Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
2.
Neurogastroenterol Motil ; 28(2): 299-305, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26564813

RESUMO

BACKGROUND: Despite the success of viral vector technology in the transduction of the central nervous system in both preclinical research and gene therapy, its potential in neurogastroenterological research remains largely unexploited. This study asked whether and to what extent myenteric and submucosal neurons in the ileum and distal colon of the mouse were transduced after neonatal systemic delivery of recombinant adeno-associated viral vectors (AAVs). METHODS: Mice were intravenously injected at postnatal day one with AAV pseudotypes AAV8 or AAV9 carrying a cassette encoding enhanced green fluorescent protein (eGFP) as a reporter under the control of a cytomegalovirus promoter. At postnatal day 35, transduction of the myenteric and submucosal plexuses of the ileum and distal colon was evaluated in whole-mount preparations, using immunohistochemistry to neurochemically identify transduced enteric neurons. KEY RESULTS: The pseudotypes AAV8 and AAV9 showed equal potential in transducing the enteric nervous system (ENS), with 25-30% of the neurons expressing eGFP. However, the percentage of eGFP-expressing colonic submucosal neurons was significantly lower. Neurochemical analysis showed that all enteric neuron subtypes, but not glia, expressed the reporter protein. Intrinsic sensory neurons were most efficiently transduced as nearly 80% of calcitonin gene-related peptide-positive neurons expressed the transgene. CONCLUSIONS & INFERENCES: The pseudotypes AAV8 and AAV9 can be employed for gene delivery to both the myenteric and the submucosal plexus, although the transduction efficiency in the latter is region-dependent. These findings open perspectives for novel preclinical applications aimed at manipulating and imaging the ENS in the short term, and in gene therapy in the longer term.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Plexo Mientérico/virologia , Neurônios/virologia , Plexo Submucoso/virologia , Transdução Genética , Animais , Colo , Dependovirus , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Injeções Intravenosas , Intestino Delgado , Camundongos , Modelos Animais
3.
Gene Ther ; 20(12): 1158-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24005577

RESUMO

Ex vivo gene therapy during coronary artery bypass grafting (CABG) holds great potential to prevent excessive smooth muscle cell (SMC) proliferation, neointima formation and graft failure. The most successful preclinical strategies to date have utilised vectors based on the species C adenovirus, Ad5, which engages the Coxsackie and Adenovirus receptor (CAR) as its primary attachment receptor. Profiling receptors on human SMCs demonstrated the absence of CAR but substantial expression of the species B receptor CD46. We performed transduction experiments using Ad5 and the CD46-utilising adenovirus Ad35, and found Ad35 significantly more efficient at transducing SMCs. To evaluate whether transduction could be further augmented, we evaluated chimeric CD46-utilising Ad5/Ad35 vectors comprising the Ad5 capsid pseudotyped with the Ad35 fibre alone (Ad5/F35) or in combination with the Ad35 penton (Ad5/F35/P35). In human smooth muscle cells (hSMCs), Ad5/F35/P35 mediated significantly higher levels of transduction than either parental vector or Ad5/F35. Ex vivo transduction experiments using mouse aortas from CD46 transgenics demonstrated that Ad5/F35/P35 was significantly more efficient at transducing SMCs than the other vectors tested. Finally, ex vivo transduction and immunofluorescent colocalisation experiments using human tissue from CABG procedures confirmed the preclinical potential of Ad5/F35/P35 as an efficient vector for vascular transduction during CABG.


Assuntos
Adenovírus Humanos/genética , Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Transdução Genética , Adenovírus Humanos/classificação , Animais , Aorta , Células Cultivadas , Ponte de Artéria Coronária , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/cirurgia , Doença da Artéria Coronariana/terapia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Terapia Genética , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/virologia , Neointima/patologia , Neointima/terapia
4.
Vet Microbiol ; 162(1): 62-7, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22986055

RESUMO

The aim of this study was to assess the pathogenicity and infection kinetics of Bluetongue virus serotype 26 (BTV-26) in goats. Out of a group of six goats housed in insect free accommodation, five were experimentally infected with BTV-26 and one was kept uninfected as an in-contact control. Samples taken throughout the study were used to determine the kinetics of infection using a pan specific BTV real time RT-PCR assay and a group specific ELISA. The five infected goats did not show clinical signs of BTV, however high levels of viral RNA were detected and virus was isolated from the blood of all 5 goats. Antibodies against BTV were first detected between 7 and 11 dpi in all 5 experimentally infected goats. Interestingly at 21 dpi viral RNA was detected in, and virus was isolated from, the blood of the in-contact control goat, which also seroconverted. These results suggest that BTV-26 replicates to high levels in goats, causing no obvious clinical disease, suggesting that goats may be the natural host for this virus. Preliminary evidence also indicates that BTV-26 may be spread by contact transmission between goats, however a more detailed study is required in order to confirm this observation.


