individual patient while simultaneously serving the public interest in rigorously determining in a scientific fashion if gene therapy can be effective to any degree in treating Parkinson's disease.


Subject(s)
Clinical Protocols , Electric Stimulation Therapy/methods , Gene Transfer Techniques , Genetic Therapy/legislation & jurisprudence , Genetic Therapy/methods , Glutamate Decarboxylase/genetics , Parkinson Disease/therapy , Cell Nucleus/metabolism , Combined Modality Therapy , Dependovirus/genetics , Genetic Vectors , Glutamate Decarboxylase/chemistry , Humans , Protein Isoforms
13.
Cancer Res ; 61(12): 4827-36, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406559

ABSTRACT

The fragile histidine triad (FHIT) gene is a tumor suppressor gene that is altered by deletion in a large fraction of human tumors, including pancreatic cancer. To evaluate the potential of FHIT gene therapy, we developed recombinant adenoviral and adenoassociated viral (AAV) FHIT vectors and tested these vectors in vitro and in vivo for activity against human pancreatic cancer cells. Our data show that viral FHIT gene delivery results in apoptosis by activation of the caspase pathway. Furthermore, Fhit overexpression enhances the susceptibility of pancreatic cancer cells to exogenous inducers of apoptosis. In vivo results show that FHIT gene transfer delays tumor growth and prolongs survival in a murine model mimicking human disease.


Subject(s)
Acid Anhydride Hydrolases , Apoptosis/genetics , Neoplasm Proteins , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proteins/genetics , Adenoviridae/genetics , Animals , Caspases/metabolism , Cell Cycle/physiology , Cell Division/genetics , DNA Fragmentation , Female , Gene Transfer Techniques , Genes, Tumor Suppressor , Genetic Therapy , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Mitochondria/physiology , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Signal Transduction/physiology , Transduction, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Proc Natl Acad Sci U S A ; 98(6): 3346-51, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248081

ABSTRACT

The tumor suppressor gene FHIT spans a common fragile site and is highly susceptible to environmental carcinogens. FHIT inactivation and loss of expression is found in a large fraction of premaligant and malignant lesions. In this study, we were able to inhibit tumor development by oral gene transfer, using adenoviral or adenoassociated viral vectors expressing the human FHIT gene, in heterozygous Fhit(+/-) knockout mice, that are prone to tumor development after carcinogen exposure. We therefore suggest that FHIT gene therapy could be a novel clinical approach not only in treatment of early stages of cancer, but also in prevention of human cancer.


Subject(s)
Acid Anhydride Hydrolases , Dimethylnitrosamine/analogs & derivatives , Genes, Tumor Suppressor/physiology , Genetic Therapy/methods , Neoplasm Proteins/genetics , Neoplasms, Experimental/prevention & control , Adenoviridae/genetics , Animals , Carcinogens/adverse effects , Dependovirus/genetics , Dimethylnitrosamine/adverse effects , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
15.
Mol Ther ; 3(2): 225-32, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11237679

ABSTRACT

Recombinant adeno-associated viruses (rAAV) are highly efficient vectors for gene transfer into the central nervous system (CNS). However, a major hurdle for gene delivery to the mammalian brain is to achieve high-level transduction in target cells beyond the immediate injection site. Therefore, in addition to improvements in expression cassettes and viral titers, optimal injection parameters need to be defined. Here, we show that previous studies of somatic cell gene transfer to the mammalian brain have used suboptimal injection parameters, with even the lowest reported perfusion rates still excessively fast. Moreover, we evaluated the effect of local administration of mannitol to further enhance transgene expression and vector spread. Ultraslow microperfusion of rAAV, i.e., <33 nl/min, resulted in significantly higher gene expression and less injury of surrounding tissue than the previously reported rates of 100 nl/min or faster. Co-infusion of mannitol facilitated gene transfer to neurons, increasing both the total number and the distribution of transduced cells by 200-300%. Gene transfer studies in the CNS using rAAV should use very slow infusion rates and combined injection with mannitol to maximize transduction efficiency and spread.


Subject(s)
Brain/metabolism , Dependovirus/genetics , Diuretics, Osmotic/therapeutic use , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Mannitol/therapeutic use , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lectins/metabolism , Luciferases/metabolism , Male , Models, Genetic , Perfusion , Plasmids/metabolism , Rats , Rats, Wistar , Transduction, Genetic
16.
Proc Natl Acad Sci U S A ; 98(5): 2676-81, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226298

ABSTRACT

Fabry disease is a lysosomal storage disorder caused by a deficiency of the lysosomal enzyme alpha-galactosidase A (alpha-gal A). This enzyme deficiency leads to impaired catabolism of alpha-galactosyl-terminal lipids such as globotriaosylceramide (Gb3). Patients develop painful neuropathy and vascular occlusions that progressively lead to cardiovascular, cerebrovascular, and renal dysfunction and early death. Although enzyme replacement therapy and bone marrow transplantation have shown promise in the murine analog of Fabry disease, gene therapy holds a strong potential for treating this disease in humans. Delivery of the normal alpha-gal A gene (cDNA) into a depot organ such as liver may be sufficient to elicit corrective circulating levels of the deficient enzyme. To investigate this possibility, a recombinant adeno-associated viral vector encoding human alpha-gal A (rAAV-AGA) was constructed and injected into the hepatic portal vein of Fabry mice. Two weeks postinjection, alpha-gal A activity in the livers of rAAV-AGA-injected Fabry mice was 20-35% of that of the normal mice. The transduced animals continued to show higher alpha-gal A levels in liver and other tissues compared with the untouched Fabry controls as long as 6 months after treatment. In parallel to the elevated enzyme levels, we see significant reductions in Gb3 levels to near normal at 2 and 5 weeks posttreatment. The lower Gb3 levels continued in liver, spleen, and heart, up to 25 weeks with no significant immune response to the virus or alpha-gal A. Also, no signs of liver toxicity occurred after the rAAV-AGA administration. These findings suggest that an AAV-mediated gene transfer may be useful for the treatment of Fabry disease and possibly other metabolic disorders.