Assuntos
Vírus Bluetongue/classificação , Vírus Bluetongue/patogenicidade , Bluetongue/virologia , Doenças das Cabras/virologia , Animais , Bluetongue/imunologia , Bluetongue/transmissão , Vírus Bluetongue/genética , Vírus Bluetongue/imunologia , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Doenças das Cabras/imunologia , Doenças das Cabras/patologia , Doenças das Cabras/transmissão , Cabras , Cinética , Masculino , Testes de Neutralização , RNA Viral/análise , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ovinos , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/patologia , Doenças dos Ovinos/transmissão , Doenças dos Ovinos/virologia
5.
Gene Ther ; 20(1): 69-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22278413

RESUMO

Correction of perinatally lethal neurogenetic diseases requires efficient transduction of several cell types within the relatively inaccessible CNS. Intravenous AAV9 delivery in mouse has achieved development stage-specific transduction of neuronal cell types, with superior neuron-targeting efficiency demonstrated in prenatal compared with postnatal recipients. Because of the clinical relevance of the non-human primate (NHP) model, we investigated the ability of AAV9 to transduce the NHP CNS following intrauterine gene therapy (IUGT). We injected two macaque fetuses at 0.9 G with 1 × 10(13) vg scAAV9-CMV-eGFP through the intrahepatic continuation of the umbilical vein. Robust green fluorescent protein (GFP) expression was observed for up to 14 weeks in the majority of neurons (including nestin-positive cells), motor neurons and oligodendrocytes throughout the CNS, with a significantly lower rate of transduction in astrocytes. Photoreceptors and neuronal cell bodies in the plexiform and ganglionic retinal layers were also transduced. In the peripheral nervous system (PNS), widespread transduction of neurons was observed. Tissues harvested at 14 weeks showed substantially lower vector copy number and GFP levels, although the percentage of GFP-expressing cells remained stable. Thus, AAV9-IUGT in late gestation efficiently transduces both the CNS and PNS with neuronal predilection, of translational relevance to hereditary disorders characterized by perinatal onset of neuropathology.


Assuntos
Córtex Cerebral/metabolismo , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Sistema Nervoso Periférico/metabolismo , Transdução Genética , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Citomegalovirus/genética , Feminino , Feto/metabolismo , Terapia Genética , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Macaca , Oligodendroglia/metabolismo , Gravidez , Retina/metabolismo
6.
Gene Ther ; 19(9): 936-46, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22071970

RESUMO

The efficient delivery of genetic material to the developing fetal brain represents a powerful research tool and a means to supply therapy in a number of neonatal lethal neurological disorders. In this study, we have delivered vectors based upon adenovirus serotype 5 (Ad5) and adeno-associated virus (AAV) pseudotypes 2/5, 2/8 and 2/9 expressing green fluorescent protein to the E16 fetal mouse brain. One month post injection, widespread caudal to rostral transduction of neural cells was observed. In discrete areas of the brain these vectors produced differential transduction patterns. AAV2/8 and 2/9 produced the most extensive gene delivery and had similar transduction profiles. All AAV pseudotypes preferentially transduced neurons whereas Ad5 transduced both neurons and glial cells. None of the vectors elicited any significant microglia-mediated immune response when compared with control uninjected mice. Whole-body imaging and immunohistological evaluation of brains 9 months post injection revealed long-term expression using these non-integrating vectors. These data will be useful in targeting genetic material to discrete or widespread areas of the fetal brain with the purpose of devising therapies for early neonatal lethal neurodegenerative disease and for studying brain development.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Animais , Encéfalo/embriologia , Feminino , Proteínas de Fluorescência Verde/genética , Camundongos , Neuroglia/metabolismo , Neurônios/metabolismo , Transdução Genética
7.
Vet Microbiol ; 157(1-2): 119-24, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22177889

RESUMO

Bluetongue virus serotype 26 (BTV-26) has recently been isolated from sheep in Kuwait. The aim of this study was to assess the pathogenicity and infection kinetics of BTV-26 in Dorset Poll sheep. Six sheep were experimentally infected with BTV-26 and samples taken throughout the study were used to determine the kinetics of infection using a pan specific BTV real time RT-PCR assay and two group specific ELISAs. Five of the six sheep showed mild clinical signs characteristic of bluetongue including conjunctivitis, reddening of the mouth mucosal membranes, slight oedema of the face and nasal discharge. Viral RNA was detected in 5 of the 6 sheep by real time RT-PCR, however the levels of viral RNA detected in the samples were lower and of shorter duration than seen with other field strains of BTV. Virus was isolated from the blood of infected animals at the peak of viraemia at around 9 dpi. Antibodies against BTV were first detected by 7 dpi using the early detection BTV ELISA and a little later (7-14 dpi) using a BTV specific competitive ELISA. Four of the five remaining sheep developed neutralising antibodies to BTV-26, measured by a serum neutralisation test (SNT), with titres (log(10)) ranging from 1.40 to 2.08.


Assuntos
Vírus Bluetongue/patogenicidade , Bluetongue/imunologia , Carneiro Doméstico/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bluetongue/patologia , Bluetongue/virologia , Vírus Bluetongue/genética , Vírus Bluetongue/imunologia , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Kuweit , Masculino , Testes de Neutralização , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Ovinos/imunologia , Ovinos/virologia , Carneiro Doméstico/virologia , Viremia
8.
Gene Ther ; 16(4): 509-20, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158847

RESUMO

Non-integrating lentiviral vectors show considerable promise for gene therapy applications as they persist as long-term episomes in non-dividing cells and diminish risks of insertional mutagenesis. In this study, non-integrating lentiviral vectors were evaluated for their use in the adult and fetal central nervous system of rodents. Vectors differentially pseudotyped with vesicular stomatitis virus, rabies and baculoviral envelope proteins allowed targeting of varied cell populations. Efficient gene delivery to discrete areas of the brain and spinal cord was observed following stereotactic administration. Furthermore, after direct in utero administration (E14), sustained and strong expression was observed 4 months into adulthood. Quantification of transduction and viral copy number was comparable when using non-integrating lentivirus and conventional integrating vector. These data support the use of non-integrating lentiviral vectors as an effective alternative to their integrating counterparts in gene therapy applications, and highlight their potential for treatment of inherited and acquired neurological disorders.


Assuntos
Encéfalo/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Lentivirus/genética , Animais , Corpo Estriado/metabolismo , Terapias Fetais/métodos , Terapia Genética/métodos , Lentivirus/fisiologia , Camundongos , Ratos , Medula Espinal/metabolismo , Técnicas Estereotáxicas , Transdução Genética , Integração Viral
9.
Gene Ther ; 15(24): 1593-605, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18633447

RESUMO

An ideal gene therapy vector should enable persistent transgene expression without limitations of safety and reproducibility. Here we report the development of a non-viral episomal plasmid DNA (pDNA) vector that appears to fulfil these criteria. This pDNA vector combines a scaffold/matrix attachment region (S/MAR) with a human liver-specific promoter (alpha1-antitrypsin (AAT)) in such a way that long-term expression is enabled in murine liver following hydrodynamic injection. Long-term expression is demonstrated by monitoring the longitudinal luciferase expression profile for up to 6 months by means of in situ bioluminescent imaging. All relevant control pDNA constructs expressing luciferase are unable to sustain significant transgene expression beyond 1 week post-administration. We establish that this shutdown of expression is due to promoter methylation. In contrast, the S/MAR element appears to inhibit methylation of the AAT promoter thereby preventing transgene silencing. Although this vector appears to be maintained as an episome throughout, we have no evidence for its establishment as a replicating entity. We conclude that the combination of a mammalian, tissue-specific promoter with the S/MAR element is sufficient to drive long-term episomal pDNA expression of genes in vivo.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Fígado/metabolismo , Regiões de Interação com a Matriz/genética , Plasmídeos/administração & dosagem , alfa 1-Antitripsina/genética , Animais , Metilação de DNA/genética , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Hepatectomia , Humanos , Imuno-Histoquímica , Injeções , Luciferases/análise , Luciferases/genética , Camundongos , Camundongos Endogâmicos , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Fatores de Tempo , Transfecção/métodos , Transgenes
11.
Gene Ther ; 15(16): 1167-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18432275