Subject(s)
Dependovirus/genetics , Fabry Disease/enzymology , Gene Transfer Techniques , Genetic Vectors , alpha-Galactosidase/metabolism , Animals , Cell Line , Fabry Disease/immunology , Fabry Disease/therapy , Humans , Liver/enzymology , Liver/physiopathology , Mice , Mice, Inbred C57BL , Microscopy, Electron , alpha-Galactosidase/genetics
17.
Gene Ther ; 8(22): 1675-81, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11892834

ABSTRACT

Recombinant AAV is increasingly becoming the vector of choice for many gene therapy applications in the CNS, due to its lack of toxicity and high level of sustained expression. With recent improvements in the generation of pure, high titer vector stocks, the regulation of gene expression is now a key issue for successful translation of gene therapy-based treatments to the clinic. The level of the transgene protein may need to be maintained within a narrow therapeutic window for the successful treatment of human disease. The doxycycline responsive system directs a dose-responsive, tightly regulated level of gene expression and has been used successfully in transgenic mouse models. Here, we have optimized an autoregulatory, bidirectional doxycyline responsive cassette specifically for use in rAAV. We minimized the size of the cassette and decreased the basal leakiness of the system, leading to tight regulation in the rat


Subject(s)
Brain/metabolism , Dependovirus/genetics , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Animals , Anti-Bacterial Agents/therapeutic use , Brain/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Doxycycline/therapeutic use , Genetic Vectors/genetics , Green Fluorescent Proteins , Luciferases/genetics , Luminescent Proteins/genetics , Male , Rats , Rats, Wistar , Trans-Activators
19.
J Virol ; 74(24): 11456-63, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090141

ABSTRACT

Recombinant adeno-associated virus (rAAV) is capable of directing long-term, high-level transgene expression without destructive cell-mediated immune responses. However, traditional packaging methods for rAAV vectors are generally inefficient and contaminated with replication-competent AAV (rcAAV) particles. Although wild-type AAV is not associated with any known human diseases, contaminating rcAAV particles may affect rAAV gene expression and are an uncontrolled variable in many AAV gene transfer studies. In the current study, a novel strategy was designed to both optimize AAV rep gene expression and increase vector yield, as well as simultaneously to diminish the potential of generating rcAAV particles from the helper plasmid. The strategy is based on the insertion of an additional intron in the AAV genome. In the AAV infectious clone, the intron insertion had no effects on the properties of Rep proteins expressed. Normal levels of both Rep and Cap proteins were expressed, and the replication of the AAV genome was not impaired. However, the generation of infectious rcAAV particles using intronized AAV helper was greatly diminished, which was due to the oversized AAV genome caused by the insertion of the artificial introns. Moreover, the rAAV packaging was significantly improved with the appropriate choice of intron and insertion position. The intron is another element that can regulate the rep and cap gene expression from the helper plasmid. This study provides for a novel AAV packaging system which is highly versatile and efficient. It can not only be combined with other AAV packaging systems, including rep-containing cell lines and herpes simplex virus hybrid packaging methods, but also be used in other vector systems as well.


Subject(s)
Dependovirus , Genetic Vectors , Dependovirus/genetics , Humans , Introns , Plasmids , Recombination, Genetic , Transfection
20.
Neurobiol Dis ; 7(4): 362-74, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10964607

ABSTRACT

We hypothesized that overexpression of specific glutamate receptors within the hippocampus would induce seizures and the associated cellular changes seen in temporal lobe epilepsy (TLE). The GluR6 kainate receptor was overexpressed by injecting rat hippocampi with HSVGluR6, a viral vector transducing fully edited GluR6. These animals experienced limbic seizures approximately 4 h following the injection. Control animals injected with HSVlac, a vector expressing beta-galactosidase, did not have seizures. Recordings from hippocampal CA1 pyramidal cells were performed 12 to 48 h and 1 week to 1 month postinjection. We observed nonsynaptic Na(+)-mediated bursting in 77.5% of cells 12 to 48 h following injection of HSVGluR6 but not HSVlac. The synaptic responses were normal in both groups. However, the physiological properties of cells from HSVGluR6-injected hippocampi changed over time. Two weeks following HSVGluR6 injection, synaptic bursts could be evoked, but intrinsic bursting became rare. These changes persisted for at least 1 month. We postulate that this transition from intrinsic to synaptic hyperexcitability may be important in the development of TLE.


Subject(s)
Action Potentials/physiology , Hippocampus/metabolism , Neurons/metabolism , Receptors, Kainic Acid/metabolism , Seizures/etiology , Action Potentials/drug effects , Animals , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Genetic Vectors/pharmacology , Herpesvirus 1, Human , Hippocampus/drug effects , Neurons/drug effects , Rats , Receptors, Kainic Acid/drug effects , Seizures/metabolism , Sodium Channels/drug effects , Sodium Channels/metabolism , Transfection , GluK2 Kainate Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...