RESUMO

Gene transfer for cystic fibrosis (CF) airway disease has been hampered by the lung's innate refractivity to pathogen infection. We hypothesized that early intervention with an integrating gene transfer vector capable of transducing the lung via the lumen may be a successful therapeutic approach. An HIV-based lentiviral vector pseudotyped with the baculovirus gp64 envelope was applied to the fetal, neonatal or adult airways. Fetal intra-amniotic administration resulted in transduction of approximately 14% of airway epithelial cells, including both ciliated and non-ciliated epithelia of the upper, mid and lower airways; there was negligible alveolar or nasal transduction. Following neonatal intra-nasal administration we observed significant transduction of the airway epithelium (approximately 11%), although mainly in the distal lung, and substantial alveolar transduction. This expression was still detectable at 1 year after application. In the adult, the majority of transduction was restricted to the alveoli. In contrast, vesicular stomatitis virus glycoprotein pseudotyped virus transduced only alveoli after adult and neonatal application and no transduction was observed after fetal administration. Repeat administration did not increase transduction levels of the conducting airway epithelia. These data demonstrate that application at early developmental stages in conjunction with an appropriately pseudotyped virus provides efficient, high-level transgene expression in the murine lung. This may provide a modality for treatment for lung disease in CF.


Assuntos
Baculoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , HIV/genética , Transdução Genética/métodos , Proteínas do Envelope Viral/genética , Animais , Animais Recém-Nascidos , Fibrose Cística/metabolismo , Fibrose Cística/terapia , Feminino , Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Luciferases/análise , Luciferases/genética , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Animais , Tempo , Transgenes
12.
Mol Ther ; 16(5): 819-24, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18388933

RESUMO

Due to its early onset and severe prognosis, cystic fibrosis (CF) has been suggested as a candidate disease for in utero gene therapy. In 1997, a study was published claiming that to how transient prenatal expression of CF transmembrane conductance regulator (CFTR) from an in utero-injected adenovirus vector could achieve permanent reversal of the CF intestinal pathology in adult CF knockout mice, despite the loss of CFTR transgene expression by birth. This would imply that the underlying cause of CF is a prenatal defect for which lifelong cure can be achieved by transient prenatal expression of CFTR. Despite criticism at the time of publication, no independent verification of this contentious finding has been published so far. This is vital for the development of future therapeutic strategies as it may determine whether CF gene therapy should be performed prenatally or postnatally. We therefore reinvestigated this finding with an identical adenoviral vector and a knockout CF mouse line (Cftr(tmlCam)) with a completely inbred genetic background to eliminate any effects due to genetic variation. After delivery of the CFTR-expressing adenovirus to the fetal mouse, both vector DNA and transgenic CFTR expression were detected in treated animals postpartum but statistically no significant difference in survival was observed between the Cftr(-/-) mice treated with the CFTR-adenovirus and those treated with the control vector.


Assuntos
Adenoviridae/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Fibrose Cística/terapia , Regulação da Expressão Gênica , Terapia Genética/métodos , Líquido Amniótico/metabolismo , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Gravidez , Prenhez , Reprodutibilidade dos Testes
13.
Gene Ther ; 13(23): 1665-76, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16871230

RESUMO

Prenatal gene therapy has been considered for Herlitz junctional epidermolysis bullosa (H-JEB), a lethal genodermatosis caused by the absence of any of the three subunits of laminin-5, resulting from birth in widespread blistering and erosions of skin and mucosae. To investigate this strategy in an animal model, adenovirus type 5- and adeno-associated virus (AAV) type 2-derived vectors carrying a beta-galactosidase reporter gene or LAMB3 cDNA encoding the beta3 chain of laminin-5 were generated, tested for stability in amniotic fluid and evaluated in vitro on murine H-JEB keratinocytes, and in vivo by prenatal injection into the amniotic cavities of laminin-5 beta3-deficient mice. The different vectors were administered individually or combined at maximum doses on day 14 post coitum. Adenoviral vectors infected preferentially the foetal epidermis, whereas AAV delivered the transgene mainly to mucous membranes of the airways and the upper digestive tract. The LAMB3 transgene was expressed in target epithelia of newborn laminin-5 beta3-deficient mice, and the transgenic beta3 chain was shown to assemble with its endogenous partner chains, resulting in detectable amounts of laminin-5 in the basement membranes of skin and mucosae and in a lower extent of tissue separation in the skin. However, only combined delivery of the two vector types led to a minor increase of the life span of H-JEB mice. Failure to rescue diseased animals was, at least in part, due to abandonment of any conspicuous pup by the heterozygous mother. This is the first study of a prenatal gene therapy approach to a heritable blistering disorder. Although our findings indicate that prenatal combined administration of adenoviral and adeno-associated LAMB3 vectors provides therapeutic benefit to H-JEB mice, this animal model appears unsuitable for long-term investigations of the therapeutic concept.


Assuntos
Moléculas de Adesão Celular/genética , Epidermólise Bolhosa Juncional/embriologia , Epidermólise Bolhosa Juncional/prevenção & controle , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Transdução Genética/métodos , Adenoviridae/genética , Âmnio , Animais , Animais Recém-Nascidos , Membrana Basal/química , Membrana Basal/metabolismo , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/metabolismo , Dependovirus/genética , Epidermólise Bolhosa Juncional/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/genética , Injeções , Camundongos , Camundongos Knockout , Modelos Animais , Gravidez , Pele/metabolismo , Transgenes , Calinina
14.
Hum Gene Ther ; 17(7): 767-79, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16839275

RESUMO

Targeting gene therapy vectors to the fetal intestinal tract could provide a novel means toward prevention of the early postnatal intestinal pathology of cystic fibrosis and other conditions, such as congenital enteropathy, that cause intestinal failure. Among these conditions, cystic fibrosis is by far the most common lethal genetic disease. It is caused by a functional absence or deficiency of the cystic fibrosis transmembrane conductance regulator and manifests in the gut as meconium ileus. Prenatal treatment of genetic disease may avoid early-onset tissue damage and immune sensitization, and may target cells that are less accessible in the adult. We investigated gene transfer to the fetal gut, using a minimally invasive injection technique. First-generation replication-deficient adenoviral vectors encoding the beta-galactosidase gene and transduction-enhancing agents were injected into the stomach of early-gestation fetal sheep (n = 8, 60 days of gestation; term, 145 days) under ultrasound guidance. Reporter gene expression was observed 2 days after injection in the villi of the gastrointestinal epithelia after 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside staining and beta-galactosidase immunohistochemistry of fetal tissues. Expression of beta-galactosidase, as measured by enzyme-linked immunosorbent assay, was enhanced after pretreatment of the fetal gut with sodium caprate, which opens tight junctions, and after adenovirus complexation with DEAE-dextran, which confers a positive charge to the virus. Instillation of the fluorocarbon perflubron after virus delivery resulted in tissue transduction from the fetal stomach to the colon. Using a clinically relevant technique, we have demonstrated widespread gene transfer to the fetal gastrointestinal epithelia.


Assuntos
Fibrose Cística/prevenção & controle , Fetoscopia/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Enteropatias/prevenção & controle , Mucosa Intestinal/metabolismo , Adenoviridae/genética , Animais , Feminino , Feto/metabolismo , Mucosa Gástrica/metabolismo , Genes Reporter , Vetores Genéticos/genética , Intestinos/embriologia , Intestinos/enzimologia , Ovinos , Estômago/enzimologia , Distribuição Tecidual , beta-Galactosidase/análise , beta-Galactosidase/genética
15.
Gene Ther ; 13(2): 117-26, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16163377

RESUMO

Immune responses against an introduced transgenic protein are a potential risk in many gene replacement strategies to treat genetic disease. We have developed a gene delivery approach for hemophilia B based on lentiviral expression of human factor IX in purified hematopoietic stem cells. In both normal C57Bl/6J and hemophilic 129/Sv recipient mice, we observed the production of therapeutic levels of human factor IX, persisting for at least a year with tolerance to human factor IX antigen. Secondary and tertiary recipients also demonstrate long-term production of therapeutic levels of human factor IX and tolerance, even at very low levels of donor chimerism. Furthermore, in hemophilic mice, partial functional correction of treated mice and phenotypic rescue is achieved. These data show the potential of a stem cell approach to gene delivery to tolerize recipients to a secreted foreign transgenic protein and, with appropriate modification, may be of use in developing treatments for other genetic disorders.


Assuntos
Fator IX/genética , Terapia Genética/métodos , HIV-1/genética , Hemofilia B/terapia , Transplante de Células-Tronco/métodos , Animais , Antígenos/imunologia , Células Cultivadas , Fator IX/metabolismo , Expressão Gênica , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Genótipo , Proteínas de Fluorescência Verde/genética , Hemofilia B/sangue , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/metabolismo , Células-Tronco/virologia , Fatores de Tempo , Transdução Genética/métodos , Transgenes
16.
Gene Ther ; 12(22): 1601-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16136161

RESUMO

Somatic gene delivery in utero is a novel approach to gene therapy for genetic disease based on the hypothesis that prenatal intervention may avoid the development of severe manifestations of early-onset disease, allow targeting of otherwise inaccessible tissues including expanding stem cell populations, induce tolerance against the therapeutic transgenic protein and thereby provide permanent somatic gene correction. This approach is particularly relevant in relation to prenatal screening programmes for severe genetic diseases as it could offer prevention as a third option to families faced with the prenatal diagnosis of a genetically affected child. Most investigations towards in utero gene therapy have been performed on mice and sheep fetuses as model animals for human disease and for the application of clinically relevant intervention techniques such as vector delivery by minimally invasive ultrasound guidance. Other animals such as dogs may serve as particular disease models and primates have to be considered in immediate preparation for clinical trials. Proof of principle for the hypothesis of fetal gene therapy has been provided during the last 2 years in mouse models for Crigler Najjar Disease, Leber's congenital amaurosis, Pompe's disease and haemophilia B showing long-term postnatal therapeutic effects and tolerance of the transgenic protein after in utero gene delivery. However, recently we have also observed a high incidence of liver tumours after in utero application of an early form of third-generation equine infectious anaemia virus vectors with SIN configuration. These findings highlight the need for more investigations into the safety and the ethical aspects of in utero gene therapy as well as for science-based public information on risks and benefits of this preventive gene therapy approach before application in humans can be contemplated.


Assuntos
Doenças Fetais/terapia , Doenças Genéticas Inatas/terapia , Terapia Genética/métodos , Animais , Feminino , Doenças Fetais/genética , Previsões , Doenças Genéticas Inatas/embriologia , Doenças Genéticas Inatas/genética , Terapia Genética/efeitos adversos , Terapia Genética/tendências , Neoplasias Hepáticas/etiologia , Camundongos , Modelos Animais , Gravidez , Primatas , Projetos de Pesquisa , Ovinos , Transgenes
17.
Mol Ther ; 12(3): 484-92, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16099411

RESUMO

Intra-amniotic injection of adenovirus allows transduction of the fetal airways following natural fetal breathing movements. This administration method is promising for use in gene therapy for cystic fibrosis and other diseases for which the main target for exogenous gene expression is the lung. Here we have investigated factors that may affect the efficacy of gene transfer to the murine fetal lung. We examined marker compound distribution and transgene expression (from a first-generation adenoviral vector) at different stages of development. This demonstrated that fetal breathing movements at 15-16 days of gestation are of sufficient intensity to carry marker/vector into the fetal lungs. These movements can be significantly stimulated by the combination of intra-amniotic theophylline administration and postoperative exposure of the dam to elevated CO(2) levels. However, the most important factor for efficient and consistent pulmonary transgene delivery is the dose of adenoviral vector used, as both the degree of transduction and the percentage of lungs transduced increases with escalating viral dose.


Assuntos
Adenoviridae/genética , Vetores Genéticos , Pulmão/embriologia , Traqueia/embriologia , Adenosina/metabolismo , Animais , Carbono/química , Dióxido de Carbono/metabolismo , Coloides/química , Fibrose Cística/terapia , Ensaio de Imunoadsorção Enzimática , Técnicas de Transferência de Genes , Genes Reporter , Camundongos , Teofilina/administração & dosagem , Fatores de Tempo , Transgenes
18.
Gene Ther ; 11 Suppl 1: S92-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15454963

RESUMO

The current approaches to gene therapy of monogenetic diseases into mature organisms are confronted with several problems including the following: (1) the underlying genetic defect may have already caused irreversible pathological changes; (2) the level of sufficient protein expression to ameliorate or prevent the disease requires prohibitively large amounts of gene delivery vector; (3) adult tissues may be poorly infected by conventional vector systems dependent upon cellular proliferation for optimal infection, for example, oncoretrovirus vectors; (4) immune responses, either pre-existing or developing following vector delivery, may rapidly eliminate transgenic protein expression and prevent future effective intervention. Early gene transfer, in the neonatal or even fetal period, may overcome some or all of these obstacles. The mammalian fetus enjoys a uniquely protected environment in the womb, bathed in a biochemically and physically supportive fluid devoid of myriad extra-uterine pathogens. Strong physical and chemical barriers to infection might, perhaps, impede the frenetic cell division. The physical support and the biochemical support provided by the fetal-maternal placental interface may, therefore, minimize the onset of genetic diseases manifest early in life. The fetal organism must prepare itself for birth, but lacking a mature adaptive immune system may depend upon more primordial immune defences. It is the nature of these defences, and the vulnerabilities they protect, that are poorly understood in the context of gene therapy and might provide useful information for approaches to gene therapy in the young, as well as perhaps the mature organism.


Assuntos
Doenças Fetais/terapia , Doenças Genéticas Inatas/terapia , Terapia Genética/métodos , Animais , Doenças Fetais/imunologia , Marcação de Genes , Técnicas de Transferência de Genes , Doenças Genéticas Inatas/imunologia , Terapia Genética/efeitos adversos , Humanos , Sistema Imunitário/fisiologia , Recém-Nascido
19.
Br J Radiol ; 77(919): 557-61, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15238401

RESUMO

The requirement of the Ionising Radiation (Medical Exposure) Regulation 2000 [IR(ME)R] of justifying all exposures to ionizing radiation includes those from radiotherapy double exposure portal images resulting in exposure to normal tissues outside the treatment volume. Typical effective doses were calculated for a range of common sites using CT data to outline those parts of specific organs subject to concomitant radiation and generate dose-volume histograms. The product of the mean dose and the relative probability of inducing a fatal cancer in specific organs was used to determine a representative total effective dose in mSv per monitor unit for each site. A table of representative effective doses, ranging from 0.32 mSv to 2.56 mSv per monitor unit, was produced, which may be used to monitor cumulative effective doses of individual patients from double exposure portal images, in addition to those received from localization procedures.


Assuntos
Neoplasias Induzidas por Radiação/etiologia , Segunda Neoplasia Primária/etiologia , Doses de Radiação , Radioterapia Conformacional/efeitos adversos , Feminino , Humanos , Masculino , Neoplasias da Próstata/radioterapia , Medição de Risco/métodos , Tomografia Computadorizada por Raios X
20.
Gene Ther ; 11(14): 1117-25, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15141156

RESUMO

Gene therapy for Duchenne muscular dystrophy has so far not been successful because of the difficulty in achieving efficient and permanent gene transfer to the large number of affected muscles and the development of immune reactions against vector and transgenic protein. In addition, the prenatal onset of disease complicates postnatal gene therapy. We have therefore proposed a fetal approach to overcome these barriers. We have applied beta-galactosidase expressing equine infectious anaemia virus (EIAV) lentiviruses pseudotyped with VSV-G by single or combined injection via different routes to the MF1 mouse fetus on day 15 of gestation and describe substantial gene delivery to the musculature. Highly efficient gene transfer to skeletal muscles, including the diaphragm and intercostal muscles, as well as to cardiac myocytes was observed and gene expression persisted for at least 15 months after administration of this integrating vector. These findings support the concept of in utero gene delivery for therapeutic and long-term prevention/correction of muscular dystrophies and pave the way for a future application in the clinic.


Assuntos
Feto/metabolismo , Terapia Genética/métodos , Vírus da Anemia Infecciosa Equina/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , beta-Galactosidase/genética , Animais , Feminino , Feto/imunologia , Expressão Gênica , Engenharia Genética , Injeções , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/embriologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...